Network


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

Hotspot


Dive into the research topics where Nando Dulal Das is active.

Publication


Featured researches published by Nando Dulal Das.


Phytotherapy Research | 2011

Terminalia chebula extract acts as a potential nf-κb inhibitor in human lymphoblastic t cells

Nando Dulal Das; Kyoung Hwa Jung; Ji Hyun Park; Md. Abdul Mojid Mondol; Hee Jae Shin; Hyi-Seung Lee; Kyoung Sun Park; Mi Ran Choi; Kyung Suk Kim; Moo Sung Kim; Sang Rin Lee; Young Gyu Chai

Terminalia chebula (TC) is native to southern Asia to southwestern China and is used in traditional medicine for the treatment of human ailments including malignant tumors and diabetes. This plant also has antibacterial and immunomodulatory properties. Nuclear factor kappa‐light chain‐enhancer of activated B cells (NF‐κB) is responsible for the expression of numerous genes involved in cell survival, proliferation, angiogenesis, inflammation, invasion and metastasis, among other processes. This study aims to assess the NF‐κB inhibitory effect of TC extract in human lymphoblastic T (Jurkat) cells. The effects of TC extract were investigated using the FRET‐based Gene Blazer technique in transfected Jurkat‐NF‐κB‐RE‐bla cells. The concentration of TC extract required for NF‐κB inhibition was determined by a cell proliferation assay. Treatment with TC extract (50 μg/mL) inhibited NF‐κB activity and protected against IκBα degradation and strongly suppressed IκBα phosphorylation in Jurkat‐NF‐κB‐RE‐bla cells. This treatment might be crucial for inhibiting NF‐κB translocation and activation. In addition, the TC extract downregulated certain NF‐κB regulated genes, including IL‐8 and MCP‐1, in Jurkat‐NF‐κB‐RE‐bla cells. Moreover, gallic acid was identified from the TC extract demonstrating its ability to inhibit NF‐κB activity in Jurkat‐NF‐κB‐RE‐bla cells. Further studies to identify the role of gallic acid in NF‐κB inhibition may uncover the crucial antiinflammatory and antitumor properties of the TC extract. Copyright


Cell Biochemistry and Function | 2012

Gene networking and inflammatory pathway analysis in a JMJD3 knockdown human monocytic cell line.

Nando Dulal Das; Kyoung Hwa Jung; Mi Ran Choi; Hyun Soo Yoon; Seung Hyun Kim; Young Gyu Chai

JMJD3, a Jumonji C family histone demethylase, is induced by transcription factor, nuclear factor‐kappa B (NF‐κB), in response to various stimuli. JMJD3 is crucial for erasing histone‐3 lysine‐27 trimethylation (H3K27me3), a modification associated with transcriptional repression and is responsible for the activation of a diverse set of genes. Here, we identify the genes in human leukaemia monocyte (THP‐1) human monocytic cells that are significantly affected by the stable knockdown (kd) of JMJD3. Global gene expression levels were detected in stable JMJD3 knockdown THP‐1 cells and in tumor necrosis factor‐alpha (TNF‐α)‐stimulated JMJD3‐kd THP‐1 cells by using a 12‐plex NimbleGen human whole genome array. In addition, datasets were analysed by using Ingenuity Pathway Analysis. Stable knockdown of JMJD3 in THP‐1 cells affected particularly in expression levels and in downstream effects on inflammatory signalling pathways. JMJD3 attenuation down‐regulates various key genes in NF‐κB, chemokine and CD40 signalling, and mostly affects inflammatory disease response molecules. In addition, chromatin immunoprecipitation revealed that JMJD3‐kd could inhibit several NF‐κB‐regulated inflammatory genes by recruiting repressive histone‐3 lysine‐27 trimethylation to their promoters. Moreover, this study significantly highlights the connexion of NF‐κB with JMJD3, which suggests an epigenetic regulation in different signalling pathways. Finally, this study establishes novel JMJD3 targets through Ingenuity Pathway Analysis. Copyright


PLOS ONE | 2010

The Role of NF-κB and H3K27me3 Demethylase, Jmjd3, on the Anthrax Lethal Toxin Tolerance of RAW 264.7 Cells

Nando Dulal Das; Kyoung Hwa Jung; Young Gyu Chai

Background In Bacillus anthracis, lethal toxin (LeTx) is a critical virulence factor that causes immune suppression and toxic shock in the infected host. NF-κB is a key mediator of the inflammatory response and is crucial for the plasticity of first level immune cells such as macrophages, monocytes and neutrophils. In macrophages, this inflammatory response, mediated by NF-κB, can regulate host defense against invading pathogens. A Jumonji C family histone 3 lysine-27 (H3K27) demethylase, Jmjd3, plays a crucial role in macrophage plasticity and inflammation. Here we report that NF-κB and Jmjd3 can modulate the LeTx intoxication resistance of RAW 264.7 cells. Principal Findings This study showed that a 2 h exposure of macrophages to LeTx caused substantial cell death with a survival rate of around 40%. The expression of the Jmjd3 gene was induced 8-fold in intoxication-resistant cells generated by treatment with lipopolysaccharides of RAW 264.7 cells. These intoxication-resistant cell lines (PLx intox and PLxL intox) were maintained for 8 passages and had a survival rate of around 100% on secondary exposure to LeTx and lipopolysaccharides. Analysis of NF-κB gene expression showed that the expression of p100, p50 and p65 was induced around 20, 7 and 4 fold, respectively, in both of the intoxication-resistant cell lines following a 2 h treatment with PLxL (0.1+0.1+1 µg/ml). In contrast, these NF-κB genes were not induced following treatment with PLx treatment at the same concentrations. Conclusions Although LeTx influences macrophage physiology and causes defects of some key signaling pathways such as GSK3β which contributes to cytotoxicity, these results indicate that modulation of NF-κB by p50, p100 and Jmjd3 could be vital for the recovery of murine macrophages from exposure to the anthrax lethal toxin.


Journal of Chemical Neuroanatomy | 2012

Effect of fluoxetine on the expression of tryptophan hydroxylase and 14-3-3 protein in the dorsal raphe nucleus and hippocampus of rat.

Mi Ran Choi; Sejin Hwang; Geu Meum Park; Kyung Hwa Jung; Seok Hyeon Kim; Nando Dulal Das; Young Gyu Chai

The serotonergic system is one of the major systems targeted in the pharmacological treatment of mood disorders including depression. Fluoxetine, one of the selective serotonin reuptake inhibitors (SSRIs), has been reported to induce the expression of tryptophan hydroxylase (TPH), the rate-limiting enzyme in the biosynthesis of serotonin. The 14-3-3 protein family not only activates neuronal enzymes, including TPH, but also plays a role in a wide variety of cell signaling. The aim of the present study was to determine whether fluoxetine regulates both the interaction of TPH and 14-3-3 proteins as well as the increase of those proteins in the dorsal raphe nucleus and the hippocampus. Sprague-Dawley rats were administered fluoxetine or vehicle for 5 and 14 days and sacrificed at 5 and 14 days after initial treatment. The intensity of immunoreactivity for TPH and 14-3-3 proteins in the dorsal raphe nucleus of the midbrain and in the hippocampus was measured, and the colocalization of both proteins was observed with double-labeling immunofluorescence. At 5 days after initial treatment with fluoxetine, immunoreactivity of 14-3-3 protein increased in both the dorsal raphe nucleus and the hippocampus, while that of TPH did not change in either region. In addition, at 14 days after initial treatment with fluoxetine, immunoreactivity of 14-3-3 protein significantly increased in both the dorsal raphe nucleus and hippocampus, while that of TPH showed few changes in either region. Colocalization of TPH and 14-3-3 proteins was observed in the cell bodies of dorsal raphe nucleus, whereas it was not observed in the hippocampus. These results suggest that the time-dependent regulation of 14-3-3 protein may be one of the various factors associated with delayed pharmacological effects of SSRIs.


Neurotoxicity Research | 2013

Functional Analysis of Histone Demethylase Jmjd2b on Lipopolysaccharide-Treated Murine Neural Stem Cells (NSCs)

Nando Dulal Das; Mi Ran Choi; Kyoung Hwa Jung; Ji Hyun Park; Hyung Tae Lee; Amitabh Das; Seung Hyun Kim; Young Gyu Chai

Neural stem cell (NSC) neurogenesis is the formation of new neurons by which the brain maintains its lifelong plasticity in response to extrinsic and intrinsic changes. Here, we show the effect of lipopolysaccharides (LPS) as an in vitro model of inflammation on NSCs to determine whether the inflammatory mediators can epigenetically affect NSCs and alter their proliferation and differentiation abilities. To study the effect of LPS on NSCs, we used an immortalized mouse neuroectodermal stem cell line, NE-4C. We found that Jmjd2b, histone-3 lysine-9 di-/tri-methyl (H3K9me2/3) demethylase, is functional following LPS treatment and is crucial in multiple signaling pathways and biological processes. The global gene expression levels were detected in Jmjd2b-knockdown (kd) NE-4C cells and in LPS-stimulated Jmjd2b-kd NE-4C cells using an Affymetrix GeneChip® Mouse Gene 1.0 ST Array. In addition, the datasets were analyzed using MetaCore Pathway Analysis (GeneGo). The attenuation of Jmjd2b in NE-4C cells significantly affected the p65, iNOS, Bcl2, and TGF-β expression levels and had downstream effects on related signaling pathways. In addition, chromatin immunoprecipitation revealed that Jmjd2b-kd could inhibit the Notch1, IL-1β, and IL-2 genes by recruiting repressive H3K9me3 to their promoters. Moreover, this study highlights Jmjd2b role in LPS-mediated inflammation, which suggests an epigenetic regulation in NE-4C cells. Finally, this study establishes novel Jmjd2b targets that potentiate a biological rationale involving Jmjd2b in NSC inflammation.


