Network


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

Hotspot


Dive into the research topics where Huating Wang is active.

Publication


Featured researches published by Huating Wang.


BMC Genomics | 2013

iSeeRNA: identification of long intergenic non-coding RNA transcripts from transcriptome sequencing data

Kun Sun; Xiaona Chen; Peiyong Jiang; Xiaofeng Song; Huating Wang; Hao Sun

BackgroundLong intergenic non-coding RNAs (lincRNAs) are emerging as a novel class of non-coding RNAs and potent gene regulators. High-throughput RNA-sequencing combined with de novo assembly promises quantity discovery of novel transcripts. However, the identification of lincRNAs from thousands of assembled transcripts is still challenging due to the difficulties of separating them from protein coding transcripts (PCTs).ResultsWe have implemented iSeeRNA, a support vector machine (SVM)-based classifier for the identification of lincRNAs. iSeeRNA shows better performance compared to other software. A public available webserver for iSeeRNA is also provided for small size dataset.ConclusionsiSeeRNA demonstrates high prediction accuracy and runs several magnitudes faster than other similar programs. It can be integrated into the transcriptome data analysis pipelines or run as a web server, thus offering a valuable tool for lincRNA study.


PLOS ONE | 2012

Inhibition of miR-29 by TGF-beta-Smad3 Signaling through Dual Mechanisms Promotes Transdifferentiation of Mouse Myoblasts into Myofibroblasts

Liang Zhou; Lijun Wang; Leina Lu; Peiyong Jiang; Hao Sun; Huating Wang

MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression in post-transcriptional fashion, and emerging studies support their importance in regulating many biological processes, including myogenic differentiation and muscle development. miR-29 is a promoting factor during myogenesis but its full spectrum of impact on muscle cells has yet to be explored. Here we describe an analysis of miR-29 affected transcriptome in C2C12 muscle cells using a high throughput RNA-sequencing platform. The results reveal that miR-29 not only functions to promote myogenic differentiation but also suppresses the transdifferentiation of myoblasts into myofibroblasts. miR-29 inhibits the fibrogenic differentiation through down-regulating both extracellular matrix genes and cell adhesion genes. We further demonstrate that miR-29 is under negative regulation by TGF-beta (TGF-β)–Smad3 signaling via dual mechanisms of both inhibiting MyoD binding and enhancing Yin Yang 1 (YY1)-recruited Polycomb association. Together, these results identify miR-29 as a pleiotropic molecule in both myogenic and fibrogenic differentiation of muscle cells.


Cell Research | 2015

LncRNA Dum interacts with Dnmts to regulate Dppa2 expression during myogenic differentiation and muscle regeneration

Lijun Wang; Yu Zhao; Xichen Bao; Xihua Zhu; Yvonne Ka Yin Kwok; Kun Sun; Xiaona Chen; Yongheng Huang; Ralf Jauch; Miguel A. Esteban; Hao Sun; Huating Wang

Emerging studies document the roles of long non-coding RNAs (LncRNAs) in regulating gene expression at chromatin level but relatively less is known how they regulate DNA methylation. Here we identify an lncRNA, Dum (developmental pluripotency-associated 2 (Dppa2) Upstream binding Muscle lncRNA) in skeletal myoblast cells. The expression of Dum is dynamically regulated during myogenesis in vitro and in vivo. It is also transcriptionally induced by MyoD binding upon myoblast differentiation. Functional analyses show that it promotes myoblast differentiation and damage-induced muscle regeneration. Mechanistically, Dum was found to silence its neighboring gene, Dppa2, in cis through recruiting Dnmt1, Dnmt3a and Dnmt3b. Furthermore, intrachromosomal looping between Dum locus and Dppa2 promoter is necessary for Dum/Dppa2 interaction. Collectively, we have identified a novel lncRNA that interacts with Dnmts to regulate myogenesis.


PLOS ONE | 2010

TNF Inhibits Notch-1 in Skeletal Muscle Cells by Ezh2 and DNA Methylation Mediated Repression: Implications in Duchenne Muscular Dystrophy

Swarnali Acharyya; Sudarshana M. Sharma; Alfred S.L. Cheng; Katherine J. Ladner; Wei He; William Kline; Huating Wang; Michael C. Ostrowski; Tim H M Huang; Denis C. Guttridge

Background Classical NF-κB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFα on skeletal muscle differentiation are mediated in part through sustained NF-κB activity. In dystrophic muscles, NF-κB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFα that is also under IKKβ and NF-κB control. Methodology/Principal Findings Based on these findings we speculated that in DMD, TNFα secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFα is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-κB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFα stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. Conclusions/Significance We propose that in dystrophic muscles, elevated levels of TNFα and NF-κB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.


The EMBO Journal | 2013

Genome-wide survey by ChIP-seq reveals YY1 regulation of lincRNAs in skeletal myogenesis.

Leina Lu; Kun Sun; Xiaona Chen; Yu Zhao; Lijun Wang; Liang Zhou; Hao Sun; Huating Wang

Skeletal muscle differentiation is orchestrated by a network of transcription factors, epigenetic regulators, and non‐coding RNAs. The transcription factor Yin Yang 1 (YY1) silences multiple target genes in myoblasts (MBs) by recruiting Ezh2 (Enhancer of Zeste Homologue2). To elucidate genome‐wide YY1 binding in MBs, we performed chromatin immunoprecipitation (ChIP)‐seq and found 1820 specific binding sites in MBs with a large portion residing in intergenic regions. Detailed analysis demonstrated that YY1 acts as an activator for many loci in addition to its known repressor function. No significant co‐occupancy was found between YY1 and Ezh2, suggesting an additional Ezh2‐independent function for YY1 in MBs. Further analysis of intergenic binding sites showed that YY1 potentially regulates dozens of large intergenic non‐coding RNAs (lincRNAs), whose function in myogenesis is underexplored. We characterized a novel muscle‐associated lincRNA (Yam‐1) that is positively regulated by YY1. Yam‐1 is downregulated upon differentiation and acts as an inhibitor of myogenesis. We demonstrated that Yam‐1 functions through in cis regulation of miR‐715, which in turn targets Wnt7b. Our findings not only provide the first genome‐wide picture of YY1 association in muscle cells, but also uncover the functional role of lincRNA Yam‐1.


Cell Research | 2015

The p53-induced lincRNA-p21 derails somatic cell reprogramming by sustaining H3K9me3 and CpG methylation at pluripotency gene promoters.

Xichen Bao; Haitao Wu; Xihua Zhu; Xiangpeng Guo; Andrew Paul Hutchins; Zhiwei Luo; Hong Song; Yongqiang Chen; Keyu Lai; Menghui Yin; Lingxiao Xu; Liang Zhou; Jiekai Chen; Dongye Wang; Baoming Qin; Jon Frampton; Hung-Fat Tse; Duanqing Pei; Huating Wang; Biliang Zhang; Miguel A. Esteban

Recent studies have boosted our understanding of long noncoding RNAs (lncRNAs) in numerous biological processes, but few have examined their roles in somatic cell reprogramming. Through expression profiling and functional screening, we have identified that the large intergenic noncoding RNA p21 (lincRNA-p21) impairs reprogramming. Notably, lincRNA-p21 is induced by p53 but does not promote apoptosis or cell senescence in reprogramming. Instead, lincRNA-p21 associates with the H3K9 methyltransferase SETDB1 and the maintenance DNA methyltransferase DNMT1, which is facilitated by the RNA-binding protein HNRNPK. Consequently, lincRNA-p21 prevents reprogramming by sustaining H3K9me3 and/or CpG methylation at pluripotency gene promoters. Our results provide insight into the role of lncRNAs in reprogramming and establish a novel link between p53 and heterochromatin regulation.


Molecular Therapy | 2012

Loss of miR-29 in Myoblasts Contributes to Dystrophic Muscle Pathogenesis

Lijun Wang; Liang Zhou; Peiyong Jiang; Leina Lu; Xiaona Chen; Huiyao Lan; Denis C. Guttridge; Hao Sun; Huating Wang

microRNAs (miRNAs) are noncoding RNAs that regulate gene expression in post-transcriptional fashion, and emerging studies support their importance in a multitude of physiological and pathological processes. Here, we describe the regulation and function of miR-29 in Duchenne muscular dystrophy (DMD) and its potential use as therapeutic target. Our results demonstrate that miR-29 expression is downregulated in dystrophic muscles of mdx mice, a model of DMD. Restoration of its expression by intramuscular and intravenous injection improved dystrophy pathology by both promoting regeneration and inhibiting fibrogenesis. Mechanistic studies revealed that loss of miR-29 in muscle precursor cells (myoblasts) promotes their transdifferentiation into myofibroblasts through targeting extracellular molecules including collagens and microfibrillar-associated protein 5 (Mfap5). We further demonstrated that miR-29 is under negative regulation by transforming growth factor-β (TGF-β) signaling. Together, these results not only identify TGF-β-miR-29 as a novel regulatory axis during myoblasts conversion into myofibroblasts which constitutes a novel contributing route to muscle fibrogenesis of DMD but also implicate miR-29 replacement therapy as a promising treatment approach for DMD.


PLOS ONE | 2012

A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network

Leina Lu; Liang Zhou; Eric Z. Chen; Kun Sun; Peiyong Jiang; Lijun Wang; Xiaoxi Su; Hao Sun; Huating Wang

microRNAs (miRNAs) are non-coding RNAs that regulate gene expression post-transcriptionally, and mounting evidence supports the prevalence and functional significance of their interplay with transcription factors (TFs). Here we describe the identification of a regulatory circuit between muscle miRNAs (miR-1, miR-133 and miR-206) and Yin Yang 1 (YY1), an epigenetic repressor of skeletal myogenesis in mouse. Genome-wide identification of potential down-stream targets of YY1 by combining computational prediction with expression profiling data reveals a large number of putative miRNA targets of YY1 during skeletal myoblasts differentiation into myotubes with muscle miRs ranking on top of the list. The subsequent experimental results demonstrate that YY1 indeed represses muscle miRs expression in myoblasts and the repression is mediated through multiple enhancers and recruitment of Polycomb complex to several YY1 binding sites. YY1 regulating miR-1 is functionally important for both C2C12 myogenic differentiation and injury-induced muscle regeneration. Furthermore, we demonstrate that miR-1 in turn targets YY1, thus forming a negative feedback loop. Together, these results identify a novel regulatory circuit required for skeletal myogenesis and reinforce the idea that regulatory circuitries involving miRNAs and TFs are prevalent mechanisms.


Journal of Biological Chemistry | 2012

A Novel Target of MicroRNA-29, Ring1 and YY1-binding Protein (Rybp), Negatively Regulates Skeletal Myogenesis

Liang Zhou; Lijun Wang; Leina Lu; Peiyong Jiang; Hao Sun; Huating Wang

Background: MicroRNA-29-YY1 regulatory circuitry functions during skeletal myogenesis. Results: A genome-wide search revealed Rybp as a novel target of miR-29, and it silences myogenic loci together with YY1. Conclusion: Rybp functions as a repressor of myogenesis. Significance: This study identifies a novel regulatory circuitry underlying muscle formation and highlights the intimate interplay among transcription factors, epigenetic regulators, and microRNAs. Skeletal muscle cell differentiation (myogenesis) is a process orchestrated by a complex network involving transcription factors, epigenetic regulators, and microRNAs. Previous studies identified miR-29 as a pro-myogenic factor that interacts with components of Polycomb repressive complex, YY1 and Ezh2. In a genome-wide survey of miR-29-mediated transcriptome changes in C2C12 myoblasts, many epigenetic factors were found to be down-regulated by miR-29. Among them, Rybp was shown to be a direct target of miR-29 through binding to its 3′ UTR. Functional studies demonstrated that Rybp is down-regulated during myogenesis and acts as a negative regulator of skeletal myogenesis both in vitro during C2C12 differentiation and in vivo in injury-induced muscle regeneration. Furthermore, we found that Rybp and YY1 co-occupy several myogenic loci, including miR-29 itself, to silence their expression, thus forming a Rybp-miR-29 feedback loop. Rybp overexpression was found to enhance the enrichment of Ezh2 and trimethylation of H3K27 at target loci, suggesting it may facilitate the recruitment or stabilization of the Polycomb repressive complex. Collectively, our results identify Rybp as a novel regulator of myogenesis that co-acts with YY1 to silence miR-29 and other myogenic loci.


Cancer Research | 2014

A Novel Wnt Regulatory Axis in Endometrioid Endometrial Cancer

Yu Zhao; Yihua Yang; Jone Trovik; Kun Sun; Liang Zhou; Peiyong Jiang; Tat-San Lau; Erling A. Hoivik; Helga B. Salvesen; Hao Sun; Huating Wang

The Protocadherin 10 (PCDH10) is inactivated often by promoter hypermethylation in various human tumors, but its possible functional role as a tumor suppressor gene is not established. In this study, we identify PCDH10 as a novel Wnt pathway regulatory element in endometrioid endometrial carcinoma (EEC). PCDH10 was downregulated in EEC tumor cells by aberrant methylation of its promoter. Restoring PCDH10 levels suppressed cell growth and triggered apoptosis in EEC cells and tumor xenografts. Gene expression profiling revealed as part of the transcriptomic changes induced by PCDH10 a reduction in levels of MALAT1, a long noncoding RNA, that mediated tumor suppression functions of PCDH10 in EEC cells. We found that MALAT1 transcription was regulated by Wnt/β-catenin signaling via TCF promoter binding and PCDH10 decreased MALAT1 by modulating this pathway. Clinically, MALAT1 expression was associated with multiple parameters in patients with EEC. Taken together, our findings establish a novel PCDH10-Wnt/β-catenin-MALAT1 regulatory axis that contributes to EEC development. Cancer Res; 74(18); 5103-17. ©2014 AACR.

Collaboration


Dive into the Huating Wang's collaboration.

Top Co-Authors

Avatar

Hao Sun

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Kun Sun

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Leina Lu

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Liang Zhou

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Yu Zhao

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Lijun Wang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Peiyong Jiang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Jiajian Zhou

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Xiaona Chen

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Xichen Bao

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge