1
|
Muhammad Abdur Rahman H, Javaid S, Ashraf W, Fawad Rasool M, Saleem H, Ali Khan S, Ul-Haq Z, Muhammad Muneeb Anjum S, Ahmad T, Alqahtani F, Ur Rehman A, Imran I. Effects of long-term Ailanthus altissima extract supplementation on fear, cognition and brain antioxidant levels. Saudi Pharm J 2023; 31:191-206. [PMID: 36942273 PMCID: PMC10023549 DOI: 10.1016/j.jsps.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Ailanthus altissima is an indigenous plant known for various remedial properties. The present study aimed to evaluate the neuroprotective potential of methanolic extract Ailanthus altissima (AA) bark as current scientific trend is searching plant for neurodegenerative diseases, worldwide. Methodology In in-vitro experiments, the AA was analyzed for phenols, flavonoids, antioxidative and cholinesterase inhibitory properties with subsequent detailed characterization for secondary metabolites. The in-vivo neurological effects were evaluated in rats through behavioral assessment for anxiety and memory after chronic administration (28 days) of 50-200 mg/kg of AA. At the end of behavior studies, isolated brains were biochemically tested to determine antioxidant enzyme activity. Results AA was found rich in phenols/flavonoids and active in radical scavenging with the presence of 13 secondary metabolites in UHPLC-MS analysis. The AA yielded anxiolytic effects dose-dependently in the open field, light/dark and elevated-plus maze tests as animals significantly (P < 0.05 vs control group) preferred open arena, illuminated zone and exposed arms of maze. Similarly, the animals treated with AA showed significant (P < 0.05 vs amnesic group) increase in spontaneous alternation, discrimination index in y-maze, novel object recognition tests. Further, AA.Cr treated rats showed noticeably shorter escape latencies in Morris water maze tests.In biochemical analysis, the dissected brains AA treated rats showed reduced levels of AChE and malondialdehyde with increased levels of first-line antioxidant enzymes i.e. glutathione peroxidase and superoxide dismutase. These observed biological effects might be attributed to phenols and flavonoids constituents owned by AA. -The in-silico studies showed thatconessine and lophirone J phytocompounds have good blood-brain barrier permeability and interaction with AChE. Conclusion The outcomes of this study validate that bark of Ailanthus altissima might work as a source of bioactive phytochemicals of neuroprotective potential.
Collapse
Affiliation(s)
| | - Sana Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
- Department of Pharmacy, The Women University, Multan 60000, Pakistan
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Hammad Saleem
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore, Pakistan, 75270, Pakistan
| | - Salman Ali Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Third World Center for Science and Technology, H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Syed Muhammad Muneeb Anjum
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore, Pakistan, 75270, Pakistan
| | - Tanveer Ahmad
- Institut pour l’Avancée des Biosciences, Centre de Recherche UGA / INSERM U1209 / CNRS 5309, Université Grenoble Alpes, France
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Corresponding authors.
| | - Anees Ur Rehman
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
- Corresponding authors.
| |
Collapse
|
2
|
Abo-Hiemad HM, Nassar AY, Shatat AR, Mohamed MA, Soliman M, Abdelrady YA, Sayed AM. Protective effect of copper II-albumin complex against aflatoxin B1- induced hepatocellular toxicity: The impact of Nrf2, PPAR-γ, and NF-kB in these protective effects. J Food Biochem 2022; 46:e14160. [PMID: 35338511 DOI: 10.1111/jfbc.14160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 12/29/2022]
Abstract
Copper II-Albumin complex (Cu-II-Albumin complex) is a novel therapeutic target that has been used as anti-inflammatory, antioxidant, and anti-gastrointestinal toxicity. In this study, 40 rats were divided into four groups, normal control (NC), aflatoxicosed group (AF) that received Aflatoxin B1 (AFB1) (50 μg/kg of the AFB1 daily for 3 weeks), AFB1-Cu-II-Albumin prophylactic group (AF/CUC-P) that subjected to intermittent treatment between AFB1 and Cu-II-Albumin complex (0.05 g/kg Cu-II-Albumin complex) day after day for 3 weeks and AFB1-Cu-II-albumin treatment group (AF/CUC-T) that received AFB1 for 3 weeks and Cu-II-albumin complex for another 3 weeks. The hepatocellular protective effect of the Cu-II-albumin complex was assessed by evaluating the liver functions markers, hepatic histopathology, reactive oxygen species (ROS) levels (Nitric Oxide (NO) and malondialdehyde (MDA)), apoptotic genes (caspase-3 and tumor necrosis factor receptor 1 [TNF-R1]) expressions, and serological and molecular biomarkers of hepatocellular carcinoma (histamine and Glucose-Regulated Protein 78 [GRP78], respectively). Our finding showed that Cu-II-Albumin Complex administration had restored liver function, oxidative stress levels, enhanced liver tissue recovery, and reduced the expression of the apoptotic genes of the aflatoxicosed rats. In conclusion, the current study results demonstrated the protective effect of Cu-II-albumin complex against AFB1-induced hepatocellular toxicity. PRACTICAL APPLICATIONS: The protective effect of Cu-II-Albumin Complex against AFB1-induced hepatocellular toxicity by assessing oxidative stress, liver biomarkers, inflammation, and histological changes of liver tissues. The protective mechanism of the Cu-II-albumin complex was also investigated. More clinical studies are required to evaluate the potential of using the Cu-II-albumin complex as a therapeutic agent against hepatocellular toxicity.
Collapse
Affiliation(s)
- Hend M Abo-Hiemad
- Biochemistry Division, Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Ahmed Y Nassar
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed R Shatat
- Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mona A Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mahmoud Soliman
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | | | - Ahmed M Sayed
- Biochemistry Laboratory, Faculty of Science, Chemistry Department, Assiut University, Assiut, Egypt
| |
Collapse
|
3
|
Shang C, Cao Y, Sun C, Li Y. Comparison of the excited-state proton transfer and single electron transfer mechanisms of the natural antioxidant Juglone and its dimer 3,3′-bijuglone. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.113825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
4
|
Umaya SR, Vijayalakshmi YC, Sejian V. Exploration of plant products and phytochemicals against aflatoxin toxicity in broiler chicken production: Present status. Toxicon 2021; 200:55-68. [PMID: 34228958 DOI: 10.1016/j.toxicon.2021.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/30/2021] [Accepted: 06/26/2021] [Indexed: 12/28/2022]
Abstract
Aflatoxins (AFs) are a class of mycotoxins produced by the toxigenic Aspergillus fungi and are common contaminants of foods and feeds. Aflatoxin B1 (AFB1), the most potent aflatoxin, is well characterized to reduce productive performance and mortality in broilers. This exclusive review summarizes the efficacy of various plant products and phytochemicals to counteract AFB1 toxicity in broilers. The biochemical and molecular mode of action of AFB1 to induce liver damage, genotoxicity, immunosuppression and the protective effect of plant products against such mechanisms and their toxic effects are discussed. The link between antioxidant, immunomodulatory and hepatoprotective functions of plant products; oxidative stress and AFB1 macromolecular adducts mediated AFB1 toxicity are covered. Efficacy of Satureja khuzistanica, Zataria multiflora Boiss, Thymus vulgaris, Sauropsus androgynus, Hemidesmus indicus, Leucas aspera, Moringa oleifera, Eclipta alba, Curcuma longa, Silybum marianum, Urtica dioica, and citrus fruit are summarized. The anti-aflatoxic effect of water-soluble substances of wheat, grape seed proanthocyanidin extract and phytochemicals like thymol, carvarol, piperine, transcinnamaldehyde, resveratrol, curcumin, and silymarin are also discussed. Specific plant products and phytochemicals are shown to be effective against AF toxicity in broilers and could represent an important tool to reduce health and economic losses associated with AFB1 exposure.
Collapse
Affiliation(s)
- Suganthi R Umaya
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560 030, Karnataka, India.
| | - Y C Vijayalakshmi
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560 030, Karnataka, India
| | - V Sejian
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560 030, Karnataka, India
| |
Collapse
|
5
|
Hua Z, Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, Li W, Li W, Lu C, Liu Y. Contamination of Aflatoxins Induces Severe Hepatotoxicity Through Multiple Mechanisms. Front Pharmacol 2021; 11:605823. [PMID: 33505311 PMCID: PMC7830880 DOI: 10.3389/fphar.2020.605823] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
Aflatoxins (AFs) are commonly contaminating mycotoxins in foods and medicinal materials. Since they were first discovered to cause “turkey X” disease in the United Kingdom in the early 1960s, the extreme toxicity of AFs in the human liver received serious attention. The liver is the major target organ where AFs are metabolized and converted into extremely toxic forms to engender hepatotoxicity. AFs influence mitochondrial respiratory function and destroy normal mitochondrial structure. AFs initiate damage to mitochondria and subsequent oxidative stress. AFs block cellular survival pathways, such as autophagy that eliminates impaired cellular structures and the antioxidant system that copes with oxidative stress, which may underlie their high toxicities. AFs induce cell death via intrinsic and extrinsic apoptosis pathways and influence the cell cycle and growth via microribonucleic acids (miRNAs). Furthermore, AFs induce the hepatic local inflammatory microenvironment to exacerbate hepatotoxicity via upregulation of NF-κB signaling pathway and inflammasome assembly in the presence of Kupffer cells (liver innate immunocytes). This review addresses the mechanisms of AFs-induced hepatotoxicity from various aspects and provides background knowledge to better understand AFs-related hepatoxic diseases.
Collapse
Affiliation(s)
- Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guangzhi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chenxi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yurong Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwen Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wen Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Weifeng Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Zhu L, Huang C, Yang X, Zhang B, He X, Xu W, Huang K. Proteomics reveals the alleviation of zinc towards aflatoxin B1-induced cytotoxicity in human hepatocyes (HepG2 cells). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 198:110596. [PMID: 32353602 DOI: 10.1016/j.ecoenv.2020.110596] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 05/24/2023]
Abstract
Aflatoxin B1 (AFB1) is a known carcinogen found in contaminated food and designated by the World Health Organization as a class I carcinogenic substance. AFB1 presents with carcinogenicity, teratogenicity, and mutagenicity, and the liver is the human organ most susceptible to AFB1. Zinc (Zn), which is one of the essential nutrient elements that could protect the cells from biological toxins, heavy metals, hydrogen peroxide, metal chelators and radiation, is assessed in this study for its potential to alleviate AFB1-induced cytotoxicity. Samples were divided into three groups, namely CK, AFB1, and AFB1+Zn. Protein expressions were analyzed by two-way electrophoresis combined with flight mass spectrometry, with 41 differentially expressed proteins identified in the results, mainly related to oxidative stress, cell apoptosis, DNA damage, and energy metabolism. Zn was found to regulate the expression of peroxidases (peroxiredoxin-1, peroxiredoxin-5, peroxiredoxin-6) to relieve AFB1-induced oxidative stress. Moreover, Zn could decrease the expression of pro-apoptotic genes (cleaved-caspase-3, caspase-9, and Bax) and increase the expression of anti-apoptotic genes (Bcl-2 and Bcl-xl) to alleviate the cell apoptosis induced by AFB1. In addition, AFB1 reduced intracellular ATP levels, whereas Zn supplementation boosted ATP levels and maintained homeostasis and a steady state of cellular energy metabolism by modulating AMPK-ACC phosphorylation levels, while many zinc finger proteins changed after AFB1 treatment. These results, therefore, indicate that Zn could alleviate AFB1-induced cytotoxicity by changing the expressions of zinc finger proteins in liver hepatocellular carcinoma (HepG2 cells).
Collapse
Affiliation(s)
- Liye Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Chuchu Huang
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Xuan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Boyang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China
| | - Kunlun Huang
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
7
|
Ajiboye TO, Aliyu NO, Ajala-Lawal RA. Lophirones B and C induce oxidative cellular death pathway in Acinetobacter baumannii by inhibiting DNA gyrase. Microb Pathog 2019; 130:226-231. [PMID: 30872146 DOI: 10.1016/j.micpath.2019.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/02/2019] [Accepted: 03/07/2019] [Indexed: 10/27/2022]
Abstract
We evaluated the inactivation of DNA gyrase on the oxidative stress response and sensitivity of A. baumannii to lophirones B and C. The sensitivity of parental and the mutant strains of A. baumannii to lophirones B and C was determined using minimum inhibitory concentration (MIC) and time-kill sensitivity. Inactivation of sodB, katG, recA enhanced the sensitivity of A. baumannii to lophirones B and C. Furthermore, this inactivation increased the accumulation of superoxide anion radical and hydrogen peroxide in lophirones B and C-treated A. baumannii, which was reversed in the presence of thiourea. Inactivation of gyrA stalled lophirones B and C-mediated ROS accumulation in A. baumannii. In addition, lophirones B and C raised the Fe2+ contents of A. baumannii. Dipyridyl (Fe chelator) reversed the sensitivity of A. baumannii to lophirones B and C. Lophirones significantly lowered the NAD+/NADH ratio of A. baumannii. The results of this study revealed that the impact of DNA gyrase in lophirones B and C-mediated ROS accumulation, Fe2+ release and cell death.
Collapse
Affiliation(s)
- T O Ajiboye
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences, Nile University of Nigeria, FCT-Abuja, Nigeria.
| | - N O Aliyu
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences, Nile University of Nigeria, FCT-Abuja, Nigeria
| | - R A Ajala-Lawal
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences, Nile University of Nigeria, FCT-Abuja, Nigeria
| |
Collapse
|
8
|
Engin AB, Engin A. DNA damage checkpoint response to aflatoxin B1. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 65:90-96. [PMID: 30594067 DOI: 10.1016/j.etap.2018.12.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/20/2018] [Accepted: 12/07/2018] [Indexed: 05/28/2023]
Abstract
Although most countries regulate the aflatoxin levels in food by legislations, high amounts of aflatoxin B1 (AFB1)-DNA adducts can still be detected in normal and tumorous tissue obtained from cancer patients. AFB1 cannot directly interact with DNA unless it is biotransformed to AFB1-8, 9-epoxide via cytochrome p450 enzymes. This metabolite spontaneously and irreversibly attaches to guanine residues to generate highly mutagenic DNA adducts. AFB1-induced mutation of ATM kinase results in the deterioration of the cell cycle checkpoint activation at the G2/M checkpoint site. Genomic instability and increased cancer risk due to A-T mutation is the result of diminished repair of DNA double strand breaks. The major point mutation caused by AFB1 is G-to-T transversion that is related with the high frequency of p53 mutation. Majority of AFB1 associated hepatocellular cancer cases carry TP53 mutant DNA, which is an indicator of AFB1 exposure, as well as hepatocellular cancer risk.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| |
Collapse
|
9
|
Abdel-Wahhab MA, El-Nekeety AA, Hassan NS, Gibriel AAY, Abdel-Wahhab KG. Encapsulation of cinnamon essential oil in whey protein enhances the protective effect against single or combined sub-chronic toxicity of fumonisin B 1 and/or aflatoxin B 1 in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:29144-29161. [PMID: 30112645 DOI: 10.1007/s11356-018-2921-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 08/06/2018] [Indexed: 05/17/2023]
Abstract
Fumonisin B1 (FB1) and aflatoxin B1 (AFB1) are fungal metabolites that frequently co-occur in foodstuffs and are responsible for mycotoxicosis and several primary cancers. Cinnamon essential oil (CEO) has a spacious range of benefit effects but also has some limitations owing to its strong taste or its interaction with some drugs. This study aimed to use the cinnamon oil emulsion droplets (COED) for the protection against oxidative stress, cytotoxicity, and reproductive toxicity in male Sprague-Dawley rats sub-chronically exposed to FB1 and/or AFB1. The composition of CEO was identified using GC-MS then was encapsulated using whey protein as wall material. Male rats were divided into eight groups and treated orally for 8 weeks as follows: control group, AFB1-trreated group (80 μg/kg b.w), FB1-treated group (100 mg/kg b.w), FB1 plus AFB1-treated group, and the groups treated with COED plus FB1 and/or AFB1. Blood and samples of the kidney, liver, and testis were collected for different analysis and histopathological examination. The GC-MS analysis revealed that cinnamaldehyde, α-copaene, trans-cinnamaldehyde, caryophyllene, and delta-cadinene were the main compounds in COE. The average size of COED was 235 ± 1.4 nm and the zeta potential was - 6.24 ± 0.56. Treatment with FB1 and/or AFB1 induced significant disturbances in the serum biochemical analysis, oxidative stress parameters, DNA fragmentation, gene expression, and testosterone and severe pathological changes in the tested organs. Moreover, treatment with both mycotoxins induced synergistic toxic effects. COED did not induce toxic effects and could normalize the majority of the tested parameters and improve the histological picture in rats treated with FB1 and/or AFB1. It could be concluded that COED induce potential protective effects against the single or combined exposure to FB1 and AFB1.
Collapse
Affiliation(s)
- Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Center, Dokki, Cairo, Egypt.
| | - Aziza A El-Nekeety
- Food Toxicology & Contaminants Department, National Research Center, Dokki, Cairo, Egypt
| | - Nabila S Hassan
- Pathology Department, National Research Center, Dokki, Cairo, Egypt
| | - Abdullah A Y Gibriel
- Biochemistry & Molecular Biology Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
- Center of Drug Research & Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | | |
Collapse
|
10
|
Yilmaz S, Kaya E, Karaca A, Karatas O. Aflatoxin B1 induced renal and cardiac damage in rats: Protective effect of lycopene. Res Vet Sci 2018; 119:268-275. [DOI: 10.1016/j.rvsc.2018.07.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/10/2018] [Accepted: 07/21/2018] [Indexed: 01/08/2023]
|
11
|
Aliyu NO, Ajala-Lawal RA, Ajiboye TO. Lophirones B and C halt acetaminophen hepatotoxicity by upregulating redox transcription factor Nrf-2 through Akt, PI3K, and PKC pathways. J Biochem Mol Toxicol 2018; 32:e22055. [DOI: 10.1002/jbt.22055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/26/2018] [Accepted: 03/27/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Najeeb O. Aliyu
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences; Nile University of Nigeria; Abuja Nigeria
| | - Rafiat A. Ajala-Lawal
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences; Nile University of Nigeria; Abuja Nigeria
| | - Taofeek O. Ajiboye
- Antioxidants, Redox Biology and Toxicology Research Group, Department of Medical Biochemistry, College of Health Sciences; Nile University of Nigeria; Abuja Nigeria
| |
Collapse
|
12
|
Karimi-Sales E, Mohaddes G, Alipour MR. Chalcones as putative hepatoprotective agents: Preclinical evidence and molecular mechanisms. Pharmacol Res 2018; 129:177-187. [DOI: 10.1016/j.phrs.2017.11.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 02/08/2023]
|
13
|
Müller LK, da Silva AS, Baldissera MD, Santurio JM, Glombowsky P, Gugel J, Campigotto G, Gloria EM, Paiano D, Machado G. Effects of supplementation with spray-dried porcine plasma on blood variables on piglets feed with diet contaminated by mycotoxins. Microb Pathog 2017; 110:464-470. [DOI: 10.1016/j.micpath.2017.07.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/12/2017] [Accepted: 07/17/2017] [Indexed: 12/24/2022]
|
14
|
Abas H, Linsdall SM, Mamboury M, Rzepa HS, Spivey AC. Total Synthesis of (+)-Lophirone H and Its Pentamethyl Ether Utilizing an Oxonium–Prins Cyclization. Org Lett 2017; 19:2486-2489. [DOI: 10.1021/acs.orglett.7b00642] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hossay Abas
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, U.K
| | - Sean M. Linsdall
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, U.K
| | - Mathias Mamboury
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, U.K
| | - Henry S. Rzepa
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, U.K
| | - Alan C. Spivey
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, U.K
| |
Collapse
|