RNA splicing factor RBFOX2 is a key factor in the progression of cancer and cardiomyopathy

Jinze Shen , Jianqiao Shentu , Chenming Zhong , Qiankai Huang , Shiwei Duan

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (9) : e1788

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (9) : e1788 DOI: 10.1002/ctm2.1788
REVIEW

RNA splicing factor RBFOX2 is a key factor in the progression of cancer and cardiomyopathy

Author information +
History +
PDF

Abstract

Background: Alternative splicing of pre-mRNA is a fundamental regulatory process in multicellular eukaryotes, significantly contributing to the diversification of the human proteome. RNA-binding fox-1 homologue 2 (RBFOX2), a member of the evolutionarily conserved RBFOX family, has emerged as a critical splicing regulator, playing a pivotal role in the alternative splicing of pre-mRNA. This review provides a comprehensive analysis of RBFOX2, elucidating its splicing activity through direct and indirect binding mechanisms. RBFOX2 exerts substantial influence over the alternative splicing of numerous transcripts, thereby shaping essential cellular processes such as differentiation and development.

Main body of the abstract: Dysregulation of RBFOX2-mediated alternative splicing has been closely linked to a spectrum of cardiovascular diseases and malignant tumours, underscoring its potential as a therapeutic target. Despite significant progress, current research faces notable challenges. The complete structural characterisation of RBFOX2 remains elusive, limiting in-depth exploration beyond its RNA-recognition motif. Furthermore, the scarcity of studies focusing on RBFOX2-targeting drugs poses a hindrance to translating research findings into clinical applications.

Conclusion: This review critically assesses the existing body of knowledge on RBFOX2, highlighting research gaps and limitations. By delineating these areas, this analysis not only serves as a foundational reference for future studies but also provides strategic insights for bridging these gaps. Addressing these challenges will be instrumental in unlocking the full therapeutic potential of RBFOX2, paving the way for innovative and effective treatments in various diseases.

Keywords

alternative splicing / pre-mRNA / RBFOX2 / therapeutic targeting

Cite this article

Download citation ▾
Jinze Shen, Jianqiao Shentu, Chenming Zhong, Qiankai Huang, Shiwei Duan. RNA splicing factor RBFOX2 is a key factor in the progression of cancer and cardiomyopathy. Clinical and Translational Medicine, 2024, 14(9): e1788 DOI:10.1002/ctm2.1788

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

He S, Valkov E, Cheloufi S, Murn J. The nexus between RNA-binding proteins and their effectors. Nat Rev Genet. 2023; 24(5): 276-294.

[2]

Corley M, Burns MC, Yeo GW. How RNA-binding proteins interact with RNA: molecules and mechanisms. Mol Cell. 2020; 78(1): 9-29.

[3]

Xu W, Biswas J, Singer RH, Rosbash M. Targeted RNA editing: novel tools to study post-transcriptional regulation. Mol Cell. 2022; 82(2): 389-403.

[4]

Conboy JG. Developmental regulation of RNA processing by Rbfox proteins. Wiley Interdiscip Rev RNA. 2017; 8(2).

[5]

Jin Y, Suzuki H, Maegawa S, et al. A vertebrate RNA-binding protein Fox-1 regulates tissue-specific splicing via the pentanucleotide GCAUG. Embo J. 2003; 22(4): 905-912.

[6]

van der Werf I, Mondala PK, Steel SK, et al. Detection and targeting of splicing deregulation in pediatric acute myeloid leukemia stem cells. Cell Rep Med. 2023; 4(3): 100962.

[7]

Naro C, De Musso M, Delle Monache F, Panzeri V, de la Grange P, Sette C. The oncogenic kinase NEK2 regulates an RBFOX2-dependent pro-mesenchymal splicing program in triple-negative breast cancer cells. J Exp Clin Cancer Res. 2021; 40(1): 397.

[8]

Hu J, Gao C, Wei C, et al. RBFox2-miR-34a-Jph2 axis contributes to cardiac decompensation during heart failure. Proc Natl Acad Sci U S A. 2019; 116(13): 6172-6180.

[9]

Danan-Gotthold M, Golan-Gerstl R, Eisenberg E, Meir K, Karni R, Levanon EY. Identification of recurrent regulated alternative splicing events across human solid tumors. Nucleic Acids Res. 2015; 43(10): 5130-5144.

[10]

Cao J, Verma SK, Jaworski E, et al. RBFOX2 is critical for maintaining alternative polyadenylation patterns and mitochondrial health in rat myoblasts. Cell Rep. 2021; 37(5): 109910.

[11]

Norris JD, Fan D, Sherk A, McDonnell DP. A negative coregulator for the human ER. Mol Endocrinol. 2002; 16(3): 459-468.

[12]

Auweter SD, Fasan R, Reymond L, et al. Molecular basis of RNA recognition by the human alternative splicing factor Fox-1. Embo J. 2006; 25(1): 163-173.

[13]

Kuroyanagi H. Fox-1 family of RNA-binding proteins. Cell Mol Life Sci. 2009; 66(24): 3895-3907.

[14]

Lambert N, Robertson A, Jangi M, McGeary S, Sharp PA, Burge CB. RNA Bind-n-Seq: quantitative assessment of the sequence and structural binding specificity of RNA binding proteins. Mol Cell. 2014; 54(5): 887-900.

[15]

Damianov A, Ying Y, Lin CH, et al. Rbfox proteins regulate splicing as part of a large multiprotein complex LASR. Cell. 2016; 165(3): 606-619.

[16]

Meng EC, Goddard TD, Pettersen EF, et al. UCSF ChimeraX: tools for structure building and analysis. Protein Sci. 2023; 32(11): e4792.

[17]

Kim KK, Yang Y, Zhu J, Adelstein RS, Kawamoto S. Rbfox3 controls the biogenesis of a subset of microRNAs. Nat Struct Mol Biol. 2014; 21(10): 901-910.

[18]

Park C, Choi S, Kim YE, et al. Stress granules contain Rbfox2 with cell cycle-related mRNAs. Sci Rep. 2017; 7(1): 11211.

[19]

Kedersha N, Stoecklin G, Ayodele M, et al. Stress granules and processing bodies are dynamically linked sites of mRNP remodeling. J Cell Biol. 2005; 169(6): 871-884.

[20]

Lee JA, Damianov A, Lin CH, et al. Cytoplasmic Rbfox1 regulates the expression of synaptic and autism-related genes. Neuron. 2016; 89(1): 113-128.

[21]

Nakahata S, Kawamoto S. Tissue-dependent isoforms of mammalian Fox-1 homologs are associated with tissue-specific splicing activities. Nucleic Acids Res. 2005; 33(7): 2078-2089.

[22]

Damianov A, Black DL. Autoregulation of Fox protein expression to produce dominant negative splicing factors. RNA. 2010; 16(2): 405-416.

[23]

Choi S, Cho N, Kim KK. Non-canonical splice junction processing increases the diversity of RBFOX2 splicing isoforms. Int J Biochem Cell Biol. 2022; 144: 106172.

[24]

Cho N, Kim JO, Lee S, et al. Alternative splicing induces cytoplasmic localization of RBFOX2 protein in calcific tendinopathy. Exp Mol Pathol. 2019; 109: 36-41.

[25]

Wenzel M, Schüle M, Casanovas S, Strand D, Strand S, Winter J. Identification of a classic nuclear localization signal at the N terminus that regulates the subcellular localization of Rbfox2 isoforms during differentiation of NMuMG and P19 cells. FEBS Lett. 2016; 590(24): 4453-4460.

[26]

Misra C, Bangru S, Lin F, et al. Aberrant expression of a non-muscle RBFOX2 isoform triggers cardiac conduction defects in myotonic dystrophy. Dev Cell. 2020; 52(6): 748-763. e6.

[27]

Ying Y, Wang XJ, Vuong CK, Lin CH, Damianov A, Black DL. Splicing activation by Rbfox requires self-aggregation through its tyrosine-rich domain. Cell. 2017; 170(2): 312-323. e10.

[28]

Wei C, Xiao R, Chen L, et al. RBFox2 binds nascent RNA to globally regulate polycomb complex 2 targeting in mammalian genomes. Mol Cell. 2016; 62(6): 875-889.

[29]

Dou X, Xiao Y, Shen C, et al. RBFOX2 recognizes N(6)-methyladenosine to suppress transcription and block myeloid leukaemia differentiation. Nat Cell Biol. 2023; 25(9): 1359-1368.

[30]

Wu R, Sun C, Chen X, et al. NSUN5/TET2-directed chromatin-associated RNA modification of 5-methylcytosine to 5-hydroxymethylcytosine governs glioma immune evasion. Proc Natl Acad Sci U S A. 2024; 121(14): e2321611121.

[31]

Davidovich C, Zheng L, Goodrich KJ, Cech TR. Promiscuous RNA binding by Polycomb repressive complex 2. Nat Struct Mol Biol. 2013; 20(11): 1250-1257.

[32]

Liu J, Gao M, He J, et al. The RNA m(6)A reader YTHDC1 silences retrotransposons and guards ES cell identity. Nature. 2021; 591(7849): 322-326.

[33]

Marasco LE, Kornblihtt AR. The physiology of alternative splicing. Nat Rev Mol Cell Biol. 2023; 24(4): 242-254.

[34]

Kim KK, Adelstein RS, Kawamoto S. Isoform-specific proteasomal degradation of Rbfox3 during chicken embryonic development. Biochem Biophys Res Commun. 2014; 450(4): 1662-1667.

[35]

Gehman LT, Meera P, Stoilov P, et al. The splicing regulator Rbfox2 is required for both cerebellar development and mature motor function. Genes Dev. 2012; 26(5): 445-460.

[36]

Zhou D, Couture S, Scott MS, Elela SA. RBFOX2 alters splicing outcome in distinct binding modes with multiple protein partners. Nucleic Acids Res. 2021; 49(14): 8370-8383.

[37]

Zhou Y, Fan J, Zhu H, et al. Aberrant splicing induced by dysregulated Rbfox2 produces enhanced function of Ca(V)1.2 calcium channel and vascular myogenic tone in hypertension. Hypertension. 2017; 70(6): 1183-1192.

[38]

Gu M, Zhao Y, Wang H, et al. Suppression of RBFox2 by multiple miRNAs in pressure overload-induced heart failure. Int J Mol Sci. 2023; 24(2): 1283.

[39]

Vecellio Reane D, Cerqua C, Sacconi S, Salviati L, Trevisson E, Raffaello A. The splicing of the mitochondrial calcium uniporter genuine activator MICU1 is driven by RBFOX2 splicing factor during myogenic differentiation. Int J Mol Sci. 2022; 23(5): 2517.

[40]

Cao J, Routh AL, Kuyumcu-Martinez MN. Nanopore sequencing reveals full-length Tropomyosin 1 isoforms and their regulation by RNA-binding proteins during rat heart development. J Cell Mol Med. 2021; 25(17): 8352-8362.

[41]

Wan X, Belanger K, Widen SG, Kuyumcu-Martinez MN, Garg NJ. Genes of the cGMP-PKG-Ca(2+) signaling pathway are alternatively spliced in cardiomyopathy: role of RBFOX2. Biochim Biophys Acta Mol Basis Dis. 2020; 1866(3): 165620.

[42]

Cooper AM, Nutter CA, Kuyumcu-Martinez MN, Wright CW. Alternative splicing of the aryl hydrocarbon receptor nuclear translocator (ARNT) is regulated by RBFOX2 in lymphoid malignancies. Mol Cell Biol. 2022; 42(5): e0050321.

[43]

Kim JH, Jeong K, Li J, et al. SON drives oncogenic RNA splicing in glioblastoma by regulating PTBP1/PTBP2 switching and RBFOX2 activity. Nat Commun. 2021; 12(1): 5551.

[44]

Saulnier O, Guedri-Idjouadiene K, Aynaud MM, et al. ERG transcription factors have a splicing regulatory function involving RBFOX2 that is altered in the EWS-FLI1 oncogenic fusion. Nucleic Acids Res. 2021; 49(9): 5038-5056.

[45]

Zhang J, Chen S, Wei S, et al. CircRAPGEF5 interacts with RBFOX2 to confer ferroptosis resistance by modulating alternative splicing of TFRC in endometrial cancer. Redox Biol. 2022; 57: 102493.

[46]

Lu B, Li X, Miao W, et al. LncRNA ZFAS1 promotes laryngeal cancer progression through RBFOX2-mediated MENA alternative splicing. Environ Toxicol. 2023; 38(3): 522-533.

[47]

Londero M, Gallo A, Cattaneo C, et al. NF-YAl drives EMT in Claudin(low) tumours. Cell Death Dis. 2023; 14(1): 65.

[48]

Choi S, Lee HS, Cho N, et al. RBFOX2-regulated TEAD1 alternative splicing plays a pivotal role in Hippo-YAP signaling. Nucleic Acids Res. 2022; 50(15): 8658-8673.

[49]

Lu Y, Xu W, Ji J, et al. Alternative splicing of the cell fate determinant Numb in hepatocellular carcinoma. Hepatology. 2015; 62(4): 1122-1131.

[50]

Nakura T, Ozoe A, Narita Y, et al. Rbfox2 mediates exon 11 inclusion in insulin receptor pre-mRNA splicing in hepatoma cells. Biochimie. 2021; 187: 25-32.

[51]

Jbara A, Lin KT, Stossel C, et al. RBFOX2 modulates a metastatic signature of alternative splicing in pancreatic cancer. Nature. 2023; 617(7959): 147-153.

[52]

Paterson HAB, Yu S, Artigas N, et al. Liver RBFOX2 regulates cholesterol homeostasis via Scarb1 alternative splicing in mice. Nat Metab. 2022; 4(12): 1812-1829.

[53]

Seth P, Yeowell HN. Fox-2 protein regulates the alternative splicing of scleroderma-associated lysyl hydroxylase 2 messenger RNA. Arthritis Rheum. 2010; 62(4): 1167-1175.

[54]

Verma SK, Deshmukh V, Nutter CA, et al. Rbfox2 function in RNA metabolism is impaired in hypoplastic left heart syndrome patient hearts. Sci Rep. 2016; 6: 30896.

[55]

Supadmanaba IGP, Mantini G, Randazzo O, et al. Interrelationship between miRNA and splicing factors in pancreatic ductal adenocarcinoma. Epigenetics. 2022; 17(4): 381-404.

[56]

Li P, Qin D, Chen T, et al. Dysregulated Rbfox2 produces aberrant splicing of Ca(V)1.2 calcium channel in diabetes-induced cardiac hypertrophy. Cardiovasc Diabetol. 2023; 22(1): 168.

[57]

Gallagher TL, Arribere JA, Geurts PA, et al. Rbfox-regulated alternative splicing is critical for zebrafish cardiac and skeletal muscle functions. Dev Biol. 2011; 359(2): 251-261.

[58]

Yin QF, Yang L, Zhang Y, et al. Long noncoding RNAs with snoRNA ends. Mol Cell. 2012; 48(2): 219-230.

[59]

Yang Y, Lee GC, Nakagaki-Silva E, et al. Cell-type specific regulator RBPMS switches alternative splicing via higher-order oligomerization and heterotypic interactions with other splicing regulators. Nucleic Acids Res. 2023; 51(18): 9961-9982.

[60]

Weischenfeldt J, Waage J, Tian G, et al. Mammalian tissues defective in nonsense-mediated mRNA decay display highly aberrant splicing patterns. Genome Biol. 2012; 13(5): R35.

[61]

Jangi M, Boutz PL, Paul P, Sharp PA. Rbfox2 controls autoregulation in RNA-binding protein networks. Genes Dev. 2014; 28(6): 637-651.

[62]

Han J, An O, Ren X, et al. Multilayered control of splicing regulatory networks by DAP3 leads to widespread alternative splicing changes in cancer. Nat Commun. 2022; 13(1): 1793.

[63]

García-Moreno JF, Romão L. Perspective in alternative splicing coupled to nonsense-mediated mRNA decay. Int J Mol Sci. 2020; 21(24): 9424.

[64]

Yang G, Huang SC, Wu JY. Regulated Fox-2 isoform expression mediates protein 4.1R splicing during erythroid differentiation. Blood. 2008; 111(1): 392-401.

[65]

Braeutigam C, Rago L, Rolke A, Waldmeier L, Christofori G, Winter J. The RNA-binding protein Rbfox2: an essential regulator of EMT-driven alternative splicing and a mediator of cellular invasion. Oncogene. 2014; 33(9): 1082-1092.

[66]

Van Nostrand EL, Freese P, Pratt GA, et al. A large-scale binding and functional map of human RNA-binding proteins. Nature. 2020; 583(7818): 711-719.

[67]

Singh RK, Kolonin AM, Fiorotto ML, Cooper TA. Rbfox-splicing factors maintain skeletal muscle mass by regulating Calpain3 and proteostasis. Cell Rep. 2018; 24(1): 197-208.

[68]

Wei C, Qiu J, Zhou Y, et al. Repression of the central splicing regulator RBFox2 is functionally linked to pressure overload-induced heart failure. Cell Rep. 2015; 10(9): 1521-1533.

[69]

Homsy J, Zaidi S, Shen Y, et al. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies. Science. 2015; 350(6265): 1262-1266.

[70]

Krach F, Wheeler EC, Regensburger M, et al. Aberrant NOVA1 function disrupts alternative splicing in early stages of amyotrophic lateral sclerosis. Acta Neuropathol. 2022; 144(3): 413-435.

[71]

Leong DP, Joseph PG, McKee M, et al. Reducing the global burden of cardiovascular disease, part 2: prevention and treatment of cardiovascular disease. Circ Res. 2017; 121(6): 695-710.

[72]

McKean DM, Homsy J, Wakimoto H, et al. Loss of RNA expression and allele-specific expression associated with congenital heart disease. Nat Commun. 2016; 7: 12824.

[73]

Nutter CA, Jaworski EA, Verma SK, et al. Dysregulation of RBFOX2 is an early event in cardiac pathogenesis of diabetes. Cell Rep. 2016; 15(10): 2200-2213.

[74]

Ajoolabady A, Chiong M, Lavandero S, Klionsky DJ, Ren J. Mitophagy in cardiovascular diseases: molecular mechanisms, pathogenesis, and treatment. Trends Mol Med. 2022; 28(10): 836-849.

[75]

Walsh R, Offerhaus JA, Tadros R, Bezzina CR. Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol. 2022; 19(3): 151-167.

[76]

Reynolds JO, Chiang DY, Wang W, et al. Junctophilin-2 is necessary for T-tubule maturation during mouse heart development. Cardiovasc Res. 2013; 100(1): 44-53.

[77]

Hofmann F, Flockerzi V, Kahl S, Wegener JW. L-type CaV1.2 calcium channels: from in vitro findings to in vivo function. Physiol Rev. 2014; 94(1): 303-326.

[78]

Xue GL, Li DS, Wang ZY, et al. Interleukin-17 upregulation participates in the pathogenesis of heart failure in mice via NF-kappaB-dependent suppression of SERCA2a and Cav1.2 expression. Acta Pharmacol Sin. 2021; 42(11): 1780-1789.

[79]

Zhao Z, Wen H, Fefelova N, et al. Revisiting the ionic mechanisms of early afterdepolarizations in cardiomyocytes: predominant by Ca waves or Ca currents? Am J Physiol Heart Circ Physiol. 2012; 302(8): H1636-H1644.

[80]

Tang ZZ, Hong X, Wang J, Soong TW. Signature combinatorial splicing profiles of rat cardiac-and smooth-muscle Cav1.2 channels with distinct biophysical properties. Cell Calcium. 2007; 41(5): 417-428.

[81]

Wang J, Thio SS, Yang SS, et al. Splice variant specific modulation of CaV1.2 calcium channel by galectin-1 regulates arterial constriction. Circ Res. 2011; 109(11): 1250-1258.

[82]

Fici P, Gallerani G, Morel AP, et al. Splicing factor ratio as an index of epithelial-mesenchymal transition and tumor aggressiveness in breast cancer. Oncotarget. 2017; 8(2): 2423-2436.

[83]

Nystoriak MA, Murakami K, Penar PL, Wellman GC. Ca(v)1.2 splice variant with exon 9* is critical for regulation of cerebral artery diameter. Am J Physiol Heart Circ Physiol. 2009; 297(5): H1820-1828.

[84]

Wu W, Ziemann M, Huynh K, et al. Activation of Hippo signaling pathway mediates mitochondria dysfunction and dilated cardiomyopathy in mice. Theranostics. 2021; 11(18): 8993-9008.

[85]

Zhuang L, Jia K, Chen C, et al. DYRK1B–STAT3 drives cardiac hypertrophy and heart failure by impairing mitochondrial bioenergetics. Circulation. 2022; 145(11): 829-846.

[86]

Graham BH, Waymire KG, Cottrell B, Trounce IA, MacGregor GR, Wallace DC. A mouse model for mitochondrial myopathy and cardiomyopathy resulting from a deficiency in the heart/muscle isoform of the adenine nucleotide translocator. Nat Genet. 1997; 16(3): 226-234.

[87]

Fan M, Zhang J, Tsai CW, et al. Structure and mechanism of the mitochondrial Ca(2+) uniporter holocomplex. Nature. 2020; 582(7810): 129-133.

[88]

Singh RK, Xia Z, Bland CS, et al. Rbfox2-coordinated alternative splicing of Mef2d and Rock2 controls myoblast fusion during myogenesis. Mol Cell. 2014; 55(4): 592-603.

[89]

Runfola V, Sebastian S, Dilworth FJ, Gabellini D. Rbfox proteins regulate tissue-specific alternative splicing of Mef2D required for muscle differentiation. J Cell Sci. 2015; 128(4): 631-637.

[90]

Verma SK, Deshmukh V, Liu P, et al. Reactivation of fetal splicing programs in diabetic hearts is mediated by protein kinase C signaling. J Biol Chem. 2013; 288(49): 35372-35386.

[91]

Liu L, Kryvokhyzha D, Rippe C, et al. Myocardin regulates exon usage in smooth muscle cells through induction of splicing regulatory factors. Cell Mol Life Sci. 2022; 79(8): 459.

[92]

Gazzara MR, Mallory MJ, Roytenberg R, et al. Ancient antagonism between CELF and RBFOX families tunes mRNA splicing outcomes. Genome Res. 2017; 27(8): 1360-1370.

[93]

Dasgupta T, Ladd AN. The importance of CELF control: molecular and biological roles of the CUG-BP, Elav-like family of RNA-binding proteins. Wiley Interdiscip Rev RNA. 2012; 3(1): 104-121.

[94]

Ferlay J, Colombet M, Soerjomataram I, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer. 2018; 103: 356-387.

[95]

van den Bosch QCC, Nguyen JQN, Brands T, et al. FOXD1 is a transcription factor important for uveal melanocyte development and associated with high-risk uveal melanoma. Cancers (Basel). 2022; 14(15): 3668.

[96]

Xie S, Li Z, Zhong Y, et al. RBFOX2 confers tumor growth by PI3K/AKT and MAPK signaling in gastric cancer. Eur J Cancer Prev. 2023; 32(5): 468-477.

[97]

Choi S, Sa M, Cho N, Kim KK, Park SH. Rbfox2 dissociation from stress granules suppresses cancer progression. Exp Mol Med. 2019; 51(4): 1-12.

[98]

Ahuja N, Ashok C, Natua S, et al. Hypoxia-induced TGF-β-RBFOX2-ESRP1 axis regulates human MENA alternative splicing and promotes EMT in breast cancer. NAR Cancer. 2020; 2(3): zcaa021.

[99]

Mochizuki Y, Funayama R, Shirota M, et al. Alternative microexon splicing by RBFOX2 and PTBP1 is associated with metastasis in colorectal cancer. Int J Cancer. 2021; 149(10): 1787-1800.

[100]

Maurin M, Ranjouri M, Megino-Luque C, et al. RBFOX2 deregulation promotes pancreatic cancer progression and metastasis through alternative splicing. Nat Commun. 2023; 14(1): 8444.

[101]

Ahuja N, Ashok C, Natua S, et al. Hypoxia-induced TGF-beta-RBFOX2-ESRP1 axis regulates human MENA alternative splicing and promotes EMT in breast cancer. NAR Cancer. 2020; 2(3): zcaa021.

[102]

Ou X, Zhou X, Li J, et al. p53-Induced LINC00893 regulates RBFOX2 stability to suppress gastric cancer progression. Front Cell Dev Biol. 2021; 9: 796451.

[103]

Li S, Chen Y, Xie Y, et al. FBXO7 confers mesenchymal properties and chemoresistance in glioblastoma by controlling Rbfox2-mediated alternative splicing. Adv Sci. 2023; 10(33): e2303561.

[104]

Gordon MA, Babbs B, Cochrane DR, Bitler BG, Richer JK. The long non-coding RNA MALAT1 promotes ovarian cancer progression by regulating RBFOX2-mediated alternative splicing. Mol Carcinog. 2019; 58(2): 196-205.

[105]

Chen Y, Zubovic L, Yang F, et al. Rbfox proteins regulate microRNA biogenesis by sequence-specific binding to their precursors and target downstream Dicer. Nucleic Acids Res. 2016; 44(9): 4381-4395.

[106]

Robertson AG, Rendina LM. Gadolinium theranostics for the diagnosis and treatment of cancer. Chem Soc Rev. 2021; 50(7): 4231-4244.

[107]

Polyak K. Heterogeneity in breast cancer. J Clin Invest. 2011; 121(10): 3786-3788.

[108]

Marusyk A, Polyak K. Tumor heterogeneity: causes and consequences. Biochim Biophys Acta. 2010; 1805(1): 105-117.

[109]

Innocenti M, Zucconi A, Disanza A, et al. Abi1 is essential for the formation and activation of a WAVE2 signalling complex. Nat Cell Biol. 2004; 6(4): 319-327.

[110]

Zhu G, Herlyn M, Yang X. TRIM15 and CYLD regulate ERK activation via lysine-63-linked polyubiquitination. Nat Cell Biol. 2021; 23(9): 978-991.

[111]

Tracz M, Bialek W. Beyond K48 and K63: non-canonical protein ubiquitination. Cell Mol Biol Lett. 2021; 26(1): 1.

[112]

Long L, Assaraf YG, Lei ZN, et al. Genetic biomarkers of drug resistance: a compass of prognosis and targeted therapy in acute myeloid leukemia. Drug Resist Updat. 2020; 52: 100703.

[113]

Bazvand F, Mahdizad Z, Mohammadi N, et al. Tamoxifen retinopathy. Surv Ophthalmol. 2023; 68(4): 628-640.

[114]

Johansson A, Dar H, van ’t Veer LJ, et al. Twenty-year benefit from adjuvant goserelin and tamoxifen in premenopausal patients with breast cancer in a controlled randomized clinical trial. J Clin Oncol. 2022; 40(35): 4071-4082.

[115]

Hong S, Chang J, Jeong K, Lee W. Raloxifene as a treatment option for viral infections. J Microbiol. 2021; 59(2): 124-131.

[116]

Sledge GW Jr, Toi M, Neven P, et al. The effect of abemaciclib plus fulvestrant on overall survival in hormone receptor-positive, ERBB2-negative breast cancer that progressed on endocrine therapy-MONARCH 2: a randomized clinical trial. JAMA Oncol. 2020; 6(1): 116-124.

[117]

Desai K, McManus JM, Sharifi N. Hormonal therapy for prostate cancer. Endocr Rev. 2021; 42(3): 354-373.

[118]

Arya AD, Wilson DI, Baralle D, Raponi M. RBFOX2 protein domains and cellular activities. Biochem Soc Trans. 2014; 42(4): 1180-1183.

[119]

Crews LA, Ma W, Ladel L, et al. Reversal of malignant ADAR1 splice isoform switching with Rebecsinib. Cell Stem Cell. 2023; 30(3): 250-263. e6.

[120]

Julio AR, Backus KM. New approaches to target RNA binding proteins. Curr Opin Chem Biol. 2021; 62: 13-23.

[121]

Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022; 21(3): 181-200.

[122]

Ghidini A, Cléry A, Halloy F, Allain FHT, Hall J. RNA-PROTACs: degraders of RNA-binding proteins. Angew Chem Int Ed Engl. 2021; 60(6): 3163-3169.

[123]

Xu Y, Yuan Y, Fu DQ, et al. The aptamer-based RNA-PROTAC. Bioorg Med Chem. 2023; 86: 117299.

[124]

Chuang SH, Lee YE, Huang LYL, et al. Discovery of T-1101 tosylate as a first-in-class clinical candidate for Hec1/Nek2 inhibition in cancer therapy. Eur J Med Chem. 2020; 191: 112118.

[125]

Xi JB, Fang YF, Frett B, et al. Structure-based design and synthesis of imidazo[1, 2-a]pyridine derivatives as novel and potent Nek2 inhibitors with in vitro and in vivo antitumor activities. Eur J Med Chem. 2017; 126: 1083-1106.

[126]

Wang H, Chen Y, Gu X, et al. Design, synthesis, and structure activity relationship (SAR) studies of novel imidazo[1, 2-a] pyridine derivatives as Nek2 inhibitors. Bioorg Med Chem. 2020; 28(23): 115775.

[127]

Zhang C, Zhang Z, Castle J, et al. Defining the regulatory network of the tissue-specific splicing factors Fox-1 and Fox-2. Genes Dev. 2008; 22(18): 2550-2563.

[128]

Cibi DM, Mia MM, Guna Shekeran S, et al. Neural crest-specific deletion of Rbfox2 in mice leads to craniofacial abnormalities including cleft palate. Elife. 2019; 8: e45418.

RIGHTS & PERMISSIONS

2024 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

163

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/