Review Article

可溶性鸟苷酸环化酶的治疗靶向

卷 26, 期 15, 2019

页: [2730 - 2747] 页: 18

弟呕挨: 10.2174/0929867326666190108095851

价格: $65

摘要

可溶性鸟苷酸环化酶(sGC)是一氧化氮的生理传感器,其功能的改变主动涉及多种病理生理条件。过去20年来的大量研究工作提供了有关其监管的重要信息,最终合理开发了经批准的药物或研究性铅分子,这些分子通过新机制靶向并与sGC相互作用。但是,有许多问题仍然没有答案。正在进行的研究,结构化学研究的关键帮助,试图进一步阐明酶的结构特征,分别定义血红素部分存在或不存在时sGC的“刺激物”或“激活剂”的关联,以及精确的构象属性,将允许设计更具创新性和有效的药物。该综述涉及在理解该酶的功能方面取得的进展,特别是在过去10年中所取得的进展,并着重于a)其在疾病情况下的治疗靶向的基本原理和结果,取决于酶的状态(氧化与否,血红蛋白携带与否)和b)最近的结构研究,应该允许改进设计未来的治疗分子,旨在直接上调sGC的活动。

关键词: 6可溶性鸟苷酸环化酶(sGC),环磷酸鸟苷(cGMP),一氧化氮(NO),sGC激活剂,sGC刺激剂,H-NOX结构域,血红素,Fe。

[1]
Feil, R.; Kemp-Harper, B. cGMP signalling: From bench to bedside. Conference on cGMP generators, effectors and therapeutic implications. EMBO Rep., 2006, 7(2), 149-153.
[http://dx.doi.org/10.1038/sj.embor.7400627] [PMID: 16439998]
[2]
Hobbs, A.J. Soluble guanylate cyclase: An old therapeutic target re-visited. Br. J. Pharmacol., 2002, 136(5), 637-640.
[http://dx.doi.org/10.1038/sj.bjp.0704779] [PMID: 12086972]
[3]
Alderton, W.K.; Cooper, C.E.; Knowles, R.G. Nitric oxide synthases: Structure, function and inhibition. Biochem. J., 2001, 357(Pt 3), 593-615.
[http://dx.doi.org/10.1042/bj3570593] [PMID: 11463332]
[4]
Stuehr, D.J. Structure-function aspects in the nitric oxide synthases. Annu. Rev. Pharmacol. Toxicol., 1997, 37, 339-359.
[http://dx.doi.org/10.1146/annurev.pharmtox.37.1.339] [PMID: 9131257]
[5]
Sanders, K.M.; Ward, S.M. Nitric oxide as a mediator of nonadrenergic noncholinergic neurotransmission. Am. J. Physiol., 1992, 262(3 Pt 1), G379-G392.
[http://dx.doi.org/10.1152/ajpgi.1992.262.3.G379] [PMID: 1347974]
[6]
Erusalimsky, J.D.; Moncada, S. Nitric oxide and mitochondrial signaling: From physiology to pathophysiology. Arterioscler. Thromb. Vasc. Biol., 2007, 27(12), 2524-2531.
[http://dx.doi.org/10.1161/ATVBAHA.107.151167] [PMID: 17885213]
[7]
Lima, B.; Forrester, M.T.; Hess, D.T.; Stamler, J.S. S-nitrosylation in cardiovascular signaling. Circ. Res., 2010, 106(4), 633-646.
[http://dx.doi.org/10.1161/CIRCRESAHA.109.207381] [PMID: 20203313]
[8]
Ashman, D.F.; Lipton, R.; Melicow, M.M.; Price, T.D. Isolation of adenosine 3′, 5′-monophosphate and guanosine 3′, 5′-monophosphate from rat urine. Biochem. Biophys. Res. Commun., 1963, 11, 330-334.
[http://dx.doi.org/10.1016/0006-291X(63)90566-7] [PMID: 13965190]
[9]
Robison, G.A.; Butcher, R.W.; Sutherland, E.W. Cyclic AMP. Annu. Rev. Biochem., 1968, 37, 149-174.
[http://dx.doi.org/10.1146/annurev.bi.37.070168.001053] [PMID: 4299844]
[10]
Hardman, J.G.; Robison, G.A.; Sutherland, E.W. Cyclic nucleotides. Annu. Rev. Physiol., 1971, 33, 311-336.
[http://dx.doi.org/10.1146/annurev.ph.33.030171.001523] [PMID: 4157117]
[11]
Furchgott, R.F.; Zawadzki, J.V. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature, 1980, 288(5789), 373-376.
[http://dx.doi.org/10.1038/288373a0] [PMID: 6253831]
[12]
Ignarro, L.J.; Byrns, R.E.; Buga, G.M.; Wood, K.S.; Chaudhuri, G. Pharmacological evidence that endothelium-derived relaxing factor is nitric oxide: Use of pyrogallol and superoxide dismutase to study endothelium-dependent and nitric oxide-elicited vascular smooth muscle relaxation. J. Pharmacol. Exp. Ther., 1988, 244(1), 181-189.
[PMID: 2826766]
[13]
Mittal, C.K.; Arnold, W.P.; Murad, F. Characterization of protein inhibitors of guanylate cyclase activation from rat heart and Bovine lung., Liver, 1978, 37(54.8), 1.5.
[14]
Koesling, D.; Russwurm, M.; Mergia, E.; Mullershausen, F.; Friebe, A. Nitric oxide-sensitive guanylyl cyclase: Structure and regulation. Neurochem. Int., 2004, 45(6), 813-819.
[http://dx.doi.org/10.1016/j.neuint.2004.03.011] [PMID: 15312975]
[15]
Yuen, P.S.; Potter, L.R.; Garbers, D.L. A new form of guanylyl cyclase is preferentially expressed in rat kidney. Biochemistry, 1990, 29(49), 10872-10878.
[http://dx.doi.org/10.1021/bi00501a002] [PMID: 1980215]
[16]
Harteneck, C.; Wedel, B.; Koesling, D.; Malkewitz, J.; Böhme, E.; Schultz, G. Molecular cloning and expression of a new alpha-subunit of soluble guanylyl cyclase. Interchangeability of the alpha-subunits of the enzyme. FEBS Lett., 1991, 292(1-2), 217-222.
[http://dx.doi.org/10.1016/0014-5793(91)80871-Y] [PMID: 1683630]
[17]
Koesling, D. Studying the structure and regulation of soluble guanylyl cyclase. Methods, 1999, 19(4), 485-493.
[http://dx.doi.org/10.1006/meth.1999.0891] [PMID: 10581148]
[18]
Ma, X.; Sayed, N.; Baskaran, P.; Beuve, A.; van den Akker, F. PAS-mediated dimerization of soluble guanylyl cyclase revealed by signal transduction histidine kinase domain crystal structure. J. Biol. Chem., 2008, 283(2), 1167-1178.
[http://dx.doi.org/10.1074/jbc.M706218200] [PMID: 18006497]
[19]
Rothkegel, C.; Schmidt, P.M.; Atkins, D.J.; Hoffmann, L.S.; Schmidt, H.H.; Schröder, H.; Stasch, J.P. Dimerization region of soluble guanylate cyclase characterized by bimolecular fluorescence complementation in vivo. Mol. Pharmacol., 2007, 72(5), 1181-1190.
[http://dx.doi.org/10.1124/mol.107.036368] [PMID: 17715400]
[20]
Underbakke, E.S.; Iavarone, A.T.; Chalmers, M.J.; Pascal, B.D.; Novick, S.; Griffin, P.R.; Marletta, M.A. Nitric oxide-induced conformational changes in soluble guanylate cyclase. Structure, 2014, 22(4), 602-611.
[http://dx.doi.org/10.1016/j.str.2014.01.008] [PMID: 24560804]
[21]
Winger, J.A.; Marletta, M.A. Expression and characterization of the catalytic domains of soluble guanylate cyclase: Interaction with the heme domain. Biochemistry, 2005, 44(10), 4083-4090.
[http://dx.doi.org/10.1021/bi047601d] [PMID: 15751985]
[22]
Gerzer, R.; Böhme, E.; Hofmann, F.; Schultz, G. Soluble guanylate cyclase purified from bovine lung contains heme and copper. FEBS Lett., 1981, 132(1), 71-74.
[http://dx.doi.org/10.1016/0014-5793(81)80429-2] [PMID: 6117479]
[23]
Gerzer, R.; Radany, E.W.; Garbers, D.L. The separation of the heme and apoheme forms of soluble guanylate cyclase. Biochem. Biophys. Res. Commun., 1982, 108(2), 678-686.
[http://dx.doi.org/10.1016/0006-291X(82)90883-X] [PMID: 6128979]
[24]
Humbert, P.; Niroomand, F.; Fischer, G.; Mayer, B.; Koesling, D.; Hinsch, K.D.; Gausepohl, H.; Frank, R.; Schultz, G.; Böhme, E. Purification of soluble guanylyl cyclase from bovine lung by a new immunoaffinity chromatographic method. Eur. J. Biochem., 1990, 190(2), 273-278.
[http://dx.doi.org/10.1111/j.1432-1033.1990.tb15572.x] [PMID: 1973095]
[25]
Tomita, T.; Ogura, T.; Tsuyama, S.; Imai, Y.; Kitagawa, T. Effects of GTP on bound nitric oxide of soluble guanylate cyclase probed by resonance Raman spectroscopy. Biochemistry, 1997, 36(33), 10155-10160.
[http://dx.doi.org/10.1021/bi9710131] [PMID: 9254612]
[26]
Zhao, Y.; Marletta, M.A. Localization of the heme binding region in soluble guanylate cyclase. Biochemistry, 1997, 36(50), 15959-15964.
[http://dx.doi.org/10.1021/bi971825x] [PMID: 9398330]
[27]
Wedel, B.; Humbert, P.; Harteneck, C.; Foerster, J.; Malkewitz, J.; Böhme, E.; Schultz, G.; Koesling, D. Mutation of His-105 in the beta 1 subunit yields a nitric oxide-insensitive form of soluble guanylyl cyclase. Proc. Natl. Acad. Sci. USA, 1994, 91(7), 2592-2596.
[http://dx.doi.org/10.1073/pnas.91.7.2592] [PMID: 7908439]
[28]
Schmidt, P.M.; Schramm, M.; Schröder, H.; Wunder, F.; Stasch, J.P. Identification of residues crucially involved in the binding of the heme moiety of soluble guanylate cyclase. J. Biol. Chem., 2004, 279(4), 3025-3032.
[http://dx.doi.org/10.1074/jbc.M310141200] [PMID: 14570894]
[29]
Schmidt, P.M.; Rothkegel, C.; Wunder, F.; Schröder, H.; Stasch, J.P. Residues stabilizing the heme moiety of the nitric oxide sensor soluble guanylate cyclase. Eur. J. Pharmacol., 2005, 513(1-2), 67-74.
[http://dx.doi.org/10.1016/j.ejphar.2005.02.046] [PMID: 15878710]
[30]
Iyer, L.M.; Anantharaman, V.; Aravind, L. Ancient conserved domains shared by animal soluble guanylyl cyclases and bacterial signaling proteins. BMC Genomics, 2003, 4(1), 5.
[http://dx.doi.org/10.1186/1471-2164-4-5] [PMID: 12590654]
[31]
Karow, D.S.; Pan, D.; Tran, R.; Pellicena, P.; Presley, A.; Mathies, R.A.; Marletta, M.A. Spectroscopic characterization of the soluble guanylate cyclase-like heme domains from Vibrio cholerae and Thermoanaerobacter tengcongensis. Biochemistry, 2004, 43(31), 10203-10211.
[http://dx.doi.org/10.1021/bi049374l] [PMID: 15287748]
[32]
Boon, E.M.; Davis, J.H.; Tran, R.; Karow, D.S.; Huang, S.H.; Pan, D.; Miazgowicz, M.M.; Mathies, R.A.; Marletta, M.A. Nitric oxide binding to prokaryotic homologs of the soluble guanylate cyclase beta1 H-NOX domain. J. Biol. Chem., 2006, 281(31), 21892-21902.
[http://dx.doi.org/10.1074/jbc.M600557200] [PMID: 16728401]
[33]
Pellicena, P.; Karow, D.S.; Boon, E.M.; Marletta, M.A.; Kuriyan, J. Crystal structure of an oxygen-binding heme domain related to soluble guanylate cyclases. Proc. Natl. Acad. Sci. USA, 2004, 101(35), 12854-12859.
[http://dx.doi.org/10.1073/pnas.0405188101] [PMID: 15326296]
[34]
Antoniades, C.; Shirodaria, C.; Crabtree, M.; Rinze, R.; Alp, N.; Cunnington, C.; Diesch, J.; Tousoulis, D.; Stefanadis, C.; Leeson, P.; Ratnatunga, C.; Pillai, R.; Channon, K.M. Altered plasma versus vascular biopterins in human atherosclerosis reveal relationships between endothelial nitric oxide synthase coupling, endothelial function, and inflammation. Circulation, 2007, 116(24), 2851-2859.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.107.704155] [PMID: 18040031]
[35]
Ellulu, M.S.; Patimah, I.; Khaza’ai, H.; Rahmat, A.; Abed, Y.; Ali, F. Atherosclerotic cardiovascular disease: A review of initiators and protective factors. Inflammopharmacology, 2016, 24(1), 1-10.
[http://dx.doi.org/10.1007/s10787-015-0255-y] [PMID: 26750181]
[36]
Papapetropoulos, A.; Hobbs, A.J.; Topouzis, S. Extending the translational potential of targeting NO/cGMP-regulated pathways in the CVS. Br. J. Pharmacol., 2015, 172(6), 1397-1414.
[http://dx.doi.org/10.1111/bph.12980] [PMID: 25302549]
[37]
Stasch, J.P.; Pacher, P.; Evgenov, O.V. Soluble guanylate cyclase as an emerging therapeutic target in cardiopulmonary disease. Circulation, 2011, 123(20), 2263-2273.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.110.981738] [PMID: 21606405]
[38]
Follmann, M.; Griebenow, N.; Hahn, M.G.; Hartung, I.; Mais, F.J.; Mittendorf, J.; Schäfer, M.; Schirok, H.; Stasch, J.P.; Stoll, F.; Straub, A. The chemistry and biology of soluble guanylate cyclase stimulators and activators. Angew. Chem. Int. Ed. Engl., 2013, 52(36), 9442-9462.
[http://dx.doi.org/10.1002/anie.201302588] [PMID: 23963798]
[39]
Buechler, W.A.; Ivanova, K.; Wolfram, G.; Drummer, C.; Heim, J.M.; Gerzer, R. Soluble guanylyl cyclase and platelet function. Ann. N. Y. Acad. Sci., 1994, 714, 151-157.
[http://dx.doi.org/10.1111/j.1749-6632.1994.tb12039.x] [PMID: 7912486]
[40]
Denninger, J.W.; Marletta, M.A. Guanylate cyclase and the. NO/cGMP signaling pathway. Biochim. Biophys. Acta, 1999, 1411(2-3), 334-350.
[http://dx.doi.org/10.1016/S0005-2728(99)00024-9] [PMID: 10320667]
[41]
Warner, T.D.; Mitchell, J.A.; Sheng, H.; Murad, F. Effects of cyclic GMP on smooth muscle relaxation. Adv. Pharmacol., 1994, 26, 171-194.
[http://dx.doi.org/10.1016/S1054-3589(08)60054-X] [PMID: 7913615]
[42]
Behrends, S. Drugs that activate specific nitric oxide sensitive guanylyl cyclase isoforms independent of nitric oxide release. Curr. Med. Chem., 2003, 10(4), 291-301.
[http://dx.doi.org/10.2174/0929867033368286] [PMID: 12570702]
[43]
Evgenov, O.V.; Pacher, P.; Schmidt, P.M.; Haskó, G.; Schmidt, H.H.; Stasch, J.P. NO-independent stimulators and activators of soluble guanylate cyclase: Discovery and therapeutic potential. Nat. Rev. Drug Discov., 2006, 5(9), 755-768.
[http://dx.doi.org/10.1038/nrd2038] [PMID: 16955067]
[44]
Stasch, J.P.; Hobbs, A.J. NO-independent, haem-dependent soluble guanylate cyclase stimulators. Handb. Exp. Pharmacol., 2009, (191), 277-308.
[http://dx.doi.org/10.1007/978-3-540-68964-5_13] [PMID: 19089334]
[45]
Friebe, A.; Schultz, G.; Koesling, D. Sensitizing soluble guanylyl cyclase to become a highly CO-sensitive enzyme. EMBO J., 1996, 15(24), 6863-6868.
[http://dx.doi.org/10.1002/j.1460-2075.1996.tb01078.x] [PMID: 9003762]
[46]
Schmidt, H.H.; Schmidt, P.M.; Stasch, J.P. NO- and haem-independent soluble guanylate cyclase activators. Handb. Exp. Pharmacol., 2009, (191), 309-339.
[http://dx.doi.org/10.1007/978-3-540-68964-5_14] [PMID: 19089335]
[47]
Martin, F.; Baskaran, P.; Ma, X.; Dunten, P.W.; Schaefer, M.; Stasch, J.P.; Beuve, A.; van den Akker, F. Structure of cinaciguat (BAY 58-2667) bound to Nostoc H-NOX domain reveals insights into heme-mimetic activation of the soluble guanylyl cyclase. J. Biol. Chem., 2010, 285(29), 22651-22657.
[http://dx.doi.org/10.1074/jbc.M110.111559] [PMID: 20463019]
[48]
Evgenov, O.V.; Pacher, P.; Schmidt, P.M.; Haskó, G.; Schmidt, H.H.; Stasch, J-P. NO-independent stimulators and activators of soluble guanylate cyclase: discovery and therapeutic potential. Nat. Rev. Drug Discov., 2006, 5(9), 755-768.
[http://dx.doi.org/10.1038/nrd2038] [PMID: 16955067]
[49]
Sayed, N.; Kim, D.D.; Fioramonti, X.; Iwahashi, T.; Durán, W.N.; Beuve, A. Nitroglycerin-induced S-nitrosylation and desensitization of soluble guanylyl cyclase contribute to nitrate tolerance. Circ. Res., 2008, 103(6), 606-614.
[http://dx.doi.org/10.1161/CIRCRESAHA.108.175133] [PMID: 18669924]
[50]
Stasch, J-P.; Schmidt, P.M.; Nedvetsky, P.I.; Nedvetskaya, T.Y. H s, A.K.; Meurer, S.; Deile, M.; Taye, A.; Knorr, A.; Lapp, H.; Müller, H.; Turgay, Y.; Rothkegel, C.; Tersteegen, A.; Kemp-Harper, B.; Müller-Esterl, W.; Schmidt, H.H. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J. Clin. Invest., 2006, 116(9), 2552-2561.
[http://dx.doi.org/10.1172/JCI28371] [PMID: 16955146]
[51]
Stasch, J.; Hobbs, A.J. NO-independent, haem-dependent soluble guanylate cyclase stimulators. Handb. Exp. Pharmacol., 2009, 277-308.
[http://dx.doi.org/10.1007/978-3-540-68964-5_13]
[52]
Stasch, J.P.; Evgenov, O.V. Soluble guanylate cyclase stimulators in pulmonary hypertension. Pharmacother. Pulmon. Hyperten., 2013, 218, 279-313.
[http://dx.doi.org/10.1007/978-3-662-45805-1_12] [PMID: 24092345]
[53]
Ghofrani, H.A.; D’Armini, A.M.; Grimminger, F.; Hoeper, M.M.; Jansa, P.; Kim, N.H.; Mayer, E.; Simonneau, G.; Wilkins, M.R.; Fritsch, A.; Neuser, D.; Weimann, G.; Wang, C. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N. Engl. J. Med., 2013, 369(4), 319-329.
[http://dx.doi.org/10.1056/NEJMoa1209657] [PMID: 23883377]
[54]
Ghofrani, H.A.; Galiè, N.; Grimminger, F.; Grünig, E.; Humbert, M.; Jing, Z.C.; Keogh, A.M.; Langleben, D.; Kilama, M.O.; Fritsch, A.; Neuser, D.; Rubin, L.J. Riociguat for the treatment of pulmonary arterial hypertension. N. Engl. J. Med., 2013, 369(4), 330-340.
[http://dx.doi.org/10.1056/NEJMoa1209655] [PMID: 23883378]
[55]
Schermuly, R.T.; Stasch, J-P.; Pullamsetti, S.S.; Middendorff, R.; Müller, D.; Schlüter, K-D.; Dingendorf, A.; Hackemack, S.; Kolosionek, E.; Kaulen, C.; Dumitrascu, R.; Weissmann, N.; Mittendorf, J.; Klepetko, W.; Seeger, W.; Ghofrani, H.A.; Grimminger, F. Expression and function of soluble guanylate cyclase in pulmonary arterial hypertension. Eur. Respir. J., 2008, 32(4), 881-891.
[http://dx.doi.org/10.1183/09031936.00114407] [PMID: 18550612]
[56]
Stasch, J.P.; Pacher, P.; Evgenov, O.V. Soluble guanylate cyclase as an emerging therapeutic target in cardiopulmonary disease. Circulation, 2011, 123(20), 2263-2273.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.110.981738] [PMID: 21606405]
[57]
Lang, M.; Kojonazarov, B.; Tian, X.; Kalymbetov, A.; Weissmann, N.; Grimminger, F.; Kretschmer, A.; Stasch, J-P.; Seeger, W.; Ghofrani, H.A.; Schermuly, R.T. The soluble guanylate cyclase stimulator riociguat ameliorates pulmonary hypertension induced by hypoxia and SU5416 in rats. PLoS One, 2012, 7(8)e43433
[http://dx.doi.org/10.1371/journal.pone.0043433] [PMID: 22912874]
[58]
Halank, M.; Hoeper, M.M.; Ghofrani, H-A.; Meyer, F.J.; Stähler, G.; Behr, J.; Ewert, R.; Fletcher, M.; Colorado, P.; Nikkho, S.; Grimminger, F. Riociguat for pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension: Results from a phase II long-term extension study. Respir. Med., 2017, 128, 50-56.
[http://dx.doi.org/10.1016/j.rmed.2017.05.008] [PMID: 28610669]
[59]
Benza, R.; Mathai, S.; Nathan, S.D. sGC stimulators: Evidence for riociguat beyond groups 1 and 4 pulmonary hypertension. Respir. Med., 2017, 122(Suppl. 1), S28-S34.
[http://dx.doi.org/10.1016/j.rmed.2016.11.010] [PMID: 27890470]
[60]
Huntgeburth, M.; Kießling, J.; Weimann, G.; Kiepsel, V.; Saleh, S.; Hunzelmann, N.; Rosenkranz, S. The sGC-stimulator Riociguat for the treatment of Raynaud’s phenomenon: A single-dose, double-blind, randomized, placebo-controlled cross-over study (DIGIT). BMC Pharmacol. Toxicol., 2015, 16(1), A58.
[http://dx.doi.org/10.1186/2050-6511-16-S1-A58]
[61]
Follmann, M.; Ackerstaff, J.; Redlich, G.; Wunder, F.; Lang, D.; Kern, A.; Fey, P.; Griebenow, N.; Kroh, W.; Becker-Pelster, E.M.; Kretschmer, A.; Geiss, V.; Li, V.; Straub, A.; Mittendorf, J.; Jautelat, R.; Schirok, H.; Schlemmer, K.H.; Lustig, K.; Gerisch, M.; Knorr, A.; Tinel, H.; Mondritzki, T.; Trübel, H.; Sandner, P.; Stasch, J.P. Discovery of the soluble guanylate cyclase stimulator vericiguat (bay 1021189) for the treatment of chronic heart failure. J. Med. Chem., 2017, 60(12), 5146-5161.
[http://dx.doi.org/10.1021/acs.jmedchem.7b00449] [PMID: 28557445]
[62]
Pieske, B.; Maggioni, A.P.; Lam, C.S.P.; Pieske-Kraigher, E.; Filippatos, G.; Butler, J.; Ponikowski, P.; Shah, S.J.; Solomon, S.D.; Scalise, A.V.; Mueller, K.; Roessig, L.; Gheorghiade, M. Vericiguat in patients with worsening chronic heart failure and preserved ejection fraction: Results of the soluble guanylate cyclase stimulator in heart failure patients with preserved EF (socrates-preserved) study. Eur. Heart J., 2017, 38(15), 1119-1127.
[http://dx.doi.org/10.1093/eurheartj/ehw593] [PMID: 28369340]
[63]
Filippatos, G.; Maggioni, A.P.; Lam, C.S.P.; Pieske-Kraigher, E.; Butler, J.; Spertus, J.; Ponikowski, P.; Shah, S.J.; Solomon, S.D.; Scalise, A.V.; Mueller, K.; Roessig, L.; Bamber, L.; Gheorghiade, M.; Pieske, B. Patient-reported outcomes in the soluble guanylate cyclase stimulator in heart failure patients with preserved ejection fraction (socrates-preserved) study. Eur. J. Heart Fail., 2017, 19(6), 782-791.
[http://dx.doi.org/10.1002/ejhf.800] [PMID: 28586537]
[64]
Gheorghiade, M.; Greene, S.J.; Butler, J.; Filippatos, G.; Lam, C.S.; Maggioni, A.P.; Ponikowski, P.; Shah, S.J.; Solomon, S.D.; Kraigher-Krainer, E.; Samano, E.T.; Müller, K.; Roessig, L.; Pieske, B. Effect of vericiguat, a soluble guanylate cyclase stimulator, on natriuretic peptide levels in patients with worsening chronic heart failure and reduced ejection fraction: The socrates-reduced randomized trial. JAMA, 2015, 314(21), 2251-2262.
[http://dx.doi.org/10.1001/jama.2015.15734] [PMID: 26547357]
[65]
Pieske, B.; Butler, J.; Filippatos, G.; Lam, C.; Maggioni, A.P.; Ponikowski, P.; Shah, S.; Solomon, S.; Kraigher-Krainer, E.; Samano, E.T.; Scalise, A.V.; Müller, K.; Roessig, L.; Gheorghiade, M. Rationale and design of the soluble guanylate cyclase stimulator in heart failure studies (socrates). Eur. J. Heart Fail., 2014, 16(9), 1026-1038.
[http://dx.doi.org/10.1002/ejhf.135] [PMID: 25056511]
[66]
Armstrong, P.W.; Roessig, L.; Patel, M.J.; Anstrom, K.J.; Butler, J.; Voors, A.A.; Lam, C.S.; Ponikowski, P.; Temple, T.; Pieske, B.; Multicenter, A. Randomized, double-blind, placebo-controlled trial of the efficacy and safety of the oral soluble guanylate cyclase stimulator: The victoria trial. JACC Heart Fail., 2017.
[http://dx.doi.org/ 10.1016/j.jchf.2017.08.013] [PMID: 29032136]
[67]
Gheorghiade, M.; Greene, S.J.; Filippatos, G.; Erdmann, E.; Ferrari, R.; Levy, P.D.; Maggioni, A.; Nowack, C.; Mebazaa, A. Cinaciguat, a soluble guanylate cyclase activator: Results from the randomized, controlled, phase IIb COMPOSE programme in acute heart failure syndromes. Eur. J. Heart Fail., 2012, 14(9), 1056-1066.
[http://dx.doi.org/10.1093/eurjhf/hfs093] [PMID: 22713287]
[68]
Erdmann, E.; Semigran, M.J.; Nieminen, M.S.; Gheorghiade, M.; Agrawal, R.; Mitrovic, V.; Mebazaa, A. Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute decompensated heart failure. Eur. Heart J., 2013, 34(1), 57-67.
[http://dx.doi.org/10.1093/eurheartj/ehs196] [PMID: 22778174]
[69]
Stasch, J.P.; Schlossmann, J.; Hocher, B. Renal effects of soluble guanylate cyclase stimulators and activators: A review of the preclinical evidence. Curr. Opin. Pharmacol., 2015, 21, 95-104.
[http://dx.doi.org/10.1016/j.coph.2014.12.014] [PMID: 25645316]
[70]
Kumar, V.; Martin, F.; Hahn, M.G.; Schaefer, M.; Stamler, J.S.; Stasch, J.P.; van den Akker, F. Insights into BAY 60-2770 activation and S-nitrosylation-dependent desensitization of soluble guanylyl cyclase via crystal structures of homologous nostoc H-NOX domain complexes. Biochemistry, 2013, 52(20), 3601-3608.
[http://dx.doi.org/10.1021/bi301657w] [PMID: 23614626]
[71]
Rekowski, M.V.W.; Kumar, V.; Zhou, Z.; Moschner, J.; Marazioti, A.; Bantzi, M.; Spyroulias, G.A.; van den Akker, F.; Giannis, A.; Papapetropoulos, A. Insights into soluble guanylyl cyclase activation derived from improved heme-mimetics. J. Med. Chem., 2013, 56(21), 8948-8952.
[http://dx.doi.org/10.1021/jm400539d] [PMID: 24090476]
[72]
Martin, F.; Baskaran, P.; Ma, X.; Dunten, P.W.; Schaefer, M.; Stasch, J.P.; Beuve, A.; van den Akker, F. Structure of cinaciguat (BAY 58-2667) bound to Nostoc H-NOX domain reveals insights into heme-mimetic activation of the soluble guanylyl cyclase. J. Biol. Chem., 2010, 285(29), 22651-22657.
[http://dx.doi.org/10.1074/jbc.M110.111559] [PMID: 20463019]
[73]
Ma, X. Sayed, N.; Beuve, A.; van den Akker, F. NO and CO differentially activate soluble guanylyl cyclase via a heme pivot-bend mechanism. EMBO J., 2007, 26(2), 578-588.
[http://dx.doi.org/10.1038/sj.emboj.7601521] [PMID: 17215864]
[74]
Hespen, C.W.; Bruegger, J.J.; Phillips-Piro, C.M.; Marletta, M.A. Bruegger, J. J.; Phillips-Piro, C. M.; Marletta, M. A., Structural and functional evidence indicates selective oxygen signaling in Caldanaerobacter subterraneus H-NOX. ACS Chem. Biol., 2016, 11(8), 2337-2346.
[http://dx.doi.org/10.1021/acschembio.6b00431] [PMID: 27328180]
[75]
Herzik, M.A., Jr; Jonnalagadda, R.; Kuriyan, J.; Marletta, M.A. Structural insights into the role of iron-histidine bond cleavage in nitric oxide-induced activation of H-NOX gas sensor proteins. Proc. Natl. Acad. Sci. USA, 2014, 111(40), E4156-E4164.
[http://dx.doi.org/10.1073/pnas.1416936111] [PMID: 25253889]
[76]
Alexandropoulos, I.I.; Argyriou, A.I.; Marousis, K.D.; Topouzis, S.; Papapetropoulos, A.; Spyroulias, G.A. (1)H, (13)C, (15)N backbone and side-chain resonance assignment of Nostoc sp. C139A variant of the heme-nitric oxide/oxygen binding (H-NOX) domain. Biomol. NMR Assign., 2016, 10(2), 395-400.
[http://dx.doi.org/10.1007/s12104-016-9707-6] [PMID: 27614467]
[77]
Mülsch, A.; Bauersachs, J.; Schäfer, A.; Stasch, J.P.; Kast, R.; Busse, R. Effect of YC-1, an NO-independent, superoxide-sensitive stimulator of soluble guanylyl cyclase, on smooth muscle responsiveness to nitrovasodilators. Br. J. Pharmacol., 1997, 120(4), 681-689.
[http://dx.doi.org/10.1038/sj.bjp.0700982] [PMID: 9051308]
[78]
Stasch, J.P.; Becker, E.M.; Alonso-Alija, C.; Apeler, H.; Dembowsky, K.; Feurer, A.; Gerzer, R.; Minuth, T.; Perzborn, E.; Pleiss, U.; Schröder, H.; Schroeder, W.; Stahl, E.; Steinke, W.; Straub, A.; Schramm, M. NO-independent regulatory site on soluble guanylate cyclase. Nature, 2001, 410(6825), 212-215.
[http://dx.doi.org/10.1038/35065611] [PMID: 11242081]
[79]
Ko, F.N.; Wu, C.C.; Kuo, S.C.; Lee, F.Y.; Teng, C.M. YC-1, a novel activator of platelet guanylate cyclase. Blood, 1994, 84(12), 4226-4233.
[PMID: 7527671]
[80]
Straub, A.; Stasch, J.P.; Alonso-Alija, C.; Benet-Buchholz, J.; Ducke, B.; Feurer, A.; Fürstner, C. NO-independent stimulators of soluble guanylate cyclase. Bioorg. Med. Chem. Lett., 2001, 11(6), 781-784.
[http://dx.doi.org/10.1016/S0960-894X(01)00073-7] [PMID: 11277519]
[81]
Mittendorf, J.; Weigand, S.; Alonso-Alija, C.; Bischoff, E.; Feurer, A.; Gerisch, M.; Kern, A.; Knorr, A.; Lang, D.; Muenter, K.; Radtke, M.; Schirok, H.; Schlemmer, K.H.; Stahl, E.; Straub, A.; Wunder, F.; Stasch, J.P. Discovery of riociguat (BAY 63-2521): A potent, oral stimulator of soluble guanylate cyclase for the treatment of pulmonary hypertension. ChemMedChem, 2009, 4(5), 853-865.
[http://dx.doi.org/10.1002/cmdc.200900014] [PMID: 19263460]
[82]
Frey, R.; Mück, W.; Unger, S.; Artmeier-Brandt, U.; Weimann, G.; Wensing, G. Single-dose pharmacokinetics, pharmacodynamics, tolerability, and safety of the soluble guanylate cyclase stimulator BAY 63-2521: An ascending-dose study in healthy male volunteers. J. Clin. Pharmacol., 2008, 48(8), 926-934.
[http://dx.doi.org/10.1177/0091270008319793] [PMID: 18519919]
[83]
Shah, P.; Westwell, A.D. The role of fluorine in medicinal chemistry. J. Enzyme Inhib. Med. Chem., 2007, 22(5), 527-540.
[http://dx.doi.org/10.1080/14756360701425014] [PMID: 18035820]
[84]
Selwood, D.L.; Brummell, D.G.; Budworth, J.; Burtin, G.E.; Campbell, R.O.; Chana, S.S.; Charles, I.G.; Fernandez, P.A.; Glen, R.C.; Goggin, M.C.; Hobbs, A.J.; Kling, M.R.; Liu, Q.; Madge, D.J.; Meillerais, S.; Powell, K.L.; Reynolds, K.; Spacey, G.D.; Stables, J.N.; Tatlock, M.A.; Wheeler, K.A.; Wishart, G.; Woo, C.K. Synthesis and biological evaluation of novel pyrazoles and indazoles as activators of the nitric oxide receptor, soluble guanylate cyclase. J. Med. Chem., 2001, 44(1), 78-93.
[http://dx.doi.org/10.1021/jm001034k] [PMID: 11141091]
[85]
Nakai, T.; Perl, N.R.; Barden, T.C.; Carvalho, A.; Fretzen, A.; Germano, P. Im, G.Y.; Jin, H.; Kim, C.; Lee, T.W.; Long, K.; Moore, J.; Rohde, J.M.; Sarno, R.; Segal, C.; Solberg, E.O.; Tobin, J.; Zimmer, D.P.; Currie, M.G. Discovery of IWP-051, a novel orally bioavailable sGC stimulator with once-daily dosing potential in humans. ACS Med. Chem. Lett., 2016, 7(5), 465-469.
[http://dx.doi.org/10.1021/acsmedchemlett.5b00479] [PMID: 27190594]
[86]
Nakane, M.; Kolasa, T.; Chang, R.; Miller, L.N.; Moreland, R.B.; Brioni, J.D. Acrylamide analog as a novel nitric oxide-independent soluble guanylyl cyclase activator. J. Pharmacol. Sci., 2006, 102(2), 231-238.
[http://dx.doi.org/10.1254/jphs.FPJ06017X] [PMID: 17050951]
[87]
Lamothe, M.; Chang, F-J.; Balashova, N.; Shirokov, R.; Beuve, A. Functional characterization of nitric oxide and YC-1 activation of soluble guanylyl cyclase: Structural implication for the YC-1 binding site? Biochemistry, 2004, 43(11), 3039-3048.
[http://dx.doi.org/10.1021/bi0360051] [PMID: 15023055]
[88]
Yazawa, S.; Tsuchiya, H.; Hori, H.; Makino, R. Functional characterization of two nucleotide-binding sites in soluble guanylate cyclase. J. Biol. Chem., 2006, 281(31), 21763-21770.
[http://dx.doi.org/10.1074/jbc.M508983200] [PMID: 16754683]
[89]
Hu, X.; Murata, L.B.; Weichsel, A.; Brailey, J.L.; Roberts, S.A.; Nighorn, A.; Montfort, W.R. Allostery in recombinant soluble guanylyl cyclase from Manduca sexta. J. Biol. Chem., 2008, 283(30), 20968-20977.
[http://dx.doi.org/10.1074/jbc.M801501200] [PMID: 18515359]
[90]
Ibrahim, M.; Derbyshire, E.R.; Marletta, M.A.; Spiro, T.G. Probing soluble guanylate cyclase activation by CO and YC-1 using resonance Raman spectroscopy. Biochemistry, 2010, 49(18), 3815-3823.
