Network


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

Hotspot


Dive into the research topics where Leina Lu is active.

Publication


Featured researches published by Leina Lu.


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.


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.


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.


Cell Stem Cell | 2012

Pax3/7BP Is a Pax7- and Pax3-Binding Protein that Regulates the Proliferation of Muscle Precursor Cells by an Epigenetic Mechanism

Yarui Diao; Xing Guo; Yanfeng Li; Kun Sun; Leina Lu; Lei Jiang; Xinrong Fu; Han Zhu; Hao Sun; Huating Wang; Zhenguo Wu

In mouse skeletal muscles, Pax7 uniquely marks muscle satellite cells and plays some important yet unknown functions at the perinatal stage. To elucidate its in vivo functions, we initiated a yeast two-hybrid screening to look for Pax7-interacting proteins and identified a previously uncharacterized Pax7- and Pax3-binding protein (Pax3/7BP). Pax3/7BP is a ubiquitously expressed nuclear protein, enriched in Pax7+ muscle precursor cells (MPCs), and serves as an indispensable adaptor for Pax7 to recruit the histone 3 lysine 4 (H3K4) methyltransferase (HMT) complex by bridging Pax7 and Wdr5. Knockdown of Pax3/7BP abolished the Pax3/7-associated H3K4 HMT activity and inhibited the proliferation of Pax7+ MPCs from young mice both in culture and in vivo. Id3 and Cdc20 were direct target genes of Pax7 and Pax3/7BP involved in the proliferation of Pax7+ MPCs. Collectively, our work establishes Pax3/7BP as an essential adaptor linking Pax3/7 with the H3K4 HMT to regulate the proliferation of MPCs.


Oncogene | 2013

A novel miR-193a-5p-YY1-APC regulatory axis in human endometrioid endometrial adenocarcinoma

Yuh Cheng Yang; Liangfu Zhou; Leina Lu; Lijun Wang; Xiaoxing Li; Peiyong Jiang; Loucia K.Y. Chan; Tao Zhang; Jun Yu; Joseph Kwong; Tak-Hong Cheung; Tony K.H. Chung; Kinglun Kingston Mak; Hao Sun; Huating Wang

Aberrant expression and altered function of transcription factors (TFs) have vital roles in many aspects of tumor development and progression. In this study, we investigated the functional significance of a TF, Yin Yang1 (YY1) in tumorigenesis of endometrioid endometrial carcinoma (EEC). We demonstrated that YY1 is upregulated in EEC cell lines and primary tumors; and its expression is associated with tumor stages. Depletion of YY1 inhibits EEC cell proliferation and migration both in vitro and in vivo, whereas overexpression of YY1 promotes EEC cell growth. These results suggest that YY1 functions as an oncogenic factor in EEC. Transcriptome analysis revealed a significant effect of YY1 on critical aspects of EEC tumorigenesis through inhibition of APC expression. Further mechanistic investigation uncovered a new epigenetic silencing mode of APC by YY1 through recruitment of EZH2 and trimethylation of histone 3 lysine 27 on its promoter region. Moreover, YY1 overexpression was found to be a consequence of miR-193a-5p downregulation through direct miR-193a-5p-YY1 interplay. Our results therefore establish a novel miR-193a-5p-YY1-APC axis, which contributes to EEC development, and may serve as future intervention target.


Nature Communications | 2015

Linc-YY1 promotes myogenic differentiation and muscle regeneration through an interaction with the transcription factor YY1

Liang Zhou; Kun Sun; Yu Zhao; Suyang Zhang; Xuecong Wang; Yuying Li; Leina Lu; Xiaona Chen; Fengyuan Chen; Xichen Bao; Xihua Zhu; Lijun Wang; Ling Yin Tang; Miguel A. Esteban; Chi Chiu Wang; Ralf Jauch; Hao Sun; Huating Wang

Little is known how lincRNAs are involved in skeletal myogenesis. Here we describe the discovery of Linc-YY1 from the promoter of the transcription factor (TF) Yin Yang 1 (YY1) gene. We demonstrate that Linc-YY1 is dynamically regulated during myogenesis in vitro and in vivo. Gain or loss of function of Linc-YY1 in C2C12 myoblasts or muscle satellite cells alters myogenic differentiation and in injured muscles has an impact on the course of regeneration. Linc-YY1 interacts with YY1 through its middle domain, to evict YY1/Polycomb repressive complex (PRC2) from target promoters, thus activating the gene expression in trans. In addition, Linc-YY1 also regulates PRC2-independent function of YY1. Finally, we identify a human Linc-YY1 orthologue with conserved function and show that many human and mouse TF genes are associated with lincRNAs that may modulate their activity. Altogether, we show that Linc-YY1 regulates skeletal myogenesis and uncover a previously unappreciated mechanism of gene regulation by lincRNA.


Gut | 2013

Zinc-finger protein 545 is a novel tumour suppressor that acts by inhibiting ribosomal RNA transcription in gastric cancer

Shiyan Wang; Yingduan Cheng; Wan Du; Leina Lu; Liang Zhou; Huating Wang; Wei Kang; Xiaoxing Li; Qian Tao; Joseph J.Y. Sung; Jun Yu

Objective Zinc-finger protein 545 (ZNF545) is a member of the family of Krüppel-associated box-containing zinc-finger proteins. The aim of this study was to clarify its biological function as a tumour suppressor in gastric cancer. Design The biological function of ZNF545 was determined by cell growth and apoptosis assays. The ZNF545 target signal pathway was identified by promoter luciferase assay, northern blot, run-on transcription assay, chromatin immunoprecipitation and coimmunoprecipitation assays. The clinical application of ZNF545 was assessed in primary gastric cancers. Results ZNF545 was silenced or reduced in 16 out of 18 gastric cancer cell lines by promoter hypermethylation. Restoration of ZNF545 expression in gastric cancer cell lines suppressed cell proliferation and induced apoptosis. These effects of ZNF545 were attributed to inhibition of ribosomal RNA (rRNA) transcription. Inhibition of rRNA transcription by ZNF545 was further revealed to be associated with direct ribosomal DNA (rDNA) promoter binding, recruitment of the corepressor, heterochromatin protein 1β, and reduction of trimethylated histone H3 at the Lys4 residue at the rDNA locus. ZNF545 methylation was detected in 51.9% (41/79) of gastric cancer tissues, 27.0% (20/74) of adjacent non-tumour gastric tissues (p=0.001), but none of 20 normal controls. Multivariate analysis revealed that patients with ZNF545 methylation had a significant decrease in overall survival. Kaplan–Meier survival curves showed that ZNF545 methylation was significantly associated with shortened survival in patients with stage I–II gastric cancer. Conclusions ZNF545 acts as a functional tumour suppressor in gastric cancer by inhibiting rRNA transcription. Its methylation at early stages of gastric carcinogenesis is an independent prognostic factor.


Cell Stem Cell | 2014

Transcriptional Pause Release Is a Rate-Limiting Step for Somatic Cell Reprogramming

Longqi Liu; Yan Xu; Minghui He; Meng Zhang; Fenggong Cui; Leina Lu; Mingze Yao; Weihua Tian; Christina Benda; Qiang Zhuang; Zhijian Huang; Wenjuan Li; Xiangchun Li; Ping Zhao; Wenxia Fan; Zhiwei Luo; Yuan Li; Yasong Wu; Andrew Paul Hutchins; Dongye Wang; Hung-Fat Tse; Axel Schambach; Jon Frampton; Baoming Qin; Xichen Bao; Hongjie Yao; Biliang Zhang; Hao Sun; Duanqing Pei; Huating Wang

Reactivation of the pluripotency network during somatic cell reprogramming by exogenous transcription factors involves chromatin remodeling and the recruitment of RNA polymerase II (Pol II) to target loci. Here, we report that Pol II is engaged at pluripotency promoters in reprogramming but remains paused and inefficiently released. We also show that bromodomain-containing protein 4 (BRD4) stimulates productive transcriptional elongation of pluripotency genes by dissociating the pause release factor P-TEFb from an inactive complex containing HEXIM1. Consequently, BRD4 overexpression enhances reprogramming efficiency and HEXIM1 suppresses it, whereas Brd4 and Hexim1 knockdown do the opposite. We further demonstrate that the reprogramming factor KLF4 helps recruit P-TEFb to pluripotency promoters. Our work thus provides a mechanism for explaining the reactivation of pluripotency genes in reprogramming and unveils an unanticipated role for KLF4 in transcriptional pause release.

Collaboration


Dive into the Leina Lu's collaboration.

Top Co-Authors

Avatar

Huating Wang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Hao Sun

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

Lijun Wang

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

Peiyong Jiang

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

Yu Zhao

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Xichen Bao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Eric Z. Chen

The Chinese University of Hong Kong

View shared research outputs
Researchain Logo
Decentralizing Knowledge