51
|
The developmental journey of therapies targeting purine receptors: from basic science to clinical trials. Purinergic Signal 2022; 18:435-450. [PMID: 36173587 PMCID: PMC9832190 DOI: 10.1007/s11302-022-09896-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/21/2022] [Indexed: 01/14/2023] Open
Abstract
Since the discovery of ATP as an extracellular signalling molecule in 1972, purinergic signalling, mediated by extracellular purines and pyrimidines has been identified in virtually all mammalian tissues and is implicated in regulating fundamental cellular processes. In recent years, there has been an increasing focus on the pathophysiology and potential therapeutic interventions based on purinergic signalling. A vast range of compounds targeting purine receptors are in clinical development, and many more are in preclinical studies, which highlights the fast growth in this research field. As a tribute to Professor Geoffrey Burnstock's legacy in purinergic signalling, we present here a brief review of compounds targeting purine receptors that are in different stages of clinical trials. The review highlights the 50-year journey from basic research on purinergic receptors to clinical applications of therapies targeting purine receptors.
Collapse
|
52
|
Dangelmaier C, Kunapuli SP. Evidence for a PI3-kinase independent pathway in the regulation of Rap1b activation downstream of the P2Y12 receptor in platelets. Platelets 2022; 33:1301-1306. [PMID: 35514261 PMCID: PMC9547944 DOI: 10.1080/09537104.2022.2071855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
Platelet activation by adenosine diphosphate (ADP) is mediated through two G-protein-coupled receptors, P2Y1 and P2Y12, which signal through Gq and Gi, respectively. P2Y1 stimulation leads to phospholipase C activation and an increase in cytosolic calcium necessary for CalDAG-GEF1 activation. Engagement of P2Y12 inhibits adenylate cyclase, which reduces cAMP, and activation of PI3-kinase, which inhibits RASA3 resulting in sustained activated Rap1b. In this study we activated human platelets with 2-MeSADP in the presence of LY294002, a PI3-kinase inhibitor, AR-C69931MX, a P2Y12 antagonist or MRS2179, a P2Y1 antagonist. We measured the phosphorylation of Akt on Ser473 as an indicator of PI3-kinase activity. As previously shown, LY294002 and ARC69931MX abolished 2MeSADP-induced Akt phosphorylation. MRS2179 reduced ADP-induced Akt phosphorylation but did not abolish it. Rap1b activation, however, was only reduced, but not ablated, using LY294002 and was completely inhibited by ARC69931MX or MRS2179. Furthermore, 2MeSADP-induced Rap1b activation was abolished in either P2Y1 or P2Y12 null platelets. These data suggest that ADP-induced Rap1b activation requires both P2Y1 and P2Y12. In addition, although stimulation of P2Y12 results in PI3-kinase activation leading to Akt phosphorylation and Rap1b activation, Rap1b activation can occur independently of PI3-kinase downstream of P2Y12. Thus, we propose that the P2Y12 receptor can regulate Rap1b, possibly through RASA3, in a pathway independent of PI3-kinase.
Collapse
Affiliation(s)
- Carol Dangelmaier
- Sol Sherry Thrombosis Research Center and the Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and the Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
53
|
Ksenofontov AA, Bocharov PS, Antina EV, Shevchenko OG, Samorodov AV, Gilfanov IR, Pavelyev RS, Ostolopovskaya OV, Startseva VA, Fedyunina IV, Azizova ZR, Gaysin SI, Pestova SV, Izmest’ev ES, Rubtsova SA, Khelkhal MA, Nikitina LE. Thioterpenoids as Potential Antithrombotic Drugs: Molecular Docking, Antiaggregant, Anticoagulant and Antioxidant Activities. Biomolecules 2022; 12:1599. [PMID: 36358949 PMCID: PMC9687754 DOI: 10.3390/biom12111599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 01/09/2024] Open
Abstract
Natural monoterpenes and their derivatives are widely considered as effective ingredients for the design and production of new biologically active compounds with high antioxidant, antimicrobial and anti-protozoa properties. In this study, we synthesized two series of thiotherpenoids "sulfide-sulfoxide-sulfone", with different bicyclic monoterpene skeleton (bornane and pinane) structures. The effect of the obtained compounds on platelet aggregation was investigated by using the molecular docking technique. The obtained data revealed that all the synthesized compounds may act as potential inhibitors of platelet aggregation. Moreover, the studied sulfides have shown high antioxidant activity as revealed by lipid peroxidation (LPO) process inhibition in a non-cellular substrate containing animal lipids. The sulfides were able to inhibit erythrocyte oxidative hemolysis, to reduce the accumulation of secondary LPO products in cells and to prevent the oxidation of native oxyhemoglobin. Additionally, the corresponding sulfones and sulfoxides exhibited insignificant antioxidant activity. However, the sulfides were found to exhibit significant antiaggregant and anticoagulant effects. These findings suggest as well that the sulfides could serve as a leader compound for future research and possible practical applications.
Collapse
Affiliation(s)
- Alexander A. Ksenofontov
- G.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 1 Akademicheskaya Street, 153045 Ivanovo, Russia
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Pavel S. Bocharov
- G.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 1 Akademicheskaya Street, 153045 Ivanovo, Russia
| | - Elena V. Antina
- G.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 1 Akademicheskaya Street, 153045 Ivanovo, Russia
| | - Oksana G. Shevchenko
- Center of Collective Usage «Molecular Biology», Institute of Biology, Komi Science Centre, Ural Branch of Russian Academy of Sciences, 28 Kommunisticheskaya Street, 167982 Syktyvkar, Russia
| | - Aleksandr V. Samorodov
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- Department of Pharmacology, Bashkir State Medical University, 3 Lenina str., 450008 Ufa, Russia
| | - Ilmir R. Gilfanov
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- Varnishes and Paints Department, Kazan National Research Technological University, 68 K. Marksa Street, 420015 Kazan, Russia
| | - Roman S. Pavelyev
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Olga V. Ostolopovskaya
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- General and Organic Chemistry Department, Kazan State Medical University, 49 Butlerova Street, 420012 Kazan, Russia
| | - Valeriya A. Startseva
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- General and Organic Chemistry Department, Kazan State Medical University, 49 Butlerova Street, 420012 Kazan, Russia
| | - Inna V. Fedyunina
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- General and Organic Chemistry Department, Kazan State Medical University, 49 Butlerova Street, 420012 Kazan, Russia
| | - Zulfiya R. Azizova
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Salavat I. Gaysin
- General and Organic Chemistry Department, Kazan State Medical University, 49 Butlerova Street, 420012 Kazan, Russia
| | - Svetlana V. Pestova
- Medical Chemistry Laboratory, Institute of Chemistry, Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 48 Pervomaiskaya Street, 167000 Syktyvkar, Russia
| | - Evgeniy S. Izmest’ev
- Medical Chemistry Laboratory, Institute of Chemistry, Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 48 Pervomaiskaya Street, 167000 Syktyvkar, Russia
| | - Svetlana A. Rubtsova
- Medical Chemistry Laboratory, Institute of Chemistry, Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 48 Pervomaiskaya Street, 167000 Syktyvkar, Russia
| | - Mohammed A. Khelkhal
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
| | - Liliya E. Nikitina
- Biologically Active Terpenoids Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Street, 420008 Kazan, Russia
- General and Organic Chemistry Department, Kazan State Medical University, 49 Butlerova Street, 420012 Kazan, Russia
| |
Collapse
|
54
|
Tang S, Wu Z, Xu L, Wen Q, Zhang X. Adverse reaction signals mining and hemorrhagic signals comparison of ticagrelor and clopidogrel: A pharmacovigilance study based on FAERS. Front Pharmacol 2022; 13:970066. [PMID: 36263117 PMCID: PMC9573960 DOI: 10.3389/fphar.2022.970066] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Ticagrelor and clopidogrel are commonly used antiplatelet agents, and we conducted a pharmacovigilance analysis using the Food and Drug Administration Adverse Event Reporting System (FAERS) to provide a reference for safe and reasonable clinical use.Methods: Data were collected in FAERS from 2012 Q1 to 2022 Q2 for data cleaning. We used system organ classes (SOCs) and prefer terms (PTs) from the Medical Dictionary of Regulatory Activity (MedDRA version 25.1). Adverse event reports were retrieved at the PT level. Adverse reaction (ADR) signals of ticagrelor and clopidogrel were mined by calculating reporting odds ratios (ROR), proportional reporting ratios (PRR), information component (IC) and empirical Bayesian geometric mean (EBGM). After that, further analysis of the hemorrhagic signals and their clinical information were performed.Results: The number of ADR reports where the primary suspect (PS) drugs were 15,133 for ticagrelor and 23,860 for clopidogrel. Significant ADR signals were identified by the SOC analysis for ticagrelor including cardiac disorders (ROR 4.87, PRR 4.46), respiratory disorders (ROR 2.45, PRR 2.28), and vascular disorders (ROR 2.22, PRR 2.16). Clopidogrel included blood disorders (ROR 2.86, PRR 2.77), vascular disorders (ROR 2.71, PRR 2.61), and cardiac disorders (ROR 2.29, PRR 2.22). At the PT level, the more frequent ADR signals for ticagrelor were dyspnoea, contusion, and haemorrhage, while clopidogrel were gastrointestinal haemorrhage, anaemia, and drug interaction. The hemorrhagic signals of both were mainly focused on the SOC level of gastrointestinal disorders, injury disorders and vascular disorders and nervous system disorders. The death and life-threatening rate of ticagrelor was 7.76 percentage higher than that of clopidogrel.Conclusion: Clinicians need to pay attention to not only common ADRs but also be alert to new ADR signals when choosing to use ticagrelor and clopidogrel. This study provides a reference for the reasonable and safe clinical use of ticagrelor and clopidogrel.
Collapse
Affiliation(s)
- Shu Tang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shu Tang, ; Xiaojian Zhang,
| | - Zhanshen Wu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liqing Xu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shu Tang, ; Xiaojian Zhang,
| |
Collapse
|
55
|
Cao X, Du X, Jiao H, An Q, Chen R, Fang P, Wang J, Yu B. Carbohydrate-based drugs launched during 2000 -2021. Acta Pharm Sin B 2022; 12:3783-3821. [PMID: 36213536 PMCID: PMC9532563 DOI: 10.1016/j.apsb.2022.05.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/18/2022] [Accepted: 05/12/2022] [Indexed: 01/09/2023] Open
Abstract
Carbohydrates are fundamental molecules involved in nearly all aspects of lives, such as being involved in formating the genetic and energy materials, supporting the structure of organisms, constituting invasion and host defense systems, and forming antibiotics secondary metabolites. The naturally occurring carbohydrates and their derivatives have been extensively studied as therapeutic agents for the treatment of various diseases. During 2000 to 2021, totally 54 carbohydrate-based drugs which contain carbohydrate moities as the major structural units have been approved as drugs or diagnostic agents. Here we provide a comprehensive review on the chemical structures, activities, and clinical trial results of these carbohydrate-based drugs, which are categorized by their indications into antiviral drugs, antibacterial/antiparasitic drugs, anticancer drugs, antidiabetics drugs, cardiovascular drugs, nervous system drugs, and other agents.
Collapse
Affiliation(s)
- Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Xiaojing Du
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Heng Jiao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Quanlin An
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Ruoxue Chen
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Pengfei Fang
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jing Wang
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
56
|
Chen J, Tang Y, Zhong Y, Wei B, Huang XR, Tang PMK, Xu A, Lan HY. P2Y12 inhibitor clopidogrel inhibits renal fibrosis by blocking macrophage-to-myofibroblast transition. Mol Ther 2022; 30:3017-3033. [PMID: 35791881 PMCID: PMC9481993 DOI: 10.1016/j.ymthe.2022.06.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Clopidogrel, a P2Y12 inhibitor, is a novel anti-fibrosis agent for chronic kidney disease (CKD), but its mechanisms remain unclear, which we investigated by silencing P2Y12 or treating unilateral ureteral obstruction (UUO) in LysM-Cre/Rosa Tomato mice with clopidogrel in vivo and in vitro. We found that P2Y12 was significantly increased and correlated with progressive renal fibrosis in CKD patients and UUO mice. Phenotypically, up to 82% of P2Y12-expressing cells within the fibrosing kidney were of macrophage origin, identified by co-expressing CD68/F4/80 antigens or a macrophage-lineage-tracing marker Tomato. Unexpectedly, more than 90% of P2Y12-expressing macrophages were undergoing macrophage-to-myofibroblast transition (MMT) by co-expressing alpha smooth muscle actin (α-SMA), which was also confirmed by single-cell RNA sequencing. Functionally, clopidogrel improved the decline rate of the estimated glomerular filtration rate (eGFR) in patients with CKD and significantly inhibited renal fibrosis in UUO mice. Mechanistically, P2Y12 expression was induced by transforming growth factor β1 (TGF-β1) and promoted MMT via the Smad3-dependent mechanism. Thus, silencing or pharmacological inhibition of P2Y12 was capable of inhibiting TGF-β/Smad3-mediated MMT and progressive renal fibrosis in vivo and in vitro. In conclusion, P2Y12 is highly expressed by macrophages in fibrosing kidneys and mediates renal fibrosis by promoting MMT via TGF-β/Smad3 signaling. Thus, P2Y12 inhibitor maybe a novel and effective anti-fibrosis agent for CKD.
Collapse
Affiliation(s)
- Junzhe Chen
- Department of Nephrology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China; Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ying Tang
- Department of Nephrology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yu Zhong
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Biao Wei
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Ru Huang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Patrick Ming-Kuen Tang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.
| | - Anping Xu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Hui-Yao Lan
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China.
| |
Collapse
|
57
|
Ma BB, Montgomery AP, Chen B, Kassiou M, Danon JJ. Strategies for targeting the P2Y 12 receptor in the central nervous system. Bioorg Med Chem Lett 2022; 71:128837. [PMID: 35640763 DOI: 10.1016/j.bmcl.2022.128837] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 11/28/2022]
Abstract
The purinergic 2Y type 12 receptor (P2Y12R) is a well-known biological target for anti-thrombotic drugs due to its role in platelet aggregation and blood clotting. While the importance of the P2Y12R in the periphery has been known for decades, much less is known about its expression and roles in the central nervous system (CNS), where it is expressed exclusively on microglia - the first responders to brain insults and neurodegeneration. Several seminal studies have shown that P2Y12 is a robust, translatable biomarker for anti-inflammatory and neuroprotective microglial phenotypes in models of degenerative diseases such as multiple sclerosis and Alzheimer's disease. An enduring problem for studying this receptor in vivo, however, is the lack of selective, high-affinity small molecule ligands that can bypass the blood-brain barrier and accumulate in the CNS. In this Digest, we discuss previous attempts by researchers to target the P2Y12R in the CNS and opine on strategies that may be employed to design and assess the suitability of novel P2Y12 ligands for this purpose going forward.
Collapse
Affiliation(s)
- Ben B Ma
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Biling Chen
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan J Danon
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
58
|
Sykes DL, Zhang M, Morice AH. Treatment of chronic cough: P2X3 receptor antagonists and beyond. Pharmacol Ther 2022; 237:108166. [DOI: 10.1016/j.pharmthera.2022.108166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
|
59
|
Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15091039. [PMID: 36145259 PMCID: PMC9505853 DOI: 10.3390/ph15091039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing environmental distress is associated with a growing asthma incidence; no treatments are available but montelukast (MTK)—an antagonist of the cysteinyl leukotrienes receptor 1—is widely used in the management of symptoms among adults and children. Recently, new molecular targets have been identified and MTK has been proposed for repurposing in other therapeutic applications, with several ongoing clinical trials. The proposed applications include neuroinflammation control, which could be explored in some neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases (AD and PD). However, this drug has been associated with an increasing number of reported neuropsychiatric adverse drug reactions (ADRs). Besides, and despite being on the market since 1998, MTK metabolism is still poorly understood and the mechanisms underlying neuropsychiatric ADRs remain unknown. We review the role of MTK as a modulator of leukotriene pathways and systematize the current knowledge about MTK metabolism. Known toxic effects of MTK are discussed, and repurposing applications are presented comprehensively, with a focus on AD and PD.
Collapse
Affiliation(s)
- Cátia F. Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Maria Matilde Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Gonçalo C. Justino
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
60
|
Zhao X, Cooper M, Michael JV, Yarman Y, Baltz A, Chuprun JK, Koch WJ, McKenzie SE, Tomaiuolo M, Stalker TJ, Zhu L, Ma P. GRK2 regulates ADP signaling in platelets via P2Y1 and P2Y12. Blood Adv 2022; 6:4524-4536. [PMID: 35793439 PMCID: PMC9636328 DOI: 10.1182/bloodadvances.2022007007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/10/2022] [Indexed: 11/20/2022] Open
Abstract
The critical role of G protein-coupled receptor kinase 2 (GRK2) in regulating cardiac function has been well documented for >3 decades. Targeting GRK2 has therefore been extensively studied as a novel approach to treating cardiovascular disease. However, little is known about its role in hemostasis and thrombosis. We provide here the first evidence that GRK2 limits platelet activation and regulates the hemostatic response to injury. Deletion of GRK2 in mouse platelets causes increased platelet accumulation after laser-induced injury in the cremaster muscle arterioles, shortens tail bleeding time, and enhances thrombosis in adenosine 5'-diphosphate (ADP)-induced pulmonary thromboembolism and in FeCl3-induced carotid injury. GRK2-/- platelets have increased integrin activation, P-selectin exposure, and platelet aggregation in response to ADP stimulation. Furthermore, GRK2-/- platelets retain the ability to aggregate in response to ADP restimulation, indicating that GRK2 contributes to ADP receptor desensitization. Underlying these changes in GRK2-/- platelets is an increase in Ca2+ mobilization, RAS-related protein 1 activation, and Akt phosphorylation stimulated by ADP, as well as an attenuated rise of cyclic adenosine monophosphate levels in response to ADP in the presence of prostaglandin I2. P2Y12 antagonist treatment eliminates the phenotypic difference in platelet accumulation between wild-type and GRK2-/- mice at the site of injury. Pharmacologic inhibition of GRK2 activity in human platelets increases platelet activation in response to ADP. Finally, we show that GRK2 binds to endogenous Gβγ subunits during platelet activation. Collectively, these results show that GRK2 regulates ADP signaling via P2Y1 and P2Y12, interacts with Gβγ, and functions as a signaling hub in platelets for modulating the hemostatic response to injury.
Collapse
Affiliation(s)
- Xuefei Zhao
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Matthew Cooper
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - James V. Michael
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Yanki Yarman
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Aiden Baltz
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - J. Kurt Chuprun
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Walter J. Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Steven E. McKenzie
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Maurizio Tomaiuolo
- Vickie and Jack Farber Vision Research Center, Wills Eye Hospital, Philadelphia, PA
| | - Timothy J. Stalker
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Li Zhu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
61
|
Shen CL, Wu YF. Flow cytometry for evaluating platelet immunophenotyping and function in patients with thrombocytopenia. Tzu Chi Med J 2022; 34:381-387. [PMID: 36578648 PMCID: PMC9791859 DOI: 10.4103/tcmj.tcmj_117_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/10/2022] [Accepted: 06/08/2022] [Indexed: 01/19/2023] Open
Abstract
Platelets play an essential role in primary hemostasis through bleeding and thromboembolism. Thus, the diagnosis or evaluation of impaired hereditary, acquired, and drug-related platelet dysfunction has become imperative. The assessment of the platelet function is too complex for routine platelet function study. The major methods involved in platelet function study include platelet function analyzer testing, thromboelastography, thromboelastometry, light transmission aggregometry, and flow cytometry. The current review article focuses on the methods with flow cytometry for immunophenotyping of platelet and evaluating platelet function for platelet disorders, especially in patients with thrombocytopenia. According to the consensus published by the International Society on Thrombosis and Haemostasis, for inherited and acquired platelet disorders, the two major measures by which flow cytometry determines platelet function are glycoprotein IIb/IIIa/P-selectin (CD62p) expression and percentage of leukocyte-platelet aggregates. Using flow cytometry to determine platelet function has several advantages, including good sensitivity to low platelet counts, small blood volume required, and the nonnecessity of centrifugation. However, flow cytometry has still many limitations and challenges, with standardization for routine laboratory testing also proving difficult. Although flow cytometry is available for multipurpose and sensitive study of platelet functions at the same time, the challenging analysis gradually increases and needs to be addressed before reality.
Collapse
Affiliation(s)
- Chih-Lung Shen
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yi-Feng Wu
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Yi-Feng Wu, Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Section 3, Chung-Yang Road, Hualien, Taiwan. E-mail:
| |
Collapse
|
62
|
Kuszynski DS, Lauver DA. Pleiotropic effects of clopidogrel. Purinergic Signal 2022; 18:253-265. [PMID: 35678974 DOI: 10.1007/s11302-022-09876-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/26/2022] [Indexed: 01/04/2023] Open
Abstract
Clopidogrel is a widely prescribed prodrug with anti-thrombotic activity through irreversible inhibition of the P2Y12 receptor on platelets. It is FDA-approved for the clinical management of thrombotic diseases like unstable angina, myocardial infarction, stroke, and during percutaneous coronary interventions. Hepatic clopidogrel metabolism generates several distinct metabolites. Only one of these metabolites is responsible for inhibiting the platelet P2Y12 receptor. Importantly, various non-hemostatic effects of clopidogrel therapy have been described. These non-hemostatic effects are perhaps unsurprising, as P2Y12 receptor expression has been reported in multiple tissues, including osteoblasts, leukocytes, as well as vascular endothelium and smooth muscle. While the "inactive" metabolites have been commonly thought to be biologically inert, recent findings have uncovered P2Y12 receptor-independent effects of clopidogrel treatment that may be mediated by understudied metabolites. In this review, we summarize both the P2Y12 receptor-mediated and non-P2Y12 receptor-mediated effects of clopidogrel and its metabolites in various tissues.
Collapse
Affiliation(s)
- Dawn S Kuszynski
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, 1355 Bogue Street, B336 Life Science, East Lansing, MI, USA.,Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - D Adam Lauver
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, 1355 Bogue Street, B336 Life Science, East Lansing, MI, USA.
| |
Collapse
|
63
|
Zhou J, Zhang H, Fareed MS, He Y, Lu Y, Yang C, Wang Z, Su J, Wang P, Yan W, Wang K. An Injectable Peptide Hydrogel Constructed of Natural Antimicrobial Peptide J-1 and ADP Shows Anti-Infection, Hemostasis, and Antiadhesion Efficacy. ACS NANO 2022; 16:7636-7650. [PMID: 35533290 DOI: 10.1021/acsnano.1c11206] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Postoperative adhesion is a common complication of abdominal surgery, which always has many adverse effects in patients. At present, there is still a lack of effective treatment measures and materials to prevent adhesion in the clinics. Herein, we report the potential use of J-1-ADP hydrogel formed by natural antimicrobial peptide jelleine-1 (J-1) self-assembling in adenosine diphosphate (ADP) sodium solution to prevent postsurgery adhesion formation. J-1-ADP hydrogel was found to have good antimicrobial activity against the bacteria and fungi tested and can be used to prevent tissue infection, which was thought to be one of the incitements of adhesion. Due to ADP being a platelet-activating factor, J-1-ADP hydrogel showed significant hemostatic activity in vitro verified by whole blood coagulation, plasma coagulation, platelet activation, and platelet adhesion assays. Further, it showed potent hemostatic activity in a mouse liver hemorrhage model. Bleeding was believed to be a cause of the formation of postsurgery adhesion. J-1-ADP hydrogel had a significant antiadhesion effect in a rat side wall defect-cecum abrasion model. In addition, it had good biocompatibility and degradation properties. So the present study may provide an alternative strategy for designing antimicrobial peptide hydrogel material to prevent postoperative adhesion formation in the clinic.
Collapse
Affiliation(s)
| | - Hanru Zhang
- Department of Obstetrics & Gynecology, Gansu Provincial Maternity and Child Care Hospital, Lanzhou 730000, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Fernando H, McFadyen JD, Wang X, Shaw J, Stub D, Peter K. P2Y12 Antagonists in Cardiovascular Disease—Finding the Best Balance Between Preventing Ischemic Events and Causing Bleeding. Front Cardiovasc Med 2022; 9:854813. [PMID: 35647068 PMCID: PMC9133423 DOI: 10.3389/fcvm.2022.854813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022] Open
Abstract
Dual antiplatelet therapy comprising of aspirin and oral P2Y12 receptor antagonists are an established cornerstone of therapy in acute coronary syndromes and percutaneous coronary intervention. As a result, the platelet P2Y12 receptor remains a key therapeutic target in cardiovascular medicine since pharmacological antagonists were first developed in the 1990’s. With a greater understanding of platelet biology and the role played by the P2Y12 receptor in the amplification of platelet activation and thrombus formation, there has been progressive refinement in the development of P2Y12 receptor antagonists with greater potency and consistency of antiplatelet effect. However, challenges remain in the utilization of these agents particularly in balancing the need for greater protection from ischemic events whilst minimizing the bleeding risk and present a real opportunity for the institution of individualized medicine. Future drug developments will provide clinicians with greater avenues to achieve this.
Collapse
Affiliation(s)
- Himawan Fernando
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
| | - James D. McFadyen
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Thrombosis and Hemostasis Unit, Department of Clinical Hematology, The Alfred Hospital, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| | - James Shaw
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Dion Stub
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
- Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
- *Correspondence: Karlheinz Peter,
| |
Collapse
|
65
|
Medical Gas Plasma—A Potent ROS-Generating Technology for Managing Intraoperative Bleeding Complications. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12083800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cold medical gas plasmas are under pre-clinical investigation concerning their hemostatic activity and could be applied for intra-operative bleeding control in the future. The technological leap innovation was their generation at body temperature, thereby causing no thermal harm to the tissue and ensuring tissue integrity. This directly contrasts with current techniques such as electrocautery, which induces hemostasis by carbonizing the tissue using a heated electrode. However, the necrotized tissue is prone to fall, raising the risk of post-operative complications such as secondary bleedings or infection. In recent years, various studies have reported on the ability of medical gas plasmas to induce blood coagulation, including several suggestions concerning their mode of action. As non-invasive and gentle hemostatic agents, medical gas plasmas could be particularly eligible for vulnerable tissues, e.g., colorectal surgery and neurosurgery. Further, their usage could be beneficial regarding the prevention of post-operative bleedings due to the absence or sloughing of eschar. However, no clinical trials or individual healing attempts for medical gas plasmas have been reported to pave the way for clinical approvement until now, despite promising results in experimental animal models. In this light, the present mini-review aims to emphasize the potential of medical gas plasmas to serve as a hemostatic agent in clinical procedures. Providing a detailed overview of the current state of knowledge, feasible application fields are discussed, and possible obstacles are addressed.
Collapse
|
66
|
Morrell CN, Mix D, Aggarwal A, Bhandari R, Godwin M, Owens Iii AP, Lyden SP, Doyle A, Krauel K, Rondina MT, Mohan A, Lowenstein CJ, Shim S, Stauffer S, Josyula VP, Ture SK, Yule DI, Wagner Iii LE, Ashton JM, Elbadawi A, Cameron SJ. Platelet olfactory receptor activation limits platelet reactivity and growth of aortic aneurysms. J Clin Invest 2022; 132:152373. [PMID: 35324479 PMCID: PMC9057618 DOI: 10.1172/jci152373] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 03/16/2022] [Indexed: 11/28/2022] Open
Abstract
As blood transitions from steady laminar flow (S-flow) in healthy arteries to disturbed flow (D-flow) in aneurysmal arteries, platelets are subjected to external forces. Biomechanical platelet activation is incompletely understood and is a potential mechanism behind antiplatelet medication resistance. Although it has been demonstrated that antiplatelet drugs suppress the growth of abdominal aortic aneurysms (AAA) in patients, we found that a certain degree of platelet reactivity persisted in spite of aspirin therapy, urging us to consider additional antiplatelet therapeutic targets. Transcriptomic profiling of platelets from patients with AAA revealed upregulation of a signal transduction pathway common to olfactory receptors, and this was explored as a mediator of AAA progression. Healthy platelets subjected to D-flow ex vivo, platelets from patients with AAA, and platelets in murine models of AAA demonstrated increased membrane olfactory receptor 2L13 (OR2L13) expression. A drug screen identified a molecule activating platelet OR2L13, which limited both biochemical and biomechanical platelet activation as well as AAA growth. This observation was further supported by selective deletion of the OR2L13 ortholog in a murine model of AAA that accelerated aortic aneurysm growth and rupture. These studies revealed that olfactory receptors regulate platelet activation in AAA and aneurysmal progression through platelet-derived mediators of aortic remodeling.
Collapse
Affiliation(s)
- Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - Doran Mix
- Department of Surgery, Division of Vascular Surgery, University of Rochester School of Medicine, Rochester, United States of America
| | - Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Rohan Bhandari
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Matthew Godwin
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - A Phillip Owens Iii
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, United States of America
| | - Sean P Lyden
- Department of Vascular Surgery, Cleveland Clinic, Cleveland, United States of America
| | - Adam Doyle
- Department of Surgery, Division of Vascular Surgery, University of Rochester School of Medicine, Rochester, United States of America
| | - Krystin Krauel
- Department of Molecular Medicine, University of Utah, Salt Lake City, United States of America
| | - Matthew T Rondina
- Department of Internal Medicine, University of Utah, Salt Lake City, United States of America
| | - Amy Mohan
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - Charles J Lowenstein
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, United States of America
| | - Sharon Shim
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Shaun Stauffer
- Center for Therapeutics Discovery, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Vara Prasad Josyula
- Center for Therapeutics Discovery, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| | - Sara K Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, United States of America
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, United States of America
| | - Larry E Wagner Iii
- Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, United States of America
| | - John M Ashton
- Department of Biomedical Genetics, University of Rochester School of Medicine, Rochester, United States of America
| | - Ayman Elbadawi
- Department of Cardiovascular Medicine, University of Texas Medical Branch, Galveston, United States of America
| | - Scott J Cameron
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, United States of America
| |
Collapse
|
67
|
Kenkhuis B, Somarakis A, Kleindouwel LRT, van Roon-Mom WMC, Höllt T, van der Weerd L. Co-expression patterns of microglia markers Iba1, TMEM119 and P2RY12 in Alzheimer's disease. Neurobiol Dis 2022; 167:105684. [PMID: 35247551 DOI: 10.1016/j.nbd.2022.105684] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022] Open
Abstract
Microglia have been identified as key players in Alzheimer's disease pathogenesis, and other neurodegenerative diseases. Iba1, and more specifically TMEM119 and P2RY12 are gaining ground as presumedly more specific microglia markers, but comprehensive characterization of the expression of these three markers individually as well as combined is currently missing. Here we used a multispectral immunofluorescence dataset, in which over seventy thousand microglia from both aged controls and Alzheimer patients have been analysed for expression of Iba1, TMEM119 and P2RY12 on a single-cell level. For all markers, we studied the overlap and differences in expression patterns and the effect of proximity to β-amyloid plaques. We found no difference in absolute microglia numbers between control and Alzheimer subjects, but the prevalence of specific combinations of markers (phenotypes) differed greatly. In controls, the majority of microglia expressed all three markers. In Alzheimer patients, a significant loss of TMEM119+-phenotypes was observed, independent of the presence of β-amyloid plaques in its proximity. Contrary, phenotypes showing loss of P2RY12, but consistent Iba1 expression were increasingly prevalent around β-amyloid plaques. No morphological features were conclusively associated with loss or gain of any of the markers or any of the identified phenotypes. All in all, none of the three markers were expressed by all microglia, nor can be wholly regarded as a pan- or homeostatic marker, and preferential phenotypes were observed depending on the surrounding pathological or homeostatic environment. This work could help select and interpret microglia markers in previous and future studies.
Collapse
Affiliation(s)
- Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Antonios Somarakis
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lynn R T Kleindouwel
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Thomas Höllt
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Intelligent Systems, Delft University of Technology, Delft, the Netherlands
| | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
68
|
Monooxygenase- and Dioxygenase-Catalyzed Oxidative Dearomatization of Thiophenes by Sulfoxidation, cis-Dihydroxylation and Epoxidation. Int J Mol Sci 2022; 23:ijms23020909. [PMID: 35055091 PMCID: PMC8777831 DOI: 10.3390/ijms23020909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Enzymatic oxidations of thiophenes, including thiophene-containing drugs, are important for biodesulfurization of crude oil and drug metabolism of mono- and poly-cyclic thiophenes. Thiophene oxidative dearomatization pathways involve reactive metabolites, whose detection is important in the pharmaceutical industry, and are catalyzed by monooxygenase (sulfoxidation, epoxidation) and dioxygenase (sulfoxidation, dihydroxylation) enzymes. Sulfoxide and epoxide metabolites of thiophene substrates are often unstable, and, while cis-dihydrodiol metabolites are more stable, significant challenges are presented by both types of metabolite. Prediction of the structure, relative and absolute configuration, and enantiopurity of chiral metabolites obtained from thiophene enzymatic oxidation depends on the substrate, type of oxygenase selected, and molecular docking results. The racemization and dimerization of sulfoxides, cis/trans epimerization of dihydrodiol metabolites, and aromatization of epoxides are all factors associated with the mono- and di-oxygenase-catalyzed metabolism of thiophenes and thiophene-containing drugs and their applications in chemoenzymatic synthesis and medicine.
Collapse
|
69
|
Chen X, Zhu W, Liu H, Deng F, Wang W, Qin L. Preparation of injectable clopidogrel loaded submicron emulsion for enhancing physicochemical stability and anti-thrombotic efficacy. Int J Pharm 2022; 611:121323. [PMID: 34848363 DOI: 10.1016/j.ijpharm.2021.121323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 11/24/2021] [Indexed: 01/13/2023]
Abstract
Due to the superior safety and therapeutic efficacy, clopidogrel (CLP) has been widely used to prevent postoperative thrombosis. However, limitations of delayed absorption and metabolic activation of clopidogrel after oral administration hinder its clinic use for acute thrombosis treatment in percutaneous coronary intervention (PCI). Although clopidogrel aqueous injection systems were designed and developed, chemical instability under physiological condition or vascular irritation remains to be solved. In this study, we aim to prepare an injectable clopidogrel loaded submicron emulsion to overcome the drawbacks of conventional clopidogrel aqueous formulation and improve the antiplatelet aggregation effects. Results showed that this delivery system exerted inspiring features including uniform particle size, higher drug loading capacity and sustained drug release behavior. It can dramatically upgrade the formulation stability and prevent the drug degradation under sterilization or higher pH environments. No remarkable droplet size increase or drug content decrease was observed during storage. Compared to CLP tablet, the peak drug concentration (Cmax) and area under the curve (AUC) of CLP emulsion increased by 12.01-fold and 4.69-fold, respectively. Most importantly, it exerted excellent in vivo anti-thrombotic effect on numerous designed animal models. Conclusively, submicron emulsion is a promising delivery system for improving clopidogrel stability and anti-thrombotic efficacy.
Collapse
Affiliation(s)
- Xuehong Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; The Information Area of Xihu Industrial Base Shilong Town, Dongguan 523000, China
| | - Wanye Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huan Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Fengjian Deng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wanting Wang
- HEC Industrial Development Co. Changan Town, Dongguan 523000, China.
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
70
|
|
71
|
P2Y 12-dependent activation of hematopoietic stem and progenitor cells promotes emergency hematopoiesis after myocardial infarction. Basic Res Cardiol 2022; 117:16. [PMID: 35353230 PMCID: PMC8967792 DOI: 10.1007/s00395-022-00927-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 02/28/2022] [Accepted: 03/17/2022] [Indexed: 01/31/2023]
Abstract
Emergency hematopoiesis is the driving force of the inflammatory response to myocardial infarction (MI). Increased proliferation of hematopoietic stem and progenitor cells (LSK) after MI enhances cell production in the bone marrow (BM) and replenishes leukocyte supply for local cell recruitment to the infarct. Decoding the regulation of the inflammatory cascade after MI may provide new avenues to improve post-MI remodeling. In this study, we describe the influence of adenosine diphosphate (ADP)-dependent P2Y12-mediated signaling on emergency hematopoiesis and cardiac remodeling after MI. Permanent coronary ligation was performed to induce MI in a murine model. BM activation, inflammatory cell composition and cardiac function were assessed using global and platelet-specific gene knockout and pharmacological inhibition models for P2Y12. Complementary in vitro studies allowed for investigation of ADP-dependent effects on LSK cells. We found that ADP acts as a danger signal for the hematopoietic BM and fosters emergency hematopoiesis by promoting Akt phosphorylation and cell cycle progression. We were able to detect P2Y12 in LSK, implicating a direct effect of ADP on LSK via P2Y12 signaling. P2Y12 knockout and P2Y12 inhibitor treatment with prasugrel reduced emergency hematopoiesis and the excessive inflammatory response to MI, translating to lower numbers of downstream progeny and inflammatory cells in the blood and infarct. Ultimately, P2Y12 inhibition preserved cardiac function and reduced chronic adverse cardiac remodeling after MI. P2Y12-dependent signaling is involved in emergency hematopoiesis after MI and fuels post-ischemic inflammation, proposing a novel, non-canonical value for P2Y12 antagonists beyond inhibition of platelet-mediated atherothrombosis.
Collapse
|
72
|
Wang J, Zheng B, Yang S, Fan J, Sun H, Wang J. Mesoporous Silica Nanoparticles Carrying MicroRNA-124 to Target P2Y 12 Facilitates Cerebral Angiogenesis in Lacunar Cerebral Infarction Through Stem Cell Factor/c-Kit Signaling Pathway. J Biomed Nanotechnol 2022; 18:218-224. [PMID: 35180915 DOI: 10.1166/jbn.2022.3240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
MicroRNA (miRNA)-124 inhibitor may enhance cerebral blood vessel formation in lacunar cerebral infarction (LCI) and mesoporous silica nanoparticles are highlighted as a drug carrier which improves patients' outcome. This study explored the impact of miR-124 and its promising targeted gene P2Y12 encapsulated by mesoporous silica nanoparticles (MSNs) on progression of LCI, and its interaction between SCF/c-kit signaling pathway. After establishment of animal models, the animals were divided into 6 groups, namely: model group, blank group, empty carrier group, carrier + miR-124 inhibitor group, SCF/c-kit inhibitor group, and P2Y12 agonist group. Western blot analysis and microscope determined the expression level of miR-124 in the rat brain tissue slices. MVD, SCF and c-kit P2Y12 protein expression levels were detected and their targeting relationship was verified. miR-124 was poorly expressed in the cells of rats with LCI upon injection of MSNs carrying miR-124-inhibitor. The LCI model group had the highest number of VEGF-positive. Compared with the model group, the number in the carrier + miR-124 inhibitor group was lowest. Moreover, treatment with SCF/c-kit inhibitor and P2Y12 agonist also obtained reduction in the number of VEGF-positive cells with less prominent effect (P < 0.05). With elevation of MVD in the LCI rats, injection of P2Y12 agonist or SCF/c-kit inhibitor significantly decreased the amount of MVD, while miR-124 inhibitor-loaded MSNs better reduced the MVD level. Besides, the LCI rats exhibited up-regulated level of P2Y12 protein. Injection of P2Y12 agonist or SCF/c-kit inhibitor dramatically decreased the level of P2Y12, where the level was still higher than that of carrier + miR-124 inhibitor group. Moreover, administration of miR-124 inhibitor-loaded MSNs resulted in increased SCF and c-kit protein level, and SCF/c-kit inhibitor group and P2Y12 agonist group also had increased SCF and c-kit protein level, compared to the model group. Mechanistically, the miR-124 was indicated to target P2Y12 with stronger fluorescence intensity in mutant plasmid (P < 0.05). MSN-encapsulated miR-124 inhibitor increased the expression of SCF/c-kit protein by targeting P2Y12, thereby enhancing regeneration of cerebral blood vessels in LCI.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Ya'an Peoples Hospital, Ya'an, Sichuan, 625000, China
| | - Bo Zheng
- Department of Neurology, Ya'an Peoples Hospital, Ya'an, Sichuan, 625000, China
| | - Shu Yang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610000, China
| | - Jian Fan
- Department of Neurology, The First People's Hospital of Shuangliu District, Chengdu, Sichuan, 610299, China
| | - Hao Sun
- Department of Neurology, The First People's Hospital of Shuangliu District, Chengdu, Sichuan, 610299, China
| | - Jianhong Wang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610000, China
| |
Collapse
|
73
|
Wu Z, He K, Chen Y, Li H, Pan S, Li B, Liu T, Xi F, Deng F, Wang H, Du J, Jing M, Li Y. A sensitive GRAB sensor for detecting extracellular ATP in vitro and in vivo. Neuron 2021; 110:770-782.e5. [PMID: 34942116 DOI: 10.1016/j.neuron.2021.11.027] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/31/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022]
Abstract
The purinergic transmitter ATP (adenosine 5'-triphosphate) plays an essential role in both the central and peripheral nervous systems, and the ability to directly measure extracellular ATP in real time will increase our understanding of its physiological functions. Here, we developed a sensitive GPCR activation-based ATP sensor called GRABATP1.0, with a robust fluorescence response to extracellular ATP when expressed in several cell types. This sensor has sub-second kinetics, has ATP affinity in the range of tens of nanomolar, and can be used to localize ATP release with subcellular resolution. Using this sensor, we monitored ATP release under a variety of in vitro and in vivo conditions, including stimuli-induced and spontaneous ATP release in primary hippocampal cultures, injury-induced ATP release in a zebrafish model, and lipopolysaccharides-induced ATP-release events in individual astrocytes in the mouse cortex. Thus, the GRABATP1.0 sensor is a sensitive, versatile tool for monitoring ATP release and dynamics under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China.
| | - Kaikai He
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Yue Chen
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Hongyu Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Sunlei Pan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Bohan Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Tingting Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Fengxue Xi
- Chinese Institute for Brain Research, Beijing 102206, China; School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fei Deng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Miao Jing
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
74
|
Jourdi G, Lordkipanidzé M, Philippe A, Bachelot-Loza C, Gaussem P. Current and Novel Antiplatelet Therapies for the Treatment of Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms222313079. [PMID: 34884884 PMCID: PMC8658271 DOI: 10.3390/ijms222313079] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decades, antiplatelet agents, mainly aspirin and P2Y12 receptor antagonists, have significantly reduced morbidity and mortality associated with arterial thrombosis. Their pharmacological characteristics, including pharmacokinetic/pharmacodynamics profiles, have been extensively studied, and a significant number of clinical trials assessing their efficacy and safety in various clinical settings have established antithrombotic efficacy. Notwithstanding, antiplatelet agents carry an inherent risk of bleeding. Given that bleeding is associated with adverse cardiovascular outcomes and mortality, there is an unmet clinical need to develop novel antiplatelet therapies that inhibit thrombosis while maintaining hemostasis. In this review, we present the currently available antiplatelet agents, with a particular focus on their targets, pharmacological characteristics, and patterns of use. We will further discuss the novel antiplatelet therapies in the pipeline, with the goal of improved clinical outcomes among patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: (G.J.); (P.G.)
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Aurélien Philippe
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Christilla Bachelot-Loza
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
| | - Pascale Gaussem
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
- Correspondence: (G.J.); (P.G.)
| |
Collapse
|
75
|
Liu Y, Hu X, Song P, Li H, Li M, Du Y, Li M, Ma Q, Peng L, Song M, Chen X. Influence of GAS5/MicroRNA-223-3p/P2Y12 Axis on Clopidogrel Response in Coronary Artery Disease. J Am Heart Assoc 2021; 10:e021129. [PMID: 34713722 PMCID: PMC8751826 DOI: 10.1161/jaha.121.021129] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Dual antiplatelet therapy based on aspirin and P2Y12 receptor antagonists such as clopidogrel is currently the primary treatment for coronary artery disease (CAD). However, a percentage of patients exhibit clopidogrel resistance, in which genetic factors play vital roles. This study aimed to investigate the roles of GAS5 (growth arrest-specific 5) and its rs55829688 polymorphism in clopidogrel response in patients with CAD. Methods and Results A total of 444 patients with CAD receiving dual antiplatelet therapy from 2017 to 2018 were enrolled to evaluate the effect of GAS5 single nucleotide polymorphism rs55829688 on platelet reactivity index. Platelets from 37 patients of these patients were purified with microbeads to detect GAS5 and microRNA-223-3p (miR-223-3p) expression. Platelet-rich plasma was isolated from another 17 healthy volunteers and 46 newly diagnosed patients with CAD to detect GAS5 and miR-223-3p expression. A dual-luciferase reporter assay was performed to explore the interaction between miR-223-3p and GAS5 or P2Y12 3'-UTR in (human embryonic kidney 293 cell line that expresses a mutant version of the SV40 large T antigen) HEK 293T and (megakaryoblastic cell line derived in 1983 from the bone marrow of a chronic myeloid leukemia patient with megakaryoblastic crisis) MEG-01 cells. Loss-of-function and gain-of-function experiments were performed to reveal the regulation of GAS5 toward P2Y12 via miR-223-3p in MEG-01 cells. We observed that rs55829688 CC homozygotes showed significantly decreased platelet reactivity index than TT homozygotes in CYP2C19 poor metabolizers. Platelet GAS5 expression correlated positively with both platelet reactivity index and P2Y12 mRNA expressions, whereas platelet miR-223-3p expression negatively correlated with platelet reactivity index. Meanwhile, a negative correlation between GAS5 and miR-223-3p expressions was observed in platelets. MiR-223-3p mimic reduced while the miR-223-3p inhibitor increased the expression of GAS5 and P2Y12 in MEG-01 cells. Knockdown of GAS5 by siRNA increased miR-223-3p expression and decreased P2Y12 expression, which could be reversed by the miR-223-3p inhibitor. Meanwhile, overexpression of GAS5 reduced miR-223-3p expression and increased P2Y12 expression, which could be reversed by miR-223-3p mimic. Conclusions GAS5 rs55829688 polymorphism might affect clopidogrel response in patients with CAD with the CYP2C19 poor metabolizer genotypes, and GAS5 regulates P2Y12 expression and clopidogrel response by acting as a competitive endogenous RNA for miR-223-3p.
Collapse
Affiliation(s)
- Yan‐Ling Liu
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Xiao‐Lei Hu
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Pei‐Yuan Song
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - He Li
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Mu‐Peng Li
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Yin‐Xiao Du
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Mo‐Yun Li
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
| | - Qi‐Lin Ma
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Li‐Ming Peng
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ming‐Yu Song
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
- Department of NeurologyXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiao‐Ping Chen
- Department of Clinical PharmacologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of PharmacogeneticsChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
76
|
Olgar Y, Durak A, Degirmenci S, Tuncay E, Billur D, Ozdemir S, Turan B. Ticagrelor alleviates high-carbohydrate intake induced altered electrical activity of ventricular cardiomyocytes by regulating sarcoplasmic reticulum-mitochondria miscommunication. Mol Cell Biochem 2021; 476:3827-3844. [PMID: 34114148 DOI: 10.1007/s11010-021-04205-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022]
Abstract
Metabolic syndrome (MetS) is associated with additional cardiovascular risk in mammalians while there are relationships between hyperglycemia-associated cardiovascular dysfunction and increased platelet P2Y12 receptor activation. Although P2Y12 receptor antagonist ticagrelor (Tica) plays roles in reduction of cardiovascular events, its beneficial mechanism remains poorly understood. Therefore, we aimed to clarify whether Tica can exert a direct protective effect in ventricular cardiomyocytes from high-carbohydrate diet-induced MetS rats, at least, through affecting sarcoplasmic reticulum (SR)-mitochondria (Mit) miscommunication. Tica treatment of MetS rats (150 mg/kg/day for 15 days) significantly reversed the altered parameters of action potentials by reversing sarcolemmal ionic currents carried by voltage-dependent Na+ and K+ channels, and Na+/Ca2+-exchanger in the cells, expressed P2Y12 receptors. The increased basal-cytosolic Ca2+ level and depressed SR Ca2+ load were also reversed in Tica-treated cells, at most, though recoveries in the phosphorylation levels of ryanodine receptors and phospholamban. Moreover, there were marked recoveries in Mit structure and function (including increases in both autophagosomes and fragmentations) together with recoveries in Mit proteins and the factors associated with Ca2+ transfer between SR-Mit. There were further significant recoveries in markers of both ER stress and oxidative stress. Taken into consideration the Tica-induced prevention of ER stress and mitochondrial dysfunction, our data provided an important document on the pleiotropic effects of Tica in the electrical activity of the cardiomyocytes from MetS rats. This protective effect seems through recoveries in SR-Mit miscommunication besides modulation of different sarcolemmal ion-channel activities, independent of P2Y12 receptor antagonism.
Collapse
Affiliation(s)
- Yusuf Olgar
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey
| | - Aysegul Durak
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey
| | - Sinan Degirmenci
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey
| | - Erkan Tuncay
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey
| | - Deniz Billur
- Faculty of Medicine, Department of Histology and Embryology, Ankara University, Ankara, Turkey
| | - Semir Ozdemir
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey.
| | - Belma Turan
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey.
- Faculty of Medicine, Department of Biophysics, Lokman Hekim University, Ankara, Turkey.
| |
Collapse
|
77
|
Zhang X, Zhao W, Zhao Y, Zhao Z, Lv Z, Zhang Z, Ren H, Wang Q, Liu M, Qian M, Du B, Qin J. Inflammatory macrophages exacerbate neutrophil-driven joint damage through ADP/P2Y 1 signaling in rheumatoid arthritis. SCIENCE CHINA-LIFE SCIENCES 2021; 65:953-968. [PMID: 34480694 DOI: 10.1007/s11427-020-1957-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that primarily affects the joints and is associated with excessive immune cell infiltration. However, the complex interactions between the immune cell populations in the RA synovium remain unknown. Here, we demonstrate that inflammatory macrophages in the synovium exacerbate neutrophil-driven joint damage in RA through ADP/P2Y1 signaling. We show that extracellular ADP (eADP) and its receptors are obviously increased in synovial tissues of RA patients as well as collagen-induced arthritis (CIA) mice, and eADP enhances neutrophil infiltration into joints through macrophages producing the chemokine CXCL2, aggravating disease development. Accordingly, the arthritis mouse model had more neutrophils in inflamed joints following ADP injection, whereas P2Y1 deficiency and pharmacologic inhibition restored arthritis severity to basal levels, suggesting a dominant role of ADP/P2Y1 signaling in RA pathology. Moreover, cellular activity of ADP/P2Y1-mediated CXCL2 production was dependent on the Gαq/Ca2+-NF-κB/NFAT pathway in macrophages. Overall, this study reveals a non-redundant role of eADP as a trigger in the pathogenesis of RA through neutrophil recruitment and disrupted tissue homeostasis and function.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.,Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 510275, China
| | - Wenxiang Zhao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Yihan Zhao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Zeda Zhao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Zhangsheng Lv
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Zhen Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Hua Ren
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Qin Wang
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Shanghai, 201499, China
| | - Mingyao Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Min Qian
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China
| | - Bing Du
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.
| | - Juliang Qin
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
78
|
P 2Y 12 receptor blockers are anti-inflammatory drugs inhibiting both circulating monocytes and macrophages including THP-1 cells. Sci Rep 2021; 11:17459. [PMID: 34465804 PMCID: PMC8408182 DOI: 10.1038/s41598-021-95710-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/22/2021] [Indexed: 01/11/2023] Open
Abstract
P2Y12 blockade improves patient outcomes after myocardial infarction. As well as antithrombotic effects, anti-inflammatory effects may contribute to this beneficial clinical outcome. Here we aimed to identify potential anti-inflammatory effects of P2Y12 receptor blockers on monocytes and macrophages. Using flow cytometry, migration assays, flow chambers and RNA microarrays, we investigated the effects of adenosine diphosphate (ADP) and P2Y12 receptor blockers on blood monocytes, THP-1 monocytes and THP-1 monocytes after differentiation to macrophages. P2Y12 -expressing platelets can form aggregates with monocytes in circulating blood. Mediated by platelets, ADP results in activation of the integrin receptor Mac-1 on blood monocytes, as detected by the conformation-specific single-chain antibody MAN-1. Via the same association with platelets, THP-1 monocyte adhesion to the endothelial intercellular adhesion molecule 1 (ICAM-1) is induced by ADP. P2Y12 receptor blockers prevent these ADP effects on monocytes. Interestingly, in contrast to THP-1 monocytes, THP-1 monocytes, after differentiation to macrophages, directly expressed the P2Y12 receptor and consequently ADP was found to be a potent chemoattractant. Again, P2Y12 receptor blockers antagonised this effect. Accordingly, stimulation of THP-1 macrophages with ADP caused a substantial change in gene expression pattern and upregulation of several genes associated with inflammation and atherogenesis. These data establish novel anti-inflammatory effects of P2Y12 receptor blockers on monocytes and macrophages, which are expected to contribute to cardiovascular risk reduction.
Collapse
|
79
|
Lier J, Streit WJ, Bechmann I. Beyond Activation: Characterizing Microglial Functional Phenotypes. Cells 2021; 10:cells10092236. [PMID: 34571885 PMCID: PMC8464670 DOI: 10.3390/cells10092236] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022] Open
Abstract
Classically, the following three morphological states of microglia have been defined: ramified, amoeboid and phagocytic. While ramified cells were long regarded as “resting”, amoeboid and phagocytic microglia were viewed as “activated”. In aged human brains, a fourth, morphologically novel state has been described, i.e., dystrophic microglia, which are thought to be senescent cells. Since microglia are not replenished by blood-borne mononuclear cells under physiological circumstances, they seem to have an “expiration date” limiting their capacity to phagocytose and support neurons. Identifying factors that drive microglial aging may thus be helpful to delay the onset of neurodegenerative diseases, such as Alzheimer’s disease (AD). Recent progress in single-cell deep sequencing methods allowed for more refined differentiation and revealed regional-, age- and sex-dependent differences of the microglial population, and a growing number of studies demonstrate various expression profiles defining microglial subpopulations. Given the heterogeneity of pathologic states in the central nervous system, the need for accurately describing microglial morphology and expression patterns becomes increasingly important. Here, we review commonly used microglial markers and their fluctuations in expression in health and disease, with a focus on IBA1 low/negative microglia, which can be found in individuals with liver disease.
Collapse
Affiliation(s)
- Julia Lier
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
- Department of Neurology, University of Leipzig, 04109 Leipzig, Germany
- Correspondence:
| | - Wolfgang J. Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32611, USA;
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
| |
Collapse
|
80
|
Zhao Z, Wang Y, Tian N, Yan H, Wang J. Synthesis and biological evaluation of N 6 derivatives of 8-azapurine as novel antiplatelet agents. RSC Med Chem 2021; 12:1414-1427. [PMID: 34458743 PMCID: PMC8372213 DOI: 10.1039/d1md00128k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/19/2021] [Indexed: 11/21/2022] Open
Abstract
Two series of novel N 6 derivatives of 8-azapurine I and II were designed as antiplatelet agents. Series I and II were N 6 amino derivatives and N 6 hydrazone derivatives of 8-azapurine, respectively. The compounds were synthesized in acceptable yields via conventional procedures, including nucleophilic substitution, diazotization, and amination or hydrazonation with amino alcohol and 4,6-dichloropyrimidine as starting materials. To assess the ability of the synthesized compounds as antiplatelet agents, the ADP-induced platelet aggregation assay of Born was performed both in vitro and in vivo using ticagrelor as a reference control substance. The analysis of the structure-activity relationship and molecular docking were also discussed in detail. The results demonstrated that series I and II compounds exhibited antiplatelet activity in vitro and IIh was the most active compound (IC50 = 0.20 μM) among the target compounds, being almost 4-fold better than ticagrelor (IC50 = 0.74 μM). For a preliminary assessment of the safety profile, a bleeding test (mouse tail) and a single-dose toxicity test were conducted. The use of compound IIh resulted in a shorter bleeding time, less blood loss and lower acute toxicity compared to ticagrelor. In addition, a molecular docking study was performed to investigate the binding capacity and binding mode between IIh and P2Y12.
Collapse
Affiliation(s)
- Zhichang Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology Beijing 100124 P. R. China
| | - Yeming Wang
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Econnomi Technological Development Area (BDA) Beijing 100176 China
| | - Nana Tian
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology Beijing 100124 P. R. China
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Econnomi Technological Development Area (BDA) Beijing 100176 China
| | - Hong Yan
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology Beijing 100124 P. R. China
| | - Juan Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology Beijing 100124 P. R. China
| |
Collapse
|
81
|
LncRNA TCONS_00041002 improves neurological outcomes in neonatal rats with hypoxic-ischemic encephalopathy by inhibiting apoptosis and promoting neuron survival. Exp Neurol 2021; 346:113835. [PMID: 34390705 DOI: 10.1016/j.expneurol.2021.113835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 07/12/2021] [Accepted: 08/05/2021] [Indexed: 02/05/2023]
Abstract
It has been reported that Neonatal hypoxic-ischemic encephalopathy (HIE) could induce apoptosis in neonates and result in cognitive and sensory impairments, which are associated with poor developmental outcomes. Despite the improvement in neonatology, there is still no clinically effective treatment for HIE presently. Long non-coding RNAs (lncRNAs) play important roles in cellular homeostasis. Nevertheless, their effects in developing rat brains with HI is little known. Here, we established HIE model in neonate rats and explored the expression and function of lncRNAs in HI, and found the expression of 19 lncRNAs was remarkably changed in the brains of HI rats, compared to the sham group. Among them, three lncRNAs (TCONS_00041002, TCONS_00070547, TCONS_00045572) were enriched in the apoptotic process via gene ontology (GO) and pathway analysis, which were selected for the further qRT-PCR verification. Through lentivirus-mediated overexpression of these three lncRNAs, we found that overexpression of TCONS_00041002 attenuated the cell apoptosis, and increased the vitality of neurons after oxygen-glucose deprivation (OGD), therefore reduced the brain infarction and further promoted the neuron survival as well as improved the neurological disorders in the rats subjected to HIE. What's more, ceRNA network prediction and co-expression verification showed that the expression of TCONS_00041002 was positively associated with Foxe1, Pawr and Nfkbiz. Altogether, this study has exhibited that lncRNA TCONS_00041002 participates in the cell apoptosis and neuronal survival of HIE and represents a potential new target for the treatment of HIE.
Collapse
|
82
|
Aminoshariae A, Donaldson M, Horan M, Kulild JC, Baur D. Perioperative Antiplatelet and Anticoagulant Management with Endodontic Microsurgical Techniques. J Endod 2021; 47:1557-1565. [PMID: 34265324 DOI: 10.1016/j.joen.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/23/2021] [Accepted: 07/01/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The purpose of this study was to review evidence-based recommendations for the safe perioperative management of patients undergoing endodontic microsurgery who are currently taking antiplatelet or anticoagulant medications. Using the PICO (Population, Intervention, Comparison, Outcome) format, the following scientific question was asked: In patients taking anticoagulant or antiplatelet agents, what is the available evidence in the management of endodontic microsurgery? METHODS MEDLINE, Scopus, Cochrane Library, and ClinicalTrials.gov databases were searched to identify current recommendations regarding the management of antiplatelet and anticoagulant medications in the context of outpatient dental surgical procedures. Additionally, the authors hand searched the bibliographies of all relevant articles, the gray literature, and textbooks. Because of the lack of clinical studies and evidence on this subject, articles and guidelines from other organizations and association position statements were included. RESULTS Because any minor surgery can become a major surgery, the treating doctor needs to best assess the risk of bleeding, especially if the surgery is anticipated to take longer than 45 minutes. Every patient should be stratified on a case-by-case basis. Consultation with the patient's physician is highly recommended. CONCLUSIONS In order to maximize the effects of these medications (to prevent thrombosis) while minimizing the potential risks (procedural hemorrhage), clinicians should be aware of the best available evidence when considering continuation or discontinuation of antiplatelet and anticoagulant agents perioperatively for endodontic microsurgery. Ideally, a joint effort from an expert panel for microsurgery would be warranted.
Collapse
Affiliation(s)
- Anita Aminoshariae
- Departments of Endodontics, Case Western Reserve University, School of Dental Medicine, Cleveland, Ohio.
| | - Mark Donaldson
- School of Pharmacy, University of Montana, Missoula, Montana; School of Dentistry, Oregon Health and Sciences University, Portland, Oregon
| | - Michael Horan
- Oral and Maxillofacial Surgery, Case Western Reserve University, School of Dental Medicine, Cleveland, Ohio
| | - James C Kulild
- UKMC Dental School, University of Missouri-Kansas City, Kansas City, Missouri
| | - Dale Baur
- Oral and Maxillofacial Surgery, Case Western Reserve University, School of Dental Medicine, Cleveland, Ohio
| |
Collapse
|
83
|
Wang A, Shi X, Yu R, Qiao B, Yang R, Xu C. The P2X 7 Receptor Is Involved in Diabetic Neuropathic Pain Hypersensitivity Mediated by TRPV1 in the Rat Dorsal Root Ganglion. Front Mol Neurosci 2021; 14:663649. [PMID: 34163328 PMCID: PMC8215290 DOI: 10.3389/fnmol.2021.663649] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022] Open
Abstract
The purinergic 2X7 (P2X7) receptor expressed in satellite glial cells (SGCs) is involved in the inflammatory response, and transient receptor potential vanilloid 1 (TRPV1) participates in the process of neurogenic inflammation, such as that in diabetic neuropathic pain (DNP) and peripheral neuralgia. The main purpose of this study was to explore the role of the P2X7 receptor in DNP hypersensitivity mediated by TRPV1 in the rat and its possible mechanism. A rat model of type 2 diabetes mellitus-related neuropathic pain (NPP) named the DNP rat model was established in this study. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of DNP rats were increased after intrathecal injection of the P2X7 receptor antagonist A438079, and the mRNA and protein levels of TRPV1 in the dorsal root ganglion (DRG) were decreased in DNP rats treated with A438079 compared to untreated DNP rats; in addition, A438079 also decreased the phosphorylation of p38 and extracellular signal-regulated kinase 1/2 (ERK1/2) in the DNP group. Based on these results, the P2X7 receptor might be involved in DNP mediated by TRPV1.
Collapse
Affiliation(s)
- Anhui Wang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
| | - Xiangchao Shi
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Ruoyang Yu
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Bao Qiao
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Runan Yang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, China
| |
Collapse
|
84
|
BAC transgenic mice to study the expression of P2X2 and P2Y 1 receptors. Purinergic Signal 2021; 17:449-465. [PMID: 34050505 PMCID: PMC8410928 DOI: 10.1007/s11302-021-09792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/19/2021] [Indexed: 11/30/2022] Open
Abstract
Extracellular purines are important signaling molecules involved in numerous physiological and pathological processes via the activation of P2 receptors. Information about the spatial and temporal P2 receptor (P2R) expression and its regulation remains crucial for the understanding of the role of P2Rs in health and disease. To identify cells carrying P2X2Rs in situ, we have generated BAC transgenic mice that express the P2X2R subunits as fluorescent fusion protein (P2X2-TagRFP). In addition, we generated a BAC P2Y1R TagRFP reporter mouse expressing a TagRFP reporter for the P2RY1 gene expression. We demonstrate expression of the P2X2R in a subset of DRG neurons, the brain stem, the hippocampus, as well as on Purkinje neurons of the cerebellum. However, the weak fluorescence intensity in our P2X2R-TagRFP mouse precluded tracking of living cells. Our P2Y1R reporter mice confirmed the widespread expression of the P2RY1 gene in the CNS and indicate for the first time P2RY1 gene expression in mouse Purkinje cells, which so far has only been described in rats and humans. Our P2R transgenic models have advanced the understanding of purinergic transmission, but BAC transgenic models appeared not always to be straightforward and permanent reliable. We noticed a loss of fluorescence intensity, which depended on the number of progeny generations. These problems are discussed and may help to provide more successful animal models, even if in future more versatile and adaptable nuclease-mediated genome-editing techniques will be the methods of choice.
Collapse
|
85
|
Cao D, Chandiramani R, Chiarito M, Claessen BE, Mehran R. Evolution of antithrombotic therapy in patients undergoing percutaneous coronary intervention: a 40-year journey. Eur Heart J 2021; 42:339-351. [PMID: 33367641 DOI: 10.1093/eurheartj/ehaa824] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/02/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Since its introduction in 1977, percutaneous coronary intervention has become one of the most commonly performed therapeutic procedures worldwide. Such widespread diffusion, however, would have not been possible without a concomitant evolution of the pharmacotherapies associated with this intervention. Antithrombotic agents are fundamental throughout the management of patients undergoing coronary stent implantation, starting from the procedure itself to the long-term prevention of cardiovascular events. The last 40 years of interventional cardiology have seen remarkable improvements in both drug therapies and device technologies, which largely reflected a progressive understanding of the pathophysiological mechanisms of coronary artery disease, as well as procedure- and device-related adverse events. The purpose of this article is to provide an overview of the important milestones in antithrombotic pharmacology that have shaped clinical practice of today while also providing insights into knowledge gaps and future directions.
Collapse
Affiliation(s)
- Davide Cao
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Rishi Chandiramani
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Mauro Chiarito
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy.,Cardio Center, Humanitas Clinical and Research Hospital IRCCS, Via Alessandro Manzoni 56, 20090 Rozzano, Milan, Italy
| | - Bimmer E Claessen
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA
| |
Collapse
|
86
|
Klaver D, Thurnher M. Control of Macrophage Inflammation by P2Y Purinergic Receptors. Cells 2021; 10:1098. [PMID: 34064383 PMCID: PMC8147772 DOI: 10.3390/cells10051098] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages comprise a phenotypically and functionally diverse group of hematopoietic cells. Versatile macrophage subsets engage to ensure maintenance of tissue integrity. To perform tissue stress surveillance, macrophages express many different stress-sensing receptors, including purinergic P2X and P2Y receptors that respond to extracellular nucleotides and their sugar derivatives. Activation of G protein-coupled P2Y receptors can be both pro- and anti-inflammatory. Current examples include the observation that P2Y14 receptor promotes STAT1-mediated inflammation in pro-inflammatory M1 macrophages as well as the demonstration that P2Y11 receptor suppresses the secretion of tumor necrosis factor (TNF)-α and concomitantly promotes the release of soluble TNF receptors from anti-inflammatory M2 macrophages. Here, we review macrophage regulation by P2Y purinergic receptors, both in physiological and disease-associated inflammation. Therapeutic targeting of anti-inflammatory P2Y receptor signaling is desirable to attenuate excessive inflammation in infectious diseases such as COVID-19. Conversely, anti-inflammatory P2Y receptor signaling must be suppressed during cancer therapy to preserve its efficacy.
Collapse
Affiliation(s)
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
87
|
Lovászi M, Branco Haas C, Antonioli L, Pacher P, Haskó G. The role of P2Y receptors in regulating immunity and metabolism. Biochem Pharmacol 2021; 187:114419. [PMID: 33460626 DOI: 10.1016/j.bcp.2021.114419] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
P2Y receptors are G protein-coupled receptors whose physiological agonists are the nucleotides ATP, ADP, UTP, UDP and UDP-glucose. Eight P2Y receptors have been cloned in humans: P2Y1R, P2Y2R, P2Y4R, P2Y6R, P2Y11R, P2Y12R, P2Y13R and P2Y14R. P2Y receptors are expressed in lymphoid tissues such as thymus, spleen and bone marrow where they are expressed on lymphocytes, macrophages, dendritic cells, neutrophils, eosinophils, mast cells, and platelets. P2Y receptors regulate many aspects of immune cell function, including phagocytosis and killing of pathogens, antigen presentation, chemotaxis, degranulation, cytokine production, and lymphocyte activation. Consequently, P2Y receptors shape the course of a wide range of infectious, autoimmune, and inflammatory diseases. P2Y12R ligands have already found their way into the therapeutic arena, and we envision additional ligands as future drugs for the treatment of diseases caused by or associated with immune dysregulation.
Collapse
Affiliation(s)
- Marianna Lovászi
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | | | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA.
| |
Collapse
|
88
|
Huang B, Qian Y, Xie S, Ye X, Chen H, Chen Z, Zhang L, Xu J, Hu H, Ma S, Héroux P, Wang D, Shen HM, Wu Y, Xia D. Ticagrelor inhibits the NLRP3 inflammasome to protect against inflammatory disease independent of the P2Y 12 signaling pathway. Cell Mol Immunol 2021; 18:1278-1289. [PMID: 32523112 PMCID: PMC8093290 DOI: 10.1038/s41423-020-0444-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 04/12/2020] [Indexed: 12/16/2022] Open
Abstract
Ticagrelor is the first reversibly binding oral P2Y12 receptor antagonist to inhibit platelet activation and has been approved by the Food and Drug Administration for the treatment of coronary artery disease. At present, the other pharmacological functions of ticagrelor remain poorly understood. The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays a critical role in the innate immune system, but its excessive activation also contributes to the pathogenesis of complex diseases. In this study, we systematically examined the effects of ticagrelor on the NLRP3 inflammasome and found that ticagrelor inhibits NLRP3 inflammasome activation in macrophages independent of its classic inhibitory effect on the P2Y12 signaling pathway. Further mechanistic studies demonstrate that ticagrelor attenuates the oligomerization of apoptosis-associated speck-like protein containing a CARD (ASC) by blocking chloride efflux, an effect achieved through the degradation of chloride intracellular channel proteins (CLICs) and blockade of the translocation of CLICs to the plasma membrane. Moreover, experiments on lipopolysaccharide-induced sepsis and alum-induced peritonitis in mice confirmed that ticagrelor mitigates the severity of systemic inflammation independent of P2Y12 receptor antagonism. Importantly, oral administration of ticagrelor rapidly and strongly inhibited NLRP3 inflammasome activation in peripheral blood mononuclear cells from patients with acute coronary syndrome. Overall, our study reveals a novel pharmacological function of ticagrelor in addition to its classic antiplatelet properties, which suggests that ticagrelor may serve as a potential therapeutic agent for use in NLRP3-associated diseases.
Collapse
Affiliation(s)
- Bo Huang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufeng Qian
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shujun Xie
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Translational Medicine Research Center, and Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianhua Ye
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hanwen Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhifeng Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihuan Zhang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinming Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hu Hu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenglin Ma
- Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Paul Héroux
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
89
|
Improved Practical Synthesis of DY‐038, an Oral Available Antiplatelet Agent. ChemistrySelect 2021. [DOI: 10.1002/slct.202100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
90
|
Luquero A, Badimon L, Borrell-Pages M. PCSK9 Functions in Atherosclerosis Are Not Limited to Plasmatic LDL-Cholesterol Regulation. Front Cardiovasc Med 2021; 8:639727. [PMID: 33834043 PMCID: PMC8021767 DOI: 10.3389/fcvm.2021.639727] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/01/2021] [Indexed: 12/31/2022] Open
Abstract
The relevance of PCSK9 in atherosclerosis progression is demonstrated by the benefits observed in patients that have followed PCSK9-targeted therapies. The impact of these therapies is attributed to the plasma lipid-lowering effect induced when LDLR hepatic expression levels are recovered after the suppression of soluble PCSK9. Different studies show that PCSK9 is involved in other mechanisms that take place at different stages during atherosclerosis development. Indeed, PCSK9 regulates the expression of key receptors expressed in macrophages that contribute to lipid-loading, foam cell formation and atherosclerotic plaque formation. PCSK9 is also a regulator of vascular inflammation and its expression correlates with pro-inflammatory cytokines release, inflammatory cell recruitment and plaque destabilization. Furthermore, anti-PCSK9 approaches have demonstrated that by inhibiting PCSK9 activity, the progression of atherosclerotic disease is diminished. PCSK9 also modulates thrombosis by modifying platelets steady-state, leukocyte recruitment and clot formation. In this review we evaluate recent findings on PCSK9 functions in cardiovascular diseases beyond LDL-cholesterol plasma levels regulation.
Collapse
Affiliation(s)
- Aureli Luquero
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación en Red- Área Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación en Red- Área Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
91
|
Manjuprasanna VN, Urs AP, Rudresha GV, Milan Gowda MD, Jayachandra K, Hiremath V, Rajaiah R, Vishwanath BS. Drupin, a thrombin-like protease prompts platelet activation and aggregation through protease-activated receptors. J Cell Biochem 2021; 122:870-881. [PMID: 33748988 DOI: 10.1002/jcb.29917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/10/2022]
Abstract
Hemostasis is a proteolytically regulated process that requires activation of platelets and the blood coagulation cascade upon vascular injury. Activated platelets create a thrombogenic environment and amplify the coagulation process. Plant latex proteases (PLPs) have been used as therapeutic components to treat various ailments by folk healers. One of the main applications of plant latices is to stop bleeding from minor injuries and to enhance wound healing activity. Although many studies have reported the pro-coagulant activities of PLPs, an in-depth investigation is required to understand the mechanism of action of PLPs on platelets. Here, the effect of PLPs on platelet aggregation was studied systematically to validate the observed pharmacological effect by folk healers. Among 29 latices from the Ficus genus tested, Ficus drupacea exhibited potent pro-coagulant and thrombin-like activity. Drupin, a thrombin-like cysteine protease responsible for platelet aggregation was purified from F. drupacea latex. Drupin exhibits pro-coagulant activity and reduces the bleeding time in mice tail. It induces platelet aggregation by activating mitogen-activated protein kinases and the nuclear factor-κB and PI3K/Akt signalling cascade, which, in turn, phosphorylats, cytosolic phospholipase A2 leading to the release of thromboxane A2 from the granules to activate the nearby platelets to aggregate. Furthermore, we investigated the involvement of protease-activated receptors in drupin-induced platelet aggregation using specific protease activated receptor 1 (PAR1) and PAR4 receptor antagonists. The results confirmed that the drupin-induced platelet aggregation was mediated by both PAR1 and PAR4, synergistically. Overall, drupin reduces the bleeding time by exerting pro-coagulant activity and induces platelet aggregation by activating the intracellular signalling cascade.
Collapse
Affiliation(s)
| | - Amog P Urs
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Gotravalli V Rudresha
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | | | - Krishnegowda Jayachandra
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Vilas Hiremath
- Vijayashree Diagnostics, Specialized Coagulation Lab, Bengaluru, Karnataka, India
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India.,Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| |
Collapse
|
92
|
Ferrari D, la Sala A, Milani D, Celeghini C, Casciano F. Purinergic Signaling in Controlling Macrophage and T Cell Functions During Atherosclerosis Development. Front Immunol 2021; 11:617804. [PMID: 33664731 PMCID: PMC7921745 DOI: 10.3389/fimmu.2020.617804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a hardening and narrowing of arteries causing a reduction of blood flow. It is a leading cause of death in industrialized countries as it causes heart attacks, strokes, and peripheral vascular disease. Pathogenesis of the atherosclerotic lesion (atheroma) relies on the accumulation of cholesterol-containing low-density lipoproteins (LDL) and on changes of artery endothelium that becomes adhesive for monocytes and lymphocytes. Immunomediated inflammatory response stimulated by lipoprotein oxidation, cytokine secretion and release of pro-inflammatory mediators, worsens the pathological context by amplifying tissue damage to the arterial lining and increasing flow-limiting stenosis. Formation of thrombi upon rupture of the endothelium and the fibrous cup may also occur, triggering thrombosis often threatening the patient’s life. Purinergic signaling, i.e., cell responses induced by stimulation of P2 and P1 membrane receptors for the extracellular nucleotides (ATP, ADP, UTP, and UDP) and nucleosides (adenosine), has been implicated in modulating the immunological response in atherosclerotic cardiovascular disease. In this review we will describe advancements in the understanding of purinergic modulation of the two main immune cells involved in atherogenesis, i.e., monocytes/macrophages and T lymphocytes, highlighting modulation of pro- and anti-atherosclerotic mediated responses of purinergic signaling in these cells and providing new insights to point out their potential clinical significance.
Collapse
Affiliation(s)
- Davide Ferrari
- Department of Life Science and Biotechnology, Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
| | - Andrea la Sala
- Certification Unit, Health Directorate, Bambino Gesù Pediatric Hospital, IRCCS, Rome, Italy
| | - Daniela Milani
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Claudio Celeghini
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
93
|
Nurden P, Stritt S, Favier R, Nurden AT. Inherited platelet diseases with normal platelet count: phenotypes, genotypes and diagnostic strategy. Haematologica 2021; 106:337-350. [PMID: 33147934 PMCID: PMC7849565 DOI: 10.3324/haematol.2020.248153] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
Inherited platelet disorders resulting from platelet function defects and a normal platelet count cause a moderate or severe bleeding diathesis. Since the description of Glanzmann thrombasthenia resulting from defects of ITGA2B and ITGB3, new inherited platelet disorders have been discovered, facilitated by the use of high throughput sequencing and genomic analyses. Defects of RASGRP2 and FERMT3 responsible for severe bleeding syndromes and integrin activation have illustrated the critical role of signaling molecules. Important are mutations of P2RY12 encoding the major ADP receptor causal for an inherited platelet disorder with inheritance characteristics that depend on the variant identified. Interestingly, variants of GP6 encoding the major subunit of the collagen receptor GPVI/FcRγ associate only with mild bleeding. The numbers of genes involved in dense granule defects including Hermansky-Pudlak and Chediak Higashi syndromes continue to progress and are updated. The ANO6 gene encoding a Ca2+-activated ion channel required for phospholipid scrambling is responsible for the rare Scott syndrome and decreased procoagulant activity. A novel EPHB2 defect in a familial bleeding syndrome demonstrates a role for this tyrosine kinase receptor independent of the classical model of its interaction with ephrins. Such advances highlight the large diversity of variants affecting platelet function but not their production, despite the difficulties in establishing a clear phenotype when few families are affected. They have provided insights into essential pathways of platelet function and have been at the origin of new and improved therapies for ischemic disease. Nevertheless, many patients remain without a diagnosis and requiring new strategies that are now discussed.
Collapse
Affiliation(s)
| | - Simon Stritt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala
| | - Remi Favier
- French National Reference Center for Inherited Platelet Disorders, Armand Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris
| | | |
Collapse
|
94
|
Maeda J, Minamihisamatsu T, Shimojo M, Zhou X, Ono M, Matsuba Y, Ji B, Ishii H, Ogawa M, Akatsu H, Kaneda D, Hashizume Y, Robinson JL, Lee VMY, Saito T, Saido TC, Trojanowski JQ, Zhang MR, Suhara T, Higuchi M, Sahara N. Distinct microglial response against Alzheimer's amyloid and tau pathologies characterized by P2Y12 receptor. Brain Commun 2021; 3:fcab011. [PMID: 33644757 PMCID: PMC7901060 DOI: 10.1093/braincomms/fcab011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/11/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Microglia are the resident phagocytes of the central nervous system, and microglial activation is considered to play an important role in the pathogenesis of neurodegenerative diseases. Recent studies with single-cell RNA analysis of CNS cells in Alzheimer's disease and diverse other neurodegenerative conditions revealed that the transition from homeostatic microglia to disease-associated microglia was defined by changes of gene expression levels, including down-regulation of the P2Y12 receptor gene (P2Y12R). However, it is yet to be clarified in Alzheimer's disease brains whether and when this down-regulation occurs in response to amyloid-β and tau depositions, which are core pathological processes in the disease etiology. To further evaluate the significance of P2Y12 receptor alterations in the neurodegenerative pathway of Alzheimer's disease and allied disorders, we generated an anti-P2Y12 receptor antibody and examined P2Y12 receptor expressions in the brains of humans and model mice bearing amyloid-β and tau pathologies. We observed that the brains of both Alzheimer's disease and non-Alzheimer's disease tauopathy patients and tauopathy model mice (rTg4510 and PS19 mouse lines) displayed declined microglial P2Y12 receptor levels in regions enriched with tau inclusions, despite an increase in the total microglial population. Notably, diminution of microglial immunoreactivity with P2Y12 receptor was noticeable prior to massive accumulations of phosphorylated tau aggregates and neurodegeneration in rTg4510 mouse brains, despite a progressive increase of total microglial population. On the other hand, Iba1-positive microglia encompassing compact and dense-cored amyloid-β plaques expressed P2Y12 receptor at varying levels in amyloid precursor protein (APP) mouse models (APP23 and AppNL-F/NL-F mice). By contrast, neuritic plaques in Alzheimer's disease brains were associated with P2Y12 receptor-negative microglia. These data suggest that the down-regulation of microglia P2Y12 receptor, which is characteristic of disease-associated microglia, is intimately associated with tau rather than amyloid-β pathologies from an early stage and could be a sensitive index for neuroinflammatory responses to Alzheimer's disease-related neurodegenerative processes.
Collapse
Affiliation(s)
- Jun Maeda
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takeharu Minamihisamatsu
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masafumi Shimojo
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Xiaoyun Zhou
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yukio Matsuba
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Bin Ji
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hideki Ishii
- Department of Advanced Nuclear Medicine Science, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Science, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroyasu Akatsu
- Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Aichi, Japan.,Department of Community-based Medical Education, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Daita Kaneda
- Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Aichi, Japan
| | - Yoshio Hashizume
- Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Aichi, Japan
| | - John L Robinson
- Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-2674, USA
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-2674, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-2674, USA
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Science, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan.,Department of Advanced Nuclear Medicine Science, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Aichi, Japan.,Department of Community-based Medical Education, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan.,Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-2674, USA.,Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| |
Collapse
|
95
|
Cao X, Ye X, Zhang S, Wang L, Xu Y, Peng S, Zhou Y, Peng Y, Li J, Zhang X, Han X, Huang WY, Jia W, Ye J. ADP Induces Blood Glucose Through Direct and Indirect Mechanisms in Promotion of Hepatic Gluconeogenesis by Elevation of NADH. Front Endocrinol (Lausanne) 2021; 12:663530. [PMID: 33986729 PMCID: PMC8111448 DOI: 10.3389/fendo.2021.663530] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Extracellular ADP, a derivative of ATP, interacts with the purinergic receptors in the cell membrane to regulate cellular activities. This signaling pathway remains unknown in the regulation of blood glucose in vivo. We investigated the acute activity of ADP in mice through a peritoneal injection. In the lean mice, in response to the ADP treatment, the blood glucose was elevated, and pyruvate tolerance was impaired. Hepatic gluconeogenesis was enhanced with elevated expression of glucogenic genes (G6pase and Pck1) in the liver. An elevation was observed in NADH, cAMP, AMP, GMP and citrate in the liver tissue in the targeted metabolomics assay. In the primary hepatocytes, ADP activated the cAMP/PKA/CREB signaling pathway, which was blocked by the antagonist (2211) of the ADP receptor P2Y13. In the circulation, gluconeogenic hormones including glucagon and corticosterone were elevated by ADP. Insulin and thyroid hormones (T3 and T4) were not altered in the blood. In the diet-induced obese (DIO) mice, NADH was elevated in the liver tissue to match the hepatic insulin resistance. Insulin resistance was intensified by ADP for further impairment in insulin tolerance. These data suggest that ADP induced the blood glucose through direct and indirect actions in liver. One of the potential pathways involves activation of the P2Y13/cAMP/PKA/CREB signaling pathway in hepatocytes and the indirect pathway may involve induction of the gluconeogenic hormones. NADH is a signal for gluconeogenesis in the liver of both DIO mice and lean mice.
Collapse
Affiliation(s)
- Xinyu Cao
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaotong Ye
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Shuang Zhang
- Core Facility Center of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Wang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yanhong Xu
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shiqiao Peng
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yang Zhou
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yue Peng
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- College of Physical Education, Jiangxi Normal University, Nanchang City, China
| | - Junhua Li
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaoying Zhang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Wen-ying Huang
- College of Physical Education, Jiangxi Normal University, Nanchang City, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jianping Ye
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Jianping Ye,
| |
Collapse
|
96
|
Schöneberg T, Liebscher I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol Rev 2021; 73:89-119. [PMID: 33219147 DOI: 10.1124/pharmrev.120.000011] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There are approximately 800 annotated G protein-coupled receptor (GPCR) genes, making these membrane receptors members of the most abundant gene family in the human genome. Besides being involved in manifold physiologic functions and serving as important pharmacotherapeutic targets, mutations in 55 GPCR genes cause about 66 inherited monogenic diseases in humans. Alterations of nine GPCR genes are causatively involved in inherited digenic diseases. In addition to classic gain- and loss-of-function variants, other aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, pseudogenes, gene fusion, and gene dosage, contribute to the repertoire of GPCR dysfunctions. However, the spectrum of alterations and GPCR involvement is probably much larger because an additional 91 GPCR genes contain homozygous or hemizygous loss-of-function mutations in human individuals with currently unidentified phenotypes. This review highlights the complexity of genomic alteration of GPCR genes as well as their functional consequences and discusses derived therapeutic approaches. SIGNIFICANCE STATEMENT: With the advent of new transgenic and sequencing technologies, the number of monogenic diseases related to G protein-coupled receptor (GPCR) mutants has significantly increased, and our understanding of the functional impact of certain kinds of mutations has substantially improved. Besides the classical gain- and loss-of-function alterations, additional aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, uniparental disomy, pseudogenes, gene fusion, and gene dosage, need to be elaborated in light of GPCR dysfunctions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| |
Collapse
|
97
|
|
98
|
Aparna A, Kumar YS, Bhikshapathi DVRN. Formulation and In Vivo Evaluation of Ticagrelor Self-nanoemulsifying Drug Delivery Systems. Pharm Nanotechnol 2021; 9:61-69. [PMID: 32640972 DOI: 10.2174/2211738508666200708150151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/23/2020] [Accepted: 06/12/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ticagrelor (TGR), being an antiplatelet agent, belongs to BCS class IV drug with low solubility and permeability that undergoes first-pass metabolism, leading to reduced bioavailability of 36%. OBJECTIVE The main objective of this study is to develop TGR SNEDDS for enhancing solubility and oral bioavailability. METHODS An oil, surfactant and co-surfactant (miglyol 810, brij 35 and lauro glycol FCC) are chosen based on the maximum solubility of TGR. The selected vehicles are mixed in different ratios and are agitated mildly. Transmittance values that are more than 80 were noted and are used for constructing pseudo ternary phase diagram. Formulations that passed stability testing were evaluated for % transmission, drug content and in vitro drug release analysis. In vivo bioavailability studies of optimized SNEDDS are performed in Wistar rats. RESULTS From evaluation studies of TGR, formulation F13 with maximum drug release of 98.99% in 60 minutes, that is higher than 31.99% of the pure drug is considered as an optimised formulation. The particle size, Z average and zeta potential of the optimized TGR formulation F13 was 289.6 nm, 185.1 nm and -18.3 mV respectively. The FTIR and SEM studies do not indicate any drug excipient interaction and confirm nano size which is stable for 3 months. From in vivo bioavailability studies in rats, the Cmax of optimized TGR SNEDDS (302.43±4.78 ng/ml) is higher than pure TGR suspension (47.32±2.75 ng/ml) and optimized SNEDDS exhibited 5 folds increase in oral bioavailability when compared to pure drug. CONCLUSION Hence the results reveal that, application of SNEDDS formulation technique for TGR Increases solubility and oral bioavailability.
Collapse
Affiliation(s)
- Adella Aparna
- Mewar University, Chittorgarh-312901, Rajasthan, India
| | | | | |
Collapse
|
99
|
Woo SH, Trinh TN. P2 Receptors in Cardiac Myocyte Pathophysiology and Mechanotransduction. Int J Mol Sci 2020; 22:ijms22010251. [PMID: 33383710 PMCID: PMC7794727 DOI: 10.3390/ijms22010251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 12/30/2022] Open
Abstract
ATP is a major energy source in the mammalian cells, but it is an extracellular chemical messenger acting on P2 purinergic receptors. A line of evidence has shown that ATP is released from many different types of cells including neurons, endothelial cells, and muscle cells. In this review, we described the distribution of P2 receptor subtypes in the cardiac cells and their physiological and pathological roles in the heart. So far, the effects of external application of ATP or its analogues, and those of UTP on cardiac contractility and rhythm have been reported. In addition, specific genetic alterations and pharmacological agonists and antagonists have been adopted to discover specific roles of P2 receptor subtypes including P2X4-, P2X7-, P2Y2- and P2Y6-receptors in cardiac cells under physiological and pathological conditions. Accumulated data suggest that P2X4 receptors may play a beneficial role in cardiac muscle function, and that P2Y2- and P2Y6-receptors can induce cardiac fibrosis. Recent evidence further demonstrates P2Y1 receptor and P2X4 receptor as important mechanical signaling molecules to alter membrane potential and Ca2+ signaling in atrial myocytes and their uneven expression profile between right and left atrium.
Collapse
|
100
|
PLGA-PEG Nanoparticles Show Minimal Risks of Interference with Platelet Function of Human Platelet-Rich Plasma. Int J Mol Sci 2020; 21:ijms21249716. [PMID: 33352749 PMCID: PMC7767100 DOI: 10.3390/ijms21249716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 01/14/2023] Open
Abstract
The expansion of nanotechnology for drug delivery applications has raised questions regarding the safety of nanoparticles (NPs) due to their potential for interacting at molecular and cellular levels. Although polymeric NPs for drug delivery are formulated using FDA-approved polymers such as lactide- and glycolide-based polymers, their interactions with blood constituents, remain to be identified. The aim of this study was to determine the impact of size-selected Poly-lactide-co-glycolide-polyethylene glycol (PLGA-PEG) NPs on platelet activity. The NPs of 113, 321, and 585 nm sizes, were formulated and their effects at concentrations of 0–2.2 mg/mL on the activation and aggregation of platelet-rich plasma (PRP) were investigated. The results showed that NPs of 113 nm did not affect adenosine diphosphate (ADP)-induced platelet aggregation at any NP concentration studied. The NPs of 321 and 585 nm, at concentrations ≥0.25 mg/mL, reduced ADP-activated platelet aggregation. The platelet activation profile remained unchanged in the presence of investigated NPs. Confocal microscopy revealed that NPs were attached to or internalised by platelets in both resting and activated states, with no influence on platelet reactivity. The results indicate minimal risks of interference with platelet function for PLGA-PEG NPs and that these NPs can be explored as nanocarriers for targeted drug delivery to platelets.
Collapse
|