1
|
Rani BU, Vasantharekha R, Santosh W, Swarnalingam T, Barathi S. Endocrine-Disrupting Chemicals and the Effects of Distorted Epigenetics on Preeclampsia: A Systematic Review. Cells 2025; 14:493. [PMID: 40214447 PMCID: PMC11987890 DOI: 10.3390/cells14070493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Preeclampsia (PE) is a critical complication of pregnancy that affects 3% to 5% of all pregnancies and has been linked to aberrant placentation, causing severe maternal and fetal illness and death. OBJECTIVES This systematic review aims to elucidate the association of in-utero endocrine-disrupting chemical (EDC) exposure and microRNAs and their imprinted genes from prenatal and maternal circulation of PE patients. METHODS Databases such as PubMed, PubMed Central, ScienceDirect, the Comparative Toxicogenomics Database (CTD), ProQuest, EBSCOhost, and Google Scholar were utilized to search for articles that investigate the relationships between selected EDCs and epigenetic events such as DNA methylation and microRNAs that are associated with PE. RESULTS A total of 29 studies were included in the database search. Altered expression of microRNAs (miR-15a-5p, miR-142-3p, and miR-185) in the placenta of PE patients was positively associated with the urinary concentration of phthalates and phenols in the development of the disease in the first trimester. EDCs such as phenols, phthalates, perfluoroalkyl substances (PFOAs), polybrominated diphenyl ethers (PBDEs), and organochlorine phosphates (OCPs) have been reported to be associated with hypertensive disorders in pregnancy. miRNA-31, miRNA-144, miRNA-145, miRNA-210, placental specific clusters (C14MC, and C19MC) may be used as possible targets for PE because of their potential roles in the onset and progression of PE. CONCLUSIONS Prenatal EDC exposure, including exposure to BPA, showed association with signaling pathways including estrogen, sFlt-1/PlGF, ErbB, MAPK/ERK, and cholesterol mechanisms with placental hemodynamics. Even low EDC exposures leave altered epigenetic marks throughout gestation, which might cause PE complications.
Collapse
Affiliation(s)
- Balu Usha Rani
- Endocrine Disruption and Reproductive Toxicology Laboratory (EDART), Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (B.U.R.); (R.V.)
| | - Ramasamy Vasantharekha
- Endocrine Disruption and Reproductive Toxicology Laboratory (EDART), Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (B.U.R.); (R.V.)
| | - Winkins Santosh
- Toxicology Research on Endocrine Disruptors (TRENDS) Laboratory, PG & Research Department of Advanced Zoology and Biotechnology, Government Arts College, Nandanam, Chennai 600035, India;
| | - Thangavelu Swarnalingam
- Department of Critical Care Medicine, SRM Medical College Hospital & Research Centre, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Seetharaman Barathi
- Endocrine Disruption and Reproductive Toxicology Laboratory (EDART), Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (B.U.R.); (R.V.)
| |
Collapse
|
2
|
Qu J, Li W, Jia C, Jiang Q, Tang R, Yin Y, Wang X, Long P, Wu T, Yuan Y. Persistent organic pollutants and plasma microRNAs: A community-based profiling analysis. ENVIRONMENT INTERNATIONAL 2025; 197:109328. [PMID: 39999486 DOI: 10.1016/j.envint.2025.109328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/23/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Whether exposure to persistent organic pollutants (POPs) may incur microRNAs (miRNAs) dysregulation remains largely unclear. OBJECTIVES We aim to identify the miRNA signature and related pathways of low-level POPs exposure in a community-based population. METHODS We used general linear regression to model the association of POPs with plasma miRNAs, adjusting for age, gender, smoking, alcohol consumption, body mass index, triglyceride, and total cholesterol levels. We performed pathway enrichment analysis based on 11 experimentally validated and prediction-based databases, and performed tissue specificity analysis. RESULTS We identified 19 POPs significantly associated with 23 miRNAs at false discovery rate-adjusted P value < 0.2, most of which were related to organophosphorus and organochlorine pesticides. Pathway enrichment of the associated miRNAs highlighted chromosome segregation, RNA splicing, autophagy regulation, lipid metabolism, cell growth, development and differentiation, cell cycle regulation, neural network construction, and signal transduction. Notably, 13 POPs were positively associated with miR-6810-3p, a miRNA enriched in temporal lobe with high tissue specificity (Tissue Specificity Index = 0.78). Pathway analysis revealed that miR-6810-3p contributes to the positive regulation of autophagy and multiple cellular functions related to the nervous system such as transport along microtubules, maintenance of cell polarity, and synaptic transmission. CONCLUSION In conclusion, we identified POPs-related miRNA signatures in community-dwelling adults, and highlighted miR-6810-3p in association with multiple POPs, with a potential role in nervous system regulation.
Collapse
Affiliation(s)
- Jingli Qu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wending Li
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, NY 10032, USA
| | - Chengyong Jia
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qin Jiang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Tang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Yin
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pinpin Long
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tangchun Wu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yu Yuan
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
3
|
Panneerselvam D, Murugesan A, Raveendran SK, Kumar JS, Venkataraman P. Examining the hidden dangers: Understanding how microplastics affect pregnancy. Eur J Obstet Gynecol Reprod Biol 2025; 304:53-62. [PMID: 39580908 DOI: 10.1016/j.ejogrb.2024.11.024] [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: 07/22/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Microplastics, a fast-growing environmental concern, play a crucial role in developing the major pollution crisis that affects nearly the entire surface of the planet. Microplastics are tiny particles, measuring less than 5 mm which are ubiquitous, in occurrence, and found in a wide array of products including plastic packaging, synthetic textiles, seafood, fruits, vegetables, salt, sugar, bottled water, and even personal care products. The presence of microplastics in our environment and the potential adverse health effects they may cause have made them a significant perturbation in recent years. Pregnancy is a potentially life-changing experience that entails several apprehensions and new responsibilities for women. For expectant mothers, it is imperative to be aware of the implications of microplastics during pregnancy. One threatened concern is the potential transfer of microplastics across the placenta, which could expose the developing fetus to these particles. Although research on the impact of microplastics on pregnancy is still in its early stages, preliminary findings indicate potential risks that expectant mothers should be aware of. The timing of exposure during pregnancy may play a significant role in the potential risks associated with these tiny particles. In this review, we will delve into the topic, exploring how microplastics enter the body and the potential mechanism by which they pose risks to pregnancy outcomes.
Collapse
Affiliation(s)
- Deboral Panneerselvam
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anuradha Murugesan
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| | - Sajeetha Kumari Raveendran
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Janardanan Subramonia Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - P Venkataraman
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
4
|
Lepeule J, Broséus L, Jedynak P, Masdoumier C, Philippat C, Guilbert A, Nakamura A. [Environmental exposures and epigenome changes within the first 1000 days of life]. Med Sci (Paris) 2024; 40:947-954. [PMID: 39705565 DOI: 10.1051/medsci/2024178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
Early environmental exposures can have long-term effects on child's development and health. Epigenetic modifications may partly explain these effects, and studying them could lead to significant advances in our understanding of the underlying mechanisms. This review summarises recent data on epigenetic and environmental epidemiology during the first 1000 days of life for several common exposures, including tobacco, phenols and phthalates, air pollutants, ambient temperature and vegetation.
Collapse
Affiliation(s)
- Johanna Lepeule
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Lucile Broséus
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Paulina Jedynak
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Chloé Masdoumier
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Claire Philippat
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Ariane Guilbert
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| | - Aurélie Nakamura
- Université Grenoble Alpes, Inserm, CNRS, Team of environmental epidemiology applied to development and respiratory health, IAB, Grenoble, France
| |
Collapse
|
5
|
Rosen EM, Stevens DR, McNell EE, Wood ME, Engel SM, Keil AP, Calafat AM, Botelho JC, Sinkovskaya E, Przybylska A, Saade G, Abuhamad A, Ferguson KK. Longitudinal associations between urinary biomarkers of phthalates and replacements with novel in vivo measures of placental health. Hum Reprod 2024; 39:2104-2114. [PMID: 38970902 PMCID: PMC11373341 DOI: 10.1093/humrep/deae152] [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: 11/13/2023] [Revised: 06/10/2024] [Indexed: 07/08/2024] Open
Abstract
STUDY QUESTION What is the longitudinal association between gestational phthalate exposure and in vivo placental outcomes? SUMMARY ANSWER Phthalates were adversely associated with placental microvasculature, stiffness, and presence of calcification, with different metabolites associated with different outcomes. WHAT IS KNOWN ALREADY Phthalate exposure is ubiquitous and implicated as a contributor to adverse pregnancy outcomes, possibly through impacts on the placenta. STUDY DESIGN, SIZE, DURATION A total of 303 women were recruited in early pregnancy and prospectively followed for up to eight visits across gestation in the Human Placenta and Phthalates study. PARTICIPANTS/MATERIALS, SETTING, METHODS At each visit, women provided urine samples and underwent placental ultrasounds. Urine was analyzed for 18 metabolites of phthalates and replacements. We took the geometric mean of repeated measurements to reflect pregnancy-averaged phthalate or replacement exposure for each participant (n = 303). Placental microvasculature, stiffness, and microcalcification presence were quantified from ultrasounds at each visit. Higher scores reflected worse placental function for all measures. Generalized linear mixed models were created to estimate the association between pregnancy-averaged exposure biomarker concentrations and repeated outcome measurements for microvasculature and stiffness. Gestational age at the time of calcification detection was modeled using Cox proportional hazards models. MAIN RESULTS AND THE ROLE OF CHANCE Monocarboxyisononyl phthalate and summed di(2-ethylhexyl) phthalate metabolites were associated with impaired microvasculature development, such that an interquartile range increase in concentration was associated with 0.11 standard deviation increase in the microvasculature ratio, indicating poorer vascularization (95% CI: 0.00, 0.22); 0.11 [95% CI: -0.01, 0.22], respectively. Monoethyl phthalate was associated with increased placental stiffness (0.09 [95% CI: -0.01, 0.19]) while summed di-iso-butyl phthalate metabolites and monobenzyl phthalate were associated with increased hazard of calcification detection (hazard ratios: 1.18 [95% CI: 0.98, 1.42]; 1.13 [95% CI: 0.96, 1.34]). LIMITATIONS, REASONS FOR CAUTION Outcomes used in this study are novel and further investigation is needed to provide clinical context and relevance. WIDER IMPLICATIONS OF THE FINDINGS We found evidence of associations between select phthalate biomarkers and various aspects of in vivo placental health, although we did not observe consistency across placental outcomes. These findings could illustrate heterogeneous effects of phthalate exposure on placental function. STUDY FUNDING/COMPETING INTEREST(S) This research was supported in part by the Intramural Research Program of the NIH, National Institute of Environmental Health Sciences (ZIA ES103344), and NIEHS T32ES007018. The authors declare that they have no competing interests to disclose. The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the Centers for Disease Control and Prevention. Use of trade names is for identification only and does not imply endorsement by the CDC, the Public Health Service, or the US Department of Health and Human Services. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Emma M Rosen
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Danielle R Stevens
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Erin E McNell
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Mollie E Wood
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie M Engel
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Alexander P Keil
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Julianne Cook Botelho
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Elena Sinkovskaya
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ann Przybylska
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alfred Abuhamad
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Kelly K Ferguson
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| |
Collapse
|
6
|
Rafeletou A, Niemi JVL, Lagunas-Rangel FA, Liu W, Kudłak B, Schiöth HB. The exposure to UV filters: Prevalence, effects, possible molecular mechanisms of action and interactions within mixtures. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 928:170999. [PMID: 38458461 DOI: 10.1016/j.scitotenv.2024.170999] [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: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024]
Abstract
Substances that can absorb sunlight and harmful UV radiation such as organic UV filters are widely used in cosmetics and other personal care products. Since humans use a wide variety of chemicals for multiple purposes it is common for UV filters to co-occur with other substances either in human originating specimens or in the environment. There is increasing interest in understanding such co-occurrence in form of potential synergy, antagonist, or additive effects of biological systems. This review focuses on the collection of data about the simultaneous occurrence of UV filters oxybenzone (OXYB), ethylexyl-methoxycinnamate (EMC) and 4-methylbenzylidene camphor (4-MBC) as well as other classes of chemicals (such as pesticides, bisphenols, and parabens) to understand better any such interactions considering synergy, additive effect and antagonism. Our analysis identified >20 different confirmed synergies in 11 papers involving 16 compounds. We also highlight pathways (such as transcriptional activation of estrogen receptor, promotion of estradiol synthesis, hypothalamic-pituitary-gonadal (HPG) axis, and upregulation of thyroid-hormone synthesis) and proteins (such as Membrane Associated Progesterone Receptor (MAPR), cytochrome P450, and heat shock protein 70 (Hsp70)) that can act as important key nodes for such potential interactions. This article aims to provide insight into the molecular mechanisms on how commonly used UV filters act and may interact with other chemicals.
