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










Publication year range
1.
BMC Cancer ; 21(1): 903, 2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34364387

ABSTRACT

BACKGROUND: 5-Fluorouracil (5-FU) is regarded as the first line treatment for colorectal cancer; however, its effectiveness is limited by drug resistance. The ultimate goal of cancer therapy is induction of cancer cell death to achieve an effective outcome with minimal side effects. The present work aimed to assess the anti-cancer activities of mitocans which can be considered as an effective anticancer drug due to high specificity in targeting cancer cells. METHODS: MTT (3-4,5-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide) assay was performed to determine the effects of our mitocans on cell viability and cell death. Apoptosis and necrosis, caspase 3 activity, mitochondrial membrane potential and ROS production in HT29 cell lines were analyzed by ApopNexin™ FITC/PI Kit, Caspase- 3 Assay Kit, MitoTracker Green and DCFH-DA, respectively. Moreover, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression level of pro-apoptotic (Bax) and anti-apoptotic (Bcl-2) genes in HT29 cell lines. RESULTS: Treatment with mitocans (3Br-P + DCA) inhibited the growth of HT29. Moreover, 3Br-P + DCA significantly induced apoptosis and necrosis, activation of caspase 3 activity, depolarize the mitochondrial membrane potential, and ROS production. At a molecular level, 3Br-P + DCA treatment remarkably down-regulated the expression of Bcl-2, while up-regulated the expression of Bax. CONCLUSION: Mitocans, in particular the combined drug, 3Br-P + DCA, could be regarded and more evaluated as a safe and effective compound for CRC treatment. Targeting hexokinase and pyruvate dehydrogenase kinase enzymes may be an option to overcome 5-FU -mediated chemo-resistant in colorectal cancer.


Subject(s)
Apoptosis/drug effects , Dichloroacetic Acid/pharmacology , Glucose/metabolism , Metabolic Networks and Pathways/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Pyruvates/pharmacology , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms , Drug Synergism , Fluorouracil , HT29 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Models, Biological
2.
Avicenna J Phytomed ; 11(4): 394-406, 2021.
Article in English | MEDLINE | ID: mdl-34290970

ABSTRACT

OBJECTIVE: Bisphenol A (BPA) is an organic synthetic compound, often used in manufacturing polycarbonate plastics. Researches have shown the role of BPA as an endocrine disruptor. The present study intended to evaluate the hepatoprotective properties of naringin, an active flavanone glycoside present in many citrus fruit, against hepatotoxicity induced by BPA. MATERIALS AND METHODS: Male Wistar rats were orally treated with 50 mg/kg BPA for 30 consecutive days for induction of toxicity and 40, 80 and 160 mg/kg naringin for the same period along with BPA or alone. RESULTS: This study demonstrated that BPA significantly increased serum levels of triglyceride, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), lipid peroxidation, and aspartate aminotransferase (AST) and significantly reduced catalase, glutathione peroxidase (GPx) and superoxide dismutase (SOD) activity, glutathione (GSH) and caused periportal inflammation and microvesicular steatosis in rat tissue. However, BPA did not change serum levels of high-density lipoprotein-cholesterol (HDL-C), total cholesterol, alanine aminotransferase (ALT), or low-density lipoprotein-cholesterol (LDL-C). Furthermore, the results displayed that administration of 80 and 160 mg/kg naringin improved hepatotoxicity and altered lipid peroxidation level, serum values of triglyceride and liver enzymes, and oxidative stress factors that were induced by BPA. The effect of two doses of 80 and 160 mg/kg naringin was more noticeable than that of dose 40 mg/kg. CONCLUSION: The findings suggested the protective effects of naringin against BPA-induced hepatotoxicity via ameliorating liver histopathological alteration, suppressing oxidative stress and lipid-lowering properties.

3.
Vet Res Forum ; 12(1): 77-85, 2021.
Article in English | MEDLINE | ID: mdl-33953877

ABSTRACT

Increasing applications of carbon nanotubes (CNTs) indicate the necessity to examine their toxicity. According to previous studies, CNTs caused oxidative stress that impaired ß-cell functions and reduced insulin secretion. Our previous study indicated that single-walled carbon nanotubes (SWCNTs) could induce oxidative stress in pancreatic islets. However, there is no study on the effects of multi-walled carbon nanotubes (MWCNTs) on islets and ß-cells. Therefore, the present study aims to evaluate effects of MWCNTs on the oxidative stress of islets and the protective effects of caffeic acid (CA) as an antioxidant. The effects of MWCNTs and CA on islets were investigated using MTT assay, reactive oxygen species (ROS), malondialdehyde (MDA), activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), the content of glutathione (GSH) and mitochondrial membrane potential (MMP) and insulin secretion measurements. The lower viability of islet cells was dose-dependent due to the exposure to MWCNTs according to the MTT assay. Further studies revealed that MWCNTs decreased insulin secretion and MMP, induced ROS creation, increased the MDA level, and decreased activities of SOD, GSH-Px, CAT, and content of GSH. Furthermore, the pretreatment of islets with CA returned the changes. These findings indicated that MWCNTs might induce the oxidative stress of pancreatic islets occurring diabetes and protective CA effects that were mediated by the augmentation of the antioxidant defense system of islets. Our research suggested the necessity of conducting further studies on effects of MWCNTs and CA on the diabetes.

4.
Environ Sci Pollut Res Int ; 27(12): 13447-13456, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32026367

ABSTRACT

Multi-walled carbon nanotubes (MWCNTs) are material with exclusive features that can be applied in different fields including industrial and medicine. It has been determined that the accumulation of MWCNTs in the organs is along with genotoxic and cytotoxic injuries. Previous studies have shown mitochondrial dysfunction in MWCNTs exposure with cell lines, but their exact mechanisms with isolated mitochondria have remained unclear. The present study evaluated toxicity induced by MWCNTs in isolated rat heart mitochondria and protective effect of naringin. Our results showed that MWCNTs toxicity caused the prevention of heart mitochondrial complex II activity. Treatment of isolated heart mitochondria with MWCNTs led to an increase in mitochondrial reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP) collapse, and mitochondrial malondialdehyde (MDA) and a decrease in mitochondrial glutathione (GSH) level and mitochondrial catalase (CAT) activity. Pretreatment of isolated heart mitochondria with naringin decreased mitochondrial oxidative damage through decreasing lipid peroxidation, returned mitochondrial complex II changes, decreasing MMP collapse and ROS production, and restoration of GSH level and CAT activity. Our findings indicated that MWCNTs had toxic effects on isolated heart mitochondria by inducing oxidative stress and possibly apoptosis pathway. The protection effects of naringin may be accompanied by mitochondrial conservation by its antioxidant property or due to its free radical scavenging. Our findings indicated that naringin had a possible role in preventing the mitochondria complaints in the heart.


Subject(s)
Mitochondria, Heart , Nanotubes, Carbon , Animals , Apoptosis , Flavanones , Oxidative Stress , Rats , Reactive Oxygen Species
5.
Drug Chem Toxicol ; 43(1): 85-95, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30264589

ABSTRACT

Bisphenol A (BPA), which is an applied endocrine disrupting chemical in industry for producing epoxy resins and polycarbonate plastics and naringin, is an active flavanone glycoside of grapefruit and many citrus fruits. The present study evaluated the protective effect of naringin against cardiotoxicity induced by BPA. Male Wistar rats were divided into six groups. Control group received oral olive oil; and BPA group orally were administrated 50 mg/kg of BPA for 30 d consecutively to induce toxicity. 40, 80, and 160 mg/kg of naringin were orally administered for 30 consecutive, along with BPA. Naringin group orally received 160 mg/kg of naringin for 30 d consecutively. Animals were sacrificed and their biochemical, histological, and oxidative stress parameters were measured 24 h after the last treatment. Heart injury was induced by BPA as an evidence with a significant increase in levels of aspartate aminotransferase, lactate dehydrogenase, creatine kinase-MB, triglyceride, lipid peroxidation, and a significant decrease in levels of glutathione, superoxide dismutase, catalase, and glutathione peroxidase and triggered myocardial disorganization, myofibrillar loss, congestion of red blood cells, and the inflammation. However, there were not any changes in the total cholesterol, low-density lipoprotein-cholesterol, high-density lipoprotein-cholesterol, and alanine aminotransferase. Moreover, our results indicated that administering 80 and 160 mg/kg of naringin significantly altered all examined endpoints that were induced by BPA. Both concentrations of 80 and 160 mg/kg of naringin were more effective than 40 mg/kg. These findings indicated that naringin had a protective effect against cardiotoxicity induced by BPA through lipid-lowering properties, antioxidant activity, and suppressed lipid peroxidation.


Subject(s)
Benzhydryl Compounds/toxicity , Cardiotoxicity/prevention & control , Flavanones/pharmacology , Oxidative Stress/drug effects , Phenols/toxicity , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cardiotoxicity/etiology , Dose-Response Relationship, Drug , Endocrine Disruptors/toxicity , Flavanones/administration & dosage , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar
6.
Metab Brain Dis ; 35(2): 305-313, 2020 02.
Article in English | MEDLINE | ID: mdl-31630319

ABSTRACT

Opioid-induced neuroinflammation plays a role in the development of opioid physical dependence. Moreover, nitric oxide (NO) has been implicated in several oxidative and inflammatory pathologies. Here, we sought to determine whether treatment with venlafaxine during the development of morphine dependence could inhibit naloxone-precipitated withdrawal symptoms. The involvement of neuro-inflammation related cytokines, oxidative stress, and L-arginine (L-arg)-NO pathway in these effects were also investigated. Mice received morphine (50 mg/kg/daily; s.c.), plus venlafaxine (5 and 40 mg/kg, i.p.) once a day for 3 consecutive days. In order to evaluate the possible role of L-arg-NO on the effects caused by venlafaxine, animals received L-arg, L-NAME or aminoguanidine with venlafaxine (40 mg/kg, i.p.) 30 min before each morphine injection for 3 consecutive days. On 4th day of experiment, behavioral signs of morphine-induced physical dependence were evaluated after i.p. naloxone injection. Then, brain levels of tissue necrosis factor-alpha (TNF-α), interleukin-1-beta (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), brain-derived neurotrophic factor (BDNF), NO and oxidative stress factors including; total thiol, malondialdehyde (MDA) contents and glutathione peroxidase (GPx) activity were determined. Co-administration of venlafaxine (40 mg/kg) with morphine not only inhibited the naloxone-precipitated withdrawal signs including jumping and weight loss, but also reduced the up-regulation of TNF-α, IL-1ß, IL-6, NO and MDA contents in mice brain tissue. However, repeated administration of venlafaxine inhibited the decrease in the brain levels of BDNF, total thiol and GPx. Pre-administration of L-NAME and aminoguanidine improved, while L-arg antagonized the venlafaxine-induced effects. These results provide evidences that venlafaxine could be used as a candidate drug to inhibit morphine withdrawal through the involvement of inflammatory cytokines and l-arginine-NO in mice.


Subject(s)
Cytokines/antagonists & inhibitors , Morphine/adverse effects , Naloxone/toxicity , Nitric Oxide/antagonists & inhibitors , Substance Withdrawal Syndrome/drug therapy , Venlafaxine Hydrochloride/therapeutic use , Animals , Cytokines/metabolism , Male , Mice , Morphine Dependence/drug therapy , Morphine Dependence/metabolism , Nitric Oxide/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Serotonin and Noradrenaline Reuptake Inhibitors/therapeutic use , Substance Withdrawal Syndrome/metabolism , Venlafaxine Hydrochloride/pharmacology
7.
Can J Physiol Pharmacol ; 97(12): 1176-1184, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31491344

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a promising anticancer cytokine with minimal toxicity towards normal cells. Nevertheless, most primary cancers are often intrinsically TRAIL-resistant or can acquire resistance after TRAIL therapy. This study aimed to investigate the inhibitory effect of co-treatment of 3-bromopyruvate (3-BP) as a potent anticancer agent with TRAIL on colon cancer cells (HT-29). The results of present study indicated that combined treatment with 3-BP and TRAIL inhibited the proliferation of HT-29 cells to a greater extent (88.4%) compared with 3-BP (54%) or TRAIL (11%) treatment alone. In contrast, the combination of 3-BP and TRAIL had no significant inhibitory effect on the proliferation of normal cells (HEK-293) (8.4%). At a cellular mechanistic level, the present study showed that 3-BP sensitized human colon cancer cells to TRAIL-induced apoptosis via reactive oxygen species generation, upregulation of Bax, downregulation of Bcl-2 and survivin, release of cytochrome c into the cytosol, and activation of caspase-3. In normal cells, 3-BP, TRAIL, or combination of both had no significant effect on the reactive oxygen species levels, release of cytochrome c, and caspase-3 activity. Therefore, the combination of 3-BP and TRAIL can be a promising therapeutic strategy for treatment of colon cancer.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Colonic Neoplasms/pathology , Mitochondria/drug effects , Pyruvates/pharmacology , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antineoplastic Agents/pharmacology , Cytochromes c/metabolism , Down-Regulation/drug effects , Drug Synergism , Enzyme Activation/drug effects , HEK293 Cells , HT29 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
8.
Metab Brain Dis ; 34(6): 1595-1606, 2019 12.
Article in English | MEDLINE | ID: mdl-31422513

ABSTRACT

Both arsenic (As) and obesity are associated with brain disorders. However, long term studies to evaluate their concomitant adverse effects on the brain functions are lacking. Present study was conducted to evaluate the long term co-exposure of As and high fat diet (HFD) on memory and brain mitochondrial function in mice. Male mice were randomly divided into 7 groups fed with HFD or ordinary diet (OD) and instantaneously exposed to As (25 or 50 ppm) in drinking water for, 4, 8, 12, 16 or 20 weeks. Step-down passive avoidance method was used for memory assessment and post exposure various parameters including mitochondrial damage, level of reactive oxygen species (ROS), malondialdeid (MDA) and glutathione (GSH) were determined. Results indicated that the retention latency decreased in As (25 and 50 ppm) and HFD received mice after 12 and 16 weeks respectively. Same results were observed at significantly shorter duration (8th week) when As was administered along with HFD as compared to control group. In the HFD alone fed mice increased the mitochondrial membrane damage, levels of ROS and MDA were observed while GSH contents decreased significantly. Concomitant administration of HFD and As amplified those mentioned toxic effects (p < 0.001). In conclusion, our findings demonstrated that the simultaneous HFD and As impaired memory at least three times more than exposing each one alone. These toxic effects could be due to the mitochondria originated oxidative stress along with the depleted antioxidant capacity of the brain of mice.


Subject(s)
Arsenic Poisoning/metabolism , Arsenites/pharmacology , Diet, High-Fat/adverse effects , Memory Disorders/metabolism , Memory/drug effects , Mitochondria/drug effects , Sodium Compounds/pharmacology , Animals , Avoidance Learning/drug effects , Brain/drug effects , Brain/metabolism , Glutathione/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mice , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
9.
Toxicol Ind Health ; 35(7): 497-506, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31272286

ABSTRACT

Single-walled carbon nanotubes (SWCNTs) and multiwalled carbon nanotubes (MWCNTs) are broadly applicable across a variety of industrial fields. Despite their usefulness in many different applications, oxidative stress-induced toxicity of SWCNTs and MWCNTs has not been widely investigated. The present study examined the effects of SWCNTs and MWCNTs on rat brain mitochondria using the 3,4 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and indices of reactive oxygen species (ROS), based on measurements of malondialdehyde (MDA), glutathione (GSH), and mitochondrial membrane potential. Based on the MTT assay, exposure to SWCNTs and MWCNTs decreased mitochondrial survival and viability in a dose-dependent manner. Findings also indicated that MWCNTs and SWCNTs could damage mitochondrial membranes and induce the formation of ROS, as indicated by increased levels of MDA and decreased GSH content. The results of this study suggest that SWCNTs and MWCNTs likely damage brain tissue mitochondria by increasing oxidative stress and possibly activating the apoptosis pathway as well as other pathways of cytotoxicity.


Subject(s)
Brain/drug effects , Mitochondria/drug effects , Nanotubes, Carbon/toxicity , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Cell Survival , Dose-Response Relationship, Drug , Male , Membrane Potential, Mitochondrial/drug effects , Rats , Rats, Wistar
10.
Iran J Basic Med Sci ; 22(3): 315-523, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31156794

ABSTRACT

OBJECTIVES: Bisphenol A (BPA) as a synthetic compound is applied in many plastic industries. BPA has been reported to have endocrine-disrupting feature with cytotoxic effects. The study aimed to evaluate the efficiency of Naringin against testicular toxicity induced by BPA in adult rats. MATERIALS AND METHODS: The animals were assigned into six groups of control, BPA-treated (50 mg/kg), BPA+Naringin-administrated (40, 80, 160 mg/kg) and Naringin-treated (160 mg/kg) for 30 days. At the end of experiments, testicular weight, total testicular protein, epididymal sperm count, testicular enzymes, serum follicle-stimulating hormone (FSH), luteinizing hormone (LH), testosterone and estradiol, testicular enzymatic and non-enzymatic antioxidants and histopathology of testis tissue were evaluated by their own methods. RESULTS: The results showed a reduction in testicular weight, total testicular protein, epididymal sperm count, testicular enzymes (alkaline phosphatase and lactate dehydrogenase) and decrease in the serum TSH, LH, testosterone and estradiol in BPA-administrated rats. Furthermore, BPA reduced the enzyme activities of glutathione peroxidase, superoxide dismutase, and catalase in testis tissue. Also, BPA caused an induction in lipid peroxidation and increase in reactive oxygen species levels, whereas it decreased the glutathione content of testis tissue. Histological findings exhibited seminiferous tubules vacuoles, atrophy and separation of the germinal epithelium in BPA-administrated rats. Oral administration of Naringin along with BPA normalized the biochemical, morphological and histological changes and reduced the testicular toxic condition. CONCLUSION: These results demonstrated that Naringin significantly managed male reproductive toxicity by antioxidant capabilities, preventing morphological modifications and escalating defense mechanism, thereby reducing oxidative stress from BPA-induced damage.

11.
J Family Reprod Health ; 13(4): 181-190, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32518568

ABSTRACT

Objective: Obesity is associated with reproductive disorders. Arsenic disrupts male reproduction by direct effects on the male gonads or androgens secretion. So, the present study was conducted to evaluate the toxic effects of chronic concomitant administration of high-fat diet (HF) and arsenic on the reproductive system of the male mouse. Materials and methods: In this experimental study, 72 adult male mice were randomly divided into 6 groups: low-fat diet (LF0), LF+arsenic 25 ppm, LF+arsenic 50ppm, HF0, HF+arsenic 25 ppm and, HF+arsenic 50 ppm. 24 hours after the last experimental day, plasma samples, the cauda of epididymis and testis were prepared and removed for hormonal, sperm count and histopathological assessments. Results: Testis weight and volume increased in HF0 than other groups except for LF0. Plasma LH and testosterone levels decreased in LF50, HF0, HF25, and HF50 compared to LF0. A similar effect was observed in plasma FSH levels of HF0, HF25 and HF50 groups compared with LF0. Plasma level of estradiol increased in LF50 versus to other groups. Testosterone to estradiol ratio and sperm count decreased in all groups compared to LF0. Reduced interstitial cells and large numbers of vacuoles were observed in germinal epithelium of HF0 group, that these changes were more intense in both concentrations of arsenic-treated mice. Conclusion: Present study indicated that chronic exposure to HF and arsenic-induced hypogonadotropic hypogonadism concomitant with sperm count reduction and testicular damage.

12.
Int J Mol Cell Med ; 8(2): 141-153, 2019.
Article in English | MEDLINE | ID: mdl-32215265

ABSTRACT

Bisphenol A (BPA) is one of the highest volume chemicals produced worldwide, which is used in many plastic industries. The present study aimed to evaluate the effect of BPA on cognitive functions and oxidative stress, and determine whether the naringin (NG) co-administration can modify the effect of this compound on cognitive functions and inhibit any possible oxidative stress in the brain tissue of rats. Adult male Wistar rats were divided into six groups. Group I: control, Group II: BPA-treated rats (50 mg/kg/day), Group III, IV, V: BPA+NG (40, 80, 160 mg/kg/day), Group VI: NG (160 mg/kg/day) alone. Cognitive functions were evaluated using step-down latency (SDL) on a passive avoidance apparatus, and transfer latency (TL) in elevated plus-maze. A significant decrease in SDL, prolongation of TL, noticeable oxidative impairment and increase in acetylcholinesterase activity were observed in the BPA-treated in comparison with the control group. Also, the co-administration of NG (160 mg/kg) antagonized the effect of BPA on SDL and TL, attenuated oxidative damage by lowering malondialdehyde and nitrite concentrations and restored superoxide dismutase, catalase, and glutathione S-transferase activities. On the other hand, acetylcholinesterase activity was reduced in the groups co-administred with NG (80 or 160 mg/kg) and BPA in comparison with the BPA alone-treated group. The present study highlighted the therapeutic potential of NG against BPA-induced cognitive impairment and oxidative damage.

13.
Behav Brain Res ; 361: 86-94, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30550947

ABSTRACT

Here, we aimed to investigate the effects of fluoxetine on morphine-induced analgesia, as well as preventive effects of it on morphine induced tolerance and dependence in mice. We also elucidate the involvement of L-type Ca2+ channels in these phenomena. To induce morphine tolerance, mice were treated with morphine (50 mg/kg) for 3 consecutive days. To evaluate the involvement of the calcium channel in the effects of fluoxetine (5, 20 mg/kg), combination ineffective doses of the two L-type calcium channel blockers, nimodipine (5 mg/kg) or diltiazem (20 mg/kg) with flouxetine were used with each morphine dose. Nociceptive behavior was evaluated using hot-plate test, while physical dependence assessed by naloxone-precipitated withdrawal on the fourth day of experiment. The expression of Cav1.2 and Cav1.3 subunits of the L-type calcium channels in cortex and mesolimbic tissues were measured using western immunoassay. Results showed that co-administration of fluoxetine (20 mg/kg) with morphine increased its acute analgesia effect and prevented the induction of morphine antinociceptive tolerance and physical dependence in mice. Moreover, these effects was potentiated by pre-treatment with diltiazem or nimodipine. Results also showed up-regulation of the Cav1.3 and Cav1.2 expression in the cerebral cortex and mesolimbic regions through the development of morphine dependence. Moreover, chronic administration of fluoxetine with morphine reduced the observed up-regulation of Cav1.3 and Cav1.2 expression in cortex and mesolimbic tissues. Our data indicated that co-administering of fluoxetine with morphine could potentiate the antinociceptive effect of morphine, prevent morphine analgesia tolerance and attenuated the morphine withdrawal signs during induction phases. Moreover, we also pointed out for the first time the role of L-type Ca2+ channel channels in the modulatory effects of fluoxetine on the morphine-related effects.


Subject(s)
Calcium Channels, L-Type/drug effects , Fluoxetine/pharmacology , Substance-Related Disorders/prevention & control , Analgesics, Opioid/pharmacology , Animals , Diltiazem/pharmacology , Drug Tolerance/physiology , Male , Mice , Morphine/pharmacology , Morphine Dependence/metabolism , Nimodipine/pharmacology , Pain/drug therapy , Substance-Related Disorders/physiopathology
14.
Sci Rep ; 8(1): 11960, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097599

ABSTRACT

The key features of type 2 diabetes mellitus (T2DM) caused by high fat diet (HFD) in combination with arsenic (As) exposure (pronounced glucose intolerance despite a significant decrease in insulin resistance) are different from those expected for T2DM. Autophagy has been considered as a possible link between insulin resistance and obesity. Therefore in this study, we utilized autophagy gene expression profiling via real-time RT-PCR array analysis in livers of NMRI mice exposed to an environmentally relevant and minimally cytotoxic concentration of arsenite (50 ppm) in drinking water while being fed with a HFD for 20 weeks. Out of 84 genes associated with autophagy under study, 21 genes were related to autophagy machinery components of which 13 genes were downregulated when HDF diet was applied. In this study, for the first time, it was shown that the exposure to arsenic in the livers of mice chronically fed with HFD along with increased oxidative stress resulted in the restoration of autophagy [upregulation of genes involved in the early phase of phagophore formation, phagophore expansion and autophagosome-lysosome linkage stages]. Considering the role of arsenic in the induction of autophagy; it can be argued that reduced insulin resistance in HFD - As induced diabetes may be mediated by autophagy upregulation.


Subject(s)
Autophagy , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Animals , Arsenic/adverse effects , Autophagy/genetics , Biomarkers , Blood Glucose , Diabetes Mellitus, Type 2/blood , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation , Insulin/blood , Insulin/metabolism , Liver/metabolism , Liver/pathology , Male , Metabolic Networks and Pathways , Mice , Models, Biological , Oxidative Stress , Signal Transduction , Ubiquitination
15.
Monaldi Arch Chest Dis ; 88(1): 903, 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29741077

ABSTRACT

Arsenic is a common environmental and occupational contaminant worldwide which can influence the development of respiratory diseases. In recent years, alteration in the lifestyle as well as food habits have led to increased consumption of food containing high levels of fat. The present study was designed to evaluate the effects of chronic exposure to a high-fat diet (HFD) on arsenic-induced damages and oxidative stress in the lung tissue of mice. This is the first study to reveal the effect of diet-induced obesity on arsenic-induced lung damages. Seventy-two male Naval Medical Research Institute (NMRI) mice were divided into six groups and fed an HFD or standard diet (SD) while being exposed to 25 or 50 ppm of arsenic through drinking water for 20 weeks. At the end of the experiment, the lung weight to body weight ratio; oxidative stress markers, nitrite level, and hydroxyproline content in the lung tissue; and lung histology were evaluated. The results demonstrated that arsenic exposure leads to a significant decrease in the glutathione level and catalase enzyme activity, and significantly increased reactive oxygen species, malondialdehyde, and nitrite level, but it did not affect the superoxide dismutase activity and hydroxyproline content in the lung tissue. Consequently, all the parameters studied aggravated when HFD was consumed along with arsenic. These findings were confirmed by histological examination. Our study showed that HFD increased arsenic-induced lung damages through oxidative stress in mice. These findings could be important for clinical research to protect against arsenic-induced respiratory toxicity in humans.

16.
Iran J Pharm Res ; 17(1): 164-183, 2018.
Article in English | MEDLINE | ID: mdl-29755549

ABSTRACT

Obesity is a main reason of type 2 diabetes and also chronic exposure to arsenic (As) can produce diabetic symptoms. In previous studies, the association between high-fat diet and arsenic in the incidence of diabetes was found, but the role of beta cells activity, liver mitochondrial oxidative stress, and hepatic enzymes (leptin, adiponectin and beta amylase) was unclear. Thus, present study was conducted to evaluate the diabetogenic mechanism of arsenic followed by concomitant administration of high-fat diet (HFD) in male mice. In this experimental study, the mice consumed with HFD or low-fat diet (LFD) while exposed to As 25 or 50 ppm in drinking water for 20 weeks. At the end of experiments, hyperglycemia, insulin resistance variables, lipid profile, hepatic enzymes, liver mitochondrial oxidative stress, islet insulin secretion, liver, and pancreas histopathology were evaluated in all mice by their own methods. Control HFD fed mice showed a significant increase in FBG, OGTT, HOMA-IR, ITT, lipid profile, leptin, ß-amylase, liver mitochondrial oxidative stress, hepatic enzymes and decreased FPI, HOMA-ß, adiponectin, and islet insulin secretion or content. However, exposure to HFD concomitant with Arsenic revealed an impressive reduction in FBG, FPI, HOMA-IR, HOMA-ß, ITT, lipid profile, and islet insulin secretion or content. This exposure enhanced OGTT, leptin, adiponectin, liver mitochondrial oxidative stress, and hepatic enzymes. In conclusion, HFD and arsenic concomitant administration induced impairment of OGTT and islet insulin secretion or content through the mitochondrial oxidative stress.

17.
Korean J Physiol Pharmacol ; 22(3): 257-267, 2018 May.
Article in English | MEDLINE | ID: mdl-29719448

ABSTRACT

The present study aimed to evaluate the cinnamic acid effect on memory impairment, oxidative stress, and cholinergic dysfunction in streptozotocin (STZ)-induced diabetic model in mice. In this experimental study, 48 male Naval Medical Research Institute (NMRI) mice (30-35 g) were chosen and were randomly divided into six groups: control, cinnamic acid (20 mg/kg day, i.p. ), diabetic, and cinnamic acid-treated diabetic (10, 20 and 40 mg/kg day, i.p. ). Memory was impaired by administering an intraperitoneal STZ injection of 50 mg/kg. Cinnamic acid was injected for 40 days starting from the 21st day after confirming STZ-induced dementia to observe its therapeutic effect. Memory function was assessed using cross-arm maze, morris water maze and passive avoidance test. After the administration, biochemical parameters of oxidative stress and cholinergic function were estimated in the brain. Present data indicated that inducing STZ caused significant memory impairment, whereas administration of cinnamic acid caused significant and dose-dependent memory improvement. Assessment of brain homogenates indicated cholinergic dysfunction, increase in lipid peroxidation and reactive oxygen species (ROS) levels, and decrease in glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT) activities in the diabetic group compared to the control animals, whereas cinnamic acid administration ameliorated these indices in the diabetic mice. The present study demonstrated that cinnamic acid improves memory by reducing the oxidative stress and cholinergic dysfunction in the brain of diabetic mice.

18.
Exp Neurol ; 303: 134-141, 2018 05.
Article in English | MEDLINE | ID: mdl-29453978

ABSTRACT

Opioid-induced neuroinflammation and the nitric oxide (NO) signal-transduction pathway are involved in the development of opioid analgesic tolerance. The antidepressant venlafaxine (VLF) modulates NO in nervous tissues, and so we investigated its effect on induced tolerance to morphine, neuroinflammation, and oxidative stress in mice. Tolerance to the analgesic effects of morphine were induced by injecting mice with morphine (50 mg/kg) once a day for three consecutive days; the effect of co-administration of VLF (5 or 40 mg/kg) with morphine was similarly tested in a separate group. To determine if the NO precursor l-arginine hydrochloride (l-arg) or NO are involved in the effects rendered by VLF, animals were pre-treated with l-arg (200 mg/kg), or the NO synthesis inhibitors N(ω)-nitro-l-arginine methyl ester (L-NAME; 30 mg/kg) or aminoguanidine hydrochloride (AG; 100 mg/kg), along with VLF (40 mg/kg) for three days before receiving morphine for another three days. Nociception was assessed with a hot-plate test on the fourth day, and the concentration of tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1ß), interleukin-6 (IL-6), interleukin-10, brain-derived neurotrophic factor, NO, and oxidative stress factors such as total thiol, malondialdehyde content, and glutathione peroxidase (GPx) activity in the brain was also determined. Co-administration of VLF with morphine attenuated morphine-induced analgesic tolerance and prevented the upregulation of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6), NO, and malondialdehyde in brains of mice with induced morphine tolerance; chronic VLF administration inhibited this decrease in brain-derived neurotrophic factor, total thiol, and GPx levels. Moreover, repeated administration of l-arg before receipt of VLF antagonized the effects induced by VLF, while L-NAME and AG potentiated these effects. VLF attenuates morphine-induced analgesic tolerance, at least partly because of its anti-inflammatory and antioxidative properties. VLF also appears to suppress the development of morphine-induced analgesic tolerance through an l-arg-NO-mediated mechanism.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Cytokines/metabolism , Morphine Dependence/prevention & control , Oxidative Stress/drug effects , Signal Transduction/drug effects , Venlafaxine Hydrochloride/pharmacology , Analgesics, Opioid/adverse effects , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Morphine/adverse effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitrites/metabolism , Pain Measurement
19.
Res Pharm Sci ; 13(1): 47-56, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29387111

ABSTRACT

Diet is one of the important risk factors that could potentially affect arsenic-induced cardiotoxicity. The present study was undertaken to investigate the effect of high fat diet on arsenic-induced cardiotoxicity in mice. Mice were divided into six different groups (n = 12), two control groups received either low fat diet (LFD) or high fat diet (HFD) along with deionized drinking water and four test groups given LFD + 25 ppm arsenic, LFD + 50 ppm arsenic, HFD + 25 ppm arsenic, and HFD + 50 ppm arsenic in drinking water for 5 months. The body weight, heart weight to body weight ratio, cardiac biochemical markers, lipid profile, and histological examination of heart were evaluated. The results demonstrated that arsenic exposure led to a significant decrease in heart glutathione level, catalase enzyme activity, and a significant increase in reactive oxygen species (ROS), malondialdehyde levels, and biochemical enzymes. The administration of HFD resulted in above-mentioned changes as well as an alteration in lipid profile; however, arsenic exposure alone or along with HFD caused a reduction in lipid profile factors, except HDL level. Our results revealed that HFD increased arsenic-induced heart injury in the mice. This effect may be because of reduction in antioxidant activities and/or increase in oxidative stress and ROS in mice heart tissues. These findings could be important for clinical intervention to protect against or prevent arsenic-induced cardiotoxicity in humans.

20.
Can J Physiol Pharmacol ; 96(2): 165-172, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28787580

ABSTRACT

The present study was conducted to evaluate the local antinociceptive actions of fluoxetine, a selective serotonin reuptake inhibitor, and the possible involvement of the l-arginine/NO/cGMP/KATP channel pathway in this effect using the formalin test in rats. To elucidate the underlying mechanisms, animals were pre-treated with l-NAME, aminoguanidine, methylene blue, glibenclamide, l-arginine, sodium nitroprusside, or diazoxide. Local ipsilateral, but not contralateral, administration of fluoxetine (10-300 µg/paw) dose-dependently suppressed flinching number during both early and late phases of the test, and this was comparable with morphine also given peripherally. Pre-treatment with l-NAME, aminoguanidine, methylene blue, or glibenclamide dose-dependently prevented fluoxetine (100 µg/paw)-induced antinociception in the late phase. In contrast, administration of l-arginine, sodium nitroprusside, and diazoxide significantly enhanced the antinociception caused by fluoxetine in the late phase of the test. However, these treatments had no significant effect on the antinociceptive response of fluoxetine in the early phase of the formalin test. Our data demonstrate that local peripheral antinociception of fluoxetine during the late phase of the formalin test could be due to activation of l-arginine/NO/cGMP/KATP channel pathway. The peripheral action of fluoxetine raises the possibility that topical application of this drug (e.g., as a cream, ointment, or jelly) may be a useful method for relieving the inflammatory pain states.


Subject(s)
Arginine/metabolism , Biological Assay , Fluoxetine/pharmacology , KATP Channels/metabolism , Nitric Oxide/metabolism , Nociception/drug effects , Signal Transduction/drug effects , Animals , Cyclic GMP/metabolism , Diazoxide/pharmacology , Formaldehyde/chemistry , Glyburide/pharmacology , Guanidines/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...