Thromb Haemost 2023; 123(02): 207-218
DOI: 10.1055/a-1962-1613
Cellular Haemostasis and Platelets

Impaired Platelet Function and Thrombus Formation in PDE5A-Deficient Mice

Xiang Gui#
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Xiang Chu#
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Yuwei Du
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Yuhan Wang
4   School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
,
Sixuan Zhang
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Yangyang Ding
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Huan Tong
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Mengdi Xu
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Yue Li
4   School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
,
Wen Ju
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Zengtian Sun
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Zhenyu Li
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Lingyu Zeng*
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
4   School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
,
Kailin Xu*
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
,
Jianlin Qiao*
1   Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China
2   Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
3   Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
› Author Affiliations
Funding This work was supported by the National Natural Science Foundation of China (grant no. 82170130, 81970124, 81400082, 81641151, and 81700178), the Natural Science Foundation of Jiangsu Province (grant no. BK20140219 and BK20170259), the funding for the Distinguished Professorship Program of Jiangsu Province, the Shuangchuang Project of Jiangsu Province, the Six Talent Peaks Project of Jiangsu Province (WSN-133), the 333 projects of Jiangsu Province (BRA2017542), the Natural Science Foundation of the Jiangsu Higher Education Institutions of China (18KJA320010 and 17KJA320008), Jiangsu Province's Key Provincial Talents Program (ZDRCA2016054), Jiangsu Province's Graduate Scientific Research Innovation Program (KYCX22-2896 and KYCX21-2691), and Youth Science and Technology Innovation Team of Xuzhou Medical University.


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.

Author Contributions

X. Gui and X. Chu performed research, analyzed data, and wrote the manuscript. S. Zhang, Y. Wang, Y. Du, Y. Ding, H. Tong, M. Xu, Y. Li, W. Ju, Z. Sun, and Z. Li performed research and analyzed data, L. Zeng, K. Xu, and J. Qiao conceived and designed the study and wrote the manuscript.


# These authors share first authorship.


* These authors share senior authorship.


Supplementary Material



Publication History

Received: 29 June 2022

Accepted: 11 October 2022

Accepted Manuscript online:
17 October 2022

Article published online:
30 December 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Estevez B, Du X. New concepts and mechanisms of platelet activation signaling. Physiology (Bethesda) 2017; 32 (02) 162-177
  • 2 Grover SP, Bergmeier W, Mackman N. Platelet signaling pathways and new inhibitors. Arterioscler Thromb Vasc Biol 2018; 38 (04) e28-e35
  • 3 Durrant TN, van den Bosch MT, Hers I. Integrin αIIbβ3 outside-in signaling. Blood 2017; 130 (14) 1607-1619
  • 4 Nagy Z, Smolenski A. Cyclic nucleotide-dependent inhibitory signaling interweaves with activating pathways to determine platelet responses. Res Pract Thromb Haemost 2018; 2 (03) 558-571
  • 5 Stefanini L, Bergmeier W. Negative regulators of platelet activation and adhesion. J Thromb Haemost 2018; 16 (02) 220-230
  • 6 Smolenski A. Novel roles of cAMP/cGMP-dependent signaling in platelets. J Thromb Haemost 2012; 10 (02) 167-176
  • 7 Altschuler D, Lapetina EG. Mutational analysis of the cAMP-dependent protein kinase-mediated phosphorylation site of Rap1b. J Biol Chem 1993; 268 (10) 7527-7531
  • 8 Manganello JM, Djellas Y, Borg C, Antonakis K, Le Breton GC. Cyclic AMP-dependent phosphorylation of thromboxane A(2) receptor-associated Galpha(13). J Biol Chem 1999; 274 (39) 28003-28010
  • 9 El-Daher SS, Patel Y, Siddiqua A. et al. Distinct localization and function of (1,4,5)IP(3) receptor subtypes and the (1,3,4,5)IP(4) receptor GAP1(IP4BP) in highly purified human platelet membranes. Blood 2000; 95 (11) 3412-3422
  • 10 Schinner E, Salb K, Schlossmann J. Signaling via IRAG is essential for NO/cGMP-dependent inhibition of platelet activation. Platelets 2011; 22 (03) 217-227
  • 11 Mullershausen F, Friebe A, Feil R, Thompson WJ, Hofmann F, Koesling D. Direct activation of PDE5 by cGMP: long-term effects within NO/cGMP signaling. J Cell Biol 2003; 160 (05) 719-727
  • 12 Butt E, Abel K, Krieger M. et al. cAMP- and cGMP-dependent protein kinase phosphorylation sites of the focal adhesion vasodilator-stimulated phosphoprotein (VASP) in vitro and in intact human platelets. J Biol Chem 1994; 269 (20) 14509-14517
  • 13 Conti M, Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem 2007; 76: 481-511
  • 14 Omori K, Kotera J. Overview of PDEs and their regulation. Circ Res 2007; 100 (03) 309-327
  • 15 Haslam RJ, Dickinson NT, Jang EK. Cyclic nucleotides and phosphodiesterases in platelets. Thromb Haemost 1999; 82 (02) 412-423
  • 16 Dunkern TR, Hatzelmann A. The effect of Sildenafil on human platelet secretory function is controlled by a complex interplay between phosphodiesterases 2, 3 and 5. Cell Signal 2005; 17 (03) 331-339
  • 17 Feijge MA, Ansink K, Vanschoonbeek K, Heemskerk JW. Control of platelet activation by cyclic AMP turnover and cyclic nucleotide phosphodiesterase type-3. Biochem Pharmacol 2004; 67 (08) 1559-1567
  • 18 Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev 2006; 58 (03) 488-520
  • 19 Kass DA, Champion HC, Beavo JA. Phosphodiesterase type 5: expanding roles in cardiovascular regulation. Circ Res 2007; 101 (11) 1084-1095
  • 20 Berkels R, Klotz T, Sticht G, Englemann U, Klaus W. Modulation of human platelet aggregation by the phosphodiesterase type 5 inhibitor sildenafil. J Cardiovasc Pharmacol 2001; 37 (04) 413-421
  • 21 Halcox JP, Nour KR, Zalos G. et al. The effect of sildenafil on human vascular function, platelet activation, and myocardial ischemia. J Am Coll Cardiol 2002; 40 (07) 1232-1240
  • 22 Arthur JF, Qiao J, Shen Y. et al. ITAM receptor-mediated generation of reactive oxygen species in human platelets occurs via Syk-dependent and Syk-independent pathways. J Thromb Haemost 2012; 10 (06) 1133-1141
  • 23 Qiao J, Wu X, Luo Q. et al. NLRP3 regulates platelet integrin αIIbβ3 outside-in signaling, hemostasis and arterial thrombosis. Haematologica 2018; 103 (09) 1568-1576
  • 24 Wang X, Zhang S, Ding Y. et al. p47phox deficiency impairs platelet function and protects mice against arterial and venous thrombosis. Redox Biol 2020; 34: 101569
  • 25 Wei G, Xu X, Tong H. et al. Salidroside inhibits platelet function and thrombus formation through AKT/GSK3β signaling pathway. Aging (Albany NY) 2020; 12 (09) 8151-8166
  • 26 Montoro-García S, Schindewolf M, Stanford S, Larsen OH, Thiele T. The role of platelets in venous thromboembolism. Semin Thromb Hemost 2016; 42 (03) 242-251
  • 27 Flevaris P, Stojanovic A, Gong H, Chishti A, Welch E, Du X. A molecular switch that controls cell spreading and retraction. J Cell Biol 2007; 179 (03) 553-565
  • 28 Clark EA, Shattil SJ, Ginsberg MH, Bolen J, Brugge JS. Regulation of the protein tyrosine kinase pp72syk by platelet agonists and the integrin alpha IIb beta 3. J Biol Chem 1994; 269 (46) 28859-28864
  • 29 Suzuki-Inoue K, Hughes CE, Inoue O. et al. Involvement of Src kinases and PLCgamma2 in clot retraction. Thromb Res 2007; 120 (02) 251-258
  • 30 Abbasian N, Millington-Burgess SL, Chabra S, Malcor JD, Harper MT. Supramaximal calcium signaling triggers procoagulant platelet formation. Blood Adv 2020; 4 (01) 154-164
  • 31 Arachiche A, Kerbiriou-Nabias D, Garcin I, Letellier T, Dachary-Prigent J. Rapid procoagulant phosphatidylserine exposure relies on high cytosolic calcium rather than on mitochondrial depolarization. Arterioscler Thromb Vasc Biol 2009; 29 (11) 1883-1889
  • 32 Chakrabarti S, Clutton P, Varghese S, Cox D, Mascelli MA, Freedman JE. Glycoprotein IIb/IIIa inhibition enhances platelet nitric oxide release. Thromb Res 2004; 113 (3–4): 225-233
  • 33 Clutton P, Miermont A, Freedman JE. Regulation of endogenous reactive oxygen species in platelets can reverse aggregation. Arterioscler Thromb Vasc Biol 2004; 24 (01) 187-192
  • 34 Gresele P, Momi S, Falcinelli E. Anti-platelet therapy: phosphodiesterase inhibitors. Br J Clin Pharmacol 2011; 72 (04) 634-646
  • 35 Rondina MT, Weyrich AS. Targeting phosphodiesterases in anti-platelet therapy. Handb Exp Pharmacol 2012; (210) 225-238
  • 36 Brito FC, Kummerle AE, Lugnier C, Fraga CA, Barreiro EJ, Miranda AL. Novel thienylacylhydrazone derivatives inhibit platelet aggregation through cyclic nucleotides modulation and thromboxane A2 synthesis inhibition. Eur J Pharmacol 2010; 638 (1–3): 5-12
  • 37 Lima LM, Ormelli CB, Brito FF, Miranda AL, Fraga CA, Barreiro EJ. Synthesis and antiplatelet evaluation of novel aryl-sulfonamide derivatives, from natural safrole. Pharm Acta Helv 1999; 73 (06) 281-292
  • 38 Sun B, Li H, Shakur Y. et al. Role of phosphodiesterase type 3A and 3B in regulating platelet and cardiac function using subtype-selective knockout mice. Cell Signal 2007; 19 (08) 1765-1771
  • 39 Tani T, Sakurai K, Kimura Y, Ishikawa T, Hidaka H. Pharmacological manipulation of tissue cyclic AMP by inhibitors. Effects of phosphodiesterase inhibitors on the functions of platelets and vascular endothelial cells. Adv Second Messenger Phosphoprotein Res 1992; 25: 215-227
  • 40 De Bon E, Bonanni G, Saggiorato G, Bassi P, Cella G. Effects of tadalafil on platelets and endothelium in patients with erectile dysfunction and cardiovascular risk factors: a pilot study. Angiology 2010; 61 (06) 602-606
  • 41 Manoury B, Idres S, Leblais V, Fischmeister R. Ion channels as effectors of cyclic nucleotide pathways: functional relevance for arterial tone regulation. Pharmacol Therapeut 2020; 209: 107499
  • 42 Morgado M, Cairrão E, Santos-Silva AJ, Verde I. Cyclic nucleotide-dependent relaxation pathways in vascular smooth muscle. Cell Mol Life Sci 2012; 69 (02) 247-266
  • 43 Zhang G, Xiang B, Dong A. et al. Biphasic roles for soluble guanylyl cyclase (sGC) in platelet activation. Blood 2011; 118 (13) 3670-3679
  • 44 Weiss HJ. Impaired platelet procoagulant mechanisms in patients with bleeding disorders. Semin Thromb Hemost 2009; 35 (02) 233-241
  • 45 Zwaal RF, Schroit AJ. Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood 1997; 89 (04) 1121-1132
  • 46 Reddy EC, Rand ML. Procoagulant Phosphatidylserine-Exposing Platelets in vitro and in vivo . Front Cardiovasc Med 2020; 7: 15
  • 47 Agbani EO, Poole AW. Procoagulant platelets: generation, function, and therapeutic targeting in thrombosis. Blood 2017; 130 (20) 2171-2179
  • 48 Toque HA, Teixeira CE, Priviero FB, Morganti RP, Antunes E, De Nucci G. Vardenafil, but not sildenafil or tadalafil, has calcium-channel blocking activity in rabbit isolated pulmonary artery and human washed platelets. Br J Pharmacol 2008; 154 (04) 787-796
  • 49 Ahmed WS, Geethakumari AM, Biswas KH. Phosphodiesterase 5 (PDE5): structure-function regulation and therapeutic applications of inhibitors. Biomed Pharmacother 2021; 134: 111128
  • 50 Shi J, Ross CR, Leto TL, Blecha F. PR-39, a proline-rich antibacterial peptide that inhibits phagocyte NADPH oxidase activity by binding to Src homology 3 domains of p47 phox. Proc Natl Acad Sci U S A 1996; 93 (12) 6014-6018
  • 51 Son Y, Cheong YK, Kim NH, Chung HT, Kang DG, Pae HO. Mitogen-activated protein kinases and reactive oxygen species: how can ROS activate MAPK pathways?. J Signal Transduct 2011; 2011: 792639