Skip to main content
Top
Published in: Molecular Cancer 1/2024

Open Access 01-12-2024 | Prostate Cancer | Research

Co-transcriptional R-loops-mediated epigenetic regulation drives growth retardation and docetaxel chemosensitivity enhancement in advanced prostate cancer

Authors: Yufan Ying, Yuqing Wu, Fenghao Zhang, Yijie Tang, Jiahe Yi, Xueyou Ma, Jiangfeng Li, Danni Chen, Xiao Wang, Xiaoyan Liu, Ben Liu, Jindan Luo, Xiangyi Zheng, Liping Xie

Published in: Molecular Cancer | Issue 1/2024

Login to get access

Abstract

R-loops are prevalent three-stranded nucleic acid structures, comprising a DNA-RNA hybrid and a displaced single-stranded DNA, that frequently form during transcription and may be attributed to genomic stability and gene expression regulation. It was recently discovered that RNA modification contributes to maintain the stability of R-loops such as N6-methyladenosine (m6A). Yet, m6A-modified R-loops in regulating gene transcription remains poorly understood. Here, we demonstrated that insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) recognize R-loops in an m6A-dependent way. Consequently, IGF2BPs overexpression leads to increased overall R-loop levels, cell migration inhibition, and cell growth retardation in prostate cancer (PCa) via precluding the binding of DNA methyltransferase 1(DNMT1) to semaphorin 3 F (SEMA3F) promoters. Moreover, the K homology (KH) domains of IGF2BPs are required for their recognition of m6A-containing R-loops and are required for tumor suppressor functions. Overexpression of SEMA3F markedly enhanced docetaxel chemosensitivity in prostate cancer via regulating Hippo pathway. Our findings point to a distinct R-loop resolution pathway mediated by IGF2BPs, emphasizing the functional importance of IGF2BPs as epigenetic R-loop readers in transcriptional genetic regulation and cancer biology.
The manuscript summarizes the new role of N6-methyladenosine in epigenetic regulation, we introduce the distinct R-loop resolution mediated by IGF2BP proteins in an m6A-dependent way, which probably lead to the growth retardation and docetaxel chemotherapy resistance in prostate cancer. Moreover, our findings first emphasized the functional importance of IGF2BPs as epigenetic R-loop readers in transcriptional genetic regulation and cancer biology. In addition, our research provides a novel RBM15/IGF2BPs/DNMT1 trans-omics regulation m6A axis, indicating the new crosstalk between RNA m6A methylation and DNA methylation in prostate cancer.

Graphical Abstract

Appendix
Available only for authorised users
Literature
2.
go back to reference Garcia-Muse T, Aguilera A. R loops: from physiological to pathological roles. Cell. 2019;179(3):604–18.CrossRefPubMed Garcia-Muse T, Aguilera A. R loops: from physiological to pathological roles. Cell. 2019;179(3):604–18.CrossRefPubMed
4.
go back to reference Aguilera A, Garcia-Muse T. R loops: from transcription byproducts to threats to genome stability. Mol Cell. 2012;46(2):115–24.CrossRefPubMed Aguilera A, Garcia-Muse T. R loops: from transcription byproducts to threats to genome stability. Mol Cell. 2012;46(2):115–24.CrossRefPubMed
5.
go back to reference Santos-Pereira JM, Aguilera A. R loops: new modulators of genome dynamics and function. Nat Rev Genet. 2015;16(10):583–97.CrossRefPubMed Santos-Pereira JM, Aguilera A. R loops: new modulators of genome dynamics and function. Nat Rev Genet. 2015;16(10):583–97.CrossRefPubMed
6.
go back to reference Ginno PA, Lott PL, Christensen HC, Korf I, Chedin F. R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters. Mol Cell. 2012;45(6):814–25.CrossRefPubMedPubMedCentral Ginno PA, Lott PL, Christensen HC, Korf I, Chedin F. R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters. Mol Cell. 2012;45(6):814–25.CrossRefPubMedPubMedCentral
7.
go back to reference Grunseich C, Wang IX, Watts JA, Burdick JT, Guber RD, Zhu Z, et al. Senataxin Mutation reveals how R-Loops promote transcription by blocking DNA methylation at Gene promoters. Mol Cell. 2018;69(3):426-37 e7.CrossRefPubMedPubMedCentral Grunseich C, Wang IX, Watts JA, Burdick JT, Guber RD, Zhu Z, et al. Senataxin Mutation reveals how R-Loops promote transcription by blocking DNA methylation at Gene promoters. Mol Cell. 2018;69(3):426-37 e7.CrossRefPubMedPubMedCentral
8.
go back to reference Arab K, Karaulanov E, Musheev M, Trnka P, Schafer A, Grummt I, et al. GADD45A binds R-loops and recruits TET1 to CpG island promoters. Nat Genet. 2019;51(2):217–23.CrossRefPubMedPubMedCentral Arab K, Karaulanov E, Musheev M, Trnka P, Schafer A, Grummt I, et al. GADD45A binds R-loops and recruits TET1 to CpG island promoters. Nat Genet. 2019;51(2):217–23.CrossRefPubMedPubMedCentral
9.
go back to reference Batista PJ, Molinie B, Wang J, Qu K, Zhang J, Li L, et al. M(6)a RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014;15(6):707–19.CrossRefPubMedPubMedCentral Batista PJ, Molinie B, Wang J, Qu K, Zhang J, Li L, et al. M(6)a RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014;15(6):707–19.CrossRefPubMedPubMedCentral
10.
go back to reference Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)a methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–45.CrossRefPubMedPubMedCentral Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)a methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–45.CrossRefPubMedPubMedCentral
11.
go back to reference Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.CrossRefPubMed Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.CrossRefPubMed
12.
go back to reference Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, et al. RNA m(6)a methylation regulates the ultraviolet-induced DNA damage response. Nature. 2017;543(7646):573–6.CrossRefPubMedPubMedCentral Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, et al. RNA m(6)a methylation regulates the ultraviolet-induced DNA damage response. Nature. 2017;543(7646):573–6.CrossRefPubMedPubMedCentral
13.
go back to reference Abakir A, Giles TC, Cristini A, Foster JM, Dai N, Starczak M, et al. N(6)-methyladenosine regulates the stability of RNA:DNA hybrids in human cells. Nat Genet. 2020;52(1):48–55.CrossRefPubMed Abakir A, Giles TC, Cristini A, Foster JM, Dai N, Starczak M, et al. N(6)-methyladenosine regulates the stability of RNA:DNA hybrids in human cells. Nat Genet. 2020;52(1):48–55.CrossRefPubMed
14.
go back to reference Kang HJ, Cheon NY, Park H, Jeong GW, Ye BJ, Yoo EJ, et al. TonEBP recognizes R-loops and initiates m6A RNA methylation for R-loop resolution. Nucleic Acids Res. 2021;49(1):269–84.CrossRefPubMed Kang HJ, Cheon NY, Park H, Jeong GW, Ye BJ, Yoo EJ, et al. TonEBP recognizes R-loops and initiates m6A RNA methylation for R-loop resolution. Nucleic Acids Res. 2021;49(1):269–84.CrossRefPubMed
15.
go back to reference Zhang C, Chen L, Peng D, Jiang A, He Y, Zeng Y, et al. METTL3 and N6-Methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA-RNA hybrid accumulation. Mol Cell. 2020;79(3):425-42 e7.CrossRefPubMed Zhang C, Chen L, Peng D, Jiang A, He Y, Zeng Y, et al. METTL3 and N6-Methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA-RNA hybrid accumulation. Mol Cell. 2020;79(3):425-42 e7.CrossRefPubMed
16.
go back to reference Yang X, Liu QL, Xu W, Zhang YC, Yang Y, Ju LF, et al. M(6)a promotes R-loop formation to facilitate transcription termination. Cell Res. 2019;29(12):1035–8.CrossRefPubMedPubMedCentral Yang X, Liu QL, Xu W, Zhang YC, Yang Y, Ju LF, et al. M(6)a promotes R-loop formation to facilitate transcription termination. Cell Res. 2019;29(12):1035–8.CrossRefPubMedPubMedCentral
17.
go back to reference Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95.CrossRefPubMedPubMedCentral Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95.CrossRefPubMedPubMedCentral
18.
go back to reference Ying Y, Ma X, Fang J, Chen S, Wang W, Li J, et al. EGR2-mediated regulation of m(6)a reader IGF2BP proteins drive RCC tumorigenesis and metastasis via enhancing S1PR3 mRNA stabilization. Cell Death Dis. 2021;12(8):750.CrossRefPubMedPubMedCentral Ying Y, Ma X, Fang J, Chen S, Wang W, Li J, et al. EGR2-mediated regulation of m(6)a reader IGF2BP proteins drive RCC tumorigenesis and metastasis via enhancing S1PR3 mRNA stabilization. Cell Death Dis. 2021;12(8):750.CrossRefPubMedPubMedCentral
19.
go back to reference Wang IX, Grunseich C, Fox J, Burdick J, Zhu Z, Ravazian N, Hafner M, Cheung VG. Human proteins that interact with RNA/DNA hybrids. Genome Res. 2018;28(9):1405–14.CrossRefPubMedPubMedCentral Wang IX, Grunseich C, Fox J, Burdick J, Zhu Z, Ravazian N, Hafner M, Cheung VG. Human proteins that interact with RNA/DNA hybrids. Genome Res. 2018;28(9):1405–14.CrossRefPubMedPubMedCentral
20.
go back to reference Cristini A, Groh M, Kristiansen MS, Gromak N. RNA/DNA hybrid interactome identifies DXH9 as a molecular player in transcriptional termination and R-loop-associated DNA damage. Cell Rep. 2018;23(6):1891–905.CrossRefPubMedPubMedCentral Cristini A, Groh M, Kristiansen MS, Gromak N. RNA/DNA hybrid interactome identifies DXH9 as a molecular player in transcriptional termination and R-loop-associated DNA damage. Cell Rep. 2018;23(6):1891–905.CrossRefPubMedPubMedCentral
21.
go back to reference Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, et al. METTL3 facilitates tumor progression via an m(6)A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.CrossRefPubMedPubMedCentral Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, et al. METTL3 facilitates tumor progression via an m(6)A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.CrossRefPubMedPubMedCentral
22.
go back to reference Yang H, Wang Y, Xiang Y, Yadav T, Ouyang J, Phoon L, et al. FMRP promotes transcription-coupled homologous recombination via facilitating TET1-mediated m5C RNA modification demethylation. Proc Natl Acad Sci U S A. 2022;119(12):e2116251119.CrossRefPubMedPubMedCentral Yang H, Wang Y, Xiang Y, Yadav T, Ouyang J, Phoon L, et al. FMRP promotes transcription-coupled homologous recombination via facilitating TET1-mediated m5C RNA modification demethylation. Proc Natl Acad Sci U S A. 2022;119(12):e2116251119.CrossRefPubMedPubMedCentral
23.
go back to reference Sanz LA, Chedin F. High-resolution, strand-specific R-loop mapping via S9.6-based DNA-RNA immunoprecipitation and high-throughput sequencing. Nat Protoc. 2019;14(6):1734–55.CrossRefPubMedPubMedCentral Sanz LA, Chedin F. High-resolution, strand-specific R-loop mapping via S9.6-based DNA-RNA immunoprecipitation and high-throughput sequencing. Nat Protoc. 2019;14(6):1734–55.CrossRefPubMedPubMedCentral
24.
go back to reference Li J, Xie H, Ying Y, Chen H, Yan H, He L, et al. YTHDF2 mediates the mRNA degradation of the tumor suppressors to induce AKT phosphorylation in N6-methyladenosine-dependent way in prostate cancer. Mol Cancer. 2020;19(1):152.CrossRefPubMedPubMedCentral Li J, Xie H, Ying Y, Chen H, Yan H, He L, et al. YTHDF2 mediates the mRNA degradation of the tumor suppressors to induce AKT phosphorylation in N6-methyladenosine-dependent way in prostate cancer. Mol Cancer. 2020;19(1):152.CrossRefPubMedPubMedCentral
25.
go back to reference Ma X, Ying Y, Sun J, Xie H, Li J, He L, et al. circKDM4C enhances bladder cancer invasion and metastasis through miR-200bc-3p/ZEB1 axis. Cell Death Discov. 2021;7(1):365.CrossRefPubMedPubMedCentral Ma X, Ying Y, Sun J, Xie H, Li J, He L, et al. circKDM4C enhances bladder cancer invasion and metastasis through miR-200bc-3p/ZEB1 axis. Cell Death Discov. 2021;7(1):365.CrossRefPubMedPubMedCentral
26.
go back to reference Lai CJ, Lin CY, Liao WY, Hour TC, Wang HD, Chuu CP. CD44 promotes migration and invasion of docetaxel-resistant prostate cancer cells likely via induction of hippo-yap signaling. Cells. 2019;8(4):295.CrossRefPubMedPubMedCentral Lai CJ, Lin CY, Liao WY, Hour TC, Wang HD, Chuu CP. CD44 promotes migration and invasion of docetaxel-resistant prostate cancer cells likely via induction of hippo-yap signaling. Cells. 2019;8(4):295.CrossRefPubMedPubMedCentral
27.
go back to reference Matsuda Y, Narita S, Nara T, Mingguo H, Sato H, Koizumi A, et al. Impact of nuclear YAP1 expression in residual cancer after neoadjuvant chemohormonal therapy with docetaxel for high-risk localized prostate cancer. BMC Cancer. 2020;20(1):302.CrossRefPubMedPubMedCentral Matsuda Y, Narita S, Nara T, Mingguo H, Sato H, Koizumi A, et al. Impact of nuclear YAP1 expression in residual cancer after neoadjuvant chemohormonal therapy with docetaxel for high-risk localized prostate cancer. BMC Cancer. 2020;20(1):302.CrossRefPubMedPubMedCentral
28.
go back to reference Wang Y, Lieberman R, Pan J, Zhang Q, Du M, Zhang P, et al. miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol Cancer. 2016;15(1):70.CrossRefPubMedPubMedCentral Wang Y, Lieberman R, Pan J, Zhang Q, Du M, Zhang P, et al. miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol Cancer. 2016;15(1):70.CrossRefPubMedPubMedCentral
29.
go back to reference Hanniford D, Ulloa-Morales A, Karz A, Berzoti-Coelho MG, Moubarak RS, Sanchez-Sendra B, et al. Epigenetic silencing of CDR1as drives IGF2BP3-mediated melanoma invasion and metastasis. Cancer Cell. 2020;37(1):55-e7015.CrossRefPubMedPubMedCentral Hanniford D, Ulloa-Morales A, Karz A, Berzoti-Coelho MG, Moubarak RS, Sanchez-Sendra B, et al. Epigenetic silencing of CDR1as drives IGF2BP3-mediated melanoma invasion and metastasis. Cancer Cell. 2020;37(1):55-e7015.CrossRefPubMedPubMedCentral
30.
go back to reference Li B, Zhu L, Lu C, Wang C, Wang H, Jin H, et al. circNDUFB2 inhibits non-small cell lung cancer progression via destabilizing IGF2BPs and activating anti-tumor immunity. Nat Commun. 2021;12(1):295.CrossRefPubMedPubMedCentral Li B, Zhu L, Lu C, Wang C, Wang H, Jin H, et al. circNDUFB2 inhibits non-small cell lung cancer progression via destabilizing IGF2BPs and activating anti-tumor immunity. Nat Commun. 2021;12(1):295.CrossRefPubMedPubMedCentral
31.
go back to reference Nguyen LH, Robinton DA, Seligson MT, Wu L, Li L, Rakheja D, et al. Lin28b is sufficient to drive liver cancer and necessary for its maintenance in murine models. Cancer Cell. 2014;26(2):248–61.CrossRefPubMedPubMedCentral Nguyen LH, Robinton DA, Seligson MT, Wu L, Li L, Rakheja D, et al. Lin28b is sufficient to drive liver cancer and necessary for its maintenance in murine models. Cancer Cell. 2014;26(2):248–61.CrossRefPubMedPubMedCentral
32.
go back to reference Okuda H, Miyamoto R, Takahashi S, Kawamura T, Ichikawa J, Harada I, et al. RNA-binding proteins of KHDRBS and IGF2BP families control the oncogenic activity of MLL-AF4. Nat Commun. 2022;13(1):6688.CrossRefPubMedPubMedCentral Okuda H, Miyamoto R, Takahashi S, Kawamura T, Ichikawa J, Harada I, et al. RNA-binding proteins of KHDRBS and IGF2BP families control the oncogenic activity of MLL-AF4. Nat Commun. 2022;13(1):6688.CrossRefPubMedPubMedCentral
Metadata
Title
Co-transcriptional R-loops-mediated epigenetic regulation drives growth retardation and docetaxel chemosensitivity enhancement in advanced prostate cancer
Authors
Yufan Ying
Yuqing Wu
Fenghao Zhang
Yijie Tang
Jiahe Yi
Xueyou Ma
Jiangfeng Li
Danni Chen
Xiao Wang
Xiaoyan Liu
Ben Liu
Jindan Luo
Xiangyi Zheng
Liping Xie
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2024
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-024-01994-0

Other articles of this Issue 1/2024

Molecular Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine