Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Article in English | MEDLINE | ID: mdl-37721630

ABSTRACT

Globally, lung cancer contributes significantly to the public health burden-associated mortality. As this form of cancer is insidious in nature, there is an inevitable diagnostic delay leading to chronic tumor development. Non-small cell lung cancer (NSCLC) constitutes 80-85% of all lung cancer cases, making this neoplasia form a prevalent subset of lung carcinoma. One of the most vital aspects for proper diagnosis, prognosis, and adequate therapy is the precise classification of non-small cell lung cancer based on biomarker expression profiling. This form of biomarker profiling has provided opportunities for improvements in patient stratification, mechanistic insights, and probable druggable targets. However, numerous patients have exhibited numerous toxic side effects, tumor relapse, and development of therapy-based chemoresistance. As a result of these exacting situations, there is a dire need for efficient and effective new cancer therapeutics. De novo drug development approach is a costly and tedious endeavor, with an increased attrition rate, attributed, in part, to toxicity-related issues. Drug repurposing, on the other hand, when combined with computer-assisted systems biology approach, provides alternatives to the discovery of new, efficacious, and safe drugs. Therefore, in this review, we focus on a comparison of the conventional therapy-based chemoresistance mechanisms with the repurposed anti-cancer drugs from three different classes-anti-parasitic, anti-depressants, and anti-psychotics for cancer treatment with a primary focus on NSCLC therapeutics. Certainly, amalgamating these novel therapeutic approaches with that of the conventional drug regimen in NSCLC-affected patients will possibly complement/synergize the existing therapeutic modalities. This approach has tremendous translational significance, since it can combat drug resistance and cytotoxicity-based side effects and provides a relatively new strategy for possible application in therapy of individuals with NSCLC.

3.
Article in English | MEDLINE | ID: mdl-35035034

ABSTRACT

The novel coronavirus strain SARS-CoV-2 is the virus responsible for the recent global health crisis, as it causes the coronavirus disease-19 (COVID-19) in humans. Due to its high rate of spreading and significant fatality rates, the situation has escalated to a pandemic, which is the cause of immense disruption in daily life. In this study, we have taken a docking-based virtual screening approach to select natural molecules (from plants) with possible therapeutic potential. For this purpose, AUTODOCK Vina-based determination of binding affinity values (blind and active-site oriented) was obtained to short-list molecules with possible inhibitory potential against the main Mpro in SARS-CoV-2 (PDB ID 6Y2F -the monomeric form). The 4 molecules selected were Chebuloside (-8.2; -8.2), Acetoside (-8.0; -8.0), Corilagin (-8.1; -7.7) and Arjunolic Acid (-8.0; -7.6) (blind and active-site oriented docking scores (Kcal/mol) in parenthesis, respectively). Further, a comparative search, with FDA-approved drugs, has shown that Ouabain was comparable to Chebuloside with a similarity score of 0.227. This in silico finding with respect to Ouabain is significant, since this polycyclic glycoside has been shown to treat COVID-19 positive patients with a cardiovascular disease. Hydrocortisone was similar to Arjunolic acid with a score of 0.539. Again, this likeness is worthy of mention, since hydrocortisone has been used earlier for the treatment of SARS-CoV1 and MERS. However, further experimentation and validation of the results, in suitable biological model systems, are necessary to gain more insight and relevance as well as provide corroborative evidence for our in-silico findings. Supplementary Information: The online version contains supplementary material available at 10.1007/s40011-021-01292-5.

4.
Chemosphere ; 267: 128923, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33190912

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) often co-exist with the other co-contaminants like antibiotics. The antibiotics can potentially modify the toxic effects of the co-contaminants like the NPs in the environment. Hence, the present study aims to understand the toxic potential of a binary mixture of tetracycline (TC) and TiO2 NPs to a model freshwater alga - Scenedesmus obliquus. Since, TiO2 NPs are known to be photo-catalytically active, non-irradiated (NI-TiO2 NPs), UVA pre-irradiated (UVA-TiO2 NPs), and UVB pre-irradiated (UVB-TiO2 NPs) TiO2 NPs was mixed separately with TC and their toxicity evaluated. It was observed that the cell viability for the three experimental groups decreased significantly (p < 0.001) with respect to the individual NPs-treated algae. Abbott's model suggested that the interaction between TC and Ni-TiO2 NPs was additive for all the concentrations of NI-TiO2 NPs tested. However, in the case of both the UV pre-irradiated NPs, the interaction was additive for the lower concentration (1.56 µM) and synergistic for both the higher concentrations (3.13, and 6.26 µM). At the concentrations tested the cell membrane damage and intracellular uptake of NPs increased significantly (p < 0.05) for the mixture in comparison with the individual NPs treated algae. This study suggested that even a non-lethal concentration of TC (EC10 = 0.135 µM) increased the toxic potential of the TiO2 NPs significantly and when this antibiotic was used in combination with the UV pre-irradiated NPs, toxicity even increased to a higher level.


Subject(s)
Metal Nanoparticles , Nanoparticles , Scenedesmus , Water Pollutants, Chemical , Anti-Bacterial Agents/toxicity , Fresh Water , Metal Nanoparticles/toxicity , Titanium/toxicity , Water Pollutants, Chemical/toxicity
5.
Trends Biotechnol ; 36(5): 476-477, 2018 05.
Article in English | MEDLINE | ID: mdl-29477234

ABSTRACT

Models of tumor heterogeneity should mimic perfusion in the tumor mass and consider cancer stem cell hierarchy and convertibility. Other important factors include epigenomic alterations, enhanced permeation and retention effects, and the reticuloendothelial system. New stimuli-responsive delivery systems, active targeting-based methods, and corona characterization strategies can deliver genes more precisely.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Drug Delivery Systems , Genetic Therapy , Humans , Nanotechnology
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 193: 523-527, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29304487

ABSTRACT

The study focused on the analysis of the nature and site of binding of Phenytoin (PHT) -(a model hydrophobic drug) with Bovine Serum Albumin (BSA) (a model protein used as a surrogate for HSA). Interactions with defined amounts of Phenytoin and BSA demonstrated a blue shift (hypsochromic -change in the microenvironment of the tryptophan residue with decrease in the polar environment and more of hydrophobicity) with respect to the albumin protein and a red shift (bathochromic -hydrophobicity and polarity related changes) in the case of the model hydrophobic drug. This shift, albeit lower in magnitude, has been substantiated by a fairly convincing, Phenytoin-mediated quenching of the endogenous fluorophore in BSA. Spectral shifts studied at varying pH, temperatures and incubation periods (at varying concentrations of PHT with a defined/constant BSA concentration) showed no significant differences (data not shown). FTIR analysis provided evidence of the interaction of PHT with BSA with a stretching vibration of 1737.86cm-1, apart from the vibrations characteristically associated with the amine and carboxyl groups respectively. Our in vitro findings were extended to molecular docking of BSA with PHT (with the different ionized forms of the drug) and the subsequent LIGPLOT-based analysis. In general, a preponderance of hydrophobic interactions was observed. These hydrophobic interactions corroborate the tryptophan-based spectral shifts and the fluorescence quenching data. These results substantiates our hitherto unreported in vitro/in silico experimental flow and provides a basis for screening other hydrophobic drugs in its class.


Subject(s)
Anticonvulsants/metabolism , Phenytoin/metabolism , Serum Albumin, Bovine/metabolism , Spectrometry, Fluorescence/methods , Spectrophotometry, Ultraviolet/methods , Animals , Anticonvulsants/chemistry , Binding Sites , Cattle , Computer Simulation , Molecular Docking Simulation , Phenytoin/chemistry , Serum Albumin, Bovine/chemistry
8.
J Cancer Res Ther ; 12(4): 1224-1233, 2016.
Article in English | MEDLINE | ID: mdl-28169232

ABSTRACT

Major histocompatibility complex Class I-related chain A/chain B (MICA/MICB) is stress-inducible, highly polymorphic ligands whose expression at the transcript level has been detected in all tissues except the central nervous system. However, their restricted protein expression is due to their regulation at the posttranslational level. Its levels are elevated in virally infected and neoplastically transformed cells. Membrane expression of this NKG2DL marks the aberrant cells for elimination by those immune effector cells that express the cognate NKG2D receptor. Among the evasion strategies developed by tumors, the metalloprotease-dependent shedding of MICA/MICB from tumors (either the free or the exosome form) can contribute to the inhibition of cytolysis by the immune effector cells (all NK cells, most NKT cells; γδ CD8+ T cells and αß CD8+ T cells, as well as some αß CD4+ T cells). There are micro-RNA clusters that regulate surface expression and shedding. Polymorphic variants can be used as susceptibility/associative markers and can also possibly be used to correlate with tumor survival as well as staging/grading of tumors. Variations in the expression level require quantification of this marker for diagnostic/prognostic and therapeutic purposes. Mechanism-based studies would provide a better tumor-specific understanding of their relative roles in the processes of tumor cell elimination versus growth and progression. Last but not least, conventional, interlaboratory validated assays (for, e.g., antibody-based methods) should be replaced by robust, reproducible, feasible biophysics-based methods using tumor biopsies. Further, correlative DNA polymorphism-based studies can be done using biological fluids (for, e.g., human saliva) that can be sampled by minimally invasive means.


Subject(s)
Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , Cell Transformation, Viral/genetics , Cell Transformation, Viral/immunology , Cytotoxicity, Immunologic , Drug Discovery , Gene Expression Regulation, Neoplastic/drug effects , Genetic Variation , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/immunology , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Protein Transport , Signal Transduction , Tumor Escape/drug effects , Tumor Escape/genetics , Tumor Escape/immunology
9.
In Vitro Cell Dev Biol Anim ; 51(10): 1072-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26427706

ABSTRACT

Recent studies have demonstrated the role of caspase-14 in terminally differentiated keratinocytes, and its expression may decrease the magnitude of tumors in the epidermis. In the present study, we assessed the potential of luteolin (LUT) to elicit the expression of caspase-14 in terminal differentiation of human keratinocytes. The semi-qualitative RT-PCR data revealed a significant level of caspase-14 expression in LUT-treated human immortalized keratinocytes (HaCaT) with respect to untreated cells. The quantitative data (ELISA) further supported the potency of LUT to induce caspase-14 expression at 3.19 ng/ml when compared to 1.29 ng/ml of vitamin D3 (positive control). Further, the enhanced expression of human involucrin gene in LUT-treated HaCaT cells confirmed its ability to drive terminal differentiation in these cells. These preliminary results provide first-hand information about the in vitro potential of LUT to elicit the expression of caspase-14, thereby inducing terminal differentiation in human keratinocytes.


Subject(s)
Caspase 14/biosynthesis , Cell Differentiation/drug effects , Epidermal Cells , Keratinocytes/cytology , Luteolin/pharmacology , Caspase 14/metabolism , Cell Line , Cholecalciferol/pharmacology , Humans , Protein Precursors/biosynthesis
10.
J Food Sci Technol ; 52(4): 2328-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25829616

ABSTRACT

Annona muricata is a naturally occurring edible plant with wide array of therapeutic potentials. In India, it has a long history of traditional use in treating various ailments. The present investigation was carried out to characterize the phytochemicals present in the methanolic and aqueous leaf extracts of A. muricata, followed by validation of its radical scavenging and DNA protection activities. The extracts were also analyzed for its total phenolic contents and subjected to HPLC analysis to determine its active metabolites. The radical scavenging activities were premeditated by various complementary assays (DRSA, FRAP and HRSA). Further, its DNA protection efficacy against H2O2 induced toxicity was evaluated using pBR322 plasmid DNA. The results revealed that the extracts were highly rich in various phytochemicals including luteolin, homoorientin, tangeretin, quercetin, daidzein, epicatechin gallate, emodin and coumaric acid. Both the extracts showed significant (p < 0.05) radical scavenging activities, while methanolic extract demonstrated improved protection against H2O2-induced DNA damage when compared to aqueous extract. A strong positive correlation was observed for the estimated total phenolic contents and radical scavenging potentials of the extracts. Further HPLC analysis of the phyto-constituents of the extracts provides a sound scientific basis for compound isolation.

11.
Gene ; 542(2): 89-97, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24698776

ABSTRACT

Endometriosis is a complex disorder of the female reproductive system where endometrial tissue embeds and grows at extrauterine location leading to inflammation and pain. Hundreds of polymorphisms in several genes have been studied as probable risk factors of this debilitating disease. Bioinformatics tools have come a long way in augmenting the search for putative functional polymorphisms in human diseases. In this study we have explored 16 genes involved in the detoxification of xenobiotic chemicals that are implicated in endometriosis by utilising publically available programs like SIFT, Polyphen, Panther, FastSNP, SNPeffect and PhosSNP. The variations among different ethnic populations of the SNPs were studied. We then calculated the extent to which bioinformatics based predictions are concurrent with real world epidemiological, genotyping studies using a set of SNPs that have been studied in endometriosis case-control studies. Our study shows that there is a significant positive correlation (r=0.569, p<0.005) between the summary of the predicted scores taken from 4 different servers and the odds ratio found from epidemiological studies. This report has identified and catalogued various deleterious SNPs that could be important in endometriosis and could aid in further analysis by in vitro and in vivo methods for the better understanding of the disease pathophysiology.


Subject(s)
Algorithms , Endometriosis/genetics , Inactivation, Metabolic/genetics , Polymorphism, Single Nucleotide , Computational Biology/methods , Computer Simulation , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Xenobiotics/metabolism
12.
Toxicol Ind Health ; 30(8): 765-76, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23081860

ABSTRACT

The aryl hydrocarbon receptor (AHR) is one of the principal xenobiotic, nuclear receptor that is responsible for the early events involved in the transcription of a complex set of genes comprising the CYP450 gene family. In the present computational study, homology modelling and molecular docking were carried out with the objective of predicting the relationship between the binding efficiency and the lipophilicity of different polychlorinated biphenyl (PCB) congeners and the AHR in silico. Homology model of the murine AHR was constructed by several automated servers and assessed by PROCHECK, ERRAT, VERIFY3D and WHAT IF. The resulting model of the AHR by MODWEB was used to carry out molecular docking of 36 PCB congeners using PatchDock server. The lipophilicity of the congeners was predicted using the XLOGP3 tool. The results suggest that the lipophilicity influences binding energy scores and is positively correlated with the same. Score and Log P were correlated with r = +0.506 at p = 0.01 level. In addition, the number of chlorine (Cl) atoms and Log P were highly correlated with r = +0.900 at p = 0.01 level. The number of Cl atoms and scores also showed a moderate positive correlation of r = +0.481 at p = 0.01 level. To the best of our knowledge, this is the first study employing PatchDock in the docking of AHR to the environmentally deleterious congeners and attempting to correlate structural features of the AHR with its biochemical properties with regards to PCBs. The result of this study are consistent with those of other computational studies reported in the previous literature that suggests that a combination of docking, scoring and ranking organic pollutants could be a possible predictive tool for investigating ligand-mediated toxicity, for their subsequent validation using wet lab-based studies.


Subject(s)
Polychlorinated Biphenyls/chemistry , Polychlorinated Biphenyls/metabolism , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/metabolism , Binding Sites , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Docking Simulation , Protein Binding , Software
13.
Sci Pharm ; 82(4): 857-72, 2014.
Article in English | MEDLINE | ID: mdl-26171330

ABSTRACT

A novel, colloidal nanogold-based drug delivery system for phenytoin, a well-known anti-epileptic drug with an enhanced efflux via P-glycoprotein, has been proposed in this study. The vital physical properties that would aid in predicting the biological interaction of this system were profiled using various techniques such as UV-Vis, DLS, and TEM in corroboration with theoretical calculations. It was significant to note that the binding of phenytoin to colloidal nanogold was strongly pH-dependent with the optimum at pH 5.5 and a consistently reproducible spectral shift. Analysis of the conjugate by FTIR revealed that the imide functional group of phenytoin mediated a dative coordinate bond to colloidal nanogold at the optimum pH. The amount of the drug bound to the gold was quantified to be 85.8±2.5% (w/v) by HPLC. Hypothetically, the surface charge of the conjugate could possibly imply charge-mediated uptake across the cell membrane. Further, the novel conjugate was screened for its cytotoxicity in two cell lines and the dosage range was identified. Subsequent development, thorough evaluations in suitable model systems, and the potential for bioimaging to track the payload would validate our hypothesis on the conjugate for better intracellular retention at the site of action, and thereby achieve the targeted delivery.

15.
Int J Pharm ; 449(1-2): 28-36, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23583708

ABSTRACT

Lipophilicity vs hydrophicility physicochemical traits are extremely important variables that are active considerations for optimizing drug delivery systems. The comparative anti-inflammatory delivery potential of dexamethasone (dex) in an encapsulation-based (liposome-lipophilic) and poly (amidoamine) (PAMAM) dendrimer prodrug conjugation-based delivery systems (hydrophilic) was performed in this work. Dendrimer prodrug conjugates were characterized by (1)H NMR. The drug encapsulation efficiency for drug in liposomes was observed to be 14.02% and this was correlated with a dose-dependent tumor necrosis factor (TNF)-α inhibition (39-57% inhibition). The biological evaluation of nanocarriers for drug was demonstrated in a standard, conventionally used in vitro cell-based system for TNF-α inhibition. This served as a comparative tool to demonstrate a quantitatively higher TNF-α inhibition (67-71.48%) produced by the dendrimer-dex drug conjugate. The structure activity relationship (dose-for-dose) was inferred by relatively lesser inhibition of TNF-α by variants of PAMAM G4 (NH2) dendrimer-dex conjugates and was compared with liposomes carrying dex. In vitro results suggest that the prodrug conjugates of PAMAM dendrimer deliver dex to be more efficient in comparison with liposome-based dex in terms of higher TNF-α inhibition. This study has implications in designing efficient prodrug nanocarrier systems for delivering dex.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dendrimers/chemistry , Dexamethasone/pharmacology , Drug Delivery Systems , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Cells, Cultured , Dexamethasone/administration & dosage , Dexamethasone/chemistry , Dose-Response Relationship, Drug , Humans , Hydrophobic and Hydrophilic Interactions , Liposomes , Prodrugs , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors
16.
Indian J Biochem Biophys ; 50(1): 7-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23617068

ABSTRACT

P-glycoprotein (P-gp), an ATP-dependant efflux pump transports a wide range of substrates across cellular membranes. Earlier studies have identified drug efflux due to the over-expression of P-gp as one of the causes for the resistance of phenytoin, an anti-epileptic drug (AED). While no clear evidence exists on the specific characteristics of phenytoin association with the human P-gp, this study employed structure-based computational approaches to identify its binding site and the underlying interactions. The identified site was validated with that of rhodamine, a widely accepted reference and an experimental probe. Further, an in silico proof-of-concept for phenytoin interactions and its decreased binding affinity with the closed-state of human P-gp model was provided in comparison with other AEDs. This is the first report to provide insights into the phenytoin binding site and possibly better explain its efflux by P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/ultrastructure , Models, Chemical , Models, Molecular , Phenytoin/chemistry , Binding Sites , Catalysis , Computer Simulation , Humans , Protein Binding , Protein Conformation
17.
Appl Biochem Biotechnol ; 169(3): 911-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292904

ABSTRACT

The objective of this paper is to compare in silico data with wet lab physicochemical properties of crude laccase enzyme isolated from Rigidoporus sp. using wheat bran as solid substrate support towards dye decolorization. Molecular docking analysis of selected nine textile and non-textile dyes were performed using laccase from Rigidoporus lignosus as reference protein. Enzyme-based remediation methodology using crude enzyme enriched from solid state fermentation was applied to screen the effect of four influencing variables such as pH, temperature, dye concentration, and incubation time toward dye decolorization. The extracellular crude enzyme decolorized 69.8 % Acid Blue 113, 45.07 % Reactive Blue 19, 36.61 % Reactive Orange 122, 30.55 % Acid Red 88, 24.59 % Direct Blue 14, 18.48 % Reactive Black B, 16.49 % Reactive Blue RGB, and 11.66 % Acid Blue 9 at 100 mg/l dye concentration at their optimal pH at room temperature under static and dark conditions after 1 h of incubation without addition of any externally added mediators. Our wet lab studies approach, barring other factors, validate in silico for screening and ranking textile dyes based on their proximity to the T1 site. We are reporting for the first time a combinatorial approach involving in silico methods and wet lab-based crude laccase-mediated dye decolorization without any external mediators.


Subject(s)
Basidiomycota/enzymology , Biodegradation, Environmental , Coloring Agents/metabolism , Laccase/metabolism , Hydrogen-Ion Concentration , Temperature
18.
EXCLI J ; 12: 462-78, 2013.
Article in English | MEDLINE | ID: mdl-26417238

ABSTRACT

The aggressive nature of estrogen receptor (ER)-negative breast cancer subtype obligates for innovative targeted therapies. The present study aimed to investigate the phytoconstituents and specific anticancer activities of Rheum emodi rhizome, a known food source used locally to treat various ailments. Petroleum ether extracts (hot [PHR] and cold [PCR]) of R. emodi, exhibited significant free radical scavenging potentials through DPPH and reducing power assays, rendering them as good sources of antioxidants. The extracts, PHR and PCR had shown significant (P < 0.05) cancer-cell-specific cytotoxicity in the assayed cells (MDA-MB-231 [breast carcinoma] and WRL-68 [non-tumoral]) at 100 µg/ml, and 50 and 100 µg/ml concentrations respectively. Extracts also induced fervent apoptosis in ER-negative cells (MDA-MB-231) compared to ER-positive subtype (MCF-7), and found to involve CPP32/caspase-3 in its apoptosis induction mechanism. Moreover, extracts had an inevitable potential to inhibit the migration of metastatic breast cancer cells (MDA-MB-231) in vitro. Further, the active principles of extracts were identified through HPLC and GC-MS analysis to reveal major polyphenolics, 4,7-Dimethyl-(octahydro)indolo[4,3-fg]quinolin-10-one, 5-Oxo-isolongifolene, Valencene-2, and other quinone, quinoline and anthraquinone derivatives. The extracts are thus good candidates to target malignant ER-negative breast cancer, and the identified phytoconstituents could be eluted in further exploratory studies for use in dietary-based anti-breast cancer therapies.

19.
Asian Pac J Cancer Prev ; 13(6): 2729-34, 2012.
Article in English | MEDLINE | ID: mdl-22938449

ABSTRACT

Despite clinical advances in anticancer therapy, there is still a need for novel anticancer metabolites, with higher efficacy and lesser side effects. Oroxylum indicum (L.) Vent. is a small tree of the Bignoniaceae family which is well known for its food and medicinal properties. In present study, the chemopreventive properties of O. indicum hot and cold non-polar extracts (petroleum ether and chloroform) were investigated with MDA- MB-231 (cancer cells) and WRL-68 (non-tumor cells) by XTT assay. All the extracts, and particularly the petroleum ether hot extract (PHO), exhibited significantly (P<0.05) higher cytotoxicity in MDA-MB-231 when compared to WRL-68 cells. PHO was then tested for apoptosis induction in estrogen receptor (ER)-negative (MDA-MB-231) and ER-positive (MCF-7) breast cancer cells by cellular DNA fragmentation ELISA, where it proved more efficient in the MDA-MB-231 cells. Further, when PHO was tested for anti-metastatic potential in a cell migration inhibition assay, it exhibited beneficial effects. Thus non-polar extracts of O. indicum (especially PHO) can effectively target ER-negative breast cancer cells to induce apoptosis, without harming normal cells by cancer-specific cytotoxicity. Hence, it could be considered as an extract with candidate precursors to possibly harness or alleviate ER-negative breast cancer progression even in advanced stages of malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bignoniaceae , Breast Neoplasms/drug therapy , Phytotherapy , Plant Extracts/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Chemoprevention , Female , Humans , Receptors, Estrogen/metabolism
20.
Asian Pac J Cancer Prev ; 13(5): 2015-20, 2012.
Article in English | MEDLINE | ID: mdl-22901164

ABSTRACT

Oleanolic acid (OA) is a naturally occurring triterpenoid in food materials and is a component of the leaves and roots of Olea europaea, Viscum album L., Aralia chinensis L. and more than 120 other plant species. There are several reports validating its antitumor activity against different cancer cells apart from its hepatoprotective activity. However, antitumor activity against skin cancer has not been studied well thus far. Hence the present study of effects of OA against HaCaT (immortalized keratinocyte) cells--a cell-based epithelial model system for toxicity/ethnopharmacology-based studies--was conducted. Radical scavenging activity (DPPH·) and FRAP were determined spectrophotometrically. Proliferation was assessed by XTT assay at 24, 48 and 72 hrs with exposure to various concentrations (12.5-200 µM) of OA. Apoptotic induction potential of OA was demonstrated using a cellular DNA fragmentation ELISA method. Morphological studies were also carried out to elucidate its antitumor potential. The results revealed that OA induces apoptosis by altering cellular morphology as well as DNA integrity in HaCaT cells in a dose-dependent manner, with comparatively low cytotoxicity. The moderate toxicity observed in HaCaT cells, with induction of apoptosis, possibly suggests greater involvement of programmed-cell death-mediated mechanisms. We conclude that OA has relatively low toxicity and has the potential to induce apoptosis in HaCaT cells and hence provides a substantial and sound scientific basis for further validation studies.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Design , Keratinocytes/drug effects , Oleanolic Acid/pharmacology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescence Recovery After Photobleaching , Humans , Keratinocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...