Current Progress of 5-Methylcytosine RNA Methylation in Non-Neoplastic Kidney Diseases

ZHANGChen, ZHAOZixia, WUSi, LUANJunjun, ZHOUHua

Acta Academiae Medicinae Sinicae ›› 2025, Vol. 47 ›› Issue (1) : 86-94.

PDF(677 KB)
Home Journals Acta Academiae Medicinae Sinicae
Acta Academiae Medicinae Sinicae

Abbreviation (ISO4): Acta Academiae Medicinae Sinicae      Editor in chief: Xuetao CAO

About  /  Aim & scope  /  Editorial board  /  Indexed  /  Contact  / 
PDF(677 KB)
Acta Academiae Medicinae Sinicae ›› 2025, Vol. 47 ›› Issue (1) : 86-94. DOI: 10.3881/j.issn.1000-503X.16053
Review Articles

Current Progress of 5-Methylcytosine RNA Methylation in Non-Neoplastic Kidney Diseases

Author information +
History +

Abstract

RNA methylation is a key process in the epigenetic regulation of post-transcriptional gene expression.5-Methylcytosine(m5C)is a type of RNA methylation,commonly existing in eukaryotic mRNA and non-coding RNAs.It mainly regulates transfer RNA stability,ribosomal RNA assembly,and mRNA translation,stability,and translation.RNA methylation is dynamically reversible and regulated by methyltransferase,demethylase,and methylation recognition protein.It has been confirmed that aberrant m5C RNA methylation is involved in the pathogenesis of non-neoplastic kidney diseases.This article summarizes the current progress of m5C RNA methylation associated with non-neoplastic acute and chronic kidney diseases,aiming to provide potential targets for the diagnosis and treatment of such diseases.

Key words

epigenetics / RNA methylation / RNA / 5-methylcytosine / kidney diseases

Cite this article

Download Citations
ZHANG Chen , ZHAO Zixia , WU Si , et al . Current Progress of 5-Methylcytosine RNA Methylation in Non-Neoplastic Kidney Diseases[J]. Acta Academiae Medicinae Sinicae. 2025, 47(1): 86-94 https://doi.org/10.3881/j.issn.1000-503X.16053

References

[1]
Bouchard J, Acharya A, Cerda J, et al. A prospective international multicenter study of AKI in the intensive care unit[J]. Clin J Am Soc Nephrol, 2015, 10(8):1324-1331.DOI:10.2215/CJN.04360514.
[2]
Wang L, Xu X, Zhang M, et al. Prevalence of chronic kidney disease in China:results from the sixth China chronic disease and risk factor surveillance[J]. JAMA Intern Med, 2023, 183(4):298-310.DOI:10.1001/jamainternmed.2022.6817.
[3]
Zhang L, Wang F, Wang L, et al. Prevalence of chronic kidney disease in China:a cross-sectional survey[J]. Lancet, 2012, 379(9818):815-822.DOI:10.1016/S0140-6736(12)60033-6.
[4]
GBD Chronic Kidney Disease Collaboration. and national burden of chronic kidney disease,1990-2017:a systematic analysis for the Global Burden of Disease Study 2017[J]. Lancet, 2020, 395(10225):709-733.DOI:10.1016/S0140-6736(20)30045-3.
[5]
Ortiz A. RICORS2040:the need for collaborative research in chronic kidney disease[J]. Clin Kidney J, 2022, 15(3):372-387.DOI:10.1093/ckj/sfab170.
[6]
Luan J, Kopp JB, Zhou H. N6-methyladenine RNA methylation epigenetic modification and kidney diseases[J]. Kidney Int Rep, 2023, 8(1):36-50.DOI:10.1016/j.ekir.2022.10.009.
RNA methylation modification is a rapidly developing field in epigenetics. N6-methyladensine (mA) is the most common internal modification in eukaryotic mRNA. mA group regulates RNA splicing, stability, translocation, and translation. Enzymes catalyzing this process were termed as writers, erasers, and readers. Recent studies have focused on exploring the role of RNA methylation in human diseases. RNA methylation modifications, particularly mA, play important roles in the pathogenesis of kidney diseases. In this review, we provide a brief description of mA and summarize the impact of mA on acute and chronic kidney disease (CKD) and possible future study directions for this research.© 2022 Published by Elsevier Inc. on behalf of the International Society of Nephrology.
[7]
Cantara WA, Crain PF, Rozenski J, et al. The RNA modification database,RNAMDB:2011 update[J]. Nucleic Acids Res, 2011, 39(Database issue):D195-D201.DOI:10.1093/nar/gkq1028.
[8]
Tuorto F, Liebers R, Musch T, et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis[J]. Nat Struct Mol Biol, 2012, 19(9):900-905.DOI:10.1038/nsmb.2357.
The function of cytosine-C5 methylation, a widespread modification of tRNAs, has remained obscure, particularly in mammals. We have now developed a mouse strain defective in cytosine-C5 tRNA methylation, by disrupting both the Dnmt2 and the NSun2 tRNA methyltransferases. Although the lack of either enzyme alone has no detectable effects on mouse viability, double mutants showed a synthetic lethal interaction, with an underdeveloped phenotype and impaired cellular differentiation. tRNA methylation analysis of the double-knockout mice demonstrated complementary target-site specificities for Dnmt2 and NSun2 and a complete loss of cytosine-C5 tRNA methylation. Steady-state levels of unmethylated tRNAs were substantially reduced, and loss of Dnmt2 and NSun2 was further associated with reduced rates of overall protein synthesis. These results establish a biologically important function for cytosine-C5 tRNA methylation in mammals and suggest that this modification promotes mouse development by supporting protein synthesis.
[9]
Chan CTY, Pang YLJ, Deng W, et al. Reprogramming of tRNA modifications controls the oxidative stress response by codon-biased translation of proteins[J]. Nat Commun, 2012,3:937.DOI:10.1038/ncomms1938.
[10]
Janin M, Ortiz-Barahona V, de Moura MC, et al. Epigenetic loss of RNA-methyltransferase NSUN5 in glioma targets ribosomes to drive a stress adaptive translational program[J]. Acta Neuropathol, 2019, 138(6):1053-1074.DOI:10.1007/s00401-019-02062-4.
Tumors have aberrant proteomes that often do not match their corresponding transcriptome profiles. One possible cause of this discrepancy is the existence of aberrant RNA modification landscapes in the so-called epitranscriptome. Here, we report that human glioma cells undergo DNA methylation-associated epigenetic silencing of NSUN5, a candidate RNA methyltransferase for 5-methylcytosine. In this setting, NSUN5 exhibits tumor-suppressor characteristics in vivo glioma models. We also found that NSUN5 loss generates an unmethylated status at the C3782 position of 28S rRNA that drives an overall depletion of protein synthesis, and leads to the emergence of an adaptive translational program for survival under conditions of cellular stress. Interestingly, NSUN5 epigenetic inactivation also renders these gliomas sensitive to bioactivatable substrates of the stress-related enzyme NQO1. Most importantly, NSUN5 epigenetic inactivation is a hallmark of glioma patients with long-term survival for this otherwise devastating disease.
[11]
Yang X, Yang Y, Sun BF, et al. 5-Methylcytosine promotes mRNA export-NSUN2 as the methyltransferase and ALYREF as an m5C reader[J]. Cell Res, 2017, 27(5):606-625.DOI:10.1038/cr.2017.55.
5-methylcytosine (mC) is a post-transcriptional RNA modification identified in both stable and highly abundant tRNAs and rRNAs, and in mRNAs. However, its regulatory role in mRNA metabolism is still largely unknown. Here, we reveal that mC modification is enriched in CG-rich regions and in regions immediately downstream of translation initiation sites and has conserved, tissue-specific and dynamic features across mammalian transcriptomes. Moreover, mC formation in mRNAs is mainly catalyzed by the RNA methyltransferase NSUN2, and mC is specifically recognized by the mRNA export adaptor ALYREF as shown by in vitro and in vivo studies. NSUN2 modulates ALYREF's nuclear-cytoplasmic shuttling, RNA-binding affinity and associated mRNA export. Dysregulation of ALYREF-mediated mRNA export upon NSUN2 depletion could be restored by reconstitution of wild-type but not methyltransferase-defective NSUN2. Our study provides comprehensive mC profiles of mammalian transcriptomes and suggests an essential role for mC modification in mRNA export and post-transcriptional regulation.
[12]
Yang Y, Wang L, Han X, et al. RNA 5-methylcytosine facilitates the maternal-to-zygotic transition by preventing maternal mRNA decay[J]. Mol Cell, 2019, 75(6):1188-1202.DOI:10.1016/j.molcel.2019.06.033.
The maternal-to-zygotic transition (MZT) is a conserved and fundamental process during which the maternal environment is converted to an environment of embryonic-driven development through dramatic reprogramming. However, how maternally supplied transcripts are dynamically regulated during MZT remains largely unknown. Herein, through genome-wide profiling of RNA 5-methylcytosine (mC) modification in zebrafish early embryos, we found that mC-modified maternal mRNAs display higher stability than non-mC-modified mRNAs during MZT. We discovered that Y-box binding protein 1 (Ybx1) preferentially recognizes mC-modified mRNAs through π-π interactions with a key residue, Trp45, in Ybx1's cold shock domain (CSD), which plays essential roles in maternal mRNA stability and early embryogenesis of zebrafish. Together with the mRNA stabilizer Pabpc1a, Ybx1 promotes the stability of its target mRNAs in an mC-dependent manner. Our study demonstrates an unexpected mechanism of RNA mC-regulated maternal mRNA stabilization during zebrafish MZT, highlighting the critical role of mC mRNA modification in early development.Copyright © 2019 Elsevier Inc. All rights reserved.
[13]
Young JI, Hong EP, Castle JC, et al. Regulation of RNA splicing by the methylation-dependent transcriptional repressor methyl-CpG binding protein 2[J]. Proc Natl Acad Sci U S A, 2005, 102(49):17551-17558.DOI:10.1073/pnas.0507856102.
[14]
Chen H, Yang H, Zhu X, et al. m5C modification of mRNA serves a DNA damage code to promote homologous recombination[J]. Nat Commun, 2020, 11(1):2834.DOI:10.1038/s41467-020-16722-7.
Recruitment of DNA repair proteins to DNA damage sites is a critical step for DNA repair. Post-translational modifications of proteins at DNA damage sites serve as DNA damage codes to recruit specific DNA repair factors. Here, we show that mRNA is locally modified by mC at sites of DNA damage. The RNA methyltransferase TRDMT1 is recruited to DNA damage sites to promote mC induction. Loss of TRDMT1 compromises homologous recombination (HR) and increases cellular sensitivity to DNA double-strand breaks (DSBs). In the absence of TRDMT1, RAD51 and RAD52 fail to localize to sites of reactive oxygen species (ROS)-induced DNA damage. In vitro, RAD52 displays an increased affinity for DNA:RNA hybrids containing mC-modified RNA. Loss of TRDMT1 in cancer cells confers sensitivity to PARP inhibitors in vitro and in vivo. These results reveal an unexpected TRDMT1-mC axis that promotes HR, suggesting that post-transcriptional modifications of RNA can also serve as DNA damage codes to regulate DNA repair.
[15]
Xue S, Xu H, Sun Z, et al. Depletion of TRDMT1 affects 5-methylcytosine modification of mRNA and inhibits HEK293 cell proliferation and migration[J]. Biochem Biophys Res Commun, 2019, 520(1):60-66.DOI:10.1016/j.bbrc.2019.09.098.
[16]
Zou F, Tu R, Duan B, et al. Drosophila YBX1 homolog YPS promotes ovarian germ line stem cell development by preferentially recognizing 5-methylcytosine RNAs[J]. Proc Natl Acad Sci U S A, 2020, 117(7):3603-3609.DOI:10.1073/pnas.1910862117.
[17]
Luo Y, Feng J, Xu Q, et al. NSun2 deficiency protects endothelium from inflammation via mRNA methylation of ICAM-1[J]. Circ Res, 2016, 118(6):944-956.DOI:10.1161/CIRCRESAHA.115.307674.
[18]
Guo G, Wang H, Shi X, et al. Disease activity-associated alteration of mRNA m5C methylation in CD4+ T cells of systemic lupus erythematosus[J]. Front Cell Dev Biol, 2020,8:430.DOI:10.3389/fcell.2020.00430.
[19]
Chen X, Li A, Sun BF, et al. 5-Methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs[J]. Nat Cell Biol, 2019, 21(8):978-990.DOI:10.1038/s41556-019-0361-y.
Although 5-methylcytosine (mC) is a widespread modification in RNAs, its regulation and biological role in pathological conditions (such as cancer) remain unknown. Here, we provide the single-nucleotide resolution landscape of messenger RNA mC modifications in human urothelial carcinoma of the bladder (UCB). We identify numerous oncogene RNAs with hypermethylated mC sites causally linked to their upregulation in UCBs and further demonstrate YBX1 as an mC 'reader' recognizing mC-modified mRNAs through the indole ring of W65 in its cold-shock domain. YBX1 maintains the stability of its target mRNA by recruiting ELAVL1. Moreover, NSUN2 and YBX1 are demonstrated to drive UCB pathogenesis by targeting the mC methylation site in the HDGF 3' untranslated region. Clinically, a high coexpression of NUSN2, YBX1 and HDGF predicts the poorest survival. Our findings reveal an unprecedented mechanism of RNA mC-regulated oncogene activation, providing a potential therapeutic strategy for UCB.
[20]
Zaccara S, Ries RJ, Jaffrey SR. Reading,writing and erasing mRNA methylation[J]. Nat Rev Mol Cell Biol, 2019, 20(10):608-624.DOI:10.1038/s41580-019-0168-5.
[21]
Arguello AE, Li A, Sun X, et al. Reactivity-dependent profiling of RNA 5-methylcytidine dioxygenases[J]. Nat Commun, 2022, 13(1):4176.DOI:10.1038/s41467-022-31876-2.
Epitranscriptomic RNA modifications can regulate fundamental biological processes, but we lack approaches to map modification sites and probe writer enzymes. Here we present a chemoproteomic strategy to characterize RNA 5-methylcytidine (mC) dioxygenase enzymes in their native context based upon metabolic labeling and activity-based crosslinking with 5-ethynylcytidine (5-EC). We profile mC dioxygenases in human cells including ALKBH1 and TET2 and show that ALKBH1 is the major hmC- and fC-forming enzyme in RNA. Further, we map ALKBH1 modification sites transcriptome-wide using 5-EC-iCLIP and ARP-based sequencing to identify ALKBH1-dependent mC oxidation in a variety of tRNAs and mRNAs and analyze ALKBH1 substrate specificity in vitro. We also apply targeted pyridine borane-mediated sequencing to measure fC sites on select tRNA. Finally, we show that fC at the wobble position of tRNA-Leu-CAA plays a role in decoding Leu codons under stress. Our work provides powerful chemical approaches for studying RNA mC dioxygenases and mapping oxidative mC modifications and reveals the existence of novel epitranscriptomic pathways for regulating RNA function.© 2022. The Author(s).
[22]
Zaccara S, Jaffrey SR. A unified model for the function of YTHDF proteins in regulating m6A-modified mRNA[J]. Cell, 2020, 181(7):1582-1595.e18.DOI:10.1016/j.cell.2020.05.012.
N-methyladenosine (mA) is the most abundant mRNA nucleotide modification and regulates critical aspects of cellular physiology and differentiation. mA is thought to mediate its effects through a complex network of interactions between different mA sites and three functionally distinct cytoplasmic YTHDF mA-binding proteins (DF1, DF2, and DF3). In contrast to the prevailing model, we show that DF proteins bind the same mA-modified mRNAs rather than different mRNAs. Furthermore, we find that DF proteins do not induce translation in HeLa cells. Instead, the DF paralogs act redundantly to mediate mRNA degradation and cellular differentiation. The ability of DF proteins to regulate stability and differentiation becomes evident only when all three DF paralogs are depleted simultaneously. Our study reveals a unified model of mA function in which all mA-modified mRNAs are subjected to the combined action of YTHDF proteins in proportion to the number of mA sites.Copyright © 2020 Elsevier Inc. All rights reserved.
[23]
Liao H, Gaur A, McConie H, et al. Human NOP2/NSUN1 regulates ribosome biogenesis through non-catalytic complex formation with box C/D snoRNPs[J]. Nucleic Acids Res, 2022, 50(18):10695-10716.DOI:10.1093/nar/gkac817.
[24]
Wang Y, Wei J, Feng L, et al. Aberrant m5C hypermethylation mediates intrinsic resistance to gefitinib through NSUN2/YBX1/QSOX1 axis in EGFR-mutant non-small-cell lung cancer[J]. Mol Cancer, 2023, 22(1):81.DOI:10.1186/s12943-023-01780-4.
RNA 5-methylcytosine (mC) modification plays critical roles in the pathogenesis of various tumors. However, the function and molecular mechanism of RNA mC modification in tumor drug resistance remain unclear.The correlation between RNA mC methylation, mC writer NOP2/Sun RNA methyltransferase family member 2 (NSUN2) and EGFR-TKIs resistance was determined in non-small-cell lung cancer (NSCLC) cell lines and patient samples. The effects of NSUN2 on EGFR-TKIs resistance were investigated by gain- and loss-of-function assays in vitro and in vivo. RNA-sequencing (RNA-seq), RNA bisulfite sequencing (RNA-BisSeq) and mC methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) were performed to identify the target gene of NSUN2 involved in EGFR-TKIs resistance. Furthermore, the regulatory mechanism of NSUN2 modulating the target gene expression was investigated by functional rescue and puromycin incorporation assays.RNA mC hypermethylation and NSUN2 were significantly correlated with intrinsic resistance to EGFR-TKIs. Overexpression of NSUN2 resulted in gefitinib resistance and tumor recurrence, while genetic inhibition of NSUN2 led to tumor regression and overcame intrinsic resistance to gefitinib in vitro and in vivo. Integrated RNA-seq and mC-BisSeq analyses identified quiescin sulfhydryl oxidase 1 (QSOX1) as a potential target of aberrant mC modification. NSUN2 methylated QSOX1 coding sequence region, leading to enhanced QSOX1 translation through mC reader Y-box binding protein 1 (YBX1).Our study reveals a critical function of aberrant RNA mC modification via the NSUN2-YBX1-QSOX1 axis in mediating intrinsic resistance to gefitinib in EGFR-mutant NSCLC.© 2023. The Author(s).
[25]
Mei L, Shen C, Miao R, et al. RNA methyltransferase NSUN2 promotes gastric cancer cell proliferation by repressing p57Kip2 by an m5C-dependent manner[J]. Cell Death Dis, 2020, 11(4):270.DOI:10.1038/s41419-020-2487-z.
[26]
Blanco S, Dietmann S, Flores JV, et al. Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders[J]. EMBO J, 2014, 33(18):2020-2039.DOI:10.15252/embj.201489282.
Mutations in the cytosine-5 RNA methyltransferase NSun2 cause microcephaly and other neurological abnormalities in mice and human. How post-transcriptional methylation contributes to the human disease is currently unknown. By comparing gene expression data with global cytosine-5 RNA methylomes in patient fibroblasts and NSun2-deficient mice, we find that loss of cytosine-5 RNA methylation increases the angiogenin-mediated endonucleolytic cleavage of transfer RNAs (tRNA) leading to an accumulation of 5' tRNA-derived small RNA fragments. Accumulation of 5' tRNA fragments in the absence of NSun2 reduces protein translation rates and activates stress pathways leading to reduced cell size and increased apoptosis of cortical, hippocampal and striatal neurons. Mechanistically, we demonstrate that angiogenin binds with higher affinity to tRNAs lacking site-specific NSun2-mediated methylation and that the presence of 5' tRNA fragments is sufficient and required to trigger cellular stress responses. Furthermore, the enhanced sensitivity of NSun2-deficient brains to oxidative stress can be rescued through inhibition of angiogenin during embryogenesis. In conclusion, failure in NSun2-mediated tRNA methylation contributes to human diseases via stress-induced RNA cleavage. © 2014 The Authors. Published under the terms of the CC BY 4.0 license.
[27]
Chellamuthu A, Gray SG. The RNA methyltransferase NSUN2 and its potential roles in cancer[J]. Cells, 2020, 9(8):1758.DOI:10.3390/cells9081758.
[28]
Haag S, Sloan KE, Ranjan N, et al. NSUN3 and ABH1 modify the wobble position of mt-tRNAMet to expand codon recognition in mitochondrial translation[J]. EMBO J, 2016, 35(19):2104-2119.DOI:10.15252/embj.201694885.
Mitochondrial gene expression uses a non-universal genetic code in mammals. Besides reading the conventional AUG codon, mitochondrial (mt-)tRNA mediates incorporation of methionine on AUA and AUU codons during translation initiation and on AUA codons during elongation. We show that the RNA methyltransferase NSUN3 localises to mitochondria and interacts with mt-tRNA to methylate cytosine 34 (C34) at the wobble position. NSUN3 specifically recognises the anticodon stem loop (ASL) of the tRNA, explaining why a mutation that compromises ASL basepairing leads to disease. We further identify ALKBH1/ABH1 as the dioxygenase responsible for oxidising mC34 of mt-tRNA to generate an fC34 modification. In vitro codon recognition studies with mitochondrial translation factors reveal preferential utilisation of mC34 mt-tRNA in initiation. Depletion of either NSUN3 or ABH1 strongly affects mitochondrial translation in human cells, implying that modifications generated by both enzymes are necessary for mt-tRNA function. Together, our data reveal how modifications in mt-tRNA are generated by the sequential action of NSUN3 and ABH1, allowing the single mitochondrial tRNA to recognise the different codons encoding methionine.© 2016 The Authors. Published under the terms of the CC BY NC ND 4.0 license.
[29]
Metodiev MD, Spåhr H, Loguercio Polosa P, et al. NSUN4 is a dual function mitochondrial protein required for both methylation of 12S rRNA and coordination of mitoribosomal assembly[J]. PLoS Genet, 2014, 10(2):e1004110.DOI:10.1371/journal.pgen.1004110.
[30]
Yang L, Ren Z, Yan S, et al. Nsun4 and Mettl3 mediated translational reprogramming of Sox9 promotes BMSC chondrogenic differentiation[J]. Commun Biol, 2022, 5(1):495.DOI:10.1038/s42003-022-03420-x.
The chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) has been used in the treatment and repair of cartilage defects; however, the in-depth regulatory mechanisms by which RNA modifications are involved in this process are still poorly understood. Here, we found that Sox9, a critical transcription factor that mediates chondrogenic differentiation, exhibited enhanced translation by ribosome sequencing in chondrogenic pellets, which was accompanied by increased 5-methylcytosine (mC) and N6-methyladenosine (mA) levels. Nsun4-mediated mC and Mettl3-mediated mA modifications were required for Sox9-regulated chondrogenic differentiation. Interestingly, we showed that in the 3'UTR of Sox9 mRNA, Nsun4 catalyzed the mC modification and Mettl3 catalyzed the mA modification. Furthermore, we found that Nsun4 and Mettl3 co-regulated the translational reprogramming of Sox9 via the formation of a complex. Surface plasmon resonance (SPR) assays showed that this complex was assembled along with the recruitment of Ythdf2 and eEF1α-1. Moreover, BMSCs overexpressing Mettl3 and Nsun4 can promote the repair of cartilage defects in vivo. Taken together, our study demonstrates that mC and mA co-regulate the translation of Sox9 during the chondrogenic differentiation of BMSCs, which provides a therapeutic target for clinical implications.© 2022. The Author(s).
[31]
Heissenberger C, Liendl L, Nagelreiter F, et al. Loss of the ribosomal RNA methyltransferase NSUN5 impairs global protein synthesis and normal growth[J]. Nucleic Acids Res, 2019, 47(22):11807-11825.DOI:10.1093/nar/gkz1043.
Modifications of ribosomal RNA expand the nucleotide repertoire and thereby contribute to ribosome heterogeneity and translational regulation of gene expression. One particular m5C modification of 25S ribosomal RNA, which is introduced by Rcm1p, was previously shown to modulate stress responses and lifespan in yeast and other small organisms. Here, we report that NSUN5 is the functional orthologue of Rcm1p, introducing m5C3782 into human and m5C3438 into mouse 28S ribosomal RNA. Haploinsufficiency of the NSUN5 gene in fibroblasts from William Beuren syndrome patients causes partial loss of this modification. The N-terminal domain of NSUN5 is required for targeting to nucleoli, while two evolutionary highly conserved cysteines mediate catalysis. Phenotypic consequences of NSUN5 deficiency in mammalian cells include decreased proliferation and size, which can be attributed to a reduction in total protein synthesis by altered ribosomes. Strikingly, Nsun5 knockout in mice causes decreased body weight and lean mass without alterations in food intake, as well as a trend towards reduced protein synthesis in several tissues. Together, our findings emphasize the importance of single RNA modifications for ribosome function and normal cellular and organismal physiology.© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.
[32]
Wang Y, Jiang T, Xu J, et al. Mutations in RNA methyltransferase gene NSUN5 confer high risk of outflow tract malformation[J]. Front Cell Devl Biol, 2021,9:623394.DOI:10.3389/fcell.2021.623394.
[33]
Ding C, Lu J, Li J, et al. RNA-methyltransferase Nsun5 controls the maternal-to-zygotic transition by regulating maternal mRNA stability[J]. Clin Transl Med, 2022, 12(12):e1137.DOI:10.1002/ctm2.1137.
RNA modification-induced ovarian dysgenesis appears to be necessary for ovary development. However, how m C (5-methylcytosine)-coordinating modificatory transcripts are dynamically regulated during oogenesis, and ovarian development is unknown. The purpose of this study was to determine whether NOP2/Sun RNA methyltransferase 5 (Nsun5) deletion leads to suppression of ovarian function and arrest of embryonic development. The regulation of mRNA decay and stability by m C modification is essential at multiple stages during the maternal-to-zygotic (MZT) transition.Mouse ovaries and oocytes with Nsun5 and the KGN cell line were subjected to m C identification, alternative splicing analysis and protein expression. BS-m C-seq, real-time polymerase chain reaction, Western blot, immunofluorescence and actinomycin D treatment assays were used. In particular, BS-m C-seq revealed a dynamic pattern of m C sites and genes in the ovaries between Nsun5 and WT mice at the 2-month and 6-month stages. Diverse bioinformatic tools were employed to identify target genes for Nsun5.Here, a maternal mRNA stability study showed that deletion of the m C methyltransferase Nsun5 obstructs follicular development and ovarian function, which leads directly to inhibition of embryogenesis and embryo development. Dynamic analysis of m C revealed that the level of m C decreased in a time-dependent manner after Nsun5 knockout. Regarding the molecular mechanism, we found that Nsun5 deficiency caused a m C decline in the exon and 3'UTR regions that influenced the translation efficiency of Mitotic arrest deficient 2 like 2 (MAD2L2) and Growth differentiation factor 9 (GDF9) in the ovary. Mechanistic investigation of alternative splicing indicated that Nsun5 triggers aberrant events in the exon region of Brd8.Nsun5 loss arrests follicular genesis and development in ovarian aging, indicating that Nsun5/m C-regulated maternal mRNA stabilization is essential for MZT transition.© 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.
[34]
Hussain S. The emerging roles of cytosine-5 methylation in mRNAs[J]. Trends Genet, 2021, 37(6):498-500.DOI:10.1016/j.tig.2021.02.001.
Recent studies have unequivocally confirmed the presence of 5-methylcytosine (m5C) in mammalian mRNAs while indicating significant functional roles for this internal base modification type. Here, a brief history of m5C epitranscriptome research and a discussion of the important ways in which the field may now progress is presented.Copyright © 2021 Elsevier Ltd. All rights reserved.
[35]
Selmi T, Hussain S, Dietmann S, et al. Sequence- and structure-specific cytosine-5 mRNA methylation by NSUN6[J]. Nucleic Acids Res, 2021, 49(2):1006-1022.DOI:10.1093/nar/gkaa1193.
The highly abundant N6-methyladenosine (m6A) RNA modification affects most aspects of mRNA function, yet the precise function of the rarer 5-methylcytidine (m5C) remains largely unknown. Here, we map m5C in the human transcriptome using methylation-dependent individual-nucleotide resolution cross-linking and immunoprecipitation (miCLIP) combined with RNA bisulfite sequencing. We identify NSUN6 as a methyltransferase with strong substrate specificity towards mRNA. NSUN6 primarily targeted three prime untranslated regions (3'UTR) at the consensus sequence motif CTCCA, located in loops of hairpin structures. Knockout and rescue experiments revealed enhanced mRNA and translation levels when NSUN6-targeted mRNAs were methylated. Ribosome profiling further demonstrated that NSUN6-specific methylation correlated with translation termination. While NSUN6 was dispensable for mouse embryonic development, it was down-regulated in human tumours and high expression of NSUN6 indicated better patient outcome of certain cancer types. In summary, our study identifies NSUN6 as a methyltransferase targeting mRNA, potentially as part of a quality control mechanism involved in translation termination fidelity.© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.
[36]
Haag S, Warda AS, Kretschmer J, et al. NSUN6 is a human RNA methyltransferase that catalyzes formation of m5C72 in specific tRNAs[J]. RNA, 2015, 21(9):1532-1543.DOI:10.1261/rna.051524.115.
Many cellular RNAs require modification of specific residues for their biogenesis, structure, and function. 5-methylcytosine (m(5)C) is a common chemical modification in DNA and RNA but in contrast to the DNA modifying enzymes, only little is known about the methyltransferases that establish m(5)C modifications in RNA. The putative RNA methyltransferase NSUN6 belongs to the family of Nol1/Nop2/SUN domain (NSUN) proteins, but so far its cellular function has remained unknown. To reveal the target spectrum of human NSUN6, we applied UV crosslinking and analysis of cDNA (CRAC) as well as chemical crosslinking with 5-azacytidine. We found that human NSUN6 is associated with tRNAs and acts as a tRNA methyltransferase. Furthermore, we uncovered tRNA(Cys) and tRNA(Thr) as RNA substrates of NSUN6 and identified the cytosine C72 at the 3' end of the tRNA acceptor stem as the target nucleoside. Interestingly, target recognition in vitro depends on the presence of the 3'-CCA tail. Together with the finding that NSUN6 localizes to the cytoplasm and largely colocalizes with marker proteins for the Golgi apparatus and pericentriolar matrix, our data suggest that NSUN6 modifies tRNAs in a late step in their biogenesis. © 2015 Haag et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.
[37]
Aguilo F, Li S, Balasubramaniyan N, et al. Deposition of 5-methylcytosine on enhancer RNAs enables the coactivator function of PGC-1α[J]. Cell Rep, 2016, 14(3):479-492.DOI:10.1016/j.celrep.2015.12.043.
The Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) is a transcriptional co-activator that plays a central role in adapted metabolic responses. PGC-1α is dynamically methylated and unmethylated at the residue K779 by the methyltransferase SET7/9 and the Lysine Specific Demethylase 1A (LSD1), respectively. Interactions of methylated PGC-1α[K779me] with the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex, the Mediator members MED1 and MED17, and the NOP2/Sun RNA methytransferase 7 (NSUN7) reinforce transcription, and are concomitant with the m(5)C mark on enhancer RNAs (eRNAs). Consistently, loss of Set7/9 and NSun7 in liver cell model systems resulted in depletion of the PGC-1α target genes Pfkl, Sirt5, Idh3b, and Hmox2, which was accompanied by a decrease in the eRNAs levels associated with these loci. Enrichment of m(5)C within eRNA species coincides with metabolic stress of fasting in vivo. Collectively, these findings illustrate the complex epigenetic circuitry imposed by PGC-1α at the eRNA level to fine-tune energy metabolism.Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.
[38]
Ortiz-Barahona V, Soler M, Davalos V, et al. Epigenetic inactivation of the 5-methylcytosine RNA methyltransferase NSUN7 is associated with clinical outcome and therapeutic vulnerability in liver cancer[J]. Mol Cancer, 2023, 22(1):83.DOI:10.1186/s12943-023-01785-z.
RNA modifications are important regulators of transcript activity and an increasingly emerging body of data suggests that the epitranscriptome and its associated enzymes are altered in human tumors.Combining data mining and conventional experimental procedures, NSUN7 methylation and expression status was assessed in liver cancer cell lines and primary tumors. Loss-of-function and transfection-mediated recovery experiments coupled with RNA bisulfite sequencing and proteomics determined the activity of NSUN7 in downstream targets and drug sensitivity.In this study, the initial screening for genetic and epigenetic defects of 5-methylcytosine RNA methyltransferases in transformed cell lines, identified that the NOL1/NOP2/Sun domain family member 7 (NSUN7) undergoes promoter CpG island hypermethylation-associated with transcriptional silencing in a cancer-specific manner. NSUN7 epigenetic inactivation was common in liver malignant cells and we coupled bisulfite conversion of cellular RNA with next-generation sequencing (bsRNA-seq) to find the RNA targets of this poorly characterized putative RNA methyltransferase. Using knock-out and restoration-of-function models, we observed that the mRNA of the coiled-coil domain containing 9B (CCDC9B) gene required NSUN7-mediated methylation for transcript stability. Most importantly, proteomic analyses determined that CCDC9B loss impaired protein levels of its partner, the MYC-regulator Influenza Virus NS1A Binding Protein (IVNS1ABP), creating sensitivity to bromodomain inhibitors in liver cancer cells exhibiting NSUN7 epigenetic silencing. The DNA methylation-associated loss of NSUN7 was also observed in primary liver tumors where it was associated with poor overall survival. Interestingly, NSUN7 unmethylated status was enriched in the immune active subclass of liver tumors.The 5-methylcytosine RNA methyltransferase NSUN7 undergoes epigenetic inactivation in liver cancer that prevents correct mRNA methylation. Furthermore, NSUN7 DNA methylation-associated silencing is associated with clinical outcome and distinct therapeutic vulnerability.© 2023. The Author(s).
[39]
Delatte B, Wang F, Ngoc LV, et al. RNA biochemistry.Transcriptome-wide distribution and function of RNA hydroxymethylcytosine[J]. Science, 2016, 351(6270):282-285.DOI:10.1126/science.aac5253.
[40]
Shen Q, Zhang Q, Shi Y, et al. TET2 promotes pathogen infection-induced myelopoiesis through mRNA oxidation[J]. Nature, 2018, 554(7690):123-127.DOI:10.1038/nature25434.
[41]
Shen H, Ontiveros RJ, Owens MC, et al. TET-mediated 5-methylcytosine oxidation in tRNA promotes translation[J]. J Biol Chem, 2021,296:100087.DOI:10.1074/jbc.RA120.014226.
[42]
Wang JZ, Zhu W, Han J, et al. The role of the HIF-1α/ALYREF/PKM2 axis in glycolysis and tumorigenesis of bladder cancer[J]. Cancer Commun(Lond), 2021, 41(7):560-575.DOI:10.1002/cac2.12158.
[43]
Li M, Tao Z, Zhao Y, et al. 5-Methylcytosine RNA methyltransferases and their potential roles in cancer[J]. J Transl Med, 2022, 20(1):214.DOI:10.1186/s12967-022-03427-2.
In recent years, 5-methylcytosine (mC) RNA modification has emerged as a key player in regulating RNA metabolism and function through coding as well as non-coding RNAs. Accumulating evidence has shown that mC modulates the stability, translation, transcription, nuclear export, and cleavage of RNAs to mediate cell proliferation, differentiation, apoptosis, stress responses, and other biological functions. In humans, mC RNA modification is catalyzed by the NOL1/NOP2/sun (NSUN) family and DNA methyltransferase 2 (DNMT2). These RNA modifiers regulate the expression of multiple oncogenes such as fizzy-related-1, forkhead box protein C2, Grb associated-binding protein 2, and TEA domain transcription factor 1, facilitating the pathogenesis and progression of cancers. Furthermore, the aberrant expression of methyltransferases have been identified in various cancers and used to predict the prognosis of patients. In this review, we present a comprehensive overview of mC RNA methyltransferases. We specifically highlight the potential mechanism of action of mC in cancer. Finally, we discuss the prospect of mC-relative studies.© 2022. The Author(s).
[44]
Tao W, Zhao D, Sun M, et al. Intestinal absorption and activation of decitabine amino acid ester prodrugs mediated by peptide transporter PEPT1 and enterocyte enzymes[J]. Int J Pharm, 2018, 541(1-2):64-71.DOI:10.1016/j.ijpharm.2018.02.033.
[45]
Lio CJ, Rao A. TET enzymes and 5hmC in adaptive and innate immune systems[J]. Front Immunol, 2019,10:210.DOI:10.3389/fimmu.2019.00210.
[46]
Zhang Q, Liu F, Chen W, et al. The role of RNA m5C modification in cancer metastasis[J]. Int J Biol Sci, 2021, 17(13):3369-3380.DOI:10.7150/ijbs.61439.
[47]
Alkrekshi A, Wang W, Rana PS, et al. A comprehensive review of the functions of YB-1 in cancer stemness,metastasis and drug resistance[J]. Cellular Signalling, 2021,85:110073.DOI:10.1016/j.cellsig.2021.110073.
[48]
Dai X, Gonzalez G, Li L, et al. YTHDF2 binds to 5-methylcytosine in RNA and modulates the maturation of ribosomal RNA[J]. Anal Chem, 2020, 92(1):1346-1354.DOI:10.1021/acs.analchem.9b04505.
5-Methylcytosine is found in both DNA and RNA; although its functions in DNA are well established, the exact role of 5-methylcytidine (mC) in RNA remains poorly defined. Here we identified, by employing a quantitative proteomics method, multiple candidate recognition proteins of mC in RNA, including several YTH domain-containing family (YTHDF) proteins. We showed that YTHDF2 could bind directly to mC in RNA, albeit at a lower affinity than that toward -methyladenosine (mA) in RNA, and this binding involves Trp, a conserved residue located in the hydrophobic pocket of YTHDF2 that is also required for mA recognition. RNA bisulfite sequencing results revealed that, after CRISPR-Cas9-mediated knockout of the gene, the majority of mC sites in rRNA (rRNA) exhibited substantially augmented levels of methylation. Moreover, we found that YTHDF2 is involved in pre-rRNA processing in cells. Together, our data expanded the functions of the YTHDF2 protein in post-transcriptional regulations of RNA and provided novel insights into the functions of mC in RNA biology.
[49]
Yan H, Tan L, Liu Y, et al. Ten-eleven translocation methyl-cytosine dioxygenase 2 deficiency exacerbates renal ischemia-reperfusion injury[J]. Clin Epigenetics, 2020, 12(1):98.DOI:10.1186/s13148-020-00892-8.
Ten-eleven translocation (Tet) methyl-cytosine dioxygenases (including Tet1/2/3)-mediated 5mC oxidation and DNA demethylation play important roles in embryonic development and adult tissue homeostasis. The expression of Tet2 and Tet3 genes are relatively abundant in the adult murine kidneys while Tet1 gene is expressed at a low level. Although Tet3 has been shown to suppress kidney fibrosis, the role of Tet2 in kidney physiology as well as renal ischemia-reperfusion (IR) injury is still largely unknown.Tet2 mice displayed normal kidney morphology and renal function as WT mice while the expression of genes associated with tight junction and adherens junction was impaired. At 24 h post-renal IR, Tet2 mice showed higher SCr and BUN levels, more severe tubular damage, and elevated expression of Kim1 and Ngal genes in the kidney in comparison with WT mice. Moreover, the transcriptomic analysis revealed augmented inflammatory response in the kidneys of Tet2 mice.Tet2 is dispensable for kidney development and function at baseline condition while protects against renal IR injury possibly through repressing inflammatory response. Our findings suggest that Tet2 may be a potential target for the intervention of IR-induced acute kidney injury (AKI).
[50]
Huang N, Tan L, Xue Z, et al. Reduction of DNA hydroxymethylation in the mouse kidney insulted by ischemia reperfusion[J]. Biochem Biophys Res Commun, 2012, 422(4):697-702.DOI:10.1016/j.bbrc.2012.05.061.
[51]
Bao Y, Bai M, Zhu H, et al. DNA demethylase TET2 suppresses cisplatin-induced acute kidney injury[J]. Cell Death Discov, 2021, 7(1):167.DOI:10.1038/s41420-021-00528-7.
Demethylase Tet2 plays a vital role in the immune response. Acute kidney injury (AKI) initiation and maintenance phases are marked by inflammatory responses and leukocyte recruitment in endothelial and tubular cell injury processes. However, the role of Tet2 in AKI is poorly defined. Our study determined the degree of renal tissue damage associated with Tet2 gene expression levels in a cisplatin-induced AKI mice model. Tet2-knockout (KO) mice with cisplatin treatment experienced severe tubular necrosis and dilatation, inflammation, and AKI markers' expression levels than the wild-type mice. In addition, the administration of Tet2 plasmid protected Tet2-KO mice from cisplatin-induced nephrotoxicity, but not Tet2-catalytic-dead mutant. Tet2 KO was associated with a change in metabolic pathways like retinol, arachidonic acid, linolenic acid metabolism, and PPAR signaling pathway in the cisplatin-induced mice model. Tet2 expression is also downregulated in other AKI mice models and clinical samples. Thus, our results indicate that Tet2 has a renal protective effect during AKI by regulating metabolic and inflammatory responses through the PPAR signaling pathway.
[52]
Tampe B, Steinle U, Tampe D, et al. Low-dose hydralazine prevents fibrosis in a murine model of acute kidney injury-to-chronic kidney disease progression[J]. Kidney Int, 2017, 91(1):157-176.DOI:10.1016/j.kint.2016.07.042.
Acute kidney injury (AKI) and progressive chronic kidney disease (CKD) are intrinsically tied syndromes. In this regard, the acutely injured kidney often does not achieve its full regenerative capacity and AKI directly transitions into progressive CKD associated with tubulointerstitial fibrosis. Underlying mechanisms of such AKI-to-CKD progression are still incompletely understood and specific therapeutic interventions are still elusive. Because epigenetic modifications play a role in maintaining tissue fibrosis, we used a murine model of ischemia-reperfusion injury to determine whether aberrant promoter methylation of RASAL1 contributes causally to the switch between physiological regeneration and tubulointerstitial fibrogenesis, a hallmark of AKI-to-CKD progression. It is known that the antihypertensive drug hydralazine has demethylating activity, and that its optimum demethylating activity occurs at concentrations below blood pressure-lowering doses. Administration of low-dose hydralazine effectively induced expression of hydroxylase TET3, which catalyzed RASAL1 hydroxymethylation and subsequent RASAL1 promoter demethylation. Hydralazine-induced CpG promoter demethylation subsequently attenuated renal fibrosis and preserved excretory renal function independent of its blood pressure-lowering effects. In comparison, RASAL1 demethylation and inhibition of tubulointerstitial fibrosis was not detected upon administration of the angiotensin-converting enzyme inhibitor Ramipril in this model. Thus, RASAL1 promoter methylation and subsequent transcriptional RASAL1 suppression plays a causal role in AKI-to-CKD progression.Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[53]
Hermert D, Martin IV, Reiss LK, et al. The nucleic acid binding protein YB-1-controlled expression of CXCL-1 modulates kidney damage in liver fibrosis[J]. Kidney Int, 2020, 97(4):741-752.DOI:10.1016/j.kint.2019.10.024.
[54]
Hanssen L, Frye BC, Ostendorf T, et al. Y-box binding protein-1 mediates profibrotic effects of calcineurin inhibitors in the kidney[J]. J Immunol, 2011, 187(1):298-308.DOI:10.4049/jimmunol.1100382.
The immunosuppressive calcineurin inhibitors (CNIs) cyclosporine A (CsA) and tacrolimus are widely used in transplant organ recipients, but in the kidney allograft, they may cause tubulointerstitial as well as mesangial fibrosis, with TGF-β believed to be a central inductor. In this study, we report that the cold-shock protein Y-box binding protein-1 (YB-1) is a TGF-β independent downstream effector in CsA- as well as in tacrolimus- but not in rapamycin-mediated activation of rat mesangial cells (rMCs). Intracellular content of YB-1 is several-fold increased in MCs following CNI treatment in vitro and in vivo in mice. This effect ensues in a time-dependent manner, and the operative concentration range encompasses therapeutically relevant doses for CNIs. The effect of CNI on cellular YB-1 content is abrogated by specific blockade of translation, whereas retarding the transcription remains ineffective. The activation of rMCs by CNIs is accomplished by generation of reactive oxygen species. In contrast to TGF-β-triggered reactive oxygen species generation, hydrogen peroxide especially could be identified as a potent inductor of YB-1 accumulation. In line with this, hindering TGF-β did not influence CNI-induced YB-1 upregulation, whereas ERK/Akt pathways are involved in CNI-mediated YB-1 expression. CsA-induced YB-1 accumulation results in mRNA stabilization and subsequent generation of collagen. Our results provide strong evidence for a CNI-dependent induction of YB-1 in MCs that contributes to renal fibrosis via regulation of its own and collagen translation.
[55]
Fraser DJ, Phillips AO, Zhang X, et al. Y-box protein-1 controls transforming growth factor-beta1 translation in proximal tubular cells[J]. Kidney Int, 2008, 73(6):724-732.DOI:10.1038/sj.ki.5002719.
Transforming growth factor-beta1 (TGF-beta1) mRNA has low basal translational efficiency in proximal tubule cells; however, its translation is stimulated by profibrotic cytokines. We studied the role of the multifunctional Y-box protein-1 (YB-1) in regulating proximal tubule cell TGF-beta1 translation. Using RNA-electrophoretic mobility shift assays and ultraviolet crosslinking, we found two protein complexes of 50 and 100 kDa, which bound to the TGF-beta1 mRNA 5'-untranslated region. Supershift studies using antibodies to YB-1 showed that both sites contained YB-1 as did studies with recombinant YB-1, which demonstrated that it was sufficient to form both complexes. RNA competition experiments confirmed YB-1 binding to the two predicted binding sites; one with high affinity and the other with lower affinity. Strong basal YB-1 association with TGF-beta1 mRNA was found in proximal tubule cells, which decreased when platelet-derived growth factor was used to activate TGF-beta1 translation. In contrast, knockdown of proximal tubule cell YB-1 expression abrogated TGF-beta1 synthesis. Our results suggest that TGF-beta1 translation in proximal tubule cells requires YB-1 binding to a high-affinity site in the 5'-untranslated region of its mRNA; however, binding to a low-affinity site inhibits basal translation.
[56]
Mertens PR, Harendza S, Pollock AS, et al. Glomerular mesangial cell-specific transactivation of matrix metalloproteinase 2 transcription is mediated by YB-1[J]. J Biol Chem, 1997, 272(36):22905-22912.DOI:10.1074/jbc.272.36.22905.
Mesangial cell (MC) activation plays a pivotal role in the development of the end stage sclerotic lesion characteristic of most forms of chronic glomerular disease. We have previously demonstrated that MC activation is directly linked to high level expression of the matrix metalloproteinase-2 (MMP-2) enzyme (Turck, J., Pollock, A. S., Lee, L., Marti, H.-P., and Lovett, D. H. (1996) J. Biol. Chem. 25, 15074-15083), the transcription of which is regulated in a tissue-specific fashion. Recent studies (Harendza, S., Pollock, A., Mertens, P. R., and Lovett, D. H. (1995) J. Biol. Chem. 270, 18786-18796) delineated a strong cis-acting enhancer element, designated MMP-2 RE1, within the 5'-flanking region of the rat MMP-2 gene. Gel shift, DNA footprint, and transcriptional analyses mapped the enhancer element to a unique 40-base pair (bp) sequence located at -1322 to -1282 bp relative to the translational start site. Bromodeoxyuridine-substituted UV cross-linking of the 40-bp enhancer element with MC nuclear extracts yielded a single protein of 52 kDa, while Southwestern blot analysis with MMP-2 RE1 demonstrated three hybridizing nuclear proteins of 52, 62, and 86 kDa size. Screening of a human MC cDNA expression library with MMP-2 RE1 exclusively yielded clones with the identical sequence of the transcription factor YB-1. Western blot and supershift gel analysis of MC nuclear extracts with an anti-YB-1 antibody confirmed the presence of YB-1 within the shifted complex. Examination of the MMP-2 RE1 sequence revealed an incomplete Y-box sequence (CTGCTGGGCAAG), which specifically interacted with recombinant YB-1 on DMS protection footprinting analysis. YB-1 protein preferentially bound the single-stranded components of the 40-bp MMP-2 RE1 and, with increasing concentrations, formed multimeric complexes. Co-transfection of YB-1 in MC increased the enhancer activity within the context of the native MMP-2 promoter, while transfection of non-MMP-2-synthesizing glomerular epithelial cells with YB-1 led to transcriptional suppression. This study indicates that YB-1 is a major, cell type-specific transactivator of MMP-2 transcription by glomerular mesangial cells.
[57]
Bhreathnach U, Griffin B, Brennan E, et al. Profibrotic IHG-1 complexes with renal disease associated HSPA5 and TRAP1 in mitochondria[J]. Biochim Biophys Acta Mol Basis Dis, 2017, 1863(4):896-906.DOI:10.1016/j.bbadis.2017.01.015.
[58]
Wang J, Gibbert L, Djudjaj S, et al. Therapeutic nuclear shuttling of YB-1 reduces renal damage and fibrosis[J]. Kidney Int, 2016, 90(6):1226-1237.DOI:10.1016/j.kint.2016.07.008.
Virtually all chronic kidney diseases progress towards tubulointerstitial fibrosis. In vitro, Y-box protein-1 (YB-1) acts as a central regulator of gene transcription and translation of several fibrosis-related genes. However, it remains to be determined whether its pro- or antifibrotic propensities prevail in disease. Therefore, we investigated the outcome of mice with half-maximal YB-1 expression in a model of renal fibrosis induced by unilateral ureteral obstruction. Yb1 animals displayed markedly reduced tubular injury, immune cell infiltration and renal fibrosis following ureteral obstruction. The increase in renal YB-1 was limited to a YB-1 variant nonphosphorylated at serine 102 but phosphorylated at tyrosine 99. During ureteral obstruction, YB-1 localized to the cytoplasm, directly stabilizing Col1a1 mRNA, thus promoting fibrosis. Conversely, the therapeutic forced nuclear compartmentalization of phosphorylated YB-1 by the small molecule HSc025 mediated repression of the Col1a1 promoter and attenuated fibrosis following ureteral obstruction. Blunting of these effects in Yb1 mice confirmed involvement of YB-1. HSc025 even reduced tubulointerstitial damage when applied at later time points during maximum renal damage. Thus, phosphorylation and subcellular localization of YB-1 determines its effect on renal fibrosis in vivo. Hence, induced nuclear YB-1 shuttling may be a novel antifibrotic treatment strategy in renal diseases with the potential of damage reversal.Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[59]
Kato M, Wang L, Putta S, et al. Post-transcriptional up-regulation of Tsc-22 by YBX1,a target of miR-216a,mediates TGF-β-induced collagen expression in kidney cells[J]. J Biol Chem, 2010, 285(44):34004-34015.DOI:10.1074/jbc.M110.165027.
[60]
Bernhardt A, Fehr A, Brandt S, et al. Inflammatory cell infiltration and resolution of kidney inflammation is orchestrated by the cold-shock protein Y-box binding protein-1[J]. Kidney Int, 2017, 92(5):1157-1177.DOI:10.1016/j.kint.2017.03.035.
Tubular cells recruit monocytic cells in inflammatory tubulointerstitial kidney diseases. The cell-cell communication that establishes pro- or anti-inflammatory activities is mainly influenced by cytokines, reactive oxygen species, nitric oxide, and phagocytosis. Key proteins orchestrating these processes such as cold-shock proteins linked with chemoattraction and cell maturation have been identified. The prototypic member of the cold-shock protein family, Y-box binding protein (YB)-1, governs specific phenotypic alterations in monocytic cells and was explored in the present study. Following tubulointerstitial injury by unilateral ureteral obstruction, increased inflammatory cell infiltration and tubular cell CCL5 expression was found in conditional Ybx1 knockout animals with specific depletion in monocytes/macrophages (YB-1). Furthermore, YB-1 mice exhibit enhanced tissue damage, myofibroblast activation, and fibrosis. To investigate relevant molecular mechanism(s), we utilized bone marrow-derived macrophage cultures and found that YB-1-deficient macrophages display defects in cell polarization and function, including reduced proliferation and nitric oxide production, loss of phagocytic activity, and failure to upregulate IL-10 and CCL5 expression in response to inflammatory stimuli. Co-culture with primary tubular cells confirmed these findings. Thus, monocytic YB-1 has prominent and distinct roles for cellular feed-forward crosstalk and resolution of inflammatory processes by its ability to regulate cell differentiation and cytokine/chemokine synthesis.Copyright © 2017 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[61]
Tan Y, Cao H, Li Q, et al. The role of transcription factor Ap1 in the activation of the Nrf2/ARE pathway through TET1 in diabetic nephropathy[J]. Cell Biol Int, 2021, 45(8):1654-1665.DOI:10.1002/cbin.11599.
[62]
Veloso Pereira BM, Charleaux de Ponte M, Malavolta Luz AP, et al. DNA methylation enzymes in the kidneys of male and female BTBR ob/ob mice[J]. Front Endocrinol(Lausanne), 2023,14:1167546.DOI:10.3389/fendo.20231167546.
[63]
Liu D, Sun H, Li K, et al. HIF-1α mediates renal fibrosis by regulating metabolic remodeling of renal tubule epithelial cells[J]. Biochem Biophys Res Commun, 2022, 618:15-23.DOI:10.1016/j.bbrc.2022.06.008.
[64]
Yang L, Zhang Q, Wu Q, et al. Effect of TET2 on the pathogenesis of diabetic nephropathy through activation of transforming growth factor β1 expression via DNA demethylation[J]. Life Sci, 2018, 207:127-137.DOI:10.1016/j.lfs.2018.04.044.
[65]
Tampe B, Tampe D, Müller CA, et al. TET3-mediated hydroxymethylation of epigenetically silenced genes contributes to bone morphogenic protein 7-induced reversal of kidney fibrosis[J]. J Am Soc Nephrol, 2014, 25(5):905-912.DOI:10.1681/ASN.2013070723.
Methylation of CpG island promoters is an epigenetic event that can effectively silence transcription over multiple cell generations. Hypermethylation of the Rasal1 promoter contributes to activation of fibroblasts and progression of kidney fibrosis. Here, we explored whether such causative hypermethylation could be reversed through endogenous mechanisms and whether such reversal of hypermethylation is a constituent of the antifibrotic activity of bone morphogenic protein 7 (BMP7). We show that successful inhibition of experimental kidney fibrosis through administration of BMP7 associates with normalization of Rasal1 promoter hypermethylation. Furthermore, this reversal of pathologic hypermethylation was achieved specifically through Tet3-mediated hydroxymethylation. Collectively, our findings reveal a new mechanism that may be exploited to facilitate therapeutic DNA demethylation to reverse kidney fibrosis.Copyright © 2014 by the American Society of Nephrology.
[66]
Xu X, Tan X, Tampe B, et al. High-fidelity CRISPR/Cas9-based gene-specific hydroxymethylation rescues gene expression and attenuates renal fibrosis[J]. Nat Commun, 2018, 9(1):3509.DOI:10.1038/s41467-018-05766-5.
[67]
Rana R, Manoharan J, Elwakiel A, et al. Glomerular-tubular crosstalk via cold shock Y-box binding protein-1 in the kidney[J]. Kidney Int, 2024, 105(1):65-83.DOI:10.1016/j.kint.2023.09.014.
PDF(677 KB)

Accesses

Citation

Detail

Sections
Recommended

/