1
|
Lai Y, Li M, Liao X, Zou L. Smartphone-Assisted Colorimetric Detection of Glutathione and Glutathione Reductase Activity in Human Serum and Mouse Liver Using Hemin/G-Quadruplex DNAzyme. Molecules 2021; 26:5016. [PMID: 34443603 PMCID: PMC8398021 DOI: 10.3390/molecules26165016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/26/2022] Open
Abstract
Abnormal levels of reduced glutathione (GSH) and glutathione reductase (GR) are usually related to a variety of diseases, so it is of great significance to determine the GSH concentration and GR activity. We herein develop a smartphone-assisted colorimetric biosensor for the detection of GSH and GR activity in human serum and mouse liver using hemin/G-quadruplex DNAzyme. Firstly, an obvious color change from colorless to green can be observed, owing to the high peroxidase-like activity of hemin/G-quadruplex DNAzyme toward 2,2'-azino-bis(3-ethylbenzothiozoline-6-sulfonic acid) (ABTS). With the addition of GSH or GR, the H2O2-mediated oxidation of ABTS catalyzed by hemin/G-quadruplex DNAzyme is significantly inhibited, resulting in remarkable color fading. Therefore, the detection of GSH and GR activity can be achieved by observing the color transition or measuring the absorbance at 420 nm. The detection limit was estimated to be as low as 0.1 μM and 10 μU/mL for GSH and GR, respectively. More interestingly, the RGB values of the sensing system can be identified by the smartphone application (APP, color collect), which makes it an ideal format for on-site determination and point-of-care testing (POCT). In addition, the proposed method shows excellent selectivity and acceptable applicability for the determination of GSH concentration and GR activity in human serum samples and mouse liver tissues, which might hold great application potential in clinical diagnosis and drug screening.
Collapse
Affiliation(s)
| | | | | | - Li Zou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Y.L.); (M.L.); (X.L.)
| |
Collapse
|
2
|
Bahrami A, Sathyapalan T, Moallem SA, Sahebkar A. Counteracting arsenic toxicity: Curcumin to the rescue? JOURNAL OF HAZARDOUS MATERIALS 2020; 400:123160. [PMID: 32574880 DOI: 10.1016/j.jhazmat.2020.123160] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Arsenicosis leads to various irreversible damages in several organs and is considered to be a carcinogen. The effects of chronic arsenic poisoning are a result of an imbalance between pro- and antioxidant homeostasis, oxidative stress, as well as DNA and protein damage. Curcumin, the polyphenolic pigment extracted from the rhizome of Curcuma longa, is well-known for its pleiotropic medicinal effects. Curcumin has been shown to have ameliorative effects in arsenic-induced genotoxicity, nephrotoxicity, hepatotoxicity, angiogenesis, skin diseases, reproductive toxicity, neurotoxicity, and immunotoxicity. This review aims to summarize the scientific evidence on arsenic toxicity in various organs and the ameliorative effects of curcumin on the arsenic toxicity.
Collapse
Affiliation(s)
- Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, HU3 2JZ, UK
| | - Seyed Adel Moallem
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Kim J, Hyun SW, Lee IS, Jo K, Kim YS, Kim JS, Kim CS. Aster koraiensis extract lowers postprandial glucose in normoglycemic and high-fat-diet-induced obese mice. Food Sci Biotechnol 2019; 28:563-568. [PMID: 30956869 PMCID: PMC6431321 DOI: 10.1007/s10068-018-0497-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/08/2018] [Accepted: 10/16/2018] [Indexed: 11/28/2022] Open
Abstract
The Aster koraiensis extract (ASKO) is a newly developed dietary herbal supplement. In this study, the potent blood glucose-lowering activity of ASKO in vitro and in vivo was investigated. In an in vitro glucose uptake assay, ASKO was found to enhance glucose transport in 3T3-L1 adipocytes. Oral administration of ASKO significantly reduced glucose levels in normoglycemic mice during oral glucose tolerance tests (OGTTs). In a long-term efficacy study, 4 weeks of oral ASKO treatment significantly attenuated blood glucose levels during OGTTs in diet-induced obese (DIO) mice. ASKO also enhanced plasma insulin levels after glucose loading, leading to a reduction in blood glucose levels. In addition, ASKO normalized glucose transporter-4 mRNA expression in the muscles of DIO mice. These results indicate that ASKO has postprandial glucose-lowering effects and could be beneficial in the management of prediabetes or type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Junghyun Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
- Department of Oral Pathology, School of Dentistry, Chonbuk National University, Jeonju, 54896 Republic of Korea
| | - Soo-Wang Hyun
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
| | - Ik Soo Lee
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
| | - Kyuhyung Jo
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
| | - Young Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
| | - Chan-Sik Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yusengdae-ro, Daejeon, 34054 Republic of Korea
- Korean Medicine Life Science, University of Science Technology (UST), Daejeon, 34113 Republic of Korea
| |
Collapse
|
4
|
Krishnan V, Loganathan C, Thayumanavan P. Green synthesized selenium nanoparticles using Spermacoce hispida as carrier of s-allyl glutathione: to accomplish hepatoprotective and nephroprotective activity against acetaminophen toxicity. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:56-63. [DOI: 10.1080/21691401.2018.1543192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Vennila Krishnan
- Department of Biochemistry, Molecular Therapeutics Laboratory, Periyar University, Salem, India
| | - Chitra Loganathan
- Department of Biochemistry, Molecular Therapeutics Laboratory, Periyar University, Salem, India
| | - Palvannan Thayumanavan
- Department of Biochemistry, Molecular Therapeutics Laboratory, Periyar University, Salem, India
| |
Collapse
|
5
|
Amin KA, Hashem KS, Alshehri FS, Awad ST, Hassan MS. Antioxidant and Hepatoprotective Efficiency of Selenium Nanoparticles Against Acetaminophen-Induced Hepatic Damage. Biol Trace Elem Res 2017; 175:136-145. [PMID: 27220627 DOI: 10.1007/s12011-016-0748-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/16/2016] [Indexed: 12/27/2022]
Abstract
Overdoses of acetaminophen (APAP), a famous and widely used drug, may have hepatotoxic effects. Nanoscience is a novel scientific discipline that provides specific tools for medical science problems including using nano trace elements in hepatic diseases. Our study aimed to assess the hepatoprotective role of selenium nanoparticles (Nano-Se) against APAP-induced hepatic injury. Twenty-four male rats were classified into three equal groups: a control group that received 0.9 % NaCl, an APAP-treated group (oral administration), and a group treated with Nano-Se (10-20 nm, intraperitoneal (i.p.) injection) and APAP (oral administration). APAP overdose induced significant elevations in liver function biomarkers, hepatic lipid peroxidation, hepatic catalase, and superoxide dismutase (SOD), decreased the reduced glutathione (GSH) content and glutathione reductase (GR) activity, and stimulated significant DNA damage in hepatocytes, compared to control rats. Nano-Se administration improved the hepatic antioxidant protection mechanism and decreased cellular sensitivity to DNA fragmentation. Nano-Se exhibits a protective effect against APAP-induced hepatotoxicity through improved liver function and oxidative stress mediated by catalase, SOD, and GSH and decreases hepatic DNA fragmentation, a hepatic biomarker of cell death. Nano-Se could be a novel hepatoprotective strategy to inhibit oxidative stress.
Collapse
Affiliation(s)
- Kamal Adel Amin
- Department of Chemistry, College of Science, University of Dammam, PO Box 383, Dammam, 31113, Dammam, Saudi Arabia.
- Biochemistry Department, Faculty of Veterinary Medicine, Beni Suef University, Beni-Suef City, Egypt.
| | - Khalid Shaban Hashem
- Biochemistry Department, Faculty of Veterinary Medicine, Beni Suef University, Beni-Suef City, Egypt
| | - Fawziah Saleh Alshehri
- Department of Chemistry, College of Science, University of Dammam, PO Box 383, Dammam, 31113, Dammam, Saudi Arabia
| | - Said T Awad
- Biochemistry Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mohammed S Hassan
- Internal Medicine Department, Faculty of Veterinary Medicine, Beni Suef University, Beni-Suef City, Egypt
| |
Collapse
|
6
|
Abu El-Saad AM, Al-Kahtani MA, Abdel-Moneim AM. N-acetylcysteine and meso-2,3-dimercaptosuccinic acid alleviate oxidative stress and hepatic dysfunction induced by sodium arsenite in male rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3425-3434. [PMID: 27799742 PMCID: PMC5076801 DOI: 10.2147/dddt.s115339] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Environmental exposure to arsenic represents a serious challenge to humans and other animals. The aim of the present study was to test the protective effect of antioxidant N-acetylcysteine (NAC) either individually or in combination with a chelating agent, meso-2,3-dimercaptosuccinic acid (DMSA), against sodium arsenite oral toxicity in male rats. Five groups were used: control; arsenic group (orally administrated in a concentration of 2 mg/kg body weight [b.w.]); the other three groups were orally administrated sodium arsenite in a concentration of 2 mg/kg b.w. followed by either NAC (10 mg/kg b.w., intraperitoneally [i.p.]), DMSA (50 mg/kg b.w., i.p.) or NAC plus DMSA. Arsenic toxicity caused significant rise in serum aspartate aminotransferase, alanine aminotransferase and total bilirubin, and a significant decrease in total protein (TP) and albumin levels after 3 weeks of experimental period. In addition, arsenic-treated rats showed significantly higher arsenic content in liver and significant rise in hepatic malondialdehyde level. By contrast, sharp decreases in glutathione content and catalase and glutathione reductase activities were discernible. NAC and/or DMSA counteracted most of these physiologic and biochemical defects. NAC monotherapy was more effective than DMSA in increasing TP, while DMSA was more effective in decreasing alanine aminotransferase. The combined treatment was superior over monotherapies in recovery of TP and glutathione. Biochemical data were well supported by histopathological and ultrastructural findings. In conclusion, the combination therapy of NAC and DMSA may be an ideal choice against oxidative insult induced by arsenic poisoning.
Collapse
Affiliation(s)
- Ahmed M Abu El-Saad
- Department of Biology, Faculty of Medicine, Dammam University, Dammam, Saudi Arabia; Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohammed A Al-Kahtani
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Ashraf M Abdel-Moneim
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
7
|
Xiaohua D, Jin Z, Hui W, Haifeng C, Chao Z, Zepu Y. Effect of Yajieshaba, a preparation of Dai indigenous medicine, on enhanced liver detoxification. J TRADIT CHIN MED 2015; 35:197-205. [PMID: 25975053 DOI: 10.1016/s0254-6272(15)30028-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To explore the mechanistic effects of Yajieshaba (YJSB) on enhanced liver detoxification. METHODS The effects of YJSB on alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were assayed in five acute chemical liver injury models [carbon tetrachloride (CCl4), D-galactosamine (D-Glan), 4-acetamidophenol (AAP), thioacetamide (TAA) and 1-naphthyl isothiocyanate (ANIT)]. Sleep latency and sleep time of pentobarbital sodium were tested in control mice and CCl4 model miceafter oral YJSB administration. The effects of YJSB on drug metabolism enzymes of liver microsomes were tested in control rats and CCl4 model rats. The levels of cytochrome P450 (CYP450) and Cyt b5 in liver microsomes were assayed using the method by Omura and Sato, and activities of erythromycin N-demethylase (ERD) and aminopyrine N-demethyl (ADM) were evaluated by Nash colorimetry. Probe substrate-based high performance liquid chromatography (HPLC) methods were established for CYP3A4 and CYP1A2. RESULTS The level of serum ALT was reduced by YJSB at 3.51 g/kg in the five models as follows: CCl4 > D-Glan, AAP, ANIT > TAA. YJSB treatment did not reduce the level of serum AST. YJSB at 3.51 g/kg prolonged the sleep latency in control mice and shortened the sleep time of control mice and CCl4 model mice. For control rats, YJSB at 2.43 g/kg increased the levels of CYP450 and Cyt b5 and induced the activities of ERD and ADM; for liver injuries induced by CCl4 in rats, YJSB at 2.43 g/kg increased the levels of CYP450 and Cyt b5. These results suggest that YJSB at 2.43 g/kg induces CYP3A4 and CYP1A2. CONCLUSION These results suggest that YJSB enhanced liver detoxification and the mechanisms may be partially related to CYP3A4 and CYP1A2 induction.
Collapse
|
8
|
Muthumani M, Miltonprabu S. Ameliorative efficacy of tetrahydrocurcumin against arsenic induced oxidative damage, dyslipidemia and hepatic mitochondrial toxicity in rats. Chem Biol Interact 2015; 235:95-105. [DOI: 10.1016/j.cbi.2015.04.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/10/2015] [Accepted: 04/07/2015] [Indexed: 12/14/2022]
|
9
|
Sankar P, Gopal Telang A, Kalaivanan R, Karunakaran V, Manikam K, Sarkar SN. Effects of nanoparticle-encapsulated curcumin on arsenic-induced liver toxicity in rats. ENVIRONMENTAL TOXICOLOGY 2015; 30:628-637. [PMID: 24347089 DOI: 10.1002/tox.21940] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 06/03/2023]
Abstract
We investigated the therapeutic effectiveness of the nanoparticle-encapsulated curcumin (CUR-NP) against sodium arsenite-induced hepatic oxidative damage in rats. The CUR-NP prepared by emulsion technique was spherical in shape with an encapsulation efficiency of 86.5%. The particle size ranged between 120 and 140 nm with the mean particle size being 130.8 nm. Rats were divided into five groups of six each. Group 1 served as control. Group 2 rats were exposed to sodium arsenite (25 ppm) daily through drinking water for 42 days. Groups 3, 4, and 5 were treated with arsenic as in group 2, however, they were administered, empty nanoparticles, curcumin (100 mg/kg bw) and CUR-NP (100 mg/kg bw), respectively, by oral gavage during the last 14 days of arsenic exposure. Arsenic increased the activities of serum alanine aminotransferase and aspartate aminotransferase and caused histological alterations in liver indicating hepatotoxicity. Arsenic increased lipid peroxidation, depleted reduced glutathione and decreased the activities of superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase in liver. All these effects of arsenic were attenuated with both curcumin and CUR-NP. However, the magnitude of amelioration was more pronounced with CUR-NP. The results indicate that curcumin given in nano-encapsulated form caused better amelioration than free curcumin. © 2013 Wiley Periodicals, Inc. Environ Toxicol 30: 628-637, 2015.
Collapse
Affiliation(s)
- Palanisamy Sankar
- Department of Veterinary Pharmacology and Toxicology, Veterinary College and Research Institute, Orathanadu, 614 625, Tamil Nadu, India
| | - Avinash Gopal Telang
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Ramya Kalaivanan
- Department of Veterinary Epidemiology and Preventive Medicine, Veterinary College and Research Institute, Namakkal, 637002, Tamil Nadu, India
| | - Vijayakaran Karunakaran
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Kesavan Manikam
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Souvendra Nath Sarkar
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| |
Collapse
|
10
|
Majhi CR, Khan S, Leo MDM, Prawez S, Kumar A, Sankar P, Telang AG, Sarkar SN. Acetaminophen increases the risk of arsenic-mediated development of hepatic damage in rats by enhancing redox-signaling mechanism. ENVIRONMENTAL TOXICOLOGY 2014; 29:187-198. [PMID: 22120977 DOI: 10.1002/tox.20785] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 10/15/2011] [Indexed: 05/31/2023]
Abstract
We evaluated whether the commonly used analgesic-antipyretic drug acetaminophen can modify the arsenic-induced hepatic oxidative stress and also whether withdrawal of acetaminophen administration during the course of long-term arsenic exposure can increase susceptibility of liver to arsenic toxicity. Acetaminophen was co-administered orally to rats for 3 days following 28 days of arsenic pre-exposure (Phase-I) and thereafter, acetaminophen was withdrawn, but arsenic exposure was continued for another 28 days (Phase-II). Arsenic increased lipid peroxidation and reactive oxygen species (ROS) generation, depleted glutathione (GSH), and decreased superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), and glutathione reductase (GR) activities. Acetaminophen caused exacerbation of arsenic-mediated lipid peroxidation and ROS generation and further enhancement of serum alanine aminotransferase and aspartate aminotransferase activities. In Phase-I, acetaminophen caused further GSH depletion and reduction in SOD, catalase, GPx and GR activities, but in Phase-II, only GPx and GR activities were more affected. Arsenic did not alter basal and inducible nitric oxide synthase (iNOS)-mediated NO production, but decreased constitutive NOS (cNOS)-mediated NO release. Arsenic reduced expression of endothelial NOS (eNOS) and iNOS genes. Acetaminophen up-regulated eNOS and iNOS expression and NO production in Phase-I, but reversed these effects in Phase-II. Results reveal that acetaminophen increased the risk of arsenic-mediated hepatic oxidative damage. Withdrawal of acetaminophen administration also increased susceptibility of liver to hepatotoxicity. Both ROS and NO appeared to mediate lipid peroxidation in Phase-I, whereas only ROS appeared responsible for peroxidative damage in Phase-II.
Collapse
Affiliation(s)
- Chhaya Rani Majhi
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar 243 122, Uttar Pradesh, India
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Anwar-Mohamed A, El-Sherbeni A, Kim SH, Elshenawy OH, Althurwi HN, Zordoky BNM, El-Kadi AOS. Acute arsenic treatment alters cytochrome P450 expression and arachidonic acid metabolism in lung, liver and kidney of C57Bl/6 mice. Xenobiotica 2013; 43:719-29. [DOI: 10.3109/00498254.2012.754113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
12
|
Inkielewicz-Stępniak I, Knap N. Effect of exposure to fluoride and acetaminophen on oxidative/nitrosative status of liver and kidney in male and female rats. Pharmacol Rep 2012; 64:902-11. [DOI: 10.1016/s1734-1140(12)70885-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 04/16/2012] [Indexed: 10/25/2022]
|
13
|
Anwar-Mohamed A, El-Sherbeni AA, Kim SH, Althurwi HN, Zordoky BNM, El-Kadi AOS. Acute arsenic toxicity alters cytochrome P450 and soluble epoxide hydrolase and their associated arachidonic acid metabolism in C57Bl/6 mouse heart. Xenobiotica 2012; 42:1235-47. [PMID: 22680237 DOI: 10.3109/00498254.2012.693971] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute arsenic (As(III)) exposure has been reported to cause cardiac toxicity, however this toxicity was never linked to the disturbance in cytochrome P450 (P450)-mediated arachidonic acid metabolism. Therefore, we investigated the effect of acute As(III) toxicity on the expression of P450 and soluble epoxide hydrolase (sEH) and their associated arachidonic acid metabolism in mice hearts. As(III) toxicity was induced by a single intraperitoneal injection of 12.5 mg/kg of As(III). Our results showed that As(III) treatment caused a significant induction of the cardiac hypertrophic markers in addition to Cyp1b1, Cyp2b, Cyp2c, Cyp4f, and sEH gene expression in mice hearts. Furthermore, As(III) increased sEH protein expression and activity in hearts with a consequent decrease in 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs) formation. Whereas the formation of 8,9-, 11,12-, 14,15-dihydroxyeicosatrienoic acids (DHETs) was significantly increased. As(III) also increased sEH mRNA and protein expression levels in addition to the hypertrophic markers which was reversed by knockdown of sEH in H9c2 cells. In conclusion, acute As(III) toxicity alters the expression of several P450s and sEH enzymes with a consequent decrease in the cardioprotective EETs which may represent a novel mechanism by which As(III) causes progressive cardiotoxicity. Furthermore, inhibiting sEH might represent a novel therapeutic approach to prevent As(III)-induced hypertrophy.
Collapse
Affiliation(s)
- Anwar Anwar-Mohamed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
14
|
Gardner CR, Mishin V, Laskin JD, Laskin DL. Exacerbation of acetaminophen hepatotoxicity by the anthelmentic drug fenbendazole. Toxicol Sci 2011; 125:607-12. [PMID: 22048645 DOI: 10.1093/toxsci/kfr301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Fenbendazole is a broad-spectrum anthelmintic drug widely used to prevent or treat nematode infections in laboratory rodent colonies. Potential interactions between fenbendazole and hepatotoxicants such as acetaminophen are unknown, and this was investigated in this study. Mice were fed a control diet or a diet containing fenbendazole (8-12 mg/kg/day) for 7 days prior to treatment with acetaminophen (300 mg/kg) or phosphate buffered saline. In mice fed a control diet, acetaminophen administration resulted in centrilobular hepatic necrosis and increases in serum transaminases, which were evident within 12 h. Acetaminophen-induced hepatotoxicity was markedly increased in mice fed the fenbendazole-containing diet, as measured histologically and by significant increases in serum transaminase levels. Moreover, in mice fed the fenbendazole-containing diet, but not the control diet, 63% mortality was observed within 24 h of acetaminophen administration. Fenbendazole by itself had no effect on liver histology or serum transaminases. To determine if exaggerated hepatotoxicity was due to alterations in acetaminophen metabolism, we analyzed sera for the presence of free acetaminophen and acetaminophen-glucuronide. We found that there were no differences in acetaminophen turnover. We also measured cytochrome P450 (cyp) 2e1, cyp3a, and cyp1a2 activity. Whereas fenbendazole had no effect on the activity of cyp2e1 or cyp3a, cyp1a2 was suppressed. A prolonged suppression of hepatic glutathione (GSH) was also observed in acetaminophen-treated mice fed the fenbendazole-containing diet when compared with the control diet. These data demonstrate that fenbendazole exacerbates the hepatotoxicity of acetaminophen, an effect that is related to persistent GSH depletion. These findings are novel and suggest a potential drug-drug interaction that should be considered in experimental protocols evaluating mechanisms of hepatotoxicity in rodent colonies treated with fenbendazole.
Collapse
Affiliation(s)
- Carol R Gardner
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA.
| | | | | | | |
Collapse
|
15
|
Majhi CR, Khan S, Leo MDM, Manimaran A, Sankar P, Sarkar SN. Effects of acetaminophen on reactive oxygen species and nitric oxide redox signaling in kidney of arsenic-exposed rats. Food Chem Toxicol 2011; 49:974-82. [PMID: 21219961 DOI: 10.1016/j.fct.2011.01.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 12/04/2010] [Accepted: 01/04/2011] [Indexed: 01/01/2023]
Abstract
We examined whether acetaminophen could alter renal oxidative stress induced by arsenic; also whether withdrawal of acetaminophen treatment can increase susceptibility of kidney to arsenic toxicity. Acetaminophen (400 and 1600 mg/kg) was co-administered orally to rats for 3 days after preexposure to arsenic (25 ppm) for 28 days (Phase-I) and thereafter, acetaminophen was withdrawn, but arsenic exposure was continued for another 28 days (Phase-II). Acetaminophen enhanced arsenic-induced lipid peroxidation, GSH depletion and ROS production and further decreased superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase activities. Increased peroxidation did not alter kidney weight, but increased serum urea nitrogen and creatinine. Arsenic did not alter basal, iNOS-mediated NO production or iNOS expression. Arsenic decreased cNOS-mediated NO release and eNOS expression in Phase-II. Acetaminophen increased their expressions and NO production in Phase-I. In Phase-II, arsenic-mediated effects on NO remained mostly unaffected with acetaminophen. Results reveal that acetaminophen enhanced the risk of arsenic-mediated oxidative stress in kidney. Discontinuation of acetaminophen administration also increased the susceptibility of kidney to nephrotoxic effect of arsenic. It appeared ROS were primarily responsible for oxidative stress in both the phases. NO may have a minor role in Phase-I, but does not contribute to redox signaling mechanism in Phase-II.
Collapse
Affiliation(s)
- Chhaya Rani Majhi
- Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Izatnagar 243 122, Bareilly, Uttar Pradesh, India
| | | | | | | | | | | |
Collapse
|