Kakoli Mukherjee
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Kakoli Mukherjee.
Infection and Immunity | 2003
Sture Karlsson; Bruno Dupuy; Kakoli Mukherjee; Elisabeth Norin; Lars G. Burman; Thomas Åkerlund
ABSTRACT Growth temperature was found to control the expression of toxins A and B in Clostridium difficile VPI 10463, with a maximum at 37°C and low levels at 22 and 42°C in both peptone yeast (PY) and defined media. The up-regulation of toxin A and B mRNA and protein levels upon temperature upshift from 22 to 37°C followed the same kinetics, showing that temperature control occurred at the level of transcription. Experiments with Clostridium perfringens using gusA as a reporter gene demonstrated that both toxin gene promoters were temperature controlled and that their high activity at 37°C was dependent on the alternative sigma factor TcdD. Furthermore, tcdD was found to be autoinduced at 37°C. Glucose down-regulated all these responses in the C. perfringens constructs, similar to its impact on toxin production in C. difficile PY broth cultures. C. difficile proteins induced at 37°C and thus coregulated with the toxins by temperature were demonstrated by two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis and identified as enzymes involved in butyric acid production and as electron carriers in oxidation-reduction reactions. The regulation of toxin production in C. difficile by temperature is a novel finding apparently reflecting an adaptation of the expression of its virulence to mammalian hosts.
Microbiology | 2002
Kakoli Mukherjee; Sture Karlsson; Lars G. Burman; Thomas Åkerlund
The mechanism by which toxins A and B are released by Clostridium difficile is unknown and information about the other extracellular proteins of this bacterium is limited. The authors identified exported proteins from C. difficile strain VPI 10463 during conditions promoting high toxin production. Toxins A and B were released in a 1:1 ratio and the proportion of toxin in the extracellular fraction reached 50% during the stationary phase as compared to a proportion of <1% for typical cytoplasmic proteins, showing that toxin export was not due to bacterial lysis. A 47 kDa protein, released with similar kinetics to the toxins, was processed and showed weak similarity to the channel-forming protein TolC. Another protein released during high toxin production was unprocessed and showed similarity to XkdK encoded by the prophage PBSX in Bacillus subtilis, a protein supposedly exported via phage-specific holins. The two most abundant extracellular C. difficile proteins, found during both high and low toxin production, were processed and identified as shed S-layer proteins. As shown by N-terminal sequencing and PCR-based methods, there was a considerable sequence variation of the S-layer gene slpA in different serogroup reference strains. To conclude, C. difficile uses the classical Sec-dependent and probably also holin-like pathways to secrete a comparatively small repertoire of proteins.
Antimicrobial Agents and Chemotherapy | 2006
Jeanette W. P. Teo; Pamela Thayalan; David Beer; Amelia S. L. Yap; Mahesh Nanjundappa; Xinyi Ngew; Jeyaraj Duraiswamy; Sarah Liung; Véronique Dartois; Mark Schreiber; Samiul Hasan; Michael H. Cynamon; Neil S. Ryder; Xia Yang; Beat Weidmann; Kathryn Rene Bracken; Thomas Dick; Kakoli Mukherjee
ABSTRACT Peptide deformylase (PDF) catalyzes the hydrolytic removal of the N-terminal formyl group from nascent proteins. This is an essential step in bacterial protein synthesis, making PDF an attractive target for antibacterial drug development. Essentiality of the def gene, encoding PDF from Mycobacterium tuberculosis, was demonstrated through genetic knockout experiments with Mycobacterium bovis BCG. PDF from M. tuberculosis strain H37Rv was cloned, expressed, and purified as an N-terminal histidine-tagged recombinant protein in Escherichia coli. A novel class of PDF inhibitors (PDF-I), the N-alkyl urea hydroxamic acids, were synthesized and evaluated for their activities against the M. tuberculosis PDF enzyme as well as their antimycobacterial effects. Several compounds from the new class had 50% inhibitory concentration (IC50) values of <100 nM. Some of the PDF-I displayed antibacterial activity against M. tuberculosis, including MDR strains with MIC90 values of <1 μM. Pharmacokinetic studies of potential leads showed that the compounds were orally bioavailable. Spontaneous resistance towards these inhibitors arose at a frequency of ≤5 × 10−7 in M. bovis BCG. DNA sequence analysis of several spontaneous PDF-I-resistant mutants revealed that half of the mutants had acquired point mutations in their formyl methyltransferase gene (fmt), which formylated Met-tRNA. The results from this study validate M. tuberculosis PDF as a drug target and suggest that this class of compounds have the potential to be developed as novel antimycobacterial agents.
Journal of Medicinal Chemistry | 2014
Shahul Hameed P; Vikas Patil; Suresh Solapure; Umender Sharma; Prashanti Madhavapeddi; Anandkumar Raichurkar; Murugan Chinnapattu; Praveena Manjrekar; Gajanan Shanbhag; Jayashree Puttur; Vikas Shinde; Sreenivasaiah Menasinakai; Suresh Rudrapatana; Vijayashree Achar; Disha Awasthy; Radha Nandishaiah; Vaishali Humnabadkar; Anirban Ghosh; Chandan Narayan; V. K. Ramya; Parvinder Kaur; Sreevalli Sharma; Jim Werngren; Sven Hoffner; C. N. Naveen Kumar; Jitendar Reddy; Mahesh Kumar Kn; Samit Ganguly; Ugarkar Bheemarao; Kakoli Mukherjee
DNA gyrase is a clinically validated target for developing drugs against Mycobacterium tuberculosis (Mtb). Despite the promise of fluoroquinolones (FQs) as anti-tuberculosis drugs, the prevalence of pre-existing resistance to FQs is likely to restrict their clinical value. We describe a novel class of N-linked aminopiperidinyl alkyl quinolones and naphthyridones that kills Mtb by inhibiting the DNA gyrase activity. The mechanism of inhibition of DNA gyrase was distinct from the fluoroquinolones, as shown by their ability to inhibit the growth of fluoroquinolone-resistant Mtb. Biochemical studies demonstrated this class to exert its action via single-strand cleavage rather than double-strand cleavage, as seen with fluoroquinolones. The compounds are highly bactericidal against extracellular as well as intracellular Mtb. Lead optimization resulted in the identification of potent compounds with improved oral bioavailability and reduced cardiac ion channel liability. Compounds from this series are efficacious in various murine models of tuberculosis.
Bioorganic & Medicinal Chemistry Letters | 2013
Manoranjan Panda; Kakoli Mukherjee; Nilanjana Roy Choudhury; Subramanyam J. Tantry; Chaitanya Kumar Kedari; Sreevalli Sharma; V. K. Ramya; Supreeth Guptha; Vasan K. Sambandamurthy
Imidazo[1,2-a]pyridine-8-carboxamides as a novel antimycobacterial lead were generated by whole cell screening of a focused library against Mycobacterium tuberculosis. Herein, we describe the synthesis and structure activity relationship evaluation of this class of inhibitors and the optimization of physicochemical properties. These are selective inhibitors of Mycobacterium tuberculosis with no activity on either gram positive or gram negative pathogens.
Nature Communications | 2015
Shahul Hameed P; Suresh Solapure; Vikas Patil; Philipp P. Henrich; Pamela Magistrado; Kannan Murugan; Pavithra Viswanath; Jayashree Puttur; Abhishek Srivastava; Eknath Bellale; Gajanan Shanbag; Disha Awasthy; Sudhir Landge; Sapna Morayya; Krishna Koushik; Ramanatha Saralaya; Anandkumar Raichurkar; Nikhil Rautela; Nilanjana Roy Choudhury; Anisha Ambady; Radha Nandishaiah; Jitendar Reddy; K. R. Prabhakar; Sreenivasaiah Menasinakai; Suresh Rudrapatna; Monalisa Chatterji; María Belén Jiménez-Díaz; María Santos Martínez; Laura Sanz; Olivia Coburn-Flynn
The widespread emergence of Plasmodium falciparum (Pf) strains resistant to frontline agents has fuelled the search for fast-acting agents with novel mechanism of action. Here, we report the discovery and optimization of novel antimalarial compounds, the triaminopyrimidines (TAPs), which emerged from a phenotypic screen against the blood stages of Pf. The clinical candidate (compound 12) is efficacious in a mouse model of Pf malaria with an ED99 <30 mg kg−1 and displays good in vivo safety margins in guinea pigs and rats. With a predicted half-life of 36 h in humans, a single dose of 260 mg might be sufficient to maintain therapeutic blood concentration for 4–5 days. Whole-genome sequencing of resistant mutants implicates the vacuolar ATP synthase as a genetic determinant of resistance to TAPs. Our studies highlight the potential of TAPs for single-dose treatment of Pf malaria in combination with other agents in clinical development.
Antimicrobial Agents and Chemotherapy | 2014
Shahul Hameed P; Suresh Solapure; Kakoli Mukherjee; Vrinda Nandi; David Waterson; Radha Shandil; Meenakshi Balganesh; Vasan K. Sambandamurthy; Anand Kumar V. Raichurkar; Abhijeet Deshpande; Anirban Ghosh; Disha Awasthy; Gajanan Shanbhag; Gulebahar Sheikh; Helen McMiken; Jayashree Puttur; Jitendar Reddy; Jim Werngren; Jon Read; Mahesh Kumar; Manjunatha R; Murugan Chinnapattu; Prashanti Madhavapeddi; Praveena Manjrekar; Reetobrata Basu; Sheshagiri Gaonkar; Sreevalli Sharma; Sven Hoffner; Vaishali Humnabadkar; Venkita Subbulakshmi
ABSTRACT Moxifloxacin has shown excellent activity against drug-sensitive as well as drug-resistant tuberculosis (TB), thus confirming DNA gyrase as a clinically validated target for discovering novel anti-TB agents. We have identified novel inhibitors in the pyrrolamide class which kill Mycobacterium tuberculosis through inhibition of ATPase activity catalyzed by the GyrB domain of DNA gyrase. A homology model of the M. tuberculosis H37Rv GyrB domain was used for deciphering the structure-activity relationship and binding interactions of inhibitors with mycobacterial GyrB enzyme. Proposed binding interactions were later confirmed through cocrystal structure studies with the Mycobacterium smegmatis GyrB ATPase domain. The most potent compound in this series inhibited supercoiling activity of DNA gyrase with a 50% inhibitory concentration (IC50) of <5 nM, an MIC of 0.03 μg/ml against M. tuberculosis H37Rv, and an MIC90 of <0.25 μg/ml against 99 drug-resistant clinical isolates of M. tuberculosis. The frequency of isolating spontaneous resistant mutants was ∼10−6 to 10−8, and the point mutation mapped to the M. tuberculosis GyrB domain (Ser208 Ala), thus confirming its mode of action. The best compound tested for in vivo efficacy in the mouse model showed a 1.1-log reduction in lung CFU in the acute model and a 0.7-log reduction in the chronic model. This class of GyrB inhibitors could be developed as novel anti-TB agents.
Microbiology | 2010
Disha Awasthy; Anisha Ambady; Jyothi Bhat; Gulebahar Sheikh; Sudha Ravishankar; Venkita Subbulakshmi; Kakoli Mukherjee; Vasan K. Sambandamurthy; Umender Sharma
Pantothenate kinase, an essential enzyme in bacteria and eukaryotes, is involved in catalysing the first step of conversion of pantothenate to coenzyme A (CoA). Three isoforms (type I, II and III) of this enzyme have been reported from various organisms, which can be differentiated from each other on the basis of their biochemical and structural characteristics. Though most bacteria carry only one of the isoforms of pantothenate kinases, some of them possess two isoforms. The physiological relevance of the presence of two types of isozymes in a single organism is not clear. Mycobacterium tuberculosis, an intracellular pathogen, possesses two isoforms of pantothenate kinases (CoaA and CoaX) belonging to type I and III. In order to determine which pantothenate kinase is essential in mycobacteria, we performed gene inactivation of coaA and coaX of M. tuberculosis individually. It was found that coaA could only be inactivated in the presence of an extra copy of the gene, while coaX could be inactivated in the wild-type cells, proving that CoaA is the essential pantothenate kinase in M. tuberculosis. Additionally, the coaA gene of M. tuberculosis was able to complement a temperature-sensitive coaA mutant of Escherichia coli at a non-permissive temperature while coaX could not. The coaX deletion mutant showed no growth defects in vitro, in macrophages or in mice. Taken together, our data suggest that CoaX, which is essential in Bacillus anthracis and thus had been suggested to be a drug target in this organism, might not be a valid target in M. tuberculosis. We have established that the type I isoform, CoaA, is the essential pantothenate kinase in M. tuberculosis and thus can be explored as a drug target.
Journal of Medicinal Chemistry | 2014
Shahul Hameed P; Murugan Chinnapattu; Gajanan Shanbag; Praveena Manjrekar; Krishna Koushik; Anandkumar Raichurkar; Vikas Patil; Sandesh Jatheendranath; Suresh Rudrapatna; Shubhada Pramod Barde; Nikhil Rautela; Disha Awasthy; Sapna Morayya; Chandan Narayan; Stefan Kavanagh; Ramanatha Saralaya; Pavithra Viswanath; Kakoli Mukherjee; Balachandra Bandodkar; Abhishek Srivastava; Jitender Reddy; K. R. Prabhakar; Achyut Sinha; María Belén Jiménez-Díaz; María Santos Martínez; Iñigo Angulo-Barturen; Santiago Ferrer; Laura Sanz; Francisco Javier Gamo; Sandra Duffy
Whole-cell high-throughput screening of the AstraZeneca compound library against the asexual blood stage of Plasmodium falciparum (Pf) led to the identification of amino imidazoles, a robust starting point for initiating a hit-to-lead medicinal chemistry effort. Structure-activity relationship studies followed by pharmacokinetics optimization resulted in the identification of 23 as an attractive lead with good oral bioavailability. Compound 23 was found to be efficacious (ED90 of 28.6 mg·kg(-1)) in the humanized P. falciparum mouse model of malaria (Pf/SCID model). Representative compounds displayed a moderate to fast killing profile that is comparable to that of chloroquine. This series demonstrates no cross-resistance against a panel of Pf strains with mutations to known antimalarial drugs, thereby suggesting a novel mechanism of action for this chemical class.
Journal of Biological Chemistry | 2013
Christofer Björkelid; Terese Bergfors; Anand Kumar V. Raichurkar; Kakoli Mukherjee; Krishnan Malolanarasimhan; Balachandra Bandodkar; T. Alwyn Jones
Background: Pantothenate kinase (PanK), an essential enzyme for Mycobacterium tuberculosis, catalyzes the rate-limiting step in the CoA pathway. Results: Structures of M. tuberculosis PanK, complexed with new inhibitors, were determined, and their inhibitions were evaluated biochemically. Conclusion: Inhibitor binding overlaps with the substrate/product sites; also, an alternative mode of ATP binding is proposed. Significance: These are the first structures of a type I PanK complexed with inhibitors. Mycobacterium tuberculosis, the bacterial causative agent of tuberculosis, currently affects millions of people. The emergence of drug-resistant strains makes development of new antibiotics targeting the bacterium a global health priority. Pantothenate kinase, a key enzyme in the universal biosynthesis of the essential cofactor CoA, was targeted in this study to find new tuberculosis drugs. The biochemical characterizations of two new classes of compounds that inhibit pantothenate kinase from M. tuberculosis are described, along with crystal structures of their enzyme-inhibitor complexes. These represent the first crystal structures of this enzyme with engineered inhibitors. Both classes of compounds bind in the active site of the enzyme, overlapping with the binding sites of the natural substrate and product, pantothenate and phosphopantothenate, respectively. One class of compounds also interferes with binding of the cofactor ATP. The complexes were crystallized in two crystal forms, one of which is in a new space group for this enzyme and diffracts to the highest resolution reported for any pantothenate kinase structure. These two crystal forms allowed, for the first time, modeling of the cofactor-binding loop in both open and closed conformations. The structures also show a binding mode of ATP different from that previously reported for the M. tuberculosis enzyme but similar to that in the pantothenate kinases of other organisms.