Network


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

Hotspot


Dive into the research topics where Jacqui Mendez is active.

Publication


Featured researches published by Jacqui Mendez.


ACS Chemical Biology | 2008

HaloTag: a novel protein labeling technology for cell imaging and protein analysis.

Georgyi V. Los; Lance P. Encell; Mark McDougall; Danette Hartzell; Natasha Karassina; Chad Zimprich; Monika G. Wood; Randy Learish; Rachel Friedman Ohana; Marjeta Urh; Dan Simpson; Jacqui Mendez; Kris Zimmerman; Paul Otto; Gediminas Vidugiris; Ji Zhu; Aldis Darzins; Dieter Klaubert; Robert F. Bulleit; Keith V. Wood

We have designed a modular protein tagging system that allows different functionalities to be linked onto a single genetic fusion, either in solution, in living cells, or in chemically fixed cells. The protein tag (HaloTag) is a modified haloalkane dehalogenase designed to covalently bind to synthetic ligands (HaloTag ligands). The synthetic ligands comprise a chloroalkane linker attached to a variety of useful molecules, such as fluorescent dyes, affinity handles, or solid surfaces. Covalent bond formation between the protein tag and the chloroalkane linker is highly specific, occurs rapidly under physiological conditions, and is essentially irreversible. We demonstrate the utility of this system for cellular imaging and protein immobilization by analyzing multiple molecular processes associated with NF-kappaB-mediated cellular physiology, including imaging of subcellular protein translocation and capture of protein--protein and protein--DNA complexes.


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.


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.


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.


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.


Nature Chemical Biology | 2016

Potent and selective bivalent inhibitors of BET bromodomains

Michael J. Waring; Huawei Chen; Alfred A. Rabow; Graeme Walker; Romel Bobby; Scott Boiko; Rob H. Bradbury; Rowena Callis; Edwin Clark; Ian L. Dale; Danette L. Daniels; Austin Dulak; Liz Flavell; Geoff Holdgate; Thomas A. Jowitt; Alexey Kikhney; Mark S. McAlister; Jacqui Mendez; Derek Ogg; Joe Patel; Philip Petteruti; Graeme R. Robb; Matthew B. Robers; Sakina Saif; Natalie Stratton; Dmitri I. Svergun; Wenxian Wang; David Whittaker; David Wilson; Yi Yao

Proteins of the bromodomain and extraterminal (BET) family, in particular bromodomain-containing protein 4 (BRD4), are of great interest as biological targets. BET proteins contain two separate bromodomains, and existing inhibitors bind to them monovalently. Here we describe the discovery and characterization of probe compound biBET, capable of engaging both bromodomains simultaneously in a bivalent, in cis binding mode. The evidence provided here was obtained in a variety of biophysical and cellular experiments. The bivalent binding results in very high cellular potency for BRD4 binding and pharmacological responses such as disruption of BRD4-mediator complex subunit 1 foci with an EC50 of 100 pM. These compounds will be of considerable utility as BET/BRD4 chemical probes. This work illustrates a novel concept in ligand design-simultaneous targeting of two separate domains with a drug-like small molecule-providing precedent for a potentially more effective paradigm for developing ligands for other multi-domain proteins.


Journal of Proteome Research | 2012

Examining the complexity of human RNA polymerase complexes using HaloTag technology coupled to label free quantitative proteomics

Danette L. Daniels; Jacqui Mendez; Amber L. Mosley; Sreenivasa R. Ramisetty; Nancy Murphy; Hélène A Benink; Keith V. Wood; Marjeta Urh; Michael P. Washburn

Efficient determination of protein interactions and cellular localization remains a challenge in higher order eukaryotes and creates a need for robust technologies for functional proteomics studies. To address this, the HaloTag technology was developed for highly efficient and rapid isolation of intracellular complexes and correlative in vivo cellular imaging. Here we demonstrate the strength of this technology by simultaneous capture of human eukaryotic RNA polymerases (RNAP) I, II, and III using a shared subunit, POLR2H, fused to the HaloTag. Affinity purifications showed successful isolation, as determined using quantitative proteomics, of all RNAP core subunits, even at expression levels near endogenous. Transient known RNAP II interacting partners were identified as well as three previously uncharacterized interactors. These interactions were validated and further functionally characterized using cellular imaging. The multiple capabilities of the HaloTag technology demonstrate the ability to efficiently isolate highly challenging multiprotein complexes, discover new interactions, and characterize cellular localization.


BMC Genomics | 2009

A functional analysis of the CREB signaling pathway using HaloCHIP-chip and high throughput reporter assays

Danette Hartzell; Nathan D. Trinklein; Jacqui Mendez; Nancy Murphy; Shelley Force Aldred; Keith V. Wood; Marjeta Urh

BackgroundRegulation of gene expression is essential for normal development and cellular growth. Transcriptional events are tightly controlled both spatially and temporally by specific DNA-protein interactions. In this study we finely map the genome-wide targets of the CREB protein across all known and predicted human promoters, and characterize the functional consequences of a subset of these binding events using high-throughput reporter assays. To measure CREB binding, we used HaloCHIP, an antibody-free alternative to the ChIP method that utilizes the HaloTag fusion protein, and also high-throughput promoter-luciferase reporter assays, which provide rapid and quantitative screening of promoters for transcriptional activation or repression in living cells.ResultsIn analysis of CREB genome-wide binding events using a comprehensive DNA microarray of human promoters, we observe for the first time that CREB has a strong preference for binding at bidirectional promoters and unlike unidirectional promoters, these binding events often occur downstream of transcription start sites. Comparison between HaloCHIP-chip and ChIP-chip data reveal this to be true for both methodologies, indicating it is not a bias of the technology chosen. Transcriptional data obtained from promoter-luciferase reporter arrays also show an unprecedented, high level of activation of CREB-bound promoters in the presence of the co-activator protein TORC1.ConclusionThese data suggest for the first time that TORC1 provides directional information when CREB is bound at bidirectional promoters and possible pausing of the CREB protein after initial transcriptional activation. Also, this combined approach demonstrates the ability to more broadly characterize CREB protein-DNA interactions wherein not only DNA binding sites are discovered, but also the potential of the promoter sequence to respond to CREB is evaluated.

Collaboration


Dive into the Jacqui Mendez's collaboration.

Researchain Logo
Decentralizing Knowledge