Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bin Jiang is active.

Publication


Featured researches published by Bin Jiang.


Molecular and Cellular Biology | 2014

Methylation by NSun2 Represses the Levels and Function of MicroRNA 125b

Shuai Yuan; Hao Tang; Junyue Xing; Xiuqin Fan; Xiaoyu Cai; Qiu Li; Pei Han; Yuhong Luo; Zhuojun Zhang; Bin Jiang; Yali Dou; Myriam Gorospe; Wengong Wang

ABSTRACT Methylation is a prevalent posttranscriptional modification of RNAs. However, whether mammalian microRNAs are methylated is unknown. Here, we show that the tRNA methyltransferase NSun2 methylates primary (pri-miR-125b), precursor (pre-miR-125b), and mature microRNA 125b (miR-125b) in vitro and in vivo. Methylation by NSun2 inhibits the processing of pri-miR-125b2 into pre-miR-125b2, decreases the cleavage of pre-miR-125b2 into miR-125, and attenuates the recruitment of RISC by miR-125, thereby repressing the function of miR-125b in silencing gene expression. Our results highlight the impact of miR-125b function via methylation by NSun2.


BMC Molecular Biology | 2013

Loss of CARM1 is linked to reduced HuR function in replicative senescence

Lijun Pang; Haiyan Tian; Na Chang; Jie Yi; Lixiang Xue; Bin Jiang; Myriam Gorospe; Xiaowei Zhang; Wengong Wang

BackgroundThe co-activator-associated arginine methyltransferase 1 (CARM1) catalyzes the methylation of HuR. However, the functional impact of this modification is not fully understood. Here, we investigated the influence of HuR methylation by CARM1 upon the turnover of HuR target mRNAs encoding senescence-regulatory proteins.ResultsChanging the methylation status of HuR in HeLa cells by either silencing CARM1 or mutating the major methylation site (R217K) greatly diminished the effect of HuR in regulating the turnover of mRNAs encoding cyclin A, cyclin B1, c-fos, SIRT1, and p16. Although knockdown of CARM1 or HuR individually influenced the expression of cyclin A, cyclin B1, c-fos, SIRT1, and p16, joint knockdown of both CARM1 and HuR did not show further effect. Methylation by CARM1 enhanced the association of HuR with the 3′UTR of p16 mRNA, but not with the 3′UTR of cyclin A, cyclin B1, c-fos, or SIRT1 mRNAs. In senescent human diploid fibroblasts (HDFs), reduced CARM1 was accompanied by reduced HuR methylation. In addition, knockdown of CARM1 or mutation of the major methylation site of HuR in HDF markedly impaired the ability of HuR to regulate the expression of cyclin A, cyclin B1, c-fos, SIRT1, and p16 as well to maintain a proliferative phenotype.ConclusionCARM1 represses replicative senescence by methylating HuR and thereby enhancing HuR’s ability to regulate the turnover of cyclin A, cyclin B1, c-fos, SIRT1, and p16 mRNAs.


Journal of Cellular Biochemistry | 2010

Hydrogen peroxide induces p16INK4a through an AUF1‐dependent manner

Gai E Guo; Li Wei Ma; Bin Jiang; Jie Yi; Tanjun Tong; Wen Gong Wang

Elevation of p16INK4a has been described as an important mechanism for hydrogen peroxide (H2O2)‐induced replicative senescence. However, the mechanisms underlying remain unknown. In this study, we demonstrate an important role of RNA‐binding protein AUF1‐mediated mRNA turnover in H2O2‐induced p16INK4a expression. The induction of p16 by H2O2 was accompanied with declined cytoplasmic AUF1 level. Accordingly, exposure of cells to H2O2 remarkably reduced the binding of AUF1 to p16 3′UTR and increased the half‐life of an EGFP‐p16‐3′UTR chimeric transcript. In AUF1‐silenced cells, the effect of H2O2 on p16 induction was abolished. Furthermore, in cells co‐transfected with vectors expressing AUF1s, treatment with H2O2 failed to significantly reduce the expression of AUF1 and subsequently elevate the levels of p16. Moreover, HeLa cells overexpressing AUF1s were resistant to H2O2‐induced senescence. Our results indicate that AUF1 is critical for H2O2‐induced p16 expression and cellular senescence. J. Cell. Biochem. 109: 1000–1005, 2010.


Molecular and Cellular Biology | 2015

NSun2 Promotes Cell Growth via Elevating Cyclin-Dependent Kinase 1 Translation

Junyue Xing; Jie Yi; Xiaoyu Cai; Hao Tang; Zhenyun Liu; Xiaotian Zhang; Jennifer L. Martindale; Xiaoling Yang; Bin Jiang; Myriam Gorospe; Wengong Wang

ABSTRACT The tRNA methytransferase NSun2 promotes cell proliferation, but the molecular mechanism has not been elucidated. Here, we report that NSun2 regulates cyclin-dependent kinase 1 (CDK1) expression in a cell cycle-dependent manner. Knockdown of NSun2 decreased the CDK1 protein level, while overexpression of NSun2 elevated it without altering CDK1 mRNA levels. Further studies revealed that NSun2 methylated CDK1 mRNA in vitro and in cells and that methylation by NSun2 enhanced CDK1 translation. Importantly, NSun2-mediated regulation of CDK1 expression had an impact on the cell division cycle. These results provide new insight into the regulation of CDK1 during the cell division cycle.


Oncotarget | 2016

RNA methyltransferase NSUN2 promotes stress-induced HUVEC senescence.

Xiaoyu Cai; Yuanyuan Hu; Hao Tang; Han Hu; Lijun Pang; Junyue Xing; Zhenyun Liu; Yuhong Luo; Bin Jiang; Te Liu; Myriam Gorospe; Chuan Chen; Wengong Wang

The tRNA methyltransferase NSUN2 delays replicative senescence by regulating the translation of CDK1 and CDKN1B mRNAs. However, whether NSUN2 influences premature cellular senescence remains untested. Here we show that NSUN2 methylates SHC mRNA in vitro and in cells, thereby enhancing the translation of the three SHC proteins, p66SHC, p52SHC, and p46SHC. Our results further show that the elevation of SHC expression by NSUN2-mediated mRNA methylation increased the levels of ROS, activated p38MAPK, thereby accelerating oxidative stress- and high-glucose-induced senescence of human vascular endothelial cells (HUVEC). Our findings highlight the critical impact of NSUN2-mediated mRNA methylation in promoting premature senescence.


Aging Cell | 2014

let-7-repressesed Shc translation delays replicative senescence

Fang Xu; Lijun Pang; Xiaoyu Cai; Xinwen Liu; Shuai Yuan; Xiuqin Fan; Bin Jiang; Xiaowei Zhang; Yali Dou; Myriam Gorospe; Wengong Wang

The p66Shc adaptor protein is an important regulator of lifespan in mammals, but the mechanisms responsible are still unclear. Here, we show that expression of p66Shc, p52Shc, and p46Shc is regulated at the post‐transcriptional level by the microRNA let‐7a. The levels of let‐7a correlated inversely with the levels of Shc proteins without affecting Shc mRNA levels. We identified ‘seedless’ let‐7a interaction elements in the coding region of Shc mRNA; mutation of the ‘seedless’ interaction sites abolished the regulation of Shc by let‐7a. Our results further revealed that repression of Shc expression by let‐7a delays senescence of human diploid fibroblasts (HDFs). In sum, our findings link let‐7a abundance to the expression of p66Shc, which in turn controls the replicative lifespan of HDFs.


Journal of Cellular Biochemistry | 2017

NSUN2-Mediated m5C Methylation and METTL3/METTL14-Mediated m6A Methylation Cooperatively Enhance p21 Translation

Qiu Li; Xiu Li; Hao Tang; Bin Jiang; Yali Dou; Myriam Gorospe; Wengong Wang

N6‐methyladenosine (m6A) and m5C methylation are two major types of RNA methylation, but the impact of joint modifications on the same mRNA is unknown. Here, we show that in p21 3′UTR, NSUN2 catalyzes m5C modification and METTL3/METTL14 catalyzes m6A modification. Interestingly, methylation at m6A by METTL3/METTL14 facilitates the methylation of m5C by NSUN2, and vice versa. NSUN2‐mediated m5C and METTL3/METTL14‐mediated m6A methylation synergistically enhance p21 expression at the translational level, leading to elevated expression of p21 in oxidative stress‐induced cellular senescence. Our findings on p21 mRNA methylation and expression reveal that joint m6A and m5C modification of the same RNA may influence each other, coordinately affecting protein expression patterns. J. Cell. Biochem. 118: 2587–2598, 2017.


Nature Communications | 2018

HuR regulates telomerase activity through TERC methylation

Hao Tang; Hu Wang; Xiaolei Cheng; Xiuqin Fan; Fan Yang; Mengmeng Zhang; Yanlian Chen; Yuyang Tian; Cihang Liu; Dongxing Shao; Bin Jiang; Yali Dou; Yusheng Cong; Junyue Xing; Xiaotian Zhang; Xia Yi; Zhou Songyang; Wenbin Ma; Yong Zhao; Xian Wang; Jinbiao Ma; Myriam Gorospe; Zhenyu Ju; Wengong Wang

Telomerase consists of the catalytic protein TERT and the RNA TERC. Mutations in TERC are linked to human diseases, but the underlying mechanisms are poorly understood. Here we report that the RNA-binding protein HuR associates with TERC and promotes the assembly of the TERC/TERT complex by facilitating TERC C106 methylation. Dyskeratosis congenita (DC)-related TERC U100A mutation impair the association of HuR with TERC, thereby reducing C106 methylation. Two other TERC mutations linked to aplastic anemia and autosomal dominant DC, G107U, and GC107/108AG, likewise disrupt methylation at C106. Loss-of-HuR binding and hence lower TERC methylation leads to decreased telomerase activity and telomere shortening. Furthermore, HuR deficiency or mutation of mTERC HuR binding or methylation sites impair the renewal of mouse hematopoietic stem cells, recapitulating the bone marrow failure seen in DC. Collectively, our findings reveal a novel function of HuR, linking HuR to telomerase function and TERC-associated DC.Mutations in the RNA component TERC can cause telomerase dysfunction but the underlying mechanisms are largely unknown. Here, the authors show that RNA-binding protein HuR regulates telomerase function by enhancing the methylation of TERC, which is impaired by several disease-relevant TERC mutations.


Nature Communications | 2018

Author Correction: HuR regulates telomerase activity through TERC methylation

Hao Tang; Hu Wang; Xiaolei Cheng; Xiuqin Fan; Fan Yang; Mengmeng Zhang; Yanlian Chen; Yuyang Tian; Cihang Liu; Dongxing Shao; Bin Jiang; Yali Dou; Yusheng Cong; Junyue Xing; Xiaotian Zhang; Xia Yi; Zhou Songyang; Wenbin Ma; Yong Zhao; Xian Wang; Jinbiao Ma; Myriam Gorospe; Zhenyu Ju; Wengong Wang

In the original version of this Article, the affiliation details for Fan Yang were incorrectly given as ‘Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China’ and ‘Leibniz Institute for Age Research - Fritz Lipmann Institute, Friedrich-Schiller University of Jena, Jena, 07745, Germany’. This has now been corrected in both the PDF and HTML versions of the Article.


Aging (Albany NY) | 2015

NSun2 delays replicative senescence by repressing p27 (KIP1) translation and elevating CDK1 translation

Hao Tang; Xiuqin Fan; Junyue Xing; Zhenyun Liu; Bin Jiang; Yali Dou; Myriam Gorospe; Wengong Wang

Collaboration


Dive into the Bin Jiang's collaboration.

Top Co-Authors

Avatar

Myriam Gorospe

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wengong Wang

University of Connecticut

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yali Dou

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge