6 research outputs found

    Effects of Albumin–Chlorogenic Acid Nanoparticles on Apoptosis and PI3K/Akt/mTOR Pathway Inhibitory Activity in MDA-MB-435s Cells

    No full text
    In this study, we synthesized, characterized, and explored the anti-microbial and anti-cancer effects of albumin–chlorogenic acid nanoparticles (NPs). Characterization studies with a UV-vis spectrophotometer, FTIR, PL spectrum, TEM, FESEM, XRD, and DLA analysis showed patterns confirming the physio–chemical nature of biogenic nanocomposites. Further, anti-microbial studies using bacterial strains Staphylococcus aureus, Streptococcus pneumonia, Bacillus subtilis, Escherichia coli, Pseudomonas aeruginosa, Vibrio cholera, and fungal strain Candida albicans showed significant (p < 0.05) anti-bacterial and anti-fungal activities. Next, we used MDA-MB-435s, a human cell line, to evaluate the anti-cancer effects of albumin–chlorogenic acid NPs. Cytotoxic studies revealed its IC50 concentration at 24 μg/mL after a 24 h treatment of MDA-MB-435s cells. We chose this IC50 dose to analyze albumin–chlorogenic acid NPs anti-cancer effects in vitro. MDA-MB-435s cells exposed to our NPs were studied via AO/EtBr staining, cell cycle analyses via PI staining, the status of whole genomic damage via comet assay, levels of apoptotic cells via annexin V/PI staining, ROS generation via DCFH-DA staining, an assay of antioxidant enzymes catalase, superoxide dismutase, and antioxidant GSH, via ELISA analyses of apoptotic markers caspase-3, 8, 9, Bax, Bcl-2, CytC, and p53, PI3/AKT/mTOR pathway. Our results collectively showed albumin–chlorogenic acid NPs induced apoptosis via p53-dependent and PI3/AKT/mTOR inhibition in MDA-MB-435s cells. Our results denote albumin–chlorogenic acid NPs can be used as an effective candidate for anti-microbial and anti-cancer applications; however, further in vivo confirmatory studies are warranted

    Therapeutic Potential of Albumin Nanoparticles Encapsulated Visnagin in MDA-MB-468 Triple-Negative Breast Cancer Cells

    No full text
    Breast cancer is among the most recurrent malignancies, and its prevalence is rising. With only a few treatment options available, there is an immediate need to search for better alternatives. In this regard, nanotechnology has been applied to develop potential chemotherapeutic techniques, particularly for cancer therapy. Specifically, albumin-based nanoparticles are a developing platform for the administration of diverse chemotherapy drugs owing to their biocompatibility and non-toxicity. Visnagin, a naturally derived furanochromone, treats cancers, epilepsy, angina, coughs, and inflammatory illnesses. In the current study, the synthesis and characterization of albumin visnagin (AV) nanoparticles (NPs) using a variety of techniques such as transmission electron microscopy, UV-visible, Fourier transform infrared, energy dispersive X-ray composition analysis, field emission scanning electron microscopy, photoluminescence, X-Ray diffraction, and dynamic light scattering analyses have been carried out. The MTT test, dual AO/EB, DCFH-DA, Annexin-V-FITC/PI, Propidium iodide staining techniques as well as analysis of apoptotic proteins, antioxidant enzymes, and PI3K/Akt/mTOR signaling analysis was performed to examine the NPs’ efficacy to suppress MDA-MB-468 cell lines. The NPs decreased cell viability increased the amount of ROS in the cells, disrupted membrane integrity, decreased the level of antioxidant enzymes, induced cell cycle arrest, and activated the PI3K/Akt/mTOR signaling cascade, ultimately leading to cell death. Thus, AV NPs possesses huge potential to be employed as a strong anticancer therapy alternative

    In vitro anti-cancer and antimicrobial effects of manganese oxide nanoparticles synthesized using the Glycyrrhiza uralensis leaf extract on breast cancer cell lines

    No full text
    In this study, we evaluated the antiproliferative and apoptotic properties of Pluronic-F127-containing manganese oxide nanoparticles (PF-127-coated Mn2O3 NPs) derived from the leaf extract of Glycyrrhiza uralensis (GU) on breast adenocarcinoma, MCF7, and MDA-MB-231 cell lines. The leaf extract of GU contains bioactive molecules that act as a reducing or capping agent to form Mn2O3 NPs. Various analytical techniques were used to characterize the physiochemical properties of PF-127-coated Mn2O3 NPs, including spectroscopy (ultralight-Vis, Fourier transform infrared, photoluminescence), electron microscopy (field emission scanning electron microscopy and transmission electron microscopy), X-ray diffraction (XRD), electron diffracted X-ray spectroscopy (EDAX), and dynamic light scattering. The average crystallite size of Mn2O3 NPs was estimated to be 80 nm, and the NPs had a cubic crystalline structure. PF127-encapsulated Mn2O3 NPs significantly reduce MDA-MB-231 and MCF-7 cell proliferation, while increasing endogenous ROS and lowering mitochondrial matrix protein levels. DAPI, EtBr/AO dual staining, and Annexin-V-FITC-based flow cytometry analysis revealed that PF127-coated Mn2O3 NP-treated breast cancer cells exhibit nuclear damage and apoptotic cell death, resulting in cell cycle arrest in the S phase. Furthermore, PF127-encapsulated Mn2O3 NPs show strong antimicrobial efficacy against various strains. As a result, we can conclude that PF127-coated Mn2O3 NPs may be effective as future anticancer agents and treatment options for breast cancer

    Manganese and copper-coated nickel oxide nanoparticles synthesized from Carica papaya leaf extract induce antimicrobial activity and breast cancer cell death by triggering mitochondrial caspases and p53

    No full text
    In the present work, manganese–copper co-infused nickel oxide nanoparticles (MnCu co-doped NiO NPs) were formulated via a green process using Carica papaya extract. The MnCu co-doped NiO NPs were characterized by X-ray diffraction (XRD), UV–Vis, Fourier transform infrared, field emission scanning electron microscope, energy dispersive X-ray analysis, and photoluminescence (PL) spectrum. The XRD pattern demonstrated that synthesized MnCu codoped NiO NPs exhibit cubic structure. On the PL spectrum, various surface defects were identified. MnCu co-doped NiO NPs exhibited ferromagnetic properties at 37°C. The antimicrobial activity of green synthesis MnCu co-doped NiO NPs against human pathogens (Escherichia coli, Streptococcus pneumoniae, Bacillus megaterium, Bacillus subtilis, Shigella dysenteriae, Pseudomonas aeruginosa) and Candida albicans as fungal strains were demonstrated. The MnCu co-doped NiO NPs treatment considerably reduced MDA-MB-231 cell viability while not disturbing HBL-100 cell viability. Different fluorescent staining analyses revealed that MnCu co-doped NiO NPs induced nuclear and mitochondrial damage to improve free radical production, altering mitochondrial membrane protein potential, which led to apoptotic cell death in MDA-MB-231 cells. The MnCu co-doped NiO NP treatment enhanced pro-apoptotic protein expression and inhibited the cell cycle at the S phase in MDA-MB-231 cells. This makes it easy, cheap, and environmentally friendly to make MnCu co-doped NiO NPs using C. papaya extract, which has excellent antimicrobial properties

    Manganese and copper-coated nickel oxide nanoparticles synthesized from Carica papaya leaf extract induce antimicrobial activity and breast cancer cell death by triggering mitochondrial caspases and p53

    No full text
    In the present work, manganese–copper co-infused nickel oxide nanoparticles (MnCu co-doped NiO NPs) were formulated via a green process using Carica papaya extract. The MnCu co-doped NiO NPs were characterized by X-ray diffraction (XRD), UV–Vis, Fourier transform infrared, field emission scanning electron microscope, energy dispersive X-ray analysis, and photoluminescence (PL) spectrum. The XRD pattern demonstrated that synthesized MnCu co-doped NiO NPs exhibit cubic structure. On the PL spectrum, various surface defects were identified. MnCu co-doped NiO NPs exhibited ferromagnetic properties at 37°C. The antimicrobial activity of green synthesis MnCu co-doped NiO NPs against human pathogens (Escherichia coli, Streptococcus pneumoniae, Bacillus megaterium, Bacillus subtilis, Shigella dysenteriae, Pseudomonas aeruginosa) and Candida albicans as fungal strains were demonstrated. The MnCu co-doped NiO NPs treatment considerably reduced MDA-MB-231 cell viability while not disturbing HBL-100 cell viability. Different fluorescent staining analyses revealed that MnCu co-doped NiO NPs induced nuclear and mitochondrial damage to improve free radical production, altering mitochondrial membrane protein potential, which led to apoptotic cell death in MDA-MB-231 cells. The MnCu co-doped NiO NP treatment enhanced pro-apoptotic protein expression and inhibited the cell cycle at the S phase in MDA-MB-231 cells. This makes it easy, cheap, and environmentally friendly to make MnCu co-doped NiO NPs using C. papaya extract, which has excellent antimicrobial properties

    Enhanced apoptotic activity of Pluronic F127 polymer-encapsulated chlorogenic acid nanoparticles through the PI3K/Akt/mTOR signaling pathway in liver cancer cells and in vivo toxicity studies in zebrafish

    No full text
    In this study, chlorogenic acid nanoparticles encapsulated in Pluronic F127 polymer were synthesized and characterized to determine if they could treat human liver cancer. The nanoparticles were synthesized using standard procedures and characterized using physical and biological techniques such as X-ray diffraction, Fourier transform infrared spectroscopy, UV-Vis, dynamic light scattering, Photoluminescence, scanning electron microscopy, and transmission electron microscopy. The anticancer effects were assessed using MTT analysis, acridine orange/ethidium bromide, reactive oxygen species (ROS), COMET assay, annexin-V/FITC, cell cycle analysis, and expression of marker genes against HepG2 cell lines. The results showed significant cytotoxicity, apoptosis induction, and increased ROS production in treated cells compared to control cells. The nanoparticles also activated the apoptotic cascade and regulated the PI3K/AKT/mTOR pathways. The nanocomposites exhibited unique characteristics such as anticancer efficacy in vitro. Further research was conducted using zebrafish to model hematological parameters, liver enzymes, and histopathology to study effectiveness. Green-synthesized Pluronic F127–chlorogenic acid nanoparticles can be considered potential cancer therapy agents
    corecore