Network


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

Hotspot


Dive into the research topics where Christopher R. Vakoc is active.

Publication


Featured researches published by Christopher R. Vakoc.


Cell | 2011

BET Bromodomain Inhibition as a Therapeutic Strategy to Target c-Myc

Jake Delmore; Ghayas C Issa; Madeleine E. Lemieux; Peter B. Rahl; Junwei Shi; Hannah M. Jacobs; Efstathios Kastritis; Timothy Gilpatrick; Ronald M. Paranal; Jun Qi; Marta Chesi; Anna C. Schinzel; Michael R. McKeown; Timothy P. Heffernan; Christopher R. Vakoc; P. Leif Bergsagel; Irene M. Ghobrial; Paul G. Richardson; Richard A. Young; William C. Hahn; Kenneth C. Anderson; Andrew L. Kung; James E. Bradner; Constantine S. Mitsiades

MYC contributes to the pathogenesis of a majority of human cancers, yet strategies to modulate the function of the c-Myc oncoprotein do not exist. Toward this objective, we have targeted MYC transcription by interfering with chromatin-dependent signal transduction to RNA polymerase, specifically by inhibiting the acetyl-lysine recognition domains (bromodomains) of putative coactivator proteins implicated in transcriptional initiation and elongation. Using a selective small-molecule bromodomain inhibitor, JQ1, we identify BET bromodomain proteins as regulatory factors for c-Myc. BET inhibition by JQ1 downregulates MYC transcription, followed by genome-wide downregulation of Myc-dependent target genes. In experimental models of multiple myeloma, a Myc-dependent hematologic malignancy, JQ1 produces a potent antiproliferative effect associated with cell-cycle arrest and cellular senescence. Efficacy of JQ1 in three murine models of multiple myeloma establishes the therapeutic rationale for BET bromodomain inhibition in this disease and other malignancies characterized by pathologic activation of c-Myc.


Nature | 2011

RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia

Johannes Zuber; Junwei Shi; Eric Wang; Amy R. Rappaport; Harald Herrmann; Edward Allan R. Sison; Daniel Magoon; Jun Qi; Katharina Blatt; Mark Wunderlich; Meredith J. Taylor; Christopher Johns; Agustin Chicas; James C. Mulloy; Scott C. Kogan; Patrick Brown; Peter Valent; James E. Bradner; Scott W. Lowe; Christopher R. Vakoc

Epigenetic pathways can regulate gene expression by controlling and interpreting chromatin modifications. Cancer cells are characterized by altered epigenetic landscapes, and commonly exploit the chromatin regulatory machinery to enforce oncogenic gene expression programs. Although chromatin alterations are, in principle, reversible and often amenable to drug intervention, the promise of targeting such pathways therapeutically has been limited by an incomplete understanding of cancer-specific dependencies on epigenetic regulators. Here we describe a non-biased approach to probe epigenetic vulnerabilities in acute myeloid leukaemia (AML), an aggressive haematopoietic malignancy that is often associated with aberrant chromatin states. By screening a custom library of small hairpin RNAs (shRNAs) targeting known chromatin regulators in a genetically defined AML mouse model, we identify the protein bromodomain-containing 4 (Brd4) as being critically required for disease maintenance. Suppression of Brd4 using shRNAs or the small-molecule inhibitor JQ1 led to robust antileukaemic effects in vitro and in vivo, accompanied by terminal myeloid differentiation and elimination of leukaemia stem cells. Similar sensitivities were observed in a variety of human AML cell lines and primary patient samples, revealing that JQ1 has broad activity in diverse AML subtypes. The effects of Brd4 suppression are, at least in part, due to its role in sustaining Myc expression to promote aberrant self-renewal, which implicates JQ1 as a pharmacological means to suppress MYC in cancer. Our results establish small-molecule inhibition of Brd4 as a promising therapeutic strategy in AML and, potentially, other cancers, and highlight the utility of RNA interference (RNAi) screening for revealing epigenetic vulnerabilities that can be exploited for direct pharmacological intervention.


Molecular and Cellular Biology | 2006

Profile of Histone Lysine Methylation across Transcribed Mammalian Chromatin

Christopher R. Vakoc; Mira M. Sachdeva; Hongxin Wang; Gerd A. Blobel

ABSTRACT Complex patterns of histone lysine methylation encode distinct functions within chromatin. We previously reported that trimethylation of lysine 9 of histone H3 (H3K9) occurs at both silent heterochromatin and at the transcribed regions of active mammalian genes, suggesting that the extent of histone lysine methylation involved in mammalian gene activation is not completely defined. To identify additional sites of histone methylation that respond to mammalian gene activity, we describe here a comparative assessment of all six known positions of histone lysine methylation and relate them to gene transcription. Using several model loci, we observed high trimethylation of H3K4, H3K9, H3K36, and H3K79 in the transcribed region, consistent with previous findings. We identify H4K20 monomethylation, a modification previously linked with repression, as a mark of transcription elongation in mammalian cells. In contrast, H3K27 monomethylation, a modification enriched at pericentromeric heterochromatin, was observed broadly distributed throughout all euchromatic sites analyzed, with selective depletion in the vicinity of the transcription start sites at active genes. Together, these results underscore that similar to other described methyl-lysine modifications, H4K20 and H3K27 monomethylation are versatile and dynamic with respect to gene activity, suggesting the existence of novel site-specific methyltransferases and demethylases coupled to the transcription cycle.


Molecular and Cellular Biology | 2008

DOT1L/KMT4 Recruitment and H3K79 Methylation Are Ubiquitously Coupled with Gene Transcription in Mammalian Cells

David J. Steger; Martina I. Lefterova; Lei Ying; Aaron J. Stonestrom; Michael Schupp; David Zhuo; Adam L. Vakoc; Ja Eun Kim; Junjie Chen; Mitchell A. Lazar; Gerd A. Blobel; Christopher R. Vakoc

ABSTRACT The histone H3 lysine 79 methyltransferase DOT1L/KMT4 can promote an oncogenic pattern of gene expression through binding with several MLL fusion partners found in acute leukemia. However, the normal function of DOT1L in mammalian gene regulation is poorly understood. Here we report that DOT1L recruitment is ubiquitously coupled with active transcription in diverse mammalian cell types. DOT1L preferentially occupies the proximal transcribed region of active genes, correlating with enrichment of H3K79 di- and trimethylation. Furthermore, Dot1l mutant fibroblasts lacked H3K79 di- and trimethylation at all sites examined, indicating that DOT1L is the sole enzyme responsible for these marks. Importantly, we identified chromatin immunoprecipitation (ChIP) assay conditions necessary for reliable H3K79 methylation detection. ChIP-chip tiling arrays revealed that levels of all degrees of genic H3K79 methylation correlate with mRNA abundance and dynamically respond to changes in gene activity. Conversion of H3K79 monomethylation into di- and trimethylation correlated with the transition from low- to high-level gene transcription. We also observed enrichment of H3K79 monomethylation at intergenic regions occupied by DNA-binding transcriptional activators. Our findings highlight several similarities between the patterning of H3K4 methylation and that of H3K79 methylation in mammalian chromatin, suggesting a widespread mechanism for parallel or sequential recruitment of DOT1L and MLL to genes in their normal “on” state.


Molecular Cell | 2014

The Mechanisms behind the Therapeutic Activity of BET Bromodomain Inhibition

Junwei Shi; Christopher R. Vakoc

The bromodomain and extraterminal (BET) protein Brd4 recruits transcriptional regulatory complexes to acetylated chromatin. While Brd4 is considered to be a general transcriptional regulator, pharmacological inhibition of BET proteins shows therapeutic activity in a variety of different pathologies, particularly in models of cancer and inflammation. Such effects have been attributed to a specific set of downstream target genes whose expression is disproportionately sensitive to pharmacological targeting of BET proteins. Emerging evidence links the transcriptional consequences of BET inhibition to the association of Brd4 with enhancer elements, which tend to be involved in lineage-specific gene regulation. Furthermore, Brd4 engages in direct regulatory interactions with several DNA-binding transcription factors to influence their disease-relevant functions. Here we review the current understanding of molecular mechanisms that underlie the promising therapeutic effects of BET bromodomain inhibition.


Nature Biotechnology | 2015

Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains

Junwei Shi; Eric Wang; Joseph P. Milazzo; Zhihua Wang; Justin B. Kinney; Christopher R. Vakoc

CRISPR-Cas9 genome editing technology holds great promise for discovering therapeutic targets in cancer and other diseases. Current screening strategies target CRISPR-Cas9–induced mutations to the 5′ exons of candidate genes, but this approach often produces in-frame variants that retain functionality, which can obscure even strong genetic dependencies. Here we overcome this limitation by targeting CRISPR-Cas9 mutagenesis to exons encoding functional protein domains. This generates a higher proportion of null mutations and substantially increases the potency of negative selection. We also show that the magnitude of negative selection can be used to infer the functional importance of individual protein domains of interest. A screen of 192 chromatin regulatory domains in murine acute myeloid leukemia cells identifies six known drug targets and 19 additional dependencies. A broader application of this approach may allow comprehensive identification of protein domains that sustain cancer cells and are suitable for drug targeting.


The EMBO Journal | 2005

FOG-1 recruits the NuRD repressor complex to mediate transcriptional repression by GATA-1.

Wei Hong; Minako Nakazawa; Ying-Yu Chen; Rajashree Kori; Christopher R. Vakoc; Carrie Rakowski; Gerd A. Blobel

Transcription factor GATA‐1 and its cofactor FOG‐1 coordinate erythroid cell maturation by activating erythroid‐specific genes and repressing genes associated with the undifferentiated state. Here we show that FOG‐1 binds to the NuRD corepressor complex in vitro and in vivo. The interaction is mediated by a small conserved domain at the extreme N‐terminus of FOG‐1 that is necessary and sufficient for NuRD binding. This domain defines a novel repression module found in diverse transcriptional repressors. NuRD is present at GATA‐1/FOG‐1‐repressed genes in erythroid cells in vivo. Point mutations near the N‐terminus of FOG‐1 that abrogate NuRD binding block gene repression by FOG‐1. Finally, the ability of GATA‐1 to repress transcription was impaired in erythroid cells expressing mutant forms of FOG‐1 that are defective for NuRD binding. Together, these studies show that FOG‐1 and likely other FOG‐like proteins are corepressors that link GATA factors to histone deacetylation and nucleosome remodeling.


Molecular Cell | 2008

Exchange of GATA Factors Mediates Transitions in Looped Chromatin Organization at a Developmentally Regulated Gene Locus

Huie Jing; Christopher R. Vakoc; Lei Ying; Sean Mandat; Hongxin Wang; Xingwu Zheng; Gerd A. Blobel

Enhancers can regulate designate promoters over long distances by forming chromatin loops. Whether chromatin loops are lost or reconfigured during gene repression is largely unexplored. We examined the chromosome conformation of the Kit gene that is expressed during early erythropoiesis but is downregulated upon cell maturation. Kit expression is controlled by sequential occupancy of two GATA family transcription factors. In immature cells, a distal enhancer bound by GATA-2 is in physical proximity with the active Kit promoter. Upon cell maturation, GATA-1 displaces GATA-2 and triggers a loss of the enhancer/promoter interaction. Moreover, GATA-1 reciprocally increases the proximity in nuclear space among distinct downstream GATA elements. GATA-1-induced transitions in chromatin conformation are not simply the consequence of transcription inhibition and require the cofactor FOG-1. This work shows that a GATA factor exchange reconfigures higher-order chromatin organization, and suggests that de novo chromatin loop formation is employed by nuclear factors to specify repressive outcomes.


Genes & Development | 2013

Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation

Junwei Shi; Warren A. Whyte; Cinthya J. Zepeda-Mendoza; Joseph P. Milazzo; Chen Shen; Jae-Seok Roe; Jessica Minder; Fatih Mercan; Eric Wang; Mélanie A. Eckersley-Maslin; Amy E. Campbell; Shinpei Kawaoka; Sarah Shareef; Zhu Zhu; Jude Kendall; Matthias Muhar; Christian Haslinger; Ming Yu; Robert G. Roeder; Michael Wigler; Gerd A. Blobel; Johannes Zuber; David L. Spector; Richard A. Young; Christopher R. Vakoc

Cancer cells frequently depend on chromatin regulatory activities to maintain a malignant phenotype. Here, we show that leukemia cells require the mammalian SWI/SNF chromatin remodeling complex for their survival and aberrant self-renewal potential. While Brg1, an ATPase subunit of SWI/SNF, is known to suppress tumor formation in several cell types, we found that leukemia cells instead rely on Brg1 to support their oncogenic transcriptional program, which includes Myc as one of its key targets. To account for this context-specific function, we identify a cluster of lineage-specific enhancers located 1.7 Mb downstream from Myc that are occupied by SWI/SNF as well as the BET protein Brd4. Brg1 is required at these distal elements to maintain transcription factor occupancy and for long-range chromatin looping interactions with the Myc promoter. Notably, these distal Myc enhancers coincide with a region that is focally amplified in ∼3% of acute myeloid leukemias. Together, these findings define a leukemia maintenance function for SWI/SNF that is linked to enhancer-mediated gene regulation, providing general insights into how cancer cells exploit transcriptional coactivators to maintain oncogenic gene expression programs.


Nature | 2015

Transcriptional plasticity promotes primary and acquired resistance to BET inhibition

Philipp Rathert; Mareike Roth; Tobias Neumann; Felix Muerdter; Jae-Seok Roe; Matthias Muhar; Sumit Deswal; Sabine Cerny-Reiterer; Barbara Peter; Julian Jude; Thomas Hoffmann; Łukasz M. Boryń; Elin Axelsson; Norbert Schweifer; Ulrike Tontsch-Grunt; Lukas E. Dow; Davide Gianni; Mark Pearson; Peter Valent; Alexander Stark; Norbert Kraut; Christopher R. Vakoc; Johannes Zuber

Following the discovery of BRD4 as a non-oncogene addiction target in acute myeloid leukaemia (AML), bromodomain and extra terminal protein (BET) inhibitors are being explored as a promising therapeutic avenue in numerous cancers. While clinical trials have reported single-agent activity in advanced haematological malignancies, mechanisms determining the response to BET inhibition remain poorly understood. To identify factors involved in primary and acquired BET resistance in leukaemia, here we perform a chromatin-focused RNAi screen in a sensitive MLL–AF9;NrasG12D-driven AML mouse model, and investigate dynamic transcriptional profiles in sensitive and resistant mouse and human leukaemias. Our screen shows that suppression of the PRC2 complex, contrary to effects in other contexts, promotes BET inhibitor resistance in AML. PRC2 suppression does not directly affect the regulation of Brd4-dependent transcripts, but facilitates the remodelling of regulatory pathways that restore the transcription of key targets such as Myc. Similarly, while BET inhibition triggers acute MYC repression in human leukaemias regardless of their sensitivity, resistant leukaemias are uniformly characterized by their ability to rapidly restore MYC transcription. This process involves the activation and recruitment of WNT signalling components, which compensate for the loss of BRD4 and drive resistance in various cancer models. Dynamic chromatin immunoprecipitation sequencing and self-transcribing active regulatory region sequencing of enhancer profiles reveal that BET-resistant states are characterized by remodelled regulatory landscapes, involving the activation of a focal MYC enhancer that recruits WNT machinery in response to BET inhibition. Together, our results identify and validate WNT signalling as a driver and candidate biomarker of primary and acquired BET resistance in leukaemia, and implicate the rewiring of transcriptional programs as an important mechanism promoting resistance to BET inhibitors and, potentially, other chromatin-targeted therapies.

Collaboration


Dive into the Christopher R. Vakoc's collaboration.

Top Co-Authors

Avatar

Junwei Shi

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Gerd A. Blobel

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Johannes Zuber

Research Institute of Molecular Pathology

View shared research outputs
Top Co-Authors

Avatar

Jae-Seok Roe

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Eric Wang

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Scott W. Lowe

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anand S. Bhagwat

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Chen Shen

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar

Joseph P. Milazzo

Cold Spring Harbor Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge