Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Radiat Biol ; : 1-11, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953870

ABSTRACT

PURPOSE: Radiation-induced alterations in gene expression show great promise for dose reconstruction and for severity prediction of acute health effects. Among several genes explored as potential biomarkers, FDXR is widely used due to high upregulation in white blood cells following radiation exposure. Nonetheless, the absence of a standardized protocols for gene expression-based biodosimetry is a notable gap that warrants attention to enhance the accuracy, reproducibility and reliability. The objective of this study was to evaluate the sensitivity of transcriptional biodosimetry to differences in protocols used by different laboratories and establish guidelines for the calculation of calibration curve using FDXR expression data. MATERIAL AND METHODS: Two sets of irradiated blood samples generated during RENEB exercise were used. The first included samples irradiated with known doses including: 0, 0.25, 0.5, 1, 2, 3 and 4 Gy. The second set consisted of three 'blind' samples irradiated with 1.8 Gy, 0.4 Gy and a sham-irradiated sample. After irradiation, samples were incubated at 37 °C over 24 h and sent to participating laboratories, where RNA isolation and FDXR expression analysis by qPCR were performed using sets of primers/probes and reference genes specific for each laboratory. Calibration curves based on FDXR expression data were generated using non-linear and linear regression and used for dose estimation of 'blind' samples. RESULTS: Dose estimates for sham-irradiated sample (0.020-0.024 Gy) and sample irradiated with 0.4 Gy (0.369-0.381 Gy) showed remarkable consistency across all laboratories, closely approximating the true doses regardless variation in primers/probes and reference genes used. For sample irradiated with 1.8 Gy the dose estimates were less precise (1.198-2.011 Gy) but remained within an acceptable margin for triage within the context of high dose range. CONCLUSION: Methodological differences in reference genes and primers/probes used for FDXR expression measurement do not have a significant impact on the dose estimates generated, provided that all reference genes performed as expected and the primers/probes target a similar set of transcript variants. The preferred method for constructing a calibration curve based on FDXR expression data involves employing linear regression to establish a function that describes the relationship between the logarithm of absorbed dose and FDXR ΔCt values. However, one should be careful with using non-irradiated sample data as these cannot be accurately represented on a logarithmic scale. A standard curve generated using this approach can give reliable dose estimations in a dose range from 50 mGy to 4 Gy at least.

2.
Molecules ; 29(7)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38611909

ABSTRACT

Dinitrosyl iron complexes (DNICs) stabilize nitric oxide in cells and tissues and constitute an important form of its storage and transportation. DNICs may comprise low-molecular-weight ligands, e.g., thiols, imidazole groups in chemical compounds with low molecular weight (LMWDNICs), or high-molecular-weight ligands, e.g., peptides or proteins (HMWDNICs). The aim of this study was to investigate the role of low- and high-molecular-weight ligands in DNIC formation. Lysosomal and proteasomal proteolysis was inhibited by specific inhibitors. Experiments were conducted on human erythroid K562 cells and on K562 cells overexpressing a heavy chain of ferritin. Cell cultures were treated with •NO donor. DNIC formation was monitored by electron paramagnetic resonance. Pretreatment of cells with proteolysis inhibitors diminished the intensity and changed the shape of the DNIC-specific EPR signal in a treatment time-dependent manner. The level of DNIC formation was significantly influenced by the presence of protein degradation products. Interestingly, formation of HMWDNICs depended on the availability of LMWDNICs. The extent of glutathione involvement in the in vivo formation of DNICs is minor yet noticeable, aligning with our prior research findings.


Subject(s)
Nitric Oxide , Nitrogen Oxides , Humans , Proteolysis , Nitrogen Oxides/pharmacology , Iron
3.
Materials (Basel) ; 16(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38068008

ABSTRACT

The interest in nanoparticles (NPs) and their effects on living organisms has been continuously growing in the last decades. A special interest is focused on the effects of NPs on the central nervous system (CNS), which seems to be the most vulnerable to their adverse effects. Non-metallic NPs seem to be less toxic than metallic ones; thus, the application of non-metallic NPs in medicine and industry is growing very fast. Hence, a closer look at the impact of non-metallic NPs on neural tissue is necessary, especially in the context of the increasing prevalence of neurodegenerative diseases. In this review, we summarize the current knowledge of the in vitro and in vivo neurotoxicity of non-metallic NPs, as well as the mechanisms associated with negative or positive effects of non-metallic NPs on the CNS.

4.
Materials (Basel) ; 16(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37297299

ABSTRACT

The importance of epigenetic changes as a measurable endpoint in nanotoxicological studies is getting more and more appreciated. In the present work, we analyzed the epigenetic effects induced by citrate- and PEG-coated 20 nm silver nanoparticles (AgNPs) in a model consisting of 4T1 breast cancer tumors in mice. Animals were administered with AgNPs intragastrically (1 mg/kg b.w. daily-total dose 14 mg/kg b.w.) or intravenously (administration twice with 1 mg/kg b.w.-total dose 2 mg/kg b.w.). We observed a significant decrease in 5-methylcytosine (5-mC) level in tumors from mice treated with citrate-coated AgNPs regardless of the route of administration. For PEG-coated AgNPs, a significant decrease in DNA methylation was observed only after intravenous administration. Moreover, treatment of 4T1 tumor-bearing mice with AgNPs decreased histone H3 methylation in tumor tissue. This effect was the most pronounced for PEG-coated AgNPs administered intravenously. No changes in histone H3 Lys9 acetylation were observed. The decrease in methylation of DNA and histone H3 was accompanied by changes in expression of genes encoding chromatin-modifying enzymes (Setd4, Setdb1, Smyd3, Suv39h1, Suv420h1, Whsc1, Kdm1a, Kdm5b, Esco2, Hat1, Myst3, Hdac5, Dnmt1, Ube2b, and Usp22) and genes related to carcinogenesis (Akt1, Brca1, Brca2, Mlh1, Myb, Ccnd1, and Src). The significance of the observed changes and the mechanisms responsible for their development are unclear, and more research in this area is warranted. Nevertheless, the present work points to the epigenetic effects as an important level of interaction between nanomaterials and biological systems, which should always be taken into consideration during analysis of the biological activity of nanomaterials and development of nanopharmaceuticals.

5.
Materials (Basel) ; 15(11)2022 May 27.
Article in English | MEDLINE | ID: mdl-35683135

ABSTRACT

The potential anticancer activity of different silver nanoformulations is increasingly recognized. In the present work, we use the model of 4T1 tumor in BALB/ccmdb immunocompetent mice to analyze the impact of citrate- and PEG-coated silver nanoparticles (AgNPs) on the development and metastatic potential of breast cancer. One group of mice was intragastrically administered with 1 mg/kg body weight (b.w.) of AgNPs daily from day 1 to day 14 after cancer cells implantation (total dose 14 mg/kg b.w.). The second group was intravenously administered twice with 1 or 5 mg/kg b.w. of AgNPs. A tendency for lowering tumor volume on day 21 (mean volumes 491.31, 428.88, and 386.83 mm3 for control, AgNPs-PEG, and AgNPs-citrate, respectively) and day 26 (mean volumes 903.20, 764.27, and 672.62 mm3 for control, AgNPs-PEG, and AgNPs-citrate, respectively) has been observed in mice treated intragastrically, but the effect did not reach the level of statistical significance. Interestingly, in mice treated intragastrically with citrate-coated AgNPs, the number of lung metastases was significantly lower, as compared to control mice (the mean number of metastases 18.89, 14.90, and 8.03 for control, AgNPs-PEG, and AgNPs-citrate, respectively). No effect of AgNPs treatment on the number of lung metastases was observed after intravenous administration (the mean number of metastases 12.44, 9.86, 12.88, 11.05, and 10.5 for control, AgNPs-PEG 1 mg/kg, AgNPs-PEG 5 mg/kg, AgNPs-citrate 1 mg/kg, and AgNPs-citrate 5 mg/kg, respectively). Surprisingly, inhibition of metastasis was not accompanied by changes in the expression of genes associated with epithelial-mesenchymal transition. Instead, changes in the expression of inflammation-related genes were observed. The presented results support the antitumor activity of AgNPs in vivo, but the effect was limited to the inhibition of metastasis. Moreover, our results clearly point to the importance of AgNPs coating and route of administration for its anticancer activity. Finally, our study supports the previous findings that antitumor AgNPs activity may depend on the activation of the immune system and not on the direct action of AgNPs on cancer cells.

6.
Int J Mol Sci ; 22(11)2021 May 28.
Article in English | MEDLINE | ID: mdl-34071406

ABSTRACT

Coralyne is a synthetic analog of berberine related to protoberberine-isoquinoline alkaloids. Isoquinoline derivatives and analogs are renowned as potent radiosensitizers with potential medical application. In the present study, we investigated the effect of coralyne on the cell death, cytoskeletal changes and cell cycle progression of irradiated A549 cells. A clonogenic assay revealed that coralyne pretreatment decreased the viability of A549 cells in a time- and dose-dependent manner. Moreover, exposure to coralyne and ionizing radiation (IR) markedly altered the filamentous actin cytoskeletal architecture and integrin-ß binding sites of A549 cells. Treatment with 1-25 µM coralyne in combination with 2 Gy of IR significantly reduced the percentage of cells in G2/M phase compared with 2 Gy IR alone. These results indicate that coralyne is a potent radiosensitizing agent that may find an application in medicine.


Subject(s)
Berberine Alkaloids/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Up-Regulation/drug effects , A549 Cells , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , G2 Phase Cell Cycle Checkpoints/radiation effects , Humans , Microscopy, Confocal , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology
7.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34071854

ABSTRACT

Metastatic castration-resistant prostate cancer (mCRPC) is a progressive and incurable disease with poor prognosis for patients. Despite introduction of novel therapies, the mortality rate remains high. An attractive alternative for extension of the life of mCRPC patients is PSMA-based targeted radioimmunotherapy. In this paper, we extended our in vitro study of 223Ra-labeled and PSMA-targeted NaA nanozeolites [223RaA-silane-PEG-D2B] by undertaking comprehensive preclinical in vitro and in vivo research. The toxicity of the new compound was evaluated in LNCaP C4-2, DU-145, RWPE-1 and HPrEC prostate cells and in BALB/c mice. The tissue distribution of 133Ba- and 223Ra-labeled conjugates was studied at different time points after injection in BALB/c and LNCaP C4-2 tumor-bearing BALB/c Nude mice. No obvious symptoms of antibody-free and antibody-functionalized nanocarriers cytotoxicity and immunotoxicity was found, while exposure to 223Ra-labeled conjugates resulted in bone marrow fibrosis, decreased the number of WBC and platelets and elevated serum concentrations of ALT and AST enzymes. Biodistribution studies revealed high accumulation of 223Ra-labeled conjugates in the liver, lungs, spleen and bone tissue. Nontargeted and PSMA-targeted radioconjugates exhibited a similar, marginal uptake in tumour lesions. In conclusion, despite the fact that NaA nanozeolites are safe carriers, the intravenous administration of NaA nanozeolite-based radioconjugates is dubious due to its high accumulation in the lungs, liver, spleen and bones.


Subject(s)
Immunoconjugates/pharmacokinetics , Nanoparticles , Prostatic Neoplasms/therapy , Radiopharmaceuticals/pharmacokinetics , Radium , Theranostic Nanomedicine , Zeolites , Animals , Antibodies, Monoclonal , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Chemical Phenomena , Chemistry Techniques, Synthetic , Disease Models, Animal , Drug Design , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Isotope Labeling , Male , Mice , Mice, Nude , Molecular Structure , Nanoparticles/chemistry , Prostatic Neoplasms/diagnosis , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemistry , Radium/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays , Zeolites/chemistry
8.
Int J Mol Sci ; 22(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33653008

ABSTRACT

Gold nanoparticles (AuNPs) are foreseen as a promising tool in nanomedicine, both as drug carriers and radiosensitizers. They have been also proposed as a potential anticancer drug due to the anti-angiogenic effect in tumor tissue. In this work we investigated the effect of citrate-coated AuNPs of nominal diameter 20 nm on the growth and metastatic potential of 4T1 cells originated from a mouse mammary gland tumor inoculated into the mammary fat pad of Balb/ccmdb mice. To evaluate whether AuNPs can prevent the tumor growth, one group of inoculated mice was intragastrically (i.g.) administered with 1 mg/kg of AuNPs daily from day 1 to day 14 after cancer cell implantation. To evaluate whether AuNPs can attenuate the tumor growth, the second group was intravenously (i.v.) administered with 1 or 5 mg/kg of AuNPs, twice on day 5 and day 14 after inoculation. We did not observe any anticancer activity of i.v. nor i.g. administered AuNPs, as they did not affect neither the primary tumor growth rate nor the number of lung metastases. Unexpectedly, both AuNP treatment regimens caused a marked vasodilating effect in the tumor tissue. As no change of potential angiogenic genes (Fgf2, Vegfa) nor inducible nitric oxygenase (Nos2) was observed, we proposed that the vasodilation was caused by AuNP-dependent decomposition of nitrosothiols and direct release of nitric oxide in the tumor tissue.


Subject(s)
Citric Acid/therapeutic use , Gold/therapeutic use , Mammary Neoplasms, Animal/blood supply , Metal Nanoparticles/therapeutic use , Animals , Cell Line, Tumor , Citric Acid/administration & dosage , Female , Gold/administration & dosage , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred BALB C , Nanomedicine , Particle Size , Vasodilation
9.
Dis Markers ; 2020: 8885189, 2020.
Article in English | MEDLINE | ID: mdl-33224316

ABSTRACT

BACKGROUND: Heart failure patients presenting with iron deficiency can benefit from systemic iron supplementation; however, there is the potential for iron overload to occur, which can seriously damage the heart. Therefore, myocardial iron (M-Iron) content should be precisely balanced, especially in already failing hearts. Unfortunately, the assessment of M-Iron via repeated heart biopsies or magnetic resonance imaging is unrealistic, and alternative serum markers must be found. This study is aimed at assessing M-Iron in patients with advanced heart failure (HF) and its association with a range of serum markers of iron metabolism. METHODS: Left ventricle (LV) myocardial biopsies and serum samples were collected from 33 consecutive HF patients (25 males) with LV dysfunction (LV ejection fraction 22 (11) %; NT-proBNP 5464 (3308) pg/ml) during heart transplantation. Myocardial ferritin (M-FR) and soluble transferrin receptor (M-sTfR1) were assessed by ELISA, and M-Iron was determined by Instrumental Neutron Activation Analysis in LV biopsies. Nonfailing hearts (n = 11) were used as control/reference tissue. Concentrations of serum iron-related proteins (FR and sTfR1) were assessed. RESULTS: LV M-Iron load was reduced in all HF patients and negatively associated with M-FR (r = -0.37, p = 0.05). Of the serum markers, sTfR1/logFR correlated with (r = -0.42; p = 0.04) and predicted (in a step-wise analysis, R 2 = 0.18; p = 0.04) LV M-Iron. LV M-Iron load (µg/g) can be calculated using the following formula: 210.24-22.869 × sTfR1/logFR. CONCLUSIONS: The sTfR1/logFR ratio can be used to predict LV M-Iron levels. Therefore, serum FR and sTfR1 levels could be used to indirectly assess LV M-Iron, thereby increasing the safety of iron repletion therapy in HF patients.


Subject(s)
Antigens, CD/blood , Biomarkers/blood , Ferritins/blood , Heart Failure/metabolism , Iron/metabolism , Receptors, Transferrin/blood , Female , Heart Failure/physiopathology , Heart Ventricles/metabolism , Humans , Male , Middle Aged , Ventricular Function, Left
10.
Materials (Basel) ; 13(14)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698417

ABSTRACT

Microglial cells clear the brain of pathogens and harmful debris, including amyloid-ß (Aß) deposits that are formed during Alzheimer's disease (AD). We studied the expression of Msr1, Ager and Cd36 receptors involved in Aß uptake and expression of Cd33 protein, which is considered a risk factor in AD. The effect of silver nanoparticles (AgNPs) and cadmium telluride quantum dots (CdTeQDs) on the expression of the above receptors and Aß uptake by microglial cells was investigated. Absorption of Aß and NPs was confirmed by confocal microscopy. AgNPs, but not CdTeQDs, caused a decrease in Aß accumulation. By using a specific inhibitor-polyinosinic acid-we demonstrated that Aß and AgNPs compete for scavenger receptors. Real-time PCR showed up-regulation of Cd33 and Cd36 gene expression after treatment with CdTeQDs for 24 h. Analysis of the abundance of the receptors on the cell surface revealed that AgNP treatment significantly reduced the presence of Msr1, Cd33, Ager and Cd36 receptors (6 and 24 h), whereas CdTeQDs increased the levels of Msr1 and Cd36 (24 h). To summarize, we showed that AgNP uptake competes with Aß uptake by microglial cells and consequently can impair the removal of the aggregates. In turn, CdTeQD treatment led to the accumulation of proinflammatory Cd36 protein on the cell surface.

11.
Ann Agric Environ Med ; 27(2): 231-234, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32588598

ABSTRACT

INTRODUCTION AND OBJECTIVE: Nuclear factor kappa B (NF-κB) signalling pathway plays a central role in the regulation of cellular response to stress. The aim of the study was to investigate the ability of silver nanoparticles (AgNPs), gold nanoparticles (AuNPs), CdTe quantum dots (CdTeQDs) or their binary mixtures to stimulate NF-κB binding in HepG2 cells. A dual luciferase reporter system was used to investigate NF-κB binding. MATERIAL AND METHODS: Cells were transiently transfected with a firefly luciferase reporter system and Renilla luciferase expression plasmid as a transfection efficiency control. Twenty- four hours after transfection, the cells were treated with nanoparticles (10 µg/cm3 AgNPs, 10 µg/cm3 AuNPs, 3 µg/cm3 CdTeQDs) or with 10 ng/cm3 TNFα as a positive control. Six hours later, the cells were lysed and the activities of the luminescence of firefly and Renilla luciferases were measured using the Dual-Luciferase Reporter Assay System. RESULTS: AuNPs and CdTeQDs alone significantly inhibited NF-κB binding activity. Co-treatment with AgNPs and CdTeQDs resulted in an additive effect, whereas the presence of AgNPs diminished the inhibitory effect of AuNPs. Interestingly, significant antagonism was observed between AuNPs and CdTeQDs, suggesting a similar mode of action. CONCLUSIONS: Comparison of the NF-κB binding activity induced by the mixtures of NPs suggests that in some cases NF-κB binding activity might differ from that observed for the NPs alone.


Subject(s)
Cadmium Compounds/metabolism , Gold/metabolism , Metal Nanoparticles , NF-kappa B/metabolism , Quantum Dots/metabolism , Silver/metabolism , Tellurium/metabolism , Hep G2 Cells , Humans
12.
Materials (Basel) ; 12(7)2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30934809

ABSTRACT

The increasing use of nanoparticles (NPs) in various applications entails the need for reliable assessment of their potential toxicity for humans. Originally, studies concerning the toxicity of NPs focused on cytotoxic and genotoxic effects, but more recently, attention has been paid to epigenetic changes induced by nanoparticles. In the present research, we analysed the DNA methylation status of genes related to inflammation and apoptosis as well as the expression of miRNAs related to these processes in response to silver (AgNPs), gold (AuNPs), and superparamagnetic iron oxide nanoparticles (SPIONs) at low cytotoxic doses in HepG2 cells. There were no significant differences between treated and control cells in the DNA methylation status. We identified nine miRNAs, the expression of which was significantly altered by treatment with nanoparticles. The highest number of changes was induced by AgNPs (six miRNAs), followed by AuNPs (four miRNAs) and SPIONs (two miRNAs). Among others, AgNPs suppressed miR-34a expression, which is of particular interest since it may be responsible for the previously observed AgNPs-mediated HepG2 cells sensitisation to tumour necrosis factor (TNF). Most of the miRNAs affected by NP treatment in the present study have been previously shown to inhibit cell proliferation and tumourigenesis. However, based on the observed changes in miRNA expression we cannot draw definite conclusions regarding the pro- or anti-tumour nature of the NPs under study. Further research is needed to fully elucidate the relation between observed changes in miRNA expression and the effect of NPs observed at the cellular level. The results of the present study support the idea of including epigenetic testing during the toxicological assessment of the biological interaction of nanomaterials.

13.
Pestic Biochem Physiol ; 154: 17-22, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30765052

ABSTRACT

Skin, the organ responsible for vitamin D synthesis, is fully exposed to many xenobiotics, e.g. polycyclic aromatic hydrocarbons and pesticides. A broad spectrum organophosphorus insecticides (OP's), such as chlorpyrifos (CPS), are commonly used in agriculture and to control domestic insects. Thus, the aim of this study was to investigate the effect of chlorpyrifos, on the expression of vitamin D3 receptor (VDR) in human keratinocytes cell line HaCaT and fibroblasts cell line BJ. The impact of CPS and UVB radiation on cell viability were examined by Neutral Red assay. The effect of CPS on VDR expression was evaluated by RT-qPCR and flow cytometry (FC). The presented study demonstrated that exposure to CPS and UVB significantly affects the viability of HaCaT and BJ cells lines. Results also revealed that exposure to CPS induced the expression at mRNA and protein level of VDR nuclear receptor in both cell lines exposed to UVB. In HaCaT incubated with 250 µM CPS and 15 mJ/cm2 UVB, the relative VDR expression was ∼2-fold higher; whereas in BJ incubated with 250 µM CPS and 20 mJ/cm2, UVB was∼3-fold higher. Results from FC confirmed this result, as VDR expression increased by ~250% in HaCaT incubated with 250 µM CPS and 20 mJ/cm2 UVB, and in BJ incubated with 250 µM CPS, and 20 mJ/cm2 UVB cells VDR expression increased by ~190%, compared with control. It can therefore be concluded that OPs pesticide might interfere with vitamin D3 metabolism in skin cells.


Subject(s)
Chlorpyrifos/toxicity , Fibroblasts/drug effects , Fibroblasts/radiation effects , Insecticides/toxicity , Keratinocytes/drug effects , Keratinocytes/radiation effects , Receptors, Calcitriol/physiology , Ultraviolet Rays , Cell Line , Fibroblasts/physiology , Humans , Keratinocytes/physiology
14.
Pol Arch Intern Med ; 129(1): 6-11, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30600310

ABSTRACT

INTRODUCTION Although the coexistence of type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) may be attributed to environmental risk factors common for both diseases, a genetic background should also be considered. Data on the role of genetic factors in the development of T2DM in patients with OSA are lacking. OBJECTIVES The study was aimed to evaluate the prevalence of polymorphisms of selected genes that are known to be associated with diabetes or obesity in patients with OSA and concomitant T2DM and to assess these polymorphisms in the context of OSA severity. PATIENTS AND METHODS Consecutive patients with newly diagnosed OSA confirmed by polysomnography underwent genotyping for the following single nucleotide polymorphisms (SNPs): SREBF1 rs11868035, HIF1A rs11549465, APOA5 rs3135506, TCF7L2 rs7903146, and FTO rs16945088. The frequency of genotypes was compared between patients with and without concomitant T2DM and was analyzed with regard to OSA severity. RESULTS A total of 600 patients with newly diagnosed OSA were enrolled to the study. Of these, 121 patients (20.2%) were diagnosed with T2DM (97 men and 24 women; median age, 58 years; range, 52-64 years). The prevalence of T2DM was significantly lower in APOA5 rs3135506 GG homozygotes than in CG heterozygotes (18.8% vs 33.3%, P = 0.02). APOA5 rs3135506 CG heterozygotes were at higher risk for developing T2DM (adjusted odds ratio, 2.64; 95% confidence interval,1.38-5.04; P = 0.003). No significant differences were found for the genotype distribution of the other investigated SNPs. CONCLUSIONS Our study shows a possible link between the polymorphism of the gene encoding APOA5 and T2DM in patients with OSA.


Subject(s)
Apolipoprotein A-V , Diabetes Mellitus, Type 2/diagnosis , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Sleep Apnea, Obstructive/complications , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Female , Humans , Male , Middle Aged
15.
Oncol Lett ; 16(5): 6831-6837, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30405827

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is often accompanied by lung cancer. In our previous work, it was observed that matrix metalloproteinase-3 and haptoglobin (HP) polymorphisms were potential markers of enhanced susceptibility to lung cancer development among male COPD subjects. Here, results are reported on blood serum levels of several proteins involved in iron metabolism, inflammation and the oxidative stress response compared between the same groups of subjects. The blood serum levels of tumor necrosis factor α (TNFα), transferrin, hepcidin, ferritin, soluble transferrin receptor and 8-oxo-2'-deoxyguanosine were compared, as well as total iron-binding capacity (TIBC) and ceruloplasmin ferroxidase activity in two groups of subjects: Male COPD patients (54 subjects) and male COPD patients diagnosed with lung cancer (53 subjects). Statistically significant differences were identified between the two groups in transferrin and TNFα levels, as well as in TIBC; all three parameters were lower in the group consisting of COPD patients diagnosed with lung cancer (P<0.01). It was also revealed that HP genotype 1/2 was concomitant with low transferrin blood level in subjects with COPD; this apparent dependence was absent in the COPD + cancer subjects. The results indicate a role of iron metabolism in the susceptibility to lung cancer in COPD-affected subjects. They also emphasize the importance of individual capacity for an effective response to oxidative stress during the pathogenic process as HP is a plasma protein that binds free hemoglobin and its polymorphism results in proteins with altered hemoglobin-binding capacity and different antioxidant and iron-recycling functions.

16.
Toxicol Appl Pharmacol ; 356: 140-150, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30096344

ABSTRACT

Metallic nanomaterials are utilized in an increasing number of applications in medicine and industry. Their general toxicity was tested in numerous reports both in vitro and in vivo but limited data exist on how nanomaterials affect the activity of cellular signaling pathways activated by growth factors and cytokines. The aim of the present work was to test the hypothesis predicting that silver, gold and superparamagnetic iron oxide nanoparticles may interfere with cellular signaling activated by tumor necrosis factor (TNF) and change the final cellular outcome of TNF action. Such interference may result in disruption of homeostasis and contribute to the development of malignancies such as cancer or autoimmune diseases. Experiments were performed on HepG2 and A549 cell lines. We did not observe any interaction between nanoparticles and TNF at the level of clonogenic growth, apoptosis/necrosis induction or cell cycle. At all these endpoints, the effects of TNF and nanoparticles were additive. In contrast, gene expression analysis revealed synergistic effects. A group of genes was significantly affected only by simultaneous treatment with TNF and nanoparticles and not by any of the factors alone. Observed synergistic effect on IL10 and IL8 expression seems to be of particular importance since these cytokines are often expressed by tumor cells to inhibit tumor-targeted immune response. The observed synergistic effects of TNF and nanoparticles on cytokines expression may have significant consequences for tissue homeostasis and tumor promotion and therefore should be taken into account during development of new nanoparticle-based anticancer therapies.


Subject(s)
Ferric Compounds/pharmacology , Gold/pharmacology , Metal Nanoparticles , Silver/pharmacology , Tumor Necrosis Factor Inhibitors , A549 Cells , Cell Survival/drug effects , Colony-Forming Units Assay , Ferric Compounds/metabolism , Gold/metabolism , Hep G2 Cells , Humans , Interleukin-10/biosynthesis , Interleukin-8/biosynthesis , Magnetics , Signal Transduction/drug effects , Silver/metabolism
18.
Environ Sci Pollut Res Int ; 24(31): 24223-24234, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28889235

ABSTRACT

Epidemiological data indicate that exposure to diesel exhaust particles (DEPs) from traffic emissions is associated with higher risk of morbidity and mortality related to cardiovascular and pulmonary diseases, accelerated progression of atherosclerotic plaques, and possible lung cancer. While the impact of DEPs from combustion of fossil diesel fuel on human health has been extensively studied, current knowledge of DEPs from combustion of biofuels provides limited and inconsistent information about its mutagenicity and genotoxicity, as well as possible adverse health risks. The objective of the present work was to compare the genotoxicity of DEPs from combustion of two first-generation fuels, 7% fatty acid methyl esters (FAME) (B7) and 20% FAME (B20), and a second-generation 20% FAME/hydrotreated vegetable oil (SHB: synthetic hydrocarbon biofuel) fuel. Our results revealed that particulate engine emissions from each type of biodiesel fuel induced genotoxic effects in BEAS-2B and A549 cells, manifested as the increased levels of single-strand breaks, the increased frequencies of micronuclei, or the deregulated expression of genes involved in DNA damage signaling pathways. We also found that none of the tested DEPs showed the induction of oxidative DNA damage and the gamma-H2AX-detectable double-strand breaks. The most pronounced differences concerning the tested particles were observed for the induction of single-strand breaks, with the greatest genotoxicity being associated with the B7-derived DEPs. The differences in other effects between DEPs from the different biodiesel blend percentage and biodiesel feedstock were also observed, but the magnitude of these variations was limited.


Subject(s)
Biofuels/analysis , DNA Damage , Gasoline/toxicity , Vehicle Emissions/toxicity , A549 Cells , Humans
19.
Environ Sci Pollut Res Int ; 24(23): 19357-19374, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28674953

ABSTRACT

Biodiesels represent more carbon-neutral fuels and are introduced at an increasing extent to reduce emission of greenhouse gases. However, the potential impact of different types and blend concentrations of biodiesel on the toxicity of diesel engine emissions are still relatively scarce and to some extent contradictory. The objective of the present work was to compare the toxicity of diesel exhaust particles (DEP) from combustion of two 1st-generation fuels: 7% fatty acid methyl esters (FAME; B7) and 20% FAME (B20) and a 2nd-generation 20% FAME/HVO (synthetic hydrocarbon biofuel (SHB)) fuel. Our findings indicate that particulate emissions of each type of biodiesel fuel induce cytotoxic effects in BEAS-2B and A549 cells, manifested as cell death (apoptosis or necrosis), decreased protein concentrations, intracellular ROS production, as well as increased expression of antioxidant genes and genes coding for DNA damage-response proteins. The different biodiesel blend percentages and biodiesel feedstocks led to marked differences in chemical composition of the emitted DEP. The different DEPs also displayed statistically significant differences in cytotoxicity in A549 and BEAS-2B cells, but the magnitude of these variations was limited. Overall, it seems that increasing biodiesel blend concentrations from the current 7 to 20% FAME, or substituting 1st-generation FAME biodiesel with 2nd-generation HVO biodiesel (at least below 20% blends), affects the in vitro toxicity of the emitted DEP to some extent, but the biological significance of this may be moderate.


Subject(s)
Biofuels , Particulate Matter/chemistry , Particulate Matter/toxicity , Vehicle Emissions/toxicity , Air Pollutants/analysis , Air Pollutants/chemistry , Air Pollutants/toxicity , Biofuels/analysis , Cell Line , Cell Survival/drug effects , Humans , Hydrocarbons/analysis , Hydrocarbons/chemistry , Hydrocarbons/toxicity , Particulate Matter/analysis , Vehicle Emissions/analysis
20.
Int J Radiat Biol ; 93(1): 87-98, 2017 01.
Article in English | MEDLINE | ID: mdl-27626709

ABSTRACT

PURPOSE: This collaboration of five established European gene expression labs investigated the potential impact of culture conditions on the transcriptional response of peripheral blood to radiation exposure. MATERIALS AND METHODS: Blood from one healthy donor was exposed ex vivo to a Cobalt 60 source to produce a calibration curve in addition to four unknown doses. After exposure, the blood samples were either diluted with RPMI medium or left untouched. After 24-h incubation at 37 °C the diluted blood samples were lysed, while the undiluted samples were mixed with the preservative RNALater and all samples were shipped frozen to the participating labs. Samples were processed by each lab using microarray (one lab) and QRT-PCR (four labs). RESULTS: We show that although culture conditions affect the total amount of RNA recovered (p < .0001) and its integrity (p < .0001), it does not significantly affect dose estimates (except for the true dose at 1.1 Gy). Most importantly, the different analysis approaches provide comparable mean absolute difference of estimated doses relative to the true doses (p = .9) and number of out of range (>0.5 Gy) measurements (p = .6). CONCLUSION: This study confirms the robustness of gene expression as a method for biological dosimetry.


Subject(s)
Biological Assay/methods , Blood Proteins/analysis , Blood Specimen Collection/methods , Gene Expression Profiling/methods , Radiation Exposure/analysis , Radiation Monitoring/methods , Blood Chemical Analysis/methods , Europe , Radiation Dosage , Reproducibility of Results , Sensitivity and Specificity , Single-Blind Method
SELECTION OF CITATIONS
SEARCH DETAIL
...