Network


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

Hotspot


Dive into the research topics where Deependra Kumar Singh is active.

Publication


Featured researches published by Deependra Kumar Singh.


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.


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.


Scientific Reports | 2017

A Novel Benzocoumarin-Stilbene Hybrid as a DNA ligase i inhibitor with in vitro and in vivo anti-tumor activity in breast cancer models

Mohd. Kamil Hussain; Deependra Kumar Singh; Akhilesh Singh; Mohd. Asad; Mohd. Imran Ansari; Mohammad Shameem; Shagun Krishna; Guru R. Valicherla; Vishal Makadia; Sanjeev Meena; Amit Laxmikant Deshmukh; Jiaur R. Gayen; Mohammad Imran Siddiqi; Dipak Datta; Kanchan Hajela; Dibyendu Banerjee

Existing cancer therapies are often associated with drug resistance and toxicity, which results in poor prognosis and recurrence of cancer. This necessitates the identification and development of novel therapeutics against existing as well as novel cellular targets. In this study, a novel class of Benzocoumarin-Stilbene hybrid molecules were synthesized and evaluated for their antiproliferative activity against various cancer cell lines followed by in vivo antitumor activity in a mouse model of cancer. The most promising molecule among the series, i.e. compound (E)-4-(3,5-dimethoxystyryl)-2H-benzo[h]chromen-2-one (19) showed maximum antiproliferative activity in breast cancer cell lines (MDA-MB-231 and 4T1) and decreased the tumor size in the in-vivo 4T1 cell-induced orthotopic syngeneic mouse breast cancer model. The mechanistic studies of compound 19 by various biochemical, cell biology and biophysical approaches suggest that the compound binds to and inhibits the human DNA ligase I enzyme activity that might be the cause for significant reduction in tumor growth and may constitute a promising next-generation therapy against breast cancers.


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.


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.


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.


ACS Medicinal Chemistry Letters | 2014

Discovery of 3-Arylcoumarin-tetracyclic Tacrine Hybrids as Multifunctional Agents against Parkinson’s Disease

Koneni V. Sashidhara; Ram K. Modukuri; Pooja Jadiya; K. Bhaskara Rao; Tanuj Sharma; Rizwanul Haque; Deependra Kumar Singh; Dibyendu Banerjee; Mohammad Imran Siddiqi; Aamir Nazir


DNA Repair | 2016

Dynamics of replication proteins during lagging strand synthesis: A crossroads for genomic instability and cancer

Amit Laxmikant Deshmukh; Chandan Kumar; Deependra Kumar Singh; Pooja Maurya; Dibyendu Banerjee


Molecular BioSystems | 2017

Identification of human flap endonuclease 1 (FEN1) inhibitors using a machine learning based consensus virtual screening

Amit Laxmikant Deshmukh; Sharat Chandra; Deependra Kumar Singh; Mohammad Imran Siddiqi; Dibyendu Banerjee

Collaboration


Dive into the Deependra Kumar Singh's collaboration.

Top Co-Authors

Avatar

Dibyendu Banerjee

Council of Scientific and Industrial Research

View shared research outputs
Top Co-Authors

Avatar

Amit Laxmikant Deshmukh

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

Mohammad Shameem

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dipak Datta

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kanchan Hajela

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Komal Sharma

Council of Scientific and Industrial Research

View shared research outputs
Top Co-Authors

Avatar

Mohd. Kamil Hussain

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Pooja Maurya

Central Drug Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge