Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Iran J Basic Med Sci ; 26(3): 316-325, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36865037

ABSTRACT

Objectives: Autism is a complicated neurodevelopmental disorder characterized by social interaction deficiencies, hyperactivity, anxiety, communication disorders, and a limited range of interests. The zebrafish (Danio rerio) is a social vertebrate used as a biomedical research model to understand social behavior mechanisms. Materials and Methods: After spawning, the eggs were exposed to sodium valproate for 48 hr, after which the eggs were divided into eight groups. Except for the positive and control groups, there were six treatment groups based on oxytocin concentration (25, 50, and 100 µM) and time point (24 and 48 hr). Treatment was performed on days 6 and 7, examined by labeling oxytocin with fluorescein-5-isothiocyanate (FITC) and imaging with confocal microscopy and the expression levels of potential genes associated with the qPCR technique. Behavioral studies, including light-dark background preference test, shoaling behavior, mirror test, and social preference, were performed on 10, 11, 12, and 13 days post fertilization (dpf), respectively. Results: The results showed that the most significant effect of oxytocin was at the concentration of 50 µM and the time point of 48 hr. Increased expression of shank3a, shank3b, and oxytocin receptor genes was also significant at this oxytocin concentration. Light-dark background preference results showed that oxytocin in the concentration of 50 µM significantly increased the number of crosses between dark and light areas compared with valproic acid (positive group). Also, oxytocin showed an increase in the frequency and time of contact between the two larvae. We showed a decrease in the distance in the larval group and an increase in time spent at a distance of one centimeter from the mirror. Conclusion: Our findings showed that the increased gene expression of shank3a, shank3b, and oxytocin receptors improved autistic behavior. Based on this study some indications showed that oxytocin administration in the larval stage could significantly improve the autism-like spectrum.

2.
Mini Rev Med Chem ; 23(13): 1390-1411, 2023.
Article in English | MEDLINE | ID: mdl-36515022

ABSTRACT

A class of organic chemicals known as polychlorinated biphenyls (PCBs) consists of chlorine, hydrogen, and carbon atoms. High boiling points, chemical stability, non-flammability, and insulating properties have enabled them to be used in various industries. Because of their high toxicity, PCBs were one of the first industrial compounds to be banned from production. These compounds have high-fat solubility with bioaccumulation and biomagnification properties in the environment, food chain, and individuals. Hence, they may have an impact not only on individual organisms but ultimately on whole ecosystems. The main sources of PCB exposure are food and environmental pollutants. In the toxicology of PCBs, oxidative stress plays the most influential function. The induction of CYP1A1 due to the high affinity of PCBs for aryl hydrocarbon receptors is considered a trigger for oxidative stress. Production of reactive oxygen species and depletion of glutathione occur due to phase Ⅰ and Ⅱ metabolism, respectively. Thus, cellular redox balance may be disrupted in the presence of PCBs and their metabolites. Chronic and long-term exposure to these compounds can often lead to life-threatening diseases, like diabetes, obesity, cardiovascular and neurological diseases, cancer, and reproductive and endocrine disorders. We present the current knowledge of the routes of PCB exposure and bioaccumulation, the outlook regarding environmental and food safety, the potential role of PCBs in various diseases, the principal mechanisms responsible for PCB toxicity, and the main detection techniques used for PCBs.


Subject(s)
Neoplasms , Polychlorinated Biphenyls , Humans , Polychlorinated Biphenyls/toxicity , Polychlorinated Biphenyls/chemistry , Polychlorinated Biphenyls/metabolism , Ecosystem , Environmental Monitoring , Food Safety
3.
Front Endocrinol (Lausanne) ; 13: 1022989, 2022.
Article in English | MEDLINE | ID: mdl-36303864

ABSTRACT

Background: Diabetes-induced reproductive complications can lead to subfertility and infertility, raising the need to protect reproductive organs. There are limited medications used to improve reproductive health in diabetic patients. Melatonin, mainly produced by the pineal gland, may improve diabetes-associated reproductive complications through various mechanisms and may be a preferred candidate to protect the reproductive system. The present review aims to elucidate the underlying mechanisms of melatonin's effect on the reproductive system adversely affected by diabetes mellitus (DM). Methods: A comprehensive systematic literature electronic search was done using the PRISMA guidelines. Web of Science, PubMed, Embase, and Scopus were searched for publications up to June 2022. Search terms were selected based on the study purpose and were explored in titles and abstracts. After screening, out of a total of 169 articles, 14 pertinent articles were included based on our inclusion and exclusion criteria. Results: The results of studies using rats and mice suggest that DM adversely affects reproductive tissues, including testes and epididymis, prostate, corpus cavernosum, and ovary leading to alterations in histological and biochemical parameters compared to the normal groups. Treatment with melatonin improves oxidative stress, blocks apoptosis induced by endoplasmic reticulum stress and caspase activation, reduces pro-inflammation cytokines, and enhances steroidogenesis. Conclusion: Melatonin exerted a protective action on the impaired reproductive system in in-vivo and in-vitro models of DM. The topic has to be followed up in human pregnancy cases that will need more time to be collected and approved.


Subject(s)
Diabetes Mellitus , Melatonin , Male , Female , Humans , Rats , Mice , Animals , Melatonin/pharmacology , Melatonin/therapeutic use , Oxidative Stress , Antioxidants/pharmacology , Antioxidants/therapeutic use , Reproduction/physiology , Diabetes Mellitus/drug therapy
4.
Curr Pharm Des ; 28(22): 1843-1853, 2022.
Article in English | MEDLINE | ID: mdl-35570565

ABSTRACT

BACKGROUND: Various anticancer drugs are effective therapeutic agents for cancer treatment; however, they cause severe toxicity in body organs. Cardiotoxicity is one of the most critical side effects of these drugs. Based on various findings, turmeric extract has positive effects on cardiac cells. OBJECTIVE: This study aims to evaluate how curcumin, as the main component of turmeric, may affect chemotherapy- induced cardiotoxicity. METHODS: A database search was performed up to April 2021 using "curcumin OR turmeric OR Curcuma longa" and "chemotherapy-induced cardiac disease", including their equivalents and similar terms. After screening the total articles obtained from the electronic databases, 25 relevant articles were included in this systematic review. RESULTS: The studies demonstrate lower body weight and increased mortality rates due to doxorubicin administration. Besides, cancer therapeutic agents induced various morphological and biochemical abnormalities compared to the non-treated groups. Based on most of the obtained results, curcumin at nontoxic doses can protect the cardiac cells mainly through modulating antioxidant capacity, regulation of cell death, and antiinflammatory effects. Nevertheless, according to a minority of findings, curcumin increases the susceptibility of the rat cardiomyoblast cell line (H9C2) to apoptosis triggered by doxorubicin. CONCLUSION: According to most nonclinical studies, curcumin could potentially have cardioprotective effects against chemotherapy-induced cardiotoxicity. However, based on limited, contradictory findings demonstrating the function of curcumin in potentiating doxorubicin-induced cardiotoxicity, well-designed studies are needed to evaluate the safety and effectiveness of treatment with new formulations of this compound during cancer therapy.


Subject(s)
Antineoplastic Agents , Curcumin , Animals , Apoptosis , Cardiotoxicity , Curcuma , Doxorubicin , Rats
5.
Toxicol Mech Methods ; 32(4): 288-301, 2022 May.
Article in English | MEDLINE | ID: mdl-34711111

ABSTRACT

Aluminum phosphide (AlP) poisoning is common in many countries responsible for high mortality. The heart is the main target organ in AlP poisoning. Several studies have reported the beneficial effects of cannabidiol (CBD) in reducing heart injuries. This study aimed to investigate the possible protective effect of CBD on cardiac toxicity caused by AlP poisoning. Study groups included almond oil, normal saline, sole CBD (100 µg/kg), AlP (11.5 mg/kg), and four groups of AlP + CBD (following AlP gavage, CBD administrated at doses of 5, 25, 50, and 100 µg/kg via intravenous (iv) injection). Thirty minutes after AlP treatment, an electronic cardiovascular device (PowerLab) was used to record electrocardiographic (ECG) changes, heart rate (HR), and blood pressure (BP) for three hours. Cardiac tissue was examined for the activities of mitochondrial complexes, ADP/ATP ratio, the release of cytochrome C, mitochondrial membrane potential (MMP), apoptosis, oxidative stress parameter, and cardiac biomarkers at 12 and 24 hours time points. AlP administration caused abnormal ECG, decreased HR, and BP. AlP also significantly reduced mitochondrial complex I and IV activity and ADP/ATP ratio. The level of cytochrome C release, apoptosis, oxidative stress, and cardiac biomarkers was considerably increased by AlP, which was compensated following CBD administration. CBD was able to improve hemodynamic function to some extent in AlP poisoned rats. CBD restored ATP levels and mitochondrial function and decreased oxidative damage and thus, prevented the heart cells from entering the apoptotic stage. Further clinical trials are needed to explore any possible benefits of CBD in AlP-poisoned patients.


Subject(s)
Cannabidiol , Phosphines , Animals , Cannabidiol/toxicity , Electrocardiography , Heart Rate , Humans , Mitochondria , Phosphines/toxicity , Rats , Rats, Wistar
6.
Drug Res (Stuttg) ; 72(2): 109-118, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34788887

ABSTRACT

BACKGROUND: Aluminum phosphide (AlP) toxicity is associated with a high risk of death due to heart, liver, and kidney failure as the target organs. Phosphine gas released due to the ingestion is the main factor involved in the multi-organ failure with various mechanisms. Levosimendan (LEV) is a calcium sensitizer with a pleiotropic effect on multiple organs. This study aimed to investigate whether LEV can alleviate AlP-induced nephrotoxicity in the rat model. METHOD: Six groups included control group (almond oil only), sole LEV group (48 µg/kg), AlP group (LD50=10 µg/kg), and the poisoned groups treated with LEV at doses of 12, 24, and 48 µg/kg 30 min after AlP gavage. After 24 hours of treatment, serum and kidney samples were taken for biochemical and histopathological analyses. RESULT: Biochemical analysis of the AlP group showed that the activity of complexes I, II, and IV was significantly reduced, while the levels of lipid peroxidation (LPO) and reactive oxygen species (ROS), lactate, and myeloperoxidase (MPO) activity significantly increased. Also, AlP reduced live renal cells and elevated necrosis. However, the levels of serum creatinine and blood urea nitrogen were not affected by the poisoning. LEV co-treatment could increase mitochondrial complex activity and reduce MPO activity, LPO, ROS, and lactate levels. Additionally, the histopathological analysis showed the detrimental effects of AlP on kidney tissue, which was mitigated by LEV administration. CONCLUSION: Our findings showed that LEV can potentially improve oxidative stress, imbalance in the redox status, necrosis, and pathological injuries in kidney tissue following AlP-poisoning.


Subject(s)
Heart , Oxidative Stress , Animals , Kidney , Phosphines , Rats , Reactive Oxygen Species , Simendan
7.
Hum Exp Toxicol ; 40(12_suppl): S381-S396, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34569344

ABSTRACT

Aluminum phosphide (AlP) poisoning can be deadly in most cases targeting the heart. To overcome AlP toxicity, exenatide has been studied in the present study due to its pleiotropic effects on cardiac damages. In this study, the rats were exposed to LD50 of AlP (10 mg/kg) by gavage, and exenatide at doses (0.05, 0.1, and 0.2 mg/kg) injected intraperitoneally 30 min after poisoning. The cardiac parameters including heart rate (HR), blood pressure (BP), QRS, corrected QT (QTc), and ST were monitored for 180 min. Blood glucose level was measured in the study groups 30 min after exenatide injection. Evaluation of biochemical parameters including mitochondrial complexes I, II, and IV activities, adenosine diphosphate (ADP)/adenosine triphosphate (ATP) ratio, malondialdehyde (MDA), apoptosis, lactate, troponin I, and brain natriuretic peptide (BNP) was done on heart tissues after 12 and 24 h. Additionally, the tissues were analyzed for any pathological damages including necrosis, hemorrhage, or hyperemia 24 h post-treatment. Our results showed that AlP-induced HR, BP, and electrocardiographic changes were improved by exenatide at all doses. The blood glucose levels of poisoned animals reached control levels after exenatide treatment. Besides, treatment with exenatide at all doses improved complexes I and IV activity, ADP/ATP ratio, and apoptosis. Malondialdehyde, lactate, troponin I, and BNP levels were also diminished after exenatide co-treatment in poisoned animals. On the other hand, administration of exenatide doses improved the histopathology of AlP-induced tissues. Based on our findings, exenatide has a protective effect against phosphine-induced cardiotoxicity in an almost dose-dependent way. However, further investigations are needed on the potential clinical use of exenatide in this poisoning.


Subject(s)
Aluminum Compounds/toxicity , Blood Pressure/drug effects , Electrocardiography , Exenatide/pharmacology , Heart Rate/drug effects , Incretins/pharmacology , Phosphines/toxicity , Animals , Blood Glucose/drug effects , Dose-Response Relationship, Drug , Exenatide/administration & dosage , Lethal Dose 50 , Lipid Peroxidation , Male , Random Allocation , Rats , Rats, Wistar
8.
Toxicol Mech Methods ; 31(9): 631-643, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34219611

ABSTRACT

Aluminum phosphide (AlP) causes serious poisoning in which severe cardiac suppression is the significant lethal consequence. According to evidence, levosimendan can exert outstanding cardiac support and protection in different pathological conditions. This study aimed to investigate the mechanisms by which levosimendan may alleviate cardiovascular toxicity due to AlP intoxication in the rat model. The groups included control group (normal saline only), sole levosimendan groups (12, 24, 48 µg/kg), AlP group (10 mg/kg), and AlP + levosimendan groups receiving 12, 24, 48 µg/kg levosimendan intraperitoneally 30 min after AlP administration. Electrocardiographic (ECG) parameters (QRS and PR duration and ST height), heart rate, and blood pressure were monitored for 180 minutes. Also, after 24 h of poisoning, echocardiography was applied to assess left ventricle function. Evaluation of the biochemical parameters in heart tissue, including mitochondrial complexes I, II, IV activity, ADP/ATP ratio, the rate of apoptosis, malondialdehyde (MDA), lactate, and troponin I levels, were done after 12 and 24 h. AlP-induced ECG abnormalities (PR duration and ST height), reduction in heart rate, blood pressure, cardiac output, ejection fraction, and stroke volume were improved by levosimendan administration. Besides, levosimendan significantly improved complex IV activity, the ADP/ATP ratio, apoptosis, MDA, lactate, and troponin I level following AlP-poisoning. Our results suggest that levosimendan might alleviate AlP-induced cardiotoxicity by modulating mitochondrial activity and improving cardiac function. However, the potential clinical use of levosimendan in this toxicity needs more investigations.


Subject(s)
Echocardiography , Electrocardiography , Animals , Phosphines , Rats , Rats, Wistar , Simendan
9.
Food Chem Toxicol ; 154: 112347, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34139304

ABSTRACT

BACKGROUND: Aluminum phosphide (AlP) causes severe cardiotoxicity. Taurine has been chosen for the present study because of its positive known effects on cardiac injuries. METHOD: To evaluate AlP-induced cardiotoxicity, the animals were divided into seven groups, including the control group, the taurine group (500 mg/kg), AlP with LD50 dose, AlP + taurine 20, 50, 100, and 200 mg/kg group. To assess cardiac hemodynamic parameters, Wistar rats received taurine intraperitoneally 60 min after AlP gavage. Cardiac hemodynamic parameters were evaluated for 180 min. To study biochemical parameters, 24 h after AlP treatment, the animals were sacrificed, and heart tissues were collected. RESULT: ECG, BP, and HR abnormalities of AlP poisoning were improved by taurine treatment. AlP induced biochemical alterations including complexes I and IV activities, the ADP/ATP ratio, mitochondrial membrane potential, cytochrome C release, and oxidative stress biomarkers ameliorated by taurine. Moreover, taurine improved apoptosis, as well as lessened CK-MB and troponin I levels. Also, there were no significant changes between taurine 500 mg/kg and the control group in tests. CONCLUSION: The present findings showed that taurine could be a possible candidate for AlP cardiotoxicity treatment via the effect on mitochondrial electron transfer chain and maintaining intracellular ATP balance.


Subject(s)
Aluminum Compounds/toxicity , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Phosphines/toxicity , Taurine/therapeutic use , Animals , Blood Pressure/drug effects , Cardiotoxicity/metabolism , Creatine Kinase/metabolism , Electrocardiography/drug effects , Electron Transport Chain Complex Proteins/metabolism , Heart/drug effects , Heart Rate/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Myocardium/enzymology , Oxidative Stress/drug effects , Rats, Wistar , Troponin I/metabolism
10.
Life Sci ; 265: 118813, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33275984

ABSTRACT

AIMS: Although chemotherapeutic agents have highly beneficial effects against cancer, they disturb the body's normal homeostasis. One of the critical side effects of chemotherapeutic agents is their deleterious effect on the cardiac system, which causes limitations of their clinical usage. Taurine constitutes more than 50% of the amino acids in the heart. The use of taurine might prevent chemotherapy-induced cardiotoxicity. This systematic study aims to evaluate the protective role of taurine against cardiotoxicity induced by chemotherapy. METHODS: A systematic search was performed in databases up to November 2020, and the review is designed on PRISMA guidelines. The search keywords were selected based on our study target and were searched in the title and abstract. After the consecutive screening, out of a whole of 94 articles, 8 articles were included according to our inclusion and exclusion criteria. KEY FINDINGS: According to the study results, chemotherapy decreases body and heart weight and increases mortality. Also, it induces some biochemical and histological changes compared to the control group. By co-administration of taurine with chemotherapy, alterations returned near to the average level. These protective effects of taurine are mediated through anti-oxidant, anti-inflammatory, and anti-apoptotic properties. SIGNIFICANCE: Based on evaluated non-clinical studies, taurine ameliorates chemotherapy-induced cardiotoxicity, but its possible interaction with the efficacy of anti-cancer medicines that mostly act through induction of oxidants remains to be elucidated in the future. This needs conducting well-designed studies to assess the effectiveness and safety of this combination simultaneously.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/prevention & control , Taurine/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antioxidants/administration & dosage , Antioxidants/pharmacology , Apoptosis/drug effects , Cardiotoxicity/etiology , Humans , Neoplasms/drug therapy , Taurine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...