Network


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

Hotspot


Dive into the research topics where Henry W. Long is active.

Publication


Featured researches published by Henry W. Long.


Cell | 2013

HIF1A Employs CDK8-Mediator to Stimulate RNAPII Elongation in Response to Hypoxia

Matthew D. Galbraith; Mary A. Allen; Claire L. Bensard; Xiaoxing Wang; Marie K. Schwinn; Bo Qin; Henry W. Long; Danette L. Daniels; William C. Hahn; Robin D. Dowell; Joaquín M. Espinosa

The transcription factor HIF1A is a key mediator of the cellular response to hypoxia. Despite the importance of HIF1A in homeostasis and various pathologies, little is known about how it regulates RNA polymerase II (RNAPII). We report here that HIF1A employs a specific variant of the Mediator complex to stimulate RNAPII elongation. The Mediator-associated kinase CDK8, but not the paralog CDK19, is required for induction of many HIF1A target genes. HIF1A induces binding of CDK8-Mediator and the super elongation complex (SEC), containing AFF4 and CDK9, to alleviate RNAPII pausing. CDK8 is dispensable for HIF1A chromatin binding and histone acetylation, but it is essential for binding of SEC and RNAPII elongation. Global analysis of active RNAPII reveals that hypoxia-inducible genes are paused and active prior to their induction. Our results provide a mechanistic link between HIF1A and CDK8, two potent oncogenes, in the cellular response to hypoxia.


Science | 2017

Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance

Sheng Yu Ku; Spencer Rosario; Yanqing Wang; Ping Mu; Mukund Seshadri; Zachary W. Goodrich; Maxwell M. Goodrich; David P. Labbé; Eduardo Cortes Gomez; Jianmin Wang; Henry W. Long; Bo Xu; Myles Brown; Massimo Loda; Charles L. Sawyers; Leigh Ellis; David W. Goodrich

Evading cancer drugs by identity fraud Prostate cancer growth is fueled by male hormones called androgens. Drugs targeting the androgen receptor (AR) are initially efficacious, but most tumors eventually become resistant (see the Perspective by Kelly and Balk). Mu et al. found that prostate cancer cells escaped the effects of androgen deprivation therapy through a change in lineage identity. Functional loss of the tumor suppressors TP53 and RB1 promoted a shift from AR-dependent luminal epithelial cells to AR-independent basal-like cells. In related work, Ku et al. found that prostate cancer metastasis, lineage switching, and drug resistance were driven by the combined loss of the same tumor suppressors and were accompanied by increased expression of the epigenetic regulator Ezh2. Ezh2 inhibitors reversed the lineage switch and restored sensitivity to androgen deprivation therapy in experimental models. Science, this issue p. 84, p. 78; see also p. 29 Prostate cancer cells escape androgen deprivation therapy by morphing into a cell type that does not require androgens. Prostate cancer relapsing from antiandrogen therapies can exhibit variant histology with altered lineage marker expression, suggesting that lineage plasticity facilitates therapeutic resistance. The mechanisms underlying prostate cancer lineage plasticity are incompletely understood. Studying mouse models, we demonstrate that Rb1 loss facilitates lineage plasticity and metastasis of prostate adenocarcinoma initiated by Pten mutation. Additional loss of Trp53 causes resistance to antiandrogen therapy. Gene expression profiling indicates that mouse tumors resemble human prostate cancer neuroendocrine variants; both mouse and human tumors exhibit increased expression of epigenetic reprogramming factors such as Ezh2 and Sox2. Clinically relevant Ezh2 inhibitors restore androgen receptor expression and sensitivity to antiandrogen therapy. These findings uncover genetic mutations that enable prostate cancer progression; identify mouse models for studying prostate cancer lineage plasticity; and suggest an epigenetic approach for extending clinical responses to antiandrogen therapy.


Nature Methods | 2014

Refined DNase-seq protocol and data analysis reveals intrinsic bias in transcription factor footprint identification.

Housheng Hansen He; Clifford A. Meyer; Sheng'en Shawn Hu; Mei-Wei Chen; Chongzhi Zang; Yin Liu; Prakash Rao; Teng Fei; Han Xu; Henry W. Long; X. Shirley Liu; Myles Brown

Sequencing of DNase I hypersensitive sites (DNase-seq) is a powerful technique for identifying cis-regulatory elements across the genome. We studied the key experimental parameters to optimize performance of DNase-seq. Sequencing short fragments of 50–100 base pairs (bp) that accumulate in long internucleosome linker regions was more efficient for identifying transcription factor binding sites compared to sequencing longer fragments. We also assessed the potential of DNase-seq to predict transcription factor occupancy via generation of nucleotide-resolution transcription factor footprints. In modeling the sequence-specific DNase I cutting bias, we found a strong effect that varied over more than two orders of magnitude. This indicates that the nucleotide-resolution cleavage patterns at many transcription factor binding sites are derived from intrinsic DNase I cleavage bias rather than from specific protein-DNA interactions. In contrast, quantitative comparison of DNase I hypersensitivity between states can predict transcription factor occupancy associated with particular biological perturbations.


Nature | 2014

Broadly permissive intestinal chromatin underlies lateral inhibition and cell plasticity

Tae-Hee Kim; Fugen Li; Isabel Ferreiro-Neira; Li-Lun Ho; Annouck Luyten; Kodandaramireddy Nalapareddy; Henry W. Long; Michael P. Verzi; Ramesh A. Shivdasani

Cells differentiate when transcription factors bind accessible cis-regulatory elements to establish specific gene expression programs. In differentiating embryonic stem cells, chromatin at lineage-restricted genes becomes sequentially accessible, probably by means of ‘pioneer’ transcription factor activity, but tissues may use other strategies in vivo. Lateral inhibition is a pervasive process in which one cell forces a different identity on its neighbours, and it is unclear how chromatin in equipotent progenitors undergoing lateral inhibition quickly enables distinct, transiently reversible cell fates. Here we report the chromatin and transcriptional underpinnings of differentiation in mouse small intestine crypts, where notch signalling mediates lateral inhibition to assign progenitor cells into absorptive or secretory lineages. Transcript profiles in isolated LGR5+ intestinal stem cells and secretory and absorptive progenitors indicated that each cell population was distinct and the progenitors specified. Nevertheless, secretory and absorptive progenitors showed comparable levels of H3K4me2 and H3K27ac histone marks and DNase I hypersensitivity—signifying accessible, permissive chromatin—at most of the same cis-elements. Enhancers acting uniquely in progenitors were well demarcated in LGR5+ intestinal stem cells, revealing early priming of chromatin for divergent transcriptional programs, and retained active marks well after lineages were specified. On this chromatin background, ATOH1, a secretory-specific transcription factor, controls lateral inhibition through delta-like notch ligand genes and also drives the expression of numerous secretory lineage genes. Depletion of ATOH1 from specified secretory cells converted them into functional enterocytes, indicating prolonged responsiveness of marked enhancers to the presence or absence of a key transcription factor. Thus, lateral inhibition and intestinal crypt lineage plasticity involve interaction of a lineage-restricted transcription factor with broadly permissive chromatin established in multipotent stem cells.


Nature Genetics | 2015

The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis

Mark Pomerantz; Fugen Li; David Y. Takeda; Romina Lenci; Apurva Chonkar; Matthew S. Chabot; Paloma Cejas; Francisca Vazquez; Jennifer Cook; Ramesh A. Shivdasani; Michaela Bowden; Rosina T. Lis; William C. Hahn; Philip W. Kantoff; Myles Brown; Massimo Loda; Henry W. Long; Matthew L. Freedman

Master transcription factors interact with DNA to establish cell type identity and to regulate gene expression in mammalian cells. The genome-wide map of these transcription factor binding sites has been termed the cistrome. Here we show that the androgen receptor (AR) cistrome undergoes extensive reprogramming during prostate epithelial transformation in man. Using human prostate tissue, we observed a core set of AR binding sites that are consistently reprogrammed in tumors. FOXA1 and HOXB13 colocalized at the reprogrammed AR binding sites in human tumor tissue. Introduction of FOXA1 and HOXB13 into an immortalized prostate cell line reprogrammed the AR cistrome to resemble that of a prostate tumor, functionally linking these specific factors to AR cistrome reprogramming. These findings offer mechanistic insights into a key set of events that drive normal prostate epithelium toward transformation and establish the centrality of epigenetic reprogramming in human prostate tumorigenesis.


Cancer Cell | 2016

The Public Repository of Xenografts Enables Discovery and Randomized Phase II-like Trials in Mice

Elizabeth Townsend; Mark A. Murakami; Alexandra N. Christodoulou; Amanda L. Christie; Johannes Köster; Tiffany DeSouza; Elizabeth A. Morgan; Scott P. Kallgren; Huiyun Liu; Shuo-Chieh Wu; Olivia Plana; Joan Montero; Kristen E. Stevenson; Prakash Rao; Raga Vadhi; Michael Andreeff; Philippe Armand; Karen K. Ballen; Patrizia Barzaghi-Rinaudo; Sarah Cahill; Rachael A. Clark; Vesselina G. Cooke; Matthew S. Davids; Daniel J. DeAngelo; David M. Dorfman; Hilary Eaton; Benjamin L. Ebert; Julia Etchin; Brant Firestone; David C. Fisher

More than 90% of drugs with preclinical activity fail in human trials, largely due to insufficient efficacy. We hypothesized that adequately powered trials of patient-derived xenografts (PDX) in mice could efficiently define therapeutic activity across heterogeneous tumors. To address this hypothesis, we established a large, publicly available repository of well-characterized leukemia and lymphoma PDXs that undergo orthotopic engraftment, called the Public Repository of Xenografts (PRoXe). PRoXe includes all de-identified information relevant to the primary specimens and the PDXs derived from them. Using this repository, we demonstrate that large studies of acute leukemia PDXs that mimic human randomized clinical trials can characterize drug efficacy and generate transcriptional, functional, and proteomic biomarkers in both treatment-naive and relapsed/refractory disease.


Cancer Discovery | 2016

Oncogenic deregulation of EZH2 as an opportunity for targeted therapy in lung cancer

Hailei Zhang; Jun Qi; Jaime Reyes; Liren Li; Prakash Rao; Fugen Li; Charles Y. Lin; Jennifer A. Perry; Matthew A. Lawlor; Alexander J. Federation; De Raedt T; Yvonne Y. Li; Yin Liu; Melissa Duarte; Yunyu Zhang; Grit S. Herter-Sprie; Eiki Kikuchi; Julian Carretero; Charles M. Perou; Jakob B. Reibel; Joshiawa Paulk; Roderick T. Bronson; Hideo Watanabe; Christine Fillmore Brainson; Carla F. Kim; Peter S. Hammerman; Myles Brown; Karen Cichowski; Henry W. Long; James E. Bradner

UNLABELLED As a master regulator of chromatin function, the lysine methyltransferase EZH2 orchestrates transcriptional silencing of developmental gene networks. Overexpression of EZH2 is commonly observed in human epithelial cancers, such as non-small cell lung carcinoma (NSCLC), yet definitive demonstration of malignant transformation by deregulated EZH2 remains elusive. Here, we demonstrate the causal role of EZH2 overexpression in NSCLC with new genetically engineered mouse models of lung adenocarcinoma. Deregulated EZH2 silences normal developmental pathways, leading to epigenetic transformation independent of canonical growth factor pathway activation. As such, tumors feature a transcriptional program distinct from KRAS- and EGFR-mutant mouse lung cancers, but shared with human lung adenocarcinomas exhibiting high EZH2 expression. To target EZH2-dependent cancers, we developed a potent open-source EZH2 inhibitor, JQEZ5, that promoted the regression of EZH2-driven tumors in vivo, confirming oncogenic addiction to EZH2 in established tumors and providing the rationale for epigenetic therapy in a subset of lung cancer. SIGNIFICANCE EZH2 overexpression induces murine lung cancers that are similar to human NSCLC with high EZH2 expression and low levels of phosphorylated AKT and ERK, implicating biomarkers for EZH2 inhibitor sensitivity. Our EZH2 inhibitor, JQEZ5, promotes regression of these tumors, revealing a potential role for anti-EZH2 therapy in lung cancer. Cancer Discov; 6(9); 1006-21. ©2016 AACR.See related commentary by Frankel et al., p. 949This article is highlighted in the In This Issue feature, p. 932.


Bioinformatics | 2012

CistromeMap: a knowledgebase and web server for ChIP-Seq and DNase-Seq studies in mouse and human

Bo Qin; Meng Zhou; Ying Ge; Len Taing; Tao Liu; Qian Wang; Su Wang; Junsheng Chen; Lingling Shen; Xikun Duan; Shengen Hu; Wei Li; Henry W. Long; Yong Zhang; X. Shirley Liu

SUMMARY Transcription and chromatin regulators, and histone modifications play essential roles in gene expression regulation. We have created CistromeMap as a web server to provide a comprehensive knowledgebase of all of the publicly available ChIP-Seq and DNase-Seq data in mouse and human. We have also manually curated metadata to ensure annotation consistency, and developed a user-friendly display matrix for quick navigation and retrieval of data for specific factors, cells and papers. Finally, we provide users with summary statistics of ChIP-Seq and DNase-Seq studies.


Cell Reports | 2015

Somatic Cell Fusions Reveal Extensive Heterogeneity in Basal-like Breast Cancer

Ying Su; Ashim Subedee; Noga Bloushtain-Qimron; Virginia Savova; Marcin Krzystanek; Lewyn Li; Andriy Marusyk; Doris P. Tabassum; Alexander Zak; Mary Jo Flacker; Mei Li; Jessica J. Lin; Saraswati Sukumar; Hiromu Suzuki; Henry W. Long; Zoltan Szallasi; Alexander A. Gimelbrant; Reo Maruyama; Kornelia Polyak

Basal-like and luminal breast tumors have distinct clinical behavior and molecular profiles, yet the underlying mechanisms are poorly defined. To interrogate processes that determine these distinct phenotypes and their inheritance pattern, we generated somatic cell fusions and performed integrated genetic and epigenetic (DNA methylation and chromatin) profiling. We found that the basal-like trait is generally dominant and is largely defined by epigenetic repression of luminal transcription factors. Definition of super-enhancers highlighted a core program common in luminal cells but a high degree of heterogeneity in basal-like breast cancers that correlates with clinical outcome. We also found that protein extracts of basal-like cells are sufficient to induce a luminal-to-basal phenotypic switch, implying a trigger of basal-like autoregulatory circuits. We determined that KDM6A might be required for luminal-basal fusions, and we identified EN1, TBX18, and TCF4 as candidate transcriptional regulators of the luminal-to-basal switch. Our findings highlight the remarkable epigenetic plasticity of breast cancer cells.


Human Molecular Genetics | 2015

Integration of multiethnic fine-mapping and genomic annotation to prioritize candidate functional SNPs at prostate cancer susceptibility regions

Ying Han; Dennis J. Hazelett; Fredrik Wiklund; Fredrick R. Schumacher; Daniel O. Stram; Sonja I. Berndt; Zhaoming Wang; Kristin A. Rand; Robert N. Hoover; Mitchell J. Machiela; M. Yeager; Laurie Burdette; Charles C. Chung; Amy Hutchinson; Kai Yu; Jianfeng Xu; Ruth C. Travis; Timothy J. Key; Afshan Siddiq; Federico Canzian; Atsushi Takahashi; Michiaki Kubo; Janet L. Stanford; Suzanne Kolb; Susan M. Gapstur; W. Ryan Diver; Victoria L. Stevens; Sara S. Strom; Curtis A. Pettaway; Ali Amin Al Olama

Interpretation of biological mechanisms underlying genetic risk associations for prostate cancer is complicated by the relatively large number of risk variants (n = 100) and the thousands of surrogate SNPs in linkage disequilibrium. Here, we combined three distinct approaches: multiethnic fine-mapping, putative functional annotation (based upon epigenetic data and genome-encoded features), and expression quantitative trait loci (eQTL) analyses, in an attempt to reduce this complexity. We examined 67 risk regions using genotyping and imputation-based fine-mapping in populations of European (cases/controls: 8600/6946), African (cases/controls: 5327/5136), Japanese (cases/controls: 2563/4391) and Latino (cases/controls: 1034/1046) ancestry. Markers at 55 regions passed a region-specific significance threshold (P-value cutoff range: 3.9 × 10(-4)-5.6 × 10(-3)) and in 30 regions we identified markers that were more significantly associated with risk than the previously reported variants in the multiethnic sample. Novel secondary signals (P < 5.0 × 10(-6)) were also detected in two regions (rs13062436/3q21 and rs17181170/3p12). Among 666 variants in the 55 regions with P-values within one order of magnitude of the most-associated marker, 193 variants (29%) in 48 regions overlapped with epigenetic or other putative functional marks. In 11 of the 55 regions, cis-eQTLs were detected with nearby genes. For 12 of the 55 regions (22%), the most significant region-specific, prostate-cancer associated variant represented the strongest candidate functional variant based on our annotations; the number of regions increased to 20 (36%) and 27 (49%) when examining the 2 and 3 most significantly associated variants in each region, respectively. These results have prioritized subsets of candidate variants for downstream functional evaluation.

Collaboration


Dive into the Henry W. Long's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge