1
|
Baalbaki G, Lim V, Gillet AP, Verner MA, Vaillancourt C, Caron-Beaudoin E, Delbes G. Trace elements alone or in mixtures associated with unconventional natural gas exploitation affect rat fetal steroidogenesis and testicular development in vitro. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 357:124393. [PMID: 38901820 DOI: 10.1016/j.envpol.2024.124393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Biomonitoring studies have shown that pregnant women living in regions of unconventional natural gas (UNG) exploitation have higher levels of trace elements. Whether developmental endocrine disruption can be expected at these exposure levels during pregnancy is unclear. In this study, we aimed to test the impact of five trace elements alone or in mixtures using in vitro cell- and tissue-based assays relevant to endocrine disruption and development. Manganese, aluminum, strontium, barium, and cobalt were tested at concentrations including those representatives of human fetal exposure. Using transactivation assays, none of the tested elements nor their mixture altered the human estrogen receptor 1 or androgen receptor genomic signalling. In the rat fetal testis assay, an organ culture system, cobalt (5 μg/l), barium (500 μg/l) and strontium (500 μg/l) significantly increased testosterone secretion. Cobalt and strontium were associated with hyperplasia and/or hypertrophy of fetal Leydig cells. Mixing the five elements at concentrations where none had an effect individually stimulated testosterone secretion by the rat fetal testis paralleled by the significant increase of 3β-hydroxysteroid dehydrogenase protein level in comparison to the vehicle control. The mechanisms involved may be specific to the fetal testis as no effect was observed in the steroidogenic H295R cells. Our data suggest that some trace elements in mixture at concentrations representative of human fetal exposure can impact testis development and function. This study highlights the potential risk posed by UNG operations, especially for the most vulnerable populations, pregnant individuals, and their fetus.
Collapse
Affiliation(s)
- Ghida Baalbaki
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Victoria Lim
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Antoine P Gillet
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Marc-André Verner
- Université de Montréal, Department of Occupational and Environmental Health, Montreal, QC, Canada; Centre de Recherche en Santé Publique, Université de Montréal et CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montreal, QC, Canada
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada; Research Center, CIUSSS du Nord-de-l'Île-de-Montréal, Montreal, QC, Canada
| | - Elyse Caron-Beaudoin
- Department of Health and Society, University of Toronto Scarborough, Toronto, ON, Canada; Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Geraldine Delbes
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada.
| |
Collapse
|
2
|
Erradhouani C, Bortoli S, Aït‐Aïssa S, Coumoul X, Brion F. Metabolic disrupting chemicals in the intestine: the need for biologically relevant models: Zebrafish: what can we learn from this small environment-sensitive fish? FEBS Open Bio 2024; 14:1397-1419. [PMID: 39218795 PMCID: PMC11492336 DOI: 10.1002/2211-5463.13878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Although the concept of endocrine disruptors first appeared almost 30 years ago, the relatively recent involvement of these substances in the etiology of metabolic pathologies (obesity, diabetes, hepatic steatosis, etc.) has given rise to the concept of Metabolic Disrupting Chemicals (MDCs). Organs such as the liver and adipose tissue have been well studied in the context of metabolic disruption by these substances. The intestine, however, has been relatively unexplored despite its close link with these organs. In vivo models are useful for the study of the effects of MDCs in the intestine and, in addition, allow investigations into interactions with the rest of the organism. In the latter respect, the zebrafish is an animal model which is used increasingly for the characterization of endocrine disruptors and its use as a model for assessing effects on the intestine will, no doubt, expand. This review aims to highlight the importance of the intestine in metabolism and present the zebrafish as a relevant alternative model for investigating the effect of pollutants in the intestine by focusing, in particular, on cytochrome P450 3A (CYP3A), one of the major molecular players in endogenous and MDCs metabolism in the gut.
Collapse
Affiliation(s)
- Chedi Erradhouani
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
- Université Paris CitéFrance
- Inserm UMR‐S 1124ParisFrance
| | | | - Selim Aït‐Aïssa
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
| | | | - François Brion
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
| |
Collapse
|
3
|
Sriram S, Macedo T, Mavinkurve‐Groothuis A, van de Wetering M, Looijenga LHJ. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front. Clin Transl Sci 2024; 17:e13866. [PMID: 38965809 PMCID: PMC11224131 DOI: 10.1111/cts.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure.
Collapse
Affiliation(s)
- Sruthi Sriram
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Tiago Macedo
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | |
Collapse
|
4
|
Li Y, Li P, Tao Q, Abuqeis IJA, Xiyang Y. Role and limitation of cell therapy in treating neurological diseases. IBRAIN 2024; 10:93-105. [PMID: 38682022 PMCID: PMC11045202 DOI: 10.1002/ibra.12152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 05/01/2024]
Abstract
The central role of the brain in governing systemic functions within human physiology underscores its paramount significance as the focal point of physiological regulation. The brain, a highly sophisticated organ, orchestrates a diverse array of physiological processes encompassing motor control, sensory perception, cognition, emotion, and the regulation of vital functions, such as heartbeat, respiration, and hormonal equilibrium. A notable attribute of neurological diseases manifests as the depletion of neurons and the occurrence of tissue necrosis subsequent to injury. The transplantation of neural stem cells (NSCs) into the brain exhibits the potential for the replacement of lost neurons and the reconstruction of neural circuits. Furthermore, the transplantation of other types of cells in alternative locations can secrete nutritional factors that indirectly contribute to the restoration of nervous system equilibrium and the mitigation of neural inflammation. This review summarized a comprehensive investigation into the role of NSCs, hematopoietic stem cells, mesenchymal stem cells, and support cells like astrocytes and microglia in alleviating neurological deficits after cell infusion. Moreover, a thorough assessment was undertaken to discuss extant constraints in cellular transplantation therapies, concurrently delineating indispensable model-based methodologies, specifically on organoids, which were essential for guiding prospective research initiatives in this specialized field.
Collapse
Affiliation(s)
- Yu‐Qi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Peng‐Fei Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Qian Tao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | | | - Yan‐Bin Xiyang
- School of Basic MedicineKunming Medical UniversityKunmingChina
- Department of Pharmacology and Toxicology, College of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
5
|
Yu Y, Ren Z, Wang H, Sang J, Chen Y, Zhang M, Zhu Y, Wang Y, Ge RS. Benzene ring bisphenol A substitutes potently inhibit human, rat, and mouse gonadal 3β-hydroxysteroid dehydrogenases: Structure-activity relationship and in silico docking analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115461. [PMID: 37703809 DOI: 10.1016/j.ecoenv.2023.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023]
Abstract
Bisphenol A (BPA) is a chemical used in the production of certain plastics and resins. Recent research has found that BPA can inhibit the activity of 3β-hydroxysteroid dehydrogenase/Δ5,4-isomerases (3β-HSDs). Whether benzene ring BPA substitutes can inhibit human, rat, and mouse gonadal 3β-HSDs, the structure-activity relationship and the underlying mechanism remain unclear. In this study, we compared 6 benzene ring BPA substitutes to BPA in the inhibition of human, rat, and mouse gonadal 3β-HSDs and conducted structure-activity relationship and in silico docking analysis. The inhibitory activity (IC50) of human 3β-HSD2 in KGN cells ranged from about 0.02 μM for bisphenol H to 8.75 μM for BPA, that of rat 3β-HSD1 in testicular microsomes ranged from 0.099 μM for bisphenol H to 31.32 μM for BPA, and that of mouse 3β-HSD6 ranged from 0.021 μM for BPH to ineffectiveness for 100 μM BPA. These compounds acted as mixed inhibitors with LogP inversely correlated with IC50 and ΔG positively correlated with IC50 value. Docking analysis showed that these compounds bind to the steroid active site of the 3β-HSD enzymes. In conclusion, some benzene ring BPA substitutes potently inhibit gonadal 3β-HSD in various species, and lipophilicity and binding affinity determine their inhibitory strength.
Collapse
Affiliation(s)
- Yang Yu
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zheyuan Ren
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hong Wang
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianmin Sang
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ya Chen
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Reproductive Medicine Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minjie Zhang
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yang Zhu
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiyan Wang
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ren-Shan Ge
- Department of Anaesthesiology of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Reproductive Medicine Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, and Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325000, Zhejiang Province, China.
| |
Collapse
|
6
|
Tardif S, Rwigemera A, Letourneau N, Robaire B, Delbes G. Reproductive toxicity of emerging plasticizers, flame retardants, and bisphenols, using culture of the rat fetal testis†. Biol Reprod 2023; 108:837-848. [PMID: 36780129 PMCID: PMC10183361 DOI: 10.1093/biolre/ioad018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/10/2023] [Accepted: 02/01/2023] [Indexed: 02/14/2023] Open
Abstract
The use of bis (2-ethylhexyl) phthalate (DEHP), 2,2'4,4'-tetrabromodiphenyl ether (BDE47), and bisphenol A (BPA), as plasticizers, flame retardants, and epoxy resins, respectively, has been regulated due to their endocrine disrupting activities. Replacements for these chemicals are found in human matrices, yet the endocrine disrupting potential of these emerging contaminants is poorly characterized. We compared the effects of legacy chemicals with those of their replacements using fetal rat testis organ culture. Fetal testes sampled at gestation day 15 were grown ex vivo, and the impact was evaluated after a 3-day exposure to 10 μM of each legacy chemical; two BPA analogs (bisphenol M and bisphenol TMC); three replacements for DEHP/MEHP (2,2,4-trimethyl-1,3-pentanediol diisobutyrate, diisononyl-phthalate, and diisodecyl adipate); or two replacements for BDE47 (tributoxyethyl phosphate and isopropylated triphenyl phosphate). We showed that only BPA and MEHP significantly decrease testosterone secretions after 24 h, while BPM and BPTMC have the opposite effect. Luteinizing hormone-stimulated testosterone was reduced by BPA and MEHP but was increased by BPTMC. After exposure, testes were used for immunofluorescent staining of germ cells, Sertoli cells, and Leydig cells. Interestingly, exposures to BPM or BPTMC induced a significant increase in the Leydig cell density and surface area. A decrease in germ cell density was observed only after treatment with MEHP or BDE47. MEHP also significantly decreased Sertoli cell proliferation. These studies show that some replacement chemicals can affect testicular function, while others appear to show little toxicity in this model. These findings provide essential information regarding the need for their regulation.
Collapse
Affiliation(s)
- Sarah Tardif
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Arlette Rwigemera
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Natasha Letourneau
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| | - Bernard Robaire
- Department of Pharmacology and Therapeutic, McGill University, Montreal, Quebec, Canada
- Department of Obstetrics & Gynecology, McGill University, Montreal, Quebec, Canada
| | - Geraldine Delbes
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Quebec, Canada
| |
Collapse
|
7
|
Astuto MC, Benford D, Bodin L, Cattaneo I, Halldorsson T, Schlatter J, Sharpe RM, Tarazona J, Younes M. Applying the adverse outcome pathway concept for assessing non-monotonic dose responses: biphasic effect of bis(2-ethylhexyl) phthalate (DEHP) on testosterone levels. Arch Toxicol 2023; 97:313-327. [PMID: 36336711 DOI: 10.1007/s00204-022-03409-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
Male reproduction is one of the primary health endpoints identified in rodent studies for some phthalates, such as DEHP (Bis(2-ethylhexyl) phthalate), DBP (Dibutyl phthalate), and BBP (Benzyl butyl phthalate). The reduction in testosterone level was used as an intermediate key event for grouping some phthalates and to establish a reference point for risk assessment. Phthalates, and specifically DEHP, are one of the chemicals for which the greatest number of non-monotonic dose responses (NMDRs) are observed. These NMDRs cover different endpoints and situations, often including testosterone levels. The presence of NMDR has been the subject of some debate within the area of chemical risk assessment, which is traditionally anchored around driving health-based guidance values for apical endpoints that typically follow a clear monotonic dose-response. The consequence of NMDR for chemical risk assessment has recently received considerable attention amongst regulatory agencies, which confirmed its relevance particularly for receptor-mediated effects. The present review explores the relationship between DEHP exposure and testosterone levels, investigating the biological plausibility of the observed NMDRs. The Adverse Outcome Pathway (AOP) concept is applied to integrate NMDRs into Key Event Relationships (KERs) for exploring a mechanistic understanding of initial key events and possibly associated reproductive and non-reproductive adverse outcomes.
Collapse
Affiliation(s)
- M C Astuto
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy.
| | - D Benford
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - L Bodin
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - I Cattaneo
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - T Halldorsson
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy.,Faculty of Food Science and Nutrition, University of Iceland, Reykjavik, Iceland
| | - J Schlatter
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - R M Sharpe
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - J Tarazona
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| | - M Younes
- European Food Safety Authority, Methodology and Scientific Support Unit and Working Group on Non-Monotonic Dose Responses, Parma, Italy
| |
Collapse
|
8
|
Shepherd A, Brunckhorst O, Ahmed K, Xu Q. Botanicals in health and disease of the testis and male fertility: A scoping review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154398. [PMID: 36049429 DOI: 10.1016/j.phymed.2022.154398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 07/18/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Male factor infertility often results from testicular disorders leading to inadequate sperm quantity and quality. Both beneficial and detrimental effects of botanical products, especially herbal medicines, on testicular functions and male fertility have been reported in the literature. PURPOSE This scoping review aims to map the main clinical evidence on different impacts of botanical entities on the testis and to critically appraise relevant randomized controlled trials (RCTs) published in the recent 5 years, so as to inform the future. METHODS Systematic reviews, meta-analyses and RCT reports on botanical impacts on testicular functions and male fertility were retrieved and synthesized from Pubmed, Web of Science, Scopus, Embase, ProQuest, Cochrane Library and Google Scholar up to 10th May 2022. RCTs published since 2018 were critically appraised against good practice guidelines for RCT and for reporting herbal studies. RESULTS We identified 24 systematic reviews and meta-analyses published since 2005, by authors from Iran (25%), China (21%), USA (12.5%) and 9 other countries. All but two were published in English. Only 3 systematic review protocols were identified, all published in English from China in the recent 3 years. We identified 125 RCTs published in six languages, mainly English (55%) and Chinese (42%). They were published since 1994 from 23 countries on all the six inhabitable continents, with China (46%), Australia (8%), USA (8%), India (7%) and Iran (5%) being the leading contributors. 72% and 28% RCTs published in English were on efficacy (botanicals vs placebo) and comparative effectiveness (a botanical vs other treatments), respectively. In contrast, 98% RCT reports in Chinese were on comparative effectiveness, with merely 2% on efficacy. Among all the 125 RCTs, 57% were studies in patients with semen abnormality and/or male infertility, 22% investigated herbal effects in healthy men, 14% were on patients with male sexual dysfunction and hypogonadism, and 7% were conducted in men with non-sexual disorders. Since 2018, 32 RCTs have been published, in English (69%) or Chinese (31%). Nineteen RCT reports from China, India, Japan and Korea all studied herbal formulae while the 13 RCT reports from Australia, Brazil, Czech and Italy, Iran, Malaysia, Spain, the UK and the USA all exclusively studied extracts of a single species. Putting geo-cultural differences aside, gossypol and extracts of Tripterygium wilfordii Hook. f. were found to be detrimental to the testis and male fertility, while the extracts of Withania somnifera (L.) Dunal and traditional Chinese medicine Qilin Pill, etc., might improve testosterone levels and semen parameters, thus could be therapeutic for male sexual dysfunction and infertility. However, all still require further evaluation in view of recurring weaknesses in quality control of herbal materials, RCT design and reporting. For example, only 9%-23% of the RCTs published since 2018 provided information on voucher samples, chemical profiling, herbal authentication and herbal extraction. CONCLUSION Research on botanicals and the testis has been reported worldwide, demonstrating clear geo-cultural differences in studied plant species, botanical types, study objectives and quality of research design, implementation and reporting. Due to a few recurring weaknesses in the literature, this study is unable to recommend the use of any specific botanicals, however, current evidence does indicate that botanicals can be double-edged swords to the testis and male fertility. To secure better clinical evidence, future studies must faithfully implement existing and emerging good practice guidelines.
Collapse
Affiliation(s)
- Adam Shepherd
- GKT School of Medical Education, King's College London, London, United Kingdom
| | - Oliver Brunckhorst
- MRC Centre for Transplantation, Guy's Hospital Campus, King's College London, King's Health Partners, London, United Kingdom
| | - Kamran Ahmed
- MRC Centre for Transplantation, Guy's Hospital Campus, King's College London, King's Health Partners, London, United Kingdom; Department of Urology, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates; Department of Epidemiology and Public Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Qihe Xu
- Renal Sciences and Integrative Chinese Medicine Laboratory, Department of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.
| |
Collapse
|
9
|
Ma Y, Chen J, Li H, Xu F, Chong T, Wang Z, Zhang L. Immature rat testis sustained long-term development using an integrative model. Biol Res 2022; 55:30. [PMID: 36195947 PMCID: PMC9531454 DOI: 10.1186/s40659-022-00398-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Xenotransplantation has been primarily performed using fresh donor tissue to study testicular development for about 20 years, and whether the cultured tissue would be a suitable donor is unclear. In this study, we combined testicular culture and xenotransplantation into an integrative model and explored whether immature testicular tissue would survive and continue to develop in this model. METHODS In the new integrative model group, the testes of neonatal rats on postnatal day 8 (PND 8) were cultured for 4 days ex vivo and then were transplanted under the dorsal skin of castrated nude mice. The xenografted testes were resected on the 57th day after xenotransplantation and the testes of rats in the control group were harvested on PND 69. The survival state of testicular tissue was evaluated from morphological and functional perspectives including H&E staining, immunohistochemical staining of 8-OH-dG, immunofluorescence staining, TUNEL assay, ultrastructural study, gene expression and protein analysis. RESULTS (a) We found that complete spermatogenesis was established in the testes in the new integrative model group. Compared with the control in the same stage, the seminiferous epithelium in some tubules was a bit thinner and there were vacuoles in part of the tubules. Immunofluorescence staining revealed some ACROSIN-positive spermatids were present in seminiferous tubule of xenografted testes. TUNEL detection showed apoptotic cells and most of them were germ cells in the new integrative model group. 8-OH-dG immunohistochemistry showed strongly positive-stained in the seminiferous epithelium after xenotransplantation in comparison with the control group; (b) Compared with the control group, the expressions of FOXA3, DAZL, GFRα1, BOLL, SYCP3, CDC25A, LDHC, CREM and MKI67 in the new integrative model group were significantly elevated (P < 0.05), indicating that the testicular tissue was in an active differentiated and proliferative state; (c) Antioxidant gene detection showed that the expression of Nrf2, Keap1, NQO1 and SOD1 in the new integrative model group was significantly higher than those in the control group (P < 0.05), and DNA methyltransferase gene detection showed that the expression of DNMT3B was significantly elevated as well (P < 0.05). CONCLUSION The new integrative model could maintain the viability of immature testicular tissue and sustain the long-term survival in vivo with complete spermatogenesis. However, testicular genes expression was altered, vacuolation and thin seminiferous epithelium were still apparent in this model, manifesting that oxidative damage may contribute to the testicular development lesion and it needs further study in order to optimize this model.
Collapse
Affiliation(s)
- Yubo Ma
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China
| | - Juan Chen
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China
| | - Hecheng Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China
| | - Fangshi Xu
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China
| | - Ziming Wang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China.
| | - Liandong Zhang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, China.
| |
Collapse
|
10
|
Zeng JY, Chen PP, Liu C, Deng YL, Miao Y, Zhang M, Cui FP, Lu TT, Shi T, Yang KD, Liu CJ, Zeng Q. Bisphenol A analogues in associations with serum hormone levels among reproductive-aged Chinese men. ENVIRONMENT INTERNATIONAL 2022; 167:107446. [PMID: 35940031 DOI: 10.1016/j.envint.2022.107446] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Bisphenol A (BPA) as an endocrine disrupting chemical has been shown to alter reproductive endocrine function, but little is known on its analogues such as bisphenol F (BPF) and bisphenol S (BPS) with increasing usage and exposure. OBJECTIVE To explore the associations between exposures to BPA, BPF and BPS and serum reproductive hormones among reproductive-aged Chinese men. METHODS We measured BPA, BPF and BPS concentrations in repeated urine samples and multiple reproductive hormones in the serum samples collected from 462 men attending an infertility clinic in Wuhan, China. Linear regression models were applied to assess the associations between averaged urinary BPA, BPF and BPS levels and serum hormone concentrations, and restricted cubic spline (RCS) models were further utilized to explore potential non-linear associations. We also examined potential modifying effects by age and body mass index (BMI). RESULTS There was little evidence of associations between BPA exposure and altered reproductive hormones. However, we found that elevated BPF and BPS exposures were in negative associations with estrogen (E2) levels and E2/T (total testosterone) ratio (all P for trends < 0.05), and that elevated BPS exposure was negatively associated with SHBG levels (P for trend = 0.09). Based on the RCS models, these linear negative associations except that between BPS exposure and E2/T ratio were further confirmed. In stratified analyses, BPF and BPS exposures in relation to reduced E2 and E2/T ratio were more pronounced among men aged > 30 years, whereas their associations with reduced SHBG levels were more pronounced among men aged ≤ 30. Also, BPS exposure in negative association with FSH only emerged among men with BMI ≥ 24 kg/m2 (P for interaction = 0.03). CONCLUSION BPF and BPS exposures were negatively associated with male serum E2, E2/T ratio and SHBG levels, and these associations varied by age and BMI.
Collapse
Affiliation(s)
- Jia-Yue 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, 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-Pan Chen
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu Miao
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Min Zhang
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Fei-Peng Cui
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Ting-Ting 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Tian Shi
- 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Ke-Di 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, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chang-Jiang Liu
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, 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, 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
|
11
|
Boizet-Bonhoure B, Déjardin S, Rossitto M, Poulat F, Philibert P. Using Experimental Models to Decipher the Effects of Acetaminophen and NSAIDs on Reproductive Development and Health. FRONTIERS IN TOXICOLOGY 2022; 4:835360. [PMID: 35295217 PMCID: PMC8915900 DOI: 10.3389/ftox.2022.835360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 01/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin (acetylsalicylic acid), diclofenac and ibuprofen (IBU), and analgesic drugs, such as acetaminophen (APAP, or paracetamol), are widely used to treat inflammation and pain. APAP and IBU are over-the-counter drugs and are among the most commonly taken drugs in the first trimester of pregnancy, even in combination. Furthermore, these drugs and their metabolites are released in the environment, and can be frequently detected in wastewater, surface water, and importantly in drinking water. Although their environmental concentrations are much lower than the therapeutics doses, this suggests an uncontrolled low-dose exposure of the general population, including pregnant women and young children, two particularly at risk populations. Epidemiological studies show that exposure to these molecules in the first and second trimester of gestation can favor genital malformations in new-born boys. To investigate the cellular, molecular and mechanistic effects of exposure to these molecules, ex vivo studies with human or rodent gonadal explants and in vivo experiments in rodents have been performed in the past years. This review recapitulates recent data obtained in rodent models after in utero or postnatal exposure to these drugs. The first part of this review discusses the mechanisms by which NSAIDs and analgesics may impair gonadal development and maturation, puberty development, sex hormone production, maturation and function of adult organs, and ultimately fertility in the exposed animals and their offspring. Like other endocrine disruptors, NSAIDs and APAP interfere with endocrine gland function and may have inter/transgenerational adverse effects. Particularly, they may target germ cells, resulting in reduced quality of male and female gametes, and decreased fertility of exposed individuals and their descendants. Then, this review discusses the effects of exposure to a single drug (APAP, aspirin, or IBU) or to combinations of drugs during early embryogenesis, and the consequences on postnatal gonadal development and adult reproductive health. Altogether, these data may increase medical and public awareness about these reproductive health concerns, particularly in women of childbearing age, pregnant women, and parents of young children.
Collapse
Affiliation(s)
- Brigitte Boizet-Bonhoure
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- *Correspondence: Brigitte Boizet-Bonhoure,
| | - Stéphanie Déjardin
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | | | - Francis Poulat
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - Pascal Philibert
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- Laboratory of Biochemistry and Molecular Biology, Carèmeau Hospital, Nîmes University Hospital, Nîmes, France
| |
Collapse
|
12
|
Delbes G, Blázquez M, Fernandino JI, Grigorova P, Hales BF, Metcalfe C, Navarro-Martín L, Parent L, Robaire B, Rwigemera A, Van Der Kraak G, Wade M, Marlatt V. Effects of endocrine disrupting chemicals on gonad development: Mechanistic insights from fish and mammals. ENVIRONMENTAL RESEARCH 2022; 204:112040. [PMID: 34509487 DOI: 10.1016/j.envres.2021.112040] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 06/13/2023]
Abstract
Over the past century, evidence has emerged that endocrine disrupting chemicals (EDCs) have an impact on reproductive health. An increased frequency of reproductive disorders has been observed worldwide in both wildlife and humans that is correlated with accidental exposures to EDCs and their increased production. Epidemiological and experimental studies have highlighted the consequences of early exposures and the existence of key windows of sensitivity during development. Such early in life exposures can have an immediate impact on gonadal and reproductive tract development, as well as on long-term reproductive health in both males and females. Traditionally, EDCs were thought to exert their effects by modifying the endocrine pathways controlling reproduction. Advances in knowledge of the mechanisms regulating sex determination, differentiation and gonadal development in fish and rodents have led to a better understanding of the molecular mechanisms underlying the effects of early exposure to EDCs on reproduction. In this manuscript, we review the key developmental stages sensitive to EDCs and the state of knowledge on the mechanisms by which model EDCs affect these processes, based on the roadmap of gonad development specific to fish and mammals.
Collapse
Affiliation(s)
- G Delbes
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, Canada.
| | - M Blázquez
- Institute of Marine Sciences (ICM-CSIC), Barcelona, Spain
| | - J I Fernandino
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Argentina
| | | | - B F Hales
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - C Metcalfe
- School of Environment, Trent University, Trent, Canada
| | - L Navarro-Martín
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Barcelona, Spain
| | - L Parent
- Université TELUQ, Montréal, Canada
| | - B Robaire
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | - A Rwigemera
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, Canada
| | - G Van Der Kraak
- Department of Integrative Biology, University of Guelph, Guelph, Canada
| | - M Wade
- Environmental Health Science & Research Bureau, Health Canada, Ottawa, Canada
| | - V Marlatt
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
13
|
Vunjak-Novakovic G, Ronaldson-Bouchard K, Radisic M. Organs-on-a-chip models for biological research. Cell 2021; 184:4597-4611. [PMID: 34478657 PMCID: PMC8417425 DOI: 10.1016/j.cell.2021.08.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
We explore the utility of bioengineered human tissues-individually or connected into physiological units-for biological research. While much smaller and simpler than their native counterparts, these tissues are complex enough to approximate distinct tissue phenotypes: molecular, structural, and functional. Unlike organoids, which form spontaneously and recapitulate development, "organs-on-a-chip" are engineered to display some specific functions of whole organs. Looking back, we discuss the key developments of this emerging technology. Thinking forward, we focus on the challenges faced to fully establish, validate, and utilize the fidelity of these models for biological research.
Collapse
|
14
|
Wu H, Wang J, Xiang Y, Li L, Qie H, Ren M, Lin A, Qi F. Effects of tetrabromobisphenol A (TBBPA) on the reproductive health of male rodents: A systematic review and meta-analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 781:146745. [PMID: 33794456 DOI: 10.1016/j.scitotenv.2021.146745] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
Tetrabromobisphenol A (TBBPA) is a type of brominated flame retardant widely detected in the environment and organisms. It has been reported to cause cytotoxicity and disrupt endocrine system of animals. However, the effect of TBBPA on the reproductive system of male rodents is still controversial. Hence, this meta-analysis aims to determine whether TBBPA exposure damage to the reproductive system of male rodents. In this study, a thorough search of literatures was undertaken to select papers published before December 1st, 2020. The standard mean difference (SMD) and 95% confidence interval (CI) were calculated by random model. The results showed a statistically significant association between TBBPA exposure and the reproductive system health of male rodents (SMD = -0.35, 95% CI -0.50 to -0.19). The SMD for the reproductive system index organ weight, sperm quality, hormone levels, and gene expression were 0.03 (95% CI -0.18 to 0.23), -0.47 (95% CI -0.78 to -0.16), -0.51 (95% CI -0.75 to -0.27), and -0.98 (95% CI -1.36 to -0.60), respectively. There was a significant dose-effect relationship between TBBPA exposure and the reproductive health of male rodents, with the SMD values of low, medium, and high doses -0.20 (95% CI -0.34 to -0.05), -0.24 (95% CI -0.56 to 0.07), and -0.48 (95% CI -0.83 to -0.13), respectively. For exposure duration of TBBPA, an exposure time of >10 weeks (SMD = -0.33, 95% CI -0.54 to -0.12) showed more significant effect than an exposure time of ≤10 weeks (SMD = -0.22, 95% CI -0.43 to -0.02). Moreover, TBBPA exposure exhibited significant negative effects on sperm count (SMD = -0.49, 95% CI -0.82 to -0.17) while also reduced the content of triiodothyronine (T3), thyroxine (T4), and thyroid stimulating hormone (TSH) hormones. To summarize, our meta-analysis indicated that TBBPA had a toxicity effect to the reproductive system of male rodents.
Collapse
Affiliation(s)
- Huihui Wu
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Jinhang Wang
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Ying Xiang
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Lu Li
- Chinese Academy for Environmental Planning, Beijing 100012, PR China
| | - Hantong Qie
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Meng Ren
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Aijun Lin
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Fangjie Qi
- Global Centre for Environmental Remediation, ATC Building, Faculty of Science, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia.
| |
Collapse
|
15
|
Androgens and the masculinization programming window: human-rodent differences. Biochem Soc Trans 2021; 48:1725-1735. [PMID: 32779695 PMCID: PMC7458408 DOI: 10.1042/bst20200200] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023]
Abstract
Human male reproductive disorders are common and may have a fetal origin - the testicular dysgenesis syndrome (TDS) hypothesis. In rats, experimentally induced TDS disorders result from disruption of fetal androgen production/action specifically in the masculinization programming window (MPW). MPW androgen action also programs longer anogenital distance (AGD) in male versus female rats; shorter male AGD is correlated with risk and severity of induced TDS disorders. AGD thus provides a lifelong, calibrated readout of MPW androgen exposure and predicts likelihood of reproductive dysfunction. Pregnant rat exposure to environmental chemicals, notably certain phthalates (e.g. diethyl hexl phthalate, DEHP; dibutyl phthalate, DBP), pesticides or paracetamol, can reduce fetal testis testosterone and AGD and induce TDS disorders, provided exposure includes the MPW. In humans, AGD is longer in males than females and the presumptive MPW is 8-14 weeks' gestation. Some, but not all, epidemiological studies of maternal DEHP (or pesticides) exposure reported shorter AGD in sons, but this occurred at DEHP exposure levels several thousand-fold lower than are effective in rats. In fetal human testis culture/xenografts, DEHP/DBP do not reduce testosterone production, whereas therapeutic paracetamol exposure does. In humans, androgen production in the MPW is controlled differently (human chorionic gonadotrophin-driven) than in rats (paracrine controlled), and other organs (placenta, liver, adrenals) contribute to MPW androgens, essential for normal masculinization, via the 'backdoor pathway'. Consequently, early placental dysfunction, which is affected by maternal lifestyle and diet, and maternal painkiller use, may be more important than environmental chemical exposures in the origin of TDS in humans.
Collapse
|
16
|
Baert Y, Ruetschle I, Cools W, Oehme A, Lorenz A, Marx U, Goossens E, Maschmeyer I. A multi-organ-chip co-culture of liver and testis equivalents: a first step toward a systemic male reprotoxicity model. Hum Reprod 2021; 35:1029-1044. [PMID: 32390056 DOI: 10.1093/humrep/deaa057] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/30/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Is it possible to co-culture and functionally link human liver and testis equivalents in the combined medium circuit of a multi-organ chip? SUMMARY ANSWER Multi-organ-chip co-cultures of human liver and testis equivalents were maintained at a steady-state for at least 1 week and the co-cultures reproduced specific natural and drug-induced liver-testis systemic interactions. WHAT IS KNOWN ALREADY Current benchtop reprotoxicity models typically do not include hepatic metabolism and interactions of the liver-testis axis. However, these are important to study the biotransformation of substances. STUDY DESIGN, SIZE, DURATION Testicular organoids derived from primary adult testicular cells and liver spheroids consisting of cultured HepaRG cells and hepatic stellate cells were loaded into separate culture compartments of each multi-organ-chip circuit for co-culture in liver spheroid-specific medium, testicular organoid-specific medium or a combined medium over a week. Additional multi-organ-chips (single) and well plates (static) were loaded only with testicular organoids or liver spheroids for comparison. Subsequently, the selected type of medium was supplemented with cyclophosphamide, an alkylating anti-neoplastic prodrug that has demonstrated germ cell toxicity after its bioactivation in the liver, and added to chip-based co-cultures to replicate a human liver-testis systemic interaction in vitro. Single chip-based testicular organoids were used as a control. Experiments were performed with three biological replicates unless otherwise stated. PARTICIPANTS/MATERIALS, SETTING, METHODS The metabolic activity was determined as glucose consumption and lactate production. The cell viability was measured as lactate dehydrogenase activity in the medium. Additionally, immunohistochemical and real-time quantitative PCR end-point analyses were performed for apoptosis, proliferation and cell-specific phenotypical and functional markers. The functionality of Sertoli and Leydig cells in testicular spheroids was specifically evaluated by measuring daily inhibin B and testosterone release, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Co-culture in multi-organ chips with liver spheroid-specific medium better supported the metabolic activity of the cultured tissues compared to other media tested. The liver spheroids did not show significantly different behaviour during co-culture compared to that in single culture on multi-organ-chips. The testicular organoids also developed accordingly and produced higher inhibin B but lower testosterone levels than the static culture in plates with testicular organoid-specific medium. By comparison, testosterone secretion by testicular organoids cultured individually on multi-organ-chips reached a similar level as the static culture at Day 7. This suggests that the liver spheroids have metabolised the steroids in the co-cultures, a naturally occurring phenomenon. The addition of cyclophosphamide led to upregulation of specific cytochromes in liver spheroids and loss of germ cells in testicular organoids in the multi-organ-chip co-cultures but not in single-testis culture. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The number of biological replicates included in this study was relatively small due to the limited availability of individual donor testes and the labour-intensive nature of multi-organ-chip co-cultures. Moreover, testicular organoids and liver spheroids are miniaturised organ equivalents that capture key features, but are still simplified versions of the native tissues. Also, it should be noted that only the prodrug cyclophosphamide was administered. The final concentration of the active metabolite was not measured. WIDER IMPLICATIONS OF THE FINDINGS This co-culture model responds to the request of setting up a specific tool that enables the testing of candidate reprotoxic substances with the possibility of human biotransformation. It further allows the inclusion of other human tissue equivalents for chemical risk assessment on the systemic level. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by research grants from the Scientific Research Foundation Flanders (FWO), Universitair Ziekenhuis Brussel (scientific fund Willy Gepts) and the Vrije Universiteit Brussel. Y.B. is a postdoctoral fellow of the FWO. U.M. is founder, shareholder and CEO of TissUse GmbH, Berlin, Germany, a company commercializing the Multi-Organ-Chip platform systems used in the study. The other authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- Y Baert
- Biology of the Testis (BITE) Research Group, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - I Ruetschle
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - W Cools
- Interfaculty Center Data Processing and Statistics (ICDS), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - A Oehme
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - A Lorenz
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - U Marx
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - E Goossens
- Biology of the Testis (BITE) Research Group, Department of Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - I Maschmeyer
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| |
Collapse
|
17
|
Hess RA, Sharpe RM, Hinton BT. Estrogens and development of the rete testis, efferent ductules, epididymis and vas deferens. Differentiation 2021; 118:41-71. [PMID: 33441255 PMCID: PMC8026493 DOI: 10.1016/j.diff.2020.11.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 02/07/2023]
Abstract
Estrogen has always been considered the female hormone and testosterone the male hormone. However, estrogen's presence in the testis and deleterious effects of estrogen treatment during development have been known for nearly 90 years, long before estrogen receptors (ESRs) were discovered. Eventually it was learned that testes actually synthesize high levels of estradiol (E2) and sequester high concentrations in the reproductive tract lumen, which seems contradictory to the overwhelming number of studies showing reproductive pathology following exogenous estrogen exposures. For too long, the developmental pathology of estrogen has dominated our thinking, even resulting in the "estrogen hypothesis" as related to the testicular dysgenesis syndrome. However, these early studies and the development of an Esr1 knockout mouse led to a deluge of research into estrogen's potential role in and disruption of development and function of the male reproductive system. What is new is that estrogen action in the male cannot be divorced from that of androgen. This paper presents what is known about components of the estrogen pathway, including its synthesis and target receptors, and the need to achieve a balance between androgen- and estrogen-action in male reproductive tract differentiation and adult functions. The review focuses on what is known regarding development of the male reproductive tract, from the rete testis to the vas deferens, and examines the expression of estrogen receptors and presence of aromatase in the male reproductive system, traces the evidence provided by estrogen-associated knockout and transgenic animal models and discusses the effects of fetal and postnatal exposures to estrogens. Hopefully, there will be enough here to stimulate discussions and new investigations of the androgen:estrogen balance that seems to be essential for development of the male reproductive tract.
Collapse
Affiliation(s)
- Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, IL, 61802 USA and Epivara, Inc., Research Park, 60 Hazelwood Dr., Suite 230G, Champaign, IL, 61820, USA.
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Barry T Hinton
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
18
|
Munier M, Ayoub M, Suteau V, Gourdin L, Henrion D, Reiter E, Rodien P. In vitro effects of the endocrine disruptor p,p'DDT on human choriogonadotropin/luteinizing hormone receptor signalling. Arch Toxicol 2021; 95:1671-1681. [PMID: 33638691 DOI: 10.1007/s00204-021-03007-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/16/2021] [Indexed: 10/22/2022]
Abstract
Dichlorodiphenyltrichloroethane (p,p'DDT) is an endocrine-disrupting chemical (EDC). Several studies showed an association between p,p'DDT exposure and reprotoxic effects. We showed that p,p'DDT was a positive allosteric modulator of human follitropin receptor (FSHR). In contrast, we demonstrated that p,p'DDT decreased the cyclic AMP (cAMP) production induced by human choriogonadotropin (hCG). This study evaluated further the effects of p,p'DDT on Gs-, β-arrestin 2- and steroidogenesis pathways induced by hCG or luteinizing hormone (LH). We used Chinese hamster ovary cells line stably expressing hCG/LHR. The effects of 10-100 µM p,p'DDT on cAMP production and on β-arrestin 2 recruitment were measured using bioluminescence and time-resolved resonance energy transfer technology. The impact of 100 µM of p,p'DDT on steroid secretion was analysed in murine Leydig tumor cell line (mLTC-1). In cAMP assays, 100 µM p,p'DDT increased the EC50 by more than 300% and reduced the maximum response of the hCG/LHR to hCG and hLH by 30%. This inhibitory effect was also found in human granulosa cells line and in mLTC-1 cells. Likewise, 100 µM p,p'DDT decreased the hCG- and hLH-promoted β-arrestin 2 recruitment down to 14.2 and 26.6%, respectively. Moreover, 100 µM p,p'DDT decreased by 30 and 47% the progesterone secretion induced by hCG or hLH, respectively, without affecting testosterone secretion. This negative effect of p,p'DDT was independent of cytotoxicity. p,p'DDT acted as a negative allosteric modulator of the hCG/LHR signalling. This emphasizes the importance of analyzing all receptor-downstream pathways to fully understand the deleterious effects of EDC on human health.
Collapse
Affiliation(s)
- Mathilde Munier
- UMR CNRS 6015, INSERM 1083, 3 Rue Roger Amsler - Angers University, 49000, Angers, France. .,Department of Endocrinology, University Hospital, 4 Rue Larrey, 49933, Angers, France. .,Reference Center for Rare Diseases of Thyroid and Hormone Receptors, University Hospital, 4 Rue Larrey, 49933, Angers, France.
| | - Mohammed Ayoub
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates.,Reproductive and Behavioural Physiology, INRAE, CNRS, IFCE, Tours University, 37380, Nouzilly, France
| | - Valentine Suteau
- UMR CNRS 6015, INSERM 1083, 3 Rue Roger Amsler - Angers University, 49000, Angers, France.,Department of Endocrinology, University Hospital, 4 Rue Larrey, 49933, Angers, France
| | - Louis Gourdin
- UMR CNRS 6015, INSERM 1083, 3 Rue Roger Amsler - Angers University, 49000, Angers, France.,Reference Center for Rare Diseases of Thyroid and Hormone Receptors, University Hospital, 4 Rue Larrey, 49933, Angers, France
| | - Daniel Henrion
- UMR CNRS 6015, INSERM 1083, 3 Rue Roger Amsler - Angers University, 49000, Angers, France
| | - Eric Reiter
- Reproductive and Behavioural Physiology, INRAE, CNRS, IFCE, Tours University, 37380, Nouzilly, France
| | - Patrice Rodien
- UMR CNRS 6015, INSERM 1083, 3 Rue Roger Amsler - Angers University, 49000, Angers, France.,Department of Endocrinology, University Hospital, 4 Rue Larrey, 49933, Angers, France.,Reference Center for Rare Diseases of Thyroid and Hormone Receptors, University Hospital, 4 Rue Larrey, 49933, Angers, France
| |
Collapse
|
19
|
Companion animals get close to the toxic aspects of antropogenic world: cytotoxicity of phthalates and bisphenol A on dog testicular primary cells. Cytotechnology 2020; 72:629-638. [PMID: 32435861 DOI: 10.1007/s10616-020-00401-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/11/2020] [Indexed: 10/24/2022] Open
Abstract
Phthalates, which are among the most abundant plasticizers, have detrimental effects on the reproductive system. Similar to human, dogs are prominently exposed to phthalates in daily routines at low concentrations; while toys, training devices and commercial dog foods are considered as the primary sources of exposure. This study aimed to reveal and compare the cytotoxic effects of selected phthalates (Benzyl butyl phthalate (BBP), Diethyl phthalate (DEP), Bis (2-ethylhexyl) phthalate (DEHP), Di-'isobutyl' phthalate (DIBP), Di-'isodecyl' phthalate (-DIDP) Di-'isononyl' phthalate (DINP), Dimethyl phthalate (DMP), Di-n-octyl phthalate (DNOP)), and Bisphenol A (BPA) following 24 h exposure on primary testicular parenchymal cells of dog in vitro at concentrations between 0.001 and 2.5 nM. According to cytotoxicity results, DEHP was found to be the most toxic phthalate with IC50 at 22.53 µM; while DMP was the least (169.17 nM). IC50 of BPA was 161.81 nM, less than the average (61.95 nM) of phthalates. In addition, dog primary testicular cells were found more susceptible to the high molecular weight phthalates (DNOP, DEHP, DINP, DIDP) than low molecular weight phthalates (DMP, DEP, DIBP, BBP). Further studies should focus on morphological, physiological and molecular differences to comprehend the mechanisms involved as well as decreasing the risk for impaired spermatogenesis caused by environmental toxicants in companion animal medicine.
Collapse
|
20
|
Viguié C, Chaillou E, Gayrard V, Picard-Hagen N, Fowler PA. Toward a better understanding of the effects of endocrine disrupting compounds on health: Human-relevant case studies from sheep models. Mol Cell Endocrinol 2020; 505:110711. [PMID: 31954824 DOI: 10.1016/j.mce.2020.110711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 11/25/2022]
Abstract
There are many challenges to overcome in order to properly understand both the exposure to, and effects of, endocrine disruptors (EDs). This is particularly true with respect to fetal life where ED exposures are a major issue requiring toxicokinetic studies of materno-fetal exchange and identification of pathophysiological consequences. The sheep, a large, monotocous, species, is very suitable for in utero fetal catheterization allowing a modelling approach predictive of human fetal exposure. Predicting adverse effects of EDs on human health is frequently impeded by the wide interspecies differences in the regulation of endocrine functions and their effects on biological processes. Because of its similarity to humans as regards gestational and thyroid physiologies and brain ontogeny, the sheep constitutes a highly appropriate model to move one step further on thyroid disruptor hazard assessment. As a grazing animal, the sheep has also proven to be useful in the evaluation of the consequences of chronic environmental exposure to "real-life" complex mixtures at different stages of the reproductive life cycle.
Collapse
Affiliation(s)
- Catherine Viguié
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France.
| | - Elodie Chaillou
- PRC, INRAE Val de Loire, UMR85 Physiologie de la Reproduction et des Comportements, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Véronique Gayrard
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Nicole Picard-Hagen
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
21
|
Spade DJ, Hall SJ, Wortzel JD, Reyes G, Boekelheide K. All-trans Retinoic Acid Disrupts Development in Ex Vivo Cultured Fetal Rat Testes. II: Modulation of Mono-(2-ethylhexyl) Phthalate Toxicity. Toxicol Sci 2020; 168:149-159. [PMID: 30476341 DOI: 10.1093/toxsci/kfy283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Humans are universally exposed to low levels of phthalate esters (phthalates), which are used to plasticize polyvinyl chloride. Phthalates exert adverse effects on the development of seminiferous cords in the fetal testis through unknown toxicity pathways. To investigate the hypothesis that phthalates alter seminiferous cord development by disrupting retinoic acid (RA) signaling in the fetal testis, gestational day 15 fetal rat testes were exposed for 1-3 days to 10-6 M all-trans retinoic acid (ATRA) alone or in combination with 10-6-10-4 M mono-(2-ethylhexyl) phthalate (MEHP) in ex vivo culture. As previously reported, exogenous ATRA reduced seminiferous cord number. This effect was attenuated in a concentration-dependent fashion by MEHP co-exposure. ATRA and MEHP-exposed testes were depleted of DDX4-positive germ cells but not Sertoli cells. MEHP alone enhanced the expression of the RA receptor target Rbp1 and the ovary development-associated genes Wnt4 and Nr0b1, and suppressed expression of the Leydig cell marker, Star, and the germ cell markers, Ddx4 and Pou5f1. In co-exposures, MEHP predominantly enhanced the gene expression effects of ATRA, but the Wnt4 and Nr0b1 concentration-responses were nonlinear. Similarly, ATRA increased the number of cells expressing the granulosa cell marker FOXL2 in testis cultures, but this induction was attenuated by addition of MEHP. These results indicate that MEHP can both enhance and inhibit actions of ATRA during fetal testis development and provide evidence that RA signaling is a target for phthalate toxicity in the fetal testis.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Jeremy D Wortzel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Gerardo Reyes
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912.,Division of Natural Sciences, College of Mount Saint Vincent, Riverdale, New York 10471
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
22
|
Arzuaga X, Walker T, Yost EE, Radke EG, Hotchkiss AK. Use of the Adverse Outcome Pathway (AOP) framework to evaluate species concordance and human relevance of Dibutyl phthalate (DBP)-induced male reproductive toxicity. Reprod Toxicol 2019; 96:445-458. [PMID: 31260805 PMCID: PMC10067323 DOI: 10.1016/j.reprotox.2019.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 06/05/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022]
Abstract
Dibutyl phthalate (DBP) is a phthalate ester used as a plasticizer, and solvent. Studies using rats consistently report that DBP exposure disrupts normal development of the male reproductive system in part via inhibition of androgen synthesis. However, studies using xenograft models report that in human fetal testis DBP exposure is unlikely to impair testosterone synthesis. These results question the validity of the rat model for assessment of male reproductive effects caused by DBP. The Adverse Outcome Pathway (AOP) framework was used to evaluate the available evidence for DBP-induced toxicity to the male reproductive system. Three relevant biological elements were identified: 1) fetal rats are more sensitive than other rodents and human fetal xenografts to DBP-induced anti-androgenic effects, 2) DBP-induced androgen-independent adverse outcomes are conserved amongst different mammalian models and human fetal testis xenografts, and 3) DBP-induced anti-androgenic effects are conserved in different mammalian species when exposure occurs during postnatal life stages.
Collapse
Affiliation(s)
- Xabier Arzuaga
- U.S. Environmental Protection Agency, National Center for Environmental Assessment, Washington, DC, United States of America.
| | - Teneille Walker
- U.S. Environmental Protection Agency, National Center for Environmental Assessment, Washington, DC, United States of America
| | - Erin E Yost
- U.S. Environmental Protection Agency, National Center for Environmental Assessment, Research Triangle Park, NC, United States of America
| | - Elizabeth G Radke
- U.S. Environmental Protection Agency, National Center for Environmental Assessment, Washington, DC, United States of America
| | - Andrew K Hotchkiss
- U.S. Environmental Protection Agency, National Center for Environmental Assessment, Research Triangle Park, NC, United States of America
| |
Collapse
|
23
|
Mayerhofer A. Peritubular cells of the human testis: prostaglandin E 2 and more. Andrology 2019; 8:898-902. [PMID: 31237067 DOI: 10.1111/andr.12669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Several layers of slender, smooth muscle-like, peritubular cells and extracellular matrix (ECM) form the peritubular compartment of the human testis. Peritubular cells are the least explored testicular cells. MATERIALS AND METHODS Human testicular peritubular cells (HTPCs) can be isolated from small testicular fragments of patients and studied in vitro. We have used this cellular model, in combination with human testicular samples, to examine how peritubular cells may contribute to male (in)fertility. RESULTS Human testicular peritubular cells (HTPCs) retain contractile abilities in vitro and secrete many proteins. Among them are factors, which serve intra-testicular roles, for example, glial cell line-derived neurotrophic factor (GDNF), thought to be important for the renewal of spermatogonial stem cells (SSCs). Studies in mutant mice indicated that peritubular cell-derived GDNF is crucial for lifelong spermatogenesis. Thus, peritubular cells are a functional part of the SSC niche. Peritubular cells of mice and men express androgen receptors (AR). In mouse peritubular cells, androgens enhanced GDNF production, but not in HTPCs. Rather, AR activation increased the levels of AR and smooth muscle proteins and thereby enhanced the smooth muscle-like phenotype. Following the lead of a proteomic analysis, which identified the key prostaglandin (PG)-synthesizing enzyme (PTGS1 = COX1), we found that HTPCs secrete PGE2 . COX1, and PGE2 receptors (EP1, 2, and 4) were identified in peritubular cells in situ, supporting in vivo relevance. In HTPCs, activation of EP1/4 increased GDNF and a smooth muscle protein. Ibuprofen is a nonsteroidal anti-inflammatory drug (NSAID), which blocks PG synthesis. Added to HTPCs it reduced PGE2 and GDNF production and lowered smooth muscle protein levels. If applicable to the in vivo situation, the results suggest that ibuprofen and possibly other NSAIDs may impair important peritubular cell functions and consequently testicular functions. CONCLUSION The few examples highlighted, together with others not mentioned here, indicate that HTPCs provide an experimental window into the human testis.
Collapse
Affiliation(s)
- Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology - Anatomy III, Ludwig-Maximilians-Universität (LMU), Planegg-Martinsried, Germany
| |
Collapse
|
24
|
Paschou SA, Wolffenbuttel BHR. Metformin use during pregnancy: Is it really safe? J Diabetes 2018; 10:984-985. [PMID: 29987873 DOI: 10.1111/1753-0407.12813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Stavroula A Paschou
- Division of Endocrinology and Diabetes, "Aghia Sophia" Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
25
|
Rey-Ares V, Rossi SP, Dietrich KG, Köhn FM, Schwarzer JU, Welter H, Frungieri MB, Mayerhofer A. Prostaglandin E 2 (PGE 2) is a testicular peritubular cell-derived factor involved in human testicular homeostasis. Mol Cell Endocrinol 2018; 473:217-224. [PMID: 29408603 DOI: 10.1016/j.mce.2018.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/06/2017] [Accepted: 01/29/2018] [Indexed: 10/18/2022]
Abstract
In man, blockage of prostaglandin (PG)-production e.g. by non-steroidal anti-inflammatory drug (NSAIDs) may have negative testicular side effects, implying beneficial actions of PGs in the testis. We examined human testicular samples and isolated human testicular peritubular cells (HTPCs) to explore sites of PG-synthesis and targets. HTPCs express cyclooxygenase 1 (COX1) and secrete PGE2. Receptors (EP1, 2, 4) were specifically identified in peritubular cells. In HTPCs PGE2 significantly increased mRNA levels of the contractility protein calponin, but did not induce contractions. PGE2, as well as EP1 and EP4 receptor agonists, significantly increased glia cell line derived neurotrophic factor (GDNF) mRNA and/or protein levels. Importantly, the NSAID ibuprofen reduced PGE2 and this action also lowered SMA and calponin mRNA levels and levels of secreted GDNF protein. The results reveal an unknown PGE2 system in the human testis, in involving peritubular cells, which may be prone to interference by NSAIDs.
Collapse
Affiliation(s)
- Verónica Rey-Ares
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | - Soledad Paola Rossi
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany; Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Kim-Gwendolyn Dietrich
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | | | | | - Harald Welter
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilian-University (LMU), D-82152 Planegg, Germany.
| |
Collapse
|
26
|
Anand-Ivell R, Cohen A, Nørgaard-Pedersen B, Jönsson BAG, Bonde JP, Hougaard DM, Lindh CH, Toft G, Lindhard MS, Ivell R. Amniotic Fluid INSL3 Measured During the Critical Time Window in Human Pregnancy Relates to Cryptorchidism, Hypospadias, and Phthalate Load: A Large Case-Control Study. Front Physiol 2018; 9:406. [PMID: 29740335 PMCID: PMC5928321 DOI: 10.3389/fphys.2018.00406] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022] Open
Abstract
The period of the first to second trimester transition in human pregnancy represents a sensitive window for fetal organogenesis, particularly in regard to the development of the male reproductive system. This is a time of relative analytical inaccessibility. We have used a large national biobank of amniotic fluid samples collected at routine amniocentesis to determine the impacts of exogenous endocrine disruptor load on specific fetal biomarkers at this critical time. While adrenal and testicular steroids are highly correlated, they are also mostly positively influenced by increasing phthalate load, represented by the metabolites 7cx-MMeHP and 5cx-MEPP, by perfluorooctane sulfonate (PFOS) exposure, and by smoking, suggesting an adrenal stress response. In contrast, the testis specific biomarkers insulin-like peptide 3 (INSL3) and androstenedione are negatively impacted by the phthalate endocrine disruptors. Using a case-control design, we show that cryptorchidism and hypospadias are both significantly associated with increased amniotic concentration of INSL3 during gestational weeks 13-16, and some, though not all steroid biomarkers. Cases are also linked to a specifically increased variance in the Leydig cell biomarker INSL3 compared to controls, an effect exacerbated by maternal smoking. No influence of phthalate metabolites or PFOS was evident on the distribution of cases and controls. Considering that several animal and human studies have shown a negative impact of phthalate load on fetal and cord blood INSL3, respectively, the present results suggest that such endocrine disruptors may rather be altering the relative dynamics of testicular development and consequent hormone production, leading to a desynchronization of tissue organization during fetal development. Being born small for gestational age appears not to impact on the testicular biomarker INSL3 in second trimester amniotic fluid.
Collapse
Affiliation(s)
| | - Arieh Cohen
- Section of Neonatal Screening and Hormones, Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Bent Nørgaard-Pedersen
- Section of Neonatal Screening and Hormones, Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Bo A. G. Jönsson
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Jens-Peter Bonde
- Department of Occupational and Environmental Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - David M. Hougaard
- Section of Neonatal Screening and Hormones, Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Christian H. Lindh
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Gunnar Toft
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten S. Lindhard
- Perinatal Epidemiology Research Unit, Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - Richard Ivell
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
27
|
Ronaldson-Bouchard K, Vunjak-Novakovic G. Organs-on-a-Chip: A Fast Track for Engineered Human Tissues in Drug Development. Cell Stem Cell 2018; 22:310-324. [PMID: 29499151 PMCID: PMC5837068 DOI: 10.1016/j.stem.2018.02.011] [Citation(s) in RCA: 409] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Organs-on-a-chip (OOCs) are miniature tissues and organs grown in vitro that enable modeling of human physiology and disease. The technology has emerged from converging advances in tissue engineering, semiconductor fabrication, and human cell sourcing. Encompassing innovations in human stem cell technology, OOCs offer a promising approach to emulate human patho/physiology in vitro, and address limitations of current cell and animal models. Here, we review the design considerations for single and multi-organ OOCs, discuss remaining challenges, and highlight the potential impact of OOCs as a fast-track opportunity for tissue engineering to advance drug development and precision medicine.
Collapse
Affiliation(s)
- Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University in the City of New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University in the City of New York, NY 10032, USA; Department of Medicine, Columbia University in the City of New York, NY 10032, USA.
| |
Collapse
|
28
|
Gao HT, Xu R, Cao WX, Di QN, Li RX, Lu L, Xu Q, Yu SQ. Combined effects of simultaneous exposure to six phthalates and emulsifier glycerol monosterate on male reproductive system in rats. Toxicol Appl Pharmacol 2018; 341:87-97. [DOI: 10.1016/j.taap.2018.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 02/02/2023]
|
29
|
Eladak S, Moison D, Guerquin MJ, Matilionyte G, Kilcoyne K, N’Tumba-Byn T, Messiaen S, Deceuninck Y, Pozzi-Gaudin S, Benachi A, Livera G, Antignac JP, Mitchell R, Rouiller-Fabre V, Habert R. Effects of environmental Bisphenol A exposures on germ cell development and Leydig cell function in the human fetal testis. PLoS One 2018; 13:e0191934. [PMID: 29385186 PMCID: PMC5791995 DOI: 10.1371/journal.pone.0191934] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/15/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Using an organotypic culture system termed human Fetal Testis Assay (hFeTA) we previously showed that 0.01 μM BPA decreases basal, but not LH-stimulated, testosterone secreted by the first trimester human fetal testis. The present study was conducted to determine the potential for a long-term antiandrogenic effect of BPA using a xenograft model, and also to study the effect of BPA on germ cell development using both the hFETA and xenograft models. METHODS Using the hFeTA system, first trimester testes were cultured for 3 days with 0.01 to 10 μM BPA. For xenografts, adult castrate male nude mice were injected with hCG and grafted with first trimester testes. Host mice received 10 μM BPA (~ 500 μg/kg/day) in their drinking water for 5 weeks. Plasma levels of total and unconjugated BPA were 0.10 μM and 0.038 μM respectively. Mice grafted with second trimester testes received 0.5 and 50 μg/kg/day BPA by oral gavage for 5 weeks. RESULTS With first trimester human testes, using the hFeTA model, 10 μM BPA increased germ cell apoptosis. In xenografts, germ cell density was also reduced by BPA exposure. Importantly, BPA exposure significantly decreased the percentage of germ cells expressing the pluripotency marker AP-2γ, whilst the percentage of those expressing the pre-spermatogonial marker MAGE-A4 significantly increased. BPA exposure did not affect hCG-stimulated androgen production in first and second trimester xenografts as evaluated by both plasma testosterone level and seminal vesicle weight in host mice. CONCLUSIONS Exposure to BPA at environmentally relevant concentrations impairs germ cell development in first trimester human fetal testis, whilst gonadotrophin-stimulated testosterone production was unaffected in both first and second trimester testis. Studies using first trimester human fetal testis demonstrate the complementarity of the FeTA and xenograft models for determining the respective short-term and long term effects of environmental exposures.
Collapse
Affiliation(s)
- Soria Eladak
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Delphine Moison
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Marie-Justine Guerquin
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Gabriele Matilionyte
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, Scotland, United Kingdom
| | - Karen Kilcoyne
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, Scotland, United Kingdom
| | - Thierry N’Tumba-Byn
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Sébastien Messiaen
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Yoann Deceuninck
- Laboratoire d’Etude des Résidus et Contaminants dans les Aliments (LABERCA), Ecole Nationale Vétérinaire Agroalimentaire et de l’Alimentation Nantes Atlantique (ONIRIS), Nantes, France
| | - Stéphanie Pozzi-Gaudin
- Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Hôpital A. Béclère, Université Paris Sud, Clamart, France
| | - Alexandra Benachi
- Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Hôpital A. Béclère, Université Paris Sud, Clamart, France
| | - Gabriel Livera
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - Jean-Philippe Antignac
- Laboratoire d’Etude des Résidus et Contaminants dans les Aliments (LABERCA), Ecole Nationale Vétérinaire Agroalimentaire et de l’Alimentation Nantes Atlantique (ONIRIS), Nantes, France
| | - Rod Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, Scotland, United Kingdom
| | - Virginie Rouiller-Fabre
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| | - René Habert
- Univ. Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay aux Roses, France
| |
Collapse
|
30
|
Svechnikov K, Savchuk I, Morvan ML, Antignac JP, Le Bizec B, Söder O. Phthalates Exert Multiple Effects on Leydig Cell Steroidogenesis. Horm Res Paediatr 2018; 86:253-263. [PMID: 26559938 DOI: 10.1159/000440619] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/21/2015] [Indexed: 11/19/2022] Open
Abstract
Humans are significantly exposed to phthalates via food packaging, cosmetics and medical devices such as tubings and catheters. Testicular Leydig cells (LCs) are suggested to be among the main targets of phthalate toxicity in the body. However, their sensitivity to phthalates is species-dependent. This paper describes the response of the LCs from different species (mouse, rat and human) to phthalate exposure in different experimental paradigms (in vivo, ex vivo and in vitro), with particular focus on mechanisms of phthalate action on LC steroidogenesis. A comprehensive analysis of the impact of phthalate diesters and phthalate monoesters on LCs in different stages of their development is presented and possible mechanisms of phthalates action are discussed. Finally novel, not yet fully elucidated sites of action of phthalate monoesters on the backdoor pathway of 5α-dihydrotestosterone biosynthesis in immature mouse LCs and their effects on steroidogenesis and redox state in adult mouse LCs are reported.
Collapse
Affiliation(s)
- Konstantin Svechnikov
- Department of Women's and Children's Health, Pediatric Endocrinology Unit, Karolinska Institute and University Hospital, Q2:08, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
31
|
Arendrup FS, Mazaud-Guittot S, Jégou B, Kristensen DM. EDC IMPACT: Is exposure during pregnancy to acetaminophen/paracetamol disrupting female reproductive development? Endocr Connect 2018; 7:149-158. [PMID: 29305399 PMCID: PMC5776669 DOI: 10.1530/ec-17-0298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 12/19/2022]
Abstract
Concern has been raised over chemical-induced disruption of ovary development during fetal life resulting in long-lasting consequences only manifesting themselves much later during adulthood. A growing body of evidence suggests that prenatal exposure to the mild analgesic acetaminophen/paracetamol can cause such a scenario. Therefore, in this review, we discuss three recent reports that collectively indicate that prenatal exposure in a period of 13.5 days post coitum in both rats and mouse can result in reduced female reproductive health. The combined data show that the exposure results in the reduction of primordial follicles, irregular menstrual cycle, premature absence of corpus luteum, as well as reduced fertility, resembling premature ovarian insufficiency syndrome in humans that is linked to premature menopause. This could especially affect the Western parts of the world, where the age for childbirth is continuously being increased and acetaminophen is recommended during pregnancy for pain and fever. We therefore highlight an urgent need for more studies to verify these data including both experimental and epidemiological approaches.
Collapse
Affiliation(s)
| | - Severine Mazaud-Guittot
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
| | - Bernard Jégou
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
- EHESP-School of Public HealthRennes, France
| | - David Møbjerg Kristensen
- Department of NeurologyDanish Headache Center, Rigshospitalet, University of Copenhagen, Denmark
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
| |
Collapse
|
32
|
Forcato S, Novi DRBDS, Costa NO, Borges LI, Góes MLMD, Ceravolo GS, Gerardin DCC. In utero and lactational exposure to metformin induces reproductive alterations in male rat offspring. Reprod Toxicol 2017; 74:48-58. [DOI: 10.1016/j.reprotox.2017.08.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023]
|
33
|
Ivell R, Agoulnik AI, Anand‐Ivell R. Relaxin-like peptides in male reproduction - a human perspective. Br J Pharmacol 2017; 174:990-1001. [PMID: 27933606 PMCID: PMC5406299 DOI: 10.1111/bph.13689] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/10/2016] [Accepted: 12/05/2016] [Indexed: 12/26/2022] Open
Abstract
The relaxin family of peptide hormones and their cognate GPCRs are becoming physiologically well-characterized in the cardiovascular system and particularly in female reproductive processes. Much less is known about the physiology and pharmacology of these peptides in male reproduction, particularly as regards humans. H2-relaxin is involved in prostate function and growth, while insulin-like peptide 3 (INSL3) is a major product of the testicular Leydig cells and, in the adult, appears to modulate steroidogenesis and germ cell survival. In the fetus, INSL3 is a key hormone expressed shortly after sex determination and is responsible for the first transabdominal phase of testicular descent. Importantly, INSL3 is becoming a very useful constitutive biomarker reflecting both fetal and post-natal development. Nothing is known about roles for INSL4 in male reproduction and only very little about relaxin-3, which is mostly considered as a brain peptide, or INSL5. The former is expressed at very low levels in the testes, but has no known physiology there, whereas the INSL5 knockout mouse does exhibit a testicular phenotype with mild effects on spermatogenesis, probably due to a disruption of glucose homeostasis. INSL6 is a major product of male germ cells, although it is relatively unexplored with regard to its physiology or pharmacology, except that in mice disruption of the INSL6 gene leads to a disruption of spermatogenesis. Clinically, relaxin analogues may be useful in the control of prostate cancer, and both relaxin and INSL3 have been considered as sperm adjuvants for in vitro fertilization. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- Richard Ivell
- School of BiosciencesUniversity of NottinghamNottinghamLE12 5RDUK
- School of Veterinary and Medical SciencesUniversity of NottinghamNottinghamLE12 5RDUK
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | | |
Collapse
|
34
|
Hoffmann M, Gebauer S, Nüchter M, Baber R, Ried J, von Bergen M, Kiess W. Endokrine Modulatoren. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2017; 60:640-648. [DOI: 10.1007/s00103-017-2551-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
35
|
Rwigemera A, Joao F, Delbes G. Fetal testis organ culture reproduces the dynamics of epigenetic reprogramming in rat gonocytes. Epigenetics Chromatin 2017; 10:19. [PMID: 28413450 PMCID: PMC5387332 DOI: 10.1186/s13072-017-0127-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Background Epigenetic reprogramming is a critical step in male germ cell development that occurs during perinatal life. It is characterized by the remodeling of different epigenetic marks such as DNA methylation (5mC) and methylation of histone H3. It has been suggested that endocrine disruptors can affect the male germline epigenome by altering epigenetic reprogramming, but the mechanisms involved are still unknown. We have previously used an organ culture system that maintains the development of the different fetal testis cell types, to evaluate the effects of various endocrine disruptors on gametogenesis and steroidogenesis in the rat. We hypothesize that this culture model can reproduce the epigenetic reprogramming in gonocytes. Our aim was to establish the kinetics of three epigenetic marks throughout perinatal development in rats in vivo and compare them after different culture times. Results Using immunofluorescence, we showed that H3K4me2 transiently increased in gonocytes at 18.5 days post-coitum (dpc), while H3K4me3 displayed a stable increase in gonocytes from 18.5 dpc until after birth. 5mC progressively increased from 20.5 dpc until after birth. Using GFP-positive gonocytes purified from GCS-EGFP rats, we established the chronology of re-methylation of H19 and Snrpn in rat gonocytes. Most importantly, using testis explanted at 16.5 or 18.5 dpc and cultured for 2–4 days, we demonstrated that the kinetics of changes in H3K4me2, H3K4me3, global DNA methylation and on parental imprints can generally be reproduced ex vivo with the model of organ culture without the addition of serum. Conclusions This study reveals the chronology of three epigenetic marks (H3K4me2, H3K4me3 and 5mC) and the patterns of methylation of H19 and Snrpn differentially methylated regions in rat gonocytes during perinatal development. Most importantly, our results suggest that the organ culture can reproduce the process of epigenetic reprogramming and can be used to study the impact of environmental chemicals on the establishment of the male germ cell epigenome. Electronic supplementary material The online version of this article (doi:10.1186/s13072-017-0127-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arlette Rwigemera
- Institut National de la Recherche Scientifique, Centre INRS - Institut Armand-Frappier, 531, boulevard des Prairies, Laval, QC H7V 1B7 Canada
| | - Fabien Joao
- Institut National de la Recherche Scientifique, Centre INRS - Institut Armand-Frappier, 531, boulevard des Prairies, Laval, QC H7V 1B7 Canada
| | - Geraldine Delbes
- Institut National de la Recherche Scientifique, Centre INRS - Institut Armand-Frappier, 531, boulevard des Prairies, Laval, QC H7V 1B7 Canada
| |
Collapse
|
36
|
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: 8.8] [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
|
37
|
Koneva LA, Vyas AK, McEachin RC, Puttabyatappa M, H-S W, Sartor MA, Padmanabhan V. Developmental programming: Interaction between prenatal BPA and postnatal overfeeding on cardiac tissue gene expression in female sheep. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:4-18. [PMID: 28079927 PMCID: PMC5730970 DOI: 10.1002/em.22071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 05/23/2023]
Abstract
Epidemiologic studies and studies in rodents point to potential risks from developmental exposure to BPA on cardiometabolic diseases. Furthermore, it is becoming increasingly evident that the manifestation and severity of adverse outcomes is the result of interaction between developmental insults and the prevailing environment. Consistent with this premise, recent studies in sheep found prenatal BPA treatment prevented the adverse effects of postnatal obesity in inducing hypertension. The gene networks underlying these complex interactions are not known. mRNA-seq of myocardium was performed on four groups of four female sheep to assess the effects of prenatal BPA exposure, postnatal overfeeding and their interaction on gene transcription, pathway perturbations and functional effects. The effects of prenatal exposure to BPA, postnatal overfeeding, and prenatal BPA with postnatal overfeeding all resulted in transcriptional changes (85-141 significant differentially expressed genes). Although the effects of prenatal BPA and postnatal overfeeding did not involve dysregulation of many of the same genes, they affected a remarkably similar set of biological pathways. Furthermore, an additive or synergistic effect was not found in the combined treatment group, but rather prenatal BPA treatment led to a partial reversal of the effects of overfeeding alone. Many genes previously known to be affected by BPA and involved in obesity, hypertension, or heart disease were altered following these treatments, and AP-1, EGR1, and EGFR were key hubs affected by BPA and/or overfeeding. Environ. Mol. Mutagen. 58:4-18, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- LA Koneva
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI
| | - AK Vyas
- Department of Pediatrics, Texas Tech Health Sciences Permian Basin, Odessa, TX
| | - RC McEachin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI
| | - M Puttabyatappa
- Department of Pediatrics, University of Michigan, Ann Arbor MI
| | - Wang H-S
- Department of Environmental Health, University of Cincinnati, Cincinnati OH
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, Cincinnati OH
| | - MA Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI
| | - V Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor MI
| |
Collapse
|
38
|
Koneva LA, Vyas AK, McEachin RC, Puttabyatappa M, Wang HS, Sartor MA, Padmanabhan V. Developmental programming: Interaction between prenatal BPA and postnatal overfeeding on cardiac tissue gene expression in female sheep. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:4-18. [PMID: 28079927 DOI: 10.1002/em] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 05/23/2023]
Abstract
Epidemiologic studies and studies in rodents point to potential risks from developmental exposure to BPA on cardiometabolic diseases. Furthermore, it is becoming increasingly evident that the manifestation and severity of adverse outcomes is the result of interaction between developmental insults and the prevailing environment. Consistent with this premise, recent studies in sheep found prenatal BPA treatment prevented the adverse effects of postnatal obesity in inducing hypertension. The gene networks underlying these complex interactions are not known. mRNA-seq of myocardium was performed on four groups of four female sheep to assess the effects of prenatal BPA exposure, postnatal overfeeding and their interaction on gene transcription, pathway perturbations and functional effects. The effects of prenatal exposure to BPA, postnatal overfeeding, and prenatal BPA with postnatal overfeeding all resulted in transcriptional changes (85-141 significant differentially expressed genes). Although the effects of prenatal BPA and postnatal overfeeding did not involve dysregulation of many of the same genes, they affected a remarkably similar set of biological pathways. Furthermore, an additive or synergistic effect was not found in the combined treatment group, but rather prenatal BPA treatment led to a partial reversal of the effects of overfeeding alone. Many genes previously known to be affected by BPA and involved in obesity, hypertension, or heart disease were altered following these treatments, and AP-1, EGR1, and EGFR were key hubs affected by BPA and/or overfeeding. Environ. Mol. Mutagen. 58:4-18, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- L A Koneva
- Departments of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - A K Vyas
- Department of Pediatrics, Texas Tech Health Sciences Permian Basin, Odessa, Texas
| | - R C McEachin
- Departments of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - M Puttabyatappa
- Departments of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - H-S Wang
- Departments of Environmental Health, University of Cincinnati, Cincinnati, Ohio
- Departments of Pharmacology and Cell Biophysics, University of Cincinnati, Cincinnati, Ohio
| | - M A Sartor
- Departments of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - V Padmanabhan
- Departments of Pediatrics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
39
|
Cardoso AM, Alves MG, Mathur PP, Oliveira PF, Cavaco JE, Rato L. Obesogens and male fertility. Obes Rev 2017; 18:109-125. [PMID: 27776203 DOI: 10.1111/obr.12469] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 12/28/2022]
Abstract
In the last decades, several studies evidenced a decrease in male fertility in developed countries. Although the aetiology of this trend in male reproductive health remains a matter of debate, environmental compounds that predispose to weight gain, namely obesogens, are appointed as contributors because of their action as endocrine disruptors. Obesogens favour adipogenesis by an imbalance of metabolic processes and can be found virtually everywhere. These compounds easily accumulate in tissues with high lipid content. Obesogens change the functioning of male reproductive axis, and, consequently, the testicular physiology and metabolism that are pivotal for spermatogenesis. The disruption of these tightly regulated metabolic pathways leads to adverse reproductive outcomes. Notably, adverse effects of obesogens may also promote disturbances in the metabolic performance of the following generations, through epigenetic modifications passed by male gametes. Thus, unveiling the molecular pathways by which obesogens induce toxicity that may end up in epigenetic modifications is imperative. Otherwise, a transgenerational susceptibility to metabolic diseases may be favoured. We present an up-to-date overview of the impact of obesogens on testicular physiology, with a particular focus on testicular metabolism. We also address the effects of obesogens on male reproductive parameters and the subsequent consequences for male fertility.
Collapse
Affiliation(s)
- A M Cardoso
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - M G Alves
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - P P Mathur
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India.,KIIT University, Bhubaneswar, India
| | - P F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - J E Cavaco
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - L Rato
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
40
|
Liang S, Yin L, Shengyang Yu K, Hofmann MC, Yu X. High-Content Analysis Provides Mechanistic Insights into the Testicular Toxicity of Bisphenol A and Selected Analogues in Mouse Spermatogonial Cells. Toxicol Sci 2017; 155:43-60. [PMID: 27633978 PMCID: PMC5216646 DOI: 10.1093/toxsci/kfw178] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Bisphenol A (BPA), an endocrine-disrupting compound, was found to be a testicular toxicant in animal models. Bisphenol S (BPS), bisphenol AF (BPAF), and tetrabromobisphenol A (TBBPA) were recently introduced to the market as alternatives to BPA. However, toxicological data of these compounds in the male reproductive system are still limited so far. This study developed and validated an automated multi-parametric high-content analysis (HCA) using the C18-4 spermatogonial cell line as a model. We applied these validated HCA, including nuclear morphology, DNA content, cell cycle progression, DNA synthesis, cytoskeleton integrity, and DNA damage responses, to characterize and compare the testicular toxicities of BPA and 3 selected commercial available BPA analogues, BPS, BPAF, and TBBPA. HCA revealed BPAF and TBBPA exhibited higher spermatogonial toxicities as compared with BPA and BPS, including dose- and time-dependent alterations in nuclear morphology, cell cycle, DNA damage responses, and perturbation of the cytoskeleton. Our results demonstrated that this specific culture model together with HCA can be utilized for quantitative screening and discriminating of chemical-specific testicular toxicity in spermatogonial cells. It also provides a fast and cost-effective approach for the identification of environmental chemicals that could have detrimental effects on reproduction.
Collapse
Affiliation(s)
- Shenxuan Liang
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | - Lei Yin
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
- ReproTox Biotech LLC, 111 Riverbend Drive, Athens, Georgia
| | - Kevin Shengyang Yu
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia & Hormonal Disorders, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit no. 1105 Houston, Texas
| | - Xiaozhong Yu
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia;
| |
Collapse
|
41
|
Effects of long-term endocrine disrupting compound exposure on Macaca mulatta embryonic stem cells. Reprod Toxicol 2016; 65:382-393. [PMID: 27614199 DOI: 10.1016/j.reprotox.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/23/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022]
Abstract
Endocrine disrupting chemicals (EDCs) exert significant effects on health and physiology, many traceable to effects on stem cell programming underlying development. Understanding risk of low-level, chronic EDC exposure will be enhanced by knowledge of effects on stem cells. We exposed rhesus monkey embryonic stem cells to low levels of five EDCs [bisphenol A (BPA), atrazine (ATR), tributyltin (TBT), perfluorooctanoic acid (PFOA), and di-(2-ethylhexyl) phthalate (DEHP)] for 28days, and evaluated effects on gene expression by RNAseq transcriptome profiling. We observed little effect of BPA, and small numbers of affected genes (≤119) with other EDCs. There was substantial overlap in effects across two, three, or four treatments. Ingenuity Pathway analysis indicated suppression of cell survival genes and genes downstream of several stress response mediators, activation of cell death genes, and modulations in several genes regulating pluripotency, differentiation, and germ layer development. Potential adverse effects of these changes on development are discussed.
Collapse
|
42
|
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: 21] [Impact Index Per Article: 2.3] [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
|
43
|
The Epigenetic Consequences of Paternal Exposure to Environmental Contaminants and Reproductive Toxicants. Curr Environ Health Rep 2016; 3:202-13. [DOI: 10.1007/s40572-016-0101-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
44
|
Hrubik J, Glisic B, Tubic A, Ivancev-Tumbas I, Kovacevic R, Samardzija D, Andric N, Kaisarevic S. Toxicological and chemical investigation of untreated municipal wastewater: Fraction- and species-specific toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2016; 127:153-162. [PMID: 26829069 DOI: 10.1016/j.ecoenv.2016.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 06/05/2023]
Abstract
Absence of a municipal wastewater (WW) treatment plant results in the untreated WW discharge into the recipient. The present study investigated toxic effects and chemical composition of water extracts and fractions from untreated WW and recipient Danube River (DR). Samples were prepared by solid-phase extraction and silica gel fractionation and screened for EROD activity and cytotoxicity using aquatic models, comprising of fish liver cells (PLHC-1) and a model of the early development of zebrafish embryos, while rat (H4IIE) and human (HepG2) hepatoma cells served as mammalian models. Polar fraction caused cytotoxicity and increased the EROD activity in PLHC-1 cells, and increased mortality and developmental abnormalities in developing zebrafish embryos. In H4IIE, polar fraction induced inhibition of cell growth and increased EROD activity, whereas HepG2 exerted low or no response to the exposure. Non-polar and medium-polar fractions were ineffective. Tentative identification by GC/MS showed that WW is characterized by the hydrocarbons, alkylphenols, plasticizers, and a certain number of benzene derivatives and organic acids. In DR, smaller number of organic compounds was identified and toxicity was less pronounced than in WW treatments. The present study revealed the potent toxic effect of polar fraction of untreated WW, with biological responses varying in sensitivity across organisms. Obtained results confirmed that fraction- and species-specific toxicity should be considered when assessing health risk of environmental pollution.
Collapse
Affiliation(s)
- Jelena Hrubik
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia
| | - Branka Glisic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia
| | - Aleksandra Tubic
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Novi Sad, Serbia
| | - Ivana Ivancev-Tumbas
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Novi Sad, Serbia
| | - Radmila Kovacevic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia
| | - Dragana Samardzija
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia
| | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia
| | - Sonja Kaisarevic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Laboratory for Ecotoxicology, Novi Sad, Serbia.
| |
Collapse
|
45
|
Buck Louis GM, Barr DB, Kannan K, Chen Z, Kim S, Sundaram R. Paternal exposures to environmental chemicals and time-to-pregnancy: overview of results from the LIFE study. Andrology 2016; 4:639-47. [PMID: 27061873 DOI: 10.1111/andr.12171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/12/2016] [Accepted: 01/16/2016] [Indexed: 11/30/2022]
Abstract
Published findings from the Longitudinal Investigation of Fertility and the Environment (LIFE) Study regarding the relation between environmental chemicals and couple fecundity, as measured by time-to-pregnancy (TTP), are reviewed with a particular focus on role of the male partner. The LIFE Study recruited 501 couples from 16 counties in two U.S. states upon discontinuing contraception for purposes of becoming pregnant. Upon enrollment, couples provided a blood and urine sample for the quantification of persistent and non-persistent environmental chemicals, respectively, and then completed daily journals until pregnant or up to one year of trying. Female partners used fertility monitors to aid the timing of intercourse relative to ovulation, and digital home pregnancy test kits on the day of expected menses. Chemical classes included: metals, persistent organic pollutants, environmental phenols, and phthalates that were quantified using inductively coupled plasma mass spectrometry or isotope dilution high-resolution or tandem mass spectrometry. Time-to-pregnancy (TTP) was defined as the number of prospectively observed menstrual cycles required for pregnancy. Fecundability odds ratios (FORs) and 95% confidence intervals (CIs) were estimated for each chemical and partner after adjusting for potential confounders and accounting for right censoring and time off contraception. FORs < 1 are suggestive of diminished fecundity or a longer TTP. Significant reductions (ranging from 17-31%) in couple fecundity were observed for male partners' concentration of lead (0.83; 0.70, 0.98), 2,2',4,4'-tetrahydroxybenzophenone (0.69; 0.49, 0.97), monobenzyl (0.80; 0.67, 0.97), and monomethyl (0.81; 0.70, 0.94) phthalates after adjusting for the female partners' concentrations. Seven PCB congeners quantified in men's serum were associated with a 17-29% reduction in couple fecundity. Our findings underscore the importance of a couple-based exposure design, inclusive of the male partner, when assessing couple-dependent outcomes such as TTP to avoid misinterpretation of results based only upon the female partner.
Collapse
Affiliation(s)
- G M Buck Louis
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, The National Institutes of Health, Rockville, MD, USA
| | - D B Barr
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - K Kannan
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Z Chen
- Biostatistics and Bioinformatics Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, The National Institutes of Health, Rockville, MD, USA
| | - S Kim
- Biostatistics and Bioinformatics Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, The National Institutes of Health, Rockville, MD, USA
| | - R Sundaram
- Biostatistics and Bioinformatics Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, The National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
46
|
Martinez-Arguelles DB, Papadopoulos V. Prenatal phthalate exposure: epigenetic changes leading to lifelong impact on steroid formation. Andrology 2016; 4:573-84. [DOI: 10.1111/andr.12175] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/11/2016] [Accepted: 01/28/2016] [Indexed: 01/02/2023]
Affiliation(s)
- D. B. Martinez-Arguelles
- The Research Institute of the McGill University Health Centre; McGill University; Montreal QC Canada
- Department of Medicine; McGill University; Montreal QC Canada
| | - V. Papadopoulos
- The Research Institute of the McGill University Health Centre; McGill University; Montreal QC Canada
- Department of Medicine; McGill University; Montreal QC Canada
- Department of Biochemistry; McGill University; Montreal QC Canada
- Department of Pharmacology & Therapeutics; McGill University; Montreal Quebec Canada
| |
Collapse
|
47
|
Odermatt A, Strajhar P, Engeli RT. Disruption of steroidogenesis: Cell models for mechanistic investigations and as screening tools. J Steroid Biochem Mol Biol 2016; 158:9-21. [PMID: 26807866 DOI: 10.1016/j.jsbmb.2016.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/31/2015] [Accepted: 01/20/2016] [Indexed: 02/03/2023]
Abstract
In the modern world, humans are exposed during their whole life to a large number of synthetic chemicals. Some of these chemicals have the potential to disrupt endocrine functions and contribute to the development and/or progression of major diseases. Every year approximately 1000 novel chemicals, used in industrial production, agriculture, consumer products or as pharmaceuticals, are reaching the market, often with limited safety assessment regarding potential endocrine activities. Steroids are essential endocrine hormones, and the importance of the steroidogenesis pathway as a target for endocrine disrupting chemicals (EDCs) has been recognized by leading scientists and authorities. Cell lines have a prominent role in the initial stages of toxicity assessment, i.e. for mechanistic investigations and for the medium to high throughput analysis of chemicals for potential steroidogenesis disrupting activities. Nevertheless, the users have to be aware of the limitations of the existing cell models in order to apply them properly, and there is a great demand for improved cell-based testing systems and protocols. This review intends to provide an overview of the available cell lines for studying effects of chemicals on gonadal and adrenal steroidogenesis, their use and limitations, as well as the need for future improvements of cell-based testing systems and protocols.
Collapse
Affiliation(s)
- Alex Odermatt
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Petra Strajhar
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Roger T Engeli
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
48
|
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: 9.9] [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
|
49
|
Skakkebaek NE, Rajpert-De Meyts E, Buck Louis GM, Toppari J, Andersson AM, Eisenberg ML, Jensen TK, Jørgensen N, Swan SH, Sapra KJ, Ziebe S, Priskorn L, Juul A. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol Rev 2016; 96:55-97. [PMID: 26582516 DOI: 10.1152/physrev.00017.2015] [Citation(s) in RCA: 618] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
It is predicted that Japan and European Union will soon experience appreciable decreases in their populations due to persistently low total fertility rates (TFR) below replacement level (2.1 child per woman). In the United States, where TFR has also declined, there are ethnic differences. Caucasians have rates below replacement, while TFRs among African-Americans and Hispanics are higher. We review possible links between TFR and trends in a range of male reproductive problems, including testicular cancer, disorders of sex development, cryptorchidism, hypospadias, low testosterone levels, poor semen quality, childlessness, changed sex ratio, and increasing demand for assisted reproductive techniques. We present evidence that several adult male reproductive problems arise in utero and are signs of testicular dysgenesis syndrome (TDS). Although TDS might result from genetic mutations, recent evidence suggests that it most often is related to environmental exposures of the fetal testis. However, environmental factors can also affect the adult endocrine system. Based on our review of genetic and environmental factors, we conclude that environmental exposures arising from modern lifestyle, rather than genetics, are the most important factors in the observed trends. These environmental factors might act either directly or via epigenetic mechanisms. In the latter case, the effects of exposures might have an impact for several generations post-exposure. In conclusion, there is an urgent need to prioritize research in reproductive physiology and pathophysiology, particularly in highly industrialized countries facing decreasing populations. We highlight a number of topics that need attention by researchers in human physiology, pathophysiology, environmental health sciences, and demography.
Collapse
Affiliation(s)
- Niels E Skakkebaek
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Germaine M Buck Louis
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Jorma Toppari
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Michael L Eisenberg
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Tina Kold Jensen
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Shanna H Swan
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Katherine J Sapra
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Søren Ziebe
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Lærke Priskorn
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth & Reproduction and EDMaRC, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Division of Epidemiology, Statistics and Prevention Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Physiology & Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Male Reproductive Medicine & Surgery Program, Stanford University, Stanford, California; Icahn School of Medicine at Mount Sinai, New York, New York; and The Fertility Clinic, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
50
|
Veiga-Lopez A, Moeller J, Sreedharan R, Singer K, Lumeng C, Ye W, Pease A, Padmanabhan V. Developmental programming: interaction between prenatal BPA exposure and postnatal adiposity on metabolic variables in female sheep. Am J Physiol Endocrinol Metab 2016; 310:E238-47. [PMID: 26646100 PMCID: PMC4888526 DOI: 10.1152/ajpendo.00425.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 01/05/2023]
Abstract
Among potential contributors for the increased incidence of metabolic diseases is the developmental exposure to endocrine-disrupting chemicals such as bisphenol A (BPA). BPA is an estrogenic chemical used in a variety of consumer products. Evidence points to interactions of BPA with the prevailing environment. The aim of this study was to assess the effects of prenatal exposure to BPA on postnatal metabolic outcomes, including insulin resistance, adipose tissue distribution, adipocyte morphometry, and expression of inflammatory markers in adipose tissue as well as to assess whether postnatal overfeeding would exacerbate these effects. Findings indicate that prenatal BPA exposure leads to insulin resistance in adulthood in the first breeder cohort (study 1), but not in the second cohort (study 2), which is suggestive of potential differences in genetic susceptibility. BPA exposure induced adipocyte hypertrophy in the visceral fat depot without an accompanying increase in visceral fat mass or increased CD68, a marker of macrophage infiltration, in the subcutaneous fat depot. Cohens effect size analysis found the ratio of visceral to subcutaneous fat depot in the prenatal BPA-treated overfed group to be higher compared with the control-overfed group. Altogether, these results suggest that exposure to BPA during fetal life at levels found in humans can program metabolic outcomes that lead to insulin resistance, a forerunner of type 2 diabetes, with postnatal obesity failing to manifest any interaction with prenatal BPA relative to insulin resistance and adipocyte hypertrophy.
Collapse
Affiliation(s)
- Almudena Veiga-Lopez
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan; Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| | - Jacob Moeller
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Rohit Sreedharan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | - Carey Lumeng
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Wen Ye
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan; and
| | - Anthony Pease
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan
| | | |
Collapse
|