Network


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

Hotspot


Dive into the research topics where Lijing Yao is active.

Publication


Featured researches published by Lijing Yao.


Nature Neuroscience | 2015

The PsychENCODE project

Schahram Akbarian; Chunyu Liu; James A. Knowles; Flora M. Vaccarino; Peggy J. Farnham; Gregory E. Crawford; Andrew E. Jaffe; Dalila Pinto; Stella Dracheva; Daniel H. Geschwind; Jonathan Mill; Angus C. Nairn; Alexej Abyzov; Sirisha Pochareddy; Shyam Prabhakar; Sherman M. Weissman; Patrick F. Sullivan; Matthew W. State; Zhiping Weng; Mette A. Peters; Kevin P. White; Mark Gerstein; Anahita Amiri; Chris Armoskus; Allison E. Ashley-Koch; Taejeong Bae; Andrea Beckel-Mitchener; Benjamin P. Berman; Gerhard A. Coetzee; Gianfilippo Coppola

Recent research on disparate psychiatric disorders has implicated rare variants in genes involved in global gene regulation and chromatin modification, as well as many common variants located primarily in regulatory regions of the genome. Understanding precisely how these variants contribute to disease will require a deeper appreciation for the mechanisms of gene regulation in the developing and adult human brain. The PsychENCODE project aims to produce a public resource of multidimensional genomic data using tissue- and cell type–specific samples from approximately 1,000 phenotypically well-characterized, high-quality healthy and disease-affected human post-mortem brains, as well as functionally characterize disease-associated regulatory elements and variants in model systems. We are beginning with a focus on autism spectrum disorder, bipolar disorder and schizophrenia, and expect that this knowledge will apply to a wide variety of psychiatric disorders. This paper outlines the motivation and design of PsychENCODE.


Genome Biology | 2012

Cell type-specific binding patterns reveal that TCF7L2 can be tethered to the genome by association with GATA3

Seth Frietze; Rui Wang; Lijing Yao; Yu Gyoung Tak; Zhenqing Ye; Malaina Gaddis; Heather Witt; Peggy J. Farnham; Victor X. Jin

BackgroundThe TCF7L2 transcription factor is linked to a variety of human diseases, including type 2 diabetes and cancer. One mechanism by which TCF7L2 could influence expression of genes involved in diverse diseases is by binding to distinct regulatory regions in different tissues. To test this hypothesis, we performed ChIP-seq for TCF7L2 in six human cell lines.ResultsWe identified 116,000 non-redundant TCF7L2 binding sites, with only 1,864 sites common to the six cell lines. Using ChIP-seq, we showed that many genomic regions that are marked by both H3K4me1 and H3K27Ac are also bound by TCF7L2, suggesting that TCF7L2 plays a critical role in enhancer activity. Bioinformatic analysis of the cell type-specific TCF7L2 binding sites revealed enrichment for multiple transcription factors, including HNF4alpha and FOXA2 motifs in HepG2 cells and the GATA3 motif in MCF7 cells. ChIP-seq analysis revealed that TCF7L2 co-localizes with HNF4alpha and FOXA2 in HepG2 cells and with GATA3 in MCF7 cells. Interestingly, in MCF7 cells the TCF7L2 motif is enriched in most TCF7L2 sites but is not enriched in the sites bound by both GATA3 and TCF7L2. This analysis suggested that GATA3 might tether TCF7L2 to the genome at these sites. To test this hypothesis, we depleted GATA3 in MCF7 cells and showed that TCF7L2 binding was lost at a subset of sites. RNA-seq analysis suggested that TCF7L2 represses transcription when tethered to the genome via GATA3.ConclusionsOur studies demonstrate a novel relationship between GATA3 and TCF7L2, and reveal important insights into TCF7L2-mediated gene regulation.


Genome Biology | 2014

Global loss of DNA methylation uncovers intronic enhancers in genes showing expression changes

Adam Blattler; Lijing Yao; Heather Witt; Yu Guo; Charles M. Nicolet; Benjamin P. Berman; Peggy J. Farnham

BackgroundGene expression is epigenetically regulated by a combination of histone modifications and methylation of CpG dinucleotides in promoters. In normal cells, CpG-rich promoters are typically unmethylated, marked with histone modifications such as H3K4me3, and are highly active. During neoplastic transformation, CpG dinucleotides of CG-rich promoters become aberrantly methylated, corresponding with the removal of active histone modifications and transcriptional silencing. Outside of promoter regions, distal enhancers play a major role in the cell type-specific regulation of gene expression. Enhancers, which function by bringing activating complexes to promoters through chromosomal looping, are also modulated by a combination of DNA methylation and histone modifications.ResultsHere we use HCT116 colorectal cancer cells with and without mutations in DNA methyltransferases, the latter of which results in a 95% reduction in global DNA methylation levels. These cells are used to study the relationship between DNA methylation, histone modifications, and gene expression. We find that the loss of DNA methylation is not sufficient to reactivate most of the silenced promoters. In contrast, the removal of DNA methylation results in the activation of a large number of enhancer regions as determined by the acquisition of active histone marks.ConclusionsAlthough the transcriptome is largely unaffected by the loss of DNA methylation, we identify two distinct mechanisms resulting in the upregulation of distinct sets of genes. One is a direct result of DNA methylation loss at a set of promoter regions and the other is due to the presence of new intragenic enhancers.


Nature Communications | 2014

Functional annotation of colon cancer risk SNPs

Lijing Yao; Yu Gyoung Tak; Benjamin P. Berman; Peggy J. Farnham

Colorectal cancer (CRC) is a leading cause of cancer-related deaths in the United States. Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) associated with increased risk for CRC. A molecular understanding of the functional consequences of this genetic variation has been complicated because each GWAS SNP is a surrogate for hundreds of other SNPs, most of which are located in non-coding regions. Here we use genomic and epigenomic information to test the hypothesis that the GWAS SNPs and/or correlated SNPs are in elements that regulate gene expression, and identify 23 promoters and 28 enhancers. Using gene expression data from normal and tumour cells, we identify 66 putative target genes of the risk-associated enhancers (10 of which were also identified by promoter SNPs). Employing CRISPR nucleases, we delete one risk-associated enhancer and identify genes showing altered expression. We suggest that similar studies be performed to characterize all CRC risk-associated enhancers.


Genome Biology | 2015

Inferring regulatory element landscapes and transcription factor networks from cancer methylomes

Lijing Yao; Hui Shen; Peter W. Laird; Peggy J. Farnham; Benjamin P. Berman

Recent studies indicate that DNA methylation can be used to identify transcriptional enhancers, but no systematic approach has been developed for genome-wide identification and analysis of enhancers based on DNA methylation. We describe ELMER (Enhancer Linking by Methylation/Expression Relationships), an R-based tool that uses DNA methylation to identify enhancers and correlates enhancer state with expression of nearby genes to identify transcriptional targets. Transcription factor motif analysis of enhancers is coupled with expression analysis of transcription factors to infer upstream regulators. Using ELMER, we investigated more than 2,000 tumor samples from The Cancer Genome Atlas. We identified networks regulated by known cancer drivers such as GATA3 and FOXA1 (breast cancer), SOX17 and FOXA2 (endometrial cancer), and NFE2L2, SOX2, and TP63 (squamous cell lung cancer). We also identified novel networks with prognostic associations, including RUNX1 in kidney cancer. We propose ELMER as a powerful new paradigm for understanding the cis-regulatory interface between cancer-associated transcription factors and their functional target genes.


Epigenetics & Chromatin | 2013

ZBTB33 binds unmethylated regions of the genome associated with actively expressed genes

Adam Blattler; Lijing Yao; Yao Wang; Zhenqing Ye; Victor X. Jin; Peggy J. Farnham

BackgroundDNA methylation and repressive histone modifications cooperate to silence promoters. One mechanism by which regions of methylated DNA could acquire repressive histone modifications is via methyl DNA-binding transcription factors. The zinc finger protein ZBTB33 (also known as Kaiso) has been shown in vitro to bind preferentially to methylated DNA and to interact with the SMRT/NCoR histone deacetylase complexes. We have performed bioinformatic analyses of Kaiso ChIP-seq and DNA methylation datasets to test a model whereby binding of Kaiso to methylated CpGs leads to loss of acetylated histones at target promoters.ResultsOur results suggest that, contrary to expectations, Kaiso does not bind to methylated DNA in vivo but instead binds to highly active promoters that are marked with high levels of acetylated histones. In addition, our studies suggest that DNA methylation and nucleosome occupancy patterns restrict access of Kaiso to potential binding sites and influence cell type-specific binding.ConclusionsWe propose a new model for the genome-wide binding and function of Kaiso whereby Kaiso binds to unmethylated regulatory regions and contributes to the active state of target promoters.


Critical Reviews in Biochemistry and Molecular Biology | 2015

Demystifying the secret mission of enhancers: linking distal regulatory elements to target genes

Lijing Yao; Benjamin P. Berman; Peggy J. Farnham

Abstract Enhancers are short regulatory sequences bound by sequence-specific transcription factors and play a major role in the spatiotemporal specificity of gene expression patterns in development and disease. While it is now possible to identify enhancer regions genomewide in both cultured cells and primary tissues using epigenomic approaches, it has been more challenging to develop methods to understand the function of individual enhancers because enhancers are located far from the gene(s) that they regulate. However, it is essential to identify target genes of enhancers not only so that we can understand the role of enhancers in disease but also because this information will assist in the development of future therapeutic options. After reviewing models of enhancer function, we discuss recent methods for identifying target genes of enhancers. First, we describe chromatin structure-based approaches for directly mapping interactions between enhancers and promoters. Second, we describe the use of correlation-based approaches to link enhancer state with the activity of nearby promoters and/or gene expression. Third, we describe how to test the function of specific enhancers experimentally by perturbing enhancer–target relationships using high-throughput reporter assays and genome editing. Finally, we conclude by discussing as yet unanswered questions concerning how enhancers function, how target genes can be identified, and how to distinguish direct from indirect changes in gene expression mediated by individual enhancers.


Nucleic Acids Research | 2016

Effects on the transcriptome upon deletion of a distal element cannot be predicted by the size of the H3K27Ac peak in human cells

Yu Gyoung Tak; Yuli Hung; Lijing Yao; Matthew R. Grimmer; Albert Do; Mital S. Bhakta; Henriette O'Geen; David J. Segal; Peggy J. Farnham

Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) associated with increased risk for colorectal cancer (CRC). A molecular understanding of the functional consequences of this genetic variation is complicated because most GWAS SNPs are located in non-coding regions. We used epigenomic information to identify H3K27Ac peaks in HCT116 colon cancer cells that harbor SNPs associated with an increased risk for CRC. Employing CRISPR/Cas9 nucleases, we deleted a CRC risk-associated H3K27Ac peak from HCT116 cells and observed large-scale changes in gene expression, resulting in decreased expression of many nearby genes. As a comparison, we showed that deletion of a robust H3K27Ac peak not associated with CRC had minimal effects on the transcriptome. Interestingly, although there is no H3K27Ac peak in HEK293 cells in the E7 region, deletion of this region in HEK293 cells decreased expression of several of the same genes that were downregulated in HCT116 cells, including the MYC oncogene. Accordingly, deletion of E7 causes changes in cell culture assays in HCT116 and HEK293 cells. In summary, we show that effects on the transcriptome upon deletion of a distal regulatory element cannot be predicted by the size or presence of an H3K27Ac peak.


Epigenetics & Chromatin | 2016

Identification of activated enhancers and linked transcription factors in breast, prostate, and kidney tumors by tracing enhancer networks using epigenetic traits

Suhn Kyong Rhie; Yu Guo; Yu Gyoung Tak; Lijing Yao; Hui Shen; Gerhard A. Coetzee; Peter W. Laird; Peggy J. Farnham

BackgroundAlthough technological advances now allow increased tumor profiling, a detailed understanding of the mechanisms leading to the development of different cancers remains elusive. Our approach toward understanding the molecular events that lead to cancer is to characterize changes in transcriptional regulatory networks between normal and tumor tissue. Because enhancer activity is thought to be critical in regulating cell fate decisions, we have focused our studies on distal regulatory elements and transcription factors that bind to these elements.ResultsUsing DNA methylation data, we identified more than 25,000 enhancers that are differentially activated in breast, prostate, and kidney tumor tissues, as compared to normal tissues. We then developed an analytical approach called Tracing Enhancer Networks using Epigenetic Traits that correlates DNA methylation levels at enhancers with gene expression to identify more than 800,000 genome-wide links from enhancers to genes and from genes to enhancers. We found more than 1200 transcription factors to be involved in these tumor-specific enhancer networks. We further characterized several transcription factors linked to a large number of enhancers in each tumor type, including GATA3 in non-basal breast tumors, HOXC6 and DLX1 in prostate tumors, and ZNF395 in kidney tumors. We showed that HOXC6 and DLX1 are associated with different clusters of prostate tumor-specific enhancers and confer distinct transcriptomic changes upon knockdown in C42B prostate cancer cells. We also discovered de novo motifs enriched in enhancers linked to ZNF395 in kidney tumors.ConclusionsOur studies characterized tumor-specific enhancers and revealed key transcription factors involved in enhancer networks for specific tumor types and subgroups. Our findings, which include a large set of identified enhancers and transcription factors linked to those enhancers in breast, prostate, and kidney cancers, will facilitate understanding of enhancer networks and mechanisms leading to the development of these cancers.


Clinical Cancer Research | 2016

Abstract 22: Inferring regulatory element landscapes and transcription factor networks from cancer methylomes

Lijing Yao; Hui Shen; Peter W. Laird; Peggy J. Farnham; Berman Benjamin

Genome-wide DNA methylation studies have established that transcriptional enhancers can be detected from changes in methylation patterns. However, no systematic approach has been developed to infer the function of these elements within larger transcription factor networks. We developed ELMER (Enhancer Linking by Methylation/Expression Relationships), a computational tool that combines DNA methylation and gene expression data from tissue samples, along with annotations from epigenomic repositories, to infer multi-level cis-regulatory networks. ELMER uses DNA methylation to identify enhancers that differ between normal and disease tissues, and correlates these with gene expression to identify a target gene or genes (typically not the nearest gene). Transcription factor (TF) binding site analysis is then used in conjunction with TF expression patterns to infer the upstream regulators that drive enhancer changes. We applied ELMER to Infinium HM450k methylation and RNA-seq data for 2,000+ tumor samples and ten cancer types from The Cancer Genome Atlas (TCGA). Our multi-level analysis identified enhancer networks regulated by known cancer driver transcription factors, such as GATA3 and FOXA1 in breast cancer, FOXA2 and SOX17 in endometrial cancer, and NFE2L2, SOX2, and TP63 in squamous cell lung cancer. We also identified several novel networks with prognostic associations, including RUNX1 in kidney cancer, and putative target genes of these transcription factors in primary tumor. Citation Format: Lijing Yao, Hui Shen, Peter Laird, Peggy Farnham, Berman Benjamin. Inferring regulatory element landscapes and transcription factor networks from cancer methylomes. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Integrating Clinical Genomics and Cancer Therapy; Jun 13-16, 2015; Salt Lake City, UT. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(1_Suppl):Abstract nr 22.

Collaboration


Dive into the Lijing Yao's collaboration.

Top Co-Authors

Avatar

Peggy J. Farnham

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Benjamin P. Berman

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yu Gyoung Tak

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam Blattler

University of California

View shared research outputs
Top Co-Authors

Avatar

Dennis J. Hazelett

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gerhard A. Coetzee

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Heather Witt

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Simon G. Coetzee

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge