Network


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

Hotspot


Dive into the research topics where Danette L. Daniels is active.

Publication


Featured researches published by Danette L. Daniels.


The EMBO Journal | 2013

TET2 and TET3 regulate GlcNAcylation and H3K4 methylation through OGT and SET1/COMPASS.

Rachel Deplus; Benjamin Delatte; Marie K. Schwinn; Matthieu Defrance; Jacqui Mendez; Nancy Murphy; Mark A. Dawson; Michael Volkmar; Pascale Putmans; Emilie Calonne; Alan H. Shih; Ross L. Levine; Olivier A. Bernard; Thomas Mercher; Eric Solary; Marjeta Urh; Danette L. Daniels; François Fuks

TET proteins convert 5‐methylcytosine to 5‐hydroxymethylcytosine, an emerging dynamic epigenetic state of DNA that can influence transcription. Evidence has linked TET1 function to epigenetic repression complexes, yet mechanistic information, especially for the TET2 and TET3 proteins, remains limited. Here, we show a direct interaction of TET2 and TET3 with O‐GlcNAc transferase (OGT). OGT does not appear to influence hmC activity, rather TET2 and TET3 promote OGT activity. TET2/3–OGT co‐localize on chromatin at active promoters enriched for H3K4me3 and reduction of either TET2/3 or OGT activity results in a direct decrease in H3K4me3 and concomitant decreased transcription. Further, we show that Host Cell Factor 1 (HCF1), a component of the H3K4 methyltransferase SET1/COMPASS complex, is a specific GlcNAcylation target of TET2/3–OGT, and modification of HCF1 is important for the integrity of SET1/COMPASS. Additionally, we find both TET proteins and OGT activity promote binding of the SET1/COMPASS H3K4 methyltransferase, SETD1A, to chromatin. Finally, studies in Tet2 knockout mouse bone marrow tissue extend and support the data as decreases are observed of global GlcNAcylation and also of H3K4me3, notably at several key regulators of haematopoiesis. Together, our results unveil a step‐wise model, involving TET–OGT interactions, promotion of GlcNAcylation, and influence on H3K4me3 via SET1/COMPASS, highlighting a novel means by which TETs may induce transcriptional activation.


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.


Cell | 2013

KDM4A Lysine Demethylase Induces Site-Specific Copy Gain and Rereplication of Regions Amplified in Tumors

Joshua C. Black; Amity L. Manning; Capucine Van Rechem; Jaegil Kim; Brendon Ladd; Juok Cho; Cristiana M. Pineda; Nancy Murphy; Danette L. Daniels; Cristina Montagna; Peter W. Lewis; Kimberly Glass; C. David Allis; Nicholas J. Dyson; Gad Getz; Johnathan R. Whetstine

Acquired chromosomal instability and copy number alterations are hallmarks of cancer. Enzymes capable of promoting site-specific copy number changes have yet to be identified. Here, we demonstrate that H3K9/36me3 lysine demethylase KDM4A/JMJD2A overexpression leads to localized copy gain of 1q12, 1q21, and Xq13.1 without global chromosome instability. KDM4A-amplified tumors have increased copy gains for these same regions. 1q12h copy gain occurs within a single cell cycle, requires S phase, and is not stable but is regenerated each cell division. Sites with increased copy number are rereplicated and have increased KDM4A, MCM, and DNA polymerase occupancy. Suv39h1/KMT1A or HP1γ overexpression suppresses the copy gain, whereas H3K9/K36 methylation interference promotes gain. Our results demonstrate that overexpression of a chromatin modifier results in site-specific copy gains. This begins to establish how copy number changes could originate during tumorigenesis and demonstrates that transient overexpression of specific chromatin modulators could promote these events.


ACS Chemical Biology | 2015

NanoBRET--A Novel BRET Platform for the Analysis of Protein-Protein Interactions.

Thomas Machleidt; Carolyn C. Woodroofe; Marie K. Schwinn; Jacqui Mendez; Matthew B. Robers; Kris Zimmerman; Paul Otto; Danette L. Daniels; Thomas A. Kirkland; Keith V. Wood

Dynamic interactions between proteins comprise a key mechanism for temporal control of cellular function and thus hold promise for development of novel drug therapies. It remains technically challenging, however, to quantitatively characterize these interactions within the biologically relevant context of living cells. Although, bioluminescence resonance energy transfer (BRET) has often been used for this purpose, its general applicability has been hindered by limited sensitivity and dynamic range. We have addressed this by combining an extremely bright luciferase (Nanoluc) with a means for tagging intracellular proteins with a long-wavelength fluorophore (HaloTag). The small size (19 kDa), high emission intensity, and relatively narrow spectrum (460 nm peak intensity) make Nanoluc luciferase well suited as an energy donor. By selecting an efficient red-emitting fluorophore (635 nm peak intensity) for attachment onto the HaloTag, an overall spectral separation exceeding 175 nm was achieved. This combination of greater light intensity with improved spectral resolution results in substantially increased detection sensitivity and dynamic range over current BRET technologies. Enhanced performance is demonstrated using several established model systems, as well as the ability to image BRET in individual cells. The capabilities are further exhibited in a novel assay developed for analyzing the interactions of bromodomain proteins with chromatin in living cells.


Cancer Research | 2015

Generation of a Selective Small Molecule Inhibitor of the CBP/p300 Bromodomain for Leukemia Therapy

Sarah Picaud; Oleg Fedorov; Angeliki Thanasopoulou; Katharina Leonards; Katherine Louise Jones; Julia Meier; Heidi Olzscha; Octovia P. Monteiro; Sarah Martin; Martin Philpott; Anthony Tumber; Panagis Filippakopoulos; Clarence Yapp; Christopher Wells; Ka Hing Che; Andrew J. Bannister; Samuel Robson; Umesh Kumar; Nigel James Parr; Kevin Lee; Dave Lugo; Philip Jeffrey; Simon Taylor; Matteo Vecellio; C. Bountra; Paul E. Brennan; Alison O'Mahony; Sharlene Velichko; Susanne Müller; Duncan Hay

The histone acetyltransferases CBP/p300 are involved in recurrent leukemia-associated chromosomal translocations and are key regulators of cell growth. Therefore, efforts to generate inhibitors of CBP/p300 are of clinical value. We developed a specific and potent acetyl-lysine competitive protein-protein interaction inhibitor, I-CBP112, that targets the CBP/p300 bromodomains. Exposure of human and mouse leukemic cell lines to I-CBP112 resulted in substantially impaired colony formation and induced cellular differentiation without significant cytotoxicity. I-CBP112 significantly reduced the leukemia-initiating potential of MLL-AF9(+) acute myeloid leukemia cells in a dose-dependent manner in vitro and in vivo. Interestingly, I-CBP112 increased the cytotoxic activity of BET bromodomain inhibitor JQ1 as well as doxorubicin. Collectively, we report the development and preclinical evaluation of a novel, potent inhibitor targeting CBP/p300 bromodomains that impairs aberrant self-renewal of leukemic cells. The synergistic effects of I-CBP112 and current standard therapy (doxorubicin) as well as emerging treatment strategies (BET inhibition) provide new opportunities for combinatorial treatment of leukemia and potentially other cancers.


Chemistry & Biology | 2014

EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation.

William D. Bradley; Shilpi Arora; Jennifer Busby; Srividya Balasubramanian; Victor S. Gehling; Christopher G. Nasveschuk; Rishi G. Vaswani; Chih-Chi Yuan; Charlie Hatton; Feng Zhao; Kaylyn E. Williamson; Priyadarshini Iyer; Jacqui Mendez; Robert E. Campbell; Nico Cantone; Shivani Garapaty-Rao; James E. Audia; Andrew Simon Cook; Les A. Dakin; Brian K. Albrecht; Jean-Christophe Harmange; Danette L. Daniels; Richard T. Cummings; Barbara M. Bryant; Emmanuel Normant; Patrick Trojer

The histone lysine methyltransferase (MT) Enhancer of Zeste Homolog 2 (EZH2) is considered an oncogenic driver in a subset of germinal center B-cell-like diffuse large B cell lymphoma (GCB-DLBCL) and follicular lymphoma due to the presence of recurrent, monoallelic mutations in the EZH2 catalytic domain. These genomic data suggest that targeting the EZH2 MT activity is a valid therapeutic strategy for the treatment of lymphoma patients with EZH2 mutations. Here we report the identification of highly potent and selective EZH2 small molecule inhibitors, their validation by a cellular thermal shift assay, application across a large cell panel representing various non-Hodgkins lymphoma (NHL) subtypes, and their efficacy in EZH2mutant-containing GCB-DLBCL xenograft models. Surprisingly, our EZH2 inhibitors selectively affect the turnover of trimethylated, but not monomethylated histone H3 lysine 27 at pharmacologically relevant doses. Importantly, we find that these inhibitors are broadly efficacious also in NHL models with wild-type EZH2.


Angewandte Chemie | 2015

LP99: Discovery and Synthesis of the First Selective BRD7/9 Bromodomain Inhibitor

Peter G. K. Clark; Lucas C. C. Vieira; Cynthia Tallant; Oleg Fedorov; Dean C. Singleton; Catherine Rogers; Octovia P. Monteiro; James M. Bennett; Roberta Baronio; Susanne Müller; Danette L. Daniels; Jacqui Mendez; Stefan Knapp; Paul E. Brennan; Darren J. Dixon

The bromodomain-containing proteins BRD9 and BRD7 are part of the human SWI/SNF chromatin-remodeling complexes BAF and PBAF. To date, no selective inhibitor for BRD7/9 has been reported despite its potential value as a biological tool or as a lead for future therapeutics. The quinolone-fused lactam LP99 is now reported as the first potent and selective inhibitor of the BRD7 and BRD9 bromodomains. Development of LP99 from a fragment hit was expedited through balancing structure-based inhibitor design and biophysical characterization against tractable chemical synthesis: Complexity-building nitro-Mannich/lactamization cascade processes allowed for early structure–activity relationship studies whereas an enantioselective organocatalytic nitro-Mannich reaction enabled the synthesis of the lead scaffold in enantioenriched form and on scale. This epigenetic probe was shown to inhibit the association of BRD7 and BRD9 to acetylated histones in vitro and in cells. Moreover, LP99 was used to demonstrate that BRD7/9 plays a role in regulating pro-inflammatory cytokine secretion.


Nature Communications | 2015

Target engagement and drug residence time can be observed in living cells with BRET

Matthew B. Robers; Melanie Dart; Carolyn C. Woodroofe; Chad Zimprich; Thomas A. Kirkland; Thomas Machleidt; Kevin R. Kupcho; Sergiy Levin; James Robert Hartnett; Kristopher Zimmerman; Andrew L. Niles; Rachel Friedman Ohana; Danette L. Daniels; Michael R. Slater; Monika G. Wood; Mei Cong; Yi-Qiang Cheng; Keith V. Wood

The therapeutic action of drugs is predicated on their physical engagement with cellular targets. Here we describe a broadly applicable method using bioluminescence resonance energy transfer (BRET) to reveal the binding characteristics of a drug with selected targets within intact cells. Cell-permeable fluorescent tracers are used in a competitive binding format to quantify drug engagement with the target proteins fused to Nanoluc luciferase. The approach enabled us to profile isozyme-specific engagement and binding kinetics for a panel of histone deacetylase (HDAC) inhibitors. Our analysis was directed particularly to the clinically approved prodrug FK228 (Istodax/Romidepsin) because of its unique and largely unexplained mechanism of sustained intracellular action. Analysis of the binding kinetics by BRET revealed remarkably long intracellular residence times for FK228 at HDAC1, explaining the protracted intracellular behaviour of this prodrug. Our results demonstrate a novel application of BRET for assessing target engagement within the complex milieu of the intracellular environment.


Current Chemical Genomics | 2013

Development of a dehalogenase-based protein fusion tag capable of rapid, selective and covalent attachment to customizable ligands.

Lance P. Encell; Rachel Friedman Ohana; Kris Zimmerman; Paul Otto; Gediminas Vidugiris; Monika G. Wood; Georgyi V. Los; Mark McDougall; Chad Zimprich; Natasha Karassina; Randall D. Learish; James Robert Hartnett; Sarah Wheeler; Pete Stecha; Jami English; Kate Zhao; Jacqui Mendez; Hélène A Benink; Nancy Murphy; Danette L. Daniels; Michael R. Slater; Marjeta Urh; Aldis Darzins; Dieter Klaubert; Robert F. Bulleit; Keith V. Wood

Our fundamental understanding of proteins and their biological significance has been enhanced by genetic fusion tags, as they provide a convenient method for introducing unique properties to proteins so that they can be examinedin isolation. Commonly used tags satisfy many of the requirements for applications relating to the detection and isolation of proteins from complex samples. However, their utility at low concentration becomes compromised if the binding affinity for a detection or capture reagent is not adequate to produce a stable interaction. Here, we describe HaloTag® (HT7), a genetic fusion tag based on a modified haloalkane dehalogenase designed and engineered to overcome the limitation of affinity tags by forming a high affinity, covalent attachment to a binding ligand. HT7 and its ligand have additional desirable features. The tag is relatively small, monomeric, and structurally compatible with fusion partners, while the ligand is specific, chemically simple, and amenable to modular synthetic design. Taken together, the design features and molecular evolution of HT7 have resulted in a superior alternative to common tags for the overexpression, detection, and isolation of target proteins.


Journal of Medicinal Chemistry | 2015

9H-purine scaffold reveals induced-fit pocket plasticity of the BRD9 bromodomain

Sarah Picaud; Maria Strocchia; Stefania Terracciano; Gianluigi Lauro; Jacqui Mendez; Danette L. Daniels; Raffaele Riccio; Giuseppe Bifulco; Ines Bruno; Panagis Filippakopoulos

The 2-amine-9H-purine scaffold was identified as a weak bromodomain template and was developed via iterative structure based design into a potent nanomolar ligand for the bromodomain of human BRD9 with small residual micromolar affinity toward the bromodomain of BRD4. Binding of the lead compound 11 to the bromodomain of BRD9 results in an unprecedented rearrangement of residues forming the acetyllysine recognition site, affecting plasticity of the protein in an induced-fit pocket. The compound does not exhibit any cytotoxic effect in HEK293 cells and displaces the BRD9 bromodomain from chromatin in bioluminescence proximity assays without affecting the BRD4/histone complex. The 2-amine-9H-purine scaffold represents a novel template that can be further modified to yield highly potent and selective tool compounds to interrogate the biological role of BRD9 in diverse cellular systems.

Collaboration


Dive into the Danette L. Daniels's collaboration.

Researchain Logo
Decentralizing Knowledge