Molecular Immunology | 2013

Proteomic changes induced by histone demethylase JMJD3 in TNF alpha-treated human monocytic (THP-1) cells.

Amitabh Das; Nando Dulal Das; Kyoung Hwa Jung; Ji Hyun Park; Hyung Tae Lee; Dalmuri Han; Mi Ran Choi; Sung Chul Kang; Young Gyu Chai

JMJD3, a Jumonji C family histone demethylase, plays an important role in the regulation of inflammation induced by the transcription factor nuclear factor-kappa B (NF-κB) in response to various stimuli. JMJD3 is a histone-3 lysine-27 trimethylation (H3K27me3) demethylase, a histone mark associated with transcriptional repression and activation of a diverse set of genes. The present study assessed stable JMJD3 knockdown (KD)-dependent proteomic profiling in human leukemia monocyte (THP-1) cells to analyze the JMJD3-mediated differential changes of marker expression in inflammatory cells. To analyze the protein expression profile of tumor necrosis factor-alpha (TNF-α)-stimulated JMJD3-kd THP-1 cells, we employed matrix-assisted-laser-desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF MS). Additionally, Ingenuity Pathways Analysis (IPA) was applied to establish the molecular networks. A comparative proteomic profile was determined in TNF-α-treated both of JMJD3-kd THP-1 cells and THP-1 scrambled (sc) cells. The expression of tripartite motif protein (TRIM5), glutathione peroxidase (GPx), glia maturation factor-γ (GMFG), caspase recruitment domain family, member 14 (CARMA2), and dUTP pyrophosphatase were significantly down-regulated, whereas heat shock protein beta-1 (HspB1) and prohibition were significantly up-regulated in JMJD3-kd THP-1 cells. The molecular and signaling networks of the differentially expressed proteins in JMJD3-kd THP-1 cells were determined by IPA. The molecular network signatures and functional proteomics obtained in this study may facilitate the suppression of different key inflammatory regulators through JMJD3-attenuation, which would be crucial to evaluate potential therapeutic targets and to elucidate the molecular mechanism of JMJD3-kd dependent effects in THP-1 cells.


Psychiatry Investigation | 2012

Fluoxetine Increases the Expression of NCAM140 and pCREB in Rat C6 Glioma Cells

Mi Ran Choi; Dong Hoon Oh; Seok Hyeon Kim; Kyoung Hwa Jung; Nando Dulal Das; Young Gyu Chai

Objective Dysfunction of neural plasticity in the brain is known to alter neural networks, resulting in depression. To understand how fluoxetine regulates molecules involved in neural plasticity, the expression levels of NCAM, NCAM140, CREB and pCREB, in rat C6 glioma cells after fluoxetine treatment were examined. Methods C6 cells were cultured after 20 min or after 6, 24 or 72 h treatments with 10 µM fluoxetine. Immunocytochemistry was used to determine the effect of fluoxetine on the expression of NCAM. Western blot analysis was used to measure the expression levels of NCAM140 and CREB and the induction of pCREB after fluoxetine treatment. Results NCAM expression following 72-h fluoxetine treatment was significantly increased around cell membranes compared to control cells. Cells treated with fluoxetine for 6 and 72 h showed a significant increase in NCAM140 expression compared to cells treated for 20 min. The level of pCREB in the cells treated with fluoxetine for 72 h not only increased more than 60%, but was also significantly different when compared with the other treatment times. The 72-h fluoxetine treatment led to the increase of NCAM140 and the phosphorylation of CREB in C6 cells. Conclusion Our findings indicate that fluoxetine treatment regulates neuronal plasticity and neurite outgrowth by phosphorylating and activating CREB via the NCAM140 homophilic interaction-induced activation of the Ras-MAPK pathway.


Neuroscience Letters | 2011

Changes in the expression of transthyretin and protein kinase Cγ genes in the prefrontal cortex in response to naltrexone

Jaehak Yu; Debasish Halder; Mi Na Baek; Nando Dulal Das; Mi Ran Choi; Dong Yul Oh; Ihn Geun Choi; Kyoung Hwa Jung; Young Gyu Chai

Naltrexone, an opioid receptor antagonist, has been approved for clinical use in the treatment of alcohol dependence. In the present study, we examined the underlying mechanisms of naltrexone by investigating the pharmacogenomic variations in the brain regions associated with alcohol consumption. A complementary DNA microarray analysis was used to profile gene expression changes in the hippocampus and prefrontal cortex (PFC) of C57BL/6 mice injected with naltrexone following ethanol treatment. Intraperitoneal administration of 200μl (16mg/kg) of naltrexone for 4 weeks caused alterations in the expression of a wide range of hippocampal (394) and PFC (566) genes in ethanol-treated mice. Ingenuity Pathway Analysis (IPA) software was used to search for biological pathways and interrelationships between gene networks in the subsets of candidate genes that were altered in the naltrexone-treated PFC and hippocampus. We found gene networks associated with cell morphology, cell death, nervous system development and function, and neurological disease. Confirmation studies using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed that the expression of transthyretin (TTR) and protein kinase C (PKC)γ were increased in the PFC but not in the hippocampus of naltrexone-treated mice. In conclusion, the present study demonstrates a pharmacogenomic response to naltrexone in the brains of ethanol-consuming mice. These findings provide a basis for conducting pharmacogenetic research on the effect of naltrexone in specific brain areas, which would enhance our understanding of the underlying causes and possible treatments of alcohol use disorders.


Scientific Reports | 2015

Dual transcriptome sequencing reveals resistance of TLR4 ligand-activated bone marrow-derived macrophages to inflammation mediated by the BET inhibitor JQ1

Amitabh Das; Jin Choul Chai; Chul-Su Yang; Young Seek Lee; Nando Dulal Das; Kyoung Hwa Jung; Young Gyu Chai

Persistent macrophage activation is associated with the expression of various pro-inflammatory genes, cytokines and chemokines, which may initiate or amplify inflammatory disorders. A novel synthetic BET inhibitor, JQ1, was proven to exert immunosuppressive activities in macrophages. However, a genome-wide search for JQ1 molecular targets has not been undertaken. The present study aimed at evaluating the anti-inflammatory function and underlying genes that are targeted by JQ1 in LPS-stimulated primary bone marrow-derived macrophages (BMDMs) using global transcriptomic RNA sequencing and quantitative real-time PCR. Among the annotated genes, transcriptional sequencing of BMDMs that were treated with JQ1 revealed a selective effect on LPS-induced gene expression in which the induction of cytokines/chemokines, interferon-stimulated genes, and prominent (transcription factors) TFs was suppressed. Additionally, we found that JQ1 reduced the expression of previously unidentified genes that are important in inflammation. Importantly, these inflammatory genes were not affected by JQ1 treatment alone. Furthermore, we confirmed that JQ1 reduced cytokines/chemokines in the supernatants of LPS treated BMDMs. Moreover, the biological pathways and gene ontology of the differentially expressed genes were determined in the JQ1 treatment of BMDMs. These unprecedented results suggest that the BET inhibitor JQ1 is a candidate for the prevention or therapeutic treatment of inflammatory disorders.


Journal of Medicinal Food | 2012

Proteomic Analysis of Terminalia chebula Extract-Dependent Changes in Human Lymphoblastic T Cell Protein Expression

Nando Dulal Das; Kyoung Hwa Jung; Ji Hyun Park; Mi Ran Choi; Hyung Tae Lee; Moo Sung Kim; Sang Rin Lee; Young Gyu Chai

Terminalia chebula is a native plant from southern Asia to southwestern China that is used in traditional medicine for the treatment of malignant tumors and diabetes. This plant also has antibacterial and immunomodulatory properties. The present study assessed T. chebula extract-dependent protein expression changes in Jurkat cells. Matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry and Ingenuity Pathways Analysis (IPA) were performed to assess protein expression and networks, respectively. A comparative proteomic profile was determined in T. chebula extract (50 μg/mL)-treated and control cells; the expressions of β-tubulin, ring finger and CHY zinc finger domain containing 1, and insulin-like growth factor 1 receptor kinase were significantly down-regulated in T. chebula extract-treated Jurkat cells. Moreover, the molecular basis for the T. chebula extract-dependent protein expression changes in Jurkat cells was determined by IPA. Treatment with the T. chebula extract significantly inhibited nuclear factor-κB activity and affected the proteomic profile of Jurkat cells. The molecular network signatures and functional proteomics obtained in this study may facilitate the evaluation of potential antitumor therapeutic targets and elucidate the molecular mechanism of T. chebula extract-dependent effects in Jurkat cells.

Collaboration


Dive into the Nando Dulal Das's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge