1
|
Alahmadi H, Martinez S, Farrell R, Bikienga R, Arinzeh N, Potts C, Li Z, Warner GR. Mixtures of phthalates disrupt expression of genes related to lipid metabolism and peroxisome proliferator-activated receptor signaling in mouse granulosa cells. Toxicol Sci 2024; 202:69-84. [PMID: 39150890 PMCID: PMC11514836 DOI: 10.1093/toxsci/kfae105] [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] [Indexed: 08/18/2024] Open
Abstract
Phthalates are a class of known endocrine-disrupting chemicals that are found in common everyday products. Several studies associate phthalate exposure with detrimental effects on ovarian function, including growth and development of the follicle and production of steroid hormones. We hypothesized that dysregulation of the ovary by phthalates may be mediated by phthalate toxicity towards granulosa cells, a major cell type in ovarian follicles responsible for key steps of hormone production and nourishing the developing oocyte. To test the hypothesis that phthalates target granulosa cells, we harvested granulosa cells from adult CD-1 mouse ovaries and cultured them for 96 h in vehicle control, a phthalate mixture, or a phthalate metabolite mixture (0.1 to 100 μg/ml). After culture, we measured metabolism of the phthalate mixture into monoester metabolites by the granulosa cells, finding that granulosa cells do not significantly contribute to ovarian metabolism of phthalates. Immunohistochemistry of phthalate metabolizing enzymes in whole ovaries confirmed that these enzymes are not strongly expressed in granulosa cells of antral follicles and that ovarian metabolism of phthalates likely occurs primarily in the stroma. RNA sequencing of treated granulosa cells identified 407 differentially expressed genes, with overrepresentation of genes from lipid metabolic processes, cholesterol metabolism, and peroxisome proliferator-activated receptor (PPAR) signaling pathways. Expression of significantly differentially expressed genes related to these pathways was confirmed using qPCR. Our results agree with previous findings that phthalates and phthalate metabolites have different effects on the ovary, but both interfere with PPAR signaling in granulosa cells.
Collapse
Affiliation(s)
- Hanin Alahmadi
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Stephanie Martinez
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Rivka Farrell
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Rafiatou Bikienga
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Nneka Arinzeh
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Courtney Potts
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Zhong Li
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Genoa R Warner
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| |
Collapse
|
2
|
Alahmadi H, Martinez S, Farrell R, Bikienga R, Arinzeh N, Potts C, Li Z, Warner GR. Mixtures of phthalates disrupt expression of genes related to lipid metabolism and peroxisome proliferator-activated receptor signaling in mouse granulosa cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592217. [PMID: 38746167 PMCID: PMC11092572 DOI: 10.1101/2024.05.02.592217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Phthalates are a class of known endocrine disrupting chemicals that are found in common everyday products. Several studies associate phthalate exposure with detrimental effects on ovarian functions, including growth and development of the follicle and production of steroid hormones. We hypothesized that dysregulation of the ovary by phthalates may be mediated by phthalate toxicity towards granulosa cells, a major cell type in ovarian follicles responsible for key steps of hormone production and nourishing the developing oocyte. To test the hypothesis that phthalates target granulosa cells, we harvested granulosa cells from adult CD-1 mouse ovaries and cultured them for 96 hours in vehicle control, a phthalate mixture, or a phthalate metabolite mixture (0.1-100 μg/mL). After culture, we measured metabolism of the phthalate mixture into monoester metabolites by the granulosa cells, finding that granulosa cells do not significantly contribute to ovarian metabolism of phthalates. Immunohistochemistry of phthalate metabolizing enzymes in whole ovaries confirmed that these enzymes are not strongly expressed in granulosa cells of antral follicles and that ovarian metabolism of phthalates likely occurs primarily in the stroma. RNA sequencing of treated granulosa cells identified 407 differentially expressed genes, with overrepresentation of genes from lipid metabolic processes, cholesterol metabolism, and peroxisome proliferator-activated receptor (PPAR) signaling pathways. Expression of significantly differentially expressed genes related to these pathways were confirmed using qPCR. Our results agree with previous findings that phthalates and phthalate metabolites have different effects on the ovary and interfere with PPAR signaling in granulosa cells.
Collapse
|
3
|
Ardenkjær-Skinnerup J, Saar D, Petersen PSS, Pedersen M, Svingen T, Kragelund BB, Hadrup N, Ravn-Haren G, Emanuelli B, Brown KA, Vogel U. PPARγ antagonists induce aromatase transcription in adipose tissue cultures. Biochem Pharmacol 2024; 222:116095. [PMID: 38423186 DOI: 10.1016/j.bcp.2024.116095] [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: 10/31/2023] [Revised: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens and a key risk factor for hormone receptor-positive breast cancer. In postmenopausal women, estrogens synthesized in adipose tissue promotes the growth of estrogen receptor positive breast cancers. Activation of peroxisome proliferator-activated receptor gamma (PPARγ) in adipose stromal cells (ASCs) leads to decreased expression of aromatase and differentiation of ASCs into adipocytes. Environmental chemicals can act as antagonists of PPARγ and disrupt its function. This study aimed to test the hypothesis that PPARγ antagonists can promote breast cancer by stimulating aromatase expression in human adipose tissue. Primary cells and explants from human adipose tissue as well as A41hWAT, C3H10T1/2, and H295R cell lines were used to investigate PPARγ antagonist-stimulated effects on adipogenesis, aromatase expression, and estrogen biosynthesis. Selected antagonists inhibited adipocyte differentiation, preventing the adipogenesis-associated downregulation of aromatase. NMR spectroscopy confirmed direct interaction between the potent antagonist DEHPA and PPARγ, inhibiting agonist binding. Short-term exposure of ASCs to PPARγ antagonists upregulated aromatase only in differentiated cells, and a similar effect could be observed in human breast adipose tissue explants. Overexpression of PPARG with or without agonist treatment reduced aromatase expression in ASCs. The data suggest that environmental PPARγ antagonists regulate aromatase expression in adipose tissue through two mechanisms. The first is indirect and involves inhibition of adipogenesis, while the second occurs more acutely.
Collapse
Affiliation(s)
- Jacob Ardenkjær-Skinnerup
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | - Daniel Saar
- REPIN and Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Patricia S S Petersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen N, Denmark
| | - Mikael Pedersen
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Terje Svingen
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Birthe B Kragelund
- REPIN and Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Niels Hadrup
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | - Gitte Ravn-Haren
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Brice Emanuelli
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen N, Denmark
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Ulla Vogel
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark.
| |
Collapse
|
4
|
Gao M, Shen H, Li Q, Gu X, Jia T, Wang Y. Perfluorooctane sulfonate (PFOS) induces apoptosis and autophagy by inhibition of PI3K/AKT/mTOR pathway in human granulosa cell line KGN. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123333. [PMID: 38211877 DOI: 10.1016/j.envpol.2024.123333] [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: 10/17/2023] [Revised: 12/22/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is recognized as an environmental endocrine disruptor with widespread use in industrial manufacturing and daily life, contributing to various public health concerns. However, the precise impacts of PFOS on the ovary and its regulatory mechanisms remain unclear. This study aims to delineate the ovarian toxicity of PFOS and scrutinize its effects on apoptosis and autophagy through modulation of the PI3K/AKT/mTOR pathway in the human granulosa cell line (KGN). Cell viability, assessed via the Cell Counting Kit-8 (CCK8), revealed a dose-dependent reduction in cell viability upon PFOS exposure. Flow cytometry analysis demonstrated an elevated proportion of apoptotic cells following PFOS treatment. Western blot analyses unveiled increased expression of Bax, Cyt c, cleaved caspase-9, and LC3-II/I, coupled with decreased expression of Bcl-2 and p62. Transmission electron microscopy (TEM) observations illustrated a heightened number of autophagosomes induced by PFOS. Molecular docking investigations, in conjunction with Western blot experiments, substantiated PFOS's significant inhibition of the PI3K/AKT/mTOR signaling pathway. These findings collectively underscore that PFOS induces apoptosis and autophagy in KGN cells through modulation of the PI3K/AKT/mTOR pathway, providing experimental evidence for PFOS-induced ovarian toxicity and elucidating the underlying regulatory mechanisms in KGN cells.
Collapse
Affiliation(s)
- Min Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haofei Shen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiuyuan Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuzhao Gu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tianyu Jia
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Yiqing Wang
- The First School of Clinical Medicine & Research Unit of Peptide Science, Chinese Academy of Medical Science, 2019RU066, Lanzhou University, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Reproductive Medicine Transformation Application, Key Laboratory for Reproductive Medicine and Embryo of Gansu Province & Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China.
| |
Collapse
|
5
|
Gao C, Sun N, Xie J, Li J, Tao L, Guo L, Shi L, He X, Shen X, Wang H, Yang P, Covaci A, Huang Y. Co-exposure to 55 endocrine-disrupting chemicals linking diminished sperm quality: Mixture effect, and the role of seminal plasma docosapentaenoic acid. ENVIRONMENT INTERNATIONAL 2024; 185:108571. [PMID: 38471262 DOI: 10.1016/j.envint.2024.108571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Isolated effects of single endocrine-disrupting chemicals (EDCs) on male reproductive health have been studied extensively, but their mixture effect remains unelucidated. Previous research has suggested that consuming diet enriched in omega-3 polyunsaturated fatty acids (PUFA) might be beneficial for reproductive health, whether omega-3 PUFA could moderate the effect of EDCs mixture on semen quality remains to be explored. In this study of 155 male recruited from a reproductive health center in China, we used targeted-exposomics to simultaneously measure 55 EDCs in the urine for exposure burden. Regression analyses were restricted to highly detected EDCs (≥55%, n = 34), and those with consistently elevated risk were further screened and brought into mixture effect models (Bisphenol A, ethyl paraben, methyl paraben [MeP], benzophenone-1 [BP1], benzophenone-3, mono(3-carboxypropyl) phthalate [MCPP]). Bayesian Kernel Machine Regression (BKMR) and quantile-based g-computation (QGC) models demonstrated that co-exposure to top-ranked EDCs was related to reduced sperm total (β = -0.18, 95%CI: -0.29 - -0.07, P = 0.002) and progressive motility (β = -0.27, 95%CI: -0.43 - -0.10, P = 0.002), but not to lower semen volume. BP1, MeP and MCPP were identified as the main effect driver for deteriorated sperm motion parameters using mixture model analyses. Seminal plasma fatty acid profiling showed that high omega-3 PUFA status, notably elevated docosapentaenoic acid (DPA, C22:5n-3) status, moderated the association between MCPP and sperm motion parameters (total motility: β = 0.26, 95%CI: 0.01 - -0.51, Pinteraction = 0.047; progressive motility: β = 0.64, 95%CI: 0.23 - 1.05, Pinteraction = 0.003). Co-exposure to a range of EDCs is mainly associated with deteriorated sperm quality, but to a lesser extent on sperm quantity, high seminal plasma DPA status might be protective against the effect. Our work emphasizes the importance of exposomic approach to assess chemical exposures and highlighted a new possible intervention target for mitigating the potential adverse effect of EDCs on semen quality.
Collapse
Affiliation(s)
- Chang Gao
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China
| | - Nan Sun
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China
| | - Jinying Xie
- Department of Public Health and Preventive Medicine, China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China
| | - Jiehao Li
- Department of Public Health and Preventive Medicine, China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China
| | - Lin Tao
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China
| | - Lijuan Guo
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China
| | - Lan Shi
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaojin He
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui Province, China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoting Shen
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China
| | - Pan Yang
- Department of Public Health and Preventive Medicine, China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China.
| | - Adrian Covaci
- Toxicological Center, University of Antwerp, Wilrijk, Belgium
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Center for Big Data and Population Health of IHM, Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, China; Clinical Research Center, Suzhou Hospital of Anhui Medical University, Anhui Medical University, Suzhou, China.
| |
Collapse
|
6
|
Zhang Y, Yang Y, Tao Y, Guo X, Cui Y, Li Z. Phthalates (PAEs) and reproductive toxicity: Hypothalamic-pituitary-gonadal (HPG) axis aspects. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132182. [PMID: 37557049 DOI: 10.1016/j.jhazmat.2023.132182] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
Phthalates (PAEs) are widely used for their excellent ability to improve plastic products. As an essential endocrine axis that regulates the reproductive system, whether dysfunction of the hypothalamic-pituitary-gonadal (HPG) axis is involved in reproductive toxicity mediated by environmental endocrine disruptors PAEs has become a hot topic of widespread concern. This study systematically reviewed the adverse effects of multiple PAEs on the HPG axis in different models and objectively discussed the possible underlying mechanisms. The abnormal release of gonadotropin-releasing hormone and gonadotropin, dysfunction of sex hormone receptors and steroid hormone synthesis, and general damage, including cell proliferation, oxidative stress, apoptosis, and autophagy have been confirmed to be involved in this process. Although it is widely established that PAEs induce HPG axis dysfunction, the specific mechanisms involved remain unclear. From a systematic review of relevant publications, it appears that the abnormal expression of peroxisome proliferator-activated, aryl hydrocarbon, and insulin receptors mediated by PAEs is key upstream event that induces these adverse outcomes; however, this inference needs to be further verified. Overall, this study aimed to provide reliable potential biomarkers for future environmental risk assessment and epidemiological investigation of PAEs.
Collapse
Affiliation(s)
- Ying Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China.
| | - Yang Yang
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Tao
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiangyong Guo
- Fuyu County Agricultural Technology Extension Center, Qiqihar 161200, PR China
| | - Yunhe Cui
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Zixu Li
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
7
|
Sukur N, Milošević N, Pogrmic-Majkic K, Stanic B, Andric N. Predicting chemicals' toxicity pathway of female reproductive disorders using AOP7 and deep neural networks. Food Chem Toxicol 2023; 180:114013. [PMID: 37683992 DOI: 10.1016/j.fct.2023.114013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/05/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023]
Abstract
Experimental evidence shows that certain chemicals, particularly endocrine disrupting chemicals, may negatively affect the female reproductive system, thereby lowering women's fertility. However, humans are constantly exposed to a number of different chemicals with limited or no experimental data regarding their effect and the mechanism of action in the female reproductive system. To predict chemical hazards to the female reproductive system, we used a previously defined adverse outcome pathway (AOP) that links activation of the peroxisome proliferator-activated receptor γ to the reproductive toxicity in adult females (AOP7) and the Convolutional Deep Neural Network models that produce meaningful predictions when trained on a significant amount of data. The models trained using CompTox assays with intended molecular and biological targets corresponding to AOP7 achieved high performance (over 90% validation accuracy). The integration of AOP7 and Deep Neural Network identified chemicals that could negatively affect female reproduction through the mechanism described in AOP7. We provide a solution to quickly analyze the data and produce machine learning models to identify potentially active chemicals in the female reproductive system. Although we focused on the female reproductive system, this approach could be valid for a number of other chemicals and AOPs if the right data exist.
Collapse
Affiliation(s)
- Nataša Sukur
- University of Novi Sad, Faculty of Sciences, Department of Mathematics and Informatics, Trg Dositeja Obradovica 4, Novi Sad, 21000, Serbia.
| | - Nemanja Milošević
- University of Novi Sad, Faculty of Sciences, Department of Mathematics and Informatics, Trg Dositeja Obradovica 4, Novi Sad, 21000, Serbia
| | - Kristina Pogrmic-Majkic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Trg Dositeja Obradovica 2, Novi Sad, 21000, Serbia
| | - Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Trg Dositeja Obradovica 2, Novi Sad, 21000, Serbia
| | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Trg Dositeja Obradovica 2, Novi Sad, 21000, Serbia
| |
Collapse
|
8
|
Zhu L, Yao X, Mo Y, Chen MW, Li SC, Liu JQ, Liu HY. miR-4433a-3p promotes granulosa cell apoptosis by targeting peroxisome proliferator-activated receptor alpha and inducing immune cell infiltration in polycystic ovarian syndrome. J Assist Reprod Genet 2023; 40:1447-1459. [PMID: 37204637 PMCID: PMC10310670 DOI: 10.1007/s10815-023-02815-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/24/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Granulosa cell (GC) proliferation and apoptosis are critical events of the ovum energy supply, which lead to follicular growth retardation or atresia, and various ovulatory obstacles, eventually resulting in the development of ovarian disorders such as polycystic ovarian syndrome (PCOS). Apoptosis and dysregulated miRNA expression in GCs are manifestations of PCOS. miR-4433a-3p has been reported to be involved in apoptosis. However, there is no study reporting the roles of miR-4433a-3p in GC apoptosis and PCOS progression. METHODS miR-4433a-3p and peroxisome proliferator-activated receptor alpha (PPAR-α) levels in GCs of PCOS patients or in tissues of a PCOS rat model were examined by quantitative polymerase chain reaction and immunohistochemistry. Bioinformatics analyses and luciferase assays were used to examine the association between miR-4433a-3p and PPAR-α, as well as PPAR-α and immune cell infiltration, in PCOS patients. RESULTS miR-4433a-3p expression in GCs of PCOS patients was increased. miR-4433a-3p overexpression inhibited the growth of the human granulosa-like tumor cell line (KGN) and promoted apoptosis, while co-treatment with PPAR-α and miR-4433a-3p mimic rescued miR-4433a-3p-induced apoptosis. PPAR-α was a direct target of miR-4433a-3p and its expression was decreased in PCOS patients. PPAR-α expression was also positively correlated with the infiltration of activated CD4+ T cells, eosinophils, B cells, gamma delta T cells, macrophages, and mast cells, but negatively correlated with the infiltration of activated CD8+ T cells, CD56+ bright natural killer cells, immature dendritic cells, monocytes, plasmacytoid dendritic cells, neutrophils, and type 1 T helper cells in PCOS patients. CONCLUSION The miR-4433a-3p/PPAR-α/immune cell infiltration axis may function as a novel cascade to alter GC apoptosis in PCOS.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xi Yao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Mo
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ming-Wei Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Si-Chen Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian-Qiao Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Hai-Ying Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Mao X, Li H, Zheng J. Effects of xenobiotics on CYP1 enzyme-mediated biotransformation and bioactivation of estradiol. Drug Metab Rev 2023; 55:1-49. [PMID: 36823774 DOI: 10.1080/03602532.2023.2177671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Endogenous estradiol (E2) exerts diverse physiological and pharmacological activities, commonly used for hormone replacement therapy. However, prolonged and excessive exposure to E2 potentially increases estrogenic cancer risk. Reportedly, CYP1 enzyme-mediated biotransformation of E2 is largely concerned with its balance between detoxification and carcinogenic pathways. Among the three key CYP1 enzymes (CYP1A1, CYP1A2, and CYP1B1), CYP1A1 and CYP1A2 mainly catalyze the formation of nontoxic 2-hydroxyestradiol (2-OH-E2), while CYP1B1 specifically catalyzes the formation of genotoxic 4-hydroxyestradiol (4-OH-E2). 4-OH-E2 can be further metabolized to electrophilic quinone intermediates accompanied by the generation of reactive oxygen species (ROS), triggering DNA damage. Since abnormal alterations in CYP1 activities can greatly affect the bioactivation process of E2, regulatory effects of xenobiotics on CYP1s are essential for E2-associated cancer development. To date, thousands of natural and synthetic compounds have been found to show potential inhibition and/or induction actions on the three CYP1 members. Generally, these chemicals share similar planar polycyclic skeletons, the structural motifs and substituent groups of which are important for their inhibitory/inductive efficiency and selectivity toward CYP1 enzymes. This review comprehensively summarizes these known inhibitors and/or inductors of E2-metabolizing CYP1s based on chemical categories and discusses their structure-activity relationships, which would contribute to better understanding of the correlation between xenobiotic-regulated CYP1 activities and estrogenic cancer susceptibility.
Collapse
Affiliation(s)
- Xu Mao
- Department of Pharmaceutical Analysis, College of Pharmacy, Mudanjiang Medical University, Mudanjiang, China
| | - Hui Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
10
|
Cannarella R, Gül M, Rambhatla A, Agarwal A. Temporal decline of sperm concentration: role of endocrine disruptors. Endocrine 2023; 79:1-16. [PMID: 36194343 DOI: 10.1007/s12020-022-03136-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/03/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Male infertility is a widespread disease with an etiology that is not always clear. A number of studies have reported a decrease in sperm production in the last forty years. Although the reasons are still undefined, the change in environmental conditions and the higher exposure to endocrine-disrupting chemicals (EDCs), namely bisphenol A, phthalates, polychlorinated biphenyls, polybrominated diphenyl esters, dichlorodiphenyl-dichloroethylene, pesticides, and herbicides, organophosphates, and heavy metals, starting from prenatal life may represent a possible factor justifying the temporal decline in sperm count. AIM The aim of this study is to provide a comprehensive description of the effects of the exposure to EDCs on testicular development, spermatogenesis, the prevalence of malformations of the male genital tract (cryptorchidism, testicular dysgenesis, and hypospadias), testicular tumor, and the mechanisms of testicular EDC-mediated damage. NARRATIVE REVIEW Animal studies confirm the deleterious impact of EDCs on the male reproductive apparatus. EDCs can compromise male fertility by binding to hormone receptors, dysregulating the expression of receptors, disrupting steroidogenesis and hormonal metabolism, and altering the epigenetic mechanisms. In humans, exposure to EDCs has been associated with poor semen quality, increased sperm DNA fragmentation, increased gonadotropin levels, a slightly increased risk of structural abnormalities of the genital apparatus, such as cryptorchidism and hypospadias, and development of testicular tumor. Finally, maternal exposure to EDCs seems to predispose to the risk of developing testicular tumors. CONCLUSION EDCs negatively impact the testicular function, as suggested by evidence in both experimental animals and humans. A prenatal and postnatal increase to EDC exposure compared to the past may likely represent one of the factors leading to the temporal decline in sperm counts.
Collapse
Affiliation(s)
- Rossella Cannarella
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Murat Gül
- Department of Urology, Selcuk University School of Medicine, Konya, Turkey
| | | | - Ashok Agarwal
- American Center for Reproductive Medicine (Virtual Research), Global Andrology Forum, Cleveland, OH, USA.
| |
Collapse
|
11
|
Franssen D, Johansson HKL, Lopez-Rodriguez D, Lavergne A, Terwagne Q, Boberg J, Christiansen S, Svingen T, Parent AS. Perinatal exposure to the fungicide ketoconazole alters hypothalamic control of puberty in female rats. Front Endocrinol (Lausanne) 2023; 14:1140886. [PMID: 37077353 PMCID: PMC10108553 DOI: 10.3389/fendo.2023.1140886] [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: 01/09/2023] [Accepted: 03/03/2023] [Indexed: 04/05/2023] Open
Abstract
INTRODUCTION Estrogenic endocrine disrupting chemicals (EDCs) such as diethylstilbestrol (DES) are known to alter the timing of puberty onset and reproductive function in females. Accumulating evidence suggests that steroid synthesis inhibitors such as ketoconazole (KTZ) or phthalates may also affect female reproductive health, however their mode of action is poorly understood. Because hypothalamic activity is very sensitive to sex steroids, we aimed at determining whether and how EDCs with different mode of action can alter the hypothalamic transcriptome and GnRH release in female rats. DESIGN Female rats were exposed to KTZ or DES during perinatal (DES 3-6-12μg/kg.d; KTZ 3-6-12mg/kg.d), pubertal or adult periods (DES 3-12-48μg/kg.d; KTZ 3-12-48mg/kg.d). RESULTS Ex vivo study of GnRH pulsatility revealed that perinatal exposure to the highest doses of KTZ and DES delayed maturation of GnRH secretion before puberty, whereas pubertal or adult exposure had no effect on GnRH pulsatility. Hypothalamic transcriptome, studied by RNAsequencing in the preoptic area and in the mediobasal hypothalamus, was found to be very sensitive to perinatal exposure to all doses of KTZ before puberty with effects persisting until adulthood. Bioinformatic analysis with Ingenuity Pathway Analysis predicted "Creb signaling in Neurons" and "IGF-1 signaling" among the most downregulated pathways by all doses of KTZ and DES before puberty, and "PPARg" as a common upstream regulator driving gene expression changes. Deeper screening ofRNAseq datasets indicated that a high number of genes regulating the activity of the extrinsic GnRH pulse generator were consistently affected by all the doses of DES and KTZ before puberty. Several, including MKRN3, DNMT3 or Cbx7, showed similar alterations in expression at adulthood. CONCLUSION nRH secretion and the hypothalamic transcriptome are highly sensitive to perinatal exposure to both DES and KTZ. The identified pathways should be exploredfurther to identify biomarkers for future testing strategies for EDC identification and when enhancing the current standard information requirements in regulation.
Collapse
Affiliation(s)
- Delphine Franssen
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
- *Correspondence: Delphine Franssen,
| | | | | | - Arnaud Lavergne
- GIGA-Bioinformatics, GIGA Institute, Université de Liège, Liège, Belgium
| | - Quentin Terwagne
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Julie Boberg
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Sofie Christiansen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
- Department of Pediatrics, University Hospital Liege, Liege, Belgium
| |
Collapse
|
12
|
Jozkowiak M, Piotrowska-Kempisty H, Kobylarek D, Gorska N, Mozdziak P, Kempisty B, Rachon D, Spaczynski RZ. Endocrine Disrupting Chemicals in Polycystic Ovary Syndrome: The Relevant Role of the Theca and Granulosa Cells in the Pathogenesis of the Ovarian Dysfunction. Cells 2022; 12:cells12010174. [PMID: 36611967 PMCID: PMC9818374 DOI: 10.3390/cells12010174] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common heterogeneous endocrine disorder among women of reproductive age. The pathogenesis of PCOS remains elusive; however, there is evidence suggesting the potential contribution of genetic interactions or predispositions combined with environmental factors. Among these, endocrine disrupting chemicals (EDCs) have been proposed to potentially contribute to the etiology of PCOS. Granulosa and theca cells are known to cooperate to maintain ovarian function, and any disturbance can lead to endocrine disorders, such as PCOS. This article provides a review of the recent knowledge on PCOS pathophysiology, the role of granulosa and theca cells in PCOS pathogenesis, and the evidence linking exposure to EDCs with reproductive disorders such as PCOS.
Collapse
Affiliation(s)
- Malgorzata Jozkowiak
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland
- Correspondence: ; Tel.: +48-61847-0721
| | - Dominik Kobylarek
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland
| | - Natalia Gorska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Bartosz Kempisty
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Chalubinskiego 6a, 50-368 Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland
| | - Dominik Rachon
- Department of Clinical and Experimental Endocrinology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Robert Z. Spaczynski
- Center for Gynecology, Obstetrics and Infertility Treatment Pastelova, Pastelowa 8, 60-198 Poznan, Poland
| |
Collapse
|
13
|
Li Y, Yao Y, Xiao N, Liu Y, Du Y, Liu M, Zhang Q, Zhao H, Zhang T, Zhang H, Wang L, Luo H, Zhang Y, Sun H. The association of serum phthalate metabolites with biomarkers of ovarian reserve in women of childbearing age. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113909. [PMID: 35999756 DOI: 10.1016/j.ecoenv.2022.113909] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Phthalates (PAEs) are widely used plasticizers drawing increasing concern due to reproductive toxicity. However, studies on serum PAEs metabolites (mPAEs) and their associations with human ovarian function remain very scarce. In this study, from April 2019 to August 2020, a total of 297 women of childbearing age were recruited in Tianjin, China. Eleven mPAEs were analyzed in serum samples and eight mPAEs were detected at frequencies > 65% with median concentrations of 0.43-15.3 ng/mL. In multinomial logistic analysis, an increase in serum mono (2-isobutyl) phthalate (miBP) was associated with decline in antral follicle count (AFC) (OR=1.26, 95% CI: 0.99, 1.61) and 5-mono-(2-ethyl-5-hydroxyhexyl) phthalate (mEHHP) was significantly associated with AFC increase (OR=1.43, 95% CI: 1.06, 1.92), which were aligned with the associations found between mPAEs and AMH through generalized linear regression. In multiple linear regression models, per 10% increase in serum mono (2-ethylhexyl) phthalate (mEHP), mono (2-ethyl-5-oxohexyl) phthalate (mEOHP) (oxo-mEHP), and principal component 1 featured for high concentrations of mono-n-butyl phthalate (mBP), miBP and mEHP were associated with 0.15 (95% CI: -0.29, -0.02), 0.01 (95% CI: -0.01, 0.00) and 0.01 (95% CI: -0.02, 0.00) ln-unit decrease in estradiol (E2) levels, respectively, while mono-[(2-carboxymethyl) hexyl] phthalate (mCMHP) (carboxymethyl-mEHP) was positively associated with 0.05 ln-unit increase of E2 (95% CI: 0.02, 0.08). The observed negative associations between mPAEs and the Anti-Müllerian hormone (AMH) also aligned with the change in AFC. Generalized linear regression also revealed nonlinear associations between mono-ethyl phthalate (mEP), mCMHP and follicle-stimulating hormone (FSH). Overall, serum mEHP and its metabolites were negatively associated with E2. miBP was negatively associated with AFC. The nonlinear associations between mPAEs and FSH, and AMH need further study.
Collapse
Affiliation(s)
- Yongcheng Li
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Yiming Yao
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Nan Xiao
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics / Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Yarui Liu
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Yongrui Du
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics / Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Min Liu
- Tianjin Medical University, Tianjin 300070, China
| | - Qiuyue Zhang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Hongzhi Zhao
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Tao Zhang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Huajing Zhang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Lei Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China
| | - Haining Luo
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics / Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China.
| | - Yunshan Zhang
- Department of Center for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics / Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300100, China
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China.
| |
Collapse
|
14
|
Pogrmic-Majkic K, Samardzija Nenadov D, Tesic B, Fa Nedeljkovic S, Kokai D, Stanic B, Andric N. Mapping DEHP to the adverse outcome pathway network for human female reproductive toxicity. Arch Toxicol 2022; 96:2799-2813. [PMID: 35790550 PMCID: PMC9352620 DOI: 10.1007/s00204-022-03333-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/15/2022] [Indexed: 01/09/2023]
Abstract
Adverse outcome pathways (AOPs) and AOP networks are tools for mechanistic presentation of toxicological effects across different levels of biological organization. These tools are used to better understand how chemicals impact human health. In this study, a four-step workflow was used to derive the AOP network of human female reproductive toxicity (HFRT-AOP) from five AOPs available in the AOP-Wiki and ten AOPs obtained from the literature. Standard network analysis identified key events (KEs) that are point of convergence and divergence, upstream and downstream KEs, and bottlenecks across the network. To map di-(2-ethylhexyl) phthalate (DEHP) to the HFRT-AOP network, we extracted DEHP target genes and proteins from the Comparative Toxicogenomic and the CompTox Chemicals Dashboard databases. Enriched GO terms analysis was used to identify relevant biological processes in the ovary that are DEHP targets, whereas screening of scientific literature was performed manually and automatically using AOP-helpFinder. We combined this information to map DEHP to HFRT-AOP network to provide insight on the KEs and system-level perturbations caused by this endocrine disruptor and the emergent paths. This approach can enable better understanding of the toxic mechanism of DEHP-induced human female reproductive toxicity and reveal potential novel DEHP female reproductive targets for experimental studies.
Collapse
Affiliation(s)
- Kristina Pogrmic-Majkic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia.
| | - Dragana Samardzija Nenadov
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| | - Biljana Tesic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| | - Svetlana Fa Nedeljkovic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| | - Dunja Kokai
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| | - Bojana Stanic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| | - Nebojsa Andric
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 2, 21000, Novi Sad, Serbia
| |
Collapse
|
15
|
Tseng HH, Li CY, Wu ST, Su HH, Wong TH, Wu HE, Chang YW, Huang SK, Tsai EM, Suen JL. Di-(2-ethylhexyl) Phthalate Promotes Allergic Lung Inflammation by Modulating CD8α + Dendritic Cell Differentiation via Metabolite MEHP-PPARγ Axis. Front Immunol 2022; 13:581854. [PMID: 35663974 PMCID: PMC9160748 DOI: 10.3389/fimmu.2022.581854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a common plasticizer, is a ubiquitous environmental pollutant that can disrupt endocrine function. Epidemiological studies suggest that chronic exposure to DEHP in the environment is associated with the prevalence of childhood allergic diseases; however, the underlying causal relationship and immunological mechanism remain unclear. This study explored the immunomodulatory effect of DEHP on allergic lung inflammation, while particularly focusing on the impact of DEHP and its metabolite on dendritic cell differentiation and activity of peroxisome proliferator-activated receptor gamma (PPARγ). The results showed that exposure to DEHP at a human tolerable daily intake dose exacerbated allergic lung inflammation in mice. Ex vivo flow cytometric analysis revealed that DEHP-exposed mice displayed a significantly decreased number of CD8α+ dendritic cells (DCs) in spleens and DC progenitors in the bone marrow, as well as, less interleukin-12 production in splenic DCs and increased T helper 2 polarization. Pharmacological experiments showed that mono-(2-ethylhexyl) phthalate (MEHP), the main metabolite of DEHP, significantly hampered the differentiation of CD8α+ DCs from Fms-like tyrosine kinase 3 ligand-differentiated bone marrow culture, by modulating PPARγ activity. These results suggested that chronic exposure to DEHP at environmentally relevant levels, promotes allergic lung inflammation, at least in part, by altering DC differentiation through the MEHP-PPARγ axis. This study has crucial implications for the interaction(s) between environmental pollutants and innate immunity, with respect to the development of allergic asthma.
Collapse
Affiliation(s)
- Hsin-Han Tseng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shin-Ting Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiang-Han Su
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Hsuan Wong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Wei Chang
- Department of Laboratory, Taitung Hospital, Ministry of Health and Welfare, Taitung, Taiwan
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan.,Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eing Mei Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
16
|
Phthalate monoesters act through peroxisome proliferator-activated receptors in the mouse ovary. Reprod Toxicol 2022; 110:113-123. [PMID: 35421560 PMCID: PMC9749796 DOI: 10.1016/j.reprotox.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022]
Abstract
Widespread use of phthalates as solvents and plasticizers leads to everyday human exposure. The mechanisms by which phthalate metabolites act as ovarian toxicants are not fully understood. Thus, this study tested the hypothesis that the phthalate metabolites monononyl phthalate (MNP), monoisononyl phthalate (MiNP), mono(2-ethylhexyl) phthalate (MEHP), monobenzyl phthalate (MBzP), monobutyl phthalate (MBP), monoisobutyl phthalate (MiBP), and monoethyl phthalate (MEP) act through peroxisome proliferator-activated receptors (PPARs) in mouse granulosa cells. Primary granulosa cells were isolated from CD-1 mice and cultured with vehicle control (dimethyl sulfoxide) or MNP, MiNP, MEHP, MBzP, MBP, MiBP, or MEP (0.4-400 μM) for 24 h. Following culture, qPCR was performed for known PPAR targets, Fabp4 and Cd36. Treatment with the phthalate metabolites led to significant changes in Fabp4 and Cd36 expression relative to control in dose-dependent or nonmonotonic fashion. Primary granulosa cell cultures were also transfected with a DNA plasmid containing luciferase expressed under the control of a consensus PPAR response element. MNP, MiNP, MEHP, and MBzP caused dose-dependent changes in expression of luciferase, indicating the presence of functional endogenous PPAR receptors in the granulosa cells that respond to phthalate metabolites. The effects of phthalate metabolites on PPAR target genes were inhibited in most of the cultures by co-treatment with the PPAR-γ inhibitor, T0070907, or with the PPAR-α inhibitor, GW6471. Collectively, these data suggest that some phthalate metabolites may act through endogenous PPAR nuclear receptors in the ovary and that the differing structures of the phthalates result in different levels of activity.
Collapse
|
17
|
Basso CG, de Araujo-Ramos AT, Martino-Andrade AJ. Exposure to phthalates and female reproductive health: a literature review. Reprod Toxicol 2022; 109:61-79. [DOI: 10.1016/j.reprotox.2022.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022]
|
18
|
Prenatal exposure to the phthalate DEHP impacts reproduction-related gene expression in the pituitary. Reprod Toxicol 2022; 108:18-27. [PMID: 34954075 PMCID: PMC8882145 DOI: 10.1016/j.reprotox.2021.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/08/2021] [Accepted: 12/21/2021] [Indexed: 11/23/2022]
Abstract
Phthalates are chemicals used in products including plastics, personal care products, and building materials, leading to widespread contact. Previous studies on prenatal exposure to Di-(2-ethylhexyl) phthalate (DEHP) in mice and humans demonstrated pubertal timing and reproductive performance could be affected in exposed offspring. However, the impacts at the pituitary, specifically regarding signaling pathways engaged and direct effects on the gonadotropins LH and FSH, are unknown. We hypothesized prenatal exposure to DEHP during a critical period of embryonic development (e15.5 to e18.5) will cause sex-specific disruptions in reproduction-related mRNA expression in offspring's pituitary due to interference with androgen and aryl hydrocarbon receptor (AhR) signaling. We found that prenatal DEHP exposure in vivo caused a significant increase in Fshb specifically in males, while the anti-androgen flutamide caused significant increases in both Lhb and Fshb in males. AhR target gene Cyp1b1 was increased in both sexes in DEHP-exposed offspring. In embryonic pituitary cultures, the DEHP metabolite MEHP increased Cyp1a1 and Cyp1b1 mRNA in both sexes and Cyp1b1 induction was reduced by co-treatment with AhR antagonist. AhR reporter assay in GHFT1 cells confirmed MEHP can activate AhR signaling. Lhb, Fshb and Gnrhr mRNA were significantly decreased in both sexes by MEHP, but co-treatment with AhR antagonist did not restore mRNA levels in pituitary culture. In summary, our data suggest phthalates can directly affect the function of the pituitary by activating AhR signaling and altering gonadotropin expression. This indicates DEHP's impacts on the pituitary could contribute to reproductive dysfunctions observed in exposed mice and humans.
Collapse
|
19
|
Welch BM, Keil AP, Bommarito PA, van T' Erve TJ, Deterding LJ, Williams JG, Lih FB, Cantonwine DE, McElrath TF, Ferguson KK. Longitudinal exposure to consumer product chemicals and changes in plasma oxylipins in pregnant women. ENVIRONMENT INTERNATIONAL 2021; 157:106787. [PMID: 34314981 PMCID: PMC8490329 DOI: 10.1016/j.envint.2021.106787] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Exposure to consumer product chemicals during pregnancy may increase susceptibility to pregnancy disorders by influencing maternal inflammation. However, effects on specific inflammatory pathways have not been well characterized. Oxylipins are a diverse class of lipids that act as important mediators and biomarkers of several biological pathways that regulate inflammation. Adverse pregnancy outcomes have been associated with circulating oxylipin levels in pregnancy. In this study, we aimed to determine the longitudinal associations between plasma oxylipins and urinary biomarkers of three classes of consumer product chemicals among pregnant women. METHODS Data come from a study of 90 pregnant women nested within the LIFECODES cohort. Maternal plasma and urine were collected at three prenatal visits. Plasma was analyzed for 61 oxylipins, which were grouped according to biosynthetic pathways that we defined by upstream: 1) fatty acid precursor, including linoleic, arachidonic, docosahexaenoic, or eicosapentaenoic acid; and 2) enzyme pathway, including cyclooxygenase (COX), lipoxygenase (LOX), or cytochrome P450 (CYP). Urine was analyzed for 12 phenol, 12 phthalate, and 9 organophosphate ester (OPE) biomarkers. Linear mixed effect models were used for single-pollutant analyses. We implemented a novel extension of quantile g-computation for longitudinal data to examine the joint effect of class-specific chemical mixtures on individual plasma oxylipin concentrations. RESULTS We found that urinary biomarkers of consumer product chemicals were positively associated with pro-inflammatory oxylipins from several biosynthetic pathways. Importantly, these associations depended upon the chemical class of exposure biomarker. We estimated positive associations between urinary phenol biomarkers and oxylipins produced from arachidonic acid by LOX enzymes, including several important pro-inflammatory hydroxyeicosatetraenoic acids (HETEs). On average, mean concentrations of oxylipin produced from the arachidonic acid/LOX pathway were 48%-71% higher per quartile increase in the phenol biomarker mixture. For example, a simultaneous quartile increase in all urinary phenols was associated with 53% higher (95% confidence interval [CI]: 11%, 111%) concentrations of 12-HETE. The positive associations among phenols were primarily driven by methyl paraben, 2,5-dichlorophenol, and triclosan. Additionally, we observed that phthalate and OPE metabolites were associated with higher concentrations of oxylipins produced from linoleic acid by CYP enzymes, including the pro-inflammatory dihydroxy-octadecenoic acids (DiHOMEs). Associations among DiHOME oxylipins were driven by metabolites of benzylbutyl and di-isodecyl phthalate, and by the metabolite of tris(1,3-dichloro-2-propyl) phosphate among OPEs. We also observed inverse associations between phthalate and OPE metabolites and oxylipins produced from other pathways; however, adjusting for a plasma indicator of dietary fatty acid intake attenuated those results. CONCLUSIONS Our findings support the hypothesis that consumer product chemicals may have diverse impacts on inflammation processes in pregnancy. Certain pro-inflammatory oxylipins were generally higher among participants with higher urinary chemical biomarker concentrations. Associations varied by class of chemical and by the biosynthetic pathway of oxylipin production, indicating potential specificity in the inflammatory effects of these environmental chemicals during pregnancy that warrant investigation in larger studies.
Collapse
Affiliation(s)
- Barrett M Welch
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States
| | - Alexander P Keil
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States; Department of Epidemiology, University of North Carolina, United States
| | - Paige A Bommarito
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States
| | | | | | - Jason G Williams
- Mass Spectrometry Research and Support Group, NIEHS, United States
| | - Fred B Lih
- Mass Spectrometry Research and Support Group, NIEHS, United States
| | - David E Cantonwine
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Kelly K Ferguson
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States.
| |
Collapse
|
20
|
Bottalico LN, Korlyakova J, Weljie AM, Habibi HR. Seasonally Related Disruption of Metabolism by Environmental Contaminants in Male Goldfish ( Carassius auratus). FRONTIERS IN TOXICOLOGY 2021; 3:750870. [PMID: 35295106 PMCID: PMC8915895 DOI: 10.3389/ftox.2021.750870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/13/2021] [Indexed: 12/24/2022] Open
Abstract
Endocrine disrupting chemicals mimic or disrupt action of the natural hormones, adversely impacting hormonal function as well as cardiovascular, reproductive, and metabolic health. Goldfish are seasonal breeders with an annual reproductive cycle regulated by neuroendocrine signaling which involves allocation of metabolic energy to sustain growth and reproduction. We hypothesize that seasonal changes in physiology alter overall vulnerability of goldfish to metabolic perturbation induced by environmental contaminants. In this study, we assess effects of endogenous hormones, individual contaminants and their mixture on metabolism of goldfish at different reproductive stages. Exposure effects were assessed using 1H-NMR metabolomics profiling of male goldfish midbrain, gonad and liver harvested during early recrudescence (October), mid-recrudescence (February) and late recrudescence (June). Compounds assessed include bisphenol A, nonylphenol, bis(2-ethylhexyl) phthalate, fucosterol and a tertiary mixture (DEHP + NP + FS). Metabolome-level responses induced by contaminant exposure across tissues and seasons were benchmarked against responses induced by 17β-estradiol, testosterone and thyroid hormone (T3). We observe a clear seasonal dependence to metabolome-level alteration induced by hormone or contaminant exposures, with February (mid-recrudescence) the stage at which male goldfish are most vulnerable to metabolic perturbation. Responses induced by contaminant exposures differed from those induced by the natural hormones in a season-specific manner. Exposure to the tertiary mixture induced a functional gain at the level of biochemical pathways modeling over responses induced by individual components in select tissues and seasons. We demonstrate the importance of seasonally driven changes in physiology altering overall vulnerability of goldfish to metabolic perturbation induced by environmental contaminants, the relevance of which likely extends to other seasonally-breeding species.
Collapse
Affiliation(s)
- Lisa N. Bottalico
- Department of Systems Pharmacology and Translational Therapeutics, Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Julia Korlyakova
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hamid R Habibi
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada,*Correspondence: Hamid R Habibi,
| |
Collapse
|
21
|
Sharma P, Bilkhiwal N, Chaturvedi P, Kumar S, Khetarpal P. Potential environmental toxicant exposure, metabolizing gene variants and risk of PCOS-A systematic review. Reprod Toxicol 2021; 103:124-132. [PMID: 34126208 DOI: 10.1016/j.reprotox.2021.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/12/2021] [Accepted: 06/02/2021] [Indexed: 10/21/2022]
Abstract
Exposure of environmental toxicants such as potentially toxic metals and pesticides have largely been attributed to produce adverse effects on general women's health and to be more precise on the reproductive system. In order to explore exposure of toxicants and metabolizing gene variants as risk factor for polycystic ovarian syndrome (PCOS), literature search was carried out using the databases PubMed, Central Cochrane Library, Google Scholar, Science Direct with appropriate keywords upto 6 December 2020. While most of the studies indicate higher serum Cu concentration and lower concentration of Mn as risk factor, studies also report presence of higher pesticide concentration in PCOS women. Genes such as MTHFR, CYPs participate in the metabolism of toxicants and may show different response due to underlying genetic variants. Thus, toxicant exposure are to some extent responsible for the pathogenesis of syndrome through oxidative stress and endocrine disruption, but the susceptibility may vary due to the underlying genetic polymorphism of the exposed population.
Collapse
Affiliation(s)
- Priya Sharma
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Nisha Bilkhiwal
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Pragya Chaturvedi
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Sachin Kumar
- Department of Mathematics and Statistics, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India.
| | - Preeti Khetarpal
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
22
|
Chou Y, Tzeng C. The impact of phthalate on reproductive function in women with endometriosis. Reprod Med Biol 2021; 20:159-168. [PMID: 33850448 PMCID: PMC8022091 DOI: 10.1002/rmb2.12364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Endometriosis is a common gynecological condition in which stromal or glandular epithelium is implanted in extrauterine locations. Endometriosis causes detrimental effects on the granulosa cells, and phthalate interferes with the biological and reproductive function of endometrial cells at a molecular level. METHODS This article retrospectively reviewed the studies on phthalate exposure and its relationship with endometriosis. A literature search was performed for scientific articles using the keywords "phthalate and endometriosis," "endometriosis and granulosa cells," "phthalate and granulosa cells," and "phthalates and endometrial cells." RESULTS Endometriosis can affect cytokine production, steroidogenesis, cell cycle progression, expression of estrogen receptor-α (ER-α)/progesterone receptor (PR), and cause endoplasmic reticulum stress, senescence, apoptosis, autophagy, and oxidative stress in the granulosa cells. Mono-n-butyl phthalate (MnBP) alters the expression of cytokines, cell cycle-associated genes, ovarian stimulation, steroidogenesis, and progesterone production. Several in vitro studies have demonstrated that phthalate caused inflammation, invasion, change in cytokines, increased oxidative stress, viability, resistance to hydrogen peroxide, and proliferation of endometrial cells. CONCLUSION This might provide new insights about the impact of phthalate on the pathogenesis of endometriosis and its consequences on the ovarian function.
Collapse
Affiliation(s)
- Ya‐Ching Chou
- Department of Biological Science and TechnologyCollege of Biological Science and TechnologyNational Chiao Tung UniversityHsinchuTaiwan
- Center for Intelligent Drug Systems and Smart Bio‐devices (IDSB)National Chiao Tung UniversityHsinchuTaiwan
| | - Chii‐Ruey Tzeng
- Department of Obstetrics and GynecologyTaipei Medical UniversityTaipeiTaiwan
- Taipei Fertility Center, TaipeiTaiwan
| |
Collapse
|
23
|
Sedha S, Lee H, Singh S, Kumar S, Jain S, Ahmad A, Bin Jardan YA, Sonwal S, Shukla S, Simal-Gandara J, Xiao J, Huh YS, Han YK, Bajpai VK. Reproductive toxic potential of phthalate compounds - State of art review. Pharmacol Res 2021; 167:105536. [PMID: 33677105 DOI: 10.1016/j.phrs.2021.105536] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 02/05/2023]
Abstract
Phthalates are pervasive compounds, and due to the ubiquitous usage of phthalates, humans or even children are widely exposed to them. Since phthalates are not chemically bound to the plastic matrix, they can easily leach out to contaminate the peripheral environment. Various animal and human studies have raised vital health concern including developmental and reproductive toxicity of phthalate exposure. The present review is based upon the available literature on phthalates with respect to their reproductive toxic potential. Common reproductive effects such as declined fertility, reduced testis weight, variations in accessory sex organs and several female reproductive disorders appeared to be largely associated with the transitional phthalates. Among the higher molecular weight phthalates (≥ C7), di-isononyl phthalate (DINP) produces some minor effects on development of male reproductive tract and among low molecular weight phthalates (≤C3), di-methyl (DMP) and di-isobutyl (DIBP) phthalate produce some adverse effects on male reproductive system. Whereas transitional phthalates such as di-butyl phthalate, benzyl butyl phthalate, and di-(2-ethylhexyl) phthalate have shown adverse effects on female reproductive system. Owing to these, non-toxic alternatives to phthalates may be developed and use of phthalates could be rationalized as an important issue where human reproduction system is involved. Though, more epidemiological studies are needed to substantiate the reported findings on phthalates.
Collapse
Affiliation(s)
- Sapna Sedha
- Department of Biotechnology, Dr Hari Singh Gour Vishwavidyalaya, Sagar 470003, MP, India
| | - Hoomin Lee
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, 100 Inha-ro, Nam-gu, Incheon 22212, South Korea
| | - Siddhartha Singh
- Government Girls P.G. College for Excellence, Sagar 470002, MP, India
| | - Sunil Kumar
- National Institute of Occupational Health - ICMR, Meghaninagar, Ahmedabad 380016, Gujarat, India
| | - Subodh Jain
- Department of Biotechnology, Dr Hari Singh Gour Vishwavidyalaya, Sagar 470003, MP, India
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sonam Sonwal
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, 100 Inha-ro, Nam-gu, Incheon 22212, South Korea
| | - Shruti Shukla
- Department of Food Science and Technology, National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonipat, Haryana 131028, India
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense E-32004, Spain
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense E-32004, Spain.
| | - Yun Suk Huh
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, 100 Inha-ro, Nam-gu, Incheon 22212, South Korea.
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul 04620, South Korea.
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul 04620, South Korea.
| |
Collapse
|
24
|
Suriyakalaa U, Ramachandran R, Doulathunnisa JA, Aseervatham SB, Sankarganesh D, Kamalakkannan S, Kadalmani B, Angayarkanni J, Akbarsha MA, Achiraman S. Upregulation of Cyp19a1 and PPAR-γ in ovarian steroidogenic pathway by Ficus religiosa: A potential cure for polycystic ovary syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113540. [PMID: 33152430 DOI: 10.1016/j.jep.2020.113540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 08/03/2020] [Accepted: 10/29/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Quite a few plants are in use to treat female infertility and associated problems. Availing the cues from traditional knowledge, phytochemical studies and ethnopharmacological evidences, the aphrodisiac plant Ficus religiosa (F. religiosa) is widely in use to cure infertility in women. For instance, the juice of leaf and aerial root of F. religiosa is reported to normalize the dysregulated menstrual cycle in women. Besides, it is believed that regular circumambulation of F. religiosa during the early hours of the morning helps women in alleviating infertility which could be attributed to the potential phytovolatiles released from F. religiosa. However, the evidences for therapeutic potential of F. religiosa in treating female infertility are arbitrary and mostly anecdotal. AIM OF THE STUDY The present study was aimed at examining if extracts of fresh and/or dry leaf of F. religiosa would cure polycystic ovary syndrome (PCOS) in the rat model. METHODS Rats were divided into seven groups; control (Group I), PCOS-induced (P.O, Letrozole -1 mg/kg BW for 21 days) and untreated (Group II), PCOS-induced and treated with the leaf extracts of F. religiosa (Groups III-VI), and, PCOS-induced and treated with pioglitazone (Group VII). The estrous intervals, body and organ weights (ovary and uterus), and serum hormones (testosterone, luteinizing hormone [LH], estrogen, and progesterone) were measured, and the expression of Cyp19a1 (aromatase), and Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) were assessed in the experimental rats. The levels of 3β-hydroxysteroid dehydrogenase (3β-HSD), 17β-hydroxysteroid dehydrogenase (17β-HSD), and antioxidants (MDA, GSH, GPx, SOD, and CAT) were also quantified. Besides, the putative volatile compounds in the esterified leaf extracts were identified using Gas Chromatography-Mass Spectrometry (GC-MS). RESULTS Letrozole treatment induced irregular estrous and altered weight of organs and hormonal milieu, which were reverted to normal in leaf extracts-treated PCOS-induced rats. Remarkably, fresh leaf treatment up-regulated Cyp19a1and PPAR-γ and increased the levels of 3β-HSD and 17β-HSD. We found 3-acetoxy-3-hydroxy-propionic acid in fresh and dry leaf extracts, which we attribute to efficacy of the extracts in alleviating PCOS. CONCLUSION Put together, our findings suggest the leaves of F. religiosa as potential in alleviating PCOS, mainly due to the presence of putative volatile molecules. Further screening of the leaves of F. religiosa is recommended to identify other key molecules and to develop a systematic therapeutic intervention for PCOS.
Collapse
Affiliation(s)
- Udhayaraj Suriyakalaa
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, 620024, Tamilnadu, India; Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India; PG and Research Department of Biotechnology, Hindusthan College of Arts and Science, Coimbatore, 641028, Tamilnadu, India
| | - Rajamanickam Ramachandran
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, 620024, Tamilnadu, India; Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India
| | - Jaffar Ali Doulathunnisa
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Smilin Bell Aseervatham
- PG and Research Department of Biotechnology and Bioinformatics, Holy Cross College (Autonomous), Tiruchirappalli, 620002, Tamilnadu, India
| | - Devaraj Sankarganesh
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India; Department of Biotechnology, School of Bio- and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, 626126, Tamilnadu, India
| | | | - Balamuthu Kadalmani
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, 620024, Tamilnadu, India
| | - Jayaraman Angayarkanni
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India
| | | | - Shanmugam Achiraman
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, 620024, Tamilnadu, India.
| |
Collapse
|
25
|
Yang L, Yin W, Li P, Hu C, Hou J, Wang L, Yuan J, Yu Z. Seasonal exposure to phthalates and inflammatory parameters: A pilot study with repeated measures. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111633. [PMID: 33396153 DOI: 10.1016/j.ecoenv.2020.111633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 06/12/2023]
Abstract
Exposure to phthalates poses potential to damage multiple organs and system in the body. However, limited data are available regarding effects of seasonal exposure levels of phthalates and indicators reflecting inflammatory response. We designed a pilot study with repeated measures. We recruited 106 eligible habitants from Wuhan city, China. They completed questionnaires, physical examinations and provided urine specimens in winter and summer seasons. We found that urinary levels of low-molecular-weight phthalate metabolites were higher in summer than in winter (all P < 0.01). In winter, an interquartile range increase (1.264 μg/L) in 3-day moving average of high-molecular-weight phthalate metabolites corresponded to a 13.634% (95% CI: -22.331, -3.941) decrease in mean platelet volume, 25.879% (95% CI: -37.424, -12.204) for lymphocyte count or 10.862% (95% CI: -18.716, -2.125) for platelet count (P < 0.05 or P < 0.01). However, in summer, an interquartile range increase (1.215 μg/L) in urinary levels of high-molecular-weight phthalate metabolites corresponded to an 8.743% (95% CI: 4.217, 13.467) increase in platelet distribution width value or a 4.597% (95% CI: 2.335, 6.780) for mean platelet volume value at 3-day lag (both P < 0.01). In conclusion, phthalate exposure exhibited the potential for the activation of platelet function, particularly in winter. Seasonal variations of phthalate exposure should be considered when assessing health risk.
Collapse
Affiliation(s)
- Luoyao Yang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13 Wuhan 430030, PR China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Wenjun Yin
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13 Wuhan 430030, PR China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China; Wuhan Prevention and Treatment Center for Occupational Diseases, Wuhan 430015, Hubei, PR China
| | - Pei Li
- State Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China
| | - Chen Hu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13 Wuhan 430030, PR China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Jian Hou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13 Wuhan 430030, PR China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Lin Wang
- Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China
| | - Jing Yuan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13 Wuhan 430030, PR China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, State Environmental Protection Key Laboratory of Environment and Health (Wuhan) and State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, Hubei, PR China.
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, PR China.
| |
Collapse
|
26
|
Akın L, Kendirci M, Narin F, Kurtoglu S, Hatipoglu N, Elmalı F. Endocrine Disruptors and Polycystic Ovary Syndrome: Phthalates. J Clin Res Pediatr Endocrinol 2020; 12:393-400. [PMID: 32431137 PMCID: PMC7711640 DOI: 10.4274/jcrpe.galenos.2020.2020.0037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE We aimed to investigate a possible role of the endocrine disruptors phthalates, di-2-ethylhexyl phthalate (DEHP) and mono (2-ethylhexyl) phthalate (MEHP), in polycystic ovary syndrome (PCOS) aetiopathogenesis. We also wished to evaluate the relationship between phthalates and metabolic disturbances in adolescents with PCOS. METHODS A total of 124 adolescents were included. Serum MEHP and DEHP levels were determined by high-performance liquid chromatography. Insulin resistance was evaluated using homeostasis model assessment-insulin resistance, quantitative Insulin Sensitivity Check Index, fasting glucose/insulin ratio, Matsuda index, and total insulin levels during oral glucose tolerance test. Participants were further subdivided into lean and obese subgroups according to body mass index (BMI). RESULTS Sixty-three PCOS and 61 controls, (mean age 15.2±1.5; range: 13-19 years) were enrolled. Serum DEHP and MEHP concentrations were not significantly different between PCOS and control groups. The mean (95% confidence interval) values of DEHP and MEHP were 2.62 (2.50-2.75) μg/mL vs 2.71 (2.52-2.90) μg/mL and 0.23 (0.19-0.29) μg/mL vs 0.36 (0.18-0.54) μg/mL in PCOS and the control groups respectively, p>0.05. Correlation analysis, adjusted for BMI, showed that both phthalates significantly correlated with insulin resistance indices and serum triglycerides in adolescents with PCOS. CONCLUSION Serum DEHP and MEHP concentrations were not different between adolescents with or without PCOS. However, these phthalates are associated with metabolic disturbances such as dyslipidemia and insulin resistance, independently of obesity, in girls with PCOS.
Collapse
Affiliation(s)
- Leyla Akın
- Erciyes University Faculty of Medicine, Department of Pediatric Endocrinology, Kayseri, Turkey,Ondokuz Mayıs University Faculty of Medicine, Department of Pediatric Endocrinology, Samsun, Turkey,* Address for Correspondence: Ondokuz Mayıs University Faculty of Medicine, Department of Pediatric Endocrinology, Samsun, Turkey Phone: +90 533 240 16 43 E-mail:
| | - Mustafa Kendirci
- Erciyes University Faculty of Medicine, Department of Pediatric Endocrinology, Kayseri, Turkey
| | - Figen Narin
- Erciyes University Faculty of Medicine, Department of Biochemistry, Kayseri; İzmir Kâtip Çelebi University, Department of Medical Biochemistry, İzmir, Turkey
| | - Selim Kurtoglu
- Erciyes University Faculty of Medicine, Department of Pediatric Endocrinology, Kayseri, Turkey
| | - Nihal Hatipoglu
- Erciyes University Faculty of Medicine, Department of Pediatric Endocrinology, Kayseri, Turkey
| | - Ferhan Elmalı
- Erciyes University Faculty of Medicine, Department of Biochemistry, Kayseri; İzmir Kâtip Çelebi University, Department of Medical Biochemistry, İzmir, Turkey
| |
Collapse
|
27
|
Shan A, Leng L, Li J, Luo XM, Fan YJ, Yang Q, Xie QH, Chen YS, Ni CS, Guo LM, Tang H, Chen X, Tang NJ. TCDD-induced antagonism of MEHP-mediated migration and invasion partly involves aryl hydrocarbon receptor in MCF7 breast cancer cells. JOURNAL OF HAZARDOUS MATERIALS 2020; 398:122869. [PMID: 33027880 DOI: 10.1016/j.jhazmat.2020.122869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 06/11/2023]
Abstract
Evidence has shown that the activation of AhR (aryl hydrocarbon receptor) can promote cancer cell metastasis. However, limited studies have been carried out on mixed exposure to endocrine-disrupting chemicals (EDCs), especially in human breast cancer. Therefore, using MCF7 human breast cancer cells, we investigated the effects of coexposure to MEHP (mono 2-ethylhexyl phthalate) and TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) on cell migration and invasion, as well as the roles of AhR and the MMP/slug pathway. Our data suggest that MEHP or TCDD can induce migration and invasion in MCF7 cells, and the promotion is partly AhR dependent. We also observed that MEHP antagonized TCDD to reduce AhR-mediated CYP1A1 expression. Subsequently, we revealed that MEHP recruited AhR to dioxin response element (DRE) sequences and decreased TCDD-induced AhR-DRE binding in CYP1A1 genes. Overall, MEHP is a potential AHR agonist, capable of decreasing TCDD-induced AhR-DRE binding in CYP1A1 genes. The antagonizing effect of coexposure led to the inhibition of the epithelial-mesenchymal transition (EMT) in MCF7 cells. Our study provides new evidence for the potential mechanisms involved in EDCs exposure and their interactions in EMT.
Collapse
Affiliation(s)
- Anqi Shan
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Ling Leng
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Department of Cell Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China.
| | - Jing Li
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Xiu-Mei Luo
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Ya-Jiao Fan
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Qiaoyun Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Qun-Hui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Yang-Sheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Chun-Sheng Ni
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China.
| | - Li-Ming Guo
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Hua Tang
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Xi Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Nai-Jun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
28
|
Tao Y, Yang Y, Jiao Y, Wu S, Zhu G, Akindolie MS, Zhu T, Qu J, Wang L, Zhang Y. Monobutyl phthalate (MBP) induces energy metabolism disturbances in the gills of adult zebrafish (Danio rerio). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115288. [PMID: 32795888 DOI: 10.1016/j.envpol.2020.115288] [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: 03/29/2020] [Revised: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
Monobutyl phthalate (MBP) is a primary metabolite of an environmental endocrine disruptor dibutyl phthalate (DBP), which poses a potential threat to living organisms. In this research, the acute toxicity of MBP on energy metabolism in zebrafish gills was studied. Transmission electron microscopy (TEM) results show that 10 mg L-1 MBP can induce mitochondrial structural damage of chloride cells after 96 h of continuous exposure. The activity of ion ATPase and the expression level of oxidative phosphorylation-related genes suggest that MBP interferes with ATP synthesis and ion transport. Further leading to a decrease in mitochondrial membrane potential (MMP) and cell viability, thereby mediating early-stage cell apoptosis. Through a comprehensive analysis of principal component analysis (PCA) and integrated biomarker response (IBR) scores, atp5a1, a subunit of mitochondrial ATP synthase, is mainly inhibited by MBP, followed by genes encoding ion ATPase (atp1b2 and atp2b1). Importantly, MBP inhibits aerobic metabolism by inhibiting the key enzyme malate dehydrogenase (MDH) in the TCA cycle, forcing zebrafish to maintain ATP supply by enhancing anaerobic metabolism.
Collapse
Affiliation(s)
- Yue Tao
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yang Yang
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yaqi Jiao
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Song Wu
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Guangxue Zhu
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Modupe Sarah Akindolie
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Tong Zhu
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jianhua Qu
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Lei Wang
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China
| | - Ying Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
29
|
Li XN, Li HX, Yang TN, Li XW, Huang YQ, Zhu SY, Li JL. Di-(2-ethylhexyl) phthalate induced developmental abnormalities of the ovary in quail (Coturnix japonica) via disruption of the hypothalamic-pituitary-ovarian axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 741:140293. [PMID: 32610232 DOI: 10.1016/j.scitotenv.2020.140293] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 06/11/2023]
Abstract
An increasing number of epidemiologic studies show that women have a special exposure profile to phthalates, and the exposures have attracted attention regarding their potential health hazards. Here, we developed a model for studying the ovarian action of di-(2-ethylhexyl) phthalate (DEHP) and its major metabolite monoethylhexyl phthalate (MEHP). In vivo, treatment with DEHP (250, 500, and 1000 mg kg^-1) induced decreased thickness of ovarian granulosa cell layer and mitochondrial damage in quail, caused oxidative stress, interfered with the transcription of hypothalamic-pituitary-ovarian axis (HPOA) steroid hormone-related factors (increased transcription of StAR, 3β-HSD, P450scc, and LH and decreased transcription of 17β-HSD, P450arom, FSH, and ERβ), and blocked the secretion of steroid hormones (decreased FSH, E2, and T levels and increased LH, P, and PRL levels). In vitro, granulosa cells were cultured with MEHP (50, 100, and 200 μM), activator of PPARγ (rosiglitazone, 50 μM), or antagonist of PPARγ (GW9662, 10 μM) for 24 h and gene and protein expression were analyzed by real time RT-PCR and western blot. Rosiglitazone, like MEHP, significantly decreased mRNA and protein levels of P450arom. Antagonist GW9662 partially blocked the suppression of P450arom by MEHP, suggesting that MEHP acts through PPARγ, but not exclusively. Our model shows that MEHP acts on granulosa cells in quail by stimulating PPARs, which leads to decreased gene and protein expression of P450arom. Therefore, the environmental endocrine disruptor DEHP and its major metabolite MEHP act through a receptor-mediated signaling pathway to inhibit the production of estradiol, interfere with the modulation of HPOA, suppress the synthesis of sex hormones, and cause sex hormone secretion disorders, resulting in severe toxicity in the female reproductive system. A framework for an adverse outcome pathway of DEHP/MEHP-induced ovarian toxicity was constructed, which can facilitate an improved understanding of the mechanism of female reproductive toxicity.
Collapse
Affiliation(s)
- Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hui-Xin Li
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150086, PR China
| | - Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiao-Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue-Qiang Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
30
|
Park CG, Sung B, Ryu CS, Kim YJ. Mono-(2-ethylhexyl) phthalate induces oxidative stress and lipid accumulation in zebrafish liver cells. Comp Biochem Physiol C Toxicol Pharmacol 2020; 230:108704. [PMID: 31927120 DOI: 10.1016/j.cbpc.2020.108704] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/02/2020] [Accepted: 01/04/2020] [Indexed: 12/18/2022]
Abstract
The purpose of the present study was to examine the antioxidant and oxidative stress changes in zebrafish liver (ZFL) cells in the presence of mono-(2-ethylhexyl) phthalate (MEHP). When reactive oxygen species (ROS) and antioxidant levels were measured by immunoassay, significant differences were observed between MEHP-treated and control cells, while catalase levels did not change in any group. MEHP-treated cells had higher levels of ROS, glutathione peroxidase (GPx), glutathione S-transferase (GST), glutathione, and superoxide dismutase (SOD) than control cells. However, lower levels of lipid peroxidation were observed in MEHP-treated cells compared to control cells. After 24 h of MEHP treatment, ROS, SOD, GPx, and GST activity increased in a dose-dependent manner. Cellular lipid droplet formation and endoplasmic reticulum stress were both induced in the presence of MEHP. These findings demonstrated the potential impacts of the association of MEHP with adverse outcomes in fish liver. Future studies will focus on clarifying the molecular mechanism of phthalate toxicity via oxidative stress and peroxisome proliferator activated receptor as the major mechanistic pathway.
Collapse
Affiliation(s)
- Chang Gyun Park
- Environmental Safety Group, JRC-APT, KIST Europe Forschungsgesellschaft mbH, 66123 Saarbrücken, Germany
| | - Baeckkyoung Sung
- Environmental Safety Group, JRC-APT, KIST Europe Forschungsgesellschaft mbH, 66123 Saarbrücken, Germany; Division of Energy & Environment Technology, University of Science and Technology, 34113 Daejeon, Korea
| | - Chang Seon Ryu
- Environmental Safety Group, JRC-APT, KIST Europe Forschungsgesellschaft mbH, 66123 Saarbrücken, Germany.
| | - Young Jun Kim
- Environmental Safety Group, JRC-APT, KIST Europe Forschungsgesellschaft mbH, 66123 Saarbrücken, Germany; Division of Energy & Environment Technology, University of Science and Technology, 34113 Daejeon, Korea.
| |
Collapse
|
31
|
Xi Y, Zhang Y, Zhu S, Luo Y, Xu P, Huang Z. PPAR-Mediated Toxicology and Applied Pharmacology. Cells 2020; 9:cells9020352. [PMID: 32028670 PMCID: PMC7072218 DOI: 10.3390/cells9020352] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 01/26/2020] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor family, attract wide attention as promising therapeutic targets for the treatment of multiple diseases, and their target selective ligands were also intensively developed for pharmacological agents such as the approved drugs fibrates and thiazolidinediones (TZDs). Despite their potent pharmacological activities, PPARs are reported to be involved in agent- and pollutant-induced multiple organ toxicity or protective effects against toxicity. A better understanding of the protective and the detrimental role of PPARs will help to preserve efficacy of the PPAR modulators but diminish adverse effects. The present review summarizes and critiques current findings related to PPAR-mediated types of toxicity and protective effects against toxicity for a systematic understanding of PPARs in toxicology and applied pharmacology.
Collapse
Affiliation(s)
- Yue Xi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yunhui Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Sirui Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuping Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence: (P.X.); (Z.H.); Tel.: +1-412-708-4694(P.X.); +86-20-39943092 (Z.H.)
| | - Zhiying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Correspondence: (P.X.); (Z.H.); Tel.: +1-412-708-4694(P.X.); +86-20-39943092 (Z.H.)
| |
Collapse
|
32
|
Tao T, Wang Y, Xu B, Mao X, Sun Y, Liu W. Role of adiponectin/peroxisome proliferator-activated receptor alpha signaling in human chorionic gonadotropin-induced estradiol synthesis in human luteinized granulosa cells. Mol Cell Endocrinol 2019; 493:110450. [PMID: 31116958 DOI: 10.1016/j.mce.2019.110450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/11/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022]
Abstract
Impaired steroid production in polycystic ovary syndrome (PCOS) may result from adiponectin system dysfunction. However, adiponectin's role in ovulatory dysfunction remains unclear. We aimed to determine whether human chorionic gonadotropin (hCG) and adiponectin affect progesterone and estradiol secretion by granulosa cells (GCs) from overweight or obese women with PCOS or normal ovulation. ADIPOR2 expression was higher in hCG-treated GCs from PCOS patients than in those from normovulatory women. hCG may upregulate ADIPOR2 expression through cAMP/PKA signaling in GCs. GCs from both groups expressed PPARA. Estradiol levels were lower in hCG + adiponectin-treated GCs from PCOS patients than in those from normovulatory women. hCG + adiponectin decreased P450 aromatase expression through adiponectin/PPARα signaling in GCs. Adiponectin downregulates hCG-induced estradiol levels in GCs from overweight or obese women through gonadotropin-adiponectin crosstalk. Changes in gonadotropin and adiponectin signaling in the ovarian microenvironment may improve symptoms in women with PCOS.
Collapse
Affiliation(s)
- Tao Tao
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Yuying Wang
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Bing Xu
- Shanghai Key laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Xiuying Mao
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Yun Sun
- Shanghai Key laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Wei Liu
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China; Shanghai Key laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
33
|
Fetal programming by androgen excess in rats affects ovarian fuel sensors and steroidogenesis. J Dev Orig Health Dis 2019; 10:645-658. [PMID: 31122307 DOI: 10.1017/s2040174419000126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fetal programming by androgen excess is hypothesized as one of the main factors contributing to the development of polycystic ovary syndrome (PCOS). PCOS is more than a reproductive disorder, as women with PCOS also show metabolic and other endocrine alterations. Since both ovarian and reproductive functions depend on energy balance, the alterations in metabolism may be related to reproductive alterations. The present study aimed to evaluate the effect of androgen excess during prenatal life on ovarian fuel sensors and its consequences on steroidogenesis. To this end, pregnant rats were hyperandrogenized with testosterone and the following parameters were evaluated in their female offspring: follicular development, PPARG levels, adipokines (including leptin, adiponectin, and chemerin as ovarian fuel sensors), serum gonadotropins (LH and FSH), the mRNA of their ovarian receptors, and the expression of steroidogenic mediators. At 60 days of age, the prenatally hyperandrogenized (PH) female offspring displayed both an irregular ovulatory phenotype and an anovulatory phenotype with altered follicular development and the presence of cysts. Both PH groups showed altered levels of both proteins and mRNA of PPARG and a different expression pattern of the adipokines studied. Although serum gonadotropins were not impaired, there were alterations in the mRNA levels of their ovarian receptors. The steroidogenic mediators Star, Cyp11a1, Cyp17a1, and Cyp19a1 were altered differently in each of the PH groups. We concluded that androgen excess during prenatal life leads to developmental programming effects that affect ovarian fuel sensors and steroidogenesis in a phenotype-specific way.
Collapse
|
34
|
Wen R, Gan X, Hu S, Gao S, Deng Y, Qiu J, Sun W, Li L, Han C, Hu J, Wang J. Evidence for the existence of de novo lipogenesis in goose granulosa cells. Poult Sci 2019; 98:1023-1030. [PMID: 30376078 DOI: 10.3382/ps/pey400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 10/10/2018] [Indexed: 12/21/2022] Open
Abstract
De novo lipogenesis (DNL) is an important physiological mechanism, but it is poorly understood in avian follicles. The protein distribution patterns of three key genes related to DNL (i.e., FAS, ACC, and PPARγ) were firstly determined in geese developing follicles using immunohistochemistry, and our results showed that all three proteins were present in both prehierarchical and hierarchical follicles. Furthermore, it was revealed by qPCR that transcripts of these three genes were widely expressed in theca and granulosa layers of all staged follicles; however, the expression of DNL-related genes in granulosa cell changed significantly (P < 0.05) after follicle selection (FAS and PPARγ) and before ovulation (FAS). It is suggested that the DNL mechanism may be closely related to the follicular selection, while FAS may be closely associated with ovulation and steroidogenesis. These results suggested that DNL exists throughout follicle development and it potentially have an important role in the process of follicular selection, development, steroidogenesis, and ovulation, especially in their granulosa layers. To further demonstrate this point, granulosa cells isolated from hierarchical follicles were cultured in vitro. By analyzing the mRNA and protein expression patterns of these three genes, the fatty acid synthase enzyme activity, the contents of extracellular triglyceride, and intracellular lipids, as well as the cell activity at different time points of in vitro culture (0, 6, 12, and 18 h). These findings not only ensured the existence of DNL in the granulosa cells of goose follicles, but also suggested the complex process of lipid metabolism that associated with DNL, may play an important role in cell proliferation and physiological functions. Taken together, we first confirmed the existence of lipid metabolism, especially the DNL in goose follicles, and further suggested its role in the follicles, especially in the granulosa cells.
Collapse
Affiliation(s)
| | | | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Shanyan Gao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiamin Qiu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenqiang Sun
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
35
|
Xu D, He H, Jiang X, Hua R, Chen H, Yang L, Cheng J, Duan J, Li Q. SIRT2 plays a novel role on progesterone, estradiol and testosterone synthesis via PPARs/LXRα pathways in bovine ovarian granular cells. J Steroid Biochem Mol Biol 2019; 185:27-38. [PMID: 30009951 DOI: 10.1016/j.jsbmb.2018.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/27/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022]
Abstract
SIRT2 has been shown to possess NAD+-dependent deacetylase and desuccinylase enzymatic activities, it also regulates metabolism homeostasis in mammals. Previous data has suggested that resveratrol, a potential activator of Sirtuins, played a stimulation role in steroidogenesis. Unfortunately, to date, the physiological roles of SIRT2 in ovarian granular cells (GCs) are largely unknown. Here, we studied the function and molecular mechanisms of SIRT2 on steroid hormone synthesis in GCs from Qinchuan cattle. Immunohistochemistry and western blotting showed that SIRT2 was expressed not only in GCs and cumulus cells, but also in oocytes and theca cells. We found that the secretion of progesterone was induced, whereas that of estrogen and testosterone secretion was suppressed by treatment with the SIRT2 inhibitor (Thiomyristoyl or SirReal2) or siRNA. Additionally, the PPARs/LXRα signaling pathways were suppressed by SIRT2 siRNA or inhibitors. The mRNA expression of CYP17, aromatase and StAR was suppressed, but the abundance of CYP11A1 mRNA was induced by SIRT2 inhibition. Furthermore, the PPARα agonist or PPARγ antagonist could mimic the effects of SIRT2 inhibition on hormones levels and gene expression associated with steroid hormone biosynthesis. In turn, those effects were abolished by the LXRα agonist (LXR-623). Together, these data support the hypothesis that SIRT2 regulates steroid hormone synthesis via the PPARs/LXRα pathways in GCs.
Collapse
Affiliation(s)
- Dejun Xu
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China. -
| | - Huanshan He
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Xiaohan Jiang
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Rongmao Hua
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Huali Chen
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Li Yang
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Jianyong Cheng
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Jiaxin Duan
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| | - Qingwang Li
- Northwest A&F University, College of Animal Science and Technology, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
36
|
Junaid M, Jia PP, Tang YM, Xiong WX, Huang HY, Strauss PR, Li WG, Pei DS. Mechanistic toxicity of DEHP at environmentally relevant concentrations (ERCs) and ecological risk assessment in the Three Gorges Reservoir Area, China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:1939-1949. [PMID: 30055792 DOI: 10.1016/j.envpol.2018.07.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 07/15/2018] [Accepted: 07/15/2018] [Indexed: 06/08/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) associated in vitro/vivo toxicity at current environmentally relevant concentration (ERC) with attendant ecological risks in the Three Gorges Reservoir Area (TGRA) is still elusive. Responding to this challenge, a novel integrated study based on analytical and biological assays was designed to elucidate the underlying mechanisms for toxicity of DEHP and its ecological risks at ERC. In this study, GC-MS analysis showed that the highest environmental concentration of DEHP in the TGRA surface water was nearly double that of WHO and USEPA standards. Both distribution and ecological risk decreased from the upper to middle and lower reaches of the TGRA. In vitro toxicity was assessed by cell viability and DNA damage assays: DEHP exposure at ERCs (100-800 μg/L) caused significant reduction in cell viability and elevated DNA damage. Further, DEHP exposure above 400 μg/L resulted in enhanced migration behavior of cancer cells. For in vivo toxicity assessment, short term acute exposure (7 d, 400 μg/L) apparently activated the PI3K-AKT-mTOR pathway, and chronic low-level exposure (3 months, 10-33 μg/L) suppressed the hypothalamus pituitary thyroid (HPT) axis pathway in zebrafish. In addition, acute low-level exposure (5 d, 33-400 μg/L) to DEHP increased aryl hydrocarbon receptor (AhR) activity in Tg(cyp1a:gfp) zebrafish in a concentration-dependent manner. In short, DEHP at ERC has extended potential to induce diverse in vitro and in vivo toxicity at concentrations that also cause impairment of biochemical function in aquatic species of the TGRA.
Collapse
Affiliation(s)
- Muhammad Junaid
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pan-Pan Jia
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Mei Tang
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Xu Xiong
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Hai-Yang Huang
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Phyllis R Strauss
- Department of Biology, College of Science, Northeastern University, Boston, MA 02115, USA
| | - Wei-Guo Li
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - De-Sheng Pei
- Key Laboratory of Reservoir Aquatic Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; College of Life Science, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
37
|
Zarezadeh R, Mehdizadeh A, Leroy JLMR, Nouri M, Fayezi S, Darabi M. Action mechanisms of n-3 polyunsaturated fatty acids on the oocyte maturation and developmental competence: Potential advantages and disadvantages. J Cell Physiol 2018; 234:1016-1029. [PMID: 30073662 DOI: 10.1002/jcp.27101] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Infertility is a growing problem worldwide. Currently, in vitro fertilization (IVF) is widely performed to treat infertility. However, a high percentage of IVF cycles fails, due to the poor developmental potential of the retrieved oocyte to generate viable embryos. Fatty acid content of the follicular microenvironment can affect oocyte maturation and the subsequent developmental competence. Saturated and monounsaturated fatty acids are mainly used by follicle components as primary energy sources whereas polyunsaturated fatty acids (PUFAs) play a wide range of roles. A large body of evidence supports the beneficial effects of n-3 PUFAs in prevention, treatment, and amelioration of some pathophysiological conditions including heart diseases, cancer, diabetes, and psychological disorders. Nevertheless, current findings regarding the effects of n-3 PUFAs on reproductive outcomes in general and on oocyte quality more specifically are inconsistent. This review attempts to provide a comprehensive overview of potential molecular mechanisms by which n-3 PUFAs affect oocyte maturation and developmental competence, particularly in the setting of IVF and thereby aims to elucidate the reasons behind current discrepancies around this topic.
Collapse
Affiliation(s)
- Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jo L M R Leroy
- Department of Veterinary Sciences, Gamete Research Center, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Belgium
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Infertility and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Université de Nice Sophia Antipolis, Inserm U1091 - CNRS U7277, Nice 06034, France
| | - Masoud Darabi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
Rehman S, Usman Z, Rehman S, AlDraihem M, Rehman N, Rehman I, Ahmad G. Endocrine disrupting chemicals and impact on male reproductive health. Transl Androl Urol 2018; 7:490-503. [PMID: 30050807 PMCID: PMC6043754 DOI: 10.21037/tau.2018.05.17] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) have been known to adversely affect the endocrine system leading to compromised functions of hormones. The presence of these compounds in everyday products such as canned food, water bottles, plastics, cosmetics, fertilizers, kid’s toys and many others goods is a greater concern for general population. The persistent and long-term use of EDCs has deleterious effects on human reproductive health by interfering with the synthesis and mechanism of action of sex hormones. Any change during the synthesis or action of the sex hormones may result in abnormal reproductive functions which includes developmental anomalies in the reproductive tract and decline in semen quality. The present paper provides an overview of the EDCs and their possible impact on male reproductive health with major focus on semen quality which leads to male infertility.
Collapse
Affiliation(s)
- Saba Rehman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Zeenat Usman
- Department of Physiology, University of Health Sciences, Lahore, Pakistan
| | - Sabeen Rehman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - Noor Rehman
- Department of Biological Sciences, Binghamton University, NY, USA
| | - Ibraheem Rehman
- Department of Biological Sciences, Cornell University, Ithaca, NY, USA
| | - Gulfam Ahmad
- Department of Physiology, University of Health Sciences, Lahore, Pakistan.,Human Reproduction Unit, Kolling Institute, Sydney Medical School, Sydney University, Sydney, Australia
| |
Collapse
|
39
|
Chaparro-Ortega A, Betancourt M, Rosas P, Vázquez-Cuevas FG, Chavira R, Bonilla E, Casas E, Ducolomb Y. Endocrine disruptor effect of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) on porcine ovarian cell steroidogenesis. Toxicol In Vitro 2018; 46:86-93. [DOI: 10.1016/j.tiv.2017.09.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 11/17/2022]
|
40
|
Rattan S, Brehm E, Gao L, Niermann S, Flaws JA. Prenatal exposure to di(2-ethylhexyl) phthalate disrupts ovarian function in a transgenerational manner in female mice. Biol Reprod 2018; 98:130-145. [PMID: 29165555 PMCID: PMC5803793 DOI: 10.1093/biolre/iox154] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/26/2017] [Accepted: 11/17/2017] [Indexed: 01/04/2023] Open
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a plasticizer found in polyvinyl chloride products such as vinyl flooring, plastic food containers, medical devices, and children's toys. DEHP is a ubiquitous environmental contaminant and is a known endocrine disrupting chemical. Little is known about the effects of prenatal DEHP exposure on the ovary and whether effects occur in subsequent generations. Thus, we tested the hypothesis that prenatal exposure to DEHP disrupts ovarian functions in the F1, F2, and F3 generations of female mice. To test this hypothesis, pregnant CD-1 mice were orally dosed with corn oil (vehicle control) or DEHP (20 and 200 μg/kg/day and 200, 500, and 750 mg/kg/day) daily from gestation day 10.5 until birth (7-28 dams/treatment group). F1 females were mated with untreated males to obtain the F2 generation, and F2 females were mated with untreated males to produce the F3 generation. On postnatal days 1, 8, 21, and 60, ovaries were collected and used for histological evaluation of follicle numbers and sera were used to measure progesterone, testosterone, 17β-estradiol, luteinizing hormone, and follicle stimulating hormone levels. In the F1 generation, prenatal exposure to DEHP disrupted body and organ weights, decreased folliculogenesis, and increased serum 17β-estradiol levels. In the F2 generation, exposure to DEHP decreased body and organ weights, dysregulated folliculogenesis, and disrupted serum progesterone levels. In the F3 generation, DEHP exposure accelerated folliculogenesis. These data suggest that prenatal exposure to DEHP leads to adverse multigenerational and transgenerational effects on ovarian function.
Collapse
Affiliation(s)
| | | | | | - Sarah Niermann
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Illinois, USA
| |
Collapse
|
41
|
Cocci P, Capriotti M, Mosconi G, Palermo FA. Effects of endocrine disrupting chemicals on estrogen receptor alpha and heat shock protein 60 gene expression in primary cultures of loggerhead sea turtle (Caretta caretta) erythrocytes. ENVIRONMENTAL RESEARCH 2017; 158:616-624. [PMID: 28719870 DOI: 10.1016/j.envres.2017.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/16/2017] [Accepted: 07/10/2017] [Indexed: 06/07/2023]
Abstract
The loggerhead turtle (Caretta caretta) can be considered a good indicator species for studying the ecological impact of endocrine disrupting chemicals (EDCs) on wildlife. However, the effect of these environmental pollutants on nuclear steroid hormone signaling has not yet been addressed in sea turtles mainly due to the legal constraints of their endangered status. Here we describe the use of primary erythrocyte cell cultures as in vitro models for evaluating the effects of different EDCs on the expression of estrogen receptor α (ERα). In addition, we evaluated erythrocyte toxicity caused by EDCs using Alamar Blue assay and heat shock proteins 60 (HSP60) expression. Primary cultures of erythrocytes were exposed to increasing concentrations of 4-nonylphenol (4NP), Diisodecyl phthalate (DiDP), Tri-m-cresyl phosphate (TMCP) and Tributyltin (TBT) for 48h. Alamar Blue demonstrated that exposure of erythrocytes to each contaminant for up to 48h led to a significant impairment of cellular metabolic activity at 100μM, with the exception of TBT. Moreover, our data indicate that loggerhead erythrocytes constitutively express ERα and HSP60 at the transcript level and respond to EDCs by up-regulating their expression. In this regard, ERα was up-regulated in a dose-dependent manner after 48h exposure to both 4NP and TMCP. Interestingly, the dosage-dependent effects of DiDP on ERα expression were opposite in comparison to that obtained following exposure to the other tested compounds. This work provides the first indication regarding the potential of primary erythrocytes as study models for evaluating the effects of EDCs on sea turtles.
Collapse
Affiliation(s)
- Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy.
| | - Martina Capriotti
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Gilberto Mosconi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III Da Varano, I-62032 Camerino, MC, Italy
| |
Collapse
|
42
|
Mediating Roles of PPARs in the Effects of Environmental Chemicals on Sex Steroids. PPAR Res 2017; 2017:3203161. [PMID: 28819354 PMCID: PMC5551527 DOI: 10.1155/2017/3203161] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/23/2017] [Accepted: 06/21/2017] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear receptors that are widely involved in various physiological functions. They are widely expressed through the reproductive system. Their roles in the metabolism and function of sex steroids and thus the etiology of reproductive disorders receive great concern. Various kinds of exogenous chemicals, especially environmental pollutants, exert their adverse impact on the reproductive system through disturbing the PPAR signaling pathway. Chemicals could bind to PPARs and modulate the transcription of downstream genes containing PPRE (peroxisome proliferator response element). This will lead to altered expression of genes related to metabolism of sex steroids and thus the abnormal physiological function of sex steroids. In this review, various kinds of environmental ligands are summarized and discussed. Their interactions with three types of PPARs are classified by various data from transcript profiles, PPRE reporter in cell line, in silico docking, and gene silencing. The review will contribute to the understanding of the roles of PPARs in the reproductive toxicology of environmental chemicals.
Collapse
|
43
|
Chen Q, Yang H, Zhou N, Sun L, Bao H, Tan L, Chen H, Ling X, Zhang G, Huang L, Li L, Ma M, Yang H, Wang X, Zou P, Peng K, Liu T, Shi X, Feng D, Zhou Z, Ao L, Cui Z, Cao J. Phthalate exposure, even below US EPA reference doses, was associated with semen quality and reproductive hormones: Prospective MARHCS study in general population. ENVIRONMENT INTERNATIONAL 2017; 104:58-68. [PMID: 28441547 DOI: 10.1016/j.envint.2017.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/13/2017] [Accepted: 04/11/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Environment-Protection-Agency Reference Doses (EPA RfDs) for phthalate intakes are based on limited evidence, especially regarding low-dose male-reproductive toxicity. This study investigates the association between phthalate exposure and semen parameters and reproductive hormones in a general population with low phthalate exposure compared to the EPA RfDs. METHODS The MARHCS (Male-Reproductive-Health-in-Chongqing-College-Students) cohort recruited 796 male students, who experienced a relocation of campuses and shifting environmental exposure. Urine, semen and blood before and after the relocation was collected and investigated for: (1) the associations between 13 urinary phthalate metabolites and 11 semen/hormone outcomes (five semen parameters including semen volume, sperm concentration, total sperm number, progressive motility, normal morphology) and six serum reproductive hormones including estradiol, follicle-stimulating hormone, luteinizing hormone, prolactin, progesterone, testosterone; (2) re-analysis of the metabolite-outcome associations in the subjects with estimated phthalate intakes below the RfDs; (3) a change in phthalate metabolites and change in semen/hormone outcomes after the relocation; (4) the association between these changes. RESULTS (1) All but two semen/hormone outcomes were associated with at least one phthalate metabolite, e.g., each quartile monoethyl phthalate was associated with a 5.3%, 5.7% and 2.6% decrease of sperm concentration, total sperm number and progressive motility respectively. (2) In the subjects with phthalate intakes below the RfDs, these metabolite-outcome associations remained significant. (3) All metabolites except mono(2-ethylhexyl) phthalate declined after relocation (P<0.001 respectively); at the same time, semen volume, normal morphology, estradiol and luteinizing hormone increased (by 5.9%, 25.0%, 34.2% and 10.0%) and testosterone decreased (by 7.0%). (4) The changes in semen volume, normal morphology, estradiol and testosterone, but not the change in luteinizing hormone after relocation, were associated with the changes in the phthalate metabolites. CONCLUSIONS Phthalate exposure is associated with interrupted semen quality and reproductive hormones in the human population even with a dose given below the RfDs. These effects, however, may only partially revert back when exposure decreases, thus emphasizing the urgency of stricter phthalate administration.
Collapse
Affiliation(s)
- Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Huan Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Niya Zhou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Lei Sun
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Huaqiong Bao
- Chongqing Institute of Science and Technology for Population and Family Planning, Yangheercun 5, Chongqing 401147, China
| | - Lu Tan
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Hongqiang Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Xi Ling
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Guowei Zhang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Linping Huang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Lianbing Li
- Chongqing Institute of Science and Technology for Population and Family Planning, Yangheercun 5, Chongqing 401147, China
| | - Mingfu Ma
- Chongqing Institute of Science and Technology for Population and Family Planning, Yangheercun 5, Chongqing 401147, China
| | - Hao Yang
- Chongqing Institute of Science and Technology for Population and Family Planning, Yangheercun 5, Chongqing 401147, China
| | - Xiaogang Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Peng Zou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Kaige Peng
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Taixiu Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Xiefei Shi
- National Institute of Measurement and Testing Technology, Yushuang St 10, Chengdu, Sichuan 610021, China
| | - Dejian Feng
- National Institute of Measurement and Testing Technology, Yushuang St 10, Chengdu, Sichuan 610021, China
| | - Ziyuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Zhihong Cui
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China.
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China.
| |
Collapse
|
44
|
Kishi R, Araki A, Minatoya M, Hanaoka T, Miyashita C, Itoh S, Kobayashi S, Ait Bamai Y, Yamazaki K, Miura R, Tamura N, Ito K, Goudarzi H. The Hokkaido Birth Cohort Study on Environment and Children's Health: cohort profile-updated 2017. Environ Health Prev Med 2017; 22:46. [PMID: 29165157 PMCID: PMC5664568 DOI: 10.1186/s12199-017-0654-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/25/2017] [Indexed: 12/11/2022] Open
Abstract
The Hokkaido Study on Environment and Children's Health is an ongoing study consisting of two birth cohorts of different population sizes: the Sapporo cohort and the Hokkaido cohort. Our primary study goals are (1) to examine the effects of low-level environmental chemical exposures on birth outcomes, including birth defects and growth retardation; (2) to follow the development of allergies, infectious diseases, and neurobehavioral developmental disorders and perform a longitudinal observation of child development; (3) to identify high-risk groups based on genetic susceptibility to environmental chemicals; and (4) to identify the additive effects of various chemicals, including tobacco smoking. The purpose of this report is to update the progress of the Hokkaido Study, to summarize the recent results, and to suggest future directions. In particular, this report provides the basic characteristics of the cohort populations, discusses the population remaining in the cohorts and those who were lost to follow-up at birth, and introduces the newly added follow-up studies and case-cohort study design. In the Sapporo cohort of 514 enrolled pregnant women, various specimens, including maternal and cord blood, maternal hair, and breast milk, were collected for the assessment of exposures to dioxins, polychlorinated biphenyls, organochlorine pesticides, perfluoroalkyl substances, phthalates, bisphenol A, and methylmercury. As follow-ups, face-to-face neurobehavioral developmental tests were conducted at several different ages. In the Hokkaido cohort of 20,926 enrolled pregnant women, the prevalence of complicated pregnancies and birth outcomes, such as miscarriage, stillbirth, low birth weight, preterm birth, and small for gestational age were examined. The levels of exposure to environmental chemicals were relatively low in these study populations compared to those reported previously. We also studied environmental chemical exposure in association with health outcomes, including birth size, neonatal hormone levels, neurobehavioral development, asthma, allergies, and infectious diseases. In addition, genetic and epigenetic analyses were conducted. The results of this study demonstrate the effects of environmental chemical exposures on genetically susceptible populations and on DNA methylation. Further study and continuous follow-up are necessary to elucidate the combined effects of chemical exposure on health outcomes.
Collapse
Affiliation(s)
- Reiko Kishi
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan.
| | - Atsuko Araki
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Machiko Minatoya
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Tomoyuki Hanaoka
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Chihiro Miyashita
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Sachiko Itoh
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Sumitaka Kobayashi
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Yu Ait Bamai
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Keiko Yamazaki
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Ryu Miura
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
| | - Naomi Tamura
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Kumiko Ito
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
- Department of Public Health, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Houman Goudarzi
- Hokkaido University Center for Environmental and Health Sciences, Kita 12, Nishi 7, Kita-ku, Sapporo, 060-0812, Japan
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
45
|
Hu J, Cao X, Pang D, Luo Q, Zou Y, Feng B, Li L, Chen Z, Huang C. Tumor grade related expression of neuroglobin is negatively regulated by PPARγ and confers antioxidant activity in glioma progression. Redox Biol 2017; 12:682-689. [PMID: 28410531 PMCID: PMC5390670 DOI: 10.1016/j.redox.2017.03.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
Neuroglobin (NGB), distributed mainly in central and peripheral nervous systems, is a nerve globin with neuroprotective effects against oxidative stress resulting from hypoxia and ischemia. Recent studies have indicated that the expression of NGB is related to neurodegenerative disorders and cancers, but the molecular mechanisms for its transcriptional regulation and protection are not well defined. Here, we report that the expression of NGB in glioma is grade related and is negatively regulated by PPARγ. Specific PPARγ agonist reduces the expression of NGB, while its inhibitor enhances the expression. Moreover, NGB participates in regulating the phosphorylation of AKT in glioma cells, which may contribute to the glioma progression where accumulating oxidative pressure presents. Overexpression of NGB could protect glioma cells against 4-HNE induced cell death, and partially reverse PPARγ's pro-apoptotic and anti-proliferative abilities. These results display an important role of NGB in glioma progression and a mechanism for its transcriptional regulation, and suggest that the treatment on glioma through PPARγ agonist appears to be triggered by the modulation of NGB.
Collapse
Affiliation(s)
- Jing Hu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Department of Neurology, Shuangnan hospital of Chengdu, Chengdu 610000, China
| | - Xiyue Cao
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dejiang Pang
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, West China hospital, Sichuan University, Chengdu 610041, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
46
|
Araki A, Mitsui T, Goudarzi H, Nakajima T, Miyashita C, Itoh S, Sasaki S, Cho K, Moriya K, Shinohara N, Nonomura K, Kishi R. Prenatal di(2-ethylhexyl) phthalate exposure and disruption of adrenal androgens and glucocorticoids levels in cord blood: The Hokkaido Study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 581-582:297-304. [PMID: 28043700 DOI: 10.1016/j.scitotenv.2016.12.124] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/17/2016] [Accepted: 12/18/2016] [Indexed: 05/21/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is known for its endocrine disrupting properties. We previously demonstrated that prenatal DEHP exposure is associated with decreased progesterone levels and testosterone/estradiol ratio in the cord blood. However, evidence of the effects of prenatal DEHP exposure on adrenal androgen and glucocorticoids in infants is scarce. Thus, the objectives of this study were to investigate the association between prenatal DEHP exposure and adrenal androgen and glucocorticoids, and to discuss its effects on steroid hormone profiles in infants. This is part of a birth cohort study: The Hokkaido Study on Environment and Children's Health, Sapporo Cohort. Among the 514 participants, 202 mother-infant pairs with available data on maternal mono(2-ethylhexyl) phthalate (MEHP), adrenal androgen (dehydroepiandrostenedione [DHEA] and androstenedione) and glucocorticoid (cortisol and cortisone) cord blood levels were included in this study. After adjusting for potential confounders, a linear regression analysis showed that maternal MEHP levels were associated with reduced cortisol and cortisone levels and glucocorticoid/adrenal androgen ratio, whereas increased DHEA levels and DHEA/androstenedione ratio. In a quartile model, when comparing the adjusted least square means in the 4th quartile of MEHP with those in the 1st quartile, cortisol and cortisone levels and glucocorticoid/adrenal androgen ratio decreased, whereas DHEA/androstenedione and cortisol/cortisone ratios increased. Significant p-value trends for cortisol and cortisone levels, cortisol/cortisone ratio, and glucocorticoid/adrenal androgen ratio were observed. In combination with the previous results of reduced progesterone levels and testosterone/estradiol ratio, prenatal exposure to DEHP altered the steroid hormone profiles of infants. Further studies investigating the long-term effects of DEHP exposure on growth, neurodevelopment, and gonad and reproductive function are required.
Collapse
Affiliation(s)
- Atsuko Araki
- Center for Environmental and Health Sciences, Hokkaido University, Kita 12, Nishi 7, Sapporo, Hokkaido, Japan
| | - Takahiko Mitsui
- Department of Urology, Hokkaido University Hospital, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan; Yamanashi University, 1110, Shimogato, Chuo, Yamanashi, Japan
| | - Houman Goudarzi
- Center for Environmental and Health Sciences, Hokkaido University, Kita 12, Nishi 7, Sapporo, Hokkaido, Japan; Division of Respiratory Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan
| | - Tamie Nakajima
- Graduate School of Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, Japan; College of Life and Health Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi, Japan
| | - Chihiro Miyashita
- Center for Environmental and Health Sciences, Hokkaido University, Kita 12, Nishi 7, Sapporo, Hokkaido, Japan
| | - Sachiko Itoh
- Center for Environmental and Health Sciences, Hokkaido University, Kita 12, Nishi 7, Sapporo, Hokkaido, Japan
| | - Seiko Sasaki
- Department of Public Health, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan
| | - Kazutoshi Cho
- Department of Obstetrics and Gynecology, Hokkaido University Hospital, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan
| | - Kimihiko Moriya
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan
| | - Katsuya Nonomura
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Sapporo, Hokkaido, Japan; Kushiro Rosai Hospital, 13-23, Nakazono-cho, Kushiro, Hokkaido, Japan
| | - Reiko Kishi
- Center for Environmental and Health Sciences, Hokkaido University, Kita 12, Nishi 7, Sapporo, Hokkaido, Japan.
| |
Collapse
|
47
|
Hinson JT, Chopra A, Lowe A, Sheng CC, Gupta RM, Kuppusamy R, O'Sullivan J, Rowe G, Wakimoto H, Gorham J, Burke MA, Zhang K, Musunuru K, Gerszten RE, Wu SM, Chen CS, Seidman JG, Seidman CE. Integrative Analysis of PRKAG2 Cardiomyopathy iPS and Microtissue Models Identifies AMPK as a Regulator of Metabolism, Survival, and Fibrosis. Cell Rep 2016; 17:3292-3304. [PMID: 28009297 PMCID: PMC5193246 DOI: 10.1016/j.celrep.2016.11.066] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/19/2016] [Accepted: 11/21/2016] [Indexed: 01/20/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a metabolic enzyme that can be activated by nutrient stress or genetic mutations. Missense mutations in the regulatory subunit, PRKAG2, activate AMPK and cause left ventricular hypertrophy, glycogen accumulation, and ventricular pre-excitation. Using human iPS cell models combined with three-dimensional cardiac microtissues, we show that activating PRKAG2 mutations increase microtissue twitch force by enhancing myocyte survival. Integrating RNA sequencing with metabolomics, PRKAG2 mutations that activate AMPK remodeled global metabolism by regulating RNA transcripts to favor glycogen storage and oxidative metabolism instead of glycolysis. As in patients with PRKAG2 cardiomyopathy, iPS cell and mouse models are protected from cardiac fibrosis, and we define a crosstalk between AMPK and post-transcriptional regulation of TGFβ isoform signaling that has implications in fibrotic forms of cardiomyopathy. Our results establish critical connections among metabolic sensing, myocyte survival, and TGFβ signaling.
Collapse
Affiliation(s)
- J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Cardiology Center, University of Connecticut Health, Farmington, CT 06030, USA.
| | - Anant Chopra
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Andre Lowe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Calvin C Sheng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rajat M Gupta
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rajarajan Kuppusamy
- Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John O'Sullivan
- Division of Cardiovascular Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Glenn Rowe
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Burke
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Kiran Musunuru
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Cardiovascular Medicine, Beth Israel Deaconess Hospital, Boston, MA 02115, USA
| | - Sean M Wu
- Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
48
|
Increased Urinary Phthalate Levels in Women with Uterine Leiomyoma: A Case-Control Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13121247. [PMID: 27983712 PMCID: PMC5201388 DOI: 10.3390/ijerph13121247] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/09/2016] [Accepted: 12/11/2016] [Indexed: 01/05/2023]
Abstract
We assessed the urinary concentration of 16 phthalate metabolites in 57 women with and without uterine leiomyoma (n = 30 and 27; respectively) to determine the association between phthalate exposure and uterine leiomyoma. To evaluate exposure to di-(2-ethylhexyl) phthalate (DEHP); we calculated the molar sum of DEHP metabolites; ∑3-DEHP (combining mono-(2-ethylhexyl) phthalate (MEHP); mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP); and mono-(2-ethyl-5-oxohexyl) phthalate); ∑4-DEHP (∑3-DEHP plus mono-(2-ethyl-5-carboxypentyl) phthalate); and ∑5-DEHP (∑4-DEHP plus mono (2-(carboxylmethyl)hexyl) phthalate (2cx-MMHP)). The log transformed urinary levels of MEHP; MEHHP; 2cx-MMHP; ∑3-DEHP; ∑4-DEHP; and ∑5-DEHP in the leiomyoma group were significantly higher than those of controls. When we adjusted for age; waist circumference; and parity using multiple logistic regression analyses; we found log ∑3-DEHP (OR = 10.82; 95% CI = 1.25; 93.46) and ∑4-DEHP (OR = 8.78; 95% CI = 1.03; 75.29) were significantly associated with uterine leiomyoma. Our findings suggest an association between phthalate exposure and uterine leiomyoma. However; larger studies are needed to investigate potential interactions between phthalate exposure and uterine leiomyoma.
Collapse
|
49
|
Hwang DY, Cho JS, Oh JH, Shim SB, Jee SW, Lee SH, Seo SJ, Kang HG, Sheen YY, Lee SH, Kim YK. An In Vivo Bioassay for Detecting Antiandrogens Using Humanized Transgenic Mice Coexpressing the Tetracycline-Controlled Transactivator and Human CYP1B1 Gene. Int J Toxicol 2016; 24:157-64. [PMID: 16040568 DOI: 10.1080/10915810590948370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The typical strategy used in analysis of antiandrogens involves the morphological changes of a marker in castrated rats Hershberger assay for the prostate, seminal vesicle, levator ani plus bulbocavernosus muscles (LABC), Cowper’s gland, and glans penis. However, there are disadvantages to this approach, such as the time required, and the results may not correspond to those in actual human exposure. To evaluate its ability for detecting antiandrogens, in vivo the dose effect of di-(2-ethylhexyl) phthalate (DEHP) and time effect of five antiandrogens, DEHP, di-n-butyl phthalate (DBP), diethyl phthalate (DEP), linuron (3-(4-dichlorophenyl)-methoxy-1-methylurea), and 2,4′-DDE (1,1-dichloro-2-( p-chlorophenyl)-2-( o-chlorophenyl)ethylene), were investigated using humanized transgenic mice coexpressing tetracycline-controlled transactivator (tTA) and the human cytochrome P450 (CYP) enzyme CYP1B1 (hCYP1B1). Adult transgenic males were treated with each of the five antiandrogens, and their tTA-driven hCYP1B1 expressions analyzed by real-time polymerase chain reaction (PCR) and/or Western blot and for O-debenzylation activity. Herein, the treatments of adult males with the five antiandrogens were shown to affect the increased levels of tTA-driven hCYP1B1 expression in both dose-dependent and repeated experiments. Thus, this novel in vivo bioassay, using humanized transgenic mice, is useful for measuring antiandrogens, and is a means to a more relevant bioas-say relating to actual human exposure.
Collapse
Affiliation(s)
- Dae Y Hwang
- Division of Laboratory Animal Resources, Korea Food and Drug Administration, National Institute of Toxicological Research, Eunpyng-ku, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mariana M, Feiteiro J, Verde I, Cairrao E. The effects of phthalates in the cardiovascular and reproductive systems: A review. ENVIRONMENT INTERNATIONAL 2016; 94:758-776. [PMID: 27424259 DOI: 10.1016/j.envint.2016.07.004] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 05/20/2023]
Abstract
Every year millions of tons of plastic are produced around the world and humans are increasingly exposed to them. This constant exposure to plastics has raised some concerns against human health, particularly when it comes to phthalates. These compounds have endocrine-disrupting properties, as they have the ability to bind molecular targets in the body and interfere with hormonal function and quantity. The main use of phthalates is to give flexibility to polyvinyl chloride (PVC) polymers. Phthalates are found in a variety of industrial and consumer products, and as they are not covalently bound to the plastic, phthalates contaminate the environment from which human exposure occurs. Studies in human and animal populations suggest a correlation between phthalate exposure and adverse health outcomes, particularly at the reproductive and cardiovascular systems, however there is much less information about the phthalate toxicity of the later. Thus, the main purpose of this review is to present the studies relating the effects already stated of phthalates on the cardiovascular and reproductive systems, and also present the link between these two systems.
Collapse
Affiliation(s)
- Melissa Mariana
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Joana Feiteiro
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Ignacio Verde
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Elisa Cairrao
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|