Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 485: 116875, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38437957

ABSTRACT

Cisplatin is an effective and commonly used chemotherapeutic drug; however, its use is accompanied by several adverse effects, including chemobrain. Ondansetron is a 5-HT3 antagonist, commonly used in prophylactic against chemotherapy-induced nausea and vomiting. Moreover, it has been identified as a novel neuroprotective agent in different animal models. However, its protective role against chemotherapy-induced chemobrain has not been investigated. The current study was the first study that explored the potential neuroprotective effect of ondansetron against cisplatin-induced chemobrain in rats. Cisplatin (5 mg/Kg) was injected intraperitoneally, once weekly, for 4 weeks with the daily administration of ondansetron (0.5 and 1 mg/Kg). Compared to the cisplatin-treated group, ondansetron administration showed a significant decrease in the latency time and a significant increase in ambulation, rearing, and grooming frequency in the open field test (OFT). Moreover, a significant improvement in the latency time in the rotarod and passive avoidance tests, following ondansetron administration. In addition, ondansetron treatment increased the percentage of alternation in the Y-maze test. Also, ondansetron showed a remarkable enhancement in the biochemical parameters in the hippocampus. It increased the acetylcholine (Ach) level and decreased the level of the acetylcholine esterase enzyme (AchE). Ondansetron significantly decreased interleukin-1ß (Il-1ß), tumor necrosis factor-alpha (TNF-α), toll-like receptor-4 (TLR-4), NOD-like receptor-3 (NLRP3) inflammasome as well as caspase-1 and caspase-3 levels. Furthermore, ondansetron significantly decreased the levels of copper transporter-1(CTR1) expression in the hippocampus. Collectively, these findings suggest that ondansetron may exhibit a neuroprotective and therapeutic activity against cisplatin-induced chemobrain.


Subject(s)
Behavior, Animal , Cisplatin , Inflammasomes , Ondansetron , Animals , Ondansetron/pharmacology , Cisplatin/toxicity , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , Behavior, Animal/drug effects , Rats , Down-Regulation/drug effects , Neuroprotective Agents/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Wistar , Hippocampus/drug effects , Hippocampus/metabolism , Antineoplastic Agents/toxicity , Signal Transduction/drug effects , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Chemotherapy-Related Cognitive Impairment/drug therapy
4.
J Diet Suppl ; 20(3): 391-410, 2023.
Article in English | MEDLINE | ID: mdl-34633271

ABSTRACT

Depression is a severely debilitating psychiatric disorder that influences more than 15% of the population worldwide. It has been demonstrated that it is associated with a high risk of developing other diseases such as cardiovascular diseases, diabetes, stroke, epilepsy, and cancer. The current study examines the possibility of chrysin and lycopene having an antidepressant effect in a rat model of depression induced by clonidine, as well as the mechanisms underlying this effect, including the role of neuroinflammation and oxidative stress. Rats were allotted into seven groups. The rats in group 1 served as a control. Group 2 received lycopene only. Group 3 was provided chrysin only. Group 4 was administered clonidine and served as the model. Group 5 was offered lycopene and clonidine. Group 6 was administered chrysin and clonidine. Group 7 was given FLX and clonidine and represented the standard. The experiment lasted two weeks, during which behavioral, biochemical, histopathological, and immunohistochemical measurements were performed. Lycopene and chrysin were used to correct the concentrations of noradrenaline and serotonin hippocampal tissue concentrations. These findings were also improved by immunohistochemical analysis of GFAP, VEGF, caspase3, and histopathological examinations, in which pretreatment of rats with lycopene and chrysin reversed all clonidine-induced alterations. The current research demonstrates that lycopene and chrysin have an auspicious antidepressant effect against clonidine that provoked behavioral hopelessness in rats. Manipulating oxidative stress, inflammation, and apoptosis may partially represent the corrective mechanism for the neuroprotective actions against the depressive effect of clonidine.


Subject(s)
Clonidine , Depression , Rats , Animals , Lycopene/pharmacology , Clonidine/pharmacology , Depression/chemically induced , Depression/drug therapy , Neuroinflammatory Diseases , Oxidative Stress , Flavonoids/pharmacology , Antidepressive Agents/pharmacology , Antioxidants/pharmacology
5.
Inflammopharmacology ; 30(6): 2197-2209, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35948811

ABSTRACT

BACKGROUND AND AIM: Multiple sclerosis (MS) is a demyelinating neurodegenerative inflammatory disease affecting mainly young adults. Microgliosis-derived neuroinflammation represents a key hallmark in MS pathology and progression. Nebivolol (Neb) demonstrated antioxidant, anti-inflammatory and neuroprotective properties in several brain pathologies. This study was conducted to investigate the potential neuroprotective effect of Neb in the cuprizone (Cup) model of MS. METHODS: C57Bl/6 mice were fed 0.2% Cup mixed into rodent chow for 5 weeks. Neb (5 and 10 mg/kg/day) was administered by oral gavage during the last 2 weeks. RESULTS: Neb prevented Cup-induced weight loss and motor deficits as evidenced by increased latency to fall in the rotarod test and enhanced locomotor activity as compared to Cup-intoxicated mice. Neb reversed Cup-induced demyelination as confirmed by Luxol fast blue staining and myelin basic protein western blotting. Administration of Neb modulated microglial activation status by suppressing M1 markers (Iba-1, CD86, iNOS, NO and TNF-α) and increasing M2 markers (Arg-1 and IL-10) as compared to Cup-fed mice. Furthermore, Neb hindered NLRP3/caspase-1/IL-18 inflammatory cascade and alleviated oxidative stress by reducing lipid peroxidation, as well as increasing catalase and superoxide dismutase activities. CONCLUSION: These findings suggest the potential neuroprotective effect of Neb in the Cup-induced model of MS in mice, at least partially by virtue of shifting microglia towards M2 phenotype, mitigation of NLRP3 inflammasome activation and alleviation of oxidative stress.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Nebivolol , Neuroprotective Agents , Animals , Mice , Cuprizone/adverse effects , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Disease Models, Animal , Inflammasomes/metabolism , Mice, Inbred C57BL , Microglia , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Nebivolol/pharmacology , Neuroprotective Agents/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Macrophages , Cell Polarity
6.
J Pharm Pharmacol ; 74(7): 1051-1060, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35512548

ABSTRACT

CONTEXT: Renal toxicity correlated with cisplatin administration curbs its clinical application. Accordingly, the identification of novel protective agents is important. Forskolin provides anti-inflammatory, anti-oxidant as well as anti-cancer effects. OBJECTIVES: This study aimed to explore the nephroprotective effect of forskolin in a model of cisplatin-induced acute renal toxicity in rats in addition to exploring the possible mechanisms. METHODS: Rats were sorted into four groups: control group, cisplatin group, cisplatin/forskolin group that was given forskolin (10 mg/kg, i.p.) 1 week before cisplatin and forskolin-only group. Nephrotoxicity markers were tested in the blood. Tissues were used to assess histopathology, oxidative stress, inflammation and apoptosis. KEY FINDINGS: In cisplatin-injected rats, the nephrotoxicity indices were particularly increased. Cisplatin markedly reduced the levels of reduced glutathione and superoxide dismutase. Also, malondialdehyde and Nicotinamide adenine dinucleotide phosphate oxidase were increased. In addition, the pro-inflammatory cytokines and caspase-3 were elevated. Moreover, the epidermal growth factor expression was significantly reduced. Furthermore, marked histopathological changes were noted in the tissues of cisplatin-injected rats. Forskolin attenuated nephrotoxicity markers, inflammation, oxidative stress and apoptotic insults provoked via cisplatin. Moreover, cisplatin cytotoxic activity was not modulated by forskolin in human cultured cancerous cell lines. CONCLUSION: Forskolin provides significant protection from cisplatin-evoked nephrotoxicity enhancing its therapeutic index.


Subject(s)
Acute Kidney Injury , Cisplatin , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Antioxidants/metabolism , Apoptosis , Biomarkers/metabolism , Cisplatin/adverse effects , Colforsin/metabolism , Colforsin/pharmacology , Colforsin/therapeutic use , Humans , Inflammation/drug therapy , Kidney , Oxidative Stress , Rats
7.
Toxicol Appl Pharmacol ; 435: 115853, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34973289

ABSTRACT

Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder. Although mounting studies have been conducted, no effective therapy is available to halt its progression. Indole-3-carbinol (I3C) is a naturally occurring compound obtained by ß-thioglucosidase-mediated autolysis of glucobrassicin in cruciferous vegetables. Besides its powerful antioxidant activity, I3C has shown neuroprotection against depression and chemically induced neurotoxicity via its anti-inflammatory and antiapoptotic effects. This study aimed to investigate the neuroprotective effects of I3C against rotenone (ROT)-induced PD in male albino rats. The possible protective mechanisms were also explored. PD was induced by subcutaneous administration of ROT (2 mg/kg) for 28 days. The effects of I3C (25, 50, and 100 mg/kg/day) were assessed by catalepsy test (bar test), spontaneous locomotor activity, rotarod test, weight change, tyrosine hydroxylase (TH) expression, α-synuclein (α-Syn) expression, striatal dopamine (DA) content, and histological examination. The highest dose of I3C (100 mg/kg) was the most effective to prevent ROT-mediated motor dysfunctions and amend striatal DA decrease, weight loss, neurodegeneration, TH expression reduction, and α-Syn expression increase in both the midbrain and striatum. Further mechanistic investigations revealed that the neuroprotective effects of I3C are partially attributed to its anti-inflammatory and antiapoptotic effects and the activation of the sirtuin 1/AMP-activated protein kinase pathway. Altogether, these results suggested that I3C could attenuate biochemical, molecular, and functional changes in a rat PD model with following repeated rotenone exposures.


Subject(s)
Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/prevention & control , Rotenone , Sirtuin 1/metabolism , Uncoupling Agents , Animals , Body Weight/drug effects , Catalepsy/chemically induced , Catalepsy/prevention & control , Dopamine/metabolism , Male , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Parkinson Disease, Secondary/psychology , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Sirtuin 1/drug effects , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/biosynthesis , alpha-Synuclein/drug effects
8.
Eur J Pharmacol ; 912: 174602, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34710367

ABSTRACT

Depression is a widespread, withering illness, resulting in a massive personal suffering and economic loss. The chronic exposure to stress may be involved in the etiology of human psychiatric disorders; such as depression. In the current study, the animals were subjected to chronic unpredictable mild stress (CUMS) for 14 days. Saxagliptin (SAXA) is a member of dipeptidyl peptidase-4 (DPP-4) inhibitors class. The current study was the first one to examine the anti-depressive effect of SAXA in an experimental model of CUMS-induced depression in rats and the possible underlying mechanisms. Animals were orally treated with SAXA (0.5, 1 and 2 mg/kg) for 14 days. SAXA treatment reversed the CUMS-induced alterations in the behavioral, biochemical as well as histopathological parameters. Moreover, it hindered the CUMS-induced increase in the oxidative stress, inflammatory, and apoptotic markers. On the other hand, it increased the monoamines levels and the neurogenic brain derived neurotrophic factor (BDNF). In addition, SAXA treatment increased the incretin hormones, glucagon like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP), which are linked to the activation of protein kinase B (AKT)/phosphatidylinositol3-kinase (PI3K) pathway. In conclusion, the current study revealed that the modulation of the interplay between the key events involved in depression, including oxidative stress, inflammation, and GLP-1/PI3K/AKT signaling pathway, can explain the anti-depressant activity of SAXA.


Subject(s)
Adamantane/analogs & derivatives , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/metabolism , Dipeptides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , Depression/etiology , Dipeptides/therapeutic use , Disease Models, Animal , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Incretins/pharmacology , Incretins/therapeutic use , Inflammation/metabolism , Male , Oxidative Stress/drug effects , Rats , Signal Transduction/drug effects , Stress, Psychological/complications
9.
Clin Exp Pharmacol Physiol ; 48(10): 1358-1370, 2021 10.
Article in English | MEDLINE | ID: mdl-34081810

ABSTRACT

Rosuvastatin has been shown to activate PI3K/Akt/Nrf2/HO-1 pathway, which promotes cell survival in the myocardium. This study investigated the therapeutic benefit of adding rosuvastatin to low-dose carvedilol in protection against myocardial infarction (MI). Rosuvastatin (RSV) and carvedilol (CAR) were given for 7 consecutive days with concurrent administration of two doses of isoprenaline (ISP) on 6th and 7th days to induce MI. Isoprenaline injections caused detrimental alterations in the myocardial architecture and electrocardiogram (ECG) pattern and significantly increased the infarct size, heart index and serum levels of cardiotoxicity markers compared to the control group. ISP induced oxidative damage, inflammatory and apoptotic events and downregulated PI3K/Akt/Nrf2/HO-1 signalling pathway compared to the control values. Treatment with low-dose CAR and/or RSV prevented the ECG and histopathological alterations induced by ISP, and also reduced the infarct size, heart index, serum creatine kinase-MB, cardiac troponin-I and C-reactive protein levels compared to ISP group. CAR and/or RSV treatment restored the activity of superoxide dismutase and total antioxidant capacity with a consequent reduction in lipid peroxides level. Further, they decreased the expression of nuclear factor (NF)-κB (p65) and increased the phosphorylated PI3K and Akt, which may activate the anti-apoptotic signalling as evidenced by the decreased active caspase 3 level. The combination therapy has a more significant effect in the most studied parameters than their monotherapy, which may be because of the activation of PI3K/Akt Nrf2/HO-1 pro-survival signalling pathway. This study highlights the potential benefits of combining RSV with low-dose CAR in case of MI.


Subject(s)
Carvedilol/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Isoproterenol/toxicity , Myocardial Infarction/drug therapy , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rosuvastatin Calcium/pharmacology , Adrenergic beta-Agonists/toxicity , Adrenergic beta-Antagonists/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Apoptosis/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Male , Myocardial Infarction/chemically induced , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
10.
Toxicol Lett ; 347: 23-35, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33961984

ABSTRACT

Liver fibrosis is the conjoint consequence of almost all chronic liver diseases. Cholestatic liver injury is a significant stimulus for fibrotic liver. This study was conducted to investigate the hepatoprotective effect of niclosamide as a NOTCH inhibitor and on the Wnt pathway against cholestatic liver fibrosis (CLF) which was experimentally induced by bile duct ligation (BDL). Rats were randomly divided into five main groups (6 per group): sham, BDL, BDL/niclosamide 5, BDL/niclosamide 10 and niclosamide 10 only group. Niclosamide was administered intraperitoneally (i.p.) for 4 weeks starting at the same day of surgery at doses 5 and 10 mg/kg. Liver function, cholestasis, oxidative stress, inflammation, liver fibrosis, NOTCH signaling pathway and Wnt pathway markers were assessed. Niclosamide (5 and 10 mg/kg) significantly reduced liver enzymes levels, oxidative stress, inflammation and phosphorylated signal transducer and activator of transcription3 (p-STAT3). Niclosamide (5 and 10 mg/kg) also significantly reduced NOTCH pathway (Jagged1, NOTCH2, NOTCH3, HES1, SOX9), Wnt pathway (Wnt5B, and Wnt10A), and fibrosis (transforming growth factor-beta1 (TGF-ß1), alpha smooth muscle actin (α-SMA) and collagen deposition with more prominent effect of the higher dose 10 mg/kg. So, this study presents nicloamide as a promising antifibrotic agent in CLF through inhibition of NOTCH and Wnt pathways.


Subject(s)
Liver Cirrhosis, Biliary/prevention & control , Liver/drug effects , Niclosamide/pharmacology , Receptors, Notch/metabolism , Wnt Signaling Pathway/drug effects , Animals , Bile Ducts/surgery , Cell Proliferation/drug effects , Collagen/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Ligation , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Male , Oxidative Stress/drug effects , Phosphorylation , Rats, Wistar , SOX9 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism
11.
Int Immunopharmacol ; 94: 107431, 2021 May.
Article in English | MEDLINE | ID: mdl-33578261

ABSTRACT

Parkinson's disease (PD) is a disabling progressive neurodegenerative disease. So far, PD's treatment remains symptomatic with no curative effects. Aside from its blatant analgesic and antipyretic efficacy, recent studies highlighted the endowed neuroprotective potentials of paracetamol (PCM). To this end: the present study investigated: (1) Possible protective role of PCM against rotenone-induced PD-like neurotoxicity in rats, and (2) the mechanisms underlying its neuroprotective actions including cannabinoid receptors' modulation. A dose-response study was conducted using three doses of PCM (25, 50, and 100 mg/kg/day, i.p.) and their effects on body weight changes, spontaneous locomotor activity, rotarod test, tyrosine hydroxylase (TH) and α-synuclein expression, and striatal dopamine (DA) content were evaluated. Results revealed that PCM (100 mg/kg/day, i.p.) halted PD motor impairment, prevented rotenone-induced weight loss, restored normal histological tissue structure, reversed rotenone-induced reduction in TH expression and striatal DA content, and markedly decreased midbrain and striatal α-synuclein expression in rotenone-treated rats. Accordingly, PCM (100 mg/kg/day, i.p.) was selected for further mechanistic investigations, where it ameliorated rotenone-induced oxidative stress, neuro-inflammation, apoptosis, and disturbed cannabinoid receptors' expression. In conclusion, our findings imply a multi-target neuroprotective effect of PCM in PD which could be attributed to its antioxidant, anti-inflammatory and anti-apoptotic activities, in addition to cannabinoid receptors' modulation.


Subject(s)
Acetaminophen/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Acetaminophen/pharmacology , Animals , Apoptosis/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Endocannabinoids , Male , Mesencephalon/drug effects , Mesencephalon/pathology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Rotenone , alpha-Synuclein/metabolism
12.
Front Pharmacol ; 11: 567852, 2020.
Article in English | MEDLINE | ID: mdl-33381027

ABSTRACT

Cisplatin is one of the standard anti-cancer agents that are used to treat variety of solid tumors. Nevertheless, due to the accumulation of cisplatin in the renal epithelial cells, nephrotoxicity was found to be the main side effect that limits its clinical use. The current study was conducted to assess the potential nephroprotective effect of dibenzazepine, a Notch inhibitor, against cisplatin-induced nephrotoxicity in rats as well as the possible mechanisms underlying this nephroprotection. The rats were pre-treated with 2 mg/kg dibenzazepine for 7 days before giving a single nephrotoxic dose of cisplatin (7 mg/kg). Cisplatin induced acute nephrotoxicity, where blood urea nitrogen and serum creatinine levels were significantly increased. Besides, lipid peroxidation was markedly elevated and the levels of reduced glutathione and catalase were significantly reduced. Also, the tissue levels of the pro-inflammatory mediators; IL-1ß, TNF-α, and NF-kB, were significantly increased in the cisplatin group. The pre-treatment with dibenzazepine significantly mitigated the nephrotoxic effects of cisplatin, the oxidative stress and inflammatory status as well as decreased caspase-3 expression, as compared to the cisplatin group. Furthermore, the up-regulation of Notch-1 and Hes-1 was found to be involved in cisplatin-induced nephrotoxicity and their expression was significantly reduced by dibenzazepine. The nephroprotective effect of dibenzazepine was further confirmed by the histopathological assessment. Moreover, dibenzazepine pre-treatment of hela and PC3 cells in vitro did not antagonize the cisplatin anti-cancer activity. In conclusion, these findings show that dibenzazepine provides protection against cisplatin-induced nephrotoxicity. Moreover, the up-regulation of the Notch pathway was shown to play a role in the pathogenesis of cisplatin-induced renal injury.

13.
Biochem Pharmacol ; 180: 114140, 2020 10.
Article in English | MEDLINE | ID: mdl-32652141

ABSTRACT

Depression is a common mental illness that possesses a noteworthy effect on patients' lives. Many theories are recently studied for their plausible involvement in depression pathogenesis, especially oxidative stress and inflammation. Morin (2',3,4',5,7-pentahydroxyflavone), a natural flavonoid, is characterized by its potent anti-inflammatory, and anti-oxidant activities. Accordingly, the aim of the current study was to investigate its potential protective anti-depressant effect in the model of chronic unpredictable mild stress (CUMS) in experimental rats. Moreover, the conceivable neuro-protective mechanisms, especially those related to the inflammasome pathway, were explored. Several, mild, unpredictable stressors were applied for 4 weeks concomitantly with the oral administration of morin (15 and 30 mg/Kg). Morin hydrate supplementations exhibited a significant improvement in the scores of the forced swimming and sucrose preference tests. In addition, it prompted a marked elevation in the ambulation, rearing as well as grooming scores of the open field test. The morin-treated groups showed a great improvement in the biochemical parameters in both the cortex and hippocampus, where it significantly elevated the serotonin, epinephrine, and norepinephrine levels. Also, it significantly increased reduced glutathione levels and decreased malondialdehyde levels. Regarding the inflammasome pathway, morin significantly decreased the tissue levels of tumor necrosis factor-alpha, toll-like receptor-4, interleukin-1beta, NOD-like receptor pyrin domain-containing protein-3, and caspase-1 levels. Morin also significantly decreased the level of the key apoptotic marker, caspase-3. In conclusion, these findings propose that morin might show a promising anti-depressant effect.


Subject(s)
Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Depression/drug therapy , Flavonoids/therapeutic use , Inflammasomes/metabolism , Stress, Psychological/drug therapy , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Depression/psychology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Male , Rats, Sprague-Dawley , Signal Transduction , Stress, Psychological/psychology
14.
Front Pharmacol ; 11: 218, 2020.
Article in English | MEDLINE | ID: mdl-32256346

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a pathological accumulation of triglycerides (TGs) in the hepatocyte in the absence of alcohol intake. Untreated NAFLD is expected to progress into liver fibrosis. Cranberry is rich in polyphenols with antioxidant and anti-inflammatory activities. HYPOTHESIS: The present study was performed to evaluate our hypothesis of the possible anti-fibrotic effect of cranberry nutraceuticals in a high fat cholesterol diet induced (HFCD)-NAFLD in rats, focusing on improving insulin sensitivity and modulating the expression of nuclear factor erythroid-2-related factor-2 (Nrf2) (a transcription factor responsible for regulating cellular redox balance). METHOD: Male albino wistar rats (12 weeks) received HFCD and/or cranberry (50 and 100 mg/kg/day, three times/week) orally for 8 consecutive weeks. RESULTS: In comparison to the HFCD group, cranberry treated groups (50 and 100 mg/kg) showed marked hepatoprotection, where it significantly decreased liver enzymes (alanine transaminases by 49 and 64% and aspartate transaminases by 45 and 64%; respectively), TGs, and ameliorated the histopathological alterations (such as inflammatory cells infiltration and ballooning degeneration) induced by HFCD. Cranberry also alleviated oxidative stress (malondialdehyde, glutathione, catalase and superoxide dismutase) and inflammation (tumor necrosis factor- alpha, interleukine-6 and nuclear factor kappa-b) and significantly reduced the HOMA-IR and TyG index. On the other hand, cranberry treated groups (50 and 100 mg/kg) showed a marked increase in the expression of adiponectin, by 8 and 13-fold, insulin receptor substrate-2 by 21 and 79%, and Nrf2 by 13 and 61%, respectively. Notably, cranberry significantly reduced the fibrotic markers, TGF-ß and α-SMA expression and collagen deposition. CONCLUSION: The present study showed that cranberry significantly attenuated NAFLD, in a dose dependent manner, which could be partially recognized by its antioxidant, anti-inflammatory activities, and its ability to improve insulin sensitivity. Notably, our study proves for the first time that the anti-fibrotic activity of cranberry is promising.

15.
Chem Biol Interact ; 308: 137-146, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31103702

ABSTRACT

Cisplatin, a platinum chemotherapeutic agent, is used in a diversity of malignancies; nevertheless, the excessive nephrotoxicity following cisplatin treatment is the dose-limiting devastating reaction. This study was designed to explore the possible nephroprotective impact of wogonin, a forceful anti-oxidant, anti-inflammatory, and anti-tumor agent, in a rat model of cisplatin-induced renal injury. The potential nephroprotective mechanisms were additionally investigated. Wogonin was given at a dose of 40 mg/kg. Acute nephrotoxicity was indicated by a significant rise in BUN, and serum creatinine levels in cisplatin-injected rats. Also, cisplatin enhanced the lipid peroxidation, diminished GSH, catalase, and PPAR-γ levels. Additionally, cisplatin-injected rats showed a significant rise in tissue levels of IL-1ß, TNF-α, NF-kB, and caspase-3 enzymatic activity. Notably, the pre-treatment with wogonin ameliorated the nephrotoxicity indices, oxidative stress, inflammation, and apoptosis induced by cisplatin. Also, wogonin up-regulated PPAR-γ expression. The involvement of Wnt/ß-catenin pathway was debatable; however, our findings showed that it was significantly induced by cisplatin. Wogonin pre-treatment markedly attenuated Wnt/ß-catenin pathway. Collectively, these findings imply that wogonin is a promising nephroprotective agent that improves the therapeutic index of cisplatin via reducing oxidative stress, inflammation as well as inducing PPAR-γ. Also, Wnt/ß-catenin pathway is partially involved in the pathogenesis of cisplatin nephrotoxicity.


Subject(s)
Apoptosis/drug effects , Cisplatin/toxicity , Flavanones/pharmacology , PPAR gamma/metabolism , Protective Agents/pharmacology , Wnt Signaling Pathway/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Animals , Flavanones/therapeutic use , Glutathione/metabolism , Inflammation/prevention & control , Interleukin-1beta/analysis , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis , Up-Regulation/drug effects
16.
Drug Discov Ther ; 12(2): 58-67, 2018.
Article in English | MEDLINE | ID: mdl-29760339

ABSTRACT

Liver fibrosis results from chronic inflammation that precipitates excessive accumulation of extracellular matrix. Oxidative stress is involved in its pathogenesis. This study aimed to elucidate the potential antifibrotic effect of the NADPH oxidase (NOX) inhibitor, apocynin against concanavalin A (ConA)-induced immunological model of liver fibrosis, and to investigate the ability of the antioxidant, alpha-lipoic acid (α-LA) to potentiate this effect. Rats were treated with apocynin and/or α-LA for six weeks. Hepatotoxicity indices, oxidative stress, insulin, NOXs, inflammatory and liver fibrosis markers were assessed. Treatment of animals with apocynin and α-LA significantly ameliorated the changes in liver functions and histopathological architecture induced by ConA. Liver fibrosis induced by ConA was evident where alpha-smooth muscle actin and transforming growth factor- beta1 were elevated, which was further confirmed by Masson's trichrome stain and increased hydroxyproline. Co-treatment with apocynin and α-LA significantly reduced their expression. Besides, apocynin and α-LA significantly ameliorated oxidative stress injury evoked by ConA, as evidenced by enhancing reduced glutathione content, antioxidant enzymes activities and decreasing lipid peroxides. ConA induced a significant elevation in serum insulin level and inflammatory markers; tumor necrosis factor-alpha, interleukin-6 and nuclear factor kappa b. Furthermore, the mRNA tissue expression of NOXs 1 and 4 was found to be elevated in the ConA group. All these elevations were significantly reduced by apocynin and α-LA co-treatment. These findings indicate that using apocynin and α-LA in combination possess marked antifibrotic effects, and that NOX enzymes are partially involved in the pathogenesis of ConA-induced liver fibrosis.


Subject(s)
Acetophenones/administration & dosage , Concanavalin A/adverse effects , Liver Cirrhosis/drug therapy , NADPH Oxidase 1/genetics , NADPH Oxidase 4/genetics , Thioctic Acid/administration & dosage , Acetophenones/pharmacology , Animals , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Gene Expression Regulation, Enzymologic/drug effects , Humans , Liver/physiopathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/enzymology , Liver Function Tests , Male , NADPH Oxidase 1/antagonists & inhibitors , NADPH Oxidase 4/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Thioctic Acid/pharmacology
17.
Mol Neurobiol ; 55(8): 6572-6588, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29327204

ABSTRACT

All current treatments of Parkinson's disease (PD) focus on enhancing the dopaminergic effects and providing symptomatic relief; however, they cannot delay the disease progression. Filgrastim, a recombinant methionyl granulocyte colony-stimulating factor, demonstrated neuroprotection in many neurodegenerative and neurological diseases. This study aimed to assess the neuroprotective effects of filgrastim in rotenone-induced rat model of PD and investigate the potential underlying mechanisms of filgrastim actions. The effects of two doses of filgrastim (20 and 40 µg/kg) on spontaneous locomotion, catalepsy, body weight, histology, and striatal dopamine (DA) content, as well as tyrosine hydroxylase (TH) and α-synuclein expression, were evaluated. Then, the effective dose was further tested for its potential anti-inflammatory, neurotrophic, and antiapoptotic effects. Filgrastim (40 µg/kg) prevented rotenone-induced motor deficits, weight reduction, striatal DA depletion, and histological damage. Besides, it significantly inhibited rotenone-induced decrease in TH expression and increase in α-synuclein immunoreactivity in the midbrains and striata of the rats. These effects were associated with reduction of rotenone-induced neuroinflammation, apoptosis, and brain-derived neurotrophic factor depletion. Collectively, these results suggest that filgrastim might be a good candidate for management of PD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Filgrastim/pharmacology , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/pathology , Animals , Body Weight , Corpus Striatum/pathology , Humans , Inflammation/pathology , Male , Mesencephalon/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Parkinson Disease/physiopathology , Rats, Wistar , Rotenone , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism , bcl-2-Associated X Protein/metabolism
18.
Sci Rep ; 6: 32733, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27612096

ABSTRACT

Sorafenib is the only chemotherapeutic agent currently approved for unresectable hepatocellular carcinoma (HCC). However, poor response rates have been widely reported. Indole-3-carbinol (I3C) is a potential chemopreventive phytochemical. The present study aimed to explore the potential chemomodulatory effects of I3C on sorafenib in HCC cells as well as the possible underlying mechanisms. I3C exhibited a greater cytotoxicity in HepG2 cells compared to Huh-7 cells (p < 0.0001). Moreover, the co-treatment of HepG2 cells with I3C and sorafenib was more effective (p = 0.002). Accordingly, subsequent mechanistic studies were carried on HepG2 cells. The results show that the ability of I3C to enhance sorafenib cytotoxicity in HCC cells could be partially attributed to increasing the apoptotic activity and decreasing the angiogenic potentials. The combination had a negative effect on epithelial-mesenchymal transition (EMT). Increased NOX-1 expression was also observed which may indicate the involvement of NOX-1 in I3C chemomodulatory effects. Additionally, the combination induced cell cycle arrest at the G0/G1 phase. In conclusion, these findings provide evidence that I3C enhances sorafenib anti-cancer activity in HCC cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Indoles/pharmacology , Liver Neoplasms/metabolism , NADPH Oxidase 1/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Niacinamide/pharmacology , Sorafenib
19.
Br J Pharmacol ; 173(22): 3248-3260, 2016 11.
Article in English | MEDLINE | ID: mdl-27590029

ABSTRACT

BACKGROUND AND PURPOSE: Liver fibrosis is one of the leading causes of morbidity and mortality worldwide with very limited therapeutic options. Given the pivotal role of activated hepatic stellate cells in liver fibrosis, attention has been directed towards the signalling pathways underlying their activation and fibrogenic functions. Recently, the hedgehog (Hh) signalling pathway has been identified as a potentially important therapeutic target in liver fibrosis. The present study was designed to explore the antifibrotic effects of the potent Hh signalling inhibitor, forskolin, and the possible molecular mechanisms underlying these effects. EXPERIMENTAL APPROACH: Male Sprague-Dawley rats were treated with either CCl4 and/or forskolin for 6 consecutive weeks. Serum hepatotoxicity markers were determined, and histopathological evaluation was performed. Hepatic fibrosis was assessed by measuring α-SMA expression and collagen deposition by Masson's trichrome staining and hydroxyproline content. The effects of forskolin on oxidative stress markers (GSH, GPx, lipid peroxides), inflammatory markers (NF-κB, TNF-α, COX-2, IL-1ß), TGF-ß1 and Hh signalling markers (Ptch-1, Smo, Gli-2) were also assessed. KEY RESULTS: Hepatic fibrosis induced by CCl4 was significantly reduced by forskolin, as indicated by decreased α-SMA expression and collagen deposition. Forskolin co-treatment significantly attenuated oxidative stress and inflammation, reduced TGF-ß1 levels and down-regulated mRNA expression of Ptch-1, Smo and Gli-2 through cAMP-dependent PKA activation. CONCLUSION AND IMPLICATIONS: In our model, forskolin exerted promising antifibrotic effects which could be partly attributed to its antioxidant and anti-inflammatory effects, as well as to its inhibition of Hh signalling, mediated by cAMP-dependent activation of PKA.


Subject(s)
Carbon Tetrachloride/antagonists & inhibitors , Colforsin/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Signal Transduction/drug effects , Animals , Colforsin/administration & dosage , Hedgehog Proteins/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley
20.
Sci Rep ; 6: 29857, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417335

ABSTRACT

Nephrotoxicity associated with the clinical use of the anticancer drug cisplatin is a limiting problem. Thus, searching for new protective measures is required. Indole-3-carbinol is a powerful anti-oxidant, anti-inflammatory and anti-tumor agent. The present study aimed to investigate the potential protective effect of indole-3-carbinol against cisplatin-induced acute nephrotoxicity in rats. Rats were pre-treated with 20 mg/kg indole-3-carbinol orally before giving cisplatin (7 mg/kg). Cisplatin-induced acute nephrotoxicity was demonstrated where relative kidney weight, BUN and serum creatinine were significantly increased. Increased oxidative stress was evident in cisplatin group where GSH and SOD tissue levels were significantly depleted. Also, lipid peroxidation and NOX-1 were increased as compared to the control. Additionally, renal expression of pro-inflammatory mediators was induced by cisplatin. Cisplatin-induced cell death was shown by increased caspase-3 and decreased expression of EGF, IGF-1 and IGF-1 receptor. Nephrotoxicity, oxidative stress, inflammation and apoptotic effects induced by cisplatin were significantly ameliorated by indole-3-carbinol pre-treatment. Besides, the role of CGRP in cisplatin-induced nephrotoxicity was explored. Furthermore, cisplatin cytotoxic activity was significantly enhanced by indole-3-carbinol pre-treatment in vitro. In conclusion, indole-3-carbinol provides protection against cisplatin-induced nephrotoxicity. Also, reduced expression of CGRP may play a role in the pathogenesis of cisplatin-induced renal injury.


Subject(s)
Acute Kidney Injury/drug therapy , Calcitonin Gene-Related Peptide/genetics , Cisplatin/adverse effects , Indoles/administration & dosage , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Calcitonin Gene-Related Peptide/metabolism , Cisplatin/administration & dosage , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...