Collapse
Affiliation(s)
- Alexandra Rafeletou
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Jenni Viivi Linnea Niemi
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | | | - Wen Liu
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Błażej Kudłak
- Department of Analytical Chemistry, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza Str., Gdańsk 80-233, Poland
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Mitra T, Gulati R, Ramachandran K, Rajiv R, Enninga EAL, Pierret CK, Kumari R S, Janardhanan R. Endocrine disrupting chemicals: gestational diabetes and beyond. Diabetol Metab Syndr 2024; 16:95. [PMID: 38664841 PMCID: PMC11046910 DOI: 10.1186/s13098-024-01317-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Gestational Diabetes Mellitus (GDM) has been on the rise for the last two decades along with the growing incidence of obesity. The ubiquitous use of Endocrine-Disrupting Chemicals (EDCs) worldwide has been associated with this increase in GDM incidence. Epigenetic modifications such as DNA methylation, histone acetylation, and methylation have been associated with prenatal exposure to EDCs. EDC exposure can also drive a sustained disruption of the hypothalamus-pituitary-thyroid axis and various other signaling pathways such as thyroid signaling, PPARγ signaling, PI3K-AKT signaling. This disruption leads to impaired glucose metabolism, insulin resistance as well as β-cell dysfunction, which culminate into GDM. Persistent EDC exposure in pregnant women also increases adipogenesis, which results in gestational weight gain. Importantly, pregnant mothers transfer these EDCs to the fetus via the placenta, thus leading to other pregnancy-associated complications such as intrauterine growth restriction (IUGR), and large for gestational age neonates. Furthermore, this early EDC exposure of the fetus increases the susceptibility of the infant to metabolic diseases in early life. The transgenerational impact of EDCs is also associated with higher vascular tone, cognitive aberrations, and enhanced susceptibility to lifestyle disorders including reproductive health anomalies. The review focuses on the impact of environmental toxins in inducing epigenetic alterations and increasing the susceptibility to metabolic diseases during pregnancy needs to be extensively studied such that interventions can be developed to break this vicious cycle. Furthermore, the use of EDC-associated ExomiRs from the serum of patients can help in the early diagnosis of GDM, thereby leading to triaging of patients based on increasing risk factor of the clinicopathological condition.
Collapse
Affiliation(s)
- Tridip Mitra
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Richa Gulati
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Krithika Ramachandran
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Rohan Rajiv
- Dietrich School of Arts and Sciences, University of Pittsburgh, 15260, Pittsburgh, PA, USA
| | | | - Chris K Pierret
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Sajeetha Kumari R
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India
| | - Rajiv Janardhanan
- Division of Medical Research, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, 603 203, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
8
|
Yu J, Tang L, Yang L, Zheng M, Yu H, Luo Y, Liu J, Xu J. Role and mechanism of MiR-542-3p in regulating TLR4 in nonylphenol-induced neuronal cell pyroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155123. [PMID: 37976699 DOI: 10.1016/j.phymed.2023.155123] [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: 02/14/2023] [Revised: 07/26/2023] [Accepted: 09/27/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND This study aimed to investigate the spatial learning/memory and motor abilities of rats and the alteration of miR-542-3p and pyroptosis in the midbrain nigrostriatal area in vivo after nonylphenol (NP) gavage and to explore the mechanism of miR-542-3p regulation of Toll-like receptor 4 (TLR4) in NP-induced pyroptosis in BV2 microglia in vitro. METHODS In vivo: Thirty-six specific-pathogen-free-grade Sprague-Dawley rats were divided into three equal groups: blank control group (treated with pure corn oil), NP group (treated with NP, 80 mg/kg body weight per day for 90 days), and positive control group [treated with lipopolysaccharide (LPS), 2 mg/kg body weight for 7 days]. In vitro: The first part of the experiment was divided into blank group (control, saline), LPS group [1 µg/ml + 1 mM adenosine triphosphate (ATP)], and NP group (40 µmol/L). The second part was divided into mimics NC (negative control) group, miR-542-3p mimics group, mimics NC + NP group, and miR-542-3p mimics + NP group. RESULTS In vivo: Behaviorally, the spatial learning/memory and motor abilities of rats after NP exposure declined, as detected via Y-maze, open field, and rotarod tests. Some microglia in the substantia nigra of the NP-treated rats were activated. The downregulation of miR-542-3p was observed in rat brain tissue after NP exposure. The mRNA/protein expression of pyroptosis-related indicators (TLR4), NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), gasdermin-D (GSDMD), cysteinyl aspartate-specific proteinase-1 (caspase-1), and interleukin-1β (IL-1β) in the substantia nigra of the midbrain increased after NP exposure. In vitro: ASC fluorescence intensity increased in BV2 cells after NP exposure. The mRNA and/or protein expression of pyroptosis-related indicators (TLR4, NLRP3, GSDMD, caspase-1, and IL-1β) in BV2 cells was upregulated after NP exposure. The transfection of miR-542-3p mimics inhibited NP-induced ASC expression in BV2 cells. The overexpression of miR-542-3p, followed by NP exposure, significantly reduced TLR4, NLRP3, ASC, caspase-1, and IL-1β gene and/or protein expression. CONCLUSIONS This study suggested that NP exposure caused a decline in spatial learning memory and whole-body motor ability in rats. Our study was novel in reporting that the upregulation of miR-542-3p targeting and regulating TLR4 could inhibit NLRP3 inflammatory activation and alleviate NP-induced microglia pyroptosis.
Collapse
Affiliation(s)
- Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Lan Tang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China; Department of Nosocomial Infection Control, Guizhou Provincial People's Hospital, Guiyang City, Guizhou Province, 550002, PR China
| | - Lilin Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Mucong Zheng
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Huawen Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Ya Luo
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Jinqing Liu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China.
| |
Collapse
|
9
|
Merrill AK, Sobolewski M, Susiarjo M. Exposure to endocrine disrupting chemicals impacts immunological and metabolic status of women during pregnancy. Mol Cell Endocrinol 2023; 577:112031. [PMID: 37506868 PMCID: PMC10592265 DOI: 10.1016/j.mce.2023.112031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Affiliation(s)
- Alyssa K Merrill
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA.
| |
Collapse
|
10
|
Basak S, Varma S, Duttaroy AK. Modulation of fetoplacental growth, development and reproductive function by endocrine disrupters. Front Endocrinol (Lausanne) 2023; 14:1215353. [PMID: 37854189 PMCID: PMC10579913 DOI: 10.3389/fendo.2023.1215353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Maternal endocrine homeostasis is vital to a successful pregnancy, regulated by several hormones such as human chorionic gonadotropin, estrogen, leptin, glucocorticoid, insulin, prostaglandin, and others. Endocrine stress during pregnancy can modulate nutrient availability from mother to fetus, alter fetoplacental growth and reproductive functions. Endocrine disrupters such as bisphenols (BPs) and phthalates are exposed in our daily life's highest volume. Therefore, they are extensively scrutinized for their effects on metabolism, steroidogenesis, insulin signaling, and inflammation involving obesity, diabetes, and the reproductive system. BPs have their structural similarity to 17-β estradiol and their ability to bind as an agonist or antagonist to estrogen receptors to elicit an adverse response to the function of the endocrine and reproductive system. While adults can negate the adverse effects of these endocrine-disrupting chemicals (EDCs), fetuses do not equip themselves with enzymatic machinery to catabolize their conjugates. Therefore, EDC exposure makes the fetoplacental developmental window vulnerable to programming in utero. On the one hand prenatal BPs and phthalates exposure can impair the structure and function of the ovary and uterus, resulting in placental vascular defects, inappropriate placental expression of angiogenic growth factors due to altered hypothalamic response, expression of nutrient transporters, and epigenetic changes associated with maternal endocrine stress. On the other, their exposure during pregnancy can affect the offspring's metabolic, endocrine and reproductive functions by altering fetoplacental programming. This review highlights the latest development in maternal metabolic and endocrine modulations from exposure to estrogenic mimic chemicals on subcellular and transgenerational changes in placental development and its effects on fetal growth, size, and metabolic & reproductive functions.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Saikanth Varma
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Maitre L, Jedynak P, Gallego M, Ciaran L, Audouze K, Casas M, Vrijheid M. Integrating -omics approaches into population-based studies of endocrine disrupting chemicals: A scoping review. ENVIRONMENTAL RESEARCH 2023; 228:115788. [PMID: 37004856 DOI: 10.1016/j.envres.2023.115788] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 05/16/2023]
Abstract
Health effects of endocrine disrupting chemicals (EDCs) are challenging to detect in the general population. Omics technologies become increasingly common to identify early biological changes before the apparition of clinical symptoms, to explore toxic mechanisms and to increase biological plausibility of epidemiological associations. This scoping review systematically summarises the application of omics in epidemiological studies assessing EDCs-associated biological effects to identify potential gaps and priorities for future research. Ninety-eight human studies (2004-2021) were identified through database searches (PubMed, Scopus) and citation chaining and focused on phthalates (34 studies), phenols (19) and PFASs (17), while PAHs (12) and recently-used pesticides (3) were less studied. The sample sizes ranged from 10 to 12,476 (median = 159), involving non-pregnant adults (38), pregnant women (11), children/adolescents (15) or both latter populations studied together (23). Several studies included occupational workers (10) and/or highly exposed groups (11) focusing on PAHs, PFASs and pesticides, while studies on phenols and phthalates were performed in the general population only. Analysed omics layers included metabolic profiles (30, including 14 targeted analyses), miRNA (13), gene expression (11), DNA methylation (8), microbiome (5) and proteins (3). Twenty-one studies implemented targeted multi-assays focusing on clinical routine blood lipid traits, oxidative stress or hormones. Overall, DNA methylation and gene expression associations with EDCs did not overlap across studies, while some EDC-associated metabolite groups, such as carnitines, nucleotides and amino acids in untargeted metabolomic studies, and oxidative stress markers in targeted studies, were consistent across studies. Studies had common limitations such as small sample sizes, cross-sectional designs and single sampling for exposure biomonitoring. In conclusion, there is a growing body of evidence evaluating the early biological responses to exposure to EDCs. This review points to a need for larger longitudinal studies, wider coverage of exposures and biomarkers, replication studies and standardisation of research methods and reporting.
Collapse
Affiliation(s)
- Léa Maitre
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| | - Paulina Jedynak
- ISGlobal, Barcelona, Spain; University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Marta Gallego
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Laura Ciaran
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Karine Audouze
- Université Paris Cité, T3S, INSERM UMR-S 1124, 45 Rue des Saints Pères, Paris, France
| | - Maribel Casas
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
12
|
Wang JQ, Liang CM, Hu YB, Xia X, Li ZJ, Gao H, Sheng J, Huang K, Wang SF, Zhu P, Hao JH, Tao FB. The effect of phthalates exposure during pregnancy on asthma in infants aged 0 to 36 months: a birth cohort study. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:1951-1974. [PMID: 35751763 DOI: 10.1007/s10653-022-01320-x] [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: 12/29/2021] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
This cohort study sought to investigate the effects of phthalates exposure during pregnancy on offspring asthma and its association with placental stress and inflammatory factor mRNA expression levels. A total of 3474 pregnant women from the China Ma'anshan birth cohort participated in this study. Seven phthalate metabolites were detected in urine samples during pregnancy by solid phase extraction-high-performance liquid chromatography tandem mass spectrometry. Placenta stress and inflammation mRNA expression were assessed by real-time quantitative polymerase chain reaction (RT-qPCR). Early pregnancy may be the critical period when phthalates exposure increases the risk of asthma in infants and young children, and there is a certain gender difference in the risk of asthma in infants and young children. Moreover, through the placenta stress and inflammatory factor associated with infant asthma found anti-inflammatory factor of interleukin-10 (IL-10) mRNA expression will reduce the risk of 36-month-old male infant asthma. The expression of interleukin-4(IL-4) and macrophage (M2) biomarker cluster of differentiation 206(CD206) mRNA reduced the risk of asthma in 18-month-old female infants. Placental stress and inflammatory response were analyzed using mediating effects. Tumor necrosis factor-α (TNFα) showed a complete mediating effect between mono-benzyl phthalate (MBzP) exposure in early pregnancy and asthma in 12-month-old males, and IL-10 also showed a complete mediating effect between mono-n-butyl phthalate (MBP) exposure in early and late pregnancy and asthma in 36-month-old males. In summary, exposure to phthalates during pregnancy may contribute to the development of asthma in infants, which may be associated with placental stress and inflammation.
Collapse
Affiliation(s)
- Jian-Qing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Chun-Mei Liang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ya-Bin Hu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xun Xia
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Zhi-Juan Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hui Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Jie Sheng
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Su-Fang Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peng Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jia-Hu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
13
|
Rudge MVC, Alves FCB, Hallur RLS, Oliveira RG, Vega S, Reyes DRA, Floriano JF, Prudencio CB, Garcia GA, Reis FVDS, Emanueli C, Fuentes G, Cornejo M, Toledo F, Valenzuela-Hinrichsen A, Guerra C, Grismaldo A, Valero P, Barbosa AMP, Sobrevia L. Consequences of the exposome to gestational diabetes mellitus. Biochim Biophys Acta Gen Subj 2023; 1867:130282. [PMID: 36436753 DOI: 10.1016/j.bbagen.2022.130282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 11/26/2022]
Abstract
The exposome is the cumulative measure of environmental influences and associated biological responses throughout the lifespan, including those from the environment, diet, behaviour, and endogenous processes. The exposome concept and the 2030 Agenda for the Sustainable Development Goals (SDGs) from the United Nations are the basis for understanding the aetiology and consequences of non-communicable diseases, including gestational diabetes mellitus (GDM). Pregnancy may be developed in an environment with adverse factors part of the immediate internal medium for fetus development and the external medium to which the pregnant woman is exposed. The placenta is the interface between maternal and fetal compartments and acts as a protective barrier or easing agent to transfer exposome from mother to fetus. Under and over-nutrition in utero, exposure to adverse environmental pollutants such as heavy metals, endocrine-disrupting chemicals, pesticides, drugs, pharmaceuticals, lifestyle, air pollutants, and tobacco smoke plays a determinant role in the development of GDM. This phenomenon is worsened by metabolic stress postnatally, such as obesity which increases the risk of GDM and other diseases. Clinical risk factors for GDM development include its aetiology. It is proposed that knowledge-based interventions to change the potential interdependent ecto-exposome and endo-exposome could avoid the occurrence and consequences of GDM.
Collapse
Affiliation(s)
- Marilza V C Rudge
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil.
| | - Fernanda C B Alves
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Raghavendra L S Hallur
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Centre for Biotechnology, Pravara Institute of Medical Sciences (DU), Loni-413736, Rahata Taluk, Ahmednagar District, Maharashtra, India
| | - Rafael G Oliveira
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Sofia Vega
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - David R A Reyes
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Juliana F Floriano
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Caroline B Prudencio
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Gabriela A Garcia
- São Paulo State University (UNESP), School of Sciences, Postgraduate Program in Materials Science and Technology (POSMAT), 17033-360 Bauru, São Paulo, Brazil
| | - Fabiana V D S Reis
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Gonzalo Fuentes
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Marcelo Cornejo
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 02800, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Andrés Valenzuela-Hinrichsen
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Catalina Guerra
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Adriana Grismaldo
- Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León 64710, Mexico; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Paola Valero
- Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Angelica M P Barbosa
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Department of Physiotherapy and Occupational Therapy, School of Philosophy and Sciences, São Paulo State University (UNESP), 17525-900 Marília, São Paulo, Brazil
| | - Luis Sobrevia
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León 64710, Mexico; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| |
Collapse
|
14
|
Yan Y, Guo F, Liu K, Ding R, Wang Y. The effect of endocrine-disrupting chemicals on placental development. Front Endocrinol (Lausanne) 2023; 14:1059854. [PMID: 36896182 PMCID: PMC9989293 DOI: 10.3389/fendo.2023.1059854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs) or endocrine disruptors are substances that are either naturally occurring or artificial and are released into the natural environment. Humans are exposed to EDCs through ingestion, inhalation, and skin contact. Many everyday household items, such as plastic bottles and containers, the liners of metal food cans, detergents, flame retardants, food, gadgets, cosmetics, and pesticides, contain endocrine disruptors. Each hormone has a unique chemical makeup and structural attributes. The way that endocrine hormones connect to receptors is described as a "lock and key" mechanism, with each hormone serving as the key (lock). This mechanism is enabled by the complementary shape of receptors to their hormone, which allows the hormone to activate the receptors. EDCs are described as exogenous chemicals or compounds that have a negative impact on organisms' health by interacting with the functioning of the endocrine system. EDCs are associated with cancer, cardiovascular risk, behavioural disorders, autoimmune abnormalities, and reproductive disorders. EDCs exposure in humans is highly harmful during critical life stages. Nonetheless, the effect of EDCs on the placenta is often underestimated. The placenta is especially sensitive to EDCs due to its abundance of hormone receptors. In this review, we evaluated the most recent data on the effects of EDCs on placental development and function, including heavy metals, plasticizers, pesticides, flame retardants, UV filters and preservatives. The EDCs under evaluation have evidence from human biomonitoring and are found in nature. Additionally, this study indicates important knowledge gaps that will direct future research on the topic.
Collapse
Affiliation(s)
- Yan Yan
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, China
| | - Fengjun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Kexin Liu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rixin Ding
- Department of Cardiovascular Medicine, Changchun Central Hospital, Changchun, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Yichao Wang,
| |
Collapse
|
15
|
Schrott R, Song A, Ladd-Acosta C. Epigenetics as a Biomarker for Early-Life Environmental Exposure. Curr Environ Health Rep 2022; 9:604-624. [PMID: 35907133 DOI: 10.1007/s40572-022-00373-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW There is interest in evaluating the developmental origins of health and disease (DOHaD) which emphasizes the role of prenatal and early-life environments on non-communicable health outcomes throughout the life course. The ability to rigorously assess and identify early-life risk factors for later health outcomes, including those with childhood onset, in large population samples is often limited due to measurement challenges such as impractical costs associated with prospective studies with a long follow-up duration, short half-lives for some environmental toxicants, and lack of biomarkers that capture inter-individual differences in biologic response to external environments. RECENT FINDINGS Epigenomic patterns, and DNA methylation in particular, have emerged as a potential objective biomarker to address some of these study design and exposure measurement challenges. In this article, we summarize the literature to date on epigenetic changes associated with specific prenatal and early-life exposure domains as well as exposure mixtures in human observational studies and their biomarker potential. Additionally, we highlight evidence for other types of epigenetic patterns to serve as exposure biomarkers. Evidence strongly supports epigenomic biomarkers of exposure that are detectable across the lifespan and across a range of exposure domains. Current and future areas of research in this field seek to expand these lines of evidence to other environmental exposures, to determine their specificity, and to develop predictive algorithms and methylation scores that can be used to evaluate early-life risk factors for health outcomes across the life span.
Collapse
Affiliation(s)
- Rose Schrott
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ashley Song
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
16
|
Shree N, Ding Z, Flaws J, Choudhury M. Role of microRNA in Endocrine Disruptor-Induced Immunomodulation of Metabolic Health. Metabolites 2022; 12:1034. [PMID: 36355117 PMCID: PMC9695656 DOI: 10.3390/metabo12111034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 01/22/2025] Open
Abstract
The prevalence of poor metabolic health is growing exponentially worldwide. This condition is associated with complex comorbidities that lead to a compromised quality of life. One of the contributing factors recently gaining attention is exposure to environmental chemicals, such as endocrine-disrupting chemicals (EDCs). Considerable evidence suggests that EDCs can alter the endocrine system through immunomodulation. More concerning, EDC exposure during the fetal development stage has prominent adverse effects later in life, which may pass on to subsequent generations. Although the mechanism of action for this phenomenon is mostly unexplored, recent reports implicate that non-coding RNAs, such as microRNAs (miRs), may play a vital role in this scenario. MiRs are significant contributors in post-transcriptional regulation of gene expression. Studies demonstrating the immunomodulation of EDCs via miRs in metabolic health or towards the Developmental Origins of Health and Disease (DOHaD) Hypothesis are still deficient. The aim of the current review was to focus on studies that demonstrate the impact of EDCs primarily on innate immunity and the potential role of miRs in metabolic health.
Collapse
Affiliation(s)
- Nitya Shree
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| | - Zehuan Ding
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| | - Jodi Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| |
Collapse
|
17
|
Micro-RNAs in Human Placenta: Tiny Molecules, Immense Power. Molecules 2022; 27:molecules27185943. [PMID: 36144676 PMCID: PMC9501247 DOI: 10.3390/molecules27185943] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 12/06/2022] Open
Abstract
Micro-RNAs (miRNAs) are short non-coding single-stranded RNAs that modulate the expression of various target genes after transcription. The expression and distribution of kinds of miRNAs have been characterized in human placenta during different gestational stages. The identified miRNAs are recognized as key mediators in the regulation of placental development and in the maintenance of human pregnancy. Aberrant expression of miRNAs is associated with compromised pregnancies in humans, and dysregulation of those miRNAs contributes to the occurrence and development of related diseases during pregnancy, such as pre-eclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), recurrent miscarriage, preterm birth (PTB) and small-for-gestational-age (SGA). Thus, having a better understanding of the expression and functions of miRNAs in human placenta during pregnancy and thereby developing novel drugs targeting the miRNAs could be a potentially promising method in the prevention and treatment of relevant diseases in future. Here, we summarize the current knowledge of the expression pattern and function regulation of miRNAs in human placental development and related diseases.
Collapse
|
18
|
Rong W, Shukun W, Xiaoqing W, Wenxin H, Mengyuan D, Chenyang M, Zhang H. Regulatory roles of non-coding RNAs and m6A modification in trophoblast functions and the occurrence of its related adverse pregnancy outcomes. Crit Rev Toxicol 2022; 52:681-713. [PMID: 36794364 DOI: 10.1080/10408444.2022.2144711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Adverse pregnancy outcomes, such as preeclampsia, gestational diabetes mellitus, fetal growth restriction, and recurrent miscarriage, occur frequently in pregnant women and might further induce morbidity and mortality for both mother and fetus. Increasing studies have shown that dysfunctions of human trophoblast are related to these adverse pregnancy outcomes. Recent studies also showed that environmental toxicants could induce trophoblast dysfunctions. Moreover, non-coding RNAs (ncRNAs) have been reported to play important regulatory roles in various cellular processes. However, the roles of ncRNAs in the regulation of trophoblast dysfunctions and the occurrence of adverse pregnancy outcomes still need to be further investigated, especially with exposure to environmental toxicants. In this review, we analyzed the regulatory mechanisms of ncRNAs and m6A methylation modification in the dysfunctions of trophoblast cells and the occurrence of adverse pregnancy outcomes and also summarized the harmful effects of environmental toxicants. In addition to DNA replication, mRNA transcription, and protein translation, ncRNAs and m6A modification might be considered as the fourth and fifth elements that regulate the genetic central dogma, respectively. Environmental toxicants might also affect these processes. In this review, we expect to provide a deeper scientific understanding of the occurrence of adverse pregnancy outcomes and to discover potential biomarkers for the diagnosis and treatment of these outcomes.
Collapse
Affiliation(s)
- Wang Rong
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Wan Shukun
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wang Xiaoqing
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huang Wenxin
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dai Mengyuan
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Mi Chenyang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
19
|
Seymore TN, Rivera-Núñez Z, Stapleton PA, Adibi JJ, Barrett ES. Phthalate Exposures and Placental Health in Animal Models and Humans: A Systematic Review. Toxicol Sci 2022; 188:153-179. [PMID: 35686923 PMCID: PMC9333406 DOI: 10.1093/toxsci/kfac060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Phthalates are ubiquitous compounds known to leach from the plastic products that contain them. Due to their endocrine-disrupting properties, a wide range of studies have elucidated their effects on reproduction, metabolism, neurodevelopment, and growth. Additionally, their impacts during pregnancy and on the developing fetus have been extensively studied. Most recently, there has been interest in the impacts of phthalates on the placenta, a transient major endocrine organ critical to maintenance of the uterine environment and fetal development. Phthalate-induced changes in placental structure and function may have significant impacts on the course of pregnancy and ultimately, child health. Prior reviews have described the literature on phthalates and placental health; however to date, there has been no comprehensive, systematic review on this topic. Here, we review 35 papers (24 human and 11 animal studies) and summarize phthalate exposures in relation to an extensive set of placental measures. Phthalate-related alterations were reported for placental morphology, hormone production, vascularization, histopathology, and gene/protein expression. The most consistent changes were observed in vascular and morphologic endpoints, including cell composition. These changes have implications for pregnancy complications such as preterm birth and intrauterine growth restriction as well as potential ramifications for children's health. This comprehensive review of the literature, including common sources of bias, will inform the future work in this rapidly expanding field.
Collapse
Affiliation(s)
- Talia N Seymore
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
| | - Zorimar Rivera-Núñez
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey 08854, USA
| | - Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
| | - Jennifer J Adibi
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Emily S Barrett
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey 08854, USA
| |
Collapse
|
20
|
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: 15] [Impact Index Per Article: 5.0] [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
|
21
|
Cui FP, Liu C, Deng YL, Chen PP, Miao Y, Luo Q, Zhang M, Yang P, Wang YX, Lu WQ, Zeng Q. Urinary and seminal plasma concentrations of phthalate metabolites in relation to spermatogenesis-related miRNA106a among men from an infertility clinic. CHEMOSPHERE 2022; 288:132464. [PMID: 34619260 DOI: 10.1016/j.chemosphere.2021.132464] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
Studies indicate that phthalates can disrupt spermatogenesis and lead to the reduction of semen quality. However, the underlying mechanisms remain unclear. This study aimed to examine the associations of phthalate exposures as individual chemicals and mixtures with spermatogenesis-related miRNA106a. We detected eight phthalate metabolites in repeated urine samples and a single seminal plasma specimen among 111 men from an infertility clinic in Wuhan, China. Spermatogenesis-related miRNA106a was measured in seminal plasma. We used multivariable linear regression and Bayesian kernel machine regression (BKMR) models to separately evaluate the associations of phthalate metabolites as individual chemicals and mixtures with spermatogenesis-related miRNA106a. Elevated tertiles of urinary mono (2-ethylhexyl) phthalate (MEHP) was associated with decreased miRNA106a (-61.71%; 95%CI: 81.92, -18.93% for the highest vs. lowest tertile; P for trend = 0.01). Similarly, an inverse exposure-response relationship between seminal plasma MEHP concentrations and miRNA106a was also observed (-59.44%; 95%CI: 79.19, -20.95% for the highest vs. lowest tertile; P for trend = 0.01). The BKMR models showed that the mixtures of seminal plasma phthalate metabolites were associated with decreased miRNA106a when the chemical mixtures were ≥35th percentile compared to their medians. Nonlinear associations with miRNA106a were estimated for urinary and seminal plasma MEHP while fixing other phthalate metabolites at their medians. Our findings suggest that mixtures of phthalate metabolites in seminal plasma were negatively associated with spermatogenesis-related miRNA106a, and individual MEHP was the major contributor to the adverse effects.
Collapse
Affiliation(s)
- Fei-Peng Cui
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pan-Pan Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu Miao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qiong Luo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Min Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Pan Yang
- Department of Occupational and Environmental Health, School of Basic Medicine and Public Health, Jinan University, Guangzhou, Guangdong, PR China
| | - Yi-Xin Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Wen-Qing Lu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
22
|
Tumolo MR, Panico A, De Donno A, Mincarone P, Leo CG, Guarino R, Bagordo F, Serio F, Idolo A, Grassi T, Sabina S. The expression of microRNAs and exposure to environmental contaminants related to human health: a review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:332-354. [PMID: 32393046 DOI: 10.1080/09603123.2020.1757043] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Environmental contaminants exposure may lead to detrimental changes to the microRNAs (miRNAs) expression resulting in several health effects. miRNAs, small non-coding RNAs that regulate gene expression, have multiple transcript targets and thereby regulate several signalling molecules. Even a minor alteration in the abundance of one miRNA can have deep effects on global gene expression. Altered patterns of miRNAs can be responsible for changes linked to various health outcomes, suggesting that specific miRNAs are activated in pathophysiological processes. In this review, we provide an overview of studies investigating the impact of air pollution, organic chemicals, and heavy metals on miRNA expression and the potential biologic effects on humans.Abbreviations: AHRR, aryl-hydrocarbon receptor repressor; AHR, aryl-hydrocarbon receptor; As, arsenic; BCL2, B-cell lymphoma 2; BCL2L11, B-cell lymphoma 2 like 11; BCL6, B-cell lymphoma 6; BPA, bisphenol A; CVD, cardiovascular diseases; CD40, cluster of differentiation 40; CCND1, Cyclin D1; CDKN1A, cyclin-dependent kinase inhibitor 1A; Cr, chromium; CTBP1, C-terminal binding protein 1; CXCL12, C-X-C motif chemokine ligand 12; DAZAP1, deleted in azoospermia associated protein 1; DEP, diesel exhaust particles; EGFR, epidermal growth factor receptor; eNOS, endothelial nitric oxide synthase; EVs, extracellular vesicles; FAK, focal adhesion kinase; FAS, fas cell surface death receptor; FOXO, forkhead box O; HbA1c, glycated hemoglobin; Hg, mercury; HLA-A, human leukocyte antigen A; HMGB, high-mobility group protein B; IFNAR2, interferon alpha receptor subunit 2; IL-6, interleukin-6; IRAK1, interleukin 1 receptor associated kinase 1; JAK/STAT, janus kinase/signal transducers and activators of transcription; MAPK, mitogen-activated protein kinase; miRNAs, microRNAs; MVs, microvesicles; NCDs, noncommunicable diseases; NFAT, nuclear factor of activated T cells; NFkB, nuclear factor kappa B; NRF2, nuclear factor, erythroid-derived 2; NRG3, neuregulin 3; O3, ozone; OP, organophosphorus pesticides; PAHs, polycyclic aromatic hydrocarbons; Pb, lead; PCBs, polychlorinated biphenyls; PDCD4, programmed cell death 4; PDGFB, platelet derived growth factor subunit beta; PDGFR, platelet-derived growth factor receptor; PI3K/Akt, phosphoinositide-3-kinase/protein kinase B; PKA, protein kinase A; PM, particulate matter; PRKCQ, protein kinase C theta; PTEN, phosphatase and tensin homolog; SORT1, sortilin 1; TGFβ, transforming growth factor-β; TLR, toll-like receptor; TNF, tumor necrosis factors; TRAF1, tumor necrosis factors-receptor associated factors 1; TRAP, traffic-related air pollution; TREM1, triggering receptor expressed on myeloid cells 1; TRIAP1, TP53 regulated inhibitor of apoptosis 1; VCAM-1, vascular cell adhesion molecule 1; VEGFA, vascular endothelial growth factor A; XRCC2, X-ray repair cross complementing 2; YBX2, Y-box-binding protein 2; ZEB1, zinc finger E-box-binding homeobox 1; ZEB2, zinc finger E-box-binding homeobox 2; 8-OH-dG, 8-hydroxy-guanine.
Collapse
Affiliation(s)
- Maria Rosaria Tumolo
- National Research Council, Institute for Research on Population and Social Policies, Research Unit of Brindisi, Brindisi, Italy
| | - Alessandra Panico
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Antonella De Donno
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Pierpaolo Mincarone
- National Research Council, Institute for Research on Population and Social Policies, Research Unit of Brindisi, Brindisi, Italy
| | - Carlo Giacomo Leo
- National Research Council, Institute of Clinical Physiology, Branch of Lecce, Lecce, Italy
| | - Roberto Guarino
- National Research Council, Institute of Clinical Physiology, Branch of Lecce, Lecce, Italy
| | - Francesco Bagordo
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Francesca Serio
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Adele Idolo
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Tiziana Grassi
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Saverio Sabina
- National Research Council, Institute of Clinical Physiology, Branch of Lecce, Lecce, Italy
| |
Collapse
|
23
|
Xiong YW, Feng YJ, Wei T, Zhang X, Tan LL, Zhang J, Dai LM, Zhu HL, Zhou GX, Liu WB, Liu ZQ, Xu XF, Gao L, Zhang C, Wang Q, Xu DX, Wang H. miR-6769b-5p targets CCND-1 to regulate proliferation in cadmium-treated placental trophoblasts: Association with the impairment of fetal growth. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 230:113109. [PMID: 34953275 DOI: 10.1016/j.ecoenv.2021.113109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
Environmental cadmium (Cd) is positively associated with placental impairment and fetal growth retardation. Nevertheless, its potential mechanisms remain unclear. microRNAs (miRNAs) are known to influence placental development and fetal growth. This work was aimed to determine which miRNAs are involved in Cd-impaired placental and fetal development based on the mRNA and miRNA expression profiles analysis. As a result, gestational Cd exposure deceased fetal and placental weight, and reduced the protein level of PCNA in human and mouse placentae. Furthermore, the results of mRNA microarray showed that Cd-downregulated mRNAs were predictively correlated with several biological processes, including cell proliferation, differentiation and motility. In addition, the results of miRNA microarray and qPCR assay demonstrated that Cd significantly increased the level of miR-6769b-5p, miR-146b-5p and miR-452-5p. Integrated analysis of Cd-upregulated miRNAs predicted target genes and Cd-downregulated mRNAs found that overlapping mRNAs, such as CCND1, CDK13, RINT1 and CDC26 were also significantly associated with cell proliferation. Further experiments showed that miR-6769b-5p inhibitor, but not miR-146b-5p and miR-452-5p, markedly reversed Cd-downregulated the expression of proliferation-related mRNAs, and thereby restored Cd-decreased the proteins level of CCND1 and PCNA in human placental trophoblasts. Dual luciferase reporter assay further revealed that miR-6769b-5p directly targets CCND1. Finally, the case-control study demonstrated that increased miR-6769b-5p level and impaired cell proliferation were observed in small-for-gestational-age human placentae. In conclusion, miR-6769b-5p targets CCND-1 to regulate proliferation in Cd-treated placental trophoblasts, which is associated with the impairment of fetal growth. Our findings imply that placental miR-6769b-5p may be used as an epigenetic marker for environmental pollutants-caused fetal growth restriction and its late-onset chronic diseases.
Collapse
Affiliation(s)
- Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yu-Jie Feng
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Tian Wei
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Xiang Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Lu-Lu Tan
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Jin Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Li-Min Dai
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Guo-Xiang Zhou
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Wei-Bo Liu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Zi-Qi Liu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, China
| | - Xiao-Feng Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, China
| | - Lan Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Qing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, China.
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China.
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China.
| |
Collapse
|
24
|
Ticiani E, Pu Y, Gingrich J, Veiga-Lopez A. Bisphenol S Impairs Invasion and Proliferation of Extravillous Trophoblasts Cells by Interfering with Epidermal Growth Factor Receptor Signaling. Int J Mol Sci 2022; 23:671. [PMID: 35054855 PMCID: PMC8776214 DOI: 10.3390/ijms23020671] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/03/2022] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
The placenta supports fetal growth and is vulnerable to exogenous chemical exposures. We have previously demonstrated that exposure to the emerging chemical bisphenol S (BPS) can alter placental endocrine function. Mechanistically, we have demonstrated that BPS interferes with epidermal growth factor receptor (EGFR) signaling, reducing placenta cell fusion. Extravillous trophoblasts (EVTs), a placenta cell type that aids with vascular remodeling, require EGF to invade into the maternal endometrium. We hypothesized that BPS would impair EGF-mediated invasion and proliferation in EVTs. Using human EVTs (HTR-8/SVneo cells), we tested whether BPS could inhibit the EGF response by blocking EGFR activation. We also evaluated functional endpoints of EGFR signaling, including EGF endocytosis, cell invasion and proliferation, and endovascular differentiation. We demonstrated that BPS blocked EGF-induced phosphorylation of EGFR by acting as a competitive antagonist to EGFR. Transwell assay and a three-dimensional microfluidic chip invasion assay revealed that BPS exposure can block EGF-mediated cell invasion. BPS also blocked EGF-mediated proliferation and endovascular differentiation. In conclusion, BPS can prevent EGF-mediated EVT proliferation and invasion through EGFR antagonism. Given the role of EGFR in trophoblast proliferation and differentiation during placental development, our findings suggest that maternal exposure to BPS may contribute to placental dysfunction via EGFR-mediated mechanisms.
Collapse
Affiliation(s)
- Elvis Ticiani
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA; (E.T.); (Y.P.)
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA; (E.T.); (Y.P.)
| | - Jeremy Gingrich
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA; (E.T.); (Y.P.)
- The Chicago Center for Health and the Environment, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
25
|
Pierri B, Buonerba C, Pierri A, Pizzolante A, Ferro A, Crispo A, Bollati V, Sanchez TR, Grazia Andreassi M, Esposito M, Cerino P. Exposure study on susceptible people - SPES: An integrative biomonitoring approach. ENVIRONMENT INTERNATIONAL 2022; 158:106931. [PMID: 34653810 DOI: 10.1016/j.envint.2021.106931] [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: 06/17/2021] [Revised: 09/08/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The evaluation of environmental exposure risk requires a global analysis of pollution phenomena, including biological effects and potentially correlated clinical outcomes in susceptible populations. Although human biomonitoring plays a fundamental role in assessing the degree of contamination, it is not effective alone in identifying a direct link between exposure, biomolecular effects and outcomes on target organisms. While toxicogenomics and epidemiology are mainly focused on the investigation of molecular reactions and clinical outcomes, the monitoring of environmental matrices works independently to characterize the territorial distribution of toxic compounds, without proving any correlated health risk for residents. OBJECTIVES We propose a new biomonitoring model based on a whole systemic analytical evaluation of environmental context. The paradigm of the method consists of identifying the sources of pollution, the migration pathways of those pollutants and their effects on target organisms. By means of this innovative, holistic epidemiological approach, we included healthy human subjects in a cohort to identify potential risks of exposure and predict possible correlated clinical outcomes. 4205 residents of the Campania region were enrolled in the "SPES" biomonitoring study, which especially focused on the areas dubbed "Land of Fires" in the recent decades. DISCUSSION The analysis of environmental exposure risk suffers the lack of data integration from various science fields, and this comes down to a limited point of view and a limited knowledge of phenomena. In implementing our model, we first constructed an analytical picture of the Real-world situation. We next conducted a comparative risk assessment, in order to identify possible correlations between pollution and health within a holistic view. CONCLUSION This type of research activities aims to support the implementation of public health interventions and to become a reference model in the evaluation of the risk of exposure to environmental pollutants.
Collapse
Affiliation(s)
- Biancamaria Pierri
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy; Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, (SA), Italy.
| | - Carlo Buonerba
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| | - Andrea Pierri
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| | - Antonio Pizzolante
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| | - Amedeo Ferro
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| | - Anna Crispo
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Tiffany R Sanchez
- Department of Environmental Health Sciences, Columbia University, 10032 New York, NY, USA
| | - Maria Grazia Andreassi
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), Pisa, Italy
| | - Mauro Esposito
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| | - Pellegrino Cerino
- Centro di Referenza Nazionale per l'analisi e studio di correlazione tra ambiente, animale e uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, (NA), Italy
| |
Collapse
|
26
|
Paquette AG, MacDonald J, Lapehn S, Bammler T, Kruger L, Day DB, Price ND, Loftus C, Kannan K, Marsit C, Mason WA, Bush NR, LeWinn KZ, Enquobahrie DA, Prasad B, Karr CJ, Sathyanarayana S. A Comprehensive Assessment of Associations between Prenatal Phthalate Exposure and the Placental Transcriptomic Landscape. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:97003. [PMID: 34478338 PMCID: PMC8415559 DOI: 10.1289/ehp8973] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
BACKGROUND Phthalates are commonly used endocrine-disrupting chemicals that are ubiquitous in the general population. Prenatal phthalate exposure may alter placental physiology and fetal development, leading to adverse perinatal and childhood health outcomes. OBJECTIVE We examined associations between prenatal phthalate exposure in the second and third trimesters and the placental transcriptome at birth, including genes and long noncoding RNAs (lncRNAs), to gain insight into potential mechanisms of action during fetal development. METHODS The ECHO PATHWAYs consortium quantified 21 urinary phthalate metabolites from 760 women enrolled in the CANDLE study (Shelby County, TN) using high-performance liquid chromatography-tandem mass spectrometry. Placental transcriptomic data were obtained using paired-end RNA sequencing. Linear models were fitted to estimate separate associations between maternal urinary phthalate metabolite concentration during the second and third trimester and placental gene expression at birth, adjusted for confounding variables. Genes were considered differentially expressed at a Benjamini-Hochberg false discovery rate (FDR) p<0.05. Associations between phthalate metabolites and biological pathways were identified using self-contained gene set testing and considered significantly altered with an FDR-adjusted p<0.2. RESULTS We observed significant associations between second-trimester phthalate metabolites mono (carboxyisooctyl) phthalate (MCIOP), mono-2-ethyl-5-carboxypentyl phthalate, and mono-2-ethyl-5-oxohexyl phthalate and 18 genes in total, including four lncRNAs. Specifically, placental expression of NEAT1 was associated with multiple phthalate metabolites. Third-trimester MCIOP and mono-isobutyl phthalate concentrations were significantly associated with placental expression of 18 genes and two genes, respectively. Expression of genes within 27 biological pathways was associated with mono-methyl phthalate, MCIOP, and monoethyl phthalate concentrations. DISCUSSION To our knowledge, this is the first genome-wide assessment of the relationship between the placental transcriptome at birth and prenatal phthalate exposure in a large and diverse birth cohort. We identified numerous genes and lncRNAs associated with prenatal phthalate exposure. These associations mirror findings from other epidemiological and in vitro analyses and may provide insight into biological pathways affected in utero by phthalate exposure. https://doi.org/10.1289/EHP8973.
Collapse
Affiliation(s)
- Alison G. Paquette
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| | | | - Samantha Lapehn
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Theo Bammler
- University of Washington, Seattle, Washington, USA
| | - Laken Kruger
- Washington State University, Spokane, Washington, USA
| | - Drew B. Day
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Nathan D. Price
- Institute For Systems Biology, Seattle, Washington, USA
- Onegevity Health, New York City, New York, USA
| | | | | | | | - W. Alex Mason
- University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Nicole R. Bush
- University of California San Francisco, San Francisco California, USA
| | - Kaja Z. LeWinn
- University of California San Francisco, San Francisco California, USA
| | | | | | | | - Sheela Sathyanarayana
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| |
Collapse
|
27
|
Warner GR, Dettogni RS, Bagchi IC, Flaws JA, Graceli JB. Placental outcomes of phthalate exposure. Reprod Toxicol 2021; 103:1-17. [PMID: 34015474 PMCID: PMC8260441 DOI: 10.1016/j.reprotox.2021.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022]
Abstract
Proper placental development and function relies on hormone receptors and signaling pathways that make the placenta susceptible to disruption by endocrine disrupting chemicals, such as phthalates. Here, we review relevant research on the associations between phthalate exposures and dysfunctions of the development and function of the placenta, including morphology, physiology, and genetic and epigenetic effects. This review covers in vitro studies, in vivo studies in mammals, and studies in humans. We also discuss important gaps in the literature. Overall, the evidence indicates that toxicity to the placental and maternal-fetal interface is associated with exposure to phthalates. Further studies are needed to better elucidate the mechanisms through which phthalates act in the placenta as well as additional human studies that assess placental disruption through pregnancy with larger sample sizes.
Collapse
Affiliation(s)
- Genoa R Warner
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA
| | | | - Indrani C Bagchi
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA
| | - Jodi A Flaws
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA.
| | - Jones B Graceli
- Dept of Morphology, Federal University of Espirito Santo, Brazil
| |
Collapse
|
28
|
Adibi JJ, Layden AJ, Birru RL, Miragaia A, Xun X, Smith MC, Yin Q, Millenson ME, O’Connor TG, Barrett ES, Snyder NW, Peddada S, Mitchell RT. First trimester mechanisms of gestational sac placental and foetal teratogenicity: a framework for birth cohort studies. Hum Reprod Update 2021; 27:747-770. [PMID: 33675653 PMCID: PMC8222765 DOI: 10.1093/humupd/dmaa063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/18/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The function of the gestational sac (GS) and the placenta in the closely related processes of embryogenesis and teratogenicity in the first trimester has been minimally described. The prevailing assumption is that direct teratogenic effects are mediated by the critical extraembryonic organ, the placenta, which either blocks or transfers exposures to the foetus. Placental transfer is a dominant mechanism, but there are other paradigms by which the placenta can mediate teratogenic effects. Knowledge of these paradigms and first trimester human developmental biology can be useful to the epidemiologist in the conduct of biomarker-based studies of both maternal and child health. OBJECTIVE AND RATIONALE Our aim is to provide a causal framework for modelling the teratogenic effects of first trimester exposures on child health outcomes mediated by the GS and placenta using biomarker data collected in the first trimester. We initially present first trimester human developmental biology for the sake of informing and strengthening epidemiologic approaches. We then propose analytic approaches of modelling placental mechanisms by way of causal diagrams using classical non-embryolethal teratogens (diethylstilboestrol [DES], folic acid deficiency and cytomegalovirus [CMV]) as illustrative examples. We extend this framework to two chronic exposures of particular current interest, phthalates and maternal adiposity. SEARCH METHODS Information on teratogens was identified by a non-systematic, narrative review. For each teratogen, we included papers that answered the five following questions: (i) why were these exposures declared teratogens? (ii) is there a consensus on biologic mechanism? (iii) is there reported evidence of a placental mechanism? (iv) can we construct a theoretical model of a placental mechanism? and (v) can this knowledge inform future work on measurement and modelling of placental-foetal teratogenesis? We prioritized literature specific to human development, the organogenesis window in the first trimester and non-embryolethal mechanisms. OUTCOMES As a result of our review of the literature on five exposures considered harmful in the first trimester, we developed four analytic strategies to address first trimester placental mechanisms in birth cohort studies: placental transfer and direct effects on the foetus (DES and maternal adiposity), indirect effects through targeted placental molecular pathways (DES and phthalates), pre-placental effects through disruptions in embryonic and extraembryonic tissue layer differentiation (folic acid deficiency), and multi-step mechanisms that involve maternal, placental and foetal immune function and inflammation (DES and CMV). WIDER IMPLICATIONS The significance of this review is to offer a causal approach to classify the large number of potentially harmful exposures in pregnancy when the exposure occurs in the first trimester. Our review will facilitate future research by advancing knowledge of the first trimester mechanisms necessary for researchers to effectively associate environmental exposures with child health outcomes.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander J Layden
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rahel L Birru
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexandra Miragaia
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan C Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Yin
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Thomas G O’Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Nathaniel W Snyder
- Department of Microbiology and Immunology, Center for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shyamal Peddada
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, UK
| |
Collapse
|
29
|
Padmanabhan V, Song W, Puttabyatappa M. Praegnatio Perturbatio-Impact of Endocrine-Disrupting Chemicals. Endocr Rev 2021; 42:295-353. [PMID: 33388776 PMCID: PMC8152448 DOI: 10.1210/endrev/bnaa035] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Indexed: 02/07/2023]
Abstract
The burden of adverse pregnancy outcomes such as preterm birth and low birth weight is considerable across the world. Several risk factors for adverse pregnancy outcomes have been identified. One risk factor for adverse pregnancy outcomes receiving considerable attention in recent years is gestational exposure to endocrine-disrupting chemicals (EDCs). Humans are exposed to a multitude of environmental chemicals with known endocrine-disrupting properties, and evidence suggests exposure to these EDCs have the potential to disrupt the maternal-fetal environment culminating in adverse pregnancy and birth outcomes. This review addresses the impact of maternal and fetal exposure to environmental EDCs of natural and man-made chemicals in disrupting the maternal-fetal milieu in human leading to adverse pregnancy and birth outcomes-a risk factor for adult-onset noncommunicable diseases, the role lifestyle and environmental factors play in mitigating or amplifying the effects of EDCs, the underlying mechanisms and mediators involved, and the research directions on which to focus future investigations to help alleviate the adverse effects of EDC exposure.
Collapse
Affiliation(s)
| | - Wenhui Song
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
30
|
Padmanabhan V, Moeller J, Puttabyatappa M. Impact of gestational exposure to endocrine disrupting chemicals on pregnancy and birth outcomes. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:279-346. [PMID: 34452689 DOI: 10.1016/bs.apha.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the advent of industrialization, humans are exposed to a wide range of environmental chemicals, many with endocrine disrupting potential. As successful maintenance of pregnancy and fetal development are under tight hormonal control, the gestational exposure to environmental endocrine disrupting chemicals (EDC) have the potential to adversely affect the maternal milieu and support to the fetus, fetal developmental trajectory and birth outcomes. This chapter summarizes the impact of exposure to EDCs both individually and as mixtures during pregnancy, the immediate and long-term consequences of such exposures on the mother and fetus, the direct and indirect mechanisms through which they elicit their effects, factors that modify their action, and the research directions to focus future investigations.
Collapse
Affiliation(s)
| | - Jacob Moeller
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
31
|
Al-Saleh I, Elkhatib R, Alrushud N, Alnuwaysir H, Alnemer M, Aldhalaan H, Shoukri M, McWalter P, Alkhenizan A. Potential health risks of maternal phthalate exposure during the first trimester - The Saudi Early Autism and Environment Study (SEAES). ENVIRONMENTAL RESEARCH 2021; 195:110882. [PMID: 33621597 DOI: 10.1016/j.envres.2021.110882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 06/12/2023]
Abstract
Phthalates are the most ubiquitous contaminants that we are exposed to daily due to their wide use as plasticizers in various consumer products. A few studies have suggested that in utero exposure to phthalates can disturb fetal growth and development in humans, because phthalates can interfere with endocrine function. We collected spot urine samples from 291 pregnant women in their first trimester (9.8 ± 2.3 gestational weeks) recruited in an ongoing prospective cohort study in Saudi Arabia. A second urine sample was collected within 1-7 d after enrollment. The aims of this study were to: (1) assess the extent of exposure to phthalates during the first trimester and (2) estimate the risk from single and cumulative exposures to phthalates. Most phthalate metabolites' urinary levels were high, several-fold higher than those reported in relevant studies from other countries. The highest median levels of monoethyl phthalate, mono-n-butyl phthalate (MnBP), mono-iso-butyl phthalate (MiBP), and mono-(2-ethylhexyl) phthalate (MEHP) in μg/l (μg/g creatinine) were 245.62 (197.23), 114.26 (99.45), 39.59 (34.02), and 23.51 (19.92), respectively. The MEHP levels were highest among three di (2-ethylhexyl) phthalate (DEHP) metabolites. %MEHP4, the ratio of MEHP to four di (2-ethylhexyl) phthalate metabolites (∑4DEHP), was 44%, indicating interindividual differences in metabolism and excretion. The hazard quotient (HQ) of individual phthalates estimated based on the reference dose (RfD) of the U.S. Environmental Protection Agency indicated that 58% (volume-based) and 37% (creatinine-based) of the women were at risk of exposure to ∑4DEHP (HQ > 1). Based on the tolerable daily intake (TDI) from the European Food Safety Authority, 35/12% (volume-/creatinine-based data) of the women were at risk of exposure to two dibutyl phthalate (∑DBP) metabolites (MiBP and MnBP). The cumulative risk was assessed using the hazard index (HI), the sum of HQs of all phthalates. The percentages of women (volume-/creatinine-based data) at health risks with an HI > 1 were 64/40% and 42/22% based on RfD and TDI, respectively. In view of these indices for assessing risk, our results for the anti-androgenic effects of exposing pregnant women to ∑4DEHP and ∑DBP early during pregnancy are alarming.
Collapse
Affiliation(s)
- Iman Al-Saleh
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia.
| | - Rola Elkhatib
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Nujud Alrushud
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Hissah Alnuwaysir
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Maha Alnemer
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Hesham Aldhalaan
- Center for Autism Research, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Mohamed Shoukri
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Patricia McWalter
- Family Medicine and Polyclinics Department, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| | - Abdullah Alkhenizan
- Family Medicine and Polyclinics Department, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, Riyadh 11211, Saudi Arabia
| |
Collapse
|
32
|
Vrachnis N, Loukas N, Vrachnis D, Antonakopoulos N, Christodoulaki C, Tsonis O, George M, Iliodromiti Z. Phthalates and fetal growth velocity: tracking down the suspected links. J Matern Fetal Neonatal Med 2021; 35:4985-4993. [PMID: 33467971 DOI: 10.1080/14767058.2021.1873943] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fetuses that have not achieved their full growth potential are associated with adverse perinatal and long-term outcomes; thus, it is essential to identify environmental factors that can potentially impair normal intrauterine development. Endocrine disrupting compounds (EDCs), substances capable of altering the homeostasis of the endocrine system, are thought to play a role in restriction of growth velocity, with phthalates being among the most common EDCs to which pregnant women are exposed. Such exposure can potentially lead to changes to the epigenome, placental structure, and hormone function and trigger oxidative stress. Given that these pathways have been linked to fetal growth restriction, we reviewed the literature on the relationship between phthalates and fetal growth. The majority of the studies, which used birth weight as an indicator of intrauterine development, showed contradictory results, the main reason being the EDCs' rapid metabolism. However, we can draw more consistent conclusions when phthalates are quantified at more than one time point during pregnancy. In this narrative review, we present current data indicating the role of phthalates, and especially di-(2-ethylhexyl) phthalate (DEHP), in abnormal fetal growth velocity.
Collapse
Affiliation(s)
- Nikolaos Vrachnis
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, Athens, Greece.,Vascular Biology, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Nikolaos Loukas
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, Athens, Greece
| | - Dionysios Vrachnis
- Endocrinology Unit, 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Aretaieio Hospital, Athens, Greece
| | - Nikolaos Antonakopoulos
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, Athens, Greece
| | - Chryssi Christodoulaki
- Department of Obstetrics and Gynecology, Chania General Hospital "St. George", Crete, Greece
| | - Orestis Tsonis
- Department of Obstetrics and Gynecology, Medical School, University of Ioannina, Ioannina, Greece
| | - Mastorakos George
- Endocrinology Unit, 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Aretaieio Hospital, Athens, Greece
| | - Zoi Iliodromiti
- Department of Neonatology, National and Kapodistrian University of Athens Medical School, Aretaieio Hospital, Athens, Greece
| |
Collapse
|
33
|
Shi Y, Qi W, Xu Q, Wang Z, Cao X, Zhou L, Ye L. The role of epigenetics in the reproductive toxicity of environmental endocrine disruptors. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2021; 62:78-88. [PMID: 33217042 DOI: 10.1002/em.22414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 06/11/2023]
Abstract
Environmental endocrine disruptors (EEDs) seriously endanger human health by interfering with the normal function of reproductive systems. In males, EEDs can affect sperm formation and semen quality as well spermatogenesis, ultimately reducing fertility. In females, EEDs can affect uterine development and the expression levels of reproduction-related genes, ultimately reducing female fertility and the normal development of the fetus. There are a large number of putative mechanisms by which EEDs can induce reproductive toxicity, and many studies have shown the involvement of epigenetics. In this review, we summarize the role of DNA methylation, noncoding RNAs, genomic imprinting, chromatin remodeling and histone modification in the reproductive toxicity of EEDs.
Collapse
Affiliation(s)
- Yanbin Shi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Wen Qi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Qi Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Zheng Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Xiaolian Cao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
34
|
Intrauterine growth restriction: Clinical consequences on health and disease at adulthood. Reprod Toxicol 2021; 99:168-176. [DOI: 10.1016/j.reprotox.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
|
35
|
Wang JQ, Hu YB, Gao H, Sheng J, Huang K, Zhang YW, Mao LJ, Zhou SS, Cai XX, Zhang LJ, Wang SF, Hao JH, Yang LQ, Tao FB. Sex-specific difference in placental inflammatory transcriptional biomarkers of maternal phthalate exposure: a prospective cohort study. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2020; 30:835-844. [PMID: 32015430 DOI: 10.1038/s41370-020-0200-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 11/10/2019] [Accepted: 11/24/2019] [Indexed: 05/08/2023]
Abstract
Previous epidemiologic research has shown that phthalate exposure in pregnant women is related to birth outcomes in a sex-specific manner. These outcomes may be mediated by placental inflammation, which is the proposed biological mechanism. This is the first study to address the relationship between phthalate exposure and gene expression in placental inflammation in a sex-specific manner. We performed quantitative PCR to measure placental inflammatory mRNAs (CRP, TNF-α, IL-1β, IL-6, IL-10, MCP-1, IL-8, CD68, and CD206) in 2469 placentae that were sampled at birth. We estimated the associations between mRNA and urinary phthalate monoesters using multiple linear regression models. Mono-n-butyl phthalate (MBP) was correlated with higher IL-1β, IL-6, and CRP expression in placentae of male fetuses and with higher IL-6, CRP, MCP-1, IL-8, IL-10, and CD68 expression in placentae of female fetuses. Mono benzyl phthalate (MBzP) increased the expression of TNF-α, MCP-1, and CD68 only in placentae of male fetuses. Mono (2-ethyl-5-oxohexyl) phthalate (MEOHP) was negatively correlated with CRP, MCP-1, and CD68 in placentae of female fetuses. Maternal phthalate exposure was associated with inflammatory variations in placental tissues. The associations were stronger in placentae of male than of female fetuses. Compared with the other metabolites, MBP plays a strong role in these associations. The placenta is worth being further investigated as a potential mediator of maternal exposure-induced disease risk in children.
Collapse
Affiliation(s)
- Jian-Qing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ya-Bin Hu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Hui Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jie Sheng
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yun-Wei Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Lei-Jing Mao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Shan-Shan Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiu-Xiu Cai
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Liang-Jian Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Su-Fang Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jia-Hu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li-Qi Yang
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
36
|
Eaves L, Phookphan P, Rager J, Bangma J, Santos HP, Smeester L, O'Shea TM, Fry R. A role for microRNAs in the epigenetic control of sexually dimorphic gene expression in the human placenta. Epigenomics 2020; 12:1543-1558. [PMID: 32901510 PMCID: PMC7607407 DOI: 10.2217/epi-2020-0062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/03/2020] [Indexed: 12/15/2022] Open
Abstract
Aim: The contribution of miRNAs as epigenetic regulators of sexually dimorphic gene expression in the placenta is unknown. Materials & methods: 382 placentas from the extremely low gestational age newborns (ELGAN) cohort were evaluated for expression levels of 37,268 mRNAs and 2,102 miRNAs using genome-wide RNA-sequencing. Differential expression analysis was used to identify differences in the expression based on the sex of the fetus. Results: Sexually dimorphic expression was observed for 128 mRNAs and 59 miRNAs. A set of 25 miRNA master regulators was identified that likely contribute to the sexual dimorphic mRNA expression. Conclusion: These data highlight sex-dependent miRNA and mRNA patterning in the placenta and provide insight into a potential mechanism for observed sex differences in outcomes.
Collapse
Affiliation(s)
- Lauren Eaves
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Preeyaphan Phookphan
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Laksi, Bangkok 10210, Thailand
| | - Julia Rager
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jacqueline Bangma
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hudson P Santos
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Nursing, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lisa Smeester
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rebecca Fry
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
37
|
Addo KA, Palakodety N, Hartwell HJ, Tingare A, Fry RC. Placental microRNAs: Responders to environmental chemicals and mediators of pathophysiology of the human placenta. Toxicol Rep 2020; 7:1046-1056. [PMID: 32913718 PMCID: PMC7472806 DOI: 10.1016/j.toxrep.2020.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/02/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are epigenetic modifiers that play an important role in the regulation of the expression of genes across the genome. miRNAs are expressed in the placenta as well as other organs, and are involved in several biological processes including the regulation of trophoblast differentiation, migration, invasion, proliferation, apoptosis, angiogenesis and cellular metabolism. Related to their role in disease process, miRNAs have been shown to be differentially expressed between normal placentas and placentas obtained from women with pregnancy/health complications such as preeclampsia, gestational diabetes mellitus, and obesity. This dysregulation indicates that miRNAs in the placenta likely play important roles in the pathogenesis of diseases during pregnancy. Furthermore, miRNAs in the placenta are susceptible to altered expression in relation to exposure to environmental toxicants. With relevance to the placenta, the dysregulation of miRNAs in both placenta and blood has been associated with maternal exposures to several toxicants. In this review, we provide a summary of miRNAs that have been assessed in the context of human pregnancy-related diseases and in relation to exposure to environmental toxicants in the placenta. Where data are available, miRNAs are discussed in their context as biomarkers of exposure and/or disease, with comparisons made across-tissue types, and conservation across studies detailed.
Collapse
Affiliation(s)
- Kezia A. Addo
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Niharika Palakodety
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Hadley J. Hartwell
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Aishani Tingare
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C. Fry
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
38
|
Dutta S, Haggerty DK, Rappolee DA, Ruden DM. Phthalate Exposure and Long-Term Epigenomic Consequences: A Review. Front Genet 2020; 11:405. [PMID: 32435260 PMCID: PMC7218126 DOI: 10.3389/fgene.2020.00405] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/30/2020] [Indexed: 12/27/2022] Open
Abstract
Phthalates are esters of phthalic acid which are used in cosmetics and other daily personal care products. They are also used in polyvinyl chloride (PVC) plastics to increase durability and plasticity. Phthalates are not present in plastics by covalent bonds and thus can easily leach into the environment and enter the human body by dermal absorption, ingestion, or inhalation. Several in vitro and in vivo studies suggest that phthalates can act as endocrine disruptors and cause moderate reproductive and developmental toxicities. Furthermore, phthalates can pass through the placental barrier and affect the developing fetus. Thus, phthalates have ubiquitous presence in food and environment with potential adverse health effects in humans. This review focusses on studies conducted in the field of toxicogenomics of phthalates and discusses possible transgenerational and multigenerational effects caused by phthalate exposure during any point of the life-cycle.
Collapse
Affiliation(s)
- Sudipta Dutta
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Diana K Haggerty
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
| | - Daniel A Rappolee
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States.,Reproductive Stress, Inc., Grosse Pointe Farms, MI, United States
| | - Douglas M Ruden
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States.,Institutes for Environmental Health Science, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
39
|
Song W, Puttabyatappa M, Zeng L, Vazquez D, Pennathur S, Padmanabhan V. Developmental programming: Prenatal bisphenol A treatment disrupts mediators of placental function in sheep. CHEMOSPHERE 2020; 243:125301. [PMID: 31726260 PMCID: PMC7243413 DOI: 10.1016/j.chemosphere.2019.125301] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/21/2019] [Accepted: 11/02/2019] [Indexed: 05/09/2023]
Abstract
Gestational Bisphenol A (BPA) exposure is associated with low birth weight. We hypothesized that the low birth weight is the consequence of reduced placental efficiency and a function of BPA-induced inflammatory, oxidative, lipotoxic, angiogenic, steroidal and fibrotic changes involving epigenetic alterations. Placentomes were collected during early (day 65) and mid (day 90) gestation (term ∼147 days) from control and BPA (gestational day 30-90)-treated pregnant sheep. BPA treatment: reduced placental efficiency and fetal weight; increased interleukin 8, lipid peroxidation marker, antioxidants, aromatase, 17 alpha-hydroxylase, estrogen receptor 2, insulin like growth factor (IGF) 2 receptor and IGF binding proteins (IGFBP), and histone deacetylase 1 and 2; reduced tumor necrosis factor alpha and IGF1 receptor at early gestation (Day 65). Gestational BPA-induced mid-gestational changes include: reduced angiogenic factor hypoxia inducible factor 1 alpha; increased IL1beta, oxidative stress markers, triglyceride, 17alpha hydroxylase, IGFBP 1, DNA methyltransferase 3 A and histone deacetylase 1. These findings indicate that gestational BPA, either acting directly or by altering steroidal input, produces early/mid-gestational-specific epigenetic changes culminating in placental disruptions at several levels, in keeping with time-specific/time-lagged pregnancy-associated changes in placental efficiency and fetal weight. The reduced early-gestational placental efficiency may be a function of increased inflammation/oxidative stress and reduced IGF bioavailability with the mid-gestational restoration of placental efficiency likely driven by improved IGF bioavailability and the time-lagged response to antioxidant increase. This compensation, the result of time-lagged response to increases in negative mediators of placental function must have failed with pregnancy advancement to explain the low birthweight outcome.
Collapse
Affiliation(s)
- Wenhui Song
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050011, PR China; Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Lixia Zeng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Delia Vazquez
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | | |
Collapse
|
40
|
Zota AR, Geller RJ, VanNoy BN, Marfori CQ, Tabbara S, Hu LY, Baccarelli AA, Moawad GN. Phthalate Exposures and MicroRNA Expression in Uterine Fibroids: The FORGE Study. Epigenet Insights 2020; 13:2516865720904057. [PMID: 32128507 PMCID: PMC7031793 DOI: 10.1177/2516865720904057] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022] Open
Abstract
Phthalates are associated with multiple, adverse reproductive outcomes including increased risk of uterine leiomyoma (fibroids). Phthalates can interact with epigenetic modifications including microRNAs (miRNAs), which help regulate processes crucial to fibroid pathogenesis. However, no prior study has examined the influence of phthalates on miRNA expression in fibroid tumors. We conducted a preliminary, cross-sectional study to examine the associations between phthalate exposures and miRNA expression levels in fibroid tumors and to explore potential effect modification by race/ethnicity. We quantified expression levels of 754 miRNAs in fibroid tumor samples and analyzed spot urine samples for phthalate metabolites collected from 45 pre-menopausal women undergoing surgery for fibroid treatment at an academic hospital. Associations between miRNA levels in fibroids and phthalate biomarkers were evaluated using linear regression adjusting for age, race/ethnicity, and body mass index (BMI). Statistical tests were adjusted for multiple comparisons. We also performed in silico Ingenuity Pathway Analysis to identify the biological pathways that are regulated by phthalate-associated miRNAs. Mono-hydroxybutyl phthalate and mono(2-ethyl-5-hydroxyhexyl) phthalate were positively associated with miR-10a-5p (β = 0.76, 95% CI = [0.40, 1.11]) and miR-577 (β = 1.06, 95% CI = [0.53, 1.59]), respectively. A total of 8 phthalate-miRNA associations varied by race/ethnicity (qinteraction < 0.10). Pathway analysis revealed that mRNA gene targets of phthalate-associated miRNAs were significantly associated with multiple fibroid-related processes including angiogenesis, apoptosis, and proliferation of connective tissues. Collectively, these data suggest that exposures to some phthalates are associated with miRNA in fibroids, and that associations may vary by race/ethnicity. Validation of these findings may provide insight into mechanisms underlying associations between phthalates and fibroids and contribute to novel hypotheses regarding racial/ethnic disparities in fibroids.
Collapse
Affiliation(s)
- Ami R Zota
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Ruth J Geller
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Brianna N VanNoy
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Cherie Q Marfori
- Department of Obstetrics & Gynecology, The George Washington University, Washington, DC, USA
| | - Sana Tabbara
- Department of Pathology, The George Washington University, Washington, DC, USA
| | - Lisa Y Hu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Gaby N Moawad
- Department of Obstetrics & Gynecology, The George Washington University, Washington, DC, USA
| |
Collapse
|
41
|
Gomez SD, Bustos PS, Sánchez VG, Ortega MG, Guiñazú N. Trophoblast toxicity of the neonicotinoid insecticide acetamiprid and an acetamiprid-based formulation. Toxicology 2020; 431:152363. [DOI: 10.1016/j.tox.2020.152363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/11/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
|
42
|
Jin C, Yao Q, Zhou Y, Shi R, Gao Y, Wang C, Tian Y. Exposure to triclosan among pregnant women in northern China: urinary concentrations, sociodemographic predictors, and seasonal variability. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:4840-4848. [PMID: 31845258 DOI: 10.1007/s11356-019-07294-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 12/04/2019] [Indexed: 06/10/2023]
Abstract
Although triclosan (TCS) use is widespread in China, little is known about levels of exposure to TCS in pregnant women and its potential predictors, sources, and seasonal variability. We assessed urinary TCS levels of 466 pregnant women in a Chinese cohort. The estimated daily intake (EDI) and hazard quotient (HQ) were calculated. Potential predictors and sources were collected through a questionnaire and the seasonal variability was recorded based on the time of sampling. The geometric mean of urinary TCS concentration was 0.81 μg/g. The 95th EDI was 0.15 μg/kg BW per day and the corresponding HQ was 3.23 × 10-3. Women with a household monthly salary between RMB (¥) 1000 and 3000 and between RMB (¥) 3000 and 5000 had 0.52 μg/g (95% CI 0.08, 0.75) and 0.58 μg/g (95% CI 0.17, 0.79) lower urinary TCS levels than those with a household monthly salary of < RMB (¥) 1000, respectively. Urine samples collected in winter had lower TCS levels (geometric mean 0.72 μg/g) than in spring, summer, and autumn (geometric mean 0.82, 0.84, and 0.86 μg/g), although they were non-significant (P = 0.648). No association was found between drinking water and food consumed during pregnancy and TCS levels. The study population was ubiquitously exposed to a relatively low and safe dose of TCS. Women with lower household income tended to be exposed to higher levels of TCS.
Collapse
Affiliation(s)
- Chenye Jin
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qian Yao
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yijun Zhou
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Rong Shi
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yu Gao
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Caifeng Wang
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
- School of Nursing, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ying Tian
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
- MOE and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
44
|
Chung FFL, Herceg Z. The Promises and Challenges of Toxico-Epigenomics: Environmental Chemicals and Their Impacts on the Epigenome. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:15001. [PMID: 31950866 PMCID: PMC7015548 DOI: 10.1289/ehp6104] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/15/2019] [Accepted: 12/16/2019] [Indexed: 05/02/2023]
Abstract
BACKGROUND It has been estimated that a substantial portion of chronic and noncommunicable diseases can be caused or exacerbated by exposure to environmental chemicals. Multiple lines of evidence indicate that early life exposure to environmental chemicals at relatively low concentrations could have lasting effects on individual and population health. Although the potential adverse effects of environmental chemicals are known to the scientific community, regulatory agencies, and the public, little is known about the mechanistic basis by which these chemicals can induce long-term or transgenerational effects. To address this question, epigenetic mechanisms have emerged as the potential link between genetic and environmental factors of health and disease. OBJECTIVES We present an overview of epigenetic regulation and a summary of reported evidence of environmental toxicants as epigenetic disruptors. We also discuss the advantages and challenges of using epigenetic biomarkers as an indicator of toxicant exposure, using measures that can be taken to improve risk assessment, and our perspectives on the future role of epigenetics in toxicology. DISCUSSION Until recently, efforts to apply epigenomic data in toxicology and risk assessment were restricted by an incomplete understanding of epigenomic variability across tissue types and populations. This is poised to change with the development of new tools and concerted efforts by researchers across disciplines that have led to a better understanding of epigenetic mechanisms and comprehensive maps of epigenomic variation. With the foundations now in place, we foresee that unprecedented advancements will take place in the field in the coming years. https://doi.org/10.1289/EHP6104.
Collapse
Affiliation(s)
| | - Zdenko Herceg
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
45
|
Strakovsky RS, Schantz SL. Using Experimental Models to Assess Effects of Bisphenol A (BPA) and Phthalates on the Placenta: Challenges and Perspectives. Toxicol Sci 2019; 166:250-268. [PMID: 30203063 DOI: 10.1093/toxsci/kfy224] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The placenta is critical for all aspects of fetal development. Bisphenol A (BPA) and phthalates are endocrine disruptors with ubiquitous exposure in pregnant women-their effects on the placenta is an area of growing research interest. Therefore, our objectives were to (1) summarize research related to the effects BPA or phthalates on placental outcomes in animal and cell models, and (2) evaluate the challenges for using such models to study the impacts of these chemicals on placental endpoints. Overall, studies in cells and animal models suggest that BPA and phthalates impact placental hormones, some epigenetic endpoints, increase inflammation and oxidative stress, and decrease cell viability and nutrient transfer. However, few animal or cell studies have assessed these outcomes at concentrations relevant to humans. Furthermore, it is unclear whether effects of BPA/phthalates on the placenta in animal models mediate fetal outcomes, as most studies have dosed after the earliest stages of placental and fetal development. It is also unclear whether effects of these chemicals are sex-specific, as few studies have considered placental sex. Finally, while there is substantial evidence for effects of mono-(2-ethylhexyl) phthalate (the major metabolite of di-(2-ethylhexyl) phthalate), on placental endpoints in cells, little is currently known about effects of other phthalates to which pregnant women are exposed. Moving forward, these limitations will need to be addressed to help us understand the precise mechanisms of action of these chemicals within the placenta, and how these reported perturbations impact fetal health.
Collapse
Affiliation(s)
- Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48823
| | - Susan L Schantz
- Beckman Institute for Advanced Science and Technology.,Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2347 Beckman Institute, Urbana, Illinois 61801
| |
Collapse
|
46
|
Sonkar R, Kay MK, Choudhury M. PFOS Modulates Interactive Epigenetic Regulation in First-Trimester Human Trophoblast Cell Line HTR-8/SV neo. Chem Res Toxicol 2019; 32:2016-2027. [PMID: 31508952 DOI: 10.1021/acs.chemrestox.9b00198] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Organic compounds have been linked to adverse pregnancy complications. Perfluorooctanesulfonic acid (PFOS), a man-made fluorosurfactant and global pollutant, has been shown to induce oxidative stress in various cell types. Oxidative stress plays a key role in leading several placental diseases including preeclampsia (PE), gestational diabetes, spontaneous abortion, preterm labor, and intrauterine growth restriction. Recently, epigenetic regulation such as histone modifications, DNA methylation, and microRNAs (miRNAs), are shown to be associated with oxidative stress as well as pregnancy complications such as PE. However, whether PFOS exerts its detrimental effects in the placenta through epigenetics remains to be unveiled. Therefore, we aimed to investigate the effect of PFOS-induced reactive oxygen species (ROS) generation in first trimester human trophoblast cell line (HTR-8/SVneo) and whether epigenetic regulation is involved in this process. When treated with a range of PFOS doses at 24 and 48 h, even at 10 μM, it significantly increased the ROS production and decreased gene and protein expression, respectively, of the DNA methyltransferases DNMT1 (p < 0.001; p < 0.05), DNMT3A (p < 0.001; p < 0.05), and DNMT3B (p < 0.01; p < 0.01) and the sirtuins, for example, SIRT1 (p < 0.001; p < 0.001) and SIRT3 (p < 0.001; p < 0.05), while reducing global DNA methylation (p < 0.01) and increasing protein lysine acetylation (p < 0.001) as compared to vehicle controls. Interestingly, PFOS (10 μM) significantly increased miR29-b (p < 0.01), which has been previously reported to be associated with PE. The observed epigenetic effects were shown to be dependent on the expression of miR-29b, as knockdown of miR-29b significantly alters the gene and protein expression of DNMT1, DNMT3A, DNMT3B, SIRT1, and SIRT3 and ROS production as well as global DNA methylation and protein acetylation. This study provides for the first time a novel insight into PFOS-induced ROS generation via regulation of sets of the interactive epigenetic circuit in the placenta, which may lead to pregnancy complications.
Collapse
Affiliation(s)
- Ravi Sonkar
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| | - Matthew K Kay
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy , Texas A&M Health Science Center , 312 REYN, MS 1114 , College Station , Texas 77843 , United States
| |
Collapse
|
47
|
Saenen ND, Martens DS, Neven KY, Alfano R, Bové H, Janssen BG, Roels HA, Plusquin M, Vrijens K, Nawrot TS. Air pollution-induced placental alterations: an interplay of oxidative stress, epigenetics, and the aging phenotype? Clin Epigenetics 2019; 11:124. [PMID: 31530287 PMCID: PMC6749657 DOI: 10.1186/s13148-019-0688-z] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/27/2019] [Indexed: 01/04/2023] Open
Abstract
According to the "Developmental Origins of Health and Disease" (DOHaD) concept, the early-life environment is a critical period for fetal programming. Given the epidemiological evidence that air pollution exposure during pregnancy adversely affects newborn outcomes such as birth weight and preterm birth, there is a need to pay attention to underlying modes of action to better understand not only these air pollution-induced early health effects but also its later-life consequences. In this review, we give an overview of air pollution-induced placental molecular alterations observed in the ENVIRONAGE birth cohort and evaluate the existing evidence. In general, we showed that prenatal exposure to air pollution is associated with nitrosative stress and epigenetic alterations in the placenta. Adversely affected CpG targets were involved in cellular processes including DNA repair, circadian rhythm, and energy metabolism. For miRNA expression, specific air pollution exposure windows were associated with altered miR-20a, miR-21, miR-146a, and miR-222 expression. Early-life aging markers including telomere length and mitochondrial DNA content are associated with air pollution exposure during pregnancy. Previously, we proposed the air pollution-induced telomere-mitochondrial aging hypothesis with a direct link between telomeres and mitochondria. Here, we extend this view with a potential co-interaction of different biological mechanisms on the level of placental oxidative stress, epigenetics, aging, and energy metabolism. Investigating the placenta is an opportunity for future research as it may help to understand the fundamental biology underpinning the DOHaD concept through the interactions between the underlying modes of action, prenatal environment, and disease risk in later life. To prevent lasting consequences from early-life exposures of air pollution, policy makers should get a basic understanding of biomolecular consequences and transgenerational risks.
Collapse
Affiliation(s)
- N. D. Saenen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - D. S. Martens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - K. Y. Neven
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - R. Alfano
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - H. Bové
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - B. G. Janssen
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - H. A. Roels
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - M. Plusquin
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - K. Vrijens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - T. S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Department of Public Health and Primary Care, Leuven University, Leuven, Belgium
| |
Collapse
|
48
|
Martínez-Ibarra A, Martínez-Razo LD, Vázquez-Martínez ER, Martínez-Cruz N, Flores-Ramírez R, García-Gómez E, López-López M, Ortega-González C, Camacho-Arroyo I, Cerbón M. Unhealthy Levels of Phthalates and Bisphenol A in Mexican Pregnant Women with Gestational Diabetes and Its Association to Altered Expression of miRNAs Involved with Metabolic Disease. Int J Mol Sci 2019; 20:ijms20133343. [PMID: 31284700 PMCID: PMC6650872 DOI: 10.3390/ijms20133343] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Several studies indicate that bisphenol A (BPA) and phthalates may have a role in the development of metabolic diseases using different molecular pathways, including epigenetic regulatory mechanisms. However, it is unclear whether exposure to these chemicals modifies serum levels of miRNAs associated with gestational diabetes mellitus (GDM) risk. In the present study, we evaluated the serum levels of miRNAs associated with GDM (miR-9-5p, miR-16-5p, miR-29a-3p and miR-330-3p) and urinary levels of phthalate metabolites (mono-n-butyl phthalate (MBP), mono-isobutyl phthalate (MiBP), mono-benzyl phthalate (MBzP) and mono(2-ethyl hexyl) phthalate (MEHP)) and bisphenol A in GDM patients and women without GDM during the second trimester of gestation. We observed higher levels of miR-9-5p, miR-29a-3p and miR-330-3p in sera of patients with GDM compared to non-diabetic subjects. Phthalates were detected in 97–100% of urine samples, while BPA only in 40%. Urinary MEHP and BPA concentrations were remarkably higher in both study groups compared to previously reported data. Unadjusted MEHP levels and adjusted BPA levels were higher in non-diabetics than in GDM patients (p = 0.03, p = 0.02). We found positive correlations between adjusted urinary MBzP levels and miR-16-5p expression levels (p < 0.05), adjusted MEHP concentrations and miR-29a-3p expression levels (p < 0.05). We also found negative correlations between unadjusted and adjusted MBP concentrations and miR-29a-3p expression levels (p < 0.0001, p < 0.05), unadjusted MiBP concentrations and miR-29a-3p expression levels (p < 0.01). Urinary MEHP levels reflect a striking exposure to di(2-ethylhexyl) phthalate (DEHP) in pregnant Mexican women. This study highlights the need for a regulatory strategy in the manufacture of several items containing endocrine disruptors in order to avoid involuntary ingestion of these compounds in the Mexican population.
Collapse
Affiliation(s)
- Alejandra Martínez-Ibarra
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, México
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México
| | - Luis Daniel Martínez-Razo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México
| | - Nayeli Martínez-Cruz
- Coordinación del Servicio de Endocrinología, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México 11000, México
| | - Rogelio Flores-Ramírez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Elizabeth García-Gómez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México
| | - Marisol López-López
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Ciudad de México 04960, México
| | - Carlos Ortega-González
- Coordinación del Servicio de Endocrinología, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México 11000, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, México.
| |
Collapse
|
49
|
Kamai EM, McElrath TF, Ferguson KK. Fetal growth in environmental epidemiology: mechanisms, limitations, and a review of associations with biomarkers of non-persistent chemical exposures during pregnancy. Environ Health 2019; 18:43. [PMID: 31068204 PMCID: PMC6505101 DOI: 10.1186/s12940-019-0480-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/16/2019] [Indexed: 05/18/2023]
Abstract
BACKGROUND Non-persistent chemicals, such as phthalates, environmental phenols, organophosphate pesticides, and others, are challenging to study because of their ubiquity in the environment, diverse exposure routes, and high temporal variability of biomarkers. Nonetheless, there is interest in understanding how gestational exposure to these chemicals may affect fetal growth, as perturbations to normal fetal growth are related to a plethora of adverse health outcomes in childhood and adulthood. METHODS The purpose of this review is to describe the state of the science on this topic. We searched PubMed for studies that included both 1) biomarkers of non-persistent chemicals collected during pregnancy and 2) fetal growth outcomes measured at birth (e.g., birth weight) or by ultrasound in utero (e.g., estimated fetal weight). RESULTS The bulk of the literature we found uses biomarkers measured at a single time point in pregnancy and birth weight as the primary measure of fetal growth. There is a small, but growing, body of research that uses ultrasound measures to assess fetal growth during pregnancy. In addition to summarizing the findings of the publications we identified, we describe inconsistencies in methodology, areas for improvement, and gaps in existing knowledge that can be targeted for improvement in future work. This literature is characterized by variability in methodology, likely contributing to the inconsistency of results reported. We further discuss maternal, placental, and fetal pathways by which these classes of chemicals may affect fetal growth. CONCLUSIONS To improve understanding of how everyday chemical exposures affect fetal growth, and ultimately lifelong health outcomes, mechanisms of toxicant action should be considered alongside improved study designs for future hypothesis-driven research.
Collapse
Affiliation(s)
- Elizabeth M. Kamai
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 135 Dauer Drive, 2101 McGavran-Greenberg Hall, CB #7435, Chapel Hill, NC 27599 USA
| | - Thomas F. McElrath
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Kelly K. Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Drive, Research Triangle Park, NC 27709 USA
| |
Collapse
|
50
|
Perera BPU, Svoboda L, Dolinoy DC. Genomic Tools for Environmental Epigenetics and Implications for Public Health. CURRENT OPINION IN TOXICOLOGY 2019; 18:27-33. [PMID: 31763499 DOI: 10.1016/j.cotox.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Epigenetics refers to the study of mitotically heritable and potentially reversible changes in gene expression unrelated to the DNA sequence itself, influenced by epigenetic marks including chromatin modifications, non-coding RNA and alterations to DNA itself via methylation and hydroxymethylation. Epigenetics has taken center stage in the study of diseases such as cancer, diabetes, and neurodegeneration; however, its integration into the field of environmental health sciences and toxicology (e.g. Toxicoepigenetics) is in its infancy. This review highlights the need to evaluate surrogate and target tissues in the field of toxicoepigenetics as the National Institute of Environmental Health Sciences (NIEHS) multi-phased Toxicant Exposure and Response by Genomic and Epigenomic Regulators of Transcription (TaRGET) consortia make headway, and the emergence of non-coding RNA biomarkers. The review also discusses lead (Pb) as a potential toxicoepigenetic exposure, where pre- and post-natal Pb exposure is associated with reprogramming of DNA methylation, histone modifications, and microRNA expression, representing potential biomarkers or predictors for Pb-induced health outcomes. Finally, new advances in epigenome editing, highlighting the potential of small ncRNA, will be explored for environmental health sciences research.
Collapse
Affiliation(s)
- Bambarendage P U Perera
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
| | - Laurie Svoboda
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
| | - Dana C Dolinoy
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
- University of Michigan School of Public Health, Department of Nutritional Sciences, Ann Arbor, MI
| |
Collapse
|