101
|
Ma T, Yin X, Han R, Ding J, Zhang H, Han X, Li D. Effects of In Utero Exposure to Di-n-Butyl Phthalate on Testicular Development in Rat. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14101284. [PMID: 29064414 PMCID: PMC5664784 DOI: 10.3390/ijerph14101284] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 11/16/2022]
Abstract
Humans are inevitably exposed to ubiquitous phthalate esters (PAEs). In utero exposure to di-n-butyl phthalate (DBP) induces abnormal development of the testis and reproductive tract in male offspring, which correspond closely with the human condition of testicular dysgenesis syndrome (TDS)-like syndrome. However, the underlying mechanisms have not been elucidated in detail. In this study, pregnant rats were orally exposed to either corn oil (controls) or DBP at three different doses by gavage during Gestational Days 12.5-21.5. Pathological examinations were performed for toxicity evaluation. Proliferation and apoptosis related proteins (ras related dexamethasone induced 1 (Rasd1), mitogen-activated protein kinase kinases1/2 (MEK1/2), Bcl-2, and Bax) were measured for mechanisms exploration. The results showed that different doses of DBP caused male developmental and reproductive toxicity in rats, including the decrease of anogenital distance (AGD), the histological damage of testis, and apoptosis of seminiferous tubule cells. Our data suggested that DBP played chronic and continuous toxic roles on male reproductive system by disrupting expression of Rasd1 and MEK1/2 as well as Bcl-2/Bax ratio. Further research is warranted.
Collapse
Affiliation(s)
- Tan Ma
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Xiaoqin Yin
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Ruitong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Jie Ding
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Huan Zhang
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden.
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
102
|
Benjamin S, Masai E, Kamimura N, Takahashi K, Anderson RC, Faisal PA. Phthalates impact human health: Epidemiological evidences and plausible mechanism of action. JOURNAL OF HAZARDOUS MATERIALS 2017; 340:360-383. [PMID: 28800814 DOI: 10.1016/j.jhazmat.2017.06.036] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/08/2017] [Accepted: 06/17/2017] [Indexed: 05/02/2023]
Abstract
Disregarding the rising alarm on the hazardous nature of various phthalates and their metabolites, ruthless usage of phthalates as plasticizer in plastics and as additives in innumerable consumer products continues due low their cost, attractive properties, and lack of suitable alternatives. Globally, in silico computational, in vitro mechanistic, in vivo preclinical and limited clinical or epidemiological human studies showed that over a dozen phthalates and their metabolites ingested passively by man from the general environment, foods, drinks, breathing air, and routine household products cause various dysfunctions. Thus, this review addresses the health hazards posed by phthalates on children and adolescents, epigenetic modulation, reproductive toxicity in women and men; insulin resistance and type II diabetes; overweight and obesity, skeletal anomalies, allergy and asthma, cancer, etc., coupled with the description of major phthalates and their general uses, phthalate exposure routes, biomonitoring and risk assessment, special account on endocrine disruption; and finally, a plausible molecular cross-talk with a unique mechanism of action. This clinically focused comprehensive review on the hazards of phthalates would benefit the general population, academia, scientists, clinicians, environmentalists, and law or policy makers to decide upon whether usage of phthalates to be continued swiftly without sufficient deceleration or regulated by law or to be phased out from earth forever.
Collapse
Affiliation(s)
- Sailas Benjamin
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940 2188, Japan; Enzyme Technology Laboratory, School of Biosciences, University of Calicut, Kerala 673 635, India.
| | - Eiji Masai
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940 2188, Japan
| | - Naofumi Kamimura
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940 2188, Japan
| | - Kenji Takahashi
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata 940 2188, Japan
| | - Robin C Anderson
- USDA Southern Plains Agricultural Research Center, Food and Feed Safety Research Unit, 2881 F&B Road, College Station, TX 77845, USA
| | - Panichikkal Abdul Faisal
- Enzyme Technology Laboratory, School of Biosciences, University of Calicut, Kerala 673 635, India
| |
Collapse
|
103
|
KOLATOROVA L, DUSKOVA M, VITKU J, STARKA L. Prenatal Exposure to Bisphenols and Parabens and Impacts on Human Physiology. Physiol Res 2017; 66:S305-S315. [DOI: 10.33549/physiolres.933723] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In modern societies, living organisms are exposed daily to multiform pollution from industrial chemical products. Some of these substances have been shown to affect the endocrine system, and have been termed endocrine disruptors (EDs). Bisphenol A (BPA), which can leach from plastics, and parabens, used in cosmetic products, are among the most well-studied. Prenatal development is a vulnerable phase of human life, and disruptions during this period may have lifelong consequences. Since EDs are known to cross the placental barrier and BPA may accumulate in the fetus, “BPA-free” products have been introduced to the market. However, such products often contain alternative bisphenols (e.g. BPS, BPF) that have not yet been extensively examined or regulated. Moreover, alternative bisphenols often occur together with BPA. The human organism is thus exposed to a mixture of EDs, some of which can have additive or synergic effects. Recent findings have also shown that paraben exposure can alter bisphenol pharmacokinetics. Taking into account the widespread occurrence of various EDs and the potential multiplicity of their effects, doses of EDs currently considered safe may not actually be as safe as they appear, especially during pregnancy.
Collapse
Affiliation(s)
- L. KOLATOROVA
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czech Republic
| | | | | | | |
Collapse
|
104
|
Karačonji IB, Jurica SA, Lasić D, Jurica K. Facts about phthalate toxicity in humans and their occurrence in alcoholic beverages. Arh Hig Rada Toksikol 2017; 68:81-92. [DOI: 10.1515/aiht-2017-68-2951] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/01/2017] [Indexed: 11/15/2022] Open
Abstract
Abstract
Phthalates are esters of phthalic acid and aliphatic alcohol added to plastic to improve its softness, flexibility, and extensibility. They easily migrate from plastic products into the environment because of their physical and chemical properties. This review summarises their characteristics, distribution in the environment, monitoring, use, toxic effects on human health, regulatory limits in different matrices and products, and tolerable daily intake. The studies we have reviewed suggest that phthalates have a potential to affect reproduction and development in humans. Due to the inconsistent data, further studies are needed and, in the meantime, precautionary policies must be implemented. Here we draw attention to the methods of determining phthalate levels in alcoholic beverages and reported levels in plum spirits produced in Croatia. Legally produced and moderately consumed plum spirits do not seem to increase the risk of phthalate toxicity for human health. We conclude with recommendations for the effective monitoring of phthalate exposure in humans and for the implementation of alternative materials in alcohol production.
Collapse
Affiliation(s)
- Irena Brčić Karačonji
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, Zagreb , Croatia
| | - Sonja Anić Jurica
- Department of Gynaecology and Obstetrics, University Hospital Centre Zagreb, Zagreb , Croatia
| | - Dario Lasić
- Department of Environmental Protection and Health Ecology, Andrija Štampar Teaching Institute of Public Health, Zagreb , Croatia
| | - Karlo Jurica
- PhD, Ministry of the Interior, Ulica grada Vukovara 33, 10000 Zagreb , Croatia
| |
Collapse
|
105
|
Dibutyl phthalate induced testicular dysgenesis originates after seminiferous cord formation in rats. Sci Rep 2017; 7:2521. [PMID: 28566680 PMCID: PMC5451485 DOI: 10.1038/s41598-017-02684-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/18/2017] [Indexed: 11/08/2022] Open
Abstract
Administration of dibutyl phthalate (DBP) to pregnant rats causes reproductive disorders in male offspring, resulting from suppression of intratesticular testosterone, and is used as a model for human testicular dysgenesis syndrome (TDS). DBP exposure in pregnancy induces focal dysgenetic areas in fetal testes that appear between e19.5–e21.5, manifesting as focal aggregation of Leydig cells and ectopic Sertoli cells (SC). Our aim was to identify the origins of the ectopic SC. Time-mated female rats were administered 750 mg/kg/day DBP in three different time windows: full window (FW; e13.5–e20.5), masculinisation programming window (MPW; e15.5–e18.5), late window (LW; e19.5–e20.5). We show that DBP-MPW treatment produces more extensive and severe dysgenetic areas, with more ectopic SC and germ cells (GC) than DBP-FW treatment; DBP-LW induces no dysgenesis. Our findings demonstrate that ectopic SC do not differentiate de novo, but result from rupture of normally formed seminiferous cords beyond e20.5. The more severe testis dysgenesis in DBP-MPW animals may result from the presence of basally migrating GC and a weakened basal lamina, whereas GC migration was minimal in DBP-FW animals. Our findings provide the first evidence for how testicular dysgenesis can result after normal testis differentiation/development and may be relevant to understanding TDS in human patients.
Collapse
|
106
|
Yang Y, Zhang Z, Zhang H, Hong K, Tang W, Zhao L, Lin H, Liu D, Mao J, Wu H, Jiang H. Effects of maternal acrolein exposure during pregnancy on testicular testosterone production in fetal rats. Mol Med Rep 2017; 16:491-498. [PMID: 28560422 PMCID: PMC5482093 DOI: 10.3892/mmr.2017.6624] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Acrolein has been reported to have diverse toxic effects on various organs, including the reproductive system. However, little is known regarding the effects of maternal acrolein exposure on testicular steroidogenesis in male offspring. The present study investigated the effects of acrolein on fetal testosterone production and associated genes. Pregnant Sprague-Dawley rats were intraperitoneally injected with vehicle (normal saline) or 1, 2 or 5 mg/kg acrolein from gestational day (GD) 14–20, and fetal testes were examined on GD 21. Fetal body and testicular weights were markedly reduced in pups following exposure to high doses of acrolein (5 mg/kg) in late pregnancy. Notably, in utero exposure of 5 mg/kg acrolein significantly decreased the testicular testosterone level and downregulated the expression levels of steroidogenic acute regulatory protein (StAR) and 3β-hydroxysteroid dehydrogenase (3β-HSD), whereas the levels of other steroidogenic enzymes, including scavenger receptor class B, cholesterol side-chain cleavage enzyme and steroid 17 alpha-hydroxylase/17,20 lyase, were unaffected. Furthermore, the 3β-HSD immunoreactive area in the interstitial region of the fetal testes was reduced at a 5 mg/kg dose, whereas the protein expression levels of 4-hydroxynonenalwere dose-dependently increased following maternal exposure to acrolein. mRNA expression levels of insulin-like factor 3, a critical gene involved in testicular descent, were unaltered following maternal acrolein exposure. Taken together, the results of the present study suggested that maternal exposure to high doses of acrolein inhibited fetal testosterone synthesis, and abnormal expression of StAR and 3β-HSD may be associated with impairment of the steroidogenic capacity.
Collapse
Affiliation(s)
- Yuzhuo Yang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hongliang Zhang
- Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Kai Hong
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Wenhao Tang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Lianming Zhao
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Defeng Liu
- Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Jiaming Mao
- Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Han Wu
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
107
|
Wong KH, Durrani TS. Exposures to Endocrine Disrupting Chemicals in Consumer Products-A Guide for Pediatricians. Curr Probl Pediatr Adolesc Health Care 2017; 47:107-118. [PMID: 28526231 DOI: 10.1016/j.cppeds.2017.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endocrine disrupting chemicals, a group of exogenous chemicals that can interfere with hormone action in the body, have been implicated in disrupting endocrine function, which negatively affects human health and development. Endocrine disrupting chemicals are ubiquitously detected in consumer products, foods, beverages, personal care products, and household cleaning products. Due to concerns about their negative effects on human health, several professional health provider societies have recommended the reduction of common endocrine disrupting chemical exposures. The purpose of this review is to provide a brief overview of common endocrine disrupting chemicals (bisphenol A, phthalates, triclosan, polybrominated ethers, and parabens) and potential effects on child development and health. In addition, we aim to provide guidance and resources for pediatricians and other health care providers with counseling strategies to help patients to minimize exposures to common endocrine disrupting chemicals.
Collapse
Affiliation(s)
- Katelyn H Wong
- University of Connecticut School of Medicine, Farmington, CT
| | - Timur S Durrani
- Division of Occupational and Environmental Medicine, University of California San Francisco, San Francisco, CA; Western States Pediatric Environmental Health Specialty Unit, San Francisco, CA
| |
Collapse
|
108
|
Thistle JE, Graubard BI, Braunlin M, Vesper H, Trabert B, Cook MB, McGlynn KA. Marijuana use and serum testosterone concentrations among U.S. males. Andrology 2017; 5:732-738. [PMID: 28395129 DOI: 10.1111/andr.12358] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/24/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023]
Abstract
Marijuana has been reported to have several effects on the male reproductive system. Marijuana has previously been linked to reduced adult testosterone, however, a study in Denmark reported increased testosterone concentrations among marijuana users. This study was performed to estimate the effect of marijuana use on testosterone in U.S. males. Data on serum testosterone, marijuana use, and covariates for 1577 men from the 2011-2012 U.S. National Health and Nutrition Examination Survey (NHANES) were analyzed. Information on marijuana use was collected by a self-administered computer-assisted questionnaire. Serum testosterone was determined using isotope dilution liquid chromatography tandem mass spectrometry. The effects of marijuana use on serum testosterone concentrations were examined by frequency, duration, and recency of use. Adjusted means and 95% confidence intervals (CI) of serum testosterone across levels of marijuana use were estimated using multiple linear regression weighted by the survey weights. The majority (66.2%) of the weighted study population reported ever using marijuana with 26.6% reporting current marijuana use. There was no difference in serum testosterone between ever users (adjusted mean = 3.69 ng/mL, 95% CI: 3.46, 3.93) and never users (adjusted mean = 3.70 ng/mL, 95% CI: 3.45, 3.98) upon multivariable analysis. However, serum testosterone was inversely associated with time since last regular use of marijuana (p-value for trend = 0.02). When restricted to men aged 18-29 years, this relationship strengthened (p-value for trend <0.01), and serum testosterone was also inversely associated with time since last use (p-value for trend <0.01), indicating that recency of use, and not duration or frequency, had the strongest relationship with testosterone levels. Serum testosterone concentrations were higher in men with more recent marijuana use. Studies are needed to determine the extent to which circulating testosterone concentrations mediate the relationship of marijuana use with male reproductive outcomes.
Collapse
Affiliation(s)
- J E Thistle
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - B I Graubard
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - M Braunlin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - H Vesper
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - B Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - M B Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - K A McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
109
|
van den Driesche S, Kilcoyne KR, Wagner I, Rebourcet D, Boyle A, Mitchell R, McKinnell C, Macpherson S, Donat R, Shukla CJ, Jorgensen A, Meyts ERD, Skakkebaek NE, Sharpe RM. Experimentally induced testicular dysgenesis syndrome originates in the masculinization programming window. JCI Insight 2017; 2:e91204. [PMID: 28352662 DOI: 10.1172/jci.insight.91204] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The testicular dysgenesis syndrome (TDS) hypothesis, which proposes that common reproductive disorders of newborn and adult human males may have a common fetal origin, is largely untested. We tested this hypothesis using a rat model involving gestational exposure to dibutyl phthalate (DBP), which suppresses testosterone production by the fetal testis. We evaluated if induction of TDS via testosterone suppression is restricted to the "masculinization programming window" (MPW), as indicated by reduction in anogenital distance (AGD). We show that DBP suppresses fetal testosterone equally during and after the MPW, but only DBP exposure in the MPW causes reduced AGD, focal testicular dysgenesis, and TDS disorders (cryptorchidism, hypospadias, reduced adult testis size, and compensated adult Leydig cell failure). Focal testicular dysgenesis, reduced size of adult male reproductive organs, and TDS disorders and their severity were all strongly associated with reduced AGD. We related our findings to human TDS cases by demonstrating similar focal dysgenetic changes in testes of men with preinvasive germ cell neoplasia (GCNIS) and in testes of DBP-MPW animals. If our results are translatable to humans, they suggest that identification of potential causes of human TDS disorders should focus on exposures during a human MPW equivalent, especially if negatively associated with offspring AGD.
Collapse
Affiliation(s)
- Sander van den Driesche
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Karen R Kilcoyne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ida Wagner
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Diane Rebourcet
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ashley Boyle
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rod Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris McKinnell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Sheila Macpherson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Roland Donat
- Edinburgh Urological Cancer Group, Department of Urology, Western General Hospital, Edinburgh, United Kingdom
| | - Chitranjan J Shukla
- Edinburgh Urological Cancer Group, Department of Urology, Western General Hospital, Edinburgh, United Kingdom
| | - Anne Jorgensen
- Department of Growth & Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Niels E Skakkebaek
- Department of Growth & Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
110
|
Manibusan MK, Touart LW. A comprehensive review of regulatory test methods for endocrine adverse health effects. Crit Rev Toxicol 2017; 47:433-481. [PMID: 28617201 DOI: 10.1080/10408444.2016.1272095] [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] [Indexed: 01/22/2023]
Abstract
Development of new endocrine disruption-relevant test methods has been the subject of intensive research efforts for the past several decades, prompted in part by mandates in the 1996 Food Quality Protection Act (FQPA). While scientific understanding and test methods have advanced, questions remain on whether current scientific methods are capable of adequately addressing the complexities of the endocrine system for regulatory health and ecological risk assessments. The specific objective of this article is to perform a comprehensive, detailed evaluation of the adequacy of current test methods to inform regulatory risk assessments of whether a substance has the potential to perturb endocrine-related pathways resulting in human adverse effects. To that end, approximately 42 existing test guidelines (TGs) were considered in the evaluation of coverage for endocrine-related adverse effects. In addition to evaluations of whether test methods are adequate to capture endocrine-related effects, considerations of further enhancements to current test methods, along with the need to develop novel test methods to address existing test method gaps are described. From this specific evaluation, up to 35 test methods are capable of informing whether a chemical substance perturbs known endocrine related biological pathways. Based on these findings, it can be concluded that current validated test methods are adequate to discern substances that may perturb the endocrine system, resulting in an adverse health effect. Together, these test methods predominantly form the core data requirements of a typical food-use pesticide registration submission. It is recognized, however, that the current state of science is rapidly advancing and there is a need to update current test methods to include added enhancements to ensure continued coverage and public health and environmental protection.
Collapse
Affiliation(s)
| | - L W Touart
- b Equiparent Consulting , Woodbridge , VA , USA
| |
Collapse
|
111
|
Shen H, Liao K, Wu HF, Lu HC, Li Y, Li Z, Zhang W. In utero exposure of high-dose di-n-butyl phthalate resulted in opposite effects on testicular cell apoptosis in late embryonic and pubertal male rat offspring. Hum Exp Toxicol 2017; 36:1236-1247. [DOI: 10.1177/0960327116685886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: To investigate the effects of in utero exposure to high-dose di- n-butyl phthalate (DBP) on testicular cell apoptosis in late embryonic and pubertal male rat offspring. Methods: Twenty pregnant Sprague-Dawley (SD) rats were divided into two groups. During gestation day (GD) 12 to GD 19, control group was given 1 ml day−1 of olive oil and experimental group was given DBP 500 mg kg−1 day−1 by gavage. On GD 19.5 and postnatal day (PND) 45, the testes were removed. Morphological analysis of the testes was observed by transmission electron microscopy and hematoxylin and eosin (H&E) staining. Testicular cell apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). The expression of Bcl-2, Bax, and p53 was presented by immunohistochemistry (IHC) and western blot. Data of the two groups was compared using independent samples t-test and Mann–Whitney test by SPSS 20.0. Results: H&E staining showed that spermatogenetic cells were significantly decreased in DBP exposed pubertal rat testis. The apoptosis index of testes in DBP-treated group was significantly lower on GD 19.5 but higher on PND 45 than that of the controls ( p < 0.01). IHC and western blot revealed significantly increased expression of Bcl-2 in GD 19.5 rat testis and Bax and p53 in PND 45 rat testis after DBP exposure, compared with the control ( p < 0.05). Conclusion: In utero exposure of high-dose DBP resulted in opposite effects on testicular cell apoptosis in late embryonic and pubertal rat offspring. The overexpression of Bcl-2, Bax, and p53 might be related to the occurrence of abnormal apoptosis and finally produce male infertility.
Collapse
Affiliation(s)
- H Shen
- Department of Urology, BenQ Medical Center, Nanjing Medical University, Nanjing, China
| | - K Liao
- Department of Urology, BenQ Medical Center, Nanjing Medical University, Nanjing, China
| | - H-F Wu
- Department of Urology, BenQ Medical Center, Nanjing Medical University, Nanjing, China
| | - H-C Lu
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Y Li
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Z Li
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - W Zhang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
112
|
Haraux E, Braun K, Buisson P, Stéphan-Blanchard E, Devauchelle C, Ricard J, Boudailliez B, Tourneux P, Gouron R, Chardon K. Maternal Exposure to Domestic Hair Cosmetics and Occupational Endocrine Disruptors Is Associated with a Higher Risk of Hypospadias in the Offspring. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 14:ijerph14010027. [PMID: 28036072 PMCID: PMC5295278 DOI: 10.3390/ijerph14010027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 01/01/2023]
Abstract
Pregnant women are exposed to various chemical products at home and at work. Some of these products contain endocrine-disrupting chemicals (EDCs) such as cosmetics, pesticides, industrial chemicals, heavy metals, plastics or medications that could alter sexual differentiation and increase the risk of hypospadias. We evaluated maternal occupational and household exposures that could constitute risk factors for hypospadias. From 2011 to 2014, we enrolled 57 full-term newborns with hypospadias and three randomly selected controls per case (162 control newborns), matched for gestational age, from 11 maternity units in Picardy, France. Neonatal and parental data were collected at birth (personal characteristics, maternal lifestyle, and medical history). Maternal occupational exposure was assessed by a job-exposure matrix for EDCs from a job history questionnaire completed by mothers. Odds ratios (OR) and 95% confidence intervals (CI) were calculated with univariate and multivariable logistic regression, and adjusted for relevant covariates. Multivariate analysis showed a strong association between hypospadias and potential maternal occupational exposure to EDCs and maternal household use of hair cosmetics (OR 6.1, 95% CI: 1.1–34.9; OR: 9.6, 95% CI: 1.4–66.1, respectively). Our results suggest that maternal occupational exposure to EDCs is a risk factor for hypospadias and suggests a possible influence of household use of hair cosmetics during early pregnancy on the incidence of hypospadias in the offspring. A larger study with more accurate exposure assessment should evaluate the impact of EDCs in hair cosmetics on the incidence of hypospadias.
Collapse
Affiliation(s)
- Elodie Haraux
- Department of Paediatric Surgery, Amiens University Hospital, 80054 Amiens, France.
| | - Karine Braun
- Department of Paediatrics, Amiens University Hospital, 80054 Amiens, France.
| | - Philippe Buisson
- Department of Paediatric Surgery, Amiens University Hospital, 80054 Amiens, France.
| | | | | | - Jannick Ricard
- Department of Paediatric Surgery, Amiens University Hospital, 80054 Amiens, France.
| | - Bernard Boudailliez
- Department of Paediatrics, Amiens University Hospital, 80054 Amiens, France.
| | - Pierre Tourneux
- Department of Paediatric Intensive Care Unit, Amiens University Hospital, 80054 Amiens, France.
| | - Richard Gouron
- Department of Paediatric Surgery, Amiens University Hospital, 80054 Amiens, France.
| | - Karen Chardon
- PériTox-INERIS Laboratory, Jules Verne University of Picardy, 80054 Amiens, France.
| |
Collapse
|
113
|
Howdeshell KL, Hotchkiss AK, Gray LE. Cumulative effects of antiandrogenic chemical mixtures and their relevance to human health risk assessment. Int J Hyg Environ Health 2016; 220:179-188. [PMID: 27923611 DOI: 10.1016/j.ijheh.2016.11.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 11/28/2022]
Abstract
Toxicological studies of defined chemical mixtures assist human health risk assessment by establishing how chemicals interact with one another to induce an effect. This paper reviews how antiandrogenic chemical mixtures can alter reproductive tract development in rats with a focus on the reproductive toxicant phthalates. The reviewed studies compare observed mixture data to mathematical mixture model predictions based on dose addition or response addition to determine how the individual chemicals in a mixture interact (e.g., additive, greater, or less than additive). Phthalate mixtures were observed to act in a dose additive manner based on the relative potency of the individual phthalates to suppress fetal testosterone production. Similar dose additive effects have been reported for mixtures of phthalates with antiandrogenic pesticides of differing mechanisms of action. Overall, data from these phthalate experiments in rats can be used in conjunction with human biomonitoring data to determine individual hazard indices, and recent cumulative risk assessments in humans indicate an excess risk to antiandrogenic chemical mixtures that include phthalates only or phthalates in combination with other antiandrogenic chemicals.
Collapse
Affiliation(s)
- Kembra L Howdeshell
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), PO Box 12233, Mail Drop K2-04, Research Triangle Park, NC 27709-2233, USA.
| | - Andrew K Hotchkiss
- National Center for Environmental Assessment (NCEA), National Health and Environmental Effects Research Laboratories (NHEERL), Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - L Earl Gray
- Toxicology Assessment Division, National Health and Environmental Effects Research Laboratories (NHEERL), Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
114
|
Wang Y, Bilandzic M, Ooi GT, Findlay JK, Stenvers KL. Endogenous inhibins regulate steroidogenesis in mouse TM3 Leydig cells by altering SMAD2 signalling. Mol Cell Endocrinol 2016; 436:68-77. [PMID: 27465829 DOI: 10.1016/j.mce.2016.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/28/2022]
Abstract
This study tested the hypothesis that inhibins act in an autocrine manner on Leydig cells using a pre-pubertal Leydig cell line, TM3, as a model of immature Leydig cells. The expression of Inha, Inhba, and Inhbb in TM3 cells was determined by RT-PCR and the production of the inhibin-alpha subunit was confirmed by western blot. Knockdown of Inha expression resulted in significant decreases in the expression of Leydig cell markers Cyp17a1, Cyp11a1, Nr5a1, and Insl3. Western blot showed that activin A, TGFβ1 and TGFβ2 activated SMAD2, and that knockdown of Inha expression in TM3 cells enhanced both activin A- and TGFβ-induced SMAD2 activation. SB431542, a chemical inhibitor of the TGFβ/activin type I receptors, blocked ligand-induced SMAD2 activation and the downregulation of Cyp17a1 expression. Our findings demonstrate that TGFβs and activin A negatively regulate steroidogenic gene expression in TM3 cells via ALK4/5 and SMAD2 and endogenous inhibins can counter this regulation.
Collapse
Affiliation(s)
- Yao Wang
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia.
| | - Maree Bilandzic
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia
| | - Guck T Ooi
- Sun BioMedical Technologies, 209 W. Ridgecrest Blvd, Suite A, Ridgecrest, CA, 93555, USA
| | - Jock K Findlay
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia
| | - Kaye L Stenvers
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Victoria, 3168, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3168, Australia
| |
Collapse
|
115
|
Dekant W, Bridges J. Assessment of reproductive and developmental effects of DINP, DnHP and DCHP using quantitative weight of evidence. Regul Toxicol Pharmacol 2016; 81:397-406. [DOI: 10.1016/j.yrtph.2016.09.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
|
116
|
Lardone MC, Argandoña F, Flórez M, Parada-Bustamante A, Ebensperger M, Palma C, Piottante A, Castro A. Overexpression of CYP19A1
aromatase in Leydig cells is associated with steroidogenic dysfunction in subjects with Sertoli cell-only syndrome. Andrology 2016; 5:41-48. [DOI: 10.1111/andr.12289] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022]
Affiliation(s)
- M. C. Lardone
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
| | - F. Argandoña
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
| | - M. Flórez
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
| | - A. Parada-Bustamante
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
| | - M. Ebensperger
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
- Urology Department; San Borja Arriarán Clinical Hospital; Santiago Chile
| | - C. Palma
- Urology Department; José Joaquín Aguirre Clinical Hospital; School of Medicine; University of Chile; Santiago Chile
- Urology Department; Clínica Las Condes; Santiago Chile
| | - A. Piottante
- Pathology Department; Clínica Las Condes; Santiago Chile
| | - A. Castro
- Institute of Maternal and Child Research; School of Medicine; University of Chile; Santiago Chile
| |
Collapse
|
117
|
Boisvert A, Jones S, Issop L, Erythropel HC, Papadopoulos V, Culty M. In vitro functional screening as a means to identify new plasticizers devoid of reproductive toxicity. ENVIRONMENTAL RESEARCH 2016; 150:496-512. [PMID: 27423704 DOI: 10.1016/j.envres.2016.06.033] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/19/2016] [Accepted: 06/20/2016] [Indexed: 06/06/2023]
Abstract
Plasticizers are indispensable additives providing flexibility and malleability to plastics. Among them, several phthalates, including di (2-ethylhexyl) phthalate (DEHP), have emerged as endocrine disruptors, leading to their restriction in consumer products and creating a need for new, safer plasticizers. The goal of this project was to use in vitro functional screening tools to select novel non-toxic plasticizers suitable for further in vivo evaluation. A panel of novel compounds with satisfactory plasticizer properties and biodegradability were tested, along with several commercial plasticizers, such as diisononyl-cyclohexane-1,2-dicarboxylate (DINCH®). MEHP, the monoester metabolite of DEHP was also included as reference compound. Because phthalates target mainly testicular function, including androgen production and spermatogenesis, we used the mouse MA-10 Leydig and C18-4 spermatogonial cell lines as surrogates to examine cell survival, proliferation, steroidogenesis and mitochondrial integrity. The most promising compounds were further assessed on organ cultures of rat fetal and neonatal testes, corresponding to sensitive developmental windows. Dose-response studies revealed the toxicity of most maleates and fumarates, while identifying several dibenzoate and succinate plasticizers as innocuous on Leydig and germ cells. Interestingly, DINCH®, a plasticizer marketed as a safe alternative to phthalates, exerted a biphasic effect on steroid production in MA-10 and fetal Leydig cells. MEHP was the only plasticizer inducing the formation of multinucleated germ cells (MNG) in organ culture. Overall, organ cultures corroborated the cell line data, identifying one dibenzoate and one succinate as the most promising candidates. The adoption of such collaborative approaches for developing new chemicals should help prevent the development of compounds potentially harmful to human health.
Collapse
Affiliation(s)
- Annie Boisvert
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Medicine, McGill University, Montreal, Quebec, Canada H4A 3J1
| | - Steven Jones
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Medicine, McGill University, Montreal, Quebec, Canada H4A 3J1
| | - Leeyah Issop
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Medicine, McGill University, Montreal, Quebec, Canada H4A 3J1
| | - Hanno C Erythropel
- Department of Chemical Engineering, McGill University, Montreal, Quebec, Canada H4A 3J1
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Medicine, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada H4A 3J1
| | - Martine Culty
- The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Medicine, McGill University, Montreal, Quebec, Canada H4A 3J1; Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada H4A 3J1.
| |
Collapse
|
118
|
Hart RJ, Doherty DA, Keelan JA, McLachlan R, Skakkebaek NE, Norman RJ, Dickinson JE, Pennell CE, Newnham JP, Hickey M, Handelsman DJ. Early Life Events Predict Adult Testicular Function; Data Derived From the Western Australian (Raine) Birth Cohort. J Clin Endocrinol Metab 2016; 101:3333-44. [PMID: 27340882 DOI: 10.1210/jc.2016-1646] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT The impact of early life events on testicular function in adulthood is not well understood. OBJECTIVE To study the early influences of fetal growth, exposures to cigarette smoke in utero and cord blood estrogens, and the influences of growth and adiposity in childhood through adolescence; on testicular function in adulthood. DESIGN Male members of the Western Australian Pregnancy Cohort (Raine) were contacted at 20-22 years of age. Of 913 contacted, 423 (56%) agreed to participate; 404 underwent a testicular ultrasound, 365 provided a semen sample, and reproductive hormones were measured (384). Fetal growth measurements (n = 137), umbilical cord estrogen concentrations (n = 128), cord testosterone (T) (n = 125), and child-adulthood growth charts (n = 395) were available. RESULTS Median sperm output for the 18.6% of men exposed in utero to smoking was lower than nonexposed (82.4 × 10(6) vs 123.1 × 10(6); P = .029). Sperm output in adulthood was inversely correlated with cord serum estradiol (P = .019) and estrone (P = .018). The sperm output of men whose cord blood estradiol and estrone were less than 50th percentile vs more than 50th percentile was 191.1 × 10(6) vs 100.5 × 10(6) (P = .002) and 190.0 × 10(6) vs 106.0 × 10(6) (P = .012), respectively. Men with favorable fetal growth patterns in utero were less likely to have total motile sperm counts within the lowest quartile (P = .011), and men born prematurely had reduced serum T levels in adulthood (13.4 vs 16.6nmol/L, P = .024). Consistent height above the 50th percentile for age through childhood was associated with larger adult mean testicular volume (P < .001). Optimal body mass index trajectory through childhood and adolescence was associated with larger testicular volume (P = .009) and higher serum inhibin B (P = .010) and T (P = .003) in adulthood. CONCLUSIONS Exposures to maternal smoking and higher cord blood estrogens at delivery were associated with a reduced sperm output in adulthood. Optimal adult testicular function depends on being born at or above average weight, and maintaining optimal growth and adiposity into adulthood.
Collapse
Affiliation(s)
- Roger J Hart
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Dorota A Doherty
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Jeffrey A Keelan
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Rob McLachlan
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Niels E Skakkebaek
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Robert J Norman
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Jan E Dickinson
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Craig E Pennell
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - John P Newnham
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Martha Hickey
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - David J Handelsman
- School of Women's and Infants' Health (R.J.H., D.A.D., J.A.K., J.E.D., C.E.P., J.P.N.), The University of Western Australia, Perth, Western Australia 6008, Australia; Fertility Specialists of Western Australia (R.J.H.), Bethesda Hospital, Claremont, Western Australia 6010, Australia; Women and Infants Research Foundation (D.A.D., J.A.K., C.E.P., J.P.N.), King Edward Memorial Hospital, Perth, Western Australia 6008, Australia; Centre for Endocrinology and Metabolism (R.M.), Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; Monash University (R.M.), Clayton, Victoria 3168, Australia; University Department of Growth and Reproduction (N.E.S.), Rigshospitalet, Department of Growth and Reproduction, 2100 Copenhagen, Denmark; Robinson Institute (R.J.N.), University of Adelaide, Adelaide, South Australia 5000, Australia; Department of Obstetrics and Gynaecology (M.H.), The University of Melbourne, The Royal Women's Hospital, Melbourne, Victoria 3052, Australia; and ANZAC Research Institute (D.J.H.), University of Sydney, Concord Hospital, Sydney, New South Wales 2139, Australia
| |
Collapse
|
119
|
Polland A, Berookhim BM. Fertility concerns in men with genitourinary malignancies: Treatment dilemmas, fertility options, and medicolegal considerations. Urol Oncol 2016; 34:399-406. [DOI: 10.1016/j.urolonc.2016.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 04/10/2016] [Accepted: 05/04/2016] [Indexed: 12/11/2022]
|
120
|
Maternal treatment with dexamethasone during gestation alters sexual development markers in the F1 and F2 male offspring of Wistar rats. J Dev Orig Health Dis 2016; 8:101-112. [DOI: 10.1017/s2040174416000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal treatment with dexamethasone (Dex) in threatening preterm delivery alters activities at the hypothalamic–pituitary–adrenal axis in the offspring. This alteration may interfere with reproductive function. The impact of gestational Dex exposure on male reproductive function of the offspring was investigated. A total of 25 pregnant rats randomly assigned to five groups (n=5) were treated with normal saline (control), Dex (100 μg/kg/day sc) during gestation days (GD) 1–7, 8–14, 15–21 and 1–21, respectively. Birth weight, anogenital distance (AGD), pubertal age, sperm parameters, hormonal profile and histopathology of testis and epididymis were determined in the F1 and F2 offspring. Results showed a significant increase (P<0.05) in pubertal age, serum corticosterone and gonadotropin-releasing hormone (GnRH) levels in the male offspring of DexGD 15–21 and 1–21 groups and a significant decrease (P<0.05) in serum testosterone, luteinizing hormone, birth weight and AGD at birth in the male F1 offspring. In the F2 offspring, there was a significant reduction (P<0.05) in serum corticosterone, testosterone, follicle-stimulating hormone and GnRH when compared with the control. Dex treatment at GD 15–21 and 1–21 significantly reduced (P<0.05) sperm motility and normal morphology in the F1 and F2 offspring. Maternal Dex treatment in rats during late gestation may disrupt sexual development markers in the F1 and F2 male offspring.
Collapse
|
121
|
Lea RG, Byers AS, Sumner RN, Rhind SM, Zhang Z, Freeman SL, Moxon R, Richardson HM, Green M, Craigon J, England GCW. Environmental chemicals impact dog semen quality in vitro and may be associated with a temporal decline in sperm motility and increased cryptorchidism. Sci Rep 2016; 6:31281. [PMID: 27503122 PMCID: PMC4977511 DOI: 10.1038/srep31281] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 07/15/2016] [Indexed: 11/15/2022] Open
Abstract
Adverse temporal trends in human semen quality and cryptorchidism in infants have been associated with exposure to environmental chemicals (ECs) during development. Here we report that a population of breeding dogs exhibit a 26 year (1988-2014) decline in sperm quality and a concurrent increased incidence of cryptorchidism in male offspring (1995-2014). A decline in the number of males born relative to the number of females was also observed. ECs, including diethylhexyl phthalate (DEHP) and polychlorinated biphenyl 153 (PCB153), were detected in adult dog testes and commercial dog foods at concentrations reported to perturb reproductive function in other species. Testicular concentrations of DEHP and PCB153 perturbed sperm viability, motility and DNA integrity in vitro but did not affect LH stimulated testosterone secretion from adult testis explants. The direct effects of chemicals on sperm may therefore contribute to the decline in canine semen quality that parallels that reported in the human.
Collapse
Affiliation(s)
- Richard G. Lea
- School of Veterinary Medicine and Science, University of Nottingham, UK
- School of Animal Rural and Environmental Sciences, Nottingham Trent University, UK
| | - Andrew S. Byers
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rebecca N. Sumner
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Stewart M. Rhind
- Environmental and Biochemical Sciences, The James Hutton Institute, UK
| | - Zulin Zhang
- Environmental and Biochemical Sciences, The James Hutton Institute, UK
| | - Sarah L. Freeman
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rachel Moxon
- National Breeding Centre, Guide Dogs for the Blind Association, UK
| | | | - Martin Green
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Jim Craigon
- School of Biosciences, University of Nottingham, UK
| | | |
Collapse
|
122
|
Barlow PW. Why so many sperm cells? Not only a possible means of mitigating the hazards inherent to human reproduction but also an indicator of an exaptation. Commun Integr Biol 2016; 9:e1204499. [PMID: 27574542 PMCID: PMC4988455 DOI: 10.1080/19420889.2016.1204499] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/17/2016] [Indexed: 11/10/2022] Open
Abstract
Redundancy—the excess of supply over necessity—has recently been proposed for human sperm cells. However, the apparent superfluity of cell numbers may be necessary in order to circumvent the hazards, many of which can be quantified, that can occur during the transition from gametogenesis within the testes to zygosis within the female reproductive tract. Sperm cell numbers are directly related to testicular volume, and it is owing to a redundancy, and the possible exaptation, of this latter parameter that a putative excess of sperm cells is perceived.
Collapse
Affiliation(s)
- Peter W Barlow
- School of Biological Sciences, University of Bristol , Bristol, UK
| |
Collapse
|
123
|
Leung MC, Phuong J, Baker NC, Sipes NS, Klinefelter GR, Martin MT, McLaurin KW, Setzer RW, Darney SP, Judson RS, Knudsen TB. Systems Toxicology of Male Reproductive Development: Profiling 774 Chemicals for Molecular Targets and Adverse Outcomes. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1050-61. [PMID: 26662846 PMCID: PMC4937872 DOI: 10.1289/ehp.1510385] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/24/2015] [Indexed: 05/21/2023]
Abstract
BACKGROUND Trends in male reproductive health have been reported for increased rates of testicular germ cell tumors, low semen quality, cryptorchidism, and hypospadias, which have been associated with prenatal environmental chemical exposure based on human and animal studies. OBJECTIVE In the present study we aimed to identify significant correlations between environmental chemicals, molecular targets, and adverse outcomes across a broad chemical landscape with emphasis on developmental toxicity of the male reproductive system. METHODS We used U.S. EPA's animal study database (ToxRefDB) and a comprehensive literature analysis to identify 774 chemicals that have been evaluated for adverse effects on male reproductive parameters, and then used U.S. EPA's in vitro high-throughput screening (HTS) database (ToxCastDB) to profile their bioactivity across approximately 800 molecular and cellular features. RESULTS A phenotypic hierarchy of testicular atrophy, sperm effects, tumors, and malformations, a composite resembling the human testicular dysgenesis syndrome (TDS) hypothesis, was observed in 281 chemicals. A subset of 54 chemicals with male developmental consequences had in vitro bioactivity on molecular targets that could be condensed into 156 gene annotations in a bipartite network. CONCLUSION Computational modeling of available in vivo and in vitro data for chemicals that produce adverse effects on male reproductive end points revealed a phenotypic hierarchy across animal studies consistent with the human TDS hypothesis. We confirmed the known role of estrogen and androgen signaling pathways in rodent TDS, and importantly, broadened the list of molecular targets to include retinoic acid signaling, vascular remodeling proteins, G-protein coupled receptors (GPCRs), and cytochrome P450s. CITATION Leung MC, Phuong J, Baker NC, Sipes NS, Klinefelter GR, Martin MT, McLaurin KW, Setzer RW, Darney SP, Judson RS, Knudsen TB. 2016. Systems toxicology of male reproductive development: profiling 774 chemicals for molecular targets and adverse outcomes. Environ Health Perspect 124:1050-1061; http://dx.doi.org/10.1289/ehp.1510385.
Collapse
Affiliation(s)
- Maxwell C.K. Leung
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
- Address correspondence to M.C.K. Leung, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27711 USA. Telephone: (919) 541-2721. E-mail: , or T.B. Knudsen, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27711 USA. Telephone: (919) 541-9776. E-mail:
| | - Jimmy Phuong
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | | | - Nisha S. Sipes
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - Gary R. Klinefelter
- National Health and Environmental Effects Research Laboratory, U.S. EPA, Research Triangle Park, North Carolina
| | - Matthew T. Martin
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - Keith W. McLaurin
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - R. Woodrow Setzer
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - Sally Perreault Darney
- National Health and Environmental Effects Research Laboratory, U.S. EPA, Research Triangle Park, North Carolina
| | - Richard S. Judson
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - Thomas B. Knudsen
- National Center for Computational Toxicology, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
- Address correspondence to M.C.K. Leung, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27711 USA. Telephone: (919) 541-2721. E-mail: , or T.B. Knudsen, U.S. Environmental Protection Agency, 109 T.W. Alexander Dr., Research Triangle Park, NC 27711 USA. Telephone: (919) 541-9776. E-mail:
| |
Collapse
|
124
|
Comments on Li et al. Effects of in Utero Exposure to Dicyclohexyl Phthalate on Rat Fetal Leydig Cells. Int. J. Environ. Res. Public Health 2016, 13, 246. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13060532. [PMID: 27231928 PMCID: PMC4923989 DOI: 10.3390/ijerph13060532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/03/2016] [Accepted: 05/23/2016] [Indexed: 01/26/2023]
Abstract
Profiling the expression levels of genes or proteins in tissues comprising two or more cell types is commonplace in biological sciences. Such analyses present particular challenges, however, for example a potential shift in cellular composition, or ‘cellularity’, between specimens. That is, does an observed change in expression level represent what occurs within individual cells, or does it represent a shift in the ratio of different cell types within the tissue? This commentary attempts to highlight the importance of considering cellularity when interpreting quantitative expression data, using the mammalian testis and a recent study on the effects of phthalate exposure on testis function as an example.
Collapse
|
125
|
Singal AK, Jain VG, Gazali Z, Shekhawat P. Shorter anogenital distance correlates with the severity of hypospadias in pre-pubertal boys. Hum Reprod 2016; 31:1406-10. [DOI: 10.1093/humrep/dew115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/22/2016] [Indexed: 12/18/2022] Open
|
126
|
|
127
|
Nistal M, Paniagua R, González-Peramato P, Reyes-Múgica M. Perspectives in Pediatric Pathology, Chapter 14. Natural History of Undescended Testes. Pediatr Dev Pathol 2016; 19:183-201. [PMID: 25105691 DOI: 10.2350/14-05-1483-pb.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cryptorchidism is one of the most frequent problems encountered in pediatric urology. Its causes, associated lesions, and prognosis in terms of fertility have been a source of interest and discrepancies for pediatric pathologists and urological surgeons.
Collapse
Affiliation(s)
- Manuel Nistal
- 1 Department of Pathology, Hospital La Paz, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo No. 2, Madrid 28029, Spain
| | - Ricardo Paniagua
- 2 Department of Cell Biology, Universidad de Alcala, Madrid, Spain
| | - Pilar González-Peramato
- 1 Department of Pathology, Hospital La Paz, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo No. 2, Madrid 28029, Spain
| | - Miguel Reyes-Múgica
- 3 Department of Pathology, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
128
|
Adverse effects of endocrine disruptors on the foetal testis development: focus on the phthalates. Folia Histochem Cytobiol 2016; 47:S67-74. [PMID: 20067897 DOI: 10.2478/v10042-009-0056-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There are great concerns about the increasing incidence of abnormalities in male reproductive function. Human sperm counts have markedly dropped and the rate of testicular cancer has clearly augmented over the past four decades. Moreover, the prevalence rates of cryptorchidism and hypospadias are also probably increasing. It has been hypothesized that all these adverse trends in male reproduction result from abnormalities in the development of the testis during foetal and neonatal life. Furthermore, many recent epidemiological, clinical and experimental data suggest that these male reproductive disorders could be due to the effects of xenobiotics termed endocrine disruptors, which are becoming more and more concentrated and prevalent in our environment. Among these endocrine disruptors, we chose to focus this review on the phthalates for different reasons: 1) they are widespread in the environment; 2) their concentrations in many human biological fluids have been measured; 3) the experimental data using rodent models suggesting a reprotoxicity are numerous and are the most convincing; 4) their deleterious effects on the in vivo and in vitro development and function of the rat foetal testis have been largely studied; 5) some epidemiological data in humans suggest a reprotoxic effect at environmental concentrations at least during neonatal life. However, the direct effects of phthalates on human foetal testis have never been explored. Thus, as we did for the rat in the 1990s, we recently developed and validated an organ culture system which allows maintenance of the development of the different cell types of human foetal testis. In this system, addition of 10-4 M MEHP (mono-2-ethylhexyl phthalate), the most produced phthalate, had no effect on basal or LH-stimulated production of testosterone, but it reduced the number of germ cells by increasing their apoptosis, without modification of their proliferation. This is the first experimental demonstration that phthalates alter the development of the foetal testis in humans. Using our organotypic culture system, we and others are currently investigating the effect of MEHP in the mouse and the rat, and it will be interesting to compare the results between these species to analyse the relevance of toxicological tests based on rodent models.
Collapse
|
129
|
Zhou N, Sun L, Yang H, Chen Q, Wang X, Yang H, Tan L, Chen H, Zhang G, Ling X, Huang L, Zou P, Peng K, Liu T, Liu J, Ao L, Zhou Z, Cui Z, Cao J. Anogenital distance is associated with serum reproductive hormones, but not with semen quality in young men. Hum Reprod 2016; 31:958-67. [PMID: 27052617 DOI: 10.1093/humrep/dew052] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/26/2016] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is anogenital distance associated with semen parameters and serum reproductive hormone levels in males? SUMMARY ANSWER Anogenital distance is associated with serum reproductive hormones, but not with semen quality. WHAT IS KNOWN ALREADY Epidemiological studies have suggested that anogenital distance (AGD) may be associated with testicular dysfunction in adult men. However, the role of AGD in estimating male reproductive function remains unclear. STUDY DESIGN, SIZE, DURATION We examined the associations between AGD and semen parameters and reproductive hormones levels in 656 young college students in a Male Reproductive Health in Chongqing College Students (MARHCSs) cohort study in June of 2014. PARTICIPANTS/MATERIALS, SETTING, METHODS In this study, two variants of AGD (AGDAP and AGDAS) were measured in 656 university students. Serum levels of testosterone (T), estradiol (E2), progesterone (P), prolactin (PRL), luteinizing hormone (LH), follicle-stimulating hormone (FSH), sex hormone-binding globulin (SHBG) and inhibin-B; and semen quality outcomes, including semen volume, sperm concentration, total sperm number, sperm progressive motility, total motility and morphology, were assessed. The associations between AGD and semen parameters/reproductive hormones levels were analyzed using multiple regression analysis. MAIN RESULTS AND THE ROLE OF CHANCE Both AGDAS and AGDAP were not associated with any semen parameters. In the non-parametric correlation analysis, AGDAP were correlated with sperm progressive motility and reproductive hormones of E2, testosterone, SHBG and the testosterone/LH ratio. However, body mass index (BMI) also significantly correlated with serum testosterone ( ITALIC! r = -0.216, ITALIC! P = <0.0001) and SHBG ( ITALIC! r = -0.229, ITALIC! P = <0.001). In the multiple regression models, AGDAP was negatively associated with the serum E2 level (95% CI, -0.198 to -0.043; ITALIC! P = 0.002) and positively associated with the ratio of T/E2 (95% CI, 0.004-0.011; ITALIC! P = 0.001) after an adjustment for BMI and other confounders. LIMITATIONS, REASONS FOR CAUTION Using only a single semen sample to predict male reproductive function over a longer period is a potential limitation of the present study. The other limitation is the cross-sectional nature of the study design. Longitudinal data from an extended follow-up on a large cohort would be more definitive. WIDER IMPLICATIONS OF THE FINDINGS Our results do not support previous studies where AGD is associated with male semen quality. The utility of AGD in predicting reproductive outcomes in adult males should thus be considered prudently. STUDY FUNDING/COMPETING INTERESTS This study was supported by the Key Program of Natural Science Funding of China (no. 81130051), Young Scientist Program of NSFC (no. 81502788) and the National Scientific and Technological Support Program of China (no. 2013BAI12B02). None of authors had any competing interests to declare.
Collapse
Affiliation(s)
- Niya Zhou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Lei Sun
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Huan Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Xiaogang Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Hao Yang
- Chongqing Institute of Science and Technology for Population and Family Planning, Chongqing, PR China
| | - Lu Tan
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Hongqiang Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Guowei Zhang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Xi Ling
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Linping Huang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Peng Zou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Kaige Peng
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Taixiu Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Jinyi Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Ziyuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Zhihong Cui
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
130
|
Wang YX, Zeng Q, Sun Y, Yang P, Wang P, Li J, Huang Z, You L, Huang YH, Wang C, Li YF, Lu WQ. Semen phthalate metabolites, semen quality parameters and serum reproductive hormones: A cross-sectional study in China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 211:173-182. [PMID: 26766535 DOI: 10.1016/j.envpol.2015.12.052] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/05/2015] [Accepted: 12/23/2015] [Indexed: 06/05/2023]
Abstract
Exposure to phthalates has been found to have adverse effects on male reproductive function in animals. However, the findings from human studies are inconsistent. Here we examined the associations of phthalate exposure with semen quality and reproductive hormones in a Chinese population using phthalate metabolite concentrations measured in semen as biomarkers. Semen (n = 687) and blood samples (n = 342) were collected from the male partners of sub-fertile couples who presented to the Reproductive Center of Tongji Hospital in Wuhan, China. Semen quality parameters and serum reproductive hormone levels were determined. Semen concentrations of 8 phthalate metabolites were assessed using high-performance liquid chromatography and tandem mass spectrometry. Associations of the semen phthalate metabolites with semen quality parameters and serum reproductive hormones were assessed using confounder-adjusted linear and logistic regression models. Semen phthalate metabolites were significantly associated with decreases in semen volume [mono-n-butyl phthalate (MBP), mono-(2-ethylhexyl) phthalate (MEHP), mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), mono(2-ethyl-5-oxohexyl) phthalate (MEOHP)], sperm curvilinear velocity [monobenzyl phthalate (MBzP), MEHP, the percentage of di-(2-ethylhexyl)-phthalate metabolites excreted as MEHP (%MEHP)], and straight-line velocity (MBzP, MEHP, %MEHP), and also associated with an increased percentage of abnormal heads and tails (MBzP) (all p for trend <0.05). These associations remained suggestive or significant after adjustment for multiple testing. There were no significant associations between semen phthalate metabolites and serum reproductive hormones. Our findings suggest that environmental exposure to phthalates may impair human semen quality.
Collapse
Affiliation(s)
- Yi-Xin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yang Sun
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pan Yang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Peng Wang
- Department of Biostatistics, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jin Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Zhen Huang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Ling You
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yue-Hui Huang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Cheng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wen-Qing Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
131
|
In utero betamethasone affects 3β-hydroxysteroid dehydrogenase and inhibin-α immunoexpression during testis development. J Dev Orig Health Dis 2016; 7:342-9. [PMID: 27019950 DOI: 10.1017/s2040174416000118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Prenatal glucocorticoids, commonly used in women at risk of preterm delivery, can predispose the newborn to disease in later life. Since male reproductive function is likely to reflect testis development during fetal life, we studied the effects of prenatal glucocorticoids on two key intra-testicular factors that play roles in cellular proliferation and differentiation, 3β-hydroxysteroid dehydrogenase (3β-HSD) and inhibin-α. Pregnant sheep (n=42) were treated with betamethasone (0.5 mg/kg) or saline (control) at 104, 111 and 118 days of gestation (DG). Testicular tissue was sampled from fetuses at 121 and 132DG, and from lambs at 45 and 90 postnatal days (PD). Within the betamethasone treated group, 3β-HSD immunostaining area was greater at 121DG than at 90PD (P=0.04), but the intensity of immunostaining was higher at 90PD than at 121DG (P=0.04), 132DG (P=0.04) and 45PD (P=0.03). Control animals showed no changes in 3β-HSD area or intensity of immunostaining. No significant differences were observed between treated and control animals in immunostaining area, but immunostaining was more intense in the treated group than in the control group at 90PD (P=0.03). For inhibin-α, the proportion of immunostaining area declined in treated offspring from 121DG to 45PD, in contrast to control values, but recovered fully by 90PD, concomitantly with the onset of spermatogenesis. In conclusion, prenatal betamethasone increased the postnatal testicular expression of inhibin-α but reduced the expression of 3β-HSD. These effects could compromise androgen-mediated testicular development and therefore adult capacity for spermatogenesis.
Collapse
|
132
|
van den Driesche S, Macdonald J, Anderson RA, Johnston ZC, Chetty T, Smith LB, Mckinnell C, Dean A, Homer NZ, Jorgensen A, Camacho-Moll ME, Sharpe RM, Mitchell RT. Prolonged exposure to acetaminophen reduces testosterone production by the human fetal testis in a xenograft model. Sci Transl Med 2016; 7:288ra80. [PMID: 25995226 DOI: 10.1126/scitranslmed.aaa4097] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Most common male reproductive disorders are linked to lower testosterone exposure in fetal life, although the factors responsible for suppressing fetal testosterone remain largely unknown. Protracted use of acetaminophen during pregnancy is associated with increased risk of cryptorchidism in sons, but effects on fetal testosterone production have not been demonstrated. We used a validated xenograft model to expose human fetal testes to clinically relevant doses and regimens of acetaminophen. Exposure to a therapeutic dose of acetaminophen for 7 days significantly reduced plasma testosterone (45% reduction; P = 0.025) and seminal vesicle weight (a biomarker of androgen exposure; 18% reduction; P = 0.005) in castrate host mice bearing human fetal testis xenografts, whereas acetaminophen exposure for just 1 day did not alter either parameter. Plasma acetaminophen concentrations (at 1 hour after the final dose) in exposed host mice were substantially below those reported in humans after a therapeutic oral dose. Subsequent in utero exposure studies in rats indicated that the acetaminophen-induced reduction in testosterone likely results from reduced expression of key steroidogenic enzymes (Cyp11a1, Cyp17a1). Our results suggest that protracted use of acetaminophen (1 week) may suppress fetal testosterone production, which could have adverse consequences. Further studies are required to establish the dose-response and treatment-duration relationships to delineate the maximum dose and treatment period without this adverse effect.
Collapse
Affiliation(s)
- Sander van den Driesche
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Joni Macdonald
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Zoe C Johnston
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Tarini Chetty
- Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, EH9 1LF, Scotland, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Chris Mckinnell
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Afshan Dean
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Natalie Z Homer
- Edinburgh CRF Mass Spectrometry Core, Centre for Cardiovascular Science, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Anne Jorgensen
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.,University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Maria-Elena Camacho-Moll
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.,Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, EH9 1LF, Scotland, UK
| |
Collapse
|
133
|
Effects of in Utero Exposure to Dicyclohexyl Phthalate on Rat Fetal Leydig Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13030246. [PMID: 26907321 PMCID: PMC4808909 DOI: 10.3390/ijerph13030246] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/04/2022]
Abstract
Dicyclohexyl phthalate (DCHP) is one of the phthalate plasticizers. The objective of the present study was to investigate the effects of DCHP on fetal Leydig cell distribution and function as well as testis development. Female pregnant Sprague Dawley dams orally received vehicle (corn oil, control) or DCHP (10, 100, and 500 mg/kg/day) from gestational day (GD) 12 to GD 21. At GD 21.5, testicular testosterone production, fetal Leydig cell number and distribution, testicular gene and protein expression levels were examined. DCHP administration produced a dose-dependent increase of the incidence of multinucleated gonocytes at ≥100 mg/kg. DCHP dose-dependently increased abnormal fetal Leydig cell aggregation and decreased fetal Leydig cell size, cytoplasmic size, and nuclear size at ≥10 mg/kg. DCHP reduced the expression levels of steroidogenesis-related genes (including Star, Hsd3b1, and Hsd17b3) and testis-descent related gene Insl3 as well as protein levels of 3β-hydroxysteroid dehydrogenase 1 (HSD3B1) and insulin-like 3 (INSL3) at ≥10 mg/kg. DCHP significantly inhibited testicular testosterone levels at ≥100 mg/kg. The results indicate that in utero exposure to DCHP affects the expression levels of fetal Leydig cell steroidogenic genes and results in the occurrence of multinucleated gonocytes and Leydig cell aggregation.
Collapse
|
134
|
Zhang L, Ding S, Qiao P, Dong L, Yu M, Wang C, Zhang M, Zhang L, Li Y, Tang N, Chang B. n-butylparaben induces male reproductive disorders via regulation of estradiol and estrogen receptors. J Appl Toxicol 2016; 26:1223-1234. [DOI: 10.1002/jat.3291] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Linyuan Zhang
- Key Laboratory of Chemical Safety and Health; National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention; No. 29 Nanwei Road , Xicheng district Beijing 100050 China
| | - Sijin Ding
- Key Laboratory of Chemical Safety and Health; National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention; No. 29 Nanwei Road , Xicheng district Beijing 100050 China
| | - Peihuan Qiao
- Key Laboratory of Chemical Safety and Health; National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention; No. 29 Nanwei Road , Xicheng district Beijing 100050 China
| | - Li Dong
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Miao Yu
- Key Laboratory of Chemical Safety and Health; National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention; No. 29 Nanwei Road , Xicheng district Beijing 100050 China
| | - Chong Wang
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Ming Zhang
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Lixia Zhang
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Yimin Li
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Ning Tang
- Department of Environmental Toxicology; Institute of Environmental Health and Related Product Safety, Chinese Center for Disease Control and Prevention; No. 7 Panjiayuan nanli , Chaoyang district Beijing 100021 China
| | - Bing Chang
- Key Laboratory of Chemical Safety and Health; National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention; No. 29 Nanwei Road , Xicheng district Beijing 100050 China
| |
Collapse
|
135
|
Elzinga-Tinke JE, Dohle GR, Looijenga LH. Etiology and early pathogenesis of malignant testicular germ cell tumors: towards possibilities for preinvasive diagnosis. Asian J Androl 2016; 17:381-93. [PMID: 25791729 PMCID: PMC4430936 DOI: 10.4103/1008-682x.148079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Malignant testicular germ cell tumors (TGCT) are the most frequent cancers in Caucasian males (20-40 years) with an 70% increasing incidence the last 20 years, probably due to combined action of (epi)genetic and (micro)environmental factors. It is expected that TGCT have carcinoma in situ(CIS) as their common precursor, originating from an embryonic germ cell blocked in its maturation process. The overall cure rate of TGCT is more than 90%, however, men surviving TGCT can present long-term side effects of systemic cancer treatment. In contrast, men diagnosed and treated for CIS only continue to live without these long-term side effects. Therefore, early detection of CIS has great health benefits, which will require an informative screening method. This review described the etiology and early pathogenesis of TGCT, as well as the possibilities of early detection and future potential of screening men at risk for TGCT. For screening, a well-defined risk profile based on both genetic and environmental risk factors is needed. Since 2009, several genome wide association studies (GWAS) have been published, reporting on single-nucleotide polymorphisms (SNPs) with significant associations in or near the genes KITLG, SPRY4, BAK1, DMRT1, TERT, ATF7IP, HPGDS, MAD1L1, RFWD3, TEX14, and PPM1E, likely to be related to TGCT development. Prenatal, perinatal, and postnatal environmental factors also influence the onset of CIS. A noninvasive early detection method for CIS would be highly beneficial in a clinical setting, for which specific miRNA detection in semen seems to be very promising. Further research is needed to develop a well-defined TGCT risk profile, based on gene-environment interactions, combined with noninvasive detection method for CIS.
Collapse
Affiliation(s)
| | | | - Leendert Hj Looijenga
- Department of Pathology, Laboratory of Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
136
|
Jeje SO, Akindele OO, Balogun ME, Raji Y. Maternal treatment with dexamethasone during lactation delays male puberty and disrupts reproductive functions via hypothalamic-pituitary-gonadal axis alterations. ACTA ACUST UNITED AC 2016; 23:43-9. [PMID: 26774541 DOI: 10.1016/j.pathophys.2015.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/25/2015] [Accepted: 12/26/2015] [Indexed: 01/09/2023]
Abstract
The effects of maternal treatment with dexamethasone during lactation on pubertal timing, serum hormonal profile and sperm indices in the male offspring were assessed. Twenty lactating dams were divided into 4 groups (n=5). Group 1 was administered subcutaneously 0.02ml/100g/day normal saline at lactation days 1-21. Groups 2-4 were administered subcutaneously 100μg/kg/day dexamethasone (Dex) at lactation days 1-7, 1-14, and 1-21 respectively. Results showed that there was significant reduction in serum testosterone in the DexLD 1-7 (p<0.05), DexLD 1-14 (p<0.01) and DexLD 1-21 (p<0.001) relative to control. In addition there was a significant reduction in serum FSH and LH in the DexLD 1-7 (p<0.01), DexLD 1-14 (p<0.001) and DexLD 1-21 (p<0.001) when compared with the control. Treatment with dexamethasone during lactation significantly increased the days of preputial separation in the DexLD 1-7 (p<0.05), DexLD 1-14 (p<0.05) and DexLD 1-21 (p<0.001) relative to control. Maternal treatment with dexamethasone throughout lactation period also significantly reduced sperm counts (p<0.001), motility (p<0.01) and increased percentage abnormal sperm (p<0.001) in the offspring when compared with the control. In conclusion, maternal treatment with dexamethasone during lactation may induce delayed puberty and disrupt reproductive functions by altering activities at hypothalamic-pituitary-gonadal axis in the male offspring.
Collapse
Affiliation(s)
- S O Jeje
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria; Department of Human Physiology, Cross River University of Technology, Okuku Campus, Cross River State, Nigeria.
| | - O O Akindele
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - M E Balogun
- Department of Physiology, Faculty of Medicine, College of Health Sciences, Ebonyi State University, Abakaliki, Nigeria
| | - Y Raji
- Laboratory for Reproductive Physiology and Developmental Programming, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
137
|
Gong T, Wei Q, Mao D, Shi F. Expression patterns of taste receptor type 1 subunit 3 and α-gustducin in the mouse testis during development. Acta Histochem 2016; 118:20-30. [PMID: 26589384 DOI: 10.1016/j.acthis.2015.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 02/04/2023]
Abstract
Taste receptor type 1 subunit 3 (T1R3) and its associated heterotrimeric G protein α-gustducin (Gα) are involved in sweet and umami sensing in taste cells. They are also strongly expressed in the testis and sperm, but their expression patterns and potential roles involved were previously unknown. In present study, we investigated the expression patterns of T1R3 and Gα in the mouse testis at critical stages of postnatal life, and throughout the spermatogenic cycle. Our results indicated that T1R3 and Gα exhibited a stage-dependent expression pattern during mouse development, and a cell-specific pattern during the spermatogenic cycle. Their expressions have been increased significantly from prepubertal to pubertal periods (P<005), and decreased significantly in aged mice (P<005). The changes were mainly attributed to the differential expression of T1R3 or Gα in elongated spermatids and Leydig cells at different stages of the spermatogenic cycle. In addition, the expression of T1R3 and Gα were first observed in residual bodies of spermatozoa and endothelial cells of blood vessels at post-pubertal mice, while Gα was located in apoptotic spermatogonia of postnatal mice. These novel expression patterns suggest a role of T1R3 and Gα in the onset of spermatogenesis, pace of spermatogenic cycle, and aging of the testis.
Collapse
|
138
|
Kalfa N, Paris F, Philibert P, Orsini M, Broussous S, Fauconnet-Servant N, Audran F, Gaspari L, Lehors H, Haddad M, Guys JM, Reynaud R, Alessandrini P, Merrot T, Wagner K, Kurzenne JY, Bastiani F, Bréaud J, Valla JS, Lacombe GM, Dobremez E, Zahhaf A, Daures JP, Sultan C. Is Hypospadias Associated with Prenatal Exposure to Endocrine Disruptors? A French Collaborative Controlled Study of a Cohort of 300 Consecutive Children Without Genetic Defect. Eur Urol 2015; 68:1023-30. [DOI: 10.1016/j.eururo.2015.05.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
|
139
|
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: 1318] [Impact Index Per Article: 146.4] [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
|
140
|
Busada JT, Geyer CB. The Role of Retinoic Acid (RA) in Spermatogonial Differentiation. Biol Reprod 2015; 94:10. [PMID: 26559678 PMCID: PMC4809555 DOI: 10.1095/biolreprod.115.135145] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/06/2015] [Indexed: 12/22/2022] Open
Abstract
Retinoic acid (RA) directs the sequential, but distinct, programs of spermatogonial differentiation and meiotic differentiation that are both essential for the generation of functional spermatozoa. These processes are functionally and temporally decoupled, as they occur in distinct cell types that arise over a week apart, both in the neonatal and adult testis. However, our understanding is limited in terms of what cellular and molecular changes occur downstream of RA exposure that prepare differentiating spermatogonia for meiotic initiation. In this review, we describe the process of spermatogonial differentiation and summarize the current state of knowledge regarding RA signaling in spermatogonia.
Collapse
Affiliation(s)
- Jonathan T Busada
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
141
|
Wang Y, Liu W, Yang Q, Yu M, Zhang Z. Di (2-ethylhexyl) phthalate exposure during pregnancy disturbs temporal sex determination regulation in mice offspring. Toxicology 2015. [DOI: 10.1016/j.tox.2015.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
142
|
Howdeshell KL, Rider CV, Wilson VS, Furr JR, Lambright CR, Gray LE. Dose Addition Models Based on Biologically Relevant Reductions in Fetal Testosterone Accurately Predict Postnatal Reproductive Tract Alterations by a Phthalate Mixture in Rats. Toxicol Sci 2015; 148:488-502. [PMID: 26350170 DOI: 10.1093/toxsci/kfv196] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Challenges in cumulative risk assessment of anti-androgenic phthalate mixtures include a lack of data on all the individual phthalates and difficulty determining the biological relevance of reduction in fetal testosterone (T) on postnatal development. The objectives of the current study were 2-fold: (1) to test whether a mixture model of dose addition based on the fetal T production data of individual phthalates would predict the effects of a 5 phthalate mixture on androgen-sensitive postnatal male reproductive tract development, and (2) to determine the biological relevance of the reductions in fetal T to induce abnormal postnatal reproductive tract development using data from the mixture study. We administered a dose range of the mixture (60, 40, 20, 10, and 5% of the top dose used in the previous fetal T production study consisting of 300 mg/kg per chemical of benzyl butyl (BBP), di(n)butyl (DBP), diethyl hexyl phthalate (DEHP), di-isobutyl phthalate (DiBP), and 100 mg dipentyl (DPP) phthalate/kg; the individual phthalates were present in equipotent doses based on their ability to reduce fetal T production) via gavage to Sprague Dawley rat dams on GD8-postnatal day 3. We compared observed mixture responses to predictions of dose addition based on the previously published potencies of the individual phthalates to reduce fetal T production relative to a reference chemical and published postnatal data for the reference chemical (called DAref). In addition, we predicted DA (called DAall) and response addition (RA) based on logistic regression analysis of all 5 individual phthalates when complete data were available. DA ref and DA all accurately predicted the observed mixture effect for 11 of 14 endpoints. Furthermore, reproductive tract malformations were seen in 17-100% of F1 males when fetal T production was reduced by about 25-72%, respectively.
Collapse
Affiliation(s)
- Kembra L Howdeshell
- *Division of the National Toxicology Program (NTP), National Institute of Environmental Health Sciences (NIEHS), PO Box 12233, Research Triangle Park (RTP), North Carolina 27709 and
| | - Cynthia V Rider
- *Division of the National Toxicology Program (NTP), National Institute of Environmental Health Sciences (NIEHS), PO Box 12233, Research Triangle Park (RTP), North Carolina 27709 and
| | - Vickie S Wilson
- Reproductive Toxicology Branch, Toxicology Assessment Division (TAD), National Health and Environmental Effects Research Laboratories, Office of Research and Development, US Environmental Protection Agency (US EPA), RTP, North Carolina 27711
| | - Johnathan R Furr
- Reproductive Toxicology Branch, Toxicology Assessment Division (TAD), National Health and Environmental Effects Research Laboratories, Office of Research and Development, US Environmental Protection Agency (US EPA), RTP, North Carolina 27711
| | - Christy R Lambright
- Reproductive Toxicology Branch, Toxicology Assessment Division (TAD), National Health and Environmental Effects Research Laboratories, Office of Research and Development, US Environmental Protection Agency (US EPA), RTP, North Carolina 27711
| | - L Earl Gray
- Reproductive Toxicology Branch, Toxicology Assessment Division (TAD), National Health and Environmental Effects Research Laboratories, Office of Research and Development, US Environmental Protection Agency (US EPA), RTP, North Carolina 27711
| |
Collapse
|
143
|
Axelsson J, Rylander L, Rignell-Hydbom A, Lindh CH, Jönsson BAG, Giwercman A. Prenatal phthalate exposure and reproductive function in young men. ENVIRONMENTAL RESEARCH 2015; 138:264-70. [PMID: 25743932 DOI: 10.1016/j.envres.2015.02.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 05/21/2023]
Abstract
BACKGROUND Prenatal exposure to phthalates is suggested to negatively impact male reproductive function, but human data are lacking. OBJECTIVES To study associations between prenatal exposure to diethylhexyl phthalate (DEHP) and diisononyl phthalate (DiNP), and reproductive parameters of adolescent men. METHODS Using linear regression models adjusted for potential confounders, we studied associations between levels of DEHP- and DiNP metabolites in maternal sera from mean 12 weeks of pregnancy, and testicular size, semen quality and reproductive hormones in 112 adolescent sons, recruited from the general population. RESULTS Men in the highest exposure tertile of one DiNP metabolite [mono-(carboxy-iso-octyl) phthalate], compared with men in the lowest tertile had: 4.3mL (95% CI: 0.89, 7.6mL; p<0.001) lower total testicular volume; 30% (95% CI: 3.6, 63%; p=0.02) higher levels of follicle-stimulating hormone; and 0.87mL (95% CI: 0.28, 1.5mL; p=0.004) lower semen volume. Men in the highest exposure tertile of one DEHP metabolite [mono-(2-ethyl-5-hydroxylhexyl) phthalate] had 0.70mL (95% CI: 0.090, 1.3mL; p=0.03) lower semen volume than men in the lowest exposure tertile. The levels of two DiNP metabolites [mono-(hydroxy-iso-nonyl) phthalate and mono-(oxo-iso-nonyl) phthalate] were linearly associated with luteinizing hormone (p<0.01). CONCLUSION Prenatal levels of some metabolites of DEHP and DiNP seemed negatively associated with reproductive function of adolescent men.
Collapse
Affiliation(s)
- Jonatan Axelsson
- Molecular Reproductive Medicine, Skåne University Hospital Malmö, Lund University, 205 02 Malmö, Sweden.
| | - Lars Rylander
- Division of Occupational and Environmental Medicine, Lund University, 221 85 Lund, Sweden
| | - Anna Rignell-Hydbom
- Division of Occupational and Environmental Medicine, Lund University, 221 85 Lund, Sweden
| | - Christian H Lindh
- Division of Occupational and Environmental Medicine, Lund University, 221 85 Lund, Sweden
| | - Bo A G Jönsson
- Division of Occupational and Environmental Medicine, Lund University, 221 85 Lund, Sweden
| | - Aleksander Giwercman
- Molecular Reproductive Medicine, Skåne University Hospital Malmö, Lund University, 205 02 Malmö, Sweden
| |
Collapse
|
144
|
van den Driesche S, McKinnell C, Calarrão A, Kennedy L, Hutchison GR, Hrabalkova L, Jobling MS, Macpherson S, Anderson RA, Sharpe RM, Mitchell RT. Comparative effects of di(n-butyl) phthalate exposure on fetal germ cell development in the rat and in human fetal testis xenografts. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:223-30. [PMID: 25514601 PMCID: PMC4348744 DOI: 10.1289/ehp.1408248] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 12/12/2014] [Indexed: 05/02/2023]
Abstract
BACKGROUND Phthalate exposure induces germ cell effects in the fetal rat testis. Although experimental models have shown that the human fetal testis is insensitive to the steroidogenic effects of phthalates, the effects on germ cells have been less explored. OBJECTIVES We sought to identify the effects of phthalate exposure on human fetal germ cells in a dynamic model and to establish whether the rat is an appropriate model for investigating such effects. METHODS We used immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction to examine Sertoli and germ cell markers on rat testes and human fetal testis xenografts after exposure to vehicle or di(n-butyl) phthalate (DBP). Our study included analysis of germ cell differentiation markers, proliferation markers, and cell adhesion proteins. RESULTS In both rat and human fetal testes, DBP exposure induced similar germ cell effects, namely, germ cell loss (predominantly undifferentiated), induction of multinucleated gonocytes (MNGs), and aggregation of differentiated germ cells, although the latter occurred rarely in the human testes. The mechanism for germ cell aggregation and MNG induction appears to be loss of Sertoli cell-germ cell membrane adhesion, probably due to Sertoli cell microfilament redistribution. CONCLUSIONS Our findings provide the first comparison of DBP effects on germ cell number, differentiation, and aggregation in human testis xenografts and in vivo in rats. We observed comparable effects on germ cells in both species, but the effects in the human were muted compared with those in the rat. Nevertheless, phthalate effects on germ cells have potential implications for the next generation, which merits further study. Our results indicate that the rat is a human-relevant model in which to explore the mechanisms for germ cell effects.
Collapse
Affiliation(s)
- Sander van den Driesche
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Boeira SP, Funck VR, Borges Filho C, Del'Fabbro L, de Gomes MG, Donato F, Royes LFF, Oliveira MS, Jesse CR, Furian AF. Lycopene protects against acute zearalenone-induced oxidative, endocrine, inflammatory and reproductive damages in male mice. Chem Biol Interact 2015; 230:50-7. [PMID: 25682699 DOI: 10.1016/j.cbi.2015.02.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/20/2015] [Accepted: 02/05/2015] [Indexed: 10/24/2022]
Abstract
Male mice received lycopene for 10 days before a single oral administration of zearalenone (ZEA). After 48 h testes and blood were collected. Mice treated with lycopene/ZEA exhibited amelioration of the hematological changes. Lycopene prevented the reduction in the number and motility of spermatozoa and testosterone levels, indicating a protective effect in the testicular damage induced by ZEA. Lycopene was also effective in protecting against the decrease in glutathione-S-transferase, glutathione peroxidase, glutathione reductase and δ-aminolevulinic acid dehydratase activities caused by ZEA in the testes. Exposure of animals to ZEA induced modification of antioxidant and inflammatory status with increase of reduced glutathione (GSH) levels and increase of the oxidized glutathione, interleukins 1β, 2, 6, 10, tumor necrosis factor-α and bilirubin levels. Lycopene prevented ZEA-induced changes in GSH levels and inhibited the processes of inflammation, reducing the damage induced by ZEA. Altogether, our results indicate that lycopene was able to prevent ZEA-induced damage in the mice.
Collapse
Affiliation(s)
- Silvana Peterini Boeira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil; Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Vinícius Rafael Funck
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Carlos Borges Filho
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, 97500-970 Uruguaiana, RS, Brazil
| | - Lucian Del'Fabbro
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, 97500-970 Uruguaiana, RS, Brazil
| | - Marcelo Gomes de Gomes
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, 97500-970 Uruguaiana, RS, Brazil
| | - Franciele Donato
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, 97500-970 Uruguaiana, RS, Brazil
| | - Luiz Fernando Freire Royes
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Cristiano Ricardo Jesse
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas (LaftamBio Pampa), Universidade Federal do Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, 97500-970 Uruguaiana, RS, Brazil
| | - Ana Flávia Furian
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil; Programa de Pós-Graduação em Ciência e Tecnologia de Alimentos, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
146
|
Stukenborg JB, Kjartansdóttir KR, Reda A, Colon E, Albersmeier JP, Söder O. Male germ cell development in humans. Horm Res Paediatr 2015; 81:2-12. [PMID: 24356336 DOI: 10.1159/000355599] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/12/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Germ cells are unique cells that possess the ability to transmit genetic information between generations. Detailed knowledge about the molecular and cellular mechanisms determining the fate of human male germ cells still remains sparse. This is partially due to ethical issues limiting the access to research material. Therefore, the mechanisms of proliferation, differentiation and apoptosis of human male germ cells still remain challenging study objectives. METHODS This review focuses on using English articles accessible in PubMed as well as personal files on the current knowledge of the molecular and cellular mechanisms connected with human testicular germ cell development, maturation failure and the possibility of fertility preservation in patients in whom there is a risk of gonadal failure. However, since rodents, particularly mice, offer the possibility of studying germ cell development by use of genetic modification techniques, some studies using animal models are also discussed. CONCLUSION This mini review focuses on the current knowledge about male germ cells. However, the reader is referred to two previous mini reviews focusing on testicular somatic cells, i.e. on Sertoli cells and Leydig cells.
Collapse
Affiliation(s)
- Jan-Bernd Stukenborg
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
147
|
Neufeld K, Ezell K, Grow WA. Plastic Additives Decrease Agrin-Induced Acetylcholine Receptor Clusters and Myotube Formation in C2C12 Skeletal Muscle Cell Culture. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/cellbio.2015.41002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
148
|
Rouiller-Fabre V, Guerquin MJ, N’Tumba-Byn T, Muczynski V, Moison D, Tourpin S, Messiaen S, Habert R, Livera G. Nuclear receptors and endocrine disruptors in fetal and neonatal testes: a gapped landscape. Front Endocrinol (Lausanne) 2015; 6:58. [PMID: 25999913 PMCID: PMC4423451 DOI: 10.3389/fendo.2015.00058] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/07/2015] [Indexed: 11/28/2022] Open
Abstract
During the last decades, many studies reported that male reproductive disorders are increasing among humans. It is currently acknowledged that these abnormalities can result from fetal exposure to environmental chemicals that are progressively becoming more concentrated and widespread in our environment. Among the chemicals present in the environment (air, water, food, and many consumer products), several can act as endocrine disrupting compounds (EDCs), thus interfering with the endocrine system. Phthalates, bisphenol A (BPA), and diethylstilbestrol (DES) have been largely incriminated, particularly during the fetal and neonatal period, due to their estrogenic and/or anti-androgenic properties. Indeed, many epidemiological and experimental studies have highlighted their deleterious impact on fetal and neonatal testis development. As EDCs can affect many different genomic and non-genomic pathways, the mechanisms underlying the adverse effects of EDC exposure are difficult to elucidate. Using literature data and results from our laboratory, in the present review, we discuss the role of classical nuclear receptors (genomic pathway) in the fetal and neonatal testis response to EDC exposure, particularly to phthalates, BPA, and DES. Among the nuclear receptors, we focused on some of the most likely candidates, such as peroxisome-proliferator activated receptor (PPAR), androgen receptor (AR), estrogen receptors (ERα and β), liver X receptors (LXR), and small heterodimer partner (SHP). First, we describe the expression and potential functions (based on data from studies using receptor agonists and mouse knockout models) of these nuclear receptors in the developing testis. Then, for each EDC studied, we summarize the main evidences indicating that the reprotoxic effect of each EDC under study is mediated through a specific nuclear receptor(s). We also point-out the involvement of other receptors and nuclear receptor-independent pathways.
Collapse
Affiliation(s)
- Virginie Rouiller-Fabre
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
- *Correspondence: Virginie Rouiller-Fabre, Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, CEA, DSV, iRCM, SCSR, LDG, BP6, Fontenay aux Roses F-92265, France,
| | - Marie Justine Guerquin
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Thierry N’Tumba-Byn
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Vincent Muczynski
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Delphine Moison
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Sophie Tourpin
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Sébastien Messiaen
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - René Habert
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Gabriel Livera
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| |
Collapse
|
149
|
Mitchell RT, Mungall W, McKinnell C, Sharpe RM, Cruickshanks L, Milne L, Smith LB. Anogenital distance plasticity in adulthood: implications for its use as a biomarker of fetal androgen action. Endocrinology 2015; 156:24-31. [PMID: 25375036 PMCID: PMC4272396 DOI: 10.1210/en.2014-1534] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Androgen action during the fetal masculinization programming window (MPW) determines the maximum potential for growth of androgen-dependent organs (eg, seminal vesicles, prostate, penis, and perineum) and is reflected in anogenital distance (AGD). As such, determining AGD in postnatal life has potential as a lifelong easily accessible biomarker of overall androgen action during the MPW. However, whether the perineum remains androgen responsive in adulthood and thus responds plastically to perturbed androgen drive remains unexplored. To determine this, we treated adult male rats with either the antiandrogen flutamide or the estrogen diethylstilbestrol (DES) for 5 weeks, followed by a 4-week washout period of no treatment. We determined AGD and its correlate anogenital index (AGI) (AGD relative to body weight) at weekly intervals across this period and compared these with normal adult rats (male and female), castrated male rats, and appropriate vehicle controls. These data showed that, in addition to reducing circulating testosterone and seminal vesicle weight, castration significantly reduced AGD (by ∼17%), demonstrating that there is a degree of plasticity in AGD in adulthood. Flutamide treatment increased circulating testosterone yet also reduced seminal vesicle weight due to local antagonism of androgen receptor. Despite this suppression, surprisingly, flutamide treatment had no effect on AGD at any time point. In contrast, although DES treatment suppressed circulating testosterone and reduced seminal vesicle weight, it also induced a significant reduction in AGD (by ∼11%), which returned to normal 1 week after cessation of DES treatment. We conclude that AGD in adult rats exhibits a degree of plasticity, which may be mediated by modulation of local androgen/estrogen action. The implications of these findings regarding the use of AGD as a lifelong clinical biomarker of fetal androgen action are discussed.
Collapse
Affiliation(s)
- Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
150
|
Spade DJ, McDonnell EV, Heger NE, Sanders JA, Saffarini CM, Gruppuso PA, De Paepe ME, Boekelheide K. Xenotransplantation models to study the effects of toxicants on human fetal tissues. ACTA ACUST UNITED AC 2014; 101:410-22. [PMID: 25477288 DOI: 10.1002/bdrb.21131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/14/2014] [Indexed: 12/11/2022]
Abstract
Many diseases that manifest throughout the lifetime are influenced by factors affecting fetal development. Fetal exposure to xenobiotics, in particular, may influence the development of adult diseases. Established animal models provide systems for characterizing both developmental biology and developmental toxicology. However, animal model systems do not allow researchers to assess the mechanistic effects of toxicants on developing human tissue. Human fetal tissue xenotransplantation models have recently been implemented to provide human-relevant mechanistic data on the many tissue-level functions that may be affected by fetal exposure to toxicants. This review describes the development of human fetal tissue xenotransplant models for testis, prostate, lung, liver, and adipose tissue, aimed at studying the effects of xenobiotics on tissue development, including implications for testicular dysgenesis, prostate disease, lung disease, and metabolic syndrome. The mechanistic data obtained from these models can complement data from epidemiology, traditional animal models, and in vitro studies to quantify the risks of toxicant exposures during human development.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | | | | | | | | | | | | | | |
Collapse
|