1
|
Whitlock BD, Ma Y, Conseil G, O'Brien AR, Banerjee M, Swanlund DP, Lin ZP, Wang Y, Le XC, Schuetz JD, Cole SPC, Leslie EM. Differential Selectivity of Human and Mouse ABCC4/Abcc4 for Arsenic Metabolites. Drug Metab Dispos 2024; 52:1417-1428. [PMID: 39313329 PMCID: PMC11585317 DOI: 10.1124/dmd.124.001852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Millions of people globally are exposed to the proven human carcinogen arsenic at unacceptable levels in drinking water. In contrast, arsenic is a poor rodent carcinogen, requiring >100-fold higher doses for tumor induction, which may be explained by toxicokinetic differences between humans and mice. The human ATP-binding cassette subfamily C (ABCC) transporter hABCC4 mediates the cellular efflux of a diverse array of metabolites, including the glutathione (GSH) conjugate of the highly toxic monomethylarsonous acid (MMAIII), monomethylarsenic diglutathione [MMA(GS)2], and the major human urinary arsenic metabolite dimethylarsinic acid (DMAV). Our objective was to determine if mouse Abcc4 (mAbcc4) protected against and/or transported the same arsenic species as hABCC4. The anti-ABCC4 antibody M4I-10 epitope was first mapped to an octapeptide (411HVQDFTA418F) present in both hABCC4 and mAbcc4, enabling quantification of relative amounts of hABCC4/mAbcc4. mAbcc4 expressed in human embryonic kidney (HEK)293 cells did not protect against any of the six arsenic species tested [arsenite, arsenate, MMAIII, monomethylarsonic acid, dimethylarsinous acid, or DMAV], despite displaying remarkable resistance against the antimetabolite 6-mercaptopurine (>9-fold higher than hABCC4). Furthermore, mAbcc4-enriched membrane vesicles prepared from transfected HEK293 cells did not transport MMA(GS)2 or DMAV despite a >3-fold higher transport activity than hABCC4-enriched vesicles for the prototypic substrate 17β-estradiol-17-(β-D-glucuronide). Abcc4(+/+) mouse embryonic fibroblasts (MEFs) were ∼3-fold more resistant to arsenate than Abcc4(-/-) MEFs; however, further characterization indicated that this was not mAbcc4 mediated. Thus, under the conditions tested, arsenicals are not transported by mAbcc4, and differences between the substrate selectivity of hABCC4 and mAbcc4 seem likely to contribute to arsenic toxicokinetic differences between human and mouse. SIGNIFICANCE STATEMENT: Toxicokinetics of the carcinogen arsenic differ among animal species. Arsenic methylation is known to contribute to this, whereas arsenic transporters have not been considered. Human ATP-binding cassette subfamily C member 4 (hABCC4) is a high-affinity transporter of toxicologically important arsenic metabolites. Here we used multiple approaches to demonstrate that mouse Abcc4 does not protect cells against or transport any arsenic species tested. Thus, differences between hABCC4 and mAbcc4 substrate selectivity likely contribute to differences in human and mouse arsenic toxicokinetics.
Collapse
Affiliation(s)
- Brayden D Whitlock
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Yingze Ma
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Gwenaëlle Conseil
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Alicia R O'Brien
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Mayukh Banerjee
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Diane P Swanlund
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Z Ping Lin
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Yao Wang
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - X Chris Le
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - John D Schuetz
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Susan P C Cole
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| | - Elaine M Leslie
- Department of Physiology (B.D.W., Y.M., A.R.O., M.B., D.P.S., E.M.L.), Membrane Protein Disease Research Group (B.D.W., Y.M., M.B., D.P.S., E.M.L.), and Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology (X.C.L., E.M.L.), University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Molecular Medicine, Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, Ontario, Canada (G.C., S.P.C.C.); Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut (Z.P.L.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (Y.W., J.D.S.)
| |
Collapse
|
2
|
Doroudian M, Pourzadi N, Gautam A, Gailer J. Translational toxicology of metal(loid) species: linking their bioinorganic chemistry in the bloodstream to organ damage onset. Biometals 2024; 37:739-753. [PMID: 37815752 DOI: 10.1007/s10534-023-00537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/08/2023] [Indexed: 10/11/2023]
Abstract
The quantification of arsenic, mercury, cadmium and lead in the human bloodstream is routinely used today to assess exposure to these toxic metal(loid)s, but the interpretation of the obtained data in terms of their cumulative health relevance remains problematic. Seemingly unrelated to this, epidemiological studies strongly suggest that the simultaneous chronic exposure to these environmental pollutants is associated with the etiology of autism, type 2 diabetes, irritable bowel disease and other diseases. This from a public health point of view undesirable situation urgently requires research initiatives to establish functional connections between human exposure to multiple toxic metal(loid) species and adverse health effects. One way to establish causal exposure-response relationships is a molecular toxicology approach, which requires one to unravel the biomolecular mechanisms that unfold after individual toxic metal(loid)s enter the bloodstream/organ nexus as these interactions ultimately determine which metabolites impinge on target organs and thus provide mechanistic links to diseases of unknown etiology. In an attempt to underscore the importance of the toxicological chemistry of metal(loid)s in the bloodstream, this review summarizes recent progress into relevant bioinorganic processes that are implicated in the etiology of adverse organ-based health effects and possibly diseases. A better understanding of these bioinorganic processes will not only help to improve the regulatory framework to better protect humans from the adverse effects of toxic metal(loid) species, but also represents an important starting point for the development of treatments to ameliorate pollution-induced adverse health effects on human populations, including pregnant women, the fetus and children.
Collapse
Affiliation(s)
- Maryam Doroudian
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Negar Pourzadi
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Astha Gautam
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Jürgen Gailer
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
3
|
Cheff DM, Skröder H, Akhtar E, Cheng Q, Hall MD, Raqib R, Kippler M, Vahter M, Arnér ES. Arsenic exposure and increased C-reactive protein are independently associated with lower erythrocyte glutathione peroxidase activity in Bangladeshi children. REDOX BIOCHEMISTRY AND CHEMISTRY 2023; 5-6:100015. [PMID: 37908807 PMCID: PMC10613583 DOI: 10.1016/j.rbc.2023.100015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Toxic metal contaminants present in food and water have widespread effects on health and disease. Chalcophiles, such as arsenic, cadmium, and mercury, show a high affinity to selenium and exposure to these metals could have a modulating effect on enzymes dependent on selenocysteine in their active sites. The aim of this study was to assess the influence of these metals on the activity of the selenoprotein glutathione peroxidase 1 (GPX1) in erythrocytes of 100 children residing in rural Bangladesh, where drinking water often contains arsenic. GPX1 expression, as measured using high-throughput immunoblotting, showed little correlation with GPX activity (rs = 0.02, p = 0.87) in blood samples. Toxic metals and selenium measured in erythrocytes using inductively coupled plasma mass spectrometry (ICP-MS) and C-reactive protein (CRP) measured in plasma, were all considered as effectors of this divergence in GPX enzymatic activity. Arsenic concentrations in erythrocytes were most influential for GPX1 activity (rs = -0.395, p < 0.0001), and CRP levels also negatively impacted GPX1 activity (rs = -0.443, p < 0.0001). These effects appear independent of each other as arsenic concentrations and CRP showed no correlation (rs = 0.124, p = 0.2204). Erythrocyte selenium, cadmium, and mercury did not show any correlation with GPX1 activity, nor with CRP or arsenic. Our findings suggest that childhood exposure to inorganic arsenic, as well as inflammation triggering the release of CRP, may negatively affect GPX1 activity in erythrocytes.
Collapse
Affiliation(s)
- Dorian M. Cheff
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Helena Skröder
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE, 171 77, Stockholm, Sweden
| | - Evana Akhtar
- International Center for Diarrheal Disease Research, GPO Box 128, Dhaka, 1000, Bangladesh
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden
| | - Matthew D. Hall
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Rubhana Raqib
- International Center for Diarrheal Disease Research, GPO Box 128, Dhaka, 1000, Bangladesh
| | - Maria Kippler
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE, 171 77, Stockholm, Sweden
| | - Marie Vahter
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE, 171 77, Stockholm, Sweden
| | - Elias S.J. Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| |
Collapse
|
4
|
Cai Z, Zhang Y, Zhang W, Ye J, Ling Q, Xing Z, Zhang S, Hoffmann PR, Liu Y, Yang W, Huang Z. Arsenic retention in erythrocytes and excessive erythrophagocytosis is related to low selenium status by impaired redox homeostasis. Redox Biol 2022; 52:102321. [PMID: 35500533 PMCID: PMC9065714 DOI: 10.1016/j.redox.2022.102321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/03/2022] [Accepted: 04/18/2022] [Indexed: 11/09/2022] Open
Abstract
Arsenic (As) contamination in drinking water is a global public health problem. Epidemiological studies have shown that selenium (Se) deficiency is associated with an increasing risk of arsenism. However, the association between Se status and As retention in erythrocytes and mechanisms underlying this association have not been fully investigated. In the present study, a total of 165 eligible subjects were recruited and As was found to accumulate in blood mainly by retention in erythrocytes. Retention of As in erythrocytes was negatively correlated with Se status, antioxidant parameters related to Se and As methylation capacity, but positively correlated with the protein-binding capacity of As. Additionally, erythrocytes isolated from subjects with low Se status exhibited cellular damage along with lower protein levels of CD47, which could be aggravated by hydrogen peroxide treatment. Consistent with the human study, the erythrocytes from mice with sub-chronic As exposure exhibited similar cellular damage and shown to be phagocytosed by splenic macrophages, and these effects were mitigated by dietary Se supplementation. Furthermore, hydrogen peroxide treatment induced excessive phagocytosis of erythrocytes with As exposure by splenic macrophages, while co-treating erythrocytes with the reducing agent, N-Acetyl-l-cysteine, mitigated this excessive erythrophagocytosis. Hyperactivation of the NFκB pathway was also detected in splenic macrophages after excessive erythrophagocytosis. In conclusion, this study found that low Se status involving impaired redox homeostasis increased As retention in erythrocytes, which were subsequently phagocytosed by splenic macrophages and led to an increased inflammatory status of splenic macrophages. These findings provide insight into physiological features of arsenism related to Se status and redox homeostasis.
Collapse
Affiliation(s)
- Zhihui Cai
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China
| | - Yutian Zhang
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China
| | - Weijie Zhang
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China
| | - Jinmin Ye
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China
| | - Qinjie Ling
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhi Xing
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Youbin Liu
- Department of Cardiology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| | - Weidong Yang
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China.
| | - Zhi Huang
- Department of Biotechnology, Jinan University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
5
|
Zhou JR, Kaur G, Ma Y, Arutyunov D, Lu X, Le XC, Leslie EM. Biliary excretion of arsenic by human HepaRG cells is stimulated by selenide and mediated by the multidrug resistance protein 2 (MRP2/ABCC2). Biochem Pharmacol 2021; 193:114799. [PMID: 34678219 DOI: 10.1016/j.bcp.2021.114799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
Millions of people worldwide are exposed to unacceptable levels of arsenic, a proven human carcinogen, in drinking water. In animal models, arsenic and selenium are mutually protective through formation and biliary excretion of seleno-bis (S-glutathionyl) arsinium ion [(GS)2AsSe]-. Selenium-deficient humans living in arsenic-endemic regions are at increased risk of arsenic-induced diseases, and may benefit from selenium supplementation. The influence of selenium on human arsenic hepatobiliary transport has not been studied using optimal human models. HepaRG cells, a surrogate for primary human hepatocytes, were used to investigate selenium (selenite, selenide, selenomethionine, and methylselenocysteine) effects on arsenic hepatobiliary transport. Arsenite + selenite and arsenite + selenide at different molar ratios revealed mutual toxicity antagonism, with the latter being higher. Significant levels of arsenic biliary excretion were detected with a biliary excretion index (BEI) of 14 ± 8%, which was stimulated to 32 ± 7% by selenide. Consistent with the formation and biliary efflux of [(GS)2AsSe]-, arsenite increased the BEI of selenide from 0% to 24 ± 5%. Arsenic biliary excretion was lost in the presence of selenite, selenomethionine, and methylselenocysteine. Sinusoidal export of arsenic was stimulated ∼1.6-fold by methylselenocysteine, but unchanged by other selenium forms. Arsenic canalicular and sinusoidal transport (±selenide) was temperature- and GSH-dependent and inhibited by MK571. Knockdown experiments revealed that multidrug resistance protein 2 (MRP2/ABCC2) accounted for all detectable biliary efflux of arsenic (±selenide). Overall, the chemical form of selenium and human MRP2 strongly influenced arsenic hepatobiliary transport, information critical for human selenium supplementation in arsenic-endemic regions.
Collapse
Affiliation(s)
- Janet R Zhou
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada
| | - Gurnit Kaur
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada
| | - Yingze Ma
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Denis Arutyunov
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Xiufen Lu
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada
| | - X Chris Le
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada
| | - Elaine M Leslie
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada.
| |
Collapse
|
6
|
Farkhondeh T, Naseri K, Esform A, Aramjoo H, Naghizadeh A. Drinking water heavy metal toxicity and chronic kidney diseases: a systematic review. REVIEWS ON ENVIRONMENTAL HEALTH 2021; 36:359-366. [PMID: 33128529 DOI: 10.1515/reveh-2020-0110] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/03/2020] [Indexed: 06/11/2023]
Abstract
Heavy metals in drinking water can threat human health and may induce several diseases. The association between heavy metals exposure and chronic kidney disease (CKD) has been indicated by few epidemiological studies. We conducted a systematic review of the epidemiologic publications of the association between exposure to heavy metals through drinking water and CKD. Keywords related to heavy metals and kidney diseases on MeSH were identified and searched in PubMed, Google Scholar, Scopus, Ovid-Medline and Web of Science until July 2020. 14 publications met our inclusion criteria and included in the current review. The included articles were conducted on the association between arsenic, cadmium, lead and chromium in drinking water and CKD. Our study could not find strong evidence between heavy exposure to through drinking water and CKD, except for arsenic. The negative association was found between arsenic and lead and glomerular filtration rate (eGFR). The positive correlation was observed between cadmium exposure and urinary N-acetyl-β-d-glucosaminidase (NAG) concentrations, and also arsenic and chromium exposure and kidney injury molecule (KIM-1). Assessment of studies showed an association between arsenic, cadmium, lead and chromium and albuminuria and proteinuria, without CKD outcomes. Current systematic study showed few evidence for exposure to arsenic, cadmium, lead and chromium through drinking water and incidence of kidney problems. However, more epidemiological studies are required to confirm this association.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Kobra Naseri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Adeleh Esform
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Student Research Committee, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Hamed Aramjoo
- Student Research Committee, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Ali Naghizadeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Department of Environmental Health Engineering, Faculty of Health, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
7
|
Chaturvedi A, Bhattacharjee S, Kumar V, Singh AK. A study on the interdependence of heavy metals while contributing to groundwater pollution index. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:25798-25807. [PMID: 33471306 DOI: 10.1007/s11356-021-12352-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/02/2021] [Indexed: 06/12/2023]
Abstract
The contribution of heavy metal to the groundwater pollution index (m-HPI) is dependent on other heavy metals present in it. This contribution may be synergistic or anti-synergistic (antagonistic) depending upon the constituent matrix. Both heavy metal type and its concentration are important. m-HPI, a variant of heavy metal pollution index in water, may be calibrated against USEPA hazard index (HI) using a generic multivariate non-linear regression (MVNLR) model. Excellent correlation may be obtained between HI and m-HPI through optimization of normalized weightage factors of constituent metals that contribute to m-HPI. MVNLR model was employed on groundwater samples of ten sites having different heavy metal matrix. The synergistic/antagonistic contribution of heavy metals to m-HPI was well discernible at each site. This study clearly showed that the individual contribution of a particular heavy metal to pollution index might be altered (enhanced or reduced) in the presence of other heavy metals. A calibrated MVNLR model was successfully used for predicting the hazard index (HI) of water samples.
Collapse
Affiliation(s)
- Aaditya Chaturvedi
- Natural Resource and Environmental Management Group, CSIR - Central Institute of Mining and Fuel Research, Barwa Road, Dhanbad, 826015, India
- Department Environmental Science & Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, 826004, India
| | - Santanu Bhattacharjee
- Natural Resource and Environmental Management Group, CSIR - Central Institute of Mining and Fuel Research, Barwa Road, Dhanbad, 826015, India
| | - Vipin Kumar
- Department Environmental Science & Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, 826004, India
| | - Abhay Kumar Singh
- Natural Resource and Environmental Management Group, CSIR - Central Institute of Mining and Fuel Research, Barwa Road, Dhanbad, 826015, India.
| |
Collapse
|
8
|
Wang Y, Gu Y, Yang Y, Sun K, Li H. Glutathione transmembrane transmission gated by light-switches. J Photochem Photobiol A Chem 2021. [DOI: 10.1016/j.jphotochem.2020.112954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
9
|
Selenium-doped calcium phosphate biomineral reverses multidrug resistance to enhance bone tumor chemotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 32:102322. [PMID: 33186694 DOI: 10.1016/j.nano.2020.102322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/19/2020] [Accepted: 10/15/2020] [Indexed: 01/17/2023]
Abstract
The construction of a functional drug delivery system to reverse the multidrug resistance (MDR) of bone tumors in cases of failed chemotherapy remains a challenge. Herein, we demonstrate a selenium-doped calcium phosphate (Se-CaP) biomineral with high biocompatibility, biodegradability and pH-sensitive drug release properties. Se-CaP may not only serve as an effective drug-carrier to enhance the uptake of doxorubicin (DOX), but may also synchronously induce caspases-mediated apoptosis of osteosarcoma by generating intracellular reactive oxygen species (ROS). Furthermore, in vitro and in vivo studies obviously demonstrate that Se-CaP can reverse the MDR of osteosarcoma by down-regulating the expression of MDR-related ABC (ATP binding cassette) transporters proteins (ABCB1 and ABCC1). Finally, DOX-loaded Se-CaP can significantly inhibit DOX-resistant MG63 (MG63/DXR) tumor growth in nude mice. Considering its biomimetic chemical properties, the Se-CaP biomineral, with the multiple functions mentioned above, could be a promising candidate for treating bone tumors with MDR characteristics.
Collapse
|
10
|
Kaur G, Javed W, Ponomarenko O, Shekh K, Swanlund DP, Zhou JR, Summers KL, Casini A, Wenzel MN, Casey JR, Cordat E, Pickering IJ, George GN, Leslie EM. Human red blood cell uptake and sequestration of arsenite and selenite: Evidence of seleno-bis(S-glutathionyl) arsinium ion formation in human cells. Biochem Pharmacol 2020; 180:114141. [PMID: 32652143 DOI: 10.1016/j.bcp.2020.114141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 01/07/2023]
Abstract
Over 200 million people worldwide are exposed to the human carcinogen, arsenic, in contaminated drinking water. In laboratory animals, arsenic and the essential trace element, selenium, can undergo mutual detoxification through the formation of the seleno-bis(S-glutathionyl) arsinium ion [(GS)2AsSe]-, which undergoes biliary and fecal elimination. [(GS)2AsSe]-, formed in animal red blood cells (RBCs), sequesters arsenic and selenium, and slows the distribution of both compounds to peripheral tissues susceptible to toxic effects. In human RBCs, the influence of arsenic on selenium accumulation, and vice versa, is largely unknown. The study aims were to characterize arsenite (AsIII) and selenite (SeIV) uptake by human RBCs, to determine if SeIV and AsIII increase the respective accumulation of the other in human RBCs, and ultimately to determine if this occurs through the formation and sequestration of [(GS)2AsSe]-. 75SeIV accumulation was temperature and Cl--dependent, inhibited by 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonic acid (H2DIDS) (IC50 1 ± 0.2 µM), and approached saturation at 30 µM, suggesting uptake is mediated by the erythrocyte anion-exchanger 1 (AE1 or Band 3, gene SLC4A1). HEK293 cells overexpressing AE1 showed concentration-dependent 75SeIV uptake. 73AsIII uptake by human RBCs was temperature-dependent, partly reduced by aquaglyceroporin 3 inhibitors, and not saturated. AsIII increased 75SeIV accumulation (in the presence of albumin) and SeIV increased 73AsIII accumulation in human RBCs. Near-edge X-ray absorption spectroscopy revealed the formation of [(GS)2AsSe]- in human RBCs exposed to both AsIII and SeIV. The sequestration of [(GS)2AsSe]- in human RBCs potentially slows arsenic distribution to susceptible tissues and could reduce arsenic-induced disease.
Collapse
Affiliation(s)
- Gurnit Kaur
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada
| | - Warda Javed
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Olena Ponomarenko
- Department of Geological Sciences, University of Saskatchewan, Canada
| | - Kamran Shekh
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Diane P Swanlund
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Janet R Zhou
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada
| | - Kelly L Summers
- Department of Geological Sciences, University of Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Canada
| | - Angela Casini
- School of Chemistry, Cardiff University, UK; Department of Chemistry, Technical University of Munich, Germany
| | | | - Joseph R Casey
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada; Department of Biochemistry, University of Alberta, Canada
| | - Emmanuelle Cordat
- Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada
| | - Ingrid J Pickering
- Department of Geological Sciences, University of Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Canada
| | - Graham N George
- Department of Geological Sciences, University of Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Canada
| | - Elaine M Leslie
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Canada; Department of Physiology, University of Alberta, Canada.
| |
Collapse
|
11
|
Kaur G, Ponomarenko O, Zhou JR, Swanlund DP, Summers KL, Dolgova NV, Antipova O, Pickering IJ, George GN, Leslie EM. Studies of selenium and arsenic mutual protection in human HepG2 cells. Chem Biol Interact 2020; 327:109162. [PMID: 32524993 DOI: 10.1016/j.cbi.2020.109162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 10/24/2022]
Abstract
Hundreds of millions of people worldwide are exposed to unacceptable levels of carcinogenic inorganic arsenic. Animal models have shown that selenium and arsenic are mutually protective through the formation and elimination of the seleno-bis(S-glutathionyl) arsinium ion [(GS)2AsSe]-. Consistent with this, human selenium deficiency in arsenic-endemic regions is associated with arsenic-induced disease, leading to the initiation of human selenium supplementation trials. In contrast to the protective effect observed in vivo, in vitro studies have suggested that selenite increases arsenite cellular retention and toxicity. This difference might be explained by the rapid conversion of selenite to selenide in vivo. In the current study, selenite did not protect the human hepatoma (HepG2) cell line against the toxicity of arsenite at equimolar concentrations, however selenide increased the IC50 by 2.3-fold. Cytotoxicity assays of arsenite + selenite and arsenite + selenide at different molar ratios revealed higher overall mutual antagonism of arsenite + selenide toxicity than arsenite + selenite. Despite this protective effect, in comparison to 75Se-selenite, HepG2 cells in suspension were at least 3-fold more efficient at accumulating selenium from reduced 75Se-selenide, and its accumulation was further increased by arsenite. X-ray fluorescence imaging of HepG2 cells also showed that arsenic accumulation, in the presence of selenide, was higher than in the presence of selenite. These results are consistent with a greater intracellular availability of selenide relative to selenite for protection against arsenite, and the formation and retention of a less toxic product, possibly [(GS)2AsSe]-.
Collapse
Affiliation(s)
- Gurnit Kaur
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Olena Ponomarenko
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Janet R Zhou
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Diane P Swanlund
- Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Kelly L Summers
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Natalia V Dolgova
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Olga Antipova
- Advanced Photon Source, Argonne National Laboratory, Argonne, Illinois, USA
| | - Ingrid J Pickering
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Graham N George
- Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elaine M Leslie
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
12
|
Hirano S. Biotransformation of arsenic and toxicological implication of arsenic metabolites. Arch Toxicol 2020; 94:2587-2601. [PMID: 32435915 DOI: 10.1007/s00204-020-02772-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
Arsenic is a well-known environmental carcinogen and chronic exposure to arsenic through drinking water has been reported to cause skin, bladder and lung cancers, with arsenic metabolites being implicated in the pathogenesis. In contrast, arsenic trioxide (As2O3) is an effective therapeutic agent for the treatment of acute promyelocytic leukemia, in which the binding of arsenite (iAsIII) to promyelocytic leukemia (PML) protein is the proposed initial step. These findings on the two-edged sword characteristics of arsenic suggest that after entry into cells, arsenic reaches the nucleus and triggers various nuclear events. Arsenic is reduced, conjugated with glutathione, and methylated in the cytosol. These biotransformations, including the production of reactive metabolic intermediates, appear to determine the intracellular dynamics, target organs, and biological functions of arsenic.
Collapse
Affiliation(s)
- Seishiro Hirano
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305-8506, Japan.
| |
Collapse
|
13
|
Sobh A, Loguinov A, Yazici GN, Zeidan RS, Tagmount A, Hejazi NS, Hubbard AE, Zhang L, Vulpe CD. Functional Profiling Identifies Determinants of Arsenic Trioxide Cellular Toxicity. Toxicol Sci 2020; 169:108-121. [PMID: 30815697 DOI: 10.1093/toxsci/kfz024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Arsenic exposure is a worldwide health concern associated with an increased risk of skin, lung, and bladder cancer but arsenic trioxide (AsIII) is also an effective chemotherapeutic agent. The current use of AsIII in chemotherapy is limited to acute promyelocytic leukemia (APL). However, AsIII was suggested as a potential therapy for other cancer types including chronic myeloid leukemia (CML), especially when combined with other drugs. Here, we carried out a genome-wide CRISPR-based approach to identify modulators of AsIII toxicity in K562, a human CML cell line. We found that disruption of KEAP1, the inhibitory partner of the key antioxidant transcription factor Nrf2, or TXNDC17, a thioredoxin-like protein, markedly increased AsIII tolerance. Loss of the water channel AQP3, the zinc transporter ZNT1 and its regulator MTF1 also enhanced tolerance to AsIII whereas loss of the multidrug resistance protein ABCC1 increased sensitivity to AsIII. Remarkably, disruption of any of multiple genes, EEFSEC, SECISBP2, SEPHS2, SEPSECS, and PSTK, encoding proteins involved in selenocysteine metabolism increased resistance to AsIII. Our data suggest a model in which an intracellular interaction between selenium and AsIII may impact intracellular AsIII levels and toxicity. Together this work revealed a suite of cellular components/processes which modulate the toxicity of AsIII in CML cells. Targeting such processes simultaneously with AsIII treatment could potentiate AsIII in CML therapy.
Collapse
Affiliation(s)
- Amin Sobh
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida.,Department of Nutritional Sciences & Toxicology, Comparative Biochemistry Program, University of California, Berkeley, Berkeley, California
| | - Alex Loguinov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Gulce Naz Yazici
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida.,Department of Histology and Embryology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Rola S Zeidan
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Nima S Hejazi
- Division of Biostatistics and Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, California.,Center for Computational Biology, University of California, Berkeley, Berkeley, California
| | - Alan E Hubbard
- Division of Biostatistics and Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, California
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, California
| | - Chris D Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida.,Department of Nutritional Sciences & Toxicology, Comparative Biochemistry Program, University of California, Berkeley, Berkeley, California
| |
Collapse
|
14
|
Painefilú JC, Pascual MM, Bieczynski F, Laspoumaderes C, González C, Villanueva SSM, Luquet CM. Ex vivo and in vivo effects of arsenite on GST and ABCC2 activity and expression in the middle intestine of the rainbow trout Oncorhynchus mykiss. Comp Biochem Physiol C Toxicol Pharmacol 2019; 225:108566. [PMID: 31301398 DOI: 10.1016/j.cbpc.2019.108566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/07/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
In fish of freshwaters environments, the accumulation and toxic effects of arsenite (AsIII) can be attenuated by detoxification proteins such as GST and ABCC transporters. We studied the effects of AsIII on the middle intestine of O. mykiss in ex-vivo and in vivo/ex vivo assays. For the ex vivo assays, we measured the transport rate of the ABCC substrate DNP-SG and GST activity in intestinal strips and everted sacs. AsIII inhibited DNP-SG transport in a concentration-dependent manner, specifically when we applied it on the basolateral side. GST activity increased when we applied a maximum concentration of AsIII. For the in vivo/ex vivo assays, we kept fish in water with or without 7.7 μmol L-1 of AsIII for 48 h. Then, we measured DNP-SG transport rate, GST activity, and PP1 activity in intestine strips during one hour. For PP1 activity, we incubated the strips with or without microcystin-LR (MCLR), a toxin excreted through ABCC2 proteins. We also analyzed Abcc2 and Gst-π mRNA expression in intestine and liver tissue. In the group exposed in vivo to AsIII, DNP-SG transport rate and GST activity were higher and the effect of MCLR over PP1 activity was attenuated. AsIII significantly induced only Abcc2 mRNA expression in both middle intestine and liver. Our results suggest that, in the middle intestine of O. mykiss, AsIII is absorbed mainly at the basolateral side of the enterocytes, excreted to the lumen by ABCC2 transporters, and is capable of modulating Abcc2 mRNA expression by a transcriptional mechanism.
Collapse
Affiliation(s)
- Julio C Painefilú
- Laboratorio de Ecotoxicología Acuática, INIBIOMA (CONICET-UNCo) - CEAN, ruta provincial no. 61, km 3, CCP 7, Junín de los Andes, 8371 Neuquén, Argentina
| | - Mariano M Pascual
- Laboratorio de Ecotoxicología Acuática, INIBIOMA (CONICET-UNCo) - CEAN, ruta provincial no. 61, km 3, CCP 7, Junín de los Andes, 8371 Neuquén, Argentina
| | - Flavia Bieczynski
- Instituto de Biotecnologia Agropecuaria del Comahue, Sub-Sede Ibac, CITAAC (CONICET-UNCo) Universidad Nacional del Comahue, Buenos Aires 1400. Neuquén 8300, Argentina
| | - Cecilia Laspoumaderes
- Laboratorio de Limnología, INIBIOMA, CONICET-Universidad Nacional del Comahue, Quintral 1250, 8400, San Carlos de Bariloche, Rìo Negro, Argentina; Shelf Sea System Ecology, Alfred-Wegener-Institut, Helmholtz-Zentrum für Polar- und Meeresforschung (AWI), Biologische Anstalt Helgoland, Helgoland, Germany
| | - Carolina González
- C I Agua y Saneamientos Argentinos, Tucumán 752, 1049 Buenos Aires, Argentina; Laboratorio de Limnología, Facultad de Ciencias Exactas y Naturales, UBA, Argentina
| | - Silvina S M Villanueva
- Instituto de Fisiología Experimental, IFISE-CONICET, Facultad de Ciencias Bioquimicas y Farmaceúticas, Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Santa Fe, Argentina
| | - Carlos M Luquet
- Laboratorio de Ecotoxicología Acuática, INIBIOMA (CONICET-UNCo) - CEAN, ruta provincial no. 61, km 3, CCP 7, Junín de los Andes, 8371 Neuquén, Argentina.
| |
Collapse
|
15
|
Maimaitiyiming Y, Wang C, Xu S, Islam K, Chen YJ, Yang C, Wang QQ, Naranmandura H. Role of arsenic (+3 oxidation state) methyltransferase in arsenic mediated APL treatment: an in vitro investigation. Metallomics 2019; 10:828-837. [PMID: 29774349 DOI: 10.1039/c8mt00057c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Arsenic (+3 oxidation state) methyltransferase (AS3MT) is a key enzyme responsible for arsenic metabolism in humans, which facilitates conversion of arsenic trioxide (As2O3) to more reactive metabolites such as monomethylarsonous acid (MMAIII) and dimethylarsinous acid (DMAIII). However, it is unclear whether the biotransformation of arsenic by AS3MT contributes to the promotion of acute promyelocytic leukemia (APL) therapy. In order to understand the probable role of AS3MT in APL patients, we evaluated the effects of arsenite (iAsIII) and three mixed arsenicals (i.e., iAsIII, MMAIII and DMAIII, to mimic active arsenic species in the blood) on NB4 cell differentiation and apoptosis. Although the mixed arsenicals exhibited about 2 fold less effect on the induction of NB4 cell differentiation and PML-RARα fusion protein degradation, they showed 5 times stronger ability to induce apoptosis when compared with iAsIII. More importantly, the proliferation of NB4 cells was significantly (p < 0.05) inhibited in a transwell system co-cultured with AS3MT-transfected HepG2 cells after exposure to iAsIII, suggesting that the generation of methylated metabolites restrained cell proliferation. These findings indicate that the therapeutic efficacy of As2O3 (i.e., iAsIII) in APL patients is probably associated with the production of methylated arsenic metabolites (i.e., MMAIII and DMAIII) by AS3MT.
Collapse
Affiliation(s)
- Yasen Maimaitiyiming
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Stajnko A, Šlejkovec Z, Mazej D, France-Štiglic A, Briški AS, Prpić I, Špirić Z, Horvat M, Falnoga I. Arsenic metabolites; selenium; and AS3MT, MTHFR, AQP4, AQP9, SELENOP, INMT, and MT2A polymorphisms in Croatian-Slovenian population from PHIME-CROME study. ENVIRONMENTAL RESEARCH 2019; 170:301-319. [PMID: 30612060 DOI: 10.1016/j.envres.2018.11.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 06/09/2023]
Abstract
The relationships between inorganic arsenic (iAs) metabolism, selenium (Se) status, and genetic polymorphisms of various genes, commonly studied in populations exposed to high levels of iAs from drinking water, were studied in a Croatian-Slovenian population from the wider PHIME-CROME project. Population consisted of 136 pregnant women in the 3rd trimester and 176 non-pregnant women with their children (n = 176, 8-9 years old). Their exposure to iAs, defined by As (speciation) analyses of biological samples, was low. The sums of biologically active metabolites (arsenite + arsenate + methylated As forms) for pregnant women, non-pregnant women, and children, respectively were: 3.23 (2.84-3.68), 1.83 (1.54-2.16) and 2.18 (1.86-2.54) ng/mLSG; GM (95 CI). Corresponding plasma Se levels were: 54.8 (52.8-56.9), 82.3 (80.4-84.0) and 65.8 (64.3-67.3) ng/mL; GM (95 CI). As methylation efficiency indexes confirmed the relationship between pregnancy/childhood and better methylation efficiency. Archived blood and/or saliva samples were used for single nucleotide polymorphism (SNP) genotyping of arsenic(3+) methyltransferase - AS3MT (rs7085104, rs3740400, rs3740393, rs3740390, rs11191439, rs10748835, rs1046778 and the corresponding AS3MT haplotype); methylene tetrahydrofolate reductase - MTHFR (rs1801131, rs1801133); aquaporin - AQP 4 and 9 (rs9951307 and rs2414539); selenoprotein P1 - SELENOP (rs7579, rs3877899); indolethylamine N-methyltransferase - INMT (rs6970396); and metallothionein 2A - MT2A (rs28366003). Associations of SNPs with As parameters and urine Se were determined through multiple regression analyses adjusted using appropriate confounders (blood As, plasma Se, ever smoking, etc.). SNPs' influence on As methylation, defined particularly by the secondary methylation index (SMI), confirmed the 'protective' role of minor alleles of six AS3MT SNPs and their haplotype only among non-pregnant women. Among the other investigated genes, the carriers of AQP9 (rs2414539) were associated with more efficient As methylation and higher urine concentration of As and Se among non-pregnant women; poorer methylation was observed for carriers of AQP4 (rs9951307) among pregnant women and SELENOP (rs7579) among non-pregnant women; MT2A (rs28366003) was associated with higher urine concentration of AsIII regardless of the pregnancy status; and INMT (rs6970396) was associated with higher As and Se concentration in non-pregnant women. Among confounders, the strongest influence was observed for plasma Se; it reduced urine AsIII concentration during pregnancy and increased secondary methylation index among non-pregnant women. In the present study of populations with low As exposure, we observed a few new As-gene associations (particularly with AQPs). More reliable interpretations will be possible after their confirmation in larger populations with higher As exposure levels.
Collapse
Affiliation(s)
- Anja Stajnko
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana, Slovenia
| | - Zdenka Šlejkovec
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia
| | - Darja Mazej
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia
| | - Alenka France-Štiglic
- Institute of Clinical Chemistry and Biochemistry, University Medical Centre Ljubljana, Njegoševa 4, Ljubljana, Slovenia
| | - Alenka Sešek Briški
- Institute of Clinical Chemistry and Biochemistry, University Medical Centre Ljubljana, Njegoševa 4, Ljubljana, Slovenia
| | - Igor Prpić
- Department of Pediatrics, University Hospital Centre Rijeka, Krešimirova 42, Rijeka, Croatia; Faculty of Medicine, University of Rijeka, Ul. Braće Branchetta 20/1, Rijeka, Croatia
| | - Zdravko Špirić
- Green infrastructure ltd., Fallerovo šetalište 22, Zagreb, Croatia
| | - Milena Horvat
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana, Slovenia
| | - Ingrid Falnoga
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Redox metabolism of ingested arsenic: Integrated activities of microbiome and host on toxicological outcomes. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2018.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
18
|
Vähäkangas K, Loikkanen J, Sahlman H, Karttunen V, Repo J, Sieppi E, Kummu M, Huuskonen P, Myöhänen K, Storvik M, Pasanen M, Myllynen P, Pelkonen O. Biomarkers of Toxicity in Human Placenta. BIOMARKERS IN TOXICOLOGY 2019:303-339. [DOI: 10.1016/b978-0-12-814655-2.00018-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Skröder H, Kippler M, De Loma J, Raqib R, Vahter M. Predictors of selenium biomarker kinetics in 4-9-year-old Bangladeshi children. ENVIRONMENT INTERNATIONAL 2018; 121:842-851. [PMID: 30342415 DOI: 10.1016/j.envint.2018.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Biomarker selenium concentrations vary greatly between studies. Concentrations in erythrocytes, urine, and hair vary even at similar plasma concentrations, suggesting that unknown factors influence the distribution of selenium between body compartments. OBJECTIVE To assess predictors of the different selenium biomarkers in children. DESIGN We used a mother-child cohort, nested in a population-based supplementation trial in rural Bangladesh (MINIMat), established for evaluation of arsenic toxicity. Selenium was measured in plasma (n = 223), erythrocytes, urine, and hair at 9 years (n = 395) and in erythrocytes and urine at 4.5 years (n = 259) using inductively coupled plasma mass spectrometry. We also measured concentrations of arsenic (all biospecimen) and cadmium (erythrocytes and urine). Genotyping for INMT, a methyltransferase involved in selenium metabolism, was performed using TaqMan probes. RESULTS At 9 years, the selenium concentrations ranged 51-139 μg/L in plasma, 128-281 μg/L in erythrocytes, 2.2-55 μg/L in urine, and 258-723 μg/kg in hair. Correlations (rS) between biomarkers ranged 0.12-0.37, and were strongest between blood compartments and between erythrocytes and hair (long-term markers). In multivariable-adjusted linear regression analyses, plasma selenium differed by sampling season (highest in food-secure pre-monsoon season) and was inversely associated with plasma arsenic (range < 0.0080-20 μg/L; B = -1.1, 95% CI: -1.8, -0.41). In contrast, erythrocyte selenium was positively associated with erythrocyte arsenic (range 0.95-50 μg/L; B = 0.58, 95% CI: 0.26, 0.91) and inversely associated with erythrocyte cadmium (range 0.27-3.1 μg/L; B = -12, 95% CI: -17, -6.9). These associations were similar at 4.5 years. Only selenium in hair and urine were influenced by INMT polymorphisms. Finally, chronic malnutrition seemed to increase selenium retention, measured as the ratio plasma/urinary selenium. CONCLUSIONS Selenium biomarkers seem to be influenced by malnutrition, genetics, and exposure to metal pro-oxidants. This might affect the evaluation of deficiency/sufficiency, normally assessed by selenium in plasma/serum.
Collapse
Affiliation(s)
- Helena Skröder
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Maria Kippler
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Jessica De Loma
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Rubhana Raqib
- International Center for Diarrheal Disease Research, GPO Box 128, Dhaka 1000, Bangladesh
| | - Marie Vahter
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
20
|
Sarpong-Kumankomah S, Gibson MA, Gailer J. Organ damage by toxic metals is critically determined by the bloodstream. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
21
|
Banerjee M, Kaur G, Whitlock BD, Carew MW, Le XC, Leslie EM. Multidrug Resistance Protein 1 (MRP1/ABCC1)-Mediated Cellular Protection and Transport of Methylated Arsenic Metabolites Differs between Human Cell Lines. Drug Metab Dispos 2018; 46:1096-1105. [DOI: 10.1124/dmd.117.079640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/09/2018] [Indexed: 12/28/2022] Open
|
22
|
Molin M, Ulven SM, Dahl L, Lundebye AK, Holck M, Alexander J, Meltzer HM, Ydersbond TA. Arsenic in seafood is associated with increased thyroid-stimulating hormone (TSH) in healthy volunteers - A randomized controlled trial. J Trace Elem Med Biol 2017; 44:1-7. [PMID: 28965562 DOI: 10.1016/j.jtemb.2017.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Exposure to exogenous elements like arsenic (As) may influence thyroid enzymes, thyroid-stimulating hormone (TSH), and the two principal thyroid hormones, free thyroxine (FT4) and free triiodothyronine (FT3), but little is known about how this is related to organic arsenicals, the main form in seafood. AIM To investigate whether a high intake of dietary arsenic from seafood can impact thyroid function and thyroid hormones by examining possible associations with changes in TSH, FT4, FT3 and the FT4:FT3-ratio in plasma. METHODS Thirty-eight healthy subjects were randomized into four groups. During a 14-day semi-controlled dietary study, the subjects ingested daily portions of either 150g cod, salmon, blue mussels or potato (control). Plasma concentrations of total As, FT3, FT4, TSH and selenium (Se), and urinary concentrations of iodine were monitored. RESULTS Plasma concentrations of TSH increased significantly in all seafood groups. The change in plasma As, with different coefficients for each seafood group, was the dominant factor in the optimal multiple regression model for change in TSH (R2=0.47). Plasma Se and iodine were negative and positive factors, respectively. There were also indications of changes in FT4, FT3 and the FT4:FT3 ratio consistent with a net inhibiting effect of As on FT4 to FT3 conversion. CONCLUSION Ingestion of seafood rich in various organic As species was strongly associated with an increase of the TSH concentrations in plasma. Change in TSH was positively associated with total plasma As, but varied with the type of seafood ingested. These findings indicate that organic dietary As, apparently depending on chemical form, may influence thyroid hormones and function.
Collapse
Affiliation(s)
- M Molin
- Oslo and Akershus University College of Applied Sciences, PO Box 4 St. Olavs Plass, N-0130 Oslo, Norway; Bjorknes University College, Lovisenberggata 13, N-0456 Oslo, Norway.
| | - S M Ulven
- Oslo and Akershus University College of Applied Sciences, PO Box 4 St. Olavs Plass, N-0130 Oslo, Norway; Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, PO Box 1046 Blindern, 0317 Oslo, Norway
| | - L Dahl
- National Institute of Nutrition and Seafood Research, PO Box 2029 Nordnes, N-5817 Bergen, Norway
| | - A-K Lundebye
- National Institute of Nutrition and Seafood Research, PO Box 2029 Nordnes, N-5817 Bergen, Norway
| | - M Holck
- Oslo and Akershus University College of Applied Sciences, PO Box 4 St. Olavs Plass, N-0130 Oslo, Norway
| | - J Alexander
- Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403 Oslo, Norway
| | - H M Meltzer
- Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403 Oslo, Norway
| | - T A Ydersbond
- Statistics Norway, PO Box 8131 Dep, N-0033 Oslo, Norway
| |
Collapse
|
23
|
Ponomarenko O, La Porte PF, Singh SP, Langan G, Fleming DEB, Spallholz JE, Alauddin M, Ahsan H, Ahmed S, Gailer J, George GN, Pickering IJ. Selenium-mediated arsenic excretion in mammals: a synchrotron-based study of whole-body distribution and tissue-specific chemistry. Metallomics 2017; 9:1585-1595. [PMID: 29058732 DOI: 10.1039/c7mt00201g] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Arsenicosis, a syndrome caused by ingestion of arsenic contaminated drinking water, currently affects millions of people in South-East Asia and elsewhere. Previous animal studies revealed that the toxicity of arsenite essentially can be abolished if selenium is co-administered as selenite. Although subsequent studies have provided some insight into the biomolecular basis of this striking antagonism, many details of the biochemical pathways that ultimately result in the detoxification and excretion of arsenic using selenium supplements have yet to be thoroughly studied. To this end and in conjunction with the recent Phase III clinical trial "Selenium in the Treatment of Arsenic Toxicity and Cancers", we have applied synchrotron X-ray techniques to elucidate the mechanisms of this arsenic-selenium antagonism at the tissue and organ levels using an animal model. X-ray fluorescence imaging (XFI) of cryo-dried whole-body sections of laboratory hamsters that had been injected with arsenite, selenite, or both chemical species, provided insight into the distribution of both metalloids 30 minutes after treatment. Co-treated animals showed strong co-localization of arsenic and selenium in the liver, gall bladder and small intestine. X-ray absorption spectroscopy (XAS) of freshly frozen organs of co-treated animals revealed the presence in liver tissues of the seleno bis-(S-glutathionyl) arsinium ion, which was rapidly excreted via bile into the intestinal tract. These results firmly support the previously postulated hepatobiliary excretion of the seleno bis-(S-glutathionyl) arsinium ion by providing the first data pertaining to organs of whole animals.
Collapse
Affiliation(s)
- Olena Ponomarenko
- Molecular and Environmental Science Research Group, Department of Geological Sciences, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E2, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Transporters in proximal renal tubules contribute to the disposition of numerous drugs. Furthermore, the molecular mechanisms of tubular secretion have been progressively elucidated during the past decades. Organic anions tend to be secreted by the transport proteins OAT1, OAT3 and OATP4C1 on the basolateral side of tubular cells, and multidrug resistance protein (MRP) 2, MRP4, OATP1A2 and breast cancer resistance protein (BCRP) on the apical side. Organic cations are secreted by organic cation transporter (OCT) 2 on the basolateral side, and multidrug and toxic compound extrusion (MATE) proteins MATE1, MATE2/2-K, P-glycoprotein, organic cation and carnitine transporter (OCTN) 1 and OCTN2 on the apical side. Significant drug-drug interactions (DDIs) may affect any of these transporters, altering the clearance and, consequently, the efficacy and/or toxicity of substrate drugs. Interactions at the level of basolateral transporters typically decrease the clearance of the victim drug, causing higher systemic exposure. Interactions at the apical level can also lower drug clearance, but may be associated with higher renal toxicity, due to intracellular accumulation. Whereas the importance of glomerular filtration in drug disposition is largely appreciated among clinicians, DDIs involving renal transporters are less well recognized. This review summarizes current knowledge on the roles, quantitative importance and clinical relevance of these transporters in drug therapy. It proposes an approach based on substrate-inhibitor associations for predicting potential tubular-based DDIs and preventing their adverse consequences. We provide a comprehensive list of known drug interactions with renally-expressed transporters. While many of these interactions have limited clinical consequences, some involving high-risk drugs (e.g. methotrexate) definitely deserve the attention of prescribers.
Collapse
Affiliation(s)
- Anton Ivanyuk
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland.
| | - Françoise Livio
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Jérôme Biollaz
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Thierry Buclin
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| |
Collapse
|
25
|
Orr SE, Bridges CC. Chronic Kidney Disease and Exposure to Nephrotoxic Metals. Int J Mol Sci 2017; 18:ijms18051039. [PMID: 28498320 PMCID: PMC5454951 DOI: 10.3390/ijms18051039] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/26/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic kidney disease (CKD) is a common progressive disease that is typically characterized by the permanent loss of functional nephrons. As injured nephrons become sclerotic and die, the remaining healthy nephrons undergo numerous structural, molecular, and functional changes in an attempt to compensate for the loss of diseased nephrons. These compensatory changes enable the kidney to maintain fluid and solute homeostasis until approximately 75% of nephrons are lost. As CKD continues to progress, glomerular filtration rate decreases, and remaining nephrons are unable to effectively eliminate metabolic wastes and environmental toxicants from the body. This inability may enhance mortality and/or morbidity of an individual. Environmental toxicants of particular concern are arsenic, cadmium, lead, and mercury. Since these metals are present throughout the environment and exposure to one or more of these metals is unavoidable, it is important that the way in which these metals are handled by target organs in normal and disease states is understood completely.
Collapse
Affiliation(s)
- Sarah E Orr
- Mercer University School of Medicine, Division of Basic Medical Sciences, 1550 College St., Macon, GA 31207, USA.
| | - Christy C Bridges
- Mercer University School of Medicine, Division of Basic Medical Sciences, 1550 College St., Macon, GA 31207, USA.
| |
Collapse
|
26
|
Rahman MT, De Ley M. Arsenic Induction of Metallothionein and Metallothionein Induction Against Arsenic Cytotoxicity. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 240:151-168. [PMID: 27115674 DOI: 10.1007/398_2016_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Human exposure to arsenic (As) can lead to oxidative stress that can become evident in organs such as the skin, liver, kidneys and lungs. Several intracellular antioxidant defense mechanisms including glutathione (GSH) and metallothionein (MT) have been shown to minimize As cytotoxicity. The current review summarizes the involvement of MT as an intracellular defense mechanism against As cytotoxicity, mostly in blood. Zinc (Zn) and selenium (Se) supplements are also proposed as a possible remediation of As cytotoxicity. In vivo and in vitro studies on As toxicity were reviewed to summarize cytotoxic mechanisms of As. Intracellular antioxidant defense mechanisms of MT are linked in relation to As cytotoxicity. Arsenic uses a different route, compared to major metal MT inducers such as Zn, to enter/exit blood cells. A number of in vivo and in vitro studies showed that upregulated MT biosynthesis in blood components are related to toxic levels of As. Despite the cysteine residues in MT that aid to bind As, MT is not the preferred binding protein for As. Nonetheless, intracellular oxidative stress due to As toxicity can be minimized, if not eliminated, by MT. Thus MT induction by essential metals such as Zn and Se supplementation could be beneficial to fight against As toxicity.
Collapse
Affiliation(s)
| | - Marc De Ley
- Laboratorium voor Biochemie, Katholieke Universiteit Leuven, Celestijnenlaan 200G, Postbus 2413, 3001, Leuven-Heverlee, Belgium
| |
Collapse
|
27
|
Roggenbeck BA, Banerjee M, Leslie EM. Cellular arsenic transport pathways in mammals. J Environ Sci (China) 2016; 49:38-58. [PMID: 28007179 DOI: 10.1016/j.jes.2016.10.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 06/06/2023]
Abstract
Natural contamination of drinking water with arsenic results in the exposure of millions of people world-wide to unacceptable levels of this metalloid. This is a serious global health problem because arsenic is a Group 1 (proven) human carcinogen and chronic exposure is known to cause skin, lung, and bladder tumors. Furthermore, arsenic exposure can result in a myriad of other adverse health effects including diseases of the cardiovascular, respiratory, neurological, reproductive, and endocrine systems. In addition to chronic environmental exposure to arsenic, arsenic trioxide is approved for the clinical treatment of acute promyelocytic leukemia, and is in clinical trials for other hematological malignancies as well as solid tumors. Considerable inter-individual variability in susceptibility to arsenic-induced disease and toxicity exists, and the reasons for such differences are incompletely understood. Transport pathways that influence the cellular uptake and export of arsenic contribute to regulating its cellular, tissue, and ultimately body levels. In the current review, membrane proteins (including phosphate transporters, aquaglyceroporin channels, solute carrier proteins, and ATP-binding cassette transporters) shown experimentally to contribute to the passage of inorganic, methylated, and/or glutathionylated arsenic species across cellular membranes are discussed. Furthermore, what is known about arsenic transporters in organs involved in absorption, distribution, and metabolism and how transport pathways contribute to arsenic elimination are described.
Collapse
Affiliation(s)
- Barbara A Roggenbeck
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| | - Mayukh Banerjee
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Elaine M Leslie
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB, T6G 2H7, Canada; Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2G3, Canada.
| |
Collapse
|
28
|
Polymorphic variants of MRP4/ABCC4 differentially modulate the transport of methylated arsenic metabolites and physiological organic anions. Biochem Pharmacol 2016; 120:72-82. [PMID: 27659809 DOI: 10.1016/j.bcp.2016.09.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/16/2016] [Indexed: 01/17/2023]
Abstract
Broad inter-individual variation exists in susceptibility to arsenic-induced tumours, likely involving differences in the ability of individuals to eliminate this metalloid. We recently identified human multidrug resistance protein 4 (MRP4/ABCC4) as a novel pathway for the cellular export of dimethylarsinic acid (DMAV), the major urinary arsenic metabolite in humans, and the diglutathione conjugate of the highly toxic monomethylarsonous acid [MMA(GS)2]. These findings, together with the basolateral and apical membrane localization of MRP4 in hepatocytes and renal proximal tubule cells, respectively, suggest a role for MRP4 in the urinary elimination of hepatic arsenic metabolites. Accordingly, we have now investigated the influence of non-synonymous single nucleotide polymorphisms (SNPs) on MRP4 levels, cellular localization, and arsenical transport. Of eight MRP4 variants (C171G-, G187W-, K304N-, G487E-, Y556C-, E757K-, V776I- and C956S-MRP4) characterized, two (V776I- and C956S-MRP4) did not localize appropriately to the plasma membrane of HEK293T and LLC-PK1 cells. Characterization of the six correctly localized mutants revealed that MMA(GS)2 transport by C171G-, G187W-, and K304N-MRP4 was 180%, 73%, and 30% of WT-MRP4 activity, respectively, whereas DMAV transport by K304N- and Y556C-MRP4 was 30% and 184% of WT-MRP4, respectively. Transport of the prototypical physiological MRP4 substrates prostaglandin E2 and 17β-estradiol 17-(β-d-glucuronide) by the six variants was also differentially affected. Thus, MRP4 variants have differing abilities to transport arsenic and endogenous metabolites through both altered function and membrane localization. Further investigation is warranted to determine if genetic variations in ABCC4 contribute to inter-individual differences in susceptibility to arsenic-induced (and potentially other) diseases.
Collapse
|
29
|
Shukalek CB, Swanlund DP, Rousseau RK, Weigl KE, Marensi V, Cole SPC, Leslie EM. Arsenic Triglutathione [As(GS)3] Transport by Multidrug Resistance Protein 1 (MRP1/ABCC1) Is Selectively Modified by Phosphorylation of Tyr920/Ser921 and Glycosylation of Asn19/Asn23. Mol Pharmacol 2016; 90:127-39. [PMID: 27297967 DOI: 10.1124/mol.116.103648] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/10/2016] [Indexed: 11/22/2022] Open
Abstract
The ATP-binding cassette (ABC) transporter multidrug resistance protein 1 (MRP1/ABCC1) is responsible for the cellular export of a chemically diverse array of xenobiotics and endogenous compounds. Arsenic, a human carcinogen, is a high-affinity MRP1 substrate as arsenic triglutathione [As(GS)3]. In this study, marked differences in As(GS)3 transport kinetics were observed between MRP1-enriched membrane vesicles prepared from human embryonic kidney 293 (HEK) (Km 3.8 µM and Vmax 307 pmol/mg per minute) and HeLa (Km 0.32 µM and Vmax 42 pmol/mg per minute) cells. Mutant MRP1 lacking N-linked glycosylation [Asn19/23/1006Gln; sugar-free (SF)-MRP1] expressed in either HEK293 or HeLa cells had low Km and Vmax values for As(GS)3, similar to HeLa wild-type (WT) MRP1. When prepared in the presence of phosphatase inhibitors, both WT- and SF-MRP1-enriched membrane vesicles had a high Km value for As(GS)3 (3-6 µM), regardless of the cell line. Kinetic parameters of As(GS)3 for HEK-Asn19/23Gln-MRP1 were similar to those of HeLa/HEK-SF-MRP1 and HeLa-WT-MRP1, whereas those of single glycosylation mutants were like those of HEK-WT-MRP1. Mutation of 19 potential MRP1 phosphorylation sites revealed that HEK-Tyr920Phe/Ser921Ala-MRP1 transported As(GS)3 like HeLa-WT-MRP1, whereas individual HEK-Tyr920Phe- and -Ser921Ala-MRP1 mutants were similar to HEK-WT-MRP1. Together, these results suggest that Asn19/Asn23 glycosylation and Tyr920/Ser921 phosphorylation are responsible for altering the kinetics of MRP1-mediated As(GS)3 transport. The kinetics of As(GS)3 transport by HEK-Asn19/23Gln/Tyr920Glu/Ser921Glu were similar to HEK-WT-MRP1, indicating that the phosphorylation-mimicking substitutions abrogated the influence of Asn19/23Gln glycosylation. Overall, these data suggest that cross-talk between MRP1 glycosylation and phosphorylation occurs and that phosphorylation of Tyr920 and Ser921 can switch MRP1 to a lower-affinity, higher-capacity As(GS)3 transporter, allowing arsenic detoxification over a broad concentration range.
Collapse
Affiliation(s)
- Caley B Shukalek
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Diane P Swanlund
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Rodney K Rousseau
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Kevin E Weigl
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Vanessa Marensi
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Susan P C Cole
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| | - Elaine M Leslie
- Department of Physiology (C.B.S., D.P.S., R.K.R., V.M., E.M.L.) and Membrane Protein Disease Research Group (C.B.S., D.P.S., R.K.R., V.M., E.M.L.), University of Alberta, Edmonton, Alberta, Canada. Department of Pathology and Molecular Medicine and Division of Cancer Biology and Genetics (K.E.W., S.P.C.C.), Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
30
|
George GN, Gailer J, Ponomarenko O, La Porte PF, Strait K, Alauddin M, Ahsan H, Ahmed S, Spallholz J, Pickering IJ. Observation of the seleno bis-(S-glutathionyl) arsinium anion in rat bile. J Inorg Biochem 2016; 158:24-29. [DOI: 10.1016/j.jinorgbio.2016.01.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/30/2015] [Accepted: 01/18/2016] [Indexed: 12/01/2022]
|
31
|
Skröder Löveborn H, Kippler M, Lu Y, Ahmed S, Kuehnelt D, Raqib R, Vahter M. Arsenic Metabolism in Children Differs From That in Adults. Toxicol Sci 2016; 152:29-39. [PMID: 27056082 PMCID: PMC4922540 DOI: 10.1093/toxsci/kfw060] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Arsenic toxicity in adults is associated with methylation efficiency, influenced by factors such as gender, genetics, and nutrition. The aim of this study was to evaluate influencing factors for arsenic metabolism in children. For 488 children (9 years), whose mothers participated in a study on arsenic exposure during pregnancy (nested into the MINIMat trial) in rural Bangladesh, we measured urinary concentrations of inorganic arsenic (iAs) and its metabolites methylarsonic acid (MMA) and dimethylarsinic acid (DMA) by HPLC-HG-ICPMS. Methylation efficiency was assessed by relative amounts (%) of the metabolites. We evaluated the impact of factors such as maternal urinary metabolite pattern, arsenic exposure, gender, socioeconomic status, season of sampling, and nutritional factors, including erythrocyte selenium (Ery-Se), and plasma folate and vitamin B12. Children had higher %DMA and lower %iAs in urine compared to their mothers, unrelated to their lower exposure [median urinary arsenic (U-As) 53 vs 78 µg/l]. Surprisingly, selenium status (Ery-Se) was strongly associated with children’s arsenic methylation; an increase in Ery-Se from the 5–95th percentile was associated with: +1.8 percentage points (pp) for %iAs (P = .001), +1.4 pp for %MMA (P = .003), and −3.2 pp for %DMA (P < .001). Despite this, Ery-Se was positively associated with U-As (5–95th percentile: +41 µg/l, P = .026). As expected, plasma folate was inversely associated with %iAs (5–95th percentile: −1.9 pp, P = .001) and positively associated with %DMA (5–95th percentile: +2.2 pp, P = .008). Children methylated arsenic more efficiently than their mothers. Also influencing factors, mainly selenium and folate, differed. This warrants further research.
Collapse
Affiliation(s)
| | - Maria Kippler
- *Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77, Sweden;
| | - Ying Lu
- *Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Sultan Ahmed
- *Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77, Sweden; International Center for Diarrhoeal Disease Research, Bangladesh (Icddr,B), Dhaka 1000, Bangladesh
| | - Doris Kuehnelt
- Institute of Chemistry, Analytical Chemistry, NAWI Graz, University of Graz, Universitaetsplatz 1, Graz 8010, Austria
| | - Rubhana Raqib
- International Center for Diarrhoeal Disease Research, Bangladesh (Icddr,B), Dhaka 1000, Bangladesh
| | - Marie Vahter
- *Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77, Sweden;
| |
Collapse
|
32
|
Fleming DE, Groves JW, Gherase MR, George GN, Pickering IJ, Ponomarenko O, Langan G, Spallholz JE, Alauddin M, Ahsan H, Ahmed S, La Porte PF. Soft tissue measurement of arsenic and selenium in an animal model using portable X-ray fluorescence. Radiat Phys Chem Oxf Engl 1993 2015. [DOI: 10.1016/j.radphyschem.2015.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Yuan B, Yoshino Y, Fukushima H, Markova S, Takagi N, Toyoda H, Kroetz DL. Multidrug resistance-associated protein 4 is a determinant of arsenite resistance. Oncol Rep 2015; 35:147-54. [PMID: 26497925 PMCID: PMC6918809 DOI: 10.3892/or.2015.4343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/19/2015] [Indexed: 12/25/2022] Open
Abstract
Although arsenic trioxide (arsenite, AsIII) has shown a remarkable efficacy in the treatment of acute promyelocytic leukemia patients, multidrug resistance is still a major concern for its clinical use. Multidrug resistance-associated protein 4 (MRP4), which belongs to the ATP-binding cassette (ABC) superfamily of transporters, is localized to the basolateral membrane of hepatocytes and the apical membrane of renal proximal tubule cells. Due to its characteristic localization, MRP4 is proposed as a candidate in the elimination of arsenic and may contribute to resistance to AsIII. To test this hypothesis, stable HEK293 cells overexpressing MRP4 or MRP2 were used to establish the role of these two transporters in AsIII resistance. The IC50 values of AsIII in MRP4 cells were approximately 6-fold higher than those in MRP2 cells, supporting an important role for MRP4 in resistance to AsIII. The capacity of MRP4 to confer resistance to AsIII was further confirmed by a dramatic decrease in the IC50 values with the addition of MK571, an MRP4 inhibitor, and cyclosporine A, a well-known broad-spectrum inhibitor of ABC transporters. Surprisingly, the sensitivity of the MRP2 cells to AsIII was similar to that of the parent cells, although insufficient formation of glutathione and/or Se conjugated arsenic compounds in the MRP2 cells might limit transport. Given that MRP4 is a major contributor to arsenic resistance in vitro, further investigation into the correlation between MRP4 expression and treatment outcome of leukemia patients treated with arsenic-based regimens is warranted.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yuta Yoshino
- Department of Clinical Molecular Genetics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hisayo Fukushima
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Svetlana Markova
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroo Toyoda
- Department of Clinical Molecular Genetics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
34
|
Roggenbeck BA, Carew MW, Charrois GJ, Douglas DN, Kneteman NM, Lu X, Le XC, Leslie EM. Characterization of arsenic hepatobiliary transport using sandwich-cultured human hepatocytes. Toxicol Sci 2015; 145:307-20. [PMID: 25752797 DOI: 10.1093/toxsci/kfv051] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Arsenic is a proven human carcinogen and is associated with a myriad of other adverse health effects. This metalloid is methylated in human liver to monomethylarsonic acid (MMA(V)), monomethylarsonous acid (MMA(III)), dimethylarsinic acid (DMA(V)), and dimethylarsinous acid (DMA(III)) and eliminated predominantly in urine. Hepatic basolateral transport of arsenic species is ultimately critical for urinary elimination; however, these pathways are not fully elucidated in humans. A potentially important human hepatic basolateral transporter is the ATP-binding cassette (ABC) transporter multidrug resistance protein 4 (MRP4/ABCC4) that in vitro is a high-affinity transporter of DMA(V) and the diglutathione conjugate of MMA(III) [MMA(GS)(2)]. In rats, the related canalicular transporter Mrp2/Abcc2 is required for biliary excretion of arsenic as As(GS)(3) and MMA(GS)(2). The current study used sandwich cultured human hepatocytes (SCHH) as a physiological model of human arsenic hepatobiliary transport. Arsenic efflux was detected only across the basolateral membrane for 9 out of 14 SCHH preparations, 5 had both basolateral and canalicular efflux. Basolateral transport of arsenic was temperature- and GSH-dependent and inhibited by the MRP inhibitor MK-571. Canalicular efflux was completely lost after GSH depletion suggesting MRP2-dependence. Treatment of SCHH with As(III) (0.1-1 µM) dose-dependently increased MRP2 and MRP4 levels, but not MRP1, MRP6, or aquaglyceroporin 9. Treatment of SCHH with oltipraz (Nrf2 activator) increased MRP4 levels and basolateral efflux of arsenic. In contrast, oltipraz increased MRP2 levels without increasing biliary excretion. These results suggest arsenic basolateral transport prevails over biliary excretion and is mediated at least in part by MRPs, most likely including MRP4.
Collapse
Affiliation(s)
- Barbara A Roggenbeck
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7 *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Michael W Carew
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7 *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Gregory J Charrois
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Donna N Douglas
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Norman M Kneteman
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Xiufen Lu
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - X Chris Le
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Elaine M Leslie
- *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7 *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7 *Department of Physiology, Membrane Protein Disease Research Group, Department of Laboratory Medicine and Pathology, and Department of Surgery, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| |
Collapse
|
35
|
Tan S, Li H, Jin Y, Yu H. In vitro and in vivo effects of sublethal cadmium on the expression of MT2 and ABCC2 genes in grass carp (Ctenopharyngodon idellus). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2014; 108:258-264. [PMID: 25103569 DOI: 10.1016/j.ecoenv.2014.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 06/03/2023]
Abstract
To gain more knowledge about the physiological regulation of metal pollutant detoxification in grass carp, we examined Cd concentration and its the potential influence on the expression of metallothionein 2 (MT2) and multidrug resistance protein 2 (ABCC2) mRNA in the liver and kidney, using in vitro and in vivo experiments. First, the full-length of MT2 cDNA and partial ABCC2 cDNA was obtained, consisting 183bp and 366bp respectively. In vivo approach, grass carp received 96h exposure of Cd (1/10 LD50), and MT2 and ABCC2 mRNA expression were determined by qRT-PCR. The Cd treatment resulted in an increase of MT2 mRNA level in the liver with Cd accumulation. Nonetheless, the elevation ABCC2 mRNA in the liver was appeared at 48h after Cd exposure, as well as the expression of MT2 and ABCC2 mRNA in the kidney. The in vitro experiment was carried out using the hepatocyte (L86) and nephroblasts (CIK). The qRT-RCR results showed that MT2 and ABCC2 mRNA dramatically increased following Cd exposure (1/10 LD50); however, ABCC2 mRNA expression was suppressed in the L86 cell line at first (6h). In conclusion, this result suggested that both MT2 and ABCC2 mRNA may play important roles in the detoxification of toxic metals, and MT2 gene was more sensitive to Cd induction.
Collapse
Affiliation(s)
- Shuwen Tan
- College of Life Science, Foshan University, No. 1 Xianhu University Road, Nanhai, Foshan, Guangdong 528231, China; Holdone Aquaculture Breeding Limited Company, Foshan, Guangdong 528231, China
| | - Hua Li
- College of Life Science, Foshan University, No. 1 Xianhu University Road, Nanhai, Foshan, Guangdong 528231, China; Holdone Aquaculture Breeding Limited Company, Foshan, Guangdong 528231, China
| | - Ying Jin
- College of Biophotonics, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Hui Yu
- College of Life Science, Foshan University, No. 1 Xianhu University Road, Nanhai, Foshan, Guangdong 528231, China; Holdone Aquaculture Breeding Limited Company, Foshan, Guangdong 528231, China.
| |
Collapse
|
36
|
Pushie MJ, Pickering I, Korbas M, Hackett MJ, George GN. Elemental and chemically specific X-ray fluorescence imaging of biological systems. Chem Rev 2014; 114:8499-541. [PMID: 25102317 PMCID: PMC4160287 DOI: 10.1021/cr4007297] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Indexed: 12/13/2022]
Affiliation(s)
- M. Jake Pushie
- Molecular
and Environmental Sciences Research Group, Department of Geological
Sciences, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Ingrid
J. Pickering
- Molecular
and Environmental Sciences Research Group, Department of Geological
Sciences, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
- Toxicology
Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
- Department
of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| | - Malgorzata Korbas
- Canadian
Light Source Inc., 44
Innovation Boulevard, Saskatoon, SK S7N 2V3, Canada
- Department
of Anatomy and Cell Biology, University
of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Mark J. Hackett
- Molecular
and Environmental Sciences Research Group, Department of Geological
Sciences, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Graham N. George
- Molecular
and Environmental Sciences Research Group, Department of Geological
Sciences, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
- Toxicology
Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
- Department
of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| |
Collapse
|
37
|
Banerjee M, Carew MW, Roggenbeck BA, Whitlock BD, Naranmandura H, Le XC, Leslie EM. A Novel Pathway for Arsenic Elimination: Human Multidrug Resistance Protein 4 (MRP4/ABCC4) Mediates Cellular Export of Dimethylarsinic Acid (DMAV) and the Diglutathione Conjugate of Monomethylarsonous Acid (MMAIII). Mol Pharmacol 2014; 86:168-79. [DOI: 10.1124/mol.113.091314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
38
|
Metabolomic study in plasma, liver and kidney of mice exposed to inorganic arsenic based on mass spectrometry. Anal Bioanal Chem 2014; 406:1455-69. [DOI: 10.1007/s00216-013-7564-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/29/2013] [Accepted: 12/07/2013] [Indexed: 11/25/2022]
|
39
|
Nakano T, Sekine S, Ito K, Horie T. Ezrin regulates the expression of Mrp2/Abcc2 and Mdr1/Abcb1 along the rat small intestinal tract. Am J Physiol Gastrointest Liver Physiol 2013; 305:G807-17. [PMID: 24091598 DOI: 10.1152/ajpgi.00187.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Multidrug resistance-associated protein 2 (MRP2)/ATP-binding cassette protein C2 (ABCC2) and multidrug resistance protein 1 (MDR1)/ABCB1 are well-known efflux transporters located on the brush border membrane of the small intestinal epithelia, where they limit the absorption of a broad range of substrates. The expression patterns of MRP2/ABCC2 and MDR1/ABCB1 along the small intestinal tract are tightly regulated. Several reports have demonstrated the participation of ERM (ezrin/radixin/moesin) proteins in the posttranslational modulation of MRP2/ABCC2 and MDR1/ABCB1, especially with regard to their membrane localization. The present study focused on the in vivo expression profiles of MRP2/ABCC2, MDR1/ABCB1, ezrin, and phosphorylated ezrin to further elucidate the relationship between the efflux transporters and the ERM proteins. The current results showed good correlation between the phosphorylation status of ezrin and Mrp2/Abcc2 expression along the gastrointestinal tract of rats and between the expression profiles of both ezrin and Mdr1/Abcb1 in the small intestine. We also demonstrated the involvement of conventional protein kinase C isoforms in the regulation of ezrin phosphorylation. Furthermore, experiments conducted with wild-type (WT) ezrin and a T567A (Ala substituted Thr) dephosphorylated mutant showed a decrease in membrane surface-localized and total expressed MRP2/ABCC2 in T567A-expressing vs. WT ezrin-expressing Caco-2 cells. In contrast, T567A- and WT-expressing cells both showed an increase in membrane surface-localized and total expressed MDR1/ABCB1. These findings suggest that the phosphorylation status and the expression profile of ezrin differentially direct MRP2/ABCC2 and MDR1/ABCB1 expression, respectively, along the small intestinal tract.
Collapse
Affiliation(s)
- Takafumi Nakano
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, 4-21-2 Nakano, Nakano-ku, Tokyo 164-8530, Japan.
| | | | | | | |
Collapse
|
40
|
Gribble MO, Voruganti VS, Cropp CD, Francesconi KA, Goessler W, Umans JG, Silbergeld EK, Laston SL, Haack K, Kao WHL, Fallin MD, Maccluer JW, Cole SA, Navas-Acien A. SLCO1B1 variants and urine arsenic metabolites in the Strong Heart Family Study. Toxicol Sci 2013; 136:19-25. [PMID: 23970802 DOI: 10.1093/toxsci/kft181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Arsenic species patterns in urine are associated with risk for cancer and cardiovascular diseases. The organic anion transporter coded by the gene SLCO1B1 may transport arsenic species, but its association with arsenic metabolites in human urine has not yet been studied. The objective of this study is to evaluate associations of urine arsenic metabolites with variants in the candidate gene SLCO1B1 in adults from the Strong Heart Family Study. We estimated associations between % arsenic species biomarker traits and 5 single-nucleotide polymorphisms (SNPs) in the SLCO1B1 gene in 157 participants, assuming additive genetics. Linear regression models for each SNP accounted for kinships and were adjusted for sex, body mass index, and study center. The minor allele of rs1564370 was associated with lower %MMA (p = .0003) and higher %DMA (p = .0002), accounting for 8% of the variance for %MMA and 9% for %DMA. The rs1564370 minor allele homozygote frequency was 17% and the heterozygote frequency was 43%. The minor allele of rs2291075 was associated with lower %MMA (p = .0006) and higher %DMA (p = .0014), accounting for 7% of the variance for %MMA and 5% for %DMA. The frequency of rs2291075 minor allele homozygotes was 1% and of heterozygotes was 15%. Common variants in SLCO1B1 were associated with differences in arsenic metabolites in a preliminary candidate gene study. Replication of this finding in other populations and analyses with respect to disease outcomes are needed to determine whether this novel candidate gene is important for arsenic-associated disease risks.
Collapse
Affiliation(s)
- Matthew O Gribble
- * Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Placental ABC transporters, cellular toxicity and stress in pregnancy. Chem Biol Interact 2013; 203:456-66. [PMID: 23524238 DOI: 10.1016/j.cbi.2013.03.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/25/2022]
Abstract
The human placenta, in addition to its roles as a nutrient transfer and endocrine organ, functions as a selective barrier to protect the fetus against the harmful effects of exogenous and endogenous toxins. Members of the ATP-binding cassette (ABC) family of transport proteins limit the entry of xenobiotics into the fetal circulation via vectorial efflux from the placenta to the maternal circulation. Several members of the ABC family, including proteins from the ABCA, ABCB, ABCC and ABCG subfamilies, have been shown to be functional in the placenta with clinically significant roles in xenobiotic efflux. However, recent findings suggest that these transporters also protect placental tissue by preventing the cellular accumulation of cytotoxic compounds such as lipids, sterols and their derivatives. Such protective functions are likely to be particularly important in pregnancies complicated by inflammatory or oxidative stress, where the generation of toxic metabolites is enhanced. For example, ABC transporters have been shown to protect against the harmful effects of hypoxia and oxidative stress through increased expression and efflux of oxysterols and glutathione conjugated xenobiotics. However, this protective capacity may be diminished in response to the same stressors. Several studies in primary human trophoblast cells and animal models have demonstrated decreased expression and activity of placental ABC transporters with inflammatory, oxidative or metabolic stress. Several clinical studies in pregnancies complicated by inflammatory conditions such as preeclampsia and gestational diabetes support these findings, although further studies are required to determine the clinical relevance of the relationships between placental ABC transporter expression and activity, and placental function in stressed pregnancies. Such studies are necessary to fully understand the consequences of pregnancy disorders on placental function and viability in order to optimise pregnancy care and maximise fetal growth and health.
Collapse
|
42
|
Canet MJ, Hardwick RN, Lake AD, Kopplin MJ, Scheffer GL, Klimecki WT, Gandolfi AJ, Cherrington NJ. Altered arsenic disposition in experimental nonalcoholic fatty liver disease. Drug Metab Dispos 2012; 40:1817-24. [PMID: 22699396 DOI: 10.1124/dmd.112.046177] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is represented by a spectrum of liver pathologies ranging from simple steatosis to nonalcoholic steatohepatitis (NASH). Liver damage sustained in the progressive stages of NAFLD may alter the ability of the liver to properly metabolize and eliminate xenobiotics. The purpose of the current study was to determine whether NAFLD alters the disposition of the environmental toxicant arsenic. C57BL/6 mice were fed either a high-fat or a methionine-choline-deficient diet to model simple steatosis and NASH, respectively. At the conclusion of the dietary regimen, all mice were given a single oral dose of either sodium arsenate or arsenic trioxide. Mice with NASH excreted significantly higher levels of total arsenic in urine (24 h) compared with controls. Total arsenic in the liver and kidneys of NASH mice was not altered; however, NASH liver retained significantly higher levels of the monomethyl arsenic metabolite, whereas dimethyl arsenic was retained significantly less in the kidneys of NASH mice. NASH mice had significantly higher levels of the more toxic trivalent form in their urine, whereas the pentavalent form was preferentially retained in the liver of NASH mice. Moreover, hepatic protein expression of the arsenic biotransformation enzyme arsenic (3+ oxidation state) methyltransferase was not altered in NASH animals, whereas protein expression of the membrane transporter multidrug resistance-associated protein 1 was increased, implicating cellular transport rather than biotransformation as a possible mechanism. These results suggest that NASH alters the disposition of arsenical species, which may have significant implications on the overall toxicity associated with arsenic in NASH.
Collapse
Affiliation(s)
- Mark J Canet
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Della Torre C, Zaja R, Loncar J, Smital T, Focardi S, Corsi I. Interaction of ABC transport proteins with toxic metals at the level of gene and transport activity in the PLHC-1 fish cell line. Chem Biol Interact 2012; 198:9-17. [PMID: 22580103 DOI: 10.1016/j.cbi.2012.04.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/09/2012] [Accepted: 04/27/2012] [Indexed: 01/17/2023]
Abstract
The aim of this study was to investigate the interaction of four toxic metals with ABC transport proteins in piscine cell line PLHC-1. Cells were exposed for 24 h to 0.01-1 μM of CdCl(2), HgCl(2), As(2)O(3), or K(2)Cr(2)O(7) and the expression of a series of ABC genes (abcb1, abcc1-4) was determined using qRT-PCR. Using the fluorescent model substrates calcein-AM and monochlorbimane we measured interaction of metals with the transport activity of ABC transporters. P-glycoprotein (P-gp) activity was measured in PLHC-1/dox (P-gp overexpressing cells) while activity and interactions of metals with MRPs was measured in PLHC-1/wt cells. After 24 h exposure, abcc2-4 genes were dose-dependently up-regulated by all metals, while abcb1 and abcc1 were less affected. Up-regulation of abcc2 was more pronounced, with up to 8-fold increase in expression. Abcc3 and abcc4 were moderately inducible by HgCl(2) with 3.3-fold and 2.2-fold, respectively. All metals caused a significant inhibition of both P-gp (2.9- to 4-fold vs. controls) and MRP (1.3- to 1.8-fold) transport activities. Modulation of ABC genes and transport activities was further investigated in PLHC-1/wt cells exposed to 1 μM HgCl(2) for 72 h and in Hg resistant cells selected by long term cultivation of PLHC-1/wt cells in increasing concentrations of HgCl(2). Exposure to HgCl(2) for 72 h induced MRP genes expression and efflux activity. The long term cultivation of PLHC-1/wt cells in HgCl(2), did not cause prolonged up-regulation of the tested abc genes but resulted in higher MRP transport activities as determined by the increased sensitivity of these cells to MK571 (MRP specific inhibitor). Results of the present study indicated specific interaction of metals with selected ABC transport proteins. Modulation of ABC transporters takes place at both transcriptional and functional level. An active involvement of efflux pumps in Hg clearance in fish is suggested.
Collapse
Affiliation(s)
- Camilla Della Torre
- Department of Environmental Sciences G. Sarfatti Siena University, Siena, Italy.
| | | | | | | | | | | |
Collapse
|
44
|
Maciaszczyk-Dziubinska E, Wawrzycka D, Wysocki R. Arsenic and antimony transporters in eukaryotes. Int J Mol Sci 2012; 13:3527-3548. [PMID: 22489166 PMCID: PMC3317726 DOI: 10.3390/ijms13033527] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 02/29/2012] [Accepted: 03/07/2012] [Indexed: 12/27/2022] Open
Abstract
Arsenic and antimony are toxic metalloids, naturally present in the environment and all organisms have developed pathways for their detoxification. The most effective metalloid tolerance systems in eukaryotes include downregulation of metalloid uptake, efflux out of the cell, and complexation with phytochelatin or glutathione followed by sequestration into the vacuole. Understanding of arsenic and antimony transport system is of high importance due to the increasing usage of arsenic-based drugs in the treatment of certain types of cancer and diseases caused by protozoan parasites as well as for the development of bio- and phytoremediation strategies for metalloid polluted areas. However, in contrast to prokaryotes, the knowledge about specific transporters of arsenic and antimony and the mechanisms of metalloid transport in eukaryotes has been very limited for a long time. Here, we review the recent advances in understanding of arsenic and antimony transport pathways in eukaryotes, including a dual role of aquaglyceroporins in uptake and efflux of metalloids, elucidation of arsenic transport mechanism by the yeast Acr3 transporter and its role in arsenic hyperaccumulation in ferns, identification of vacuolar transporters of arsenic-phytochelatin complexes in plants and forms of arsenic substrates recognized by mammalian ABC transporters.
Collapse
Affiliation(s)
- Ewa Maciaszczyk-Dziubinska
- Department of Genetics and Cell Physiology, Institute of Plant Biology, University of Wroclaw, Kanonia 6/8, 50-328 Wroclaw, Poland; E-Mail:
| | - Donata Wawrzycka
- Department of Genetics and Cell Physiology, Institute of Plant Biology, University of Wroclaw, Kanonia 6/8, 50-328 Wroclaw, Poland; E-Mail:
| | - Robert Wysocki
- Department of Genetics and Cell Physiology, Institute of Plant Biology, University of Wroclaw, Kanonia 6/8, 50-328 Wroclaw, Poland; E-Mail:
| |
Collapse
|
45
|
Fardel O, Kolasa E, Le Vee M. Environmental chemicals as substrates, inhibitors or inducers of drug transporters: implication for toxicokinetics, toxicity and pharmacokinetics. Expert Opin Drug Metab Toxicol 2011; 8:29-46. [DOI: 10.1517/17425255.2012.637918] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
46
|
Gailer J. Probing the bioinorganic chemistry of toxic metals in the mammalian bloodstream to advance human health. J Inorg Biochem 2011; 108:128-32. [PMID: 22209021 DOI: 10.1016/j.jinorgbio.2011.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 12/03/2011] [Accepted: 12/05/2011] [Indexed: 11/17/2022]
Abstract
The etiology of numerous grievous human diseases, including Alzheimer's and Parkinson's Disease is not well understood. Conversely, the concentration toxic metals and metalloids, such as As, Cd, Hg and Pb in human blood of the average population is well established, yet we know strikingly little about the role that they might play in the etiology of disease processes. Establishing functional connections between the chronic exposure of humans to these and other inorganic pollutants and the etiology of certain human diseases is therefore viewed by many as one of the greatest challenges in the post-genomic era. Conceptually, this task requires us to uncover hitherto unknown biomolecular mechanisms which must explain how small doses of a toxic metal/metalloid compound (low μg per day) - or mixtures thereof - may eventually result in a particular human disease. The biological complexity that is inherently associated with mammals, however, makes the discovery of these mechanisms a truly monumental task. Recent findings suggest that a better understanding of the bioinorganic chemistry of inorganic pollutants in the mammalian bloodstream represents a fruitful strategy to unravel relevant biomolecular mechanisms. The adverse effect(s) that toxic metals/metalloid compounds exert on the transport of essential ultratrace elements to internal organs appear particularly pertinent. A brief overview of the effect that arsenite and Hg(2+) exert on the mammalian metabolism of selenium is presented.
Collapse
Affiliation(s)
- Jürgen Gailer
- Department of Chemistry, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
47
|
Leslie EM. Arsenic-glutathione conjugate transport by the human multidrug resistance proteins (MRPs/ABCCs). J Inorg Biochem 2011; 108:141-9. [PMID: 22197475 DOI: 10.1016/j.jinorgbio.2011.11.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 10/14/2011] [Accepted: 11/11/2011] [Indexed: 12/13/2022]
Abstract
Millions of people world-wide are chronically exposed to inorganic forms of the environmental toxicant arsenic in drinking water. This has led to a public health crisis because arsenic is a human carcinogen, and causes a myriad of other adverse health effects. In order to prevent and treat arsenic-induced toxicity it is critical to understand the cellular handling of this metalloid. A large body of literature describes the importance of the cellular tripeptide glutathione (γ-Glu-Cys-Gly,GSH/GS) in the excretion of arsenic. The triglutathione conjugate of arsenite [As(III)(GS)(3)] and the diglutathione conjugate of monomethylarsonous acid [MMA(III)(GS)(2)] have been isolated from rat bile and mouse urine, and account for the majority of excreted arsenic, suggesting these are important transportable forms. The ATP-binding cassette (ABC) transporter proteins, multidrug resistance protein 1 (MRP1/ABCC1) and the related protein MRP2 (ABCC2), are thought to play an important role in arsenic detoxification through the cellular efflux of arsenic-GSH conjugates. Current knowledge on the cellular handling of arsenic with a special emphasis on the transport pathways of the arsenic-GSH conjugates As(III)(GS)(3), MMA(III)(GS)(2), and dimethylarsenic glutathione DMA(III)(GS), as well as, the seleno-bis(S-glutathionyl) arsinium ion [(GS)(2)AsSe](-) are reviewed.
Collapse
Affiliation(s)
- Elaine M Leslie
- Department of Physiology, University of Alberta, Edmonton, AB, Canada,
| |
Collapse
|
48
|
Yin J, Meng Q. Use of primary rat hepatocytes in the gel entrapment culture to predictin vivobiliary excretion. Xenobiotica 2011; 42:417-28. [DOI: 10.3109/00498254.2011.633716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Carew MW, Naranmandura H, Shukalek CB, Le XC, Leslie EM. Monomethylarsenic diglutathione transport by the human multidrug resistance protein 1 (MRP1/ABCC1). Drug Metab Dispos 2011; 39:2298-304. [PMID: 21918036 DOI: 10.1124/dmd.111.041673] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ATP-binding cassette (ABC) transporter protein multidrug resistance protein 1 (MRP1; ABCC1) plays an important role in the cellular efflux of the high-priority environmental carcinogen arsenic as a triglutathione conjugate [As(GS)(3)]. Most mammalian cells can methylate arsenic to monomethylarsonous acid (MMA(III)), monomethylarsonic acid (MMA(V)), dimethylarsinous acid (DMA(III)), and dimethylarsinic acid (DMA(V)). The trivalent forms MMA(III) and DMA(III) are more reactive and toxic than their inorganic precursors, arsenite (As(III)) and arsenate (As(V)). The ability of MRP1 to transport methylated arsenicals is unknown and was the focus of the current study. HeLa cells expressing MRP1 (HeLa-MRP1) were found to confer a 2.6-fold higher level of resistance to MMA(III) than empty vector control (HeLa-vector) cells, and this resistance was dependent on GSH. In contrast, MRP1 did not confer resistance to DMA(III), MMA(V), or DMA(V). HeLa-MRP1 cells accumulated 4.5-fold less MMA(III) than HeLa-vector cells. Experiments using MRP1-enriched membrane vesicles showed that transport of MMA(III) was GSH-dependent but not supported by the nonreducing GSH analog, ophthalmic acid, suggesting that MMA(III)(GS)(2) was the transported form. MMA(III)(GS)(2) was a high-affinity, high-capacity substrate for MRP1 with apparent K(m) and V(max) values of 11 μM and 11 nmol mg(-1)min(-1), respectively. MMA(III)(GS)(2) transport was osmotically sensitive and inhibited by several MRP1 substrates, including 17β-estradiol 17-(β-D-glucuronide) (E(2)17βG). MMA(III)(GS)(2) competitively inhibited the transport of E(2)17βG with a K(i) value of 16 μM, indicating that these two substrates have overlapping binding sites. These results suggest that MRP1 is an important cellular protective pathway for the highly toxic MMA(III) and have implications for environmental and clinical exposure to arsenic.
Collapse
Affiliation(s)
- Michael W Carew
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
50
|
Long Y, Li Q, Zhong S, Wang Y, Cui Z. Molecular characterization and functions of zebrafish ABCC2 in cellular efflux of heavy metals. Comp Biochem Physiol C Toxicol Pharmacol 2011; 153:381-91. [PMID: 21266201 DOI: 10.1016/j.cbpc.2011.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/09/2011] [Accepted: 01/18/2011] [Indexed: 11/23/2022]
Abstract
Multidrug-resistance associated protein 2 (MRP2/ABCC2) plays crucial roles in bile formation and detoxification by transporting a wide variety of endogenous compounds and xenobiotics, but its functions in zebrafish (Danio rerio) remain to be characterized. In this study, we obtained the full-length cDNA of zebrafish abcc2, analyzed its expression in developing embryos and adult tissues, investigated its transcriptional response to heavy metals, and evaluated its roles in efflux of heavy metals including cadmium, mercury and lead. Zebrafish abcc2 gene is located on chromosome 13 and composed of 32 exons. The deduced polypeptide of zebrafish ABCC2 consists of 1567 amino acids and possesses most of functional domains and critical residues defined in human ABCC2. Zebrafish abcc2 gene is not maternally expressed and its earliest expression was detected in embryos at 72hpf. In larval zebrafish, abcc2 gene was found to be exclusively expressed in liver, intestine and pronephric tubules. In adult zebrafish, the highest expression of abcc2 gene was found in intestine followed by those in liver and kidney, while relative low expression was detected in brain and muscle. Expression of abcc2 in excretory organs including kidney, liver and intestine of zebrafish larvae was induced by exposure to 0.5μM mercury or 5μM lead. Moreover, exposure to 0.125-1μM of mercury or lead also significantly induced abcc2 expression in these excretory organs of adult zebrafish. Furthermore, overexpression of zebrafish ABCC2 in ZF4 cells and zebrafish embryos decreased the cellular accumulation of heavy metals including cadmium, mercury and lead as determined by MRE (metal responsive element)- or EPRE (electrophile response element)-driven luciferase reporters and atomic absorption spectrometry. These results suggest that zebrafish ABCC2/MRP2 is capable of effluxing heavy metals from cells and may play important roles in the detoxification of toxic metals.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- Biological Transport
- Cell Line
- Dose-Response Relationship, Drug
- Embryo, Nonmammalian/drug effects
- Embryo, Nonmammalian/metabolism
- Female
- Gene Expression Regulation, Developmental/drug effects
- Genes, Reporter
- Larva/drug effects
- Larva/growth & development
- Larva/metabolism
- Male
- Metals, Heavy/administration & dosage
- Metals, Heavy/pharmacokinetics
- Metals, Heavy/toxicity
- Molecular Sequence Data
- Multidrug Resistance-Associated Protein 2
- Multidrug Resistance-Associated Proteins/chemistry
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- Organ Specificity
- Phylogeny
- RNA, Messenger/metabolism
- Random Allocation
- Sequence Alignment
- Water Pollutants, Chemical/administration & dosage
- Water Pollutants, Chemical/pharmacokinetics
- Water Pollutants, Chemical/toxicity
- Zebrafish/growth & development
- Zebrafish/metabolism
- Zebrafish Proteins/chemistry
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Yong Long
- Key Laboratory of Biodiversity and Conservation of Aquatic Organism, Institute of Hydrobiology, Chinese Academy of Sciences, 7 Donghu Rd., Wuhan, Hubei 430072, PR China
| | | | | | | | | |
Collapse
|