Индуцибельная NO-синтаза как фармакологическая мишень противовоспалительной терапии: надежда не потеряна?

Автор: М.М. Галагудза, Ю.П. Бельский, Н.В. Бельская

Журнал: Сибирский журнал клинической и экспериментальной медицины @cardiotomsk

Рубрика: Обзоры и лекции

Статья в выпуске: 1 т.38, 2023 года.

Бесплатный доступ

Обзор посвящен текущему пониманию роли оксида азота (NO) и индуцибельной NO-синтазы в физиологических условиях и при некоторых патологических состояниях. Рассмотрены механизмы индукции экспрессии гена индуцибельной NO-синтазы и посттранскрипционной регуляции активности индуцибельной NO-синтазы, приведены сведения об эндогенных ингибиторах индуцибельной NO-синтазы. Проведен анализ статуса клинических исследований, направленных на изучение клинической эффективности ингибиторов NO-синтаз.

Оксид азота, ингибитор индуцибельной синтазы оксида азота, доклинические исследования, клинические испытания

Короткий адрес: https://sciup.org/149141576

IDR: 149141576   |   DOI: 10.29001/2073-8552-2023-38-1-13-20

Список литературы Индуцибельная NO-синтаза как фармакологическая мишень противовоспалительной терапии: надежда не потеряна?

  • Cinelli M.A., Do H.T., Miley G.P., Silverman R.B. Inducible nitric oxide synthase: regulation, structure, and inhibition. Med. Res. Rev. 2020;40(1):158–189. DOI: 10.1002/med.21599.
  • Ahmad N., Ansari M.Y., Haqqi T.M. Role of iNOS in osteoarthritis: Pathological and therapeutic aspects. J. Cell. Physiol. 2020;235:6366–6376. DOI: 10.1002/jcp.29607.
  • Ferreiro C.R., Chagas A.C.P., Carvalho M.H.C., Dantas A.P., Jatene M.B., Bento De Souza L.C. et al. Infl uence of hypoxia on nitric oxide synthase activity and gene expression in children with congenital heart disease: a novel pathophysiological adaptive mechanism. Circulation. 2001;103(18):2272–2276. DOI: 10.1161/01.cir.103.18.2272.
  • Navasardyan I., Bonavida B. Regulation of T cells in cancer by nitric oxide. Cells. 2021;10:2655. DOI: 10.3390/cells10102655.
  • Бельский Ю.П., Бельская Н.В., Данилец М.Г., Трофимова Е.С., Патрушев В.К., Агафонов В.И. Клетки опухоли Эрлиха стимулируют продукцию интерферона-γ Т-клетками и не чувствительны к аутокринному оксиду азота. Вопросы онкологии. 2004;50(6):689–692. [Bel’skiĭ Iu.P., Bel’skaia N.V., Danilets M.G., Trofi mova E.S., Patrushev V.K., Agafonov V.I. Ehrlich tumor cells stimulate T-cell production of interferon-gamma and are resistant to autocrine nitric oxide. Vopr. Oncol. 2004;50(6):689–692. (In Russ.)].
  • Pautz A., Art J., Hahn S., Nowag S., Voss C., Kleinert H. Regulation of the expression of inducible nitric oxide synthase. Nitric Oxide. 2010;23(2):75–93. DOI: 10.1016/j.niox.2010.04.007.
  • Anavia S., Tirosha O. iNOS as a metabolic enzyme under stress conditions. Free Radic. Biol. Med. 2020;146:16–35. DOI: 10.1016/j.freeradbiomed.2019.10.411.
  • Lirk P., Hoff mann G., Rieder J. Inducible nitric oxide synthase-time for reappraisal. Curr. Drug Targets Infl amm. Allergy. 2002;l(1):89–108. DOI: 10.2174/1568010023344913.
  • Данилец М.Г., Бельский Ю.П., Бельская Н.В., Трофимова Е.С., Учасова Е.Г., Агафонов В.И. Экспрессия аргиназы перитонеальными макрофагами и продукция ими оксида азота при Th1- и Th2-зависимом иммунном ответе. Бюлл. эксперим. биол. и медицины. 2007;S1:97–100. [Danilets M.G., Bel’skiĭ Iu.P., Bel’skaia N.V., Trofi mova E.S., Patrushev V.K., Agafonov V.I. Arginase expression by peritoneal macrophages and their production of nitric oxide in Th1- and Th2-dependent immune responses. Bull. Exp. Biol. Med. 2007;S1:97–100. (In Russ.)].
  • Nagy G., Koncz A., Telarico T., Fernandez D., Ersek B., Buzás E. et al. Central role of nitric oxide in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus. Arthritis Res. Ther. 2010;12(3):210. DOI: 10.1186/ar3045.
  • Burggraaf S., Bingham J., Payne J., Kimpton W.G., Lowenthal J.W., Bean A.G. Increased inducible nitric oxide synthase expression in organs is associated with a higher severity of H5N1 infl uenza virus infection. PLoS One. 2011;6(1):e14561. DOI: 10.1371/journal.pone.0014561.
  • Almeida-Souza F., Souza C., Taniwaki N., Silva J., Oliveira R., Abreu-Silva A.L. et al. Morinda citrifolia Linn. fruit (Noni) juice induces an increase in NO production and death of Leishmania amazonensis amastigotes in peritoneal macrophages from BALB/c. Nitric Oxide 2016;58:51–58. DOI: 10.1016/j.niox.2016.06.004.
  • Sharma J.N., Al-Omran A., Parvathy S.S. Role of nitric oxide in infl amatory diseases. Infl ammopharmacology. 2007;15(6):252–259. DOI: 10.1007/s10787-007-0013-x.
  • Thomas D.D., Ridnour L.A., Isenberg J.S., Flores-Santana W., Switzer C.H., Donzelli S. et al. The chemical biology of nitric oxide: Implications in cellular signaling. Free Radic. Biol. Med. 2008;45(1):18–31. DOI: 10.1016/j.freeradbiomed.2008.03.020.
  • Szabo C. Gasotransmitters in cancer: From pathophysiology to experimental therapy. Nat. Rev. Drug Discov. 2016;15(3):185–203. DOI: 10.1038/nrd.2015.1.
  • Kawasaki K., Smith R.S.Jr., Hsieh C.M., Sun J., Chao J., Liao J.K. Activation of the phosphatidylinositol 3-kinase/protein kinase Akt pathway mediates nitric oxide-induced endothelial cell migration and angiogenesis. Mol. Cell. Biol. 2003;23:5726–5737. DOI: 10.1128/MCB.23.16.5726-5737.2003.
  • Olson N., van der Vliet A. Interactions between nitric oxide and hypoxia-inducible factor signaling pathways in infl ammatory disease. Nitric Oxide. 2011;25(2):125–137. DOI: 10.1016/j.niox.2010.12.010.
  • Ambs S., Merriam W.G., Bennett W.P., Felley-Bosco E., Ogunfusika M.O., Oser S.M. et al. Frequent nitric oxide synthase-2 expression in human colon adenomas: Implication for tumor angiogenesis and colon cancer progression. Cancer Res. 1998;58(2):334–341.
  • Lee S.W., Choi H., Eun S.Y., Fukuyama S., Croft M. Nitric oxide modulates TGF-beta-directive signals to suppress Foxp3+ regulatory T cell differentiation and potentiate Th1 development. J. Immunol.2011;186(12):6972–6980. DOI: 10.4049/jimmunol.1100485.
  • Cinier J., Hubert M., Besson L., Di Roio A., Rodriguez C., Lombardi V. et al. Recruitment and expansion of Tregs cells in the tumor environment-how to target them? Cancers. 2021;13(8):1850. DOI: 10.3390/cancers13081850.
  • Schietinger A., Greenberg P.D. Tolerance and exhaustion: Defining mechanisms of T cell dysfunction. Trends Immunol. 2013;35(2):51–60. DOI: 10.1016/j.it.2013.10.001.
  • Jiang W., He Y., He W., Wu G., Zhou X., Sheng Q. et al. Exhausted CD8+Tcells in the tumor immune microenvironment: new pathways to therapy. Front. Immunol. 2021;11:622509. DOI: 10.3389/fimmu.2020.622509. 11.
  • Rodriguez P.C., Quiceno D.G., Zabaleta J., Ortiz B., Zea A.H., Piazuelo M.B. et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Canc. Res. 2004;64(16):5839–5849. DOI: 10.1158/0008-5472.CAN-04-0465.
  • Parker K.H., Beury D.W., Ostrand-Rosenberg S. Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv. Cancer. Res. 2015;128:95–139. DOI: 10.1016/bs.acr.2015.04.002.
  • Girotti A.W., Fahey J.F., Korytowski W. Role of nitric oxide in hyper-aggressiveness of tumor cells that survive various anti-cancer therapies. Crit. Rev. Oncol. Hematol. 2022;179:103805. DOI: 10.1016/j.critrevonc.2022.103805.
  • Khan F.H., Dervan E., Bhattacharyya D.D., McAuliffe J.D., Miranda K.M., Glynn S.A. The role of nitric oxide in cancer: master regulator of NoT? Int. J. Mol. Sci. 2020;21(24):9393. DOI: 10.3390/ijms21249393.
  • Vannini F., Kashfi K., Nath N. The dual role of iNOS in cancer. Redox Biol. 2015;6:334–343. DOI: 10.1016/j.redox.2015.08.009.
  • Somasundaram V., Ridnour L.A., Cheng R.Y., Walke A.J., Kedei N., Bhattacharyya D.D. et al. Systemic NOS2 depletion and COX inhibition limits TNBC disease progression and alters lymphoid cell spatial orientation and density. Redox Biol. 2022;58:102529. DOI: 10.1016/j.redox.2022.102529.
  • Miyoshi T., Li Y., Shih D.M., Wang X., Laubach V.E., Matsumoto A.H. et al. Deficiency of inducible NO synthase reduces advanced but not early atherosclerosis in apolipoprotein E-deficient mice. Life. Sci. 2006;79(6):525–531. DOI: 10.1016/j.lfs.2006.01.043.
  • Mercanoglu G., Safran N., Ahishali B.B., Uzun H., Yalcin A., Mercanoglu F. Nitric oxide mediated effects of nebivolol in myocardial infarction: the source of nitric oxide. Eur. Rev. Med. Pharmacol. Sci. 2015;19(24):4872–4889.
  • El-Awady M.S., Suddek G.M. Agmatine ameliorates atherosclerosis progression and endothelial dysfunction in high cholesterol-fed rabbits. J. Pharm. Pharmacol. 2014;66(6):835–843. DOI: 10.1111/jphp.12204.
  • Chauhan S.D., Seggara G., Vo P.A., Macallister R.J., Hobbs A.J., Ahluwalia A. Protection against lipopolysaccharide-induced endothelial dysfunction in resistance and conduit vasculature of iNOS knockout mice. FASEB. J. 2003;17(6):773–775. DOI: 10.1096/fj.02-0668fje.
  • Guo Y., Jones W.K., Xuan Y.T., Tang X.L., Bao W., Wu W.J. et al. The late phase of ischemic preconditioning is abrogated by targeted disruption of the inducible NO synthase gene. Proc. Natl. Acad. Sci. 1999;96(20):11507–11512. DOI: 10.1073/pnas.96.20.11507.
  • Mungrue I.N., Gros R., You X., Pirani A., Azad A., Csont T. et al. Cardiomyocyte overexpression of iNOS in mice results in peroxynutrite generation, heart block, and sudden death. J. Clin. Invest. 2002;109(6):735–743. DOI: 10.1172/JCI13265.
  • Lind M., Hayesa A., Caprndab M., Petrovicc D., Rodrigod L., Kruzliake P. et al. Inducible nitric oxide synthase: Good or bad? Biomed. Pharmacother. 2017;93:370–375. DOI: 10.1016/j.biopha.2017.06.036.
  • Oliveira-Paula G.H., Lacchini R., Tanus-Santos J.E. Inducible nitric oxide synthase as a possible target in hypertension. Curr. Drug Targets. 2014;15(2):164–174. DOI: 10.2174/13894501113146660227.
  • Результаты поиска по запросу: публикации, посвященные ингибиторам NOS за период с 1991 по январь 2023 гг. (по данным сайта PubMed). [Search results on demand: publications on NOS inhibitors from 1991 to January 2023 (accoding to PubMed)]. URL: https://pubmed.ncbi.nlm.nih.gov/?term=%28%28Nitric+oxide+synthase+inhibitor%5BTitle%5D%29+OR+%28Nitric+oxide+synthase+inhibitors%5BTitle%5D%29%29+AND+%28%28%221900%2F01%2F01%22%5BDate+-+Publication%5D+%3A+%223000%22%5BDate+-+Publication%5D%29%29&sort=&fil er=pubt.review&fil er=pubt.review (02.03.2023).
  • Yang Y., Yu T., Lian Y.J., Ma R., Yang S., Cho J.Y. Nitric oxide synthase inhibitors: a review of patents from 2011 to the present. Expert Opin. Ther. Pat. 2015;25(1):49–68. DOI: 10.1517/13543776.2014.979154.
  • Minhas R., Bansal Y., Bansal G. Inducible nitric oxide synthase inhibitors: A comprehensive update. Med. Res. Rev. 2020;40:823–855. DOI: 10.1002/med.21636.
  • Sorrells D.L., Friend C., Koltuksuz U., Courcoulas A., Boyle P., Garrett M. et al. Inhibition of nitric oxide with aminoguanidine reduces bacterial translocation after endotoxin challenge in vivo. Arch. Surg. 1996;131(11):1155–1163. DOI: 10.1001/archsurg.1996.01430230037007.
  • Nilsson B., Delbro D., Hedin L., Conradi N., Thune A., Friman S. et al. Role of nitric oxide in induction of inflammatory fluid secretion by the mucosa of the feline gallbladder. Gastroenterology. 1996;110(2):598–606. DOI: 10.1053/gast.1996.v110.pm8566609.
  • Lu G., Su R.B., Li J., Qin B.Y. Modulation by alpha-difluoromethyl-orn - thine and aminoguanidine of pain threshold, morphine analgesia and tolerance. Eur. J. Pharmacol. 2003;478(2–3):139–144. DOI: 10.1016/j.ejphar.2003.08.048.
  • Abdel-Zahera A.O., Hamdya M.M., Alya S.A., Abdel-Hadyb R.H., Abdel-Rahmanc S. Attenuation of morphinetolerance and dependence by aminoguanidine in mice. Europ. J. Pharmacol. 2006;540(1–3):60–66. DOI: 10.1016/j.ejphar.2006.03.059.
  • Brownlee M., Vlassara H., Kooney A., Ulrich P., Cerami A. Aminoguanidine prevents diabetes-induced arterial wall protein cross-linking. Science. 1986;232(4758):1629–1632. DOI: 10.1126/science.3487117.
  • Haddad E.K., Duclos A.J., Baines M.G. Early embryo loss is associated with local production of nitric oxide by decidual mononuclear cells. J. Exp. Med. 1995;182(4):1143–1151. DOI: 10.1084/jem.182.4.1143.
  • Worrall N.K., Lazenby W.D., Misko T.P., Lin T.S., Rodi C.P., Manning P.T. et al. Modulation of in vivo alloreactivity by inhibition of inducible nitric oxide synthase. J. Exp. Med. 1995;181(1):63–70. DOI: 10.1084/jem.181.1.63.
  • Kihara M., Schmelzer J.D., Poduslo J.F., Curran G.L., Nickander K.K., Low P.A. Aminoguanidine effects on nerve blood flo , vascular permeability, electrophysiology, and oxygen free radicals. Proc. Natl. Acad. Sci. USA. 1991;88(14):6107–6111. DOI: 10.1073/pnas.88.14.6107.
  • Onorato J.M., Jenkins A.J., Thorpe S.R., Baynes J.W. Pyridoxamine, an inhibitor of advanced glycation reactions, also inhibits advanced lipoxidation reactions. Mechanism of action of pyridoxamine. J. Biol. Chem. 2000;275(28):21177–21184. DOI: 10.1074/jbc.M003263200.
  • Viberti G., Slama G., Pozza G., Czyzyk A., Bilous R.W., Gries A. et al. Early closure of European Pimagedine trial. Steering Committee. Safety Committee. The Lancet. 1997;350(9072):214–215. DOI: 10.1016/S0140-6736(97)26029-0.
  • A curious stopping rule from Hoechst Marion Roussel (Editorial). The Lancet. 1997;350(9072):155. DOI: 10.1016/S0140-6736(97)21029-9.
  • Chung A.W., Anand K., Anselme A.C., Chan A.A., Gupta N., Venta L.A. et.al. A phase 1/2 clinical trial of the nitric oxide synthase inhibitor L-NMMA and taxane for treating chemoresistant triple-negative breast cancer. Sci. Transl. Med. 2021;13(624):eabj5070. DOI: 10.1126/scitranslmed.abj5070.
  • Dávila-González D., Choi D.S., Rosato R.R., Granados-Principal S.M., Kuhn J.G., Li W.F. et al. Pharmacological inhibition of NOS activates ASK1/JNK pathway augmenting docetaxel-mediated apoptosis in triple-negative breast cancer. Clin. Cancer Res. 2018;24(5):1152–1162. DOI: 10.1158/1078-0432.CCR-17-1437.
  • Howes L.G., Brillante D.G. Expert opinion on tilarginine in the treatment of shock. Expert. Opin. Investig. Drugs. 2008;17(10):1573–1580. DOI: 10.1517/13543784.17.10.1573.
  • Barbanti P., Egeo G., Aurilia C., Fofi L., Della-Morte D. Drugs targeting nitric oxide synthase for migraine treatment. Expert. Opin. Investig. Drugs. 2014;23(8):1141–1148. DOI: 10.1517/13543784.2014.918953.
  • Ott C., Bosch A., Winzer N., Friedrich S., Schinzel R., Tegtmeier F. et al. Effects of the nitric oxide synthase inhibitor ronopterin (VAS203) on renal function in healthy volunteers. Br. J. Clin. Pharmacol. 2019;85(5):900–907. DOI: 10.1111/bcp.13870.
Еще
Статья