20 results on '"Kausik Bishayee"'
Search Results
2. Dysfunctional Glucose Metabolism in Alzheimer's Disease Onset and Potential Pharmacological Interventions
- Author
-
Vijay Kumar, So-Hyeon Kim, and Kausik Bishayee
- Subjects
Organic Chemistry ,Brain ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Glucose ,Diabetes Mellitus, Type 2 ,Alzheimer Disease ,Humans ,Physical and Theoretical Chemistry ,Insulin Resistance ,Molecular Biology ,Spectroscopy - Abstract
Alzheimer’s disease (AD) is the most common age-related dementia. The alteration in metabolic characteristics determines the prognosis. Patients at risk show reduced glucose uptake in the brain. Additionally, type 2 diabetes mellitus increases the risk of AD with increasing age. Therefore, changes in glucose uptake in the cerebral cortex may predict the histopathological diagnosis of AD. The shifts in glucose uptake and metabolism, insulin resistance, oxidative stress, and abnormal autophagy advance the pathogenesis of AD syndrome. Here, we summarize the role of altered glucose metabolism in type 2 diabetes for AD prognosis. Additionally, we discuss diagnosis and potential pharmacological interventions for glucose metabolism defects in AD to encourage the development of novel therapeutic methods.
- Published
- 2022
3. RNA binding protein HuD promotes autophagy and tumor stress survival by suppressing mTORC1 activity and augmenting ARL6IP1 levels
- Author
-
Kausik Bishayee, Khadija Habib, Uddin Md. Nazim, Jieun Kang, Aniko Szabo, Sung-Oh Huh, and Ali Sadra
- Subjects
Male ,Cancer Research ,Cancer cell survival ,Research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Membrane Proteins ,Mice, Nude ,RNA-Binding Proteins ,ELAV-Like Protein 4 ,Mechanistic Target of Rapamycin Complex 1 ,Transfection ,ELAVL4 ,Mice ,Oncology ,Autophagy ,Animals ,Humans ,ARL6IP1 ,mTORC1 ,RC254-282 ,Adaptor Proteins, Signal Transducing ,GRB-10 - Abstract
Background Neuronal-origin HuD (ELAVL4) is an RNA binding protein overexpressed in neuroblastoma (NB) and certain other cancers. The RNA targets of this RNA binding protein in neuroblastoma cells and their role in promoting cancer survival have been unexplored. In the study of modulators of mTORC1 activity under the conditions of optimal cell growth and starvation, the role of HuD and its two substrates were studied. Methods RNA immunoprecipitation/sequencing (RIP-SEQ) coupled with quantitative real-time PCR were used to identify substrates of HuD in NB cells. Validation of the two RNA targets of HuD was via reverse capture of HuD by synthetic RNA oligoes from cell lysates and binding of RNA to recombinant forms of HuD in the cell and outside of the cell. Further analysis was via RNA transcriptome analysis of HuD silencing in the test cells. Results In response to stress, HuD was found to dampen mTORC1 activity and allow the cell to upregulate its autophagy levels by suppressing mTORC1 activity. Among mRNA substrates regulated cell-wide by HuD, GRB-10 and ARL6IP1 were found to carry out critical functions for survival of the cells under stress. GRB-10 was involved in blocking mTORC1 activity by disrupting Raptor-mTOR kinase interaction. Reduced mTORC1 activity allowed lifting of autophagy levels in the cells required for increased survival. In addition, ARL6IP1, an apoptotic regulator in the ER membrane, was found to promote cell survival by negative regulation of apoptosis. As a therapeutic target, knockdown of HuD in two xenograft models of NB led to a block in tumor growth, confirming its importance for viability of the tumor cells. Cell-wide RNA messages of these two HuD substrates and HuD and mTORC1 marker of activity significantly correlated in NB patient populations and in mouse xenografts. Conclusions HuD is seen as a novel means of promoting stress survival in this cancer type by downregulating mTORC1 activity and negatively regulating apoptosis.
- Published
- 2022
4. Targeting the Difficult-to-Drug CD71 and MYCN with Gambogic Acid and Vorinostat in a Class of Neuroblastomas
- Author
-
Kausik Bishayee, Vinay Dubey, Ali Sadra, and Sung-Oh Huh
- Subjects
Drug ,0301 basic medicine ,Physiology ,Xanthones ,media_common.quotation_subject ,Transferrin receptor ,lcsh:Physiology ,lcsh:Biochemistry ,Neuroblastoma ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,0302 clinical medicine ,Antigens, CD ,Antineoplastic Combined Chemotherapy Protocols ,Receptors, Transferrin ,medicine ,Humans ,lcsh:QD415-436 ,RNA, Neoplasm ,Viability assay ,neoplasms ,Vorinostat ,media_common ,N-Myc Proto-Oncogene Protein ,lcsh:QP1-981 ,Caspase 3 ,Chemistry ,General Neuroscience ,HEK 293 cells ,Cancer ,Transfection ,medicine.disease ,Class (biology) ,MicroRNAs ,HEK293 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Gambogic acid ,medicine.drug - Abstract
Background/aims Although neuroblastoma is a heterogeneous cancer, a substantial portion overexpresses CD71 (transferrin receptor 1) and MYCN. This study provides a mechanistically driven rationale for a combination therapy targeting neuroblastomas that doubly overexpress or have amplified CD71 and MYCN. For this subset, CD71 was targeted by its natural ligand, gambogic acid (GA), and MYCN was targeted with an HDAC inhibitor, vorinostat. A combination of GA and vorinostat was then tested for efficacy in cancer and non-cancer cells. Methods Microarray analysis of cohorts of neuroblastoma patients indicated a subset of neuroblastomas overexpressing both CD71 and MYCN. The viability with proliferation changes were measured by MTT and colony formation assays in neuroblastoma cells. Transfection with CD71 or MYCN along with quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to detect expression changes. For pathway analysis, gene ontology (GO) and Protein-protein interaction analyses were performed to evaluate the potential mechanisms of GA and vorinostat in treated cells. Results For both GA and vorinostat, their pathways were explored for specificity and dependence on their targets for efficacy. For GA-treated cells, the viability/proliferation loss due to GA was dependent on the expression of CD71 and involved activation of caspase-3 and degradation of EGFR. It relied on the JNK-IRE1-mTORC1 pathway. The drug vorinostat also reduced cell viability/proliferation in the treated cells and this was dependent on the presence of MYCN as MYCN siRNA transfection led to a blunting of vorinostat efficacy and conversely, MYCN overexpression improved the vorinostat potency in those cells. Vorinostat inhibition of MYCN led to an increase of the pro-apoptotic miR183 levels and this, in turn, reduced the viability/proliferation of these cells. The combination treatment with GA and vorinostat synergistically reduced cell survival in the MYCN and CD71 overexpressing tumor cells. The same treatment had no effect or minimal effect on HEK293 and HEF cells used as models of non-cancer cells. Conclusion A combination therapy with GA and vorinostat may be suitable for MYCN and CD71 overexpressing neuroblastomas.
- Published
- 2019
- Full Text
- View/download PDF
5. Reversing the HDAC-inhibitor mediated metabolic escape in MYCN-amplified neuroblastoma
- Author
-
Kausik Bishayee, Uddin Md. Nazim, Vijay Kumar, Jieun Kang, Jaebong Kim, Sung-Oh Huh, and Ali Sadra
- Subjects
Histone Deacetylase Inhibitors ,Pharmacology ,Mice ,N-Myc Proto-Oncogene Protein ,Neuroblastoma ,Cell Line, Tumor ,Animals ,Humans ,General Medicine ,Mechanistic Target of Rapamycin Complex 1 - Abstract
In MYCN-amplified neuroblastoma (NB), we noticed that the single compound treatment with the HDAC inhibitor vorinostat led to a reprogramming of the glycolytic pathway in these cells. This reprogramming was upregulation of fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS), allowing the cells to generate ATP, albeit at a reduced rate. This behavior was dependent on reduced levels of MYCN and a corresponding increase in the levels of PPARD transcription factors. By integrating metabolic and functional studies in NB cells and mouse xenografts, we demonstrate a compensatory upregulation of FAO/OXPHOS metabolism that promotes resistance to HDAC inhibitors. From the additional compounds that could reverse this metabolic reprogramming, the mTORC1 inhibitor sirolimus was selected. Besides both a block of glycolysis and OXPHOS, the HDAC/mTORC1 inhibitor combination produced significantly higher levels of reactive oxygen species (ROS) in the treated cells and in xenograft tumor samples, also a consequence of increased glycolytic block. The lead compounds were also tested for changes in the message levels of the glycolytic enzymes and their pathway activity, and HK2 and GPI glycolytic enzymes were most affected at their RNA message level. This combination was seen with no overall toxicity in treated mice in terms of weight loss or liver/kidney function.
- Published
- 2022
- Full Text
- View/download PDF
6. Azoxystrobin Impairs Neuronal Migration and Induces ROS Dependent Apoptosis in Cortical Neurons
- Author
-
Jieun Kang, Sung-Oh Huh, and Kausik Bishayee
- Subjects
Apoptosis ,Mitochondrion ,Mice ,Cell Movement ,Pregnancy ,neurotoxicity ,Biology (General) ,Spectroscopy ,Neurons ,chemistry.chemical_classification ,Kinase ,General Medicine ,primary cortical neuron ,Strobilurins ,Mitochondria ,Computer Science Applications ,Cell biology ,Chemistry ,mTORC1 activity ,Prenatal Exposure Delayed Effects ,Female ,Neurite ,QH301-705.5 ,Cell Survival ,Neurogenesis ,Neuronal Outgrowth ,in utero electroporation ,Caspase 3 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Catalysis ,Inorganic Chemistry ,medicine ,Animals ,Humans ,Autistic Disorder ,Physical and Theoretical Chemistry ,Kinase activity ,QD1-999 ,Molecular Biology ,Reactive oxygen species ,Organic Chemistry ,Neurotoxicity ,azoxystrobin ,medicine.disease ,Acetylcysteine ,Fungicides, Industrial ,Pyrimidines ,chemistry ,Reactive Oxygen Species - Abstract
Fungicides often cause genotoxic stress and neurodevelopmental disorders such as autism (ASD). Fungicide-azoxystrobin (AZOX) showed acute and chronic toxicity to various organisms, and remained a concern for ill effects in developing neurons. We evaluated the neurotoxicity of AZOX in developing mouse brains, and observed prenatal exposure to AZOX reduced neuronal viability, neurite outgrowth, and cortical migration process in developing brains. The 50% inhibitory concentration (IC50) of AZOX for acute (24 h) and chronic (7 days) exposures were 30 and 10 μM, respectively. Loss in viability was due to the accumulation of reactive oxygen species (ROS), and inhibited neurite outgrowth was due to the deactivation of mTORC1 kinase activity. Pretreatment with ROS scavenger- N-acetylcysteine (NAC) reserved the viability loss and forced activation of mTORC1 kinase revived the neurite outgrowth in AZOX treated neurons. Intra-amniotic injection of AZOX coupled with in utero electroporation of GFP-labelled plasmid in E15.5 mouse was performed and 20 mg/kg AZOX inhibited radial neuronal migration. Moreover, the accumulation of mitochondria was significantly reduced in AZOX treated primary neurons, indicative of mitochondrial deactivation and induction of apoptosis, which was quantified by Bcl2/Bax ratio and caspase 3 cleavage assay. This study elucidated the neurotoxicity of AZOX and explained the possible cure from it.
- Published
- 2021
- Full Text
- View/download PDF
7. 18α-Glycyrrhetinic acid lethality for neuroblastoma cells via de-regulating the Beclin-1/Bcl-2 complex and inducing apoptosis
- Author
-
Kausik Bishayee, Khadija Habib, Sung-Oh Huh, Md. Ataur Rahman, and Ali Sadra
- Subjects
0301 basic medicine ,Programmed cell death ,Small interfering RNA ,Cell Survival ,MAP Kinase Signaling System ,ATG5 ,Antineoplastic Agents ,Apoptosis ,Biology ,Biochemistry ,Neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Autophagy ,Animals ,Humans ,Phosphorylation ,Cytotoxicity ,Cell Shape ,Cell Size ,Membrane Potential, Mitochondrial ,Pharmacology ,Kinase ,HEK 293 cells ,Cadherins ,Rats ,Cell biology ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,030104 developmental biology ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Glycyrrhetinic Acid ,Beclin-1 ,RNA Interference ,Protein Processing, Post-Translational - Abstract
18α-Glycyrrhetinic acid (18-GA) is a known gap-junction inhibitor with demonstrated anticancer effects. However, the different modes of cell cytotoxicity for 18-GA remain to be characterized. In this study, 18-GA reduced the expression of cell-cell interaction proteins (N- and VE-cadherin), and led to a dose-dependent increase in cytotoxicity of the neuroblastoma cells tested, but was less toxic toward actively dividing human embryonic kidney cells. We found that 18-GA could induce both autophagy and apoptosis. 18-GA mediated autophagy was due to accumulation of Atg5, Atg7 and LC3II and degradation of p62. Individual siRNAs against Atg5 and Atg7 prevented autophagy and resulted in a further loss of viability with 18-GA. In addition, combination of 18-GA with autophagy inhibitor chloroquine produced a more significant cell death. This implied a pro-survival function for autophagy induction with 18-GA. 18-GA also led to the destabilization of Bcl-2/Beclin-1 interaction and cleavage of Beclin-1, a protein known to play role in apoptosis and autophagy induction. Treatment of cells by a pan-caspase inhibitor or a caspase-3 siRNA prevented a large portion of 18-GA mediated cytotoxicity, demonstrating that caspase-dependent apoptosis induction was responsible for most of the observed cytotoxicity. In terms of signaling, 18-GA led to reduced phosphorylation of all three classes of MAP kinases. Taken together, 18-GA or its pathways may lead to more effective, targeted therapeutics against neuroblastoma.
- Published
- 2016
- Full Text
- View/download PDF
8. Angelica polymorpha Maxim Induces Apoptosis of Human SH-SY5Y Neuroblastoma Cells by Regulating an Intrinsic Caspase Pathway
- Author
-
Kausik Bishayee, Sung-Oh Huh, and Md. Ataur Rahman
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Apoptosis ,Plant Roots ,Glycogen Synthase Kinase 3 ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Bax/Bcl-2 ratio ,Angelica polymorpha ,Angelica ,bcl-2-Associated X Protein ,Neurons ,Caspase 8 ,Caspase 3 ,General Medicine ,Cell biology ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Signal Transduction ,animal structures ,Cell Survival ,caspase ,DNA Fragmentation ,Biology ,Article ,Cell Line ,03 medical and health sciences ,neuroblastoma ,Bcl-2-associated X protein ,Cell Line, Tumor ,Autophagy ,Humans ,Viability assay ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Glycogen Synthase Kinase 3 beta ,Plant Extracts ,Cell Biology ,Fibroblasts ,Antineoplastic Agents, Phytogenic ,030104 developmental biology ,Cancer cell ,biology.protein ,Myeloid Cell Leukemia Sequence 1 Protein ,Proto-Oncogene Proteins c-akt - Abstract
Angelica polymorpha Maxim root extract (APRE) is a popular herbal medicine used for treating stomachache, abdominal pain, stomach ulcers, and rheumatism; however the effect of APRE on cancer cells has not yet been explored. Here, we examined APRE cytotoxicity seen on target neuroblastoma cells (NB) using cell viability assays, DAPI visualization of fragmented DNA, and Western blotting analysis of candidate signaling pathways involved in proliferation and apoptosis. We demonstrated that APRE reduced cell viability in NB to a greater extent than in fibroblast cells. In addition, we found that APRE could inhibit the three classes of MAPK proteins and could also down-regulate the PI3K/AKT/GSK-3β activity all being relevant for proliferation and survival. APRE could also up-regulate Bax expression and down-regulate Bcl-2 and Mcl-1. With APRE treatment, depolarization of mitochondria membrane potential and activation of caspase-3 was demonstrated in the SH-SY5Y cells. We could not found increased activity of death receptor and caspase-8 as markers of the extrinsic apoptosis pathway for the APRE treated cells. In presence of a caspase-3 siRNA and a pan-caspase inhibitor, APRE could not reduce the viability of NB cells to a significant degree. So we predicted that with APRE, the intrinsic pathway was solely responsible for inducing apoptosis as we also showed that the non-caspase autophagy pathway or ER stress-ROS mediated pathways were not involved. These findings demonstrate that an intrinsic mitochondria-mediated apoptosis pathway mediates the apoptotic effects of APRE on SH-SY5Y cells, and that APRE shows promise as a novel agent for neuroblastoma therapy.
- Published
- 2015
9. PLGA-Loaded Gold-Nanoparticles Precipitated with Quercetin Downregulate HDAC-Akt Activities Controlling Proliferation and Activate p53-ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells
- Author
-
Kausik Bishayee, Anisur Rahman Khuda-Bukhsh, and Sung-Oh Huh
- Subjects
Cell cycle checkpoint ,DNA damage ,Cell ,p53-ROS crosstalk ,Down-Regulation ,Histone Deacetylase 2 ,Metal Nanoparticles ,Histone Deacetylase 1 ,Biology ,Article ,Histone Deacetylases ,quercetin ,Polylactic Acid-Polyglycolic Acid Copolymer ,medicine ,histone deacetylation ,Humans ,Lactic Acid ,Molecular Biology ,Cell Proliferation ,Cyclin-dependent kinase 1 ,Cell growth ,Liver Neoplasms ,apoptosis ,Cell Biology ,General Medicine ,Hep G2 Cells ,Cell cycle ,Molecular biology ,gold-nanoparticles ,medicine.anatomical_structure ,Apoptosis ,Cancer cell ,Gold ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species ,Polyglycolic Acid ,HeLa Cells - Abstract
Controlled release of medications remains the most convenient way to deliver drugs. In this study, we precipitated gold nanoparticles with quercetin. We loaded gold-quercetin into poly(DL-lactide-co-glycolide) nanoparticles (NQ) and tested the biological activity of NQ on HepG2 hepatocarcinoma cells to acquire the sustained release property. We determined by circular dichroism spectroscopy that NQ effectively caused conformational changes in DNA and modulated different proteins related to epigenetic modifications and cell cycle control. The mitochondrial membrane potential (MMP), reactive oxygen species (ROS), cell cycle, apoptosis, DNA damage, and caspase 3 activity were analyzed by flow cytometry, and the expression profiles of different anti- and pro-apoptotic as well as epigenetic signals were studied by immunoblotting. A cytotoxicity assay indicated that NQ preferentially killed cancer cells, compared to normal cells. NQ interacted with HepG2 cell DNA and reduced histone deacetylases to control cell proliferation and arrest the cell cycle at the sub-G stage. Activities of cell cycle-related proteins, such as p21(WAF), cdk1, and pAkt, were modulated. NQ induced apoptosis in HepG2 cells by activating p53-ROS crosstalk and induces epigenetic modifications leading to inhibited proliferation and cell cycle arrest.
- Published
- 2015
10. Low doses of ethanolic extract of Boldo (Peumus boldus) can ameliorate toxicity generated by cisplatin in normal liver cells of mice in vivo and in WRL-68 cells in vitro, but not in cancer cells in vivo or in vitro
- Author
-
Anisur Rahman Khuda-Bukhsh, Jesmin Mondal, Kausik Bishayee, and Ashis Kumar Panigrahi
- Subjects
Male ,Antineoplastic Agents ,Biology ,Pharmacology ,HeLa ,Mice ,In vivo ,Neoplasms ,medicine ,Animals ,Humans ,Cytotoxicity ,Cells, Cultured ,Cisplatin ,Plant Extracts ,General Medicine ,biology.organism_classification ,Glutathione ,Apoptosis ,Peumus ,Toxicity ,Cancer cell ,Hepatocytes ,Female ,Boldo ,DNA Damage ,medicine.drug - Abstract
Objective Use of cisplatin, a conventional anticancer drug, is restricted because it generates strong hepatotoxicity by accumulating in liver. Therefore its anticancer potential can only be fully exploited if its own toxicity is considerably reduced. Towards this goal, ethanolic extract of the plant, Boldo ( Peumus boldus ), known for its antihepatotoxic effects, was used simultaneously with cisplatin, to test its ability to reduce cisplatin's cytotoxicity without affecting its anticancer potential. Methods The cytotoxicity of Boldo extract (BE) and cisplatin, administered alone and in combination, was determined in three cancer cell lines (A549, HeLa, and HepG2) and in normal liver cells (WRL-68). Drug-DNA interaction, DNA damage, cell cycle, apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP, ΔΨ) were also studied. Hepatotoxicity and antioxidant activity levels were determined by alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase and glutathione assays in mice. The cytotoxicity of related proteins was tested by Western blotting. Results Co-administration of BE and cisplatin increased viability of normal cells, but had no effect on the viability of cancer cells. Boldo protected liver from damage and normalized different antioxidant enzyme levels in vivo and also reduced ROS and re-polarized MMP in vitro. Bax and cytochrome c translocation was reduced with caspase 3 down-regulation. Further, a drug-DNA interaction study revealed that BE reduced cisplatin's DNA-binding capacity, resulting in a reduction in DNA damage. Conclusion Results indicated that a low dose of BE could be used beneficially in combination with cisplatin to reduce its toxicity without hampering cisplatin's anticancer effect. These findings signify a potential future use of BE in cancer therapy.
- Published
- 2014
- Full Text
- View/download PDF
11. Antihyperglycemic Drug Gymnema sylvestre Also Shows Anticancer Potentials in Human Melanoma A375 Cells via Reactive Oxygen Species Generation and Mitochondria-Dependent Caspase Pathway
- Author
-
Sourav Sikdar, Samrat Ghosh, Debrup Chakraborty, Avinaba Mukherjee, Kausik Bishayee, and Anisur Rahman Khuda-Bukhsh
- Subjects
Skin Neoplasms ,Cell Survival ,ICAD ,Poly ADP ribose polymerase ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Caspase 3 ,Pharmacology ,Rhodamine 123 ,chemistry.chemical_compound ,Humans ,Hypoglycemic Agents ,MTT assay ,Melanoma ,Cells, Cultured ,biology ,Plant Extracts ,Cytochrome c ,Gymnema sylvestre ,biology.organism_classification ,Molecular biology ,Mitochondria ,Complementary and alternative medicine ,Oncology ,chemistry ,Apoptosis ,biology.protein ,Reactive Oxygen Species ,Signal Transduction - Abstract
Objective. Ethanolic extract of G ymnema sylvestre (GS) leaves is used as a potent antidiabetic drug in various systems of alternative medicine, including homeopathy. The present study was aimed at examining if GS also had anticancer potentials, and if it had, to elucidate its possible mechanism of action. Methods. We initially tested possible anticancer potential of GS on A375 cells (human skin melanoma) through MTT assay and determined cytotoxicity levels in A375 and normal liver cells; we then thoroughly studied its apoptotic effects on A375 cells through protocols such as Hoechst 33258, H2DCFDA, and rhodamine 123 staining and conducted ELISA for cytochrome c, caspase 3, and PARP activity levels; we determined the mRNA level expression of cytochrome c, caspase 3, Bcl2, Bax, PARP, ICAD, and EGFR signaling genes through semiquantitative reverse transcriptase polymerase chain reaction and conducted Western blot analysis of caspase 3 and PARP. We also analyzed cell cycle events, determined reactive oxygen species accumulation, measured annexin V-FITC/PI and rhodamine 123 intensity by flow cytometry. Results. Compared with both normal liver cells and drug-untreated A375, the mortality of GS-treated A375 cells increased in a dose-dependent manner. Additionally, GS induced nuclear DNA fragmentation and showed an increased level of mRNA expression of apoptotic signal related genes cytochrome c, caspase 3, PARP, Bax, and reduced expression level of ICAD, EGFR, and the anti-apoptotic gene Bcl2. Conclusion. Overall results indicate GS to have significant anticancer effect on A375 cells apart from its reported antidiabetic effect, indicating possibility of its palliative use in patients with symptoms of both the diseases.
- Published
- 2013
- Full Text
- View/download PDF
12. [6]-Gingerol induces caspase 3 dependent apoptosis and autophagy in cancer cells: Drug–DNA interaction and expression of certain signal genes in HeLa cells
- Author
-
Samrat Ghosh, Anisur Rahman Khuda-Bukhsh, Raktim Biswas, Kausik Bishayee, Debrup Chakraborty, and Sushil Kumar Mandal
- Subjects
Programmed cell death ,Cell Survival ,Poly ADP ribose polymerase ,Catechols ,Biological Availability ,Antineoplastic Agents ,Apoptosis ,Caspase 3 ,DNA Fragmentation ,HeLa ,Materials Testing ,Autophagy ,Animals ,Humans ,Cell Nucleus ,Membrane Potential, Mitochondrial ,Pharmacology ,biology ,Cytochrome c ,DNA ,biology.organism_classification ,Caspase Inhibitors ,Molecular biology ,Cell biology ,Gene Expression Regulation, Neoplastic ,Proteolysis ,Cancer cell ,biology.protein ,Cattle ,Fatty Alcohols ,Poly(ADP-ribose) Polymerases ,HeLa Cells - Abstract
[6]-Gingerol, a pharmacologically important bioactive component of ginger, has been reported to have anti-hyperglycemic, anti-cancer and anti-oxidative properties, but mechanisms through which these are achieved are largely unclear. The present study focuses on apoptosis and autophagy, two key events of anti-cancer activity, in HeLa cells treated with [6]-gingerol. The treated cells showed several morphological changes, including externalization of phosphatidyl serine, degradation of DNA and increase in TUNEL positivity. Furthermore, there was depolarization of mitochondrial membrane potential, providing evidence of mitochondria mediated apoptosis. The expression of caspase 3 and PARP was increased in cells exposed to [6]-gingerol. Circular dichroism study for testing drug-DNA interaction with both calf thymus and nuclear DNA as target revealed that the drug had potential to bind with the nuclear DNA and induce conformational changes of DNA. The over-expression of NFkβ, AKT and Bcl2 genes in cancer cells was down-regulated by [6]-gingerol treatment. On the other hand the expression levels of TNFα, Bax and cytochrome c were enhanced in [6]-gingerol treated cells. Thus, overall results suggest that [6]-gingerol has potential to bind with DNA and induce cell death by autophagy and caspase 3 mediated apoptosis.
- Published
- 2012
- Full Text
- View/download PDF
13. Chelidonine isolated from ethanolic extract of Chelidonium majus promotes apoptosis in HeLa cells through p38-p53 and PI3K/AKT signalling pathways
- Author
-
Anisur Rahman Khuda-Bukhsh, Samrat Ghosh, Naoual Boujedaini, Debrup Chakraborty, Avijit Paul, Avinaba Mukherjee, Sourav Sikdar, and Kausik Bishayee
- Subjects
STAT3 Transcription Factor ,DNA damage ,Apoptosis ,p38 Mitogen-Activated Protein Kinases ,HeLa ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Botany ,Humans ,Chelidonium ,Fragmentation (cell biology) ,Protein kinase B ,Cell Proliferation ,Benzophenanthridines ,Membrane Potential, Mitochondrial ,biology ,Cell Cycle Checkpoints ,biology.organism_classification ,Molecular biology ,Comet assay ,Proto-Oncogene Proteins c-bcl-2 ,Complementary and alternative medicine ,chemistry ,Chelidonine ,Tumor Suppressor Protein p53 ,Proto-Oncogene Proteins c-akt ,HeLa Cells ,Signal Transduction - Abstract
Objective To evaluate the role of chelidonine isolated from ethanolic extract of Chelidonium majus in inducing apoptosis in HeLa cells and to assess the main signalling pathways involved. Methods Cells were initially treated with different concentrations of chelidonine for 48 h and the median lethal dose (LD50) value was selected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Morphological analysis of nuclear condensation and DNA damage and fragmentation were measured by 4',6-diamidino-2-phenylindole staining and comet assay. Further, reactive oxygen species (ROS) generation, cell cycle arrest and change in mitochondrial membrane potential were also examined and analyzed by flow cytometry. Evaluation of interaction of drug with CT DNA was investigated by circular dichroism (CD) spectral analysis to find any possible drug-CT DNA interaction. The mRNA and protein expressions of major signal proteins like p38, p53, protein kinase B (AKT), phosphatidylinositol 3-kinases (PI3K), Janus kinase 3 (JAK3), signal transducer and activator of transcription 3 (STAT3) and E6 and E7 oncoproteins as well as the pro-apoptotic genes and antiapoptotic genes were also estimated by reverse transcriptase-polymerase chain reaction and Western blotting. Results Based on LD(50) value (30 μg/mL) of chelidonine, three doses were selected, namely, 22.5 μg/mL (D1), 30.0 μg/mL (D2) and 37.5 μg/mL (D3). Results showed that chelidonine inhibited proliferation and induced apoptosis in HeLa cells through generation of ROS, cell cycle arrest at sub-G1 and G0/G1 stage, change in mitochondrial membrane potential and fragmentation of DNA. Results of CD spectra showed effective interaction between chelidonine and calf thymus DNA. Studies of signalling pathway revealed that chelidonine could efficiently induce apoptosis through up-regulation of expressions of p38, p53 and other pro-apoptotic genes and down-regulation of expressions of AKT, PI3K, JAK3, STAT3, E6, E7 and other antiapoptotic genes. Conclusion Chelidonine isolated from Chelidonium majus efficiently induced apoptosis in HeLa cells through possible alteration of p38-p53 and AKT/PI3 kinase signalling pathways.
- Published
- 2012
- Full Text
- View/download PDF
14. Oleanolic acid isolated from ethanolic extract of Phytolacca decandra induces apoptosis in A375 skin melanoma cells: drug-DNA interaction and signaling cascade
- Author
-
Kausik Bishayee, Samrat Ghosh, and Anisur Rahman Khuda-Bukhsh
- Subjects
Programmed cell death ,Skin Neoplasms ,Poly ADP ribose polymerase ,Antineoplastic Agents ,Apoptosis ,Biology ,chemistry.chemical_compound ,Cell Line, Tumor ,Humans ,Propidium iodide ,Viability assay ,Oleanolic Acid ,Melanoma ,Phytolacca ,Cell growth ,Plant Extracts ,General Medicine ,DNA, Neoplasm ,Cell cycle ,ErbB Receptors ,HaCaT ,chemistry ,Biochemistry ,Microscopy, Fluorescence ,Cancer research ,Phytotherapy ,Signal Transduction - Abstract
Objective Oleanolic acid (OA) has been reported to have anticancer effects, but the extent of its cytotoxicity, its ability to interact with nuclear DNA, its action against skin melanoma, as well as the molecular mechanism of its action against cell proliferation and in support of cell death are still unexplored. This led us to examine the efficacy of OA, a bioactive compound isolated from Phytolacca decandra , on these issues in the present investigation. Methods Studies related to analyses of cell viability, drug-DNA interaction, cell proliferation, cell cycle and epidermal growth factor receptor (EGFR) activity were performed. To investigate whether cells undergo apoptosis, studies like fluorescence microscopy, poly (ADP-ribose) polymerase (PARP) degradation, annexin V-fluorescein isothiocyanate/propidium iodide assay, alteration in mitochondrial membrane potential and activity of some relevant signaling proteins were performed. Results OA displayed a minimal and negligible cytotoxic effect on normal HaCaT cells (skin keratinocytes) and peripheral blood mononuclear cells but by contrast it reduced A375 cell viability significantly. OA interacted with nuclear DNA quickly after exposure. It acted as an antiproliferative agent. It suppressed EGFR activity. OA administration led the cells to mitochondria-dependent caspase 3-mediated apoptosis. Conclusion OA interacts with cellular DNA, inhibits proliferation possibly through modulating EGFR activity and induces mitochondria-dependent caspase 3-mediated apoptosis in A375 cells which would qualify it as a potent anticancer agent.
- Published
- 2014
15. Flavonol isolated from ethanolic leaf extract of Thuja occidentalis arrests the cell cycle at G2‐M and induces ROS‐independent apoptosis in A549 cells, targeting nuclear DNA
- Author
-
Sourav Sikdar, Naoual Boujedaini, Kausik Bishayee, Avinaba Mukherjee, and Anisur Rahman Khuda-Bukhsh
- Subjects
G2 Phase ,Lung Neoplasms ,Flavonols ,Cell Survival ,Poly ADP ribose polymerase ,Cell ,Apoptosis ,Gene mutation ,Biology ,Adenocarcinoma ,Mice ,Thuja ,Annexin ,Carcinoma, Non-Small-Cell Lung ,medicine ,Animals ,Humans ,A549 cell ,Dose-Response Relationship, Drug ,Plant Extracts ,Cytochrome c ,Cell Biology ,General Medicine ,Original Articles ,Cell Cycle Checkpoints ,DNA, Neoplasm ,Cell cycle ,Molecular biology ,Antineoplastic Agents, Phytogenic ,Plant Leaves ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,Reactive Oxygen Species ,Cell Division - Abstract
Objectives The K-ras gene mutation commonly found in lung adenocarcinomas contributes to their non-invasive expansion. Our main objective here was to develop a chemopreventive agent against K-ras-mutated lung adenocarcinoma cell line like-A549. Materials and methods We isolated flavonol from ethanolic leaf extract of Thuja occidentalis, and evaluated its apoptotic potentials on A549 cells. They were treated with 1–10 μg/ml of flavonol and viability was tested retaining normal lung cells L-132 as control. We performed assays such as TUNEL, annexin V, cell-cycle and mitochondrial membrane potentials, by FACS analysis. ROS-mediated oxidative stress and drug–DNA interactions were analysed along with gene expression studies for p53, Bax-Bcl2, cytochrome c, the caspase cascade genes and PARP. Results Flavonol reduced A549 cell viability in a dose- and time-dependent manner (IC50 value = 7.6 ± 0.05 μg/ml following 48 h incubation) sparing normal L-132 cells. It effected G2-M phase cell cycle arrest and apoptosis, as indicated by progressive increase in the sub-G1, annexin V and TUNEL-positive cell populations. Apoptotic effects appeared to be mitochondria-dependent, caspase-3-mediated, but ROS-independent. Analysis of circular dichroism data revealed that flavonol intercalated with nuclear DNA. In vivo studies on non small cell lung carcinoma (NSCLC)-induced mice confirmed anti-cancer potential of flavonol. Conclusion Flavonol-induced apoptosis apparently resulted from intercalation of cells' nuclear DNA. Flavonol inhibited growth of induced lung tumours in the mice, indicating its potential as an effective agent against NSCLC.
- Published
- 2013
16. Graveoline isolated from ethanolic extract of Ruta graveolens triggers apoptosis and autophagy in skin melanoma cells: a novel apoptosis-independent autophagic signaling pathway
- Author
-
Samrat, Ghosh, Kausik, Bishayee, and Anisur Rahman, Khuda-Bukhsh
- Subjects
Ruta ,Skin Neoplasms ,Plant Extracts ,Adenine ,Membrane Proteins ,Apoptosis ,Cell Line, Tumor ,Autophagy ,Humans ,Methoxsalen ,Beclin-1 ,Apoptosis Regulatory Proteins ,Reactive Oxygen Species ,Melanoma ,Signal Transduction - Abstract
Anti-cancer drugs generally kill cancer cells by apoptosis but fail to do so when they become resistant and escape apoptosis signals. But these resistant cells can still be killed by autophagy. Therefore, drugs having both apoptotic and autophagic abilities are solicited in effective cancer management. In search of such a drug, we examined the efficacy of graveoline, a bioactive compound isolated from Ruta graveolens on skin melanoma A375 cells through the use of specific signaling cascades and their inhibitors. Cytotoxicity of graveoline was tested by conducting MTT assay. Induction of autophagy and apoptosis was checked. Expression of related proteins and their localization were studied by conducting immunoblot assay and through confocal microscopy, respectively. We found graveoline-induced Beclin-1 associated autophagy in A375 cells and 3-methyladenine, an inhibitor of autophagy did not affect apoptosis. Conversely, caspase inhibitor that blocked apoptosis did not affect autophagic cell death, suggesting thereby that these two were independent events. Use of reactive oxygen species (ROS) scavengers inhibited cell death, but blocking autophagy did not affect graveoline-induced ROS generation, suggesting that ROS generation ensued autophagy. Thus, graveoline-induced both apoptotic and autophagic cell death in skin melanoma cells, a desirable quality in effective anti-cancer drug design.
- Published
- 2013
17. 5-lipoxygenase antagonist therapy: a new approach towards targeted cancer chemotherapy
- Author
-
Anisur Rahman Khuda-Bukhsh and Kausik Bishayee
- Subjects
Leukotrienes ,Biophysics ,Antineoplastic Agents ,Apoptosis ,Biochemistry ,Models, Biological ,Metastasis ,Neoplasms ,medicine ,Humans ,Epidermal growth factor receptor ,Enzyme Inhibitors ,Receptor ,Cell Proliferation ,Arachidonate 5-Lipoxygenase ,biology ,Cell growth ,Cancer ,General Medicine ,medicine.disease ,Biosynthetic Pathways ,Immunology ,Cancer cell ,Arachidonate 5-lipoxygenase ,biology.protein ,Cancer research ,Signal transduction - Abstract
Leukotrienes are the bioactive group of fatty acids and major constituents of arachidonic acid metabolism molded by the catalytic activity of 5-lipoxygenase (5-LOX). Evidence is accumulating in support of the direct involvement of 5-LOX in the progression of different types of cancer including prostate, lung, colon, and colorectal cancers. Several independent studies now support the correlation between the 5-LOX expression and cancer cell viability, proliferation, cell migration, invasion through extracellular matrix destruction, metastasis, and activation of anti-apoptotic signaling cascades. The involvement of epidermal growth factor receptor and 5-oxo-ETE receptor (OXER1) is the major talking point in the downstream of the 5-LOX pathway, which relates the cancer cells to the proliferative pathways. Antisense technology approaches and use of different kinds of blocker targeted to 5-LOX, FLAP (5-LOX-activating protein), and OXER1 have shown a greater efficiency in combating different cancer cell types. Lastly, suppression of 5-LOX activity that reduces the cell proliferation activity also induces intrinsic mitochondrial apoptotic pathway in either p53-dependent or independent manner. Pharmacological agents that specifically inhibit the LOX-mediated signaling pathways have been used during last few years to treat inflammatory diseases such as asthma and arthritis. Studies of these well-characterized agents are therefore warranted for their use as possible candidates for chemotherapeutic studies against the killer disease cancer.
- Published
- 2013
18. Homeopathic mother tincture of Phytolacca decandra induces apoptosis in skin melanoma cells by activating caspase-mediated signaling via reactive oxygen species elevation
- Author
-
Anisur Rahman Khuda-Bukhsh, Naoual Boujedaini, Avijit Paul, Kausik Bishayee, Avinaba Mukherjee, Sourav Sikdar, Samrat Ghosh, and Debrup Chakraborty
- Subjects
Necrosis ,Skin Neoplasms ,DNA damage ,Caspase 3 ,Apoptosis ,Pharmacology ,Cell Line, Tumor ,Medicine ,Humans ,Cytotoxicity ,Melanoma ,Phytolacca ,chemistry.chemical_classification ,Reactive oxygen species ,business.industry ,General Medicine ,Antineoplastic Agents, Phytogenic ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Cancer cell ,Immunology ,DNA fragmentation ,medicine.symptom ,business ,Reactive Oxygen Species ,Drugs, Chinese Herbal ,Phytotherapy ,Signal Transduction - Abstract
Objective Preventive measures against skin melanoma like chemotherapy are useful but suffer from chronic side effects and drug resistance. Ethanolic extract of Phytolacca decandra (PD), used in homeopathy for the treatment of various ailments like chronic rheumatism, regular conjunctivitis, psoriasis, and in some skin diseases was tested for its possible anticancer potential. Methods Cytotoxicity of the drug was tested by conducting 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay on both normal (peripheral blood mononuclear cells) and A375 cells. Fluorescence microscopic study of 4′,6-diamidino-2-phenylindole dihydrochloride-stained cells was conducted for DNA fragmentation assay, and changes in cellular morphology, if any, were also recorded. Lactate dehydrogenase activity assay was done to evaluate the percentages of apoptosis and necrosis. Reactive oxygen species (ROS) accumulation, if any, and expression study of apoptotic genes also were evaluated to pin-point the actual events of apoptosis. Results Results showed that PD administration caused a remarkable reduction in proliferation of A375 cells, without showing much cytotoxicity on peripheral blood mononuclear cells. Generation of ROS and DNA damage, which made the cancer cells prone to apoptosis, were found to be enhanced in PD-treated cells. These results were duly supported by the analytical data on expression of different cellular and nuclear proteins, as for example, by down-regulation of Akt and Bcl-2, up-regulation of p53, Bax and caspase 3, and an increase in number of cell deaths by apoptosis in A375 cells. Conclusion Overall results demonstrate anticancer potentials of PD on A375 cells through activation of caspase-mediated signaling and ROS generation.
- Published
- 2013
19. Quercetin induces cytochrome-c release and ROS accumulation to promote apoptosis and arrest the cell cycle in G2/M, in cervical carcinoma: signal cascade and drug-DNA interaction
- Author
-
Sushant G. Ghosh, Anisur Rahman Khuda-Bukhsh, Jesmin Mondal, Avinaba Mukherjee, Ratan Sadhukhan, and Kausik Bishayee
- Subjects
Cell Survival ,Cell ,Uterine Cervical Neoplasms ,Apoptosis ,DNA Fragmentation ,Biology ,HeLa ,Cell Movement ,medicine ,Humans ,MTT assay ,heterocyclic compounds ,Protein kinase B ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Caspase 3 ,Circular Dichroism ,Cytochromes c ,Cell Biology ,General Medicine ,DNA ,DNA, Neoplasm ,Original Articles ,Cell cycle ,biology.organism_classification ,Flow Cytometry ,Molecular biology ,Antineoplastic Agents, Phytogenic ,Cell biology ,Genes, bcl-2 ,Nucleosomes ,G2 Phase Cell Cycle Checkpoints ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,DNA fragmentation ,M Phase Cell Cycle Checkpoints ,Female ,Quercetin ,Signal transduction ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species ,Transcriptome ,HeLa Cells ,Signal Transduction - Abstract
Objectives Small aromatic compounds like flavonoids can intercalate with DNA molecules bringing about conformational changes leading to reduced replication and transcription. Here, we have examined one dietary flavonoid, quercetin (found in many fruit and vegetables), for possible anti-cancer effects, on HeLa cells originally derived from a case of human cervical cancer. Material and methods By circular dichroism spectroscopy we tested whether quercetin effectively interacted with DNA to bring about conformational changes that would strongly inhibit proliferation and migration of the HeLa cells. Cytotoxic effects of quercetin on cancer/normal cells, if any, were determined by MTT assay and such depolarization of mitochondrial membrane potential, as a consequence of quercetin treatment, and accumulation of reactive oxygen species (ROS) also were studied, by FACS analysis and expression profiles of different anti- and pro-apoptotic genes and their products were determined. Results Quercetin intercalated with calf thymus cell DNA and HeLa cell DNA and inhibition of anti-apoptotic AKT and Bcl-2 expression were observed. Levels of mitochondrial cytochrome-c were elevated and depolarization of mitochondrial membrane potential occurred with increase of ROS; upregulation of expression of p53 and caspase-3 activity were also noted. These alterations in signalling proteins and externalization of phosphotidyl serine residues were involved with initiation of apoptosis. Reduced AKT expression suggested reduction in cell proliferation and metastasis potential, with arrest of the cell cycle at G2/M. Conclusion Quercetin would have potential for use in cervical cancer chemotherapy.
- Published
- 2012
20. Diarylheptanoid-myricanone isolated from ethanolic extract of Myrica cerifera shows anticancer effects on HeLa and PC3 cell lines: signalling pathway and drug-DNA interaction
- Author
-
Asmita Samadder, Jayeeta Das, Kausik Bishayee, Ratan Sadhukhan, Anisur Rahman Khuda-Bukhsh, Sreemanti Das, and Avijit Paul
- Subjects
Male ,Cell Survival ,Apoptosis ,HeLa ,chemistry.chemical_compound ,Diarylheptanoids ,Cell Line, Tumor ,Spectroscopy, Fourier Transform Infrared ,Humans ,Propidium iodide ,biology ,Plant Extracts ,Cell growth ,Circular Dichroism ,Cell Cycle ,Diarylheptanoid ,DNA ,General Medicine ,Cell cycle ,biology.organism_classification ,Antineoplastic Agents, Phytogenic ,Molecular biology ,Cell biology ,Myrica ,chemistry ,Cell culture ,Cancer cell ,Female ,Signal Transduction - Abstract
Objective To test if myricanone (C 21 H 24 O 5 ), a cyclic diarylheptanoid, has anticancer effects on two different cancer cell lines HeLa and PC3. The present study was conducted with a note on the drug-DNA interaction and apoptotic signalling pathway. Methods Several studies like cytotoxicity, nuclear damage, annexin-V-fluorescein isothiocyanate (FITC)/propidium iodide (PI)-labelled apoptotic assay and cell cycle arrest, immunoblot and reverse transcriptase-polymerase chain reaction (RT-PCR) were used following standard protocols. Circular dichroism (CD) spectroscopy was also done to evaluate whether myricanone effectively interacted with DNA to bring about conformational changes that could strongly inhibit the cancer cell proliferation. Results Myricanone showed a greater cytotoxic effect on PC3 cells than on HeLa cells. Myricanone promoted G 0 /G 1 arrest in HeLa cells and S phase arrest in PC3 cells. Nuclear condensation and annexin V-FITC/PI studies revealed that myricanone promoted apoptotic cell death. CD spectroscopic data indicated that myricanone had an interaction with calf thymus DNA that changed DNA structural conformation. RT-PCR and immunoblot studies revealed that myricanone activated the apoptotic signalling cascades through down-regulation of transcription factors like nuclear factor-κB (NF-κB) (p65), and signal transducers and activators of transcription 3 (STAT3); cell cycle regulators like cyclin D1, and survivin and other signal proteins like Bcl-2 and up-regulation of Bax, caspase-9 and caspase-3. Conclusion Myricanone induced apoptosis in both types of cancer cells by triggering caspase activation, and suppression of cell proliferation by down-regulation of NF-κB and STAT3 signalling cascades, which makes it a suitable candidate for possible use in the formulation of therapeutic agent for combating cancer.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.