1
|
Feijó M, Carvalho TMA, Fonseca LRS, Vaz CV, Pereira BJ, Cavaco JEB, Maia CJ, Duarte AP, Kiss-Toth E, Correia S, Socorro S. Endocrine-disrupting chemicals as prostate carcinogens. Nat Rev Urol 2025:10.1038/s41585-025-01031-9. [PMID: 40379948 DOI: 10.1038/s41585-025-01031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 05/19/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) are natural or synthetic compounds that are ubiquitous in the environment and in daily-usage products and interfere with the normal function of the endocrine system leading to adverse health effects in humans. Exposure to these chemicals might elevate the risk of metabolic disorders, developmental and reproductive defects, and endocrine-related cancers. Prostate cancer is the most common hormone-dependent cancer in men, and the fifth leading cause of cancer-related mortality, partly owing to a lack of knowledge about the mechanisms that lead to aggressive castration-resistant forms. In addition to the dependence of early-stage prostate cancer on androgen actions, the prostate is a target of oestrogenic regulation. This hormone dependence, along with the fact that exogenous influences are major risk factors for prostate cancer, make the prostate a likely target of harmful actions from EDCs. Various sources of EDCs and their different modes of action might explain their role in prostate carcinogenesis.
Collapse
Affiliation(s)
- Mariana Feijó
- RISE-Health, Department of Chemistry, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- RISE-Health, Department of Chemistry, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Bruno J Pereira
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
- Instituto Português de Oncologia de Coimbra, Coimbra, Portugal
| | - José Eduardo B Cavaco
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cláudio J Maia
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ana P Duarte
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Endre Kiss-Toth
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Sara Correia
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| | - Sílvia Socorro
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
2
|
Plešnik H, Rekar Ž, Stevanović S, Virant-Klun I, Imamović Kumalić S, Sladič M, Mazej D, Tratnik JS, Horvat M, Kosjek T. Nontargeted Urinary Profiling Strategy for Endocrine-Disrupting Chemicals in Women with Ovarian Malignancies. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8380-8390. [PMID: 40263667 PMCID: PMC12060279 DOI: 10.1021/acs.est.4c13290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
Endocrine-disrupting chemicals (EDCs), including known and unknown parent compounds, their metabolites, and transformation products, are pervasive in daily life, posing increasing risks to human health and the environment. This study employed a high-resolution mass spectrometry-based nontargeted screening approach, integrating polar (HILIC) and reversed-phase separations to expand the chemical space coverage and, supported by open-science tools and resources, evaluated urinary chemical profiles to assess internal EDC exposure. Among 106 annotated biomarkers of exposure, six exhibited significantly higher normalized intensities in patients with ovarian malignancies compared to healthy controls (p < 0.05). This suggests their greater exposure to phthalates (diethylhexyl phthalate and diethyl phthalate), pesticides (metolachlor metabolite and 4-nitrophenol), a UV filter (benzophenone-1), and an industrial byproduct (4-methyl-2-nitrophenol). These compounds may interfere with hormonal regulation, potentially contributing to cancer development. While these findings highlight potential differences in internal EDC exposure, the study primarily demonstrates the applicability of nontargeted urinary profiling for chemical exposure assessment. By providing new insights into EDCs burden and its pathological implications, this work contributes to advancing next-generation chemical risk assessment within the European Partnership for the Assessment of Risks from Chemicals initiative and supports the development of preventive strategies to mitigate environmental cancer risks.
Collapse
Affiliation(s)
- Helena Plešnik
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
- Jožef
Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Žan Rekar
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
- Jožef
Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Stefanela Stevanović
- Faculty
of Computer and Information Science, University
of Ljubljana, 1000 Ljubljana, Slovenia
| | - Irma Virant-Klun
- Clinical
Research Centre, University Medical Centre
Ljubljana, 1000 Ljubljana, Slovenia
- Faculty
of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Senka Imamović Kumalić
- Faculty
of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Division
of Obstetrics and Gynecology, University
Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Mateja Sladič
- Faculty
of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Division
of Obstetrics and Gynecology, University
Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Darja Mazej
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
| | - Janja Snoj Tratnik
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
| | - Milena Horvat
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
- Jožef
Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Tina Kosjek
- Department
of Environmental Sciences, Jožef
Stefan Institute, 1000 Ljubljana, Slovenia
- Jožef
Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Alhosseini MN, Maddalon A, Cari L, Ronchetti S, Migliorati G, Corsini E, Nocentini G. Imbalance of human CD4 + T lymphocyte subsets following atrazine treatment. Arch Toxicol 2025; 99:1455-1469. [PMID: 40025135 PMCID: PMC11968556 DOI: 10.1007/s00204-025-03974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/29/2025] [Indexed: 03/04/2025]
Abstract
While being banned in the European Union, the herbicide atrazine (ATR) is still one of the most used herbicides in the world. ATR is classified as an endocrine disruptor, but the immunotoxic effects of ATR may also be due to its direct impact on immune cells. To study the effects of ATR on human T cells, we activated T cells present in PBMCs of 8 healthy donors in the presence of ATR (0.1-100 μM). After 4 days of culture, T cells were stained to evaluate cell growth and phenotype by flow cytometry. The results demonstrated that ATR treatment exerts an antiproliferative activity on CD4+ T cells and decreases their activation, including the percentage of cytokine-producing CD4+ T cells. Among these, the percentage of interferon (IFN)-γ- and interleukin (IL)-22-producing CD4+ T cells decreased within total CD4+ T cells. Moreover, IL-4-, IL-10- and IL-17-producing CD4+ T cells decreased within cytokine-producing CD4+ T cells. Consequently, ATR caused a dose-related decrease in Th1/Th2 ratio. Many of the effects of ATR treatment were quantitatively different in males and females, with more pronounced effects observed in females. tSNE analysis demonstrated that ATR strongly inhibited the differentiation of two subsets of IFN-γ+IL-4+CD4+ T cells from each of the healthy donors tested and promoted greater differentiation of the CD25+FoxP3+CD4+ T cell subset from seven out of the eight healthy donors tested. In conclusion, the study suggests that ATR skews CD4+ T cell activation towards Th2, a phenotype that may promote reduced immunosurveillance and increased risk of cancer, as well as Th2-related diseases such as asthma, thereby presenting an environmental and occupation-related risk to human health.
Collapse
Affiliation(s)
- Mahdieh Naghavi Alhosseini
- Department of Medicine and Surgery, Section of Pharmacology, Università degli Studi di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| | - Ambra Maddalon
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Luigi Cari
- Department of Medicine and Surgery, Section of Pharmacology, Università degli Studi di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy.
| | - Simona Ronchetti
- Department of Medicine and Surgery, Section of Pharmacology, Università degli Studi di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| | - Graziella Migliorati
- Department of Medicine and Surgery, Section of Pharmacology, Università degli Studi di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Giuseppe Nocentini
- Department of Medicine and Surgery, Section of Pharmacology, Università degli Studi di Perugia, Building D, Severi Square 1, 06129, Perugia, Italy
| |
Collapse
|
4
|
Guengerich FP, Tateishi Y, McCarty KD, Yoshimoto FK. Updates on Mechanisms of Cytochrome P450 Catalysis of Complex Steroid Oxidations. Int J Mol Sci 2024; 25:9020. [PMID: 39201706 PMCID: PMC11354347 DOI: 10.3390/ijms25169020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Cytochrome P450 (P450) enzymes dominate steroid metabolism. In general, the simple C-hydroxylation reactions are mechanistically straightforward and are generally agreed to involve a perferryl oxygen species (formally FeO3+). Several of the steroid transformations are more complex and involve C-C bond scission. We initiated mechanistic studies with several of these (i.e., 11A1, 17A1, 19A1, and 51A1) and have now established that the dominant modes of catalysis for P450s 19A1 and 51A1 involve a ferric peroxide anion (i.e., Fe3+O2¯) instead of a perferryl ion complex (FeO3+), as demonstrated with 18O incorporation studies. P450 17A1 is less clear. The indicated P450 reactions all involve sequential oxidations, and we have explored the processivity of these multi-step reactions. P450 19A1 is distributive, i.e., intermediate products dissociate and reassociate, but P450s 11A1 and 51A1 are highly processive. P450 17A1 shows intermediate processivity, as expected from the release of 17-hydroxysteroids for the biosynthesis of key molecules, and P450 19A1 is very distributive. P450 11B2 catalyzes a processive multi-step oxidation process with the complexity of a chemical closure of an intermediate to a locked lactol form.
Collapse
Affiliation(s)
- F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Yasuhiro Tateishi
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Kevin D. McCarty
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Francis K. Yoshimoto
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| |
Collapse
|
5
|
Surapinit A, Chaidee A, Pinlaor S, Kongsintaweesuk S, Charoenram N, Mahaamnad N, Sakonsinsiri C, Hongsrichan N. Atrazine promotes cholangiocarcinoma cell proliferation and migration via GPER-mediated PI3K/Akt/NF-κB pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:105988. [PMID: 39084791 DOI: 10.1016/j.pestbp.2024.105988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 08/02/2024]
Abstract
Atrazine (ATZ), an herbicide widely distributed on a global scale, possess a potential risk for the development of various cancers upon environmental exposure. However, the effect and molecular mechanism of ATZ in cholangiocarcinoma (CCA), is still unclear. This study aimed to investigate the effect of ATZ on the proliferation and migration of CCA cell in vitro. Immortalized human cholangiocytes (MMNK-1) and three CCA cell lines (KKU-055, KKU-100 and KKU-213B) were treated with 0.01 to 100 μM of ATZ and 17β-estradiol (E2). The results showed that, similar to E2, low doses (0.01 to 1 μM) of ATZ promoted the proliferation of all CCA and MMNK-1 cells. ATZ exposure increased non-genomic G protein-coupled estrogen receptor (GPER) expression in the cell membrane and cytoplasm of KKU-213B and KKU-055 cells via G2/M cell cycle accumulation. This, in turn, promoted the proliferation and migration of CCA cells. ATZ exposure induced the upregulation of GPER and increased expression levels of PI3K, p-PI3K, Akt, p-Akt, NF-κB and PCNA. In contrast, following ATZ treatment, the GPER antagonist G15 significantly downregulated the GPER/PI3K/Akt/NF-κB pathway. These results suggest that ATZ promotes CCA cell proliferation and migration through the GPER/PI3K/Akt/NF-κB pathway. This information can enhance public health awareness regarding ATZ contamination to prevent the relative risk of CCA.
Collapse
Affiliation(s)
- Achirawit Surapinit
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Apisit Chaidee
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Somchai Pinlaor
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Suppakrit Kongsintaweesuk
- Medical Sciences Program, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Naruechar Charoenram
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Narumon Mahaamnad
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nuttanan Hongsrichan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
6
|
Choi J, Rotter S, Ritz V, Kneuer C, Marx-Stoelting P, de Lourdes Marzo Solano M, Oertel A, Rudzok S, Ziková-Kloas A, Tralau T, Hensel A. Thresholds of adversity for endocrine disrupting substances: a conceptual case study. Arch Toxicol 2024; 98:2019-2045. [PMID: 38704806 PMCID: PMC11168997 DOI: 10.1007/s00204-024-03748-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/21/2024] [Indexed: 05/07/2024]
Abstract
For endocrine disrupting chemicals (EDC) the existence of "safe exposure levels", that is exposure levels that do not present an appreciable risk to human health is most controversially discussed, as is the existence of health-based reference values. Concerns have been especially raised that EDCs might not possess a threshold level such that no exposure level to EDCs can be considered safe. To explore whether or not threshold levels can be identified, we performed a screening exercise on 14 pesticidal and biocidal active substances previously identified as EDCs in the European Union. The respective substances are ideal subjects for case studies to review for endocrine activity and disruptive potential following well-defined regulatory assessment based on solid data to effectually establish adversity as consequence of endocrine disruption. Dimethomorph, metiram and propiconazole for which the weight of evidence demonstrating endocrine disruption was the strongest were used as subjects for further study. Epoxiconazole was additionally selected as its effects on the endocrine system are extensive. For all four substances, analysis of the toxicological data clearly indicated thresholds of adversity below which no adverse effects mediated through an endocrine mechanism were observed. Particular emphasis was placed on mechanistic considerations including homeostasis and the concept of adversity. As a proof of concept this study provides evidence that like other substances of toxicological concern EDCs have threshold levels for adversity. While for some EDCs the respective thresholds might indeed be very low this shows that, data allowing, for other EDCs sufficiently protective reference values can be derived.
Collapse
Affiliation(s)
- Judy Choi
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Stefanie Rotter
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Vera Ritz
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Carsten Kneuer
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | | | - Angelika Oertel
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Susanne Rudzok
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Andrea Ziková-Kloas
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Tewes Tralau
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Andreas Hensel
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| |
Collapse
|
7
|
Chen ZJ, Qu YN, Li SY, Wang HW, Ji CH, Shi XZ, Yang H, Li XS. Insight into the relationship between metabolic enzymes and oxadiazon degradation in Oryza sativa for reducing environmental risks. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116242. [PMID: 38513530 DOI: 10.1016/j.ecoenv.2024.116242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/10/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Oxadiazon (ODZ) is extensively utilized in agricultural fields for weed control owing to its strong effectiveness. However, excessive loading of ODZ in water bodies and agricultural soils can lead to various environmental concerns. Therefore, it is crucial to understand the ODZ metabolic process and associated mechanisms in crops to assess the likelihood of ODZ contamination in the environment. This study aimed to assess the effects of ODZ on the growth and toxicological responses of rice (Oryza sativa). The growth of rice tissues was notably compromised with the increase in ODZ concentrations. RNA sequencing in combination with liquid chromatography-quadrupole-time-of-flight-high-resolution mass spectrometry/mass spectrometry (LC-Q-TOF-HRMS/MS) analysis allowed for the identification of numerous transcriptional components associated with ODZ metabolism. Four libraries comprising rice roots and shoots exposed to ODZ were RNA-sequenced in triplicate. The application of environmentally realistic ODZ concentrations upregulated the expression of 844 genes in shoots and 1476 genes in roots. Gene enrichment analysis revealed the presence of multiple enzymes involved in ODZ metabolism and detoxification. These enzymes play a critical role in mitigating environmental stress and facilitating xenobiotic metabolism. Notably, among differentially expressed genes, several key enzymes were identified, including cytochrome P450s, protein kinases, aminotransferases, and ATP-binding cassette transporters involved in the metabolic process. Using LC-Q-TOF-HRMS/MS, 3 metabolites and 13 conjugates were identified in multiple metabolic pathways involving oxidation, hydrolysis, glycosylation, acetylation, and methylation. This study successfully established a potential link between the specific metabolic products of ODZ and increased activities of their corresponding enzymes. Moreover, this study considerably elucidates the detailed pathways and mechanisms involved in ODZ metabolism. The study findings provide valuable insights into the development of genotypes for reducing ODZ residues in paddy fields and minimizing their accumulation in rice crops.
Collapse
Affiliation(s)
- Zhao Jie Chen
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China.
| | - Ya Nan Qu
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China
| | - Si Ying Li
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China
| | - Hao Wen Wang
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China
| | | | - Xu Zhen Shi
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China
| | - Hong Yang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Xue Sheng Li
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China.
| |
Collapse
|
8
|
Abarikwu SO, Ezim OE, Ikeji CN, Farombi EO. Atrazine: cytotoxicity, oxidative stress, apoptosis, testicular effects and chemopreventive Interventions. FRONTIERS IN TOXICOLOGY 2023; 5:1246708. [PMID: 37876981 PMCID: PMC10590919 DOI: 10.3389/ftox.2023.1246708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/19/2023] [Indexed: 10/26/2023] Open
Abstract
Atrazine (ATZ) is an environmental pollutant that interferes with several aspects of mammalian cellular processes including germ cell development, immunological, reproductive and neurological functions. At the level of human exposure, ATZ reduces sperm count and contribute to infertility in men. ATZ also induces morphological changes similar to apoptosis and initiates mitochondria-dependent cell death in several experimental models. When in vitro experimental models are exposed to ATZ, they are faced with increased levels of reactive oxygen species (ROS), cytotoxicity and decreased growth rate at dosages that may vary with cell types. This results in differing cytotoxic responses that are influenced by the nature of target cells, assay types and concentrations of ATZ. However, oxidative stress could play salient role in the observed cellular and genetic toxicity and apoptosis-like effects which could be abrogated by antioxidant vitamins and flavonoids, including vitamin E, quercetin, kolaviron, myricetin and bioactive extractives with antioxidant effects. This review focuses on the differential responses of cell types to ATZ toxicity, testicular effects of ATZ in both in vitro and in vivo models and chemopreventive strategies, so as to highlight the current state of the art on the toxicological outcomes of ATZ exposure in several experimental model systems.
Collapse
Affiliation(s)
- Sunny O. Abarikwu
- Reproductive Biology and Molecular Toxicology Research Group, Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| | - Ogechukwu E. Ezim
- Reproductive Biology and Molecular Toxicology Research Group, Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| | - Cynthia N. Ikeji
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebenezer O. Farombi
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
9
|
Leri M, Vasarri M, Barletta E, Schiavone N, Bergonzi MC, Bucciantini M, Degl’Innocenti D. The Protective Role of Oleuropein Aglycone against Pesticide-Induced Toxicity in a Human Keratinocytes Cell Model. Int J Mol Sci 2023; 24:14553. [PMID: 37834001 PMCID: PMC10572371 DOI: 10.3390/ijms241914553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The extensive use of agricultural pesticides to improve crop quality and yield significantly increased the risk to the public of exposure to small but repeated doses of pesticides over time through various routes, including skin, by increasing the risk of disease outbreaks. Although much work was conducted to reduce the use of pesticides in agriculture, little attention was paid to prevention, which could reduce the toxicity of pesticide exposure by reducing its impact on human health. Extra virgin olive oil (EVOO), a major component of the Mediterranean diet, exerts numerous health-promoting properties, many of which are attributed to oleuropein aglycone (OleA), the deglycosylated form of oleuropein, which is the main polyphenolic component of EVOO. In this work, three pesticides with different physicochemical and biological properties, namely oxadiazon (OXA), imidacloprid (IMID), and glyphosate (GLYPHO), were compared in terms of metabolic activity, mitochondrial function and epigenetic modulation in an in vitro cellular model of human HaCaT keratinocytes to mimic the pathway of dermal exposure. The potential protective effect of OleA against pesticide-induced cellular toxicity was then evaluated in a cell pre-treatment condition. This study showed that sub-lethal doses of OXA and IMID reduced the metabolic activity and mitochondrial functionality of HaCaT cells by inducing oxidative stress and altering intracellular calcium flux and caused epigenetic modification by reducing histone acetylation H3 and H4. GLYPHO, on the other hand, showed no evidence of cellular toxicity at the doses tested. Pretreatment of cells with OleA was able to protect cells from the damaging effects of the pesticides OXA and IMID by maintaining metabolic activity and mitochondrial function at a controlled level and preventing acetylation reduction, particularly of histone H3. In conclusion, the bioactive properties of OleA reported here could be of great pharmaceutical and health interest, as they could be further studied to design new formulations for the prevention of toxicity from exposure to pesticide use.
Collapse
Affiliation(s)
- Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Marzia Vasarri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
- Department of Chemistry, University of Florence, Via U. Schiff 6, 50519 Sesto Fiorentino, Italy;
| | - Emanuela Barletta
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Maria Camilla Bergonzi
- Department of Chemistry, University of Florence, Via U. Schiff 6, 50519 Sesto Fiorentino, Italy;
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Donatella Degl’Innocenti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| |
Collapse
|
10
|
EFSA Panel on Plant Protection Products and their Residues (PPR), Hernandez‐Jerez AF, Adriaanse P, Aldrich A, Berny P, Coja T, Duquesne S, Focks A, Millet M, Pelkonen O, Pieper S, Tiktak A, Topping CJ, Widenfalk A, Wilks M, Wolterink G, Angeli K, Recordati C, Van Durseen M, Aiassa E, Lanzoni A, Lostia A, Martino L, Guajardo IPM, Panzarea M, Terron A, Marinovich M. Development of adverse outcome pathways relevant for the identification of substances having endocrine disruption properties Uterine adenocarcinoma as adverse outcome. EFSA J 2023; 21:e07744. [PMID: 36818642 PMCID: PMC9926893 DOI: 10.2903/j.efsa.2023.7744] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Development of adverse outcome pathways (AOPs) for uterine adenocarcinoma can provide a practical tool to implement the EFSA-ECHA Guidance (2018) for the identification of endocrine disruptors in the context of Regulations (EU) No 528/2012 and (EC) No 1107/2009. AOPs can give indications about the strength of the relationship between an adverse outcome (intended as a human health outcome) and chemicals (pesticides but not only) affecting the pathways. In this scientific opinion, the PPR Panel explored the development of AOPs for uterine adenocarcinoma. An evidence-based approach methodology was applied, and literature reviews were produced using a structured framework assuring transparency, objectivity, and comprehensiveness. Several AOPs were developed; these converged to a common critical node, that is increased estradiol availability in the uterus followed by estrogen receptor activation in the endometrium; therefore, a putative AOP network was considered. An uncertainty analysis and a probabilistic quantification of the weight of evidence have been carried out via expert knowledge elicitation for each set of MIEs/KEs/KERs included in individual AOPs. The collected data on the AOP network were evaluated qualitatively, whereas a quantitative uncertainty analysis for weight of the AOP network certainty has not been performed. Recommendations are provided, including exploring further the uncertainties identified in the AOPs and putative AOP network; further methodological developments for quantifying the certainty of the KERs and of the overall AOPs and AOP network; and investigating of NAMs applications in the context of some of the MIEs/KEs currently part of the putative AOP network developed.
Collapse
|
11
|
Jabłońska – Trypuć A, Wiater J. Protective effect of plant compounds in pesticides toxicity. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2022; 20:1035-1045. [PMID: 36406617 PMCID: PMC9672277 DOI: 10.1007/s40201-022-00823-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/27/2022] [Accepted: 08/09/2022] [Indexed: 06/16/2023]
Abstract
INTRODUCTION The relationship between pesticide exposure and the occurrence of many chronic diseases, including cancer, is confirmed by literature data. METHODS In this review, through the analysis of more than 70 papers, we explore an increase in oxidative stress level caused by exposure to environmental pollutants and the protective effects of plant-origin antioxidants. RESULTS AND DISCUSSION One of the molecular mechanisms, by which pesticides affect living organisms is the induction of oxidative stress. However, recently many plant-based dietary ingredients with antioxidant properties have been considered as a chemopreventive substances due to their ability to remove free radicals. Such a food component must meet several conditions: eliminate free radicals, be easily absorbed and function at an appropriate physiological level. Its main function is to maintain the redox balance and minimize the cellular damage caused by ROS. Therefore, it should be active in aqueous solutions and membrane domains. These properties are characteristic for phenolic compounds and selected plant hormones. Phenolic compounds have proven antioxidant properties, while increasing number of compounds from the group of plant hormones with a very diverse chemical structure turn out to act as antioxidants, being potential food ingredients that can eliminate negative effects of pesticides.
Collapse
Affiliation(s)
- Agata Jabłońska – Trypuć
- Faculty of Civil Engineering and Environmental Sciences, Division of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45E Street, 15-351 Białystok, Białystok, Poland
| | - Józefa Wiater
- Faculty of Civil Engineering and Environmental Sciences, Department of Agri-Food Engineering and Environmental Management, Bialystok University of Technology, Wiejska 45E Street, 15-351 Białystok, Białystok, Poland
| |
Collapse
|
12
|
Olayinka ET, Ore A, Adewole KE, Oyerinde O. Evaluation of the toxicological effects of atrazine-metolachlor in male rats: in vivo and in silico studies. Environ Anal Health Toxicol 2022; 37:e2022021-0. [PMID: 36262065 PMCID: PMC9582417 DOI: 10.5620/eaht.2022021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
The types and mechanisms of atrazine-metolachlor toxicity, an herbicide composed of atrazine (ATR) and metolachlor (MET), need to be further investigated. This study evaluated the toxic actions of ATR-MET by in vivo and in silico methods. Here, varying doses of ATR-MET were orally administered to rats once daily for twenty-one days using normal saline as control. Molecular docking was used to characterize the binding of ATR and MET with androgen receptor (AR) to predict their potential endocrine-disrupting effects, using testosterone as benchmark. ATR-MET-induced-testicular toxicity (reduced sperm motility, count, and daily sperm production and increased live/dead ratio) was accompanied with testicular oxidative stress (diminished level of reduced glutathione, activities of glutathione-S-transferase, superoxide dismutase and catalase and increased level of malondialdehyde). Furthermore, ATR-MET induced cardiovascular toxicity (increased levels of plasma total cholesterol, HDL-cholesterol, LDL-cholesterol, and triglycerides) with concomitant induction of renal toxicity (increased plasma creatinine and urea levels), and hepatotoxicity (increased plasma bilirubin, alkaline phosphatase, acid phosphatase, alanine aminotransferase and aspartate aminotransferase). Binding energy and amino acid interactions from in silico study revealed that MET possessed endocrine-disrupting capacity. In conclusion, exposure to atrazine-metolachlor could promote cardiovascular, renal, hepatic, as well as reproductive impairment in experimental male albino rats.
Collapse
Affiliation(s)
- Ebenezer Tunde Olayinka
- Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University Oyo, Oyo State
Nigeria
| | - Ayokanmi Ore
- Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University Oyo, Oyo State
Nigeria
| | - Kayode Ezekiel Adewole
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Ondo State,
Nigeria
| | - Oyepeju Oyerinde
- Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University Oyo, Oyo State
Nigeria
| |
Collapse
|
13
|
Liang H, Wu X, Yao H, Weng X, Liu S, Chen J, Li Y, Wu Y, Wen L, Chen Q, Jing C. Association of urinary metabolites of non-persistent pesticides with serum sex hormones among the US females: NHANES 2013-2014. CHEMOSPHERE 2022; 300:134577. [PMID: 35421444 DOI: 10.1016/j.chemosphere.2022.134577] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Evidence indicated the possibility of non-persistent pesticides disrupting the homeostasis of sex hormones. However, few studies have focused on this relationship in females. We aimed to explore the relationship between non-persistent pesticide exposure and sex hormones among the US females from the National Health and Nutrition Examination Survey 2013-2014. METHODS A total of 790 females, including girls (6-11 years), female adolescents (12-19 years), and adult females (>19 years), were enrolled in this study. Age stratified associations of individual non-persistent pesticide metabolites and their mixtures with sex hormones were analyzed by weighted multiple linear regression and Bayesian kernel machine regression (BKMR) using spot urinary non-persistent pesticide measurement, including 2,4-dichlorophenoxyacetic acid (2,4-D), 3,5,6-trichloropyridinol (TCPY), para-nitrophenol (PNP) and 3-phenoxybenzoic acid (3-PBA), and three serum sex hormones [total testosterone (TT), estradiol (E2) and sex hormone binding globulin (SHBG)]. RESULTS In girls, weighted multivariate linear regression indicated that both 2,4-D and PNP were negatively associated with TT, and TCPY was inversely associated with SHBG. In female adolescents, TCPY was negatively associated with TT and E2, and 3-PBA was negatively associated with SHBG; positive associations were detected both in 2,4-D with SHBG, and in PNP with TT. In adult females, a higher concentration of 3-PBA was associated with higher levels of TT. The BKMR model showed that in female adolescents, the concentrations of pesticide metabolite mixtures at or above the 55th percentile were negatively related to the levels of E2 compared with their mixtures at 50th percentile, and an inverse U-shaped exposure-response function between PNP and E2 was found. CONCLUSIONS Associations between the four non-persistent pesticide metabolites and serum sex hormones were identified in the US females from NHANES 2013-2014 and these associations were age dependent, especially in adolescents. Large-scale cohort studies are needed to confirm these findings and elucidate the potential biological mechanisms.
Collapse
Affiliation(s)
- Huanzhu Liang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Xiaomei Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Huojie Yao
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Xueqiong Weng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Shan Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Jingmin Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Yexin Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Yingying Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Lin Wen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Qian Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China
| | - Chunxia Jing
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No.601 Huangpu Ave West, Guangzhou, 510632, Guangdong, China; Guangdong Key Laboratory of Environmental Exposure and Health, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
14
|
Assessment of Five Pesticides as Endocrine-Disrupting Chemicals: Effects on Estrogen Receptors and Aromatase. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19041959. [PMID: 35206146 PMCID: PMC8871760 DOI: 10.3390/ijerph19041959] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 02/07/2023]
Abstract
Pesticides are widely applied all over the world, and pesticide exposure can induce different biological effects posing a possible threat to human health. Due to their effects on the endocrine system, some pesticides are classified as endocrine disruptors. The aim of the study is to assess the interference of five pesticides on estrogen biosynthesis and estrogen signaling. Three neonicotinoid insecticides (Acetamiprid, Clothianidin, and Thiamethoxam), a carbamate insecticide (Methiocarb) and a herbicide (Oxadiazon) were tested. The effect of pesticides on estrogen biosynthesis was studied through an ELISA assay using a recombinant form of human aromatase, the enzyme that catalyzes the transformation of androgens to estrogens. Moreover, the effect of pesticides on estrogen signaling was assessed using a gene reporter assay on MELN cells, which measures estrogen receptor-mediated estrogenic activity. The results of the ELISA assay showed that the pesticides did not alter aromatase activity (no interference with estrogen biosynthesis), while the results of the gene reporter assay showed that only Methiocarb was able to alter estrogen signaling at high doses. The estrogenic activity of Methiocarb, expressed as 17β-estradiol equivalency factor (EEF), was equal to 8.0 × 10−8. In conclusion, this study suggested that Methiocarb should be considered a potential endocrine disruptor.
Collapse
|
15
|
Sakali AK, Bargiota A, Fatouros IG, Jamurtas A, Macut D, Mastorakos G, Papagianni M. Effects on Puberty of Nutrition-Mediated Endocrine Disruptors Employed in Agriculture. Nutrients 2021; 13:nu13114184. [PMID: 34836437 PMCID: PMC8622967 DOI: 10.3390/nu13114184] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
Pesticide residues are largely found in daily consumed food because of their extensive use in farming and their long half-life, which prolongs their presence in the environment. Many of these pesticides act as endocrine-disrupting chemicals after pre- or postnatal exposure, significantly affecting, among other things, the time of puberty onset, progression, and completion. In humans, precocious or delayed puberty, and early or delayed sexual maturation, may entail several negative long-term health implications. In this review, we summarize the current evidence on the impact of endocrine-disrupting pesticides upon the timing of the landmarks of female and male puberty in both animals (vaginal opening, first estrus, and balanopreputial separation) and humans (thelarche, menarche, gonadarche). Moreover, we explore the possible mechanisms of action of the reviewed endocrine-disrupting pesticides on the human reproductive system. Access to safe, healthy, and nutritious food is fundamental for the maintenance of health and wellbeing. Eliminating the presence of hazardous chemicals in largely consumed food products may increase their nutritional value and be proven beneficial for overall health. Consequently, understanding the effects of human exposure to hazardous endocrine-disrupting pesticides, and legislating against their circulation, are of major importance for the protection of health in vulnerable populations, such as children and adolescents.
Collapse
Affiliation(s)
- Anastasia Konstantina Sakali
- Department of Endocrinology and Metabolic Diseases, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (A.K.S.); (A.B.)
| | - Alexandra Bargiota
- Department of Endocrinology and Metabolic Diseases, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (A.K.S.); (A.B.)
| | - Ioannis G. Fatouros
- Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece; (I.G.F.); (A.J.)
| | - Athanasios Jamurtas
- Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece; (I.G.F.); (A.J.)
| | - Djuro Macut
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Maria Papagianni
- Department of Nutrition and Dietetics, University of Thessaly, 42132 Trikala, Greece
- Unit of Endocrinology, 3rd Department of Pediatrics, Hippokration Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Correspondence:
| |
Collapse
|
16
|
Shepelska NR, Prodanchuk MG, Kolianchuk YV. Comparative analysis of two methodological approaches to the study of endocrine disruptor alpha-cypermethrin reproductive toxicity. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/0221100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
At present, one of the main threats to humanity is undoubtedly endocrine disruptors (ED), since they directly disrupt the processes of ensuring homeostasis, which are identical to the very essence of the concept of health, considered in valeology as the ability to maintain age-appropriate stability in conditions of sharp changes in quantitative and qualitative parameters of the triune flow of sensory, verbal and structural information. Pesticides can disrupt the physiological functioning of many endocrine chains, including the endocrine mechanisms that ensure reproductive health. The study aimed to compare the results of our studies of alpha-cypermethrin reproductive toxicity in the test system for studying gonadotoxic activity with data obtained in the test system "Three Generation Reproduction Study". The studies were performed on male and female Wistar Han rats with two generic samples of alpha-cypermethrin from different manufacturers at doses of 0.2, 1.0 and 3.0 mg/kg body weight. The exposure lasted 11 (males) and 10 (females) weeks. During the last two weeks of exposure the state of the estrous cycle, duration and frequency of each stage in females was studied. After the end of treatment functional parameters of the state of the gonads in males and the ability of animals to reproduce were examined. In males, the total sperm count, the absolute and relative number of motile germ cells, and the number of pathologically altered forms were evaluated. The results of the study showed that exposure of male and female Wistar Han rats to alpha-cypermethrin at doses of 0.2–3.0 mg/kg during gametogenesis had a toxic effect on the reproductive system characterized by impaired gonadal and reproductive functions. In our studies, alpha-cypermethrin was found to have reproductive toxicity (reduced number of corpora luteum and live fetuses, increased absolute and relative postimplantation death, reduced average weight of fetuses and litters) and endocrine-disruptive effect, having a pronounced antiandrogenic effect on males. Obvious signs of endocrine reproductive disorders (changes in testis and epididymis weight, deterioration in semen parameters, altered length of separate stages of the estrous cycle) were observed in both females and males. However, studies in a test system of three-generations did not reveal a reproductive and endocrine-disruptive effect of cypermethrin, the toxicity of which was recognized as equivalent to the toxicity of alpha-cypermethrin. The obtained results showed higher sensitivity, informative and diagnostic significance of the methods for studying gonadotoxicity than the methodology of reproductive toxicity studies in the test system “Three Generation Reproduction Study”.
Collapse
|
17
|
Enderle I, Costet N, Cognez N, Zaros C, Caudeville J, Garlantezec R, Chevrier C, Nougadere A, De Lauzon-Guillain B, Le Lous M, Beranger R. Prenatal exposure to pesticides and risk of preeclampsia among pregnant women: Results from the ELFE cohort. ENVIRONMENTAL RESEARCH 2021; 197:111048. [PMID: 33766571 DOI: 10.1016/j.envres.2021.111048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Preeclampsia is a pregnancy-specific syndrome caused by abnormal placentation. Although environmental chemicals, including some pesticides, are suspected of impairing placentation and promoting preeclampsia, its relationship with preeclampsia has been insufficiently explored. OBJECTIVES We aimed to investigate the relation between non-occupational exposure to pesticides during pregnancy and the risk of preeclampsia. METHODS The study cohort comprised 195 women with and 17,181 without preeclampsia from the ELFE birth cohort. We used toxicogenomic approaches to select 41 pesticides of interest for their possible influence on preeclampsia. We assessed household pesticide use (self-reported data), environmental exposure to agricultural pesticides (geographic information systems), and dietary exposure (food-frequency questionnaire with data from monitoring pesticide residues in food and water). Dietary exposures to pesticides were grouped into clusters of similar exposures to resolve collinearity issues. For each exposure source, pesticides were mutually adjusted, and odds ratios estimated with logistic regression models. RESULTS The quantity of prochloraz applied within a kilometer of the women's homes was higher in women with than without preeclampsia (fourth quartile vs. others; adjusted odds ratio [aOR] = 1.54; 95%CI: 1.02, 2.35), especially when preeclampsia was diagnosed before 34 weeks of gestation (aOR = 2.25; 95%CI: 1.01, 5.06). The reverse was observed with nearby cypermethrin application (aOR = 0.59, 95%CI: 0.36, 0.96). In sensitivity analyses, women with preeclampsia receiving antihypertensive treatment had a significantly higher probability of using herbicides at home during pregnancy than women without preeclampsia (aOR = 2.20; 95%CI: 1.23, 3.93). No statistically significant association was found between dietary exposure to pesticide residues and preeclampsia. DISCUSSION While the most of the associations examined remained statistically non-significant, our results suggest the possible influence on preeclampsia of residential exposures to prochloraz and some herbicides. These estimations are supported by toxicological and mechanistic data.
Collapse
Affiliation(s)
- Isabelle Enderle
- CHU Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France; Department of Obstetrics and Gynecology and Reproductive Medicine, Anne de Bretagne University Hospital, Rennes, France.
| | - Nathalie Costet
- Univ Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000, Rennes, France
| | - Noriane Cognez
- Univ Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000, Rennes, France
| | - Cécile Zaros
- French Institute for Demographic Studies (Ined), French Institute for Medical Research and Health (Inserm), French Blood Agency, ELFE Joint Unit, F-75020, Paris, France
| | - Julien Caudeville
- INERIS (French National Institute for Industrial Environment and Risks), 60550, Verneuil-en-Halatte, France
| | - Ronan Garlantezec
- CHU Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Cécile Chevrier
- Univ Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000, Rennes, France
| | - Alexandre Nougadere
- ANSES, Risk Assessment Department, 14 Rue Pierre et Marie Curie, F-94701, Maisons-Alfort, France
| | | | - Maela Le Lous
- Department of Obstetrics and Gynecology and Reproductive Medicine, Anne de Bretagne University Hospital, Rennes, France
| | - Rémi Beranger
- CHU Rennes, Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France; Department of Obstetrics and Gynecology and Reproductive Medicine, Anne de Bretagne University Hospital, Rennes, France
| |
Collapse
|
18
|
Avila R, Peris A, Eljarrat E, Vicent T, Blánquez P. Biodegradation of hydrophobic pesticides by microalgae: Transformation products and impact on algae biochemical methane potential. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 754:142114. [PMID: 32911153 DOI: 10.1016/j.scitotenv.2020.142114] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/09/2020] [Accepted: 08/29/2020] [Indexed: 06/11/2023]
Abstract
Intensive and extensive use of pesticides has contributed to their wide distribution in soil, air, and water. Due to their detrimental effects on non-target organisms, different technologies have been considered for their removal. In this work, three hydrophobic pesticide active compounds, namely, chlorpyrifos, cypermethrin, and oxadiazon, were selected to study the potential for their removal from aqueous media by a microalgae consortium. An abiotic and a killed control (thermally inactivated dead microalgae biomass) were employed to clarify their removal pathways, and pesticide content was quantified in liquid and biomass phases for 7 days. At the final time, total degradation (biodegradation plus photodegradation) contributed to the removal of 55% of oxadiazon, 35% of chlorpyrifos, and 14% of cypermethrin. Furthermore, more than 60% of chlorpyrifos and cypermethrin were removed by sorption onto microalgae biomass. Overall, the three pesticides showed high removal from the liquid phase. O,O-diethyl thiophosphate was identified in the liquid phase as a transformation product of chlorpyrifos formed by microalgae degradation. Phycoremediation was coupled with anaerobic degradation of the microalgae biomass containing the retained pesticides by sorption through biochemical methane potential tests. Anaerobic digestion was not inhibited by the pesticides as verified by methane production yields. The removal efficiency of the pesticides in the digestate was as follows: chlorpyrifos > cypermethrin > oxadiazon. These results highlight the potential of low-cost algal-based systems for the treatment of wastewater or effluents from agrochemical industries. The integration of wastewater treatment with biogas production through anaerobic digestion is a biorefinery approach that facilitates the economic feasibility of the process.
Collapse
Affiliation(s)
- Romina Avila
- Chemical, Biological and Environmental Engineering Department, Escola d'Enginyeria, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain
| | - Andrea Peris
- Water, Environmental and Food Chemistry, Dep. of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Ethel Eljarrat
- Water, Environmental and Food Chemistry, Dep. of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Teresa Vicent
- Chemical, Biological and Environmental Engineering Department, Escola d'Enginyeria, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain
| | - Paqui Blánquez
- Chemical, Biological and Environmental Engineering Department, Escola d'Enginyeria, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
19
|
Wang Q, Shen JY, Zhang R, Hong JW, Li Z, Ding Z, Wang HX, Zhang JP, Zhang MR, Xu LC. Effects and mechanisms of pyrethroids on male reproductive system. Toxicology 2020; 438:152460. [PMID: 32278050 DOI: 10.1016/j.tox.2020.152460] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022]
Abstract
Synthetic pyrethroids are used as insecticides in agriculture and a variety of household applications worldwide. Pyrethroids are widely distributed in all environmental compartments and the general populations are exposed to pyrethroids through various routes. Pyrethroids have been identified as endocrine-disrupting chemicals (EDCs) which are responsible for the male reproductive impairments. The data confirm pyrethroids cause male reproductive damages. The insecticides exert the toxic effects on male reproductive system through various complex mechanisms including antagonizing androgen receptor (AR), inhibiting steroid synthesis, affecting the hypothalamic-pituitary-gonadal (HPG) axis, acting as estrogen receptor (ER) modulators and inducing oxidative stress. The mechanisms of male reproductive toxicity of pyrethroids involve multiple targets and pathways. The review will provide further insight into pyrethroid-induced male reproductive toxicity and mechanisms, which is crucial to preserve male reproductive health.
Collapse
Affiliation(s)
- Qi Wang
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Jun-Yu Shen
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Rui Zhang
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Jia-Wei Hong
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Zheng Li
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Zhen Ding
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Heng-Xue Wang
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Jin-Peng Zhang
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Mei-Rong Zhang
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China
| | - Li-Chun Xu
- School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
20
|
Kovács T, Horváth M, Csordás A, Bátor G, Tóth-Bodrogi E. Tobacco plant as possible biomonitoring tool of red mud dust fallout and increased natural radioactivity. Heliyon 2020; 6:e03455. [PMID: 32181383 PMCID: PMC7062938 DOI: 10.1016/j.heliyon.2020.e03455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/28/2020] [Accepted: 02/18/2020] [Indexed: 11/17/2022] Open
Abstract
Exposure to toxic heavy metal content in soil and inherent naturally occurring radioactive materials (NORM) needs to be monitored, especially after industrial accidents and remediation efforts. Just such an accident happened near Ajka city in Hungary; a large quantity of red mud flooded out from a reservoir. The afflicted area was remediated, and the red mud deposition technology was changed from a wet to a dry procedure. Concerns have been raised about potential hazards from airborne NORM dust in this area. The objectives of this study were to assess the use of explanted tobacco plants as an active biomonitoring system for airborne NORM dust and to reveal weather-related correlations of Po-210 in airborne dust. In 2011, 2012 and 2014, measurements were taken of the following at six monitoring sites in the polluted area and at eight sites in unpolluted areas: soil and tobacco plant Po-210 isotope levels, airborne Rn-222, Ra-226 in soil, Th-232 and K-40 radioactivity concentrations. The transfer factors (TFs) of tobacco were calculated yearly for these isotopes. Association of data with local weather features was determined. In 2012 (the windiest and driest year), the mean Po-210 activity concentrations of tobacco samples in polluted areas were significantly higher than in 2011 and in 2014 (p = 0.044 and p = 0.024, respectively). The mean TF of samples in 2012 was also significantly higher in tobacco plants grown in the polluted area compared to ones grown in unpolluted areas (p = 0.020). These results presumably originate from red mud dust-particle adsorption on tobacco plant leaves. Tobacco plants are promising active bioindicators of airborne particulate pollution by Po-210 or other atmospheric NORM content.
Collapse
Affiliation(s)
- Tibor Kovács
- Institute of Radiochemistry and Radioecology, University of Pannonia, 10 Egyetem str., H-8200, Veszprém, Hungary
- Social Organisation for Radioecological Cleanliness, 7/a József Attila str., H-8200, Veszprém, Hungary
- Corresponding author.
| | - Mária Horváth
- Institute of Radiochemistry and Radioecology, University of Pannonia, 10 Egyetem str., H-8200, Veszprém, Hungary
- Social Organisation for Radioecological Cleanliness, 7/a József Attila str., H-8200, Veszprém, Hungary
| | - Anita Csordás
- Institute of Radiochemistry and Radioecology, University of Pannonia, 10 Egyetem str., H-8200, Veszprém, Hungary
| | - Gergő Bátor
- Institute of Radiochemistry and Radioecology, University of Pannonia, 10 Egyetem str., H-8200, Veszprém, Hungary
- Social Organisation for Radioecological Cleanliness, 7/a József Attila str., H-8200, Veszprém, Hungary
| | - Edit Tóth-Bodrogi
- Institute of Radiochemistry and Radioecology, University of Pannonia, 10 Egyetem str., H-8200, Veszprém, Hungary
| |
Collapse
|
21
|
Development of a prioritization method for chemical-mediated effects on steroidogenesis using an integrated statistical analysis of high-throughput H295R data. Regul Toxicol Pharmacol 2019; 109:104510. [PMID: 31676319 DOI: 10.1016/j.yrtph.2019.104510] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022]
Abstract
Synthesis of 11 steroid hormones in human adrenocortical carcinoma cells (H295R) was measured in a high-throughput steroidogenesis assay (HT-H295R) for 656 chemicals in concentration-response as part of the US Environmental Protection Agency's ToxCast program. This work extends previous analysis of the HT-H295R dataset and model by examining the utility of a novel prioritization metric based on the Mahalanobis distance that reduced these 11-dimensional data to 1-dimension via calculation of a mean Mahalanobis distance (mMd) at each chemical concentration screened for all hormone measures available. Herein, we evaluated the robustness of mMd values, and demonstrate that covariance and variance of the hormones measured appear independent of the chemicals screened and are inherent to the assay; the Type I error rate of the mMd method is less than 1%; and, absolute fold changes (up or down) of 1.5 to 2-fold have sufficient power for statistical significance. As a case study, we examined hormone responses for aromatase inhibitors in the HT-H295R assay and found high concordance with other ToxCast assays for known aromatase inhibitors. Finally, we used mMd and other ToxCast cytotoxicity data to demonstrate prioritization of the most selective and active chemicals as candidates for further in vitro or in silico screening.
Collapse
|
22
|
Wang F, Yang QW, Zhao WJ, Du QY, Chang ZJ. Effects of short-time exposure to atrazine on miRNA expression profiles in the gonad of common carp (Cyprinus carpio). BMC Genomics 2019; 20:587. [PMID: 31315571 PMCID: PMC6636164 DOI: 10.1186/s12864-019-5896-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atrazine is widely used in agriculture and is a known endocrine disrupting chemical. Atrazine can seep into the water body through surface, posing a potential threat to the aquatic ecological environment and human drinking water source. In vertebrate, studies have shown that it can affect reproduction and development seriously, but its molecular mechanism for aquatic animals is unknown. Aquaculture is very common in China, especially common carp, whose females grow faster than males. However, the effects of atrazine on the reproduction of carp, especially miRNA, have not been investigated. RESULTS In this study, common carp (Cyprinus carpio) at two key developmental stages were exposed to atrazine in vitro. Sex ratio was observed to analyze the effect of atrazine on the sex. MiRNA expression profiles were analysed to identify miRNAs related to gonad development and to reveal the atrazine mechanisms interfering with gonad differentiation. The results showed that the sex ratio was biased towards females. Atrazine exposure caused significant alteration of multiple miRNAs. Predicted targets of differently-expressed miRNAs were involved in many reproductive biology signalling pathways. CONCLUSIONS Our results indicate that atrazine promoted the expression of female-biased genes by decreasing miRNAs in primordial gonad. In addition, our results indicate that atrazine can up-regulate aromatase expression through miRNAs, which supports the hypothesis that atrazine has endocrine-disrupting activity by altering the gene expression profile of the Hypothalamus-Pituitary-Gonad axis through its corresponding miRNAs.
Collapse
Affiliation(s)
- Fang Wang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, People's Republic of China
| | - Qian-Wen Yang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, People's Republic of China
| | - Wen-Jie Zhao
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, People's Republic of China
| | - Qi-Yan Du
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, People's Republic of China
| | - Zhong-Jie Chang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, People's Republic of China.
| |
Collapse
|
23
|
Gaur H, Bhargava A. Glyphosate induces toxicity and modulates calcium and NO signaling in zebrafish embryos. Biochem Biophys Res Commun 2019; 513:1070-1075. [PMID: 31010672 DOI: 10.1016/j.bbrc.2019.04.074] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
Glyphosate, an herbicide used worldwide, has emerged as a pollutant. However, its toxic effects are debated by regulatory authorities. Therefore, it is essential to keep the use of such chemicals under continuous observation, and their effects must be re-evaluated. We used zebrafish embryos to evaluate the toxic effects of glyphosate and its mechanisms. We found that glyphosate induced significant toxicity in a time and concentration-dependent manner. We observed an LD50 of 66.04 ± 4.6 μg/mL after 48 h of exposure. Glyphosate significantly reduced the heartbeat in a time and concentration-dependent manner indicating cardiotoxicity. Selective downregulation of Cacana1C (L-type calcium channel) and ryr2a (Ryanodine receptor) genes along with selective upregulation of hspb11 (heat shock protein) gene was observed upon exposure to glyphosate indicating alterations in the calcium signaling. A reduction in the nitric oxide (NO) generation was also observed in the zebrafish embryos upon exposure to glyphosate. Our results indicate that glyphosate induces significant toxicity including cardiotoxicity in zebrafish embryos in a time and concentration-dependent manner. Further, cardiotoxicity may be due to changes in calcium and NO signaling.
Collapse
Affiliation(s)
- Himanshu Gaur
- Ion Channel Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502285, India
| | - Anamika Bhargava
- Ion Channel Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502285, India.
| |
Collapse
|
24
|
Souders CL, Xavier P, Perez-Rodriguez V, Ector N, Zhang JL, Martyniuk CJ. Sub-lethal effects of the triazole fungicide propiconazole on zebrafish (Danio rerio) development, oxidative respiration, and larval locomotor activity. Neurotoxicol Teratol 2019; 74:106809. [PMID: 31129159 DOI: 10.1016/j.ntt.2019.106809] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 01/22/2023]
Abstract
Propiconazole is a triazole fungicide used in agriculture. Via run-off, it can enter the aquatic environment, and can adversely affect organisms. However, data are scarce on how propiconazole may affect early developmental life stages of fish. The objectives of this study were to evaluate the potential sub-lethal effects of propiconazole during zebrafish development. Wildtype zebrafish (ABTu strain) embryos and larvae were exposed to propiconazole (0.1-100 μM) for up to 150 hours post fertilization (hpf) depending upon the endpoint measured. Propiconazole decreased survival and induced hypopigmentation in fish at 100 μM compared to the water and solvent controls. Pericardial edema was also noted in embryos and larvae (beginning at 2-3 dpf) exposed to 100 μM propiconazole. To visualize the effects of propiconazole on the circulatory system in more detail, we exposed transgenic zebrafish (globin-LCR:eGFP) to the fungicide. Hematopoietic changes were observed within 48 h of exposure to 100 μM, and localization of blood cells in the cardic region became diffuse, indicating pooling of blood in the pericardial region. We measured oxidative respiration in embryos as sufficient ATP is needed for development. Exposure to 100 μM propiconazole (~6-30 hpf) reduced basal respiration (~50%), oligomycin-induced ATP linked respiration (~70%), proton leak (~30%), and non-mitochondrial respiration (~50%), indicating compromised mitochondrial bioenergetics. A Visual Motor Response (VMR) test was used to measure dark photokinesis behavior in larval fish exposed to propiconazole for a 6-day period. Larval fish exposed to the highest concentration in the assay (10 μM) showed evidence of hypoactivity. This study demonstrates that propiconazole can induce hypopigmentation in zebrafish, disrupt mitochondrial bioenergetics, and can alter locomotor activity. However, these sub-lethal responses were observed at concentrations above what is typically detected in the environment.
Collapse
Affiliation(s)
- Christopher L Souders
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Priscilla Xavier
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Veronica Perez-Rodriguez
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Naomi Ector
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ji-Liang Zhang
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; Henan Open Laboratory of Key Subjects of Environmental and Animal Products Safety, College of Animal Science and Technology, Henan University of Science and Technology, Henan, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
25
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
26
|
Williams GP, Darbre PD. Low-dose environmental endocrine disruptors, increase aromatase activity, estradiol biosynthesis and cell proliferation in human breast cells. Mol Cell Endocrinol 2019; 486:55-64. [PMID: 30817981 DOI: 10.1016/j.mce.2019.02.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/07/2019] [Accepted: 02/18/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Phenolic endocrine-disrupting compounds (EDCs) have long been suspected of increasing human breast cancer risk, via aromatase up-regulation; however, the metabolic effects upon aromatase in human breast cells exposed to environmentally relevant concentrations of phenolic compounds, have not been addressed. OBJECTIVES To examine the mechanistic responses of aromatase CYP19A1 mRNA, aromatase activity, estradiol biosynthesis and cellular proliferation, in three human breast cell lines, exposed to seven phenolic compounds, at environmentally relevant concentrations. METHODS MCF-7 and ZR-75-1 breast cancer cells, and HMF3A breast fibroblasts were treated with specific concentrations of p,p'-DDT, methoxychlor, benzophenone-2, bisphenol A, bisphenol S, 4-phenylphenol and n-butylparaben, with and without the presence of aromatase inhibitors and estrogen receptor inhibitors. RESULTS All test EDCs up-regulated aromatase mRNA, increased aromatase activity, significantly increased the aromatase-induced biosynthesis of the breast carcinogen 17β-estradiol, and increased ERα-positive breast cell proliferation. CONCLUSION Inadvertent exposures to 'phenolic' EDCs, increase estradiol biosynthesis, and estrogen-sensitive breast cancer proliferation.
Collapse
Affiliation(s)
- Graeme P Williams
- Molecular and Cellular Medicine Group, School of Biological Sciences, University of Reading, Reading, Berkshire, RG6 6UB, United Kingdom.
| | - Philippa D Darbre
- Molecular and Cellular Medicine Group, School of Biological Sciences, University of Reading, Reading, Berkshire, RG6 6UB, United Kingdom
| |
Collapse
|
27
|
Nowak K, Jabłońska E, Ratajczak-Wrona W. Immunomodulatory effects of synthetic endocrine disrupting chemicals on the development and functions of human immune cells. ENVIRONMENT INTERNATIONAL 2019; 125:350-364. [PMID: 30743143 DOI: 10.1016/j.envint.2019.01.078] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 05/22/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are added to food, cosmetics, plastic packages, and children's toys and have thus become an integral part of the human environment. In the last decade, there has been increasing interest in the effect of EDCs on human health, including their impact on the immune system. So far, researchers have proved that EDCs (e.g. bisphenols, phthalates, triclosan, phenols, propanil, tetrachlorodibenzo-p-dioxin, diethylstilbestrol, tributyltin (TBT), and parabens) affect the development, functions, and lifespan of immune cells (e.g., monocytes, neutrophils, mast cells, eosinophils, lymphocytes, dendritic cells, and natural killers). In this review, we have summarized the current knowledge of the multivariable influence of EDCs on immune cells and underlined the novel approach to EDC studies, including dose-dependent effects and low-dose effects. We discuss critically the possible relationship between exposure to EDCs and immunity related diseases (e.g. allergy, asthma, diabetes, and lupus). Moreover, based on the literature, we construct a model of possible mechanisms of EDC action on immune cells at cellular, molecular, and epigenetic levels.
Collapse
Affiliation(s)
- Karolina Nowak
- Department of Immunology, Medical University of Bialystok, Poland.
| | - Ewa Jabłońska
- Department of Immunology, Medical University of Bialystok, Poland
| | | |
Collapse
|
28
|
Degl'Innocenti D, Ramazzotti M, Sarchielli E, Monti D, Chevanne M, Vannelli GB, Barletta E. Oxadiazon affects the expression and activity of aldehyde dehydrogenase and acylphosphatase in human striatal precursor cells: A possible role in neurotoxicity. Toxicology 2018; 411:110-121. [PMID: 30391265 DOI: 10.1016/j.tox.2018.10.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/05/2018] [Accepted: 10/30/2018] [Indexed: 12/17/2022]
Abstract
Exposure to herbicides can induce long-term chronic adverse effects such as respiratory diseases, malignancies and neurodegenerative diseases. Oxadiazon, a pre-emergence or early post-emergence herbicide, despite its low acute toxicity, may induce liver cancer and may exert adverse effects on reproductive and on endocrine functions. Unlike other herbicides, there are no indications on neurotoxicity associated with long-term exposure to oxadiazon. Therefore, we have analyzed in primary neuronal precursor cells isolated from human striatal primordium the effects of non-cytotoxic doses of oxadiazon on neuronal cell differentiation and migration, and on the expression and activity of the mitochondrial aldehyde dehydrogenase 2 (ALDH2) and of the acylphosphatase (ACYP). ALDH2 activity protects neurons against neurotoxicity induced by toxic aldehydes during oxidative stress and plays a role in neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease. ACYP is involved in ion transport, cell differentiation, programmed cell death and cancer, and increased levels of ACYP have been revealed in fibroblasts from patients affected by Alzheimer's disease. In this study we demonstrated that non-cytotoxic doses of oxadiazon were able to inhibit neuronal striatal cell migration and FGF2- and BDNF-dependent differentiation towards neuronal phenotype, and to inhibit the expression and activity of ALDH2 and to increase the expression and activity of ACYP2. In addition, we have provided evidence that in human primary neuronal precursor striatal cells the inhibitory effects of oxadiazon on cell migration and differentiation towards neuronal phenotype were achieved through modulation of ACYP2. Taken together, our findings reveal for the first time that oxadiazon could exert neurotoxic effects by impairing differentiative capabilities of primary neuronal cells and indicate that ALDH2 and ACYP2 are relevant molecular targets for the neurotoxic effects of oxadiazon, suggesting a potential role of this herbicide in the onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Donatella Degl'Innocenti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Erica Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Marta Chevanne
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | | | - Emanuela Barletta
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| |
Collapse
|
29
|
Quagliariello V, Rossetti S, Cavaliere C, Di Palo R, Lamantia E, Castaldo L, Nocerino F, Ametrano G, Cappuccio F, Malzone G, Montanari M, Vanacore D, Romano FJ, Piscitelli R, Iovane G, Pepe MF, Berretta M, D'Aniello C, Perdonà S, Muto P, Botti G, Ciliberto G, Veneziani BM, De Falco F, Maiolino P, Caraglia M, Montella M, Iaffaioli RV, Facchini G. Metabolic syndrome, endocrine disruptors and prostate cancer associations: biochemical and pathophysiological evidences. Oncotarget 2018; 8:30606-30616. [PMID: 28389628 PMCID: PMC5444769 DOI: 10.18632/oncotarget.16725] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/06/2017] [Indexed: 01/18/2023] Open
Abstract
This review summarizes the main pathophysiological basis of the relationship between metabolic syndrome, endocrine disruptor exposure and prostate cancer that is the most common cancer among men in industrialized countries. Metabolic syndrome is a cluster of metabolic and hormonal factors having a central role in the initiation and recurrence of many western chronic diseases including hormonal-related cancers and it is considered as the worlds leading health problem in the coming years. Many biological factors correlate metabolic syndrome to prostate cancer and this review is aimed to focus, principally, on growth factors, cytokines, adipokines, central obesity, endocrine abnormalities and exposure to specific endocrine disruptors, a cluster of chemicals, to which we are daily exposed, with a hormone-like structure influencing oncogenes, tumor suppressors and proteins with a key role in metabolism, cell survival and chemo-resistance of prostate cancer cells. Finally, this review will analyze, from a molecular point of view, how specific foods could reduce the relative risk of incidence and recurrence of prostate cancer or inhibit the biological effects of endocrine disruptors on prostate cancer cells. On the basis of these considerations, prostate cancer remains a great health problem in terms of incidence and prevalence and interventional studies based on the treatment of metabolic syndrome in cancer patients, minimizing exposure to endocrine disruptors, could be a key point in the overall management of this disease.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy.,Medical Oncology, Abdominal Department, National Cancer Institute G. Pascale Foundation, Napoli, Italy.,Association for Multidisciplinary Studies in Oncology and Mediterranean Diet, Piazza Nicola Amore, Naples, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy
| | - Carla Cavaliere
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Department of Onco-Ematology Medical Oncology, S.G. Moscati Hospital of Taranto, Taranto, Italy
| | - Rossella Di Palo
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Elvira Lamantia
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Luigi Castaldo
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Division of Urology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy
| | - Flavia Nocerino
- Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Gianluca Ametrano
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Francesca Cappuccio
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Gabriella Malzone
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Daniela Vanacore
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy
| | - Francesco Jacopo Romano
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale Naples, Italy
| | - Gelsomina Iovane
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy
| | - Maria Filomena Pepe
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, Aviano, Italy.,Association for Multidisciplinary Studies in Oncology and Mediterranean Diet, Piazza Nicola Amore, Naples, Italy
| | - Carmine D'Aniello
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Napoli, Italy
| | - Sisto Perdonà
- Division of Urology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Francesco De Falco
- Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maurizio Montella
- Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Napoli, Italy
| | - Rosario Vincenzo Iaffaioli
- Medical Oncology, Abdominal Department, National Cancer Institute G. Pascale Foundation, Napoli, Italy.,Association for Multidisciplinary Studies in Oncology and Mediterranean Diet, Piazza Nicola Amore, Naples, Italy
| | - Gaetano Facchini
- Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo - Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale' - IRCCS, Naples, Italy.,Association for Multidisciplinary Studies in Oncology and Mediterranean Diet, Piazza Nicola Amore, Naples, Italy
| |
Collapse
|
30
|
Martins-Santos E, Pimenta CG, Campos PRN, Oliveira AG, Mahecha GAB, Oliveira CA. Atrazine affects the morphophysiology, tissue homeostasis and aromatase expression in the efferent ductules of adult rats with mild alterations in the ventral prostate. CHEMOSPHERE 2018; 193:958-967. [PMID: 29874772 DOI: 10.1016/j.chemosphere.2017.11.124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 06/08/2023]
Abstract
The widely used herbicide atrazine is a potent endocrine disruptor known to cause increased aromatase expression and transient increase in testicular weight followed by remarkable testis atrophy. However, whether the effects of atrazine on the testes are primary or secondary to dysfunctions in other components of male reproductive tract remains unknown. Given the high sensitivity of the efferent ductules to estrogen imbalance and the similarity to alterations previously described for other disruptors of these ductules function, and the testicular alterations observed after atrazine exposure, we hypothesized that the efferent ductules could be a target for atrazine. Herein we characterized the efferent ductules and the ventral prostate of adult Wistar rats treated with 200 mg/kg/day of atrazine for 7, 15, and 40 days. Additionally, we evaluated if the effects of atrazine in these organs could be reduced after discontinuation of the treatment. Atrazine exposure resulted in mild effects on the ventral prostate, but remarkable alterations on the efferent ductules, including luminal dilation, reduced epithelial height, and disruption of the epithelial homeostasis, which coincides with increased aromatase expression. Together with our previous data, these results suggest that at least part of the testicular effects of atrazine may be secondary to the alterations in the efferent ductules.
Collapse
Affiliation(s)
- Elisângela Martins-Santos
- Department of Morphology, Universidade Federal de Minas Gerais, Cx. Postal 486, CEP 31.270-901, Belo Horizonte, MG, Brazil
| | - Cristiano Guimarães Pimenta
- Department of Morphology, Universidade Federal de Minas Gerais, Cx. Postal 486, CEP 31.270-901, Belo Horizonte, MG, Brazil
| | - Pollyana Rabelo Nunes Campos
- Department of Morphology, Universidade Federal de Minas Gerais, Cx. Postal 486, CEP 31.270-901, Belo Horizonte, MG, Brazil
| | - André Gustavo Oliveira
- Department of Morphology, Universidade Federal de Minas Gerais, Cx. Postal 486, CEP 31.270-901, Belo Horizonte, MG, Brazil
| | | | - Cleida Aparecida Oliveira
- Department of Morphology, Universidade Federal de Minas Gerais, Cx. Postal 486, CEP 31.270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
31
|
Supercritical fluid chromatography–tandem mass spectrometry-assisted methodology for rapid enantiomeric analysis of fenbuconazole and its chiral metabolites in fruits, vegetables, cereals, and soil. Food Chem 2018; 241:32-39. [DOI: 10.1016/j.foodchem.2017.08.038] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 06/22/2017] [Accepted: 08/12/2017] [Indexed: 11/23/2022]
|
32
|
Pinto CL, Markey K, Dix D, Browne P. Identification of candidate reference chemicals for in vitro steroidogenesis assays. Toxicol In Vitro 2017; 47:103-119. [PMID: 29146384 DOI: 10.1016/j.tiv.2017.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/19/2017] [Accepted: 11/11/2017] [Indexed: 11/15/2022]
Abstract
The Endocrine Disruptor Screening Program (EDSP) is transitioning from traditional testing methods to integrating ToxCast/Tox21 in vitro high-throughput screening assays for identifying chemicals with endocrine bioactivity. The ToxCast high-throughput H295R steroidogenesis assay may potentially replace the low-throughput assays currently used in the EDSP Tier 1 battery to detect chemicals that alter the synthesis of androgens and estrogens. Herein, we describe an approach for identifying in vitro candidate reference chemicals that affect the production of androgens and estrogens in models of steroidogenesis. Candidate reference chemicals were identified from a review of H295R and gonad-derived in vitro assays used in methods validation and published in the scientific literature. A total of 29 chemicals affecting androgen and estrogen levels satisfied all criteria for positive reference chemicals, while an additional set of 21 and 15 chemicals partially fulfilled criteria for positive reference chemicals for androgens and estrogens, respectively. The identified chemicals included pesticides, pharmaceuticals, industrial and naturally-occurring chemicals with the capability to increase or decrease the levels of the sex hormones in vitro. Additionally, 14 and 15 compounds were identified as potential negative reference chemicals for effects on androgens and estrogens, respectively. These candidate reference chemicals will be informative for performance-based validation of in vitro steroidogenesis models.
Collapse
Affiliation(s)
- Caroline Lucia Pinto
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831-0117, United States.
| | - Kristan Markey
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States
| | - David Dix
- U.S. EPA, Office of Chemical Safety and Pollution Prevention, Washington, D.C. 20004, United States
| | - Patience Browne
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States
| |
Collapse
|
33
|
Buñay J, Larriba E, Patiño-Garcia D, Cruz-Fernandes L, Castañeda-Zegarra S, Rodriguez-Fernandez M, del Mazo J, Moreno RD. Editor’s Highlight: Differential Effects of Exposure to Single Versus a Mixture of Endocrine-Disrupting Chemicals on Steroidogenesis Pathway in Mouse Testes. Toxicol Sci 2017; 161:76-86. [DOI: 10.1093/toxsci/kfx200] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Julio Buñay
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Eduardo Larriba
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CIB-CSIC), 28040 Madrid, Spain
| | - Daniel Patiño-Garcia
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Leonor Cruz-Fernandes
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Sergio Castañeda-Zegarra
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - María Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| | - Jesús del Mazo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CIB-CSIC), 28040 Madrid, Spain
| | - Ricardo D Moreno
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile (PUC), Santiago 8331150, Chile
| |
Collapse
|
34
|
Persistent testicular structural and functional alterations after exposure of adult rats to atrazine. Reprod Toxicol 2017; 73:201-213. [PMID: 28847621 DOI: 10.1016/j.reprotox.2017.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/11/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022]
Abstract
Atrazine is an endocrine disruptor affecting testicular steroidogenesis, and promoting testicular atrophy and 3β-HSD reduction. However, it remains unknown whether these effects are reversible or permanent. To address this issue was the aim of this study. Exposition of rats to 200mg/kg of atrazine resulted in transient increase in testicular weight, seminiferous tubules dilation and atrophy, and reduction in Leydig cell 3β-HSD. Testicular atrophy and 3β-HSD reduction were more pronounced after the recovery period of 75days. There was increase in aromatase expression after long-term exposure but it returned to control level after recovery. Moreover, there was increase in ED1-/ED2+, ED1+/ED2+ and ED1+/ED2- macrophages, in the recovery group. These macrophages were positive for 3β-HSD, thereby raising possibility of their involvement in steroidogenesis. These findings further emphasize the adverse effects of atrazine on male reproduction, highlighting that testicular damages may be irreversible even after a recovery period longer than the spermatogenic cycle.
Collapse
|
35
|
Foradori CD, Zimmerman AD, Coder PS, Peachee VL, Handa RJ, Kimber I, Pruett SB, Breckenridge CB. Lack of immunotoxic effects of repeated exposure to atrazine associated with the adaptation of adrenal gland activation. Regul Toxicol Pharmacol 2017; 89:200-214. [PMID: 28736286 DOI: 10.1016/j.yrtph.2017.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/09/2017] [Accepted: 07/19/2017] [Indexed: 11/12/2022]
Abstract
T cell-dependent IgM antibody production and natural killer cell (NKC) activity were assessed in SD rats orally administered atrazine for 28 days to males (0, 6.5, 25, or 100 mg/kg/day) or females (0, 3, 6, or 50 mg/kg/day), or 30 or 500 ppm in diet (3 or 51 mg/kg/day). Anti-asialo GM1 antibodies (NKC) and cyclophosphamide (antibody-forming cell assay [AFC]) served as positive controls. Pituitary (ACTH, prolactin), adrenal (corticosterone, progesterone, aldosterone), and gonadal (androgens, estrogens) hormones were assessed after 1, 7, and/or 28 days of treatment. Food intake and body weights were significantly reduced in the highest dosed males, and transiently affected in females. Urinary corticosterone levels were not increased in atrazine-treated groups in either sex at any time point measured (10, 22, or 24 days). Corticosterone and progesterone were elevated in males after a single atrazine dose ≥6.5 mg/kg/day, but not after 7, 14, or 28 doses. There were no effects on adrenal, pituitary, or gonadal hormones in females. Atrazine did not suppress the AFC response or decrease NKC function after 28 days in males or females. Atrazine had no effect on spleen weights or spleen cell numbers in males or females, although thymus weights were elevated in males receiving the highest dose. The lack of immunotoxic effect of atrazine was associated with diminished adrenal activation over time in males, and no effects on adrenal hormones in females.
Collapse
Affiliation(s)
- Chad D Foradori
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Arthur D Zimmerman
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Pragati S Coder
- Charles River Laboratories Ashland LLC, Ashland, OH, United States
| | | | - Robert J Handa
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Ian Kimber
- Faculty of Life Sciences, University of Manchester, UK
| | - Stephen B Pruett
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, United States
| | | |
Collapse
|
36
|
Quintaneiro C, Patrício D, Novais SC, Soares AMVM, Monteiro MS. Endocrine and physiological effects of linuron and S-metolachlor in zebrafish developing embryos. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 586:390-400. [PMID: 28209406 DOI: 10.1016/j.scitotenv.2016.11.153] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 06/06/2023]
Abstract
Evaluation of the effects of linuron and S-metolachlor on apical, biochemical and transcriptional endpoints in zebrafish (Danio rerio) early life stages was the main purpose of this work. Embryos were exposed for 96h to a range of concentrations of each herbicide to determine lethal and sub-lethal effects on apical (e.g. malformations, hatching) and biochemical parameters (cholinesterase, ChE; catalase, CAT; glutathione S-transferase, GST; lipid peroxidation, LPO and lactate dehydrogenase, LDH). To evaluate endocrine disruption effects, embryos were exposed during 96h to 0.88mg/L linuron and 9.66mg/L S-metolachlor, isolated or in binary mixture. Expression of a suite of genes involved in HPT, HPG and HPA-axis was then assessed. Highest concentration of linuron (5.0mg/L) decreased hatching rate to 5% and 70.0mg/L S-metolachlor completely inhibited hatching, about 100%. Both herbicides impaired development by inducing several malformations (100% in 5.0mg/L linuron and 70.0mg/L S-metolachlor). Linuron only affected GST and CAT at concentrations of 0.25 and 0.0025mg/L, respectively. S-metolachlor induced GST (to 256%), inhibited ChE (to 61%) and LDH (to 60%) and reduced LPO levels (to 63%). Linuron isolated treatment seems to have an estrogenic mode of action due to the observed induction of vtg1. Exposure to S-metolachlor seems to interfere with steroidogenesis and with HPT and HPA-axis, since it has inhibited cyp19a2, TSHβ and CRH gene expression. In addition to vtg1 induction and CRH inhibition, herbicide combination also induced sox9b that has a role in regulation of sexual development in zebrafish. This study pointed out adverse effects of linuron and S-metolachlor, namely impairment of neurotransmission and energy production, induction of steroidogenesis, and interference with HPT and HPA-axis. These results contributed to elucidate modes of action of linuron and S-metolachlor in zebrafish embryo model. Furthermore, gene expression patterns obtained are indicative of endocrine disruption action of these herbicides.
Collapse
Affiliation(s)
- C Quintaneiro
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - D Patrício
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - S C Novais
- MARE - Marine and Environmental Sciences Centre, ESTM, Instituto Politécnico de Leiria, 2520-641 Peniche, Portugal
| | - A M V M Soares
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | - M S Monteiro
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
37
|
Martínez-Ibarra A, Morimoto S, Cerbón M, Prado-Flores G. Effects on the reproductive parameters of two generations of Rattus norvegicus offspring from dams exposed to heptachlor during gestation and lactation. ENVIRONMENTAL TOXICOLOGY 2017; 32:856-868. [PMID: 27240701 DOI: 10.1002/tox.22285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/22/2016] [Accepted: 04/30/2016] [Indexed: 06/05/2023]
Abstract
Heptachlor has been targeted for global elimination because of its toxicity and environmental persistence, in accordance with the Stockholm Convention on Persistent Organic Pollutants (POPs). However, there is no regulation of heptachlor use in Mexico, where relatively high levels have been found in maternal breast milk. The aim of this study was to determine the effects of heptachlor on the reproductive system of offspring of two consecutive generations of rats (F1 and F2) from dams orally administered heptachlor during midgestation and lactation. Female offspring were analyzed for different phenotypic, reproductive, and molecular parameters. In the F1 generation, heptachlor treatment induced decreased body weight at weaning, increased female anogenital distance, and delayed vaginal opening. In both generations, serum progesterone levels decreased and estradiol levels remained unchanged, while overexpression of the progesterone receptor was observed in uterine epithelial cells on estrus day. In the F2 generation, expression of the estrogen receptor α increased in the glandular epithelium. Finally, heptachlor treatment did not affect apoptosis in the uterine epithelial cells. Overall, the results indicate that heptachlor induced female reproductive alterations when administered to dams during the perinatal period. Accordingly, exposure to heptachlor may represent a risk for the reproductive health of humans. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 856-868, 2017.
Collapse
Affiliation(s)
- Alejandra Martínez-Ibarra
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana-Xochimilco, D.F, México, México
| | - Sumiko Morimoto
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y de la Nutrición S.Z, D.F, México, México
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, D.F, México, México
| | - Guadalupe Prado-Flores
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana-Xochimilco, D.F, México, México
| |
Collapse
|
38
|
Wrobel MH, Mlynarczuk J. Secretory function of ovarian cells and myometrial contractions in cow are affected by chlorinated insecticides (chlordane, heptachlor, mirex) in vitro. Toxicol Appl Pharmacol 2017; 314:63-71. [PMID: 27888061 DOI: 10.1016/j.taap.2016.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/20/2016] [Accepted: 11/20/2016] [Indexed: 01/05/2023]
Abstract
The aim of the study was to investigate the effect of chlordane, heptachlor and mirex, on hormonal regulation of the force of myometrial contractions. Myometrial, endometrial, granulosa and luteal cells as well as strips of myometrium from non-pregnant cows were incubated with three insecticides at environmentally relevant doses (0.1, 1 or 10ng/ml). None of the insecticides affected the viability of studied cells. Chlordane stimulated, while heptachlor and mirex inhibited, secretion of testosterone and estradiol from granulosa cells as well as secretion of progesterone from luteal cells, respectively. Secretion of oxytocin (OT) from granulosa cells was increased after incubation with all studied insecticides. Only mirex stimulated OT secretion from luteal cells, while heptachlor inhibited this effect. None of them affected synthesis of OT in luteal cells and prostaglandins (PGF2 and PGE2) secretion from uterine cells, except PGE2 secretion from endometrial cells was decreased when the cells were incubated with 0.1ng/ml of chlordane. Basal and OT-stimulated myometrial contractions were increased by mirex and decreased by heptachlor. The data show that the insecticides altered secretory function of ovarian cells. Heptachlor and mirex affected also myometrial contractions in vitro, but uterine secretion of prostaglandins were not involved in the mechanism of that adverse effect of insecticides. The data indicate on potential of these insecticides to disturb fertilisation, blastocyst implantation or even the length of gestation.
Collapse
Affiliation(s)
- Michael Hubert Wrobel
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Street 10, 10-748 Olsztyn, Poland.
| | - Jaroslaw Mlynarczuk
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Street 10, 10-748 Olsztyn, Poland
| |
Collapse
|
39
|
Paul Friedman K, Papineni S, Marty MS, Yi KD, Goetz AK, Rasoulpour RJ, Kwiatkowski P, Wolf DC, Blacker AM, Peffer RC. A predictive data-driven framework for endocrine prioritization: a triazole fungicide case study. Crit Rev Toxicol 2016; 46:785-833. [PMID: 27347635 PMCID: PMC5044773 DOI: 10.1080/10408444.2016.1193722] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/13/2016] [Accepted: 05/21/2016] [Indexed: 10/27/2022]
Abstract
The US Environmental Protection Agency Endocrine Disruptor Screening Program (EDSP) is a tiered screening approach to determine the potential for a chemical to interact with estrogen, androgen, or thyroid hormone systems and/or perturb steroidogenesis. Use of high-throughput screening (HTS) to predict hazard and exposure is shifting the EDSP approach to (1) prioritization of chemicals for further screening; and (2) targeted use of EDSP Tier 1 assays to inform specific data needs. In this work, toxicology data for three triazole fungicides (triadimefon, propiconazole, and myclobutanil) were evaluated, including HTS results, EDSP Tier 1 screening (and other scientifically relevant information), and EPA guideline mammalian toxicology study data. The endocrine-related bioactivity predictions from HTS and information that satisfied the EDSP Tier 1 requirements were qualitatively concordant. Current limitations in the available HTS battery for thyroid and steroidogenesis pathways were mitigated by inclusion of guideline toxicology studies in this analysis. Similar margins (3-5 orders of magnitude) were observed between HTS-predicted human bioactivity and exposure values and between in vivo mammalian bioactivity and EPA chronic human exposure estimates for these products' registered uses. Combined HTS hazard and human exposure predictions suggest low priority for higher-tiered endocrine testing of these triazoles. Comparison with the mammalian toxicology database indicated that this HTS-based prioritization would have been protective for any potential in vivo effects that form the basis of current risk assessment for these chemicals. This example demonstrates an effective, human health protective roadmap for EDSP evaluation of pesticide active ingredients via prioritization using HTS and guideline toxicology information.
Collapse
Affiliation(s)
| | - Sabitha Papineni
- Human Health Assessment, Dow AgroSciences LLC,
Indianapolis,
IN,
USA
| | - M. Sue Marty
- Toxicology & Environmental Research and Consulting, The Dow Chemical Company,
Midland,
MI,
USA
| | - Kun Don Yi
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | - Amber K. Goetz
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | | | - Pat Kwiatkowski
- Human Safety, Bayer CropScience LP, Research Triangle Park,
NC,
USA
| | - Douglas C. Wolf
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| | - Ann M. Blacker
- Human Safety, Bayer CropScience LP, Research Triangle Park,
NC,
USA
| | - Richard C. Peffer
- Toxicology and Health Sciences, Syngenta Crop Protection LLC,
Greensboro,
NC,
USA
| |
Collapse
|
40
|
Da Cuña RH, Rey Vázquez G, Dorelle L, Rodríguez EM, Guimarães Moreira R, Lo Nostro FL. Mechanism of action of endosulfan as disruptor of gonadal steroidogenesis in the cichlid fish Cichlasoma dimerus. Comp Biochem Physiol C Toxicol Pharmacol 2016; 187:74-80. [PMID: 27235598 DOI: 10.1016/j.cbpc.2016.05.008] [Citation(s) in RCA: 12] [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: 02/10/2016] [Revised: 05/19/2016] [Accepted: 05/20/2016] [Indexed: 02/02/2023]
Abstract
The organochlorine pesticide endosulfan (ES) is used in several countries as a wide spectrum insecticide on crops with high commercial value. Due to its high toxicity to non-target animals, its persistence in the environment and its ability to act as an endocrine disrupting compound in fish, ES use is currently banned or restricted in many other countries. Previous studies on the cichlid fish Cichlasoma dimerus have shown that waterborne exposure to ES can lead to both decreased pituitary FSH content and histological alterations of testes. As gonadotropin-stimulated sex steroids release from gonads was inhibited by ES in vitro, the aim of the present study was to elucidate possible mechanisms of disruption of ES on gonadal steroidogenesis in C. dimerus, as well as compare the action of the active ingredient (AI) with that of currently used commercial formulations (CF). Testis and ovary fragments were incubated with ES (AI or CF) and/or steroidogenesis activators or precursors. Testosterone and estradiol levels were measured in the incubation media. By itself, ES did not affect hormone levels. Co-incubation with LH and the adenylate cyclase activator forskolin caused a decrease of the stimulated sex steroids release. When co-incubated with precursors dehydroandrostenedione and 17αhydroxyprogesterone, ES did not affect the increase caused by their addition alone. No differences were observed between the AI and CFs, suggesting that the effect on steroidogenesis disruption is mainly caused by the AI. Results indicate that action of ES takes place downstream of LH-receptor activation and upstream of the studied steroidogenic enzymes.
Collapse
Affiliation(s)
- Rodrigo H Da Cuña
- Laboratorio de Ecotoxicología Acuática. DBBE, FCEyN, Universidad de Buenos Aires, Argentina; IBBEA, CONICET-UBA, Buenos Aires, Argentina
| | - Graciela Rey Vázquez
- Laboratorio de Ecotoxicología Acuática. DBBE, FCEyN, Universidad de Buenos Aires, Argentina; IBBEA, CONICET-UBA, Buenos Aires, Argentina
| | - Luciana Dorelle
- Laboratorio de Ecotoxicología Acuática. DBBE, FCEyN, Universidad de Buenos Aires, Argentina; IBBEA, CONICET-UBA, Buenos Aires, Argentina
| | - Enrique M Rodríguez
- Laboratorio de Fisiología de Crustáceos, DBBE, FCEyN, Universidad de Buenos Aires, Argentina; IBBEA, CONICET-UBA, Buenos Aires, Argentina
| | | | - Fabiana L Lo Nostro
- Laboratorio de Ecotoxicología Acuática. DBBE, FCEyN, Universidad de Buenos Aires, Argentina; IBBEA, CONICET-UBA, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Saillenfait AM, Ndiaye D, Sabaté JP. The estrogenic and androgenic potential of pyrethroids in vitro. Review. Toxicol In Vitro 2016; 34:321-332. [DOI: 10.1016/j.tiv.2016.02.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/09/2016] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
|
42
|
Russart KL, Rhen T. Atrazine alters expression of reproductive and stress genes in the developing hypothalamus of the snapping turtle, Chelydra serpentina. Toxicology 2016; 366-367:1-9. [DOI: 10.1016/j.tox.2016.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/27/2016] [Accepted: 08/01/2016] [Indexed: 11/16/2022]
|
43
|
Jiang J, Chen Y, Yu R, Zhao X, Wang Q, Cai L. Pretilachlor has the potential to induce endocrine disruption, oxidative stress, apoptosis and immunotoxicity during zebrafish embryo development. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:125-134. [PMID: 26851375 DOI: 10.1016/j.etap.2016.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
The objectives of the present study were to investigate the toxic effects of pretilachlor on zebrafish during its embryo development. The results demonstrated that the transcription of genes involved in the hypothalamic-pituitary-gonadal/thyroid (HPG/HPT) axis was increased after exposure to 50, 100, 200 μg/L pretilachlor for 96 h, the aromatase activity, vitellogenin (VTG) and thyroid hormones T3 and T4 levels in zebrafish were also up-regulated simultaneously. Pretilachlor exposure induced a noticeable increase in ROS level, increased the transcription and level of antioxidant proteins (e.g., CAT, SOD and GPX). Moreover, the up-regulation of P53, Mdm2, Bbc3 expression and Caspase3 and Caspase9 activities in the apoptosis pathway suggested pretilachlor might trigger cell apoptosis in zebrafish. In addition, the transcription of CXCL-C1C, IL-1β and IL-8 related to the innate immunity was down-regulated after pretilachlor exposure. These data suggested that pretilachlor could simultaneously induce endocrine disruption, apoptosis, oxidative stress and immunotoxicity during zebrafish embryo development.
Collapse
Affiliation(s)
- Jinhua Jiang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Yanhong Chen
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Ruixian Yu
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Xueping Zhao
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Qiang Wang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Leiming Cai
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China.
| |
Collapse
|
44
|
Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev 2015; 36:E1-E150. [PMID: 26544531 PMCID: PMC4702494 DOI: 10.1210/er.2015-1010] [Citation(s) in RCA: 1417] [Impact Index Per Article: 141.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023]
Abstract
The Endocrine Society's first Scientific Statement in 2009 provided a wake-up call to the scientific community about how environmental endocrine-disrupting chemicals (EDCs) affect health and disease. Five years later, a substantially larger body of literature has solidified our understanding of plausible mechanisms underlying EDC actions and how exposures in animals and humans-especially during development-may lay the foundations for disease later in life. At this point in history, we have much stronger knowledge about how EDCs alter gene-environment interactions via physiological, cellular, molecular, and epigenetic changes, thereby producing effects in exposed individuals as well as their descendants. Causal links between exposure and manifestation of disease are substantiated by experimental animal models and are consistent with correlative epidemiological data in humans. There are several caveats because differences in how experimental animal work is conducted can lead to difficulties in drawing broad conclusions, and we must continue to be cautious about inferring causality in humans. In this second Scientific Statement, we reviewed the literature on a subset of topics for which the translational evidence is strongest: 1) obesity and diabetes; 2) female reproduction; 3) male reproduction; 4) hormone-sensitive cancers in females; 5) prostate; 6) thyroid; and 7) neurodevelopment and neuroendocrine systems. Our inclusion criteria for studies were those conducted predominantly in the past 5 years deemed to be of high quality based on appropriate negative and positive control groups or populations, adequate sample size and experimental design, and mammalian animal studies with exposure levels in a range that was relevant to humans. We also focused on studies using the developmental origins of health and disease model. No report was excluded based on a positive or negative effect of the EDC exposure. The bulk of the results across the board strengthen the evidence for endocrine health-related actions of EDCs. Based on this much more complete understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability, these findings can be much better translated to human health. Armed with this information, researchers, physicians, and other healthcare providers can guide regulators and policymakers as they make responsible decisions.
Collapse
Affiliation(s)
- A C Gore
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - V A Chappell
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - S E Fenton
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J A Flaws
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - A Nadal
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - G S Prins
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J Toppari
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - R T Zoeller
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
45
|
Evaluation of the reproductive toxicity of fungicide propiconazole in male rats. Toxicology 2015; 335:55-61. [DOI: 10.1016/j.tox.2015.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/30/2022]
|
46
|
Goodson WH, Lowe L, Carpenter DO, Gilbertson M, Manaf Ali A, Lopez de Cerain Salsamendi A, Lasfar A, Carnero A, Azqueta A, Amedei A, Charles AK, Collins AR, Ward A, Salzberg AC, Colacci AM, Olsen AK, Berg A, Barclay BJ, Zhou BP, Blanco-Aparicio C, Baglole CJ, Dong C, Mondello C, Hsu CW, Naus CC, Yedjou C, Curran CS, Laird DW, Koch DC, Carlin DJ, Felsher DW, Roy D, Brown DG, Ratovitski E, Ryan EP, Corsini E, Rojas E, Moon EY, Laconi E, Marongiu F, Al-Mulla F, Chiaradonna F, Darroudi F, Martin FL, Van Schooten FJ, Goldberg GS, Wagemaker G, Nangami GN, Calaf GM, Williams GP, Wolf GT, Koppen G, Brunborg G, Lyerly HK, Krishnan H, Ab Hamid H, Yasaei H, Sone H, Kondoh H, Salem HK, Hsu HY, Park HH, Koturbash I, Miousse IR, Scovassi A, Klaunig JE, Vondráček J, Raju J, Roman J, Wise JP, Whitfield JR, Woodrick J, Christopher JA, Ochieng J, Martinez-Leal JF, Weisz J, Kravchenko J, Sun J, Prudhomme KR, Narayanan KB, Cohen-Solal KA, Moorwood K, Gonzalez L, Soucek L, Jian L, D’Abronzo LS, Lin LT, Li L, Gulliver L, McCawley LJ, Memeo L, Vermeulen L, Leyns L, Zhang L, Valverde M, Khatami M, Romano MF, Chapellier M, Williams MA, Wade M, et alGoodson WH, Lowe L, Carpenter DO, Gilbertson M, Manaf Ali A, Lopez de Cerain Salsamendi A, Lasfar A, Carnero A, Azqueta A, Amedei A, Charles AK, Collins AR, Ward A, Salzberg AC, Colacci AM, Olsen AK, Berg A, Barclay BJ, Zhou BP, Blanco-Aparicio C, Baglole CJ, Dong C, Mondello C, Hsu CW, Naus CC, Yedjou C, Curran CS, Laird DW, Koch DC, Carlin DJ, Felsher DW, Roy D, Brown DG, Ratovitski E, Ryan EP, Corsini E, Rojas E, Moon EY, Laconi E, Marongiu F, Al-Mulla F, Chiaradonna F, Darroudi F, Martin FL, Van Schooten FJ, Goldberg GS, Wagemaker G, Nangami GN, Calaf GM, Williams GP, Wolf GT, Koppen G, Brunborg G, Lyerly HK, Krishnan H, Ab Hamid H, Yasaei H, Sone H, Kondoh H, Salem HK, Hsu HY, Park HH, Koturbash I, Miousse IR, Scovassi A, Klaunig JE, Vondráček J, Raju J, Roman J, Wise JP, Whitfield JR, Woodrick J, Christopher JA, Ochieng J, Martinez-Leal JF, Weisz J, Kravchenko J, Sun J, Prudhomme KR, Narayanan KB, Cohen-Solal KA, Moorwood K, Gonzalez L, Soucek L, Jian L, D’Abronzo LS, Lin LT, Li L, Gulliver L, McCawley LJ, Memeo L, Vermeulen L, Leyns L, Zhang L, Valverde M, Khatami M, Romano MF, Chapellier M, Williams MA, Wade M, Manjili MH, Lleonart ME, Xia M, Gonzalez Guzman MJ, Karamouzis MV, Kirsch-Volders M, Vaccari M, Kuemmerle NB, Singh N, Cruickshanks N, Kleinstreuer N, van Larebeke N, Ahmed N, Ogunkua O, Krishnakumar P, Vadgama P, Marignani PA, Ghosh PM, Ostrosky-Wegman P, Thompson PA, Dent P, Heneberg P, Darbre P, Leung PS, Nangia-Makker P, Cheng Q(S, Robey R, Al-Temaimi R, Roy R, Andrade-Vieira R, Sinha RK, Mehta R, Vento R, Di Fiore R, Ponce-Cusi R, Dornetshuber-Fleiss R, Nahta R, Castellino RC, Palorini R, Hamid RA, Langie SA, Eltom SE, Brooks SA, Ryeom S, Wise SS, Bay SN, Harris SA, Papagerakis S, Romano S, Pavanello S, Eriksson S, Forte S, Casey SC, Luanpitpong S, Lee TJ, Otsuki T, Chen T, Massfelder T, Sanderson T, Guarnieri T, Hultman T, Dormoy V, Odero-Marah V, Sabbisetti V, Maguer-Satta V, Rathmell W, Engström W, Decker WK, Bisson WH, Rojanasakul Y, Luqmani Y, Chen Z, Hu Z. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: the challenge ahead. Carcinogenesis 2015; 36 Suppl 1:S254-S296. [PMID: 26106142 PMCID: PMC4480130 DOI: 10.1093/carcin/bgv039] [Show More Authors] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 01/23/2015] [Accepted: 01/31/2015] [Indexed: 02/07/2023] Open
Abstract
Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety 'Mode of Action' framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.
Collapse
Affiliation(s)
- William H. Goodson
- *To whom correspondence should be addressed. William H.Goodson III, California Pacific Medical Center Research Institute, 2100 Webster Street #401, San Francisco, CA 94115, USA. Tel: +41 59 233925; Fax: +41 57 761977;
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur Street, Truro, Nova Scotia B2N 1X5, Canada
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4AP, UK
| | - David O. Carpenter
- Institute for Health and the Environment, University at Albany, 5 University Pl., Rensselaer, NY 12144, USA
| | | | - Abdul Manaf Ali
- School of Biotechnology, Faculty of Agriculture Biotechnology and Food Sciences, Sultan Zainal Abidin University, Tembila Campus, 22200 Besut, Terengganu, Malaysia
| | | | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, NJ 08854, USA
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas. Hospital Universitario Virgen del Rocio, Univ. de Sevilla., Avda Manuel Siurot sn. 41013 Sevilla, Spain
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31008, Spain
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Amelia K. Charles
- School of Biological Sciences, University of Reading, Hopkins Building, Reading, Berkshire RG6 6UB, UK
| | | | - Andrew Ward
- Department of Biochemistry and Biology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Anna C. Salzberg
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo N-0403, Norway
| | - Arthur Berg
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | | | - Binhua P. Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
| | - Carmen Blanco-Aparicio
- Spanish National Cancer Research Centre, CNIO, Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
| | - Carolyn J. Baglole
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Chenfang Dong
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Chia-Wen Hsu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892–3375, USA
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Life Sciences Institute, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS 39217, USA
| | - Colleen S. Curran
- Department of Molecular and Environmental Toxicology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dale W. Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Daniel C. Koch
- Stanford University Department of Medicine, Division of Oncology, Stanford, CA 94305, USA
| | - Danielle J. Carlin
- Superfund Research Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27560, USA
| | - Dean W. Felsher
- Department of Medicine, Oncology and Pathology, Stanford University,Stanford, CA 94305, USA
| | - Debasish Roy
- Department of Natural Science, The City University of New York at Hostos Campus, Bronx, NY 10451, USA
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523–1680, USA
| | - Edward Ratovitski
- Department of Head and Neck Surgery/Head and Neck Cancer Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523–1680, USA
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Emilio Rojas
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143–747, Korea
| | - Ezio Laconi
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Fabio Marongiu
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Ferdinando Chiaradonna
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
- SYSBIO Centre of Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Firouz Darroudi
- Human Safety and Environmental Research, Department of Health Sciences, College of North Atlantic, Doha 24449, State of Qatar
| | - Francis L. Martin
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4AP, UK
| | - Frederik J. Van Schooten
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht 6200, The Netherlands
| | - Gary S. Goldberg
- Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Gerard Wagemaker
- Hacettepe University, Center for Stem Cell Research and Development, Ankara 06640, Turkey
| | - Gladys N. Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Gloria M. Calaf
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
- Instituto de Alta Investigacion, Universidad de Tarapaca, Arica, Chile
| | - Graeme P. Williams
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK
| | - Gregory T. Wolf
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gudrun Koppen
- Environmental Risk and Health Unit, Flemish Institute for Technological Research, 2400 Mol, Belgium
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo N-0403, Norway
| | - H. Kim Lyerly
- Department of Surgery, Pathology, Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Harini Krishnan
- Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Hasiah Ab Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hemad Yasaei
- Department of Life Sciences, College of Health and Life Sciences and the Health and Environment Theme, Institute of Environment, Health and Societies, Brunel University Kingston Lane, Uxbridge, Middlesex UB8 3PH, UK
| | - Hideko Sone
- National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibraki 3058506, Japan
| | - Hiroshi Kondoh
- Department of Geriatric Medicine, Kyoto University Hospital 54 Kawaharacho, Shogoin, Sakyo-ku Kyoto, 606–8507, Japan
| | - Hosni K. Salem
- Department of Urology, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 11559, Egypt
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien 970, Taiwan
| | - Hyun Ho Park
- School of Biotechnology, Yeungnam University, Gyeongbuk 712-749, South Korea
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - A.Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - James E. Klaunig
- Department of Environmental Health, Indiana University, School of Public Health, Bloomington, IN 47405, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics Academy of Sciences of the Czech Republic, Brno, CZ-61265, Czech Republic
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Jesse Roman
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Robley Rex VA Medical Center, Louisville, KY 40202, USA
| | - John Pierce Wise
- Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth St., Portland, ME 04104, USA
| | - Jonathan R. Whitfield
- Mouse Models of Cancer Therapies Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Joseph A. Christopher
- Cancer Research UK. Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Josiah Ochieng
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | | | - Judith Weisz
- Departments of Obstetrics and Gynecology and Pathology, Pennsylvania State University College of Medicine, Hershey PA 17033, USA
| | - Julia Kravchenko
- Department of Surgery, Pathology, Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Kalan R. Prudhomme
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | | | - Karine A. Cohen-Solal
- Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Kim Moorwood
- Department of Biochemistry and Biology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Laetitia Gonzalez
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Laura Soucek
- Mouse Models of Cancer Therapies Group, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Le Jian
- School of Public Health, Curtin University, Bentley, WA 6102, Australia
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Leandro S. D’Abronzo
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Lin Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, The People’s Republic of China
| | - Linda Gulliver
- Faculty of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Lisa J. McCawley
- Department of Biomedical Engineering and Cancer Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Via Penninazzo 7, Viagrande (CT) 95029, Italy
| | - Louis Vermeulen
- Center for Experimental Molecular Medicine, Academic Medical Center, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Luc Leyns
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
| | - Mahara Valverde
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Marion Chapellier
- Centre De Recherche En Cancerologie,De Lyon, Lyon, U1052-UMR5286, France
| | - Marc A. Williams
- United States Army Institute of Public Health, Toxicology Portfolio-Health Effects Research Program, Aberdeen Proving Ground, Edgewood, MD 21010-5403, USA
| | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milano, Italy
| | - Masoud H. Manjili
- Department of Microbiology and Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Matilde E. Lleonart
- Institut De Recerca Hospital Vall D’Hebron, Passeig Vall d’Hebron, 119–129, 08035 Barcelona, Spain
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892–3375, USA
| | - Michael J. Gonzalez Guzman
- University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan 00921, Puerto Rico
| | - Michalis V. Karamouzis
- Department of Biological Chemistry, Medical School, University of Athens, Institute of Molecular Medicine and Biomedical Research, 10676 Athens, Greece
| | | | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George’s Medical University, Lucknow, Uttar Pradesh 226 003, India
| | - Nichola Cruickshanks
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nicole Kleinstreuer
- Integrated Laboratory Systems Inc., in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, RTP, NC 27709, USA
| | - Nik van Larebeke
- Analytische, Milieu en Geochemie, Vrije Universiteit Brussel, Brussel B1050, Belgium
| | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Victoria 3052, Australia
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - P.K. Krishnakumar
- Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 3126, Saudi Arabia
| | - Pankaj Vadgama
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Paola A. Marignani
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Paramita M. Ghosh
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Patricia Ostrosky-Wegman
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Patricia A. Thompson
- Department of Pathology, Stony Brook School of Medicine, Stony Brook University, The State University of New York, Stony Brook, NY 11794-8691, USA
| | - Paul Dent
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, CZ-100 00 Prague 10, Czech Republic
| | - Philippa Darbre
- School of Biological Sciences, The University of Reading, Whiteknights, Reading RG6 6UB, England
| | - Po Sing Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, The People’s Republic of China
| | | | - Qiang (Shawn) Cheng
- Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA
| | - R.Brooks Robey
- White River Junction Veterans Affairs Medical Center, White River Junction, VT 05009, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Rabeah Al-Temaimi
- Human Genetics Unit, Department of Pathology, Faculty of Medicine, Kuwait University, Jabriya 13110, Kuwait
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Rafaela Andrade-Vieira
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Ranjeet K. Sinha
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rekha Mehta
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Renza Vento
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, Palermo 90127, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Riccardo Di Fiore
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, Palermo 90127, Italy
| | | | - Rita Dornetshuber-Fleiss
- Department of Pharmacology and Toxicology, University of Vienna, Vienna A-1090, Austria
- Institute of Cancer Research, Department of Medicine, Medical University of Vienna, Wien 1090, Austria
| | - Rita Nahta
- Departments of Pharmacology and Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Robert C. Castellino
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Healthcare of Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Roberta Palorini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
- SYSBIO Centre of Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Roslida A. Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sabine A.S. Langie
- Environmental Risk and Health Unit, Flemish Institute for Technological Research, 2400 Mol, Belgium
| | - Sakina E. Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Samira A. Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Sandra Ryeom
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra S. Wise
- Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth St., Portland, ME 04104, USA
| | - Sarah N. Bay
- Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Shelley A. Harris
- Population Health and Prevention, Research, Prevention and Cancer Control, Cancer Care Ontario, Toronto, Ontario, M5G 2L7, Canada
- Departments of Epidemiology and Occupational and Environmental Health, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, M5T 3M7, Canada
| | - Silvana Papagerakis
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
| | - Staffan Eriksson
- Department of Anatomy, Physiology and Biochemistry, The Swedish University of Agricultural Sciences, PO Box 7011, VHC, Almas Allé 4, SE-756 51, Uppsala, Sweden
| | - Stefano Forte
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Via Penninazzo 7, Viagrande (CT) 95029, Italy
| | - Stephanie C. Casey
- Stanford University Department of Medicine, Division of Oncology, Stanford, CA 94305, USA
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu 705–717, South Korea,
| | - Takemi Otsuki
- Department of Hygiene, Kawasaki Medical School, Matsushima Kurashiki, Okayama 701-0192, Japan,
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Thierry Massfelder
- INSERM U1113, team 3 ‘Cell Signalling and Communication in Kidney and Prostate Cancer’, University of Strasbourg, Faculté de Médecine, 67085 Strasbourg, France
| | - Thomas Sanderson
- INRS-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada,
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy
- Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy
- National Institute of Biostructures and Biosystems, Viale Medaglie d’ Oro, 305, 00136 Roma, Italy
| | - Tove Hultman
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden
| | - Valérian Dormoy
- INSERM U1113, team 3 ‘Cell Signalling and Communication in Kidney and Prostate Cancer’, University of Strasbourg, Faculté de Médecine, 67085 Strasbourg, France
- Department of Cell and Developmental Biology, University of California, Irvine, CA 92697, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Venkata Sabbisetti
- Harvard Medical School/Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Veronique Maguer-Satta
- United States Army Institute of Public Health, Toxicology Portfolio-Health Effects Research Program, Aberdeen Proving Ground, Edgewood, MD 21010-5403, USA
| | - W.Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Wilhelm Engström
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden
| | | | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown,WV, 26506,USA
| | - Yunus Luqmani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, PO Box 24923, Safat 13110, Kuwait and
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Zhiwei Hu
- Department of Surgery, The Ohio State University College of Medicine, The James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
47
|
Triorganotin compounds - ligands for “rexinoid” inducible transcription factors: Biological effects. Toxicol Lett 2015; 234:50-8. [DOI: 10.1016/j.toxlet.2015.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 11/18/2022]
|
48
|
Abarikwu SO, Farombi EO. Atrazine induces apoptosis of SH-SY5Y human neuroblastoma cells via the regulation of Bax/Bcl-2 ratio and caspase-3-dependent pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2015; 118:90-98. [PMID: 25752436 DOI: 10.1016/j.pestbp.2014.12.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 11/13/2014] [Accepted: 12/05/2014] [Indexed: 06/04/2023]
Abstract
Atrazine (ATZ) is a well known herbicide that is frequently detected in ground and surface water at significant levels. Our objective was to study the toxic effect of ATZ on the human neuroblastoma (SH-SY5Y) cells, and the degree of cytotoxicity and morphological changes were followed during the cell death. Application of cytotoxicity bioassays indicates that ATZ (5-50 µg/mL) decreases cell viability in a dose- and time-dependent manner. The evidence of apoptosis was confirmed by an increase in caspase-3 activity, and cell death was blocked when caspase-3 activity was inhibited. Typical apoptotic phenotype that includes nuclear fragmentation, micro nuclei formation, DNA fragmentation and increase in the expressions apoptosis-associated markers Bax, p53 and p21 and decreased expression of Bcl-2 were observed in treated cells. We also observed dose-dependent increase in reactive oxygen species (ROS) levels in ATZ-treated cells. These results suggest that ATZ-induces apoptosis and ROS levels in SH-SY5Y cells, and could be implicated in human neurodegenerative disorder.
Collapse
Affiliation(s)
- Sunny O Abarikwu
- Department of Biochemistry, University of Port Harcourt, Choba, Nigeria.
| | | |
Collapse
|
49
|
Thueson LE, Emmons TR, Browning DL, Kreitinger JM, Shepherd DM, Wetzel SA. In vitro exposure to the herbicide atrazine inhibits T cell activation, proliferation, and cytokine production and significantly increases the frequency of Foxp3+ regulatory T cells. Toxicol Sci 2015; 143:418-29. [PMID: 25433234 PMCID: PMC4306722 DOI: 10.1093/toxsci/kfu242] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The herbicide atrazine (2-chloro-4-[ethylamino]-6-[isopropylamino]-s-triazine) is the most common water contaminant in the United States. Atrazine is a phosphodiesterase inhibitor and is classified as an estrogen disrupting compound because it elevates estrogen levels via induction of the enzyme aromatase. Previous studies have shown that atrazine exposure alters the function of innate immune cells such as NK cells, DC, mast cells, and macrophages. In this study we have examined the impact of in vitro atrazine exposure on the activation, proliferation, and effector cytokine production by primary murine CD4(+) T lymphocytes. We found that atrazine exposure significantly inhibited CD4(+) T cell proliferation and accumulation as well as the expression of the activation markers CD25 and CD69 in a dose-dependent manner. Interestingly, the effects were more pronounced in cells from male animals. These effects were partially mimicked by pharmacological reagents that elevate intracellular cAMP levels and addition of exogenous rmIL-2 further inhibited proliferation and CD25 expression. Consistent with these findings, atrazine exposure during T cell activation resulted in a 2- to 5-fold increase in the frequency of Foxp3(+) CD4(+) T cells.
Collapse
Affiliation(s)
- Lindsay E Thueson
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| | - Tiffany R Emmons
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| | - Dianna L Browning
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| | - Joanna M Kreitinger
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| | - David M Shepherd
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| | - Scott A Wetzel
- *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812 *Division of Biological Sciences, Center for Environmental Health Sciences and Department of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, Montana 59812
| |
Collapse
|
50
|
Honkisz E, Wójtowicz AK. Modulation of estradiol synthesis and aromatase activity in human choriocarcinoma JEG-3 cells exposed to tetrabromobisphenol A. Toxicol In Vitro 2014; 29:44-50. [PMID: 25223798 DOI: 10.1016/j.tiv.2014.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/09/2014] [Accepted: 09/05/2014] [Indexed: 11/27/2022]
Abstract
The goal of the present study was to investigate the impact of tetrabromobisphenol A (TBBPA) on human choriocarcinoma-derived placental JEG-3 cells in vitro. We determined the effect of this compound on estradiol secretion, aromatase protein expression and activity in vitro in the JEG-3 cell line. We assessed the ability of TBBPA to increase intracellular levels of cAMP as well as its effect on cell viability and proliferation. Our results indicated that TBBPA, at a wide range of concentrations (1×10(-8)-5×10(-5)M), significantly induced estradiol secretion by JEG-3 cells compared to that of controls after 24, 48 or 72 h of exposure. This effect was accompanied by an increase in the aromatase protein expression in JEG-3 cells treated with 100 nM and 10 μM of TBBPA for 24 h. Additionally, in our study, we confirmed that TBBPA-induced changes in aromatase protein expression were associated w ith the up-regulation of aromatase activity and cAMP levels. No tested doses of TBBPA inhibited JEG-3 cell proliferation, except for the highest dose of 100 μM, which had a toxic effect on cell viability at all time points. The present study clearly indicates that TBBPA alters JEG-3 cells estrogen synthesis due to its action on CYP19 protein expression and thus this compound may interfere with normal placental development during early pregnancy.
Collapse
Affiliation(s)
- Ewelina Honkisz
- Department of Animal Biotechnology, University of Agriculture, Rędzina 1B, 30-248 Cracow, Poland
| | - Anna K Wójtowicz
- Department of Animal Biotechnology, University of Agriculture, Rędzina 1B, 30-248 Cracow, Poland.
| |
Collapse
|