[http://dx.doi.org/10.1021/bi902214j] [PMID: 20353168]
[91]
Ibrahim, M.; Derbyshire, E.R.; Soldatova, A.V.; Marletta, M.A.; Spiro, T.G. Soluble guanylate cyclase is activated differently by excess NO and by YC-1: Resonance raman spectroscopic evidence. Biochemistry, 2010, 49(23), 4864-4871.
[http://dx.doi.org/10.1021/bi100506j] [PMID: 20459051]
[92]
Martin, E.; Czarnecki, K.; Jayaraman, V.; Murad, F.; Kincaid, J. Resonance raman and infrared spectroscopic studies of high-output forms of human soluble guanylyl cyclase. J. Am. Chem. Soc., 2005, 127(13), 4625-4631.
[http://dx.doi.org/10.1021/ja0440912] [PMID: 15796527]
[93]
Yoo, B.K.; Lamarre, I.; Rappaport, F.; Nioche, P.; Raman, C.S.; Martin, J.L.; Negrerie, M. Picosecond to second dynamics reveals a structural transition in Clostridium botulinum NO-sensor triggered by the activator BAY-41-2272. ACS Chem. Biol., 2012, 7(12), 2046-2054.
[http://dx.doi.org/10.1021/cb3003539] [PMID: 23009307]
[94]
Stasch, J.P.; Schmidt, P.; Alonso-Alija, C.; Apeler, H.; Dembowsky, K.; Haerter, M.; Heil, M.; Minuth, T.; Perzborn, E.; Pleiss, U.; Schramm, M.; Schroeder, W.; Schröder, H.; Stahl, E.; Steinke, W.; Wunder, F. NO- and haem-independent activation of soluble guanylyl cyclase: Molecular basis and cardiovascular implications of a new pharmacological principle. Br. J. Pharmacol., 2002, 136(5), 773-783.
[http://dx.doi.org/10.1038/sj.bjp.0704778] [PMID: 12086987]
[95]
Schmidt, P.; Schramm, M.; Schröder, H.; Stasch, J.P. Mechanisms of nitric oxide independent activation of soluble guanylyl cyclase. Eur. J. Pharmacol., 2003, 468(3), 167-174.
[http://dx.doi.org/10.1016/S0014-2999(03)01674-1] [PMID: 12754054]
[96]
Sharina, I.G.; Sobolevsky, M.; Papakyriakou, A.; Rukoyatkina, N.; Spyroulias, G.A.; Gambaryan, S.; Martin, E. The fibrate gemfibrozil is a NO- and haem-independent activator of soluble guanylyl cyclase: in vitro studies. Br. J. Pharmacol., 2015, 172(9), 2316-2329.
[http://dx.doi.org/10.1111/bph.13055] [PMID: 25536881]
[97]
Sömmer, A.; Sandner, P.; Behrends, S. BAY 60-2770 activates two isoforms of nitric oxide sensitive guanylyl cyclase: Evidence for stable insertion of activator drugs. Biochem. Pharmacol., 2017.
[PMID: 29155144]
[98]
Kollau, A.; Opelt, M.; Wölkart, G.; Gorren, A.C.; Russwurm, M.; Koesling, D.; Mayer, B.; Schrammel, A. Irreversible activation and stabilization of soluble guanylate cyclase by the protoporphyrin IX mimetic cinaciguat. Mol. Pharmacol., 2018, 93(2), 73-78.
[http://dx.doi.org/10.1124/mol.117.109918] [PMID: 29138269]
[99]
Winter, M.B.; Herzik, M.A., Jr; Kuriyan, J.; Marletta, M.A. Tunnels modulate ligand flux in a heme nitric oxide/oxygen binding (H-NOX) domain. Proc. Natl. Acad. Sci. USA, 2011, 108(43), E881-E889.
[http://dx.doi.org/10.1073/pnas.1114038108] [PMID: 21997213]
[100]
Weinert, E.E.; Phillips-Piro, C.M.; Tran, R.; Mathies, R.A.; Marletta, M.A. Controlling conformational flexibility of an O2-binding H-NOX domain. Biochemistry, 2011, 50(32), 6832-6840.
[http://dx.doi.org/10.1021/bi200788x] [PMID: 21721586]
[101]
Weinert, E.E.; Plate, L.; Whited, C.A.; Olea, C., Jr; Marletta, M.A. Determinants of ligand affinity and heme reactivity in H-NOX domains. Angew. Chem. Int. Ed. Engl., 2010, 49(4), 720-723.
[http://dx.doi.org/10.1002/anie.200904799] [PMID: 20017169]
[102]
Olea, C., Jr; Boon, E.M.; Pellicena, P.; Kuriyan, J.; Marletta, M.A. Probing the function of heme distortion in the H-NOX family. ACS Chem. Biol., 2008, 3(11), 703-710.
[http://dx.doi.org/10.1021/cb800185h] [PMID: 19032091]
[103]
Erbil, W.K.; Price, M.S.; Wemmer, D.E.; Marletta, M.A. A structural basis for H-NOX signaling in Shewanella oneidensis by trapping a histidine kinase inhibitory conformation. Proc. Natl. Acad. Sci. USA, 2009, 106(47), 19753-19760.
[http://dx.doi.org/10.1073/pnas.0911645106] [PMID: 19918063]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy