Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Balkan Med J ; 41(3): 193-205, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700358

ABSTRACT

Background: Paclitaxel (PAX) is a widely used chemotherapy drug for various cancer types but often induces significant toxicity in multiple organ systems. Silymarin (SIL), a natural flavonoid, has shown therapeutic potential due to its multiple benefits. Aims: To evaluate the therapeutic efficacy of SIL in mitigating liver and kidney damage induced by PAX in rats, focusing on oxidative stress, inflammation, and apoptosis pathways. Study Design: Experimental animal model. Methods: The study included 28 male Wistar rats aged 12-14 weeks weighing 270-300 g. The rats were divided into four groups: control, SIL, PAX, and PAX + SIL, with seven in each group. The rats received intraperitoneal (i.p.) injections at a dose of 2 mg per kilogram of body weight of PAX for 5 successive days, followed by oral gavage with 200 mg/kg body mass of SIL for 10 uninterrupted days. We examined the effect of SIL on specific serum biochemical parameters using an autoanalyzer and rat-specific kits. The spectrophotometric methods was used to investigate oxidative stress indicators in kidney and liver tissues. Aquaporin-2 (AQP-2), B-cell lymphoma-2 (Bcl-2), cysteine aspartate-specific protease-3 (caspase-3), interleukin-6 (IL-6), nuclear factor kappa B (NF-κB), and streptavidin-biotin staining were used to assess immunoreactivity in PAX-induced liver and kidney injury models. Results: SIL treatment significantly reduced serum levels of alanine aminotransferase, aspartate aminotransferase, creatinine, urea, and C-reactive protein, indicating its effectiveness in treating PAX-induced liver and kidney injury. SIL treatment significantly reduced oxidative stress by increasing essential antioxidant parameters, such as superoxide dismutase, catalase, glutathione peroxidase, and glutathione. It also reduced malondialdehyde levels in liver and kidney tissues of SIL-PAX groups (p < 0.05). SIL administration reduced NF-κB, caspase-3, and IL-6 expression while increasing Bcl-2 and AQP2 levels in liver and kidney tissues of rats treated with SIL and PAX (p < 0.05). Conclusion: Our findings indicate the potential of SIL to alleviate PAX-induced liver and kidney damage in rats by reducing oxidative stress, inflammation, and apoptotic processes.


Subject(s)
Apoptosis , Inflammation , Oxidative Stress , Paclitaxel , Rats, Wistar , Silymarin , Animals , Oxidative Stress/drug effects , Rats , Male , Apoptosis/drug effects , Inflammation/drug therapy , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Silymarin/pharmacology , Silymarin/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Liver/drug effects , Kidney/drug effects , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology
2.
Cardiovasc Toxicol ; 24(5): 519-526, 2024 May.
Article in English | MEDLINE | ID: mdl-38622332

ABSTRACT

Inflammation plays a key role in the pathogenesis of the coronary slow flow phenomenon (CSFP). The newly developed inflammatory marker, pan-immune-inflammation value (PIV), is associated with adverse cardiovascular events. This study investigated the predictive value of PIV for diagnosing CSFP in comparison to other inflammation-based markers. A total of 214 patients, 109 in the CSFP group and 105 in the normal coronary flow (NCF) group, were retrospectively included in the study. Coronary flow was calculated using the Thrombolysis in Myocardial Infarction frame count method. In addition to PIV, other inflammatory markers such as neutrophil-lymphocyte ratio, platelet-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) were calculated for the patients. The average age of patients was 50.3 ± 8.4, with a male ratio of 55.1%. Compared to the NCF group, patients in the CSFP group had higher levels of hyperlipidemia, glucose, triglyceride, NLR, PLR, SII, and PIV, while their high-density lipoprotein cholesterol (HDL-C), was lower (p < 0.05). Logistic regression analysis demonstrated that HDL-C, glucose, triglyceride, and PIV were independent predictor factors for CSFP (p < 0.05). PIV is a strong and independent predictor factor for CSFP and superior in predicting CSFP compared to other inflammatory markers.


Subject(s)
Biomarkers , Coronary Circulation , Inflammation Mediators , No-Reflow Phenomenon , Predictive Value of Tests , Humans , Male , Female , Middle Aged , No-Reflow Phenomenon/blood , No-Reflow Phenomenon/diagnosis , No-Reflow Phenomenon/physiopathology , Retrospective Studies , Biomarkers/blood , Inflammation Mediators/blood , Adult , Inflammation/diagnosis , Inflammation/blood , Inflammation/immunology , Neutrophils/immunology , Lymphocyte Count , Coronary Angiography , Lymphocytes/immunology , Platelet Count , Prognosis , Risk Factors , Blood Platelets/metabolism , Blood Flow Velocity
3.
Iran J Basic Med Sci ; 27(4): 466-474, 2024.
Article in English | MEDLINE | ID: mdl-38419883

ABSTRACT

Objectives: Oxaliplatin (OXL) is a platinum-based chemotherapeutic agent widely used in the treatment of colorectal cancer. Unfortunately, this important drug also causes unwanted side effects such as neuropathy, ototoxicity, and testicular toxicity. This study aimed to investigate the possible protective effects of naringin (NRG) against OXL-induced testicular toxicity in rats. Materials and Methods: In the present study, rats were injected with OXL (4 mg/kg, b.w./day, IP) in 5% dextrose solution 30 min after oral administration of NRG (50 and 100 mg/kg, b.w./day) on the 1st, 2nd, 5th, and 6th days. Then, the rats were sacrificed on the 7th day and the testicular tissues were removed. Results: The results showed that NRG decreased (P<0.001) lipid peroxidation, increased (P<0.001) the activities of superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), and the levels of glutathione (GSH), and also maintained the testis histological architecture and integrity. NRG decreased the levels of apoptosis-related markers such as caspase-3, Bax, and Apaf-1 and increased Bcl2 in the OXL-induced testicular toxicity (P<0.001). In addition, NRG reversed the changes in mRNA transcript levels of oxidative stress, inflammation, and endoplasmic reticulum stress parameters such as Nrf2, HO-1, NQO1, RAGE, NLRP3, MAPK-14, STAT3, NF-κB, IL-1ß, TNF-α, PERK, IRE1, ATF6, and GRP78 in OXL-induced testicular toxicity (P<0.001). Conclusion: Our results demonstrated that NRG can protect against OXL-induced testicular toxicity by enhancing the anti-oxidant defense system and suppressing apoptosis, inflammation, and endoplasmic reticulum stress.

4.
J Biochem Mol Toxicol ; 38(2): e23655, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38348715

ABSTRACT

Bisphenol A (BPA) is a synthetic environmental pollutant widely used in industry, as well as is an endocrine disrupting chemicals and has a toxic effects on heart tissue. The aim of this study is to reveal the cardioprotective effects of 18ß-glycyrretinic acid (GA) against BPA-induced cardiotoxicity in rats. In this study, 40 male rats were used and five different groups (each group includes eight rats) were formed. The rats were applied BPA (250 mg/kg b.w.) alone or with GA (50 and 100 mg/kg b.w.) for 14 days. Rats were killed on Day 15 and heart tissues were taken for analysis. GA treatment decreased serum lactate dehydrogenase and creatine kinase MB levels, reducing BPA-induced heart damage. GA treatment showed ameliorative effects against lipid peroxidation and oxidative stress caused by BPA by increasing the antioxidant enzyme activities (glutathione peroxidase, superoxide dismutase, and catalase) and GSH level of the heart tissue and decreasing the MDA level. In addition, GA showed antiapoptotic effect by increasing Bcl-2, procaspase-3, and -9 protein expression levels and decreasing Bax, cytochrome c, and P53 protein levels in heart tissue. As a result, it was found that GA has cardioprotective effects on heart tissue by exhibiting antioxidant and antiapoptotic effects against heart damage caused by BPA, an environmental pollutant. Thus, it was supported that GA could be a potential cardioprotective agent.


Subject(s)
Benzhydryl Compounds , Environmental Pollutants , Glycyrrhetinic Acid/analogs & derivatives , Heart Injuries , Phenols , Rats , Male , Animals , Antioxidants/pharmacology , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Oxidative Stress , Environmental Pollutants/pharmacology
5.
Exp Ther Med ; 27(2): 82, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38274343

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (COVID-19) represented a global public health crisis and the most significant pandemic in modern times. Transmission characteristics, and the lack of effective antiviral treatment protocol and protective vaccines, pushed healthcare systems, particularly intensive care units (ICUs), to their limits and led to extreme quarantine measures to control the pandemic. It was evident from an early stage that patient stratification approaches needed to be developed to better predict disease progression. In the present study, the predictive value of clinical and blood biomarkers for the outcomes of patients with COVID-19 hospitalized in the ICU were investigated, taking age and sex into consideration. The present study analyzed blood samples from 3,050 patients with COVID-19 hospitalized in the ICU. The analysis revealed that the levels of procalcitonin, N-terminal pro-B-type natriuretic peptide, D-dimer, ferritin, liver enzymes, C-reactive protein and lactate dehydrogenase were increased and were associated with disease progression, resulting in a prolonged hospitalization period and severe COVID-19 related complications. Additionally, significant age and sex disparities among these biomarkers were documented and discussed in specific cases. On the whole, the results of the present study suggest a potential association of the demographic characteristics and blood biomarkers with prolonged hospitalization in the ICU and the mortality of patients with COVID-19.

6.
Arch Pharm (Weinheim) ; 357(2): e2300497, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37972283

ABSTRACT

In this study, the mechanisms by which the enzymes glucose-6-phosphate dehydrogenase (G6PD), 6-phosphogluconate dehydrogenase (6PGD), glutathione reductase (GR), glutathione-S-transferase (GST), and thioredoxin reductase (TrxR) are inhibited by methotrexate (MTX) were investigated, as well as whether the antioxidant morin can mitigate or prevent these adverse effects in vivo and in silico. For 10 days, rats received oral doses of morin (50 and 100 mg/kg body weight). On the fifth day, a single intraperitoneal injection of MTX (20 mg/kg body weight) was administered to generate toxicity. Decreased activities of G6PD, 6PGD, GR, GST, and TrxR were associated with MTX-related toxicity while morin treatment increased the activity of the enzymes. The docking analysis indicated that H-bonds, pi-pi stacking, and pi-cation interactions were the dominant interactions in these enzyme-binding pockets. Furthermore, the docked poses of morin and MTX against GST were subjected to molecular dynamic simulations for 200 ns, to assess the stability of both complexes and also to predict key amino acid residues in the binding pockets throughout the simulation. The results of this study suggest that morin may be a viable means of alleviating the enzyme activities of important regulatory enzymes against MTX-induced toxicity.


Subject(s)
Flavones , Methotrexate , Thioredoxin-Disulfide Reductase , Rats , Animals , Methotrexate/pharmacology , Thioredoxin-Disulfide Reductase/metabolism , Glutathione Transferase/metabolism , Pentose Phosphate Pathway , Structure-Activity Relationship , Glutathione Reductase/metabolism , Body Weight
7.
Metab Brain Dis ; 39(4): 509-522, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38108941

ABSTRACT

Chlorpyrifos (CPF), considered one of the most potent organophosphates, causes a variety of human disorders including neurotoxicity. The current study was designed to evaluate the efficacy of hesperidin (HSP) in ameliorating CPF-induced neurotoxicity in rats. In the study, rats were treated with HSP (orally, 50 and 100 mg/kg) 30 min after giving CPF (orally, 6.75 mg/kg) for 28 consecutive days. Molecular, biochemical, and histological methods were used to investigate cholinergic enzymes, oxidative stress, inflammation, and apoptosis in the brain tissue. CPF intoxication resulted in inhibition of acetylcholinesterase (AChE) and butrylcholinesterase (BChE) enzymes, reduced antioxidant status [superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and glutathione (GSH)], and elevation of malondialdehyde (MDA) levels and carbonic anhydrase (CA) activities. CPF increased histopathological changes and immunohistochemical expressions of 8-OHdG in brain tissue. CPF also increased levels of glial fibrillary acidic protein (GFAP) and nuclear factor kappa B (NF-κB) while decreased levels of nuclear factor erythroid 2-related factor 2 (Nrf-2), heme oxygenase-1 (HO-1) and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α). Furthermore, CPF increased mRNA transcript levels of caspase-3, Bax, PARP-1, and VEGF, which are associated with apoptosis and endothelial damage in rat brain tissues. HSP treatment was found to protect brain tissue by reducing CPF-induced neurotoxicity. Overall, this study supports that HSP can be used to reduce CPF-induced neurotoxicity.


Subject(s)
Apoptosis , Chlorpyrifos , Hesperidin , Neurotoxicity Syndromes , Oxidative Stress , Animals , Oxidative Stress/drug effects , Hesperidin/pharmacology , Hesperidin/therapeutic use , Chlorpyrifos/toxicity , Apoptosis/drug effects , Rats , Male , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Rats, Wistar , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/chemically induced , Insecticides/toxicity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cholinesterase Inhibitors/pharmacology
8.
Behav Sci (Basel) ; 13(9)2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37754057

ABSTRACT

BACKGROUND: Bipolar disorder is a chronic psychiatric disorder with depression and manic episodes. It is one of the leading causes of disease-related disability worldwide. Despite the presence of various alternative drug options for bipolar disorder, some patients do not adequately benefit from the treatment. Therefore, possible underlying mechanisms need to be clarified. Recently, studies on the relationship between bipolar disorder and vitamin D (Vit D) have attracted attention. Although many studies have found an association between depression and Vit D deficiency, little is known about the relationship between manic episodes and Vit D. The aim of this study was to compare Vit D and related metabolites of bipolar manic episodes prior to treatment, bipolar remission after treatment, and healthy control groups. METHODS: This case-control study consisted of 34 bipolar manic episode patients and 34 healthy controls. Disease activity was evaluated with the Hamilton Depression Rating Scale (HAM-D) and Young Mania Rating Scale (YMRS). Firstly, serum 25-hydroxy vitamin D (25-OHD), calcium (Ca) and phosphorus (P) levels of patients in the bipolar manic episode were measured and compared with healthy control. Secondly, serum 25-OHD, Ca and P levels in the euthymic periods of the same patients were measured and compared with healthy control. RESULTS: Bipolar manic episode Vit D levels were lower when compared to healthy controls; while there was no difference in terms of Ca and P levels. There was no significant difference between the bipolar euthymic period patients and the healthy control group in terms of 25-OHD, Ca and P levels. CONCLUSION: Our results demonstrated low serum Vit D concentrations in the acute manic episode of bipolar disorder. Decreased Vit D level may play a role in the onset of the manic episode, or malnutrition and insufficient sunlight during the manic episode may have caused Vit D deficiency. Future studies are needed to exclude potential confounding factors and to compare all mood episodes.

9.
J Pers Med ; 13(9)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37763110

ABSTRACT

OBJECTIVES: Schizophrenia is a devastating and chronic mental disorder that affects 1% of the population worldwide. It is also associated with cognitive dysfunction and cardiovascular risk factors. The aim of this study is to investigate the relationship between cognitive impairment and some inflammatory markers and carotid intima-media thickness (CIMT) in schizophrenia. METHODS: The participants of this study were 51 schizophrenia and 57 healthy controls (HC). The Positive and Negative Syndrome Scale (PANSS) was used for severity of illness, and the Montreal Cognitive Assessment Scale (MoCA) was used for cognitive functioning. The MoCA scores, some biochemical and inflammatory markers, and CIMT were compared between schizophrenia and HC groups. RESULTS: Of the patients with schizophrenia, 11 were women (21.6%), and 40 were men (78.4%). MoCA scores were lower, and levels of NLR, MLR, PLR, SII, CRP, ESR, and CIMT were higher in schizophrenia compared to the HC group (respectively; p < 0.001, p < 0.001, p = 0.035, p = 0.008, p = 0.002, p < 0.001, p < 0.001, p < 0.001). In the schizophrenia group, there was no correlation between MoCA and inflammatory markers. MoCA and CIMT had a significant negative and moderate correlation (p < 0.001). CONCLUSIONS: This is the first study to show the relationship between cognitive impairment and CIMT in schizophrenia. In this study, NLR, MLR, PLR, SII, CRP, and ESR markers were higher in schizophrenia compared to HC, indicating inflammation. Our finding of elevated CIMT in schizophrenia suggests that there may be an atherosclerotic process along with the inflammatory process. The finding of a positive correlation between cognitive impairment and CIMT may be promising for new therapies targeting the atherosclerotic process in the treatment of cognitive impairment.

10.
Mol Biol Rep ; 50(4): 3479-3488, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36781607

ABSTRACT

BACKGROUND: Organ toxicity limits the therapeutic efficacy of methotrexate (MTX), an anti-metabolite therapeutic that is frequently used as an anti-cancer and immunosuppressive medicine. Hepatocellular toxicity is among the most severe side effects of long-term MTX use. The present study unveils new confirmations as regards the remedial effects of morin on MTX-induced hepatocellular injury through regulation of oxidative stress, apoptosis and MAPK signaling. METHODS AND RESULTS: Rats were subjected to oral treatment of morin (50 and 100 mg/kg body weight) for 10 days. Hepatotoxicity was induced by single intraperitoneal injection of MTX (20 mg/kg body weight) on the 5th day. MTX related hepatic injury was associated with increased MDA while decreased GSH levels, the activities of endogen antioxidants (glutathione peroxidase, superoxide dismutase and catalase) and mRNA levels of HO-1 and Nrf2 in the hepatic tissue. MTX treatment also resulted in apoptosis in the liver tissue via increasing mRNA transcript levels of Bax, caspase-3, Apaf-1 and downregulation of Bcl-2. Conversely, treatment with morin at different doses (50 and 100 mg/kg) considerably mitigated MTX-induced oxidative stress and apoptosis in the liver tissue. Morin also mitigated MTX-induced increases of ALT, ALP and AST levels, downregulated mRNA expressions of matrix metalloproteinases (MMP-2 and MMP-9), MAPK14 and MAPK15, JNK, Akt2 and FOXO1 genes. CONCLUSION: According to the findings of this study, morin may be a potential way to shield the liver tissue from the oxidative damage and apoptosis.


Subject(s)
Chemical and Drug Induced Liver Injury , Methotrexate , Rats , Animals , Methotrexate/toxicity , Methotrexate/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , NF-E2-Related Factor 2/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Rats, Wistar , Antioxidants/metabolism , Oxidative Stress , Liver/metabolism , Signal Transduction , Flavonoids/pharmacology , Flavonoids/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Body Weight
11.
Phytother Res ; 37(3): 1115-1135, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36562210

ABSTRACT

Caffeic acid phenethyl ester (CAPE), a main active component of propolis and a flavonoid, is one of the natural products that has attracted attention in recent years. CAPE, which has many properties such as anti-cancer, anti-inflammatory, antioxidant, antibacterial and anti-fungal, has shown many pharmacological potentials, including protective effects on multiple organs. Interestingly, molecular docking studies showed the possibility of binding of CAPE with replication enzyme. In addition, it was seen that in order to increase the binding security of the replication enzyme and CAPE, modifications can be made at three sites on the CAPE molecule, which leads to the possibility of the compound working more powerfully and usefully to prevent the proliferation of cancer cells and reduce its rate. Also, it was found that CAPE has an inhibitory effect against the main protease enzyme and may be effective in the treatment of SARS-CoV-2. This review covers in detail the importance of CAPE in alternative medicine, its pharmacological value, its potential as a cancer anti-proliferative agent, its dual role in radioprotection and radiosensitization, and its use against coronavirus disease 2019 (COVID-19).


Subject(s)
COVID-19 , Phenylethyl Alcohol , Humans , Molecular Docking Simulation , SARS-CoV-2 , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/metabolism , Phenylethyl Alcohol/pharmacology , Caffeic Acids/chemistry , Anti-Inflammatory Agents/pharmacology , Free Radicals
12.
Mol Biol Rep ; 50(1): 433-442, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36344803

ABSTRACT

BACKGROUND: Diclofenac (DF) is a non-steroidal anti-inflammatory drug (NSAID) generally prescribed for the treatment of pain. In spite of the widespread use of DF, hepatotoxicity has been reported after its administration. The current study discloses new evidence as regards of the curative effects of chrysin (CHR) on DF-induced hepatotoxicity by regulating oxidative stress, apoptosis, autophagy, and endoplasmic reticulum (ER) stress. METHODS: The animals were separated into five different groups. Group-I was in control. Group-II received CHR-only (50 mg/kg bw, p.o.) on all 5 days. Group-III received DF-only (50 mg/kg bw, i.p.) on 4th and 5th day. Group-IV received DF (50 mg/kg bw) + CHR (25 mg/kg, bw) and group-V received DF (50 mg/kg, bw) + CHR (50 mg/kg, bw) for 5 days. RESULTS: DF injection was associated with increased MDA while reduced GSH level, activities of superoxide dismutase, glutathione peroxidase, and catalase and mRNA levels of HO-1 and Nrf2 in the liver. DF injection caused apoptosis and autophagy in the liver by up-regulating caspase-3, Bax, LC3A, and LC3B levels and down-regulating Bcl-2. DF also caused ER stress by increasing mRNA transcript levels of ATF-6, IRE1, PERK, and GRP78. Additionally, it was observed that DF administration up-regulated MMP2 and MMP9. However, treatment with CHR at a dose of 25 and 50 mg/kg considerably ameliorated oxidative stress, apoptosis, autophagy, and ER stress in liver tissue. CONCLUSION: Overall, the data of this study indicate that liver damage associated with DF toxicity could be ameliorated by CHR administration.


Subject(s)
Chemical and Drug Induced Liver Injury , Diclofenac , Rats , Animals , Diclofenac/toxicity , Oxidative Stress , RNA, Messenger , Chemical and Drug Induced Liver Injury/drug therapy , Endoplasmic Reticulum Stress , Apoptosis , Autophagy
13.
Drug Chem Toxicol ; 46(3): 546-556, 2023 May.
Article in English | MEDLINE | ID: mdl-35450509

ABSTRACT

Favipiravir is a selective RNA polymerase inhibitor and a broad-spectrum antiviral drug, an important agent used in viral infections, including Ebola, Lassa, and COVID-19. This study aims to evaluate the potential toxicological effects of favipiravir administration on rats' liver and kidney tissues. Favipiravir was applied for five and ten days in the present study. During this period, it was aimed to determine possible toxic effects on the liver and kidney. For this purpose, the impact of favipiravir on liver and kidney tissues were examined using histopathologic and biochemical methods. The present study showed that favipiravir administration led to an elevation in the liver and kidney serum enzymes and oxidative and histopathologic damages. Favipiravir administration caused apoptotic cell death (Caspase-3 and Bcl-2), inflammation (NF-κB and IL-6), and a decrease in renal reabsorption (AQP2) levels. In the evaluation of the findings obtained in this study, it was determined that the favipiravir or metabolites caused liver and kidney damages.


Subject(s)
Amides , Antiviral Agents , Kidney , Liver , Pyrazines , Animals , Rats , Antiviral Agents/pharmacology , Antiviral Agents/toxicity , Aquaporin 2 , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Amides/pharmacology , Amides/toxicity , Pyrazines/pharmacology , Pyrazines/toxicity
14.
Mol Biol Rep ; 49(10): 9641-9649, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36057755

ABSTRACT

BACKGROUND: Methotrexate (MT) is a broadly used chemotherapeutic drug however its clinical use is confronted with several forms of toxicities containing testicular damage. The current study assessed the ameliorative effects of morin on MT-induced testicular damage with the investigation of its mechanism and the potential involvement of oxidative stress, inflammation, apoptosis and endoplasmic reticulum stress in such protection. METHODS: The animals were divided into 5 distinct groups (7 rats in each group). Group 1 was control group, group 2 received MT-only (20 mg/kg bw), group 3 received orally morin-only (100 mg/kg bw), group 4 received MT (20 mg/kg bw) + morin (50 mg/kg bw) and group 5 received MT (20 mg/kg bw) + morin (100 mg/kg). In this study, morin was administered orally for 10 days, while MT was administered intraperitoneally on the 5th day. RESULTS: MT intoxication was linked with augmented MDA while decreased GSH levels, the enzyme activities of glutathione peroxidase, superoxide dismutase, and catalase and mRNA levels of HO-1 and Nrf2 in the testis tissues. MT injection caused inflammation in the testicular tissue via up-regulation of MAPK14, NFκB, TNF-α and IL-1ß. MT application also caused apoptosis and endoplasmic reticulum stress in the testis tissue via increasing mRNA transcript levels of Bax, caspase-3, PERK, IRE1, ATF-6, GRP78 and down-regulation of Bcl-2. CONCLUSION: Treatment with morin at a dose of 50 and 100 mg/kg considerably mitigated oxidative stress, inflammation, apoptosis and endoplasmic reticulum stress in the testicular tissue indicating that testicular damage related to MT toxicity could be modulated by morin administration.


Subject(s)
Mitogen-Activated Protein Kinase 14 , Testis , Activating Transcription Factor 6 , Animals , Antioxidants/metabolism , Caspase 3/metabolism , Catalase/metabolism , Endoplasmic Reticulum Chaperone BiP , Flavones , Glutathione Peroxidase/metabolism , Inflammation/metabolism , Male , Methotrexate/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction , Superoxide Dismutase/metabolism , Testis/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
15.
Chem Biol Interact ; 365: 110073, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-35921949

ABSTRACT

Paclitaxel (PTX) is a widely used chemotherapeutic drug particularly effective against lung, breast, and ovarian cancer, though its usefulness is limited due to its multi-organ toxicity. The mechanisms underlying PTX toxicity are currently not yet known and there are no approved treatments for its control or prevention. This study aimed to investigate whether hesperidin (HSP) had a protective effect on paclitaxel-induced hepatotoxicity and nephrotoxicity from biochemical, and molecular perspectives. The rats were administered PTX 2 mg/kg, b.w. intraperitoneally for the first 5 consecutive days, then 100 or 200 mg/kg b.w. HSP orally for 10 consecutive days. Our results demonstrated that HSP decreased the PTX induced lipid peroxidation, improved the serum hepatic and renal functions (by decreasing the levels of AST, ALT, ALP, urea, and creatinine), and restored the liver and kidney antioxidant armory (SOD, CAT, GPx, and GSH). HSP also significantly reduced mRNA expression levels of NF-κB, TNF-α, IL-1ß, IL-6, MAPK 14, Caspase-3, Bax, LC3A, LC3B, MMP2, and MMP9 whereas caused an increase in levels of Nrf2, HO-1, and Bcl-2 in the kidney and liver of PTX-induced rats. In addition, caspase-3, Bax, and Bcl-2 protein levels were examined by Western blot analysis, and it was determined that HSP decreased caspase-3 and Bax protein levels, but increased Bcl-2 protein levels. The findings of the study suggest that HSP has chemopreventive potential against PTX-induced hepatorenal toxicity plausibly through the attenuation of oxidative stress, inflammation, apoptosis, and autophagy.


Subject(s)
Chemical and Drug Induced Liver Injury , Hesperidin , Animals , Rats , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Hesperidin/pharmacology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Paclitaxel/toxicity , Signal Transduction
16.
Metab Brain Dis ; 37(6): 1931-1940, 2022 08.
Article in English | MEDLINE | ID: mdl-35699857

ABSTRACT

The exposure to bisphenol A (BPA) is inevitable owing to its common use in the production of polycarbonate plastics. Studies to reduce side effects are gaining importance since BPA causes severe toxicities in important tissues such as testes, lungs, brain, liver and kidney. The current study was planned to study ameliorative effect of 18ß-glycyrrhetinic acid (18ß-GA) on BPA induced neurotoxicity. Fourty Wistar albino rats were divided into five equal groups as follows: I-Control group, II-18ß-GA group (100 mg/kg), III- BPA group (250 mg/kg), IV-250 mg/kg BPA + 50 mg/kg 18ß-GA group, V-250 mg/kg BPA + 100 mg/kg 18ß-GA group. BPA intoxication was associated with increased MDA level while reduced GSH concentration, activities of glutathione peroxidase, superoxide dismutase, and catalase. BPA supplementation caused apoptosis in the brain by up-regulating caspase-3 and Bax levels and down-regulating Bcl-2. BPA also caused endoplasmic reticulum (ER) stress by increasing mRNA transcript levels of PERK, IRE1, ATF-6 and GRP78. Additionally, it was observed that BPA administration activated JAK1/STAT1 signaling pathway and levels of TNF-α, NF-κB, p38 MAPK and JNK in the brain. However, co-treatment with 18ß-GA at a dose of 50 and 100 mg/kg considerably ameliorated oxidative stress, inflammation, apoptosis, ER stress and JAK1/STAT1 signaling pathway in brain tissue. Overall, the data of this study indicate that brain damage associated with BPA toxicity could be ameliorated by 18ß-GA administration.


Subject(s)
Endoplasmic Reticulum Stress , Neuroprotective Agents , Animals , Apoptosis , Benzhydryl Compounds , Glycyrrhetinic Acid/analogs & derivatives , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Phenols , Rats , Rats, Wistar , STAT1 Transcription Factor/pharmacology , Signal Transduction
17.
J Biochem Mol Toxicol ; 36(9): e23121, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35670529

ABSTRACT

Oxaliplatin (OXL) is a chemotherapeutic drug used for metastatic and other types of cancer, but it causes peripheral neuropathy as a dose-limiting side effect. Herein, we used the rat model of OXL-induced peripheral neuropathy to demonstrate the protective effects of naringin (NRG) in this neuropathy. In this study, rats were injected with OXL (4 mg/kg, body weight, i.p.) in 5% glucose solution 30 min after oral administration of NRG (50 and 100 mg/kg, body weight) on the 1st, 2nd, 5th, and 6th days. OXL caused sensory and motor neuropathy (as revealed by the hot plate, tail flick, rota-rod, and cold hyperalgesia tests) in the sciatic nerve of rats. Coadministration of oral NRG alleviated OXL-induced sensory and motor neuropathy. Levels of superoxide dismutase, catalase, glutathione peroxidase, nuclear factor erythroid 2-related factor 2, Heme oxygenase-1, nuclear factor-κ B, tumor necrosis factor-α, interleukin-1ß, Bax, Bcl-2, caspase-3, paraoxonase, mitogen-activated protein kinase 14, neuronal nitric oxide synthase (nNOS), acetylcholinesterase, and arginase 2 in the sciatic nerve tissues were assessed by real-time polymerase chain reaction. Moreover, the protein levels of caspase-3, Bax, Bcl-2, intercellular adhesion molecules-1, glial fibrillary acidic protein, and nNOS were examined by Western blot analysis. NRG treatment significantly improved all the above-mentioned parameters and reduced OXL-induced oxidative stress, inflammation, and apoptosis in the sciatic nerve tissue. In conclusion, this study demonstrated that NRG significantly attenuated OXL-induced peripheral neuropathy and might be considered as a new protective agent to prevent the OXL-induced peripheral neuropathy.


Subject(s)
Mitogen-Activated Protein Kinase 14 , Peripheral Nervous System Diseases , Acetylcholinesterase , Animals , Arginase/adverse effects , Aryldialkylphosphatase , Body Weight , Caspase 3 , Catalase/metabolism , Cell Adhesion Molecules , Flavanones , Glial Fibrillary Acidic Protein , Glucose/adverse effects , Glutathione Peroxidase , Heme Oxygenase-1 , Interleukin-1beta , Nitric Oxide Synthase Type I/adverse effects , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control , Protective Agents , Rats , Superoxide Dismutase , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein
18.
Cardiovasc Toxicol ; 22(8): 727-735, 2022 08.
Article in English | MEDLINE | ID: mdl-35606666

ABSTRACT

Excessive fluoride intake has been reported to cause toxicities to brain, thyroid, kidney, liver and testis tissues. Hesperidin (HSP) is an antioxidant that possesses anti-allergenic, anti-carcinogenic, anti-oxidant and anti-inflammatory activities. Presently, the studies focusing on the toxic effects of sodium fluoride (NaF) on heart tissue at biochemical and molecular level are limited. This study was designed to evaluate the ameliorative effects of HSP on toxicity of NaF on the heart of rats in vivo by observing the alterations in oxidative injury markers (MDA, SOD, CAT, GPX and GSH), pro-inflammatory markers (NF-κB, IL-1ß, TNF-α), expressions of apoptotic genes (caspase-3, -6, -9, Bax, Bcl-2, p53, cytochrome c), levels of autophagic markers (Beclin 1, LC3A, LC3B), expression levels of PI3K/Akt/mTOR and cardiac markers. HSP treatment attenuated the NaF-induced heart tissue injury by increasing activities of SOD, CAT and GPx and levels of GSH, and suppressing lipid peroxidation. In addition, HSP reversed the changes in expression of apoptotic (caspase-3, -6, -9, Bax, Bcl-2, p53, cytochrome c), levels of autophagic and inflammatory parameters (Beclin 1, LC3A, LC3B, NF-κB, IL-1ß, TNF-α), in the NaF-induced cardiotoxicity. HSP also modulated the gene expression levels of PI3K/Akt/mTOR signaling pathway and levels of cardiac markers (LDH, CK-MB). Overall, these findings reveal that HSP treatment can be used for the treatment of NaF-induced cardiotoxicity.


Subject(s)
Heart Diseases , Hesperidin , Animals , Apoptosis , Beclin-1/metabolism , Beclin-1/pharmacology , Cardiotoxicity , Caspase 3/metabolism , Cytochromes c/metabolism , Cytochromes c/pharmacology , Heart Diseases/chemically induced , Heart Diseases/drug therapy , Hesperidin/pharmacology , Hesperidin/therapeutic use , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , NF-kappa B/metabolism , Oxidative Stress , Phosphatidylinositol 3-Kinases/adverse effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Sodium Fluoride/toxicity , Superoxide Dismutase/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
19.
Neurotoxicology ; 90: 197-204, 2022 05.
Article in English | MEDLINE | ID: mdl-35413380

ABSTRACT

Fluoride is an element with toxic properties and has been proven to have some adverse effects on many soft tissues, including brain tissue. This study aims to evaluate the protective effects of hesperidin on sodium fluoride (NaF)-induced neurotoxicity in rats by biochemical and molecular methods. The animals were randomly divided into five groups of seven rats each as Control, hesperidin, NaF (600 ppm), NaF + hesperidin (100 mg/kg, b.w.), and NaF + hesperidin (200 mg/kg, b.w.), respectively; orally for two weeks. Hesperidin reduced lipid peroxidation and increased activities of SOD, CAT and GPx and levels of GSH in NaF-induced brain tissue. Hesperidin also showed anti-inflammatory and anti-autophagic effects by decreasing levels of NF-κB, IL-1B, TNF-α, Beclin-1, LC3A, and LC3B in NaF-induced brain tissue. Moreover, hesperidin was able to down-regulate the mRNA transcript levels of apoptosis and endoplasmic reticulum stress markers such as caspase-3, Bax, Bcl-2, PERK, IRE1, ATF6, and GRP78 in NaF-induced neurotoxicity. Hesperidin also reduced the adverse effects caused by NaF by modulating the PI3K/Akt/mTOR signaling pathway. These results demonstrate that hesperidin exhibits neuroprotective effects against NaF-induced neurotoxicity in rats by ameliorating inflammation, apoptosis, autophagy, and endoplasmic reticulum stress.


Subject(s)
Hesperidin , Neurotoxicity Syndromes , Animals , Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Hesperidin/pharmacology , Neuroinflammatory Diseases , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress , Phosphatidylinositol 3-Kinases , Rats , Sodium Fluoride/toxicity
20.
Metab Brain Dis ; 37(4): 1259-1269, 2022 04.
Article in English | MEDLINE | ID: mdl-35316447

ABSTRACT

Cadmium (Cd), is a heavy metal reported to be associated with oxidative stress and inflammation. In this paper, we investigated the possible protective effects of carvacrol against Cd-induced neurotoxicity in rats. Adult male Sprague Dawley rats were treated orally with Cd (25 mg/kg body weight) and with carvacrol (25 and 50 mg/kg body weight) for 7 days. Carvacrol decreased the levels of malondialdehyde (MDA), glial fibrillary acidic protein (GFAP) and monoamine oxidase (MAO), and significantly increased the levels of glutathione (GSH) and activities of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) in brain tissue. Additionally, carvacrol alleviated the in levels of inflammation and apoptosis related proteins involving p38 mitogen-activated protein kinase (p38 MAPK), cyclooxygenase-2 (COX-2), nuclear factor kappa B (NF-κB), B-cell lymphoma-3 (Bcl-3), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), myeloperoxidase (MPO), prostaglandin E2 (PGE2), neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase (iNOS), cysteine aspartate specific protease-3 (caspase-3) and Bcl-2 associated X protein (Bax) in the Cd-induced neurotoxicity. Carvacrol also decreased the mRNA expression of matrix metalloproteinases (MMP9 and MMP13), as well as 8-hydroxy-2'-deoxyguanosine (8 - OHdG) level, a marker of oxidative DNA damage. Collectively, our findings indicated that carvacrol has a beneficial effect in ameliorating the Cd-induced neurotoxicity in the brain of rats.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Animals , Apoptosis , Body Weight , Cadmium/toxicity , Cymenes , Glutathione/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Oxidative Stress , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...