1
|
Wang Z, Wang S, Wang H, Hu B, Qi Z, Zhang Y, Song P, Cai Q, Yang H, Wang J. Uncovering the selectivity mechanism of phosphodiesterase 7A/8A inhibitors through computational studies. Phys Chem Chem Phys 2024; 26:11770-11781. [PMID: 38566586 DOI: 10.1039/d3cp03913g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The expression of phosphodiesterase 7A (PDE7A) and phosphodiesterase 8A (PDE8) genes is integral to human signaling pathways, and the inhibition of PDE7A has been associated with the onset of various diseases, including effects on the immune system and nervous system. The development of PDE7 selective inhibitors can promote research on immune and nervous system diseases, such as multiple sclerosis, chronic inflammation, and autoimmune responses. PDE8A is expressed alongside PDE8B, and its inhibitory mechanism is still unclear. Studying the mechanisms of selective inhibitors against different PDE subtypes is crucial to prevent potential side effects, such as nausea and cardiac toxicity, and the sequence similarity of the two protein subtypes was 55.9%. Therefore, it is necessary to investigate the differences of both subtypes' ligand binding sites. Selective inhibitors of two proteins were chosen to summarize the reason for their selectivity through molecular docking, molecular dynamics simulation, alanine scanning mutagenesis, and MM-GBSA calculation. We found that Phe384PDE7A, Leu401PDE7A, Gln413PDE7A, Tyr419PDE7A, and Phe416PDE7A in the active site positively contribute to the selectivity towards PDE7A. Additionally, Asn729PDE8A, Phe767PDE8A, Gln778PDE8A, and Phe781PDE8A positively contribute to the selectivity towards PDE8A.
Collapse
Affiliation(s)
- Zhijian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shizun Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Hanxun Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Baichun Hu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Zhuo Qi
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yaming Zhang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Pengfei Song
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Qingkui Cai
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Huali Yang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Jian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
2
|
Watanabe T, Yamamoto Y, Kurahashi Y, Kawasoe K, Kidoguchi K, Ureshino H, Kamachi K, Yoshida-Sakai N, Fukuda-Kurahashi Y, Nakamura H, Okada S, Sueoka E, Kimura S. Reprogramming of pyrimidine nucleotide metabolism supports vigorous cell proliferation of normal and malignant T cells. Blood Adv 2024; 8:1345-1358. [PMID: 38190613 PMCID: PMC10945144 DOI: 10.1182/bloodadvances.2023011131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Abstract
ABSTRACT Adult T-cell leukemia/lymphoma (ATL) is triggered by infection with human T-cell lymphotropic virus-1 (HTLV-1). Here, we describe the reprogramming of pyrimidine biosynthesis in both normal T cells and ATL cells through regulation of uridine-cytidine kinase 2 (UCK2), which supports vigorous proliferation. UCK2 catalyzes the monophosphorylation of cytidine/uridine and their analogues during pyrimidine biosynthesis and drug metabolism. We found that UCK2 was overexpressed aberrantly in HTLV-1-infected T cells but not in normal T cells. T-cell activation via T-cell receptor (TCR) signaling induced expression of UCK2 in normal T cells. Somatic alterations and epigenetic modifications in ATL cells activate TCR signaling. Therefore, we believe that expression of UCK2 in HTLV-1-infected cells is induced by dysregulated TCR signaling. Recently, we established azacitidine-resistant (AZA-R) cells showing absent expression of UCK2. AZA-R cells proliferated normally in vitro, whereas UCK2 knockdown inhibited ATL cell growth. Although uridine and cytidine accumulated in AZA-R cells, possibly because of dysfunction of pyrimidine salvage biosynthesis induced by loss of UCK2 expression, the amount of UTP and CTP was almost the same as in parental cells. Furthermore, AZA-R cells were more susceptible to an inhibitor of dihydroorotic acid dehydrogenase, which performs the rate-limiting enzyme of de novo pyrimidine nucleotide biosynthesis, and more resistant to dipyridamole, an inhibitor of pyrimidine salvage biosynthesis, suggesting that AZA-R cells adapt to UCK2 loss by increasing de novo pyrimidine nucleotide biosynthesis. Taken together, the data suggest that fine-tuning pyrimidine biosynthesis supports vigorous cell proliferation of both normal T cells and ATL cells.
Collapse
Affiliation(s)
- Tatsuro Watanabe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuta Yamamoto
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuki Kurahashi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- OHARA Pharmaceutical Co, Ltd, Shiga, Japan
| | - Kazunori Kawasoe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Keisuke Kidoguchi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroshi Ureshino
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Kazuharu Kamachi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Nao Yoshida-Sakai
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuki Fukuda-Kurahashi
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- OHARA Pharmaceutical Co, Ltd, Shiga, Japan
| | - Hideaki Nakamura
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinya Kimura
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Saga, Japan
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
3
|
Stanger L, Yamaguchi A, Holinstat M. Antiplatelet strategies: past, present, and future. J Thromb Haemost 2023; 21:3317-3328. [PMID: 38000851 PMCID: PMC10683860 DOI: 10.1016/j.jtha.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 11/26/2023]
Abstract
Antiplatelet therapy plays a critical role in the prevention and treatment of major cardiovascular diseases triggered by thrombosis. Since the 1900s, significant progress in reducing morbidity and death caused by cardiovascular diseases has been made. However, despite the development and approval of drugs that specifically target the platelet, including inhibitors for cycloxygenase-1, P2Y12 receptor, integrin αIIbβ3, phosphodiesterases, and protease-activated receptor 1, the risk of recurrent thrombotic events remains high, and the increased risk of bleeding is a major concern. Scientific advances in our understanding of the role of platelets in haemostasis and thrombosis have revealed novel targets, such as protease-activated receptor 4 (PAR4), glycoprotein Ib (GPIb)-V-IX complex, glycoprotein VI, and 12-lipoxygenase. The antithrombotic effects and safety of the pharmacologic inhibition of these targets are currently under investigation in clinical studies. This review provides an overview of drugs in early development to target the platelet and those in current use in clinical practice. Furthermore, it describes the emerging drug targets being developed and studied to reduce platelet activity and outlines potential novel therapeutic targets in the platelet.
Collapse
Affiliation(s)
- Livia Stanger
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adriana Yamaguchi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Surgery, Division of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
4
|
ElHady AK, El-Gamil DS, Abdel-Halim M, Abadi AH. Advancements in Phosphodiesterase 5 Inhibitors: Unveiling Present and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:1266. [PMID: 37765073 PMCID: PMC10536424 DOI: 10.3390/ph16091266] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Phosphodiesterase 5 (PDE5) inhibitors presented themselves as important players in the nitric oxide/cGMP pathway, thus exerting a profound impact on various physiological and pathological processes. Beyond their well-known efficacy in treating male erectile dysfunction (ED) and pulmonary arterial hypertension (PAH), a plethora of studies have unveiled their significance in the treatment of a myriad of other diseases, including cognitive functions, heart failure, multiple drug resistance in cancer therapy, immune diseases, systemic sclerosis and others. This comprehensive review aims to provide an updated assessment of the crucial role played by PDE5 inhibitors (PDE5-Is) as disease-modifying agents taking their limiting side effects into consideration. From a medicinal chemistry and drug discovery perspective, the published PDE5-Is over the last 10 years and their binding characteristics are systemically discussed, and advancement in properties is exposed. A persistent challenge encountered with these agents lies in their limited isozyme selectivity; considering this obstacle, this review also highlights the breakthrough development of the recently reported PDE5 allosteric inhibitors, which exhibit an unparalleled level of selectivity that was rarely achievable by competitive inhibitors. The implications and potential impact of these novel allosteric inhibitors are meticulously explored. Additionally, the concept of multi-targeted ligands is critically evaluated in relation to PDE5-Is by inspecting the broader spectrum of their molecular interactions and effects. The objective of this review is to provide insight into the design of potent, selective PDE5-Is and an overview of their biological function, limitations, challenges, therapeutic potentials, undergoing clinical trials, future prospects and emerging uses, thus guiding upcoming endeavors in both academia and industry within this domain.
Collapse
Affiliation(s)
- Ahmed K. ElHady
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo 11865, Egypt;
| | - Dalia S. El-Gamil
- Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt;
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| | - Ashraf H. Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| |
Collapse
|
5
|
Kennerly R, Coleman AE, Jasper SL, Arnold RD, Brainard BM. Effects of orally administered pimobendan on platelet function in healthy adult cats. Am J Vet Res 2023; 84:ajvr.23.05.0103. [PMID: 37491004 DOI: 10.2460/ajvr.23.05.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
OBJECTIVE Several phosphodiesterase inhibitors have demonstrable antiplatelet actions when administered to human patients. Concentration-dependent inhibition of feline platelet aggregation by pimobendan has been previously demonstrated in vitro. However, there are no published reports characterizing the effect of oral pimobendan, administered at therapeutic doses, on platelet function in cats. This study aimed to evaluate the effect of orally administered pimobendan on platelet function in healthy adult cats. ANIMALS 6 healthy purpose-bred adult cats. METHODS Cats were administered pimobendan orally at a dosage of 0.625 mg/cat (low-dose) twice daily for 1 week, followed by 1.25 mg/cat (high-dose) twice daily for 1 week. Venous blood sampling for platelet testing and plasma drug concentration occurred at baseline, 1 hour postdose on the eighth day of treatment with low-dose pimobendan, 1 hour postdose on the eighth day of treatment with high-dose pimobendan, and after a 1-week washout period. Platelet function was assessed by whole blood aggregometry and by use of a platelet function analyzer (PFA-100®). Friedman tests were used to compare platelet function parameters among the 4 sampling timepoints. RESULTS After 1 week of treatment, median (range) plasma pimobendan concentrations were 15.1 ng/mL (6.89-20.2 ng/mL) and 32.8 ng/mL (23.3-44.8 ng/mL) in cats receiving low-dose and high-dose pimobendan, respectively. No significant differences in PFA closure time or any aggregometry variable were found among the treatment conditions. CLINICAL RELEVANCE Pimobendan was not associated with measurable inhibition of platelet function when administered orally to healthy adult cats at 2 clinically relevant dosages.
Collapse
Affiliation(s)
- Rebecca Kennerly
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Amanda E Coleman
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Shanese L Jasper
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Robert D Arnold
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Benjamin M Brainard
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| |
Collapse
|
6
|
Kubacka M, Mogilski S, Bednarski M, Pociecha K, Świerczek A, Nicosia N, Schabikowski J, Załuski M, Chłoń-Rzepa G, Hockemeyer J, Müller CE, Kieć-Kononowicz K, Kotańska M. Antiplatelet Effects of Selected Xanthine-Based Adenosine A 2A and A 2B Receptor Antagonists Determined in Rat Blood. Int J Mol Sci 2023; 24:13378. [PMID: 37686188 PMCID: PMC10487961 DOI: 10.3390/ijms241713378] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The platelet aggregation inhibitory activity of selected xanthine-based adenosine A2A and A2B receptor antagonists was investigated, and attempts were made to explain the observed effects. The selective A2B receptor antagonist PSB-603 and the A2A receptor antagonist TB-42 inhibited platelet aggregation induced by collagen or ADP. In addition to adenosine receptor blockade, the compounds were found to act as moderately potent non-selective inhibitors of phosphodiesterases (PDEs). TB-42 showed the highest inhibitory activity against PDE3A along with moderate activity against PDE2A and PDE5A. The antiplatelet activity of PSB-603 and TB-42 may be due to inhibition of PDEs, which induces an increase in cAMP and/or cGMP concentrations in platelets. The xanthine-based adenosine receptor antagonists were found to be non-cytotoxic for platelets. Some of the compounds showed anti-oxidative properties reducing lipid peroxidation. These results may provide a basis for the future development of multi-target xanthine derivatives for the treatment of inflammation and atherosclerosis and the prevention of heart infarction and stroke.
Collapse
Affiliation(s)
- Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.K.); (S.M.)
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.K.); (S.M.)
| | - Marek Bednarski
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
| | - Krzysztof Pociecha
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (K.P.); (A.Ś.)
| | - Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (K.P.); (A.Ś.)
| | - Noemi Nicosia
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
- Division of Neuroscience, Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Jakub Schabikowski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Michał Załuski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland;
| | - Jörg Hockemeyer
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, An der Immenburg 4, D-53121 Bonn, Germany; (J.H.); (C.E.M.)
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, An der Immenburg 4, D-53121 Bonn, Germany; (J.H.); (C.E.M.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (J.S.); (M.Z.); (K.K.-K.)
| | - Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (M.B.); (N.N.)
| |
Collapse
|
7
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
8
|
Affiliation(s)
- Zoltan Nagy
- Institute of Experimental Biomedicine, University of Würzburg, University Hospital, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University of Würzburg, University Hospital, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Gui X, Chu X, Du Y, Wang Y, Zhang S, Ding Y, Tong H, Xu M, Li Y, Ju W, Sun Z, Li Z, Zeng L, Xu K, Qiao J. Impaired Platelet Function and Thrombus Formation in PDE5A-Deficient Mice. Thromb Haemost 2023; 123:207-218. [PMID: 36252813 DOI: 10.1055/a-1962-1613] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intracellular cyclic GMP (cGMP) inhibits platelet function. Platelet cGMP levels are controlled by phosphodiesterase 5A (PDE5A)-mediated degradation. However, the exact role of PDE5A in platelet function and thrombus formation remains poorly understood. In this study, we characterized the role of PDE5A in platelet activation and function. Platelets were isolated from wild type or PDE5A-/- mice to measure platelet aggregation, activation, phosphatidylserine exposure (annexin-V binding), reactive oxygen species (ROS) generation, platelet spreading as well as clot retraction. Cytosolic calcium mobilization was measured using Fluo-4 AM by a microplate reader. Western blot was used to measure the phosphorylation of VASP, ERK1/2, p38, JNK, and AKT. FeCl3-induced arterial thrombosis and venous thrombosis were assessed to evaluate the in vivo hemostatic function and thrombus formation. Additionally, in vitro thrombus formation was assessed in a microfluidic whole-blood perfusion assay. PDE5A-deficient mice presented significantly prolonged tail bleeding time and delayed arterial and venous thrombus formation. PDE5A deficiency significantly inhibited platelet aggregation, ATP release, P-selectin expression, and integrin aIIbb3 activation. In addition, an impaired spreading on collagen or fibrinogen and clot retraction was observed in PDE5A-deficient platelets. Moreover, PDE5A deficiency reduced phosphatidylserine exposure, calcium mobilization, ROS production, and increased intracellular cGMP level along with elevated VASP phosphorylation and reduced phosphorylation of ERK1/2, p38, JNK, and AKT. In conclusion, PDE5A modulates platelet activation and function and thrombus formation, indicating that therapeutically targeting it might be beneficial for the treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Xiang Chu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuwei Du
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuhan Wang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zengtian Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China.,School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| |
Collapse
|
10
|
Protein tyrosine phosphatase PTPN22 negatively modulates platelet function and thrombus formation. Blood 2022; 140:1038-1051. [PMID: 35767715 DOI: 10.1182/blood.2022015554] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is a protein tyrosine phosphatase that negatively regulates T-cell signaling. However, whether it is expressed and functions in platelets remains unknown. Here we investigated the expression and role of PTPN22 in platelet function. We reported PTPN22 expression in both human and mouse platelets. Using PTPN22-/- mice, we showed that PTPN22 deficiency significantly shortened tail-bleeding time and accelerated arterial thrombus formation without affecting venous thrombosis and the coagulation factors VIII and IX. Consistently, PTPN22-deficient platelets exhibited enhanced platelet aggregation, granule secretion, calcium mobilization, lamellipodia formation, spreading, and clot retraction. Quantitative phosphoproteomic analysis revealed the significant difference of phosphodiesterase 5A (PDE5A) phosphorylation in PTPN22-deficient platelets compared with wild-type platelets after collagen-related peptide stimulation, which was confirmed by increased PDE5A phosphorylation (Ser92) in collagen-related peptide-treated PTPN22-deficient platelets, concomitant with reduced level and vasodilator-stimulated phosphoprotein phosphorylation (Ser157/239). In addition, PTPN22 interacted with phosphorylated PDE5A (Ser92) and dephosphorylated it in activated platelets. Moreover, purified PTPN22 but not the mutant form (C227S) possesses intrinsic serine phosphatase activity. Furthermore, inhibition of PTPN22 enhanced human platelet aggregation, spreading, clot retraction, and increased PDE5A phosphorylation (Ser92). In conclusion, our study shows a novel role of PTPN22 in platelet function and arterial thrombosis, identifying new potential targets for future prevention of thrombotic or cardiovascular diseases.
Collapse
|
11
|
Quantification of cyclic AMP and cyclic GMP levels in Krebs-Henseleit solution by LC-MS/MS: application in washed platelet aggregation samples. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1211:123472. [DOI: 10.1016/j.jchromb.2022.123472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
|
12
|
Zhang Y, Benz P, Stehle D, Yang S, Kurz H, Feil S, Nagel G, Feil R, Gao S, Bender M. Optogenetic manipulation of cyclic guanosine monophosphate to probe phosphodiesterase activities in megakaryocytes. Open Biol 2022; 12:220058. [PMID: 35975649 PMCID: PMC9382455 DOI: 10.1098/rsob.220058] [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] [Indexed: 11/23/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP) signalling plays a fundamental role in many cell types, including platelets. cGMP has been implicated in platelet formation, but mechanistic detail about its spatio-temporal regulation in megakaryocytes (MKs) is lacking. Optogenetics is a technique which allows spatio-temporal manipulation of molecular events in living cells or organisms. We took advantage of this method and expressed a photo-activated guanylyl cyclase, Blastocladiella emersonii Cyclase opsin (BeCyclop), after viral-mediated gene transfer in bone marrow (BM)-derived MKs to precisely light-modulate cGMP levels. BeCyclop-MKs showed a significantly increased cGMP concentration after illumination, which was strongly dependent on phosphodiesterase (PDE) 5 activity. This finding was corroborated by real-time imaging of cGMP signals which revealed that pharmacological PDE5 inhibition also potentiated nitric oxide-triggered cGMP generation in BM MKs. In summary, we established for the first-time optogenetics in primary MKs and show that PDE5 is the predominant PDE regulating cGMP levels in MKs. These findings also demonstrate that optogenetics allows for the precise manipulation of MK biology.
Collapse
Affiliation(s)
- Yujing Zhang
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Pascal Benz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shang Yang
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Hendrikje Kurz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| |
Collapse
|
13
|
Saeed ZM, Khattab MI, Khorshid NE, Salem AE. Ellagic acid and cilostazol ameliorate amikacin-induced nephrotoxicity in rats by downregulating oxidative stress, inflammation, and apoptosis. PLoS One 2022; 17:e0271591. [PMID: 35849599 PMCID: PMC9292089 DOI: 10.1371/journal.pone.0271591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 07/04/2022] [Indexed: 01/24/2023] Open
Abstract
Amikacin (AK) has the largest spectrum of aminoglycosides. However, its use is constrained because of nephrotoxicity and ototoxicity. Ellagic acid (EA) is a polyphenol present in plants. It has antioxidant, anticarcinogenic, and antimutagenic characteristics. Cilostazol (CTZ) is a phosphodiesterase Ш inhibitor, it is a potent vasodilator and antiplatelet drug. CTZ has an inhibitory effect on reactive oxygen species and superoxide generation in addition to hydroxyl radicals scavenging action. This study determines whether EA and cilostazol have a protective effect against AK-induced nephrotoxicity. Forty-nine rats were divided into seven equal groups: control normal; AK 400 mg/kg; EA 10 mg/kg; CTZ 10 mg/kg; AK 400 mg/kg plus EA 10 mg/kg; AK 400 mg/kg plus CTZ 10 mg/kg; AK 400 mg/kg plus EA 10 mg/kg and CTZ 10 mg/kg. For seven days, drugs were administered using gavage one hour before intramuscular injection of AK. Twenty-four hours after the last AK dosage, blood samples were collected to determine blood urea nitrogen and creatinine levels. Kidneys were removed for histopathological examination and measurement of: malondialdehyde (MDA), catalase (CAT), decreased glutathione (GSH), superoxide dismutase (SOD), interleukin 6 (IL6), tumor necrosis factor-alpha (TNFα), nuclear factor kappa B (NFκB), and Bcl-2 associated x protein (BAX). AK caused kidney damage, inflammatory mediator elevation, and oxidative stress and apoptotic markers. Rats receiving EA or CTZ indicated significant improvement in kidney function, decrease in oxidative stress and inflammation through NF-kB down-regulation and BAX expression. The combination of EA and CTZ showed a synergistic effect. In conclusion, EA and CTZ might play a beneficial role in preventing nephrotoxicity induced by AK partially by inhibition of tissue inflammation and apoptosis.
Collapse
Affiliation(s)
- Zeinab Mahmoud Saeed
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- * E-mail:
| | - Monira Ismail Khattab
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nadia Esmat Khorshid
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amal Elsayed Salem
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
14
|
Root‐Bernstein R. COVID-19 coagulopathies: Human blood proteins mimic SARS-CoV-2 virus, vaccine proteins and bacterial co-infections inducing autoimmunity: Combinations of bacteria and SARS-CoV-2 synergize to induce autoantibodies targeting cardiolipin, cardiolipin-binding proteins, platelet factor 4, prothrombin, and coagulation factors. Bioessays 2021; 43:e2100158. [PMID: 34677872 PMCID: PMC8646673 DOI: 10.1002/bies.202100158] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022]
Abstract
Severe COVID-19 is often accompanied by coagulopathies such as thrombocytopenia and abnormal clotting. Rarely, such complications follow SARS-CoV-2 vaccination. The cause of these coagulopathies is unknown. It is hypothesized that coagulopathies accompanying SARS-CoV-2 infections and vaccinations result from bacterial co-infections that synergize with virus-induced autoimmunity due to antigenic mimicry of blood proteins by both bacterial and viral antigens. Coagulopathies occur mainly in severe COVID-19 characterized by bacterial co-infections with Streptococci, Staphylococci, Klebsiella, Escherichia coli, and Acinetobacter baumannii. These bacteria express unusually large numbers of antigens mimicking human blood antigens, as do both SARS-CoV-2 and adenoviruses. Bacteria mimic cardiolipin, prothrombin, albumin, and platelet factor 4 (PF4). SARS-CoV-2 mimics complement factors, Rh antigens, platelet phosphodiesterases, Factors IX and X, von Willebrand Factor (VWF), and VWF protease ADAMTS13. Adenoviruses mimic prothrombin and platelet factor 4. Bacterial prophylaxis, avoidance of vaccinating bacterially infected individuals, and antigen deletion for vaccines may reduce coagulopathy risk. Also see the video abstract here: https://youtu.be/zWDOsghrPg8.
Collapse
|
15
|
de Havenon A, Sheth KN, Madsen TE, Johnston KC, Turan T, Toyoda K, Elm JJ, Wardlaw JM, Johnston SC, Williams OA, Shoamanesh A, Lansberg MG. Cilostazol for Secondary Stroke Prevention: History, Evidence, Limitations, and Possibilities. Stroke 2021; 52:e635-e645. [PMID: 34517768 PMCID: PMC8478840 DOI: 10.1161/strokeaha.121.035002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cilostazol is a PDE3 (phosphodiesterase III) inhibitor with a long track record of safety that is Food and Drug Administration and European Medicines Agency approved for the treatment of claudication in patients with peripheral arterial disease. In addition, cilostazol has been approved for secondary stroke prevention in several Asian countries based on trials that have demonstrated a reduction in stroke recurrence among patients with noncardioembolic stroke. The onset of benefit appears after 60 to 90 days of treatment, which is consistent with cilostazol's pleiotropic effects on platelet aggregation, vascular remodeling, blood flow, and plasma lipids. Cilostazol appears safe and does not increase the risk of major bleeding when given alone or in combination with aspirin or clopidogrel. Adverse effects such as headache, gastrointestinal symptoms, and palpitations, however, contributed to a 6% increase in drug discontinuation among patients randomized to cilostazol in a large secondary stroke prevention trial (CSPS.com [Cilostazol Stroke Prevention Study for Antiplatelet Combination]). Due to limitations of prior trials, such as open-label design, premature trial termination, large loss to follow-up, lack of functional or cognitive outcome data, and exclusive enrollment in Asia, the existing trials have not led to a change in clinical practice or guidelines in Western countries. These limitations could be addressed by a double-blind placebo-controlled randomized trial conducted in a broader population. If positive, it would increase the evidence in support of long-term treatment with cilostazol for secondary prevention in the millions of patients worldwide who have experienced a noncardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Adam de Havenon
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kevin N. Sheth
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tracy E. Madsen
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Karen C. Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tanya Turan
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kazunori Toyoda
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Jordan J. Elm
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Joanna M. Wardlaw
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - S. Claiborne Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Olajide A. Williams
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Ashkan Shoamanesh
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Maarten G. Lansberg
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| |
Collapse
|
16
|
Mencarini T, Roka-Moiia Y, Bozzi S, Redaelli A, Slepian MJ. Electrical impedance vs. light transmission aggregometry: Testing platelet reactivity to antiplatelet drugs using the MICELI POC impedance aggregometer as compared to a commercial predecessor. Thromb Res 2021; 204:66-75. [PMID: 34147831 PMCID: PMC11416791 DOI: 10.1016/j.thromres.2021.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/08/2021] [Accepted: 05/26/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Patients' responses to antiplatelet therapy significantly vary, with individuals showing high residual platelet reactivity associated with thrombosis. To personalize thrombosis management, platelet function testing has been suggested as a promising tool able to monitor the antithrombotic effect of antiplatelet agents in real-time. We have prototyped the MICELI, a miniature and easy-to-use electrical impedance aggregometer (EIA), measuring platelet aggregation in whole blood. Here, we tested the capability of the MICELI aggregometer to quantify platelet reactivity on antiplatelet agents, as compared with conventional light-transmission aggregometry (LTA). METHODS Platelet aggregation in ACD-anticoagulated whole blood and platelet-rich plasma of healthy donors (n = 30) was evaluated. The effect of clopidogrel, ticagrelor, cangrelor, cilostazol, and tirofiban on ADP-induced aggregation was tested, while aspirin was evaluated with arachidonic acid and collagen. Platelet aggregation was recorded using the MICELI or BioData PAP-8E (Bio/Data Corp.) aggregometers. RESULTS The MICELI aggregometer detected an adequate and comparable dose-dependent decrease of platelet aggregation in response to increments of drugs' concentrations, as compared to LTA (the inter-device R2 = 0.79-0.93). Platelet aggregation in platelet-rich plasma recorded by LTA showed higher sensitivity to antiplatelet agents, but it couldn't distinguish between different drug doses as indicated by saturation of the aggregatory response. CONCLUSION Platelet aggregation in whole blood as recorded by EIA represents a better model system for evaluation of platelet reactivity as compared with platelet aggregation in platelet-rich plasma as recorded by LTA, since EIA takes into consideration the modulatory effect of other blood cells on platelet hemostatic function and pharmacodynamics of antiplatelet drugs in vivo. As such, the MICELI impedance aggregometer could be potentially employed for the point-of-care monitoring of platelet function in patients on-treatment for personalized tailoring of their antiplatelet regimen.
Collapse
Affiliation(s)
- Tatiana Mencarini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Yana Roka-Moiia
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America
| | - Silvia Bozzi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marvin J Slepian
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
17
|
Tong HL, Chen H, Gong FP, Zhong LY, Zhu J, Yang SH. Components and Pharmacodynamical Mechanism of Yinfupian Based on Liquid Chromatography-Mass Spectrometry and Proteomics Analyses. Front Pharmacol 2021; 12:680640. [PMID: 34262452 PMCID: PMC8273442 DOI: 10.3389/fphar.2021.680640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/02/2021] [Indexed: 12/30/2022] Open
Abstract
Objective: According to the treatment records of Yang deficiency syndrome (YDS) with characteristic decoction pieces of lateral root of Aconitum carmichaelii—Yinfupian (YF) in traditional Chinese medicine prepare school, known as “Jianchangbang”. The aim of this study was to investigate differences in the composition and therapeutic mechanism of the unprocessed lateral root of Aconitum carmichaelii (ULRA) and its processed product (YF). Methods: Ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry and orthogonal partial least squares discriminant analysis method were used to determine and screen the main components of ULRA and YF. Changes in the histological structure and morphology of gonads in rats were observed using hematoxylin-eosin. Enzyme-linked immunosorbent assay was used to determine the contents of serum cyclic adenosine monophosphate and cyclic guanosine monophosphate in YDS rats treated with ULRA and YF. Tandem mass tag proteomics analysis was used to identify the differentially expressed proteins in YDS rats treated with ULRA and YF. Results: Both ULRA and YF exerted certain therapeutic effects on rats with YDS. They improved the gonadal morphology and increased the contents of serum cyclic adenosine monophosphate and cyclic guanosine monophosphate. After processing of ULRA into YF, the content of C19-diester-diterpenoid alkaloids decreased (converted into C19-monoester-diterpenoid alkaloids and C19-alkylol amine-diterpenoid alkaloids), whereas that of C20-diterpene alkaloids increased. Proteomics analysis showed that cytochrome P450 and aldehyde oxidase 3 (AOX3) were downregulated, whereas cathepsin G (CTSG) was upregulated in rats with YDS. Treatment with ULRA mainly downregulated the expression of α-actinin, fast skeletal troponin, creatine kinase, and myosin. Treatment with YF mainly upregulated the expression of mitochondrial ribosomal protein and mitochondrial inner membrane protein. Conclusion: ULRA and YF exerted good therapeutic effects on YDS; the main difference in components between these preparations was in C19-diterpenoid alkaloids. ULRA mainly acts on the muscle contraction-related proteins and is closely related to inflammation and myocardial injury. YF mainly acts on the mitochondrial proteins and is closely related to adenosine triphosphate energy metabolism.
Collapse
Affiliation(s)
- Heng-Li Tong
- Laboratory of Traditional Chinese Medicine Preparation, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Hao Chen
- Laboratory of Traditional Chinese Medicine Preparation, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Fei-Peng Gong
- Department of Orthopedic, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Ling-Yun Zhong
- Laboratory of Traditional Chinese Medicine Preparation, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jing Zhu
- Laboratory of Traditional Chinese Medicine Preparation, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Song-Hong Yang
- Laboratory of Traditional Chinese Medicine Preparation, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
18
|
Feil R, Lehners M, Stehle D, Feil S. Visualising and understanding cGMP signals in the cardiovascular system. Br J Pharmacol 2021; 179:2394-2412. [PMID: 33880767 DOI: 10.1111/bph.15500] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/14/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022] Open
Abstract
cGMP is an important signalling molecule in humans. Fluorescent cGMP biosensors have emerged as powerful tools for the sensitive analysis of cGMP pathways at the single-cell level. Here, we briefly outline cGMP's multifaceted role in (patho)physiology and pharmacotherapy. Then we summarise what new insights cGMP imaging has provided into endogenous cGMP signalling and drug action, with a focus on the cardiovascular system. Indeed, the use of cGMP biosensors has led to several conceptual advances, such as the discovery of local, intercellular and mechanosensitive cGMP signals. Importantly, single-cell imaging can provide valuable information about the heterogeneity of cGMP signals within and between individual cells of an isolated cell population or tissue. We also discuss current challenges and future directions of cGMP imaging, such as the direct visualisation of cGMP microdomains, simultaneous monitoring of cGMP and other signalling molecules and, ultimately, cGMP imaging in tissues and animals under close-to-native conditions.
Collapse
Affiliation(s)
- Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
Mostafa T. Could Oral Phosphodiesterase 5 Inhibitors Have a Potential Adjuvant Role in Combating COVID-19 Infection? Sex Med Rev 2021; 9:15-22. [PMID: 33077403 PMCID: PMC7833179 DOI: 10.1016/j.sxmr.2020.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The recent global outbreak of coronavirus disease 2019 (COVID-19) has become a pandemic with a lot of sufferers. Excessive inflammation, exaggerated immune response, with ultimate apoptosis contribute to COVID-19 pathology that progress to acute lung acute respiratory distress. OBJECTIVE To shed a light on the likely benefits of the oral phosphodiesterase 5 (PDE5) inhibitor adjuvant role in combating COVID-19 infection. METHODS A literature review was performed in the PubMed/Medline database, Scopus, Cochrane Library, EMBASE, Academic Search Complete, Google Scholar, and CINAHL databases using the keywords COVID-19; phosphodiesterase-5 inhibitors; cytokine storm; respiratory distress. RESULTS Despite the worsening trends of COVID-19, still no drugs are validated to have significant clinical efficacy in the treatment of patients with COVID-19 in large-scale studies. While the progress toward a curative agent and/or vaccine is certainly hopeful, the principal limiting factor in such public health emergencies is always the time. Therefore, a preexisting licensed therapeutic(s) might offer a reprieve to the healthcare systems operating at the edge of capacity. In this context, the innovation of oral PDE5 inhibitors with their valuable effects on erection have provided a breakthrough in the treatment of erectile dysfunction and opened new fields of clinical application for this class of drugs. Oral PDE5 inhibitors have been demonstrated to possess many beneficial useful additional implications with acknowledged anti-inflammatory, antioxidant, immune response regulation, and antiapoptotic properties. These properties have been elucidated through the nitric oxide/soluble guanylyl cyclase/cyclic guanylate monophosphate pathway in addition to the emerged hemeoxygenase-1 enzyme as well as hydrogen sulfide pathways. These properties could support repurposing oral PDE5 inhibitors' potential adjuvant use in targeting different aspects of COVID-19 infection. CONCLUSION Oral PDE5 inhibitors retain several acknowledged off-labeled useful implications with anti-inflammatory, antioxidant, immune response regulation, and antiapoptotic properties. These properties may support repurposing oral PDE5 inhibitors' potential adjuvant use in the protocols combating COVID-19 manifestations. Mostafa T. Could Oral Phosphodiesterase 5 Inhibitors Have a Potential Adjuvant Role in Combating Coronavirus Disease 2019 Infection? Sex Med Rev 2021;9:15-22.
Collapse
Affiliation(s)
- Taymour Mostafa
- Andrology, Sexology & STIs Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
20
|
Kim YR, Yi M, Cho SA, Kim WY, Min J, Shin JG, Lee SJ. Identification and functional study of genetic polymorphisms in cyclic nucleotide phosphodiesterase 3A (PDE3A). Ann Hum Genet 2020; 85:80-91. [PMID: 33249558 DOI: 10.1111/ahg.12411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 11/28/2022]
Abstract
Phosphodiesterase 3A (PDE3A) is an enzyme that plays an important role in the regulation of cyclic adenosine monophosphate (cAMP)-mediated intracellular signaling in cardiac myocytes and platelets. PDE3A hydrolyzes cAMP, which results in a decrease in intracellular cAMP levels and leads to platelet activation. Whole-exome sequencing of 50 DNA samples from a healthy Korean population revealed a total of 13 single nucleotide polymorphisms including five missense variants, D12N, Y497C, H504Q, C707R, and A980V. Recombinant proteins for the five variants of PDE3A (and wild-type protein) were expressed in a FreeStyle 293 expression system with site-directed mutagenesis. The expression of the recombinant PDE3A proteins was confirmed with Western blotting. Catalytic activity of the PDE3A missense variants and wild-type enzyme was measured with a PDE-based assay. Effects of the missense variants on the inhibition of PDE3A activity by cilostazol were also investigated. All variant proteins showed reduced activity (33-53%; p < .0001) compared to the wild-type protein. In addition, PDE3A activity was inhibited by cilostazol in a dose-dependent manner and was further suppressed in the missense variants. Specifically, the PDE3A Y497C showed significantly reduced activity, consistent with the predictions of in silico analyses. The present study provides evidence that individuals carrying the PDE3A Y497C variant may have lower enzyme activity for cAMP hydrolysis, which could cause interindividual variation in cAMP-mediated physiological functions.
Collapse
Affiliation(s)
- You Ran Kim
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea
| | - MyeongJin Yi
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea.,Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Sun-Ah Cho
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea
| | - Woo-Young Kim
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea
| | - JungKi Min
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Jae-Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea.,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Inje University College of Medicine, Inje University, Busan, 47392, South Korea
| | - Su-Jun Lee
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Inje University, Busan, South Korea
| |
Collapse
|
21
|
Could Oral Phosphodiesterase 5 Inhibitors Have a Potential Adjuvant Role in Combating COVID-19 Infection? Sex Med Rev 2020. [PMID: 33077403 DOI: 10.1016/j.sxmr.2020.08.006.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The recent global outbreak of coronavirus disease 2019 (COVID-19) has become a pandemic with a lot of sufferers. Excessive inflammation, exaggerated immune response, with ultimate apoptosis contribute to COVID-19 pathology that progress to acute lung acute respiratory distress. OBJECTIVE To shed a light on the likely benefits of the oral phosphodiesterase 5 (PDE5) inhibitor adjuvant role in combating COVID-19 infection. METHODS A literature review was performed in the PubMed/Medline database, Scopus, Cochrane Library, EMBASE, Academic Search Complete, Google Scholar, and CINAHL databases using the keywords COVID-19; phosphodiesterase-5 inhibitors; cytokine storm; respiratory distress. RESULTS Despite the worsening trends of COVID-19, still no drugs are validated to have significant clinical efficacy in the treatment of patients with COVID-19 in large-scale studies. While the progress toward a curative agent and/or vaccine is certainly hopeful, the principal limiting factor in such public health emergencies is always the time. Therefore, a preexisting licensed therapeutic(s) might offer a reprieve to the healthcare systems operating at the edge of capacity. In this context, the innovation of oral PDE5 inhibitors with their valuable effects on erection have provided a breakthrough in the treatment of erectile dysfunction and opened new fields of clinical application for this class of drugs. Oral PDE5 inhibitors have been demonstrated to possess many beneficial useful additional implications with acknowledged anti-inflammatory, antioxidant, immune response regulation, and antiapoptotic properties. These properties have been elucidated through the nitric oxide/soluble guanylyl cyclase/cyclic guanylate monophosphate pathway in addition to the emerged hemeoxygenase-1 enzyme as well as hydrogen sulfide pathways. These properties could support repurposing oral PDE5 inhibitors' potential adjuvant use in targeting different aspects of COVID-19 infection. CONCLUSION Oral PDE5 inhibitors retain several acknowledged off-labeled useful implications with anti-inflammatory, antioxidant, immune response regulation, and antiapoptotic properties. These properties may support repurposing oral PDE5 inhibitors' potential adjuvant use in the protocols combating COVID-19 manifestations. Mostafa T. Could Oral Phosphodiesterase 5 Inhibitors Have a Potential Adjuvant Role in Combating Coronavirus Disease 2019 Infection? Sex Med Rev 2021;9:15-22.
Collapse
|
22
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
23
|
Flora GD, Nayak MK. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr Pharm Des 2020; 25:4063-4084. [PMID: 31553287 DOI: 10.2174/1381612825666190925163827] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature death and disability in humans and their incidence is on the rise globally. Given their substantial contribution towards the escalating costs of health care, CVDs also generate a high socio-economic burden in the general population. The underlying pathogenesis and progression associated with nearly all CVDs are predominantly of atherosclerotic origin that leads to the development of coronary artery disease, cerebrovascular disease, venous thromboembolism and, peripheral vascular disease, subsequently causing myocardial infarction, cardiac arrhythmias or stroke. The aetiological risk factors leading to the onset of CVDs are well recognized and include hyperlipidaemia, hypertension, diabetes, obesity, smoking and, lack of physical activity. They collectively represent more than 90% of the CVD risks in all epidemiological studies. Despite high fatality rate of CVDs, the identification and careful prevention of the underlying risk factors can significantly reduce the global epidemic of CVDs. Beside making favorable lifestyle modifications, primary regimes for the prevention and treatment of CVDs include lipid-lowering drugs, antihypertensives, antiplatelet and anticoagulation therapies. Despite their effectiveness, significant gaps in the treatment of CVDs remain. In this review, we discuss the epidemiology and pathology of the major CVDs that are prevalent globally. We also determine the contribution of well-recognized risk factors towards the development of CVDs and the prevention strategies. In the end, therapies for the control and treatment of CVDs are discussed.
Collapse
Affiliation(s)
- Gagan D Flora
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Manasa K Nayak
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
24
|
Abdel-Aziz AM, Rifaai RA, Abdel-Gaber SA. Possible mechanisms mediating the protective effect of cilostazol in L-arginine induced acute pancreatitis in rats: role of cGMP, cAMP, and HO-1. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1859-1870. [PMID: 32424476 DOI: 10.1007/s00210-020-01897-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 05/06/2020] [Indexed: 12/22/2022]
Abstract
Acute pancreatitis (AP) is an inflammatory disorder with a high mortality rate. Cilostazol is a selective phosphodiesterase-3 inhibitor drug that is commonly used as an antiplatelet, antithrombotic, and vasodilator drug. It exhibits antioxidant, anti-inflammatory, and anti-apoptotic activities, but its effect on AP has not been fully elucidated yet. The present study aimed to investigate the effects of cilostazol on L-arginine-induced AP and the possible protective mechanisms. A rat model of AP was established by a single i.p. injection of 3-g/kg L-arginine on day 13 of the experiment. The treated groups received a single daily oral dose of either 100 or 300 mg/kg/day for 14 consecutive days. Rats with AP showed histopathological changes of pancreatic tissue injury together with increased serum amylase enzyme activity and decreased serum insulin, pancreatic adiponectin, and cGMP levels. Moreover, AP rats showed increased pancreatic inflammatory biomarker (TNF-α, VCAM-1, and MPO) levels with decreased anti-inflammatory IL-10 levels. In addition, oxidative stress biomarkers (MDA and NO) were increased in AP with decreased antioxidant SOD activity and GSH level. Moreover, HO-1 immunostaining was increased in the AP group. Cilostazol pretreatment reversed the histopathological change; decreased the amylase activity and the levels of TNF-α, VCAM-1, and MPO; and increased the levels of insulin, adiponectin, cGMP, cAMP, and IL-10. Moreover, cilostazol decreased MDA and NO but increased SOD and GSH. Lastly, cilostazol increased the HO-1 immunostaining more than in the AP group. These data suggest that cilostazol protects against L-arginine-induced AP, which may be related to an increase in cGMP, cAMP, and upregulation of HO-1 with subsequent anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
| | - Rehab Ahmed Rifaai
- Departments of Histology and cell biology, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| | - Seham A Abdel-Gaber
- Departments of Pharmacology, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| |
Collapse
|
25
|
Riley DRJ, Khalil JS, Pieters J, Naseem KM, Rivero F. Coronin 1 Is Required for Integrin β2 Translocation in Platelets. Int J Mol Sci 2020; 21:ijms21010356. [PMID: 31948107 PMCID: PMC6982036 DOI: 10.3390/ijms21010356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/23/2019] [Accepted: 01/01/2020] [Indexed: 01/22/2023] Open
Abstract
Remodeling of the actin cytoskeleton is one of the critical events that allows platelets to undergo morphological and functional changes in response to receptor-mediated signaling cascades. Coronins are a family of evolutionarily conserved proteins implicated in the regulation of the actin cytoskeleton, represented by the abundant coronins 1, 2, and 3 and the less abundant coronin 7 in platelets, but their functions in these cells are poorly understood. A recent report revealed impaired agonist-induced actin polymerization and cofilin phosphoregulation and altered thrombus formation in vivo as salient phenotypes in the absence of an overt hemostasis defect in vivo in a knockout mouse model of coronin 1. Here we show that the absence of coronin 1 is associated with impaired translocation of integrin β2 to the platelet surface upon stimulation with thrombin while morphological and functional alterations, including defects in Arp2/3 complex localization and cAMP-dependent signaling, are absent. Our results suggest a large extent of functional overlap among coronins 1, 2, and 3 in platelets, while aspects like integrin β2 translocation are specifically or predominantly dependent on coronin 1.
Collapse
Affiliation(s)
- David R. J. Riley
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (D.R.J.R.); (J.S.K.)
| | - Jawad S. Khalil
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (D.R.J.R.); (J.S.K.)
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Jean Pieters
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland;
| | - Khalid M. Naseem
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9NL, UK;
| | - Francisco Rivero
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (D.R.J.R.); (J.S.K.)
- Correspondence: ; Tel.: +44-1482-644-633
| |
Collapse
|
26
|
Su SC, Hung YJ, Huang CL, Shieh YS, Chien CY, Chiang CF, Liu JS, Lu CH, Hsieh CH, Lin CM, Lee CH. Cilostazol inhibits hyperglucose-induced vascular smooth muscle cell dysfunction by modulating the RAGE/ERK/NF-κB signaling pathways. J Biomed Sci 2019; 26:68. [PMID: 31492153 PMCID: PMC6731603 DOI: 10.1186/s12929-019-0550-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/25/2019] [Indexed: 02/08/2023] Open
Abstract
Background Increasing evidence suggests that high glucose (HG) causes abnormalities in endothelial and vascular smooth muscle cell function (VSMC) and contributes to atherosclerosis. Receptor for advanced glycation end-products (RAGE) has been linked to the pathogenesis of both the macrovascular and microvascular complications of diabetes. Cilostazol is used to treat diabetic vasculopathy by ameliorating HG-induced vascular dysfunction. Objectives In this study, we investigated whether the cilostazol suppression of HG-induced VSMC dysfunction is through RAGE signaling and its possible regulation mechanism. Method We investigated the effect of HG and cilostazol on RAGE signaling in A7r5 rat VSMCs. Aortic tissues of streptozotocin (STZ) diabetic mice were also collected. Results Aortic tissue samples from the diabetic mice exhibited a significantly decreased RAGE expression after cilostazol treatment. HG increased RAGE, focal adhesion kinase (FAK), matrix metalloproteinase-2 (MMP-2), intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expressions, and was accompanied with increased reactive oxygen species (ROS), cell proliferation, adhesion and migration. Cilostazol significantly reversed HG-induced RAGE, ROS, downstream gene expressions and cell functions. RAGE knockdown significantly reversed the expressions of HG-induced vasculopathy related gene expressions and cell functions. Cilostazol with RAGE knockdown had additive effects on downstream ERK/NF-κB signaling pathways, gene expressions and cell functions of A7r5 rat VSMCs in HG culture. Conclusions Both in vitro and in vivo experimental diabetes models showed novel signal transduction of cilostazol-mediated protection against HG-related VSMC dysfunction, and highlighted the involvement of RAGE signaling and downstream pathways.
Collapse
Affiliation(s)
- Sheng-Chiang Su
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. .,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan. .,Division of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| | - Chia-Luen Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Shing Shieh
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Department of Oral Diagnosis and Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Division of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chu-Yen Chien
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Fu Chiang
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jhih-Syuan Liu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chieh-Hua Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Hsun Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Ming Lin
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Hsing Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. .,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan. .,Division of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
27
|
Acquired platelet function disorders. Thromb Res 2019; 196:561-568. [PMID: 31229273 DOI: 10.1016/j.thromres.2019.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 06/08/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
Abstract
The possibility of an acquired platelet function disorder should be considered in patients who present with recent onset muco-cutaneous bleeding. Despite the availability of newer and faster platelet function assays, light transmission aggregometry (LTA) remains the preferred diagnostic test. This review examines and discusses the causes of acquired platelet dysfunction; most commonly drugs, dietary factors, medical disorders and procedures. In addition to well-known antiplatelet therapies, clinicians should be alert for newer drugs which can affect platelets, such as ibrutinib. There is little clinical trial evidence to guide the management of acquired platelet function defects, but we summarise commonly employed strategies, which include addressing the underlying cause, antifibrinolytic agents, desmopressin infusions, and in selected patients, platelet transfusions.
Collapse
|
28
|
The uncertain role for phosphodiesterase inhibitors in stroke prevention. Lancet Neurol 2019; 18:515-517. [DOI: 10.1016/s1474-4422(19)30165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 11/19/2022]
|
29
|
Hu M, Wang D, He T. Comparison of efficacy between trimetazidine and cilostazol in the treatment of arteriosclerosis obliterans in lower extremity. Exp Ther Med 2019; 17:4427-4434. [PMID: 31086577 PMCID: PMC6488977 DOI: 10.3892/etm.2019.7472] [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: 11/14/2018] [Accepted: 03/19/2019] [Indexed: 11/06/2022] Open
Abstract
This study compared the efficacy and long-term survival rate of trimetazidine and cilostazol in the treatment of lower extremity arteriosclerosis obliterans (ASO). A retrospectively analysis on the medical records of 206 patients with ASO who were admitted to The Central Hospital of Wuhan from January 2011 to May 2013 was performed, including 94 patients treated with trimetazidine (group A) and 112 patients treated with cilostazol (group B). On the basis of the same basic treatment, both groups were applied with these two drugs after two courses of treatments. Then the efficacy of clinical treatment, dorsal artery blood flow, anterior femoral artery, posterior tibial artery blood flow, brachial artery index, toe-brachial index, painless walking distance, maximum walking distance, adverse reactions, 5-year survival rates were compared. The total effective rate of clinical efficacy in group B was higher than group A (P<0.05). After the first course of treatment, the above indicators increased in both groups (P<0.05). After the end of the second course of treatment, the above-mentioned index values in both groups were significantly increased (P<0.05). The improvement of the above indicators in group B were better than the trimetazidine group in both the first and second treatment courses (P<0.05). In group A, there were 15 cases of patients with lethargy and hypodynamia and 9 cases of dizziness and headache. There were significant differences between the 7th and 3rd cases of patients when compared to group B (P<0.05). The 5-year survival rate of group A was lower than group B (P<0.05). The clinical efficacy of cliostazol in the treatment of ASO had a good effect, and there was only a few adverse reactions and the long-term survival rate was high. It is worthy of being promoted in clinical practice.
Collapse
Affiliation(s)
- Min Hu
- Department of Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Dile Wang
- Department of Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Tao He
- Department of Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
30
|
Nucleoside analogue activators of cyclic AMP-independent protein kinase A of Trypanosoma. Nat Commun 2019; 10:1421. [PMID: 30926779 PMCID: PMC6440977 DOI: 10.1038/s41467-019-09338-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 03/07/2019] [Indexed: 02/08/2023] Open
Abstract
Protein kinase A (PKA), the main effector of cAMP in eukaryotes, is a paradigm for the mechanisms of ligand-dependent and allosteric regulation in signalling. Here we report the orthologous but cAMP-independent PKA of the protozoan Trypanosoma and identify 7-deaza-nucleosides as potent activators (EC50 ≥ 6.5 nM) and high affinity ligands (KD ≥ 8 nM). A co-crystal structure of trypanosome PKA with 7-cyano-7-deazainosine and molecular docking show how substitution of key amino acids in both CNB domains of the regulatory subunit and its unique C-terminal αD helix account for this ligand swap between trypanosome PKA and canonical cAMP-dependent PKAs. We propose nucleoside-related endogenous activators of Trypanosoma brucei PKA (TbPKA). The existence of eukaryotic CNB domains not associated with binding of cyclic nucleotides suggests that orphan CNB domains in other eukaryotes may bind undiscovered signalling molecules. Phosphoproteome analysis validates 7-cyano-7-deazainosine as powerful cell-permeable inducer to explore cAMP-independent PKA signalling in medically important neglected pathogens.
Collapse
|
31
|
A shear-dependent NO-cGMP-cGKI cascade in platelets acts as an auto-regulatory brake of thrombosis. Nat Commun 2018; 9:4301. [PMID: 30327468 PMCID: PMC6191445 DOI: 10.1038/s41467-018-06638-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
Mechanisms that limit thrombosis are poorly defined. One of the few known endogenous platelet inhibitors is nitric oxide (NO). NO activates NO sensitive guanylyl cyclase (NO-GC) in platelets, resulting in an increase of cyclic guanosine monophosphate (cGMP). Here we show, using cGMP sensor mice to study spatiotemporal dynamics of platelet cGMP, that NO-induced cGMP production in pre-activated platelets is strongly shear-dependent. We delineate a new mode of platelet-inhibitory mechanotransduction via shear-activated NO-GC followed by cGMP synthesis, activation of cGMP-dependent protein kinase I (cGKI), and suppression of Ca2+ signaling. Correlative profiling of cGMP dynamics and thrombus formation in vivo indicates that high cGMP concentrations in shear-exposed platelets at the thrombus periphery limit thrombosis, primarily through facilitation of thrombus dissolution. We propose that an increase in shear stress during thrombus growth activates the NO-cGMP-cGKI pathway, which acts as an auto-regulatory brake to prevent vessel occlusion, while preserving wound closure under low shear. Nitric oxide (NO) inhibits thrombosis in part by stimulating cyclic guanosine monophosphate (cGMP) production and cGMP-dependent protein kinase I (cGKI) activity in platelets. Here, Wen et al. develop a cGMP sensor mouse to follow cGMP dynamics in platelets, and find that shear stress activates NO-cGMP-cGKI signaling during platelet aggregation to limit thrombosis.
Collapse
|
32
|
Wang C, Li S, Shen C, Shan J, Fan Y. Expression and significance of phosphodiesterase 4B gene in peripheral blood of patients with oral lichen planus. Int J Dermatol 2018; 58:302-310. [PMID: 30229893 DOI: 10.1111/ijd.14203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/04/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND The pathogenesis of oral lichen planus (OLP) may be related to mental factors. In this study, we investigated the molecular mechanism of mental factors in the development of OLP. METHODS The normal control group and OLP patients were tested and evaluated by Zung self-rating anxiety scale and self-rating depression scale. Secondly, Agilent human LncRNA chip technology was used to detect differential genes in the total RNA of the normal control group and OLP patients, and to screen out the differentially expressed genes related to anxiety and depression. Thirdly, we verified the genes at gene level and protein level, respectively. RESULTS Zung self-rating anxiety scale and self-rating depression scale showed that tendency of anxiety and depression in OLP patients is significantly higher than that in normal controls, but there was no significant difference between the erosion form group and the reticular form group; the duration of OLP showed significant negative correlations between degree of anxiety and depression. Microarray analysis showed there were four differential genes (PDE4B, RGS2, SYNGR1, and SYNE1) related to anxiety and depression in OLP patients; real-time qPCR confirmed the expression of PDE4B mRNA was lower in the peripheral blood of normal control group (P < 0.001). The expression of RGS2, SYNGR1, and SYNE1 mRNA was higher in the normal control group (P < 0.05, P < 0.05, P < 0.05). Wes automatic western blot confirmed that the expression of PDE4B protein was lower in the peripheral blood of the normal control group (P < 0.01). CONCLUSION PDE4B gene may play a role in the pathogenesis of OLP.
Collapse
Affiliation(s)
- Chen Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Shan Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Chen Shen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jing Shan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yuan Fan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Knowles RB, Warner TD. Anti-platelet drugs and their necessary interaction with endothelial mediators and platelet cyclic nucleotides for therapeutic efficacy. Pharmacol Ther 2018; 193:83-90. [PMID: 30081048 PMCID: PMC6325790 DOI: 10.1016/j.pharmthera.2018.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
For many millions of patients at secondary risk of coronary thrombosis pharmaceutical protection is supplied by dual anti-platelet therapy. Despite substantial therapeutic developments over the last decade recurrent thrombotic events occur, highlighting the need for further optimisation of therapies. Importantly, but often ignored, anti-platelet drugs interact with cyclic nucleotide systems in platelets and these are the same systems that mediate key endogenous pathways of platelet regulation, notably those dependent upon the vascular endothelium. The aim of this review is to highlight interactions between the anti-platelet drugs, aspirin and P2Y12 receptor antagonists and endogenous pathways of platelet regulation at the level of cyclic nucleotides. These considerations are key to concepts such as anti-platelet drug resistance and individualized anti-platelet therapy which cannot be understood by study of platelets in isolation from the circulatory environment. We also explore novel and emerging therapies that focus on preserving haemostasis and how the concepts outlined in this review could be exploited therapeutically to improve anti-thrombotic efficacy whilst reducing bleeding risk.
Collapse
Affiliation(s)
- Rebecca B Knowles
- The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Timothy D Warner
- The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
34
|
Cardarelli S, Miele AE, Zamparelli C, Biagioni S, Naro F, Malatesta F, Giorgi M, Saliola M. The oligomeric assembly of the phosphodiesterase-5 is a mixture of dimers and tetramers: A putative role in the regulation of function. Biochim Biophys Acta Gen Subj 2018; 1862:2183-2190. [PMID: 30025857 DOI: 10.1016/j.bbagen.2018.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/25/2018] [Accepted: 07/11/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Phosphodiesterases (PDEs) are a superfamily of evolutionary conserved cyclic nucleotides (cAMP/cGMP) hydrolysing enzymes, components of transduction pathways regulating crucial aspects of cell life. PDE5, one of these families, is the molecular target of several drugs used to treat erectile dysfunction and pulmonary hypertension. Despite its medical relevance, PDE5 macromolecular structure has only been solved for the isolated regulatory and catalytic domains. The definition of the quaternary structure of the full length PDE5 (MmPDE5A1), produced in large amounts in the yeast Kluyveromyces lactis, could greatly enhance the knowledge on its assembly/allosteric regulation and the development of new inhibitors for clinical-therapeutic applications. METHODS Small-angle X-ray scattering (SAXS), analytical ultracentrifugation (AUC), size exclusion chromatography (SEC), native polyacrylamide gel electrophoresis (PAGE) and western blot (WB) were used to assess the assembly of PDE5A1. RESULTS The full length MmPDE5A1 isoform is a mixture of dimers and tetramers in solution. We also report data showing that dimers and tetramers also coexist in vivo in platelets, blood components naturally containing high levels of PDE5. CONCLUSIONS This is the first time that structural studies on the full length protein evidenced the assembly of PDE5 in tetramers in addition to the expected dimers. GENERAL SIGNIFICANCE The assembly of PDE5 in tetramers in platelets, beside the dimers, opens the possibility to alternative assembly/allosteric regulation of this enzyme, as component of large signaling complexes, in all cellular districts in which PDE5 is present.
Collapse
Affiliation(s)
- Silvia Cardarelli
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Adriana Erica Miele
- Department of Biochemical Sciences 'Rossi Fanelli', Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy; UMR 5246 ICBMS - CNRS - UCBL, Université de Lyon, Campus La Doua, 43 bd 11 Novembre 1918, 69622 Villeurbanne, Cedex, France.
| | - Carlotta Zamparelli
- Department of Biochemical Sciences 'Rossi Fanelli', Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Stefano Biagioni
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic, and Orthopaedic Sciences, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Francesco Malatesta
- Department of Biochemical Sciences 'Rossi Fanelli', Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Mauro Giorgi
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| | - Michele Saliola
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
35
|
Andersson KE. PDE5 inhibitors - pharmacology and clinical applications 20 years after sildenafil discovery. Br J Pharmacol 2018; 175:2554-2565. [PMID: 29667180 DOI: 10.1111/bph.14205] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/01/2018] [Accepted: 03/05/2018] [Indexed: 11/28/2022] Open
Abstract
The discovery of the nitric oxide/cGMP pathway was the basis for our understanding of many normal physiological functions and the pathophysiology of several diseases. Since the discovery and introduction of sildenafil, inhibitors of PDE5 have been the first-line therapy for erectile dysfunction (ED). The success of sildenafil in the treatment of ED stimulated research in the field of PDE5 inhibition and led to many new applications, such as treatment of lower urinary symptoms, and pulmonary arterial hypertension, which are now approved indications. However, PDE5 inhibitors have also been used in several other disorders not discussed in this review, and the fields of clinical use are increasing. In the present review, the pharmacological basis of the NO/cGMP pathway and the rationale and clinical use of PDE5 inhibitors in different diseases are discussed.
Collapse
Affiliation(s)
- K-E Andersson
- Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA.,Institute of Laboratory Medicine, Lund University, 223 62, Lund, Sweden
| |
Collapse
|
36
|
Wang S, Li Z, Xu R. Human Cancer and Platelet Interaction, a Potential Therapeutic Target. Int J Mol Sci 2018; 19:ijms19041246. [PMID: 29677116 PMCID: PMC5979598 DOI: 10.3390/ijms19041246] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 03/30/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer patients experience a four-fold increase in thrombosis risk, indicating that cancer development and progression are associated with platelet activation. Xenograft experiments and transgenic mouse models further demonstrate that platelet activation and platelet-cancer cell interaction are crucial for cancer metastasis. Direct or indirect interaction of platelets induces cancer cell plasticity and enhances survival and extravasation of circulating cancer cells during dissemination. In vivo and in vitro experiments also demonstrate that cancer cells induce platelet aggregation, suggesting that platelet-cancer interaction is bidirectional. Therefore, understanding how platelets crosstalk with cancer cells may identify potential strategies to inhibit cancer metastasis and to reduce cancer-related thrombosis. Here, we discuss the potential function of platelets in regulating cancer progression and summarize the factors and signaling pathways that mediate the cancer cell-platelet interaction.
Collapse
Affiliation(s)
- Shike Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| | - Zhenyu Li
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, University of Kentucky, 741 South Limestone Street, Lexington, KY 40536, USA.
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
37
|
Sánchez G, Estrada O, Acha G, Cardozo A, Peña F, Ruiz MC, Michelangeli F, Alvarado-Castillo C. The norpurpureine alkaloid from Annona purpurea inhibits human platelet activation in vitro. Cell Mol Biol Lett 2018; 23:15. [PMID: 29713353 PMCID: PMC5905151 DOI: 10.1186/s11658-018-0082-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background The leaves of Annona purpurea have yielded several alkaloids with anti-aggregation activities against rabbit platelets. This is promising in the search for agents that might act against platelets and reduce the incidence of cardiovascular diseases. Since significant differences in platelet function have been reported between human and animal platelets, a study focusing on the effect of A. purpurea extracts against human platelet activation is necessary. Methods The compounds in an A. purpurea ethanolic extract underwent bio-guided fractionation and were used for in vitro human platelet aggregation assays to isolate the compounds with anti-platelet activity. The bioactive compounds were identified by spectroscopic analysis. Additional platelet studies were performed to characterize their action as inhibitors of human platelet activation. Results The benzylisoquinoline alkaloid norpurpureine was identified as the major anti-platelet compound. The IC50 for norpurpureine was 80 μM against platelets when stimulated with adenosine 5′-diphosphate (ADP), collagen and thrombin. It was pharmacologically effective from 20 to 220 μM. Norpurpureine (220 μM) exhibited its in vitro effectiveness in samples from 30 healthy human donors who did not take any drugs during the 2 weeks prior to the collection. Norpurpureine also gradually inhibited granule secretion and adhesion of activated platelets to immobilized fibrinogen. At the intra-platelet level, norpurpureine prevented agonist-stimulated calcium mobilization and cAMP reduction. Structure–activity relationship analysis indicates that the lack of a methyl group at the nitrogen seems to be key in the ability of the compound to interact with its molecular target. Conclusion Norpurpureine displays a promising in vitro pharmacological profile as an inhibitor of human platelet activation. Its molecular target could be a common effector between Ca2+ and cAMP signaling, such as the PLC-PKC-Ca2+ pathway and PDEs. This needs further evaluation at the protein isoform level. Electronic supplementary material The online version of this article (10.1186/s11658-018-0082-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Sánchez
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Omar Estrada
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Giovana Acha
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Alfonso Cardozo
- 2Laboratorio de Botánica Sistemática, Facultad de Agronomía, Universidad Central de Venezuela (UCV), Maracay, Bolivarian Republic of Venezuela
| | - Franshelle Peña
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Marie Christine Ruiz
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Fabián Michelangeli
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Claudia Alvarado-Castillo
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela.,3Laboratorio de Hemostasia y Genética Vascular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Apartado 20632, K11 de la Carretera Panamericana, Caracas, 1020-A Bolivarian Republic of Venezuela
| |
Collapse
|
38
|
Bazin HG, Bess LS, Livesay MT. Synthesis and Applications of Imidazoquinolines: A Review. ORG PREP PROCED INT 2018. [DOI: 10.1080/00304948.2018.1433427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Hélène G. Bazin
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| | - Laura S. Bess
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| | - Mark T. Livesay
- Biomedical & Pharmaceutical Science, University of Montana, 32 Campus Drive #1552, Missoula, MT 59812, USA
| |
Collapse
|
39
|
|
40
|
Warner TD, Armstrong PC, Chan MV, Knowles RB. The importance of endothelium-derived mediators to the efficacy of dual anti-platelet therapy. Expert Rev Hematol 2016; 9:223-5. [PMID: 26822235 DOI: 10.1586/17474086.2016.1140035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Timothy D Warner
- a The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry , Queen Mary University of London , London , United Kingdom
| | - Paul C Armstrong
- a The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry , Queen Mary University of London , London , United Kingdom
| | - Melissa V Chan
- a The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry , Queen Mary University of London , London , United Kingdom
| | - Rebecca B Knowles
- a The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry , Queen Mary University of London , London , United Kingdom
| |
Collapse
|
41
|
Shen K, Johnson DW, Gobe GC. The role of cGMP and its signaling pathways in kidney disease. Am J Physiol Renal Physiol 2016; 311:F671-F681. [PMID: 27413196 DOI: 10.1152/ajprenal.00042.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/10/2016] [Indexed: 01/20/2023] Open
Abstract
Cyclic nucleotide signal transduction pathways are an emerging research field in kidney disease. Activated cell surface receptors transduce their signals via intracellular second messengers such as cAMP and cGMP. There is increasing evidence that regulation of the cGMP-cGMP-dependent protein kinase 1-phosphodiesterase (cGMP-cGK1-PDE) signaling pathway may be renoprotective. Selective PDE5 inhibitors have shown potential in treating kidney fibrosis in patients with chronic kidney disease (CKD), via their downstream signaling, and these inhibitors also have known activity as antithrombotic and anticancer agents. This review gives an outline of the cGMP-cGK1-PDE signaling pathways and details the downstream signaling and regulatory functions that are modulated by cGK1 and PDE inhibitors with regard to antifibrotic, antithrombotic, and antitumor activity. Current evidence that supports the renoprotective effects of regulating cGMP-cGK1-PDE signaling is also summarized. Finally, the effects of icariin, a natural plant extract with PDE5 inhibitory function, are discussed. We conclude that regulation of cGMP-cGK1-PDE signaling might provide novel, therapeutic strategies for the worsening global public health problem of CKD.
Collapse
Affiliation(s)
- Kunyu Shen
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China; and
| | - David W Johnson
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia;
| |
Collapse
|
42
|
Raslan Z, Aburima A, Naseem KM. The Spatiotemporal Regulation of cAMP Signaling in Blood Platelets-Old Friends and New Players. Front Pharmacol 2015; 6:266. [PMID: 26617518 PMCID: PMC4639615 DOI: 10.3389/fphar.2015.00266] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/26/2015] [Indexed: 11/22/2022] Open
Abstract
Atherothrombosis, the pathology underlying numerous cardiovascular diseases, is a major cause of death globally. Hyperactive blood platelets play a key role in the atherothrombotic process through the release of inflammatory mediators and formation of thrombi. In healthy blood vessels, excessive platelet activation is restricted by endothelial-derived prostacyclin (PGI2) through cyclic adenosine-5′-monophosphate (cAMP) and protein kinase A (PKA)-dependent mechanisms. Elevation in intracellular cAMP is associated with the control of a number of distinct platelet functions including actin polymerisation, granule secretion, calcium mobilization and integrin activation. Unfortunately, in atherosclerotic disease the protective effects of cAMP are compromised, which may contribute to pathological thrombosis. The cAMP signaling network in platelets is highly complex with the presence of multiple isoforms of adenylyl cyclase (AC), PKA, and phosphodiesterases (PDEs). However, a precise understanding of the relationship between specific AC, PKA, and PDE isoforms, and how individual signaling substrates are targeted to control distinct platelet functions is still lacking. In other cells types, compartmentalisation of cAMP signaling has emerged as a key mechanism to allow precise control of specific cell functions. A-kinase anchoring proteins (AKAPs) play an important role in this spatiotemporal regulation of cAMP signaling networks. Evidence of AKAP-mediated compartmentalisation of cAMP signaling in blood platelets has begun to emerge and is providing new insights into the regulation of platelet function. Dissecting the mechanisms that allow cAMP to control excessive platelet activity without preventing effective haemostasis may unleash the possibility of therapeutic targeting of the pathway to control unwanted platelet activity.
Collapse
Affiliation(s)
- Zaher Raslan
- Centre for Cardiovascular and Metabolic Research, Hull-York Medical School, University of Hull , Hull, UK
| | - Ahmed Aburima
- Centre for Cardiovascular and Metabolic Research, Hull-York Medical School, University of Hull , Hull, UK
| | - Khalid M Naseem
- Centre for Cardiovascular and Metabolic Research, Hull-York Medical School, University of Hull , Hull, UK
| |
Collapse
|
43
|
Modulating platelet reactivity through control of RGS18 availability. Blood 2015; 126:2611-20. [PMID: 26407691 DOI: 10.1182/blood-2015-04-640037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 09/22/2015] [Indexed: 01/13/2023] Open
Abstract
Most platelet agonists activate platelets by binding to G-protein-coupled receptors. We have shown previously that a critical node in the G-protein signaling network in platelets is formed by a scaffold protein, spinophilin (SPL), the tyrosine phosphatase, Src homology region 2 domain-containing phosphatase-1 (SHP-1), and the regulator of G-protein signaling family member, RGS18. Here, we asked whether SPL and other RGS18 binding proteins such as 14-3-3γ regulate platelet reactivity by sequestering RGS18 and, if so, how this is accomplished. The results show that, in resting platelets, free RGS18 levels are relatively low, increasing when platelets are activated by thrombin. Free RGS18 levels also rise when platelets are rendered resistant to activation by exposure to prostaglandin I2 (PGI2) or forskolin, both of which increase platelet cyclic adenosine monophosphate (cAMP) levels. However, the mechanism for raising free RGS18 is different in these 2 settings. Whereas thrombin activates SHP-1 and causes dephosphorylation of SPL tyrosine residues, PGI2 and forskolin cause phosphorylation of SPL Ser94 without reducing tyrosine phosphorylation. Substituting alanine for Ser94 blocks cAMP-induced dissociation of the SPL/RGS/SHP-1 complex. Replacing Ser94 with aspartate prevents formation of the complex and produces a loss-of-function phenotype when expressed in mouse platelets. Together with the defect in platelet function we previously observed in SPL(-/-) mice, these data show that (1) regulated sequestration and release of RGS18 by intracellular binding proteins provides a mechanism for coordinating activating and inhibitory signaling networks in platelets, and (2) differential phosphorylation of SPL tyrosine and serine residues provides a key to understanding both.
Collapse
|
44
|
Kontogiorgis C, Nicolotti O, Mangiatordi GF, Tognolini M, Karalaki F, Giorgio C, Patsilinakos A, Carotti A, Hadjipavlou-Litina D, Barocelli E. Studies on the antiplatelet and antithrombotic profile of anti-inflammatory coumarin derivatives. J Enzyme Inhib Med Chem 2015; 30:925-33. [PMID: 25807297 DOI: 10.3109/14756366.2014.995180] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The interest towards coumarin-based structures stems from their polypharmacological profile. Herein, we present a series of Mannich bases and 7-azomethine-linked coumarin derivatives exhibiting antiplatelet and antithrombotic activities, in addition to the already known anti-inflammatory and antioxidant activities. Among others, compounds 15 and 16 were found to be the most potent and selective inhibitors of platelet aggregation whereas compound 3 also proved to be the most potent in the clot retraction assay. Structure-activity relationship studies were conducted to elucidate the molecular determinants responsible for the herein observed activities. The chance of inhibiting cyclooxygenase-1 was also investigated for evaluating the platelet aggregation induced by arachidonic acid. Taken together, these results suggest that the investigation of other targets connected to the antiplatelet activity, such as phosphodiesterase-3 (PDE3), could be a viable strategy to shed light on the polypharmacological profile of coumarin-based compounds. Docking simulations towards PDE3 were also carried out.
Collapse
Affiliation(s)
- Christos Kontogiorgis
- a Laboratory of Hygiene and Environmental Protection , Democritus University of Thrace , Alexandroupolis , Greece
| | - Orazio Nicolotti
- b Dipartimento di Farmacia - Scienze del Farmaco , Universita degli Studi Bari "Aldo Moro" , Bari , Italy
| | | | | | - Foteini Karalaki
- d Department of Pharmaceutical Chemistry , School of Pharmacy, Aristotle University of Thessaloniki , Thessaloniki , Greece , and
| | - Carmine Giorgio
- c Department of Pharmacy , University of Parma , Parma , Italy
| | - Alexandros Patsilinakos
- e Department of Chemistry and Drug Technologies , "Sapienza" University of Rome , Rome , Italy
| | - Angelo Carotti
- b Dipartimento di Farmacia - Scienze del Farmaco , Universita degli Studi Bari "Aldo Moro" , Bari , Italy
| | - Dimitra Hadjipavlou-Litina
- d Department of Pharmaceutical Chemistry , School of Pharmacy, Aristotle University of Thessaloniki , Thessaloniki , Greece , and
| | | |
Collapse
|
45
|
Bath PM, Wardlaw JM. Pharmacological treatment and prevention of cerebral small vessel disease: a review of potential interventions. Int J Stroke 2015; 10:469-78. [PMID: 25727737 PMCID: PMC4832291 DOI: 10.1111/ijs.12466] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/06/2015] [Indexed: 12/21/2022]
Abstract
Small vessel disease encompasses lacunar stroke, white matter hyperintensities, lacunes and microbleeds. It causes a quarter of all ischemic strokes, is the commonest cause of vascular dementia, and the cause is incompletely understood. Vascular prophylaxis, as appropriate for large artery disease and cardioembolism, includes antithrombotics, and blood pressure and lipid lowering; however, these strategies may not be effective for small vessel disease, or are already used routinely so precluding further detailed study. Further, intensive antiplatelet therapy is known to be hazardous in small vessel disease through enhanced bleeding. Whether acetylcholinesterase inhibitors, which delay the progression of Alzheimer's dementia, are relevant in small vessel disease remains unclear. Potential prophylactic and treatment strategies might be those that target brain microvascular endothelium and the blood brain barrier, microvascular function and neuroinflammation. Potential interventions include endothelin antagonists, neurotrophins, nitric oxide donors and phosphodiesterase 5 inhibitors, peroxisome proliferator‐activated receptor‐gamma agonists, and prostacyclin mimics and phosphodiesterase 3 inhibitors. Several drugs that have relevant properties are licensed for other disorders, offering the possibility of drug repurposing. Others are in development. Since influencing multiple targets may be most effective, using multiple agents and/or those that have multiple effects may be preferable. We focus on potential small vessel disease mechanistic targets, summarize drugs that have relevant actions, and review data available from randomized trials on their actions and on the available evidence for their use in lacunar stroke.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
46
|
Park Y, Jeong YH, Tantry US, Ahn JH, Kim KH, Koh JS, Park JR, Hwang SJ, Kwak CH, Hwang JY, Gurbel PA. Effect of adjunctive dipyridamole to DAPT on platelet function profiles in stented patients with high platelet reactivity. The result of the ACCEL-DIP Study. Thromb Haemost 2014; 112:1198-208. [PMID: 25182660 DOI: 10.1160/th14-01-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 07/05/2014] [Indexed: 12/27/2022]
Abstract
Adjunctive use of phosphodiesterase (PDE) inhibitor can enhance antiplatelet and vasoprotective properties in patients with cardiovascular disease. The aim of this study was to evaluate the impact of PDE5 inhibitor dipyridamole on platelet function in stented patients with high platelet reactivity (HPR) during dual antiplatelet therapy (DAPT) with aspirin and clopidogrel. Patients with HPR after 600-mg clopidogrel loading were randomly assigned to adjunctive dipyridamole 75 mg twice daily to standard DAPT (DIP group; n = 45) or double-dose clopidogrel of 150 mg daily (DOUBLE group; n = 46) for 30 days. Platelet function was assessed at baseline and 30-day follow-up with platelet reactivity index (PRI) by vasodilator-stimulated phosphoprotein-phosphorylation (VASP-P) assay and platelet aggregation (PA) by light transmittance aggregometry (LTA). Primary endpoint was PRI at 30-day follow-up. HPR was defined as PRI > 50%. Baseline platelet function did not differ between the groups. Following 30-day therapy, platelet function was significantly reduced in the DIP and DOUBLE groups (all p-values ≤ 0.004 and ≤ 0.068, respectively). PRI values were not significantly different between the two groups (mean difference: 3.1%; 95% confidence interval: -2.8% to 9.0%: p = 0.295). PA values and prevalence of HPR were similar between the groups. However, a significant number of patients still exhibited HPR in the DIP (75.6%) and DOUBLE (67.4%) groups. In conclusion, among stented HPR patients, adding dipyridamole to DAPT does not reduce platelet reactivity and prevalence of HPR compared with double-dose clopidogrel therapy, and therefore both strategies are inadequate to overcome HPR.
Collapse
Affiliation(s)
| | - Y-H Jeong
- Dr. Young-Hoon Jeong, Division of Cardiology, Department of Internal Medicine, Gyeongsang National University Hospital and, Gyeongsang National University School of Medicine, 79 Gangnam-ro, Jinju, Gyeongsangnam-do, 660-702, Korea, Tel.: +82 55 750 8873, Fax: +82 55 750 8873, E-mail:
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Shang NN, Shao YX, Cai YH, Guan M, Huang M, Cui W, He L, Yu YJ, Huang L, Li Z, Bu XZ, Ke H, Luo HB. Discovery of 3-(4-hydroxybenzyl)-1-(thiophen-2-yl)chromeno[2,3-c]pyrrol-9(2H)-one as a phosphodiesterase-5 inhibitor and its complex crystal structure. Biochem Pharmacol 2014; 89:86-98. [PMID: 24565909 DOI: 10.1016/j.bcp.2014.02.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/12/2014] [Accepted: 02/14/2014] [Indexed: 12/25/2022]
Abstract
Phosphodiesterase-5 (PDE5) inhibitors have been approved for the treatment of erectile dysfunction and pulmonary hypertension, but enthusiasm on discovery of PDE5 inhibitors continues for their potential new applications. Reported here is discovery of a series of new PDE5 inhibitors by structure-based design, molecular docking, chemical synthesis, and enzymatic characterization. The best compound, 3-(4-hydroxybenzyl)-1-(thiophen-2-yl)chromeno[2,3-c]pyrrol-9(2H)-one (57), has an IC₅₀ of 17 nM against the PDE5 catalytic domain and good selectivity over other PDE families. The crystal structure of the PDE5 catalytic domain in complex with 57 was determined at 2Å resolution and showed that 57 occupies the same pocket as other PDE5 inhibitors, but has a different binding pattern in detail. On the basis of the binding pattern of 57, a novel scaffold can be proposed as a candidate of PDE inhibitors.
Collapse
Key Words
- 3-(4-(tert-Butoxy)benzyl)-1-phenylchromeno[2,3-c]pyrrol-9(2H)-one (ACP43
- 3-(4-Hydroxybenzyl)-1-(thiophen-2-yl)chromeno[2,3-c]pyrrol-9(2H)-one (ACP57
- 3-(4-Hydroxybenzyl)-1-phenylchromeno[2,3-c]pyrrol-9(2H)-one (ACP61
- 3-(4-Hydroxybenzyl)-2-methyl-1-phenylchromeno[2,3-c]pyrrol-9(2H)-one (ACP62
- 3-(9-Oxo-1-phenyl-2,9-dihydrochromeno[2,3-c]pyrrol-3-yl)propanoic acid (ACP52
- 3-Benzyl-1-phenylchromeno[2,3-c]pyrrol-9(2H)-one (ACP42
- Crystal structure
- Guanosine 3′,5′-cyclic monophosphate
- PDE5 inhibitor
- Phosphodiesterase
- PubChem CID: 49784789)
- PubChem CID: 54770534)
- PubChem CID: 54770536)
- PubChem CID: 71738344)
- PubChem CID: 71738345)
- PubChem CID: 71765666)
- PubChem CID: 72725677)
- Structure-based molecular design
- tert-Butyl-2-(9-oxo-1-phenyl-2,9-dihydrochromeno[2,3-c]pyrrol-3-yl)acetate (ACP37
Collapse
Affiliation(s)
- Na-Na Shang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Yong-Xian Shao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Ying-Hong Cai
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Matthew Guan
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Manna Huang
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Wenjun Cui
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA
| | - Lin He
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Yan-Jun Yu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Lei Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Zhe Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Xian-Zhang Bu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China.
| | - Hengming Ke
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA.
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China.
| |
Collapse
|
48
|
Lukasik M, Owecki MK. Efficacy of Antiplatelet Treatment in Stroke Prevention: Past, Present, and Future. Drug Dev Res 2013. [DOI: 10.1002/ddr.21100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Maria Lukasik
- Department of Neurology; Poznan University of Medical Sciences; Poznan; Poland
| | - Michal K. Owecki
- Department of Neurology; Poznan University of Medical Sciences; Poznan; Poland
| |
Collapse
|