113
Views
7
CrossRef citations to date
0
Altmetric
Original Research

Nano-scaled MTCA-KKV: for targeting thrombus, releasing pharmacophores, inhibiting thrombosis and dissolving blood clots in vivo

, , , , , , , , & show all
Pages 4817-4831 | Published online: 03 Jul 2019

References

  • Moretti A, Ferrari F, Villa RF. Pharmacological therapy of acute ischaemic stroke: achievements and problems. Pharmacol Ther. 2015;146:23–34. doi:10.1016/j.pharmthera.2014.09.00325196155
  • Granger DN, Kvietys PR. Reperfusion therapy – what’s with the obstructed, leaky and broken capillaries? Pathophysiology. 2017;24:213–228. doi:10.1016/j.pathophys.2017.09.00329102280
  • Bonechi C, Lamponi S, Donati A, et al. Effect of resveratrol on platelet aggregation by fibrinogen protection. Biophys Chem. 2017;222:41. doi:10.1016/j.bpc.2016.12.00428095333
  • Bobbert P, Weikert U, Schmidt-Lucke C, et al. Platelet activation and thrombus formation relates to the presence of myocardial inflammation in patients with cardiomyopathy. J Cardiol. 2013;63(5):379–384. doi:10.1016/j.jjcc.2013.06.01824230464
  • Tesfamariam B. Distinct characteristics of neonatal platelet reactivity. Pharmacol Res. 2017;123:1–9. doi:10.1016/j.phrs.2017.06.00328625868
  • Spann AP, Campbell JE, Fitzgibbon SR, et al. The effect of hematocrit on platelet adhesion: experiments and simulations. Biophys J. 2016;111(3):577–588. doi:10.1016/j.bpj.2016.06.02427508441
  • Faggio C, Sureda A, Morabito S, et al. Flavonoids and platelet aggregation: a brief review. Eur J Pharmacol. 2017;807:91–101. doi:10.1016/j.ejphar.2017.04.00928412372
  • Ghasemzadeh M, Hosseini E. Platelet granule release is associated with reactive oxygen species generation during platelet storage: a direct link between platelet pro-inflammatory and oxidation states. Thromb Res. 2017;156:101. doi:10.1016/j.thromres.2017.06.01628623810
  • Milioli M, Ib´A˜nezvea M, Sidoli S, et al. Quantitative proteomics analysis of platelet-derived microparticles reveals distinct protein signatures when stimulated by different physiological agonists. J Proteomics. 2015;121:56–66. doi:10.1016/j.jprot.2015.03.01325835965
  • Lee W, Ku SK, Kim MA, Bae J-S. Anti-factor Xa activities of zingerone with anti-platelet aggregation. Activity. Food Chem Toxicol. 2017;105:186–193. doi:10.1016/j.fct.2017.04.01228414123
  • Fan H, Li M, Yu L, et al. Effects of Danhong injection on platelet aggregation in hyperlipidemia rats. J Ethnopharmacol. 2017;212:67–73. doi:10.1016/j.jep.2017.09.01529066405
  • Lee W, Kim MA, Park I, et al. Novel direct factor Xa inhibitory compounds from Tenebrio molitor with anti-platelet aggregation activity. Food Chem Toxicol. 2017;109:19–27. doi:10.1016/j.fct.2017.08.02628844963
  • Ottaiano TF, Andrade SS, De OC, et al. Plasma kallikrein enhances platelet aggregation response by subthreshold doses of ADP. Biochimie. 2017;135:72–81. doi:10.1016/j.biochi.2017.01.01028115185
  • Son YM, Jeong DH, Park HJ, Rhee MH. The inhibitory activity of ginsenoside Rp4 in adenosine diphosphate-induced platelet aggregation. J Ginseng Res. 2017;41:96–102. doi:10.1016/j.jgr.2016.01.00328123327
  • Grundler K, Rotter R, Tilley S, et al. The proteasome regulates collagen-induced platelet aggregation via nuclear-factor-kappa-B (NFĸB) activation. Thromb Res. 2016;148:15–22. doi:10.1016/j.thromres.2016.10.00927768934
  • Olivier CB, Weik P, Meyer M, et al. TRAP-induced platelet aggregation is enhanced in cardiovascular patients receiving dabigatran. Thromb Res. 2015;138:63–68. doi:10.1016/j.thromres.2015.10.03826610745
  • Hally KE, La Flamme AC, Larsen PD, Harding SA. Platelet Toll-like receptor (TLR) expression and TLR-mediated platelet activation in acute myocardial infarction. Thromb Res. 2017;158:8–15. doi:10.1016/j.thromres.2017.07.03128783513
  • Gupta S, Reviakine I. The sweeter aspects of platelet activation: A lectin-based assay reveals agonist-specific glycosylation patterns. Biochim Biophys Acta. 2014;1840(12):3423–3433. doi:10.1016/j.bbagen.2014.08.01025175560
  • Bijak M, Dziedzic A, Saluk-Bijak J. Flavonolignans reduce the response of blood platelet to collagenMichal. Int J Biol Macromol. 2017;106:878–884. doi:10.1016/j.ijbiomac.2017.08.09128842200
  • Tafelmeier M, Fischer A, Orsó E, et al. Mildly oxidized HDL decrease agonist-induced platelet aggregation and release of pro-coagulant platelet extracellular vesicles. J Steroid Biochem Mol Biol. 2016;169:176–188. doi:10.1016/j.jsbmb.2016.05.00327163393
  • Honda Y, Kamisato C, Morishima Y. Prevention of arterial thrombosis by edoxaban, an oral factor Xa inhibitor in rats: monotherapy and in combination with antiplatelet agents. Eur J Pharmacol. 2016;786:246–252. doi:10.1016/j.ejphar.2016.06.01127288116
  • Fuentes E, Palomo I. Antiplatelet effects of natural bioactive compounds by multiple targets: food and drug interactions. J Funct Foods. 2014;6(1):73–81. doi:10.1016/j.jff.2013.10.012
  • Navarese EP, Kolodziejczak M, Winter MP, et al. Association of PCSK9 with platelet reactivity in patients with acute coronary syndrome treated with prasugrel or ticagrelor: the PCSK9-REACT study. Int J Cardiol. 2016;227:644–649. doi:10.1016/j.ijcard.2016.10.08427810295
  • Redfors B, Généreux P, Witzenbichler B, et al. Association of PCSK9 with platelet reactivity in patients with acute coronary syndrome treated with prasugrel or ticagrelor: the PCSK9-REACT study. Int J Cardiol. 2017;250:92–97. doi:10.1016/j.ijcard.2017.10.02229074039
  • Clavijo LC, Al-Asady N, Dhillon A, et al. Prevalence of high on-treatment (aspirin and clopidogrel) platelet reactivity in patients with critical limb ischemia. Cardiovasc Revasc Med. 2018;19(5 Pt A):516–520. doi:10.1016/j.carrev.2017.10.01329221961
  • Makowski M, Smorag I, Makowska J, et al. Platelet reactivity and mean platelet volume as risk markers of thrombogenesis in atrial fibrillation. Int J Cardiol. 2017;235:1–5. doi:10.1016/j.ijcard.2017.03.02328302320
  • Rosafio F, Lelli N, Mimmi S, et al. Platelet function testing in patients with acute ischemic stroke: an observational study. J Stroke Cerebrovasc Dis. 2017;26(8):1864–1873. doi:10.1016/j.jstrokecerebrovasdis.2017.07.02828576652
  • Meves SH, Schröder KD, Endres HG, Krogias C, Krüger JC, Neubauer H. Neubauer, Clopidogrel high-on-treatment platelet reactivity in acute ischemic stroke patients. Thromb Res. 2014;133(3):396–401. doi:10.1016/j.thromres.2013.10.03224406048
  • Rao Z, Zheng H, Wang F, et al. High on-treatment platelet reactivity to adenosine diphosphate predicts ischemic events of minor stroke and transient ischemic attack. J Stroke Cerebrovasc Dis. 2017;26(10):2074–2081. doi:10.1016/j.jstrokecerebrovasdis.2017.07.02828736132
  • Qiu L, Sun Y, Wang L, et al. Influence of CYP2C19 polymorphisms on platelet reactivity and clinical outcomes in ischemic stroke patients treated with clopidogrel. Eur J Pharmacol. 2015;747:29–35. doi:10.1016/j.ejphar.2014.11.03725489921
  • Malhotra N, Abunassar J, Wells GA, et al. A pharmacodynamic comparison of a personalized strategy for anti-platelet therapy versus ticagrelor in achieving a therapeutic window. Int J Cardiol. 2015;197:318–325. doi:10.1016/j.ijcard.2015.06.01626151596
  • Girotra C, Padhye M, Mandlik G, et al. Assessment of the risk of haemorrhage and its control following minor oral surgical procedures in patients on anti-platelet therapy: a prospective study. Int J Oral Maxillofac Surg. 2014;43(1):99–106. doi:10.1016/j.ijom.2013.07.00424074486
  • Liou K, Lambros J. Delayed left ventricular apical thrombus formation following discontinuation of dual anti-platelet therapy. Heart Lung Circ. 2014;23:e237–e239. doi:10.1016/j.hlc.2013.11.01325127668
  • Kai H, Kohro T, Fukuda K, Yamazaki T, Nagai R. Impact of systolic blood pressure on hemorrhagic stroke in patients with coronary artery disease during anti-platelet therapy: the Japanese Coronary Artery Disease (JCAD) study. Int J Cardiol. 2016;224:112–113. doi:10.1016/j.ijcard.2016.09.00427648978
  • Cai TQ, Alexandra WL, Sitko G, et al. Platelet transfusion reverses bleeding evoked by triple anti-platelet therapy including vorapaxar, a novel platelet thrombin receptor antagonist. Eur J Pharmacol. 2015;758:107–114. doi:10.1016/j.ejphar.2015.03.07325857224
  • Hally KE, La Flamme AC, Larsen PD, Harding SA. Toll-like receptor 9 expression and activation in acute coronary syndrome patients on dual anti-platelet therapy. Thromb Res. 2016;148:89–95. doi:10.1016/j.thromres.2016.10.02627815972
  • Massicotte MP, Maul TM, Snyder TA, Kreuziger LB. Mechanical circulatory support and antithrombotic therapy: looking for the holy grail. Asaio J. 2017;63(1):1–4. doi:10.1097/MAT.000000000000050028030445
  • Boccalandro F, Dhindsa M, Subramaniyam P, Mok M. Feasibility of coronary fractional flow reserve with dual anti-platelet therapy in low risk coronary lesions without systemic anticoagulation-results of the SMART-FFR study. Cardiovasc Revasc Med. 2018:19(3):343–347
  • Burnouf T, Burnouf PA, Wu Y, Chuang E, Lu L, Goubran H. Circulatory-cell-mediated nanotherapeutic approaches in disease targeting. Drug Discov Today. 2018;23(5):934–943. doi:10.1016/j.drudis.2017.08.01228917821
  • Kong Y, Xu C, He Z, et al. A novel peptide inhibitor of platelet aggregation from stiff silkworm, Bombyx batryticatus. Peptides. 2014;53:70–78. doi:10.1016/j.peptides.2013.12.00424361453
  • Pawlowski CL, Wei L, Sun M, et al. Platelet microparticle-inspired clot-responsive nanomedicine for targeted fibrinolysis. Biomaterials. 2017;128:94–108. doi:10.1016/j.biomaterials.2017.03.01228314136
