International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Recent research has indicated that Y-box binding protein 1 (YBX1/YB-1) has multifaceted functions in regulating drug resistance, PANoptosis, stress response, ferroptosis and cell proliferation (1–5). YBX1 is identified as an oncogene which is overexpressed and positively associated with poor outcomes and survival in numerous cancers including liver, endometrial, non-small-cell lung cancer (NSCLC), ovarian, breast, gastric cancer, kidney renal papillary and clear cell renal cell carcinoma (4–11). YBX1 performs the roles through the regulation of nucleotide metabolism, pre-mRNA splicing, DNA repair, stress granule formation, transcription and translation, and sorting of microRNAs (miRNAs) into exosomes (12–14). Previous studies have shown that YBX1 also plays a crucial role in regulating RNA methylation modification which affects mRNA stability and translation, including N6-methyladenine (m6A) and 5-methylcytosine (m5C) RNA modifications (5,15). The performance of these functions depends on the structure and location of YBX1. The structural features of YBX1 are crucial for its diverse functions, mediating interactions with DNA, RNA and protein molecules (3,16).
SU056 has been demonstrated to specifically target and inhibit the YBX1 protein, rendering it a valuable tool for investigating YBX1 inhibition (17–20). Multiple studies have demonstrated that SU056 effectively hinders tumor progression in diverse types of cancer including pancreatic cancer (18), triple-negative breast cancer (17) and ovarian cancer (19,20).
In the present review, the focus is mainly on the function of YBX1 in RNA modification. The mechanisms by which YBX1 is involved in RNA modification, including m5C, m6A and RNA editing are clarified, and YBX1 is proposed as a potential therapeutic target. Furthermore, the roles of YBX1 in regulating anticancer drug resistance through RNA modification are discussed, and its functions on tumor progression are encompassed. This provides a research foundation for the development of anticancer drugs targeting YBX1.
YBX1 was initially isolated using double-stranded oligonucleotides in a phage λgt11 library screening as the interacting molecule of the MHC class II gene Y-box element (21). In 1992, the murine (m) CCAAT-binding protein was identified and termed mYB-1 (22). The amino acid sequence of mYB-1 has a 95% homology with human YB-1 (22). In addition, YBX1 cDNA was isolated by screening the binding site through the enhancer oligonucleotide of type 18 human papillomavirus in an expression library of HeLa (23). Researchers found that YBX1 is a nuclear protein with a molecular weight of 42 kDa (23). YBX1 is expressed in most tissues, including the liver, spleen, lung and heart (23). Shortly thereafter, it was shown that YBX1 acted as a binding protein of human multidrug resistance 1 (MDR1) gene and regulated MDR1 gene expression in response to adverse environments (24). In humans, YBX1 genes are located on chromosome 1p34 as identified by in situ fluorescence hybridization (25).
YBX1 belongs to the superfamily of the cold shock proteins and is considered to be evolutionarily conserved (26). The structure of YBX1 consists of three distinct domains, the alanine/proline-rich domain (A/P site) in the N-terminal, the cold shock domain (CSD) in the central region and the C-terminal domain (CTD; Fig. 1). The A/P site domain is involved in protein-protein interactions and mediates the assembly of ribonucleoprotein complexes. Additionally, the A/P site domain functions as a regulatory element, modulating the stabilization of RNA-binding proteins (27). The highly conserved CSD facilitates both DNA and RNA binding, plays a crucial role in mRNA translation, and regulates the adaptation of bacteria to low temperatures (28). In previous studies, CSD was identified to contribute to the identification of m5C modification, enhancing mRNA stability and promoting translation (29,30). The CTD contains two important domains that mediate the localization of YBX1, the cytoplasmic retention site (CRS) and the nuclear localization signals (NLS), regulating the location of YBX1 between the cell nucleus and cytoplasm in response to cellular stimuli. Furthermore, CTD has been shown to mediate interactions with diverse protein partners and DNA/RNA-binding proteins (27,28). YBX1 is usually localized in the cell cytoplasm due to the stronger influence of CRS compared with that of NLS. The various functions of YBX1 stem from its diverse domains, making it a key DNA/RNA-binding protein.
Research has revealed that the cytidine residues at position five of RNA could be methylated by m5C methyltransferases, such as the NOP2/Sun RNA methyltransferase (NSUN) protein family (31). During the m5C modification, numerous methyltransferases and proteins are involved in this process, including m5C writers, m5C erasers and m5C readers (31,32). In humans, m5C writers consist of the NSUN protein family 1–6 and DNA methyltransferase 2, which catalyze cytosine-5 methylation (32). Among them, NSUN2 is considered the most prominent m5C methyltransferase (32). The m5C erasers contain the ten-eleven translocation (TET) protein family and AlkB homolog 1, histone H2A dioxygenase (32,33). However, the functions of the m5C eraser and those of additional eraser proteins require further investigation (33). The m5C readers include YBX1, YTH N6-methyladenosine RNA binding protein F2 (YTHDF2) and Aly/Ref export factor (ALYREF), and assume the recognition of the m5C modification (34,35). In addition, ALYREF is required for the nuclear export of m5C-modified mRNA (34). Furthermore, YBX1 can identify m5C-modified mRNA in the cytoplasm and maintain mRNA stability (36,37). YTHDF2 can regulate the maturation of m5C-modified ribosomal RNA (rRNA) (35).
The m5C modification of RNA can affect its stability, which is important for translation (31). It is a common and conserved phenomenon in various RNAs, including non-coding RNAs (ncRNAs) and mRNAs (38). Over the past years, the RNA m5C modification has been demonstrated to be linked to diverse diseases, including cancers (32), viral infections (39) and autoimmune diseases (40). For instance, m5C-modified SKI like proto-oncogene (SKIL) may facilitate the progression of colorectal cancer (CRC) (41). Liu et al (7) revealed that the m5C modification could enhance the expression of E2F transcription factor 1, thus accelerating ovarian cancer development. These findings indicate that the RNA m5C modification plays a vital role in disease progression.
In 2019, Chen et al (29) first characterized YBX1 as an m5C reader protein on the distribution of m5C modifications in human cancer. They identified the amino acid residue W65 as a key component involved in recognizing the m5C modification. YBX1 was previously identified as a DNA/RNA-binding protein that performs a proto-oncogene role in cancers. As an m5C reader protein, YBX1 plays significant roles, making it a potential therapeutic target in cancer treatment (5,19,29,42–45). For instance, Chen et al (46) identified an NSUN2/YBX1/nuclear factor erythroid 2-related factor 2 (NRF2) axis that promotes resistance to ferroptosis in NSCLC cells. Mechanistically, NSUN2 was demonstrated to enhance NRF2 expression by adding m5C modifications to the mRNA in the 5′ untranslated region (UTR). YBX1 was shown to bind to the m5C-modified mRNA, which increased the stability of NRF2 mRNA without affecting the translation process (46). This suggests that YBX1 contributes to resistance through m5C modification of RNA rather than by influencing MDR1 expression. Similarly, YBX1 can also regulate the stability of solute carrier family 7 member 11 (SLC7A11) mRNA, impacting ferroptosis resistance via m5C modification in endometrial cancer (4). YBX1 was shown to bind to m5C-modified SLC7A11 mRNA, enhancing its stability and developing ferroptosis resistance (4). The inhibition of YBX1 through ubiquitination was demonstrated to decrease the expression of m5C-modified mRNAs, which in turn increased the sensitivity of epithelial ovarian cancer cells to cisplatin (47). Additionally, in EGFR-mutant NSCLC cells, the YBX1/quiescin sulfhydryl oxidase 1 (QSOX1) axis was responsible for mediating resistance to gefitinib. YBX1 was found to be upregulated in cells resistant to gefitinib (5). Furthermore, YBX1 recognized the m5C modification in the coding sequence of QSOX1 mRNA, which led to an increase in QSOX1 translation and a corresponding decrease in gefitinib sensitivity in NSCLC cells (5). This indicates that the YBX1-mediated m5C modification may contribute to the drug resistance observed in cancer cells, providing novel insights into potential strategies for overcoming this resistance.
YBX1 can not only impact drug resistance through m5C modification, but also promote tumor development. In lung squamous cell carcinoma (LUSC), YBX1 recognized the m5C modification in the 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 mRNA 3′UTR region, leading to strengthened stability, thereby promoting LUSC progression (48). In CRC, YBX1 stabilized SKIL mRNA by m5C modification and increased SKIL expression. SKIL then facilitated the activation of transcriptional coactivator with PDZ-binding motif, thus accelerating tumor progression (41). Furthermore, YBX1 was shown to be increased in immune cells of tumor-bearing mice, elucidating the reason for m5C upregulation in CRC blood immune cells (49). This perspective suggests that YBX1 is not only involved in the m5C modification of tumor cells, but also influences the m5C modification of immune cells. Moreover, m5C-mediated RNA modification has also been shown to promote tumor metastasis. Tetraspanin 13 (TSPAN13) promoted acute myeloid leukemia stem cell migration/homing through the C-X-C motif chemokine (CXC)R4/CXCL12 pathway. In TET-mutant cells, YBX1 stabilized TSPAN13 mRNA via the m5C modification, thus enhancing migration/homing and cell self-renewal (43). Liu et al (50) also found that m5C-modified Orai calcium release-activated calcium modulator 2 promoted gastric cancer metastasis in a YBX1-dependent manner. In addition, YBX1 could stabilize leucine-rich repeat-containing 8 VRAC subunit A mRNA in an m5C-dependent manner to inhibit cervical cancer apoptosis (51).
Although YBX1 is mainly studied in cancers, it has also been demonstrated to regulate bone formation and metabolic diseases through m5C modification (52,53). Li et al (52) demonstrated that elevated YBX1 could facilitate osteogenesis and reduce bone loss. YBX1 deletion could repress the morphology of CD31-high, endomucin-high (CD31highEMCNhigh) endothelial cells, leading to decreased bone mass in an m5C-dependent manner. YBX1 could modulate the stability of EMCN, CD31 and bone morphogenetic protein 4 (BMP4), and influence the release of BMP4, thereby controlling bone formation (52). Furthermore, YBX1 could promote adipogenesis and autophagy in an m5C-dependent manner, thus leading to obesity (45). In a previous study it was found that YBX1 directly recognized m5C-modified Unc-51 like autophagy activating kinase 1 (ULK1) mRNA and improved its stability. In addition, YBX1 enhanced ULK1-mediated autophagy and increased obesity formation (53). These findings indicate that YBX1-mediated m5C modification assumes a crucial function in various diseases.
The dysregulation of YBX1-mediated RNA m5C modification represents a potential target for therapeutic strategy in human diseases. Regulating the relationship between YBX1 and m5C-modified transcripts holds promise for developing novel therapeutic strategies aimed at restoring normal RNA metabolism and cellular homeostasis. Furthermore, elucidating the mechanisms underlying YBX1-mediated m5C modification will advance our understanding of RNA biology and disease pathogenesis. Future investigations should concentrate on unraveling the spatiotemporal dynamics of YBX1-m5C co-actions and their function in different cellular microenvironments.
In conclusion, YBX1 plays a crucial role in modulating the RNA m5C modification, which is vital for controlling gene expression essential for cellular function and adaptation, especially in cancers and drug resistance (Fig. 2). Through its interactions with m5C methyltransferases, readers and RNA molecules, YBX1 influences various aspects of RNA metabolism and cellular physiology. Dysregulation of the YBX1-mediated m5C modification has implications for human health and disease, emphasizing its potential as a therapeutic target. Current research focuses on understanding the connection between YBX1 and the m5C modification, which will enhance our knowledge of RNA regulation and offer novel strategies for disease therapy.
To date, the most extensively studied RNA modification is m6A. This modification involves the methylation of adenosine residues at position six of RNA by m6A methyltransferases (31). The m6A modification regulates various molecular events, including RNA stability, RNA transport, the splicing of pre-mRNA, transcription, post-transcription and translation (54–56). The m6A methylation is the most abundant modification of RNA, found in long ncRNA (57–62), circular RNAs (63,64), miRNAs (65,66), mRNA (58,67), and rRNA and mtRNA (68), modulating the fates of RNA and affecting a variety of biological processes including spermatogenesis (69), pluripotency of embryonic stem cells (70), cell differentiation (71), proliferation (72), tumor metastasis (73), drug resistance of cancer (74) and the metabolism of tumor cells (75).
During the m6A modification processes, a variety of enzymes and proteins are involved, including m6A methyltransferases (commonly referred to as ‘writers’), m6A demethylases (known as ‘erasers’) and m6A-recognized factors (called ‘readers’). The writers contain methyltransferase-like (METTL)3, METTL14, WT1 associated protein, METTL16 and RNA binding motif protein 15, which assume the catalysis of m6A in RNA (76–79). Notably, two proteins, AlkB homolog 5, RNA demethylase and FTO α-ketoglutarate-dependent dioxygenase, have been demonstrated to be capable of removing the m6A modification from RNA (80–83). Furthermore, YTHDF1-3, YTHDC1-2, heterogenous nuclear ribonucleoprotein A2/B1, heterogenous nuclear ribonucleoprotein A/Cand insulin-like growth factor 2 mRNA binding protein (IGF2BP)1-3 have been identified as factors that recognize and bind to m6A-modified RNA (80,84–91). Normally, methyltransferases and demethylases dynamically cooperate to regulate gene expression and determine the fate of molecules and cells (56). However, aberrant hypermethylation of m6A can lead to the development of multiple diseases, including cancers (92), intestinal inflammation (58) and liver disease (93).
YBX1 can serve as a transcription factor or DNA-binding protein that regulates the expression of multiple genes (75). Accumulating evidence has revealed that YBX1 is a regulatory protein involved in the m6A modification. In 2021, Feng et al (15) demonstrated the significance of YBX1 in myeloid leukemia cell survival via m6A modification of BCL2 apoptosis regulator (BCL2). It was found that YBX1 was notably upregulated, promoting proliferation in myeloid leukemia cells. Mechanistically, YBX1 stabilized m6A-modified BCL2 and MYC proto-oncogene, BHLH transcription factor by cooperating with IGF2BP1 and IGF2BP3. Furthermore, the loss of YBX1 led to an accelerated decay of m6A-tagged BCL2 mRNA, suggesting that YBX1 can regulate BCL2 expression in an m6A-dependent manner, thus playing a critical role in the survival of myeloid leukemia cells (15). Similarly, YBX1 is required for the survival of leukemia stem cells (94). YBX1 was revealed to interact with the IGF2BP protein to enhance the stability of m6A-tagged tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) mRNA. YBX1 deficiency downregulated the expression of YWHAZ by promoting YWHAZ mRNA degradation in an m6A-dependent manner, thereby maintaining leukemia cell survival (94). Additionally, YBX1 has been shown to affect embryonic development (95,96) and ischemic stroke (97) in an m6A-dependent manner. YBX1 was found to influence the gene expression of zygotic genome activation by m6A-modified RNA, thereby regulating early embryonic development. YBX1 fine-tuned polycomb repressive complex 2 activity to regulate embryonic neural development (95) Peng et al (97) also observed that NSC-derived exosomes loaded with YBX1 inhibited neuronal pyroptosis, thereby mitigating the development of ischemic stroke. YBX1 interacted with IGF2BP1 to heighten the stability of m6A-tagged G protein-coupled receptor 30 (GPR30) mRNA, which increased the expression of GPR30. GPR30 then facilitated the degradation of NLRP3 by ubiquitination, alleviating the progression of ischemic stroke. The inhibition of IGF2BP1 decreased YBX1 binding to GPR30, which contributed to the progression of ischemic stroke (97). YBX1 is a regulatory protein involved in the m6A modification of RNA. It stabilizes the mRNA of target genes, thereby enhancing their expression. Unlike the YBX1 regulation model in the m5C modification, the IGF2BP protein is required for YBX1 to maintain mRNA stability in the m6A modification. Furthermore, YBX1 can function as an m6A reader, dependent on the IGF2BP protein (98–100). The functions of YBX1 in N1methyladenosine (m1A) and 7methylguanosine (m7G) remain unknown. Therefore, further investigation is needed to explore the contributions and potential mechanisms of YBX1 in RNA methylation modifications.
The dysregulation of YBX1-mediated RNA m6A modification has emerged as a hallmark of numerous diseases, making it a potential target for therapeutic interventions (15,101–105). Targeting the interaction between YBX1 and m6A-modified transcripts holds promise for developing novel therapeutic approaches aimed at restoring normal RNA modification in diseased states. Furthermore, elucidating the mechanisms of the YBX1-mediated m6A modification will provide novel insights into the complex dynamics of RNA regulation. In addition, it is essential to uncover the roles of YBX1 in m6A modification and its effects on cellular processes and the development of diseases.
Research has shown that YBX1 upregulation could promote tumor development by regulating m5C and m6A modifications. SU056 is a small molecule of azopodophyllotoxin that inhibits YBX1, and helps reverse drug resistance and inhibit tumor development (17–20). YBX1 knockdown was shown to reduce gemcitabine resistance, and SU056 in combination with gemcitabine overcame gemcitabine resistance in pancreatic cancer (18). Additionally, SU056 was demonstrated to inhibit triple-negative breast cancer growth in preclinical models by targeting YB-1 to disrupt protein translation mechanisms (17). SU056 also reduced the progression of ovarian cancer while sensitizing to paclitaxel-mediated cytotoxicity (20). Furthermore, platinum-induced cell stress enhanced YBX1, which was expressed at high levels in platinum-resistant ovarian cancer. YBX1 recognized and stabilized CHD3 mRNA through m5C modification. By targeting YBX1, SU056 reversed platinum resistance and enhanced tumor cell killing (19). In addition, another YBX1 inhibitor, 2,4-dihydroxy-5-pyrimidinyl imidothiocarbomate, also exerted similar antitumor effects, although its mechanism of action remains unexplored (106). The activities and efficacy profiles of other YBX1 inhibitors also remain incompletely characterized.
Collectively, SU056 has been demonstrated to exert antitumor effects and reverse drug resistance by targeting YBX1, with its effectiveness dependent on the cellular expression of YBX1 (20). Additionally, YBX1 has been shown to facilitate tumor progression and confer drug resistance through its regulation of RNA m5C and m6A modifications (5,107–109). Therefore, it is hypothesized that the antitumor effect and reversal of drug resistance exerted by SU056 may be attributed to its inhibition of RNA m5A and m6A methylation.
YBX1, a DNA/RNA-binding protein, is implicated in DNA repair, mRNA transcription, pre-mRNA splicing, mRNA stability regulation, translation and exosome sorting (28,110), influencing the development of multiple diseases. Notably, extensive evidence has shown that YBX1 plays multifunctional roles in cancer progression and drug resistance (111,112). Furthermore, RNA methylation modifications are dysregulated in cancers and serve as potential targets of tumor therapy (31). Although YBX1 is implicated in various cancer hallmarks, its underlying mechanisms, particularly those related to RNA methylation modifications during cancer development such as m1A, m5C, m6A and m7G require further investigation. Understanding the functional role of YBX1 in RNA modifications could provide novel insights into the regulation of gene expression, cellular homeostasis and molecular pathogenesis. This knowledge may also lead to promising therapeutic strategies that target RNA modifications for cancer treatment.
Given its critical roles in cancer pathogenesis and treatment resistance, YBX1 has emerged as an attractive therapeutic target for anticancer therapy. Several approaches have been explored to modulate YBX1 activity, including the use of small molecule inhibitors. The YBX1 inhibitor SU056 has shown promising efficacy in inhibiting tumor growth, promoting cell apoptosis and sensitizing cancer cells to chemotherapy. Despite the significance of YBX1 in cancer, the clinical application of YBX1 inhibitors remains limited. Further investigations should focus on the regulatory networks between YBX1 and RNA modifications, which will broaden the understanding of the role of YBX1 in cellular activity and tumorigenesis.
In summary, YBX1 plays a pivotal role in regulating RNA modifications specifically m6A and m5C RNA modifications influencing a variety of cellular processes and diseases (Table I). Understanding the underlying mechanisms of YBX1 in RNA modification is crucial for developing effective strategies to treat cancers. Targeting YBX1 and RNA modifications may help overcome the challenges of drug resistance in the clinic, and the YBX1 inhibitor SU056 exhibits great potential as an antitumor agent.
Not applicable.
Funding: No funding was received.
Not applicable.
XX conceived and designed the review, as well as drafted and provided overall supervision of the manuscript. GX performed and contributed to specific sections of the review and participated in manuscript revision. YX was involved in revising the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Y-box |
binding protein 1 |
|
m5C |
5-methylcytosine |
|
m6A |
N6-methyladenine |
|
CDS |
cold shock domain |
|
SKIL |
SKI like proto-oncogene |
|
NSCLC |
non-small cell lung cancer |
|
NRF2 |
nuclear factor erythroid 2-related factor 2 |
|
SLC7A11 |
solute carrier family 7 member 11 |
|
TSPAN13 |
tetraspanin 13 |
|
CRC |
colorectal cancer |
|
LUSC |
lung squamous cell carcinoma |
|
QSOX1 |
quiescin sulfhydryl oxidase 1 |
|
EMCN |
endomucin |
|
BMP4 |
bone morphogenetic protein 4 |
|
ULK1 |
Unc-51 like autophagy activating kinase 1 |
|
GPR30 |
G protein-coupled receptor 30 |
|
YWHAZ |
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta |
|
BCL2 |
BCL2 apoptosis regulator |
|
Lin C, Lin P, Yao H, Liu S, Lin X, He R, Teng Z, Zuo X, Li Y, Ye J and Zhu G: Modulation of YBX1-mediated PANoptosis inhibition by PPM1B and USP10 confers chemoresistance to oxaliplatin in gastric cancer. Cancer Lett. 587:2167122024. View Article : Google Scholar : PubMed/NCBI | |
|
Mao S, Xie C, Liu Y, Zhao Y, Li M, Gao H, Xiao Y, Zou Y, Zheng Z, Gao Y, et al: Apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) promotes stress granule formation via YBX1 phosphorylation in ovarian cancer. Cell Mol Life Sci. 81:1132024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Fan JS, Li S, Yang Y, Sun P, Zhu Q, Wang J, Jiang B, Yang D and Liu M: Structural basis of DNA binding to human YB-1 cold shock domain regulated by phosphorylation. Nucleic Acids Res. 48:9361–9371. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen SJ, Zhang J, Zhou T, Rao SS, Li Q, Xiao LY, Wei ST and Zhang HF: Epigenetically upregulated NSUN2 confers ferroptosis resistance in endometrial cancer via m(5)C modification of SLC7A11 mRNA. Redox Biol. 69:1029752024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Wei J, Feng L, Li O, Huang L, Zhou S, Xu Y, An K, Zhang Y, Chen R, et al: Aberrant m5C hypermethylation mediates intrinsic resistance to gefitinib through NSUN2/YBX1/QSOX1 axis in EGFR-mutant non-small-cell lung cancer. Mol Cancer. 22:812023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Ji L, Liang Y, Zheng W, Song X, Gorshkov K, Sun Q, Lin H, Zheng X, Chen J, et al: CircRNA-SORE mediates sorafenib resistance in hepatocellular carcinoma by stabilizing YBX1. Signal Transduct Target Ther. 5:2982020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Wei Q, Yang C, Zhao H, Xu J, Mobet Y, Luo Q, Yang D, Zuo X, Chen N, et al: RNA m(5)C modification upregulates E2F1 expression in a manner dependent on YBX1 phase separation and promotes tumor progression in ovarian cancer. Exp Mol Med. 56:600–615. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Song S, He X, Wang J, Song H, Wang Y, Liu Y, Zhou Z, Yu Z, Miao D and Xue Y: A novel long noncoding RNA, TMEM92-AS1, promotes gastric cancer progression by binding to YBX1 to mediate CCL5. Mol Oncol. 15:1256–1273. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu T, Xiong M, Hong Q, Pan B, Xu M, Wang Y, Sun Y, Sun H, Pan Y, Wang S and He B: Hsa_circ_0007990 promotes breast cancer growth via inhibiting YBX1 protein degradation to activate E2F1 transcription. Cell Death Dis. 15:1532024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Cao C, Zhou CL, Li X, Miao C, Shen L, Singla RK and Lu X: Identification of a novel 5-methylcytosine-related signature for prognostic prediction of kidney renal papillary cell carcinoma and a Putative target for drug repurposing. Transl Oncol. 36:1017412023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Yin H, Chen Y, Pan C, Hang H, Lu Y, Ma W, Li X, Gan W, Guo H and Li D: Low expression of PEBP1P2 promotes metastasis of clear cell renal cell carcinoma by post-transcriptional regulation of PEBP1 and KLF13 mRNA. Exp Hematol Oncol. 11:872022. View Article : Google Scholar : PubMed/NCBI | |
|
Gandhi M, Groß M, Holler JM, Coggins SA, Patil N, Leupold JH, Munschauer M, Schenone M, Hartigan CR, Allgayer H, et al: The lncRNA lincNMR regulates nucleotide metabolism via a YBX1-RRM2 axis in cancer. Nat Commun. 11:32142020. View Article : Google Scholar : PubMed/NCBI | |
|
Lyabin DN, Eliseeva IA and Ovchinnikov LP: YB-1 protein: Functions and regulation. Wiley Interdiscip Rev RNA. 5:95–110. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Liu XM, Ma L and Schekman R: Selective sorting of microRNAs into exosomes by phase-separated YBX1 condensates. Elife. 10:e719822021. View Article : Google Scholar : PubMed/NCBI | |
|
Feng M, Xie X, Han G, Zhang T, Li Y, Li Y, Yin R, Wang Q, Zhang T, Wang P, et al: YBX1 is required for maintaining myeloid leukemia cell survival by regulating BCL2 stability in an m6A-dependent manner. Blood. 138:71–85. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Budkina K, El Hage K, Clément MJ, Desforges B, Bouhss A, Joshi V, Maucuer A, Hamon L, Ovchinnikov LP, Lyabin DN and Pastré D: YB-1 unwinds mRNA secondary structures in vitro and negatively regulates stress granule assembly in HeLa cells. Nucleic Acids Res. 49:10061–10081. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dheeraj A, Garcia Marques FJ, Tailor D, Bermudez A, Resendez A, Pandrala M, Grau B, Kumar P, Haley CB, Honkala A, et al: Inhibition of protein translational machinery in triple-negative breast cancer as a promising therapeutic strategy. Cell Rep Med. 5:1015522024. View Article : Google Scholar : PubMed/NCBI | |
|
Li B, Xing F, Wang J, Wang X, Zhou C, Fan G, Zhuo Q, Ji S, Yu X, Xu X, et al: YBX1 as a therapeutic target to suppress the LRP1-β-catenin-RRM1 axis and overcome gemcitabine resistance in pancreatic cancer. Cancer Lett. 602:2171972024. View Article : Google Scholar : PubMed/NCBI | |
|
Meng H, Miao H, Zhang Y, Chen T, Yuan L, Wan Y, Jiang Y, Zhang L and Cheng W: YBX1 promotes homologous recombination and resistance to platinum-induced stress in ovarian cancer by recognizing m5C modification. Cancer Lett. 597:2170642024. View Article : Google Scholar : PubMed/NCBI | |
|
Tailor D, Resendez A, Garcia-Marques FJ, Pandrala M, Going CC, Bermudez A, Kumar V, Rafat M, Nambiar DK, Honkala A, et al: Y box binding protein 1 inhibition as a targeted therapy for ovarian cancer. Cell Chem Biol. 28:1206–1220.e6. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Didier DK, Schiffenbauer J, Woulfe SL, Zacheis M and Schwartz BD: Characterization of the cDNA encoding a protein binding to the major histocompatibility complex class II Y box. Proc Natl Acad Sci USA. 85:7322–7326. 1988. View Article : Google Scholar : PubMed/NCBI | |
|
Gai XX, Lipson KE and Prystowsky MB: Unusual DNA binding characteristics of an in vitro translation product of the CCAAT binding protein mYB-1. Nucleic Acids Res. 20:601–606. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Spitkovsky DD, Royer-Pokora B, Delius H, Kisseljov F, Jenkins NA, Gilbert DJ, Copeland NG and Royer HD: Tissue restricted expression and chromosomal localization of the YB-1 gene encoding a 42 kD nuclear CCAAT binding protein. Nucleic Acids Res. 20:797–803. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Asakuno K, Kohno K, Uchiumi T, Kubo T, Sato S, Isono M and Kuwano M: Involvement of a DNA binding protein, MDR-NF1/YB-1, in human MDR1 gene expression by actinomycin D. Biochem Biophys Res Commun. 199:1428–1435. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Makino Y, Ohga T, Toh S, Koike K, Okumura K, Wada M, Kuwano M and Kohno K: Structural and functional analysis of the human Y-box binding protein (YB-1) gene promoter. Nucleic Acids Res. 24:1873–1878. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Kuwano M, Shibata T, Watari K and Ono M: Oncogenic Y-box binding protein-1 as an effective therapeutic target in drug-resistant cancer. Cancer Sci. 110:1536–1543. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Gao C, Xu X, Li M, Zhao X, Wang Y, Wang Y, Zhang S, Yan Z, Liu X and Wu C: FBL promotes cancer cell resistance to DNA damage and BRCA1 transcription via YBX1. EMBO Rep. 24:e562302023. View Article : Google Scholar : PubMed/NCBI | |
|
Bates M, Boland A, McDermott N and Marignol L: YB-1: The key to personalised prostate cancer management? Cancer Lett. 490:66–75. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Li A, Sun BF, Yang Y, Han YN, Yuan X, Chen RX, Wei WS, Liu Y, Gao CC, et al: 5-methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs. Nat Cell Biol. 21:978–990. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Wang L, Han X, Yang WL, Zhang M, Ma HL, Sun BF, Li A, Xia J, Chen J, et al: RNA 5-methylcytosine facilitates the maternal-to-Zygotic transition by preventing maternal mRNA. Decay Mol Cell. 75:1188–1202.e11. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Barbieri I and Kouzarides T: Role of RNA modifications in cancer. Nat Rev Cancer. 20:303–322. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Nombela P, Miguel-López B and Blanco S: The role of m(6)A, m(5)C and Ψ RNA modifications in cancer: Novel therapeutic opportunities. Mol Cancer. 20:182021. View Article : Google Scholar : PubMed/NCBI | |
|
Guo G, Pan K, Fang S, Ye L, Tong X, Wang Z, Xue X and Zhang H: Advances in mRNA 5-methylcytosine modifications: Detection, effectors, biological functions, and clinical relevance. Mol Ther Nucleic Acids. 26:575–593. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Hou X, Guan Q, Zhou H, Zhou L, Liu L, Liu J, Li F, Li W and Liu H: RNA modification in cardiovascular disease: Implications for therapeutic interventions. Signal Transduct Target Ther. 8:4122023. View Article : Google Scholar : PubMed/NCBI | |
|
Dai X, Gonzalez G, Li L, Li J, You C, Miao W, Hu J, Fu L, Zhao Y, Li R, et al: YTHDF2 Binds to 5-Methylcytosine in RNA and modulates the maturation of ribosomal RNA. Anal Chem. 92:1346–1354. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang G, Liu L, Li J, Chen Y, Wang Y, Zhang Y, Dong Z, Xue W, Sun R and Cui G: NSUN2 stimulates tumor progression via enhancing TIAM2 mRNA stability in pancreatic cancer. Cell Death Discov. 9:2192023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Wan F, Xu W, Liu Z, Wang J, Zhang H, Huang S and Ye D: Positive epigenetic regulation loop between AR and NSUN2 promotes prostate cancer progression. Clin Transl Med. 12:e10282022. View Article : Google Scholar : PubMed/NCBI | |
|
Han X, Wang M, Zhao YL, Yang Y and Yang YG: RNA methylations in human cancers. Semin Cancer Biol. 75:97–115. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Cui L, Ma R, Cai J, Guo C, Chen Z, Yao L, Wang Y, Fan R, Wang X and Shi Y: RNA modifications: Importance in immune cell biology and related diseases. Signal Transduct Target Ther. 7:3342022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang N, Tang H, Wang X, Wang W and Feng J: Homocysteine upregulates interleukin-17A expression via NSun2-mediated RNA methylation in T lymphocytes. Biochem Biophys Res Commun. 493:94–99. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zou S, Huang Y, Yang Z, Zhang J, Meng M, Zhang Y, Feng J, Sun R, Li W, Wang W, et al: NSUN2 promotes colorectal cancer progression by enhancing SKIL mRNA stabilization. Clin Transl Med. 14:e16212024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen B, Deng Y, Hong Y, Fan L, Zhai X, Hu H, Yin S, Chen Q, Xie X, Ren X, et al: Metabolic recoding of NSUN2-Mediated m(5)C modification promotes the progression of colorectal cancer via the NSUN2/YBX1/m(5)C-ENO1 positive feedback loop. Adv Sci (Weinh). 11:e23098402024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Xue M, Deng X, Nguyen LXT, Ren L, Han L, Li C, Xue J, Zhao Z, Li W, et al: TET2-mediated mRNA demethylation regulates leukemia stem cell homing and self-renewal. Cell Stem Cell. 30:1072–1090.e10. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng S, Hu C, Lin Q, Li T, Li G, Tian Q, Zhang X, Huang T, Ye Y, He R, et al: Extracellular vesicle-packaged PIAT from cancer-associated fibroblasts drives neural remodeling by mediating m5C modification in pancreatic cancer mouse models. Sci Transl Med. 16:eadi01782024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Zhang J, Su Y, Maimaitiyiming Y, Yang S, Shen Z, Lin S, Shen S, Zhan G, Wang F, et al: Distinct roles of m(5)C RNA methyltransferase NSUN2 in major gynecologic cancers. Front. Oncol. 12:7862662022.PubMed/NCBI | |
|
Chen Y, Jiang Z, Zhang C, Zhang L, Chen H, Xiao N, Bai L, Liu H and Wan J: 5-Methylcytosine transferase NSUN2 drives NRF2-mediated ferroptosis resistance in non-small cell lung cancer. J Biol Chem. 300:1067932024. View Article : Google Scholar : PubMed/NCBI | |
|
Gao W, Chen L, Lin L, Yang M, Li T, Wei H, Sha C, Xing J, Zhang M, Zhao S, et al: SIAH1 reverses chemoresistance in epithelial ovarian cancer via ubiquitination of YBX-1. Oncogenesis. 11:132022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu T, Zhang Q, Yu SK, Nie FQ, Zhang ML, Wang Q and Lu KH: THOC3 interacts with YBX1 to promote lung squamous cell carcinoma progression through PFKFB4 mRNA modification. Cell Death Dis. 14:4752023. View Article : Google Scholar : PubMed/NCBI | |
|
Yin H, Huang Z, Niu S, Ming L, Jiang H, Gu L, Huang W, Xie J, He Y and Zhang C: 5-Methylcytosine (m(5)C) modification in peripheral blood immune cells is a novel non-invasive biomarker for colorectal cancer diagnosis. Front Immunol. 13:9679212022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Xu P, Lv J, Ge H, Yan Z, Huang S, Li B, Xu H, Yang L, Xu Z and Zhang D: Peritoneal high-fat environment promotes peritoneal metastasis of gastric cancer cells through activation of NSUN2-mediated ORAI2 m5C modification. Oncogene. 42:1980–1993. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Zuo X, Wei Q, Xu J, Liu X, Liu S, Wang H, Luo Q, Wang Y, Yang Y, et al: Upregulation of LRRC8A by m(5)C modification-mediated mRNA stability suppresses apoptosis and facilitates tumorigenesis in cervical cancer. Int J Biol Sci. 19:691–704. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li YJ, Guo Q, Ye MS, Cai G, Xiao WF, Deng S and Xiao Y: YBX1 promotes type H vessel-dependent bone formation in an m5C-dependent manner. JCI Insight. 9:e1723452024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu R, Feng S, Li F, Shu G, Wang L, Gao P, Zhu X, Zhu C, Wang S and Jiang Q: Transcriptional and post-transcriptional control of autophagy and adipogenesis by YBX1. Cell Death Dis. 14:292023. View Article : Google Scholar : PubMed/NCBI | |
|
Boo SH and Kim YK: The emerging role of RNA modifications in the regulation of mRNA stability. Exp Mol Med. 52:400–408. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Delaunay S and Frye M: RNA modifications regulating cell fate in cancer. Nat Cell Biol. 21:552–559. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Yang D, Liu T, Chen J, Yu J and Yi P: N6-methyladenosine-mediated gene regulation and therapeutic implications. Trends Mol Med. 29:454–467. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fazi F and Fatica A: Interplay between N (6)-Methyladenosine (m(6)A) and non-coding RNAs in cell development and cancer. Front Cell Dev Biol. 7:1162019. View Article : Google Scholar : PubMed/NCBI | |
|
Olazagoitia-Garmendia A, Rojas-Márquez H, Sebastian-delaCruz M, Agirre-Lizaso A, Ochoa A, Mendoza-Gomez LM, Perugorria MJ, Bujanda L, Madrigal AH, Santin I and Castellanos-Rubio A: m(6) a methylated long noncoding RNA LOC339803 regulates intestinal inflammatory response. Adv Sci (Weinh). 11:e23079282024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang R, Xu X, Yang J, Chen W, Zhao J, Wang M, Zhang Y, Yang Y, Huang W and Zhang H: BPDE exposure promotes trophoblast cell pyroptosis and induces miscarriage by up-regulating lnc-HZ14/ZBP1/NLRP3 axis. J Hazard Mater. 455:1315432023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Wei T, Yan L, Zhu S, Jin W, Bai Y, Zeng Y, Zhang X, Yin Z, Yang J, et al: Hypoxia-Responsive lncRNA AC115619 encodes a micropeptide that suppresses m6A modifications and hepatocellular carcinoma progression. Cancer Res. 83:2496–2512. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng H, Zhu M, Li W, Zhou Z and Wan X: m(5) C and m(6) A modification of long noncoding NKILA accelerates cholangiocarcinoma progression via the miR-582-3p-YAP1 axis. Liver Int. 42:1144–1157. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan J, Liu J, Huang Z, Huang W and Lv J: FOXC2-AS1 stabilizes FOXC2 mRNA via association with NSUN2 in gastric cancer cells. Hum. Cell. 34:1755–1764. 2021. | |
|
Li H, Lin R, Zhang Y, Zhu Y, Huang S, Lan J, Lu N, Xie C, He S and Zhang W: N6-methyladenosine-modified circPLPP4 sustains cisplatin resistance in ovarian cancer cells via PIK3R1 upregulation. Mol Cancer. 23:52024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng K, Peng J, Xing Y, Zhang L, Zeng P, Li W, Zhang W, Pan Z, Zhou C and Lin J: A positive feedback circuit driven by m(6)A-modified circular RNA facilitates colorectal cancer liver metastasis. Mol Cancer. 22:2022023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu S, Tang W, Liu L, Wei K, Tang Y, Ma J, Li H and Ao Y: Obesity-induced downregulation of miR-192 exacerbates lipopolysaccharide-induced acute lung injury by promoting macrophage activation. Cell Mol Biol Lett. 29:362024. View Article : Google Scholar : PubMed/NCBI | |
|
van den Homberg DAL, van der Kwast R, Quax PHA and Nossent AY: N-6-Methyladenosine in Vasoactive microRNAs during Hypoxia; A novel role for METTL4. Int J Mol Sci. 23:10572022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou L, Jiang J, Huang Z, Jin P, Peng L, Luo M, Zhang Z, Chen Y, Xie N, Gao W, et al: Hypoxia-induced lncRNA STEAP3-AS1 activates Wnt/β-catenin signaling to promote colorectal cancer progression by preventing m(6)A-mediated degradation of STEAP3 mRNA. Mol Cancer. 21:1682022. View Article : Google Scholar : PubMed/NCBI | |
|
Qi YN, Liu Z, Hong LL, Li P and Ling ZQ: Methyltransferase-like proteins in cancer biology and potential therapeutic targeting. J Hematol Oncol. 16:892023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Li J, Li C, Lu S, Wei X, Li Y, Xia W, Qian C, Wang Z, Liu M, et al: Fat mass and obesity-associated factor (FTO)-mediated N6-methyladenosine regulates spermatogenesis in an age-dependent manner. J Biol Chem. 299:1047832023. View Article : Google Scholar : PubMed/NCBI | |
|
Jin KX, Zuo R, Anastassiadis K, Klungland A, Marr C and Filipczyk A: N6-methyladenosine (m(6)A) depletion regulates pluripotency exit by activating signaling pathways in embryonic stem cells. Proc Natl Acad Sci USA. 118:e21051921182021. View Article : Google Scholar : PubMed/NCBI | |
|
Dou X, Xiao Y, Shen C, Wang K, Wu T, Liu C, Li Y, Yu X, Liu J, Dai Q, et al: RBFOX2 recognizes N(6)-methyladenosine to suppress transcription and block myeloid leukaemia differentiation. Nat Cell Biol. 25:1359–1368. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma L, Zhou X, Yao S, Zhang X, Mao J, Vona B, Fan L, Lou S, Li D, Wang L and Pan Y: METTL3-dependent m(6)A modification of PSEN1 mRNA regulates craniofacial development through the Wnt/β-catenin signaling pathway. Cell Death Dis. 15:2292024. View Article : Google Scholar : PubMed/NCBI | |
|
Li B, Xiong X, Xu J, Peng D, Nie G, Wen N, Wang Y and Lu J: METTL3-mediated m(6)A modification of lncRNA TSPAN12 promotes metastasis of hepatocellular carcinoma through SENP1-depentent deSUMOylation of EIF3I. Oncogene. 43:1050–1062. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ou X, Tan Y, Xie J, Yuan J, Deng X, Shao R, Song C, Cao X, Xie X, He R, et al: Methylation of GPRC5A promotes liver metastasis and docetaxel resistance through activating mTOR signaling pathway in triple negative breast cancer. Drug Resist Updat. 73:1010632024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang T, Qi J, Xue Z, Liu B, Liu J, Hu Q, Li Y, Ren J, Song H, Xu Y, et al: The m(6)A modification mediated-lncRNA POU6F2-AS1 reprograms fatty acid metabolism and facilitates the growth of colorectal cancer via upregulation of FASN. Mol Cancer. 23:552024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Geng X, Li Q, Xu J, Tan Y, Xiao M, Song J, Liu F, Fang C and Wang H: m6A modification in RNA: biogenesis, functions and roles in gliomas. J Exp Clin Cancer Res. 39:1922020. View Article : Google Scholar : PubMed/NCBI | |
|
van Tran N, Ernst FGM, Hawley BR, Zorbas C, Ulryck N, Hackert P, Bohnsack KE, Bohnsack MT, Jaffrey SR, Graille M and Lafontaine DLJ: The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 47:7719–7733. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Li Y, Zhou L, Zhou H, Ye L, Ou T, Hong H, Zheng S, Zhou Z, Wu K, et al: The m6A Reader YTHDF2 promotes bladder cancer progression by suppressing RIG-I-Mediated immune response. Cancer Res. 83:1834–1850. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ghazi T, Nagiah S and Chuturgoon AA: Fusaric acid decreases p53 expression by altering promoter methylation and m6A RNA methylation in human hepatocellular carcinoma (HepG2) cells. Epigenetics. 16:79–91. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shulman Z and Stern-Ginossar N: The RNA modification N6-methyladenosine as a novel regulator of the immune system. Nat Immunol. 21:501–512. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng W, Li M, Zhang L, Zhou C, Yu S, Peng X and Zhang W and Zhang W: New roles of N6-methyladenosine methylation system regulating the occurrence of non-alcoholic fatty liver disease with N6-methyladenosine-modified MYC. Front. Pharmacol. 13:9731162022.PubMed/NCBI | |
|
Jin S, Li M, Chang H, Wang R, Zhang Z, Zhang J, He Y and Ma H: The m6A demethylase ALKBH5 promotes tumor progression by inhibiting RIG-I expression and interferon alpha production through the IKKε/TBK1/IRF3 pathway in head and neck squamous cell carcinoma. Mol Cancer. 21:972022. View Article : Google Scholar : PubMed/NCBI | |
|
Ye G, Li J, Yu W, Xie Z, Zheng G, Liu W, Wang S, Cao Q, Lin J, Su Z, et al: ALKBH5 facilitates CYP1B1 mRNA degradation via m6A demethylation to alleviate MSC senescence and osteoarthritis progression. Exp Mol Med. 55:1743–1756. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Y and Zhuang X: m(6)A-binding YTHDF proteins promote stress granule formation. Nat Chem Biol. 16:955–963. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Khan D, Ramachandiran I, Vasu K, China A, Khan K, Cumbo F, Halawani D, Terenzi F, Zin I, Long B, et al: Homozygous EPRS1 missense variant causing hypomyelinating leukodystrophy-15 alters variant-distal mRNA m(6)A site accessibility. Nat Commun. 15:42842024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun R, Tian X, Li Y, Zhao Y, Wang Z, Hu Y, Zhang L, Wang Y, Gao D, Zheng S and Yao J: The m6A reader YTHDF3-mediated PRDX3 translation alleviates liver fibrosis. Redox Biol. 54:1023782022. View Article : Google Scholar : PubMed/NCBI | |
|
Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S and Tavazoie SF: HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 162:1299–1308. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang L, Lin W, Zhang C, Ash PEA, Verma M, Kwan J, van Vliet E, Yang Z, Cruz AL, Boudeau S, et al: Interaction of tau with HNRNPA2B1 and N(6)-methyladenosine RNA mediates the progression of tauopathy. Mol Cell. 81:4209–4227.e12. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Wen M and Cao X: Nuclear hnRNPA2B1 initiates and amplifies the innate immune response to DNA viruses. Science. 365:eaav07582019. View Article : Google Scholar : PubMed/NCBI | |
|
Miller C, Ealy A, Gregory A, Janarthanam C, Albers W, Richardson G, Jin H, Zenitsky G, Anantharam V, Kanthasamy A and Kanthasamy AG: Pathological α-synuclein dysregulates epitranscriptomic writer METTL3 to drive neuroinflammation in microglia. Cell Rep. 44:1156182025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang T, Zhang SW, Zhang SY and Ma QQ: m(6)Aexpress-Reader: Prediction of m(6)A regulated expression genes by integrating m(6)A sites and reader binding information in specific-context. Methods. 203:167–178. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Boriack-Sjodin PA, Ribich S and Copeland RA: RNA-modifying proteins as anticancer drug targets. Nat Rev Drug Discov. 17:435–453. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wei J, Harada BT, Lu D, Ma R, Gao B, Xu Y, Montauti E, Mani N, Chaudhuri SM, Gregory S, et al: HRD1-mediated METTL14 degradation regulates m(6)A mRNA modification to suppress ER proteotoxic liver disease. Mol Cell. 81:5052–5065.e6. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chai J, Wang Q, Qiu Q, Han G, Chen Y, Li W and Zhang H: YBX1 regulates the survival of chronic myeloid leukemia stem cells by modulating m(6)A-mediated YWHAZ stability. Cell Oncol (Dordr.). 46:451–464. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Evans MK, Matsui Y, Xu B, Willis C, Loome J, Milburn L, Fan Y, Pagala V and Peng JC: Ybx1 fine-tunes PRC2 activities to control embryonic brain development. Nat Commun. 11:40602020. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang WJ, Sun MH, Li XH, Lee SH, Heo G, Zhou D and Cui XS: Y-box binding protein 1 influences zygotic genome activation by regulating N6-methyladenosine in porcine embryos. J Cell Physiol. 238:1592–1604. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, He J, Lin L, Li Y and Xia Y: Neural stem cell extracellular vesicles carrying YBX1 inhibited neuronal pyroptosis through increasing m6A-modified GPR30 stability and expression in ischemic stroke. Transl Stroke Res. 16:262–279. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al: Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 20:285–295. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ramesh-Kumar D and Guil S: The IGF2BP family of RNA binding proteins links epitranscriptomics to cancer. Semin Cancer Biol 86(Pt 3). 18–31. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ying Y, Wu Y, Zhang F, Tang Y, Yi J, Ma X, Li J, Chen D, Wang X, Liu X, et al: Co-transcriptional R-loops-mediated epigenetic regulation drives growth retardation and docetaxel chemosensitivity enhancement in advanced prostate cancer. Mol Cancer. 23:792024. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Li W, Leng Y, Xu H, Xia Z and Wang Y: ALKBH5-Mediated M(6)A demethylation of G3BP1 attenuates ferroptosis via cytoplasmic retention of YBX1/p53 in diabetic myocardial ischemia-reperfusion injury. Adv Sci (Weinh). 12:e072542025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Shen S, Kang J, Sugai-Munson A, Xiao X, Zhang Y, Zhu J, Liu Z, McKay TB, Akeju O, et al: METTL3 is essential for exercise benefits in diabetic cardiomyopathy. Circulation. 152:327–345. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Xi Q, Yang G, He X, Zhuang H, Li L, Lin B, Wang L, Wang X, Fang C, Chen Q, et al: M(6)A-mediated upregulation of lncRNA TUG1 in liver cancer cells regulates the antitumor response of CD8(+) T cells and phagocytosis of macrophages. Adv Sci (Weinh). 11:e24006952024. View Article : Google Scholar : PubMed/NCBI | |
|
Yin H, Chen L, Piao S, Wang Y, Li Z, Lin Y, Tang X, Zhang H, Zhang H and Wang X: M6A RNA methylation-mediated RMRP stability renders proliferation and progression of non-small cell lung cancer through regulating TGFBR1/SMAD2/SMAD3 pathway. Cell Death Differ. 30:605–617. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Han Y, Wu C, Wang S, Chen M, Xu Q, Wei H, Zhou X and Wang G: m6A-modified LINC02418 induces transcriptional and post-transcriptional modification of CTNNB1 via interacting with YBX1 and IGF2BP1 in colorectal cancer. Cell Death Discov. 11:1012025. View Article : Google Scholar : PubMed/NCBI | |
|
Gunasekaran VP, Nishi K, Sivakumar D, Sivaraman T and Mathan G: Identification of 2,4-dihydroxy-5-pyrimidinyl imidothiocarbomate as a novel inhibitor to Y box binding protein-1 (YB-1) and its therapeutic actions against breast cancer. Eur J Pharm Sci. 116:2–14. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
He M, Li T, Wang A, Liu Y, Wang X, Liu Z, Xie J, Wang Y, Wang Y, Ren Z, et al: MARCH8/NSUN6/ROS-mediated DNA damage positive feedback loop regulates cisplatin resistance in osteosarcoma. Cell Death Differ. Jul 19–2025.(Epub ahead of print). View Article : Google Scholar | |
|
Huang H, Fang L, Zhu C, Lv J, Xu P, Chen Z, Zhang Z, Wang J, Wang W and Xu Z: YBX1 promotes 5-Fluorouracil resistance in gastric cancer via m5C-dependent ATG9A mRNA stabilization through autophagy. Oncogene. 44:2357–2371. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Li D, Chu X, Liu W, Ma Y, Tian X and Yang Y: The regulatory roles of RNA-binding proteins in the tumour immune microenvironment of gastrointestinal malignancies. RNA Biol. 22:1–14. 2025. View Article : Google Scholar | |
|
Ma L, Singh J and Schekman R: Two RNA-binding proteins mediate the sorting of miR223 from mitochondria into exosomes. Elife. 12:e858782023. View Article : Google Scholar : PubMed/NCBI | |
|
Dinh NTM, Nguyen TM, Park MK and Lee CH: Y-Box binding protein 1: Unraveling the multifaceted role in cancer development and therapeutic potential. Int J Mol Sci. 25:7172024. View Article : Google Scholar : PubMed/NCBI | |
|
Alkrekshi A, Wang W, Rana PS, Markovic V and Sossey-Alaoui K: A comprehensive review of the functions of YB-1 in cancer stemness, metastasis and drug resistance. Cell Signal. 85:1100732021. View Article : Google Scholar : PubMed/NCBI |