121
Views
8
CrossRef citations to date
0
Altmetric
Review

Human Sirtuin 2 inhibitors, Their Mechanisms and Binding Modes

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 291-311 | Received 11 Oct 2022, Accepted 18 Jan 2023, Published online: 09 Mar 2023

References

  • Deans C , MaggertKA. What do you mean, “epigenetic”?Genetics199(4), 887–896 (2015).
  • Gujral P , MahajanV, LissamanAC, PonnampalamAP. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod. Biol. Endocrinol.18(1), 84 (2020).
  • Skvortsova K , IovinoN, BogdanovicO. Functions and mechanisms of epigenetic inheritance in animals. Nat. Rev. Mol. Cell Biol.19(12), 774–790 (2018).
  • Hogg SJ , BeavisPA, DawsonMA, JohnstoneRW. Targeting the epigenetic regulation of antitumour immunity. Nat. Rev. Drug Discov.19(11), 776–800 (2020).
  • Zhang L , LuQ, ChangC. Epigenetics in health and disease. Adv. Exp. Med. Biol.1253, 3–55 (2020).
  • Gomes RA , FornariE, SilvaRocha AC, TripodiGL, SilvaEmery FD, GoulartTrossini GH. Parasitic sirtuin 2 as an opportunity in drug discovery. Future Med. Chem.13(16), 1397–1409 (2021).
  • Toro TB , WattTJ. Critical review of non-histone human substrates of metal-dependent lysine deacetylases. FASEB J.34(10), 13140–13155 (2020).
  • Van Dyke MW . Lysine deacetylase (KDAC) regulatory pathways: an alternative approach to selective modulation. Chem. Med. Chem.9(3), 511–522 (2014).
  • Wang Q , RosaBA, NareBet al. Targeting lysine deacetylases (KDACs) in parasites. PLoS Negl. Trop. Dis.9(9), e0004026 (2015).
  • Chen X , LuW, WuD. Sirtuin 2 (SIRT2): confusing roles in the pathophysiology of neurological disorders. Front. Neurosci.15, 614107 (2021).
  • Haigis MC , SinclairDA. Mammalian sirtuins: biological insights and disease relevance. Annu. Rev. Pathol.5, 253–295 (2010).
  • Garg G , SinghAK, SinghS, RizviSI. Promising drug discovery strategies for sirtuin modulators: what lessons have we learnt?Expert Opin. Drug Discov.16(8), 915–927 (2021).
  • Samanta S , RajasinghS, CaoT, DawnB, RajasinghJ. Epigenetic dysfunctional diseases and therapy for infection and inflammation. Biochim. Biophys. Acta Mol. Basis Dis.1863(2), 518–528 (2017).
  • Blasl AT , SchulzeS, QinC, GrafLG, VogtR, LammersM. Post-translational lysine ac(et)ylation in health, ageing and disease. Biol. Chem.403(2), 151–194 (2022).
  • Grozinger CM , SchreiberSL. Deacetylase enzymes. Chem. Biol.9(1), 3–16 (2002).
  • Li G , TianY, ZhuW-G. The roles of histone deacetylases and their inhibitors in cancer therapy. Front. Cell Dev. Biol.8, 576946 (2020).
  • Avalos JL , CelicI, MuhammadS, CosgroveMS, BoekeJD, WolbergerC. Structure of a Sir2 enzyme bound to an acetylated p53 peptide. Mol. Cell10(3), 523–535 (2002).
  • Blander G , GuarenteL. The Sir2 family of protein deacetylases. Annu. Rev. Biochem.73, 417–435 (2004).
  • Chen B , ZangW, WangJet al. The chemical biology of sirtuins. Chem. Soc. Rev.44(15), 5246–5264 (2015).
  • Fang Y , LiX. Sirtuins in metabolic and epigenetic regulation of stem cells. In: Sirtuin Biology in Cancer and Metabolic DiseaseMaieseK ( Ed.). Academic Press, 25–37 (2021).
  • Houtkooper RH , PirinenE, AuwerxJ. Sirtuins as regulators of metabolism and healthspan. Nat. Rev. Mol. Cell Biol.13(4), 225–238 (2012).
  • Jing E , GestaS, KahnCR. SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation. Cell Metab.6(2), 105–114 (2007).
  • Wang Y , YangJ, HongT, ChenX, CuiL. SIRT2: controversy and multiple roles in disease and physiology. Ageing Res. Rev.55, 100961 (2019).
  • Zheng M , HuC, WuM, ChinY. Emerging role of SIRT2 in non-small cell lung cancer (review). Oncol. Lett.22(4), 731 (2021).
  • Hiratsuka M , InoueT, TodaTet al. Proteomics-based identification of differentially expressed genes in human gliomas: down-regulation of SIRT2 gene. Biochem. Biophys. Res. Commun.309(3), 558–566 (2003).
  • North BJ , VerdinE. Mitotic regulation of SIRT2 by cyclin-dependent kinase 1-dependent phosphorylation. J. Biol. Chem.282(27), 19546–19555 (2007).
  • Rack JGM , VanlindenMR, LutterT, AaslandR, ZieglerM. Constitutive nuclear localization of an alternatively spliced sirtuin-2 isoform. J. Mol. Biol.426(8), 1677–1691 (2014).
  • North BJ , MarshallBL, BorraMT, DenuJM, VerdinE. The human Sir2 ortholog, SIRT2, is an NAD+-dependent tubulin deacetylase. Mol. Cell11(2), 437–444 (2003).
  • Schemies J , UciechowskaU, SipplW, JungM. NAD(+) -dependent histone deacetylases (sirtuins) as novel therapeutic targets. Med. Res. Rev.30(6), 861–889 (2010).
  • Dryden SC , NahhasFA, NowakJE, GoustinAS, TainskyMA. Role for human SIRT2 NAD-dependent deacetylase activity in control of mitotic exit in the cell cycle. Mol. Cell. Biol.23(9), 3173–3185 (2003).
  • Osaki T , TakimotoT, FurukawaMet al. Clinical efficacy of TS-1 in refractory bone recurrence of a postoperative squamous lung cancer patient. Gan To Kagaku Ryoho33(8), 1119–1123 (2006).
  • North BJ , RosenbergMA, JeganathanKBet al. SIRT2 induces the checkpoint kinase BubR1 to increase lifespan. EMBO J.33(13), 1438–1453 (2014).
  • Inoue T , HiratsukaM, OsakiMet al. SIRT2, a tubulin deacetylase, acts to block the entry to chromosome condensation in response to mitotic stress. Oncogene26(7), 945–957 (2007).
  • Vaquero A , ScherMB, LeeDHet al. SirT2 is a histone deacetylase with preference for histone H4 Lys 16 during mitosis. Genes Dev.20(10), 1256–1261 (2006).
  • Wang F , NguyenM, QinFX, TongQ. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell6(4), 505–514 (2007).
  • Wang Y-P , ZhouL-S, ZhaoY-Zet al. Regulation of G6PD acetylation by SIRT2 and KAT9 modulates NADPH homeostasis and cell survival during oxidative stress. EMBO J.33(12), 1304–1320 (2014).
  • Quinti L , CasaleM, MoniotSet al. SIRT2- and NRF2-targeting thiazole-containing compound with therapeutic activity in Huntington's disease models. Cell Chem. Biol.23(7), 849–861 (2016).
  • Yu J , AuwerxJ. The role of sirtuins in the control of metabolic homeostasis. Ann. NY Acad. Sci.1173(Suppl. 1), E10–E19 (2009).
  • Armoni M , HarelC, KarniSet al. FOXO1 represses peroxisome proliferator-activated receptor-γ1 and -γ2 gene promoters in primary adipocytes: a novel paradigm to increase insulin sensitivity. J. Biol. Chem.281(29), 19881–19891 (2006).
  • Wang F , TongQ. SIRT2 suppresses adipocyte differentiation by deacetylating FOXO1 and enhancing FOXO1's repressive interaction with PPARgamma. Mol. Biol. Cell20(3), 801–808 (2009).
  • Wang Q , JiangL, WangJet al. Abrogation of hepatic ATP-citrate lyase protects against fatty liver and ameliorates hyperglycemia in leptin receptor-deficient mice. Hepatology49(4), 1166–1175 (2009).
  • Jung CJ , IyengarS, BlahnikKRet al. Epigenetic modulation of miR-122 facilitates human embryonic stem cell self-renewal and hepatocellular carcinoma proliferation. PLOS ONE6(11), e27740 (2011).
  • Ramakrishnan G , DavaakhuuG, KaplunLet al. Sirt2 deacetylase is a novel AKT binding partner critical for AKT activation by insulin. J. Biol. Chem.289(9), 6054–6066 (2014).
  • Finnin MS , DonigianJR, PavletichNP. Structure of the histone deacetylase SIRT2. Nature Struct. Biol.8(7), 621–625 (2001).
  • Moniot S , SchutkowskiM, SteegbornC. Crystal structure analysis of human Sirt2 and its ADP-ribose complex. J. Struct. Biol.182(2), 136–143 (2013).
  • Rumpf T , SchiedelM, KaramanBet al. Selective Sirt2 inhibition by ligand-induced rearrangement of the active site. Nat. Commun.6, 6263 (2015).
  • Hoff KG , AvalosJL, SensK, WolbergerC. Insights into the sirtuin mechanism from ternary complexes containing NAD+ and acetylated peptide. Structure14(8), 1231–1240 (2006).
  • Hong JY , PriceIR, BaiJJ, LinH. A glycoconjugated SIRT2 inhibitor with aqueous solubility allows structure-based design of SIRT2 inhibitors. ACS Chem. Biol.14(8), 1802–1810 (2019).
  • Disch JS , EvindarG, ChiuCHet al. Discovery of thieno[3,2-d]pyrimidine-6-carboxamides as potent inhibitors of SIRT1, SIRT2, and SIRT3. J. Med. Chem.56(9), 3666–3679 (2013).
  • Knyphausen P , DeBoor S, KuhlmannNet al. Insights into lysine deacetylation of natively folded substrate proteins by sirtuins. J. Biol. Chem.291(28), 14677–14694 (2016).
  • Rumpf T , GerhardtS, EinsleO, JungM. Seeding for sirtuins: microseed matrix seeding to obtain crystals of human Sirt3 and Sirt2 suitable for soaking. Acta Crystallogr. F Struct. Biol. Commun.71(Pt 12), 1498–1510 (2015).
  • Yang LL , WangHL, ZhongLet al. X-ray crystal structure guided discovery of new selective, substrate-mimicking sirtuin 2 inhibitors that exhibit activities against non-small cell lung cancer cells. Eur. J. Med. Chem.155, 806–823 (2018).
  • Avalos JL , BeverKM, WolbergerC. Mechanism of sirtuin inhibition by nicotinamide: altering the NAD(+) cosubstrate specificity of a Sir2 enzyme. Mol. Cell17(6), 855–868 (2005).
  • Gertz M , FischerF, NguyenGTet al. Ex-527 inhibits sirtuins by exploiting their unique NAD+-dependent deacetylation mechanism. Proc. Natl Acad. Sci. USA110(30), E2772–E2781 (2013).
  • Yamagata K , GotoY, NishimasuHet al. Structural basis for potent inhibition of SIRT2 deacetylase by a macrocyclic peptide inducing dynamic structural change. Structure22(2), 345–352 (2014).
  • Cao D , WangM, QiuXet al. Structural basis for allosteric, substrate-dependent stimulation of SIRT1 activity by resveratrol. Genes Dev.29(12), 1316–1325 (2015).
  • Milne JC , LambertPD, SchenkSet al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature450(7170), 712–716 (2007).
  • Nguyen GT , SchaeferS, GertzM, WeyandM, SteegbornC. Structures of human sirtuin 3 complexes with ADP-ribose and with carba-NAD+ and SRT1720: binding details and inhibition mechanism.D Biol. Crystallogr.69(Pt 8), 1423–1432 (2013).
  • Schiedel M , RumpfT, KaramanBet al. Aminothiazoles as potent and selective Sirt2 inhibitors: a structure-activity relationship study. J. Med. Chem.59(4), 1599–1612 (2016).
  • Kudo N , ItoA, ArataM, NakataA, YoshidaM. Identification of a novel small molecule that inhibits deacetylase but not defatty-acylase reaction catalysed by SIRT2. Philos. Trans. R Soc. B Bio. Sci.373(1748), 20170070 (2018).
  • Mellini P , ItohY, TsumotoHet al. Potent mechanism-based sirtuin-2-selective inhibition by an in situ-generated occupant of the substrate-binding site, “selectivity pocket” and NAD(+)-binding site. Chem. Sci.8(9), 6400–6408 (2017).
  • Moniot S , ForgioneM, LucidiAet al. Development of 1,2,4-oxadiazoles as potent and selective inhibitors of the human deacetylase sirtuin 2: structure-activity relationship, x-ray crystal structure, and anticancer activity. J. Med. Chem.60(6), 2344–2360 (2017).
  • Sundriyal S , MoniotS, MahmudZet al. Thienopyrimidinone based sirtuin-2 (SIRT2)-selective inhibitors bind in the ligand induced selectivity pocket. J. Med. Chem.60(5), 1928–1945 (2017).
  • Teng YB , JingH, AramsangtienchaiPet al. Efficient demyristoylase activity of SIRT2 revealed by kinetic and structural studies. Sci. Rep.5, 8529 (2015).
  • Nielsen AL , RajabiN, KudoNet al. Mechanism-based inhibitors of SIRT2: structure-activity relationship, x-ray structures, target engagement, regulation of alpha-tubulin acetylation and inhibition of breast cancer cell migration. RSC Chem. Biol.2(2), 612–626 (2021).
  • Feldman JL , Dittenhafer-ReedKE, KudoNet al. Kinetic and structural basis for acyl-group selectivity and NAD(+) dependence in sirtuin-catalyzed deacylation. Biochemistry54(19), 3037–3050 (2015).
  • Jin J , HeB, ZhangX, LinH, WangY. SIRT2 reverses 4-oxononanoyl lysine modification on histones. J. Am. Chem. Soc.138(38), 12304–12307 (2016).
  • Szczepankiewicz BG , DaiH, KoppetschKJet al. Synthesis of carba-NAD and the structures of its ternary complexes with SIRT3 and SIRT5. J. Org. Chem.77(17), 7319–7329 (2012).
  • Larkin MA , BlackshieldsG, BrownNPet al. Clustal W and Clustal X version 2.0. Bioinformatics23(21), 2947–2948 (2007).
  • Pettersen EF , GoddardTD, HuangCCet al. UCSF Chimera–a visualization system for exploratory research and analysis. J. Comput. Chem.25(13), 1605–1612 (2004).
  • Webb B , SaliA. Comparative protein structure modeling using MODELLER. Curr. Protoc. Bioinformatics doi: 10.1002/0471250953.bi0506s15 (2016).
  • Schrödinger LLC . The PyMOL Molecular Graphics System. (2021).
  • Abraham MJ , MurtolaT, SchulzRet al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX1-2, 19–25 (2015).
  • Lindorff-Larsen K , PianaS, PalmoKet al. Improved side-chain torsion potentials for the Amber ff99SB protein force field. Proteins78(8), 1950–1958 (2010).
  • Wang J , WolfRM, CaldwellJW, KollmanPA, CaseDA. Development and testing of a general amber force field. J. Comput. Chem.25(9), 1157–1174 (2004).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.