Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Cell Biochem Funct ; 42(7): e4113, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39223765

ABSTRACT

Due to their exceptional physicochemical features, green synthesized silver nanoparticles (AgNPs) have been of considerable interest in cancer treatment. In the present study, for the first time, we aimed to green synthesize AgNPs from Euphorbia retusa and explore their anticancer potential on human breast cancer (MCF-7) cells. First, the green synthesized AgNPs (EU-AgNPs) were well characterized by UV-visible spectroscopy, Fourier transmission infrared (FTIR) spectrum, XRD, scanning and transmission electron microscopy (SEM and TEM), and EDX techniques. The characterization data exhibited that EU-AgNPs were spherical in shape and crystalline in nature with an average size of 17.8 nm. FTIR results established the presence of active metabolites in EU-AgNPs. Second, the anticancer effect of EU-AgNPs was evaluated against MCF-7 cells by MTT and neutral red uptake (NRU) assays. Moreover, morphological changes, ROS production, MMP, and apoptotic marker genes were also studied upon exposure to cytotoxic doses of EU-AgNPs. Our results showed that EU-AgNPs induce cytotoxicity in a concentration-dependent manner, with an IC50 value of 40 µg/mL. Morphological changes in MCF-7 cells exposed to EU-AgNPs also confirm their cytotoxic effects. Increased ROS and decreased MMP levels revealed that EU-AgNPs induced oxidative stress and mitochondrial membrane dysfunction. Moreover, ROS-mediated apoptosis was confirmed by elevated levels of proapoptotic marker genes (p53, Bax, caspase-3, and caspase-9) and reduced levels of an antiapoptotic gene (Bcl-2). Altogether, these findings suggested that EU-AgNPs could induce potential anticancer effects through ROS-mediated apoptosis in MCF-7 cells.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Euphorbia , Metal Nanoparticles , Plant Extracts , Silver , MCF-7 Cells , Euphorbia/chemistry , Silver/chemistry , Silver/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Reactive Oxygen Species , Spectrum Analysis , Plant Extracts/pharmacology , Breast Neoplasms/drug therapy , Spectroscopy, Fourier Transform Infrared , Microscopy, Electron , Ascorbic Acid/pharmacology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Apoptosis , Real-Time Polymerase Chain Reaction , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
2.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125590

ABSTRACT

Ras-related Rap1A GTPase is implicated in pancreas ß-cell insulin secretion and is stimulated by the cAMP sensor Epac2, a guanine exchange factor and activator of Rap1 GTPase. In this study, we examined the differential proteomic profiles of pancreata from C57BL/6 Rap1A-deficient (Null) and control wild-type (WT) mice with nanoLC-ESI-MS/MS to assess targets of Rap1A potentially involved in insulin regulation. We identified 77 overlapping identifier proteins in both groups, with 8 distinct identifier proteins in Null versus 56 distinct identifier proteins in WT mice pancreata. Functional enrichment analysis showed four of the eight Null unique proteins, ERO1-like protein ß (Ero1lß), triosephosphate isomerase (TP1), 14-3-3 protein γ, and kallikrein-1, were exclusively involved in insulin biogenesis, with roles in insulin metabolism. Specifically, the mRNA expression of Ero1lß and TP1 was significantly (p < 0.05) increased in Null versus WT pancreata. Rap1A deficiency significantly affected glucose tolerance during the first 15-30 min of glucose challenge but showed no impact on insulin sensitivity. Ex vivo glucose-stimulated insulin secretion (GSIS) studies on isolated Null islets showed significantly impaired GSIS. Furthermore, in GSIS-impaired islets, the cAMP-Epac2-Rap1A pathway was significantly compromised compared to the WT. Altogether, these studies underscore an essential role of Rap1A GTPase in pancreas physiological function.


Subject(s)
Insulin , Mice, Inbred C57BL , Pancreas , Proteomics , Signal Transduction , rap1 GTP-Binding Proteins , Animals , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics , Mice , Proteomics/methods , Insulin/metabolism , Pancreas/metabolism , Insulin-Secreting Cells/metabolism , Mice, Knockout , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Insulin Secretion , Male , Glucose/metabolism
3.
Tissue Cell ; 90: 102519, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39141932

ABSTRACT

Ethanol is a well-known hepatotoxic agent and date fruits have been associated with their biological actions. In current study, we have investigated the hepatoprotective potential of DFE on ethanol-induced cellular damages in human hepatoma (HepG2) cells. The hepatoprotective potential was assessed by exposing the HepG2 cells to non-toxic concentrations (15, 30, and 60 µg/mL) of DFE for 24 h; then toxic concentration (500 µM) of ethanol. Our results demonstrated that pretreatment with DFE significantly prohibited ethanol-induced hepatotoxicity in HepG2 cells. We observed that DFE treatment increased cell viability, reduced LDH leakage, restored cellular morphology, and inhibited caspase-3 enzyme activity in a dose dependent way, induced by ethanol. Further DFE was also effective in restoring the LPO, GSH, and catalase levels towards normal altered by ethanol. Our results also revealed that ethanol-induced ROS generation was significantly inhibited by DFE. The ethanol-induced mRNA expression of apoptotic related genes (p53, caspase-3, caspase-7, Bax, and Bcl-2) were also normalized by pretreatment with DFE. The findings from this study indicated that DFE can significantly protect HepG2 cells against ethanol-induced hepatotoxicity. Our study also provides scientific validation for the traditional use of DFE, aiming to understand its hepatoprotective potential. Altogether, to the best of our knowledge, this is the first study demonstrated that ethanol-induced hepatotoxicity can be prohibited by the DFE. Thus, DFE has a potential application in nutraceuticals as a therapeutic agent to prevent liver diseases.


Subject(s)
Apoptosis , Ethanol , Fruit , Liver Neoplasms , Phoeniceae , Plant Extracts , Humans , Hep G2 Cells , Apoptosis/drug effects , Ethanol/toxicity , Plant Extracts/pharmacology , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Phoeniceae/chemistry , Fruit/chemistry , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Reactive Oxygen Species/metabolism , Protective Agents/pharmacology , Cell Survival/drug effects , Caspase 3/metabolism
4.
Int J Mol Sci ; 24(24)2023 Dec 16.
Article in English | MEDLINE | ID: mdl-38139390

ABSTRACT

The G protein-coupled α2-adrenoceptor subtype C (abbreviated α2C-AR) has been implicated in peripheral vascular conditions and diseases such as cold feet-hands, Raynaud's phenomenon, and scleroderma, contributing to morbidity and mortality. Microvascular α2C-adrenoceptors are expressed in specialized smooth muscle cells and mediate constriction under physiological conditions and the occlusion of blood supply involving vasospastic episodes and tissue damage under pathological conditions. A crucial step for receptor biological activity is the cell surface trafficking of intracellular receptors, triggered by cAMP-Epac-Rap1A GTPase signaling, which involves protein-protein association with the actin-binding protein filamin-2, mediated by critical amino acid residues in the last 14 amino acids of the receptor carboxyl (C)-terminus. This study assessed the role of the C-terminus in Rap1A GTPase coupled receptor trafficking by domain-swapping studies using recombinant tagged receptors in transient co-transfections and compared with wild-type receptors using immunofluorescence microscopy. We further tested the biological relevance of the α2C-AR C-terminus, when introduced as competitor peptides, to selectively inhibit intracellular α2C-AR surface translocation in transfected as well as in microvascular smooth muscle cells expressing endogenous receptors. These studies contribute to establishing proof of principle to target intracellular α2C-adrenoceptors to reduce biological activity, which in clinical conditions can be a target for therapy.


Subject(s)
Myocytes, Smooth Muscle , Peptides , Receptors, Adrenergic, alpha-2 , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Peptides/metabolism , Peptides/pharmacology , Receptors, Adrenergic/metabolism , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Signal Transduction/physiology
5.
JACC Case Rep ; 22: 101951, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37790774

ABSTRACT

Electrocardiogram changes during stress tests are well standardized and understood. We present and explain a reversible QRS morphology change at peak exercise previously unreported. (Level of Difficulty: Intermediate.).

6.
Molecules ; 28(17)2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37687120

ABSTRACT

We investigated the anticancer mechanism of a chloroform extract of marine sponge (Haliclona fascigera) (sample C) in human breast adenocarcinoma (MCF-7) cells. Viability analysis using MTT and neutral red uptake (NRU) assays showed that sample C exposure decreased the proliferation of cells. Flow cytometric data exhibited reactive oxygen species (ROS), nitric oxide (NO), dysfunction of mitochondrial potential, and apoptosis in sample C-treated MCF-7 cells. A qPCR array of sample C-treated MCF-7 cells showed crosstalk between different pathways of apoptosis, especially BIRC5, BCL2L2, and TNFRSF1A genes. Immunofluorescence analysis affirmed the localization of p53, bax, bcl2, MAPKPK2, PARP-1, and caspase-3 proteins in exposed cells. Bioassay-guided fractionation of sample C revealed Neviotin A as the most active compound triggering maximum cell death in MCF-7, indicating its pharmacological potency for the development of a drug for the treatment of human breast cancer.


Subject(s)
Gene Expression Profiling , Transcriptome , Humans , MCF-7 Cells , Cell Death , Apoptosis
7.
J Trace Elem Med Biol ; 80: 127302, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37734210

ABSTRACT

BACKGROUND: Nanotechnology and material science have developed enormously fast in recent years. Due to their excellent magnetic properties, iron oxide nanoparticles (IONPs) have been broadly applied in the field of bioengineering and biomedical. Thus, it is important to evaluate the safety issues and health effects of these nanomaterials. The present investigation was aimed to evaluate the adverse effects of IONPs on human umbilical vein endothelial cells (HUVECs). METHODS: The cytotoxic potential of IONPs was assessed by MTT and neutral red uptake (NRU) assays. The impact of IONPs on oxidative stress markers (glutathione (GSH) and lipid peroxidation (LPO)), reactive oxygen species (ROS) production, and mitochondrial membrane potential (MMP) was also examined. Furthermore, the toxic effect of IONPs was quantified by assessing DNA damage, cell cycle arrest, and apoptosis by quantitative real time PCR. RESULTS: We found that IONPs induce a dose-dependent cytotoxicity on HUVECs with IC50 value of 79.13 µg/mL. The results also displayed that IONPs induce oxidative stress, ROS production, and mitochondrial membrane dysfunction. The comet assay results exhibited IONPs induces DNA damage in HUVECs. We found significant cell cycle arrest at SubG1 phase in treated cells and consequent cell death was evidenced by microscopic analysis. Moreover, IONPs display substantial up-regulation of pro-apoptotic genes and down-regulation of anti-apoptotic gene evidenced by real time qPCR. CONCLUSION: Overall, our results clearly demonstrated that IONPs have the potential to induce cytotoxicity, DNA damage, cell cycle arrest, and apoptosis in HUVECs mediated through oxidative stress and ROS production. Thus, IONPs are cytotoxic and it should be handled with proper care.


Subject(s)
Nanoparticles , Oxidative Stress , Humans , Reactive Oxygen Species/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , DNA Damage , Cell Cycle Checkpoints , Apoptosis , Glutathione/metabolism , Magnetic Iron Oxide Nanoparticles
8.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298090

ABSTRACT

Recent studies in nanomedicine have intensively explored the prospective applications of surface-tailored graphene oxide (GO) as anticancer entity. However, the efficacy of nonfunctionalized graphene oxide nanolayers (GRO-NLs) as an anticancer agent is less explored. In this study, we report the synthesis of GRO-NLs and their in vitro anticancer potential in breast (MCF-7), colon (HT-29), and cervical (HeLa) cancer cells. GRO-NLs-treated HT-29, HeLa, and MCF-7 cells showed cytotoxicity in the MTT and NRU assays via defects in mitochondrial functions and lysosomal activity. HT-29, HeLa, and MCF-7 cells treated with GRO-NLs exhibited substantial elevations in ROS, disturbances of the mitochondrial membrane potential, an influx of Ca2+, and apoptosis. The qPCR quantification showed the upregulation of caspase 3, caspase 9, bax, and SOD1 genes in GRO-NLs-treated cells. Western blotting showed the depletion of P21, P53, and CDC25C proteins in the above cancer cell lines after GRO-NLs treatment, indicating its function as a mutagen to induce mutation in the P53 gene, thereby affecting P53 protein and downstream effectors P21 and CDC25C. In addition, there may be a mechanism other than P53 mutation that controls P53 dysfunction. We conclude that nonfunctionalized GRO-NLs exhibit prospective biomedical application as a putative anticancer entity against colon, cervical, and breast cancers.


Subject(s)
Breast Neoplasms , Tumor Suppressor Protein p53 , Humans , Female , Cell Line, Tumor , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Nanomedicine , Apoptosis , MCF-7 Cells , Colon/metabolism
9.
J Mol Recognit ; 36(7): e3021, 2023 07.
Article in English | MEDLINE | ID: mdl-37092713

ABSTRACT

Visceral leishmaniasis (VL) is caused by Leishmania donovani (Ld), and most cases occur in Brazil, East Africa, and India. The treatment for VL is limited and has many adverse effects. The development of safer and more efficacious drugs is urgently needed. Drug repurposing is one of the best processes to repurpose existing drugs. Ornithine decarboxylase (ODC) is an important target against L. donovani in the polyamine biosynthesis pathway. In this study, we have modeled the 3D structure of ODC and performed high-throughput virtual screening of 8630 ZINC database ligands against Leishmania donovani ornithine decarboxylase (Ld ODC), selecting 45 ligands based on their high binding score. It is further validated through molecular docking simulation and the selection of the top two lead molecules (ceftaroline fosamil and rimegepant) for Molecular Dynamics (MD) simulation, Density functional theory (DFT), and molecular mechanics generalized born surface area (MMGBSA) analysis. The results showed that the binding affinities of ceftaroline fosamil, and rimegepant are, respectively, -10.719 and 10.159 kcal/mol. The docking complexes of the two lead compounds, ceftaroline fosamil, and rimegepant, with the target ODC, were found stable during molecular dynamics simulations. Furthermore, the analysis of MMGBSA revealed that these compounds had a high binding free energy. The DFT analysis showed that the top lead molecules were more reactive than the standard drug (pentamidine). In-silico findings demonstrated that ceftaroline fosamil, and rimegepant might be recognized as potent antagonists against ODC for the treatment of VL.


Subject(s)
Leishmania donovani , Leishmaniasis, Visceral , Humans , Ornithine Decarboxylase Inhibitors/chemistry , Ornithine Decarboxylase Inhibitors/pharmacology , Drug Repositioning , Molecular Docking Simulation , Ornithine Decarboxylase/chemistry , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Ligands , Leishmania donovani/metabolism , Ceftaroline
10.
Toxicol In Vitro ; 85: 105460, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35998759

ABSTRACT

Nano-based products have become an apparent and effective option to treat liver cancer, which is a deadly disease, and minimize or eradicate these problems. The Core-shell ZnO microspheres composed of nanoclusters (ZnOMS-NCs) have shown that it is very worthwhile to administer the proliferation rate in HepG2 and MCF-7 cancer cells even at a very low concentration (5 µg/mL). ZnOMS-NCs were prepared through hydrothermal solution process and well characterized. The MTT assay revealed that the cytotoxic effects were dose-dependent (2.5 µg/mL-100 µg/mL) on ZnOMS-NCs. The diminished activity in cell viability induces the cytotoxicity response to the ZnOMS-NCs treatment of human cultured cells. The qPCR data showed that the cells (HepG2 and MCF-7) were exposed to ZnOMS-NCs and exhibited up-and downregulated mRNA expression of apoptotic and anti-apoptotic genes, respectively. In conclusion, flow cytometric data exhibited significant apoptosis induction in both cancer cell lines at low concentrations. The possible mechanism also describes the role of ZnOMS-NCs against cancer cells and their responses.


Subject(s)
Breast Neoplasms , Zinc Oxide , Humans , Female , Zinc Oxide/toxicity , Cysteine , Breast Neoplasms/drug therapy , MCF-7 Cells , Liver/metabolism , RNA, Messenger/metabolism , Apoptosis , Cell Proliferation
11.
J Trace Elem Med Biol ; 73: 127029, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35785590

ABSTRACT

Neodymium oxide exhibits a unique property, which facilitates and largely utilized as an industrial applications. A number of cytotoxic study is available but very limited information is available to understand their biological activity with neodymium oxide at a very low conc- entration of the material. The present work was designed to understand the cytotoxicity against liver (HepG-2) and lung (A-549) cancer cells. Initially, Neodymium oxides (Nd2O3) were prepared and characterized with various instruments. The crystallinity and morphology of Nd2O3 powder were examined with instruments such as X-Ray Diffraction (XRD), scanning electron microscope (SEM), Transmission electron microscopy (TEM), Energy Dispersive X-Ray Analysis (EDX) respectively, revealed the size of curved nanostructure are ~140 ± 2 in diameter whereas length goes upto ~700 nm with elemental composition. The cytotoxicity study was conducted with MTT, NRU assay with genotoxicity study via ROS, cell cycle and qPCR analysis. The cells cytotoxic assessment were analysed via MTT(3-(4,5-Dimethylthiazol-2-yl)- 2,5-Diphenyl tetra zolium Bromide) and Neutral Red Uptake (NRU) assay with neodymium oxide (Nd2O3), which indicates the reduction in cell viability. Additionally, cell-cycle analysis showed an increase in the apoptotic peak after a 24-h. Quantitative real-time PCR (RT-PCR) data revealed that apoptotic genes such as p53, bax, and caspase-3 were up regulated, whereas bcl-2, an anti-apoptotic gene, was down regulated; therefore, apoptosis was mediated through ROS and genotoxicity pathways. The experiments of cytotoxicity was tested and concludes that the Nd2O3 express a moderate and dose dependent effect on cancer cells. The ROS, cell cycle analysis and qPCR showed that Nd2O3 exhibit the capability to cells death via ROS generation and genotoxicity study pathways.


Subject(s)
Antineoplastic Agents , Nanostructures , Neoplasms , Antineoplastic Agents/pharmacology , Apoptosis , Humans , Neodymium/pharmacology , Oxides/pharmacology , Reactive Oxygen Species/metabolism
12.
J Cancer Res Ther ; 18(1): 109-118, 2022.
Article in English | MEDLINE | ID: mdl-35381771

ABSTRACT

Purpose: Lung cancer mostly diagnosed at advanced inoperable stages; thereby, the chemo-, radiation-, targeted or immune-therapy alone or in combination remains the treatment of choice. In chemotherapy, platinum-based compounds such as cisplatin and carboplatin and third-generation drugs such as docetaxel, paclitaxel, gemcitabine, and vinorelbine are widely used. The beneficial therapeutic outcome of the chemotherapy alone or in combination with radiation (chemoradiation) and/or development of drug resistance depends on the inter-individual genetic differences. Hence, this study was carried out to find gene biomarker that could be useful in the diagnosis of the disease and to predict the outcome of chemo/chemoradiation therapy in ethnic North Indian population. Materials and Methods: In this clinical study, lung cancer (n = 52) patients from North Indian population were recruited. All the patients were treated with carboplatin target area under curve-5 in combination with third-generation drugs (gemcitabine 1.2 mg/m2; paclitaxel 175 mg/m2; and etopside 100 mg/m2) and radiation therapy. The genomic DNA was isolated from the blood sample and performed polymerase chain reaction (PCR) and PCR-restriction fragment length polymorphism. Results: We found hazard ratio to be significantly higher for XPDLys751Gln (hazard ratio [HR] =2.11, 95% confidence interval [CI]: 0.98-4.53, P = 0.056) and IL1 ß511C/T (HR = 9.9, 95% CI: 2.55-38.40, P = 0.001). GSTT1 null (HR = 0.39, 95%CI: 0.18-0.84, P = 0.017) genotype has better response to chemotherapy. Generalized multidimensional reduction model suggested that IL1RN (cross-validation consistency [CVC] =10/10, P = 0.054) and XRCC1399Gln, GSTM1 (CVC = 10/10, P = 0.001) as best predicted model in lung cancer patients to the treatment response. Conclusion: Genetic polymorphisms and single nucleotide polymorphisms in DNA repair gene (XRCC1, XPD) and drug-metabolizing gene (GSTM1 and GSTT1) could serve as genetic biomarkers in lung cancer patients treated with the above indicated chemotherapy. Based on genotype and chemotherapy treatments, the toxicity effects can be minimized, this will help in the development of personalized medicine in future with better efficacy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Pharmacogenetics , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Treatment Outcome , X-ray Repair Cross Complementing Protein 1/genetics
13.
Int J Mol Sci ; 23(7)2022 Apr 03.
Article in English | MEDLINE | ID: mdl-35409358

ABSTRACT

Tris (2-ethylhexyl) phosphate (TEHP) is an organophosphate flame retardant (OPFRs) which is extensively used as a plasticizer and has been detected in human body fluids. Contemporarily, toxicological studies on TEHP in human cells are very limited and there are few studies on its genotoxicity and cell death mechanism in human liver cells (HepG2). Herein, we find that HepG2 cells exposed to TEHP (100, 200, 400 µM) for 72 h reduced cell survival to 19.68%, 49.83%, 58.91% and 29.08%, 47.7% and 57.90%, measured by MTT and NRU assays. TEHP did not induce cytotoxicity at lower concentrations (5, 10, 25, 50 µM) after 24 h and 48 h of exposure. Flow cytometric analysis of TEHP-treated cells elevated intracellular reactive oxygen species (ROS), nitric oxide (NO), Ca++ influx and esterase levels, leading to mitochondrial dysfunction (ΔΨm). DNA damage analysis by comet assay showed 4.67, 9.35, 13.78-fold greater OTM values in TEHP (100, 200, 400 µM)-treated cells. Cell cycle analysis exhibited 23.1%, 29.6%, and 50.8% of cells in SubG1 apoptotic phase after TEHP (100, 200 and 400 µM) treatment. Immunofluorescence data affirmed the activation of P53, caspase 3 and 9 proteins in TEHP-treated cells. In qPCR array of 84 genes, HepG2 cells treated with TEHP (100 µM, 72 h) upregulated 10 genes and downregulated 4 genes belonging to a human cancer pathway. Our novel data categorically indicate that TEHP is an oxidative stressor and carcinogenic entity, which exaggerates mitochondrial functions to induce cyto- and genotoxicity and cell death, implying its hepatotoxic features.


Subject(s)
Phosphates , Transcriptome , DNA Damage , Humans , Liver , Organophosphates/toxicity , Organophosphorus Compounds/toxicity
14.
Chemosphere ; 296: 133977, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35216979

ABSTRACT

Recent reports have confirmed that tris(2-butoxyethyl) phosphate (TBEP), an organophosphorous flame retardants (OPFRs), profoundly detected in the dust from solid waste (SW), e-waste dumping sites, landfills, and wastewater treatment facilities. Herein, we evaluated the hepatotoxic and carcinogenic potential of TBEP in human liver cells (HepG2). HepG2 cells exhibited cytotoxicity after 3 days of exposure, especially at greater concentrations (100-400 µM). TBEP induced severe DNA damage and cell cycle disturbances that trigger apoptosis in HepG2. TBEP treated cells showed an elevated level of esterase, nitric oxide (NO), reactive oxygen species (ROS), and influx of Ca2+ in exposed cells. Thereby, causing oxidative stress and proliferation inhibition. TBEP exposed HepG2 cells exhibited dysfunction in mitochondrial membrane potential (ΔΨm). Immunofluorescence analysis demonstrated cytoplasmic and nucleolar localization of DNA damage (P53) and apoptotic (caspase 3 and 9) proteins in HepG2 grown in the presence of TBEP for 3 days. Within the cohort of 84 genes of cancer pathway, 10 genes were upregulated and 3 genes were downregulated. The transcriptomic and toxicological data categorically emphasize that TBEP is hepatotoxic, and act as a putative carcinogenic agent. Thereby, direct or indirect ingestion of TBEP containing dusts by workers involved in handling and disposal of SW, as well as residents living nearby the disposal areas are prone to its adverse health risks.


Subject(s)
Flame Retardants , Carcinogens/analysis , Flame Retardants/analysis , Flame Retardants/toxicity , Humans , Organophosphates/toxicity , Organophosphorus Compounds/toxicity , Phosphates/analysis , Solid Waste/analysis
15.
Biol Trace Elem Res ; 200(12): 5042-5051, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35000107

ABSTRACT

Copper oxide nanoparticles (CuONPs) are purposefully used to inhibit the growth of bacteria, algae, and fungi. Several studies on the beneficial and harmful effects of CuONPs have been conducted in vivo and in vitro, but there are a few studies that explain the toxicity of CuONPs in human airway epithelial cells (HEp-2). As a result, the purpose of this study is to look into the dose-dependent toxicity of CuONPs in HEp-2 cells. After 24 h of exposure to 1-40 µg/ml CuONPs, the MTT and neutral red assays were used to test for cytotoxicity. To determine the mechanism(s) of cytotoxicity in HEp-2 cells, additional oxidative stress assays (LPO and GSH), the amount of ROS produced, the loss of MMP, caspase enzyme activities, and apoptosis-related genes were performed using qRT-PCR. CuONPs exhibited dose-dependent cytotoxicity in HEp-2 cells, with an IC50 value of ~ 10 µg/ml. The morphology of HEp-2 cells was also altered in a dose-dependent manner. The involvement of oxidative stress in CuONP-induced cytotoxicity was demonstrated by increased LPO levels and ROS generation, as well as decreased levels of GSH and MMP. Furthermore, activated caspase enzymes and altered apoptotic genes support CuONPs' ability to induce apoptosis in HEp-2 cells. Overall, this study demonstrated that CuONPs can cause apoptosis in HEp-2 cells via oxidative stress; therefore, CuONPs may pose a risk to human health and should be handled and used with caution.


Subject(s)
Metal Nanoparticles , Nanoparticles , Caspases/metabolism , Cell Death , Copper/toxicity , Epithelial Cells/metabolism , Humans , Metal Nanoparticles/toxicity , Nanoparticles/toxicity , Neutral Red/pharmacology , Oxidative Stress , Oxides/pharmacology , Reactive Oxygen Species/metabolism
16.
Cells ; 11(2)2022 01 07.
Article in English | MEDLINE | ID: mdl-35053312

ABSTRACT

Tris(1,3-Dichloro-2-propyl)phosphate (TDCPP) is an organophosphorus flame retardant (OPFR) widely used in a variety of consumer products (plastics, furniture, paints, foams, and electronics). Scientific evidence has affirmed the toxicological effects of TDCPP in in vitro and in vivo test models; however, its genotoxicity and carcinogenic effects in human cells are still obscure. Herein, we present genotoxic and carcinogenic properties of TDCPP in human liver cells (HepG2). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and neutral red uptake (NRU) assays demonstrated survival reduction in HepG2 cells after 3 days of exposure at higher concentrations (100-400 µM) of TDCPP. Comet assay and flow cytometric cell cycle experiments showed DNA damage and apoptosis in HepG2 cells after 3 days of TDCPP exposure. TDCPP treatment incremented the intracellular reactive oxygen species (ROS), nitric oxide (NO), Ca2+ influx, and esterase level in exposed cells. HepG2 mitochondrial membrane potential (ΔΨm) significantly declined and cytoplasmic localization of P53, caspase 3, and caspase 9 increased after TDCPP exposure. qPCR array quantification of the human cancer pathway revealed the upregulation of 11 genes and downregulation of two genes in TDCPP-exposed HepG2 cells. Overall, this is the first study to explicitly validate the fact that TDCPP bears the genotoxic, hepatotoxic, and carcinogenic potential, which may jeopardize human health.


Subject(s)
Carcinogens/toxicity , Flame Retardants/toxicity , Liver/pathology , Mutagens/toxicity , Organophosphorus Compounds/toxicity , Calcium/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , DNA Damage , Esterases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver/drug effects , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects
17.
Biol Trace Elem Res ; 200(4): 1598-1607, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34131861

ABSTRACT

In this manuscript, the grown and annealed strontium-doped nickel oxide nanoparticles (SrNiONPs) were synthesized using a precipitation method with nickel nitrate and strontium nitrate as precursor agents with trisodium citrate. Various characterization techniques, including X-ray diffraction pattern (XRD), scanning electron microscopy (SEM), transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIR), UV-visible, and zeta sizer, were used to thoroughly examine the samples. The XRD pattern (21 nm) was used to calculate the size, phases, and crystallinity of the material (SrNiONPs). In addition to characterization, the material was tested for cytotoxicity in lung cancer cells (A549). The viability test in A549 cells was performed using [3-(4, 5-dimethylthiazol-2-yl)-2, 5 diphenyltetrazolium bromide] (MTT) and Neutral Red Uptake (NRU) assay with SrNiONPs concentration ranging from 1 to 100 µg/mL. According to the MTT and NRU data, the toxicity studies are dose-dependent. SrNiONPs also increased reactive oxygen species (ROS) and were involved in apoptosis (A549 cells). Furthermore, quantitative PCR (qPCR) data revealed that the mRNA levels of apoptotic genes marker like p53, bax, and caspase-3 were upregulated, whereas bcl-2, an anti-apoptotic gene, was downregulated. As a result, apoptosis was mediated by the p53, bax, caspase3, and bcl-2 pathways, implying a potential mechanism by which SrNiONPs mediate their toxicity.


Subject(s)
Lung Neoplasms , Metal Nanoparticles , Nanoparticles , A549 Cells , Apoptosis , Humans , Metal Nanoparticles/chemistry , Nickel , Spectroscopy, Fourier Transform Infrared , Strontium , X-Ray Diffraction
18.
Stud Health Technol Inform ; 280: 141-145, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34190076

ABSTRACT

Scoliosis is a 3D deformation of the spinal column, characterized by a lateral deviation of the spine, accompanied by axial rotation of the vertebrae. Adolescent Idiopathic Scoliosis (AIS), is the most common type, affecting children between ages 8 to 18 when bone growth is at its maximum rate. The selection of the most appropriate treatment options is based on the surgeon's experience. So, developing a clinically validated patient-specific model of the spine would aid surgeons in understanding AIS in early stages and propose an efficient method of treatment for the individual patient. This project steps include: Developing a clinically validated patient-specific Reduced Order Finite Element Model (ROFEM) of the spine, predicting AIS progression using data mining and proposing a method of treatment. First we implement FE synergistically with bio-mechanical information, image processing and data science techniques to improve predictive ability. Initial geometry of the spine will be extracted from the x-ray images from different planes and imported to FEM software to generate the spine model and perform analysis. A RO model is developed based on the detailed spinal FEM. Next, a neural network is used to predict the spinal curvature. The ability to predict the severity of AIS will have an immense impact on the treatment of AIS-affected children. Access to a predictive and patient-specific model will enable the physicians to have a better understanding of spinal curvature progression. Consequently, the physicians will be able to educate families, choose the most appropriate treatment option and asses for surgical intervention.


Subject(s)
Kyphosis , Scoliosis , Adolescent , Artificial Intelligence , Child , Humans , Imaging, Three-Dimensional , Rotation , Scoliosis/diagnostic imaging , Spine/diagnostic imaging
20.
Oxid Med Cell Longev ; 2021: 6695634, 2021.
Article in English | MEDLINE | ID: mdl-33574980

ABSTRACT

Rosa damascena Mill (Damask rose), belonging to the Rosaceae family, is known for medicinal purposes in traditional medicine system. However, its anticancer activity has not been studied yet in detail. Herein, we aimed to investigate the cytotoxic effects of R. damascena hexane (RA-HE) and methanolic (RA-ME) extracts against human breast (MCF-7), lung epithelial (A-549), and cervical (HeLa) cancer cells. The RA-HE and RA-ME showed more potent cytotoxic effects against HeLa cells with an IC50 of 819.6 and 198.4 µg/ml, respectively. Further, cytotoxic concentrations of most effective extract (RA-ME) were used to evaluate the mechanism of cytotoxicity involved in HeLa cells. A concentration-dependent induction of lipid peroxidation (LPO) and reduction of glutathione (GSH) in HeLa cells treated with 250-1000 µg/ml of RA-ME confirms the association of oxidative stress. We also detected a noteworthy increase in reactive oxygen species (ROS) production and a decline in mitochondrial membrane potential (MMP) level in RA-ME-exposed HeLa cells. Flow cytometric data showed a strong dose-response relationship in cell cycle analysis between subG1 phase in HeLa cells and RA-ME treatment. Similarly, a concentration-dependent increase was recorded with Annexin V assay in HeLa cells going to late apoptosis. In conclusion, our findings suggest that RA-ME-induced cytotoxicity and apoptosis in HeLa cells are mediated by oxidative stress.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Oxidative Stress , Rosa/chemistry , Uterine Cervical Neoplasms/pathology , A549 Cells , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cytokines/metabolism , Female , Glutathione/metabolism , HeLa Cells , Hexanes/chemistry , Humans , Inhibitory Concentration 50 , Lipid Peroxidation/drug effects , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Methanol/chemistry , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL