Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 74: 105162, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33839235

ABSTRACT

Silymarin is a flavonoid complex isolated from the plant Silybum marianum which is well known for its antioxidant, hepatoprotective and immunomodulatory effects. Since little is known about its anti-inflammatory properties and healing effects, our study focused on whether or not silymarin components reduce inflammation and support epidermis regeneration. Lipopolysaccharides (LPS) and sodium dodecyl sulfate (SDS) were used to induce inflammation in normal human epidermal keratinocytes (NHEKs) and reconstructed epidermis (RHE), respectively. The expression of pro-inflammatory cytokines (IL-1, IL-6 and IL-8) in NHEKs and RHE was measured by enzyme - linked immunosorbent assay (ELISA). The expression of cytokeratin 14 and loricrin in RHE was detected by immunofluorescent analysis. Hematoxylin and eosin staining was used for the morphological evaluation of RHE. It was determined that 2, 3 - dehydrosilybin (DHSB) downregulated the production of selected pro-inflammatory cytokines produced by NHEKs. Although all layers of RHE displayed full thickness, when SDS was applied, cell detachment was seen in the stratum corneum and loricrin expression was diminished.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Epidermis/drug effects , Keratinocytes/drug effects , Quercetin/pharmacology , Silymarin/pharmacology , Cytokines/metabolism , Epidermis/metabolism , Humans , Inflammation/chemically induced , Keratin-14/metabolism , Keratinocytes/metabolism , Lipopolysaccharides/toxicity , Membrane Proteins/metabolism , Sodium Dodecyl Sulfate/toxicity
2.
Hum Exp Toxicol ; 35(9): 946-57, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26500221

ABSTRACT

Biomedical application of silver nanoparticles (AgNPs) has been rapidly increasing. Owing to their strong antimicrobial activity, AgNPs are used in dermatology in the treatment of wounds and burns. However, recent evidence for their cytotoxicity gives rise to safety concerns. This study was undertaken as a part of an ongoing programme in our laboratory to develop a topical agent for wound healing. Here, we investigated the potential toxicity of AgNPs using normal human dermal fibroblasts (NHDF) and normal human epidermal keratinocytes (NHEK) with the aim of comparing the effects of AgNPs and ionic silver (Ag-I). Besides the effect of AgNPs and Ag-I on cell viability, the inflammatory response and DNA damage in AgNPs and Ag-I-treated cells were examined. The results showed that Ag-I were significantly more toxic than AgNPs both on NHDF and NHEK. Non-cytotoxic concentrations of AgNPs and Ag-I did not induce DNA strand breaks and did not affect inflammatory markers, except for a transient increase in interleukin 6 levels in Ag-I-treated NHDF. The results showed that AgNPs are more suitable for the intended application as a topical agent for wound healing up to the concentration 25 µg/mL.


Subject(s)
Anti-Infective Agents/toxicity , Fibroblasts/drug effects , Keratinocytes/drug effects , Metal Nanoparticles/toxicity , Silver/toxicity , Skin/drug effects , Anti-Infective Agents/chemistry , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , DNA Damage , Dose-Response Relationship, Drug , Epidermis/drug effects , Epidermis/pathology , Fibroblasts/pathology , Humans , Keratinocytes/pathology , Metal Nanoparticles/chemistry , Microscopy, Electron, Transmission , Silver/chemistry , Skin/pathology , Surface Properties
3.
Pharmazie ; 71(8): 478-483, 2016 08 01.
Article in English | MEDLINE | ID: mdl-29442036

ABSTRACT

2,3-Dehydrosilybin exhibits substantial anticancer and antiangiogenic effects, which can be potentially improved by semi-synthetic modification such as esterification with gallic acid. The aim of this study was to examine the potential antiangiogenic effect of 2,3-dehydrosilybin and its galloyl esters (3-O-galloyl-2,3-dehydrosilybin; 7-O-galloyl-2,3-dehydrosilybin; 20-O-galloyl-2,3-dehydrosilybin and 23-O-galloyl-2,3-dehydrosilybin) and to determine which molecular mechanism could be responsible for their activity. The effect on cell proliferation, tube formation, signal transduction pathways (PI3K/Akt and ERK) and the cell cycle was studied in human microvascular endothelial cells (HMEC). The results showed that all compounds decreased the growth of HMEC, but the strongest effect was observed for 20-O-galloyl-2,3-dehydrosilybin at 5 µmol/l. In addition, at 5 and 10 µmol/l, this was the only compound that significantly inhibited HMEC tube formation. Based on an assessment of Akt and ERK1/2 expression, we suggest that 20-O-galloyl-2,3-dehydrosilybin influences the angiogenic process through the Akt pathway.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Silymarin/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Cell Cycle/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Esters/chemical synthesis , Esters/pharmacology , Gallic Acid/chemical synthesis , Gallic Acid/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Microtubules/drug effects , Neovascularization, Pathologic/drug therapy , Oncogene Protein v-akt/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Silybin , Silymarin/chemical synthesis , Structure-Activity Relationship
4.
Physiol Res ; 64(Suppl 4): S529-38, 2015.
Article in English | MEDLINE | ID: mdl-26681083

ABSTRACT

The aim of our study was to evaluate possible effect of ABCB1, and OPRM1 polymorphisms on the efficacy and safety of remifentanil in women undergoing elective cesarean section under general anesthesia. Women received remifentanil (1 microg/kg i.v.) 30 s prior to the induction to standardized general anesthesia. The ABCB1 (rs2032582, rs1045642) and OPRM1 (rs1799971) polymorphisms were analyzed from maternal peripheral blood. The basal hemodynamic and demographic parameters in the study population (n=54) were similar in all the subgroups. The median +/- SD increase of systolic blood pressure at 5 min from the baseline was practically completely abolished in homozygous carriers of ABCB1 variants in comparison with wild-type subjects -2.67+/-25.0 vs. 16.57+/-15.7 mm Hg, p<0.05 for rs2032582, and 2.00+/-23.9 vs. 22.13+/-16.8 mm Hg, p<0.05, for rs1045642, respectively. While no neonate belonging to ABCB1 wild-type homozygous or OPRM1 variant carrying mothers needed any resuscitative measure, 10.5 % of the neonates belonging to OPRM1 wild-type homozygous mothers received resuscitative support similarly as 11.1 %, and 12.5 % of neonates of mothers carrying variants of rs2032582, and rs1045642, respectively. Decreased stabilizing effects of remifentanil on maternal hemodynamics has been observed in ABCB1 wild type mothers, while the adaptation of the neonates was clinically worse in OPRM1 wild type, and ABCB1 variant allele carriers.


Subject(s)
Anesthetics, Intravenous/therapeutic use , Cesarean Section , Piperidines/therapeutic use , Polymorphism, Single Nucleotide/genetics , Receptors, Opioid, mu/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Anesthetics, Intravenous/adverse effects , Blood Pressure/drug effects , Blood Pressure/physiology , Cesarean Section/adverse effects , Female , Heart Rate/drug effects , Heart Rate/physiology , Humans , Infant, Newborn , Piperidines/adverse effects , Pregnancy , Remifentanil , Treatment Outcome
5.
Mater Sci Eng C Mater Biol Appl ; 33(3): 1242-6, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23827567

ABSTRACT

Hydroxyapatite layers with silver doping from 0.06 at.% to 14 at.% were prepared by laser deposition. The films' physical properties such as morphology, composition, crystallinity, Young's modulus and microhardness were measured. Films were amorphous or polycrystalline in dependence on deposition temperature (from RT to 600 °C). Antibacterial properties were tested using Escherichia coli and Bacillus subtilis cells. The antibacterial efficacy changed with silver doping from 4% to 100%. Cytotoxicity was studied by a direct contact test. Depending on doping and crystallinity the films were either non-toxic or mildly toxic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Durapatite/pharmacology , Lasers , Animals , Cell Death/drug effects , Crystallization , Elastic Modulus/drug effects , Escherichia coli/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Hardness/drug effects , Mice , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , NIH 3T3 Cells , Silicon/pharmacology , Staphylococcus aureus/drug effects , Titanium/pharmacology , X-Ray Diffraction
6.
Acta Chir Orthop Traumatol Cech ; 77(5): 416-24, 2010 Oct.
Article in Czech | MEDLINE | ID: mdl-21040654

ABSTRACT

PURPOSE OF THE STUDY: A failure of total hip or knee artroplasty is associated with an increased production of joint fluid. This contains wear particles and host cells and proteins, and is assumed to be involved in the pathogenesis of aseptic loosening and periprosthetic osteolysis. This study investigated the effect of synovial fluid from patients with aseptically failed joint prostheses on osteoblast cultures. MATERIAL AND METHODS: Synovial fluid samples were obtained from patients with failed total joint prostheses (TJP; n=36) and from control patient groups (n = 16) involving cases without TJP and osteoarthritis, without TJP but with osteoarthritis, and with stable TJP. The samples were treated in the standard manner and then cultured with the SaOS-2 cell line which shows the characteristics and behaviour of osteoblasts. Each fluid sample was also examined for the content of proteins, cells and selected cytokines (IL-1ß, TNF-α, IL-6, RANKL and OPG detected by ELISA). We tested the hypothesis assuming that the fluids from failed joints would show higher cytotoxicity to osteoblast culture and we also expected higher levels of IL-1ß, TNF-α, IL-6, and RANKL in patients with TJP failure and/ or with more severe bone loss. The statistical methods used included the Kruskal-Wallis ANOVA and Mann-Whitney U test. RESULTS: The fluids from failed TJPs showed the highest RANKL and the lowest OPG levels resulting in the highest RANKL/OPG ratio. However, there was no evidence suggesting that the joint fluids from failed TJPs would be more toxic to osteoblast culture than the fluids from control groups. In addition, no correlation was found between the fluid levels of molecules promoting inflammation and osteoclastic activity and the extent of bone loss in the hip (in terms of Saleh's classification) or the knee (AORI classification). In fact, the fluids from failed TJPs had higher protein levels in comparison with the controls, but the difference was not significant. DISCUSSION: The finding of high RANKL levels and low OPG concentrations is in agreement with the theory of aseptic loosening and periprosthetic osteolysis. The other cytokines, particularly TNF-α and IL-1ß, were found in low levels. This can be explained by the stage of particle disease at which the samples were taken for ELISA analysis. It is probable that the level of signal molecules reflects osteolytic process activity and is therefore not constant. The reason for no correlation found between cytokine levels and the extent of bone loss may also lie in the use of therapeutic classifications of bone defects that is apparently less sensitive to the biological activity of aseptic loosening and/or periprosthetic osteolysis. CONCLUSIONS: Synovial fluids from failed total hip or knee joint prostheses are not toxic to osteoblast cultures. Cytotoxicity indicators and levels of pro-inflammatory and pro-osteoclastic cytokines (IL-1ß, TNF-α, IL-6, RANKL and OPG) do not correlate well with the extent of periprosthetic bone loss. Key words: total joint replacement, arthroplasty, aseptic loosening, periprosthetic osteolysis, joint fluid, SaOS-2 cell line, cytotoxicity, cytokines, RANKL, OPG.


Subject(s)
Arthroplasty, Replacement, Hip , Arthroplasty, Replacement, Knee , Osteoblasts/cytology , Prosthesis Failure , Synovial Fluid/chemistry , Aged , Cells, Cultured , Female , Humans , Interleukin-6/analysis , Male , Middle Aged , Osteoarthritis, Hip/metabolism , Osteoarthritis, Hip/surgery , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/surgery , Osteoprotegerin/analysis , RANK Ligand/analysis , Synovial Fluid/physiology , Tumor Necrosis Factor-alpha/analysis
7.
Neoplasma ; 57(5): 438-48, 2010.
Article in English | MEDLINE | ID: mdl-20568898

ABSTRACT

Differentiation of myeloid leukemic cells may result in less sensitivity to various apoptotic stimuli. We examined whether human leukemia HL-60 cells differentiating by all-trans retinoic acid (ATRA) acquired resistance to the apoptogenic activity of two histone deacetylase (HDAC) inhibitors, butyrate and valproate. In undifferentiated cells, the cytotoxicity of both butyrate and valproate was associated with activation of the intrinsic apoptotic pathway since we observed dissipation of mitochondrial membrane potential, induction of caspase-9 and caspase-3 activities, appearance of sub-G1 DNA and loss of plasma membrane asymmetry and/or integrity. Both HDAC inhibitors were also able to induce accumulation of undifferentiated cells in the G0/G1 phase of the cell cycle. ATRA was found to enhance the apoptotic effect of both butyrate and valproate in undifferentiated cells. This aside, ATRA appeared to synergize with butyrate in the induction of the G0/G1 cell cycle arrest. In cells pretreated for 72 h with ATRA, butyrate and valproate in combination with ATRA induced lower dissipation of mitochondrial membrane potential and weaker apoptotic and/or necrotic changes in plasma membrane, whereas DNA fragmentation was not diminished compared to undifferentiated cells. Similar results were also obtained when butyrate or valproate were combined with another neutrophilic differentiation inducer, dimethyl sulfoxide. We conclude that neutrophilic differentiation modulates but does not abrogate the apoptotic response of HL-60 cells to butyrate and valproate, and nuclei are preferentially affected during apoptosis in differentiated cells.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Neutrophils/cytology , Valproic Acid/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Dimethyl Sulfoxide/pharmacology , HL-60 Cells , Humans , Membrane Potential, Mitochondrial , Tretinoin/pharmacology
8.
Food Chem Toxicol ; 48(6): 1555-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332009

ABSTRACT

The most common oral diseases have a microbial aetiology. Pathogenic bacteria liberate a number of irritating agents including a lipopolysaccharide (LPS) that activates pro-inflammatory cytokines promoting increased activity of polymorphonucleocytes (PMN). Release of PMN-derived free radicals into an infected gingival area affects gums, periodontal ligaments and alveolar bone. Berries of Lonicera caerulea L. (blue honeysuckle) are rich in phenolics, particularly phenolic acids, flavonoids and anthocyanins that have multiple biological activities in vitro and in vivo such as antiadherence, antioxidant and anti-inflammatory. Studies have shown that polyphenols suppress a number of LPS-induced signals and thus could be effective against gingivitis. Here we assessed effects of the polyphenolic fraction of L. caerulea fruits (PFLC; containing 77% anthocyanins) on LPS-induced oxidative damage and inflammation in human gingival fibroblasts. Application of PFLC (10-50mug/ml) reduced reactive oxygen species (ROS) production, intracellular glutathione (GSH) depletion as well as lipid peroxidation in LPS-treated cells. PFLC treatment also inhibited LPS-induced up-regulation of interleukin-1beta (IL-1beta), interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-alpha) and it suppressed expression of cyclooxygenase-2 (COX-2). The effects are presumably linked to its antioxidant and anti-inflammatory activities and suggest its use in attenuating the inflammatory process, including periodontal disease.


Subject(s)
Biomarkers/metabolism , Flavonoids/analysis , Gingiva/drug effects , Inflammation/metabolism , Lipopolysaccharides/toxicity , Lonicera/chemistry , Oxidative Stress/drug effects , Phenols/analysis , Cells, Cultured , Cytokines/biosynthesis , Fibroblasts/drug effects , Gingiva/cytology , Glutathione/metabolism , Humans , Inflammation/pathology , Polyphenols , Reactive Oxygen Species/metabolism
9.
Toxicol In Vitro ; 23(3): 386-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19159670

ABSTRACT

Periodontitis is a chronic disease associated with inflammation of the tooth-supporting tissues. The inflammation is initiated by a group of gram-negative anaerobic bacteria. These express a number of irritating factors including a lipopolysaccharide (LPS), which plays a key role in periodontal disease development. Plant extracts with anti-inflammatory and anti-microbial properties have been shown to inhibit bacterial plaque formation and thus prevent chronic gingivitis. In this study we tested effects of Prunella vulgaris L. extract (PVE; 5, 10, 25microg/ml) and its component rosmarinic acid (RA; 1microg/ml) on LPS-induced oxidative damage and inflammation in human gingival fibroblasts. PVE and RA reduced reactive oxygen species (ROS) production, intracellular glutathione (GSH) depletion as well as lipid peroxidation in LPS-treated cells. Treatment with PVE and RA also inhibited LPS-induced up-regulation of interleukin 1beta (IL-1beta), interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-alpha) and suppressed expression of inducible nitric oxide synthase (iNOS). The results indicate that PVE and RA are able to suppress LPS-induced biological changes in gingival fibroblasts. The effects of PVE and RA are presumably linked to their anti-inflammatory activities and thus use of PVE and RA may be relevant in modulating the inflammation process, including periodontal disease.


Subject(s)
Antioxidants/pharmacology , Cinnamates/pharmacology , Depsides/pharmacology , Fibroblasts/drug effects , Gingiva/drug effects , Prunella/chemistry , Antioxidants/analysis , Cell Survival/drug effects , Cells, Cultured , Cinnamates/analysis , Depsides/analysis , Drug Antagonism , Fibroblasts/metabolism , Fibroblasts/pathology , Gingiva/metabolism , Gingiva/pathology , Glutathione/metabolism , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Rosmarinic Acid
10.
Cell Biol Toxicol ; 24(1): 39-53, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17610032

ABSTRACT

The quaternary benzo[c]phenanthridine alkaloid chelerythrine is widely used as an inhibitor of protein kinase C (PKC). However, in biological systems chelerythrine interacts with an array of proteins. In this study, we examined the effects of chelerythrine and sanguinarine on conventional PKCs (cPKCs) and PKC upstream kinase, phosphoinositide-dependent protein kinase 1 (PDK1), under complete inhibition conditions of PKC-dependent oxidative burst. In neutrophil-like HL-60 cells, sanguinarine and chelerythrine inhibited N-formyl-Met-Leu-Phe, phorbol 12-myristate 13-acetate (PMA)-, and A23187-induced oxidative burst with IC(50) values not exceeding 4.6 micromol/L, but the inhibition of PMA-stimulated cPKC activity in intact cells required at least fivefold higher alkaloid concentrations. At concentrations below 10 micromol/L, sanguinarine and chelerythrine prevented phosphorylation of approximately 80 kDa protein and sequestered approximately 60 kDa phosphoprotein in cytosol. Moreover, neither sanguinarine nor chelerythrine impaired PMA-stimulated translocation of autophosphorylated PKCalpha/betaII isoenzymes, but both alkaloids induced dephosphorylation of the turn motif in PKCalpha/betaII. The dephosphorylation did not occur in unstimulated cells and it was not accompanied by PKC degradation. Furthermore, cell treatment with sanguinarine or chelerythrine resulted in phosphorylation of approximately 70 kDa protein by PDK1. We conclude that PKC-dependent cellular events are affected by chelerythrine primarily by multiple protein interactions rather than by inhibition of PKC activity.


Subject(s)
Benzophenanthridines/pharmacology , Isoquinolines/pharmacology , Neutrophils/drug effects , Neutrophils/enzymology , Protein Kinase C/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Alkaloids/chemistry , Alkaloids/pharmacology , Benzophenanthridines/chemistry , Cell Death/drug effects , Cell-Free System , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Isoenzymes/metabolism , Isoquinolines/chemistry , NADPH Oxidases/metabolism , Neutrophils/cytology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Respiratory Burst/drug effects , Substrate Specificity/drug effects
11.
Physiol Res ; 57(3): 427-435, 2008.
Article in English | MEDLINE | ID: mdl-17552871

ABSTRACT

Aryl hydrocarbon receptor (AhR) and glucocorticoid receptor (GR) play crucial role in the regulation of drug metabolizing enzymes and in many essential physiological processes. Cellular signaling by these receptors shares several functional and regulatory features. Here we investigated regulatory cross-talk between these two receptors. Human hepatoma cells (HepG2) were the model of choice. We analyzed the effects of dexamethasone (DEX) and dioxin (TCDD) on i) expression of AhR and GRalpha mRNAs; ii) levels of AhR and GR proteins; iii) transcriptional activities of AhR and GR in reporter assays; iv) 7-ethoxyresorufin-O-deethylase activity (EROD). We found that both DEX and TCDD affected AhR and GR mRNAs expression, proteins levels and transcriptional activities in HepG2 cells. These effects on cellular signaling by AhR and GR comprised up-/down-regulation of gene expression and ligand-dependent protein degradation. We conclude that interactive regulatory cross-talk between GR and AhR receptors in HepG2 cells defines possible implications in physiology and drug metabolism. Future research should be focused on the investigation of AhR-GR cross-talk in various normal human cells and tissues both in vitro and in vivo.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Liver Neoplasms/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Receptor Cross-Talk , Receptors, Aryl Hydrocarbon/agonists , Receptors, Glucocorticoid/agonists , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cytochrome P-450 CYP1A1/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Pilot Projects , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transfection
12.
Gen Physiol Biophys ; 26(3): 173-80, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18063844

ABSTRACT

We investigated the effects of novel microtubules interfering agents (MIAs) in primary cultures of rat hepatocytes. Cells were treated for 24 h with a known compound colchicine and newly synthesized derivatives myoseverin, tubulyzine, and E2GG. We examined the effects of MIAs on microtubules network integrity and on the polymerization capability of isolated tubulin. All tested MIAs inhibited microtubules assembly with the following IC(50) values: tubulyzine (4.4 + or - 0.9 micromol/l), myoseverin (7.0 + or - 0.8 micromol/l), E2GG (16 + or - 2 micromol/l), colchicine (2.0 + or - 0.4 micromol/l). The potency of MIAs to perturb microtubular network integrity (monitored by immune-histochemistry) increased in the order tubulyzine < myoseverin < E2GG < colchicine. We described recently deleterious effects of MIAs on the expression of drug metabolizing enzymes, including CYP1A1. Here we observed inhibitory effects of novel MIAs on dioxin-inducible expression of CYP1A1 mRNA in rat hepatocytes. We conclude that novel MIAs exert analogical biological response as classical MIAs such as colchicine or nocodazole. This further supports the hypothesis that tubulin is the primordial target of MIAs within the cell and that perturbation of microtubules dynamics and/or integrity triggers the biological effects described here.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Microtubules/chemistry , Microtubules/metabolism , Purines/administration & dosage , Triazines/administration & dosage , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Microtubules/drug effects , Protein Binding/drug effects , Rats
13.
Gen Physiol Biophys ; 26(3): 240-2, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18063853

ABSTRACT

In the present work, the effects of colchicine (COL) and/or all-trans retinoic acid (ATRA) on expression of rexinoid receptors (RXRs) (alpha, beta, gamma), thyroid hormone receptor alpha and coregulators N-CoR, SMRT and SRC-1 mRNA in primary rat hepatocytes as a model of no-proliferating cells were investigated. Treatment with these components, either alone or in combination, induced differences of the expression profiles between distinct treatment groups.


Subject(s)
Colchicine/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/metabolism , Tretinoin/administration & dosage , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Rats
14.
Placenta ; 28(10): 1004-11, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17572486

ABSTRACT

The placental trophoblast at different stages of pregnancy contains some drug transporters and xenobiotic-metabolising enzymes, as well as ligand-activated nuclear receptors, which control their inducible transcriptional regulation. Glucocorticoid receptor alpha (GRalpha) is expressed in both placental syncytiotrophoblast and cytotrophoblast. GRalpha was shown to control inducible expression of several enzymes of the cytochrome P-450 family (CYP) and the drug transporter P-glycoprotein in the liver. However, GRalpha-mediated transcriptional regulation of drug transporters and CYPs has not been studied in the placental trophoblast. In this study, we examined the expression and activity of GRalpha in the transcriptional regulation of P-glycoprotein, CYP3A4, and CYP2C9 in placental trophoblast cell lines. Employing RT-PCR, Western blotting, and luciferase gene reporter assay, we detected the expression and activity of GRalpha in JEG3 and BeWo cell lines. However, we observed that only MDR1 mRNA was up-regulated after treatment of placental cells with dexamethasone. Accordingly, only the promoter of the MDR1 gene was activated by dexamethasone in gene reporter assays in placental cells and the activation was abolished by RU486, an antagonist of GRalpha. CYP3A4 and CYP2C9 promoters were activated in placental cells only after co-transfection with hepatocyte nuclear factor 4alpha (HNF4alpha), which indicates the hepatocyte-specific character of GRalpha-mediated regulation of the genes. On the other hand, coexpression of HNF4alpha had no effect on the activation of the MDR1 gene promoter, suggesting HNF4alpha-independent regulation via GRalpha. We conclude that GRalpha may be involved in the transcriptional regulation of P-glycoprotein in the placental trophoblast. We also indicate that the CYP3A4 and CYP2C9 genes are not inducible through GRalpha in placental cell lines, due to the lack of HNF4alpha expression and possibly some additional hepatocyte-specific transcriptional factors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Receptors, Glucocorticoid/physiology , Trophoblasts/metabolism , Cell Line , Cell Line, Tumor , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Dexamethasone/pharmacology , Female , Hepatocyte Nuclear Factor 4/physiology , Humans , Pregnancy , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Transcriptional Activation/physiology
15.
Xenobiotica ; 37(5): 549-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17523056

ABSTRACT

Sanguinarine is an alkaloid with known antibiotic and anti-inflammatory activity and its pharmacokinetics have been studied in the rat after a single oral dose (10 mg kg(-1) body weight). Alkaloid determination in the plasma and liver was carried out by high-performance liquid chromatography-electrospray ionization mass spectrometry (HPLC/ESI-MS). The pharmacokinetic parameters (t(max), c(max), AUC(0-->t) and AUC(0-->infinity)) were determined for sanguinarine and dihydrosanguinarine, the major components detected in plasma. The first step in sanguinarine metabolism in the rat was the reduction of the iminium bond resulting in formation of the less toxic dihydrosanguinarine. Both compounds were completely eliminated from the plasma and liver after 24 h and not detected in urine. After a single oral dose of (3)H-sanguinarine, more than 42% of the ingested radioactivity was present in gastrointestinal tract. Benz[c]acridine, up to date the only sanguinarine metabolite referred to in the literature, was not detected in the plasma, liver or urine.


Subject(s)
Alkaloids/pharmacokinetics , Anti-Infective Agents/pharmacokinetics , Benzophenanthridines/pharmacokinetics , Isoquinolines/pharmacokinetics , Acridines/chemistry , Administration, Oral , Alkaloids/administration & dosage , Alkaloids/blood , Alkaloids/chemistry , Animals , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/blood , Anti-Infective Agents/chemistry , Benzophenanthridines/administration & dosage , Benzophenanthridines/blood , Benzophenanthridines/chemistry , Isoquinolines/administration & dosage , Isoquinolines/blood , Isoquinolines/chemistry , Male , Mass Spectrometry , Rats , Rats, Wistar , Time Factors , Tissue Distribution , Tritium
16.
Cell Biol Toxicol ; 23(2): 63-73, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16964586

ABSTRACT

Recent findings show that colchicine (COL) in submicromolar concentrations downregulates the expression of major drug-metabolizing P450 enzymes in human hepatocytes. Concomitantly, the expression of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) was diminished by COL, whereas expression of glucocorticoid receptor (GR) remained unaltered. A tentative mechanism is perturbation of the GR-PXR/CAR-CYP2/3 signaling cascade, resulting in restricted transcriptional activity of GR receptor by colchicine. In this work we focused on 10-demethylcolchicine (colchiceine; EIN), a structural analogue and a putative metabolite of COL that lacks tubulin-binding activity. We investigated the effects of EIN on the expression of PXR, CAR, and GR receptors in primary cultures of human hepatocytes. In contrast with the effects of COL, EIN does not alter the expression of PXR, CAR, and/or GR receptors mRNAs. In addition, EIN had no effects on transcriptional activities of PXR, CAR, and GR receptors in reporter gene assays using transfected cell lines. Considering that COL and EIN are structurally very close and differ only in their tubulin-binding activity, the data presented imply that the deleterious effects of COL on the GR-PXR/CAR-CYP2/3 cascade are primarily due to perturbation of the microtubule network. Our data support the idea of replacing COL by EIN, which is less toxic and does not interact with xenoreceptors.


Subject(s)
Colchicine/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Cells, Cultured , Colchicine/analogs & derivatives , Constitutive Androstane Receptor , Gene Expression/drug effects , Genes, Reporter , Humans , Pharmaceutical Preparations/metabolism , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Steroid/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transfection
17.
Cell Biol Toxicol ; 22(6): 439-53, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16964588

ABSTRACT

We compared the effects of chelerythrine (CHE) and sanguinarine (SA) on human prostate cancer cell lines (LNCaP and DU-145) and primary culture of human gingival fibroblasts. CHE and SA treatment of cell lines for 24 h resulted in (1) inhibition of cell viability in a dose-dependent manner in all tested cells (as evaluated by MTT test and bromodeoxyuridine incorporation assay); (2) dose-dependent increase in DNA damage in all tested cells (as evaluated by DNA comet assay); (3) changes in apoptosis (assessed by western blot analysis and TUNEL assay); and (4) significant induction of cyclin kinase inhibitors p21(Waf1/Cip1) and p27(Kip1) in prostate cancer cells (identified by western blot analysis). Our study demonstrates that CHE had significant cytotoxic effect, independent of p53 and androgen status, on human prostate cancer cell lines. Normal gingival fibroblasts and DU-145 cells were more sensitive to the treatment with both alkaloids than were LNCaP cells. CHE and SA may be prospective natural molecules for use in the treatment of prostate cancer owing to their involvement in apoptosis and cell cycle regulation.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Cell Cycle/drug effects , Gingiva/drug effects , Isoquinolines/pharmacology , Prostatic Neoplasms/drug therapy , Blotting, Western , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Damage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fibroblasts/drug effects , Fibroblasts/pathology , Formazans/metabolism , Gingiva/pathology , Humans , In Situ Nick-End Labeling , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tetrazolium Salts/metabolism
18.
Cell Biol Toxicol ; 22(2): 91-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16528448

ABSTRACT

The biological activity of methanolic and aqueous extracts from dehydrated hypocotyls of Lepidium meyenii (Brassicaceae, vernacular name "maca"), was studied on rat hepatocytes and human breast cancer MCF-7 cells. The extracts did not exhibit cytotoxicity in hepatocyte primary cultures up to 10 mg/ml as measured by the MTT viability test, and lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) leakage. Moreover, after 72 h, extracts inhibited LDH and AST leakage from the hepatocytes. When hepatocytes were intoxicated by t-butyl hydroperoxide, neither extract prevented oxidative damage. Both extracts showed weak antioxidant activity in the DPPH radical scavenging test with IC(50) values of 3.46 +/- 0.16 and 0.71 +/- 0.10 mg/ml, for aqueous and methanolic extracts, respectively. Thus, the observed effect on spontaneous enzyme leakage is probably mediated through mechanisms other than antioxidant activity. Both methanolic and aqueous extracts have shown estrogenic activity comparable with that of silymarin in MCF-7 cell line. Maca estrogenicity was exhibited in the range from 100 to 200 mug of extract per ml. The findings in the present study show that maca does not display in vitro hepatotoxicity. In contrast, a slight cytoprotective effect, probably not mediated by antioxidant capacity, was noted. Maca extracts exhibited estrogenic activity comparably to the effect of silymarin in MCF-7 cells.


Subject(s)
Breast Neoplasms/drug therapy , Drug Evaluation, Preclinical , Hepatocytes/drug effects , Lepidium/chemistry , Lepidium/toxicity , Plant Extracts/toxicity , Animals , Biphenyl Compounds/analysis , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Estrogens/pharmacology , Fatty Acids/analysis , Humans , Hydrazines/analysis , Male , Picrates , Plant Extracts/chemistry , Rats , Rats, Wistar , Steroids/analysis
19.
Cell Biol Toxicol ; 22(2): 81-90, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16528449

ABSTRACT

The flavonolignan silybin and its derivative dehydrosilybin have been proposed as candidate UV-protective agents in skin care products. This study addressed the effect of silybin and dehydrosilybin on the activity of cytochrome P450 isoform CYP1A1 in human keratinocytes (HaCaT) and human hepatoma cells (HepG2). CYP1A1 catalytic activity was assessed as O-deethylation of 7-ethoxyresorufin using fluorescence detection. Silybin and dehydrosylibin inhibited basal and dioxin-inducible CYP1A1 catalytic activity in both cell lines used. The inhibitory effect of tested compounds was more pronounced in HaCaT cells than in HepG2 cells, and dehydrosilybin was a much stronger inhibitor than silybin. Analyses on CYP1A1 human recombinant protein yielded IC(50) values of 22.9 +/- 4.7 micromol/L and 0.43 +/- 0.04 micromol/L for silybin and dehydrosilybin, respectively. Since CYP1A enzymes are some of the most prominent actors in the process of chemically induced carcinogenesis, the inhibitory activity of the flavonolignans tested against CYP1A1 favors their use as cytoprotective agents in terms of skin and hepatic metabolism. In addition, the capability of dehydrosilybin to inhibit CYP1A1 in submicromolar concentrations makes this compound a potential biological probe in CYP1A1 analyses.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP1A1/metabolism , Liver Neoplasms/enzymology , Silymarin/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Dioxins , Dose-Response Relationship, Drug , Enzyme Induction , Humans , Keratinocytes/enzymology , Molecular Structure , Silybin , Time Factors
20.
Food Chem Toxicol ; 44(2): 242-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16115718

ABSTRACT

Quaternary benzo[c]phenanthridine alkaloids (QBA) sanguinarine and chelerythrine exhibit a wide spectrum of biological activities whence they are used in dental care products. Recent studies indicated that cytochrome P450 CYP1A attenuates sanguinarine toxicity both in vivo [Williams, M.K., Dalvi, S., Dalvi, R.R., 2000. Influence of 3-methylcholanthrene pretreatment on sanguinarine toxicity in mice. Vet. Hum. Toxicol. 42, 196-198] and in vitro [Vrba, J., Kosina, P., Ulrichová, J., Modrianský, M., 2004. Involvement of cytochrome P450 1A in sanguinarine detoxication. Toxicol. Lett. 151, 375-387]. However, CYP1A converts sanguinarine to the products that form DNA adducts [Stiborová, M., Simánek, V., Frei, E., Hobza, P., Ulrichová, J., 2002. DNA adduct formation from quaternary benzo[c]phenanthridine alkaloids sanguinarine and chelerythrine as revealed by the 32P-postlabeling technique. Chem. Biol. Interact. 140, 231-242]. In our work we examined the effects of sanguinarine and chelerythrine on CYP1A1 expression and catalytic activity in human hepatoma cells-HepG2. Sanguinarine and chelerythrine did not affect basal and dioxin-inducible expression of CYP1A1 mRNA and protein in HepG2 cells. The enzymatic activity of CYP1A1 was assessed by the fluorescent measurement of 7-ethyxoresorufin-O-deethylase (EROD) activity. We observed a slight decrease of dioxin-induced EROD activity in HepG2 cells by sanguinarine and chelerythrine. This decrease was attributed to the inhibition of CYP1A1 catalytic activity, as revealed by enzyme kinetic studies on recombinant CYP1A1 protein. The IC50 values for the inhibition of CYP1A1 by sanguinarine and chelerythrine were 2.1 and 1.9muM, respectively. In conclusion, albeit the CYP1A modulates QBA cytotoxicity and genotoxicity, the QBA themselves do not affect CYP1A1 expression. The data indicate that studied alkaloids do not have specific cellular target and their biological effects are rather pleiotropic.


Subject(s)
Alkaloids/pharmacology , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/biosynthesis , Enzyme Inhibitors/pharmacology , Liver Neoplasms/enzymology , Phenanthridines/pharmacology , Benzophenanthridines , Catalysis , Cell Line, Tumor , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2 Inhibitors , Humans , Isoquinolines , RNA, Messenger/biosynthesis , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...