Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 22(9): 1135-1153, 2023 05.
Article in English | MEDLINE | ID: mdl-36945177

ABSTRACT

Gliomas are brain tumors associated with high morbidity, relapse and lethality despite improvement in therapeutic regimes. The hypoxic tumor microenvironment is a key feature associated with such poor outcomes in gliomas. The Hypoxia Inducible Factor (HIF) family of transcription factors are master regulators of cellular proliferation, high metabolic rates and angiogenesis via aberrant expression of downstream genes. Recent studies have implicated long non-coding RNAs (lncRNAs) as potential prognostic and diagnostic biomarkers. In this study, identification of hypoxia regulated lncRNA with a bioinformatic pipeline consisting of a newly developed tool "GenOx" was utilized for the identification of Hypoxia Response Element (HRE) and Hypoxia Ancillary Sequence (HAS) motifs in the promoter regions of lncRNAs. This was coupled with molecular, functional and interactome-based analyses of these hypoxia-relevant lncRNAs in primary tumors and cell-line models. We report on the identification of novel hypoxia regulated lncRNAs SNHG12, CASC7 and MF12-AS1. A strong association of RNA splicing mechanisms was observed with enriched lncRNAs. Several lncRNAs have emerged as prognostic biomarkers, of which TP53TG1 and SNHG1 were identified as highly relevant lncRNAs in glioma progression and validated in hypoxia cultured cells. Significantly, we determined that SNHG1 expression in tumor (vs. normal) is different from glioma stem cells, GSC (vs. tumors) and in hypoxia (vs. normoxia), positioning downregulation of SNHG1 to be associated with worsened prognosis.


Subject(s)
Glioma , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Down-Regulation , Glioma/pathology , Hypoxia/genetics , Biomarkers , Gene Expression Regulation, Neoplastic , Tumor Microenvironment
2.
Biochim Biophys Acta Mol Cell Res ; 1870(2): 119302, 2023 02.
Article in English | MEDLINE | ID: mdl-35649481

ABSTRACT

The question of how lung cancer progresses in never-smokers remains largely unanswered. In our analysis of data from 1727 lung cancer patients, we observed a difference of only 47 days in the overall survival between lung adenocarcinoma patients who were smokers vis-a-vis never-smokers - the disease has a poor prognosis irrespective of the smoking status, or gender. We have investigated the possible collaboration between the nAChR and hypoxia signaling pathway to explicate a mechanism of disease progression in never-smokers using patient-derived tumor cells. We found a previously unidentified increase in both acetylcholine and nAChR-α7 levels in non-small cell lung cancer cells in hypoxia. A similar increase in ubiquitously expressed nAChR-α7 transcripts was also observed in other cancer lines and primary tumor tissues. A direct binding of HIF-1α with the hypoxia-response element (HRE) present at -48 position preceding the transcriptional start site in nAChR-α7 promoter region was established. Crucially, the increased acetylcholine levels in hypoxia drove a feedback loop via modulation of PI3K/AKT pathway to stabilize HIF-1α in hypoxia. Further, hypoxia-mediated metastasis and induction of HIF-1α in these cells was significantly reversed by bungarotoxin, an antagonist of nAChR-α7. The nAChR-AKT-HIF network needs to be further investigated to conclusively prove its mechanism and to explore its therapeutic potential. Our study gives a plausible explanation for the equally worse prognosis of lung adenocarcinoma in never-smokers wherein the nAChR signaling is enhanced in hypoxia by acetylcholine in the absence of nicotine.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Receptors, Nicotinic , Humans , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Nicotine/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Smokers , Acetylcholine , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Cell Line, Tumor , Adenocarcinoma of Lung/genetics , Signal Transduction
3.
Asian Pac J Cancer Prev ; 23(10): 3553-3561, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36308382

ABSTRACT

OBJECTIVE: Receptor tyrosine kinases (RTK) are relevant therapeutic targets in the treatment of lung cancer. Germline susceptibility variants that influence these RTKs may provide new insights into their regulation.  rs9387478 is located in the genomic interval between two RTK-genes ROS1/DCBLD1, of which ROS1 alterations are implicated in lung carcinogenesis and treatment response while the latter remains poorly understood. MATERIALS AND METHODS: Venous blood was drawn from 100 control and 231 case subjects. Genotype was scored by restriction fragment length polymorphism (RFLP), PCR amplification followed by HindIII digestion. Logistic regression was applied to compare the association between variables. Survival curve was plotted to draw a correlation between the genotype and overall survival. Also, eQTL and chromatin state changes were analyzed and correlated with the survival of patients using available datasets. RESULTS: In our population smoking correlated significantly with lung cancer [OR= 2.607] with the presence of the minor allele 'A' enhancing the nicotine dependence [CA (OR=3.23)]. Individuals with homozygous risk allele 'A' had a higher chance of developing lung cancer [OR=2.65] than individuals with CA/CC implying a recessive model of association. Patients with CC/CA genotype had better overall survival than patients with AA genotype [161 days/142 days vs 54 days, p=0.005]. The homozygous risk allele was significantly associated with increased DCBLD1 and ROS1 expression in lung cancer, with enriched active histone marks due to the polymorphism. Interestingly, increased DCBLD1 expression was associated with poor outcomes in lung cancer. CONCLUSION: Overall, our study provides strong evidence that rs9387478 is significantly associated with both nicotine dependence and lung cancer in our North Indian cohort. The association of the SNP with prognostic genes, DCBLD1 and ROS1 make rs9387478 a promising prognostic marker in the North Indian population. The results obtained are significant, however, the study needs to be performed in a larger sample size.


Subject(s)
Lung Neoplasms , Tobacco Use Disorder , Humans , Case-Control Studies , Genetic Predisposition to Disease , Genotype , Lung , Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics
4.
Drug Dev Res ; 83(2): 525-543, 2022 04.
Article in English | MEDLINE | ID: mdl-34569640

ABSTRACT

New candidates of imidazo[1,2-a]pyridine were designed by combining 2-amino pyridine, TOSMIC and various assorted aldehydes to explore their antioxidant and antifungal potential. The design of these derivatives was based on utilizing the antifungal potential of azoles and TOSMIC moiety. These derivatives were synthesized by adopting multi-component reaction methodology, as it serves as a rapid and efficient tool to target structurally diverse heterocyclic compounds in quantitative yield. The resulting imidazo[1,2-a]pyridine derivatives were structurally verified by 1 HNMR, 13 CNMR, HRMS, and HPLC. The compounds were analyzed for their antioxidant and fluorescent properties and it was observed that compound 15 depicted highest potential. The compounds were evaluated for their antifungal potential to highlight their medical application in the area of Invasive Fungal Infections (IFI). Compound 12 gave the highest antifungal inhibition against Aspergillus fumigatus 3007 and Candida albicans 3018. To elucidate the antifungal mechanism, confocal images of treated fungi were analyzed, which depicted porous nature of fungal membrane. Estimation of fungal membrane sterols by UPLC indicated decrease in ergosterol component of fungal membrane. In silico studies further corroborated with the in vitro results as docking studies depicted interaction of synthesized heterocyclic compounds with amino acids present in the active site of target enzyme (lanosterol 14 alpha demethylase). Absorption, distribution, metabolism, and excretion (ADME) analysis was indicative of drug-likeliness of the synthesized compounds.


Subject(s)
Antifungal Agents , Antioxidants , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Cyanides , Microbial Sensitivity Tests , Molecular Docking Simulation , Pyridines/pharmacology
5.
3 Biotech ; 11(10): 451, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34631352

ABSTRACT

Investigating the therapeutic and prognostic potential of genes in the heterogeneous hypoxic niche of glioblastoma. We have analyzed RNA expression of U87MG cells cultured in hypoxia compared to normoxia. Common differentially expressed genes (DEGs) from GSE45301 and GSE18494 and their functional enrichment was performed using MetaScape and PANTHER. Hub genes and their ontology were identified using MCode cytoHubba and ClueGO and validated with GlioVis, Oncomine, HPA and PrognoScan. Using the GEO2R analysis of GSE45301 and GSE18494 datasets, we have found a total of 246 common DEGs (180 upregulated and 66 downregulated) and identified 2 significant modules involved in ribosome biogenesis and TNF signaling. Meta-analysis of key genes of each module in cytoHubba identified 17 hub genes (ATF3, BYSL, DUSP1, EGFR, JUN, ETS1, LYAR, NIP7, NOLC1, NOP2, NOP56, PNO1, RRS1, TNFAIP3, TNFRSF1B, UTP15, VEGFA). Of the 17 hub genes, ATF3, BYSL, EGFR, JUN, NIP7, NOLC1, PNO1, RRS1, TNFAIP3 and VEGFA were identified as hypoxia signatures associated with poor prognosis in Glioma. Ribosome biogenesis emerged as a vital contender of possible therapeutic potential with BYSL, NIP7, NOLC1, PNO1 and RRS1 showing prognostic value. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s13205-021-02987-2.

6.
Cell Physiol Biochem ; 54(4): 748-766, 2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32809300

ABSTRACT

BACKGROUND/AIMS: The hypoxic microenvironment in NSCLC has been widely accepted as a contributor to both therapeutic resistance and tumor progression. In this study, we have explored Allicin, a key organosulfur compound present in garlic for its previously unreported effectiveness in the heterogeneous hypoxic tumor microenvironment of NSCLC. METHODS: The effect of Allicin on the viability of NSCLC cells was determined by MTT assay. To determine the migration rate of treated cells compared to the control, scratch and transwell migration assays were performed. Flowcytometry was done to explore cell cycle distribution, apoptosis and ROS production in cells. Fluorescence microscopy was used to examine autophagy and DNA damage in cells. Dot blot was done to check genome wide methylation. RNA expression was detected by RT-PCR and protein expression by western blotting. RESULTS: Allicin significantly decreases cell viability, proliferation and migration of NSCLC cells in both normoxia and hypoxia. It elicits both apoptosis and autophagy pathway in A549 cells by ROS accumulation and facilitating S/G2-M phase arrest in both normoxia as well as hypoxia. We suggest that ROS/MAPK and ROS/JNK signaling pathway together govern the cytotoxic effect of allicin in NSCLC cells. Notably, allicin suppresses the expression of HIF-1α and HIF-2α in hypoxic cells, pointing towards a mechanism of its effectiveness in hypoxia. A long term passive demethylation was observed, with decreased mC and no change in TET expression, thereby ruling out active demethylation by allicin. Furthermore, allicin synergistically enhances growth inhibitory activity of low dose cisplatin to effectively overcome hypoxia induced cisplatin resistance in A549 cells. CONCLUSION: Altogether, our results elucidate a potential use of allicin in sensitizing hypoxic and chemoresistant NSCLC to cisplatin-based chemotherapy and provide new, affordable therapeutic strategy with reduced side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Hypoxia/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Sulfinic Acids/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disulfides , Drug Synergism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System/genetics , Methylation , Reactive Oxygen Species/metabolism , S Phase Cell Cycle Checkpoints/drug effects
7.
Anal Biochem ; 557: 111-119, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30048629

ABSTRACT

A label-free electrochemical immunosensor is fabricated using high quality single-walled carbon nanotube for early detection of leukemia cells. It is based on P-glycoprotein (P-gp) expression level detection; by effective surface immune-complex formation with the monoclonal anti-P-glycoprotein antibodies bound to an epoxy modified nanotube surface. The expression level of P-gp on the leukemia cell surface detected by cyclic voltammetry is in good agreement with immunofluorescence microscopy studies. The proposed biosensor could be used for the detection of P-gp expressing cells within a linear range of 1.5 × 103 cells/mL - 1.5 × 107 cells/mL where lowest detection limit is found to be 19 cells/mL. A calibration plot of peak current v/s the logarithm of concentration of leukemia K562 cells is found linear with a regression coefficient of 0.935. This strategy promises high sensitivity, low-cost, fast, and repeatable recognition of cancer cells. The immunosensor was stable for three weeks and showed good precision with the relative standard deviation (RSD) of 3.57% and 2.12% assayed at the cell concentrations of 1.5 × 103 and 1.5 × 105 cells mL-1 respectively. The proposed single-wall carbon nanotube based immunosensor showed better analytical performance in comparison to similar leukemia electrochemical sensors reported.


Subject(s)
Biosensing Techniques/methods , Electrochemical Techniques/instrumentation , Electrochemical Techniques/methods , Immunoassay/instrumentation , Immunoassay/methods , Leukemia/diagnosis , Nanotubes, Carbon/chemistry , Biosensing Techniques/instrumentation , Humans , K562 Cells
8.
Cancer Res ; 78(11): 2978-2989, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29535220

ABSTRACT

Inhibin is a heterodimeric TGFß family ligand that is expressed in many cancers and is a selective biomarker for ovarian cancers; however, its tumor-specific functions remain unknown. Here, we demonstrate that the α subunit of inhibin (INHA), which is critical for the functionality of dimeric inhibin A/B, correlates with microvessel density in human ovarian tissues and is predictive of poor clinical outcomes in multiple cancers. We demonstrate that inhibin-regulated angiogenesis is necessary for metastasis. Although inhibin had no direct impact on tumor cell signaling, both tumor cell-derived and recombinant inhibin elicit a strong paracrine response from endothelial cells by triggering SMAD1/5 activation and angiogenesis in vitro and in vivo Inhibin-induced angiogenesis was abrogated via anti-inhibin α antibodies. The endothelial-specific TGFß receptor complex comprising ALK1 and endoglin was a crucial mediator of inhibin signaling, offering a molecular mechanism for inhibin-mediated angiogenesis. These results are the first to define a role for inhibin in tumor metastasis and vascularization and offer an antibody-based approach for targeting inhibin therapeutically.Significance: Inhibin is a predictor of poor patient survival in multiple cancers and is a potential target for antiangiogenic therapies. Cancer Res; 78(11); 2978-89. ©2018 AACR.


Subject(s)
Inhibins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis/pathology , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Activin Receptors, Type II/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
9.
Asian Pac J Cancer Prev ; 18(11): 3073-3079, 2017 11 26.
Article in English | MEDLINE | ID: mdl-29172281

ABSTRACT

Background: The 15q24-25 loci contain genes (CHRNA5 and CHRNA3) encoding nicotinic acetylcholine receptor subunits. We here determined for the first time the association of genetic variants rs16969968 and rs3743074 in CHRNA5 and CHRNA3, respectively, on nicotine dependence and lung cancer risk in a North Indian population by a case-control approach. Methods: Venous blood samples were obtained from 324 participants (108 lung cancer patients and 216 healthy individuals). DNA was extracted and PCR amplified with primers flanking the SNPs rs16969968 and rs3743074. Amplicons were subjected to sequencing and logistic regression was used to analyze association between variables. Results: The risk variant SNP rs16969968 in both heterozygous and homozygous forms appeared to exert a significant effect on nicotine dependence [GA (OR=2.77) and AA (OR=2.53)]. As expected, smoking was strongly associated with lung cancer (OR= 2.62). Risk allele rs16969968 in CHRNA5 also showed a significant association with increased lung cancer risk in our cohort, alone (OR= 4.99) and with smoking as a co-variable (OR= 4.28). Comparison of our analysis with other populations suggested that individuals with rs16969968 risk allele in the Indian population are more susceptible to lung cancer. Conclusion: Overall, the results strongly indicated that, in our cohort North Indian population, the genetic variant rs16969968, but not rs3743074, is significantly associated with both nicotine dependence and increased risk of lung cancer. While the results are significant, there is further need to increase the sample size and improve precision of our risk prediction.

10.
Stem Cells ; 35(6): 1468-1478, 2017 06.
Article in English | MEDLINE | ID: mdl-28376560

ABSTRACT

Activation of pluripotency regulatory circuit is an important event in solid tumor progression and the hypoxic microenvironment is known to enhance the stemness feature of some cells. The distinct population of cancer stem cells (CSCs)/tumor initiating cells exist in a niche and augment invasion, metastasis, and drug resistance. Previously, studies have reported global hypomethylation and site-specific aberrant methylation in gliomas along with other epigenetic modifications as important contributors to genomic instability during glioma progression. Here, we have demonstrated the role of hypoxia-mediated epigenetic modifications in regulating expression of core pluripotency factors, OCT4 and NANOG, in glioma cells. We observe hypoxia-mediated induction of demethylases, ten-eleven-translocation (TET) 1 and 3, but not TET2 in our cell-line model. Immunoprecipitation studies reveal active demethylation and direct binding of TET1 and 3 at the Oct4 and Nanog regulatory regions. Tet1 and 3 silencing assays further confirmed induction of the pluripotency pathway involving Oct4, Nanog, and Stat3, by these paralogues, although with varying degrees. Knockdown of Tet1 and Tet3 inhibited the formation of neurospheres in hypoxic conditions. We observed independent roles of TET1 and TET3 in differentially regulating pluripotency and differentiation associated genes in hypoxia. Overall, this study demonstrates an active demethylation in hypoxia by TET1 and 3 as a mechanism of Oct4 and Nanog overexpression thus contributing to the formation of CSCs in gliomas. Stem Cells 2017;35:1468-1478.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epigenesis, Genetic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Differentiation/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , DNA Demethylation , Dioxygenases/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genetic Loci , Glioma/pathology , Humans , Mixed Function Oxygenases/metabolism , Models, Biological , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Spheroids, Cellular/metabolism
11.
Oncotarget ; 7(44): 71841-71855, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27708247

ABSTRACT

Non-small cell lung cancer (NSCLC), comprising 85% of lung cancer cases, has been associated with resistance to chemo/radiotherapy. The hypoxic tumor micro-environment, where insufficient vasculature results in poor drug penetrance and sub-optimal chemotherapy in the tumor interiors contributes heavily to this resistance. Additionally, epigenetic changes in tumorigenic cells also change their response to different forms of therapy. In our study, we have investigated the effectiveness of a combination of cisplatin with scriptaid [a pan-Histone Deacetylase inhibitor (HDACi)] in a model that mimics the tumor microenvironment of hypoxia and sub-lethal chemotherapy. Scriptaid synergistically increases the efficacy of cisplatin in normoxia as well as hypoxia, accompanied with reduced metastasis and enhanced DNA damage. Addition of scriptaid also overcomes the cisplatin resistance exhibited in lung cancer cells with stabilized hypoxia inducible factor 1 (HIF1)-α (mutant) and mutant p53. Molecular studies showed that the combination treatment increased apoptotic cell death in both normoxia and hypoxia with a dual role of p38MAPK. Together, our results suggest that the combination of low dose cisplatin and scriptaid is cytotoxic to NSCLC lines, can overcome hypoxia induced resistance and mutant p53- induced instability often associated with this cancer, and has the potential to be an effective therapeutic modality.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Count , Cisplatin/pharmacology , Genes, p53 , Histone Deacetylase Inhibitors/immunology , Hydroxylamines/pharmacology , Lung Neoplasms/drug therapy , Mutation , Quinolines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cisplatin/administration & dosage , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxylamines/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Lung Neoplasms/genetics , Quinolines/administration & dosage , Tumor Microenvironment
12.
Chem Biol Interact ; 254: 221-30, 2016 Jul 25.
Article in English | MEDLINE | ID: mdl-27270449

ABSTRACT

BACKGROUND: The transition metal oxide nanoparticles are in focus for their anti-cancer potential. In this study we have synthesized and characterized CuO, NiO and Fe2O3 nanoparticles and, investigated their cytotoxic potential in the heterogeneous tumour microenvironment. METHODS: Nanoparticles were synthesized by aqueous precipitation method and characterized with UV-Visible spectrophotometer, Fourier transform infrared spectroscopy (FTIR), Scanning electron microscopy (SEM) and X-ray diffraction (XRD). Cell viability of lung cancer cells (A549) grown in normoxia (18%O2) and hypoxia (1%O2) was determined for all nanoparticles. The mechanism of cell death was assessed by nuclear morphological analysis, flow cytometry analysis and western blotting. Generation of intracellular ROS in treated cells and its contribution to cell viability was determined. RESULTS: The synthesized metal oxide nanoparticles were successfully characterized with SEM, spectroscopy and X-ray diffraction patterns. Cell viability of lung cancer cells was compromised in both normoxia and hypoxia. ROS generation was shown to contribute to cellular toxicity in CuO, but not NiO and Fe2O3. CONCLUSION: We have shown the therapeutic potential of CuO, NiO and Fe2O3 nanoparticles in non small cell lung cancer cells cultured in hypoxia, a relevant feature of solid tumors along with normoxia. The newly synthesized nanoparticles showed efficacy in both conditions. GENERAL SIGNIFICANCE: Hypoxia drives metabolic alterations and epigenetic modifications in the tumor microenvironment. By using conditions that mimic tumour microenvironment, this study expands the possibility of using metal oxide nanoparticles as a therapeutic agent for lung cancer treatment.


Subject(s)
Apoptosis/drug effects , Metal Nanoparticles , Oxides/pharmacology , Transition Elements/pharmacology , Tumor Microenvironment , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Survival/drug effects , Flow Cytometry , Histones/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microscopy, Electron, Scanning , Poly (ADP-Ribose) Polymerase-1/metabolism , Reactive Oxygen Species/metabolism , Spectroscopy, Fourier Transform Infrared , Surface Plasmon Resonance , Thermogravimetry , Transition Elements/chemistry , X-Ray Diffraction
13.
Expert Rev Neurother ; 15(8): 935-46, 2015.
Article in English | MEDLINE | ID: mdl-26118735

ABSTRACT

Glioblastomas are the most common form of brain tumor with a very dismal prognosis. While a standard treatment regimen of surgery followed by chemo/radiotherapy is currently used, this has only marginally improved the survival time of patients with little benefit on tumor recurrence. Although many molecular targets have already been identified and tested in clinical trials, very few are approved for use in clinics. Efforts are ongoing to target newer molecules that could be used for drug development. This review provides up-to-date information on the drugs and their molecular targets, which are currently in different stages of clinical trials. Since multiple signaling pathways are deregulated, it appears that the use of combination drugs along with personalized targeting approach would provide better therapy in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Combined Modality Therapy , Humans
14.
Biochim Biophys Acta ; 1840(1): 350-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24041991

ABSTRACT

BACKGROUND: Allicin has received much attention due to its anti-proliferative activity and not-well elucidated underlying mechanism of action. This work focuses towards determining the cellular toxicity of allicin and understanding its interaction with nucleic acid at molecular level. METHODS: MTT assay was used to assess the cell viability of A549 lung cancer cells against allicin. Fourier transform infrared (FTIR) and UV-visible spectroscopy were used to study the binding parameters of nucleic acid-allicin interaction. RESULTS: Allicin inhibits the proliferation of cancer cells in a concentration dependent manner. FTIR spectroscopy exhibited that allicin binds preferentially to minor groove of DNA via thymine base. Analysis of tRNA allicin complex has also revealed that allicin binds primarily through nitrogenous bases. Some amount of external binding with phosphate backbone was also observed for both DNA and RNA. UV visible spectra of both DNA allicin and RNA allicin complexes showed hypochromic shift with an estimated binding constant of 1.2×10(4)M(-1) for DNA and 1.06×10(3)M(-1)for RNA binding. No major transition from the B-form of DNA and A-form of RNA is observed after their interaction with allicin. CONCLUSIONS: The results demonstrated that allicin treatment inhibited the proliferation of A549 cells in a dose-dependent manner. Biophysical outcomes are suggestive of base binding and helix contraction of nucleic acid structure upon binding with allicin. GENERAL SIGNIFICANCE: The results describe cytotoxic potential of allicin and its binding properties with cellular nucleic acid, which could be helpful in deciphering the complete mechanism of cell death exerted by allicin.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , DNA/metabolism , Lung Neoplasms/pathology , Sulfinic Acids/pharmacology , Antioxidants/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Survival/drug effects , Disulfides , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Tumor Cells, Cultured
15.
Cancer Genet Cytogenet ; 198(2): 126-34, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20362227

ABSTRACT

There are few reports of loss of heterozygosity (LOH) of 1p and 19q in astrocytic tumors, especially glioblastoma multiforme (GBM). We evaluated 1p and 19q (either or both) heterozygosity status in 71 astrocytomas, including 6 pediatric cases: 20 diffuse astrocytomas (DA), 9 anaplastic astrocytomas (AA), and 42 GBM. In the GBMs, p53 protein expression was assessed by immunohistochemistry and epidermal growth factor receptor (EGFR) gene amplification by fluorescence in situ hybridization; TP53 sequencing was done in 15 of the GBMs. In adults, LOH of 1p or 19q was detected in 16% of DAs and 50% of GBMs; none of the AAs showed this alteration. In GBMs, LOH of 19q was most common (26%), followed by combined 1p and 19q LOH (13%) and 1p LOH (10%). Pediatric GBMs also harbored isolated 1p and 19q LOH (50%). Notably, LOH of 1p or 19q LOH was more frequent in p53 immunopositive secondary GBMs (61%) than in primary GBMs (17%). This suggests that LOH of 1p and 19q may be acquired during progression to secondary GBMs. Thus, 1p and 19q LOH can occur in astrocytic tumors, most commonly in secondary GBMs without morphological correlation with an oligodendroglial histology. The clinical significance of recognition of this subset of GBMs is based on several recent reports of association with better prognosis, although long-term follow-up studies are required.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , Gene Amplification , Genes, erbB-1 , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Child , Female , Gene Amplification/physiology , Heterozygote , Humans , India , Loss of Heterozygosity , Male , Middle Aged , Tumor Suppressor Protein p53/metabolism , Young Adult
16.
J Cell Mol Med ; 14(11): 2646-54, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19508390

ABSTRACT

Hypoxia is an integral part of tumorigenesis and contributes extensively to the neoplastic phenotype including drug resistance and genomic instability. It has also been reported that hypoxia results in global demethylation. Because a majority of the cytosine-phosphate-guanine (CpG) islands are found within the repeat elements of DNA, and are usually methylated under normoxic conditions, we suggested that retrotransposable Alu or short interspersed nuclear elements (SINEs) which show altered methylation and associated changes of gene expression during hypoxia, could be associated with genomic instability. U87MG glioblastoma cells were cultured in 0.1% O2 for 6 weeks and compared with cells cultured in 21% O2 for the same duration. Real-time PCR analysis showed a significant increase in SINE and reverse transcriptase coding long interspersed nuclear element (LINE) transcripts during hypoxia. Sequencing of bisulphite treated DNA as well as the Combined Bisulfite Restriction Analysis (COBRA) assay showed that the SINE loci studied underwent significant hypomethylation though there was patchy hypermethylation at a few sites. The inter-alu PCR profile of DNA from cells cultured under 6-week hypoxia, its 4-week revert back to normoxia and 6-week normoxia showed several changes in the band pattern indicating increased alu mediated genomic alteration. Our results show that aberrant methylation leading to increased transcription of SINE and reverse transcriptase associated LINE elements could lead to increased genomic instability in hypoxia. This might be a cause of genetic heterogeneity in tumours especially in variegated hypoxic environment and lead to a development of foci of more aggressive tumour cells.


Subject(s)
Alu Elements/genetics , DNA Methylation , Genomic Instability , Hypoxia/genetics , Apoptosis , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Caspases/metabolism , Cell Proliferation , CpG Islands/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Hypoxia/pathology , Long Interspersed Nucleotide Elements/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Short Interspersed Nucleotide Elements/genetics , Transcription, Genetic , Tumor Cells, Cultured
17.
BMC Cancer ; 9: 5, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19126244

ABSTRACT

BACKGROUND: We had earlier used the comparison of RAPD (Random Amplification of Polymorphic DNA) DNA fingerprinting profiles of tumor and corresponding normal DNA to identify genetic alterations in primary human glial tumors. This has the advantage that DNA fingerprinting identifies the genetic alterations in a manner not biased for locus. METHODS: In this study we used RAPD-PCR to identify novel genomic alterations in the astrocytic tumors of WHO grade II (Low Grade Diffuse Astrocytoma) and WHO Grade IV (Glioblastoma Multiforme). Loss of heterozygosity (LOH) of the altered region was studied by microsatellite and Single Nucleotide Polymorphism (SNP) markers. Expression study of the gene identified at the altered locus was done by semi-quantitative reverse-transcriptase-PCR (RT-PCR). RESULTS: Bands consistently altered in the RAPD profile of tumor DNA in a significant proportion of tumors were identified. One such 500 bp band, that was absent in the RAPD profile of 33% (4/12) of the grade II astrocytic tumors, was selected for further study. Its sequence corresponded with a region of FAT, a putative tumor suppressor gene initially identified in Drosophila. Fifty percent of a set of 40 tumors, both grade II and IV, were shown to have Loss of Heterozygosity (LOH) at this locus by microsatellite (intragenic) and by SNP markers. Semi-quantitative RT-PCR showed low FAT mRNA levels in a major subset of tumors. CONCLUSION: These results point to a role of the FAT in astrocytic tumorigenesis and demonstrate the use of RAPD analysis in identifying specific alterations in astrocytic tumors.


Subject(s)
Astrocytoma/genetics , Cadherins/genetics , Central Nervous System Neoplasms/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity , Astrocytoma/pathology , Binding Sites , Central Nervous System Neoplasms/pathology , DNA Fingerprinting/methods , DNA Primers , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Polymorphism, Single Nucleotide , Random Amplified Polymorphic DNA Technique/methods , Reverse Transcriptase Polymerase Chain Reaction
18.
J Neurooncol ; 81(3): 249-55, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17019533

ABSTRACT

The mismatch repair genes, hMLH1 (3p22) and hMSH2 (2p21), are commonly associated with accumulation of mutations and microsatellite instability. However, the status of their gene loci itself is often not addressed. In astrocytic tumors, the heterozygosity status of these genes with reference to tumor grade has not yet been determined. We have analyzed the heterozygosity status and locus specific instability in 43 glial tumors comprising 22 low grades diffuses astrocytoma (WHO Grade II, DA) and 21 glioblastoma multiforme (Grade IV GBM) using 10 microsatellite markers at 2p and 3p to elucidate the involvement of these loci in astrocytic tumorigenesis. We observed a significantly higher loss of heterozygosity (LOH) in 3p markers encompassing the hMLH1 gene locus in DA when compared to GBM (P = 0.008). In DA, while the frequency of LOH was observed to be higher in markers close to the hMLH1 gene ( approximately 40%), locus specific microsatellite instability (LSI) was higher ( approximately 30%) in markers localizing further to the gene. The frequency of LOH at markers on 2p, near the hMSH2 gene was, however, similar in DA and GBM (P = 0.451). Our results suggest that in the astrocytic tumorigenesis, LOH at the hMLH1 gene locus is an early event in tumorigenesis. However, the mismatch repair protein expression may be regulated by other cellular factors.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Carrier Proteins/genetics , Loss of Heterozygosity , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing , Astrocytoma/pathology , Brain Neoplasms/pathology , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 3/genetics , DNA Primers , Humans , Microsatellite Repeats , MutL Protein Homolog 1 , Polymerase Chain Reaction
19.
Int J Cancer ; 117(4): 683-9, 2005 Nov 20.
Article in English | MEDLINE | ID: mdl-15912534

ABSTRACT

Increased genomic instability contributes to higher frequency of secondary drug resistance and neoplastic progression in tumors as well as in cells exposed to sub-lethal concentrations of chemotherapeutic agents. We have used PCR based DNA fingerprinting techniques of randomly amplified polymorphic DNA (RAPD) and inter-alu PCR to study this phenomenon in the tumor genome. The choice of the primer, either random (for RAPD) or specific (inter-alu PCR) can determine the nature of alterations being assessed. We have compared the inter-alu PCR and RAPD profiles of U87MG glioblastoma cells exposed to sequentially increasing low doses of cisplatin for 24 passages to that of untreated controls. Inter-alu PCR, with 2 primers, demonstrated a number of alterations in the treated cells, in the form of loss / gain and changes in the intensity of bands. No changes were observed by RAPD analysis with 5 primers, however, indicating a preferential increase in the alu mediated recombination frequency in the treated cells (p = 1.866 x 10(-4)). The number of changes observed with respect to the corresponding leucocyte DNA in the inter-alu PCR profile of 26 primary tumors (Grade II = 13; Grade IV = 13), resected before chemotherapy, for the 2 inter-alu primers was very small. We present a novel application of the inter-alu PCR in detecting alterations in long term cultured cells at low dose exposure to a chemotherapeutic agent. Our results suggest that alu mediated recombination may be important in cells exposed to sub-lethal doses of cisplatin but not in the genesis of primary glioma.


Subject(s)
Alu Elements , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Cisplatin/pharmacology , Glioma/genetics , Polymerase Chain Reaction/methods , Brain Neoplasms/pathology , Cell Line, Tumor , Cloning, Molecular , Dose-Response Relationship, Drug , Glioma/pathology , Humans
20.
Cancer Gene Ther ; 11(8): 525-31, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15167899

ABSTRACT

Most gene therapy strategies related to p53 concentrate on the restoration of the activity of mutant p53, as several observations indicate that tumors and cell lines having the mutant gene are resistant to chemotherapy. However, as there is also some evidence to the contrary, we studied the relationship of the p53 status to the cellular response of glioma cells that were exposed to cisplatin. At a concentration of 2.5 microg/ml (which is about half the peak pharmacological blood level reached during chemotherapy), U373MG glioma cells, which had a mutant p53 gene, were more sensitive to the drug as compared to U87MG glioma cells (with normal p53). The U373MG cells responded with apoptosis while U87MG cells responded with a G2-M arrest. In U87MG cells, blocking the p53 response by antisense oligonucleotides also sensitized the cells to 2.5 microg/ml cisplatin, and shifted the cellular response from arrest to caspase 3-mediated apoptosis. A sensitive, p53-independent, mechanism for chemotherapy-induced apoptosis suggests that, in some cases, p53 abrogation by gene therapy or small molecule-based strategies could be a viable therapeutic strategy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Glioma/drug therapy , Oligoribonucleotides, Antisense/therapeutic use , Tumor Suppressor Protein p53/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Drug Resistance, Neoplasm , G2 Phase/drug effects , Genes, p53 , Glioma/genetics , Humans , Mutation/genetics , Oligoribonucleotides, Antisense/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...