Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Biol Macromol ; 143: 665-676, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31830450

ABSTRACT

Nanoparticles (NPs) have been widely used for immobilization of wide ranges of enzymes. However, the stabilization of enzymes on NPs is a major challenge, crucial for regulating enzymatic activity and their medical applications. To overcome these challenges, it is necessary to explore how enzymes attach to nanomaterials and their properties are affected by such interactions. In this review we present an overview on the different strategies of the enzyme immobilization into the NPs and their corresponding stability against temperature and pH. The effects of surface charge, particle size, morphology, and aggregation of NPs on the stability of immobilized enzymes were summarized. The activity of immobilized enzyme into the NPs was reviewed to disclose more detail regarding the interaction of biomolecules with NPs. The combination of enzyme immobilization with prodrugs was also reviewed as a promising approach for biomedical application of enzyme in cancer therapy. Finally, the current challenges and future applications of NPs in enzyme immobilization and the utilization of immobilized enzyme toward prodrug activation in cytoplasm of cancer cells were presented. In conclusion, this review may pave the way for providing a perspective on development to the industrial and clinical translation of immobilized enzymes.


Subject(s)
Enzymes, Immobilized/metabolism , Nanostructures/chemistry , Neoplasms/drug therapy , Prodrugs/therapeutic use , Animals , Enzyme Stability , Humans , Nanostructures/ultrastructure
2.
Int J Pharm ; 572: 118824, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31715345

ABSTRACT

In this study, we formulated silymarin-HSA nanoplex and assayed its ability to reduce LPS-induced toxicity in vitro and in vivo. Silymarin molecules were encapsulated into HSA nanoplex and the loading efficiency and characterization of fabricated nanoplex were performed by using HPLC, TEM, SEM, DLS, FTIR analysis, and theoretical studies. Afterwards, their protective effect against LPS (20 µg/ml) -induced toxicity in SH-SY5Y cells was investigated by MTT, ROS, and apoptosis assays. For in vivo experiments, rats were pre-treated with either silymarin or silymarin -HSA nanoplex (200 mg/kg) orally for 3 days and at third day received LPS by IP at a dose of 0.5 mg/kg, 150 min before scarification followed by SOD and CAT activity assay. The formulation of silymarin-HSA nanoplex showed a spherical shape with an average diameter between 50 nm and 150 nm, hydrodynamic radius of 188.3 nm, zeta potential of -26.6 mV, and a drug loading of 97.3%. In LPS-treated cells, pretreatments with silymarin-HSA noncomplex recovered the cell viability and decreased the ROS level and corresponding apoptosis more significantly than free silymarin. In rats, it was also depicted that, silymarin-HSA noncomplex can increase the SOD and CAT activity in brain tissue at LPS-triggered oxidative stress model more significantly than the free counterpart. Therefore, nanoformulation of silymarin improved its capability to reduce LPS-induced oxidative stress by restoring cell viability and elevation of SOD and CAT activity in vitro and in vivo, respectively. In conclusion, formulation of silymarin may hold a great promise in the development of antioxidant agents.


Subject(s)
Antioxidants/pharmacology , Oxidative Stress/drug effects , Serum Albumin, Human/chemistry , Silymarin/pharmacology , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lipopolysaccharides/toxicity , Male , Neuroblastoma/pathology , Particle Size , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Silymarin/administration & dosage
3.
Int J Nanomedicine ; 14: 243-256, 2019.
Article in English | MEDLINE | ID: mdl-30643404

ABSTRACT

BACKGROUND: Nanoparticles (NPs) have been emerging as potential players in modern medicine with clinical applications ranging from therapeutic purposes to antimicrobial agents. However, before applications in medical agents, some in vitro studies should be done to explore their biological responses. AIM: In this study, protein binding, anticancer and antibacterial activates of zero valent iron (ZVFe) were explored. MATERIALS AND METHODS: ZVFe nanoparticles were synthesized and fully characterized by X-ray diffraction, field-emission scanning electron microscope, and dynamic light scattering analyses. Afterward, the interaction of ZVFe NPs with human serum albumin (HSA) was examined using a range of techniques including intrinsic fluorescence, circular dichroism, and UV-visible spectroscopic methods. Molecular docking study was run to determine the kind of interaction between ZVFe NPs and HSA. The anticancer influence of ZVFe NPs on SH-SY5Y was examined by MTT and flow cytometry analysis, whereas human white blood cells were used as the control cell. Also, the antibacterial effect of ZVFe NPs was examined on Pseudomonas aeruginosa (ATCC 27853), Escherichia coli (ATCC 25922), and Staphylococcus aureus (ATCC 25923). RESULTS: X-ray diffraction, transmission electron microscope, and dynamic light scattering analyses verified the synthesis of ZVFe NPs in a nanosized diameter. Fluorescence spectroscopy analysis showed that ZVFe NPs spontaneously formed a complex with HSA through hydrogen bonds and van der Waals interactions. Also, circular dichroism spectroscopy study revealed that ZVFe NPs did not change the secondary structure of HSA. Moreover, UV-visible data presented that melting temperature (Tm) of HSA in the absence and presence of ZVFe NPs was almost identical. Molecular dynamic study also showed that ZVFe NP came into contact with polar residues on the surface of HSA molecule. Cellular assays showed that ZVFe NPs can induce cell mortality in a dose-dependent manner against SH-SY5Y cells, whereas these NPs did not trigger significant cell mortality against normal white bloods in the concentration range studied (1-100 µg/mL). Antibacterial assays showed a noteworthy inhibition on both bacterial strains. CONCLUSION: In conclusion, it was revealed that ZVFe NPs did not induce a substantial influence on the structure of protein and cytotoxicity against normal cell, whereas they derived significant anticancer and antibacterial effects.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Bacteria/drug effects , Bacteria/metabolism , Iron/administration & dosage , Metal Nanoparticles/administration & dosage , Serum Albumin, Human/metabolism , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Escherichia coli/drug effects , Escherichia coli/metabolism , Humans , Iron/chemistry , Metal Nanoparticles/chemistry , Molecular Docking Simulation , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Protein Conformation , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Serum Albumin, Human/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...