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










Publication year range
1.
J Egypt Natl Canc Inst ; 33(1): 22, 2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34423383

ABSTRACT

BACKGROUND: Glufosfamide (ß-D-glucosylisophosphoramide mustard, GLU) is an alkylating cytotoxic agent in which ifosforamide mustard (IPM) is glycosidically linked to the ß-D-glucose molecule. GLU exerted its cytotoxic effect as a targeted chemotherapy. Although, its cytotoxic efficacy in a number of cell lines, there were no experimental or clinical data available on the oncolytic effect of oxazaphosphorine drugs in hepatocellular carcinoma. Therefore, the main objective of the current study is to assess the cytotoxic potential of GLU for the first time in the hepatocellular carcinoma HepG2 cell line model. METHODS: Cytotoxicity was assayed by the MTT method, and half-maximal inhibitory concentration (IC50) was calculated. Flow cytometric analysis of apoptosis frequencies was measured by using Annexin V/PI double stain, an immunocytochemical assay of caspase-9, visualization of caspase-3, and Bcl2 gene expression were undertaken as apoptotic markers. Mitochondrial membrane potential was measured using the potentiometric dye; JC-1, as a clue for early apoptosis as well as ATP production, was measured by the luciferase-chemiluminescence assay. RESULTS: Glufosfamide induced cytotoxicity in HepG2 cells in a concentration- and time-dependent manner. The IC50 values for glufosfamide were significantly lower compared to ifosfamide. The frequency of apoptosis was much higher for glufosfamide than that of ifosfamide. The contents of caspase-9 and caspase-3 were elevated following exposure to GLU more than IFO. The anti-apoptotic Bcl2 gene expression, the mitochondrial membrane potential, and the cellular ATP levels were significantly decreased than in case of ifosfamide. CONCLUSIONS: The current study reported for the first time cytotoxicity activity of glufosfamide in HepG2 cells in vitro. The obtained results confirmed the higher oncolytic activity of glufosfamide than its aglycone ifosfamide. The generated data warrants further elucidations by in vivo study.


Subject(s)
Ifosfamide , Liver Neoplasms , Glucose/analogs & derivatives , Hep G2 Cells , Humans , Ifosfamide/analogs & derivatives , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics
2.
Toxicology ; 457: 152808, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33965443

ABSTRACT

We have addressed in the current study the potential of L-carnitine (LC) to extenuate the reproductive toxic insults of carbendazim (CBZ) in male rats, and the molecular mechanisms whereby carnitine would modify the spermatogenic and steroidogenic derangements invoked by the endocrine disruptor. Herein, animals received daily doses of carbendazim (100 mg/kg) by gavage for 8 weeks. Another CBZ-challenged group was co-supplemented with LC (500 mg/kg, IP) twice weekly for 8 weeks. Sperm quantity and quality (morphology, motility and viability), serum testosterone and gonadotropins, and thyroid hormone levels were assessed. Serum tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6) and interleukin-10 (IL-10) concentrations were determined by ELISA. Oxidant/antioxidant status in rat testis was investigated via measuring testicular contents of malondialdehyde (MDA) and reduced glutathione (GSH), as well as the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx). Immunohistochemical localizations of the junctional protein; occludin, and inflammatory markers; inducible nitric oxide synthase (iNOS) and nuclear factor kappa beta (NF-κB) were further analyzed. A host of transduction genes that regulate spermatogenic and steroidogenic pathways, and their encoded proteins namely, Steroidogenic Acute Regulatory Protein (StAR), Fatty acid binding protein 9 (FABP9) and P38-mitogen activated protein kinase (P38-MAPK) were assessed by real time quantitative (RT-qPCR) and Western blot. LC improved rat spermiogram, testicular histological alterations and endocrine perturbances, and modulated genes' expressions and their respective proteins. In conclusion, LC effects appear to reside for the most part on its endocrine-preserving, anti-oxidant and anti-inflammatory properties through a myriad of interlaced signal transductions that ultimately recapitulated its beneficial effects on spermatogenesis and steroidogenesis.


Subject(s)
Benzimidazoles/toxicity , Carbamates/toxicity , Carnitine/pharmacology , Fatty Acid-Binding Proteins/biosynthesis , Oxidative Stress/physiology , Phosphoproteins/biosynthesis , Testis/metabolism , Animals , Down-Regulation/drug effects , Down-Regulation/physiology , Endocrine Disruptors/toxicity , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , Random Allocation , Rats , Sperm Count/methods , Testis/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
3.
PeerJ ; 4: e2168, 2016.
Article in English | MEDLINE | ID: mdl-27413637

ABSTRACT

Background. Glufosfamide (GLU) is a glucose conjugate of ifosfamide in which isophosphoramide mustard is glycosidically linked to the ß-D-glucose molecule. Based on GLU structure, it is considered a targeted chemotherapy with fewer side effects. The main objective of the current study is to assess the cytotoxic potential of GLU for the first time in prostate cancer (PC) cells representing different stages of the tumor. Furthermore, this study examined the potential synergistic activity of GLU in combination with docetaxel (DOC). Methods. Two different cell lines were used, LNCaP and PC-3. Concentration-response curves were assessed. The tested groups per cell line were, control, GLU, DOC and combination. Treatment duration was 72 h. Cytotoxicity was assessed using sulforhodamine B (SRB) assay and half maximal inhibitory concentration (IC50) was calculated. Synergy analyses were performed using Calcusyn(®)software. Subsequent mechanistic studies included ß-glucosidase activity assay, glucose uptake and apoptosis studies, namely annexin V-FITC assay and the protein expression of mitochondrial pathway signals including Bcl-2, Bax, Caspase 9 and 3 were assessed. Data are presented as mean ± SD; comparisons were carried out using one way analysis of variance (ANOVA) followed by Tukey-Kramer's test for post hoc analysis. Results. GLU induced cytotoxicity in both cell lines in a concentration-dependent manner. The IC50 in PC-3 cells was significantly lower by 19% when compared to that of LNCaP cells. The IC50 of combining both drugs showed comparable effect to DOC in PC-3 but was tremendously lowered by 49% compared to the same group in LNCaP cell line. ß-glucosidase activity was higher in LNCaP by about 67% compared to that determined in PC-3 cells while the glucose uptake in PC-3 cells was almost 2 folds that found in LNCaP cells. These results were directly correlated to the efficacy of GLU in each cell line. Treatment of PC cells with GLU as single agent or in combination with DOC induced significantly higher apoptosis as evidenced by Annexin V-staining. Apoptosis was significantly increased in combination group by 4.9 folds and by 2.1 Folds when compared to control in LNCaP cells and PC-3 cells; respectively. Similarly, the expression of Bcl-2 was significantly decreased while Bax, caspase 9 and 3 were significantly increased in the combined treatment groups compared to the control. Conclusion. GLU has a synergistic effect in combination with DOC as it increases the cell kill which can be attributed at least partially to apoptosis in both the tested cell lines and it is suggested as a new combination regimen to be considered in the treatment of the prostate cancer. Further experiments and clinical investigations are needed for assessment of that regimen.

4.
Invest New Drugs ; 28(3): 306-17, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19415182

ABSTRACT

Glycoconjugates represent a recent trend in cancer chemotherapy that adopts the concept of selective prodrug/drug targeting of tumor cells by selectively binding to specific transmembrane glucose transporters. Following preferential uptake of sugar conjugates into cancer cells, they are presumably subject to enzymatic cleavage by specific beta-glycosidases to liberate the free active cytotoxic aglycones that act selectively on cancer cells and spare other noncancerous ones. In this sense, the cytotoxicity of an array of newly synthesized glycoconjugates, including curcumin beta-glucoside, perillyl alcohol beta-glucoside, perillyl alcohol beta-galactoside, diethylstilbesterol beta-glucoside and diethylstilbesterol beta-galactoside have been investigated over 24-96 h in a panel of human colon cancer cells namely, Caco-2, HT29 and T84 cells. The role of beta-glycosidases and caspases in the bioactivation and cytotoxicity of these compounds has been addressed in the current study. All the glycoconjugates have proven cytotoxic efficacy in a time-dependent manner. Curcumin beta-glucoside was the most potent amongst all glycoconjugates tested. The sensitivity rank order of tumor cells towards all beta-glucosides was Caco-2 > HT29 > T84. This sensitivity ranking was well correlated with beta-glucosidase activity assessed in these cell lines. Unlike perillyl alcohol galactoside, the cytotoxicity rank order for diethylstilbesterol beta-galactoside was not coping with the beta-galactosidase activity detected. Apoptosis was assessed by fluorometric assay of caspase-3 and caspase-9 activities. Initiation and activation of apoptosis were increased in all colon cancer cells following exposure to most of the glycoconjugates, and this was well correlated with the cytotoxicity rank order of these prodrugs. Enzymatic cleavage of glycoconjugates was accomplished using a host of hydrolytic enzymes and cleavage kinetics was determined using HPLC. The glycoconjugates were only cleaved by beta-glucosidases and beta-galactosidases, but not by pancreatic lipase or hepatic esterase. Taken together, one could conclude that beta-glucosidases and beta-galactosidases are crucial for the bioactivation and cytotoxicity of these glycoconjugates. Also, initiation and activation of apoptosis in tumor cells may contribute, at least partly, for the cytotoxicity of these sugar conjugates.


Subject(s)
Colonic Neoplasms/drug therapy , Drug Delivery Systems/methods , Glycoconjugates/therapeutic use , Prodrugs/therapeutic use , beta-Galactosidase/metabolism , beta-Glucosidase/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival , Colonic Neoplasms/metabolism , Drug Screening Assays, Antitumor , Esterases/metabolism , Glycoconjugates/pharmacology , Humans , Kinetics , Lipase , Molecular Structure , Prodrugs/pharmacology
5.
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
6.
Eur J Pharmacol ; 616(1-3): 58-63, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19545561

ABSTRACT

Glycoconjugates represent a recent trend in cancer chemotherapy that adopts the concept of selective prodrug/drug targeting of tumor cells by binding to specific transmembrane glucose transporters. Following preferential uptake of sugar conjugates into cancer cells, they are presumably subject to enzymatic cleavage by specific beta-glycosidases to liberate the free active cytotoxic aglycones that act selectively on cancer cells and spare other noncancerous ones. In this sense, the role of beta-glucosidase and caspases in the bioactivation and cytotoxicity of glufosfamide has been addressed in the current study. The cytotoxicity of glufosfamide has been investigated over 24-96 h in a panel of human colon cancer cells namely, Caco-2, HT29 and T84 using a tetrazole dye; 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; MTT assay technique. Apoptosis was assessed by fluorometric assay of caspase-3 and caspase-9 activities. Enzymatic cleavage of glufosfamide was accomplished using a host of hydrolytic enzymes and cleavage kinetics was determined using HPLC. Glufosfamide has proven cytotoxic efficacy in a concentration- and time-dependent manner. The sensitivity rank order of tumor cells towards the glycoconjugate was Caco-2>HT29>T84. This sensitivity ranking was well correlated with the enzymatic activity of beta-glucosidase assessed in these cell lines. Initiation and activation of apoptosis were increased in all colon cancer cells following exposure to glufosfamide and were well correlated with the cytotoxicity rank order of the glycoconjugate. Glufosfamide was cleaved by cytosolic and lysosomal beta-glucosidases but not by other hydrolytic enzymes such as cytosolic beta-galactosidase, pancreatic lipase or hepatic esterase. In conclusion, the current data could possibly unravel the mechanistic role of beta-glucosidase and apoptotic caspases in the bioactivation and cytotoxicity of glufosfamide within colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Caspases/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Phosphoramide Mustards/pharmacology , beta-Glucosidase/metabolism , Animals , Antineoplastic Agents/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Female , Glucose/analogs & derivatives , Humans , Ifosfamide/analogs & derivatives , Male , Phosphoramide Mustards/metabolism
7.
Basic Clin Pharmacol Toxicol ; 104(5): 393-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19413659

ABSTRACT

The possible uroprotective effects of curcumin have been addressed in the current study. Haemorrhagic cystitis was induced by challenging male Swiss albino rats with a single dose of cyclophosphamide (150 mg/kg, i.p.). Curcumin (200 mg/kg, i.p.) was administered for 10 consecutive days followed by a single dose of cyclophosphamide. Haemorrhagic cystitis was well characterized morphologically and biochemically. The hallmark of this toxicity was marked congestion, oedema and extravasation in rat urinary bladder, as well as a marked desquamative damage to the urothelium and severe inflammation in the lamina propria. Leucocytic infiltration was also observed and determined by histopathological examination. Serum level of tumour necrosis factor-alpha was notably elevated associated with apparent hypokalaemia and hyponatraemia. Bladder contents of adenosine triphosphate, reduced glutathione and glutathione-S-transferase activity were markedly reduced. Malondialdehyde level, myeloperoxidase activity and urinary nitrite-nitrate levels, expressed as nitric oxide, were dramatically increased. Prior administration of curcumin ahead of cyclophosphamide challenge improved all the biochemical and histologic alterations induced by the cytotoxic drug. Based on these broad findings, it could be concluded that curcumin has proven uroprotective efficacy in this cyclophosphamide haemorrhagic cystitis model, possibly through modulating the release of inflammatory endocoids, namely tumour necrosis factor-alpha and nitric oxide, improving the energy status and restoring the oxidant/antioxidant balance.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Curcumin/therapeutic use , Cyclophosphamide/adverse effects , Cystitis/prevention & control , Hemorrhage/prevention & control , Urothelium/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Curcumin/administration & dosage , Cystitis/chemically induced , Cystitis/enzymology , Cystitis/pathology , Hemorrhage/chemically induced , Hemorrhage/enzymology , Hemorrhage/pathology , Male , Mice , Nitric Oxide/urine , Potassium/blood , Sodium/blood , Tumor Necrosis Factor-alpha/blood , Urinary Bladder/drug effects , Urinary Bladder/enzymology , Urinary Bladder/pathology , Urothelium/enzymology , Urothelium/pathology
8.
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
9.
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
10.
Arch Toxicol ; 83(2): 139-50, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18597074

ABSTRACT

We have addressed in the current study the postulate whether or not carnitine deficiency would represent a risk factor in hepatotoxicity. Carnitine-deficient male Swiss albino rats were obtained following administration of D-carnitine (500 mg/kg, IP) for 10 consecutive days. Serum and liver carnitine levels, both total and free, were assessed to confirm carnitine depletion. Hepatotoxicity was induced by challenging animals with a single dose of paracetamol (1 g/kg, IP). Serum tumor necrosis factor (TNF-alpha) concentration, and serum activities of aspartate amino transferase (AST), alanine amino transferase (ALT) and alkaline phosphatase (ALP) were undertaken as biomarkers for toxicity. Liver contents of reduced glutathione (GSH), malondialdehyde (MDA), total nitric oxide (NO) and myeloperoxidase (MPO) activities were also investigated. Histopathological examination of liver sections was achieved to confirm the biochemical alterations. D-carnitine altered all biochemical markers and also induced mild tissue inflammation with dilatation and congestion of central and portal veins. Paracetamol produced an obvious hepatotoxicity model that was well characterized biochemically and morphologically. Combined administration of D-carnitine and paracetamol synergistically provoked marked toxicity that was more profound than either agent given alone. The present work was further extended to elucidate any hepatoprotective effect of carnitine supplementation in such toxicity paradigm. It was apparent that L-carnitine notably ameliorated all biochemical markers and also mitigated the gross histologic alterations induced by paracetamol. Data obtained so far would suggest that carnitine deficiency could possibly be a sequela as well as a causative clue for paracetamol hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Carnitine/deficiency , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Acetaminophen/metabolism , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Drug Synergism , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Peroxidase/metabolism , Rats , Rats, Inbred Strains , Risk Factors , Tumor Necrosis Factor-alpha/blood
11.
Toxicology ; 254(1-2): 51-60, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18852009

ABSTRACT

We have recently shown that carnitine deficiency could represent a risk factor in paracetamol hepatotoxicity. By the same token, d-carnitine-induced carnitine deficiency aggravated carboplatin nephropathy following challenge with a single dose (35mg/kg, IP) of the platinum drug in male Swiss albino rats. The combination modality induced marked degenerative changes and severe inflammation in kidney tissues that surpassed either carboplatin or d-carnitine given alone. The combined regimen synergistically increased the serum levels of creatinine, blood urea nitrogen (BUN), tumor necrosis factor alpha (TNF-alpha), palmitate, and kidney malondialdehyde (MDA), adenosine triphosphate (ATP), nitric oxide (NO) contents as well as kidney myeloperoxidase (MPO) activity. The only parameter that has been notably decreased was the kidney reduced glutathione (GSH) level. Exaggeration by carnitine deficit of the deleterious effects of carboplatin is most probably ascribed to energy starvation. The reduction in kidney content of ATP parcels was associated with elevation of serum palmitate level that reflected debilitated fatty acid oxidation, and this further deteriorated energy resources in kidney tissues. Compromising the oxidant/anti-oxidant balance and modulating the release of some inflammatory endocoids namely, TNF-alpha and NO could also possibly account for such combinatorial detrimental toxicity. The current study was further extended to elucidate any possible nephroprotective effects of l-carnitine. Interestingly, carnitine supplementation ahead of carboplatin challenge ameliorated and almost normalized all the biochemical parameters and also mitigated the injurious effects of the cytotoxic drug. Thus, one could conclude that carnitine deficiency, whether being a causative clue or a sequela, might represent a risk factor in carboplatin nephropathy.


Subject(s)
Carboplatin/toxicity , Carnitine/deficiency , Kidney Diseases/chemically induced , Oxidants/metabolism , Adenosine Triphosphate/metabolism , Animals , Antioxidants/metabolism , Blood Urea Nitrogen , Creatinine/blood , Inflammation/metabolism , Kidney/metabolism , Male , Nitric Oxide/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
12.
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
13.
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
14.
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
15.
Toxicology ; 222(1-2): 103-13, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16567030

ABSTRACT

We have investigated in the current study, the possible modulatory effects of dexamethasone on cisplatin cytotoxicity in Ehrlich ascites carcinoma (EAC)-bearing female Swiss albino mice. Cisplatin (3.5mg/kg) was injected IP for 3 consecutive days in mice previously inoculated SC with EAC cells in the right flank. Dexamethasone (2.5mg/kg) was administered SC alone or 24h ahead of cisplatin challenge, and these regimens were given for 3 consecutive days. Dexamethasone enhanced the anti-tumor effects of cisplatin, clearly demonstrated by the increased mean tumor growth time (TGT) and tumor growth delay time (TGDT) values compared to cisplatin alone. The effects of dexamethasone on tumor angiogenesis and cell cycle distribution of EAC cells have been addressed as possible mechanisms, whereby the glucocorticoid could probably augment cisplatin cell-kill. Indeed, dexamethasone enhanced the angiostatic activity of cisplatin by 52.5%. The glucocorticoid also synchronized the EAC cells in the G2/M phase, secondary to its regulatory role on the transcriptional and translational activity in these cells, thus, exposing them to the dramatic cytotoxic potential of cisplatin. One could conclude that dexamethasone enhanced the anti-tumor effects of cisplatin via augmenting its angiostatic activity and modulating cell cycle kinetics. Also, dexamethasone did not alter cisplatin-induced nephrotoxicity, thus demonstrating an improved therapeutic potential.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Dexamethasone/pharmacology , Kidney/drug effects , Neovascularization, Pathologic/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols , Blood Urea Nitrogen , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/toxicity , Creatinine/blood , Dexamethasone/administration & dosage , Female , Glutathione/metabolism , Kidney/metabolism , Kidney/pathology , Malondialdehyde/metabolism , Mice , Tumor Burden/drug effects
16.
Pharmacol Biochem Behav ; 82(3): 574-82, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16337998

ABSTRACT

An experimental animal model of Huntington's disease (HD) phenotype was induced using the mycotoxin 3-nitropropionic acid (3-NP) and was well characterized behaviorally, neurochemically, morphometrically and histologically. Administration of 3-NP caused a reduction in prepulse inhibition (PPI) of acoustic startle response, locomotor hyper- and/or hypoactivity, bilateral striatal lesions, brain oxidative stress, and decreased striatal gamma-aminobutyric acid (GABA) levels. Taurine is a semi-essential beta-amino acid that was demonstrated to have both antioxidant and GABA-A agonistic activity. In this study, treatment with taurine (200 mg/kg daily for 3 days) prior to 3-NP administration reversed both reduced PPI response and locomotor hypoactivity caused by 3-NP injection. Taurine pretreatment also caused about 2-fold increase in GABA concentration compared to 3-NP-treated animals. In addition, taurine demonstrated antioxidant activity against oxidative stress induced by 3-NP administration as evidenced by the reduced striatal malondialdehyde (MDA) and elevated striatal glutathione (GSH) levels. Histochemical examination of striatal tissue showed that prior administration of taurine ahead of 3-NP challenge significantly increased succinate dehydrogenase (SDH) activity compared to 3-NP-treated animals. Histopathological examination further affirmed the neuroprotective effect of taurine in 3-NP-induced HD in rats. Taken together, one may conclude that taurine has neuroprotective role in the current HD paradigm due, at least partly, to its indirect antioxidant effect and GABA agonistic action.


Subject(s)
Disease Models, Animal , Huntington Disease/chemically induced , Neuroprotective Agents/pharmacology , Nitro Compounds/toxicity , Propionates/toxicity , Taurine/pharmacology , Animals , Corpus Striatum/metabolism , Male , Motor Activity , Oxidative Stress , Phenotype , Rats , Rats, Wistar , Reflex, Startle/drug effects , gamma-Aminobutyric Acid/metabolism
17.
Med Sci Monit ; 11(7): BR228-234, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15990684

ABSTRACT

BACKGROUND: Curcumin (a component of turmeric) has long been used as a spice and food-coloring agent. In experimental animals, curcumin has shown anti-diabetic, anti-inflammatory, cytotoxic and anti-oxidant properties. MATERIAL/METHODS: The possible hypolipidemic effect of curcumin was investigated in rats fed a high-cholesterol diet (HCD). The lipid profile and activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were assessed in serum, as well as anti-oxidant parameters in liver tissues. RESULTS: Feeding the animals a high cholesterol diet (HCD) for 7 consecutive days (1 ml 100 g(-1)) resulted in marked hypercholesterolemia, increased serum level of low-density lipoprotein cholesterol (LDL-C), but a decreased serum high-density lipoprotein cholesterol (HDL-C). Curcumin admixed with the diet (0.5% w/w) decreased serum total cholesterol (TC) by about 21% and LDL-C by 42.5%, but it increased serum HDL by 50%. The atherogenic indices (LDL-C/HDL-C and TC/HDL-C) were reduced by 52% and 35%, respectively. Curcumin also decreased the enzyme activities of serum AST and ALT, which were increased in HCD animals. CONCLUSIONS: Curcumin showed an obvious hypocholesterolemic effect that could be due to an effect on cholesterol absorption, degradation or elimination, but not due to an anti-oxidant mechanism. This could be supported by the finding in our study that neither HCD nor curcumin-admixed HCD had any effects on the liver content of glutathione (GSH) or superoxide dismutase (SOD) activity. Thus one could argue that ingestion of curcumin-containing spices in the diet, especially one rich in fats, could have a lipid-lowering effect.


Subject(s)
Cholesterol/blood , Curcumin/therapeutic use , Hypercholesterolemia/drug therapy , Hypolipidemic Agents/therapeutic use , Alanine Transaminase/antagonists & inhibitors , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Cholesterol, Dietary/adverse effects , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Hypercholesterolemia/etiology , Liver/drug effects , Liver/enzymology , Male , Rats
18.
Pharmacol Res ; 48(3): 285-90, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12860448

ABSTRACT

We have investigated in the current study the possible protective effects of two carnitine esters known to have powerful anti-oxidant potential namely, propionyl L-carnitine (PLC) and acetyl L-carnitine (AC) against alcohol-induced gastric lesions in rats. Both drugs were administered as a single oral dose of 200 mg kg(-1) body weight 1h before alcohol intake. Both carnitine esters could protect the gastric mucosa against the injurious effect of absolute alcohol and promote ulcer healing as evidenced from the ulcer index (UI) values. Propionyl L-carnitine prevented alcohol-induced increase in thiobarbituric acid-reactive substances (TBARS), an index of lipid peroxidation. The propionyl carnitine ester also increased the gastric content of reduced glutathione (GSH), besides it increased the enzymatic activities of gastric superoxide dismutase (SOD) and glutathione-S-transferase (GST). Likewise, AC did protect against the ulcerating effect of alcohol and mitigate most of the biochemical adverse effects induced by alcohol in gastric mucosa, but to a lesser extent than PLC. Neither PLC nor AC did affect catalase activity in gastric tissue. Based on these observations, one could conclude that carnitine esters, particularly PLC could partly protect gastric mucosa from alcohol-induced acute mucosal injury, and these gastroprotective effects might be probably induced, at least partly, through anti-oxidant mechanisms.


Subject(s)
Acetylcarnitine/therapeutic use , Carnitine/analogs & derivatives , Carnitine/therapeutic use , Ethanol/adverse effects , Stomach Ulcer/prevention & control , Acetylcarnitine/pharmacology , Administration, Oral , Animals , Carnitine/pharmacology , Catalase/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Male , Mice , Rats , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
19.
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
20.
Pharmacol Res ; 46(3): 287-93, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12220973

ABSTRACT

Dibromoacetonitrile (DBAN) is a disinfection by-product following chlorination of drinking water. Epidemiological studies indicate that it might present a potential hazard to human health. DBAN was previously found to induce oxidative stress in rat stomach as manifested by perturbation of some enzymatic and nonenzymatic antioxidant parameters. Therefore, we have investigated the oxidative stress possibly induced by DBAN in mouse stomach and possible protection by melatonin (MLT) as a free radical scavenger. In a dose-response study, mice were administered a single oral dose of DBAN (30, 60 and 120 mg kg(-1)) and were sacrificed after 1 h. DBAN significantly reduced glutathione (GSH) content that was somehow dose-related, and inhibited glutathione-S-transferase (GST) activity in gastric tissues. The highest dose of DBAN (120 mg kg(-1)) lowered GSH by 74% and induced a significant elevation of lipid peroxidation products, determined as thiobarbituric acid reactive substances (TBARS) by 69%. The same dose inhibited the gastric activities of GST, superoxide dismutase (SOD) and catalase (CAT) by 70, 57 and 23%, respectively. In a time-course study, mice were administered DBAN (60 mg kg(-1) p.o.) and sacrificed 0.5, 1, 3, 6, 12 and 24 h after treatment. GSH was dramatically depleted at 0.5, 1, 3 and 6 h (45, 38, 39 and 49% of control, respectively) and remained significantly low at 12 and 24 h. Also, DBAN caused an accumulation of TBARS in gastric tissues starting from 3 h and was maximum at 6 h (133% of the control). The enzymatic activities of GST and SOD were maximally inhibited by DBAN treatment at 0.5 h (32% for GST and 37% for SOD of the respective control). The activities of both enzymes returned to control values at 24 h. CAT activity was not affected by DBAN administration at all. Pretreatment of another group of mice with melatonin (10 mg kg(-1) per day p.o. 12 days) before administration of DBAN (60 mg kg(-1) p.o.) completely mitigated the aforementioned parameters. In conclusion, the present study indicates that DBAN induces a marked oxidative stress in mouse stomach as evidenced by GSH depletion, TBARS accumulation and GST, SOD and CAT inhibition. Melatonin could mitigate DBAN-induced oxidative stress in mouse stomach as it did almost normalize both the enzymatic and nonenzymatic antioxidant parameters.


Subject(s)
Acetonitriles/administration & dosage , Antioxidants/pharmacology , Melatonin/pharmacology , Oxidative Stress/drug effects , Stomach/drug effects , Acetonitriles/toxicity , Administration, Oral , Animals , Catalase/drug effects , Catalase/metabolism , Gastric Mucosa/metabolism , Glutathione/drug effects , Glutathione/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Male , Mice , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...