Network


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

Hotspot


Dive into the research topics where Kaustuv Sahoo is active.

Publication


Featured researches published by Kaustuv Sahoo.


Bioorganic & Medicinal Chemistry | 2010

CLEFMA—An anti-proliferative curcuminoid from structure–activity relationship studies on 3,5-bis(benzylidene)-4-piperidones

Pallavi Lagisetty; Prachi Vilekar; Kaustuv Sahoo; Shrikant Anant; Vibhudutta Awasthi

3,5-Bis(benzylidene)-4-piperidones are being advanced as synthetic analogs of curcumin for anti-cancer and anti-inflammatory properties. We performed structure-activity relationship studies, by testing several synthesized 3,5-bis(benzylidene)-4-piperidones for anti-proliferative activity in lung adenocarcinoma H441 cells. Compared to the lead compound 1, or 3,5-bis(2-fluorobenzylidene)-4-piperidone, five compounds were found to be more potent (IC(50) < 30 microM), and 16 compounds possessed reduced cell-killing efficacy (IC(50) > 50 microM). Based on the observations, we synthesized 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] (29 or CLEFMA) as a novel analog of 1. CLEFMA was evaluated for anti-proliferative activity in H441 cells, and was found to be several folds more potent than compound 1. We did not find apoptotic cell population in flow cytometry, and the absence of apoptosis was confirmed by the lack of caspase cleavage. The electron microscopy of H441cells indicated that CLEFMA and compound 1 induce autophagic cell death that was inhibited by specific autophagy inhibitor 3-methyladenine. The results suggest that the potent and novel curcuminoid, CLEFMA, offers an alternative mode of cell death in apoptosis-resistant cancers.


Colloids and Surfaces B: Biointerfaces | 2011

Cyclodextrin-mediated entrapment of curcuminoid 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] or CLEFMA in liposomes for treatment of xenograft lung tumor in rats

Hrushikesh Agashe; Kaustuv Sahoo; Pallavi Lagisetty; Vibhudutta Awasthi

We recently reported a novel curcuminoid 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] or CLEFMA as a potent anti-proliferative agent, and showed that it induces autophagic cell death in lung cancer cells. We are now reporting a drug-in-CD-in-liposome approach to formulate CLEFMA liposomes that could be labeled with Tc-99m radionuclide for non-invasive imaging of their biodistribution. CLEFMA encapsulation was enabled by hydroxypropyl-β-cyclodextrin. In vitro studies showed that CLEFMA possessed more potent anti-proliferative activity in lung adenocarcinoma H441 cells than naturally occurring curcumin. At the same time, it had no effect on the proliferative capacity of normal lung fibroblasts. CLEFMA liposomes retained the antiproliferative potency of free CLEFMA, while maintaining its non-toxic nature in normal lung fibroblasts. In nude rats bearing xenograft H441 tumors, the tumor volume significantly reduced after i.v. treatment with CLEFMA liposomes (p<0.05); the tumor inhibition was determined to be 94%. The anti-tumor activity of CLEFMA liposomes was confirmed by the observation that F-18-fluorodeoxyglucose uptake in tumors of treated rats was reduced as compared to those of control rats. Tc-99m-labeled CLEFMA liposomes accumulated in liver (33.7%); spleen showed the largest accumulation on per gram tissue basis (6.2%/g). Upon histopathological examination of liver, lung and kidney, we found no apparent toxicity from multiple CLEFMA liposome administrations. The results demonstrate the utility of liposomes to serve as a carrier for CLEFMA. This study is the first to demonstrate the efficacy of novel curcuminoid CLEFMA in a preclinical model.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacologic Suppression of Inflammation by a Diphenyldifluoroketone, EF24, in a Rat Model of Fixed-Volume Hemorrhage Improves Survival

Vivek R. Yadav; Kaustuv Sahoo; Pamela R. Roberts; Vibhudutta Awasthi

An exaggerated release of proinflammatory cytokines and accompanying inflammation contributes to the development of multiple organ failure after hemorrhagic shock. Here, we tested the nuclear factor (NF) κ-light-chain-enhancer of activated B cell (NF-κB)–mediated transcriptional control of inflammatory pathways as a target in the management of hemorrhage-induced inflammation. We performed a study in a rat model of fixed-volume hemorrhage to investigate the anti-inflammatory effects of the diphenyldifluoroketone EF24 [3,5-bis(2-fluorobenzylidene)piperidin-4-one], an NF-κB inhibitor, in lung tissue. EF24 treatment (0.4 mg/kg) significantly prevented the upregulation of inflammatory biomarkers in rats subjected to 50% hemorrhage and preserved the pulmonary histology in hemorrhaged rats. The lung tissue from treated rats showed marked suppression of the hemorrhage-mediated induction of Toll-like receptor 4, phospho-p65 NF-κB, inducible nitric-oxide synthase, heme oxygenase–1, and cyclooxygenase-2 (COX-2). The hemorrhage-induced COX-2 activity was also significantly inhibited by the EF24 treatment. At the same time, EF24 induced nuclear factor (erythroid-derived 2)-like 2–mediated protective mechanisms against oxidative stress. EF24 also reduced hemorrhage-induced lung myeloperoxidase activity. The plasma levels of proinflammatory tumor necrosis factor-α, interleukin (IL)-6, IL-1α, and IL-1β were lower in EF24-treated rats than in untreated rats. Moreover, there was a significant reduction in the pulmonary expression of high-mobility group B1 protein. These biochemical effects were accompanied by a significant improvement in the survival of rats administered with EF24 as compared with the rats receiving vehicle control (P < 0.05). Overall, the results suggest that EF24 attenuates hemorrhage-induced inflammation and could serve as a salutary anti-inflammatory agent in resuscitation strategies.


Journal of Pharmacology and Experimental Therapeutics | 2014

Remediation of Hemorrhagic Shock-Induced Intestinal Barrier Dysfunction by Treatment with Diphenyldihaloketones EF24 and CLEFMA

Vivek R. Yadav; Alamdar Hussain; Kaustuv Sahoo; Vibhudutta Awasthi

Gut is very sensitive to hypoperfusion and hypoxia, and deranged gastrointestinal barrier is implicated in systemic failure of various organs. We recently demonstrated that diphenyldihaloketone EF24 [3,5-bis(2-fluorobenzylidene)piperidin-4-one] improves survival in a rat model of hemorrhagic shock. In this study, we tested EF24 and its other analog CLEFMA (4-[3,5-bis(2-chlorobenzylidene)-4-oxo-piperidine-1-yl]-4-oxo-2-butenoic acid) for their effect on intestinal barrier dysfunction in hypovolemic shock. Hypovolemia was induced in rats by withdrawing 50% of blood. EF24 or CLEFMA (0.4 mg/kg i.p.) treatment was provided, without volume resuscitation, after 1 hour of hemorrhage. Ileum was collected 5 hours after the treatment to investigate the expression of tight junction proteins (zonula occludens, claudin, and occludin) and epithelial injury markers [myeloperoxidase, ileal lipid-binding protein (ILBP), CD163, and plasma citrulline]. The ileal permeability for dextran-fluoroisothiocyanate and Evan’s blue dye was determined. EF24 and CLEFMA reduced the hypovolemia-induced plasma citrulline levels and the ileal expression of myeloperoxidase, ILBP, and CD163. The drugs also restored the basal expression levels of zonula occludens, claudin, and occludin, which were substantially deranged by hypovolemia. In ischemic ileum, the expression of phospho(tyrosine)-zonula occludens-1 was reduced, which was reinstated by EF24 and CLEFMA. In contrast, the drug treatments maintained the hypovolemia-induced expression of phospho(threonine)-occludin, but reduced that of phospho(tyrosine)-occludin. Both EF24 and CLEFMA treatments reduced the intestinal permeability enhanced by hypovolemia. EF24 and CLEFMA attenuate hypovolemic gut pathology and protect barrier function by restoring the status of tight junction proteins. These effects were observed in unresuscitated shock, implying the benefit of EF24 and CLEFMA in prehospital care of shock.


Investigational New Drugs | 2012

The curcuminoid CLEFMA selectively induces cell death in H441 lung adenocarcinoma cells via oxidative stress

Kaustuv Sahoo; Mikhail G. Dozmorov; Shrikant Anant; Vibhudutta Awasthi

SummaryCLEFMA or 4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid] is a curcuminoid being developed as an anticancer drug. We recently reported that it potently inhibits proliferation of various cancer cells. In this project, we investigated the effect of CLEFMA on gene expression profile in H441 lung adenocarcinoma cells, and studied its mechanism of action. In microarray data, we observed a deregulation of genes involved in redox and glutamate metabolism. Based on the affected ontologies, we hypothesized that antiproliferative activity of CLEFMA could be a result of the induction of reactive oxygen species (ROS). We tested this hypothesis by determining the levels of glutathione (GSH) and ROS in H441 cells treated with CLEFMA. We observed a rapid depletion of intracellular GSH/GSSG ratio. Using a cell-permeable fluorogenic substrate, we found that CLEFMA significantly induced ROS in a time- and dose-dependent manner (p < 0.05). Flow-cytometry with a mitochondria-selective fluorescent reporter of ROS indicated that the CLEFMA-induced ROS was of mitochondrial origin. In contrast to the cancer cells, the normal lung fibroblasts (CCL-151) did not show any increase in ROS and were resistant to CLEFMA-induced cell death. Furthermore, the addition of antioxidants, such as catalase, superoxide dismutase and N-acetylcysteine, rescued cancer cells from CLEFMA-induced cell death. Gene expression pathway analysis suggested that a transcription factor regulator Nrf2 is a pivotal molecule in the CLEFMA-induced deregulation of redox pathways. The immunoblotting of Nrf2 showed that CLEFMA treatment resulted in phosphorylation and nuclear translocation of Nrf2 in a time-dependent fashion. Based on these results, we conclude that induction of ROS is critical for the antiproliferative activity of CLEFMA and the Nrf2-mediated oxidative stress response fails to salvage H441 cells.


British Journal of Pharmacology | 2013

Preclinical evaluation of 4-[3,5-bis(2-chlorobenzylidene)-4-oxo-piperidine-1-yl]-4-oxo-2-butenoic acid, in a mouse model of lung cancer xenograft.

Vivek R. Yadav; Kaustuv Sahoo; Vibhudutta Awasthi

4‐[3,5‐Bis(2‐chlorobenzylidene)‐4‐oxo‐piperidine‐1‐yl]‐4‐oxo‐2‐butenoic acid CLEFMA is a new anti‐cancer molecule. Here, we investigated changes in apoptosis and inflammatory markers during CLEFMA‐induced tumour suppression.


Chemical Biology & Drug Design | 2012

Anticancer Activity of an Imageable Curcuminoid 1-[2-Aminoethyl-(6-hydrazinopyridine-3-carbamidyl)-3,5-bis-(2-fluorobenzylidene)-4-piperidone (EFAH)

Pallavi Lagisetty; Dharmalingam Subramaniam; Kaustuv Sahoo; Shrikant Anant; Vibhudutta Awasthi

3,5‐Bis(2‐fluorobenzylidine)‐4‐piperidone or EF24 is a potent anticancer derivative of curcumin. Using an amine derivative of EF24, we synthesized a hydrazinonicotinic acid conjugate, EFAH, for Tc‐99m radiolabelling and single photon emission tomography imaging. The aqueous solubility of EFAH (3.5 mg/mL) was significantly more than that of EF24 (1.2 mg/mL); the octanol/water partition coefficient of EFAH was estimated at log P = 0.33. As an antiproliferative agent, EFAH was as effective as EF24 in suppressing the proliferation of H441, MiaPaCa‐2 and Panc‐1 cells. Daily intraperitoneal injection of EFAH (5 μg) for 3 weeks in mice carrying xenografts of Panc‐1 pancreatic cancer showed a mean tumour volume reduction of 79%; the tumour weight decreased by 82% in the treated group. For imaging and biodistribution, EFAH was labelled with Tc‐99m (98% RCY) and intravenously administered in rats. Approximately 23.7% and 14.3% of injected dose accumulated in liver and intestine, respectively, suggesting that EFAH is mostly eliminated by hepatobiliary route. The results indicate that HYNIC modification of EF24 for Tc‐99m radiolabelling does not affect its antiproliferative efficacy. For the first time, a visual biodisposition of EF24 in a live animal model has been demonstrated. Such knowledge could be of benefit in developing therapeutic curcuminoids, such as EF24.


Cancer Research | 2012

Abstract 4887: Curcuminoid CLEFMA induces autophagy-associated apoptosis in lung cancer

Kaustuv Sahoo; Vivek R. Yadav; Vibhudutta Awasthi

CLEFMA (4-[3,5-bis(2-chlorobenzylidene-4-oxo-piperidine-1-yl)-4-oxo-2-butenoic acid) is a novel curcuminoid discovered in our laboratory. Earlier we showed that CLEFMA induces oxidative stress-dependent cell death in lung adenocarcinoma H441 cells. Although apoptosis is the desired mechanism for chemotherapy-induced cell death, the associated pathways are often defective in lung cancers resulting in high levels of chemoresistance. Therefore, drugs that induce alternate modes of cell death, such as autophagy, may be beneficial. Drug-induced hyperinduction of autophagy is classified as type 2 programmed cell death. In most circumstances, autophagy parallels apoptosis, and such composite cell death is described as “autophagy-associated apoptosis”. Whether autophagy plays a synergistic or antagonistic role in overall cell death response to anticancer drugs is an important focus in cancer research. In this study, we have investigated the interaction between autophagy and apoptosis in CLEFMA mediated cell death. Methods: The human non-small cell lung carcinoma (H441) cell line was treated with CLEFMA (10 µM) and the molecular mechanisms of cell death were investigated. Growth inhibition was assessed by MTT or hexosaminidase assay. Apoptosis was measured by flow cytometry of annexin V-stained cells, RT-PCR and immunoblotting for apoptosis-related biomarkers. Induction of autophagy was established by electron microscopy and immunobloting for LC3B, ATG, beclin1, AKT and mTOR. Autophagy was inhibited by 3-methyladenine (3-MA 1 mM) as well as chloroquine and E64D+pepstatin combination. The tumor suppressive efficacy of CLEFMA was demonstrated in H441 xenograft mice intraperitoneally receiving 0, 5 and 10 µg/day of CLEFMA for 32 days. The mice were imaged for F-18-fluorodeoxyglucose uptake in tumor tissue. Results: CLEFMA suppressed the viability of H441 cell line in a dose-dependent manner (IC50 = 6.25 µM). It induced cell cycle arrest at G2/M phase with accompanying accumulation of p21 protein. Caspase 3/7 immunoblotting and annexin V flow cytometry confirmed the induction of apoptosis. At the same time, we observed induction of LC3BII and suppression of AKT/mTOR phosphorylation. The suppression of autophagy significantly increased CLEFMA-induced cell death and increased caspase cleavage. In the xenograft model, 5 and 10 µg CLEFMA significantly reduced the tumor volume by 72% and 96.2%, respectively; the tumor uptake of F-18-fluorodeoxyglucose was also reduced. Conclusions: We demonstrate the efficacy of CLEFMA as a potent tumor suppressive agent. The mechanistic results suggest that autophagy is a survival mechanism in CLEFMA-treated lung cancer cells and it antagonizes apoptosis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4887. doi:1538-7445.AM2012-4887


Journal of Nanoparticle Research | 2011

Liposome-encapsulated EF24-HPβCD inclusion complex: a preformulation study and biodistribution in a rat model

Hrushikesh Agashe; Pallavi Lagisetty; Kaustuv Sahoo; D. Bourne; B. Grady; Vibhudutta Awasthi


Journal of Nature and Science | 2015

Imaging the Electromagnetic Field of Plants (Vigna radiata) Using Iron Particles: Qualitative and quantitative correlates

Benjamin J. Scherlag; Bing Huang; Ling Zhang; Kaustuv Sahoo; Rheal A. Towner; Natalya Smith; Abraham A. Embi; Sunny S. Po

Collaboration


Dive into the Kaustuv Sahoo's collaboration.

Top Co-Authors

Avatar

Vibhudutta Awasthi

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vivek R. Yadav

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Hrushikesh Agashe

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin J. Scherlag

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alamdar Hussain

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

D. Bourne

University of Oklahoma Health Sciences Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge