Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 17: 4383-4400, 2022.
Article in English | MEDLINE | ID: mdl-36164554

ABSTRACT

Purpose: In the search for new drug delivery platforms for cardiovascular diseases and coating of medical devices, we synthesized eptifibatide-functionalized silver nanoparticles (AgNPs-EPI) and examined the pharmacological activity of AgNPs-EPI on platelets and endothelial cells in vitro and ex vivo. Methods: Spherical AgNPs linked to eptifibatide were synthesized and characterized. Cytotoxicity was measured in microvascular endothelial cells (HMEC-1), platelets and red blood cells. Platelet mitochondrial respiration was measured using the Oxygraph-2k, a high-resolution modular respirometry system. The effect of AgNPs-EPI on the aggregation of washed platelets was measured by light aggregometry and the ex vivo occlusion time was determined using a reference laboratory method. The surface amount of platelet receptors such as P-selectin and GPIIb/IIIa was measured. The influence of AgNPS-EPI on blood coagulation science was assessed. Finally, the effect of AgNPs-EPI on endothelial cells was measured by the levels of 6-keto-PGF1alpha, tPa, cGMP and vWF. Results: We describe the synthesis of AgNPs using eptifibatide as the stabilizing ligand. The molecules of this drug are directly bonded to the surface of the nanoparticles. The synthesized AgNPs-EPI did not affect the viability of platelets, endothelial cells and erythrocytes. Preincubation of platelets with AgNPs-EPI protected by mitochondrial oxidative phosphorylation capacity. AgNPs-EPI inhibited aggregation-induced P-selectin expression and GPIIb/IIIa conformational changes in platelets. AgNPs-EPI caused prolongation of the occlusion time in the presence of collagen/ADP and collagen/adrenaline. AgNPs-EPI regulated levels of 6-keto-PGF1alpha, tPa, vWf and cGMP produced in thrombin stimulated HMEC-1 cells. Conclusion: AgNPs-EPI show anti-aggregatory activity at concentrations lower than those required by the free drug acting via regulation of platelet aggregation, blood coagulation, and endothelial cell activity. Our results provide proof-of-principle evidence that AgNPs may be used as an effective delivery platform for antiplatelet drugs.


Subject(s)
Metal Nanoparticles , P-Selectin , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Blood Platelets , Collagen/metabolism , Endothelial Cells/metabolism , Epinephrine/metabolism , Epinephrine/pharmacology , Eptifibatide/pharmacology , Ligands , P-Selectin/metabolism , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Silver/metabolism , Silver/pharmacology , Thrombin/metabolism , von Willebrand Factor/metabolism
2.
Int J Nanomedicine ; 14: 7399-7417, 2019.
Article in English | MEDLINE | ID: mdl-31571858

ABSTRACT

PURPOSE: We studied the effects of silver nanoparticles (AgNPs) on human blood platelet function. We hypothesized that AgNPs, a known antimicrobial agent, can be used as blood-compatible, "ideal material'' in medical devices or as a drug delivery system. Therefore, the aim of the current study was to investigate if functionalized AgNPs affect platelet function and platelets as well as endothelial cell viability in vitro. METHODS: AgNPs, functionalized with reduced glutathione (GSH), polyethylene glycol (PEG) and lipoic acid (LA) were synthesized. Quartz crystal microbalance with dissipation was used to measure the effect of AgNPs on platelet aggregation. Platelet aggregation was measured by changes in frequency and dissipation, and the presence of platelets on the sensor surface was confirmed and imaged by phase contrast microscopy. Flow cytometry was used to detect surface abundance of platelet receptors. Lactate dehydrogenase test was used to assess the potential cytotoxicity of AgNPs on human blood platelets, endothelial cells, and fibroblasts. Commercially available ELISA tests were used to measure the levels of thromboxane B2 and metalloproteinases (MMP-1, MMP-2) released by platelets as markers of platelet activation. RESULTS: 2 nm AgNPs-GSH, 3.7 nm AgNPs-PEG both at 50 and 100 µg/mL, and 2.5 nm AgNPs-LA at 100 µg/mL reduced platelet aggregation, inhibited collagen-mediated increase in total P-selectin and GPIIb/IIIa, TXB2 formation, MMP-1, and MMP-2 release. The tested AgNPs concentrations were not cytotoxic as they did not affect, platelet, endothelial cell, or fibroblast viability. CONCLUSION: All tested functionalized AgNPs inhibited platelet aggregation at nontoxic concentrations. Therefore, functionalized AgNPs can be used as an antiplatelet agent or in design and manufacturing of blood-facing medical devices, such as vascular grafts, stents, heart valves, and catheters.


Subject(s)
Blood Platelets/drug effects , Metal Nanoparticles/chemistry , Platelet Aggregation/drug effects , Silver/pharmacology , Collagen/metabolism , Fibroblasts/drug effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Ligands , Matrix Metalloproteinases/metabolism , Metal Nanoparticles/ultrastructure , P-Selectin/metabolism , Particle Size , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Polyethylene Glycols/chemistry , Quartz Crystal Microbalance Techniques , Spectroscopy, Fourier Transform Infrared , Thromboxane B2/metabolism
3.
Int J Mol Sci ; 20(2)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30634697

ABSTRACT

Pancreatic cancer is characterized by one of the lowest five-year survival rates. In search for new treatments, some studies explored several metal complexes as potential anticancer drugs. Therefore, we investigated three newly synthesized oxidovanadium(IV) complexes with 2-methylnitrilotriacetate (bcma3-), N-(2-carbamoylethyl)iminodiacetate (ceida3-) and N-(phosphonomethyl)-iminodiacetate (pmida4-) ligands as potential anticancer compounds using pancreatic cancer cell lines. We measured: Cytotoxicity using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), neutral red (NR) and lactate dehydrogenase (LDH) assay; antiproliferative activity by bromodeoxyuridine BrdU assay; reactive oxygen species (ROS) generation and cell cycle analysis by flow cytometry; protein level by Western blot and cellular morphology by confocal laser scanning microscopy. The results showed that these oxidovanadium(IV) complexes were cytotoxic on pancreatic cancer cells (PANC-1 and MIA PaCa2), but not on non-tumor human immortalized pancreas duct epithelial cells (hTERT-HPNE) over the concentration range of 10⁻25 µM, following 48 h incubation. Furthermore, molecular mechanisms of cytotoxicity of [4-NH2-2-Me(Q)H][VO(bcma)(H2O)]2H2O (T1) were dependent on antiproliterative activity, increased ROS generation, cell cycle arrest in G2/M phase with simultaneous triggering of the p53/p21 pathway, binucleation, and induction of autophagy. Our study indicates that oxidovanadium(IV) coordination complexes containing 2-methylnitrilotriacetate ligand are good candidates for preclinical development of novel anticancer drugs targeting pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Checkpoints/drug effects , Pancreatic Neoplasms/metabolism , Vanadium Compounds/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Molecular Structure , Reactive Oxygen Species/metabolism , Vanadium Compounds/chemistry , Pancreatic Neoplasms
4.
Chem Biol Interact ; 295: 38-51, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-28641964

ABSTRACT

Nanotechnology is a rapidly developing branch of science, which studies control of phenomena and materials sized below 100 nm. Nanotechnology is applicable in many areas of life and medicine including skin care and personal hygiene. The nanoparticles (NPs) of metals and metal oxides are increasingly used in dermatology and cosmetology, especially in prevention and treatment of bacterial and fungal infections, in protection against the harmful effects of the sun and in preparations reducing the visibility of scars by accelerating the repair processes of skin cells. NPs may also be used for skin care and dermatological treatments to improve the quality of life of patients. Nanodermatology and nanocosmetology offer effective, safe, fast-acting product formulations, thus minimizing the side effects of the products used so far. The unique properties of NPs: high surface area relative to the size as well as the ability to penetrate biological membranes and barriers greatly reduces systemic dose thus potential side effects and toxicity. Recent studies show very promising clinical potential of NPs to serve as controlled release and delivery systems for drugs/active substances. In addition, NPs can be used in diagnostic imaging of skin diseases. However, NPs may also carry a risk of cytotoxicity and side effects. The present review focuses on the use of metal and metal oxide NPs in dermatology and cosmetology and their interactions with skin cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Metal Nanoparticles/chemistry , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin/cytology , Skin/drug effects , Animals , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemistry , Bacteria/drug effects , Biofilms , Fungi/drug effects , Humans , Nanotechnology
5.
PLoS One ; 11(10): e0164137, 2016.
Article in English | MEDLINE | ID: mdl-27716791

ABSTRACT

BACKGROUND: Silver nanoparticles (AgNPs) show strong antibacterial properties, making them excellent candidates to be used in orthopaedic repair and regeneration. However, there are concerns regarding the cytotoxicity of AgNPs and molecular mechanisms underlying AgNPs-induced bone cells toxicity have not been elucidated. Therefore, the aim of our study was to explore mechanisms of AgNPs-induced osteoblast cell death with particular emphasis on the role of nitric oxide (NO) generated by inducible nitric oxide synthase (iNOS). METHODS AND RESULT: Silver nanoparticles used in this study were 18.3±2.6 nm in size, uncoated, spherical, regular shape and their zeta potential was -29.1±2.4 mV as measured by transmission electron microscopy (TEM) and zetasizer. The release of silver (Ag) from AgNPs was measured in cell culture medium by atomic absorption spectroscopy (AAS). The exposure of human osteoblast cells (hFOB 1.19) to AgNPs at concentration of 30 or 60 µg/mL for 24 or 48 hours, respectively resulted in cellular uptake of AgNPs and changes in cell ultrastructure. These changes were associated with apoptosis and necrosis as shown by flow cytometry and lactate dehydrogenase (LDH) assay as well as increased levels of pro-apoptotic Bax and decreased levels of anti-apoptotic Bcl-2 mRNA and protein. Importantly, we have found that AgNPs elevated the levels of nitric oxide (NO) with concomitant upregulation of inducible nitric oxide synthase (iNOS) mRNA and protein. A significant positive correlation was observed between the concentration of AgNPs and iNOS at protein and mRNA level (r = 0.837, r = 0.721, respectively; p<0.001). Finally, preincubation of osteoblast cells with N-iminoethyl-l-lysine (L-NIL), a selective iNOS inhibitor, as well as treating cells with iNOS small interfering RNAs (siRNA) significantly attenuated AgNPs-induced apoptosis and necrosis. Moreover, we have found that AgNPs-induced cells death is not related to Ag dissolution is cell culture medium. CONCLUSION: These results unambiguously demonstrate that increased expression of iNOS and generation of NO as well as NO-derived reactive species is involved in AgNPs-induced osteoblast cell death. Our findings may help in development of new strategies to protect bone from AgNPs-induced cytotoxicity and increase the safety of orthopaedic tissue repair.


Subject(s)
Cell Death/drug effects , Metal Nanoparticles/administration & dosage , Nitric Oxide Synthase Type II/antagonists & inhibitors , Osteoblasts/drug effects , Protective Agents/administration & dosage , Silver/administration & dosage , Cell Line , Humans , Nitric Oxide/metabolism , Osteoblasts/metabolism , Particle Size , RNA, Messenger/metabolism , Up-Regulation/drug effects
6.
Toxicol In Vitro ; 29(4): 663-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25701151

ABSTRACT

Several studies have reported that CuO nanoparticles (CuONPs) have the capacity to cross the blood brain barrier and exert a toxic effect. The aims of our study were to investigate mechanisms underlying CuONPs-induced neurotoxicity in vitro and neuroprotective effects of crocetin. We investigated the toxicological effects of exposure of HT22 hippocampal cells to CuONPs (31 nm) in the presence or absence of crocetin. Crocetin is a carotenoid with wide spectrum of pharmacological effects and the ability to cross blood-brain barrier. Exposure of HT22 cells to CuONPs resulted in: (1) increased cell death in a time- and concentration-dependent manner, with a LC50 of 25.9 µg/ml after 24 h; (2) decreased antioxidant/detoxification enzymes activities: glutathione peroxidase (GPx), superoxide dismutase (SOD), glutathione S-tranferase (GST), and reduced glutathione (GSH) levels; (3) decreased gene expression of GPx and SOD; (4) reactive oxygen species (ROS) generation; (5) enhanced apoptosis; and (6) up-regulation of the pro-apoptotic genes Bax, and down-regulation of anti-apoptotic genes Bcl-2. Importantly, all these effects were significantly attenuated by co-incubation of hippocampal cells with 5 µM crocetin.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Carotenoids/pharmacology , Copper/toxicity , Hippocampus/drug effects , Metal Nanoparticles/toxicity , Animals , Cell Line , Cell Survival/drug effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hippocampus/cytology , Light , Mice , Reactive Oxygen Species/metabolism , Scattering, Radiation , Superoxide Dismutase/metabolism , Vitamin A/analogs & derivatives
7.
Int J Nanomedicine ; 10: 1095-107, 2015.
Article in English | MEDLINE | ID: mdl-25709434

ABSTRACT

Titanium dioxide (TiO2) nanoparticles (NPs) are manufactured worldwide for a variety of engineering and bioengineering applications. TiO2NPs are frequently used as a material for orthopedic implants. However, to the best of our knowledge, the biocompatibility of TiO2NPs and their effects on osteoblast cells, which are responsible for the growth and remodeling of the human skeleton, have not been thoroughly investigated. In the research reported here, we studied the effects of exposing hFOB 1.19 human osteoblast cells to TiO2NPs (5-15 nm) for 24 and 48 hours. Cell viability, alkaline phosphatase (ALP) activity, cellular uptake of NPs, cell morphology, superoxide anion (O2 (•-2)) generation, superoxide dismutase (SOD) activity and protein level, sirtuin 3 (SIR3) protein level, correlation between manganese (Mn) SOD and SIR, total antioxidant capacity, and malondialdehyde were measured following exposure of hFOB 1.19 cells to TiO2NPs. Exposure of hFOB 1.19 cells to TiO2NPs resulted in: (1) cellular uptake of NPs; (2) increased cytotoxicity and cell death in a time- and concentration-dependent manner; (3) ultrastructure changes; (4) decreased SOD and ALP activity; (5) decreased protein levels of SOD1, SOD2, and SIR3; (6) decreased total antioxidant capacity; (7) increased O2 (•-) generation; and (8) enhanced lipid peroxidation (malondialdehyde level). The linear relationship between the protein level of MnSOD and SIR3 and between O2 (•-) content and SIR3 protein level was observed. Importantly, the cytotoxic effects of TiO2NPs were attenuated by the pretreatment of hFOB 1.19 cells with SOD, indicating the significant role of O2 (•-) in the cell damage and death observed. Thus, decreased expression of SOD leading to increased oxidizing stress may underlie the nanotoxic effects of TiO2NPs on human osteoblasts.


Subject(s)
Nanoparticles , Osteoblasts , Superoxides/metabolism , Titanium , Cell Line , Cell Survival/drug effects , Humans , Nanoparticles/chemistry , Nanoparticles/toxicity , Osteoblasts/drug effects , Osteoblasts/metabolism , Titanium/chemistry , Titanium/toxicity
8.
J Biomed Nanotechnol ; 10(6): 1004-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24749395

ABSTRACT

The increasing use of gold nanoparticles in medical diagnosis and treatment has raised the concern over their blood compatibility. The interactions of nanoparticles with blood components may lead to platelet aggregation and endothelial dysfunction. Therefore, medical applications of gold nanoparticles call for increased nanoparticle stability and biocompatibility. Functionalisation of nanoparticles with polythelene glycol (PEGylation) is known to modulate cell-particle interactions. Therefore, the aim of the current study was to investigate the effects of PEGylated-gold nanoparticles on human platelet function and endothelial cells in vitro. Gold nanoparticles, 15 nm in diameter, were synthesised in water using sodium citrate as a reducing and stabilising agent. Functionalised polyethylene glycol-based thiol polymers were used to coat and stabilise pre-synthesised gold nanoparticles. The interaction of gold nanoparticles-citrate and PEGylated-gold nanoparticles with human platelets was measured by Quartz Crystal Microbalance with Dissipation. Platelet-nanoparticles interaction was imaged using phase-contrast, scanning and transmission electron microscopy. The inflammatory effects of gold nanoparticles-citrate and PEGylated-gold nanoparticles in endothelial cells were measured by quantitative real time polymerase chain reaction. PEGylated-gold nanoparticles were stable under physiological conditions and PEGylated-gold nanoparticles-5400 and PEGylated-gold nanoparticles-10800 did not affect platelet aggregation as measured by Quartz Crystal Microbalance with Dissipation. In addition, PEGylated-gold nanoparticles did not induce an inflammatory response when incubated with endothelial cells. Therefore, this study shows that PEGylated-gold nanoparticles with a higher molecular weight of the polymer chain are both platelet- and endothelium-compatible making them attractive candidates for biomedical applications.


Subject(s)
Biocompatible Materials/pharmacology , Blood Platelets/physiology , Gold/pharmacology , Metal Nanoparticles/administration & dosage , Nanocapsules/chemistry , Platelet Activation/physiology , Polyethylene Glycols/chemistry , Biocompatible Materials/chemical synthesis , Blood Platelets/cytology , Blood Platelets/drug effects , Cells, Cultured , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Particle Size , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Polyethylene Glycols/pharmacology
9.
Int J Nanomedicine ; 7: 243-55, 2012.
Article in English | MEDLINE | ID: mdl-22275839

ABSTRACT

Interactions between blood platelets and nanoparticles have both pharmacological and toxicological significance and may lead to platelet activation and aggregation. Platelet aggregation is usually studied using light aggregometer that neither mimics the conditions found in human microvasculature nor detects microaggregates. A new method for the measurement of platelet microaggregation under flow conditions using a commercially available quartz crystal microbalance with dissipation (QCM-D) has recently been developed. The aim of the current study was to investigate if QCM-D could be used for the measurement of nanoparticle-platelet interactions. Silica, polystyrene, and gold nanoparticles were tested. The interactions were also studied using light aggregometry and flow cytometry, which measured surface abundance of platelet receptors. Platelet activation was imaged using phase contrast and scanning helium ion microscopy. QCM-D was able to measure nanoparticle-induced platelet microaggregation for all nanoparticles tested at concentrations that were undetectable by light aggregometry and flow cytometry. Microaggregates were measured by changes in frequency and dissipation, and the presence of platelets on the sensor surface was confirmed and imaged by phase contrast and scanning helium ion microscopy.


Subject(s)
Blood Platelets/cytology , Nanoparticles/chemistry , Platelet Aggregation/physiology , Quartz Crystal Microbalance Techniques/methods , Blood Platelets/chemistry , Blood Platelets/metabolism , Flow Cytometry , Gold/chemistry , Humans , Microscopy , P-Selectin/analysis , Particle Size , Polystyrenes/chemistry , Silicon Dioxide/chemistry
10.
Analyst ; 136(24): 5120-6, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22029043

ABSTRACT

Platelet aggregation is essential for vascular haemostasis and thrombosis. To improve the therapy of arterial thrombotic disorders and identify novel therapeutic targets it is imperative to study basic mechanisms of platelet thrombus formation. To date most data on biology, physiology and pharmacology of platelet aggregation have been obtained by studying this phenomenon under static or quasi-dynamic conditions at the macroscale level. There is a widespread recognition for the need of new technologies that will help to further elucidate the role of platelets in physiological and pathological thrombus formation and to design more effective and specific antithrombotic drugs. Micro- and nanofluidic devices, capable of reaching nanoscale resolution, can be used for this purpose setting the scene for the development of novel methods for studying platelet function in physiology, pathology and therapeutics.


Subject(s)
Blood Platelets/physiology , Microfluidics , Nanotechnology , Blood Platelets/metabolism , Humans , Platelet Aggregation , Quartz Crystal Microbalance Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...