Network


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

Hotspot


Dive into the research topics where Weisheng Wu is active.

Publication


Featured researches published by Weisheng Wu.


Nature | 2014

Topologically associating domains are stable units of replication-timing regulation

Benjamin D. Pope; Tyrone Ryba; Vishnu Dileep; Feng Yue; Weisheng Wu; Olgert Denas; Daniel L. Vera; Yanli Wang; R. Scott Hansen; Theresa K. Canfield; Robert E. Thurman; Yong Cheng; Günhan Gülsoy; Jonathan H. Dennis; Michael Snyder; John A. Stamatoyannopoulos; James Taylor; Ross C. Hardison; Tamer Kahveci; Bing Ren; David M. Gilbert

Eukaryotic chromosomes replicate in a temporal order known as the replication-timing program. In mammals, replication timing is cell-type-specific with at least half the genome switching replication timing during development, primarily in units of 400–800 kilobases (‘replication domains’), whose positions are preserved in different cell types, conserved between species, and appear to confine long-range effects of chromosome rearrangements. Early and late replication correlate, respectively, with open and closed three-dimensional chromatin compartments identified by high-resolution chromosome conformation capture (Hi-C), and, to a lesser extent, late replication correlates with lamina-associated domains (LADs). Recent Hi-C mapping has unveiled substructure within chromatin compartments called topologically associating domains (TADs) that are largely conserved in their positions between cell types and are similar in size to replication domains. However, TADs can be further sub-stratified into smaller domains, challenging the significance of structures at any particular scale. Moreover, attempts to reconcile TADs and LADs to replication-timing data have not revealed a common, underlying domain structure. Here we localize boundaries of replication domains to the early-replicating border of replication-timing transitions and map their positions in 18 human and 13 mouse cell types. We demonstrate that, collectively, replication domain boundaries share a near one-to-one correlation with TAD boundaries, whereas within a cell type, adjacent TADs that replicate at similar times obscure replication domain boundaries, largely accounting for the previously reported lack of alignment. Moreover, cell-type-specific replication timing of TADs partitions the genome into two large-scale sub-nuclear compartments revealing that replication-timing transitions are indistinguishable from late-replicating regions in chromatin composition and lamina association and accounting for the reduced correlation of replication timing to LADs and heterochromatin. Our results reconcile cell-type-specific sub-nuclear compartmentalization and replication timing with developmentally stable structural domains and offer a unified model for large-scale chromosome structure and function.


Genome Biology | 2012

An encyclopedia of mouse DNA elements (Mouse ENCODE)

John A. Stamatoyannopoulos; Michael Snyder; Ross C. Hardison; Bing Ren; Thomas R. Gingeras; David M. Gilbert; Mark Groudine; M. A. Bender; Rajinder Kaul; Theresa K. Canfield; Erica Giste; Audra K. Johnson; Mia Zhang; Gayathri Balasundaram; Rachel Byron; Vaughan Roach; Peter J. Sabo; Richard Sandstrom; A Sandra Stehling; Robert E. Thurman; Sherman M. Weissman; Philip Cayting; Manoj Hariharan; Jin Lian; Yong Cheng; Stephen G. Landt; Zhihai Ma; Barbara J. Wold; Job Dekker; Gregory E. Crawford

To complement the human Encyclopedia of DNA Elements (ENCODE) project and to enable a broad range of mouse genomics efforts, the Mouse ENCODE Consortium is applying the same experimental pipelines developed for human ENCODE to annotate the mouse genome.


Proceedings of the National Academy of Sciences of the United States of America | 2008

A GATA-1-regulated microRNA locus essential for erythropoiesis

Louis C. Dore; Julio D. Amigo; Camila O. dos Santos; Zhe Zhang; Xiaowu Gai; John W. Tobias; Duonan Yu; Alyssa M. Klein; Christine M. Dorman; Weisheng Wu; Ross C. Hardison; Barry H. Paw; Mitchell J. Weiss

MicroRNAs (miRNAs) control tissue development, but their mechanism of regulation is not well understood. We used a gene complementation strategy combined with microarray screening to identify miRNAs involved in the formation of erythroid (red blood) cells. Two conserved miRNAs, miR 144 and miR 451, emerged as direct targets of the critical hematopoietic transcription factor GATA-1. In vivo, GATA-1 binds a distal upstream regulatory element to activate RNA polymerase II-mediated transcription of a single common precursor RNA (pri-miRNA) encoding both mature miRNAs. Zebrafish embryos depleted of miR 451 by using antisense morpholinos form erythroid precursors, but their development into mature circulating red blood cells is strongly and specifically impaired. These results reveal a miRNA locus that is required for erythropoiesis and uncover a new regulatory axis through which GATA-1 controls this process.


Nature | 2014

Principles of regulatory information conservation between mouse and human

Yong Cheng; Zhihai Ma; Bong-Hyun Kim; Weisheng Wu; Philip Cayting; Alan P. Boyle; Vasavi Sundaram; Xiaoyun Xing; Nergiz Dogan; Jingjing Li; Ghia Euskirchen; Shin Lin; Yiing Lin; Axel Visel; Trupti Kawli; Xinqiong Yang; Dorrelyn Patacsil; Cheryl A. Keller; Belinda Giardine; Anshul Kundaje; Ting Wang; Len A. Pennacchio; Zhiping Weng; Ross C. Hardison; Michael Snyder

To broaden our understanding of the evolution of gene regulation mechanisms, we generated occupancy profiles for 34 orthologous transcription factors (TFs) in human–mouse erythroid progenitor, lymphoblast and embryonic stem-cell lines. By combining the genome-wide transcription factor occupancy repertoires, associated epigenetic signals, and co-association patterns, here we deduce several evolutionary principles of gene regulatory features operating since the mouse and human lineages diverged. The genomic distribution profiles, primary binding motifs, chromatin states, and DNA methylation preferences are well conserved for TF-occupied sequences. However, the extent to which orthologous DNA segments are bound by orthologous TFs varies both among TFs and with genomic location: binding at promoters is more highly conserved than binding at distal elements. Notably, occupancy-conserved TF-occupied sequences tend to be pleiotropic; they function in several tissues and also co-associate with many TFs. Single nucleotide variants at sites with potential regulatory functions are enriched in occupancy-conserved TF-occupied sequences.


Genome Research | 2011

Dynamics of the epigenetic landscape during erythroid differentiation after GATA1 restoration

Weisheng Wu; Yong Cheng; Cheryl A. Keller; Jason Ernst; Swathi Ashok Kumar; Tejaswini Mishra; Christapher S. Morrissey; Christine M. Dorman; Kuan-Bei Chen; Daniela I. Drautz; Belinda Giardine; Yoichiro Shibata; Lingyun Song; Maxim Pimkin; Gregory E. Crawford; Terrence S. Furey; Manolis Kellis; Webb Miller; James Taylor; Stephan C. Schuster; Yu Zhang; Francesca Chiaromonte; Gerd A. Blobel; Mitchell J. Weiss; Ross C. Hardison

Interplays among lineage-specific nuclear proteins, chromatin modifying enzymes, and the basal transcription machinery govern cellular differentiation, but their dynamics of action and coordination with transcriptional control are not fully understood. Alterations in chromatin structure appear to establish a permissive state for gene activation at some loci, but they play an integral role in activation at other loci. To determine the predominant roles of chromatin states and factor occupancy in directing gene regulation during differentiation, we mapped chromatin accessibility, histone modifications, and nuclear factor occupancy genome-wide during mouse erythroid differentiation dependent on the master regulatory transcription factor GATA1. Notably, despite extensive changes in gene expression, the chromatin state profiles (proportions of a gene in a chromatin state dominated by activating or repressive histone modifications) and accessibility remain largely unchanged during GATA1-induced erythroid differentiation. In contrast, gene induction and repression are strongly associated with changes in patterns of transcription factor occupancy. Our results indicate that during erythroid differentiation, the broad features of chromatin states are established at the stage of lineage commitment, largely independently of GATA1. These determine permissiveness for expression, with subsequent induction or repression mediated by distinctive combinations of transcription factors.


Epigenetics & Chromatin | 2015

Occupancy by key transcription factors is a more accurate predictor of enhancer activity than histone modifications or chromatin accessibility

Nergiz Dogan; Weisheng Wu; Christapher S. Morrissey; Kuan-Bei Chen; Aaron J. Stonestrom; Maria Long; Cheryl A. Keller; Yong Cheng; Deepti Jain; Axel Visel; Len A. Pennacchio; Mitchell J. Weiss; Gerd A. Blobel; Ross C. Hardison

BackgroundRegulated gene expression controls organismal development, and variation in regulatory patterns has been implicated in complex traits. Thus accurate prediction of enhancers is important for further understanding of these processes. Genome-wide measurement of epigenetic features, such as histone modifications and occupancy by transcription factors, is improving enhancer predictions, but the contribution of these features to prediction accuracy is not known. Given the importance of the hematopoietic transcription factor TAL1 for erythroid gene activation, we predicted candidate enhancers based on genomic occupancy by TAL1 and measured their activity. Contributions of multiple features to enhancer prediction were evaluated based on the results of these and other studies.ResultsTAL1-bound DNA segments were active enhancers at a high rate both in transient transfections of cultured cells (39 of 79, or 56%) and transgenic mice (43 of 66, or 65%). The level of binding signal for TAL1 or GATA1 did not help distinguish TAL1-bound DNA segments as active versus inactive enhancers, nor did the density of regulation-related histone modifications. A meta-analysis of results from this and other studies (273 tested predicted enhancers) showed that the presence of TAL1, GATA1, EP300, SMAD1, H3K4 methylation, H3K27ac, and CAGE tags at DNase hypersensitive sites gave the most accurate predictors of enhancer activity, with a success rate over 80% and a median threefold increase in activity. Chromatin accessibility assays and the histone modifications H3K4me1 and H3K27ac were sensitive for finding enhancers, but they have high false positive rates unless transcription factor occupancy is also included.ConclusionsOccupancy by key transcription factors such as TAL1, GATA1, SMAD1, and EP300, along with evidence of transcription, improves the accuracy of enhancer predictions based on epigenetic features.


Genome Research | 2014

Divergent functions of hematopoietic transcription factors in lineage priming and differentiation during erythro-megakaryopoiesis

Maxim Pimkin; Andrew V. Kossenkov; Tejaswini Mishra; Christapher S. Morrissey; Weisheng Wu; Cheryl A. Keller; Gerd A. Blobel; Dongwon Lee; Michael Beer; Ross C. Hardison; Mitchell J. Weiss

Combinatorial actions of relatively few transcription factors control hematopoietic differentiation. To investigate this process in erythro-megakaryopoiesis, we correlated the genome-wide chromatin occupancy signatures of four master hematopoietic transcription factors (GATA1, GATA2, TAL1, and FLI1) and three diagnostic histone modification marks with the gene expression changes that occur during development of primary cultured megakaryocytes (MEG) and primary erythroblasts (ERY) from murine fetal liver hematopoietic stem/progenitor cells. We identified a robust, genome-wide mechanism of MEG-specific lineage priming by a previously described stem/progenitor cell-expressed transcription factor heptad (GATA2, LYL1, TAL1, FLI1, ERG, RUNX1, LMO2) binding to MEG-associated cis-regulatory modules (CRMs) in multipotential progenitors. This is followed by genome-wide GATA factor switching that mediates further induction of MEG-specific genes following lineage commitment. Interaction between GATA and ETS factors appears to be a key determinant of these processes. In contrast, ERY-specific lineage priming is biased toward GATA2-independent mechanisms. In addition to its role in MEG lineage priming, GATA2 plays an extensive role in late megakaryopoiesis as a transcriptional repressor at loci defined by a specific DNA signature. Our findings reveal important new insights into how ERY and MEG lineages arise from a common bipotential progenitor via overlapping and divergent functions of shared hematopoietic transcription factors.


Genome Research | 2014

Dynamic shifts in occupancy by TAL1 are guided by GATA factors and drive large-scale reprogramming of gene expression during hematopoiesis

Weisheng Wu; Christapher S. Morrissey; Cheryl A. Keller; Tejaswini Mishra; Maxim Pimkin; Gerd A. Blobel; Mitchell J. Weiss; Ross C. Hardison

We used mouse ENCODE data along with complementary data from other laboratories to study the dynamics of occupancy and the role in gene regulation of the transcription factor TAL1, a critical regulator of hematopoiesis, at multiple stages of hematopoietic differentiation. We combined ChIP-seq and RNA-seq data in six mouse cell types representing a progression from multilineage precursors to differentiated erythroblasts and megakaryocytes. We found that sites of occupancy shift dramatically during commitment to the erythroid lineage, vary further during terminal maturation, and are strongly associated with changes in gene expression. In multilineage progenitors, the likely target genes are enriched for hematopoietic growth and functions associated with the mature cells of specific daughter lineages (such as megakaryocytes). In contrast, target genes in erythroblasts are specifically enriched for red cell functions. Furthermore, shifts in TAL1 occupancy during erythroid differentiation are associated with gene repression (dissociation) and induction (co-occupancy with GATA1). Based on both enrichment for transcription factor binding site motifs and co-occupancy determined by ChIP-seq, recruitment by GATA transcription factors appears to be a stronger determinant of TAL1 binding to chromatin than the canonical E-box binding site motif. Studies of additional proteins lead to the model that TAL1 regulates expression after being directed to a distinct subset of genomic binding sites in each cell type via its association with different complexes containing master regulators such as GATA2, ERG, and RUNX1 in multilineage cells and the lineage-specific master regulator GATA1 in erythroblasts.


Nucleic Acids Research | 2009

Primary sequence and epigenetic determinants of in vivo occupancy of genomic DNA by GATA1

Ying Zhang; Weisheng Wu; Yong Cheng; David C. King; Robert S. Harris; James Taylor; Francesca Chiaromonte; Ross C. Hardison

DNA sequence motifs and epigenetic modifications contribute to specific binding by a transcription factor, but the extent to which each feature determines occupancy in vivo is poorly understood. We addressed this question in erythroid cells by identifying DNA segments occupied by GATA1 and measuring the level of trimethylation of histone H3 lysine 27 (H3K27me3) and monomethylation of H3 lysine 4 (H3K4me1) along a 66 Mb region of mouse chromosome 7. While 91% of the GATA1-occupied segments contain the consensus binding-site motif WGATAR, only ∼0.7% of DNA segments with such a motif are occupied. Using a discriminative motif enumeration method, we identified additional motifs predictive of occupancy given the presence of WGATAR. The specific motif variant AGATAA and occurrence of multiple WGATAR motifs are both strong discriminators. Combining motifs to pair a WGATAR motif with a binding site motif for GATA1, EKLF or SP1 improves discriminative power. Epigenetic modifications are also strong determinants, with the factor-bound segments highly enriched for H3K4me1 and depleted of H3K27me3. Combining primary sequence and epigenetic determinants captures 52% of the GATA1-occupied DNA segments and substantially increases the specificity, to one out of seven segments with the required motif combination and epigenetic signals being bound.


The FASEB Journal | 2014

Dynamic shifts in transcription factor occupancy drive reprogramming of gene expression during hematopoietic differentiation (LB181)

Ross C. Hardison; Weisheng Wu; Christapher S. Morrissey; Cheryl A. Keller; Tejaswini Mishra; Maxim Pimkin; Gerd A. Blobel; Mitchell J. Weiss; David M. Bodine

Collaboration


Dive into the Weisheng Wu's collaboration.

Top Co-Authors

Avatar

Ross C. Hardison

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Mitchell J. Weiss

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Cheryl A. Keller

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerd A. Blobel

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Maxim Pimkin

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Tejaswini Mishra

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Christine M. Dorman

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge