Jonathan A. G. Cox
University of Birmingham
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jonathan A. G. Cox.
PLOS ONE | 2012
Katherine A. Abrahams; Jonathan A. G. Cox; Vickey L. Spivey; Nicholas J. Loman; Mark J. Pallen; Chrystala Constantinidou; Raquel Fernandez; Carlos Alemparte; Modesto J. Remuiñán; David Barros; Lluis Ballell; Gurdyal S. Besra
Mycobacterium tuberculosis is a major human pathogen and the causative agent for the pulmonary disease, tuberculosis (TB). Current treatment programs to combat TB are under threat due to the emergence of multi-drug and extensively-drug resistant TB. Through the use of high throughput whole cell screening of an extensive compound library a number of imidazo[1,2-a]pyridine (IP) compounds were obtained as potent lead molecules active against M. tuberculosis and Mycobacterium bovis BCG. The IP inhibitors (1–4) demonstrated minimum inhibitory concentrations (MICs) in the range of 0.03 to 5 µM against a panel of M. tuberculosis strains. M. bovis BCG spontaneous resistant mutants were generated against IP 1, 3, and 4 at 5× MIC and subsequent whole genome sequencing identified a single nucleotide polymorphism 937ACC>937GCC (T313A) in qcrB, which encodes the b subunit of the electron transport ubiquinol cytochrome C reductase. This mutation also conferred cross-resistance against IP 1, 3 and 4 demonstrating a common target. Gene dosage experiments confirmed M. bovis BCG QcrB as the target where over-expression in M. bovis BCG led to an increase in MIC from 0.5 to >8 µM for IP 3. An acute murine model of TB infection established bacteriostatic activity of the IP series, which await further detailed characterization.
Annual Review of Microbiology | 2015
Monika Jankute; Jonathan A. G. Cox; James Harrison; Gurdyal S. Besra
Mycobacterium tuberculosis remains one of the most successful bacterial pathogens, claiming over 1.3 million lives worldwide in 2013. The emergence of multidrug-resistant and extensively drug-resistant isolates has prompted the need for new drugs and drug targets. M. tuberculosis possesses an unusual cell wall dominated by lipids and carbohydrates that provides a permeability barrier against hydrophilic drugs and is crucial for its survival and virulence. This large macromolecular structure, termed the mycolyl-arabinogalactan-peptidoglycan complex, and the phosphatidyl-myo-inositol-based lipoglycans are key features of the mycobacterial cell wall. Assembly of these cell wall components is an attractive target for the development of chemotherapeutics against tuberculosis. Herein, we focus on recent biochemical and molecular insights into these complex molecules of M. tuberculosis cell wall.
Nature microbiology | 2016
Jonathan A. G. Cox; Katherine A. Abrahams; Carlos Alemparte; Sonja Ghidelli-Disse; Joaquín Rullas; Iñigo Angulo-Barturen; Albel Singh; Sudagar S. Gurcha; Vijayashankar Nataraj; Stephen Bethell; Modesto J. Remuiñán; Lourdes Encinas; Peter J. Jervis; Nicholas Cammack; Apoorva Bhatt; Ulrich Kruse; Marcus Bantscheff; Klaus Fütterer; David Barros; Lluis Ballell; Gerard Drewes; Gurdyal S. Besra
Phenotypic screens for bactericidal compounds against drug-resistant tuberculosis are beginning to yield novel inhibitors. However, reliable target identification remains challenging. Here, we show that tetrahydropyrazo[1,5-a]pyrimidine-3-carboxamide (THPP) selectively pulls down EchA6 in a stereospecific manner, instead of the previously assigned target Mycobacterium tuberculosis MmpL3. While homologous to mammalian enoyl-coenzyme A (CoA) hydratases, EchA6 is non-catalytic yet essential and binds long-chain acyl-CoAs. THPP inhibitors compete with CoA-binding, suppress mycolic acid synthesis, and are bactericidal in a mouse model of chronic tuberculosis infection. A point mutation, W133A, abrogated THPP-binding and increased both the in vitro minimum inhibitory concentration and the in vivo effective dose 99 in mice. Surprisingly, EchA6 interacts with selected enzymes of fatty acid synthase II (FAS-II) in bacterial two-hybrid assays, suggesting essentiality may be linked to feeding long-chain fatty acids to FAS-II. Finally, our data show that spontaneous resistance-conferring mutations can potentially obscure the actual target or alternative targets of small molecule inhibitors.
PLOS ONE | 2015
Grace Mugumbate; Katherine A. Abrahams; Jonathan A. G. Cox; George Papadatos; Gerard J. P. van Westen; Joël Lelièvre; Szymon T. Calus; Nicholas J. Loman; Lluis Ballell; David Barros; John P. Overington; Gurdyal S. Besra
The lack of success in target-based screening approaches to the discovery of antibacterial agents has led to reemergence of phenotypic screening as a successful approach of identifying bioactive, antibacterial compounds. A challenge though with this route is then to identify the molecular target(s) and mechanism of action of the hits. This target identification, or deorphanization step, is often essential in further optimization and validation studies. Direct experimental identification of the molecular target of a screening hit is often complex, precisely because the properties and specificity of the hit are not yet optimized against that target, and so many false positives are often obtained. An alternative is to use computational, predictive, approaches to hypothesize a mechanism of action, which can then be validated in a more directed and efficient manner. Specifically here we present experimental validation of an in silico prediction from a large-scale screen performed against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis. The two potent anti-tubercular compounds studied in this case, belonging to the tetrahydro-1,3,5-triazin-2-amine (THT) family, were predicted and confirmed to be an inhibitor of dihydrofolate reductase (DHFR), a known essential Mtb gene, and already clinically validated as a drug target. Given the large number of similar screening data sets shared amongst the community, this in vitro validation of these target predictions gives weight to computational approaches to establish the mechanism of action (MoA) of novel screening hit.
Nature Communications | 2016
Katherine A. Abrahams; Chun-wa Chung; Sonja Ghidelli-Disse; Joaquín Rullas; María José Rebollo-López; Sudagar S. Gurcha; Jonathan A. G. Cox; Alfonso Mendoza; Elena Jimenez-Navarro; María S. Martínez-Martínez; Margarete Neu; Anthony Shillings; Paul Homes; Argyrides Argyrou; Ruth Casanueva; Nicholas J. Loman; Patrick J. Moynihan; Joël Lelièvre; Carolyn Selenski; Matthew Axtman; Laurent Kremer; Marcus Bantscheff; Iñigo Angulo-Barturen; Mónica Cacho Izquierdo; Nicholas Cammack; Gerard Drewes; Lluis Ballell; David Barros; Gurdyal S. Besra; Robert H. Bates
Phenotypic screens for bactericidal compounds are starting to yield promising hits against tuberculosis. In this regard, whole-genome sequencing of spontaneous resistant mutants generated against an indazole sulfonamide (GSK3011724A) identifies several specific single-nucleotide polymorphisms in the essential Mycobacterium tuberculosis β-ketoacyl synthase (kas) A gene. Here, this genomic-based target assignment is confirmed by biochemical assays, chemical proteomics and structural resolution of a KasA-GSK3011724A complex by X-ray crystallography. Finally, M. tuberculosis GSK3011724A-resistant mutants increase the in vitro minimum inhibitory concentration and the in vivo 99% effective dose in mice, establishing in vitro and in vivo target engagement. Surprisingly, the lack of target engagement of the related β-ketoacyl synthases (FabH and KasB) suggests a different mode of inhibition when compared with other Kas inhibitors of fatty acid biosynthesis in bacteria. These results clearly identify KasA as the biological target of GSK3011724A and validate this enzyme for further drug discovery efforts against tuberculosis.
PLOS ONE | 2014
Sudagar S. Gurcha; Veeraraghavan Usha; Jonathan A. G. Cox; Klaus Fütterer; Katherine A. Abrahams; Apoorva Bhatt; Luke J. Alderwick; Robert C. Reynolds; Nicholas J. Loman; Vijayashankar Nataraj; Carlos Alemparte; David Barros; Adrian J. Lloyd; Lluis Ballell; Judith V. Hobrath; Gurdyal S. Besra
The human pathogen Mycobacterium tuberculosis is the causative agent of pulmonary tuberculosis (TB), a disease with high worldwide mortality rates. Current treatment programs are under significant threat from multi-drug and extensively-drug resistant strains of M. tuberculosis, and it is essential to identify new inhibitors and their targets. We generated spontaneous resistant mutants in Mycobacterium bovis BCG in the presence of 10× the minimum inhibitory concentration (MIC) of compound 1, a previously identified potent inhibitor of mycobacterial growth in culture. Whole genome sequencing of two resistant mutants revealed in one case a single nucleotide polymorphism in the gene aspS at 535GAC>535AAC (D179N), while in the second mutant a single nucleotide polymorphism was identified upstream of the aspS promoter region. We probed whole cell target engagement by overexpressing either M. bovis BCG aspS or Mycobacterium smegmatis aspS, which resulted in a ten-fold and greater than ten-fold increase, respectively, of the MIC against compound 1. To analyse the impact of inhibitor 1 on M. tuberculosis AspS (Mt-AspS) activity we over-expressed, purified and characterised the kinetics of this enzyme using a robust tRNA-independent assay adapted to a high-throughput screening format. Finally, to aid hit-to-lead optimization, the crystal structure of apo M. smegmatis AspS was determined to a resolution of 2.4 Å.
Scientific Reports | 2016
Jonathan A. G. Cox; Grace Mugumbate; Laura Vela-Glez Del Peral; Monika Jankute; Katherine A. Abrahams; Peter J. Jervis; Stefan Jackenkroll; Arancha Perez; Carlos Alemparte; Jorge Esquivias; Joël Lelièvre; Fernando Ramon; David Barros; Lluis Ballell; Gurdyal S. Besra
High-throughput phenotypic screens have re-emerged as screening tools in antibiotic discovery. The advent of such technologies has rapidly accelerated the identification of ‘hit’ compounds. A pre-requisite to medicinal chemistry optimisation programmes required to improve the drug-like properties of a ‘hit’ molecule is identification of its mode of action. Herein, we have combined phenotypic screening with a biased target-specific screen. The inosine monophosphate dehydrogenase (IMPDH) protein GuaB2 has been identified as a drugable target in Mycobacterium tuberculosis, however previously identified compounds lack the desired characteristics necessary for further development into lead-like molecules. This study has identified 7 new chemical series from a high-throughput resistance-based phenotypic screen using Mycobacterium bovis BCG over-expressing GuaB2. Hit compounds were identified in a single shot high-throughput screen, validated by dose response and subjected to further biochemical analysis. The compounds were also assessed using molecular docking experiments, providing a platform for their further optimisation using medicinal chemistry. This work demonstrates the versatility and potential of GuaB2 as an anti-tubercular drug target.
Scientific Reports | 2017
Katherine A. Abrahams; Jonathan A. G. Cox; Klaus Fütterer; Joaquín Rullas; Fátima Ortega-Muro; Nicholas J. Loman; Patrick J. Moynihan; Esther Pérez-Herrán; Elena Jimenez; Jorge Esquivias; David Barros; Lluis Ballell; Carlos Alemparte; Gurdyal S. Besra
Drug discovery efforts against the pathogen Mycobacterium tuberculosis (Mtb) have been advanced through phenotypic screens of extensive compound libraries. Such a screen revealed sulfolane 1 and indoline-5-sulfonamides 2 and 3 as potent inhibitors of mycobacterial growth. Optimization in the sulfolane series led to compound 4, which has proven activity in an in vivo murine model of Mtb infection. Here we identify the target and mode of inhibition of these compounds based on whole genome sequencing of spontaneous resistant mutants, which identified mutations locating to the essential α- and β-subunits of tryptophan synthase. Over-expression studies confirmed tryptophan synthase as the biological target. Biochemical techniques probed the mechanism of inhibition, revealing the mutant enzyme complex incurs a fitness cost but does not prevent inhibitor binding. Mapping of the resistance conferring mutations onto a low-resolution crystal structure of Mtb tryptophan synthase showed they locate to the interface between the α- and β-subunits. The discovery of anti-tubercular agents inhibiting tryptophan synthase highlights the therapeutic potential of this enzyme and draws attention to the prospect of other amino acid biosynthetic pathways as future Mtb drug targets.
Scientific Reports | 2018
Valentin Trofimov; Sébastien Kicka; Sabrina Mucaria; Nabil Hanna; Fernando Ramon-Olayo; Laura Vela-Gonzalez Del Peral; Joël Lelièvre; Lluis Ballell; Leonardo Scapozza; Gurdyal S. Besra; Jonathan A. G. Cox; Thierry Soldati
Tuberculosis remains a serious threat to human health world-wide, and improved efficiency of medical treatment requires a better understanding of the pathogenesis and the discovery of new drugs. In the present study, we performed a whole-cell based screen in order to complete the characterization of 168 compounds from the GlaxoSmithKline TB-set. We have established and utilized novel previously unexplored host-model systems to characterize the GSK compounds, i.e. the amoeboid organisms D. discoideum and A. castellanii, as well as a microglial phagocytic cell line, BV2. We infected these host cells with Mycobacterium marinum to monitor and characterize the anti-infective activity of the compounds with quantitative fluorescence measurements and high-content microscopy. In summary, 88.1% of the compounds were confirmed as antibiotics against M. marinum, 11.3% and 4.8% displayed strong anti-infective activity in, respectively, the mammalian and protozoan infection models. Additionally, in the two systems, 13–14% of the compounds displayed pro-infective activity. Our studies underline the relevance of using evolutionarily distant pathogen and host models in order to reveal conserved mechanisms of virulence and defence, respectively, which are potential “universal” targets for intervention. Subsequent mechanism of action studies based on generation of over-expresser M. bovis BCG strains, generation of spontaneous resistant mutants and whole genome sequencing revealed four new molecular targets, including FbpA, MurC, MmpL3 and GlpK.
The International Journal of Mycobacteriology | 2016
Jonathan A. G. Cox
The complex and essential cell wall of Mycobacterium tuberculosis represents a plethora of new and old drug targets that collectively form an apparent mycobacterial “Achilles’ heel”. The mycolic acids are long-chain α-alkyl-β-hydroxy fatty acids (C70–90), which are unique to mycobacterial species, forming an integral component of the mycolyl–arabinogalactan–peptidoglycan complex. Their apparent uniqueness to the M. tuberculosis complex has rendered components of mycolic acid biosynthesis as powerful drug targets for specific tuberculosis (TB) chemotherapy. Here, I will discuss a contribution to TB drug discovery by deconvolution of the inhibitory mechanisms of a number of antitubercular compounds targeting mycolic acid biosynthesis. I will begin with the early days, elucidating the mode of action of ethionamide [1] and thiolactomycin [2], each targeting two separate components of the fatty acid synthase II (FAS-II) pathway. I will further discuss the recently discovered tetrahydropyrazo[1,5-a]pyrimidine-3-carboxamide compounds [3] which selectively target the essential, catalytically silent M. tuberculosis EchA6, providing a crucial lipid shunt between β-oxidation and FAS-II and supplying lipid precursors for essential mycolate biosynthesis. Finally, I will discuss the recent discovery of the mode of action of the indazole sulfonamides [4], inhibiting M. tuberculosis KasA by, a completely novel inhibitory mechanism.