8 research outputs found

    Synthesis, characterization, and antiproliferative effect of CuO-TiO2-chitosan-Amygdalin nanocomposites in human leukemic MOLT4 cells

    No full text
    The main aim of this study was to synthesize copper oxide- (CuO-) titanium oxide- (TiO2-) chitosan-amygdalin nanocomposites (CTCANc) and to characterize them physically and biologically (antimicrobial and anticancer activity using MOLT4 blood cancer cell line) to endorse their useful applications as potential drug candidates in anticancer avenues. CuO-TiO2-chitosan-amygdalin nanocomposites were synthesized according to standard, reported methods. Physical characterization of the nanocomposites was performed using methods like X-ray diffractometer (XRD), and morphological and ultrastructural analysis of nanocomposites were done using electron microscope scanning and transmission. FTIR was recorded using a Perkin-Elmer spectrometer, and photoluminescence (PL) spectra were done using the spectrometer. Further, antibacterial activities were assessed using standard bacterial cultures. To demonstrate the nanocomposite’s anticancer effects, MTT assay, morphological analysis, apoptosis studies using acridine orange/ethidium bromide (AO/EtBr) dual staining, reactive oxygen species (ROS) analysis, and levels of antioxidant enzymes were analyzed using the MOLT4 blood cancer cell line. Synthesized nanocomposites were characterized using XRD and showed various peaks, respectively, for CuO-TiO2, amygdalin, and chitosan. MTT assay indicated an IC50 value of 38.41 μg/ml concentration of CTCANc. Hence, 30 and 40 μg/ml were used for the subsequent experiments. Morphological analysis, staining for apoptosis using AO/EtBr, mitochondrial membrane potential (MMP or ΔΨm) analysis, ROS analysis, and determination of the SOD, CAT, MDA, and GSH levels were performed. Observations like a significant loss of morphology, induction of apoptosis, elevated ROS, and decreased MMP were significant in 30 and 40 μg/ml nanocomposite-treated cells when compared to control cells. The bimetallic nanocomposites exhibited typical nanocomposites characteristics and significant antibacterial and anticancer effects. The study results endorse the antibacterial, anticancer activity of CuO-TiO2-chitosan-amygdalin nanocomposites and strongly suggest that further in-depth research using CuO-TiO2-chitosan-amygdalin nanocomposites could reveal their efficacy in the clinical scenario

    CuO-TiO2-Chitosan-Berbamine nanocomposites induce apoptosis through the mitochondrial pathway with the expression of P53, BAX, and BCL-2 in the human K562 cancer cell line

    No full text
    In this study, cells from human Chronic Myelogenous Leukemia (K562) were cultivated with CuO-TiO2-Chitosan-Berbamine nanocomposites. We examined nanocomposites using XRD, DLS, FESEM, TEM, PL, EDAX, and FTIR spectroscopy, as well as MTT for cytotoxicity, and AO/EtBr for apoptotic morphology assessment. The rate of apoptosis and cell cycle arrests was determined using flow cytometry. Flow cytometry was also employed to identify pro- and antiapoptotic proteins such as Bcl2, Bad, Bax, P53, and Cyt C. The FTIR spectrum revealed that the CuO-TiO2-Chitosan-Berbamine nanocomposites were electrostatically interlocked. The nanocomposites’ XRD signals revealed a hexagonal shape. In the DLS spectrum, nanocomposites were found to have a hydrodynamic diameter. As a result of their cytotoxic action, nanocomposites displayed concentration-dependent cytotoxicity. The nanocomposites, like Doxorubicin, caused cell cycle phase arrest in K562 cells. After treatment with IC50 concentrations of CuO-TiO2-Chitosan-Berbamine nanocomposites and Doxorubicin, a substantial percentage of cells were in G2/M stage arrest. Caspase-3, -7, -8, -9, Bax, Bad, Cyt C, and P53 expression were considerably enhanced in K562 cells, whereas Bcl2 expression was decreased, indicating that these cells may have therapeutic potential against human blood cancer/leukemia-derived disorders. As a result, the nanocomposites demonstrated outstanding anticancer potential against leukemic cells. CuO-TiO2-Chitosan-Berbamine, according to our findings

    Synthesis of zinc oxide (zno)-titanium dioxide (tio2)-chitosan-farnesol nanocomposites and assessment of their anticancer potential in human leukemic molt-4 cell line

    No full text
    Leukemia is the most prevalent cancer in children and one of the most common and deadly cancers that affect adults. Several metal oxide nanoparticles, biopolymers, and phytochemicals have been discovered to target cancer cells selectively while inflicting low to no damage to healthy cells. Among the existing nanoparticle synthesis methodologies, biologically synthesized nanoparticles using phytochemicals have emerged as a straightforward, economical, and environmentally sound strategy. The synergistic antitumor potential of ZnO-TiO2-chitosan-farnesol nanocomposites (NCs) against leukemia MOLT-4 cells was investigated in the current study. After synthesizing the NCs, characterization of the same was carried out using XRD, DLS, FESEM, TEM, PL, EDX, and FTIR spectroscopy. To analyze its anticancer activity, MOLT-4 cells were cultured and treated at diverse dosages of NCs. The cell viability upon treatment was examined by MTT assay. The morphological and nuclear modifications were observed by dual staining. ROS and MMP levels were observed by DCFH-DA staining and Rh-123 dye, respectively. Furthermore, the caspase 3, 8, and 9 levels were examined by performing ELISA. The XRD patterns exhibited a hexagonal structure of the NCs. In the DLS spectrum, the hydrodynamic diameter of the NCs was observed to be 126.2 nm. The electrostatic interface between the ZnO-TiO2-chitosan-farnesol NCs was confirmed by the FTIR spectra. A significant loss of cell viability in a dosage-dependent trend confirmed the cytotoxic effect of the NCs. An elevated ROS level and MMP depletion suggested apoptosis-associated cell death via the intrinsic pathway, which was confirmed by elevated expressions of caspase 3, 8, and 9 markers. Thus, the results showed that the synthesized NCs demonstrated a remarkable anticancer potential against leukemic cells and can be potentially valuable in cancer treatments. The findings from this study conclude that this is a new approach for modifying the physicochemical characteristics of ZnO-TiO2-chitosan-farnesol composites to increase their properties and synergistically exhibit anticancer properties in human leukemic cancer cells

    Structural, optical, antibacterial, and anticancer properties of cerium oxide nanoparticles prepared by green synthesis using Morinda citrifolia leaves extract

    No full text
    Currently, new advancements in the area of nanotechnology opened up new prospects in the field of medicine that could provide us with a solution for numerous medical complications. Although a several varieties of nanoparticles is being explored to be used as nanomedicines, cerium oxide nanoparticles (CeO2 NPs) are the most attractive due to their biocompatibility and their switchable oxidation state (+3 and +4) or in other words the ability to act as prooxidant and antioxidant depending on the pH condition. Green synthesis of nanoparticles is preferred to make it more economical, eco-friendly, and less toxic. The aim of our study here is to formulate the CeO2 NPs (CeO2 NPs) using Morinda citrifolia (Noni) leaf extract and study its optical, structural, antibacterial, and anticancer abilities. Their optical and structural characterization was accomplished by employing X-ray diffractography (XRD), TEM, EDAX, FTIR, UV-vis, and photoluminescence assays. Our CeO2 NPs expressed strong antibacterial effects against Gram-positive S. aureus and S. pneumonia in addition to Gram-negative E. coli and K. pneumonia when compared with amoxicillin. The anticancer properties of the green synthesized CeO2 NPs against human acute lymphoblastic leukemia (ALL) MOLT-4 cells were further explored by the meticulous study of their ability to diminish cancer cell viability (cytotoxicity), accelerate apoptosis, escalate intracellular reactive oxygen species (ROS) accumulation, decline the mitochondria membrane potential (MMP) level, modify the cell adhesion, and shoot up the activation of proapoptotic markers, caspase-3, -8, and -9, in the tumor cells. Altogether, the outcomes demonstrated that our green synthesized CeO2 NPs are an excellent candidate for alternative cancer therapy

    Synthesis of nickel cobalt-codoped tin oxide nanoparticles from Psidium guajava with anticancer properties

    No full text
    Metal oxide nanoparticles have been found to selectively target the tumor cells while non-toxic to the normal cells. Leukemia is one of the widespread and deadly cancers in adults, as well as the most common cancer in children. Recently, the nanoparticles have evolved as a simple, economic, effective, and ecologically sound strategy among the known nanoparticle synthesis techniques. In the present study, the structural, optical, and antibacterial effects of nickel cobalt-codoped Tin oxide nanoparticles (SnNiCoO2 NPs) formulated by the green process and the anticancer potential of SnNiCoO2 NPs in Molt-4 cells have been studied. The cytotoxic potential of the NPs against Molt-4 cells was estimated by MTT assay. The ROS and MMP levels were measured using fluorescent dyes and the changes in morphology and nuclei were noted using AO/EB staining. CAT, SOD, MDA, and GSH), and Proinflammatory Cytokines (TNF-α and IL1β) were also studied. The activity of caspase-3, −9, and −8 levels was examined to analyze the apoptotic mechanism. The XRD patterns of SnNiCoO2 NPs revealed a tetragonal structure. The SnNiCoO2 NPs was revealed a diameter of 126 nm by the DLS study. The morphology and elemental composition were studied using FESEM and EDAX spectra. In the FT-IR study, the O-sn-O stretching band was found to be 615 and 542 cm-1. The antimicrobial potential of the SnNiCoO2 NPs was examined against S. aureus, E. coli, and C. Albicans strains. A tremendous reduction in the viability of MOLT-4 cells at concentration-dependent mode witnessed the cytotoxic potential of the formulated NPs. The augmented ROS accumulation, depletion of MMP status, depleted antioxidants, and increased proinflammatory cytokines (TNF-α and IL1β) were noted on the NPs exposed cells. Furthermore, the increased expressions of caspase-3, −9, and −8 was also noted in the NPs treated MOLT-4 cells. Hence, the outcomes suggest that the formulated SnNiCoO2 NPs had remarkably potent antimicrobial and anticancer properties and could potentially prove beneficial in cancer treatment. Induces mitochondrial oxidative stress with nickel–cobalt-codoped tin oxide nanoparticles from Psidium guajava, which is a potential drug candidate for the antibiotic, antifungal, and anticancer activities of plant-based nanoparticles

    Synthesis and characterization of ZnO–TiO2–chitosan–escin metallic nanocomposites: Evaluation of their antimicrobial and anticancer activities

    No full text
    This work intended to formulate bio-nanocomposites of zinc oxide (ZnO), titanium oxide (TiO2), chitosan, and escin, characterize their physical properties, and evaluate their antimicrobial and anticancer properties. X-ray diffractometers (XRD) and scanning and transmission electron microscopes were applied to characterize the morphology and ultrastructure of chemically synthesized bio-nanocomposites. To investigate the functional groups of bio-nanocomposites, we used Perkin–Elmer spectrometers for Fourier transform infrared (FTIR) analysis and photoluminescence (PL) spectroscopy for PL spectrum analysis. Antimicrobial activities against bacterial and fungal strains were tested with agar well diffusion. Bio-nanocomposites were tested for anticancer effects on a MOLT4 blood cancer cell line using morphological analysis, methyl thiazole tetrazolium assay, apoptosis by acridine orange/ethidium bromide, and mitochondrial membrane potential (ΔΨm). In XRD, FTIR, and PL, the active compounds of ZnO–TiO2, chitosan, and escin peaks were observed. Our bio-nanocomposites demonstrated antimicrobial activity against bacterial and fungal pathogens. The bio-nanocomposite was cytotoxic to MOLT4 cells at an IC50 concentration of 33.4 µg·mL−1. Bio-nanocomposites caused cytotoxicity, changes in cell morphology, and mitochondrial membrane potential degradation, all of which resulted in apoptotic cell death. MOLT4 cells were found to be responsive to bio-nanocomposites based on ZnO–TiO2–chitosan–escin

    Synthesis, Characterization, and Antimicrobial and Antiproliferative Effects of CuO-TiO2-Chitosan-Escin Nanocomposites on Human Leukemic MOLT4 Cells

    No full text
    Nanocomposites comprised of CuO-TiO2-chitosan-escin, which has adjustable physicochemical properties, provide a solution for therapeutic selectivity in cancer treatment. By controlling the intrinsic signaling primarily through the mitochondrial signaling pathway, we desired nanocomposites with enhanced anticancer activity by containing CuO-TiO2-chitosan-escin. The metal oxides CuO and TiO2, the natural polymer chitosan, and a phytochemical compound escin were combined to form CuO-TiO2-chitosan-escin nanocomposites. The synthesized nanocomposites were confirmed and characterized using FTIR spectroscopy, TEM, and UV-Vis absorption spectroscopy. A human leukemia cell line (MOLT-4) was used to assess the efficacy and selectivity of nanocomposites. Based on a cytotoxicity study, CuO-TiO2-chitosan-escin nanocomposites had inhibition concentrations (IC50) of 13.68, 8.9, and 7.14 µg/mL against human T lymphoblast cells after 24, 48, and 72 h of incubation, respectively. Compared with untreated MOLT-4 cells, CuO-TiO2-chitosan-escin nanocomposite-treated cells significantly increased (p < 0.05) caspase-3, -8, and -9 and decreased the levels of antioxidant enzymes GR, SOD, and GSH. Furthermore, MDA for lipid peroxidase and ROS levels significantly increased (p < 0.05) in the treated cells than in the untreated cells. Remarkably, CuO-TiO2-chitosan-escin nanocomposite-mediated control of cell cycles were mainly achieved through the activation of caspase-3, -8, and -9
    corecore