Please wait a minute...
浙江大学学报(医学版)  2020, Vol. 49 Issue (1): 1-19    DOI: 10.3785/j.issn.1008-9292.2020.02.21
专题报道     
靶向Cullin-RING E3泛素连接酶的抗肿瘤策略及相关药物研发进展
俞卿1,2(),熊秀芳1,2,孙毅1,2,*()
1. 浙江大学医学院附属第二医院肿瘤研究所, 浙江 杭州 310009
2. 浙江大学转化医学研究院, 浙江 杭州 310029
Targeting Cullin-RING E3 ligases for anti-cancer therapy: efforts on drug discovery
YU Qing1,2(),XIONG Xiufang1,2,SUN Yi1,2,*()
1. Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
2. Department of Translational Medicine, Zhejiang University, Hangzhou 310029, China
 全文: PDF(8974 KB)   HTML( 61 )
摘要:

Cullin-RING E3泛素连接酶(CRL)是泛素-蛋白酶体系统的重要组分,参与催化蛋白质的泛素化,促进随后的蛋白质降解,从而影响细胞周期、细胞凋亡、DNA复制、信号转导等多种细胞生理活动,且在多种肿瘤细胞中异常活化。以MLN4924为代表的拟素化抑制剂的成功研发有力地证实了CRL是可行的抗肿瘤靶点,具有很好的药物研发潜力。近年来,不断有新的研究通过高通量筛选、基于计算机辅助的虚拟筛选或基于结构的药物设计技术寻找特异的CRL抑制剂,但由于CRL复合物具有多种亚单位,呈蛋白-蛋白相互作用和多变的蛋白构象,缺乏典型的小分子药物结合位点等特性,其相关药物研发仍面临巨大挑战。截至目前,CRL小分子抑制剂主要以研究最为透彻的SCF泛素连接酶复合体的底物识别亚基F-box蛋白家族为靶点。此外,也发现数个通过靶向UBE2M-DCN1相互作用,特异性阻断CRL3/CRL1拟素化,从而抑制CRL3/CRL1泛素连接酶活性的小分子化合物。另一方面,也有CRL激动剂的报道,主要见于植物生长素吲哚乙酸和免疫调节性酰亚胺类药物。此外,靶蛋白水解嵌合体(PROTAC)是一项靶向蛋白-蛋白相互作用的新技术,其通过特异性小分子抑制剂链接一个CRL E3泛素连接酶来精确降解特定促癌靶蛋白,已成为近年来利用E3泛素连接酶设计抗肿瘤靶向药物的热点。

关键词: 泛素蛋白连接酶类抗肿瘤/治疗学Cullin-RING E3泛素连接酶酶抑制剂酶激动剂类靶蛋白水解嵌合体综述    
Abstract:

Cullin-RING E3 ligases (CRLs) are the major components of ubiquitin-proteasome system, responsible for ubiquitylation and subsequent degradation of thousands of cellular proteins. CRLs play vital roles in the regulation of multiple cellular processes, including cell cycle, cell apoptosis, DNA replication, signalling transduction among the others, and are frequently dysregulated in many human cancers. The discovery of specific neddylation inhibitors, represented by MLN4924, has validated CRLs as promising targets for anti-cancer therapies with a growing market. Recent studies have focused on the discovery of the CRLs inhibitors by a variety of approaches, including high through-put screen, virtual screen or structure-based drug design. The field is, however, still facing the major challenging, since CRLs are a large multi-unit protein family without typical active pockets to facilitate the drug design, and enzymatic activity is mainly dependent on undruggable protein-protein interactions and dynamic conformation changes. Up to now, most reported CRLs inhibitors are aiming at targeting the F-box family proteins (e.g., SKP2, β-TrCP and FBXW7), the substrate recognition subunit of SCF E3 ligases. Other studies reported few small molecule inhibitors targeting the UBE2M-DCN1 interaction, which specifically inhibits CRL3/CRL1 by blocking the cullin neddylation. On the other hand, several CRL activators have been reported, such as plant auxin and immunomodulatory imide drugs, thalidomide. Finally, proteolysis-targeting chimeras (PROTACs) has emerged as a new technology in the field of drug discovery, specifically targeting the undruggable protein-protein interaction. The technique connects the small molecule that selectively binds to a target protein to a CRL E3 via a chemical linker to trigger the degradation of target protein. The PROTAC has become a hotspot in the field of E3-ligase-based anti-cancer drug discovery.

Key words: Ubiquitin-protein ligases    Antineoplastic/therapeutic    Cullin-RING E3 ligases    Enzyme inhibitors    Enzyme activators    Proteolysis-targeting chimera    Review
收稿日期: 2019-12-20 出版日期: 2020-06-08
CLC:  R730.5  
基金资助: 国家重点研发计划(2016YFA0501800)
通讯作者: 孙毅     E-mail: yuqing@zju.edu.cn;yisun@zju.edu.cn
作者简介: 俞卿, 男, 博士研究生, 主要从事Cullin-RINGE3泛素连接酶小分子抑制剂的高通量筛选和研发; E-mail:yuqing@zju.edu.cn; https://orcid.org/0000-0002-4519-4940
服务  
把本文推荐给朋友
加入引用管理器
E-mail Alert
RSS
作者相关文章  
俞卿
熊秀芳
孙毅

引用本文:

俞卿, 熊秀芳, 孙毅. 靶向Cullin-RING E3泛素连接酶的抗肿瘤策略及相关药物研发进展[J]. 浙江大学学报(医学版), 2020, 49(1): 1-19.

YU Qing, XIONG Xiufang, SUN Yi. Targeting Cullin-RING E3 ligases for anti-cancer therapy: efforts on drug discovery. J Zhejiang Univ (Med Sci), 2020, 49(1): 1-19.

链接本文:

http://www.zjujournals.com/med/CN/10.3785/j.issn.1008-9292.2020.02.21        http://www.zjujournals.com/med/CN/Y2020/V49/I1/1

图 1  泛素-蛋白酶体系统概况和Cullin-RING E3泛素连接酶的拟素化激活
图 2  Cullin-RING E3泛素连接酶的构成及已知底物
图 3  抑制和激活Cullin-RING E3泛素连接酶(CRL)的抗肿瘤策略
名称 靶蛋白 影响的PPI 对细胞中主要底物的影响 研发途径 其他说明 参考文献
“—”无相关资料.PPI:蛋白-蛋白相互作用; FRET:荧光共振能量转移; TR-FRET:时间分辨荧光共振能量转移; HTRF:均相时间分辨荧光.
CRL E3抑制剂
Compound A SKP2 SKP2:SKP1 p27、p21、p53蛋白积累 高通量筛选 在多发性骨髓瘤细胞中可增加细胞对硼替佐米敏感性 [54]
C1、C2、C16、C20 SKP2 SKP2:Cks1 p27蛋白积累 基于配体分子的虚拟筛选 首次通过虚拟筛选获得阻断SKP2-底物相互作用的小分子 [55]
Compound #25 SKP2 SKP2:SKP1 p27、p21、Notch1蛋白积累 基于受体结构的虚拟筛选 在前列腺癌细胞系PC3中可降低肿瘤干细胞比例 [56]
NSC689857、NSC681152 SKP2 SKP2:Cks1 p27蛋白积累 基于AlphaScreen的高通量筛选 同时具有络氨酸激酶抑制活性 [57]
Eroflorin β-TrCP β-TrCP:PDCD4 PDCD4蛋白积累 基于荧光素酶报告系统的高通量筛选 天然产物 [58]
SCF-I2 Cdc4(酵母FBXW7) Cdc4:Sic1 Sic1蛋白泛素化水平降低 基于荧光偏振的高通量筛选(酵母) 第一个特异性靶向WD40结构域的小分子变构抑制剂 [59]
Suramin CUL1 CUL1:CDC34 p27、CDT1蛋白积累 基于FRET的高通量筛选 天然产物;基于FRET的K48双泛素化报告系统 [60]
SMER3 Met30(酵母F-box) Met30:Skp1 Met4蛋白泛素化水平降低 基于化学遗传学的高通量筛选(酵母) 小分子雷帕霉素增强剂 [61]
NAcM-HIT、NAcM-OPT等 DCN1 DCN1:UBE2M p27、Nrf2蛋白积累 基于TR-FRET的高通量筛选 特异性抑制CRL3/CRL1活性 [62-65]
DI-591、DI-404 DCN1 DCN1:UBE2M Nrf2蛋白积累 基于结构的药物设计 特异性抑制CRL3活性 [66-67]
WS-383 DCN1 DCN1:UBE2M p21、p27、Nrf2蛋白积累 基于HTRF的高通量筛选 特异性抑制CRL3/CRL1活性 [68]
DC-2、DC-1 DCN1 DCN1:UBE2M Nrf2蛋白积累 基于HTRF和荧光偏振的高通量筛选 特异性抑制CRL3活性 [69]
CRL E3激动剂
Auxin TIR1(植物) TIR1:Aux/IAA 促进Aux/IAA降解 非创新药研发 [70-71]
Thalidomide、Lenalidomide、Pomalidomide CRL4-CRBN CRBN:IKZF1/3 促进IKZF1和IKZF3降解 非创新药研发 [72-75]
NRX-1532、NRX-252262 β-catenin/ β-TrCP β-catenin:β-TrCP 促进S37A突变β-catenin降解 基于荧光偏振的高通量筛选 [76]
Oridonin FBXW7 尚不明确 促进c-Myc降解 非创新药研发 [77]
表 1  Cullin-RING E3泛素连接酶抑制剂和激动剂
图 4  靶向Cullin-RING E3泛素连接酶的小分子抑制剂
图 5  靶向UBE2M-DCN1抑制CRL3/CRL1的小分子抑制剂
图 6  Cullin-RING E3泛素连接酶激活剂的“分子胶水”作用原理图
图 7  靶蛋白水解嵌合体的基本原理图
名称 结构 识别的E3 靶蛋白 其他说明 参考文献
PROTAC-1 SCFβ-TrCP MetAp-2 第一个PROTAC [106]
PROTAC-4 CRL2VHL FKBP12F36V 第一个可进入细胞的PROTAC [107]
TrkAPPFRS2α CRL2VHL FRS2α 第一个磷酸化的PROTAC [108]
SARM-nutlin PROTAC MDM2 雄激素受体 第一个全小分子的PROTAC [109]
ARV-110 暂无 暂无 雄激素受体 第一个进入临床试验的PROTAC [110]
表 2  具有代表性的靶蛋白水解嵌合体(PROTAC)及相关衍生技术
1 HERSHKO A , CIECHANOVER A . The ubiquitin system[J]. Annu Rev Biochem, 1998, 67:425- 479
doi: 10.1146/annurev.biochem.67.1.425
2 KOMANDER D , RAPE M . The ubiquitin code[J]. Annu Rev Biochem, 2012, 81:203- 229
doi: 10.1146/annurev-biochem-060310-170328
3 ZHAO Y , SUN Y . Cullin-RING ligases as attractive anti-cancer targets[J]. Curr Pharm Des, 2013, 19 (18): 3215- 3225
doi: 10.2174/13816128113199990300
4 DESHAIES R J , JOAZEIRO C A . RING domain E3 ubiquitin ligases[J]. Annu Rev Biochem, 2009, 78:399- 434
doi: 10.1146/annurev.biochem.78.101807.093809
5 ZHENG N , SHABEK N . Ubiquitin ligases:structure, function, and regulation[J]. Annu Rev Biochem, 2017, 86:129- 157
doi: 10.1146/annurev-biochem-060815-014922
6 DOVE K K , KLEVIT R E . RING-between-RING E3 ligases:emerging themes amid the variations[J]. J Mol Biol, 2017, 429 (22): 3363- 3375
doi: 10.1016/j.jmb.2017.08.008
7 SOUCY T A , SMITH P G , MILHOLLEN M A et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer[J]. Nature, 2009, 458 (7239): 732- 736
doi: 10.1038/nature07884
8 RICHARDSON P G , BARLOGIE B , BERENSON J et al. A phase 2 study of bortezomib in relapsed, refractory myeloma[J]. N Engl J Med, 2003, 348 (26): 2609- 2617
doi: 10.1056/NEJMoa030288
9 SKAAR J R , PAGAN J K , PAGANO M . SCF ubiquitin ligase-targeted therapies[J]. Nat Rev Drug Discov, 2014, 13 (12): 889- 903
doi: 10.1038/nrd4432
10 SKAAR J R , FLORENS L , TSUTSUMI T et al. PARC and CUL7 form atypical cullin RING ligase complexes[J]. Cancer Res, 2007, 67 (5): 2006- 2014
doi: 10.1158/0008-5472.CAN-06-3241
11 JIN J , CARDOZO T , LOVERING R C et al. Systematic analysis and nomenclature of mammalian F-box proteins[J]. Genes Dev, 2004, 18 (21): 2573- 2580
doi: 10.1101/gad.1255304
12 LINOSSI E M , NICHOLSON S E . The SOCS box-adapting proteins for ubiquitination and proteasomal degradation[J]. IUBMB Life, 2012, 64 (4): 316- 323
doi: 10.1002/iub.1011
13 STOGIOS P J , DOWNS G S , JAUHAL J J et al. Sequence and structural analysis of BTB domain proteins[J]. Genome Biol, 2005, 6 (10):
doi: 10.1186/gb-2005-6-10-r82
14 HE Y J , MCCALL C M , HU J et al. DDB1 functions as a linker to recruit receptor WD40 proteins to CUL4-ROC1 ubiquitin ligases[J]. Genes Dev, 2006, 20 (21): 2949- 2954
doi: 10.1101/gad.1483206
15 SARIKAS A , HARTMANN T , PAN Z Q . The cullin protein family[J]. Genome Biol, 2011, 12 (4): 220
doi: 10.1186/gb-2011-12-4-220
16 ZHAO Y , MORGAN M A , SUN Y . Targeting neddylation pathways to inactivate cullin-RING ligases for anticancer therapy[J]. Antioxid Redox Signal, 2014, 21 (17): 2383- 2400
doi: 10.1089/ars.2013.5795
17 JIA L , SUN Y . SCF E3 ubiquitin ligases as anticancer targets[J]. Curr Cancer Drug Targets, 2011, 11 (3): 347- 356
doi: 10.2174/156800911794519734
18 DESHAIES R J . SCF and Cullin/Ring H2-based ubiquitin ligases[J]. Annu Rev Cell Dev Biol, 1999, 15:435- 467
doi: 10.1146/annurev.cellbio.15.1.435
19 SKAAR J R , PAGAN J K , PAGANO M . Mechanisms and function of substrate recruitment by F-box proteins[J]. Nat Rev Mol Cell Biol, 2013, 14 (6): 369- 381
doi: 10.1038/nrm3582
20 SKAAR J R , D'ANGIOLELLA V , PAGAN J K et al. SnapShot:F box proteins Ⅱ[J]. Cell, 2009, 137:1358
doi: 10.1016/j.cell.2009.05.040
21 YAN Y , ZHANG X , LEGERSKI R J . Artemis interacts with the Cul4A-DDB1DDB2 ubiquitin E3ligase and regulates degradation of the CDK inhibitor p27[J]. Cell Cycle, 2011, 10:4098- 4109
doi: 10.4161/cc.10.23.18227
22 TAN M , ZHAO Y , KIM S J et al. SAG/RBX2/ROC2 E3 ubiquitin ligase is essential for vascular and neural development by targeting NF1 for degradation[J]. Dev Cell, 2011, 21 (6): 1062- 1076
doi: 10.1016/j.devcel.2011.09.014
23 JIN J , ARIAS E E , CHEN J et al. A family of diverse Cul4-Ddb1-interacting proteins includes Cdt2, which is required for S phase destruction of the replication factor Cdt1[J]. Mol Cell, 2006, 23 (5): 709- 721
doi: 10.1016/j.molcel.2006.08.010
24 HIGA L A , MIHAYLOV I S , BANKS D P et al. Radiation-mediated proteolysis of CDT1 by CUL4-ROC1 and CSN complexes constitutes a new checkpoint[J]. Nat Cell Biol, 2003, 5
doi: 10.1038/ncb1061
25 ZHAO Y , XIONG X , SUN Y . DEPTOR, an mTOR inhibitor, is a physiological substrate of SCF(betaTrCP) E3 ubiquitin ligase and regulates survival and autophagy[J]. Mol Cell, 2011, 44:304- 316
doi: 10.1016/j.molcel.2011.08.029
26 KAMURA T , MAENAKA K , KOTOSHIBA S et al. VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases[J]. Genes Dev, 2004, 18 (24): 3055- 3065
doi: 10.1101/gad.1252404
27 IVAN M , KONDO K , YANG H et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation:implications for O2 sensing[J]. Science, 2001, 292 (5516): 464- 468
doi: 10.1126/science.1059817
28 XIE L , XIAO K , WHALEN E J et al. Oxygen-regulated beta(2)-adrenergic receptor hydroxylation by EGLN3 and ubiquitylation by pVHL[J]. Sci Signal, 2009, 2 (78): ra33
doi: 10.1126/scisignal.2000444
29 OKUDA H , SAITOH K , HIRAI S et al. The von Hippel-Lindau tumor suppressor protein mediates ubiquitination of activated atypical protein kinase C[J]. J Biol Chem, 2001, 276 (47): 43611- 43617
doi: 10.1074/jbc.M107880200
30 NA X , DUAN H O , MESSING E M et al. Identification of the RNA polymerase Ⅱ subunit hsRPB7 as a novel target of the von Hippel-Lindau protein[J]. EMBO J, 2003, 22 (16): 4249- 4259
doi: 10.1093/emboj/cdg410
31 PUGH C W , RATCLIFFE P J . The von Hippel-Lindau tumor suppressor, hypoxia-inducible factor-1(HIF-1) degradation, and cancer pathogenesis[J]. Semin Cancer Biol, 2003, 13 (1): 83- 89
doi: 10.1016/s1044-579x(02)00103-7
32 CULLINAN S B , GORDAN J D , JIN J et al. The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase:oxidative stress sensing by a Cul3-Keap1 ligase[J]. Mol Cell Biol, 2004, 24 (19): 8477- 8486
doi: 10.1128/MCB.24.19.8477-8486.2004
33 HERNANDEZ-MU?OZ I , LUND A H , VAN DER STOOP P et al. Stable X chromosome inactivation involves the PRC1 polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase[J]. Proc Natl Acad Sci U S A, 2005, 102 (21): 7635- 7640
doi: 10.1073/pnas.0408918102
34 KWON J E , LA M , OH K H et al. BTB domain-containing speckle-type POZ protein (SPOP) serves as an adaptor of Daxx for ubiquitination by Cul3-based ubiquitin ligase[J]. J Biol Chem, 2006, 281 (18): 12664- 12672
doi: 10.1074/jbc.M600204200
35 KOBAYASHI A , KANG M I , OKAWA H et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2[J]. Mol Cell Biol, 2004, 24 (16): 7130- 7139
doi: 10.1128/MCB.24.16.7130-7139.2004
36 SUGASAWA K , OKUDA Y , SAIJO M et al. UV-induced ubiquitylation of XPC protein mediated by UV-DDB-ubiquitin ligase complex[J]. Cell, 2005, 121 (3): 387- 400
doi: 10.1016/j.cell.2005.02.035
37 ABBAS T , SHIBATA E , PARK J et al. CRL4(Cdt2) regulates cell proliferation and histone gene expression by targeting PR-Set7/Set8 for degradation[J]. Mol Cell, 2010, 40 (1): 9- 21
doi: 10.1016/j.molcel.2010.09.014
38 NAKAGAWA T , XIONG Y . X-linked mental retardation gene CUL4B targets ubiquitylation of H3K4 methyltransferase component WDR5 and regulates neuronal gene expression[J]. Mol Cell, 2011, 43 (3): 381- 391
doi: 10.1016/j.molcel.2011.05.033
39 ZHOU W , XU J , LI H et al. Neddylation E2 UBE2F promotes the survival of lung cancer cells by activating CRL5 to degrade NOXA via the K11 linkage[J]. Clin Cancer Res, 2017, 23 (4): 1104- 1116
doi: 10.1158/1078-0432.CCR-16-1585
40 YU X , YU Y , LIU B et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex[J]. Science, 2003, 302:1056- 1060
doi: 10.1126/science.1089591
41 QUERIDO E , BLANCHETTE P , YAN Q et al. Degradation of p53 by adenovirus E4orf6 and E1B55K proteins occurs via a novel mechanism involving a Cullin-containing complex[J]. Genes Dev, 2001, 15:3104- 3117
doi: 10.1101/gad.926401
42 PAN Z Q . Cullin-RING E3 ubiquitin ligase 7 in growth control and cancer[J]. Adv Exp Med Biol, 2020, 1217:285- 296
doi: 10.1007/978-981-15-1025-0_17
43 XU X , SARIKAS A , DIAS-SANTAGATA D C et al. The CUL7 E3 ubiquitin ligase targets insulin receptor substrate 1 for ubiquitin-dependent degradation[J]. Mol Cell, 2008, 30:403- 414
doi: 10.1016/j.molcel.2008.03.009
44 NIKOLAEV A Y , LI M , PUSKAS N et al. Parc:a cytoplasmic anchor for p53[J]. Cell, 2003, 112 (1): 29- 40
doi: 10.1016/s0092-8674(02)01255-2
45 HUANG X , DIXIT V M . Drugging the undruggables:exploring the ubiquitin system for drug development[J]. Cell Res, 2016, 26 (4): 484- 498
doi: 10.1038/cr.2016.31
46 NAKAYAMA K I , NAKAYAMA K . Ubiquitin ligases:cell-cycle control and cancer[J]. Nat Rev Cancer, 2006, 6:369- 381
doi: 10.1038/nrc1881
47 FRESCAS D , PAGANO M . Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP:tipping the scales of cancer[J]. Nat Rev Cancer, 2008, 8 (6): 438- 449
doi: 10.1038/nrc2396
48 WEI D , SUN Y . Small RING finger proteins RBX1 and RBX2 of SCF E3 ubiquitin ligases:the role in cancer and as cancer targets[J]. Genes Cancer, 2010, 1 (7): 700- 707
doi: 10.1177/1947601910382776
49 YU Q , JIANG Y , SUN Y . Anticancer drug discovery by targeting cullin neddylation[J]. Acta Pharmaceutica Sinica B, 2019,
doi: 10.1016/j.apsb.2019.09.005
50 MILHOLLEN M A , THOMAS M P , NARAYANAN U et al. Treatment-emergent mutations in NAEbeta confer resistance to the NEDD8-activating enzyme inhibitor MLN4924[J]. Cancer Cell, 2012, 21:388- 401
doi: 10.1016/j.ccr.2012.02.009
51 TOTH J I , YANG L , DAHL R et al. A gatekeeper residue for NEDD8-activating enzyme inhibition by MLN4924[J]. Cell Rep, 2012, 1 (4): 309- 316
doi: 10.1016/j.celrep.2012.02.006
52 ZHOU Q , SUN Y . MLN4924:additional activities beyond neddylation inhibition[J]. Mol Cell Oncol, 2019, 6 (5): e1618174
doi: 10.1080/23723556.2019.1618174
53 WELCKER M , CLURMAN B E . FBW7 ubiquitin ligase:a tumour suppressor at the crossroads of cell division, growth and differentiation[J]. Nat Rev Cancer, 2008, 8 (2): 83- 93
doi: 10.1038/nrc2290
54 CHEN Q , XIE W , KUHN D J et al. Targeting the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated cell-cycle arrest and activation of autophagy[J]. Blood, 2008, 111 (9): 4690- 4699
doi: 10.1182/blood-2007-09-112904
55 WU L , GRIGORYAN A V , LI Y et al. Specific small molecule inhibitors of Skp2-mediated p27 degradation[J]. Chem Biol, 2012, 19 (12): 1515- 1524
doi: 10.1016/j.chembiol.2012.09.015
56 CHAN C H , MORROW J K , LI C F et al. Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression[J]. Cell, 2013, 154 (3): 556- 568
doi: 10.1016/j.cell.2013.06.048
57 UNGERMANNOVA D , LEE J , ZHANG G et al. High-throughput screening AlphaScreen assay for identification of small-molecule inhibitors of ubiquitin E3 ligase SCFSkp2-Cks1[J]. J Biomol Screen, 2013, 18:910- 920
doi: 10.1177/1087057113485789
58 BLEES J S , BOKESCH H R , RUBSAMEN D et al. Erioflorin stabilizes the tumor suppressor Pdcd4 by inhibiting its interaction with the E3-ligase beta-TrCP1[J]. PLoS One, 2012, 7:e46567
doi: 10.1371/journal.pone.0046567
59 ORLICKY S , TANG X , NEDUVA V et al. An allosteric inhibitor of substrate recognition by the SCF(Cdc4) ubiquitin ligase[J]. Nat Biotechnol, 2010, 28 (7): 733- 737
doi: 10.1038/nbt.1646
60 WU K , CHONG R A , YU Q et al. Suramin inhibits cullin-RING E3 ubiquitin ligases[J]. Proc Natl Acad Sci U S A, 2016, 113 (14): E2011- 2018
doi: 10.1073/pnas.1601089113
61 AGHAJAN M , JONAI N , FLICK K et al. Chemical genetics screen for enhancers of rapamycin identifies a specific inhibitor of an SCF family E3 ubiquitin ligase[J]. Nat Biotechnol, 2010, 28 (7): 738- 742
doi: 10.1038/nbt.1645
62 SCOTT D C , HAMMILL J T , MIN J et al. Blocking an N-terminal acetylation-dependent protein interaction inhibits an E3 ligase[J]. Nat Chem Biol, 2017, 13 (8): 850- 857
doi: 10.1038/nchembio.2386
63 HAMMILL J T , BHASIN D , SCOTT D C et al. Discovery of an orally bioavailable inhibitor of defective in cullin neddylation 1(DCN1)-mediated cullin neddylation[J]. J Med Chem, 2018, 61 (7): 2694- 2706
doi: 10.1021/acs.jmedchem.7b01282
64 HAMMILL J T , SCOTT D C , MIN J et al. Piperidinyl ureas chemically control defective in cullin neddylation 1(DCN1)-mediated cullin neddylation[J]. J Med Chem, 2018, 61 (7): 2680- 2693
doi: 10.1021/acs.jmedchem.7b01277
65 KIM H S , HAMMILL J T , SCOTT D C et al. Discovery of novel pyrazolo-pyridone DCN1 inhibitors controlling cullin neddylation[J]. J Med Chem, 2019, 62 (18): 8429- 8442
doi: 10.1021/acs.jmedchem.9b00410
66 ZHOU H , LU J , LIU L et al. A potent small-molecule inhibitor of the DCN1-UBC12 interaction that selectively blocks cullin 3 neddylation[J]. Nat Commun, 2017, 8 (1): 1150
doi: 10.1038/s41467-017-01243-7
67 ZHOU H , ZHOU W , ZHOU B et al. High-affinity peptidomimetic inhibitors of the DCN1-UBC12 protein-protein interaction[J]. J Med Chem, 2018, 61 (5): 1934- 1950
doi: 10.1021/acs.jmedchem.7b01455
68 WANG S , ZHAO L , SHI X J et al. Development of highly potent, selective, and cellular active triazolo[1, 5- a]pyrimidine-based inhibitors targeting the DCN1-UBC12 protein-protein interaction[J]. J Med Chem, 2019, 62 (5): 2772- 2797
doi: 10.1021/acs.jmedchem.9b00113
69 ZHOU W , MA L , DING L et al. Potent 5-cyano-6-phenyl-pyrimidin-based derivatives targeting DCN1-UBE2M interaction[J]. J Med Chem, 2019, 62 (11): 5382- 5403
doi: 10.1021/acs.jmedchem.9b00003
70 KEPINSKI S , LEYSER O . The arabidopsis F-box protein TIR1 is an auxin receptor[J]. Nature, 2005, 435 (7041): 446- 451
doi: 10.1038/nature03542
71 TAN X , CALDERON-VILLALOBOS L I , SHARON M et al. Mechanism of auxin perception by the TIR1 ubiquitin ligase[J]. Nature, 2007, 446 (7136): 640- 645
doi: 10.1038/nature05731
72 ITO T , ANDO H , SUZUKI T et al. Identification of a primary target of thalidomide teratogenicity[J]. Science, 2010, 327 (5971): 1345- 1350
doi: 10.1126/science.1177319
73 KR?NKE J , UDESHI N D , NARLA A et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells[J]. Science, 2014, 343 (6168): 301- 305
doi: 10.1126/science.1244851
74 PETZOLD G , FISCHER E S , THOMA N H . Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase[J]. Nature, 2016, 532 (7597): 127- 130
doi: 10.1038/nature16979
75 FISCHER E S , B?HM K , LYDEARD J R et al. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide[J]. Nature, 2014, 512 (7512): 49- 53
doi: 10.1038/nature13527
76 SIMONETTA K R , TAYGERLY J , BOYLE K et al. Prospective discovery of small molecule enhancers of an E3 ligase-substrate interaction[J]. Nat Commun, 2019, 10 (1): 1402
doi: 10.1038/s41467-019-09358-9
77 HUANG H L , WENG H Y , WANG L Q et al. Triggering Fbw7-mediated proteasomal degradationof c-Myc by oridonin induces cell growth inhibition and apoptosis[J]. Mol Cancer Ther, 2012, 11 (5): 1155- 1165
doi: 10.1158/1535-7163.MCT-12-0066
78 NAKAYAMA K , NAGAHAMA H , MINAMISHIMA Y A et al. Skp2-mediated degradation of p27 regulates progression into mitosis[J]. Dev Cell, 2004, 6 (5): 661- 672
doi: 10.1016/s1534-5807(04)00131-5
79 NAKAYAMA K , NAGAHAMA H , MINAMISHIMA Y A et al. Targeted disruption of Skp2 results in accumulation of cyclin E and p27(Kip1), polyploidy and centrosome overduplication[J]. EMBO J, 2000, 19:2069- 2081
doi: 10.1093/emboj/19.9.2069
80 KOSSATZ U , DIETRICH N , ZENDER L et al. Skp2-dependent degradation of p27kip1 is essential for cell cycle progression[J]. Genes Dev, 2004, 18 (21): 2602- 2607
doi: 10.1101/gad.321004
81 LI X , ELMIRA E , ROHONDIA S et al. A patent review of the ubiquitin ligase system:2015-2018[J]. Expert Opin Ther Pat, 2018, 28 (12): 919- 937
doi: 10.1080/13543776.2018.1549229
82 KULLMANN M K , GRUBBAUER C , GOETSCH K et al. The p27-Skp2 axis mediates glucocorticoid-induced cell cycle arrest in T-lymphoma cells[J]. Cell Cycle, 2013, 12 (16): 2625- 2635
doi: 10.4161/cc.25622
83 ZHAO H , BAUZON F , FU H et al. Skp2 deletion unmasks a p27 safeguard that blocks tumorigenesis in the absence of pRb and p53 tumor suppressors[J]. Cancer Cell, 2013, 24 (5): 645- 659
doi: 10.1016/j.ccr.2013.09.021
84 HULIT J , LEE R J , LI Z et al. p27Kip1 repression of ErbB2-induced mammary tumor growth in transgenic mice involves Skp2 and Wnt/beta-catenin signaling[J]. Cancer Res, 2006, 66 (17): 8529- 8541
doi: 10.1158/0008-5472.CAN-06-0149
85 RICO-BAUTISTA E , YANG C C , LU L et al. Chemical genetics approach to restoring p27Kip1 reveals novel compounds with antiproliferative activity in prostate cancer cells[J]. BMC Biol, 2010, 8:153
doi: 10.1186/1741-7007-8-153
86 RICO-BAUTISTA E , ZHU W , KITADA S et al. Small molecule-induced mitochondrial disruption directs prostate cancer inhibition via UPR signaling[J]. Oncotarget, 2013, 4 (8): 1212- 1229
doi: 10.18632/oncotarget.1130
87 OH M , LEE J H , MOON H et al. A chemical inhibitor of the Skp2/p300 interaction that promotes p53-mediated apoptosis[J]. Angew Chem Int Ed Engl, 2016, 55 (2): 602- 606
doi: 10.1002/anie.201508716
88 ZHENG N , ZHOU Q , WANG Z et al. Recent advances in SCF ubiquitin ligase complex:Clinical implications[J]. Biochim Biophys Acta, 2016, 1866 (1): 12- 22
doi: 10.1016/j.bbcan.2016.05.001
89 WEI N A , LIU S S , LEUNG T H et al. Loss of programmed cell death 4(Pdcd4) associates with the progression of ovarian cancer[J]. Mol Cancer, 2009, 8:70
doi: 10.1186/1476-4598-8-70
90 AKHOONDI S , SUN D , VON DER LEHR N et al. FBXW7/hCDC4 is a general tumor suppressor in human cancer[J]. Cancer Res, 2007, 67 (19): 9006- 9012
doi: 10.1158/0008-5472.CAN-07-1320
91 WELCKER M , ORIAN A , JIN J et al. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation[J]. Proc Natl Acad Sci U S A, 2004, 101 (24): 9085- 9090
doi: 10.1073/pnas.0402770101
92 YADA M , HATAKEYAMA S , KAMURA T et al. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7[J]. EMBO J, 2004, 23 (10): 2116- 2125
doi: 10.1038/sj.emboj.7600217
93 BUSINO L , MILLMAN S E , SCOTTO L et al. Fbxw7alpha- and GSK3-mediated degradation of p100 is a pro-survival mechanism in multiple myeloma[J]. Nat Cell Biol, 2012, 14:375- 385
doi: 10.1038/ncb2463
94 MA J , CHENG L , LIU H et al. Genistein down-regulates miR-223 expression in pancreatic cancer cells[J]. Curr Drug Targets, 2013, 14 (10): 1150- 1156
doi: 10.2174/13894501113149990187
95 MCGEARY R P , BENNETT A J , TRAN Q B et al. Suramin:clinical uses and structure-activity relationships[J]. Mini Rev Med Chem, 2008, 8 (13): 1384- 1394
doi: 10.2174/138955708786369573
96 GORELIK M , ORLICKY S , SARTORI M A et al. Inhibition of SCF ubiquitin ligases by engineered ubiquitin variants that target the Cul1 binding site on the Skp1-F-box interface[J]. Proc Natl Acad Sci U S A, 2016, 113 (13): 3527- 3532
doi: 10.1073/pnas.1519389113
97 KURZ T , OZLU N , RUDOLF F et al. The conserved protein DCN-1/Dcn1p is required for cullin neddylation in C. elegans and S. cerevisiae[J]. Nature, 2005, 435:1257- 1261
doi: 10.1038/nature03662
98 SCOTT D C , SVIDERSKIY V O , MONDA J K et al. Structure of a RING E3 trapped in action reveals ligation mechanism for the ubiquitin-like protein NEDD8[J]. Cell, 2014, 157 (7): 1671- 1684
doi: 10.1016/j.cell.2014.04.037
99 SARKARIA I S , PHAM D , GHOSSEIN R A et al. SCCRO expression correlates with invasive progression in bronchioloalveolar carcinoma[J]. Ann Thorac Surg, 2004, 78 (5): 1734- 1741
doi: 10.1016/j.athoracsur.2004.05.056
100 SARKARIA I , O-CHAROENRAT P , TALBOT S G et al. Squamous cell carcinoma related oncogene/DCUN1D1 is highly conserved and activated by amplification in squamous cell carcinomas[J]. Cancer Res, 2006, 66 (19): 9437- 9444
doi: 10.1158/0008-5472.CAN-06-2074
101 CALDERON-VILLALOBOS L I , TAN X , ZHENG N et al. Auxin perception——structural insights[J]. Cold Spring Harb Perspect Biol, 2010, 2 (7): a005546
doi: 10.1101/cshperspect.a005546
102 FRANKS M E , MACPHERSON G R , FIGG W D . Thalidomide[J]. Lancet, 2004, 363 (9423): 1802- 1811
doi: 10.1016/S0140-6736(04)16308-3
103 POLAKIS P . Wnt signaling and cancer[J]. Genes Dev, 2000, 14 (15): 1837- 1851
doi: 10.1101/gad.14.15.1837
104 ZHOU G B , CHEN S J , WANG Z Y et al. Back to the future of oridonin:again, compound from medicinal herb shows potent antileukemia efficacies in vitro and in vivo[J]. Cell Res, 2007, 17 (4): 274- 276
doi: 10.1038/cr.2007.21
105 GU S , CUI D , CHEN X et al. PROTACs:an emerging targeting technique for protein degradation in drug discovery[J]. Bioessays, 2018, 40 (4): e1700247
doi: 10.1002/bies.201700247
106 SAKAMOTO K M , KIM K B , KUMAGAI A et al. Protacs:chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation[J]. Proc Natl Acad Sci U S A, 2001, 98 (15): 8554- 8559
doi: 10.1073/pnas.141230798
107 SCHNEEKLOTH JS J R , FONSECA F N , KOLDOBSKIY M et al. Chemical genetic control of protein levels:selective in vivo targeted degradation[J]. J Am Chem Soc, 2004, 126 (12): 3748- 3754
doi: 10.1021/ja039025z
108 HINES J , GOUGH J D , CORSON T W et al. Posttranslational protein knockdown coupled to receptor tyrosine kinase activation with phosphoPROTACs[J]. Proc Natl Acad Sci U S A, 2013, 110 (22): 8942- 8947
doi: 10.1073/pnas.1217206110
109 SCHNEEKLOTH A R , PUCHEAULT M , TAE H S et al. Targeted intracellular protein degradation induced by a small molecule:En route to chemical proteomics[J]. Bioorg Med Chem Lett, 2008, 18 (22): 5904- 5908
doi: 10.1016/j.bmcl.2008.07.114
110 NEKLESA T , SNYDER L B , WILLARD R R et al. ARV-110:an oral androgen receptor PROTAC degrader for prostate cancer[J]. J Clin Oncol, 2019, 37 (7_suppl): 259
doi: 10.1200/JCO.2019.37.7_suppl.259
111 LU J , QIAN Y , ALTIERI M et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4[J]. Chem Biol, 2015, 22 (6): 755- 763
doi: 10.1016/j.chembiol.2015.05.009
112 WINTER G E , BUCKLEY D L , PAULK J et al. Drug development. Phthalimide conjugation as a strategy for in vivo target protein degradation[J]. Science, 2015, 348 (6241): 1376- 1381
doi: 10.1126/science.aab1433
113 BONDESON D P , MARES A , SMITH I E et al. Catalytic in vivo protein knockdown by small-molecule PROTACs[J]. Nat Chem Biol, 2015, 11 (8): 611- 617
doi: 10.1038/nchembio.1858
114 BUCKLEY D L , RAINA K , DARRICARRERE N et al. HaloPROTACS:use of small molecule PROTACs to induce degradation of halotag fusion proteins[J]. ACS Chem Biol, 2015, 10 (8): 1831- 1837
doi: 10.1021/acschembio.5b00442
115 ZENGERLE M , CHAN K H , CIULLI A . Selective small molecule induced degradation of the BET bromodomain protein BRD4[J]. ACS Chem Biol, 2015, 10 (8): 1770- 1777
doi: 10.1021/acschembio.5b00216
116 LEBRAUD H , WRIGHT D J , JOHNSON C N et al. Protein degradation by in-cell self-assembly of proteolysis targeting chimeras[J]. ACS Cent Sci, 2016, 2 (12): 927- 934
doi: 10.1021/acscentsci.6b00280
[1] 朱慧琦,应可净. 组织因子与肿瘤患者静脉血栓栓塞[J]. 浙江大学学报(医学版), 2020, 49(6): 772-778.
[2] 林翠翠,陈正云,王春艳,席咏梅. 基于脂质组学的子宫内膜异位症生物标志物研究进展[J]. 浙江大学学报(医学版), 2020, 49(6): 779-784.
[3] 李梦瑶,刘盼,柯越海,张雪. 放射性肺损伤中巨噬细胞作用机制的研究进展[J]. 浙江大学学报(医学版), 2020, 49(5): 623-628.
[4] 韩雪,蒋国军,石巧娟. 降血糖药对内皮祖细胞作用的研究进展[J]. 浙江大学学报(医学版), 2020, 49(5): 629-636.
[5] 段润平,许叶圣,郑利斌,姚玉峰. 病毒感染性眼病病原学诊断的研究进展[J]. 浙江大学学报(医学版), 2020, 49(5): 644-650.
[6] 吴唯,徐键. 正五聚蛋白3在多囊卵巢综合征中的作用研究进展[J]. 浙江大学学报(医学版), 2020, 49(5): 637-643.
[7] 徐清霖,楼国东,王甜甜,张力三. 发作性睡病的药物治疗进展[J]. 浙江大学学报(医学版), 2020, 49(4): 419-424.
[8] 蒋沛然,王志萍. 模式生物神经轴突再生的研究进展[J]. 浙江大学学报(医学版), 2020, 49(4): 500-507.
[9] 孔元梅,陈虹,梁黎,郑卯昵,方燕兰,王春林. 芳香化酶抑制剂联合生长激素治疗青春期身材矮小症男性患儿的临床研究[J]. 浙江大学学报(医学版), 2020, 49(3): 283-290.
[10] 王春林,梁黎. 第三代非甾体类芳香化酶抑制剂在儿科内分泌临床应用的再认识[J]. 浙江大学学报(医学版), 2020, 49(3): 275-282.
[11] 于冰青,聂敏,伍学焱,茅江峰,王曦,马婉璐,季文,黄奇彬,张睿. 来曲唑有效提高性发育异常男性患儿的睾酮水平[J]. 浙江大学学报(医学版), 2020, 49(3): 297-301.
[12] 徐德,陆文丽,王雪晴,王俊祺,谢轶雯,董治亚,王伟. 来曲唑治疗纤维性骨营养不良综合征女性患儿外周性性早熟疗效观察[J]. 浙江大学学报(医学版), 2020, 49(3): 291-296.
[13] 王倩,张书乐,马雪,李桂梅,王增敏,王凤雪. 来曲唑对21-羟化酶缺乏型先天性肾上腺皮质增生症患儿生长发育的影响[J]. 浙江大学学报(医学版), 2020, 49(3): 302-307.
[14] 李燕虹,杜敏联,马华梅,陈秋莉,陈红珊,张军. 来曲唑改善青春期特发性身材矮小症男性患儿成年身高的疗效评价[J]. 浙江大学学报(医学版), 2020, 49(3): 308-314.
[15] 赵维霞,邹炜. 神经元树突形态建成分子机制的研究进展[J]. 浙江大学学报(医学版), 2020, 49(1): 90-99.