Network


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

Hotspot


Dive into the research topics where Arghya Adhikary is active.

Publication


Featured researches published by Arghya Adhikary.


Stem Cell Research & Therapy | 2014

Curcumin inhibits breast cancer stem cell migration by amplifying the E-cadherin/β-catenin negative feedback loop

Shravanti Mukherjee; Minakshi Mazumdar; Samik Chakraborty; Argha Manna; Shilpi Saha; Poulami Khan; Pushpak Bhattacharjee; Deblina Guha; Arghya Adhikary; Sanhita Mukhjerjee; Tanya Das

IntroductionThe existence of cancer stem cells (CSCs) has been associated with tumor initiation, therapy resistance, tumor relapse, angiogenesis, and metastasis. Curcumin, a plant ployphenol, has several anti-tumor effects and has been shown to target CSCs. Here, we aimed at evaluating (i) the mechanisms underlying the aggravated migration potential of breast CSCs (bCSCs) and (ii) the effects of curcumin in modulating the same.MethodsThe migratory behavior of MCF-7 bCSCs was assessed by using cell adhesion, spreading, transwell migration, and three-dimensional invasion assays. Stem cell characteristics were studied by using flow cytometry. The effects of curcumin on bCSCs were deciphered by cell viability assay, Western blotting, confocal microscopy, and small interfering RNA (siRNA)-mediated gene silencing. Evaluations of samples of patients with breast cancer were performed by using immunohistochemistry and flow cytometry.ResultsHere, we report that bCSCs are endowed with aggravated migration property due to the inherent suppression of the tumor suppressor, E-cadherin, which is restored by curcumin. A search for the underlying mechanism revealed that, in bCSCs, higher nuclear translocation of beta-catenin (i) decreases E-cadherin/beta-catenin complex formation and membrane retention of beta-catenin, (ii) upregulates the expression of its epithelial-mesenchymal transition (EMT)-promoting target genes (including Slug), and thereby (iii) downregulates E-cadherin transcription to subsequently promote EMT and migration of these bCSCs. In contrast, curcumin inhibits beta-catenin nuclear translocation, thus impeding trans-activation of Slug. As a consequence, E-cadherin expression is restored, thereby increasing E-cadherin/beta-catenin complex formation and cytosolic retention of more beta-catenin to finally suppress EMT and migration of bCSCs.ConclusionsCumulatively, our findings disclose that curcumin inhibits bCSC migration by amplifying E-cadherin/beta-catenin negative feedback loop.


Biomaterials | 2015

PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a

Saurav Bhattacharya; Prasun Patra; Sudeshna Mukherjee; Swatilekha Ghosh; Minakshi Mazumdar; S. Chattopadhyay; Tanya Das; Dhrubajyoti Chattopadhyay; Arghya Adhikary

Thymoquinone (TQ), a major active constituent of black seeds of Nigella sativa, has potential medical applications including spectrum of therapeutic properties against different cancers. However, little is known about their effect on breast cancer cell migration, which is the cause of over 90% of deaths worldwide. Herein, we have synthesized TQ-encapsulated nanoparticles using biodegradable, hydrophilic polymers like polyvinylpyrrolidone (PVP) and polyethyleneglycol (PEG) to overcome TQs poor aqueous solubility, thermal and light sensitivity as well as consequently, minimal systemic bioavailability which can greatly improve the cancer treatment efficiency. Sizes of synthesized TQ-Nps were found to be below 50 nm and they were mostly spherical in shape with smooth surface texture. Estimation of the zeta potential also revealed that all the three TQ-Nps were negatively charged which also facilitated their cellular uptake. In the present investigation, we provide direct evidence that TQ-Nps showed more efficiency in killing cancer cells as well as proved to be less toxic to normal cells at a significantly lower dose than TQ. Interestingly, evaluation of the anti-migratory effect of the TQ-Nps, revealed that PEG4000-TQ-Nps showed much potent anti-migratory properties than the other types. Further studies indicated that PEG4000-TQ-Nps could significantly increase the expression of miR-34a through p53. Moreover, NPs mediated miR-34a up-regulation directly down-regulated Rac1 expression followed by actin depolymerisation thereby disrupting the actin cytoskeleton which leads to significant reduction in the lamellipodia and filopodia formation on cell surfaces thus retarding cell migration. Considering the biodegradability, non-toxicity and effectivity of PEG4000-TQ-Nps against cancer cell migration, TQ-Nps may provide new insights into specific therapeutic approach for cancer treatment.


Journal of Nutritional Biochemistry | 2016

Pomegranate protects against arsenic-induced p53-dependent ROS-mediated inflammation and apoptosis in liver cells

Sreetama Choudhury; Sayan Ghosh; Sudeshna Mukherjee; Payal Gupta; Saurav Bhattacharya; Arghya Adhikary; S. Chattopadhyay

Molecular mechanisms involved in arsenic-induced toxicity are complex and elusive. Liver is one of the most favored organs for arsenic toxicity as methylation of arsenic occurs mostly in the liver. In this study, we have selected a range of environmentally relevant doses of arsenic to examine the basis of arsenic toxicity and the role of pomegranate fruit extract (PFE) in combating it. Male Swiss albino mice exposed to different doses of arsenic presented marked hepatic injury as evident from histological and electron microscopic studies. Increased activities of enzymes alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase and alkaline phosphatase corroborated extensive liver damage. It was further noted that arsenic exposure initiated reactive oxygen species (ROS)-dependent apoptosis in the hepatocytes involving loss of mitochondrial membrane potential. Arsenic significantly increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor-κB (NF-κB), coupled with increase in phosphorylated Iκ-B, possibly as adaptive cellular survival strategies. Arsenic-induced oxidative DNA damage to liver cells culminated in p53 activation and increased expression of p53 targets like miR-34a and Bax. Pomegranate polyphenols are known to possess remarkable antioxidant properties and are capable of protecting normal cells from various stimuli-induced oxidative stress and toxicities. We explored the protective role of PFE in ameliorating arsenic-induced hepatic damage. PFE was shown to reduce ROS generation in hepatocytes, thereby reducing arsenic-induced Nrf2 activation. PFE also inhibited arsenic-induced NF-κB-inflammatory pathway. Data revealed that PFE reversed arsenic-induced hepatotoxicity and apoptosis by modulating the ROS/Nrf2/p53-miR-34a axis. For the first time, we have mapped the possible signaling pathways associated with arsenic-induced hepatotoxicity and its rescue by pomegranate polyphenols.


Biomaterials | 2016

Tailored-CuO-nanowire decorated with folic acid mediated coupling of the mitochondrial-ROS generation and miR425-PTEN axis in furnishing potent anti-cancer activity in human triple negative breast carcinoma cells.

Saurav Bhattacharya; Soumendu Karmakar; Ayan Mukhopadhyay; Sudeshna Mukherjee; Swatilekha Ghosh; S. Chattopadhyay; Prasun Patra; Arghya Adhikary

Metal oxide nanoparticles are the forthcoming anti-tumor therapeutics and provide a versatile platform in the development of therapeutic approaches for drug-resistant cancers such as triple negative breast cancer (TNBC). Copper oxide nanoparticles have been characterized as anti-cancer agents but its toxicity has been a matter of concern. Herein, we have developed a targeted CuO Nanowire fabricated with Folic acid (CuO-Nw-FA) that enables enhanced cellular uptake in TNBC cells without imparting significant toxicity in normal cellular system. In the present study, we enumerated that CuO-Nw-FA caused mitochondrial-dependent apoptosis in MDAMB-231 cells. Furthermore, CuO-Nw-FA mediated cytosolic retardation of NF-κB favoured inactivation of miR-425 and henceforth activated PTEN to induce apoptosis in TNBC cells. Simultaneously, CuO-Nw-FA also restricted the in-vitro cell migration through the miR-425/PTEN axis via pFAK. Studies extended to ex-ovo and in-vivo mice models further validated the efficacy of CuO-Nw-FA. Additionally, the accumulations of nanoparticles in tumor as well as different organs in mice were examined by in-vivo biodistribution and ex-vivo optical imaging studies. Thus our results cumulatively propose that CuO-Nw-FA cross-talks two distinct signalling pathways to induce apoptosis and retard migration in TNBC cells and raises the possibility for the use of CuO-Nw-FA as a potent anti-tumor agent.


Molecular Carcinogenesis | 2017

Induction of mitochondrial apoptotic pathway in triple negative breast carcinoma cells by methylglyoxal via generation of reactive oxygen species

Anirban Roy; Saurav Bhattacharya; Pravat Kumar Parida; Arghya Adhikary; Kuladip Jana; Manju Ray

Triple negative breast cancer (TNBC) tends to form aggressive tumors associated with high mortality and morbidity which urge the need for development of new therapeutic strategies. Recently, the normal metabolite Methylglyoxal (MG) has been documented for its anti‐proliferative activity against human breast cancer. However, the mode of action of MG against TNBC remains open to question. In our study, we investigated the anticancer activity of MG in MDA MB 231 and 4T1 TNBC cell lines and elucidated the underlying mechanisms. MG dose‐dependently caused cell death, induced apoptosis, and generated ROS in both the TNBC cell lines. Furthermore, such effects were attenuated in presence of ROS scavenger N‐Acetyl cysteine. MG triggered mitochondrial cytochrome c release in the cytosol and up‐regulated Bax while down‐regulated anti‐apoptotic protein Bcl‐2. Additionally, MG treatment down‐regulated phospho‐akt and inhibited the nuclear translocation of the p65 subunit of NF‐κB. MG exhibited a tumor suppressive effect in BALB/c mouse 4T1 breast tumor model as well. The cytotoxic effect was studied using MTT assay. Apoptosis, ROS generation, and mitochondrial dysfunction was evaluated by flow cytometry as well as fluorescence microscopy. Western blot assay was performed to analyze proteins responsible for apoptosis. This study demonstrated MG as a potent anticancer agent against TNBC both in vitro and in vivo. The findings will furnish fresh insights into the treatment of this subgroup of breast cancer.


RSC Advances | 2016

Towards the realization of luminescence from visible emitting trivalent lanthanides (Sm, Eu, Tb, Dy) in polar zinc sulfide nanoparticles: evaluation of in vitro cytotoxicity

Arijita Chakraborty; Gouranga H. Debnath; Saurav Bhattacharya; Priyanka Upadhyay; Arghya Adhikary; Prasun Mukherjee

This work reports on the realization of luminescence from four visible emitting trivalent lanthanide (Ln) cations [samarium (Sm), europium (Eu), terbium (Tb) and dysprosium (Dy)] in polar zinc sulfide nanoparticles, Zn(Ln)S. Among the Zn(Ln)S nanoparticles studied, noticeable lanthanide cation centered luminescence has only been realized from the Zn(Tb)S and Zn(Eu)S nanoparticles in water, whereas no such effect has been observed in the corresponding Sm and Dy containing nanoparticles. In all the nanoparticles with characteristic lanthanide luminescence, the nanoparticles were found to be acting as an optical antenna and protector matrix, in order to realize luminescence from the respective lanthanide cations. The results have been rationalized with the lanthanide cations acting as charge (hole and/or electron) traps in the semiconductor nanoparticle matrix along with associated environmental effects. A comparison of the corresponding hydrophobic Zn(Tb)S nanoparticles reveal significant differences in photophysical properties. Finally, the hydrophilic Zn(Tb)S nanoparticles have been examined for in vitro cytotoxicity and the results indicate potential anti-cancer therapy for human mammary adenocarcinoma cells (MDA-MB-231) with the capability of cellular imaging.


Frontiers in Pharmacology | 2016

Phemindole, a Synthetic Di-indole Derivative Maneuvers the Store Operated Calcium Entry (SOCE) to Induce Potent Anti-Carcinogenic Activity in Human Triple Negative Breast Cancer Cells

Supriya Chakraborty; Swatilekha Ghosh; Bhaswati Banerjee; Abhishek Santra; Arghya Adhikary; Anup Kumar Misra; Parimal C. Sen

Triple-negative breast cancer (TNBC), is a specific subtype of epithelial breast tumors that are immuno-histochemically negative for the protein expression of the estrogen receptor (ER), the progesterone receptor (PR) and lack over expression/gene amplification of HER2. This subtype of breast cancers is highly metastatic, shows poor prognosis and hence represents an important clinical challenge to researchers worldwide. Thus alternative approaches of drug development for TNBC have gained utmost importance in the present times. Dietary indole and its derivatives have gained prominence as anti-cancer agents and new therapeutic approaches are being developed to target them against TNBC. But a major drawback with 3, 3′di Indolyl methane (DIM) is their poor bioavailability and high effective concentration against TNBC. However, the Aryl methyl ring substituted analogs of DIM display interesting anti-cancer activity in breast cancer cells. In the current study we report the synthesis of a novel synthetic aryl methyl ring substituted analog of DIM, named as Phemindole as an effective anti-tumor agent against TNBC cells. Furthermore, we enumerated that Phemindole caused reactive oxygen species mediated mitochondrial-dependent apoptosis in MDAMB-231 cells. Furthermore, Phemindole mediated Store Operated Calcium Entry (SOCE) retardation favored inactivation of STIM1 and henceforth activated ER stress to induce apoptosis in TNBC cells. Simultaneously, Phemindole was also found to restrict the in vitro cell migration through its anti mitotic property and pFAK regulation. Studies extended to ex ovo and in vivo mice models further validated the efficacy of Phemindole. Thus our results cumulatively propose Phemindole as a new chemotherapeutic regime which might be effective to target the deadly aspects of the TNBC.


Colloids and Surfaces B: Biointerfaces | 2015

Simple synthesis of biocompatible biotinylated porous hexagonal ZnO nanodisc for targeted doxorubicin delivery against breast cancer cell: In vitro and in vivo cytotoxic potential

Prasun Patra; Shouvik Mitra; Amarto Das Gupta; Saheli Pradhan; Saurav Bhattacharya; Sudeshna Mukherjee; Sampad Sarkar; Subhrodeb Roy; Sreya Chattopadhyay; Arghya Adhikary; Arunava Goswami; Dhrubajyoti Chattopadhyay

Targeted drug delivery with porous materials features great promise as improved therapeutic potential for treatment of various diseases. In the present study we have attempted a microwave synthesis of porous hexagonal nanodisc of zinc oxide (PZHD) for the first time and its subsequent targeted delivery to breast cancer cells, MCF7. PZHD has been fabricated suitably with 3-aminopropyltriethoxysilane to impart additional stability and surface amines to anchor site directing ligand NHS-biotin. Biotinylated scaffold showed targeted delivery of anticancer drug doxorubicin and pH triggered release to MCF 7 cells with preferential distribution on specified domain. A detailed in vitro cytotoxicity study was associated with it to evaluate the mode of action of Dox loaded PZHD on MCF-7 cells by means of cell cycle analysis, apoptosis assays, Western blot and immuno-fluorescence image analysis. The efficacy of the Dox loaded PZHD was further validated from our in vivo tumor regression studies. Finally, the whole study has been supported by in vitro and in vivo bio-safety studies which also signified its biocompatibility with real time applications. To the best of our knowledge this is the first effort to use biotinylated PZHD for targeted delivery of doxorubicin within MCF 7 cells with a detailed study of its mechanistic application. This study might thus hold future prospects for therapeutic intervention for treatment of cancer.


Biomedicine & Pharmacotherapy | 2017

Induction of apoptosis in human colorectal cancer cell line, HCT-116 by a vanadium- Schiff base complex

Abhinaba Sinha; Kaushik Banerjee; Arpita Banerjee; Avijit Sarkar; Arghya Adhikary; Mitali Chatterjee; Soumitra Kumar Choudhuri

Vanadium compounds are well known for their therapeutic interventions against several diseases. Various biochemical attributes of vanadium complexes inspired us to evaluate the cancer cell killing efficacy of the vanadium complex, viz., vanadyl N-(2-hydroxyacetophenone) glycinate [VO(NG)2]. Previously we showed that VO(NG)2 is an effective anticancer agent in in vitro and in vivo cancer models and imposed miniscule side effects. Herein we report that VO(NG)2 is significantly cytotoxic to various cancer cell lines. Furthermore, this redox active vanadyl complex altered the redox homeostatsis of many human cancer cell lines significantly. VO(NG)2 actuates programmed cell death in human colorectal carcinoma cells(HCT-116) through mitochondrial outer membrane permeabilization but in caspase independent manner, possibly by altering cellular redox status and by inflicting DNA damage. Thus, the present work is an attempt to provide many evidences regarding the potent and selective chemotherapeutic efficacy of the novel VO(NG)2.


New Journal of Chemistry | 2015

Multifaceted core–shell nanoparticles: superparamagnetism and biocompatibility

Debasmita Sardar; Sushanto Neogi; S. Bandyopadhyay; Biswarup Satpati; Arghya Adhikary; Ruchi Jain; Chinnakonda S. Gopinath; Tanushree Bala

NicoreAgshell nanoparticles were synthesized by redox transmetallation reaction. Reduction potential match was encouraging to attempt the synthesis of the NicoreAushell system too. However, it could be achieved only after an effective surface modification on the Ni-core. Thorough characterization (UV-Vis spectroscopy, fluorescence spectroscopy, XRD, XPS, FTIR, TEM, and EDX) proved the necessity of surface modification and the success of synthesis of both types of core–shell structures. The chemical composition and topography were determined using STEM-HAADF analysis and EFTEM imaging. Fourier transform infrared (FTIR) spectroscopy confirmed the surface modification of Ni nanoparticles and the interactions involved between the ligands and metals (in the core and/or the shell) at various steps of the synthetic process. Even after the formation of the noble metal shell, the magnetic core was found to retain its superparamagnetic nature. In addition, the Au-shell protected the core from aerial oxidation and decreased toxicity as compared to pristine Ni nanoparticles as observed by MTT assay on normal cells (PBMCs).

Collaboration


Dive into the Arghya Adhikary's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sudeshna Mukherjee

College of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Payal Gupta

University of Calcutta

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sayan Ghosh

University of Calcutta

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge