Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Molecules ; 29(7)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38611847

ABSTRACT

Central and peripheral mechanisms of the endocannabinoid system (ECS) favor energy intake and storage. The ECS, especially cannabidiol (CBD) receptors, controls adipocyte differentiation (hyperplasia) and lipid accumulation (hypertrophy) in adipose tissue. In white adipose tissue, cannabidiol receptor 1 (CB1) stimulation increases lipogenesis and inhibits lipolysis; in brown adipose tissue, it decreases mitochondrial thermogenesis and biogenesis. This study compared the availability of phytocannabinoids [CBD and Δ9-tetrahydrocannabinol (THC)] and polyunsaturated fatty acids [omega 3 (ω3) and omega 6 (ω6)] in different hemp seed oils (HSO). The study also examined the effect of HSO on adipocyte lipid accumulation by suppressing cannabinoid receptors in adipogenesis-stimulated human mesenchymal stem cells (hMSCs). Most importantly, Oil-Red-O' and Nile red tests showed that HSO induced adipogenic hMSC differentiation without differentiation agents. Additionally, HSO-treated cells showed increased peroxisome proliferator-activated receptor gamma (PPARγ) mRNA expression compared to controls (hMSC). HSO reduced PPARγ mRNA expression after differentiation media (DM) treatment. After treatment with HSO, DM-hMSCs had significantly lower CB1 mRNA and protein expressions than normal hMSCs. HSO treatment also decreased transient receptor potential vanilloid 1 (TRPV1), fatty acid amide hydrolase (FAAH), and monoacylglycerol lipase (MGL) mRNAs in hMSC and DM-hMSCs. HSO treatment significantly decreased CB1, CB2, TRPV1, and G-protein-coupled receptor 55 (GPCR55) protein levels in DM-hMSC compared to hMSC in western blot analysis. In this study, HSO initiated adipogenic differentiation in hMSC without DM, but it suppressed CB1 gene and protein expression, potentially decreasing adipocyte lipid accumulation and lipogenic enzymes.


Subject(s)
Cannabidiol , Cannabinoids , Cannabis , Mesenchymal Stem Cells , Plant Extracts , Humans , Cannabinoids/pharmacology , Cannabidiol/pharmacology , PPAR gamma , Endocannabinoids , Adipose Tissue, Brown , RNA, Messenger
2.
J Sci Food Agric ; 104(3): 1813-1823, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37872732

ABSTRACT

BACKGROUND: Lipids and carbohydrates perform essential functions in foods. In recent decades, food scientists have studied the effects of carbohydrate-lipid interactions on the functional properties of food. However, the ways in which carbohydrate-lipid complex-derived materials affect the biological system are unknown. In this study, a myristic acid-potato starch complex was created using a simple cooking approach. The complex was employed as a precursor for the fabrication of myristic acid-potato starch complex-based nanostructured materials (MPS-NMs) through a liquid-liquid extraction approach. A study was conducted on the structural and cytotoxic features of the fabricated MPS-NMs. RESULTS: Transmission electron microscopy images confirmed the formation of spherical nanostructures, 3-60 nm in size. After 24 h exposure, the chloroform fraction-based and n-hexane fraction-based MPS-NMs increased cell death by ~90% and ~ 82%, respectively. Chloroform fraction-based MPS-NMs (CMPS-NMs) triggers apoptotic cell death in human mesenchymal stem cells (hMSCs). n-Hexane fraction-based MPS-NMs (HMPS-NMs) treated cells have red color-intact nuclei, attributed to necrotic cell death. The CMPS-NMs and HMPS-NMs significantly decreased the mitochondria membrane potential and increased the intracellular reactive oxygen species (ROS) levels. We observed significant downregulation in flavin-containing monooxygenase (FMO), Ataxia Telangiectasia Mutated (ATM), and uridine diphosphate glucuronosyltransferases (UGT) gene expression levels in the exposed cells of CMPS-NMs and HMPS- NMs. In addition, we found upregulation of glutathione-disulfide reductase (GSR) and glutathione S-transferase A4 (GSTA4) genes in CMPS-NMs, and HMPS-NMs exposure. CONCLUSION: The cooking process may lead to the formation of nanostructured material in food systems. Chloroform fraction-based MPS-NMs and HMPS-NMs may contribute to cell metabolic disorders. © 2023 Society of Chemical Industry.


Subject(s)
Nanostructures , Solanum tuberosum , Humans , Myristic Acid , Chloroform , Nanostructures/chemistry , Starch , Carbohydrates
3.
Biotechnol Appl Biochem ; 71(2): 326-335, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38112040

ABSTRACT

Nanostructures have been used for various biomedical applications due to their optical, antibacterial, magnetic, antioxidant, and biocompatible properties. Cancer is a prevalent disease that severely threatens human life and health. Thus, innovative and effective therapeutic approaches are urgently required for cancer. Photothermal therapy (PTT) is a promising approach to killing cancer cells. In this investigation, we developed a low-cost, simple, green technique to fabricate molybdenum trioxide nanostructures (MNs) using Opuntia ficus-indica mucilage as a template. Moreover, the MNs were functionalized with folic acid (FA) for cancer PTT. The X-ray diffractometer results revealed that the prepared MNs have an orthorhombic crystal phase. The transmission electron microscope image of MNs shows a flake shape with 20-150 nm diameter. The cytotoxicity of MNs and FA-conjugated MNs was studied in vitro. These cell viability assay results suggested that fabricated MoO3 nanostructures reduced 25% of cell viability in MCF-7 cells, even at high doses. However, even with high-dose treatment, FA/MNs do not cause significant cell death. Acridine orange/ethidium bromide (AO/EB) staining revealed DNA and chromatin condensation in MCF-7 cells exposed to MNs. Overall, the in vitro study results suggested that FA/MNs have excellent biocompatibility, which applies to biomedical applications. MNs dispersion temperature gradually increases from 26 to 58°C under 808 nm laser irradiation. We found significant mortality rates after NIR irradiation in MNs- or FA/MNs-treated MCF-7 cells. These findings suggest that FA/MNs can be used as an effective photothermal agent to treat breast cancer.


Subject(s)
Breast Neoplasms , Nanostructures , Oxides , Humans , Female , Phototherapy/methods , Breast Neoplasms/drug therapy , Nanostructures/chemistry , Molybdenum/pharmacology , Molybdenum/chemistry
4.
Environ Sci Pollut Res Int ; 30(12): 33264-33274, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36474033

ABSTRACT

Several nano-toxicological studies have assessed the prospective health risks of engineered nanostructures. Still, nanoscale ingredients from food products are not explored well, and only a few have attended to the possible effects of food additive-based nanoparticles in food. The physicochemical properties of food additives and their fate on human health are still unknown. To fill this knowledge gap, we examined the physicochemical characteristics of food product isolate E341/E551. Additionally, we assessed the consequence of these nanoscale E341 and E551 as co-exposure on human mesenchymal stem cells (hMSCs). The transmission electron microscope (TEM) images revealed that food product isolate (E341/E551) consists of nanoscale particles. The E551 and E341 have 20-50 nm and 70-200 nm diameters, respectively. Co-exposure of food additives SiO2 (E551) and Tricalcium phosphate (E341) effect on the cell viability, morphology, mitochondrial membrane potential, and reactive oxygen species (ROS) level of hMSCs were studied. The cell viability reduction, mitochondrial membrane potential loss, and ROS generation in E341/E551 co-exposed cells were observed. Our study suggests that E341/E551 co-exposure elevated the ROS level and mitochondrial membrane potential depletion at a high dose. The oxidative stress-related genes MDM3, TNFSF10, and POR have exhibited significant upregulation in the E341/E551 treatment group. These results conclude that long-term over-exposure to E341/E551 may be triggers health risks in a human. Further in vivo studies are required for food industry implications due to nanoscale ingredients in E341 and E551.


Subject(s)
Mesenchymal Stem Cells , Nanoparticles , Humans , Reactive Oxygen Species/metabolism , Silicon Dioxide/chemistry , Nanoparticles/toxicity , Nanoparticles/chemistry , Food Additives/toxicity
5.
Genes (Basel) ; 12(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34573361

ABSTRACT

The antioxidant capacity of polyphenols and flavonoids present in dietary agents aids in arresting the development of reactive oxygen species (ROS) and protecting endothelial smooth muscle cells from oxidative stress/induced necrosis. Beetroot (Beta vulgaris var. rubra L.; BVr) is a commonly consumed vegetable representing a rich source of antioxidants. Beetroot peel's bioactive compounds and their role in human umbilical vein endothelial cells (HUVECs) are still under-researched. In the present study, beetroot peel methanol extract (BPME) was prepared, and its effect on the bio-efficacy, nuclear integrity, mitochondrial membrane potential and vascular cell growth, and immunoregulation-related gene expression levels in HUVECs with induced oxidative stress were analysed. Gas chromatography-mass spectroscopy (GC-MS) results confirmed that BPME contains 5-hydroxymethylfurfural (32.6%), methyl pyruvate (15.13%), furfural (9.98%), and 2,3-dihydro-3,5-dihydroxy-6-methyl-4H-Pyran-4-one (12.4%). BPME extract effectively enhanced cell proliferation and was confirmed by MTT assay; the nuclear integrity was confirmed by propidium iodide (PI) staining assay; the mitochondrial membrane potential (Δψm) was confirmed by JC-1 staining assay. Annexin V assay confirmed that BPME-treated HUVECs showed 99% viable cells, but only 39.8% viability was shown in HUVECs treated with H2O2 alone. In addition, BPME treatment of HUVECs for 48 h reduced mRNA expression of lipid peroxide (LPO) and increased NOS-3, Nrf-2, GSK-3ß, GPX, endothelial nitric oxide synthase (eNOS) and vascular cell growth factor (VEGF) mRNA expression levels. We found that BPME treatment decreased proinflammatory (nuclear factor-κß (F-κß), tissue necrosis factor-α (TNF-α), toll-like receptor-4 (TLR-4), interleukin-1ß (IL-1ß)) and vascular inflammation (intracellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), EDN1, IL-1ß)-related mRNA expressions. In conclusion, beetroot peel treatment effectively increased vascular smooth cell growth factors and microtubule development, whereas it decreased vascular inflammatory regulators. BPME may be beneficial for vascular smooth cell regeneration, tissue repair and anti-ageing potential.


Subject(s)
Antioxidants/pharmacology , Beta vulgaris/chemistry , Plant Extracts/pharmacology , Antioxidants/isolation & purification , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Methanol/chemistry , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/metabolism
6.
Saudi J Biol Sci ; 28(10): 6009-6016, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34588918

ABSTRACT

Cancer traits dependent chemo and radiotherapy display acute toxicity and long-term side effects. Since last two decades, researchers investigated a new anticancer agents derived from plants. Cassia alata (L.) is a medicinal herb distributed in the tropical and humid regions. In this study, C. alata flower methanol extract (CME) have been prepared using cold percolation method and the phytochemical components were identified using GC-MS analysis. CME have been used to study the antiproliferative and apoptosis properties against human colon cancer HT-115 colon cancer cells, its molecular mechanism have been explored. 0.2 mg/mL dose of CME, inhibited 50% of HT-115 colon cancer cell growth after 48hr was confirmed the significant antiproliferation effect. In normal cells such as Vero cells and hMSCs, 0.2 mg/mL dose of CME shown only 4% and 5% growth inhibition confirmed the HT-115 cell specific cytotoxic effect. This effect might be due to the availability of phytoactive biomolecules in CME such as, cyclotrisiloxan, beta-sitosterol and alpha-tocopherol have been confirmed by GC-MS. Most interestingly, PI and AO/ErBr staining of CME treated HT-115 cells shown early (25%), pro (17%) and late (8%) apoptotic and 3% necrotic cells after 48 hr. Treatment with CME extract showed potential effect on the inhibition of protumorigenic inflammatory and oxidative stress genes. Protumorigenic COX-2/PGE-2 and TNF-α/NF-κB immune axis were normalized after CME treatment. Amounts of both apoptosis related mRNA p53, Bax, caspase 3 and p21 genes were upregulated, whereas it resulted in significant reduction in the anti-apoptotic marker mdm2 and Bcl-2 genes. In conclusion, bioactive compounds present in CME potentially inhibit HT-115 colon cancer cell proliferation via an inhibition of protumorigenic immune axis and stimulation of mitochondria dependent apoptotic pathway without necrotic effect.

7.
Biomed Res Int ; 2020: 3712536, 2020.
Article in English | MEDLINE | ID: mdl-32685475

ABSTRACT

The second most biggest cancer worldwide is breast cancer. There is an increasing need for safer, effective, and affordable drug candidates from natural sources to treat breast cancer. In the present investigation, the anticancer effect of Cucurbita ficifolia Bouché (C. ficifolia) fruit extract was tested on the human breast cancer cells such as MCF-7. The cells were exposed with different doses of C. ficifolia, for the assessment of IC50 concentrations on the MCF-7 cell lines for 24 hs. The effect of C. ficifolia fruit extract on morphological and apoptotic changes were evaluated by specific fluorescence staining techniques and real-time PCR in a time-dependent manner for 24 hs and 48 hs. The IC50 value for C. ficifolia fruit extract was found to be 90 µg/mL. Morphological alteration and apoptotic distinctiveness aspect like chromatin condensation and nuclear fragmentation were noticed in C. ficifolia extract exposed breast cancer cells. Further, we observed that C. ficifolia extract-induced programmed cell death in the MCF-7 cells were mediated with the elevated expression of the tumor suppressor gene such as p53 and apoptotic markers such as caspase-8, caspase-9, caspase-3, fatty acid synthase (FAS), Fas-associated protein with death domain (FADD), Bcl-2 homologous antagonist/killer (BAK), and Bcl-2-associated X protein (BAX). These observations established that C. ficifolia significantly concealed the cell division and provoked p53/caspase-mediated programmed cell death. Further, we noticed that this cell death in MCF-7 cells is concentration and time dependent. As evaluated through the comet assay, C. ficifolia induced DNA damage; further upon increasing the duration of the treatment, the DNA damage was higher than before. Thus, our study concludes that C. ficifolia could serve as an effective anticancer agent through vital gene modulation.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Caspases/metabolism , Cucurbita/chemistry , Fruit/chemistry , Plant Extracts/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Breast Neoplasms/genetics , Caspases/genetics , DNA Damage , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staining and Labeling
8.
Mater Sci Eng C Mater Biol Appl ; 93: 242-253, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30274056

ABSTRACT

Graphene nanocomposites have received attention for the therapy and detection of diseases. In this study, we developed a simple and green chemistry approach for synthesizing Cu2O/graphene nanocomposites (Cu2O/G) using date palm fruit syrup as a reducing agent. The graphene oxide surface anchored with Cu(OH)2 and reduced it to fabricate Cu2O-anchored graphene nanosheets using date palm fruit syrup. Physicochemical characteristics of the synthesized nanocomposites were analyzed. Scanning electron microscopy images revealed 50-70 nm Cu2O nanostructures anchored on the surface of crumpled graphene sheets. The Cu2O/G nanocomposites inhibited the gram-negative and gram-positive bacterial growth at 300 µg. When compared with Cu2O nanoparticles and graphene oxide nanosheets (GO), Cu2O/G nanocomposite exhibited outstanding bactericidal activity. The cytotoxic properties of the prepared nanocomposites were studied in human mesenchymal stem cells (hMSCs). The Cu2O/G nanocomposites did not reduced cell viability by up to 200 µg/mL and slightly induced cell death at high concentrations. However, Cu2O nanoparticles and GO have significantly reduced the cell viability in hMSCs. The microscopic images of cellular and nuclear morphology suggested that the Cu2O/G composites did not cause major changes to hMSCs. The Cu2O nanoparticles and GO remarkably triggers the cellular damages, nuclear condensation and DNA fragmentation in hMSCs. Our study results revealed that Cu2O/G has excellent antibacterial activity with good biocompatibility. Thus, Cu2O/G could be used as a promising antibacterial agent in various purposes.


Subject(s)
Anti-Bacterial Agents , Copper , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/growth & development , Graphite , Materials Testing , Mesenchymal Stem Cells/metabolism , Nanocomposites , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Copper/adverse effects , Copper/chemistry , Copper/pharmacology , Drug Evaluation, Preclinical , Graphite/adverse effects , Graphite/chemistry , Graphite/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Nanocomposites/adverse effects , Nanocomposites/chemistry
9.
Microb Pathog ; 120: 85-96, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29684541

ABSTRACT

The appearance of drug-resistant (DR) bacteria in the community is a crucial development, and is associated with increased morbidity, mortality, healthcare costs, and antibiotic use. Natural oil nanoemulsions (NEs) have potential for antimicrobial applications. In the present study, we determined the antimicrobial activity of an NE against DR bacterial pathogens in vitro. The NE comprised Cleome viscosa essential oil, Tween 80 nonionic surfactant, and water. We found that an NE with a droplet size of 7 nm and an oil:surfactant (v/v) ratio of 1:3 was effective against methicillin-resistant Staphylococcus aureus (MRSA), DR Streptococcus pyogenes, and DR extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Fourier-transform infrared (FTIR) spectroscopy revealed that NE treatment modified the functional groups of lipids, proteins, and nucleic acids in DR bacterial cells. Scanning electron microscopy (SEM) showed damage to the cell membranes and walls of NE-treated DR bacteria. These alterations were caused by bioactive compounds with wide-spectrum enzyme-inhibiting activity in the NE, such as ß-sitosterol, demecolcine, campesterol, and heneicosyl formate. The results suggest that the nanoemulsion is effective against DR bacteria, and acts by inhibiting the drug efflux mechanism of DR strains.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Emulsions/pharmacology , Nanostructures/chemistry , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/chemistry , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Cleome/chemistry , Demecolcine/pharmacology , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Nanostructures/ultrastructure , Oils, Volatile/pharmacology , Particle Size , Phytosterols/pharmacology , Plant Extracts/pharmacology , Polysorbates/pharmacology , Pseudomonas aeruginosa/drug effects , Sitosterols/pharmacology , Sonication , Streptococcus pyogenes/drug effects , Surface-Active Agents
10.
Biomed Pharmacother ; 87: 388-396, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28068628

ABSTRACT

Systematic analyses of plants that are used in traditional medicine may lead to the discovery of novel cytotoxic secondary metabolites. Diterpene possesses multiple bioactivities; here, epoxy clerodane diterpene (ECD) was isolated from Tinospora cordifolia (Willd.) stem and shown potential antiproliferative effect in MCF-7 human breast cancer cells. The antiproliferative effect of ECD on MCF-7 cells was systematically analyzed by cell and nuclear morphology, alterations in oxidative stress, and the expression of tumor suppressor and mitochondria-mediated apoptosis-related genes. We found that the IC50 value of ECD was 3.2µM at 24h and 2.4µM at 48h. We observed that the cytotoxicity of ECD was specific to MCF-7 cells, whereas ECD was nontoxic to normal Vero and V79 cells. ECD significantly triggered intracellular ROS generation even from the lower doses of 0.6 and 1.2µM; and it is relative to higher dose of 2.4µM. Further, we used 0.6µM, 1.2µM and 2.4µM as experimental doses to analyze the relative dose-dependent effects. Nuclear staining revealed that cells treated with the 2.4µM dose exhibited characteristic apoptotic morphological changes and that 46% of the cells were apoptotic and 4% were necrotic after 48h. ECD significantly increased the expression of mitochondria-dependent apoptotic pathway-related genes after 48h; we observed significantly (p≤0.05) increased expression of CYP1A, GPX, GSK3ß and TNF-α and downregulated expression of NF-κB. ECD also increased the expression of tumor suppressor genes such as Cdkn2A, Rb1 and p53. In addition, we observed that ECD treatment significantly (p≤0.001) upregulated the expression of apoptotic genes such as Bax, cas-3, cas-8, cas-9 and p21 and downregulated the expression of BCL-2, mdm2 and PCNA. In conclusion, ECD regulates the expression of Cdkn2A, p53 and mdm2 and induces apoptosis via the mitochondrial pathway in MCF-7 human breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p18/genetics , Diterpenes, Clerodane/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , Retinoblastoma Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Female , Gene Expression/drug effects , Gene Expression/genetics , Humans , MCF-7 Cells , Mitochondria/drug effects , Mitochondria/genetics , NF-kappa B/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
11.
Appl Biochem Biotechnol ; 181(2): 725-734, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27778152

ABSTRACT

Nanographene- and graphene-based nanohybrids have garnered attention in the biomedical community owing to their biocompatibility, excellent aqueous processability, ease of cellular uptake, facile surface functionalization, and thermal and electrical conductivities. NiO nanoparticle-graphene nanohybrid (G-NiO) was synthesized by first depositing Ni(OH)2 onto the surface of graphene oxide (GO) sheets. The Ni(OH)2-GO hybrids were then reduced to G-NiO using date palm syrup at 85 °C. The prepared G-NiO nanohybrids were characterized by X-ray diffraction (XRD), field-emission scanning electron microscopy (FESEM), Fourier transform infrared spectroscopy, and energy-dispersive X-ray spectroscopy (EDX). The NiO nanoparticles, with a diameter of approximately 20-30 nm, were uniformly dispersed over the surface of the graphene sheets. The G-NiO hybrids exhibit biocompatibility in human mesenchymal stem cells (hMSCs) up to 100 µg/mL. The nanohybrids do not cause any significant changes in cellular and nuclear morphologies in hMSCs. The as-synthesized nanohybrids show excellent biocompatibility and could be a promising material for biomedical applications.


Subject(s)
Graphite/chemistry , Mesenchymal Stem Cells/drug effects , Metal Nanoparticles/toxicity , Nickel/chemistry , Nickel/toxicity , Phoeniceae/chemistry , Absorption, Physicochemical , Cell Survival/drug effects , Coated Materials, Biocompatible/chemical synthesis , Coated Materials, Biocompatible/toxicity , Graphite/toxicity , Humans , Materials Testing , Mesenchymal Stem Cells/cytology , Metal Nanoparticles/chemistry , Plant Extracts/chemistry , Plant Extracts/toxicity
12.
Environ Toxicol Pharmacol ; 47: 19-27, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27567443

ABSTRACT

Zinc (Zn) is an essential trace elements, its deficiency is associated with increased incidence of human breast cancer. We aimed to study the effect of Zn on human breast cancer MCF-7 cells cultured in Zn depleted and Zn adequate medium. We found increased cancer cell growth in zinc depleted condition, further Zn supplementation inhibits the viability of breast cancer MCF-7 cell cultured in Zn deficient condition and the IC25, IC50 value for Zn is 6.2µM, 15µM, respectively after 48h. Zn markedly induced apoptosis through the characteristic apoptotic morphological changes and DNA fragmentation after 48h. In addition, Zn deficient cells significantly triggered intracellular ROS level and develop oxidative stress induced DNA damage; it was confirmed by elevated expression of CYP1A, GPX, GSK3ß and TNF-α gene. Zinc depleted MCF-7 cells expressed significantly (p≤0.001) decreased levels of CDKN2A, pRb1, p53 and increased the level of mdm2 expression. Zn supplementation (IC50=15µM), increased significantly CDKN2A, pRB1 & p53 and markedly reduced mdm2 expression; also protein expression levels of CDKN2A and pRb1 was significantly increased. In addition, intrinsic apoptotic pathway related genes such as Bax, caspase-3, 8, 9 & p21 expression was enhanced and finally induced cell apoptosis. In conclusion, physiological level of zinc is important to prevent DNA damage and MCF-7 cell proliferation via regulation of tumor suppressor gene.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Salivary Proline-Rich Proteins/genetics , Tumor Suppressor Protein p53/genetics , Zinc/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Culture Media , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Humans , MCF-7 Cells/drug effects , MCF-7 Cells/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Zinc/deficiency
13.
Ultrason Sonochem ; 31: 449-55, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26964971

ABSTRACT

Nigella sativa L. (NS) is a plant renowned in traditional holistic medicine systems for almost 1400 years because of its remarkable antioxidant, antimicrobial, anti-inflammatory and anti-cancer properties. The essential oil of N. sativa, in particular, possesses these significant biological properties. However, N. sativa essential oil has many insoluble constituents with properties that have not been fully explored. Nanoemulsion-based insoluble formulations are a widely used carrier system for lipophilic materials. In the present study, we used ultrasonic emulsification, polysorbate 80 and water to formulate a highly stable N. sativa essential oil nanoemulsion (NSEO-NE). To optimize the NSEO-NE preparation, we changed the surfactant concentration, the oil-surfactant mixing ratio and the emulsification time. The droplet size distribution and morphology of the prepared NE was analyzed using dynamic light scattering and scanning electron microscopy, respectively. The droplet size of the NSEO-NE was approximately 20-50 nm in diameter. The anticancer properties of the NE preparation were studied using a modified methyl-thiazolyl-diphenyl tetrazolium bromide (MTT) assay as well as cellular uptake and nuclear morphological analyses. The NSEO-NE significantly reduced the viability of Michigan Cancer Foundation-7 (MCF-7) breast cancer cells. The nucleo-cytoplasmic morphological features of NSEO-NE-treated cells included cell membrane blebbing, cytoplasmic vacuolation, marginalization of chromatin, and fragmentation of the nucleus. The results clearly indicate that NSEO-NE induced apoptosis in MCF-7 cells. These findings support the potential application of NSEO-NE in breast cancer therapy, and also merit future translational research.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Emulsions , Nanotechnology , Nigella sativa/chemistry , Oils, Volatile/pharmacology , Ultrasonics , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans
14.
Biotechnol Appl Biochem ; 63(3): 320-7, 2016 May.
Article in English | MEDLINE | ID: mdl-25779086

ABSTRACT

Aluminum oxide nanoparticles (Al2 O3 -NPs) are important ceramic materials that have been used in a variety of commercial and industrial applications. However, the impact of acute and chronic exposure to Al2 O3 -NPs on the environment and on human health has not been well studied. In this investigation, we evaluated the cytotoxic effects of Al2 O3 -NPs on human mesenchymal stem cells (hMSCs) by using a cell viability assay and observing cellular morphological changes, analyzing cell cycle progression, and monitoring the expression of cell cycle response genes (PCNA, EGR1, E2F1, CCND1, CCNC, CCNG1, and CYCD3). The Al2 O3 -NPs reduced hMSC viability in a dose- and time-dependent manner. Nuclear condensation and fragmentation, chromosomal DNA fragmentation, and cytoplasmic vacuolization were observed in Al2 O3 -NP-exposed cells. The nuclear morphological changes indicated that Al2 O3 -NPs alter cell cycle progression and gene expression. The cell cycle distribution revealed that Al2 O3 -NPs cause cell cycle arrest in the sub-G0-G1 phase, and this is associated with a reduction in the cell population in the G2/M and G0/G1 phases. Moreover, Al2 O3 -NPs induced the upregulation of cell cycle response genes, including EGR1, E2F1, and CCND1. Our results suggested that exposure to Al2 O3 -NPs could cause acute cytotoxic effects in hMSCs through cell cycle regulatory genes.


Subject(s)
Aluminum Oxide/toxicity , Cell Cycle/drug effects , Cyclin D1/genetics , Early Growth Response Protein 1/genetics , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/drug effects , Nanoparticles/chemistry , Aluminum Oxide/chemistry , Biological Transport/drug effects , Biomarkers/metabolism , Cell Survival/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
15.
Asian Pac J Cancer Prev ; 15(22): 9655-60, 2014.
Article in English | MEDLINE | ID: mdl-25520084

ABSTRACT

BACKGROUND: Nigella Sativa (NS) is an herb from the Ranunculaceae family that exhibits numerous medicinal properties and has been used as important constituent of many complementary and alternative medicines (CAMs). The ability of NS to kill cancer cells such as PC3, HeLa and hepatoma cells is well established. However, our understanding of the mode of death caused by NS remains nebulous. The objective of this study was to gain further insight into the mode and mechanism of death caused by NS in breast cancer MCF-7 cells. MATERIALS AND METHODS: Human breast cancer cells (MCF-7) were treated with a methanolic extract of NS, and a dose- and time-dependent study was performed. The IC50 was calculated using a Cell Titer Blue® viability assay assay, and evidence for DNA fragmentation was obtained by fluorescence microscopy TUNEL assay. Gene expression was also profiled for a number of apoptosis-related genes (Caspase-3, -8, -9 and p53 genes) through qPCR. RESULTS: The IC50 of MCF-7 cells was 62.8 µL/mL. When MCF-7 cells were exposed to 50 µL/mL and 100 µL/mL NS for 24 h, 48 h and 72 h, microscopic examination (TUNEL assay) revealed a dose- and time-dependent increase in apoptosis. Similarly, the expression of the Caspase-3, -8, -9 and p53 genes increased significantly according to the dose and time. CONCLUSIONS: NS induced apoptosis in MCF-7 cells through both the p53 and caspase pathways. NS could potentially represent an alternative source of medicine for breast cancer therapy.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Nigella sativa/metabolism , Plant Extracts/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/genetics , Caspase 3/biosynthesis , Caspase 3/genetics , Caspase 8/biosynthesis , Caspase 8/genetics , Caspase 9/biosynthesis , Caspase 9/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Seeds/metabolism , Tumor Suppressor Protein p53/genetics
16.
Chem Biol Interact ; 224: 157-63, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25289771

ABSTRACT

Gastric ulcer is an illness that affects a great number of people worldwide. The goal of the present research was to assess the anti-ulcerogenic activity of nymphayol (NYM), isolated from Nymphaea stellata, against an ethanol-induced ulcer model in rats. Administration of ethanol elevates the levels of the ulcer index (UI) along with causing tremendous increases in lipid peroxidation and myeloperoxidase (MPO) and significant decreases in gastric mucus, catalase (CAT), superoxide dismutase (SOD), glutathione (GSH), glutathione peroxidase (GPx), and prostaglandin E2 (PGE2). However, the NYM- (45 mg/kg) pretreated animals showed considerable increases in antioxidants, gastric mucus, and PGE2 level and significant decreases in UI, lipid peroxidation, and MPO level. Pro-inflammatory cytokines such as interleukin-6 (IL-6), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) were increased and the level of interleukin-10 (IL-10), an anti-inflammatory cytokine, was decreased in ethanol-induced ulcerated animals, and these inequalities were amended by NYM pretreatment. Pro-apoptotic markers including caspase-8, caspase-9, and caspase-3 were decreased and Bcl-2, an anti-apoptotic marker, was increased through NYM pretreatment, as compared with the ethanol-induced ulcer group. Pretreatment with indomethacin, SC560, rofecoxib, and Nω-Nitro-L-arginine methyl ester (L-NAME) considerably prevented the ulcer protective activity of NYM (45 mg/kg), indicating the involvement of cyclooxygenase (COX) and nitric oxide synthase (NOS) in NYM-mediated gastroprotection against ethanol-induced ulcer. These outcomes suggest that the gastroprotective effect of NYM might be mediated by adjustment of inflammatory mediators and apoptotic markers and increasing antioxidants.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Flowers/chemistry , Nymphaea/chemistry , Phytosterols/pharmacology , Stomach Ulcer/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Dose-Response Relationship, Drug , Ethanol , Female , Male , Molecular Conformation , Phytosterols/chemistry , Phytosterols/isolation & purification , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Structure-Activity Relationship
17.
Chin J Integr Med ; 2014 Mar 02.
Article in English | MEDLINE | ID: mdl-24584754

ABSTRACT

OBJECTIVE: To study the immunomodulatory effects of Nigella sativa (NS) on human peripheral blood mononuclear cells (PBMCs) on a Phytohemagglutinin (PHA) and a non-PHA stimulated proliferation. METHODS: Cells isolated from human PBMCs which were treated with methanolic extract of NS for 48 h into two separate environments (PHA and non-PHA stimulated). Flow cytometry (for T helper/inducer cells and natural killer cells) and real time-polymerase chain reaction (PCR) assays for a few selected proinflammatory gene expressions were performed. RESULTS: Extracts from NS had an immunostimulating effect on non-PHA-stimulated proliferation of human PBMCs. In contrast, immunosuppressive activity was observed on PHA-stimulated proliferation of human PBMCs. CONCLUSION: This in vitro study revealed the effects of NS plant extract on nonspecifific cellular immune responses.

18.
Nat Prod Commun ; 8(2): 213-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23513732

ABSTRACT

Nigella sativa (NS), also known as black cumin, has long been used in traditional medicine for treating various cancer conditions. In this study, we sought to investigate the potential anti-cancer effects of NS extract using SiHa human cervical cancer cells. NS showed an 88.3% inhibition of proliferation of SiHa human cervical cancer cells at a concentration of 125 microL/mL methanolic extract at 24 h, and an IC50 value 93.2 microL/mL. NS exposure increased the expression of caspase-3, -8 and -9 several-fold. The analysis of apoptosis by Dead End terminal transferase-mediated dUTP-digoxigenin end labeling (TUNEL) assay was used to further confirm that NS induced apoptosis. Thus, NS was concluded to induce apoptosis in SiHa cell through both p53 and caspases activation. NS could potentially be an alternative source of medicine for cervical cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Nigella sativa , Plant Extracts/pharmacology , Uterine Cervical Neoplasms/drug therapy , Caspases/analysis , Female , Humans , Seeds/chemistry , Tumor Suppressor Protein p53/physiology , Uterine Cervical Neoplasms/pathology
19.
Article in English | MEDLINE | ID: mdl-23046173

ABSTRACT

An urgent need for toxicological studies on aluminium oxide nanoparticles (Al(2) [Formula: see text]NPs) has arisen from their rapidly emerging range of applications in the food and agricultural sectors. Despite the widespread use of nanoscale aluminium and its composites in the food industry, there is a serious lack of information concerning the biological activities of Al(2) [Formula: see text]NPs (ANPs) and their impact on human health. In this preliminary study, the effects of ANPs on metabolic stress in human mesenchymal stem cells (hMSCs) were analysed. The results showed dose-dependent effects, including cellular toxicity. The mitochondrial membrane potential in the hMSCs decreased with increasing ANP concentrations after 24 h of exposure. The expression levels of oxidative stress-responsive enzymes were monitored by RT-PCR. The expression levels of CYP1A and POR were up-regulated in response to ANPs, and a significant down-regulation in the expression of the antioxidant enzyme SOD was observed. Further, dose-dependent changes in the mRNA levels of GSTM3, GPX and GSR were noted. These findings suggest that the toxicity of ANPs in hMSCs may be mediated through an increase in oxidative stress. The results of this study clearly demonstrate the nanotoxicological effects of ANPs on hMSCs, which will be useful for nanotoxicological indexing.


Subject(s)
Aluminum Oxide/toxicity , Antioxidants/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Mesenchymal Stem Cells/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Aluminum Oxide/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Particle Size , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Basic Clin Pharmacol Toxicol ; 112(6): 374-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23145928

ABSTRACT

Tea polyphenols (TPP) have potent antioxidant and anticancer properties, particularly in patients undergoing radiation or chemotherapy. However, few studies have been conducted on treatments using a combination of TPP and the conventional chemical anticancer drug cisplatin (CP). This study was designed to investigate the mechanism of the cytotoxicity of total TPP and CP, which may synergistically induce cell death in cancer cells. Here, breast cancer cells (MCF-7) were treated with various concentrations of TPP alone or in combination with the chemotherapeutic drug CP. The effect of TPP on cell growth, intracellular reactive oxygen species (ROS) level, apoptosis and gene expression of caspase-3, caspase-8 and caspase-9 and p53 was investigated. The MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay revealed that the MCF-7 cells were less sensitive to growth inhibition by TPP treatment than either CP or the combination therapy. Propidium iodide nuclear staining indicated that exposure to this combination increased the proportion of apoptotic nuclei compared with a single-agent treatment. Flow cytometry analysis was used to quantify changes in intracellular ROS. Detection of activated caspases by fluorescently labelled inhibitors of caspases (FLICA) combined with the plasma membrane permeability assay demonstrated that the percentage of early and late apoptotic/secondary necrotic cells was higher in the cells treated with the combination than in those treated with either TPP or CP alone. The combined TPP and CP treatment synergistically induced apoptosis through both caspase-8 and caspase-9 activation and p53 over-expression. This suggests that TPP plus CP may be used as an efficient antioxidant-based combination therapy for estrogen receptor (ER)-positive and p53-positive breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Camellia sinensis/chemistry , Cisplatin/pharmacology , Polyphenols/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Caspases/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/isolation & purification , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , Gene Expression/drug effects , Humans , MCF-7 Cells , Oxidation-Reduction , Polyphenols/isolation & purification , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL