Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 826: 106-113, 2018 May 05.
Article in English | MEDLINE | ID: mdl-29501870

ABSTRACT

In the present study, the functional properties of α7 nicotinic acetylcholine receptors (α7 nAChRs) and N-methyl-D-aspartate receptors (NMDARs) endogenously expressed in SH-SY5Y human neuroblastoma cells were characterized in an extracellular-signal regulated kinase (ERK) phosphorylation assay. Both choline and N-methyl-D-aspartate (NMDA) mediated robust concentration-dependent increases in ERK phosphorylation in the SH-SY5Y cells, exhibiting EC50 values in good agreement with those reported for the agonists at recombinant α7 nAChRs and NMDARs, respectively. Importantly, the responses evoked by choline (10 mM) and by NMDA (50 µM) were significantly inhibited by the α7-selective antagonist α-bungarotoxin (100 nM) and by the NMDAR-selective antagonist MK-801 (50 µM), respectively. The increased ERK phosphorylation levels observed upon co-application of choline (1, 3, 10 mM) and NMDA (50 µM) compared to those produced by the two agonists on their own were fully reconcilable with additive effects and did not reveal substantial synergy between α7 nAChR and NMDAR signaling. Interestingly, however, the responses evoked by the "choline (10 mM) - NMDA (50 µM)" combination were almost completely inhibited by α-bungarotoxin (100 nM) as well as by MK-801 (50 µM), suggesting some sort of a link between α7 nAChR- and NMDAR-mediated ERK phosphorylation. Finally, oligomeric amyloid-ß1-42 peptide (1000 nM) mediated robust inhibition of the ERK phosphorylation induced by choline (10 mM), NMDA (50 µM) and the "choline (10 mM) - NMDA (50 µM)" combination. In conclusion, ERK phosphorylation measurements in SH-SY5Y cells provides a robust assay for studies of α7 nAChR- and NMDAR-mediating signaling and putative functional interactions between the receptors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Amyloid beta-Peptides/pharmacology , Bungarotoxins/pharmacology , Cell Line, Tumor , Choline/pharmacology , Dizocilpine Maleate/pharmacology , Humans , N-Methylaspartate/pharmacology , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
2.
PLoS One ; 12(12): e0189513, 2017.
Article in English | MEDLINE | ID: mdl-29261717

ABSTRACT

α7 nicotinic acetylcholine receptors (nAChRs) and N-methyl-D-aspartate receptors (NMDARs) are key mediators of central cholinergic and glutamatergic neurotransmission, respectively. In addition to numerous well-established functional interactions between α7 nAChRs and NMDARs, the two receptors have been proposed to form a multimeric complex, and in the present study we have investigated this putative α7 nAChR/NMDAR assembly in human and murine brain tissues. By α-bungarotoxin (BGT) affinity purification, α7 and NMDAR subunits were co-purified from human and murine cortical and hippocampal homogenates, substantiating the notion that the receptors are parts of a multimeric complex in the human and rodent brain. Interestingly, the ratios between GluN1 and α7 levels in BGT pull-downs from cortical homogenates from Alzheimer's disease (AD) brains were significantly lower than those in pull-downs from non-AD controls, indicating a reduced degree of α7 nAChR/NMDAR complex formation in the diseased tissue. A similar difference in GluN1/α7 ratios was observed between pull-downs from cortical homogenates from adult 3xTg-AD and age-matched wild type (WT) mice, whereas the GluN1/α7 ratios determined in pull-downs from young 3xTg-AD and age-matched WT mice did not differ significantly. The observation that pretreatment with oligomeric amyloid-ß1-42 reduced GluN1/α7 ratios in BGT pull-downs from human cortical homogenate in a concentration-dependent manner provided a plausible molecular mechanism for this observed reduction. In conclusion, while it will be important to further challenge the existence of the putative α7 nAChR/NMDAR complex in future studies applying other methodologies than biochemical assays and to investigate the functional implications of this complex for cholinergic and glutamatergic neurotransmission, this work supports the formation of the complex and presents new insights into its regulation in healthy and diseased brain tissue.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Adult , Animals , Case-Control Studies , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Protein Binding
3.
Fitoterapia ; 90: 151-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23892001

ABSTRACT

Increased consumption of green tea (GT) without enough scientific data has raised safety concerns. Epigallocatechin 3-gallate (EGCG) is the most prominent polyphenol of GT that has antioxidant activity. However, higher doses of EGCG have been shown to cause liver injury. This study was initiated to determine the effect of GT extracts in a mouse model. We also investigated the effects of EGCG in normal and health-compromised mice. Different doses of GT fractions and EGCG were administered for 5 days to mice. Also, a single dose of lipopolysaccharide (LPS) was combined with EGCG in order to investigate its effect in the presence of fever. Plasma ALT and ALP levels were determined along with liver histopathology. Combining a single high IG dose of EGCG with a single IP dose of LPS initiated liver injury. Furthermore, repeated administration of high IG doses of EGCG showed mild liver injury, but it was augmented under febrile conditions induced by LPS. This study confirms the safety of reasonable consumption of GT over a short term. However, it highlights a caution that high doses of EGCG can lead to mild liver injury, and this may be markedly enhanced under febrile conditions.


Subject(s)
Camellia sinensis/chemistry , Catechin/analogs & derivatives , Chemical and Drug Induced Liver Injury , Liver/drug effects , Plant Extracts/adverse effects , Polyphenols/adverse effects , Animals , Camellia sinensis/adverse effects , Catechin/adverse effects , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Fever/chemically induced , Lipopolysaccharides , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred Strains , Tea
4.
Br J Pharmacol ; 166(8): 2231-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22428706

ABSTRACT

BACKGROUND AND PURPOSE: So far, there is only limited information about the regulation of the endogenous synthesis of hydrogen sulfide (H(2) S), an important gaseous signalling molecule. This study was done to evaluate the redox-dependent signalling events that regulate the expression of the H(2) S synthesising enzyme cystathionine-γ-lyase (CSE) in rat mesangial cells. EXPERIMENTAL APPROACH: The effects of platelet-derived growth factor (PDGF)-BB and antioxidants on CSE expression and activity in cultured rat renal mesangial cells were assessed. Activity of nuclear factor erythroid-2-related factor-2 (Nrf2) was measured as the binding capacity to a radiolabelled consensus element by electrophoretic mobility shift assay (EMSA). Furthermore, CSE and Nrf2 expression was analysed in a rat model of anti-Thy-1-induced glomerulonephritis by immunohistochemistry. KEY RESULTS: Treatment of mesangial cells with PDGF-BB resulted in a marked time- and dose-dependent up-regulation of CSE mRNA and protein levels, as well as CSE activity accompanied with increased formation of reactive oxygen species. Remarkably, co-administration of antioxidants, such as N-acetylcysteine, ebselen or diphenylene iodonium chloride, drastically reduced PDGF-BB-induced CSE expression. PDGF-BB induced binding of Nrf2 to a corresponding consensus antioxidant element in a redox-dependent manner. Furthermore, PDGF-BB-induced CSE expression in mouse mesangial cells was completely abolished in Nrf2 knockout mice compared with wild-type mice. In a rat model of anti-Thy-1-induced proliferative glomerulonephritis, we observed a marked up-regulation of CSE protein paralleled by a stabilization of Nrf2 protein. CONCLUSIONS AND IMPLICATIONS: PDGF-BB regulated CSE via a redox-mediated activation of Nrf2. Such action would aid the resolution of glomerular inflammatory diseases. LINKED ARTICLE: This article is commented on by Gallyas, pp. 2228-2230 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2012.01976.x.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Mesangial Cells/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Animals , Antioxidants/pharmacology , Becaplermin , Cells, Cultured , Cystathionine gamma-Lyase/genetics , Gene Expression Regulation, Enzymologic/physiology , Glomerulonephritis/chemically induced , Glomerulonephritis/metabolism , Isoantibodies/pharmacology , Macrophages , Mesangial Cells/enzymology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spleen/cytology
5.
J Pharm Pharmacol ; 64(3): 344-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22309266

ABSTRACT

OBJECTIVES: Anthracyclines are broad spectrum anticancer drugs with dose-dependent cardiotoxicity. Protein malnutrition commonly occurs in cancer patients and is considered a risk factor for development of cardiotoxicity. This study was designed to assess the modulatory effect of protein malnutrition on the pharmacokinetics and drug disposition properties of a single dose of doxorubicin and epirubicin and how these possible changes will affect the degree of cardiotoxicity of these drugs. METHODS: A single interperitoneal dose of 15 mg/kg of either doxorubicin or epirubicin was injected into rats fed with either normal protein diet or low-protein diet. The plasma concentration-time profiles of doxorubicin and epirubicin and their concentrations in different tissues were determined. Serum creatine kinase level was determined at different time intervals and histopathological examination of heart tissue was carried out. KEY FINDINGS: Protein malnutrition significantly altered the pharmacokinetics of doxorubicin and epirubicin, with a significant decrease in their elimination, and prolonged the exposure of the heart to these drugs. Histopathological examination and serum creatine kinase measurements supported the role of protein malnutrition in enhancement of anthracycline cardiotoxicity. CONCLUSIONS: If similar alteration in anthracyclines' pharmacokinetics occurs in malnourished cancer patients, protein malnutrition will be a risk factor for development of anthracycline cardiotoxicity and dose adjustment will be required in nutritionally deprived patients.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Diet, Protein-Restricted , Doxorubicin/pharmacokinetics , Epirubicin/pharmacokinetics , Heart/drug effects , Protein-Energy Malnutrition/metabolism , Animals , Area Under Curve , Creatine Kinase/blood , Drug Combinations , Food-Drug Interactions , Humans , Injections, Intraperitoneal , Male , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Random Allocation , Rats , Risk Factors
6.
Fertil Steril ; 93(1): 239-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19144333

ABSTRACT

OBJECTIVE: To evaluate the utility of gene therapy for uterine fibroids in the Eker rat model using an adenovirus-mediated delivery of a dominant-negative estrogen receptor gene (Ad-DNER). DESIGN: Animal study. SETTING: University animal laboratory. ANIMAL(S): Twenty-seven female Eker rats. INTERVENTION(S): We randomized Eker rats with magnetic resonance imaging (MRI)-confirmed uterine leiomyomas to a single treatment of direct intrafibroid injection with Ad-DNER, Ad-bacterial ss-galactosidase, or vehicle. MAIN OUTCOME MEASURE(S): Tumor volumes were determined by MRI scanning and caliper measurement. Samples of serum, fibroid tumors, and various organs were collected at 8, 15, and 30 days after treatment to assess treatment safety and efficacy. RESULT(S): The Ad-DNER treatment significantly decreased uterine fibroid volume by 45%, 80%, and 77.4% of pretreatment volume at days 8, 15, and 30, respectively, and modulated the expression of apoptosis-, proliferation-, and extracellular matrix-related genes' compared with control animals. The Ad-DNER did not produce any toxic effects in nontarget tissues. CONCLUSION(S): The Ad-DNER treatment shrinks Eker rats' fibroids, in part, via modulation of several estrogen-regulated genes. This safe gene therapy approach presents a promising conservative treatment option for women with symptomatic uterine fibroids.


Subject(s)
Adenoviridae/genetics , Estrogen Receptor alpha/genetics , Genetic Therapy/methods , Genetic Vectors , Leiomyoma/therapy , Uterine Neoplasms/therapy , Adenoviridae/immunology , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Disease Models, Animal , Estrogen Receptor alpha/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Therapy/adverse effects , Immunity, Humoral , Leiomyoma/genetics , Leiomyoma/immunology , Leiomyoma/metabolism , Leiomyoma/pathology , Magnetic Resonance Imaging , Mutation , Rats , Rats, Mutant Strains , Time Factors , Transfection , Tuberous Sclerosis Complex 2 Protein , Tumor Burden , Tumor Suppressor Proteins/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/immunology , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
7.
Basic Clin Pharmacol Toxicol ; 105(1): 30-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19371263

ABSTRACT

In the present study, we have addressed the possible protective role of acetyl-L-carnitine in caerulein-induced acute pancreatitis in male Swiss albino rats. Acute pancreatitis paradigm was developed by challenging animals with a supramaximal dose of caerulein (20 microg/kg, SC) four times at hourly intervals. Caerulein induced acute pancreatitis that was well-characterized morphologically and biochemically. Severe oedema with marked increased relative pancreatic weight, marked atrophy of acini with increased interacinar spaces, vacuolization, and extensive leucocytic infiltration were diagnostic fingerprints of the pancreatitis phenotype. A biochemical test battery that confirmed the model comprised increased plasma amylase and lipase activities, calcium levels as well as increased pancreatic enzymatic myeloperoxidase and glutathione-S-transferase activities, beside increased pancreatic contents of nitric oxide and malondialdehyde and reduced pancreatic glutathione level. Prior administration of acetyl-L-carnitine (200 mg/kg, IP) for seven consecutive days ahead of caerulein challenge alleviated all the histological and biochemical manifestations of acute pancreatitis. These results suggest a possible protective role of the carnitine ester in such a murine acute pancreatitis model probably via regulation of the oxidant/antioxidant balance, beside modulation of the myeloperoxidase and nitric oxide systems, which are involved in the inflammatory cascade that most often associate the disease.


Subject(s)
Acetylcarnitine/pharmacology , Ceruletide/toxicity , Pancreatitis/chemically induced , Acetylcarnitine/administration & dosage , Amylases/blood , Animals , Calcium/blood , Ceruletide/administration & dosage , Disease Models, Animal , Glutathione/analysis , Glutathione Transferase/metabolism , Injections, Intraperitoneal , Injections, Subcutaneous , Lipase/blood , Lipid Peroxidation/drug effects , Male , Malondialdehyde/analysis , Nitric Oxide/analysis , Pancreas/chemistry , Pancreas/enzymology , Pancreas/pathology , Peroxidase/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacology , Rats
8.
Food Chem Toxicol ; 47(6): 1311-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19285535

ABSTRACT

We have addressed in this study the possible protective role of the main principle of turmeric pigment; curcumin on a murine model of ulcerative colitis (UC). Colitis was induced by administration of dextran sulfate sodium (DSS) (3% W/V) in drinking water to male Swiss albino rats for 5 consecutive days. DSS challenge induced UC model that was well characterized morphologically and biochemically. DSS produced shrinkage of colon length and increased the relative colon weight/length ratio accompanied by mucosal edema and bloody stool. Histologically, DSS produced submucosal erosions, ulceration, inflammatory cell infiltration and crypt abscess as well as epithelioglandular hyperplasia. The model was confirmed biochemically, and the test battery entailed elevated serum tumor necrosis factor (TNF-alpha) and colonic activity of myleoperoxidase (MPO). Colonic glutathione-S-transferase (GST) activity and its substrate concentration; GSH, were notably reduced, while lipid peroxidation, expressed as malondialdehyde (MDA) level, and total nitric oxide (NO) were significantly increased. Prior administration of curcumin (100mg/kg, IP) for 7 consecutive days ahead of DSS challenge mitigated the injurious effects of DSS and ameliorated all the altered biochemical parameters. These results suggest that curcumin could possibly have a protective role in ulcerative colitis probably via regulation of oxidant/anti-oxidant balance and modulation of the release of some inflammatory endocoids, namely TNF-alpha and NO.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/prevention & control , Curcumin/therapeutic use , Dextran Sulfate/antagonists & inhibitors , Dextran Sulfate/toxicity , Animals , Colitis, Ulcerative/pathology , Colon/enzymology , Glutathione/metabolism , Glutathione Transferase/metabolism , Intestinal Mucosa/pathology , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mice , Nitric Oxide/metabolism , Peroxidase/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
9.
Am J Obstet Gynecol ; 199(2): 156.e1-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18468574

ABSTRACT

OBJECTIVE: This study was undertaken to develop a representative murine model for human leiomyoma. STUDY DESIGN: Human fibroid tumor tissues were cut into small pieces and treated with medium alone, adenoviral-beta-galactosidase, adenoviral-vascular endothelial growth factor-A, adenoviral-cyclooxygenase-2, or both adenoviral-vascular endothelial growth factor-A and adenoviral- cyclooxygenase-2. Tissue pieces were inserted subcutaneously in the flank of each severe combined immunodeficient mouse. The developed lesion was measured twice per week. Xenograft tissues were harvested after 30 days and analyzed. RESULTS: Tissue pieces transfected with both adenoviral-cyclooxygenase-2 and adenoviral-vascular endothelial growth factor-A continued to grow up to 30 days postimplantation. The number of proliferating and apoptotic cells, as well as the expression of smooth muscle actin, desmin, vimentin, estrogen receptors, and progesterone receptors was similar between retrieved grafts from that group and the original patient tissue. Furthermore, hematoxylin and eosin and Masson's Trichrome stains confirmed this similarity. CONCLUSION: Human uterine leiomyoma xenografts, pretreated with both adenoviral- cyclooxygenase-2 and adenoviral-vascular endothelial growth factor-A and implanted subcutaneously in severe combined immunodeficient mice, represent a novel model for human uterine leiomyoma.


Subject(s)
Disease Models, Animal , Leiomyoma , Uterine Neoplasms , Adenoviridae/genetics , Adenoviridae/metabolism , Adult , Animals , Apoptosis , Cell Proliferation , Collagen/biosynthesis , Cyclooxygenase 2/metabolism , Female , Genetic Vectors , Humans , Leiomyoma/metabolism , Mice , Mice, SCID , Neoplastic Processes , Neovascularization, Pathologic , Transfection , Transplantation, Heterologous , Uterine Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , beta-Galactosidase/metabolism
10.
Mutat Res ; 651(1-2): 105-13, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18069049

ABSTRACT

Aneuploidy of germ cells contributes to reduced fertility, foetal wastage and genetic defects. The possible risk of aneuploidy induction by the cancer chemotherapeutic drugs amsacrine (AMSA) and nocodazole (NOC) was investigated in male mice. Two molecular cytogenetic approaches were used: (1) the BrdU-incorporation assay to test the altered duration of meiotic divisions and (2) the sperm-FISH assay to determine aneuploidy induction during meiosis by observing hyperhaploid and diploid sperm. Sperm were sampled from the Caudae epididymes of treated and solvent control males. Single intraperitoneal injections with NOC (35 mg/kg) and AMSA (15 mg/kg) caused a meiotic delay of 24h. The timing of sperm sampling for the sperm-FISH assay was adjusted accordingly, i.e. 23 days after treatment. Mice were treated with 18, 35 and 50 mg/kg of NOC, or 5, 10, 15 and 20 mg/kg of AMSA. Significant dose-dependent increases above the concurrent controls in the frequencies of hyperhaploid sperm were found with both agents. Significant increases in the frequencies of diploid sperm were found only with AMSA. These results provide a basis for genetic counselling of patients under AMSA or NOC chemotherapy. During a period of 3-4 months after the end of chemotherapy, they may stand a higher risk of siring chromosomally abnormal offspring.


Subject(s)
Amsacrine/adverse effects , Meiosis/drug effects , Nocodazole/adverse effects , Nondisjunction, Genetic/drug effects , Spermatocytes/drug effects , Amsacrine/administration & dosage , Aneuploidy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , In Situ Hybridization, Fluorescence , Injections, Intraperitoneal , Male , Meiosis/genetics , Mice , Nocodazole/administration & dosage , Nondisjunction, Genetic/genetics , Spermatocytes/metabolism
11.
J Clin Endocrinol Metab ; 92(10): 3949-57, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17635941

ABSTRACT

CONTEXT: Human uterine leiomyomas are very common smooth muscle cell tumors that occur in reproductive-age women and are the leading reason for performing hysterectomies. The present study was conducted to explore the potential mechanism behind the effects exerted by dominant-negative estrogen receptors (DNERs) delivered by adenovirus to leiomyoma cells to ascertain the utility of DNERs as a novel strategy for treatment of uterine fibroids. OBJECTIVE AND METHODS: We investigated the ability of DNER to affect estrogen response element (ERE) activity induced by wild-type estrogen receptor (ER) by using the adenovirus ERE luciferase (Ad-ERE-luc) system in ELT3 cells and the effect of graded doses of DNER (10, 50, and 100 plaque-forming units/cell) on the expression of some selected genes controlling cultured human leiomyoma cell proliferation (cyclin D1, Cox2, PCNA, VEGF, and EGF), apoptosis (Bcl2 and Bax), estrogen metabolism (COMT), and extracellular matrix formation (MMP(1)) as well as progesterone receptors (A and B) were assessed using Western blot analysis. These genes are all regulated by estrogen and/or progesterone. RESULTS: DNER has the ability to suppress the ERE luc activity induced by wild-type ER (P < 0.01) and significantly (P < 0.05) reverse the expression of all estrogen- and progesterone-regulated genes in this study. CONCLUSIONS: These results suggest that interruption of the estrogen signaling pathway using DNER results in modulation of both estrogen- and progesterone-regulated genes that control leiomyoma cell apoptosis, proliferation, extracellular matrix formation, progesterone receptors, and estrogen metabolism, which might account for the DNER mechanism of action.


Subject(s)
Genetic Therapy/methods , Leiomyoma/therapy , Receptors, Estrogen/genetics , Uterine Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis/physiology , Catechol O-Methyltransferase/genetics , Cell Line, Tumor , Down-Regulation/physiology , Epidermal Growth Factor/metabolism , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Genes, Dominant , Leiomyoma/physiopathology , Matrix Metalloproteinase 1/genetics , Progesterone/metabolism , Rats , Receptors, Progesterone/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Transfection , Up-Regulation/physiology , Uterine Neoplasms/physiopathology , Vascular Endothelial Growth Factor A/metabolism
12.
Eur J Pharmacol ; 564(1-3): 181-9, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17462625

ABSTRACT

A murine lung fibrosis model has been induced by challenging male Swiss albino mice with a fibrotic dose of bleomycin (10 mg/kg body weight, s.c.) twice weekly for 6 weeks. The model has been characterized and confirmed biochemically, histologically and morphometrically. Keeping in mind that inflammation is the forerunner of lung fibrosis, we have investigated the possible anti-fibrotic effect of meloxicam; a selective COX-2 inhibitor, in this lung fibrosis paradigm. When administered ahead of bleomycin challenge, meloxicam significantly reduced the lung content of hydroxyproline; the backbone of collagen matrix. This was further confirmed by the lower collagen deposition as revealed by histochemical examination of lung sections. Meloxicam had also anti-oxidant effect as shown by increase in lung reduced glutathione (GSH) level and decreases in lung malonedialdehyde (MDA) content and myeloperoxidase (MPO) activity. Besides, meloxicam has shown an apparent angiostatic activity. Histologically, meloxicam lessened lung inflammation and fibrotic changes induced by bleomycin. Taken together, one could conclude that meloxicam has shown anti-fibrotic effect in the bleomycin lung fibrosis model. Apart from its well-known anti-inflammatory potential, this anti-fibrotic action of meloxicam resides most probably, at least partly, in its anti-oxidant and angiostatic effects.


Subject(s)
Antioxidants/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Thiazines/pharmacology , Thiazoles/pharmacology , Animals , Bleomycin , Collagen/drug effects , Collagen/metabolism , Disease Models, Animal , Glutathione/drug effects , Glutathione/metabolism , Hydroxyproline/drug effects , Hydroxyproline/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Meloxicam , Mice , Neovascularization, Pathologic/drug therapy , Peroxidase/drug effects , Peroxidase/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
13.
Food Chem Toxicol ; 45(1): 88-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17011106

ABSTRACT

Thymoquione (TQ), the main constituent of the volatile oil of Nigella sativa seeds, has been shown to protect mice against benzo(a)pyrene [B(a)P]-induced forestomach carcinogenesis. The present investigation was undertaken to study the possible chemopreventive activity of TQ, supplemented in the drinking water, against B(a)P-induced chromosomal aberrations (CAs) in mouse bone marrow cells. Male Swiss albino mice received TQ (0.01% in drinking water) daily for 28 days. The daily dose of TQ was estimated to be 10mg/kg based on the calculated average daily water consumption by mice. From day 9, the carcinogen, B(a)P, was given by gastric intubation at dose level of 50mg/kg on alternative days for a total of 8 doses. On day 29, all mice were transferred to a normal drinking tap water. Control groups received corn oil vehicle, TQ alone or B(a)P alone. All mice were sacrificed at 12 weeks after the end of the treatment. Chromosome preparations were made of bone marrow. Cytogenetic end points screened were the frequencies of CAs and damaged cells induced. Daily intake of TQ after and before or during exposure to B(a)P significantly reduced the frequencies of CAs and damaged cells compared to the highly clastogenic activity of B(a)P alone.


Subject(s)
Antimutagenic Agents/pharmacology , Benzo(a)pyrene/toxicity , Benzoquinones/pharmacology , Chromosome Aberrations/drug effects , Mutagens/toxicity , Nigella/chemistry , Administration, Oral , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Chemoprevention , Chromosome Aberrations/chemically induced , Male , Mice , Seeds/chemistry , Water Supply
14.
World J Gastroenterol ; 12(33): 5363-7, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16981269

ABSTRACT

AIM: To study the effects of ferulic acid on gastrointestinal motility both in vitro and in vivo. METHODS: Ferulic acid induced concentration-dependent stimulation of the basal tone of isolated guinea pig ileum (2-20 micromol/L) and isolated rat fundus (0.05-0.4 mmol/L). RESULTS: Ferulic acid significantly accelerated the gastrointestinal transit and gastric emptying in rats in a dose-dependent manner (50-200 mg/kg, po). Cisplatin (2.5-20 mg/kg, ip) induced a dose-dependent delay in gastric emptying in rats. Pretreatment with ferulic acid dose-dependently, significantly reversed the cisplatin-induced delay in gastric emptying. CONCLUSION: The endogenous prostaglandins (PGs) are involved in mediating the stimulant effects of ferulic acid. This effect of dietary ferulic acid may help improve other accompanying gastrointestinal symptoms such as abdominal discomfort and also may protect against emesis induced by cytotoxic drugs.


Subject(s)
Cholagogues and Choleretics/pharmacology , Cisplatin/pharmacology , Coumaric Acids/pharmacology , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Animals , Female , Gastrointestinal Transit/drug effects , Guinea Pigs , Ileum/drug effects , In Vitro Techniques , Male , Prostaglandins/metabolism , Rats , Rats, Wistar
15.
Br J Pharmacol ; 145(4): 415-23, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15806111

ABSTRACT

1 To clarify the mechanism of mast cell TNF secretion, especially its release process after being produced, we utilized an antiallergic drug, azelastine (4-(p-chlorobenzyl)-2-(hexahydro-1-methyl-1H-azepin-4-yl)-1-(2H)- phthalazinone), which has been reported to inhibit TNF release without affecting its production in ionomycin-stimulated RBL-2H3 cells. 2 Such inhibition was associated with the suppression of an ionomycin-induced increase in membrane-associated PKC activity rather than the suppression of Ca2+ influx, suggesting that PKC might be involved in TNF release process. 3 To see whether conventional PKC family (cPKCs) are involved, we investigated the effects of a selective cPKC inhibitor (Gö6976) and an activator (thymeleatoxin) on TNF release by adding them 1 h after cell stimulation. By this time, TNF mRNA expression had reached its maximum. Gö6976 markedly inhibited TNF release, whereas thymeleatoxin enhanced it, showing a key role of cPKC in TNF post-transcriptional process, possibly its releasing step. 4 To determine which subtype of cPKCs could be affected by azelastine, Western blotting and live imaging by confocal microscopy were conducted to detect the translocation of endogenous cPKC (alpha, betaI and betaII) and transfected GFP-tagged cPKC, respectively. Both methods clearly demonstrated that 1 microM azelastine selectively inhibits ionomycin-triggered translocation of (alpha)PKC without acting on betaI or betaIIPKC. 5 In antigen-stimulated cells, such a low concentration of azelastine did not affect either (alpha)PKC translocation or TNF release, suggesting a functional link between (alpha)PKC and the TNF-releasing step. 6 These results suggest that (alpha)PKC mediates the TNF release process and azelastine inhibits TNF release by selectively interfering with the recruitment of (alpha)PKC in the pathway activated by ionomycin in RBL-2H3 cells.


Subject(s)
Protein Kinase C/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Western , Carbazoles/pharmacology , Cell Line, Tumor , Dinitrophenols/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Indoles/pharmacology , Ionomycin/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/pathology , Microscopy, Confocal , Phthalazines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serum Albumin, Bovine/pharmacology , Transfection , Tumor Necrosis Factor-alpha/genetics
16.
Clin Exp Pharmacol Physiol ; 31(12): 856-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15659049

ABSTRACT

1. Doxorubicin (DOX), a standard chemotherapeutic anthracycline agent, causes a positive inotropic effect in guinea-pig isolated atria in a concentration-dependent manner with an ED(50) of 3.6 micromol/L. This increase in contractility is strictly related to the generation of reactive oxygen species (ROS) as a consequence of quinone metabolism. The ED(50) of DOX is significantly increased (P < 0.05) in the presence of 150 U superoxide dismutase (SOD). In the heart, DOX may be subjected to one- or two-electron reductions catalysed by flavoenzymes in the presence of suitable electron donors. Two-electron reduction is catalysed by NAD(P)H quinone acceptor oxidoreductase (DT-diaphorase; DTD). Whether DOX will be activated or detoxified by two-electron reduction is important for the understanding of the mechanism of both the toxic and antitumour actions of DOX. 2. In order to assess the role of DTD in cardiac responses to DOX, we examined the effect of both a specific inhibitor (dicoumarol) and an inducer (3-methylcholanthrene; MCA) of the enzyme on the inotropic action of DOX. 3. In guinea-pig isolated left atria, 4 micromol/L dicoumarol significantly enhanced the positive inotropic effect of DOX, especially at lower concentrations of DOX. In atria isolated from guinea-pigs treated with MCA (44 mg/kg, i.p. for 4 days), DTD activity was enhanced (approximately twice that of the control; P < 0.01), whereas the activity of glutathione S-transferase (GST) was not significantly altered. In these preparations, DOX caused a significantly lower increase in force of contraction than in atria isolated from untreated animals. 4. These results demonstrate that cardiac DTD does not contribute to ROS generation, but represents a detoxification system.


Subject(s)
Doxorubicin/pharmacology , Myocardial Contraction/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Enzyme Induction/physiology , Enzyme Inhibitors/pharmacology , Guinea Pigs , Heart Atria/drug effects , Heart Atria/enzymology , In Vitro Techniques , Inactivation, Metabolic/physiology , Male , Myocardial Contraction/physiology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Stimulation, Chemical
17.
J Biol Chem ; 278(51): 51758-69, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14523003

ABSTRACT

Renal mesangial cells express high levels of matrix metalloproteinase 9 (MMP-9) in response to inflammatory cytokines such as interleukin (IL)-1 beta. We demonstrate here that the stable ATP analog adenosine 5'-O-(thiotriphosphate) (ATP gamma S) potently amplifies the cytokine-induced gelatinolytic content of mesangial cells mainly by an increase in the MMP-9 steady-state mRNA level. A Luciferase reporter gene containing 1.3 kb of the MMP-9 5'-promoter region showed weak responses to ATP gamma S but conferred a strong ATP-dependent increase in Luciferase activity when under the additional control of the 3'-untranslated region of MMP-9. By in vitro degradation assay and actinomycin D experiments we found that ATP gamma S potently delayed the decay of MMP-9 mRNA. Gel-shift and supershift assays demonstrated that three AU-rich elements (AREs) present in the 3'-untranslated region of MMP-9 are constitutively bound by complexes containing the mRNA stabilizing factor HuR. The RNA binding of these complexes was markedly increased by ATP gamma S. Mutation of each ARE element strongly impaired the RNA binding of the HuR containing complexes. Reporter gene assays revealed that mutation of one ARE did not affect the stimulatory effects by ATP gamma S, but mutation of all three ARE motifs caused a loss of ATP-dependent increase in luciferase activity without affecting IL-1 beta-inducibility. By confocal microscopy we demonstrate that ATP gamma S increased the nucleo cytoplasmic shuttling of HuR and caused an increase in the cytosolic HuR level as shown by cell fractionation experiments. Together, our results indicate that the amplification of MMP-9 expression by extracellular ATP is triggered through mechanisms that likely involve a HuR-dependent rise in MMP-9 mRNA stability.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Antigens, Surface , Glomerular Mesangium/cytology , Interleukin-1/pharmacology , Matrix Metalloproteinase 9/biosynthesis , RNA-Binding Proteins/metabolism , Animals , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation/drug effects , Matrix Metalloproteinase 9/genetics , Protein Transport , RNA Stability/drug effects , RNA, Messenger/drug effects , Rats , Ribonucleoproteins/metabolism , Transfection
18.
Cancer Res ; 63(14): 4062-6, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12874007

ABSTRACT

The progression of malignant melanoma is characterized by overexpression of a number of matrix metalloproteinases (MMPs), especially MMP-2, which play a critical role in the degradation of basement membranes and the extracellular matrix. Consequently, we assessed a drug targeting strategy in which the protease activity of MMP-2 is exploited to release an anticancer agent from a macromolecular carrier, i.e., circulating albumin. For this purpose, a water-soluble maleimide derivative of doxorubicin (1) incorporating a MMP-2 specific peptide sequence (Gly-Pro-Leu-Gly-Ile-Ala-Gly-Gln) was developed that binds rapidly and selectively to the cysteine-34 position of circulating albumin. The albumin-bound form of 1 was efficiently and specifically cleaved by MMP-2 liberating a doxorubicin tetrapeptide (Ile-Ala-Gly-Gln-DOXO) and subsequently doxorubicin. In vivo, 1 was superior to the parent compound doxorubicin in the A375 human melanoma xenograft, which is characterized by a high expression of MMP-2.


Subject(s)
Albumins/pharmacokinetics , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/analogs & derivatives , Matrix Metalloproteinase 2/metabolism , Melanoma/drug therapy , Melanoma/enzymology , Oligopeptides/pharmacokinetics , Prodrugs/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Humans , Maleimides/pharmacokinetics , Tumor Cells, Cultured
19.
Mol Cell Biol ; 23(14): 4901-16, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832476

ABSTRACT

Dysregulation of extracellular matrix turnover is an important feature of many inflammatory processes. Rat renal mesangial cells express high levels of matrix metalloproteinase 9 (MMP-9) in response to inflammatory cytokines such as interleukin-1 beta. We demonstrate that NO does strongly destabilize MMP-9 mRNA, since different luciferase reporter gene constructs containing the MMP-9 3' untranslated region (UTR) displayed significant reduced luciferase activity in response to the presence of NO. Moreover, by use of an in vitro degradation assay we found that the cytoplasmic fractions of NO-treated cells contained a higher capacity to degrade MMP-9 transcripts than those obtained from control cells. An RNA electrophoretic mobility shift assay demonstrated that three of four putative AU-rich elements present in the 3' UTR of MMP-9 were constitutively occupied by the mRNA-stabilizing factor HuR and that the RNA binding was strongly attenuated by the presence of NO. The addition of recombinant glutathione transferase-HuR prevented the rapid decay of MMP-9 mRNA, whereas the addition of a neutralizing anti-HuR antibody caused an acceleration of MMP-9 mRNA degradation. Furthermore, the expression of HuR mRNA and protein was significantly reduced by exogenously and endogenously produced NO. These inhibitory effects were mimicked by the cGMP analog 8-bromo-cGMP and reversed by LY-83583, an inhibitor of soluble guanylyl cyclase. These results demonstrate that NO acts in a cGMP-dependent mechanism to inhibit the expression level of HuR, thereby reducing the stability of MMP-9 mRNA.


Subject(s)
Antigens, Surface , Cyclic GMP/analogs & derivatives , Matrix Metalloproteinase 9/genetics , Nitric Oxide/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Aminoquinolines/pharmacology , Animals , Base Sequence , Cells, Cultured , Cyclic GMP/pharmacology , Cytoplasm/genetics , Cytoplasm/metabolism , Dactinomycin/pharmacology , ELAV Proteins , ELAV-Like Protein 1 , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Interleukin-1/pharmacology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Matrix Metalloproteinase 9/metabolism , Molecular Mimicry , Molecular Sequence Data , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , RNA Stability/drug effects , RNA-Binding Proteins/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repetitive Sequences, Nucleic Acid , Ribonucleoproteins/metabolism
20.
Toxicology ; 187(2-3): 171-81, 2003 May 03.
Article in English | MEDLINE | ID: mdl-12699906

ABSTRACT

In the current study, we have investigated the bioeffects of repeated exposure to low-frequency (50 Hz) high-intensity (20 mT; 200 G) electromagnetic field (EMF) on some immune parameters in mice. The animals were exposed to EMF daily for 30 min three times per week for 2 weeks. We also studied the possible immunomodulatory effects of two anti-radical substances known to have non-specific immunostimulant effects namely, L-carnitine (200 mg/kg body weight i.p.) and Q10 (200 mg/kg body weight, p.o.). Both drugs were given 1 h prior to each EMF exposure. Immune endpoints included total body weight, spleen/body weight ratio, splenocytes viability, total and differential white blood cell (WBCs; lymphocytes, monocytes, neutrophils) counts, as well as the lymphocyte proliferation induced by the mitogens; phytohaemagglutinin (PHA), concanavalin-A (Con-A) and lipoploysaccharide (LPS). Magnetic field decreased splenocyte viability, WBCs count, as well as mitogens-induced lymphocyte proliferation. L-carnitine, but not Q10 could ameliorate the adverse effects of EMF on the vast majority of the immune parameters tested, suggesting a possible immunoprotective role of L-carnitine under the current experimental conditions.


Subject(s)
Antioxidants/therapeutic use , Carnitine/therapeutic use , Electromagnetic Fields/adverse effects , Radiation-Protective Agents/therapeutic use , Spleen/pathology , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use , Animals , Body Weight/radiation effects , Cell Survival/radiation effects , Coenzymes , Concanavalin A/pharmacology , Lipopolysaccharides/pharmacology , Lymphocytes/drug effects , Male , Mice , Mitogens/pharmacology , Monocytes/pathology , Monocytes/radiation effects , Neutrophils , Phytohemagglutinins/pharmacology , Spleen/radiation effects , Stimulation, Chemical , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...