  • Koudelka S, Mikulik R, Mašek J, et al. Liposomal nanocarriers for plasminogen activators. J Controlled Release. 2016;227:45. doi:10.1016/j.jconrel.2016.02.019
  • Pescador R, Capuzzi L, Mantovani M, Fulgenzi A, Ferrero ME. Defibrotide: properties and clinical use of an old/new drug. Vascul Pharmacol. 2013;59(1–2):1–10. doi:10.1016/j.vph.2013.05.00123680861
  • Khan FA, Maalik A, Iqbal Z, Malik I. Recent pharmacological developments in β-carboline alkaloid “harmaline”. Eur J Pharmacol. 2013;721(1–3):391–394. doi:10.1016/j.ejphar.2013.05.00323707188
  • Seabrooks L, Hu L. Insects: an underrepresented resource for the discovery of biologically active natural products. Acta Pharmaceutica Sinica B. 2017;7(4):409–426. doi:10.1016/j.apsb.2017.05.00128752026
  • Eguchi K, Fujiwara Y, Hayashida A, et al. a marine-derived alkaloid, inhibits accumulation of cholesterol ester in macrophages and suppresses hyperlipidemia and atherosclerosis in vivo. Bioorg Med Chem. 2013;21(13):3831–3838. doi:10.1016/j.bmc.2013.04.02523665143
  • Wang M, Rakesh KP, Leng J, et al. Amino acids/peptides conjugated heterocycles: A tool for the recent development of novel therapeutic agents. Bioorg Chem. 2018;76:113–129. doi:10.1016/j.bioorg.2017.11.00729169078
  • Hosseinkhani H, Hosseinkhani M, Khademhosseini A, Kobayashi H, Tabata Y. Enhanced angiogenesis through controlled release of basic fibroblast growth factor from peptide amphiphile for tissue regeneration. Biomaterials. 2006;27(34):5836–5844. doi:10.1016/j.biomaterials.2006.08.00316930687
  • Hosseinkhani H, Hiraoka Y, Li CH, et al. Engineering three-dimensional collagen-IKVAV matrixto mimic neural microenvironment. ACS Chem Neurosci. 2013;4(8):1229–1235. doi:10.1021/cn400008223705903
  • Jun Kameoka SSV, Liu H, And DAC, Craighead HG. Fabrication of suspended silica glass nanofibers from polymeric materials using as canned electrospinning source. Nano Lett. 2004;4(11):2105–2108. doi:10.1021/nl048840p
  • Gao S, Han L, Zhu L, et al. One-step integration of multiple genes into the oleaginous yeast Yarrowia lipolytica. Biotechnol Lett. 2014;36(12):2523. doi:10.1007/s10529-014-1634-y25216641
  • Li Z, Huang F, Wu JH, et al. Heptapeptide-based modification leading to enhancing the action of MTCA on activated platelets, P-selectin, GPIIb/IIIa. Future Medicinal Chemistry. 2018; 10(16): 1957–1970. doi:10.4155/fmc-2018-0055
  • Wu JH, Zhu HM, Yang GD, et al. Design and synthesis of nano-scaled IQCA-TAVV as a delivery capable of anti- platelet activation, targeting arterial thrombus and releasing IQCA. Int J Nanomed. 2018;13:1139–1158. doi:10.2147/IJN.S150205
  • Wu JH, Zhu HM, Zhao M, et al. IQCA-TASS: A nano-scaled P-selectin inhibitor capable of targeting thrombus and releasing IQCA/TARGD(S)S in vivo. J Mater Chem B. 2017;5:917–927. doi:10.1039/C6TB02705A
  • Ma HP, Zhao M, Wang YJ, et al. Cholyl-L-Lysine-carboxyl butyryl adriamycin produgs targeting chemically induced liver injury. J Mater Chem B. 2017;5:470–478. doi:10.1039/C6TB02205G
  • Fujita Y, Mie M, Kobatake E. Construction of nanoscale protein particle using temperature-sensitive elastin-like peptide and polyaspartic acid chain. Biomaterials. 2009;30:3450–3457. doi:10.1016/j.biomaterials.2009.03.01219324406