Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(16)2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37627094

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most aggressive malignancies, with continuously increasing cases and fatalities. Diagnosis often occurs in the advanced stages, confining patients to systemic therapies such as sorafenib. Sorafenib (SB), a multi-kinase inhibitor, has not yet demonstrated sufficient efficacy against advanced HCC. There is a strong argument in favor of studying its use in combination with other medications to optimize the therapeutic results. According to our earlier work, crocin (CR), a key bioactive component of saffron, hinders HCC development and liver cancer stemness. In this study, we investigated the therapeutic use of CR or its combination with SB in a cirrhotic rat model of HCC and evaluated how effectively SB and CR inhibited tumor growth in this model. Diethylnitrosamine (DEN) was administered intraperitoneally to rats once a week for 15 weeks, leading to cirrhosis, and then 19 weeks later, leading to multifocal HCC. After 16 weeks of cancer induction, CR (200 mg/kg daily) and SB (10 mg/kg daily) were given orally to rats for three weeks, either separately or in combination. Consistently, the combination treatment considerably decreased the incidence of dyschromatic nodules, nodule multiplicity, and dysplastic nodules when compared to the HCC group of single therapies. Combined therapy also caused the highest degree of apoptosis, along with decreased proliferating and ß-catenin levels in the tumor tissues. Additionally, when rats received combined therapy with CR, it showed anti-inflammatory characteristics where nuclear factor kappa B (NF-κB) and cyclooxygenase-2 (Cox-2) were considerably and additively lowered. As a result, CR potentiates the suppressive effects of SB on tumor growth and provides the opportunity to strengthen the therapeutic effects of SB in the treatment of HCC.

2.
Biomed Pharmacother ; 165: 115148, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37450997

ABSTRACT

Cisplatin (CIS) is a broad-spectrum anti-carcinogen that causes cytotoxic effects both in normal and cancer cells. The purpose of this study was to test whether Hibiscus sabdariffa (HS) extract can reduce CIS-induced hepatotoxicity in rodents and to assess its anticancer activity in vitro. Treatment with HS extract at daily doses of 500 mg/kg before and after a single dose of CIS (10 mg/kg) reduced hepatotoxicity in Wistar male albino rats. HS extract reduced activity of hepatic damage marker enzymes ( i.e. alanine and aspartate aminotransferases), necrosis, and apoptosis in liver tissues of CIS-treated rats. This hepatic protection was associated with reduced oxidative stress in liver tissues. The antioxidant effects of HS were manifested as a normalization of malondialdehyde levels and glutathione levels which were all raised after CIS-induction. In addition, HS treatment resulted in a decrease of catalase, and superoxide dismutase activity. The combined effects of CIS and HS were also studied in two human lung cancer cell lines (A549 and H460). Treatment with HS (20 µg /mL) enhanced the cytotoxic activity of CIS both in A549 and H460 cell lines. Interestingly, HS increased CIS-induced apoptosis and oxidative stress more clearly in A549 cells indicating that HS extract in combination with CIS could increase the efficacy of CIS in the treatment of cancer.


Subject(s)
Antineoplastic Agents , Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Hepatitis , Hibiscus , Lung Neoplasms , Humans , Rats , Male , Animals , Cisplatin/pharmacology , Rats, Wistar , Antioxidants/pharmacology , Antioxidants/metabolism , Antineoplastic Agents/toxicity , Antineoplastic Agents/metabolism , Oxidative Stress , Liver , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Hepatitis/metabolism , Apoptosis , Plant Extracts/pharmacology , Plant Extracts/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Chemical and Drug Induced Liver Injury/metabolism
3.
Int J Nanomedicine ; 17: 6095-6112, 2022.
Article in English | MEDLINE | ID: mdl-36514376

ABSTRACT

Introduction: Metal nanoparticle synthesis using plant has emerged as an eco-friendly, clean, and viable strategy alternative to chemical and physical approaches. Methods: The fruit extract of Salvadora persica (SP) was utilized as a reducing and stabilizing agent in the synthesis of gold (AuNPs) and copper (CuNPs) nanoparticles. Results: UV-Vis spectra of the AuNPs and CuNPs showed peaks at the wavelengths of 530 nm and 440 nm, respectively. Transmission electron microscopy showed that nanoparticles exhibited a mainly spherical form, with a distribution range of 100 to 113 nm in diameter for AuNPs and of 130 to 135 nm in diameter for CuNPs. While energy-dispersive X-ray spectroscopy was able to confirm the existence of AuNPs and CuNPs. The alcoholic extract of the fruit SP was analyzed by GC-MS in order to identify whether or not it contained any active phytochemicals. Fourier-transform infrared spectra confirmed the presence capping functional biomolecules of SP on the surface of nanoparticles that acts as stabilizers. Analysis of the zeta potential revealed that NPs with high degree of stability, as demonstrated by a strong negative potential value in the range of 25.2 to 28.7 mV. Results showed that both green AuNPs and CuNPs have potential antimicrobial activity against human pathogens such gram-negative bacteria and gram-positive bacteria, with CuNPs having antimicrobial activity higher than AuNPs. In addition, AuNPs and CuNPs have promising antioxidant and anticancer properties when applied to MCF-7 and MDA-MB-231 breast cancer cells. Studies of molecular docking of SP bioactive compounds were conducted against methenyl tetrahydrofolate synthetase. Among all of them, Beta - Sitosterol was the most prominent. Conclusion: These AuNPs and CuNPs are particularly appealing in a variety of applications in the pharmaceutical and medicinal industries due to their economical and environmentally friendly production.


Subject(s)
Metal Nanoparticles , Salvadoraceae , Humans , Gold/pharmacology , Gold/chemistry , Metal Nanoparticles/chemistry , Copper/pharmacology , Copper/chemistry , Fruit , Molecular Docking Simulation , Microbial Sensitivity Tests , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Bacterial Agents/chemistry , Green Chemistry Technology , Spectroscopy, Fourier Transform Infrared
4.
Biomed Pharmacother ; 147: 112666, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35124384

ABSTRACT

Naturally occurring phytochemicals especially polyphenolic compounds have received increasing attention as chemopreventive agents. The chemopreventive potential of the ethanolic extract of Salvadora persica L. fruits SP, (the arak tree or miswak) on 7,12-dimethylbenz (a) anthracene (DMBA)-induced mammary carcinogenesis in female albino rats was investigated in this work. Ethanolic extract of SP fruits was supplemented to the experimental groups at a concentration of 500 mg/kg body weight for 22 weeks. Administration of SP extract suppressed DMBA-induced mammary carcinogenesis as revealed by incidence of tumors in histological investigation. There was a significant reduction in cell proliferation and an increase in apoptosis with downregulation of estrogen receptor expression in the mammary tissue of SP-treated animals. Additionally, SP extract prevented the oxidative damage induced in breast tissues of DMBA-treated rats. SP treatment also decreased the viability of MCF-7 breast cancer cells and induced early and late apoptosis and induced S cell cycle arrest. The chemo-preventive properties and anticancer effects of SP could be attributed to its anti-oxidative and a high percentage of phenolic compounds and esters which were detected here in the SP fruit extract.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Receptors, Estrogen/drug effects , Salvadoraceae , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , MCF-7 Cells , Random Allocation , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
5.
Biomed Pharmacother ; 134: 111102, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33338743

ABSTRACT

Ginger has been proposed as quite a promising candidate for cancer prevention. The purpose of this study was to assess the chemo-preventive effects of ginger. Furthermore, this study investigated the possible mechanisms of a standardized extract drawn from the rhizomes of ginger against diethylnitrosamine (DEN)-induced liver cancer in Wistar rats. The chemo-preventive effects of ginger at doses of 75 mg/kg, 150 mg/kg and 300 mg/kg per day were determined using a liver cancer model which was induced by DEN (Ali et al., 2008) and 2-acetylaminofluorene (2-AAF) in rats. Ginger attenuated carcinogenic changes after 22 weeks of cancer induction by decreasing the quantity and occurrences of hepatic dyschromatic nodules and positive focal areas as well as decreasing the amount of placental glutathione S-transferase (GST) in the livers of DEN/2-AAF-treated rats. Moreover, in rats, ginger counteracts DEN-influenced oxidative stress and decreases myeloperoxidase, malondialdehyde and protein carbonyl concentrations in the liver. This was determined by observing the restoration of superoxide dismutase, catalase, GST and glutathione. Immunohistochemical bleaching in rat livers showed that ginger prevented the increase in cell-positive numbers for Ki-67, cyclooxygenase-2 and nuclear factor kappa B p65. Ginger also inhibited the number of positive cells in DEN/2-AAF-treated rats for TUNEL, M30 and caspase-3 liver tissues. This research shows that ginger has an important chemo-preventative impact on liver cancer by inhibiting the growth of cells and inducing apoptosis. By reducing oxidative and inflammatory damage, ginger protects rat liver against cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Animals , Antioxidants/pharmacology , Catalase/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Inflammation/drug therapy , Liver/metabolism , Liver Neoplasms/prevention & control , Male , Malondialdehyde/metabolism , Rats , Rats, Wistar , Rhizome/chemistry , Superoxide Dismutase/metabolism
6.
Front Pharmacol ; 12: 777500, 2021.
Article in English | MEDLINE | ID: mdl-35177980

ABSTRACT

Despite all efforts, an effective and safe treatment for liver cancer remains elusive. Natural products and their derived biomolecules are potential resources to mine for novel anti-cancer drugs. Chemopreventive effects of safranal, a major bioactive ingredient of the golden spice "saffron", were evaluated in this study against diethylnitrosamine (DEN)-induced liver cancer in rats. Safranal's mechanisms of action were also investigated in the human liver cancer line "HepG2". When administered to DEN-treated rats, safranal significantly inhibited proliferation (Ki-67) and also induced apoptosis (TUNEL and M30 CytoDeath). It also exhibited anti-inflammatory properties where inflammatory markers such as NF-kB, COX2, iNOS, TNF-alpha, and its receptor were significantly inhibited. Safranal's in vivo effects were further supported in HepG2 cells where apoptosis was induced and inflammation was downregulated. In summary, safranal is reported here as a potent chemopreventive agent against hepatocellular carcinoma that may soon be an important ingredient of a broad-spectrum cancer therapy.

7.
Antioxidants (Basel) ; 9(12)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255507

ABSTRACT

Hawthorn (HAW) is a herbal preparation extracted from Crataegus oxyacantha. HAW has cardioprotective, antioxidants, anti-inflammatory, and anti-hypotensive effects. HAW's effect on hepatic fibrosis remains, however, unknown. This study evaluated the impact of HAW on carbon tetrachloride (CCl4)-induced hepatic fibrosis in rats and elucidated its mechanisms. HAW reduced liver index and the serum liver enzyme markers and reduced liver damage, and fibrosis as confirmed by histopathological scoring of hematoxylin-eosin staining. Collagen deposition was reduced in HAW group compared to CCl4 group as confirmed by Masson staining, hydroxyproline content, and both mRNA and protein levels of alpha-smooth muscle actin, collagen 1 and 3. HAW also down regulated the gene expressions of inflammatory markers including interleukin-IL-1ß, tumor necrosis factor-α, transforming growth factor-ß 1, nuclear factor kappa-B, and cyclooxygenase-2 and decreased the myeloperoxidase activity. The effects of HAW was also associated with decreased levels of hepatic oxidative stress markers (malondialdehyde and P.Carbonyl) and with increased activity of superoxide dismutase. Those effects are possibly mediated by blocking the pro-oxidant machinery and down regulating the inflammatory and profibrotic responses. Finally, chlorogenic acid, epicatechin, rutin, vitexin quercetin, and iso quercetin were identified as the major species of polyphenols of the HAW herbal preparation used here. Therefore, HAW's potent protecting effects against liver fibrosis predicts a significant beneficial application.

8.
BMC Res Notes ; 13(1): 259, 2020 May 27.
Article in English | MEDLINE | ID: mdl-32460808

ABSTRACT

OBJECTIVE: This study evaluated hepatoprotective effect of aescin (AES) and diosmin (DIO), individually or in low-dose combination in chemically induced liver injury in rats. Rats were divided into 6 groups; Group 1, control, Group 2, injected with a single dose of a mixture of corn oil and carbon tetrachloride (CCl4) to induce hepatic toxicity. Before CCl4 injection, Groups 3-6 were treated daily for 14 days with silymarin (SIL) (200 mg/kg), aescin (AES; 3.6 & 1.75 mg/kg), Diosmin (DIO; 100 & 50 mg/kg). Serum samples were analyzed for different liver function, oxidative stress and antioxidant markers. Moreover, inflammation and tissue damage were confirmed by histological staining of liver tissue sections. RESULTS: Results indicated that CCl4 elevated serum levels of all assessed liver function markers and decreased levels of key antioxidants. Administration of AES and/or DIO significantly reversed all those CCl4-induced effects. Histopathological study showed disruption of the hepatic architecture, necrosis and inflammatory cells and depositions of glycogen and protein in the tissues of CCl4-treated group. Pretreatment with DIO and/or AES significantly improved histopathological structure of liver tissue. In conclusion, low-dose combination of AES and DIO exhibited significant and preferential hepatoprotective activity compared to individual treatment with AES or DIO.


Subject(s)
Carbon Tetrachloride/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Diosmin/pharmacology , Escin/pharmacology , Protective Agents/pharmacology , Silymarin/pharmacology , Animals , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Diosmin/administration & dosage , Drug Combinations , Escin/administration & dosage , Male , Protective Agents/administration & dosage , Rats , Rats, Wistar , Silymarin/administration & dosage
9.
Sci Rep ; 8(1): 9827, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29959408

ABSTRACT

This study was designed to assess whether the protective effects of bone marrow-derived mesenchymal stem cells (MSCs) against diabetes could be enhanced by pioglitazone (PIO), a PPARγ agonist. Combined MSCs and PIO treatments markedly improved fasting blood glucose, body weight, lipid profile levels, insulin level, insulin resistance, ß cell function. Those protective effects also attenuated both pancreatic lesions and fibrosis in diabetic rats and decreased the depletion of pancreatic mediators of glycemic and lipid metabolism including peroxisome proliferator-activated receptor alpha (PPARα), PGC-1α, GLP-1 and IRS-2. Cardiac biogenesis of diabetic groups was also improved with MSCs and/or PIO treatments as reflected by the enhanced up-regulation of the expressions of cardiac IRS1, Glucose transporter 4, PGC-1, PPARα and CPT-1 genes and the down-regulated expression of lipogenic gene SREBP. The combination of MSCs and PIO also potentiated the decrease of abnormal myocardial pathological lesions in diabetic rats. Similarly, the inhibitory effects of MSCs on diabetic cardiac fibrosis and on the up regulations of TGF-ß, collagen I and III gene expressions were partial but additive when combined with PIO. Therefore, combined therapy with PIO and BMCs transplantation could further potentiate the protective benefit of MSCs against diabetes and cardiac damage compared to MSCs monotherapy.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Niacinamide/toxicity , Streptozocin/toxicity , Thiazolidinediones/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Cells, Cultured , Combined Modality Therapy , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/pathology , Heart Diseases/prevention & control , Male , Rats , Rats, Wistar , Vitamin B Complex/toxicity
10.
Sci Rep ; 8(1): 1270, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352129

ABSTRACT

The purpose of this study was to investigate the anti-cancer property of grape seed extract (GSE) during early stages of developing liver cancer using a two-stage carcinogenic model combining diethylnitrosamine (DEN) and 2-Acetyl Aminofluorene (2-AAF). Administration of GSE at doses 25, 50 and 100 mg/kg per day started at the beginning of promotion periods and continued for 14 weeks. GSE dramatically inhibited pre-neoplastic foci formation as well as significantly decreased the number and the area of placental glutathione-S-transferase in livers of DEN-2AAF-treated rats by approximately 4 & 10 fold deductions, respectively. GSE's effects were associated with induced apoptosis, reduced cell proliferation, decreased oxidative stress and down regulation of histone deacetylase activity and inflammation makers, such as cyclooxygenase 2, inducible nitric oxide synthase, nuclear factor-kappa B-p65 and p- phosphorylated tumor necrosis factor receptor expressions in liver. GSE treatment also decreased the viability of HepG2 cells and induced early and late apoptosis through activating caspase-3 and Bax. Furthermore, GSE induced G2/M and G1/S cell cycle arrest. The present study provides evidence that the GSE's anticancer effect is mediated through the inhibition of cell proliferation, induction of apoptosis, modulating oxidative damage and suppressing inflammatory response.


Subject(s)
Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Grape Seed Extract/therapeutic use , Liver Neoplasms/drug therapy , 2-Acetylaminofluorene/toxicity , Animals , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Cell Cycle Checkpoints , Diethylnitrosamine/toxicity , Grape Seed Extract/pharmacology , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver Neoplasms/etiology , Male , Rats , Rats, Wistar
12.
PLoS One ; 11(11): e0167049, 2016.
Article in English | MEDLINE | ID: mdl-27880817

ABSTRACT

Cardiotoxicity is a limiting factor of doxorubicin (DOX)-based anticancer therapy. Due to its beneficial effects, we investigated whether standardized extract of Melissa officinalis (MO) can attenuate doxorubicin-induced cardiotoxicity and can potentiate the efficacy of DOX against human breast cancer cells. MO was administered orally to male albino rats once daily for 10 consecutive days at doses of 250, 500 and 750 mg/kg b.wt. DOX (15 mg/kg b.wt. i.p.) was administered on the 8th day. MO protected against DOX-induced leakage of cardiac enzymes and histopathological changes. MO ameliorated DOX-induced oxidative stress as evidenced by decreasing lipid peroxidation, protein oxidation and total oxidant capacity depletion and by increasing antioxidant capacity. Additionally, MO pretreatment inhibited inflammatory responses to DOX by decreasing the expressions of nuclear factor kappa-B, tumor necrosis factor-alpha and cyclooxygenase-2 and the activity of myeloperoxidase. MO ameliorated DOX-induced apoptotic tissue damage in heart of rats. In vitro study showed that MO augmented the anticancer efficacy of DOX in human breast cancer cells (MCF-7) and potentiated oxidative damage and apoptosis. Thus, combination of DOX and MO may prove future cancer treatment protocols safer and more efficient.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin , Plant Extracts/pharmacology , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Doxorubicin/adverse effects , Doxorubicin/agonists , Doxorubicin/pharmacology , Drug Synergism , Female , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/prevention & control , Humans , Lipid Peroxidation/drug effects , MCF-7 Cells , Male , Melissa , Plant Extracts/agonists , Rats , Rats, Wistar
13.
Toxicol Lett ; 264: 38-50, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27815112

ABSTRACT

Cisplatin-induced testicular damage is a major obstacle in the application of cisplatin as chemotherapeutic agent. However, it remains as one of the most widely employed anticancer agents in treating various solid tumors including prostate cancer. Since heme-oxygenase-1 (HO-1) is a cytoprotective enzyme with anti-oxidative stress, anti-inflammatory and anticancer activities, we investigated the effects of up-regulation of HO-1 by hemin and its inhibition by zinc protoporphyrin-IX (ZnPP) on cisplatin-induced testicular toxicity in adult rats. Furthermore, the anticancer effect of hemin and ZnPP, with and without cisplatin, was evaluated on human prostate cancer cell line, PC3. Results of the animal study showed that hemin reversed cisplatin-induced perturbations in sperm characteristics, normalized serum testosterone level, and ameliorated cisplatin-induced alterations in testicular and epididymal weights, and restored normal testicular architecture. Moreover, hemin increased the expression and activity of HO-1 protein and prevented cisplatin-induced testicular toxicity by virtue of its antioxidant and anti-inflammatory effects. This effect was evidenced by amelioration of testicular oxidative stress markers (malondialdehyde, nitric oxide, reduced glutathione contents, and catalase activity) and inflammatory mediators (tumor necrosis factor-α and nitric oxide synthase expressions). In contrast, administration of ZnPP (HO-1 inhibitor) did not show significant improvement against cisplatin-induced testicular toxicity. Finally, in vitro analyses showed that, hemin augmented the anticancer efficacy of cisplatin, while ZnPP inhibited its apoptotic effect in PC3 cells. In conclusion, the induction of HO-1 represents a potential therapeutic approach to protect the testicular tissue from the detrimental effects of cisplatin without repressing, but rather augmenting, its cytotoxic effects on PC3 cells.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Heme Oxygenase-1/biosynthesis , Hemin/pharmacology , Infertility, Male/chemically induced , Infertility, Male/prevention & control , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Induction/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , Prostatic Neoplasms/pathology , Protective Agents/pharmacology , Protoporphyrins/pharmacology , Rats
14.
Life Sci ; 141: 13-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26407473

ABSTRACT

AIM: Besides a cholesterol-lowering effect, rosuvastatin (RUV) possesses antioxidant and anti-inflammatory properties. The present study investigates the possible protective effects of RUV in diabetes-induced reproductive damage in rats. MAIN METHODS: Diabetes was induced in male Wistar rats by injecting a single dose of streptozotocin (65mg/kg, i.p.). RUV in low and high doses (5 and 10mg/kg, p.o.) were administrated to diabetic rats for 8weeks. Reproductive damage was evaluated by estimation of testes and epididymis relative weights and caudal sperm count and motility in the control, untreated and RUV-treated diabetic rats. In addition, testicular malondialdehyde, reduced glutathione and nitric oxide levels, as well as, superoxide dismutase and myeloperoxidase activities were estimated. Finally, expressions of inflammatory [inducible nitric oxide synthase (iNOS) and nuclear factor-kappa B (NF-κB)] and apoptotic (caspase-3) markers besides histological examination of testicular tissues were performed. KEY FINDINGS: Results showed that RUV improved sperm count and motility with decrease in testicular nitric oxide and malondialdehyde levels, as well as, myeloperoxidase activity and increase in reduced glutathione level and superoxide dismutase activity in diabetic rats. Further, RUV reduced testicular inflammation and cell death by decreasing the expressions of iNOS, NF-κB and caspase-3. SIGNIFICANCE: Treatment with RUV protects against diabetes-induced testicular damage, in a dose dependent manner, through antioxidant, anti-inflammatory and anti-apoptotic mechanisms.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus, Experimental/complications , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Infertility, Male/drug therapy , Infertility, Male/etiology , Inflammation/drug therapy , Oxidative Stress/drug effects , Rosuvastatin Calcium/pharmacology , Rosuvastatin Calcium/therapeutic use , Animals , Biomarkers/metabolism , Epididymis/pathology , Infertility, Male/pathology , Inflammation Mediators , Male , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Rats , Rats, Wistar , Sperm Count , Sperm Motility/drug effects , Testis/metabolism , Testis/pathology , Testosterone/metabolism
15.
Toxicol Mech Methods ; 24(6): 433-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24966012

ABSTRACT

Isotretinoin (Iso) is a widely used retinoid for the treatment of dermatologic conditions. Although it has broad side effects, there is no well-designed study about preventive effects against its hepatic toxicity. This study was undertaken to evaluate the protective effect of selenium (Se) against Iso-induced hepatotoxicity in Wistar rats. Animals were divided into four groups. The first group served as control. The second, third and fourth groups received Se, Iso and Se & Iso, respectively, for 28 days. Se was administered daily orally at a dose of 50 µg / 100 g body weight. Iso was given daily orally at a dose of 0.75 mg/ 100 g /day in olive oil. Iso caused significant increases in serum levels of alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, cholesterol, triglycerides, and high-density lipids content. Animals also showed significant rise in thiobarbituric acid reacting substance and nitric oxide content with concomitant decrease in reduced glutathione content and the antioxidant enzyme activities of superoxide dismutase and catalase in liver tissue after Iso exposure. Se administration produced a significant protection against the hepatotoxic effects of Iso and markedly alleviated alterations in these parameters. The results obtained herein clearly indicate that Iso causes induction of oxidative stress and the co-administration of Iso and Se provides protection against Iso-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Isotretinoin/toxicity , Sodium Selenite/pharmacology , Teratogens/toxicity , Trace Elements/pharmacology , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Alkaline Phosphatase/blood , Alkaline Phosphatase/metabolism , Animals , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Cholesterol/blood , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Lipid Metabolism/drug effects , Lipids/blood , Rats , Rats, Wistar , Sodium Selenite/administration & dosage , Trace Elements/administration & dosage , Triglycerides/blood
16.
Drug Chem Toxicol ; 34(2): 101-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21314459

ABSTRACT

The aim of this study was to investigate the protective action of licorice in diabetic nephropathy in male rats. Diabetes was induced in male Wistar rats by using streptozotocin (60 mg/kg body weight). Daily oral ingestion (1 g/kg body weight) of licorice extract for 60 days after the onset of diabetes reversed the adverse effect of diabetes on rats. Licorice extract alleviated blood glucose levels, restored renal function, and attenuated body-weight loss. In addition, licorice extract modulated the adverse effect of diabetes on renal malondialdehyde, glutathione, superoxide dismutase, and catalase activity. Further, licorice extract restored the total antioxidant capacity of diabetic rat kidneys. The biochemical analyses were reinforced by histologic investigations, where focal segmental glomerulosclerosis, tubular damage, and hyperemic kidney were the histologic changes seen in diabetic, but not in treated, rats. In conclusion, the biochemical analysis and the histologic investigations of diabetic rat kidneys treated with licorice extract revealed that licorice may have a potential therapeutic effect for diabetes due to its antioxidant and -hyperglycemic properties.


Subject(s)
Antioxidants/pharmacology , Diabetic Nephropathies/drug therapy , Glycyrrhiza/chemistry , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Plant Extracts/pharmacology , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Kidney/metabolism , Male , Oxidoreductases/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...