1
|
Rizan C, Rotchell JM, Eng PC, Robaire B, Ciocan C, Kapoor N, Kalra S, Sherman JD. Mitigating the environmental effects of healthcare: the role of the endocrinologist. Nat Rev Endocrinol 2025; 21:344-359. [PMID: 40082727 DOI: 10.1038/s41574-025-01098-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/16/2025]
Abstract
Human health depends on planetary health, and yet healthcare provision can have unintended consequences for the health of the planet. Emissions from the healthcare sector include greenhouse gases, air pollution and plastic pollution, alongside chemical contamination. Chemical pollution resulting in endocrine disruption has been associated with plastics, which are a source of concerning additives such as phthalates, bisphenols, perfluoroalkyl and polyfluoroalkyl substances, and flame retardants (all routinely found in healthcare products). Many endocrine-disrupting chemicals are persistent and ubiquitous in the environment (including water and food sources), with potential secondary harms for human health, including disrupting reproductive, metabolic and thyroid function. Here we review evidence-based strategies for mitigating environmental effects of healthcare delivery. We focus on what endocrinologists can do, including reducing demand for healthcare services through better preventative health, focusing on high-value care and improving sustainability of medical equipment and pharmaceuticals through adopting circular economy principles (including reduce, reuse and, as a last resort, recycle). The specific issue of endocrine-disrupting chemicals might be mitigated through responsible disposal and processing, alongside advocating for the use of alternative materials and replacing additive chemicals with those that have lower toxicity profiles, as well as tighter regulations. We must work to urgently transition to sustainable models of care provision, minimizing negative effects on human and planetary health.
Collapse
Affiliation(s)
- Chantelle Rizan
- Centre for Sustainable Medicine, National University of Singapore, Singapore, Singapore.
- Brighton and Sussex Medical School, Brighton, UK.
| | | | - Pei Chia Eng
- Department of Endocrinology, National University Hospital, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Bernard Robaire
- Faculty of Medicine and Biomedical Sciences, McGill University, Montreal, Quebec, Canada
| | - Corina Ciocan
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Nitin Kapoor
- Department of Endocrinology, Christian Medical College, Vellore, India
- The Non-Communicable Disease Unit, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Jodi D Sherman
- Yale School of Medicine, Yale University, New Haven, CT, USA
- Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
2
|
Soyer-Gobillard MO, Gaspari L, Sultan C. In utero exposure to synthetic sex hormones and their multigenerational impact on neurodevelopmental disorders: Endocrine disruptors as neuroendocrine disruptors. J Neurol Sci 2025; 472:123471. [PMID: 40187226 DOI: 10.1016/j.jns.2025.123471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Synthetic sex hormones, estrogens and/or progestogens, have been widely administered without sufficient long-term studies for decades to millions of pregnant women around the world and although most were banned in the 1970s and 1980s, some progestins continue to be prescribed. Psychiatric disorders, including psychoses such as schizophrenia, bipolar disorders, or severe depression, anxiety, eating disorders, as well as ASD (autism spectrum disorders), accompanied or not by co-morbidities, have been described in children exposed in utero. AIM In this review, we have gathered the main works concerning the harmful effects of these synthetic sex hormones on human health and especially on neurodevelopment so that they are recognized, both for the purpose of teaching medical careers and prescribers and as a precaution for the general population and future generations. METHODS A review of the literature was carried out by searching the PubMed and Google Scholar databases for the period from 2000 to 2024 following the PRISMA guidelines. Studies were identified using the following keywords: diethylstilbestrol, 17-α-ethinylestradiol, progestins, psychosis and endocrine disrupting compounds, estrogens and epigenetics, multigenerational effects. RESULTS The epigenetic nature of such disorders was demonstrated in 2017 to be linked to hypermethylation of the ZFP57 and ADAMTS9 genes that play an important role in neurodevelopment. The ensuing (third) generation of grandchildren is also impacted both on neurodevelopmental and somatic levels, revealing in particular either cognitive disorders as attention deficit hyperactivity disorders (ADHD), psychiatric disorders (bipolarity) or autism spectrum disorders, Asperger's or not. Long-term effects on the fourth generation remain largely unknown due to their young age: however, cognitive disorders (Dyspraxia) and ASD in an adolescent, as well as the warning signs of endometriosis in another adolescent have been described. CONCLUSIONS The multi- and transgenerational effects of these endocrine disruptors have been observed both at the neuropsychological and somatic level and the evidence strongly supports the negative impact of these endocrine disruptors on subsequent generations.
Collapse
Affiliation(s)
| | - Laura Gaspari
- Unité d'Endocrinologie-Gynécologie Pédiatrique, CHU Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Charles Sultan
- Unité d'Endocrinologie-Gynécologie Pédiatrique, CHU Montpellier, Université de Montpellier, 34090 Montpellier, France
| |
Collapse
|
3
|
Ran F, Chen ST, Li MY, Jin DD, Yang ZM. Effect of Diethylstilbestrol on Implantation and Decidualization in Mice. Int J Mol Sci 2025; 26:4122. [PMID: 40362361 PMCID: PMC12071748 DOI: 10.3390/ijms26094122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Diethylstilbestrol (DES) is a synthetic non-steroidal estrogen, which was widely used to prevent preterm birth and abortion from the 1940s to the 1970s. DES can increase the incidence of infertility, the abnormal reproductive tract, and autoimmune diseases. However, the mechanism underlying DES on early pregnancy in mice is unclear. This study evaluated the effects of DES on early pregnancy in mice, especially on uterine receptivity and decidualization. Newborn female mice were subcutaneously injected with 0.1 mg/kg DES, 1 mg/kg DES, or sesame oil as controls for 5 consecutive days. At 6 weeks old, these female mice were mated with 8-12-week-old fertile males to obtain pregnancy. The uteri of these mice were collected on days 4, 5, and 8 of pregnancy for further analysis. On days 5 and 8 of pregnancy, the number of implantation sites in 0.1 mg/kg DES group is similar to the control group, while almost no implantation sites are detected in the 1 mg/kg DES group. On day 4 of pregnancy, there was no significant difference in uterine receptive molecules between the control group and the 0.1 mg/kg DES group. However, the levels of uterine receptive molecules in the 1 mg/kg DES group are abnormal. In addition, 6 μM DES significantly inhibits mouse in vitro decidualization. The excessive activation of pyroptosis may lead to pregnancy failure. The pyroptosis-related molecules in the 1 mg/kg DES group were significantly up-regulated, suggesting that DES may contribute to pregnancy failure by over-activating pyroptosis.
Collapse
Affiliation(s)
- Feng Ran
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Si-Ting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Meng-Yuan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Dan-Dan Jin
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zeng-Ming Yang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
4
|
Xue Y, Shi W, Lun B, Kan M, Jia M, Wu Y, Yang L. Preclinical research models for endometrial cancer: development and selection of animal models. Front Oncol 2025; 15:1512616. [PMID: 39975595 PMCID: PMC11835666 DOI: 10.3389/fonc.2025.1512616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Endometrial cancer (EC) is the most common gynecological malignancy in developed countries, with rising incidence in recent years. Experimental animal models are crucial for studying the pathogenesis, advancing diagnostic methods, and developing new treatments. We review five main EC animal models. The use of spontaneous and chemically-induced models has decreased, with transgenic mouse and xenograft models becoming the most widely used. These models better simulate tumor molecular mechanisms and treatments, with the organoid-based patient-derived xenograft model (O-PDX) showing great promise in drug screening and personalized therapy. The application of humanized models remains limited due to technical challenges and high costs. In this review, we highlight the strengths and limitations of each model to guide researchers in their selection.
Collapse
Affiliation(s)
- Yang Xue
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Lun
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meilin Kan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengling Jia
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuelin Wu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China
| |
Collapse
|
5
|
Gómez-Olarte S, Mailänder V, Castro-Neves J, Stojanovska V, Schumacher A, Meyer N, Zenclussen AC. The ENDOMIX perspective: how everyday chemical mixtures impact human health and reproduction by targeting the immune system†. Biol Reprod 2024; 111:1170-1187. [PMID: 39446589 DOI: 10.1093/biolre/ioae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Endocrine-disrupting chemicals are natural and synthetic compounds found ubiquitously in the environment that interfere with the hormonal-immune axis, potentially impacting human health and reproduction. Exposure to endocrine-disrupting chemicals has been associated with numerous health risks, such as neurodevelopmental disorders, metabolic syndrome, thyroid dysfunction, infertility, and cancers. Nevertheless, the current approach to establishing causality between these substances and disease outcomes has limitations. Epidemiological and experimental research on endocrine-disrupting chemicals faces challenges in accurately assessing chemical exposure and interpreting non-monotonic dose response curves. In addition, most studies have focused on single chemicals or simple mixtures, overlooking complex real-life exposures and mechanistic insights, in particular regarding endocrine-disrupting chemicals' impact on the immune system. The ENDOMIX project, funded by the EU's Horizon Health Program, addresses these challenges by integrating epidemiological, risk assessment, and immunotoxicology methodologies. This systemic approach comprises the triangulation of human cohort, in vitro, and in vivo data to determine the combined effects of chemical mixtures. The present review presents and discusses current literature regarding human reproduction in the context of immunotolerance and chemical disruption mode of action. It further underscores the ENDOMIX perspective to elucidate the impact of endocrine-disrupting chemicals on immune-reproductive health.
Collapse
Affiliation(s)
- Sergio Gómez-Olarte
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Verena Mailänder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Júlia Castro-Neves
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Violeta Stojanovska
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Anne Schumacher
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Nicole Meyer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318, Leipzig, Germany
- Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| |
Collapse
|
6
|
Asimaki K, Vazakidou P, van Tol HTA, van Duursen MBM, Gadella BM. Ketoconazole blocks progesterone production without affecting other parameters of cumulus-oocyte complex maturation. Reprod Toxicol 2024; 128:108637. [PMID: 38876429 DOI: 10.1016/j.reprotox.2024.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
Ketoconazole (KTZ) is widely used as a fungicide, but it is also known to target steroid hormone formation which may affect female reproductive health. Our study aims to investigate the effects of KTZ on in vitro matured bovine cumulus-oocyte complexes (COCs), as a model for female reproductive toxicity. Cumulus cells of in vitro maturing COCs produce progesterone and pregnenolone, but exposure to 10-6 M KTZ effectively blocked the synthesis of these hormones. Exposure to lower concentrations of KTZ (i.e. 10-7 M and 10-8 M) had no such effect on steroidogenesis compared to the 0.1 % v/v DMSO vehicle control. Classical parameters of in vitro COC maturation, such as oocyte nuclear maturation to the metaphase II stage and expansion of the cumulus investment, were not affected by any KTZ concentration tested. Apoptosis and necrosis levels were also not altered in cumulus cells or oocytes exposed to KTZ. Moreover, oocytes exposed to KTZ during maturation showed normal cleavage and early embryo development up to day 8 post fertilization; albeit a statistically significant decrease was observed in day 8 blastocysts produced from oocytes exposed to the lowest concentration of 10-8 M KTZ. When unexposed mature oocytes were fertilized, followed by embryo culture for 8 days under KTZ exposure, no adverse effects in embryo cleavage and blastocyst formation were observed. In conclusion, KTZ has no major impact on in vitro bovine oocyte maturation and blastocyst formation in our study, even at concentrations blocking steroidogenesis.
Collapse
Affiliation(s)
- K Asimaki
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - P Vazakidou
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - H T A van Tol
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - M B M van Duursen
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - B M Gadella
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
7
|
Katić A, Brčić Karačonji I, Micek V, Želježić D. Endocrine-Disrupting Effects of Transplacental and Translactational Exposure to Tembotrione on Hormone Status in Wistar Rat Offspring at Different Developmental Stages: A Pilot Study. TOXICS 2024; 12:533. [PMID: 39195635 PMCID: PMC11359872 DOI: 10.3390/toxics12080533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024]
Abstract
Green agronomy promotes the implementation of natural and naturally derived substances in crop protection. In the present study, we evaluated the endocrine-disrupting potential of the allelopathic herbicide tembotrione in Wistar rats by studying the hormone status of offspring from the treated dams. Three doses of tembotrione (0.0004, 0.0007, and 4.0 mg/kg b.w./day) have been administered to dams during gestation and/or lactation. In the serum of newborn, weaning, and pubertal female and male offspring, 17β-estradiol and testosterone were determined using enzyme-linked immunosorbent assay. A decrease in 17β-estradiol and testosterone was observed in female and male weaning and pubertal offspring exposed to all doses of tembotrione during gestation and lactation. In weaning offspring exposed only during lactation, 17β-estradiol dropped significantly after exposure to the two lower doses and testosterone after exposure to the lowest dose of tembotrione. The greatest effect was observed at the lowest dose of tembotrione. In newborns, we observed increased 17β-estradiol after exposure to two lower doses of tembotrione and significantly increased testosterone after exposure to the lowest dose. The highest dose of tembotrione decreased 17β-estradiol significantly in newborn females. The obtained results suggest that tembotrione might be considered a pro-estrogenic or estrogen agonistic compound under the exposure conditions applied in this investigation.
Collapse
Affiliation(s)
- Anja Katić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000 Zagreb, Croatia; (I.B.K.); (D.Ž.)
| | - Irena Brčić Karačonji
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000 Zagreb, Croatia; (I.B.K.); (D.Ž.)
- Faculty of Health Studies, University of Rijeka, Viktora Cara Emina 5, 51000 Rijeka, Croatia
| | - Vedran Micek
- Animal Breeding Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000 Zagreb, Croatia;
| | - Davor Želježić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000 Zagreb, Croatia; (I.B.K.); (D.Ž.)
| |
Collapse
|
8
|
Hossain MF, Mustary UH, Tokumoto T. Evidence of binding between diethylstilbestrol (DES) and the goldfish ( Carassius auratus) membrane progesterone receptor α. Toxicol Mech Methods 2024; 34:563-571. [PMID: 38317456 DOI: 10.1080/15376516.2024.2311185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND In a previous study, diethylstilbestrol (DES) was shown to induce oocyte maturation in fish. In the present study, the interaction of DES on goldfish membrane progesterone receptor α (GmPRα) was investigated using a competitive binding assay with radiolabeled steroids. The results indicate that DES exerts its effects on membrane progesterone receptor alpha (mPRα) and induces oocyte maturation through nongenomic steroid mechanisms. This study provides empirical data that demonstrate the binding between DES and GmPRα. METHODS Binding of DES to GmPRα was achieved by using radiolabeled DES and recombinant GmPRα expressed in culture cells or purified GmPRα proteins that coupled to graphene quantum dots (GQDs). Additionally, the competitive binding of fluorescently labeled progesterone to GmPRα-expressing cells was evaluated. RESULTS Although significant nonspecific binding of radiolabeled DES to the cell membrane that expresses GmPRα has been observed, specific binding of DES to GmPRα has been successfully identified in the presence of digitonin. Furthermore, the specific binding of DES to GmPRα was confirmed by a binding assay using GQD-GmPRα. The radiolabeled DES was shown to bind to GQD-GmPRα. Additionally, the competition for the binding of fluorescently labeled progesterone to GmPRα-expressing cells was achieved with the DES. CONCLUSIONS The results of the experiments revealed that DES binds to GmPRα. Thus, it can be concluded that DES induces goldfish oocyte maturation by binding to GmPRα.
Collapse
Affiliation(s)
- Md Forhad Hossain
- Department of Bioscience, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Umme Habiba Mustary
- Department of Bioscience, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Toshinobu Tokumoto
- Department of Bioscience, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| |
Collapse
|
9
|
Boizet-Bonhoure B, Déjardin S, Girard M, Durix Q, Poulat F, Philibert P. Adenomyotic Lesions Are Induced in the Mouse Uterus after Exposure to NSAID and EE2 Mixtures at Environmental Doses. Int J Mol Sci 2024; 25:2003. [PMID: 38396681 PMCID: PMC10889173 DOI: 10.3390/ijms25042003] [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: 12/28/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
The aim of this study was to assess the long-term effect of exposure to environmentally relevant doses of non-steroidal anti-inflammatory drugs (NSAIDs; ibuprofen, and diclofenac) and 17β-ethinylestradiol (EE2) on the mouse uterus. NSAID-EE2 mixtures were administered in the drinking water from gestational day 8 until 8 weeks post-birth (i.e., during embryo development, lactation, puberty, and sexual maturity). The incidence of adenomyosis lesions (presence of endometrial glands in the inner myometrium) increased up to 60% in the uterus of 8-week-old exposed females (F1) and to 85% in F2 females (exposed father). Histological analysis revealed aberrant proliferation and apoptosis, vacuolization of epithelial cells, and increased incidence of abnormal glands in the luminal and glandular epithelium in F1 and F2 uteri. Moreover, myofibroblast proportion (alpha-smooth muscle actin (α-SMA) expression analysis) and collagen expression (Picrosirius red stain; a fibrosis hallmark) were increased in F1 and F2 endometrium. Connexin-43 was aberrantly distributed in the endometrial stroma and glands of F1 and F2 uteri. Conversely, uterine 17β-estradiol and progesterone levels were not affected in F1 and F2 females. These findings demonstrated that in mice, chronic exposure to NSAID and EE2 mixtures at environmental doses intergenerationally affects uterine physiology, particularly the endometrium. It may serve as a model to study the pathophysiology of human adenomyosis.
Collapse
Affiliation(s)
- Brigitte Boizet-Bonhoure
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier UMR9002, 34090 Montpellier, France; (S.D.); (M.G.); (F.P.)
| | - Stéphanie Déjardin
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier UMR9002, 34090 Montpellier, France; (S.D.); (M.G.); (F.P.)
| | - Mélissa Girard
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier UMR9002, 34090 Montpellier, France; (S.D.); (M.G.); (F.P.)
| | - Quentin Durix
- IExplore-RAM, Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, INSERM, Université de Montpellier, 34090 Montpellier, France;
| | - Francis Poulat
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier UMR9002, 34090 Montpellier, France; (S.D.); (M.G.); (F.P.)
| | - Pascal Philibert
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier UMR9002, 34090 Montpellier, France; (S.D.); (M.G.); (F.P.)
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpital Carémeau, CHU de Nîmes, 30029 Nîmes, France
| |
Collapse
|
10
|
Li YF, Lin YT, Wang YQ, Ni JY, Power DM. Ioxynil and diethylstilbestrol impair cardiac performance and shell growth in the mussel Mytilus coruscus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:166834. [PMID: 37717744 DOI: 10.1016/j.scitotenv.2023.166834] [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: 06/20/2023] [Revised: 07/30/2023] [Accepted: 09/02/2023] [Indexed: 09/19/2023]
Abstract
The herbicide ioxynil (IOX) and the synthetic estrogen diethylstilbestrol (DES) are environmentally relevant contaminants that act as endocrine disruptors (EDCs) and have recently been shown to be cardiovascular disruptors in vertebrates. Mussels, Mytilus coruscus, were exposed to low doses of IOX (0.37, 0.037 and 0.0037 mg/L) and DES (0.27, 0.027 and 0.0027 mg/L) via the water and the effect monitored by generating whole animal transcriptomes and measuring cardiac performance and shell growth. One day after IOX (0.37 and 0.037 mg/L) and DES (0.27 and 0.027 mg/L) exposure heart rate frequency was decreased in both groups and 0.27 mg/L DES significantly reduced heart rate frequency with increasing time of exposure (P < 0.05) and no acclimatization occurred. The functional effects were coupled to significant differential expression of genes of the serotonergic synapse pathway and cardiac-related genes at 0.027 mg/L DES, which suggests that impaired heart function may be due to interference with neuroendocrine regulation and direct cardiac effect genes. Multiple genes related to detoxifying xenobiotic substances were up regulated and genes related to immune function were down regulated in the DES group (vs. control), indicating that detoxification processes were enhanced, and the immune response was depressed. In contrast, IOX had a minor disrupting effect at a molecular level. Of note was a significant suppression (P < 0.05) by DES of shell growth in juveniles and lower doses (< 0.0027 mg/L) had a more severe effect. The shell growth depression in 0.0027 mg/L DES-treated juveniles was not accompanied by abundant differential gene expression, suggesting that the effect of 0.0027 mg/L DES on shell growth may be direct. The results obtained in the present study reveal for the first time that IOX and DES may act as neuroendocrine disrupters with a broad spectrum of effects on cardiac performance and shell growth, and that DES exposure had a much more pronounced effect than IOX in a marine bivalve.
Collapse
Affiliation(s)
- Yi-Feng Li
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; Centre of Marine Sciences, University of Algarve, Faro, Portugal.
| | - Yue-Tong Lin
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Yu-Qing Wang
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Ji-Yue Ni
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Deborah M Power
- International Research Centre for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; Centre of Marine Sciences, University of Algarve, Faro, Portugal.
| |
Collapse
|
11
|
Jefferson TB, Wang T, Jefferson WN, Li Y, Hamilton KJ, Wade PA, Williams CJ, Korach KS. Multiple tissue-specific epigenetic alterations regulate persistent gene expression changes following developmental DES exposure in mouse reproductive tissues. Epigenetics 2023; 18:2139986. [PMID: 36328762 PMCID: PMC9980695 DOI: 10.1080/15592294.2022.2139986] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Clinically, developmental exposure to the endocrine disrupting chemical, diethylstilboestrol (DES), results in long-term male and female infertility. Experimentally, developmental exposure to DES results in abnormal reproductive tract phenotypes in male and female mice. Previously, we reported that neonatal DES exposure causes ERα-mediated aberrations in the transcriptome and in DNA methylation in seminal vesicles (SVs) of adult mice. However, only a subset of DES-altered genes could be explained by changes in DNA methylation. We hypothesized that alterations in histone modification may also contribute to the altered transcriptome during SV development. To test this idea, we performed a series of genome-wide analyses of mouse SVs at pubertal and adult developmental stages in control and DES-exposed wild-type and ERα knockout mice. Neonatal DES exposure altered ERα-mediated mRNA and lncRNA expression in adult SV, including genes encoding chromatin-modifying proteins that can impact histone H3K27ac modification. H3K27ac patterns, particularly at enhancers, and DNA methylation were reprogrammed over time during normal SV development and after DES exposure. Some of these reprogramming changes were ERα-dependent, but others were ERα-independent. A substantial number of DES-altered genes had differential H3K27ac peaks at nearby enhancers. Comparison of gene expression changes, H3K27ac marks and DNA methylation marks between adult SV and adult uterine tissue from ovariectomized mice neonatally exposed to DES revealed that most of the epigenetic changes and altered genes were distinct in the two tissues. These findings indicate that the effects of developmental DES exposure cause reprogramming of reproductive tract tissue differentiation through multiple epigenetic mechanisms.
Collapse
Affiliation(s)
- Tanner B. Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Tianyuan Wang
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Wendy N. Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Yin Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Katherine J. Hamilton
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Paul A. Wade
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Carmen J. Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| | - Kenneth S. Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, 27709, USA
| |
Collapse
|
12
|
Chauhan R, Archibong AE, Ramesh A. Imprinting and Reproductive Health: A Toxicological Perspective. Int J Mol Sci 2023; 24:16559. [PMID: 38068882 PMCID: PMC10706004 DOI: 10.3390/ijms242316559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
This overview discusses the role of imprinting in the development of an organism, and how exposure to environmental chemicals during fetal development leads to the physiological and biochemical changes that can have adverse lifelong effects on the health of the offspring. There has been a recent upsurge in the use of chemical products in everyday life. These chemicals include industrial byproducts, pesticides, dietary supplements, and pharmaceutical products. They mimic the natural estrogens and bind to estradiol receptors. Consequently, they reduce the number of receptors available for ligand binding. This leads to a faulty signaling in the neuroendocrine system during the critical developmental process of 'imprinting'. Imprinting causes structural and organizational differentiation in male and female reproductive organs, sexual behavior, bone mineral density, and the metabolism of exogenous and endogenous chemical substances. Several studies conducted on animal models and epidemiological studies provide profound evidence that altered imprinting causes various developmental and reproductive abnormalities and other diseases in humans. Altered metabolism can be measured by various endpoints such as the profile of cytochrome P-450 enzymes (CYP450's), xenobiotic metabolite levels, and DNA adducts. The importance of imprinting in the potentiation or attenuation of toxic chemicals is discussed.
Collapse
Affiliation(s)
- Ritu Chauhan
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Anthony E. Archibong
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Aramandla Ramesh
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
13
|
DiVito Evans A, Fairbanks RA, Schmidt P, Levine MT. Histone methylation regulates reproductive diapause in Drosophila melanogaster. PLoS Genet 2023; 19:e1010906. [PMID: 37703303 PMCID: PMC10499233 DOI: 10.1371/journal.pgen.1010906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023] Open
Abstract
Fluctuating environments threaten fertility and viability. To better match the immediate, local environment, many organisms adopt alternative phenotypic states, a phenomenon called "phenotypic plasticity." Natural populations that predictably encounter fluctuating environments tend to be more plastic than conspecific populations that encounter a constant environment, suggesting that phenotypic plasticity can be adaptive. Despite pervasive evidence of such "adaptive phenotypic plasticity," gene regulatory mechanisms underlying plasticity remains poorly understood. Here we test the hypothesis that environment-dependent phenotypic plasticity is mediated by epigenetic factors. To test this hypothesis, we exploit the adaptive reproductive arrest of Drosophila melanogaster females, called diapause. Using an inbred line from a natural population with high diapause plasticity, we demonstrate that diapause is determined epigenetically: only a subset of genetically identical individuals enter diapause and this diapause plasticity is epigenetically transmitted for at least three generations. Upon screening a suite of epigenetic marks, we discovered that the active histone marks H3K4me3 and H3K36me1 are depleted in diapausing ovaries. Using ovary-specific knockdown of histone mark writers and erasers, we demonstrate that H3K4me3 and H3K36me1 depletion promotes diapause. Given that diapause is highly polygenic, that is, distinct suites of alleles mediate diapause plasticity across distinct genotypes, we also investigated the potential for genetic variation in diapause-determining epigenetic marks. Specifically, we asked if these histone marks were similarly depleted in diapause of a genotypically distinct line. We found evidence of divergence in both the gene expression program and histone mark abundance. This study reveals chromatin determinants of phenotypic plasticity and suggests that these determinants may be genotype-dependent, offering new insight into how organisms may exploit and evolve epigenetic mechanisms to persist in fluctuating environments.
Collapse
Affiliation(s)
- Abigail DiVito Evans
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Regina A. Fairbanks
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Paul Schmidt
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mia T. Levine
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
14
|
Priyadarshini E, Parambil AM, Rajamani P, Ponnusamy VK, Chen YH. Exposure, toxicological mechanism of endocrine disrupting compounds and future direction of identification using nano-architectonics. ENVIRONMENTAL RESEARCH 2023; 225:115577. [PMID: 36871939 DOI: 10.1016/j.envres.2023.115577] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Endocrine-disrupting compounds (EDC) are a group of exogenous chemicals that structurally mimic hormones and interfere with the hormonal signaling cascade. EDC interacts with hormone receptors, transcriptional activators, and co-activators, altering the signaling pathway at both genomic and non-genomic levels. Consequently, these compounds are responsible for adverse health ailments such as cancer, reproductive issues, obesity, and cardiovascular and neurological disorders. The persistent nature and increasing incidence of environmental contamination from anthropogenic and industrial effluents have become a global concern, resulting in a movement in both developed and developing countries to identify and estimate the degree of exposure to EDC. The U.S. Environment Protection Agency (EPA) has outlined a series of in vitro and in vivo assays to screen potential endocrine disruptors. However, the multidisciplinary nature and concerns over the widespread application demand alternative and practical techniques for identifying and estimating EDC. The review chronicles the state-of-art 20 years (1990-2023) of scientific literature regarding EDC's exposure and molecular mechanism, highlighting the toxicological effects on the biological system. Alteration in signaling mechanisms by representative endocrine disruptors such as bisphenol A (BPA), diethylstilbestrol (DES), and genistein has been emphasized. We further discuss the currently available assays and techniques for in vitro detection and propose the prominence of designing nano-architectonic-sensor substrates for on-site detection of EDC in the contaminated aqueous environment.
Collapse
Affiliation(s)
- Eepsita Priyadarshini
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ajith Manayil Parambil
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Vinoth Kumar Ponnusamy
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, Taiwan; Department of Chemistry, National Sun Yat-sen University (NSYSU), Kaohsiung City, 804, Taiwan; PhD Program in Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, 811, Taiwan.
| | - Yi-Hsun Chen
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
15
|
EFSA Panel on Plant Protection Products and their Residues (PPR), Hernandez‐Jerez AF, Adriaanse P, Aldrich A, Berny P, Coja T, Duquesne S, Focks A, Millet M, Pelkonen O, Pieper S, Tiktak A, Topping CJ, Widenfalk A, Wilks M, Wolterink G, Angeli K, Recordati C, Van Durseen M, Aiassa E, Lanzoni A, Lostia A, Martino L, Guajardo IPM, Panzarea M, Terron A, Marinovich M. Development of adverse outcome pathways relevant for the identification of substances having endocrine disruption properties Uterine adenocarcinoma as adverse outcome. EFSA J 2023; 21:e07744. [PMID: 36818642 PMCID: PMC9926893 DOI: 10.2903/j.efsa.2023.7744] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Development of adverse outcome pathways (AOPs) for uterine adenocarcinoma can provide a practical tool to implement the EFSA-ECHA Guidance (2018) for the identification of endocrine disruptors in the context of Regulations (EU) No 528/2012 and (EC) No 1107/2009. AOPs can give indications about the strength of the relationship between an adverse outcome (intended as a human health outcome) and chemicals (pesticides but not only) affecting the pathways. In this scientific opinion, the PPR Panel explored the development of AOPs for uterine adenocarcinoma. An evidence-based approach methodology was applied, and literature reviews were produced using a structured framework assuring transparency, objectivity, and comprehensiveness. Several AOPs were developed; these converged to a common critical node, that is increased estradiol availability in the uterus followed by estrogen receptor activation in the endometrium; therefore, a putative AOP network was considered. An uncertainty analysis and a probabilistic quantification of the weight of evidence have been carried out via expert knowledge elicitation for each set of MIEs/KEs/KERs included in individual AOPs. The collected data on the AOP network were evaluated qualitatively, whereas a quantitative uncertainty analysis for weight of the AOP network certainty has not been performed. Recommendations are provided, including exploring further the uncertainties identified in the AOPs and putative AOP network; further methodological developments for quantifying the certainty of the KERs and of the overall AOPs and AOP network; and investigating of NAMs applications in the context of some of the MIEs/KEs currently part of the putative AOP network developed.
Collapse
|
16
|
Asimaki K, Vazakidou P, Modder E, Tol HV, Duursen MV, Gadella B. P12-45 The bovine model of in vitro oocyte maturation has a potential application in the screening of EDCs eliciting reproductive toxicity via oocyte effects; diethylstilbestrol as the showcase compound. Toxicol Lett 2022. [DOI: 10.1016/j.toxlet.2022.07.524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
17
|
Ma Q, Yang X, Guo Y, Wang Y, Liu Y, Zhang S, Xie HQ, Xiang T, Li Z, Nie T, Yan Y, Qu G, Jiang G. Effect-directed analysis of estrogenic chemicals in sediments from an electronic-waste recycling area. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 306:119369. [PMID: 35513195 DOI: 10.1016/j.envpol.2022.119369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 06/14/2023]
Abstract
Electronic waste (e-waste) pollution is of great concern due to the release of hazardous chemicals during the improper e-waste disposal. Many chemicals leached from e-waste were reported to pose estrogenic effects. To date, little is known regarding the occurrence and biological effects of estrogenic chemicals in sediments near an e-waste area. In this study, an effect-directed analysis (EDA) is applied to determine the estrogenic chemicals in sediments of four sites collected from a typical e-waste recycling city in China. Following screening with the ER-CALUX assay, the extract of sample with the most potent effect was subjected in fractionation using reverse phase liquid chromatography. Based on a target analysis for the active fractions, four compounds, including estrone, 17β-estradiol, 17α-ethinylestradiol and bisphenol A, were identified, and these contributed to 17% of the total toxic effects in the sample. A further nontarget analysis screened four candidates, namely diethylstilbestrol (DES), hexestrol (HES), nandrolone and durabolin, and the total contribution was found to be 48% from the active sample. Specifically, DES and HES were only detected in the active sample and were found to be the primary drivers of estrogenic effects. An examination of the identified chemicals in the four sites indicated that these estrogenic chemicals may originate from e-waste recycling, livestock excretion and domestic waste. These findings uncovered the estrogenic pollutants in sediments from an e-waste area. Considering single endpoint in biological assay is not abundant to screen chemicals with different toxic effects, further EDA studies with multiple endpoints are required to better understand the occurrence of representative or unknown chemicals in e-waste-polluted areas.
Collapse
Affiliation(s)
- Qianchi Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| | - Yunhe Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Environmental and Resource Science, Zhejiang University, Hangzhou, 310058, China
| | - Yi Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310000, China
| | - Yanna Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Songyan Zhang
- Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tongtong Xiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Zikang Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tong Nie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Yuhao Yan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310000, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China; College of Environmental and Resource Science, Zhejiang University, Hangzhou, 310058, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310000, China
| |
Collapse
|
18
|
Asimaki K, Vazakidou P, van Tol HTA, Oei CHY, Modder EA, van Duursen MBM, Gadella BM. Bovine In Vitro Oocyte Maturation and Embryo Production Used as a Model for Testing Endocrine Disrupting Chemicals Eliciting Female Reproductive Toxicity With Diethylstilbestrol as a Showcase Compound. FRONTIERS IN TOXICOLOGY 2022; 4:811285. [PMID: 35686045 PMCID: PMC9171015 DOI: 10.3389/ftox.2022.811285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) can interfere with normal hormonal action and regulation. Exposure of women to EDCs has been associated with adverse reproductive health outcomes. The assays currently used to identify EDCs that elicit female reproductive toxicity lack screening tests that address effects on the maturation of oocytes, a process that enables them to be fertilized and develop into embryos. Here, a screening method employing the bovine model of in vitro oocyte maturation and embryo production is described. Endpoints explored address important events in oocyte maturation and developmental competence acquisition. To test the method, the effects of the known human EDC diethylstilbestrol (DES; an estrogen receptor agonist) were evaluated in a range of concentrations (10–9 M, 10–7 M, 10–5 M). Bovine oocytes were exposed to DES during in vitro maturation (IVM) or embryos were exposed during in vitro embryo culture (IVC). The endpoints evaluated included nuclear maturation, mitochondrial redistribution, cumulus cell expansion, apoptosis, and steroidogenesis. DES-exposed oocytes were fertilized to record embryo cleavage and blastocyst rates to uncover effects on developmental competence. Similarly, the development of embryos exposed to DES during IVC was monitored to assess the impact on early embryo development. Exposure to 10–9 M or 10–7 M DES did not affect the endpoints addressing oocyte maturation or embryo development. However, there were considerable detrimental effects observed in oocytes exposed to 10–5 M DES. Specifically, compared to vehicle-treated oocytes, there was a statistically significant reduction in nuclear maturation (3% vs 84%), cumulus expansion (2.8-fold vs 3.6-fold) and blastocyst rate (3% vs 32%). Additionally, progesterone and pregnenolone concentrations measured in IVM culture media were increased. The screening method described here shows that bovine oocytes were sensitive to the action of this particular chemical (i.e., DES), albeit at high concentrations. In principle, this method provides a valuable tool to assess the oocyte maturation process and early embryo development that can be used for reproductive toxicity screening and possibly EDC identification. Further studies should include EDCs with different mechanisms of action and additional endpoints to further demonstrate the applicability of the bovine oocyte model for chemical risk assessment purposes and EDC identification.
Collapse
Affiliation(s)
- K. Asimaki
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: K. Asimaki,
| | - P. Vazakidou
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - H. T. A. van Tol
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - C. H. Y. Oei
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - E. A. Modder
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - M. B. M. van Duursen
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - B. M. Gadella
- Division of Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
19
|
Plante I, Winn LM, Vaillancourt C, Grigorova P, Parent L. Killing two birds with one stone: Pregnancy is a sensitive window for endocrine effects on both the mother and the fetus. ENVIRONMENTAL RESEARCH 2022; 205:112435. [PMID: 34843719 DOI: 10.1016/j.envres.2021.112435] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Pregnancy is a complex process requiring tremendous physiological changes in the mother in order to fulfill the needs of the growing fetus, and to give birth, expel the placenta and nurse the newborn. These physiological modifications are accompanied with psychological changes, as well as with variations in habits and behaviors. As a result, this period of life is considered as a sensitive window as impaired functional and physiological changes in the mother can have short- and long-term impacts on her health. In addition, dysregulation of the placenta and of mechanisms governing placentation have been linked to chronic diseases later-on in life for the fetus, in a concept known as the Developmental Origin of Health and Diseases (DOHaD). This concept stipulates that any change in the environment during the pre-conception and perinatal (in utero life and neonatal) period to puberty, can be "imprinted" in the organism, thereby impacting the health and risk of chronic diseases later in life. Pregnancy is a succession of events that is regulated, in large part, by hormones and growth factors. Therefore, small changes in hormonal balance can have important effects on both the mother and the developing fetus. An increasing number of studies demonstrate that exposure to endocrine disrupting compounds (EDCs) affect both the mother and the fetus giving rise to growing concerns surrounding these exposures. This review will give an overview of changes that happen during pregnancy with respect to the mother, the placenta, and the fetus, and of the current literature regarding the effects of EDCs during this specific sensitive window of exposure.
Collapse
Affiliation(s)
- Isabelle Plante
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada.
| | - Louise M Winn
- Queen's University, School of Environmental Studies, Department of Biomedical and Molecular Sciences, Kingston, ON, Canada
| | | | - Petya Grigorova
- Département Science et Technologie, Université TELUQ, Montreal, QC, Canada
| | - Lise Parent
- Département Science et Technologie, Université TELUQ, Montreal, QC, Canada
| |
Collapse
|
20
|
Athar F, Templeman NM. C. elegans as a model organism to study female reproductive health. Comp Biochem Physiol A Mol Integr Physiol 2022; 266:111152. [PMID: 35032657 DOI: 10.1016/j.cbpa.2022.111152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022]
Abstract
Female reproductive health has been historically understudied and underfunded. Here, we present the advantages of using a free-living nematode, Caenorhabditis elegans, as an animal system to study fundamental aspects of female reproductive health. C. elegans is a powerful high-throughput model organism that shares key genetic and physiological similarities with humans. In this review, we highlight areas of pressing medical and biological importance in the 21st century within the context of female reproductive health. These include the decline in female reproductive capacity with increasing chronological age, reproductive dysfunction arising from toxic environmental insults, and cancers of the reproductive system. C. elegans has been instrumental in uncovering mechanistic insights underlying these processes, and has been valuable for developing and testing therapeutics to combat them. Adopting a convenient model organism such as C. elegans for studying reproductive health will encourage further research into this field, and broaden opportunities for making advancements into evolutionarily conserved mechanisms that control reproductive function.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.
| |
Collapse
|
21
|
Zhang RR, Li XJ, Sun AL, Song SQ, Shi XZ. A highly selective fluorescence nanosensor based on the dual-function molecularly imprinted layer coated quantum dots for the sensitive detection of diethylstilbestrol/cypermethrin in fish and seawater. Food Control 2022. [DOI: 10.1016/j.foodcont.2021.108438] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Lite C, Raja GL, Juliet M, Sridhar VV, Subhashree KD, Kumar P, Chakraborty P, Arockiaraj J. In utero exposure to endocrine-disrupting chemicals, maternal factors and alterations in the epigenetic landscape underlying later-life health effects. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 89:103779. [PMID: 34843942 DOI: 10.1016/j.etap.2021.103779] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
Widespread persistence of endocrine-disrupting chemicals (EDCs) in the environment has mandated the need to study their potential effects on an individual's long-term health after both acute and chronic exposure periods. In this review article a particular focus is given on in utero exposure to EDCs in rodent models which resulted in altered epigenetic programming and transgenerational effects in the offspring causing disrupted reproductive and metabolic phenotypes. The literature to date establishes the impact of transgenerational effects of EDCs potentially associated with epigenetic mediated mechanisms. Therefore, this review aims to provide a comprehensive overview of epigenetic programming and it's regulation in mammals, primarily focusing on the epigenetic plasticity and susceptibility to exogenous hormone active chemicals during the early developmental period. Further, we have also in depth discussed the epigenetic alterations associated with the exposure to selected EDCs such as Bisphenol A (BPA), di-2-ethylhexyl phthalate (DEHP) and vinclozlin upon in utero exposure especially in rodent models.
Collapse
Affiliation(s)
- Christy Lite
- Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India.
| | - Glancis Luzeena Raja
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Melita Juliet
- Department of Oral and Maxillofacial Surgery, SRM Kattankulathur Dental College and Hospital, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Vasisht Varsh Sridhar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - K Divya Subhashree
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India
| | - Praveen Kumar
- Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Paromita Chakraborty
- Environmental Science and Technology Laboratory, Department of Chemical Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Chennai 603203, Tamil Nadu, India.
| |
Collapse
|
23
|
|
24
|
Zamora-León P. Are the Effects of DES Over? A Tragic Lesson from the Past. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10309. [PMID: 34639609 PMCID: PMC8507770 DOI: 10.3390/ijerph181910309] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023]
Abstract
Diethylstilbestrol (DES), a transplacental endocrine-disrupting chemical, was prescribed to pregnant women for several decades. The number of women who took DES is hard to know precisely, but it has been estimated that over 10 million people have been exposed around the world. DES was classified in the year 2000 as carcinogenic to humans. The deleterious effects induced by DES are very extensive, such as abnormalities or cancers of the genital tract and breast, neurodevelopmental alterations, problems associated with socio-sexual behavior, and immune, pancreatic and cardiovascular disorders. Not only pregnant women but also their children and grandchildren have been affected. Epigenetic alterations have been detected, and intergenerational effects have been observed. More cohort follow-up studies are needed to establish if DES effects are transgenerational. Even though DES is not currently in use, its effects are still present, and families previously exposed and their later generations deserve the continuity of the research studies.
Collapse
Affiliation(s)
- Pilar Zamora-León
- Department of Preclinical Sciences, Faculty of Medicine, Universidad Católica del Maule, Talca 3460000, Chile
| |
Collapse
|
25
|
Mittal K, Henry PFP, Cornman RS, Maddox C, Basu N, Karouna-Renier NK. Sex- and Developmental Stage-Related Differences in the Hepatic Transcriptome of Japanese Quail (Coturnix japonica) Exposed to 17β-Trenbolone. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:2559-2570. [PMID: 34157788 DOI: 10.1002/etc.5143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
Endocrine-disrupting chemicals can cause transcriptomic changes that may disrupt biological processes associated with reproductive function including metabolism, transport, and cell growth. We investigated effects from in ovo and dietary exposure to 17β-trenbolone (at 0, 1, and 10 ppm) on the Japanese quail (Coturnix japonica) hepatic transcriptome. Our objectives were to identify differentially expressed hepatic genes, assess perturbations of biological pathways, and examine sex- and developmental stage-related differences. The number of significantly differentially expressed genes was higher in embryos than in adults. Male embryos exhibited greater differential gene expression than female embryos, whereas in adults, males and females exhibited similar numbers of differentially expressed genes (>2-fold). Vitellogenin and apovitellenin-1 were up-regulated in male adults exposed to 10 ppm 17β-trenbolone, and these birds also exhibited indications of immunomodulation. Functional grouping of differentially expressed genes identified processes including metabolism and transport of biomolecules, enzyme activity, and extracellular matrix interactions. Pathway enrichment analyses identified as perturbed peroxisome proliferator-activated receptor pathway, cardiac muscle contraction, gluconeogenesis, growth factor signaling, focal adhesion, and bile acid biosynthesis. One of the primary uses of 17β-trenbolone is that of a growth promoter, and these results identify effects on mechanistic pathways related to steroidogenesis, cell proliferation, differentiation, growth, and metabolism of lipids and proteins. Environ Toxicol Chem 2021;40:2559-2570. © 2021 SETAC. This article has been contributed to by US Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Krittika Mittal
- Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Paula F P Henry
- US Geological Survey, Eastern Ecological Science Center, Patuxent Research Refuge, Laurel, Maryland, USA
| | - Robert S Cornman
- US Geological Survey, Fort Collins Science Center, Fort Collins, Colorado, US
| | - Catherine Maddox
- US Geological Survey, Eastern Ecological Science Center, Patuxent Research Refuge, Beltsville, Maryland, USA
| | - Niladri Basu
- Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Natalie K Karouna-Renier
- US Geological Survey, Eastern Ecological Science Center, Patuxent Research Refuge, Beltsville, Maryland, USA
| |
Collapse
|
26
|
Gaspari L, Tessier B, Paris F, Bergougnoux A, Hamamah S, Sultan C, Kalfa N. Endocrine-Disrupting Chemicals and Disorders of Penile Development in Humans. Sex Dev 2021; 15:213-228. [PMID: 34438394 DOI: 10.1159/000517157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/10/2021] [Indexed: 01/09/2023] Open
Abstract
This paper reviews the current knowledge on the environmental effects on penile development in humans. The specific focus is on endocrine-disrupting chemicals (EDCs), a heterogeneous group of natural or manmade substances that interfere with endocrine function, and whether they can induce hypospadias and micropenis in male neonates. Epidemiological data and animal observations first raised suspicions about environmental effects, leading to the testis dysgenesis syndrome (TDS) hypothesis. More recent research has provided stronger indications that TDS may indeed be the result of the direct or indirect effects of EDCs. Drawing on epidemiological and toxicological studies, we also report on the effects of maternal diet and substances like pesticides, phthalates, bisphenol A, and polychlorinated biphenyls. Proximity to contamination hazards and occupational exposure are also suspected to contribute to the occurrence of hypospadias and micropenis. Lastly, the cumulative effects of EDCs and the possibility of transgenerational effects, with the penile development of subsequent generations being affected, raise concerns for long-term public health.
Collapse
Affiliation(s)
- Laura Gaspari
- Centre de Référence Maladies Rares du Développement Génital DEVGEN, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, Hôpital Arnaud-de-Villeneuve, CHU Montpellier, Université Montpellier, Montpellier, France.,Développement Embryonnaire Fertilité Environnement, INSERM 1203, Université Montpellier, Montpellier, France
| | - Benoit Tessier
- Département de Chirurgie Viscérale et Urologique Pédiatrique, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Institut Debrest de Santé Publique IDESP, UMR INSERM, Université Montpellier, Montpellier, France
| | - Françoise Paris
- Centre de Référence Maladies Rares du Développement Génital DEVGEN, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, Hôpital Arnaud-de-Villeneuve, CHU Montpellier, Université Montpellier, Montpellier, France.,Développement Embryonnaire Fertilité Environnement, INSERM 1203, Université Montpellier, Montpellier, France
| | - Anne Bergougnoux
- Centre de Référence Maladies Rares du Développement Génital DEVGEN, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Laboratoire de Génétique Moléculaire, PhyMedExp, INSERM, CNRS UMR, CHU Montpellier, Université Montpellier, Montpellier, France
| | - Samir Hamamah
- Développement Embryonnaire Fertilité Environnement, INSERM 1203, Université Montpellier, Montpellier, France.,Département de Biologie de la Reproduction, Biologie de la Reproduction/DPI et CECOS, CHU Montpellier, Université Montpellier, Montpellier, France
| | - Charles Sultan
- Centre de Référence Maladies Rares du Développement Génital DEVGEN, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France
| | - Nicolas Kalfa
- Centre de Référence Maladies Rares du Développement Génital DEVGEN, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Département de Chirurgie Viscérale et Urologique Pédiatrique, Hôpital Lapeyronie, CHU Montpellier, Université Montpellier, Montpellier, France.,Institut Debrest de Santé Publique IDESP, UMR INSERM, Université Montpellier, Montpellier, France
| |
Collapse
|
27
|
Kataoka C, Kashiwada S. Ecological Risks Due to Immunotoxicological Effects on Aquatic Organisms. Int J Mol Sci 2021; 22:8305. [PMID: 34361068 PMCID: PMC8347160 DOI: 10.3390/ijms22158305] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022] Open
Abstract
The immunotoxic effects of some anthropogenic pollutants on aquatic organisms are among the causes of concern over the presence of these pollutants in the marine environment. The immune system is part of an organism's biological defense necessarily for homeostasis. Thus, the immunotoxicological impacts on aquatic organisms are important to understand the effects of pollutant chemicals in the aquatic ecosystem. When aquatic organisms are exposed to pollutant chemicals with immunotoxicity, it results in poor health. In addition, aquatic organisms are exposed to pathogenic bacteria, viruses, parasites, and fungi. Exposure to pollutant chemicals has reportedly caused aquatic organisms to show various immunotoxic symptoms such as histological changes of lymphoid tissue, changes of immune functionality and the distribution of immune cells, and changes in the resistance of organisms to infection by pathogens. Alterations of immune systems by contaminants can therefore lead to the deaths of individual organisms, increase the general risk of infections by pathogens, and probably decrease the populations of some species. This review introduced the immunotoxicological impact of pollutant chemicals in aquatic organisms, including invertebrates, fish, amphibians, and marine mammals; described typical biomarkers used in aquatic immunotoxicological studies; and then, discussed the current issues on ecological risk assessment and how to address ecological risk assessment through immunotoxicology. Moreover, the usefulness of the population growth rate to estimate the immunotoxicological impact of pollution chemicals was proposed.
Collapse
Affiliation(s)
- Chisato Kataoka
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura, Gunma 374-0193, Japan
| | - Shosaku Kashiwada
- Department of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura, Gunma 374-0193, Japan;
- Research Centre for Life and Environmental Sciences, Toyo University, 1-1-1 Izumino, Itakura, Gunma 374-0193, Japan
| |
Collapse
|
28
|
Gaspari L, Paris F, Cassel-Knipping N, Villeret J, Verschuur A, Soyer-Gobillard MO, Carcopino-Tusoli X, Hamamah S, Kalfa N, Sultan C. Diethylstilbestrol exposure during pregnancy with primary clear cell carcinoma of the cervix in an 8-year-old granddaughter: a multigenerational effect of endocrine disruptors? Hum Reprod 2021; 36:82-86. [PMID: 33147330 DOI: 10.1093/humrep/deaa267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/10/2020] [Indexed: 11/13/2022] Open
Abstract
To date, vaginal/cervical clear cell adenocarcinoma (CCAC) has not been reported in the granddaughters of women treated with diethylstilbestrol (DES) during pregnancy. We present an 8-year-old girl with a history of severe vaginal bleeding who was diagnosed with cervical CCAC. She underwent fertility-sparing surgery and radiotherapy. No sign of recurrence was detected throughout a 10-year follow-up. Her grandmother had received DES therapy during pregnancy with the patient's mother. Although no direct causal link is demonstrated, this case raises for the first time, the hypothesis of multigenerational effects of DES in girls and strongly suggests the need to follow the granddaughters of DES-treated women.
Collapse
Affiliation(s)
- Laura Gaspari
- Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, Hôpital Arnaud-de-Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France.,Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier, Montpellier, France.,INSERM U 1203, Montpellier, France
| | - Françoise Paris
- Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, Hôpital Arnaud-de-Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France.,Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier, Montpellier, France.,INSERM U 1203, Montpellier, France
| | | | - Julia Villeret
- Hôpital Henri Duffaut, Service de Pathologie, Avignon, France
| | - Arnauld Verschuur
- Hôpital d'Enfants de la Timone, Centre de Cancérologie Pédiatrique, Marseille, France
| | | | | | - Samir Hamamah
- INSERM U 1203, Montpellier, France.,Département de Biologie de La Reproduction et DPI (ART/PGD), Hôpital A. de Villeneuve, CHU Montpellier et Université, Montpellier, France
| | - Nicolas Kalfa
- Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier, Montpellier, France.,Département de Chirurgie Pédiatrique, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier 1, Montpellier, France
| | - Charles Sultan
- Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, Hôpital Arnaud-de-Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France.,Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier, Montpellier, France
| |
Collapse
|
29
|
Vandenberg LN. Endocrine disrupting chemicals: strategies to protect present and future generations. Expert Rev Endocrinol Metab 2021; 16:135-146. [PMID: 33973826 DOI: 10.1080/17446651.2021.1917991] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022]
Abstract
Introduction: Endocrine-disrupting chemicals (EDCs) are chemicals that alter the actions of hormones. In the 21st Century, numerous expert groups of clinicians, scientists, and environmental activists have called for action to protect present and future generations from the harm induced by EDC exposures. These demands for regulatory responses come because of the strong weight of the evidence from epidemiology, wildlife, and controlled laboratory studies.Areas covered: In this review, we examine the conclusions drawn by experts from different scientific and medical disciplines. We also address several areas where recent findings or work has changed the landscape of EDC work including new approaches to identify and evaluate the evidence for EDCs using a key characteristics approach, the need to expand our understanding of vulnerable periods of development, and the increasing concern that traditional methods used to evaluate toxicity of environmental chemicals are insufficient for EDCs and how collaborative science could help to address these gaps.Expert opinion: The science is clear: there is more than enough evidence to demonstrate that EDCs affect the health of humans and wildlife. Waiting to act is a decision that puts the health of current and future generations at risk.
Collapse
Affiliation(s)
- Laura N Vandenberg
- School of Public Health & Health Sciences, Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA USA
| |
Collapse
|
30
|
Bacon ER, Brinton RD. Epigenetics of the developing and aging brain: Mechanisms that regulate onset and outcomes of brain reorganization. Neurosci Biobehav Rev 2021; 125:503-516. [PMID: 33657435 DOI: 10.1016/j.neubiorev.2021.02.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Brain development is a life-long process that encompasses several critical periods of transition, during which significant cognitive changes occur. Embryonic development, puberty, and reproductive senescence are all periods of transition that are hypersensitive to environmental factors. Rather than isolated episodes, each transition builds upon the last and is influenced by consequential changes that occur in the transition before it. Epigenetic marks, such as DNA methylation and histone modifications, provide mechanisms by which early events can influence development, cognition, and health outcomes. For example, parental environment influences imprinting patterns in gamete cells, which ultimately impacts gene expression in the embryo which may result in hypersensitivity to poor maternal nutrition during pregnancy, raising the risks for cognitive impairment later in life. This review explores how epigenetics induce and regulate critical periods, and also discusses how early environmental interactions prime a system towards a particular health outcome and influence susceptibility to disease or cognitive impairment throughout life.
Collapse
Affiliation(s)
- Eliza R Bacon
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; The Center for Precision Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; Center for Innovation in Brain Science, School of Medicine, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
31
|
Gaspari L, Paris F, Soyer-Gobillard MO, Hamamah S, Kalfa N, Sultan C. "Idiopathic" partial androgen insensitivity syndrome in 11 grandsons of women treated by diethylstilbestrol during gestation: a multi-generational impact of endocrine disruptor contamination? J Endocrinol Invest 2021; 44:379-381. [PMID: 32488725 DOI: 10.1007/s40618-020-01310-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/26/2020] [Indexed: 12/18/2022]
Affiliation(s)
- L Gaspari
- Unité d'Endocrinologie Pédiatrique, Département de Pédiatrie, Hôpital A. de Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France.
| | - F Paris
- Unité d'Endocrinologie Pédiatrique, Département de Pédiatrie, Hôpital A. de Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France
| | - M-O Soyer-Gobillard
- Association Hhorages-France, CRNS, Sorbonne Université, F-Asnières-sur-Oise, France
| | - S Hamamah
- Département de Biologie de La Reproduction et DPI (ART/PGD), Hôpital A. de Villeneuve, CHU Montpellier et Université Montpellier, INSERM U 1203, Montpellier, France
| | - N Kalfa
- Département de Chirurgie et Urologie Pédiatrique et Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, CHU Montpellier et Université Montpellier, Montpellier, France
| | - C Sultan
- Unité d'Endocrinologie Pédiatrique, Département de Pédiatrie, Hôpital A. de Villeneuve, CHU Montpellier et Université Montpellier, Montpellier, France
| |
Collapse
|
32
|
N’Diaye J, Poorahong S, Hmam O, Jiménez GC, Izquierdo R, Siaj M. Reduced Graphene Oxide-Based Foam as an Endocrine Disruptor Adsorbent in Aqueous Solutions. MEMBRANES 2020; 10:E340. [PMID: 33202720 PMCID: PMC7696572 DOI: 10.3390/membranes10110340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 11/24/2022]
Abstract
A stable and magnetic graphene oxide (GO) foam-polyethyleneimine-iron nanoparticle (GO-PEI-FeNPs) composite has been fabricated for removal of endocrine disruptors-bisphenol A, progesterone and norethisterone-from aqueous solution. The foam with porous and hierarchical structures was synthesized by reduction of graphene oxide layers coupled with co-precipitation of iron under a hydrothermal system using polyethyleneimine as a cross linker. The presence of magnetic iron nanoparticles facilitates the separation process after decontamination. The foam was fully characterized by surface and structural scanning electron microscopy, Fourier transform infrared spectroscopy, Raman spectroscopy and X-ray photoelectron spectroscopy. The foam exhibits a high adsorption capacity, and the maximum adsorption percentages are 68%, 49% and 80% for bisphenol A, progesterone and norethisterone, respectively. The adsorption process of bisphenol A is explained according to the Langmuir model, whereas the Freundlich model was used for progesterone and norethisterone adsorption.
Collapse
Affiliation(s)
- Jeanne N’Diaye
- Department of Chemistry and Biochemistry, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (J.N.); (S.P.); (O.H.); (G.C.J.)
| | - Sujittra Poorahong
- Department of Chemistry and Biochemistry, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (J.N.); (S.P.); (O.H.); (G.C.J.)
| | - Ons Hmam
- Department of Chemistry and Biochemistry, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (J.N.); (S.P.); (O.H.); (G.C.J.)
| | - Gastón Contreras Jiménez
- Department of Chemistry and Biochemistry, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (J.N.); (S.P.); (O.H.); (G.C.J.)
- Laboratorio de Microdisección Láser, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico 04510, Mexico
| | - Ricardo Izquierdo
- École de technologie supérieure, Université du Québec à Montréal, Montréal, QC H3C 1K3, Canada;
| | - Mohamed Siaj
- Department of Chemistry and Biochemistry, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (J.N.); (S.P.); (O.H.); (G.C.J.)
| |
Collapse
|
33
|
Nagahama Y, Chakraborty T, Paul-Prasanth B, Ohta K, Nakamura M. Sex determination, gonadal sex differentiation, and plasticity in vertebrate species. Physiol Rev 2020; 101:1237-1308. [PMID: 33180655 DOI: 10.1152/physrev.00044.2019] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A diverse array of sex determination (SD) mechanisms, encompassing environmental to genetic, have been found to exist among vertebrates, covering a spectrum from fixed SD mechanisms (mammals) to functional sex change in fishes (sequential hermaphroditic fishes). A major landmark in vertebrate SD was the discovery of the SRY gene in 1990. Since that time, many attempts to clone an SRY ortholog from nonmammalian vertebrates remained unsuccessful, until 2002, when DMY/dmrt1by was discovered as the SD gene of a small fish, medaka. Surprisingly, however, DMY/dmrt1by was found in only 2 species among more than 20 species of medaka, suggesting a large diversity of SD genes among vertebrates. Considerable progress has been made over the last 3 decades, such that it is now possible to formulate reasonable paradigms of how SD and gonadal sex differentiation may work in some model vertebrate species. This review outlines our current understanding of vertebrate SD and gonadal sex differentiation, with a focus on the molecular and cellular mechanisms involved. An impressive number of genes and factors have been discovered that play important roles in testicular and ovarian differentiation. An antagonism between the male and female pathway genes exists in gonads during both sex differentiation and, surprisingly, even as adults, suggesting that, in addition to sex-changing fishes, gonochoristic vertebrates including mice maintain some degree of gonadal sexual plasticity into adulthood. Importantly, a review of various SD mechanisms among vertebrates suggests that this is the ideal biological event that can make us understand the evolutionary conundrums underlying speciation and species diversity.
Collapse
Affiliation(s)
- Yoshitaka Nagahama
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Faculty of Biological Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Tapas Chakraborty
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan.,Karatsu Satellite of Aqua-Bioresource Innovation Center, Kyushu University, Karatsu, Japan
| | - Bindhu Paul-Prasanth
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidapeetham, Kochi, Kerala, India
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan
| | - Masaru Nakamura
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Research Center, Okinawa Churashima Foundation, Okinawa, Japan
| |
Collapse
|
34
|
New frontiers of developmental endocrinology opened by researchers connecting irreversible effects of sex hormones on developing organs. Differentiation 2020; 118:4-23. [PMID: 33189416 DOI: 10.1016/j.diff.2020.10.003] [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] [Received: 10/02/2020] [Revised: 10/12/2020] [Accepted: 10/25/2020] [Indexed: 01/17/2023]
Abstract
In the early 1960's, at Professor Bern's laboratory, University of California, Berkeley) in the US, Takasugi discovered ovary-independent, persistent vaginal changes in mice exposed neonatally to estrogen, which resulted in vaginal cancer later in life. Reproductive abnormalities in rodents were reported as a result of perinatal exposure to various estrogenic chemicals. Ten years later, vaginal cancers were reported in young women exposed in utero to the synthetic estrogen diethylstilbestrol (DES) and this has been called the "DES syndrome". The developing organism is particularly sensitive to developmental exposure to estrogens inducing long-term changes in various organs including the reproductive organs. The molecular mechanism underlying the persistent vaginal changes induced by perinatal estrogen exposure was partly demonstrated. Persistent phosphorylation and sustained expression of EGF-like growth factors, lead to estrogen receptor α (ESR1) activation, and then persistent vaginal epithelial cell proliferation. Agents which are weakly estrogenic by postnatal criteria may have major developmental effects, especially during a critical perinatal period. The present review outlines various studies conducted by four generations of investigators all under the influence of Prof. Bern. The studies include reports of persistent changes induced by neonatal androgen exposure, analyses of estrogen responsive genes, factors determining epithelial differentiation in the Müllerian duct, ESR and growth factor signaling, and polyovular follicles in mammals. This review is then expanded to the studies on the effects of environmental estrogens on wildlife and endocrine disruption in Daphnids.
Collapse
|
35
|
Perceptions, Knowledge, and Practices Concerning Indoor Environmental Pollution of Parents or Future Parents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17207669. [PMID: 33096680 PMCID: PMC7589174 DOI: 10.3390/ijerph17207669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/16/2022]
Abstract
Indoor pollutants can have short- and long-term health effects, especially if exposure occurs during prenatal life or early childhood. This study describe the perceptions, knowledge, and practices of adults concerning indoor environmental pollution. Adults of 18 to 45 years of age were recruited in the department of Ille-et-Vilaine (Brittany-France) in 2019 through a stratified random draw in the waiting rooms of general practitioners (GPs) (n = 554) who completed a self-questionnaire. The 71% who had already heard of this type of pollution were older (p = 0.001), predominantly women (p = 0.007), not expecting a baby (p = 0.005), and had a higher knowledge score (p < 0.001). The average knowledge score was 6.6 ± 6.6 out of 11, which was higher for participants living in a couple and with a higher level of education (p < 0.001). Some practices were well implemented (>80% of participants) (aeration during renovation) whereas others were insufficiently practiced (<60% of participants) (paying attention to the composition of cosmetic products). Factors associated differed depending on the frequency of integration: living in a couple and having a child for well implemented practices and educational level, knowledge level, and perception for those under implemented. Knowledge must be improved to modify perceptions and certain practices, making sure not to increase social inequalities in health.
Collapse
|
36
|
Nanjappa MK, Medrano TI, Mesa AM, Ortega MT, Caldo PD, Mao J, Kinkade JA, Levin ER, Rosenfeld CS, Cooke PS. Mice lacking membrane estrogen receptor 1 are protected from reproductive pathologies resulting from developmental estrogen exposure†. Biol Reprod 2020; 101:392-404. [PMID: 31141131 DOI: 10.1093/biolre/ioz090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/22/2019] [Indexed: 01/06/2023] Open
Abstract
Both membrane and nuclear fractions of estrogen receptor 1 (ESR1) mediate 17β-estradiol (E2) actions. Mice expressing nuclear (n)ESR1 but lacking membrane (m)ESR1 (nuclear-only estrogen receptor 1 [NOER] mice) show reduced E2 responsivity and reproductive abnormalities culminating in adult male and female infertility. Using this model, we investigated whether reproductive pathologies caused by the synthetic estrogen diethylstilbestrol (DES) are mitigated by mESR1 ablation. Homozygous and heterozygous wild-type (WT and HET, respectively) and NOER male and female mice were subcutaneously injected with DES (1 mg/kg body weight [BW]) or vehicle daily from postnatal day (PND) 1-5. Uterine histology was assessed in select DES-treated females at PND 5, whereas others were ovariectomized at PND 60 and treated with E2 (10 μg/kg BW) or vehicle 2 weeks later. Neonatal DES exposure resulted in ovary-independent epithelial proliferation in the vagina and uterus of WT but not NOER females. Neonatal DES treatment also induced ovary-independent adult expression of classical E2-induced transcripts (e.g., lactoferrin [Ltf] and enhancer of zeste homolog 2 [Ezh2]) in WT but not NOER mice. At PND 90, DES-treated WT and HET males showed smaller testes and a high incidence of bacterial pyogranulomatous inflammation encompassing the testes, epididymis and occasionally the ductus deferens with spread to lumbar lymph nodes; such changes were largely absent in NOER males. Results indicate that male and female NOER mice are protected from deleterious effects of neonatal DES, and thus mESR1 signaling is required for adult manifestation of DES-induced reproductive pathologies in both sexes.
Collapse
Affiliation(s)
- Manjunatha K Nanjappa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Theresa I Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Ana M Mesa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| | - Madison T Ortega
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Paul D Caldo
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Jiude Mao
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Jessica A Kinkade
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, California, USA.,Department of Veterans Affairs Medical Center, Long Beach, Long Beach, California, USA
| | - Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,Biomedical Sciences, University of Missouri, Columbia, Missouri, USA.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri, USA.,MU Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | - Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
37
|
Rattan S, Flaws JA. The epigenetic impacts of endocrine disruptors on female reproduction across generations†. Biol Reprod 2020; 101:635-644. [PMID: 31077281 DOI: 10.1093/biolre/ioz081] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/18/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Humans and animals are repeatedly exposed to endocrine disruptors, many of which are ubiquitous in the environment. Endocrine disruptors interfere with hormone action; thus, causing non-monotonic dose responses that are atypical of standard toxicant exposures. The female reproductive system is particularly susceptible to the effects of endocrine disruptors. Likewise, exposures to endocrine disruptors during developmental periods are particularly concerning because programming during development can be adversely impacted by hormone level changes. Subsequently, developing reproductive tissues can be predisposed to diseases in adulthood and these diseases can be passed down to future generations. The mechanisms of action by which endocrine disruptors cause disease transmission to future generations are thought to include epigenetic modifications. This review highlights the effects of endocrine disruptors on the female reproductive system, with an emphasis on the multi- and transgenerational epigenetic effects of these exposures.
Collapse
Affiliation(s)
- Saniya Rattan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| |
Collapse
|
38
|
Toprani SM, Kelkar Mane V. Role of DNA damage and repair mechanisms in uterine fibroid/leiomyomas: a review. Biol Reprod 2020; 104:58-70. [PMID: 32902600 DOI: 10.1093/biolre/ioaa157] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/09/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
There has been a significant annual increase in the number of cases of uterine leiomyomas or fibroids (UF) among women of all races and ages across the world. A fortune is usually spent by the healthcare sector for fibroid-related treatments and management. Molecular studies have established the higher mutational heterogeneity in UF as compared to normal myometrial cells. The contribution of DNA damage and defects in repair responses further increases the mutational burden on the cells. This in turn leads to genetic instability, associated with cancer risk and other adverse reproductive health outcomes. Such and many more growing bodies of literature have highlighted the genetic/molecular, biochemical and clinical aspects of UF; none the less there appear to be a lacuna bridging the bench to bed gap in addressing and preventing this disease. Presented here is an exhaustive review of not only the molecular mechanisms underlying the predisposition to the disease but also possible strategies to effectively diagnose, prevent, manage, and treat this disease.
Collapse
Affiliation(s)
- Sneh M Toprani
- Department of Biotechnology, University of Mumbai, Kalina, Mumbai, India
| | - Varsha Kelkar Mane
- Department of Biotechnology, University of Mumbai, Kalina, Mumbai, India
| |
Collapse
|
39
|
Salamat N, Derakhshesh N. Oxidative stress in liver cell culture from mullet, Liza klunzingeri, induced by short-term exposure to benzo[a]pyrene and nonylphenol. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1183-1197. [PMID: 32166615 DOI: 10.1007/s10695-020-00783-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/24/2020] [Indexed: 05/22/2023]
Abstract
The present investigation aimed to use primary liver cell culture obtained from mullet, Liza klunzingeri, to evaluate the toxic effects of benzo[a]pyrene (BaP) and nonylphenol (NP) on the antioxidant defense system. Liver samples taken from 20 L. klunzingeri were digested with 0.1% collagenase IV. The digested cells were then moved to Leibovitz L-15 culture medium and incubated at 25 °C for 2 weeks. 10-5 mol/l of BaP and 10-4 mol/l of NP were considered as the half maximal inhibitory concentration (IC50). Cells were then incubated with L-15 medium containing BaP (0[control], 10-6,2 × 10-6,3 × 10-6 mol/l) and NP (0[control],10-5,2 × 10-5,3 × 10-5 mol/l), and sampling was performed after 6, 12, and 24 h of incubation for measurement of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), lipid peroxidation (LPO), total antioxidant power, and total protein. The lowest concentration of BaP and NP did not have considerable toxic effects on cultivated hepatocytes. The activities of SOD, CAT, GPx, LPO, total antioxidant power, and total protein changed dose-dependently in cells treated with BaP and NP. In conclusion, based on the results, short-term exposure to BaP and NP induced the oxidative stress in cultivated liver cells of L. klunzingeri. The toxicity of both pollutants is mainly because of the induction of the reactive oxygen species (ROS), which lead to cell membrane disruption, damage of cellular metabolism, and interference with cellular macromolecules.
Collapse
Affiliation(s)
- Negin Salamat
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, Khorramshahr, Iran.
| | - Negin Derakhshesh
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, Khorramshahr, Iran
| |
Collapse
|
40
|
Ottolina J, Schimberni M, Makieva S, Bartiromo L, Fazia T, Bernardinelli L, Viganò P, Candiani M, Gentilini D. Early-life factors, in-utero exposures and endometriosis risk: a meta-analysis. Reprod Biomed Online 2020; 41:279-289. [PMID: 32532666 DOI: 10.1016/j.rbmo.2020.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/25/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This meta-analysis aimed to offer a general picture of the available data on the effects of early-life factors on the risk of developing endometriosis in adult life. An advanced, systematic search of the online medical databases PubMed, EMBASE and CINAHL was limited to full-length manuscripts published in English in peer-reviewed journals up to February 2019. Log of relative risk (RR) was employed to calculate the pooled effect sizes using both fixed and random effects modelling and I-squared tests to assess heterogeneity. Funnel plots were used to investigation publication bias. The meta-analysis was registered in PROSPERO (ID CRD42019138668). Six studies that included a total of 2360 women affected by endometriosis were analysed. The pooled results showed that the risk of developing endometriosis in adult life was significantly increased by being born prematurely (logRR 0.21, 95% CI -0.03 to 0.40), having a low birthweight (logRR 0.35, 95% CI -0.15 to 0.54), being formula-fed (logRR 0.65, 95% CI -0.35 to 0.95) and having been exposed to diethylstilbestrol (DES) in utero (logRR 0.65, 95% CI 0.26 to 1.04. Among intrauterine and early neonatal exposures, prematurity, birthweight, formula feeding and DES were risk factors for the development of endometriosis in adult life.
Collapse
Affiliation(s)
- Jessica Ottolina
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy.
| | - Matteo Schimberni
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Sofia Makieva
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Candiani
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Istituto Auxologico Italiano IRCCS, Bioinformatics and Statistical Genomics Unit, Cusano MilaninoMilan, Italy
| |
Collapse
|
41
|
Hu XL, Tang YY, Kwok ML, Chan KM, Chu KH. Impact of juvenile hormone analogue insecticides on the water flea Moina macrocopa: Growth, reproduction and transgenerational effect. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 220:105402. [PMID: 31927065 DOI: 10.1016/j.aquatox.2020.105402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/20/2019] [Accepted: 01/01/2020] [Indexed: 06/10/2023]
Abstract
The increasing quantities of insecticides that leach into water bodies severely affect the health of the aquatic environment. Juvenile hormone analogue (JHA) insecticides are endocrine disrupters that interfere with hormonal activity in insects by mimicking juvenile hormones (JHs). Because the structure and functions of methyl farnesoate in crustaceans are similar to the insect JHs, exogenous JHA insecticides may cause adverse effects on the growth and reproduction in crustaceans similar to those observed in insects. This study examined the toxic effects of two JHA insecticides, methoprene and fenoxycarb, on the water flea Moina macrocopa. The 24-h and 48-h LC50 values for fenoxycarb and methoprene were 0.53 and 0.32 mg/L and 0.70 and 0.54 mg/L, respectively. Chronic exposure to the two JHAs caused a series of toxic effects in M. macrocopa, including shortening of life expectancy, repression of body growth, reduction in fecundity, and disturbed the expression of genes involved in the JH signaling pathway, in cuticle development, and in the carbohydrate, amino acid, and ATP metabolic processes. Moreover, JHA exposure impaired the growth and reproduction of the offspring of M. macrocopa exposed to JHAs, even when the neonates were not exposed to the chemicals. In addition, changes in the expression of genes related to histone methylation indicate that epigenetic changes may promote transgenerational impairment in M. macrocopa. These results demonstrate the toxic effects of fenoxycarb and methoprene on non-target aquatic organisms. The damages done by these JHA insecticides to the aquatic environment is worthy of our attention and further studies.
Collapse
Affiliation(s)
- Xue Lei Hu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yuan Yuan Tang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Man Long Kwok
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - King Ming Chan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ka Hou Chu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
42
|
Li K, Liszka M, Zhou C, Brehm E, Flaws JA, Nowak RA. Prenatal exposure to a phthalate mixture leads to multigenerational and transgenerational effects on uterine morphology and function in mice. Reprod Toxicol 2020; 93:178-190. [PMID: 32126281 DOI: 10.1016/j.reprotox.2020.02.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/21/2020] [Accepted: 02/26/2020] [Indexed: 12/21/2022]
Abstract
Phthalates are commonly used plasticizers and additives that are found in plastic containers, children's toys and medical equipment. Phthalates are classified as endocrine-disrupting chemicals and exposure to phthalates has been associated with several human health risks including reproductive defects. Most studies focus on a single phthalate; however, humans are exposed to a mixture of phthalates daily. We hypothesized that prenatal exposure to an environmentally relevant phthalate mixture would lead to changes in uterine morphology and function in mice in a multi-generational manner. To test this hypothesis, pregnant CD-1 dams were orally dosed with vehicle or a phthalate mixture (20 μg/kg/day, 200 μg/kg/day, 200 mg/kg/day, and 500 mg/kg/day) from gestational day 10.5 to parturition. The mixture contained 35 % diethyl phthalate, 21 % di-(2-ethylhexyl) phthalate, 15 % dibutyl phthalate, 15 % diisononyl phthalate, 8% diisobutyl phthalate, and 5% benzylbutyl phthalate. The F1 pups were maintained and mated to produce two more generations (F2 and F3). At the age of 13 months, all females were euthanized and tissue samples were collected in diestrus. Our results showed that exposure to a phthalate mixture caused a decrease in progesterone levels in the treated groups in the F2 generation. The 200 mg/kg/day treatment group showed a decreased and increased luminal epithelial cell proliferation in the F1 and F2 generations respectively. In addition, these mice in the F2 generation had reduced Hand2 expression in the sub-epithelial stroma compared to the controls. A higher incidence of multilayered luminal epithelium and large dilated endometrial glands were observed in the phthalate mixture exposed groups in all generations. The mixture also caused a higher incidence of smooth muscle actin expression and collagen deposition in the endometrium compared to controls. Collectively, our results demonstrate that prenatal exposure to an environmentally relevant phthalate mixture can have adverse effects on female reproductive functions.
Collapse
Affiliation(s)
- Kailiang Li
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Monika Liszka
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Changqing Zhou
- Department of Comparative Biosciences, University of Illinois, Urbana, IL, United States
| | - Emily Brehm
- Department of Comparative Biosciences, University of Illinois, Urbana, IL, United States
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois, Urbana, IL, United States
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States.
| |
Collapse
|
43
|
Oestrogenic Endocrine Disruptors in the Placenta and the Fetus. Int J Mol Sci 2020; 21:ijms21041519. [PMID: 32102189 PMCID: PMC7073155 DOI: 10.3390/ijms21041519] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/26/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) are exogenous substances that interfere with the stability and regulation of the endocrine system of the body or its offspring. These substances are generally stable in chemical properties, not easy to be biodegraded, and can be enriched in organisms. In the past half century, EDCs have gradually entered the food chain, and these substances have been frequently found in maternal blood. Perinatal maternal hormone levels are unstable and vulnerable to EDCs. Some EDCs can affect embryonic development through the blood-fetal barrier and cause damage to the neuroendocrine system, liver function, and genital development. Some also effect cross-generational inheritance through epigenetic mechanisms. This article mainly elaborates the mechanism and detection methods of estrogenic endocrine disruptors, such as bisphenol A (BPA), organochlorine pesticides (OCPs), diethylstilbestrol (DES) and phthalates (PAEs), and their effects on placenta and fetal health in order to raise concerns about the proper use of products containing EDCs during pregnancy and provide a reference for human health.
Collapse
|
44
|
Adam AHB, de Haan LHJ, Estruch IM, Hooiveld GJEJ, Louisse J, Rietjens IMCM. Estrogen receptor alpha (ERα)-mediated coregulator binding and gene expression discriminates the toxic ERα agonist diethylstilbestrol (DES) from the endogenous ERα agonist 17β-estradiol (E2). Cell Biol Toxicol 2020; 36:417-435. [PMID: 32088792 PMCID: PMC7505815 DOI: 10.1007/s10565-020-09516-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/28/2020] [Indexed: 12/29/2022]
Abstract
Diethylstilbestrol (DES) is a synthetic estrogen and proven human teratogen and carcinogen reported to act via the estrogen receptor α (ERα). Since the endogenous ERα ligand 17β-estradiol (E2) does not show these adverse effects to a similar extent, we hypothesized that DES' interaction with the ERα differs from that of E2. The current study aimed to investigate possible differences between DES and E2 using in vitro assays that detect ERα-mediated effects, including ERα-mediated reporter gene expression, ERα-mediated breast cancer cell (T47D) proliferation and ERα-coregulator interactions and gene expression in T47D cells. Results obtained indicate that DES and E2 activate ERα-mediated reporter gene transcription and T47D cell proliferation in a similar way. However, significant differences between DES- and E2-induced binding of the ERα to 15 coregulator motifs and in transcriptomic signatures obtained in the T47D cells were observed. It is concluded that differences observed in binding of the ERα with several co-repressor motifs, in downregulation of genes involved in histone deacetylation and DNA methylation and in upregulation of CYP26A1 and CYP26B1 contribute to the differential effects reported for DES and E2.
Collapse
Affiliation(s)
- Aziza Hussein Bakheit Adam
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands.
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Ignacio Miro Estruch
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Guido J E J Hooiveld
- Division of Human Nutrition and Health, Wageningen University and Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Jochem Louisse
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, PO Box 8000, 6700 EA, Wageningen, The Netherlands
| |
Collapse
|
45
|
Pino MTL, Ronchetti SA, Cordeiro G, Bollani S, Duvilanski BH, Cabilla JP. Soluble Guanylyl Cyclase Alpha1 Subunit: A New Marker for Estrogenicity of Endocrine Disruptor Compounds. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2019; 38:2719-2728. [PMID: 31499574 DOI: 10.1002/etc.4591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/09/2019] [Accepted: 09/04/2019] [Indexed: 06/10/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) include widespread naturally occurring and synthetic substances in the environment that adversely affect humans and wildlife. Because of the increasing numbers of EDCs, screening methods and ideal biomarkers to determine EDC potencies at relevant environmental concentrations need to be drastically improved. Soluble guanylyl cyclase α1 subunit (sGCα1) is an abundant cytosolic protein ubiquitously expressed in most tissues. We previously showed that sGCα1 is specifically and highly up-regulated by estrogen (E2) in vivo and in vitro, even though it lacks estrogen-responsive elements. The aim of the present study was to evaluate sGCα1 protein expression as a potential marker for xenoestrogenic EDC exposure in the E2-responsive lactosomatotroph-derived pituitary cell line GH3. Cells were incubated with a wide variety of EDCs such as heavy metals and a metalloid, synthetic E2 derivatives, plastic byproducts, and pesticides at a range of doses including those with proven xenoestrogenic activity. We demonstrated that E2 increased sGCα1 expression in GH3 cells as well as in other E2-responsive tumor cell lines. Moreover, this effect was fully dependent on estrogen receptor (ER) activation. Importantly, sGCα1 protein levels were strongly up-regulated by all the EDCs tested, even by those exhibiting low or null ER binding capacity. We provide evidence that the in vitro sGCα1 protein assay may be a very sensitive and powerful tool to identify compounds with estrogenic activity, which could improve current mammalian-based screening methods. Environ Toxicol Chem 2019;38:2719-2728. © 2019 SETAC.
Collapse
Affiliation(s)
- María Teresa L Pino
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| | - Sonia A Ronchetti
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| | - Georgina Cordeiro
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| | - Sabrina Bollani
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| | - Beatriz H Duvilanski
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| | - Jimena P Cabilla
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
46
|
Belcher SM, Cline JM, Conley J, Groeters S, Jefferson WN, Law M, Mackey E, Suen AA, Williams CJ, Dixon D, Wolf JC. Endocrine Disruption and Reproductive Pathology. Toxicol Pathol 2019; 47:1049-1071. [PMID: 31833458 PMCID: PMC8008741 DOI: 10.1177/0192623319879903] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During the past 20 years, investigations involving endocrine active substances (EAS) and reproductive toxicity have dominated the landscape of ecotoxicological research. This has occurred in concert with heightened awareness in the scientific community, general public, and governmental entities of the potential consequences of chemical perturbation in humans and wildlife. The exponential growth of experimentation in this field is fueled by our expanding knowledge into the complex nature of endocrine systems and the intricacy of their interactions with xenobiotic agents. Complicating factors include the ever-increasing number of novel receptors and alternate mechanistic pathways that have come to light, effects of chemical mixtures in the environment versus those of single EAS laboratory exposures, the challenge of differentiating endocrine disruption from direct cytotoxicity, and the potential for transgenerational effects. Although initially concerned with EAS effects chiefly in the thyroid glands and reproductive organs, it is now recognized that anthropomorphic substances may also adversely affect the nervous and immune systems via hormonal mechanisms and play substantial roles in metabolic diseases, such as type 2 diabetes and obesity.
Collapse
Affiliation(s)
| | - J. Mark Cline
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Mac Law
- North Carolina State College of Veterinary Medicine, Raleigh, NC, USA
| | - Emily Mackey
- Michigan State University, East Lansing, MI, USA
| | | | | | | | | |
Collapse
|
47
|
Suen AA, Jefferson WN, Wood CE, Williams CJ. SIX1 Regulates Aberrant Endometrial Epithelial Cell Differentiation and Cancer Latency Following Developmental Estrogenic Chemical Exposure. Mol Cancer Res 2019; 17:2369-2382. [PMID: 31597742 DOI: 10.1158/1541-7786.mcr-19-0475] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/08/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Early-life exposure to estrogenic chemicals can increase cancer risk, likely by disrupting normal patterns of cellular differentiation. Female mice exposed neonatally to the synthetic estrogen diethylstilbestrol (DES) develop metaplastic and neoplastic uterine changes as adults. Abnormal endometrial glands express the oncofetal protein sine oculis homeobox 1 (SIX1) and contain cells with basal [cytokeratin (CK)14+/18-] and poorly differentiated features (CK14+/18+), strongly associating SIX1 with aberrant differentiation and cancer. Here, we tested whether SIX1 expression is necessary for abnormal endometrial differentiation and DES-induced carcinogenesis by using Pgr-cre to generate conditional knockout mice lacking uterine Six1 (Six1 d/d). Interestingly, corn oil (CO) vehicle-treated Six1 d/d mice develop focal endometrial glandular dysplasia and features of carcinoma in situ as compared with CO wild-type Six1 (Six1 +/+) mice. Furthermore, Six1 d/d mice neonatally exposed to DES had a 42% higher incidence of endometrial cancer relative to DES Six1 +/+ mice. Although DES Six1 d/d mice had >10-fold fewer CK14+/18- basal cells within the uterine horns as compared with DES Six1 +/+ mice, the appearance of CK14+/18+ cells remained a feature of neoplastic lesions. These findings suggest that SIX1 is required for normal endometrial epithelial differentiation, CK14+/18+ cells act as a cancer progenitor population, and SIX1 delays DES-induced endometrial carcinogenesis by promoting basal differentiation of CK14+/18+ cells. In human endometrial biopsies, 35% of malignancies showed CK14+/18+ expression, which positively correlated with tumor stage and grade and was not present in normal endometrium. IMPLICATIONS: Aberrant epithelial differentiation is a key feature in both the DES mouse model of endometrial cancer and human endometrial cancer. The association of CK14+/18+ cells with human endometrial cancer provides a novel cancer biomarker and could lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Alisa A Suen
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina. .,Oak Ridge Institute for Science and Education (ORISE) participant in the Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Wendy N Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Charles E Wood
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
48
|
Maeda N, Okumura K, Yamaguchi K, Haeno S, Yasui Y, Kimura N, Ieko T, Miyasho T, Yokota H. Rapid prolactin induction in adult male rats after treatment with diethylstilbestrol. J Neuroendocrinol 2019; 31:e12769. [PMID: 31283846 DOI: 10.1111/jne.12769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
Abstract
Diethylstilbestrol (DES) is a synthetic oestrogen known to disrupt the endocrine system and to cause reproductive toxicity mediated via the hypothalamic-pituitary-adrenal axis; however, its molecular mechanism of action is poorly understood. In the present study, we found that, after only 1 week of exposure to DES, blood testosterone dramatically decreased and that this decrease was associated with a strong induction of prolactin (PRL). Even with the increase in PRL, the luteinising hormone and follicle-stimulating hormone mRNAs slightly decreased. Our results show that, after 48 hours of a single dose of DES, there was a six-fold increase in PRL expression. After exploring the upstream mechanisms, we determined that dopamine, which inhibits PRL secretion in male rats, did not decrease in the pituitary gland of DES-treated rats, whereas vasoactive intestinal peptide (VIP), which mediates the acute release of PRL, was elevated. Serotonin (5-HT) increased in the brain of male rats 24 hours after a single DES treatment; however, PRL, VIP or 5-HT was not induced by DES in female rats. Our results indicate that DES induces the expression of pituitary PRL in male rats by stimulating VIP in the hypothalamus and 5-HT in the central nervous system.
Collapse
Affiliation(s)
- Naoyuki Maeda
- Laboratory of Meat Science and Technology, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido, Japan
- Safety Research Institute for Chemical Compounds Co., Ltd, Sapporo, Japan
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
- Japan Meat Science and Technology Institute, Shibuya-ku, Tokyo, Japan
| | - Kanako Okumura
- Safety Research Institute for Chemical Compounds Co., Ltd, Sapporo, Japan
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
| | - Kousuke Yamaguchi
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
- Graduate School of Arts and Sciences, University of Tokyo, Meguro, Tokyo, Japan
| | - Satoko Haeno
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
| | - Yumiko Yasui
- Laboratory of Veterinary Physiology and Nutrition, Department of Veterinary Science, Rakuno Gakuen University, Hokkaido, Japan
| | - Nobuya Kimura
- Public Nutrition, Department of Food Science and Human Wellness, Rakuno Gakuen University, Hokkaido, Japan
| | - Takahiro Ieko
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
| | - Taku Miyasho
- Laboratory of Animal Biological Responses, Department of Veterinary Science, Rakuno Gakuen University, Hokkaido, Japan
| | - Hiroshi Yokota
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan
| |
Collapse
|
49
|
Prusinski Fernung LE, Yang Q, Sakamuro D, Kumari A, Mas A, Al-Hendy A. Endocrine disruptor exposure during development increases incidence of uterine fibroids by altering DNA repair in myometrial stem cells. Biol Reprod 2019; 99:735-748. [PMID: 29688260 DOI: 10.1093/biolre/ioy097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/20/2018] [Indexed: 12/20/2022] Open
Abstract
Despite the major negative impact uterine fibroids (UFs) have on female reproductive health, little is known about early events that initiate development of these tumors. Somatic fibroid-causing mutations in mediator complex subunit 12 (MED12), the most frequent genetic alterations in UFs (up to 85% of tumors), are implicated in transforming normal myometrial stem cells (MSCs) into tumor-forming cells, though the underlying mechanism(s) leading to these mutations remains unknown. It is well accepted that defective DNA repair increases the risk of acquiring tumor-driving mutations, though defects in DNA repair have not been explored in UF tumorigenesis. In the Eker rat UF model, a germline mutation in the Tsc2 tumor suppressor gene predisposes to UFs, which arise due to "second hits" in the normal allele of this gene. Risk for developing these tumors is significantly increased by early-life exposure to endocrine-disrupting chemicals (EDCs), suggesting increased UF penetrance is modulated by early drivers for these tumors. We analyzed DNA repair capacity using analyses of related gene and protein expression and DNA repair function in MSCs from adult rats exposed during uterine development to the model EDC diethylstilbestrol. Adult MSCs isolated from developmentally exposed rats demonstrated decreased DNA end-joining ability, higher levels of DNA damage, and impaired ability to repair DNA double-strand breaks relative to MSCs from age-matched, vehicle-exposed rats. These data suggest that early-life developmental EDC exposure alters these MSCs' ability to repair and reverse DNA damage, providing a driver for acquisition of mutations that may promote the development of these tumors in adult life.
Collapse
Affiliation(s)
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daitoku Sakamuro
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, USA
| | - Alpana Kumari
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, USA
| | - Aymara Mas
- Reproductive Medicine Research Group, La Fe Health Research Institute, Valencia, Spain.,Igenomix, Paterna, Valencia, Spain.,Department of Obstetrics and Gynecology, Valencia University and INCLIVA, Valencia, Spain
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
50
|
Sonavane M, Gassman NR. Bisphenol A co-exposure effects: a key factor in understanding BPA's complex mechanism and health outcomes. Crit Rev Toxicol 2019; 49:371-386. [PMID: 31256736 DOI: 10.1080/10408444.2019.1621263] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bisphenol A (BPA) is an environmental endocrine disrupting chemical widely used in the production of consumer products, such as polycarbonate plastics, epoxies, and thermal receipt paper. Human exposure to BPA is ubiquitous due to its high-volume production and use. BPA exposure has been associated with obesity, diabetes, reproductive disorders, and cancer. Yet, the molecular mechanisms or modes of action underlying these disease outcomes are poorly understood due to the pleiotropic effects induced by BPA. A further confounding factor in understanding BPA's impact on human health is that co-exposure of BPA with endogenous and exogenous agents occurs during the course of daily life. Studies investigating BPA exposure effects and their relationship to adverse health outcomes often ignore interactions between BPA and other chemicals present in the environment. This review examines BPA co-exposure studies to highlight potentially unexplored mechanisms of action and their possible associations with the adverse health effects attributed to BPA. Importantly, both adverse and beneficial co-exposure effects are observed between BPA and natural chemicals or environmental stressors in in vitro and in vivo models. These interactions clearly influence cellular responses and impact endpoint measures and need to be considered when evaluating BPA exposures and their health effects.
Collapse
Affiliation(s)
- Manoj Sonavane
- Department of Oncologic Sciences, University of South Alabama Mitchell Cancer Institute, Mobile, AL, USA.,Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Natalie R Gassman
- Department of Oncologic Sciences, University of South Alabama Mitchell Cancer Institute, Mobile, AL, USA.,Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| |
Collapse
|