Network


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

Hotspot


Dive into the research topics where Mokdad Mezna is active.

Publication


Featured researches published by Mokdad Mezna.


Oncogene | 2010

Activating ROCK1 somatic mutations in human cancer

P A Lochhead; Grant Wickman; Mokdad Mezna; Michael F. Olson

Cancer cells acquire characteristics of deregulated growth, survival and increased metastatic potential. Genetic mutations that provide a selective advantage by promoting these characteristics have been termed ‘drivers,’ whereas mutations that do not contribute to disease initiation/progression are termed ‘passengers.’ The advent of high-throughput methodologies has facilitated large-scale screening of cancer genomes and the subsequent identification of novel somatic mutations. Although this approach has generated valuable results, the data remain incomplete until the functional consequences of these mutations are determined to differentiate potential drivers from passengers. ROCK1 is an essential effector kinase downstream of Rho GTPases, an important pathway involved in cell migration. The Cancer Genome Project identified three nonsynonymous mutations in the ROCK1 gene. We now show that these somatic ROCK1 mutations lead to elevated kinase activity and drive actin cytoskeleton rearrangements that promote increased motility and decreased adhesion, characteristics of cancer progression. Mapping of the kinase-interacting regions of the carboxy terminus combined with structural modeling provides an insight into how these mutations likely affect the regulation of ROCK1. Consistent with the frequency of ROCK1 mutations in human cancer, these results support the conclusion that there is selective pressure for the ROCK1 gene to acquire ‘driver’ mutations that result in kinase activation.


PLOS ONE | 2011

Co-Crystal Structures of Inhibitors with MRCKβ, a Key Regulator of Tumor Cell Invasion

Timo Heikkila; Edward Wheatley; Diane Crighton; Ewald Schröder; Alexandra Boakes; Sarah J. Kaye; Mokdad Mezna; Leon Pang; Mathew Rushbrooke; Andrew V. Turnbull; Michael F. Olson

MRCKα and MRCKβ (myotonic dystrophy kinase-related Cdc42-binding kinases) belong to a subfamily of Rho GTPase activated serine/threonine kinases within the AGC-family that regulate the actomyosin cytoskeleton. Reflecting their roles in myosin light chain (MLC) phosphorylation, MRCKα and MRCKβ influence cell shape and motility. We report further evidence for MRCKα and MRCKβ contributions to the invasion of cancer cells in 3-dimensional matrix invasion assays. In particular, our results indicate that the combined inhibition of MRCKα and MRCKβ together with inhibition of ROCK kinases results in significantly greater effects on reducing cancer cell invasion than blocking either MRCK or ROCK kinases alone. To probe the kinase ligand pocket, we screened 159 kinase inhibitors in an in vitro MRCKβ kinase assay and found 11 compounds that inhibited enzyme activity >80% at 3 µM. Further analysis of three hits, Y-27632, Fasudil and TPCA-1, revealed low micromolar IC50 values for MRCKα and MRCKβ. We also describe the crystal structure of MRCKβ in complex with inhibitors Fasudil and TPCA-1 bound to the active site of the kinase. These high-resolution structures reveal a highly conserved AGC kinase fold in a typical dimeric arrangement. The kinase domain is in an active conformation with a fully-ordered and correctly positioned αC helix and catalytic residues in a conformation competent for catalysis. Together, these results provide further validation for MRCK involvement in regulation of cancer cell invasion and present a valuable starting point for future structure-based drug discovery efforts.


Cell Communication and Signaling | 2014

A novel small-molecule MRCK inhibitor blocks cancer cell invasion.

Mathieu Unbekandt; Daniel R. Croft; Diane Crighton; Mokdad Mezna; Duncan McArthur; Patricia McConnell; Alexander W Schüttelkopf; Simone Belshaw; Andrew Pannifer; Mairi Sime; Justin Bower; Martin Drysdale; Michael F. Olson

BackgroundThe myotonic dystrophy kinase-related CDC42-binding kinases MRCKβ and MRCKβ regulate actin-myosin contractility and have been implicated in cancer metastasis. Along with the related ROCK1 and ROCK2 kinases, the MRCK proteins initiate signalling events that lead to contractile force generation which powers cancer cell motility and invasion. A potential strategy for cancer therapy is to reduce metastasis by blocking MRCK activity, either alone or in combination with ROCK inhibition. However, to date no potent small molecule inhibitors have been developed with selectivity towards MRCK.ResultsScreening a kinase-focused small molecule chemical library resulted in the identification of compounds with inhibitory activity towards MRCK. Medicinal chemistry combined with in vitro enzyme profiling led to the discovery of 4-chloro-1-(4-piperidyl)-N-[5-(2-pyridyl)-1H-pyrazol-4-yl]pyrazole-3-carboxamide (BDP00005290; abbreviated as BDP5290) as a potent MRCK inhibitor. X-ray crystallography of the MRCKβ kinase domain in complex with BDP5290 revealed how this ligand interacts with the nucleotide binding pocket. BDP5290 demonstrated marked selectivity for MRCKβ over ROCK1 or ROCK2 for inhibition of myosin II light chain (MLC) phosphorylation in cells. While BDP5290 was able to block MLC phosphorylation at both cytoplasmic actin stress fibres and peripheral cortical actin bundles, the ROCK selective inhibitor Y27632 primarily reduced MLC phosphorylation on stress fibres. BDP5290 was also more effective at reducing MDA-MB-231 breast cancer cell invasion through Matrigel than Y27632. Finally, the ability of human SCC12 squamous cell carcinoma cells to invade a three-dimensional collagen matrix was strongly inhibited by 2 μM BDP5290 but not the identical concentration of Y27632, despite equivalent inhibition of MLC phosphorylation.ConclusionsBDP5290 is a potent MRCK inhibitor with activity in cells, resulting in reduced MLC phosphorylation, cell motility and tumour cell invasion. The discovery of this compound will enable further investigations into the biological activities of MRCK proteins and their contributions to cancer progression.


Journal of Biomolecular Screening | 2012

Development of a High-Throughput Screening Method for LIM Kinase 1 Using a Luciferase-Based Assay of ATP Consumption

Mokdad Mezna; Ai Ching Wong; Margaret Ainger; Rebecca W. Scott; Tim Hammonds; Michael F. Olson

Kinases are attractive drug targets because of the central roles they play in signal transduction pathways and human diseases. Their well-formed adenosine triphosphate (ATP)–binding pockets make ideal targets for small-molecule inhibitors. For drug discovery purposes, many peptide-based kinase assays have been developed that measure substrate phosphorylation using fluorescence-based readouts. However, for some kinases these assays may not be appropriate. In the case of the LIM kinases (LIMK), an inability to phosphorylate peptide substrates resulted in previous high-throughput screens (HTS) using radioactive labeling of recombinant cofilin protein as the readout. We describe the development of an HTS-compatible assay that measures relative ATP levels using luciferase-generated luminescence as a function of LIMK activity. The assay was inexpensive to perform, and proof-of-principle screening of kinase inhibitors demonstrated that compound potency against LIMK could be determined; ultimately, the assay was used for successful prosecution of automated HTS. Following HTS, the secondary assay format was changed to obtain more accurate measures of potency and mechanism of action using more complex (and expensive) assays. The luciferase assay nonetheless provides an inexpensive and reliable primary assay for HTS that allowed for the identification of LIMK inhibitors to initiate discovery programs for the eventual treatment of human diseases.


Protein Expression and Purification | 2017

A fully automated procedure for the parallel, multidimensional purification and nucleotide loading of the human GTPases KRas, Rac1 and RalB

Christopher H. Gray; Jennifer Konczal; Mokdad Mezna; Shehab Ismail; Justin Bower; Martin Drysdale

Small GTPases regulate many key cellular processes and their role in human disease validates many proteins in this class as desirable targets for therapeutic intervention. Reliable recombinant production of GTPases, often in the active GTP loaded state, is a prerequisite for the prosecution of drug discovery efforts. The preparation of these active forms can be complex and often constricts the supply to the reagent intensive techniques used in structure base drug discovery. We have established a fully automated, multidimensional protein purification strategy for the parallel production of the catalytic G-domains of KRas, Rac1 and RalB GTPases in the active form. This method incorporates a four step chromatography purification with TEV protease-mediated affinity tag cleavage and a conditioning step that achieves the activation of the GTPase by exchanging GDP for the non-hydrolyzable GTP analogue GMPPnP. We also demonstrate that an automated method is efficient at loading of KRas with mantGDP for application in a SOS1 catalysed fluorescent nucleotide exchange assay. In comparison to more conventional manual workflows the automated method offers marked advantages in method run time and operator workload. This reduces the bottleneck in protein production while generating products that are highly purified and effectively loaded with nucleotide analogues.


Cancer Discovery | 2018

Colorectal Tumors Require NUAK1 for Protection from Oxidative Stress

Jennifer Port; Meera Raja; Fatih Ceteci; Tiziana Monteverde; Björn Kruspig; Ann Hedley; Gabriela Kalna; Sergio Lilla; Lisa J. Neilson; Martina Brucoli; Katarina Gyuraszova; Jacqueline Tait-Mulder; Mokdad Mezna; Silvija Svambaryte; Amy Bryson; David Sumpton; Allan McVie; Colin Nixon; Martin Drysdale; Hiroyasu Esumi; Graeme I. Murray; Owen J. Sansom; Sara Zanivan; Daniel J. Murphy

Exploiting oxidative stress has recently emerged as a plausible strategy for treatment of human cancer, and antioxidant defenses are implicated in resistance to chemotherapy and radiotherapy. Targeted suppression of antioxidant defenses could thus broadly improve therapeutic outcomes. Here, we identify the AMPK-related kinase NUAK1 as a key component of the antioxidant stress response pathway and reveal a specific requirement for this role of NUAK1 in colorectal cancer. We show that NUAK1 is activated by oxidative stress and that this activation is required to facilitate nuclear import of the antioxidant master regulator NRF2: Activation of NUAK1 coordinates PP1β inhibition with AKT activation in order to suppress GSK3β-dependent inhibition of NRF2 nuclear import. Deletion of NUAK1 suppresses formation of colorectal tumors, whereas acute depletion of NUAK1 induces regression of preexisting autochthonous tumors. Importantly, elevated expression of NUAK1 in human colorectal cancer is associated with more aggressive disease and reduced overall survival.Significance: This work identifies NUAK1 as a key facilitator of the adaptive antioxidant response that is associated with aggressive disease and worse outcome in human colorectal cancer. Our data suggest that transient NUAK1 inhibition may provide a safe and effective means for treatment of human colorectal cancer via disruption of intrinsic antioxidant defenses. Cancer Discov; 8(5); 632-47. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 517.


Cancer Research | 2018

Discovery of Potent and Selective MRCK Inhibitors with Therapeutic Effect on Skin Cancer.

Mathieu Unbekandt; Simone Belshaw; Justin Bower; Maeve Clarke; Jacqueline Cordes; Diane Crighton; Daniel R. Croft; Martin Drysdale; Mathew J. Garnett; Kathryn Gill; Christopher A. Gray; David A. Greenhalgh; James Hall; Jennifer Konczal; Sergio Lilla; Duncan McArthur; Patricia McConnell; Laura McDonald; Lynn McGarry; Heather McKinnon; Carol McMenemy; Mokdad Mezna; Nicholas A. Morrice; June Munro; Gregory Naylor; Nicola Rath; Alexander W Schüttelkopf; Mairi Sime; Michael F. Olson

The myotonic dystrophy-related Cdc42-binding kinases MRCKα and MRCKβ contribute to the regulation of actin-myosin cytoskeleton organization and dynamics, acting in concert with the Rho-associated coiled-coil kinases ROCK1 and ROCK2. The absence of highly potent and selective MRCK inhibitors has resulted in relatively little knowledge of the potential roles of these kinases in cancer. Here, we report the discovery of the azaindole compounds BDP8900 and BDP9066 as potent and selective MRCK inhibitors that reduce substrate phosphorylation, leading to morphologic changes in cancer cells along with inhibition of their motility and invasive character. In over 750 human cancer cell lines tested, BDP8900 and BDP9066 displayed consistent antiproliferative effects with greatest activity in hematologic cancer cells. Mass spectrometry identified MRCKα S1003 as an autophosphorylation site, enabling development of a phosphorylation-sensitive antibody tool to report on MRCKα status in tumor specimens. In a two-stage chemical carcinogenesis model of murine squamous cell carcinoma, topical treatments reduced MRCKα S1003 autophosphorylation and skin papilloma outgrowth. In parallel work, we validated a phospho-selective antibody with the capability to monitor drug pharmacodynamics. Taken together, our findings establish an important oncogenic role for MRCK in cancer, and they offer an initial preclinical proof of concept for MRCK inhibition as a valid therapeutic strategy.Significance: The development of selective small-molecule inhibitors of the Cdc42-binding MRCK kinases reveals their essential roles in cancer cell viability, migration, and invasive character. Cancer Res; 78(8); 2096-114. ©2018 AACR.


Cancer Research | 2017

Abstract LB-228: Identifying small molecule inhibitors of Fascin 1 using fragment-based drug discovery

Daniel R. Croft; Stuart Francis; Justin Bower; Andrea Gohlke; Gillian Goodwin; Christopher A. Gray; Jennifer Konczal; Sophie Macconnachie; Patricia McConnell; Laura McDonald; Heather McKinnon; Mokdad Mezna; Charles Parry; Nikki R. Paul; Angelo Pugliese; Alexander Schuettelkopf; Laura M. Machesky; Martin J. Drysdale

The actin-bundling protein fascin 1 is markedly overexpressed in a range of invasive tumors and is believed to play a critical role in cancer cell metastasis. Targeting fascin is however very challenging owing to its mechanism of protein-protein interaction and a lack of knowledge regarding the crucial actin-binding sites. By combining a fragment-based approach, biophysical assay screening and X-ray crystallography, we have been able to identify and optimize novel fascin 1 inhibitors that show nanomolar affinity in biochemical binding and bundling assays. We now show the development of functional cell assays in which the lead fascin inhibitor compounds have been tested. For the first time we disclose the structures of the most advanced compounds and show their effects in 2D and 3D cell culture. Fascin 1 binds and cross-links filamentous actin (F-actin) into parallel bundles that are used in the formation of dynamic cellular protrusions (such as lamellipodia and filopodia) used during cell migration, and in the formation of invadopodia used by tumor cell lines to degrade the tumor extracellular matrix (ECM). Whereas fascin 1 expression is low or absent in normal epithelia, its expression is dramatically increased in a variety of invasive tumor types including colon, lung, ovarian and pancreas, with increased expression being an independent prognostic indicator of poor clinical outcome in the most aggressive and metastatic tumor types. Knockdown of fascin expression has been shown to reduce tumor cell invasion both in vitro and in vivo, thereby highlighting fascin as an important drug discovery target. Small molecule inhibitors of fascin 1 were identified by screening our fragment library (~1000 compounds) using surface plasmon resonance (SPR). X-ray co-crystallography with these hit compounds showed four distinct ligand binding sites within the fascin protein. Optimized site 2 binders induce a substantial conformational change, with the deeply enclosed pocket between fascin domains 1 and 2 opening a channel that accesses the surface of the protein. A virtual screen identified the compound BDP-00010834, which binds in this pocket with an SPR Kd of 29.7µM and also inhibits (IC50=50µM) the functional activity of fascin measured in an F-actin bundling assay. Structure-based optimization in combination with X-ray co-crystallography has enabled the generation of compounds with more than 300-fold increase in both binding affinity and functional activity over this screening hit. Our current best-in-series compounds include BDP-00013544 (Kd=29nM, IC50=185nM). These lead compounds have now been tested in a number of cell based invasion assays including both 2D and 3D cultures. Citation Format: Daniel Croft, Stuart Francis, Justin Bower, Andrea Gohlke, Gillian Goodwin, Christopher Gray, Jennifer Konczal, Sophie Macconnachie, Patricia McConnell, Laura McDonald, Heather McKinnon, Mokdad Mezna, Charles Parry, Nikki Paul, Angelo Pugliese, Alexander Schuettelkopf, Laura Machesky, Martin Drysdale. Identifying small molecule inhibitors of Fascin 1 using fragment-based drug discovery [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-228. doi:10.1158/1538-7445.AM2017-LB-228


Cancer Research | 2018

Abstract 1933: A novel small molecule inhibitor of MRCK shows utility in blocking radiation induced invasion of glioblastoma cellsin vitroandin vivo

Heather McKinnon; Joanna Birch; Laura McDonald; Mairi Sime; Daniel R. Croft; Diane Crighton; Martin J. Drysdale; Lesley Gilmore; Christopher A. Gray; Jennifer Konczal; Duncan McArthur; Patricia McConnell; Mokdad Mezna; Alexander Schuettelkopf; Karen Strathdee; Justin Bower; Michael F. Olson; Anthony J. Chalmers


Cancer Research | 2018

A novel small molecule inhibitor of MRCK prevents radiation-driven invasion in glioblastoma

Joanna Birch; Karen Strathdee; Lesley Gilmour; Antoine Vallatos; Laura McDonald; Ariadni Kouzeli; Richa Vasan; Abdulrahman Qaisi; Daniel R. Croft; Diane Crighton; Kathryn Gill; Christopher H. Gray; Jennifer Konczal; Mokdad Mezna; Duncan McArthur; Alexander W Schüttelkopf; Patricia McConnell; Mairi Sime; William M. Holmes; Justin Bower; Heather McKinnon; Martin J. Drysdale; Michael F. Olson; Anthony J. Chalmers

Collaboration


Dive into the Mokdad Mezna's collaboration.

Top Co-Authors

Avatar

Michael F. Olson

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin J. Drysdale

Institute of Cancer Research

View shared research outputs
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