Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
ACS Appl Mater Interfaces ; 13(31): 37535-37544, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34324300

ABSTRACT

Combined therapeutic strategies for bacterial infection have attracted worldwide attention owing to their faster and more effective therapy with fewer side effects compared with monotherapy. In this work, gold-platinum nanodots (AuPtNDs) are simply and quickly synthesized by a one-step method. They not only exhibit powerful peroxidase-like activity but also confer a higher affinity for hydrogen peroxide (H2O2), which is 3.4 times that of horseradish peroxidase. Under 808 nm laser irradiation, AuPtNDs also have excellent photothermal conversion efficiency (50.53%) and strong photothermal stability. Excitingly, they can combat bacterial infection through the combination of chemodynamic and photothermal therapy. In vitro antibacterial results show that the combined antibacterial strategy has a broad-spectrum antibacterial property against both Escherichia coli (Gram negative, 97.1%) and Staphylococcus aureus (Gram positive, 99.3%). Animal experiments further show that nanodots can effectively promote the healing of bacterial infection wounds. In addition, owing to good biocompatibility and low toxicity, they are hardly traceable in the main organs of mice, which indicates that they can be well excreted through metabolism. These results reveal the application potential of AuPtNDs as a simple and magic multifunctional nanoparticle in antibacterial therapy and open up new applications for clinical anti-infective therapy in the near future.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Quantum Dots/therapeutic use , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/radiation effects , Anti-Bacterial Agents/toxicity , Catalysis , Escherichia coli/drug effects , Gold/chemistry , Gold/radiation effects , Gold/therapeutic use , Gold/toxicity , HEK293 Cells , Humans , Infrared Rays , Mice, Inbred BALB C , Microbial Sensitivity Tests , Photothermal Therapy , Platinum/chemistry , Platinum/radiation effects , Platinum/therapeutic use , Platinum/toxicity , Quantum Dots/chemistry , Quantum Dots/radiation effects , Quantum Dots/toxicity , Staphylococcus aureus/drug effects , Wound Healing/drug effects
2.
ACS Appl Mater Interfaces ; 13(30): 35328-35341, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34291912

ABSTRACT

The multifunctional combined nanoplatform has a wide application prospect in the synergistic treatment of cancer. Nevertheless, the traditional treatment of phototherapy is limited by the catalytic nanomaterial itself, so the effect is not satisfactory. Here, the arris of the anisotropic truncated octahedral Au (TOh Au) was coated with noble metal Pt to form a spatial separation structure, which enhanced the local surface plasmonic resonance and thus boosted the photocatalytic effect. In this system, the highly efficient photocatalysis provides a strong guarantee for oncotherapy. On the one hand, the structure of arris deposition adequately improves the efficiency of photothermal conversion, which substantially improves the effectiveness of photothermal therapy. On the other hand, in situ oxygen production of Pt ameliorates tumor hypoxia, and through the O2 self-production and sales mode, the growth and development of tumor were inhibited. Meanwhile, under the enhanced photocatalysis, more O2 were produced, which greatly evolved the treatment effect of photodynamic therapy. In the end, the addition of hyaluronic acid can specifically target osteosarcoma cells while improving the retention time and biocompatibility of the material in the body. Thus, the nanocomposite shows superexcellent synergistic enhancement of photothermal conversion efficiency and photodynamic capability in vitro and in vivo, which provides a potential possibility for osteosarcoma cure.


Subject(s)
Antineoplastic Agents/therapeutic use , Metal Nanoparticles/therapeutic use , Osteosarcoma/drug therapy , Photosensitizing Agents/therapeutic use , Animals , Anisotropy , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , Catalysis/radiation effects , Chlorophyllides , Gold/chemistry , Gold/toxicity , Hyaluronic Acid/chemistry , Hyaluronic Acid/toxicity , Infrared Rays , Metal Nanoparticles/chemistry , Metal Nanoparticles/radiation effects , Metal Nanoparticles/toxicity , Mice, Nude , Osteosarcoma/metabolism , Oxygen/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity , Photothermal Therapy , Platinum/chemistry , Platinum/toxicity , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Porphyrins/chemistry , Porphyrins/radiation effects , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Surface Plasmon Resonance
3.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32947930

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor in childhood. The different treatments available for neuroblastoma are challenged by high rates of resistance, recurrence, and progression, most notably in advanced cases and highly malignant tumors. Therefore, the development of more targeted therapies, which are biocompatible and without undesired side effects, is highly desirable. The mechanisms of actions of platinum nanoparticles (PtNPs) and retinoic acid (RA) in neuroblastoma have remained unclear. In this study, the anticancer effects of PtNPs and RA on neuroblastoma were assessed. We demonstrated that treatment of SH-SY5Y cells with the combination of PtNPs and RA resulted in improved anticancer effects. The anticancer effects of the two compounds were mediated by cytotoxicity, oxidative stress (OS), mitochondrial dysfunction, endoplasmic reticulum stress (ERS), and apoptosis-associated networks. Cytotoxicity was confirmed by leakage of lactate dehydrogenase (LDH) and intracellular protease, and oxidative stress increased the level of reactive oxygen species (ROS), 4-hydroxynonenal (HNE), malondialdehyde (MDA), and nitric oxide (NO), and protein carbonyl content (PCC). The combination of PtNPs and RA caused mitochondrial dysfunction by decreasing the mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) content, number of mitochondria, and expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Endoplasmic reticulum-mediated stress and apoptosis were confirmed by upregulation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), activating transcription factor 4 (ATF4), p53, Bax, and caspase-3 and down regulation of B-cell lymphoma 2 (BCl-2). PtNPs and RA induced apoptosis, and oxidative DNA damage was evident by the accumulation of 8-hydroxy-2-deoxyguanosine (8-OHdG) and 8-hydroxyguanosine (8-OHG). Finally, PtNPs and RA increased the differentiation and expression of differentiation markers. Differentiated SH-SY5Y cells pre-treated with PtNPs or RA or the combination of both were more sensitive to the cytotoxic effect of cisplatin than undifferentiated cells. To our knowledge, this is the first study to demonstrate the effect of the combination of PtNPs and RA in neuroblastoma cells. PtNPs may be a potential preconditioning or adjuvant compound in chemotherapeutic treatment. The results of this study provide a rationale for clinical evaluation of the combination of PtNPs and RA for the treatment of children suffering from high-risk neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Metal Nanoparticles/therapeutic use , Neuroblastoma/drug therapy , Platinum/pharmacology , Tretinoin/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antioxidants/metabolism , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , L-Lactate Dehydrogenase/analysis , Membrane Potential, Mitochondrial/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Neoplasm Proteins/metabolism , Neuroblastoma/pathology , Oxidative Stress/drug effects , Peptide Hydrolases/analysis , Platinum/administration & dosage , Platinum/toxicity , Tretinoin/administration & dosage , beta Carotene/pharmacology
4.
Free Radic Biol Med ; 160: 263-276, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32827639

ABSTRACT

Platinum-based compounds are widely used for the treatment of different malignancies due to their high effectiveness. Unfortunately, platinum-based treatment may lead to ototoxicity, an often-irreversible side effect without a known effective treatment and prevention plan. Platinum-based compound-related ototoxicity results mainly from the production of toxic levels of reactive oxygen species (ROS) rather than DNA-adduct formation, which has led to test strategies based on direct ROS scavengers to ameliorate hearing loss. However, favorable clinical results have been associated with several complications, including potential interactions with chemotherapy efficacy. To understand the contribution of the different cytotoxic mechanisms of platinum analogues on malignant cells and auditory cells, the particular susceptibility and response of both kinds of cells to molecules that potentially interfere with these mechanisms, is fundamental to develop innovative strategies to prevent ototoxicity without affecting antineoplastic effects. The n-3 long-chain polyunsaturated fatty acids (n-3 PUFAs) have been tried in different clinical settings, including with cancer patients. Nevertheless, their use to decrease cisplatin-induced ototoxicity has not been explored to date. In this hypothesis paper, we address the mechanisms of platinum compounds-derived ototoxicity, focusing on the differences between the effects of these compounds in neoplastic versus auditory cells. We discuss the basis for a strategic use of n-3 PUFAs to potentially protect auditory cells from platinum-derived injury without affecting neoplastic cells and chemotherapy efficacy.


Subject(s)
Antineoplastic Agents , Fatty Acids, Omega-3 , Ototoxicity , Antineoplastic Agents/toxicity , Carboplatin , Cisplatin/toxicity , Humans , Oxidative Stress , Platinum/toxicity
5.
J Inorg Biochem ; 178: 94-105, 2018 01.
Article in English | MEDLINE | ID: mdl-29125948

ABSTRACT

Here we present the preparation of 14 pairs of cis- and trans-diammine monochlorido platinum(II) complexes, coordinated to heterocycles (i.e., imidazole, 2-methylimidazole and pyrazole) and linked to various acylhydrazones, which were designed as potential inhibitors of the selenium-dependent enzymes glutathione peroxidase 1 (GPx-1) and thioredoxin reductase 1 (TrxR-1). However, no inhibition of bovine GPx-1 and only weak inhibition of murine TrxR-1 was observed in in vitro assays. Nonetheless, the cis configured diammine monochlorido Pt(II) complexes exhibited cytotoxic and apoptotic properties on various human cancer cell lines, whereas the trans configured complexes generally showed weaker potency with a few exceptions. On the other hand, the trans complexes were generally more likely to lack cross-resistance to cisplatin than the cis analogues. Platinum was found bound to the nuclear DNA of cancer cells treated with representative Pt complexes, suggesting that DNA might be a possible target. Thus, detailed in vitro binding experiments with DNA were conducted. Interactions of the compounds with calf thymus DNA were investigated, including Pt binding kinetics, circular dichroism (CD) spectral changes, changes in DNA melting temperatures, unwinding of supercoiled plasmids and ethidium bromide displacement in DNA. The CD results indicate that the most active cis configured pyrazole-derived complex causes unique structural changes in the DNA compared to the other complexes as well as to those caused by cisplatin, suggesting a denaturation of the DNA structure. This may be important for the antiproliferative activity of this compound in the cancer cells.


Subject(s)
Aspartic Acid/analogs & derivatives , Chondroitin/analogs & derivatives , DNA/drug effects , Glutathione Peroxidase/antagonists & inhibitors , Organoplatinum Compounds/chemical synthesis , Platinum/pharmacology , Selenium/pharmacology , Animals , Aspartic Acid/chemistry , Aspartic Acid/pharmacology , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Chondroitin/chemistry , Chondroitin/pharmacology , DNA/chemistry , Enzyme Activation/drug effects , Enzymes/metabolism , Inhibitory Concentration 50 , Mice , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Oxidation-Reduction , Platinum/chemistry , Platinum/toxicity , Selenium/chemistry , Selenium/toxicity
6.
Bone ; 53(2): 421-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23313279

ABSTRACT

Platinum group metals (PGMs), i.e., palladium (Pd), platinum (Pt) and rhodium (Rh), are found at pollutant levels in the environment and are known to accumulate in plant and animal tissues. However, little is known about PGM toxicity. Our previous studies showed that chick embryos exposed to PGM concentrations of 1mL of 5.0ppm (LD50) and higher exhibited severe skeletal deformities. This work hypothesized that 1.0ppm doses of PGMs will negatively impact the mineralization process in tibiotarsi. One milliliter of 1.0ppm of Pd(II), Pt(IV), Rh(III) aqueous salt solutions and a PGM-mixture were injected into the air sac on the 7th and 14th day of incubation. Control groups with no-injection and vehicle injections were included. On the 20th day, embryos were sacrificed to analyze the PGM effects on tibiotarsi using four spectroscopic techniques. 1) Micro-Raman imaging: Hyperspectral Raman data were collected on paraffin embedded cross-sections of tibiotarsi, and processed using in-house-written MATLAB codes. Micro-Raman univariate images that were created from the ν1(PO4(3-)) integrated areas revealed anomalous mineral inclusions within the bone marrow for the PGM-mixture treatment. The age of the mineral crystals (ν(CO3(2-))/ν1(PO4(3-))) was statistically lower for all treatments when compared to controls (p≤0.05). 2) FAAS: The percent calcium content of the chemically digested tibiotarsi in the Pd and Pt groups changed by ~45% with respect to the no-injection control (16.1±0.2%). 3) Micro-XRF imaging: Abnormal calcium and phosphorus inclusions were found within the inner longitudinal sections of tibiotarsi for the PGM-mixture treatment. A clear increase in the mineral content was observed for the outer sections of the Pd treatment. 4) ICP-OES: PGM concentrations in tibiotarsi were undetectable (<5ppb). The spectroscopic techniques gave corroborating results, confirmed the hypothesis, and explained the observed pathological (skeletal developmental abnormalities) and histological changes (tibiotarsus ischemia and nuclear fragmentation in chondrocytes).


Subject(s)
Palladium/toxicity , Platinum/toxicity , Rhodium/toxicity , Animals , Calcium/metabolism , Chick Embryo , Environmental Monitoring , Phosphorus/metabolism
7.
Audiol Neurootol ; 17(5): 321-30, 2012.
Article in English | MEDLINE | ID: mdl-22739546

ABSTRACT

OBJECTIVE: To investigate the use of hearing preservation cochlear implantation in children with partial deafness. PATIENTS AND METHODS: Five children with either drug-induced or congenital partial deafness were enrolled in a pilot study. The patients ranged in age from 13 months to 14 years. Implantation was performed using a hearing preservation technique. A Flex EAS electrode (MED-EL, Innsbruck, Austria) was used in all full insertions. RESULTS: Low frequency hearing was preserved in all patients with postoperative bone conduction within 10 dB of the preoperative hearing levels. These changes were preserved over the follow-up period of 12 months. There were significant improvements in speech perception. CONCLUSION: Hearing preservation cochlear implantation is a new effective modality in children with partial deafness.


Subject(s)
Cochlear Implantation/methods , Cochlear Implants , Deafness/chemically induced , Deafness/surgery , Acoustic Stimulation/methods , Adenocarcinoma, Clear Cell/drug therapy , Adolescent , Antineoplastic Agents/adverse effects , Cerebellar Neoplasms/drug therapy , Child , Child, Preschool , Deafness/congenital , Female , Follow-Up Studies , Goiter, Nodular/chemically induced , Goiter, Nodular/congenital , Goiter, Nodular/surgery , Hearing/physiology , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/congenital , Hearing Loss, Sensorineural/surgery , Humans , Infant , Kidney Neoplasms/drug therapy , Male , Platinum/toxicity , Prospective Studies
8.
Sci Total Environ ; 228(2-3): 219-42, 1999 Apr 05.
Article in English | MEDLINE | ID: mdl-10371055

ABSTRACT

Automobile exhaust catalytic converters emit fine dispersed elemental platinum, Pt (0), in the nanometer range coated on larger aluminium oxide carrier particles. A pre-requisite for a potential systemic toxic effect of the emitted platinum is its bioavailability which was investigated using laboratory animals. To this end, a model substance was synthesised which consisted of aluminium oxide particles < or = 5 microns onto which platinum particles > or = 4 nm were deposited by a calcination process. These particles closely resemble those emitted from automobile exhaust converters. This model substance was applied to female Lewis rats in two doses by intratracheal instillation; the animals were killed after 1, 7, 28 and 90 days. In addition, the model substance was also applied during a 90-day inhalation study. After microwave digestion of the tissues, the platinum was determined in all organs and body fluids by inductively coupled plasma/mass spectrometry (ICP/MS). Platinum was found in the blood, urine and faeces and all important organs (liver, spleen, kidneys, adrenals, stomach, femur). Based on the platinum content determined in the body fluids and all organs (except the lung and the faeces) it was calculated that up to 16% of the platinum was retained in the lung 1 day after intratracheal instillation and up to 30% of the fine dispersed platinum deposited on an average during 90 days inhalation in the lung was bioavailable. Using size exclusion chromatography (SEC) in combination with ICP/MS, it was shown that > or = 90% of the bioavailable platinum was bound to high molecular weight compounds (approximately 80-800 kDa), most likely proteins.


Subject(s)
Platinum/pharmacokinetics , Platinum/toxicity , Vehicle Emissions/toxicity , Administration, Inhalation , Aluminum Oxide , Animals , Biological Availability , Catalysis , Female , Microscopy, Electron , Models, Biological , Particle Size , Platinum/administration & dosage , Rats , Rats, Inbred Lew , Solubility , Trachea , Vehicle Emissions/analysis
9.
Int J Radiat Biol ; 66(2): 215-20, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8089631

ABSTRACT

HeLa S-3 cells were treated with 195mPt-radiolabelled cis-diaminedichloroplatinum II) (CDDP) for 60 min at various temperatures to examine the relationship between the lethal effect and the number of Pt atoms binding to DNA, RNA and protein molecules. The mean lethal concentration (Do) of CDDP for 60-min treatment at 0, 25, 37, 40, 42 and 44 degrees C was 233, 69.9, 15.9, 11.7, 8.3 and 4.7 microM respectively. By using identically treated cells, the number of Pt atoms combined with DNA, RNA and protein molecules was determined in the subcellular fractions prepared by the method of Schneider (1961). Thus, the Do's given as the drug concentrations were substituted for the number of Pt atoms combined with each fraction. Then the efficiency of the Pt atom to kill the cells was expressed as the reciprocal of the number of Pt atoms combined and was calculated for each molecule. The efficiency for DNA was 2.47, 2.75, 9.49, 9.66, 10.53 and 15.00 x 10(4) nucleotides respectively for the conditions described above. A detailed comparison of the Do's and efficiencies suggested that the supra-additive effect of the combination treatment could be explained by two mechanisms, i.e. the increased drug level in DNA (from 37 to 42 degrees C) and the increased efficiency of the Pt atoms to kill the cells (> 42 degrees C).


Subject(s)
Cisplatin/metabolism , Cisplatin/toxicity , DNA, Neoplasm/metabolism , Hyperthermia, Induced , Platinum/metabolism , Platinum/toxicity , Cell Death/drug effects , Cell Survival/drug effects , Combined Modality Therapy , DNA, Neoplasm/analysis , Dose-Response Relationship, Drug , HeLa Cells , Humans , Kinetics , Neoplasm Proteins/analysis , RNA, Neoplasm/analysis , Radioisotopes
10.
Article in English | MEDLINE | ID: mdl-1483037

ABSTRACT

In experiments with rats dose-response relationships of alimentary PtCl2 and PtCl4 were investigated. 2 x 81 animals weighing 35 g were randomly distributed among 9 treatment groups which were fed ad libitum with a synthetic diet containing various amounts of Pt during 4 weeks. Pt was added in the form of PtCl2 or PtCl4 in the amounts 0; 0.01; 0.05; 0.10; 0.50; 1.0; 5.0; 10 and 50 mg/kg diet. The Pt supplementation had no influence on life mass gain or food consumption. In the case of 50 mg/kg Pt in the form of PtCl4 the erythrocyte count and hematocrit were reduced by about 13% in comparison with the control group. Dependent on the Pt dose, the application of PtCl4 and PtCl2 induced Pt retention in nearly all tissues especially in kidney. The effects were greater with PtCl4 than with PtCl2. As a result of the higher Pt retention in the kidneys, the serum creatinine was increased for the higher doses of PtCl4.


Subject(s)
Erythrocyte Count/drug effects , Platinum Compounds , Platinum/toxicity , Animals , Body Weight/drug effects , Diet , Dose-Response Relationship, Drug , Eating/drug effects , Hematocrit , Hemoglobins/analysis , Male , Platinum/administration & dosage , Platinum/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL