Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1

Abstract Background Emerging evidence confirms that lncRNAs (long non-coding RNAs) are potential biomarkers that play vital roles in tumors. ZNF582-AS1 is a novel lncRNA that serves as a potential prognostic marker of cancers. However, the specific clinical significance and molecular mechanism of ZN...

Full description

Bibliographic Details
Main Authors: Wuping Yang, Kenan Zhang, Lei Li, Yawei Xu, Kaifang Ma, Haibiao Xie, Jingcheng Zhou, Lin Cai, Yanqing Gong, Kan Gong
Format: Article
Language:English
Published: BMC 2021-03-01
Series:Journal of Experimental & Clinical Cancer Research
Subjects:
Online Access:https://doi.org/10.1186/s13046-021-01889-8
id doaj-32c52a90dffd427e84ee9102be5c7fc5
record_format Article
spelling doaj-32c52a90dffd427e84ee9102be5c7fc52021-03-11T11:17:42ZengBMCJournal of Experimental & Clinical Cancer Research1756-99662021-03-0140112010.1186/s13046-021-01889-8Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1Wuping Yang0Kenan Zhang1Lei Li2Yawei Xu3Kaifang Ma4Haibiao Xie5Jingcheng Zhou6Lin Cai7Yanqing Gong8Kan Gong9Department of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalDepartment of Urology, Peking University First HospitalAbstract Background Emerging evidence confirms that lncRNAs (long non-coding RNAs) are potential biomarkers that play vital roles in tumors. ZNF582-AS1 is a novel lncRNA that serves as a potential prognostic marker of cancers. However, the specific clinical significance and molecular mechanism of ZNF582-AS1 in ccRCC (clear cell renal cell carcinoma) are unclear. Methods Expression level and clinical significance of ZNF582-AS1 were determined by TCGA-KIRC data and qRT-PCR results of 62 ccRCCs. DNA methylation status of ZNF582-AS1 promoter was examined by MSP, MassARRAY methylation and demethylation analysis. Gain-of-function experiments were conducted to investigate the biological roles of ZNF582-AS1 in the phenotype of ccRCC. The subcellular localization of ZNF582-AS1 was detected by RNA FISH. iTRAQ, RNA pull-down and RIP-qRT-PCR were used to identify the downstream targets of ZNF582-AS1. rRNA MeRIP-seq and MeRIP-qRT-PCR were utilized to examine the N(6)-methyladenosine modification status. Western blot and immunohistochemistry assays were used to determine the protein expression level. Results ZNF582-AS1 was downregulated in ccRCC, and decreased ZNF582-AS1 expression was significantly correlated with advanced tumor stage, higher pathological stage, distant metastasis and poor prognosis. Decreased ZNF582-AS1 expression was caused by DNA methylation at the CpG islands within its promoter. ZNF582-AS1 overexpression inhibited cell proliferative, migratory and invasive ability, and increased cell apoptotic rate in vitro and in vivo. Mechanistically, we found that ZNF582-AS1 overexpression suppressed the N(6)-methyladenosine modification of MT-RNR1 by reducing rRNA adenine N(6)-methyltransferase A8K0B9 protein level, resulting in the decrease of MT-RNR1 expression, followed by the inhibition of MT-CO2 protein expression. Furthermore, MT-RNR1 overexpression reversed the decreased MT-CO2 expression and phenotype inhibition of ccRCC induced by increased ZNF582-AS1 expression. Conclusions This study demonstrates for the first time that ZNF582-AS1 functions as a tumor suppressor gene in ccRCC and ZNF582-AS1 may serve as a potential biomarker and therapeutic target of ccRCC.https://doi.org/10.1186/s13046-021-01889-8ccRCCZNF582-AS1DNA methylationN(6)-methyladenosine modificationMT-RNR1
collection DOAJ
language English
format Article
sources DOAJ
author Wuping Yang
Kenan Zhang
Lei Li
Yawei Xu
Kaifang Ma
Haibiao Xie
Jingcheng Zhou
Lin Cai
Yanqing Gong
Kan Gong
spellingShingle Wuping Yang
Kenan Zhang
Lei Li
Yawei Xu
Kaifang Ma
Haibiao Xie
Jingcheng Zhou
Lin Cai
Yanqing Gong
Kan Gong
Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
Journal of Experimental & Clinical Cancer Research
ccRCC
ZNF582-AS1
DNA methylation
N(6)-methyladenosine modification
MT-RNR1
author_facet Wuping Yang
Kenan Zhang
Lei Li
Yawei Xu
Kaifang Ma
Haibiao Xie
Jingcheng Zhou
Lin Cai
Yanqing Gong
Kan Gong
author_sort Wuping Yang
title Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
title_short Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
title_full Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
title_fullStr Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
title_full_unstemmed Downregulation of lncRNA ZNF582-AS1 due to DNA hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the N(6)-methyladenosine modification of MT-RNR1
title_sort downregulation of lncrna znf582-as1 due to dna hypermethylation promotes clear cell renal cell carcinoma growth and metastasis by regulating the n(6)-methyladenosine modification of mt-rnr1
publisher BMC
series Journal of Experimental & Clinical Cancer Research
issn 1756-9966
publishDate 2021-03-01
description Abstract Background Emerging evidence confirms that lncRNAs (long non-coding RNAs) are potential biomarkers that play vital roles in tumors. ZNF582-AS1 is a novel lncRNA that serves as a potential prognostic marker of cancers. However, the specific clinical significance and molecular mechanism of ZNF582-AS1 in ccRCC (clear cell renal cell carcinoma) are unclear. Methods Expression level and clinical significance of ZNF582-AS1 were determined by TCGA-KIRC data and qRT-PCR results of 62 ccRCCs. DNA methylation status of ZNF582-AS1 promoter was examined by MSP, MassARRAY methylation and demethylation analysis. Gain-of-function experiments were conducted to investigate the biological roles of ZNF582-AS1 in the phenotype of ccRCC. The subcellular localization of ZNF582-AS1 was detected by RNA FISH. iTRAQ, RNA pull-down and RIP-qRT-PCR were used to identify the downstream targets of ZNF582-AS1. rRNA MeRIP-seq and MeRIP-qRT-PCR were utilized to examine the N(6)-methyladenosine modification status. Western blot and immunohistochemistry assays were used to determine the protein expression level. Results ZNF582-AS1 was downregulated in ccRCC, and decreased ZNF582-AS1 expression was significantly correlated with advanced tumor stage, higher pathological stage, distant metastasis and poor prognosis. Decreased ZNF582-AS1 expression was caused by DNA methylation at the CpG islands within its promoter. ZNF582-AS1 overexpression inhibited cell proliferative, migratory and invasive ability, and increased cell apoptotic rate in vitro and in vivo. Mechanistically, we found that ZNF582-AS1 overexpression suppressed the N(6)-methyladenosine modification of MT-RNR1 by reducing rRNA adenine N(6)-methyltransferase A8K0B9 protein level, resulting in the decrease of MT-RNR1 expression, followed by the inhibition of MT-CO2 protein expression. Furthermore, MT-RNR1 overexpression reversed the decreased MT-CO2 expression and phenotype inhibition of ccRCC induced by increased ZNF582-AS1 expression. Conclusions This study demonstrates for the first time that ZNF582-AS1 functions as a tumor suppressor gene in ccRCC and ZNF582-AS1 may serve as a potential biomarker and therapeutic target of ccRCC.
topic ccRCC
ZNF582-AS1
DNA methylation
N(6)-methyladenosine modification
MT-RNR1
url https://doi.org/10.1186/s13046-021-01889-8
work_keys_str_mv AT wupingyang downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT kenanzhang downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT leili downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT yaweixu downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT kaifangma downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT haibiaoxie downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT jingchengzhou downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT lincai downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT yanqinggong downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
AT kangong downregulationoflncrnaznf582as1duetodnahypermethylationpromotesclearcellrenalcellcarcinomagrowthandmetastasisbyregulatingthen6methyladenosinemodificationofmtrnr1
_version_ 1724225687562223616