1
|
Marty MS, Erraguntla N, North C, Barranco WT, Kirman CR, Cagen S, Rushton EK, Shen H, Koehler MW, Budinsky R. A reproductive and developmental toxicity screening study of 1,3-butadiene in Sprague-Dawley rats. Regul Toxicol Pharmacol 2021; 127:105066. [PMID: 34699959 DOI: 10.1016/j.yrtph.2021.105066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/05/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022]
Abstract
1,3 Butadiene (BD) is an industrial intermediate used primarily in product manufacturing with the greatest exposure potential via inhalation. BD was evaluated for reproductive and developmental effects in a Good Laboratory Practice (GLP)-compliant, extended OECD 421 guideline study (completed 2003). Twelve-week old rats (12/sex/dose) were exposed via whole-body inhalation to BD vapor (0, 300, 1500, 6000 ppm) for 6 h/day, 7 days/week, starting 14 days prior to mating through the day prior to euthanasia (total exposures: 83-84 days for F0 males 60-70 days for F0 females). Select F1 offspring (1/sex/litter) were dosed 7 days (postnatal days 21-27 or 28-34), then necropsied. At 1500 and 6000 ppm, treatment-related facial soiling was seen in F0 males and females with decreased body weights/gains in F0 males. F1 males and females exhibited similar effects at 1500 and 6000 ppm. Importantly, the F0 generation had no evidence of altered sperm production, testicular effects, or ovarian atrophy, which were sensitive responses in mice. The no-observed-adverse-effect-level (NOAEL) is 300 ppm due to decreased body weight/gain and facial soiling at 1500 ppm, whereas 6000 ppm serves as a NOAEL for reproductive and developmental endpoints. This study contributes to the weight-of-evidence of differential BD reproductive toxicity in rats and mice.
Collapse
Affiliation(s)
- M Sue Marty
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, Midland, MI, USA
| | | | | | | | | | | | | | - Hua Shen
- Shell Oil Company, Houston, TX, USA
| | - Matthew W Koehler
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, Midland, MI, USA.
| | - Robert Budinsky
- The Dow Chemical Company, Toxicology & Environmental Research & Consulting, Midland, MI, USA
| |
Collapse
|
2
|
Ahmadian S, Sheshpari S, Mahdipour M, Pazhang M, Tsai PSJ, Nouri M, Rahbarghazi R, Shahnazi M. Toxic effects of VCD on kidneys and liver tissues: a histopathological and biochemical study. BMC Res Notes 2019; 12:446. [PMID: 31331386 PMCID: PMC6647313 DOI: 10.1186/s13104-019-4490-y] [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: 05/11/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE We explored detrimental effects of VCD on non-ovarian tissues such as kidneys and liver 14 days post-drug administration. Twelve rats were randomly assigned into two groups. In VCD group, rats received 160 mg/kgbw VCD intraperitoneally for 15 consequent days. Control rats were injected with VCD-free normal saline. At the respective time point, rats were euthanized, blood and tissue samples were collected. H&E staining was performed to evaluate pathological changes. Serum level of ALT, AST, creatinine and urea were also measured. RESULTS Histological analysis revealed hyperemia and follicular atresia in the ovaries, indicating successful POF induction in rats. In renal tissue, extensive tubular necrosis, focal hemorrhage, hyaline casts, and interstitial nephritis were observed. Analysis of hepatic tissue showed numerous hemorrhagic foci, chronic cholangitis, and hepatocyte necrosis, indicating apparent VCD toxicity of both hepatic and renal tissues. The biochemical evaluation revealed a tendency of increase in ALT, AST, creatinine, and Urea in VCD-treated rats; however, the values did not reach significant level. In conclusion, the induction of POF in rats by VCD correlates with renal and hepatic damages. Commensurate with data from this study, any conclusions from experiments based on VCD-induced premature ovarian failure rats should be reported with caution.
Collapse
Affiliation(s)
- Shahin Ahmadian
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Sepideh Sheshpari
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Shariati St., Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran. .,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.
| | - Mohammad Pazhang
- Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Pei-Shiue Jason Tsai
- Center for Developmental Biology and Regenerative Medicine Research, National Taiwan University/NTU, Taipei, Taiwan.,Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University/NTU, Taipei, Taiwan
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Shahnazi
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Shariati St., Tabriz, Iran.
| |
Collapse
|
3
|
Filser JG, Kessler W, Artati A, Erbach E, Faller T, Kreuzer PE, Li Q, Lichtmannegger J, Numtip W, Klein D, Pütz C, Semder B, Csanády GA. Ethylene oxide in blood of ethylene-exposed B6C3F1 mice, Fischer 344 rats, and humans. Toxicol Sci 2013; 136:344-58. [PMID: 24068676 PMCID: PMC3858200 DOI: 10.1093/toxsci/kft218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/09/2013] [Indexed: 02/04/2023] Open
Abstract
The gaseous olefin ethylene (ET) is metabolized in mammals to the carcinogenic epoxide ethylene oxide (EO). Although ET is the largest volume organic chemical worldwide, the EO burden in ET-exposed humans is still uncertain, and only limited data are available on the EO burden in ET-exposed rodents. Therefore, EO was quantified in blood of mice, rats, or 4 volunteers that were exposed once to constant atmospheric ET concentrations of between 1 and 10 000 ppm (rodents) or 5 and 50 ppm (humans). Both the compounds were determined by gas chromatography. At ET concentrations of between 1 and 10 000 ppm, areas under the concentration-time curves of EO in blood (µmol × h/l) ranged from 0.039 to 3.62 in mice and from 0.086 to 11.6 in rats. At ET concentrations ≤ 30 ppm, EO concentrations in blood were 8.7-fold higher in rats and 3.9-fold higher in mice than that in the volunteer with the highest EO burdens. Based on measured EO concentrations, levels of EO adducts to hemoglobin and lymphocyte DNA were calculated for diverse ET concentrations and compared with published adduct levels. For given ET exposure concentrations, there were good agreements between calculated and measured levels of adducts to hemoglobin in rats and humans and to DNA in rats and mice. Reported hemoglobin adduct levels in mice were higher than calculated ones. Furthermore, information is given on species-specific background adduct levels. In summary, the study provides most relevant data for an improved assessment of the human health risk from exposure to ET.
Collapse
Affiliation(s)
- Johannes Georg Filser
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
- †Institut für Toxikologie und Umwelthygiene, Technische Universität München, München, Germany
| | - Winfried Kessler
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Anna Artati
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Eva Erbach
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Thomas Faller
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Paul Erich Kreuzer
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Qiang Li
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Josef Lichtmannegger
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Wanwiwa Numtip
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Dominik Klein
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
- †Institut für Toxikologie und Umwelthygiene, Technische Universität München, München, Germany
| | - Christian Pütz
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - Brigitte Semder
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
| | - György András Csanády
- *Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, 85764 Neuherberg, Germany; and
- †Institut für Toxikologie und Umwelthygiene, Technische Universität München, München, Germany
| |
Collapse
|
4
|
Rajapaksa KS, Cannady EA, Glenn Sipes I, Hoyer PB. Involvement of CYP 2E1 enzyme in ovotoxicity caused by 4-vinylcyclohexene and its metabolites. Toxicol Appl Pharmacol 2007; 221:215-21. [PMID: 17462685 PMCID: PMC2065846 DOI: 10.1016/j.taap.2007.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 03/02/2007] [Accepted: 03/05/2007] [Indexed: 11/30/2022]
Abstract
4-Vinylcyclohexene (VCH) is bioactivated by hepatic CYP 2A and 2B to a monoepoxide (VCM) and subsequently to an ovotoxic diepoxide metabolite (VCD). Studies suggest that the ovary can directly bioactivate VCH via CYP 2E1. The current study was designed to evaluate the role of ovarian CYP 2E1 in VCM-induced ovotoxicity. Postnatal day 4 B6C3F(1) and CYP 2E1 wild-type (+/+) and null (-/-) mouse ovaries were cultured (15 days) with VCD (30 microM), 1,2-VCM (125-1000 microM), or vehicle. Twenty-eight days female CYP 2E1 +/+ and -/- mice were dosed daily (15 days; ip) with VCH, 1,2-VCM, VCD or vehicle. Following culture or in vivo dosing, ovaries were histologically evaluated. In culture, VCD decreased (p<0.05) primordial and primary follicles in ovaries from all three groups of mice. 1,2-VCM decreased (p<0.05) primordial follicles in B6C3F(1) and CYP 2E1 +/+ ovaries, but not in CYP 2E1 -/- ovaries in culture. 1,2-VCM did not affect primary follicles in any group of mouse ovaries. Conversely, following in vivo dosing, primordial and primary follicles were reduced (p<0.05) by VCD and VCM in CYP2E1 +/+ and -/-, and by VCH in +/+ mice. The data demonstrate that, whereas in vitro ovarian bioactivation of VCM requires CYP 2E1 enzyme, in vivo CYP 2E1 plays a minimal role. Thus, the findings support that hepatic metabolism dominates the contribution made by the ovary in bioactivation of VCM to its ovotoxic metabolite, VCD. This study also demonstrates the use of a novel ovarian culture system to evaluate ovary-specific metabolism of xenobiotics.
Collapse
Affiliation(s)
- Kathila S. Rajapaksa
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051, USA
| | - Ellen A. Cannady
- Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly & Company, Indianapolis, Indiana, 46285, USA
| | - I. Glenn Sipes
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724-5050, USA
| | - Patricia B. Hoyer
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051, USA
| |
Collapse
|
5
|
Hoyer PB, Sipes IG. Development of an animal model for ovotoxicity using 4-vinylcyclohexene: a case study. ACTA ACUST UNITED AC 2007; 80:113-25. [PMID: 17342769 DOI: 10.1002/bdrb.20103] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND The occupational chemical 4-vinylcyclohexene (VCH) has been shown to cause destruction of small pre-antral follicles in ovaries of mice. Further, its monoepoxide metabolites, 1,2-VCH epoxide, 7,8-VCH epoxide, and the diepoxide, VCD, have been shown to cause pre-antral follicle loss in rats as well as mice. Chemicals that destroy small pre-antral follicles are of concern to women because exposure can result in premature ovarian failure (early menopause). METHODS Studies working with these chemicals over the past decade have determined a number of aspects of the mechanism(s) of small pre-antral destruction, and a variety of questions have been answered. RESULTS Specifically, it has been determined that the diepoxide (VCD) is the bioactive form and it directly targets the ovary in mice and rats. Mice are more susceptible to VCH than rats because they are capable of its metabolic bioactivation. Follicle destruction by VCD is selective for primordial and primary follicles. Mechanistic studies in rats have determined that VCD causes ovotoxicity by accelerating the natural process of atresia (apoptosis) and this requires repeated exposures. Pro-apoptotic signaling events in the Bcl-2 and mitogen activated protein kinase families have been shown to be selectively activated in fractions of small pre-antral follicles (targets for VCD). Finally, a whole ovarian culture system using neonatal mouse and rat ovaries has been developed to expand the potential for more in depth investigations into ovotoxicity caused by VCD. CONCLUSIONS This article provides an overview of the questions asked and the approaches taken in studying VCH and VCD to support these conclusions.
Collapse
Affiliation(s)
- Patricia B Hoyer
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | | |
Collapse
|
6
|
Hu X, Roberts JR, Apopa PL, Kan YW, Ma Q. Accelerated ovarian failure induced by 4-vinyl cyclohexene diepoxide in Nrf2 null mice. Mol Cell Biol 2006; 26:940-54. [PMID: 16428448 PMCID: PMC1347017 DOI: 10.1128/mcb.26.3.940-954.2006] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genetic and biochemical analyses have uncovered an essential role for nuclear factor erythroid 2-related factor 2 (Nrf2) in regulating phase II xenobiotic metabolism and antioxidant response. Here we show that Nrf2 protects against the ovarian toxicity of 4-vinylcyclohexene diepoxide (VCD) in mice. Nrf2-/- female mice exposed to VCD exhibit an age-dependent decline in reproduction leading to secondary infertility accompanied by hypergonadotropic hypogonadism after 30 weeks of age. VCD is shown to selectively destroy small ovarian follicles, resulting in early depletion of functional follicles. Treatment with VCD induces apoptotic death in cultured cells and in ovarian follicles, suggesting apoptosis as a mechanism of follicle loss. Loss of Nrf2 function blocks the basal and inducible expression of microsomal epoxide hydrolase, a key enzyme in the detoxification of VCD, and increases the oxidative stress in cells that is further exacerbated by VCD. Foxo3a, a repressor in the early stages of follicle activation, displays reduced expression in Nrf2-/- ovaries, causing accelerated growth of follicles in the absence of exposure to exogenous chemicals. Furthermore, Foxo3a is degraded through the 26S proteasome pathway in untreated cells and is induced by VCD via both Nrf2-dependent transcription and protein stabilization. This study demonstrates that Nrf2 serves as an essential sensor and regulator of chemical homeostasis in ovarian cells, protecting the cells from toxic chemicals by controlling metabolic detoxification, reactive oxygen species defense, and Foxo3a expression. In addition, these findings raise the possibility that exposure to environmental or occupational ovotoxicants plays a role in the premature ovarian failure commonly associated with infertility and premature aging in women.
Collapse
Affiliation(s)
- Xiaoming Hu
- Receptor Biology Laboratory, TMBB/HELD/NIOSH/CDC, Mailstop 3014, 1095 Willowdale Rd., Morgantown, WV 26505, USA
| | | | | | | | | |
Collapse
|
7
|
Bevan C, Keller DA, Panepinto AS, Bentley KS. Effect of 4-vinylcyclohexene on micronucleus formation in the bone marrow of rats and mice. Drug Chem Toxicol 2001; 24:273-85. [PMID: 11452399 DOI: 10.1081/dct-100103724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This study was conducted to evaluate the potential of 4-vinylcyclohexene (VCH) to induce micronuclei in the bone marrow of mice and rats. Male and female Crl:CD BR (Sprague-Dawley) rats and B6C3F1/CrBR mice were exposed to VCH 6 hr/day for 2 days or for 13 weeks. In the 2-day study, mice were exposed by inhalation to 0, 250, 500, or 1000 ppm, and rats were exposed to 0, 500, 1000, or 2000 ppm. In the 13-week study, mice were exposed to 0, 50, 250, or 1000 ppm, and rats were exposed to 0, 250, 1000, or 1500 ppm. In each study, a separate group of mice was exposed to 1000 ppm 1,3-butadiene (BD) so that a comparison could be made between the two compounds. Likewise, cyclophosphamide was also included for rats as a positive control. Bone marrow was collected from VCH-exposed animals approximately 24 h and 48 h after the final exposure. There were no statistically significant increases in micronucleatedpolychromatic erythrocytes (MN-PCEs) among VCH-treated mice and rats at any dose level or sampling interval at either 2-days or 13-weeks. Also, no statistically significant differences in the polychromatic erythrocytes (PCE) to normochromatic erythrocytes (NCE) ratios were observed in any of the VCH-treated mice and rats compared to air-exposed animals. As expected, both the butadiene-treated mice and the cyclophosphamide-treated rats showed significantly more MN-PCEs than the control animals.
Collapse
Affiliation(s)
- C Bevan
- Exxon Biomedical Sciences, Inc., East Millstone, New Jersey 08875, USA.
| | | | | | | |
Collapse
|
8
|
Hoyer PB, Devine PJ, Hu X, Thompson KE, Sipes IG. Ovarian toxicity of 4-vinylcyclohexene diepoxide: a mechanistic model. Toxicol Pathol 2001; 29:91-9. [PMID: 11215690 DOI: 10.1080/019262301301418892] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Female mammals are born with a finite number of ovarian primordial follicles that cannot be regenerated; thus, chemicals that destroy oocytes contained in these follicles can produce premature ovarian failure (early menopuase in women). Exposure of women to known ovotoxicants, such as contaminants in cigarette smoke, is associated with early menopause. Thus, the potential risks posed by ovotoxic chemicals is of concern. Our studies have focused on the environmental chemical 4-vinylcyclohexene (VCH), which is produced during the manufacture of rubber tires, flame retardants, insecticides, plasticizers, and antioxidants. Dosing of female rats and mice with the ovotoxic diepoxide metabolite of VCH, 4-vinylcyclohexene diepoxide (VCD), for 30 days destroyed the majority of ovarian primordial follicles. Using VCD in rats as a generalized model for ovotoxicity, we determined that 1) repeated daily dosing is required, 2) cell death is via apoptosis, and 3) altered expression of specific genes is involved. An integrated approach at the morphologic, biochemical, and molecular level was used to support these conclusions. Studies in isolated rat small preantral follicles (targeted for VCD-induced ovotoxicity) focused on the role of cell death genes, mitochondrion-associated events, and VCD metabolism. We also evaluated how this information relates to human risk for early menopause. These animal research results provide a better understanding of the potential risk of human exposure to environmental ovarian toxicants and greater insight as to the impact of these toxicants on reproductive health in women.
Collapse
Affiliation(s)
- P B Hoyer
- Department of Physiology, University of Arizona, Tucson 85724, USA.
| | | | | | | | | |
Collapse
|
9
|
Borman SM, VanDePol BJ, Kao S, Thompson KE, Sipes IG, Hoyer PB. A single dose of the ovotoxicant 4-vinylcyclohexene diepoxide is protective in rat primary ovarian follicles. Toxicol Appl Pharmacol 1999; 158:244-52. [PMID: 10438657 DOI: 10.1006/taap.1999.8702] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Repeated dosing of rats with the ovotoxic chemical, 4-vinylcyclohexene diepoxide (VCD), destroys primordial and primary ovarian follicles via apoptosis (physiological cell death) by accelerating the normal rate of atresia. The present study investigated the effect of a single dose (1x) of VCD. Immature (d28) female Fischer 344 rats were dosed 1x or 15x with VCD (80 mg/kg ip). Ovaries were collected 24 h or 15 days following 1x VCD or after 15x for classification and evaluation. Following 1x VCD the number of healthy primary follicles was greater (p < 0.05) than control 24 h and 15 days later. This effect reflected a slowing of the normal rate of atresia seen in control ovaries. There was no effect of a single dose on primordial or growing follicles at any time. Expression of mRNA encoding the cell death gene bax was reduced (p < 0.05) on d1 after 1x VCD in isolated primordial and primary follicles. These observations were in contrast to a decreased (p < 0. 05) number of healthy primary and primordial follicles in ovaries and increased (p < 0.05) bax mRNA in isolated follicles from rats dosed 15x for 15 days. Immunofluorescence staining revealed that, the distribution of Bax protein was similar between ovaries from controls and 1x or 15x VCD-treated rats. These data provide evidence for a "protective" response against the normal rate of atresia in primary ovarian follicles following exposure to 1x VCD. Additionally, changes in expression of bax mRNA paralleled alterations in the rate of atresia.
Collapse
Affiliation(s)
- S M Borman
- Department of Physiology, University of Arizona, Tucson, Arizona, 85724, USA
| | | | | | | | | | | |
Collapse
|
10
|
Keller DA, Carpenter SC, Cagen SZ, Reitman FA. In vitro metabolism of 4-vinylcyclohexene in rat and mouse liver, lung, and ovary. Toxicol Appl Pharmacol 1997; 144:36-44. [PMID: 9169067 DOI: 10.1006/taap.1996.8098] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
4-Vinylcyclohexene (4-VCH), the dimer of 1,3-butadiene, is an ovarian toxicant in mice due to the formation of a diepoxide metabolite, but the tissue-specific site of formation of the metabolites is unknown. Microsomal preparations from liver, lung, and ovaries obtained from female Crl:CD BR rats and female B6C3F1 mice were tested for their ability to metabolize the following reactions: 4-VCH to 4-VCH-1,2-epoxide and 4-VCH-7,8-epoxide; 4-VCH-1,2-epoxide to 4-VCH diepoxide and 4-VCH-1,2-diol; 4-VCH-7,8-epoxide to 4-VCH diepoxide and 4-VCH-7,8-diol; and hydrolysis of 4-VCH diepoxide. Microsomes were incubated with the test chemical and the reaction products were analyzed by gas chromatography. Rat liver and lung microsomes and mouse liver and lung microsomes metabolized 4-VCH to 4-VCH-1,2-epoxide at detectable rates. Mouse liver had a Vmax for the reaction that was 56-fold higher than that for rat liver (11.1 and 0.20 nmol/min/mg protein, respectively). The Vmax for mouse lung was 2-fold higher than that for rat lung. 4-VCH-1,2-epoxide formation was not detected in ovarian microsomes from rats or mice. Metabolism of 4-VCH to 4-VCH-7,8-epoxide was detected in microsomes from rat liver and mouse liver and lung, at rates very low compared to those for metabolism to the 1,2-epoxide. Rat and mouse liver had very similar K(m) and Vmax values for metabolism of 4-VCH-1,2-epoxide to 4-VCH diepoxide. The Vmax for rat liver was 3.69 and for mouse liver was 5.35 nmol/min/mg protein. Rat and mouse ovaries did not have detectable capacity to metabolize 4-VCH-1,2-epoxide to the diepoxide. Rat and mouse liver and lung have very similar K(m) and Vmax values for metabolism of 4-VCH-7,8-epoxide to the diepoxide, while ovaries did not have detectable rates for this reaction. Hydrolysis of 4-VCH-1,2-epoxide to 4-VCH-1,2-diol was at similar rates in rat and mouse liver microsomes. Hydrolysis of 4-VCH-7,8-epoxide to 4-VCH-7,8-diol was detected only in rat liver microsomes. Hydrolysis of 4-VCH diepoxide was detected in rat and mouse liver and lung, and in rat ovary microsomes. The Vmax for rat liver was 9-fold greater than that for mouse liver (5.51 and 0.63 nmol/min/mg protein, respectively), and lung and ovary tissues were not as active as rat liver. The balance of activation versus detoxication reactions in rats and mice suggests that the mouse may be more susceptible to 4-VCH toxicity because of generation of high levels of epoxide metabolites. In general, the mouse is more efficient at metabolism of 4-VCH to epoxides than is the rat. In contrast, the rat may be more efficient at hydrolysis of epoxides. Thus, the rat would tend to produce a lower concentration of epoxide metabolites than the mouse, at equal doses of 4-VCH.
Collapse
Affiliation(s)
- D A Keller
- E.I. du Pont de Nemours and Company, Haskell Laboratory for Toxicology and Industrial Medicine, Newark, Delaware 19714, USA
| | | | | | | |
Collapse
|
11
|
Cantoreggi S, Keller DA. Pharmacokinetics and metabolism of vinyl fluoride in vivo and in vitro. Toxicol Appl Pharmacol 1997; 143:130-9. [PMID: 9073601 DOI: 10.1006/taap.1996.8041] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Vinyl fluoride (VF) is an inhalation carcinogen at concentrations of 25 ppm or greater in rats and mice. The main neoplastic lesion induced in rodents was hepatic hemangiosarcomas, and mice were more sensitive than rats. In a first set of experiments, groups of three rats or five mice were exposed to VF in a closed-chamber gas uptake system at starting concentrations ranging from 50 to 250 ppm. Chamber concentrations of VF were measured every 10-12 min by gas chromatography. Partition coefficients were determined by the vial equilibration technique and used as parameters for a physiologically based pharmacokinetic (PBPK) model. Mice showed a higher whole-body metabolic capacity compared to rats (Vmax = 0.3 vs 0.1 mg/hr-kg). Both species had an estimated Km of < or = 0.02 mg/liter. The specificity for the oxidation of VF in vivo was determined by selective inhibition or induction of CYP 2E1. Inhibition with 4-methylpyrazole completely impaired VF uptake in rats and mice, whereas induction with ethanol (rats only) increased the metabolic capacity by two- to threefold. The pharmacokinetics of VF were also investigated in vitro. Microsomes from rat and mouse liver were incubated in a sealed vial with VF and an NADPH-regenerating system. Headspace concentrations (10-300 ppm) were monitored over time by gas chromatography. Consistent with the in vivo data, VF was metabolized faster by mouse microsomes than by rat microsomes (Vmax = 3.5 and 1.1 nmol/hr-mg protein, respectively). The rates of metabolism by human liver microsomes were generally in the same range as those found with rat liver microsomes (Vmax = 0.5-1.3 nmol/hr-mg protein), but one sample was similar to mice (Vmax = 3.3 nmol/ hr-mg protein). Metabolic rates in human microsomes were found to correlate with the amount of CYP 2E1 as determined by Western blotting and by chlorzoxazone 6-hydroxylation. It is concluded that the greater metabolic capacity of mice for VF both in vivo and in vitro may contribute to their greater susceptibility to tumor formation. CYP 2E1 is clearly the main isozyme involved in the oxidation of VF in all species tested. VF pharmacokinetics and metabolism in humans may depend upon the interindividual variability in the expression level of CYP 2E1. The excellent correspondence between in vivo and in vitro kinetics in rodents improves. substantially the degree of confidence for human in vivo predictions from in vitro data.
Collapse
Affiliation(s)
- S Cantoreggi
- Haskell Laboratory for Toxicology and Industrial Medicine, E.I. du Pont de Nemours and Company, Newark, Delaware 19714, USA
| | | |
Collapse
|
12
|
Doerr JK, Hollis EA, Sipes IG. Species difference in the ovarian toxicity of 1,3-butadiene epoxides in B6C3F1 mice and Sprague-Dawley rats. Toxicology 1996; 113:128-36. [PMID: 8901892 DOI: 10.1016/0300-483x(96)03437-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
1,3-Butadiene (BD) is an ovarian carcinogen in mice, but not in rats. Since species variation in metabolism of BD has been associated with the greater sensitivity of mice to BD-induced carcinogenicity, the extent of biotransformation and detoxification of BD and its epoxides may play a critical role in other ovotoxic effects of this compound. Thus, the ovotoxic potency of the BD epoxides was determined. Butadiene monoepoxide (BMO, 0.005-1.43 mmol/kg b.w.), butadiene diepoxide (BDE, 0.002-0.29 mmol/kg b.w.), or vehicle was administered i.p. to female B6C3F1 mice and Sprague-Dawley rats for 30 days. Following day 30, tissues were removed and weighed, and the number of pre-antral ovarian follicles counted. BMO was ovotoxic in mice as demonstrated by decreases in reproductive organ weights (1.43 mmol/kg b.w.) and follicular counts, but not in rats at any dose tested. In mice the ED50 values for BMO for small and growing follicles were 0.29 and 0.40 mmol/kg b.w., respectively. BDE was ovotoxic in both species, however mice were more sensitive to BDE than rats. Ovarian and uterine weights were decreased in mice at 0.14 and 0.29 mmol/kg b.w. of BDE treatment, and in rats at 0.29 mmol/kg b.w. only. The ED50 values for BDE in mice were 0.10 and 0.14 mmol/kg b.w. for small and growing follicles, respectively, ED50 values could not be determined in rats since only 32% of the follicular population was depleted at the highest concentration tested. Thus, BMO and BDE exhibited a greater ovotoxic potential in mice, as compared to rats. In addition, in each species the diepoxide was the most potent ovotoxicant.
Collapse
Affiliation(s)
- J K Doerr
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson 85721, USA
| | | | | |
Collapse
|
13
|
Abstract
Toxic doses of 1,3-butadiene (BD) have been reported to cause reproductive and/or developmental toxicity. Regardless of the strain used, mice were always affected by BD at lower doses than rats, an expected observation, based on well recognized differences in pharmacokinetic (PK) parameters in these two species. Because the mouse is particularly sensitive to BD in comparison with other laboratory species, and there are important functional and anatomical differences between humans and mice, the NOELs and LOELs identified for BD for various reproductive endpoints in mice may not be relevant to human reproductive risk. In mice, the LOELs for reproductive endpoints include developmental toxicity at 200 ppm, genotoxic effects at 500 ppm (mouse spot test), ovarian atrophy in females at 6.25 ppm (carcinogenicity study), reduced testicular weights at 200 ppm and testicular atrophy at 625 ppm BD in males (carcinogenicity studies), low incidences of abnormal sperm heads at 1000 and 5000 ppm BD (sperm head morphology study), small reversible increases in resorption at 1250/1300 ppm or 5000 ppm (dominant lethal studies), and other possible sequelae of genotoxicity resulting from exposure of male mice at 12.5 ppm BD and higher (dominant lethal study). When available, the much higher NOELs and LOELs of other species tested for the same endpoints should be considered. For example, maternal and developmental NOELs for BD in the rat were 200 and 1000 ppm, respectively, and 40 ppm in the mouse. Likewise, exposure of cohabited pairs of rats, guinea pigs and rabbits or of female dogs to BD concentrations as high as 6700 ppm for 8 months did not impair fertility or cause testicular or ovarian atrophy in these species. Thus, consideration of these remarkable species-dependent differences in toxicity is necessary. In addition, there are alternative scientific interpretations for some of the mouse studies and this review attempts to address these areas. For example, it may be incorrect to categorize results indicating weak in vivo genotoxic effects in male mice (sperm head morphology and dominant lethal studies) at 12.5 ppm BD and higher as reproductive effects because concentrations of BD as high as 5000 ppm did not affect mating, fertility or live litter sizes, even in this sensitive species. Similarly, it may be inappropriate to identify the ovary as a target organ for reproductive risk since the ovarian atrophy in mice was identified after completion of the normal reproductive life and after more than 15 months of exposure. Neither ovarian nor testicular atrophy occurred in Sprague-Dawley rats after exposure to BD concentrations as high as 8000 ppm for 105 (females) or 111 (males) weeks.
Collapse
|
14
|
Springer LN, Flaws JA, Sipes IG, Hoyer PB. Follicular mechanisms associated with 4-vinylcyclohexene diepoxide-induced ovotoxicity in rats. Reprod Toxicol 1996; 10:137-43. [PMID: 8919611 DOI: 10.1016/0890-6238(95)02056-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The mechanism of 4-vinylcyclohexene diepoxide (VCD)-induced oocyte destruction in small preantral follicles of rats and mice has not been elucidated. This study examined the effects of daily dosing of female rats with VCD on protein synthesis and follicle viability. An investigation of granulosa cells as a target for VCD was also made. Small preantral follicles (25 to 100 microns) isolated from untreated immature rats (day 28) as well as from rats injected daily for 10 d with VCD (0.57 mmol/kg, IP) or vehicle control (sesame oil) were incubated for 3, 6, or 10 h in vitro with or without VCD. Viability (trypan blue dye exclusion) or protein synthesis (3H-leucine incorporation) in follicles was measured. Large preantral follicles (100 to 250 microns), isolated oocytes or granulosa cells from small preantral follicles, hepatocytes, and adrenal cells served as controls. Viability was not compromised in small follicles isolated from untreated or VCD-injected rats. However, following in vitro incubation of small preantral follicles with VCD, there was a significant decrease in viability by 6 h. This loss in viability was observed in granulosa cells and was even greater in follicles from dosed as compared with undosed animals. The various cell types were incubated in vitro with or without VCD for 3 h and the rate of protein synthesis was measured by 3H-leucine incorporation during the last hour of incubation. Incubation of small preantral follicles from untreated animals with VCD for 3 h produced significant inhibition in the rate of protein synthesis. This effect was reversed and significantly stimulated after 6 and 10 h of incubation with VCD. Follicles from animals that had been dosed daily with VCD for 10 d demonstrated similar inhibition of protein synthesis following 3 h in vitro incubation with VCD; however, unlike those from undosed rats, follicles from dosed rats did not recover from this inhibition after 6 or 10 h of in vitro incubation with VCD. In vitro incubation with VCD stimulated the rate of protein synthesis in large preantral follicles; however, no effect on the rate of protein synthesis was observed in isolated oocytes and granulosa cells, hepatocytes, or adrenal cells. These observations suggest that VCD affects follicular viability via an effect on granulosa cells and that daily dosing of rats with VCD makes small preantral follicles more susceptible to ovotoxicity by VCD.
Collapse
Affiliation(s)
- L N Springer
- Department of Physiology, University of Arizona, Tucson, USA
| | | | | | | |
Collapse
|
15
|
Doerr JK, Sipes IG. Ovarian toxicity and metabolism of 4-vinylcyclohexene and analogues in B6C3F1 mice: structure-activity study of 4-vinylcyclohexene and analogues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1996; 387:377-84. [PMID: 8794232 DOI: 10.1007/978-1-4757-9480-9_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- J K Doerr
- University of Arizona, Department of Pharmacology and Toxicology, College of Pharmacy, Tucson 85721, USA
| | | |
Collapse
|
16
|
Hooser SB, DeMerell DG, Douds DA, Hoyer P, Sipes IG. Testicular germ cell toxicity caused by vinylcyclohexene diepoxide in mice. Reprod Toxicol 1995; 9:359-67. [PMID: 7496092 DOI: 10.1016/0890-6238(95)00022-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Vinylcyclohexene diepoxide (VCD) produces ovarian toxicity in female mice and rats, whereas testicular damage occurs only in mice. The objectives of these studies were to determine the target cell(s) and spermatogonial survival following VCD administration. In addition, the effects of 4-vinylcyclohexene (VCH) and two epoxide metabolites, vinylcyclohexene 1,2-monoepoxide and VCD were compared. Male mice were dosed daily with VCD (320 mg/kg/d, i.p.) and killed at 5, 10, 15, 20, 25, or 30 d. Two groups were dosed daily for 30 d and allowed to recover for 30 or 60 d. Decreases in testis weight began at 5 d and continued to 30 d. These decreases corresponded to progressive necrosis of germ cells. After 5 d of VCD, there was loss of Type I and B spermatogonia in Stages II to VI and of preleptotene spermatocytes in Stages VI to VIII. After 30 d of dosing, seminiferous tubules were devoid of germ cells except for spermatogonial stem cells. Following 30 d of recovery, 100% of the seminiferous tubules were repopulated. Epididymal spermatozoa were present after 60 d of recovery. Increasing doses of VCD (0 to 320 mg/kg/d) resulted in increasing testicular toxicity. Neither VCH (800 mg/kg, i.p.) nor VCM (200 mg/kg, i.p.) caused testicular damage. VCD administration initially results in destruction of spermatogonia and spermatocytes, which are undergoing DNA synthesis and cell replication, followed by loss of maturing cells. Neither VCH nor VCM caused testicular germ cell destruction, although all three compounds destroy germ cells in female mice. Therefore, further investigation will be necessary to understand these differences in chemical-induced toxicity between ovaries and testes.
Collapse
Affiliation(s)
- S B Hooser
- Department of Pharmacology/Toxicology, University of Arizona, Tucson 85721, USA
| | | | | | | | | |
Collapse
|
17
|
Flaws JA, Doerr JK, Sipes IG, Hoyer PB. Destruction of preantral follicles in adult rats by 4-vinyl-1-cyclohexene diepoxide. Reprod Toxicol 1994; 8:509-14. [PMID: 7881202 DOI: 10.1016/0890-6238(94)90033-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
4-vinyl-1-cyclohexene diepoxide (VCD) is known to destroy oocytes in ovaries of immature rats. Since ovaries functionally differ between immature and adult animals, we examined the effect of VCD on oocytes in adult rats. Adult (58 days) and immature (28 days) rats were injected daily (30 days) with vehicle or VCD. Each group contained 10 rats. During this time, cyclicity was determined daily by vaginal cytology. Animals were terminated on day 31 and tissues were collected. Oocytes were counted; livers, spleens, and uteri were weighed. VCD reduced the number of regular estrous cycles/30 days in adults, but not immature rats (n = 20, P < 0.05). VCD reduced the number of oocytes in adult and immature rats (n = 20, P < 0.05). Liver, spleen, or ovarian weights were not affected by VCD in either group. VCD reduced uterine weight in adult (n = 20, P < 0.05) but not in immature rats. These results demonstrate that VCD decreases uterine weight in adult rats and as with immature rats, selectively destroys oocytes in ovaries of adults.
Collapse
Affiliation(s)
- J A Flaws
- Department of Physiology, University of Arizona, Tucson 85724
| | | | | | | |
Collapse
|
18
|
Hooser SB, Douds DP, DeMerell DG, Hoyer PB, Sipes IG. Long-term ovarian and gonadotropin changes in mice exposed to 4-vinylcyclohexene. Reprod Toxicol 1994; 8:315-23. [PMID: 7949757 DOI: 10.1016/0890-6238(94)90047-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This study investigated the relationships between 4-vinylcyclohexene-induced follicular destruction, plasma FSH levels, and the development of ovarian preneoplastic changes. Female, 28-day-old, B6C3F1 mice were administered VCH (800 mg/kg/day, ip) or sesame seed oil, ip daily for 30 days. At 30, 60, 120, 240, and 360 days following the beginning of treatment, groups were killed, their ovaries were harvested, and plasma was collected for measurement of FSH. Ovarian weight was less and oocytes contained in preantral follicles were significantly fewer than controls at all time points. Plasma FSH concentrations in VCH-treated animals were increased significantly above controls at 240 d and 360 d. Histologically, there was oocyte loss at all times, whereas at 240 and 360 days, small to medium, irregularly shaped foci of hypertrophic cells were present. In addition, at 360 days 80% of the VCH-treated mice had a 1- to 2-mm, blood-filled cystic structure present in one or both ovaries. These studies indicate that VCH-induced oocyte destruction and follicle loss are associated with increases in plasma FSH, are associated with ovarian failure at 360 days, and are temporally related to ovarian cellular hypertrophy and the formation of blood-filled cystic ovarian structures. These events are possibly related to ovarian neoplasms produced by long-term exposure to VCH.
Collapse
Affiliation(s)
- S B Hooser
- Department of Pharmacology and Toxicology, University of Arizona, College of Pharmacy, Tucson 85721
| | | | | | | | | |
Collapse
|
19
|
Smith BJ, Mattison DR, Sipes IG. Hepatic bioactivation of 4-vinylcyclohexene to ovotoxic epoxides. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1991; 283:465-70. [PMID: 2069018 DOI: 10.1007/978-1-4684-5877-0_62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- B J Smith
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson 85721
| | | | | |
Collapse
|
20
|
Smith BJ, Mattison DR, Sipes IG. The role of epoxidation in 4-vinylcyclohexene-induced ovarian toxicity. Toxicol Appl Pharmacol 1990; 105:372-81. [PMID: 2237912 DOI: 10.1016/0041-008x(90)90141-g] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
4-Vinylcyclohexene (VCH) is present in gases discharged during synthetic rubber production. Chronic treatment of B6C3F1 mice and F-344 rats with VCH by gavage has been shown to induce ovarian tumors in mice but not in rats. Our objective was to understand the mechanism of the species difference in VCH-induced ovarian tumors. Since a critical step in the induction of ovarian tumors is destruction of the small oocyte, small oocyte counts obtained from serially sectioned ovaries were used as an index of toxicity. VCH or its epoxide metabolites [VCH-diepoxide, VCH-1,2-epoxide, and VCH-7,8-epoxide (in mice only)] were given to 28-day-old female mice and rats in corn oil, ip, at doses ranging from 0.07 to 7.4 mmol/kg body wt/day for 30 days. The dose which reduced the small oocyte count to 50% that of control was defined as the ED50. In mice, the ED50 for the reduction in small oocytes by VCH was 2.7 mmol/kg, whereas, no detectable oocyte loss occurred in rats at the highest dose of VCH (7.4 mmol/kg). The potency of the epoxides of VCH was greater than that of VCH in both species. The ED50 for oocyte loss by VCH-1,2-epoxide in mice and rats was 0.5 and 1.4 mmol/kg, respectively. In mice, VCH-7,8-epoxide had comparable potency to VCH-1,2-epoxide (ED50 = 0.7). VCH diepoxide was even more potent with ED50 values of 0.2 and 0.4 mmol/kg, in mice and rats, respectively. The dose response of the blood concentration of VCH-1,2-epoxide in mice after VCH showed that doses of VCH which caused minimal toxicity had the lowest blood level of this ovotoxic epoxide. Pretreatment of mice with the cytochrome P450 inhibitor chloramphenicol (200 mg/kg, ip) inhibited VCH epoxidation in vivo and in vitro and partially protected mice from VCH toxicity. Thus it appears that metabolism of VCH to epoxides and their subsequent destruction of oocytes are critical steps in VCH-induced ovarian tumors. Rats may be resistant to ovarian tumor induction by VCH because the amount of VCH converted to epoxides is insufficient to produce oocyte destruction.
Collapse
Affiliation(s)
- B J Smith
- Department of Pharmacology and Toxicology, University of Arizona, Tucson 85721
| | | | | |
Collapse
|