Network


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

Hotspot


Dive into the research topics where Dibyendu Banerjee is active.

Publication


Featured researches published by Dibyendu Banerjee.


Journal of Chemical Information and Modeling | 2014

Pharmacophore-Based Screening and Identification of Novel Human Ligase I Inhibitors with Potential Anticancer Activity

Shagun Krishna; Deependra Kumar Singh; Sanjeev Meena; Dipak Datta; Mohammad Imran Siddiqi; Dibyendu Banerjee

Human DNA ligases are enzymes that are indispensable for DNA replication and repair processes. Among the three human ligases, ligase I is attributed to the ligation of thousands of Okazaki fragments that are formed during lagging strand synthesis during DNA replication. Blocking ligation therefore can lead to the accumulation of thousands of single strands and subsequently double strand breaks in the DNA, which is lethal for the cells. The reports of the high expression level of ligase I protein in several cancer cells (versus the low ligase expression level and the low rate of division of most normal cells in the adult body) support the belief that ligase I inhibitors can target cancer cells specifically with minimum side effects to normal cells. Recent publications showing exciting data for a ligase IV inhibitor exhibiting antitumor activity in mouse models also strengthens the argument for ligases as valid antitumor targets. Keeping this in view, we performed a pharmacophore-based screening for potential ligase inhibitors in the Maybridge small molecule library and procured some of the top-ranking compounds for enzyme-based and cell-based in vitro screening. We report here the identification of novel ligase I inhibitors with potential anticancer activity against a colon cancer cell line.


Medicinal Research Reviews | 2014

Human DNA Ligases: A Comprehensive New Look for Cancer Therapy

Deependra Kumar Singh; Shagun Krishna; Sharat Chandra; Mohammad Shameem; Amit Laxmikant Deshmukh; Dibyendu Banerjee

Living organisms belonging to all three domains of life, viz., eubacteria, archaeabacteria, and eukaryotes encode one or more DNA ligases. DNA ligases are indispensable in various DNA repair and replication processes and a deficiency or an inhibition of their activity can lead to accumulation of DNA damage and strand breaks. DNA damage, specially strand breaks at unsustainable levels can lead to replication block and/or cell death. DNA ligases as potential anticancer targets have been realized only recently. There is enough rationale to suggest that ligases have a tremendous potential for novel therapeutics including anticancer and antibacterial therapy, specially when the world is facing acute problems of drug resistance and chemotherapy failure, with an immediate need for new therapeutic targets. Here, we review the current state of the art in the development of human ligase inhibitors, their structures, molecular mechanisms, physiological effects, and their potential in future cancer therapy. Citing examples, we focus on strategies for improving the activity and specificity of existing and novel inhibitors by using structure‐based rational approaches. In the end, we describe potential new sites on the ligase I protein that can be targeted for the development of novel inhibitors. This is the first comprehensive review to compile all known human ligase inhibitors and to provide a rationale for the further development of ligase inhibitors for cancer therapy.


Chemico-Biological Interactions | 2015

Synthetic modified pyrrolo[1,4] benzodiazepine molecules demonstrate selective anticancer activity by targeting the human ligase 1 enzyme: An in silico and in vitro mechanistic study

Mohammad Shameem; Ravi Kumar; Shagun Krishna; Chandan Kumar; Mohammad Imran Siddiqi; Bijoy Kundu; Dibyendu Banerjee

Human DNA ligase1 (hLig1) is the major replicative enzyme in proliferating mammalian cells that join Okazaki fragments of the lagging strand during DNA replication. Interruptions in the process of ligation cause DNA damage to accumulate, resulting in cytotoxicity and cell death. In the present study we demonstrate that pyrrolo[1,4] benzodiazepine (PBD) derivatives exhibit anticancer properties by targeting the nick sealing activity of hLig1 as opposed to the DNA interaction activity known for such compounds. Our in silico and in vitro assays demonstrate the binding of these molecules with amino acid residues present in the DNA binding domain (DBD) of the hLig1 enzyme. Two of these hLig1 inhibitors S010-015 and S010-018 demonstrated selective cytotoxicity against DLD-1 (colon cancer) and HepG2 (hepatic cancer) cells in a dose dependant manner. The molecules also reduced cell viability and colony formation at concentrations of ⩽20μM in DLD-1 and HepG2 cells and induced apoptotic cell death. In yet another significant finding, the molecules reduced the migration of cancer cells in wound healing experiments, indicating their anti-metastatic property. In summary, we report the anticancer activity of PBD derivatives against DLD-1 and HepG2 cells and propose a new molecular target for their activity.


MedChemComm | 2016

Design, synthesis and anticancer activity of dihydropyrimidinone–semicarbazone hybrids as potential human DNA ligase 1 inhibitors

Koneni V. Sashidhara; L. Ravithej Singh; Mohammad Shameem; Sarika Shakya; Anoop Kumar; Tulsankar Sachin Laxman; Shagun Krishna; Mohammad Imran Siddiqi; Rabi Sankar Bhatta; Dibyendu Banerjee

A series of new dihydropyrimidinone–semicarbazone hybrids were successfully synthesised by integrating regioselective multicomponent reaction with the pharmacophore hybridization approach. All the synthesised compounds were evaluated for their hLig1 inhibition potency and most of them were found to be good to moderately active. Out of the tested derivatives, compound 6f showed selective anti-proliferative activity against HepG2 cells in a dose-dependent manner with an IC50 value of 10.07 ± 1.2. It also reduced cell survival at ≤20 μM concentration. Further, analysis of treated HepG2 cell lysates by western blot assay showed increased γ-H2AX levels and upregulation of p53, leading to apoptosis. In silico docking results explain the binding modes of compound 6f to the DNA-binding domain of hLig1 enzyme thereby preventing its nick sealing activity. In addition, the favourable pharmacokinetic properties suggest that this new class of hLig1 inhibitors could be promising leads for further drug development.


RSC Advances | 2016

Discovery of monocarbonyl curcumin hybrids as a novel class of human DNA ligase I inhibitors: in silico design, synthesis and biology

Dhanaraju Mandalapu; Deependra Kumar Singh; Sonal Gupta; Vishal M. Balaramnavar; Mohammad Shafiq; Dibyendu Banerjee; Vishnu L. Sharma

A pharmacophore model was generated and validated by using known human DNA ligase inhibitors for the identification of a novel series of monocarbonyl curcumin–thiourea/thiazole hybrids as human DNA ligase I (hLigI) inhibitors. These compounds (14–49) were synthesized and their antiligase and cytotoxic activities were evaluated in vitro. Several compounds from this series have shown significant inhibition of purified hLigI activity and exhibited a low micro molar range of cytotoxic activity against one or more cancer cell lines, with IC50 values ranging from 1.3–48.8 μM. Among these, compound 23 showed antiligase activity at an IC50 value 24.9 ± 1.8 μM, and selective cytotoxicity against DLD1 cancer cell line (IC50 value 8.7 ± 1.9 μM) compared to the reference curcumin (IC50 values were 51.9 ± 8.7 μM and 33.2 ± 1.8 μM for antiligase and cytotoxic activities against DLD1 cell line, respectively), and docking studies showed considerable interactions of compound 23 with hLigI. This new class of potent hLigI inhibitors will serve as a potential lead for further optimization and drug development.


RSC Advances | 2016

Identification of a novel human DNA ligase I inhibitor that promotes cellular apoptosis in DLD-1 cells: an in silico and in vitro mechanistic study

Deependra Kumar Singh; Mohd. Kamil Hussain; Shagun Krishna; Amit Laxmikant Deshmukh; Mohammad Shameem; Pooja Maurya; Kanchan Hajela; Mohammad Imran Siddiqi; Dibyendu Banerjee

The processes of DNA replication and repair are accomplished by the concerted action of several proteins. Among them human DNA ligases play an important role during the last step of almost all DNA replication and repair processes, where they seal the nicks between DNA strands. In humans, three kinds of DNA ligases (human DNA ligase I, III, IV) are found. DNA ligase I (hLigI) is involved in both DNA replication as well as in DNA repair pathways and is reported to be over-expressed in rapidly dividing cells, including cancer cells. For this reason, in this study we have targeted hLigI for studying its response as a novel anticancer target. We have screened for ligase I inhibitors from our in-house small molecule library by a previously validated pharmacophore based virtual screening method and found a novel hLigI inhibitor. This compound (S-097/98) demonstrated antiproliferative activities specifically in DLD-1 (colon), MDA-MB-231 (triple negative breast) and HepG2 (liver) cancer cell lines at low micromolar concentrations of 6–7 μM. Mechanistic studies show that the compound can directly interacts with the hLigI protein and inhibits ligation of both the purified protein in vitro, as well as in cell lysate of DLD-1 cells treated with the inhibitor. The compound also arrests cell cycle progression at the G2/M phase and increases the nuclear size of DLD-1 cancer cells, thereby demonstrating its antiproliferative activity. Finally, the compound promotes cellular apoptosis in DLD-1 cells.


Journal of Pharmaceutical and Biomedical Analysis | 2019

Elucidation of pharmacokinetics of novel DNA ligase I inhibitor, S012-1332 in rats: Integration of in vitro and in vivo findings

Mohammed Riyazuddin; Guru R. Valicherla; Athar Husain; Mohd. Kamil Hussain; Minakshi Shukla; Roshan Katekar; Anand P. Gupta; Pragati Singh; Dibyendu Banerjee; Kanchan Hajela; Jiaur R. Gayen

&NA; S012‐1332 is the first DNA ligase I inhibitor that demonstrated in vivo anti‐breast cancer activity. The present study aimed to assess the in vivo pharmacokinetics of S012‐1332 in rats and interpret them with in vitro findings. A sensitive and selective liquid chromatography‐tandem mass spectrometry bioanalytical method was developed and validated to determine S012‐1332. Following oral administration, the absolute bioavailability was 7.04%. The absorption was prolonged which can be explained by low solubility in simulated gastric fluid and several folds higher solubility in simulated intestinal fluid. The effective permeability across the intestinal membrane in in situ single pass perfusion study for S012‐1332 was 5.58 ± 1.83 * 10−5 cm/sec compared to 5.99 ± 0.65 * 10−5 cm/sec for carbamazepine, with no significant difference, indicating S012‐1332 has high permeability. It was rapidly partitioning into plasma in blood, where it was stable. Plasma protein binding was moderate which may have attributed to the rapid distribution out of the vascular compartment. The pharmacokinetics of S012‐1332 was characterized by extensive clearance as seen with rat liver and intestinal microsomes. In vitro results elucidate the in vivo pharmacokinetic data. These findings provide crucial information for further development of S012‐1332 as anti‐breast cancer agent. Graphical abstract Figure. No caption available. HighlightsLC‐MS/MS method has been developed and validated for the first time for S012‐1332.Absolute oral bioavailability of S012‐1332 was 7.04% in rats.In vitro findings show low solubility, high metabolism, moderate protein binding.Stable in plasma and biorelevant media and highly permeable.


Molecular Informatics | 2018

Multiple Machine Learning Based-Chemoinformatics Models for Identification of Histone Acetyl Transferase Inhibitors

Shagun Krishna; Sushil Kumar; Deependra Kumar Singh; Amar Deep Lakra; Dibyendu Banerjee; Mohammad Imran Siddiqi

The histone acetyl transferase (HAT) are involved in acetylation of histones that lead to transcription activation in numerous gene regulatory mechanisms. There are very few GCN5 HAT inhibitors reported despite of their role in cancer progression. In this study, we have utilized in‐silico virtual screening approaches based on various machine learning algorithm to identify potent inhibitors of GCN5 HAT from commercially available Maybridge library. We have generated predictive chemoinformatics models based on k‐Nearest neighbour, naïve Bayesian, Random Forest and Support Vector Machine. Based on statistical parameters, the RF and SVM models have shown comparative performance. Therefore, we performed the virtual screening with these two models and the consensus hits were selected for further evaluation using molecular docking into the active site of GCN‐5 HAT. Finally, a set of 10 molecules were selected and subjected to biological evaluation. Subsequently, inhibition of acetylation shown by three out of the ten molecules in the in‐vitro experiments validated their utility as potential HAT inhibitors. Furthermore, the selected hits have also shown weak cell growth decrease in MCF‐7 cancer cell lines, which suggests that after subsequent structural optimization the identified molecules may further be explored for the development of anti‐cancer agents.


Bioorganic & Medicinal Chemistry Letters | 2018

Biofilm inhibition and anti-Candida activity of a cationic lipo-benzamide molecule with twin-nonyl chain

Tushar Jain; Prathap Reddy Muktapuram; Komal Sharma; Owk Ravi; Garima Pant; Kalyan Mitra; Surendar Reddy Bathula; Dibyendu Banerjee

A series of cationic lipo-benzamide compounds with varying lengths of hydrocarbon chains (C2M-C18M) were evaluated for anti-Candida activity. Four compounds harbouring 8-11 hydrocarbon chains demonstrated concentration-dependent inhibition of fungal cell growth with Minimum Inhibitory Concentration (MIC) of ≤6.2 µg ml-1. The most active compound (C9M) inhibited growth of both Candida albicans and non-albicans strains and is equally active against pairs of azole sensitive and resistant clinical isolates of C. albicans. Compound C9M also inhibited different stages of Candida biofilms. Scanning Electron Microscopy (SEM) of Candida cells after C9M treatment was also done and no significant cell lysis was observed. Hemolysis assay was performed and only 2.5% haemolysis was observed at MIC concentration.


ACS Applied Materials & Interfaces | 2018

siRNA Delivery Using a Cationic-Lipid-Based Highly Selective Human DNA Ligase I Inhibitor

Surendar Reddy Bathula; Komal Sharma; Deependra Kumar Singh; Muktapuram Prathap Reddy; Pushpa Ragini Sajja; Amit Laxmikant Deshmukh; Dibyendu Banerjee

The present article illustrates the serendipitous discovery of a cationic-lipid-based human DNA ligase (hLig) I inhibitor and the development of siRNA delivering, a hLigI-targeted cationic-lipid-based nonviral vector. We have tested a small in-house library of structurally similar cationic lipo-anisamides for antiligase activity, and amongst tested, N-dodecyl-N-(2-(4-methoxybenzamido)ethyl)-N-methyldodecan-1-ammonium iodide (C12M) selectively and efficiently inhibited the enzyme activity of hLigI, compared to other human ligases (hLigIIIβ and hLigIV/XRCC4) and bacterial T4 DNA ligase. Furthermore, upon hydration with equimolar cholesterol, C12M produced antiligase cationic liposomes, which transfected survivin siRNA and showed significant inhibition of tumor growth.

Collaboration


Dive into the Dibyendu Banerjee's collaboration.

Top Co-Authors

Avatar

Deependra Kumar Singh

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mohammad Imran Siddiqi

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shagun Krishna

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Amit Laxmikant Deshmukh

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mohammad Shameem

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Komal Sharma

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Koneni V. Sashidhara

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Pooja Maurya

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Surendar Reddy Bathula

Indian Institute of Chemical Technology

View shared research outputs
Top Co-Authors

Avatar

Chandan Kumar

Central Drug Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge