1
|
Hassan MH, Reiter E, Razzaq M. Automatic ovarian follicle detection using object detection models. Sci Rep 2024; 14:31856. [PMID: 39738599 DOI: 10.1038/s41598-024-82904-8] [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: 06/11/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025] Open
Abstract
Ovaries are of paramount importance in reproduction as they produce female gametes through a complex developmental process known as folliculogenesis. In the prospect of better understanding the mechanisms of folliculogenesis and of developing novel pharmacological approaches to control it, it is important to accurately and quantitatively assess the later stages of ovarian folliculogenesis (i.e. the formation of antral follicles and corpus lutea). Manual counting from histological sections is commonly employed to determine the number of these follicular structures, however it is a laborious and error prone task. In this work, we show the benefits of deep learning models for counting antral follicles and corpus lutea in ovarian histology sections. Here, we use various backbone architectures to build two one-stage object detection models, i.e. YOLO and RetinaNet. We employ transfer learning, early stopping, and data augmentation approaches to improve the generalizability of the object detectors. Furthermore, we use sampling strategy to mitigate the foreground-foreground class imbalance and focal loss to reduce the imbalance between the foreground-background classes. Our models were trained and validated using a dataset containing only 1000 images. With RetinaNet, we achieved a mean average precision of 83% whereas with YOLO of 75% on the testing dataset. Our results demonstrate that deep learning methods are useful to speed up the follicle counting process and improve accuracy by correcting manual counting errors.
Collapse
Affiliation(s)
- Maya Haj Hassan
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France
| | - Eric Reiter
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France
- Université Paris-Saclay, Inria, Inria Saclay-Île-de-France, Palaiseau, 91120, France
| | - Misbah Razzaq
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, 37380, France.
| |
Collapse
|
2
|
Camerano Spelta Rapini C, Di Berardino C, Peserico A, Capacchietti G, Barboni B. Can Mammalian Reproductive Health Withstand Massive Exposure to Polystyrene Micro- and Nanoplastic Derivatives? A Systematic Review. Int J Mol Sci 2024; 25:12166. [PMID: 39596233 PMCID: PMC11595230 DOI: 10.3390/ijms252212166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The widespread use of plastics has increased environmental pollution by micro- and nanoplastics (MNPs), especially polystyrene micro- and nanoplastics (PS-MNPs). These particles are persistent, bioaccumulative, and linked to endocrine-disrupting toxicity, posing risks to reproductive health. This review examines the effects of PS-MNPs on mammalian reproductive systems, focusing on oxidative stress, inflammation, and hormonal imbalances. A comprehensive search in the Web of Science Core Collection, following PRISMA 2020 guidelines, identified studies on the impact of PS-MNPs on mammalian fertility, including oogenesis, spermatogenesis, and folliculogenesis. An analysis of 194 publications revealed significant reproductive harm, such as reduced ovarian size, depleted follicular reserves, increased apoptosis in somatic cells, and disrupted estrous cycles in females, along with impaired sperm quality and hormonal imbalances in males. These effects were linked to endocrine disruption, oxidative stress, and inflammation, leading to cellular and molecular damage. Further research is urgently needed to understand PS-MNPs toxicity mechanisms, develop interventions, and assess long-term reproductive health impacts across generations, highlighting the need to address these challenges given the growing environmental exposure.
Collapse
Affiliation(s)
| | | | - Alessia Peserico
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (C.C.S.R.); (C.D.B.); (G.C.); (B.B.)
| | | | | |
Collapse
|
3
|
You LM, Zhang DC, Lin CS, Lan Q. Phthalate Metabolites Were Related to the Risk of High-Frequency Hearing Loss: A Cross-Sectional Study of National Health and Nutrition Examination Survey. J Multidiscip Healthc 2024; 17:5151-5161. [PMID: 39553265 PMCID: PMC11568771 DOI: 10.2147/jmdh.s481288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
Background Phthalate metabolites are pervasive in the environment and linked to various health issues. This study aimed to investigate the relationship between phthalate metabolites and hearing loss. Methods We conducted a cross-sectional study with 1713 participants based on the National Health and Nutrition Examination Survey 2015-2018. Participants were defined as speech-frequency hearing loss (SFHL) or high-frequency hearing loss (HFHL). We analyzed the baseline characteristics of participants and assessed the detection rates of phthalate metabolites in samples. Phthalate metabolites with detection rates of >85% were enrolled. Then, restricted cubic spline and multivariable logistic regression analyses were conducted to explore the association of phthalate metabolites with hearing loss. Multi-model analysis was employed to select an optimal predictive model for HFHL based on phthalate metabolites and clinical factors. Results Among participants, 24.518% had SFHL and 41.998% had HFHL, associated with older age, higher BMI, male, non-Hispanic white, lower physical activity levels, higher exposure to work noise, hypertension, and diabetes. Monobenzyl phthalate (MBZP) showed a positive linear association with both SFHL and HFHL. Multivariable logistic regression revealed MBZP as a significant risk factor for HFHL (odds ratio=1.339, 95% confidence interval, 1.053-1.707). According to the area under curve (AUC) values, the logistic regression model had the best diagnostic performance of HFHL, with the highest AUC values of 0.865 in the test set. In the model, gender, diabetes, and MBZP were the top predictors of HFHL. Conclusion The study identified a significant association between MBZP exposure and HFHL, highlighting the need to reduce phthalate exposure.
Collapse
Affiliation(s)
- Li-Mei You
- Department of Otolaryngology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People’s Republic of China
| | - De-Chang Zhang
- Department of Otolaryngology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People’s Republic of China
| | - Chang-Shui Lin
- Department of Otolaryngology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People’s Republic of China
| | - Qiong Lan
- Department of Otolaryngology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, 364000, People’s Republic of China
| |
Collapse
|
4
|
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
|
5
|
Correia LVB, de Moraes TT, Pereira AMRDS, de Aguiar GC, Viana MDB, Ribeiro DA, da Silva RCB. Tributyltin induces apoptosis in mammalian cells in vivo: a scoping review. REVIEWS ON ENVIRONMENTAL HEALTH 2024; 0:reveh-2023-0152. [PMID: 39101219 DOI: 10.1515/reveh-2023-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/03/2024] [Indexed: 08/06/2024]
Abstract
The present review aimed to evaluate the apoptotic effect of tributyltin (TBT) exposure on mammalian tissues and cells in vivo. A search was conducted in specialized literature databases including Embase, Medline, Pubmed, Scholar Google, and Scopus for all manuscripts using the following keywords: "tributyltin", "apoptosis", "mammals", "mammalian cells', "eukaryotic cells", 'rodents', "rats", "mice" and "in vivo" for all data published until September 2023. A total of 16 studies were included. The studies have demonstrated that TBT exposure induces apoptosis in cells from various mammalian organs or tissues in vivo. TBT is capable to increase apoptotic cells, to activate proapoptotic proteins such as calpain, caspases, bax and beclin-1 and to inhibit antiapoptotic protein bcl-2. Additionally, TBT alters the ratio of bcl-2/bax which favor apoptosis. Therefore, the activation of enzymes such as calpain induces apoptosis mediated by ERS and caspases through the intrinsic apoptosis pathway. This review has demonstrated that TBT exposure induces apoptosis in mammalian tissues and cells in vivo.
Collapse
Affiliation(s)
- Lucas Vilas Bôas Correia
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| | - Talita Trindade de Moraes
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| | | | - Gabriel Carvalhal de Aguiar
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| | - Milena de Barros Viana
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| | - Daniel Araki Ribeiro
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| | - Regina Cláudia Barbosa da Silva
- Department of Biosciences, Institute of Health and Society, Federal University of São Paulo, 28105 UNIFESP , Santos, SP, Brazil
| |
Collapse
|
6
|
Blaauwendraad SM, Dykgraaf RH, Gaillard R, Liu M, Laven JS, Jaddoe VW, Trasande L. Associations of bisphenol and phthalate exposure and anti-Müllerian hormone levels in women of reproductive age. EClinicalMedicine 2024; 74:102734. [PMID: 39114272 PMCID: PMC11304696 DOI: 10.1016/j.eclinm.2024.102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/07/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Background In women, exposure to endocrine disrupting chemicals might accelerate the depletion of the ovarian reserve and might be associated with accelerative reproductive aging and fertility. We examined the longitudinal associations of exposure to bisphenols and phthalates with anti-Müllerian hormone concentrations. Methods Pregnant women of 18 years or older that resided in Rotterdam between 2002 and 2006 were eligible for participation in this longitudinal prospective cohort study. We measured urinary bisphenol and phthalate concentration at three time-points in pregnancy among 1405 women, of whom 1322 women had serum Anti-Müllerian Hormone (AMH) measurements 6 and/or 9 years postpartum. We performed linear regression models to assess the association of urinary bisphenol and phthalate metabolites with AMH after 6 and 9 years, and linear mixed-effect model to assess the association with AMH over time. Models were adjusted for sociodemographic and lifestyle factors. Findings In our multivariable linear regression models we observed associations of higher urinary pregnancy-averaged mono-isobutyl phthalate (mIBP), mono-(2-ethyl-5-oxohexyl) phthalate (mEOHP), and monobenzyl phthalate (mBzBP) with lower serum AMH after both 6 and 9 years. However, these associations did not remain after adjustment for multiple testing. No significant associations of bisphenol A with AMH were present in our study sample. In our linear mixed-effects models, higher mIBP, mono-(2-ethyl-5-hydroxyhexyl) phthalate (mEHHP), mEOHP, and mBzBP were associated with lower overall AMH levels (differences -0.07 (95% CI -0.13, -0.02), -0.09 (-0.15, -0.02), -0.08 (95% CI -0.14, -0.02), and -0.08 (-0.13, -0.03) μg/L per doubling in mIBP, mEHHP, mEOHP, and mBzBP respectively) (all False Discovery Rate adjusted p-values < 0.05). Interpretation We identify decreases in indices of ovarian reserve in relationship to prenatal phthalate exposures. Studies are needed replicating our results among large multi-ethnic non-pregnant populations and assessing transgenerational effects of exposure on ovarian reserve. Funding This study was supported by the Erasmus Medical Center and Erasmus University Rotterdam, the Netherlands Organisation for Health Research and Development, the European Research Council, the Dutch Heart Foundation, the Dutch Diabetes Foundation, the European Union's Horizon 2020 Research and Innovation Program, the National Institutes of Health, Ansh Labs Webster, and the Royal Netherlands Academy of Arts and Sciences.
Collapse
Affiliation(s)
- Sophia M. Blaauwendraad
- The Generation R Study Group, Erasmus Medical Center (MC), University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ramon H.M. Dykgraaf
- Department of Obstetrics and Gynecology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Romy Gaillard
- The Generation R Study Group, Erasmus Medical Center (MC), University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Mengling Liu
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
- New York University College of Global Public Health, New York University, New York, NY, USA
| | - Joop S. Laven
- Department of Obstetrics and Gynecology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Vincent W.V. Jaddoe
- The Generation R Study Group, Erasmus Medical Center (MC), University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Leonardo Trasande
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
- New York University College of Global Public Health, New York University, New York, NY, USA
| |
Collapse
|
7
|
Yin A, Mao L, Zhang C, Du B, Xiong X, Chen A, Cheng L, Zhang Z, Li X, Zhou Y, Jiang H. Phthalate exposure and subfecundity in preconception couples: A nested case-control study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116428. [PMID: 38723384 DOI: 10.1016/j.ecoenv.2024.116428] [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: 01/30/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Phthalates (PAEs) are endocrine-disrupting chemicals ubiquitously found in the environment. This study aimed to examine the association between exposure of PAEs and subfecundity in preconception couples. METHODS This is a nested case-control study based on preconception cohort. Preconception couples with intention to conceive were enrolled and followed up until a clinically confirmed pregnancy or 12 menstrual cycles of preparation for conception. A total of 107 couples with subfecundity- time to pregnancy (TTP) more than 12 menstrual cycles, and 144 couples ≤12 cycles were included in the analysis. The levels of PAE metabolites in one spot urine samples were detected and compared between the groups. The weighted quantile sum (WQS) regression model and Bayesian kernel machine regression (BKMR) model were used to examine the joint effects of couples' exposure to PAEs on subfecundity. RESULTS Using the multivariate binary logistic regression model, compared to the lowest quartile of urinary ∑PAEs concentration group, both preconception females (aOR=2.42, 95% CI: 1.10-5.30, p=0.027) and males (aOR=2.99, 95% CI: 1.36-6.58, p=0.006) in the highest quartile group had an increased risk of subfecundity, and a dose-response relationship was observed between PAEs and the risk of subfecundity. The WQS analyses found that co-exposure to PAE mixture was a risk factor for subfecundity in preconception female (aOR=1.76, 95% CI: 1.38-2.26, p<0.001), male (aOR=1.58, 95% CI: 1.20-2.08, p=0.001), and couple (aOR=2.39, 95% CI: 1.61-3.52, p<0.001). The BKMR model found a positive combined effect of mixed exposure to PAEs on the risk of subfecundity. CONCLUSIONS PAEs increase the risk of subfecundity in preconception couples. Our research reinforced the need of monitoring PAE exposure for the purpose of improving human reproductive health.
Collapse
Affiliation(s)
- Anxin Yin
- Department of Maternal, Child and Child Health, School of Public Health, Fudan University, Key Laboratory of Health Technology Evaluation (National Health Commission), Fudan University, Shanghai 200032, China
| | - Lisha Mao
- Shenzhen Municipal Center for Disease Control and Prevention, Shenzhen, Guangdong Province 518050, China
| | - Congcong Zhang
- Department of Maternal, Child and Child Health, School of Public Health, Fudan University, Key Laboratory of Health Technology Evaluation (National Health Commission), Fudan University, Shanghai 200032, China
| | - Bingcheng Du
- Department of Statistics, University of Toronto, Canada
| | - Xu Xiong
- School of Public Health and Tropical Medicine, Tulane University, USA
| | - An Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Lu Cheng
- Department of Computer Science, Aalto University, Espoo 02150, Finland
| | - Zhichun Zhang
- School of Public Health, Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Fudan University School of Public Health, Pudong New Area Center for Disease Control and Prevention, Fudan University Pudong Institute of Preventive Medicine, Shanghai 200032, China
| | - Xingying Li
- Department of Maternal, Child and Child Health, School of Public Health, Fudan University, Key Laboratory of Health Technology Evaluation (National Health Commission), Fudan University, Shanghai 200032, China
| | - Ying Zhou
- School of Public Health, Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Fudan University School of Public Health, Pudong New Area Center for Disease Control and Prevention, Fudan University Pudong Institute of Preventive Medicine, Shanghai 200032, China.
| | - Hong Jiang
- Department of Maternal, Child and Child Health, School of Public Health, Fudan University, Key Laboratory of Health Technology Evaluation (National Health Commission), Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Giaccari C, Antonouli S, Anifandis G, Cecconi S, Di Nisio V. An Update on Physiopathological Roles of Akt in the ReprodAKTive Mammalian Ovary. Life (Basel) 2024; 14:722. [PMID: 38929705 PMCID: PMC11204812 DOI: 10.3390/life14060722] [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: 04/20/2024] [Revised: 05/19/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
The phosphoinositide 3-kinase (PI3K)/Akt pathway is a key signaling cascade responsible for the regulation of cell survival, proliferation, and metabolism in the ovarian microenvironment. The optimal finetuning of this pathway is essential for physiological processes concerning oogenesis, folliculogenesis, oocyte maturation, and embryo development. The dysregulation of PI3K/Akt can impair molecular and structural mechanisms that will lead to follicle atresia, or the inability of embryos to reach later stages of development. Due to its pivotal role in the control of cell proliferation, apoptosis, and survival mechanisms, the dysregulation of this molecular pathway can trigger the onset of pathological conditions. Among these, we will focus on diseases that can harm female fertility, such as polycystic ovary syndrome and premature ovarian failure, or women's general health, such as ovarian cancer. In this review, we report the functions of the PI3K/Akt pathway in both its physiological and pathological roles, and we address the existing application of inhibitors and activators for the balancing of the molecular cascade in ovarian pathological environments.
Collapse
Affiliation(s)
- Carlo Giaccari
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (S.A.); (G.A.)
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (S.A.); (G.A.)
| | - Sandra Cecconi
- Department of Life, Health, and Environmental Sciences, Università dell’Aquila, 67100 L’Aquila, Italy
| | - Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, SE-14186 Stockholm, Sweden;
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-14186 Stockholm, Sweden
| |
Collapse
|
9
|
Jiang Y, Xu Y, Xiao S, Zhu X, Lv H, Zang L, Lei S, Xu X, Xu B, Han X, Zhu J, Du J, Ma H, Hu Z, Ling X, Dai J, Lin Y. Phthalate and DINCH exposure and ovarian reserve markers among women seeking infertility care. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172185. [PMID: 38575009 DOI: 10.1016/j.scitotenv.2024.172185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Phthalate exposure can adversely impact ovarian reserve, yet investigation on the influence of its alternative substance, the non-phthalate plasticizer diisononyl-cyclohexane-1,2-dicarboxylate (DINCH), on ovarian reserve is very sparce. We aimed to investigate the associations of phthalate and DINCH exposure as well as their combined mixture with ovarian reserve. This present study included 657 women seeking infertility care in Jiangsu, China (2015-2018). Urine samples during enrollment prior to infertility treatment were analyzed using high-performance liquid chromatography-isotope dilution tandem mass spectrometry (UPLC-MS/MS) to quantify 17 phthalate metabolites and 3 DINCH metabolites. Multivariate linear regression models, Poisson regression models and weighted quantile sum (WQS) regression were performed to access the associations of 17 urinary phthalate metabolites and 3 DINCH metabolites with ovarian reserve markers, including antral follicle count (AFC), anti-Mullerian hormone (AMH), and follicle-stimulating hormone (FSH). We found that the most conventional phthalates metabolites (DMP, DnBP, DiBP, DBP and DEHP) were inversely associated with AFC, and the DINCH metabolites were positively associated with serum FSH levels. The WQS index of phthalate and DINCH mixtures was inversely associated with AFC (% change = -8.56, 95 % CI: -12.63, -4.31) and positively associated with FSH levels (% change =7.71, 95 % CI: 0.21, 15.78). Our findings suggest that exposure to environmental levels of phthalate and DINCH mixtures is inversely associated with ovarian reserve.
Collapse
Affiliation(s)
- Yangqian Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yiqun Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Shuxin Xiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xianxian Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hong Lv
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Lu Zang
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Shuifang Lei
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xin Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Jin Zhu
- Department of Science and Technology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, Jiangsu, China
| | - Jiangbo Du
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China
| | - Xiufeng Ling
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Reproduction, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, Jiangsu, China.
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, Jiangsu, China.
| |
Collapse
|
10
|
Otorkpa OJ, Otorkpa CO. Health effects of microplastics and nanoplastics: review of published case reports. Environ Anal Health Toxicol 2024; 39:e2024020-0. [PMID: 39054834 PMCID: PMC11294665 DOI: 10.5620/eaht.2024020] [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: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 07/27/2024] Open
Abstract
Microplastics and nanoplastics (MNPs) represent a pervasive environmental pollutant, raising significant concerns about potential health effects on humans. These tiny plastic particles have been detected across various environmental matrices, including air, water, soil, and food sources. While the adverse impacts of MNPs on wildlife and ecosystems are well-documented, understanding their effects on human health is still in its infancy. This study aims to comprehensively review existing case reports documenting adverse health outcomes associated with MNPs exposure. Through an extensive literature search, relevant articles were identified and analyzed. MNPs exposure primarily occurs through ingestion and inhalation routes. Health effects on the digestive system include oxidative stress, inflammation, dysbiosis, and metabolic disorders, with cases linking MNPs exposure to gastrointestinal injury and liver dysfunction. Respiratory system impacts include asthma exacerbation and hypersensitivity pneumonitis, particularly in industries involving plastic production. MNPs exposure has also been associated with nervous system conditions, reproductive toxicity, skeletal system interference, excretory system disruption, and cardiovascular morbidity and mortality. Despite limited case reports, the widespread presence of MNPs warrants further investigation into their potential health risks. This study underscores the urgency of understanding and mitigating the adverse health effects posed by MNPs exposure. Further research is imperative in order to comprehensively assess and address the dangers associated with MNPs contamination in the environment.
Collapse
Affiliation(s)
- Oche Joseph Otorkpa
- Department of Public Health, School of Public Health, Texila American University, Georgetown, Guyana
| | - Chinenye Oche Otorkpa
- Department of Physiology, College of Health Sciences, Federal University, Lokoja, Nigeria
| |
Collapse
|
11
|
Panagiotou EM, Damdimopoulos A, Li T, Moussaud-Lamodière E, Pedersen M, Lebre F, Pettersson K, Arnelo C, Papaikonomou K, Alfaro-Moreno E, Lindskog C, Svingen T, Damdimopoulou P. Exposure to the phthalate metabolite MEHP impacts survival and growth of human ovarian follicles in vitro. Toxicology 2024; 505:153815. [PMID: 38685446 DOI: 10.1016/j.tox.2024.153815] [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: 03/01/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
Phthalates are found in everyday items like plastics and personal care products. There is an increasing concern that continuous exposure can adversely affect female fertility. However, experimental data are lacking to establish causal links between exposure and disease in humans. To address this gap, we tested the effects of a common phthalate metabolite, mono-(2-ethylhexyl) phthalate (MEHP), on adult human ovaries in vitro using an epidemiologically determined human-relevant concentration range (2.05 nM - 20.51 mM). Histomorphological assessments, steroid and cytokine measurements were performed on human ovarian tissue exposed to MEHP for 7 days in vitro. Cell viability and gene expression profile were investigated following 7 days of MEHP exposure using the human granulosa cancer cell lines KGN, and COV434, the germline tumor cell line PA-1, and human ovarian primary cells. Selected differentially expressed genes (DEGs) were validated by RT-qPCR and immunofluorescence in human ovarian tissue. MEHP exposure reduced follicular growth (20.51 nM) and increased follicular degeneration (20.51 mM) in ovarian tissue, while not affecting steroid and cytokine production. Out of the 691 unique DEGs identified across all the cell types and concentrations, CSRP2 involved in cytoskeleton organization and YWHAE as well as CTNNB1 involved in the Hippo pathway, were chosen for further validation. CSRP2 was upregulated and CTNNB1 downregulated in both ovarian tissue and cells, whereas YWHAE was downregulated in cells only. In summary, one-week MEHP exposure of human ovarian tissue can perturb the development and survival of human follicles through mechanisms likely involving dysregulation of cytoskeleton organization and Hippo pathway.
Collapse
Affiliation(s)
- Eleftheria Maria Panagiotou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm 14186, Sweden; Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm 171 77, Sweden.
| | - Anastasios Damdimopoulos
- Bioinformatics and Expression Analysis Core Facility, Karolinska Institutet, Stockholm 14186, Sweden
| | - Tianyi Li
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm 14186, Sweden; Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Elisabeth Moussaud-Lamodière
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm 14186, Sweden; Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Mikael Pedersen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | - Filipa Lebre
- Nanosafety Group, International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Karin Pettersson
- Department of Pregnancy and Delivery, Karolinska University Hospital, Stockholm, Sweden
| | - Catarina Arnelo
- Department of Pregnancy and Delivery, Karolinska University Hospital, Stockholm, Sweden
| | - Kiriaki Papaikonomou
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm 171 77, Sweden
| | | | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala, Sweden
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm 14186, Sweden; Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm 171 77, Sweden
| |
Collapse
|
12
|
Vazakidou P, Evangelista S, Li T, Lecante LL, Rosenberg K, Koekkoek J, Salumets A, Velthut-Meikas A, Damdimopoulou P, Mazaud-Guittot S, Fowler PA, Leonards PEG, van Duursen MBM. The profile of steroid hormones in human fetal and adult ovaries. Reprod Biol Endocrinol 2024; 22:60. [PMID: 38778396 PMCID: PMC11110185 DOI: 10.1186/s12958-024-01233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Reproduction in women is at risk due to exposure to chemicals that can disrupt the endocrine system during different windows of sensitivity throughout life. Steroid hormone levels are fundamental for the normal development and function of the human reproductive system, including the ovary. This study aims to elucidate steroidogenesis at different life-stages in human ovaries. METHODS We have developed a sensitive and specific LC-MS/MS method for 21 important steroid hormones and measured them at different life stages: in media from cultures of human fetal ovaries collected from elective terminations of normally progressing pregnancy and in media from adult ovaries from Caesarean section patients, and follicular fluid from women undergoing infertility treatment. Statistically significant differences in steroid hormone levels and their ratios were calculated with parametric tests. Principal component analysis (PCA) was applied to explore clustering of the ovarian-derived steroidogenic profiles. RESULTS Comparison of the 21 steroid hormones revealed clear differences between the various ovarian-derived steroid profiles. Interestingly, we found biosynthesis of both canonical and "backdoor" pathway steroid hormones and corticosteroids in first and second trimester fetal and adult ovarian tissue cultures. 17α-estradiol, a less potent naturally occurring isomer of 17β-estradiol, was detected only in follicular fluid. PCA of the ovarian-derived profiles revealed clusters from: adult ovarian tissue cultures with relatively high levels of androgens; first trimester and second trimester fetal ovarian tissue cultures with relatively low estrogen levels; follicular fluid with the lowest androgens, but highest corticosteroid, progestogen and estradiol levels. Furthermore, ratios of specific steroid hormones showed higher estradiol/ testosterone and estrone/androstenedione (indicating higher CYP19A1 activity, p < 0.01) and higher 17-hydroxyprogesterone/progesterone and dehydroepiandrosterone /androstenedione (indicating higher CYP17A1 activity, p < 0.01) in fetal compared to adult ovarian tissue cultures. CONCLUSIONS Human ovaries demonstrate de novo synthesis of non-canonical and "backdoor" pathway steroid hormones and corticosteroids. Elucidating the steroid profiles in human ovaries improves our understanding of physiological, life-stage dependent, steroidogenic capacity of ovaries and will inform mechanistic studies to identify endocrine disrupting chemicals that affect female reproduction.
Collapse
Affiliation(s)
- Paraskevi Vazakidou
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands.
| | - Sara Evangelista
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Tianyi Li
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Laetitia L Lecante
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Kristine Rosenberg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Nova Vita Clinic, Tallinn, Estonia
| | - Jacco Koekkoek
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Andres Salumets
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
- Competence Center on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Séverine Mazaud-Guittot
- Univ Rennes, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Inserm, Rennes, F-35000, France
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Pim E G Leonards
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Majorie B M van Duursen
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
13
|
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
|
14
|
Parikh FR, Uttamchandani S, Sawkar S, Panpalia M, Naik N, Sinkar P, Kulkarni D, Parikh R. The impact of follicular fluid phthalate metabolites on the ovarian reserve and ovarian function in Indian women undergoing intracytoplasmic sperm injection. F&S SCIENCE 2024; 5:107-120. [PMID: 38219085 DOI: 10.1016/j.xfss.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVE To investigate the adverse effects of phthalate-induced ovarian toxicity on the ovarian reserve and ovarian function. To assess whether the accumulation of higher levels of selected phthalate metabolites in the follicular fluid (FF) of Indian women undergoing intracytoplasmic sperm injection (ICSI) was associated with a decline in their antral follicle count (AFC) and/or serum antimüllerian hormone (AMH) levels, suggesting a negative impact on the ovarian reserve. To evaluate the effects of follicular phthalate metabolites on peak serum estradiol (E2) levels and the total number of oocytes and mature metaphase II (MII) stage oocytes retrieved to assess the impact of phthalate toxicity on ovarian function. DESIGN A subanalysis of an ongoing prospective cohort study was conducted to examine the association between the levels of six phthalate metabolites, namely, mono-n-butyl phthalate (MBP), mono-ethyl phthalate (MEP), mono-isononyl phthalate (MiNP), mono-isodecyl phthalate (MiDP), mono(2-ethyl-5-oxohexyl) phthalate, and mono(2-ethyl-5-hydroxyhexyl) phthalate, in the FF of Indian women undergoing ICSI and their ovarian reserve markers (AFC and serum AMH levels). To investigate the association of these follicular phthalate metabolite levels with the peak E2 levels and the total number of oocytes and number of MII stage oocytes retrieved. SETTING In vitro fertilization center in a referral hospital in India. PATIENT(S) A total of 245 consenting Indian women who had undergone oocyte retrieval between April 2017 and mid-March 2020 were included. Each woman contributed one FF sample to the study. This was screened for six phthalate metabolites. The samples were collected before the coronavirus disease 2019 pandemic. INTERVENTION(S) Using liquid chromatography-tandem mass spectrometry, the total levels of six phthalate metabolites were quantified in the FF of 245 women. Using linear regression models that were unadjusted and adjusted for maternal age and body mass index (BMI), we evaluated the association between the follicular metabolites in these women and their AFC, serum AMH levels, peak E2 levels, total number of oocytes, and MII stage oocytes. MAIN OUTCOME MEASURE(S) To evaluate the impact of phthalate-induced ovarian toxicity on the ovarian reserve and ovarian function in Indian women undergoing ICSI by studying their accumulated levels in their FF. RESULT(S) For MiNP (a metabolite of di-isononyl phthalate), in linear regression models adjusted for age and BMI, we found that with increasing quartiles of follicular MiNP, there was a significant trend in the decrease in mean AFC (P-trend = 0.023) and a suggestive trend in the decrease in mean serum AMH levels (P-trend = 0.077). For MiDP (a metabolite of di-isodecyl phthalate), in the unadjusted regression model, we found that with increasing quartiles of follicular MiDP, there was a significant trend in the decrease in mean serum AMH levels (P-trend = 0.045). For MBP (a metabolite of dibutyl phthalate), in linear regression models adjusted for age and BMI, we found that with increasing quartiles of follicular MBP, there were significant trends in the decrease in the mean number of total oocytes retrieved (P-trend = 0.003), a decrease in the mean number of MII stage oocytes retrieved, (P-trend = 0.003) and a decrease in the mean peak E2 levels (P-trend = 0.016). Although we found that with increasing quartiles of follicular mono(2-ethyl-5-oxohexyl) phthalate there was a decrease in the mean number of total and MII stage oocytes retrieved and higher follicular MEP levels were negatively associated with the mean AFC and serum AMH levels, neither trend was statistically significant. We also found that although follicular MEP levels did not show an adverse impact on ovarian function, follicular mono(2-ethyl-5-hydroxyhexyl) phthalate levels did not show an adverse impact on both the ovarian reserve and function. CONCLUSION In this study of 245 Indian women, higher accumulated FF levels of MiNP and MiDP were negatively associated with AFC and serum AMH levels, suggesting an adverse effect on the ovarian reserve. Higher accumulated FF levels of MBP were negatively associated with the total number of oocytes, MII stage oocytes, and peak E2 values, suggesting a negative impact on ovarian function. Although we found that phthalate-induced ovarian toxicity was statistically significant for selected phthalate metabolites, the role of the cumulative effect of multiple phthalates in the ovarian microenvironment cannot be ruled out and needs to be investigated further.
Collapse
Affiliation(s)
- Firuza Rajesh Parikh
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India.
| | - Shonali Uttamchandani
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India
| | - Sujatha Sawkar
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India
| | - Madhavi Panpalia
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India
| | - Nandkishor Naik
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India
| | | | - Dhananjaya Kulkarni
- Jaslok-FertilTree International Fertility Centre, Department of Assisted Reproduction & Genetics, Jaslok Hospital and Research Centre, Mumbai, India
| | - Rajesh Parikh
- Department of Neuropsychiatry, Jaslok Hospital and Research Centre, Mumbai, India
| |
Collapse
|
15
|
Wu X, Du Z, Ma R, Zhang X, Yang D, Liu H, Zhang Y. Qualitative and quantitative studies of phthalates in extra virgin olive oil (EVOO) by surface-enhanced Raman spectroscopy (SERS) combined with long short term memory (LSTM) neural network. Food Chem 2024; 433:137300. [PMID: 37657163 DOI: 10.1016/j.foodchem.2023.137300] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 08/13/2023] [Accepted: 08/25/2023] [Indexed: 09/03/2023]
Abstract
Phthalates are commonly used plasticizers in the plastics industry, and have received extensive attention due to their reproductive toxicity. Since phthalates are lipophilic solutions, phthalates can easily migrate from packaging to edible oils. This study synthesized stable and sensitive Gold Nanostars as SERS substrates to conduct qualitative and quantitative analysis of two common phthalates, dibutyl phthalate and di(2-ethylhexyl) phthalate. Two ethanol standard solutions and actual oil solutions of phthalates at different concentrations (10, 5, 1, 0.5, 0.1, 0.02 mg/kg) were prepared. After dimension reduction, LSTM achieved the accuracy of 98% for pure EVOO and EVOO adulterated with different types of phthalates. In terms of quantification, LSTM demonstrates great predictive performance with Rp2 greater than 0.97 and the ratio of performance to deviation greater than 5. These results have certain guiding significance for the analysis of plasticizers in edible oil.
Collapse
Affiliation(s)
- Xijun Wu
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China
| | - Zherui Du
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China.
| | - Renqi Ma
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China.
| | - Xin Zhang
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China
| | - Daolin Yang
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China
| | - Hailong Liu
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China
| | - Yungang Zhang
- Measurement Technology & Instrumentation Key Laboratory of Hebei Province, Institute of Electrical Engineering, Yanshan University, Qinhuangdao 066004 China
| |
Collapse
|
16
|
Guo QC, Yao W, Liu C, Deng TR, Li J, Liao HM, Tian WQ, Wang Y, Du YY, Li YF. Associations of personal care products use with reproductive outcomes of IVF/ICSI treatment. Front Endocrinol (Lausanne) 2024; 14:1320893. [PMID: 38327901 PMCID: PMC10847553 DOI: 10.3389/fendo.2023.1320893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction Personal care products (PCPs) contain a number of endocrine-disrupting chemicals (EDCs) that could potentially affect the reproductive function in women of childbearing age. However, studies focused on the effects of PCPs use on reproductive outcomes are very limited. The current study aimed to explore the relationships between PCPs use patterns and reproductive outcomes in women undergoing in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment. Methods A total of 1500 women from the Tongji Reproductive and Environmental (TREE) study between December 2018 and January 2020 were included in this study. Participants provided characteristics of PCPs use within the previous three months. Retrieved oocyte number, mature oocyte number, two distinct pronuclei (2PN) zygote number, fertilization rate, cleavage rate, blastocyst formation rate, implantation, clinical pregnancy, miscarriage, and live birth were followed up as reproductive endpoints. Generalized linear regression model was utilized to assess the associations between various categories of PCPs use and reproductive endpoints of IVF/ICSI. Results After adjusting for relevant covariates, women who used skin care products ≥14 times per week had a reduction of 22.4% in the maturation rate (95% CI: -39.2%, -1.6%) compared to participants who did not use skin care products. After transferring fresh embryos, women who used cosmetics 1-2 times per week (adjusted OR = 2.2, 95% CI: 1.0, 4.8) or 3-7 times per week (adjusted OR = 2.5, 95% CI: 1.2, 5.2) had a higher possibility of miscarriage than those who did not use cosmetics. There was negative association between the use of gel or soap and the cleavage rate among women aged < 30 years old (P for interaction = 0.01). Among women with BMI ≥ 24 kg/m2, the use of gel or soap was negatively associated with the blastocyst formation rate (P for interaction = 0.04), while cosmetics use was negatively associated with the maturation rate (P for interaction = 0.001). Conclusion Our findings suggest that the use of PCPs in women of reproductive age have a potential adverse impact on IVF/ICSI outcomes, particularly skin care and cosmetic products.
Collapse
Affiliation(s)
- Qing-Chun Guo
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Yao
- Department of Reproductive Medicine, General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Chong Liu
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao-Ran Deng
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Li
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong-Mei Liao
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen-Qu Tian
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wang
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yao-Yao Du
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu-Feng Li
- Department of Reproductive Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
17
|
Pan J, Liu P, Yu X, Zhang Z, Liu J. The adverse role of endocrine disrupting chemicals in the reproductive system. Front Endocrinol (Lausanne) 2024; 14:1324993. [PMID: 38303976 PMCID: PMC10832042 DOI: 10.3389/fendo.2023.1324993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
Reproductive system diseases pose prominent threats to human physical and mental well-being. Besides being influenced by genetic material regulation and changes in lifestyle, the occurrence of these diseases is closely connected to exposure to harmful substances in the environment. Endocrine disrupting chemicals (EDCs), characterized by hormone-like effects, have a wide range of influences on the reproductive system. EDCs are ubiquitous in the natural environment and are present in a wide range of industrial and everyday products. Currently, thousands of chemicals have been reported to exhibit endocrine effects, and this number is likely to increase as the testing for potential EDCs has not been consistently required, and obtaining data has been limited, partly due to the long latency of many diseases. The ability to avoid exposure to EDCs, especially those of artificially synthesized origin, is increasingly challenging. While EDCs can be divided into persistent and non-persistent depending on their degree of degradation, due to the recent uptick in research studies in this area, we have chosen to focus on the research pertaining to the detrimental effects on reproductive health of exposure to several EDCs that are widely encountered in daily life over the past six years, specifically bisphenol A (BPA), phthalates (PAEs), polychlorinated biphenyls (PCBs), parabens, pesticides, heavy metals, and so on. By focusing on the impact of EDCs on the hypothalamic-pituitary-gonadal (HPG) axis, which leads to the occurrence and development of reproductive system diseases, this review aims to provide new insights into the molecular mechanisms of EDCs' damage to human health and to encourage further in-depth research to clarify the potentially harmful effects of EDC exposure through various other mechanisms. Ultimately, it offers a scientific basis to enhance EDCs risk management, an endeavor of significant scientific and societal importance for safeguarding reproductive health.
Collapse
Affiliation(s)
- Jing Pan
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Pengfei Liu
- Gynecology Department, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, China
| | - Xiao Yu
- Gynecology Department, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, China
| | - Zhongming Zhang
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Jinxing Liu
- Gynecology Department, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, China
| |
Collapse
|
18
|
Lazzaretti C, Roy N, Paradiso E, Capponi C, Ferrari T, Reggianini F, Sperduti S, Perri C, Baschieri L, Mascolo E, Varani M, Canu G, Trenti T, Nicoli A, Morini D, Iannotti F, Villani MT, Vicini E, Simoni M, Casarini L. Benzo[a]pyrene disrupts LH/hCG-dependent mouse Leydig cell steroidogenesis through receptor/Gαs protein targeting. Sci Rep 2024; 14:844. [PMID: 38191651 PMCID: PMC10774265 DOI: 10.1038/s41598-024-51516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/06/2024] [Indexed: 01/10/2024] Open
Abstract
Steroidogenesis of gonadal cells is tightly regulated by gonadotropins. However, certain polycyclic aromatic hydrocarbons, including Benzo[a]pyrene (BaP), induce reproductive toxicity. Several existing studies have considered higher than environmentally relevant concentrations of BaP on male and female steroidogenesis following long-term exposure. Also, the impact of short-term exposure to BaP on gonadotropin-stimulated cells is understudied. Therefore, we evaluated the effect of 1 nM and 1 µM BaP on luteinizing hormone/choriogonadotropin (LH/hCG)-mediated signalling in two steroidogenic cell models, i.e. the mouse tumor Leydig cell line mLTC1, and the human primary granulosa lutein cells (hGLC) post 8- and 24-h exposure. Cell signalling studies were performed by homogeneous time-resolved fluorescence (HTRF) assay, bioluminescence energy transfer (BRET) and Western blotting, while immunostainings and immunoassays were used for intracellular protein expression and steroidogenesis analyses, respectively. BaP decreased cAMP production in gonadotropin-stimulated mLTC1 interfering with Gαs activation. Therefore, decrease in gonadotropin-mediated CREB phosphorylation in mLTC1 treated with 1 μM BaP was observed, while StAR protein levels in gonadotropin-stimulated mLTC1 cells were unaffected by BaP. Further, BaP decreased LH- and hCG-mediated progesterone production in mLTC1. Contrastingly, BaP failed to mediate any change in cAMP, genes and proteins of steroidogenic machinery and steroidogenesis of gonadotropin-treated hGLC. Our results indicate that short-term exposure to BaP significantly impairs steroidogenic signalling in mLTC1 interfering with Gαs. These findings could have a significant impact on our understanding of the mechanism of reproductive toxicity by endocrine disruptors.
Collapse
Affiliation(s)
- Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy.
| | - Neena Roy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Chiara Capponi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Tommaso Ferrari
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Francesca Reggianini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125, Modena, Italy
| | - Carmela Perri
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Lara Baschieri
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125, Modena, Italy
| | - Elisa Mascolo
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
| | - Manuela Varani
- Department of Laboratory Medicine and Pathology, Azienda USL/Azienda Ospedaliero-Universitaria di Modena, 41126, Modena, Italy
| | - Giulia Canu
- Department of Laboratory Medicine and Pathology, Azienda USL/Azienda Ospedaliero-Universitaria di Modena, 41126, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Azienda USL/Azienda Ospedaliero-Universitaria di Modena, 41126, Modena, Italy
| | - Alessia Nicoli
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Daria Morini
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Francesca Iannotti
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Maria Teresa Villani
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Elena Vicini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, La Sapienza University, Rome, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, via Pietro Giardini 1355, 41126, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125, Modena, Italy
| |
Collapse
|
19
|
Li T, Boberg J, Johansson HKL, Di Nisio V, Christiansen S, Svingen T, Damdimopoulou P. Quantitative analysis of ovarian surface photographs as a tool for assessment of chemical effects on folliculogenesis and ovulation in rats. Reprod Toxicol 2023; 119:108416. [PMID: 37268149 DOI: 10.1016/j.reprotox.2023.108416] [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: 03/18/2023] [Revised: 05/16/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023]
Abstract
Female reproductive toxicity assessments rely on histological evaluation of ovaries by hematoxylin & eosin (H&E)-stained cross-sections. This is time-consuming, labor-intensive and costly, thus alternative methods for ovarian toxicity assessment could be valuable. Here, we report on an improved method based on quantification of antral follicles (AF) and corpora lutea (CL) using ovarian surface photographs, called 'surface photo counting' (SPC). To validate a potential utility for the method to detect effects on folliculogenesis in toxicity studies, we investigated ovaries from rats exposed to two well-known endocrine disrupting chemicals (EDCs), diethylstilbestrol (DES) and ketoconazole (KTZ). Animals were exposed to DES (0.003, 0.012, 0.048 mg/kg body weight (bw)/day) or KTZ (3, 12, 48 mg/kg bw/day) during puberty or adulthood. At the end of the exposure, ovaries were photographed under stereomicroscope and subsequently processed for histological assessments to allow for direct comparison between the two methods by quantifying AF and CL. There was a significant correlation between the SPC and histology methods, albeit CL counts correlated better than AF counts, potentially due to their larger size. Effects of DES and KTZ were found by both methods, suggesting applicability of the SPC method to chemical hazard and risk assessment. Based on our study, we propose that SPC can be employed as a fast and cheap tool for assessment of ovarian toxicity in in vivo studies to prioritize chemical exposure groups for further histological assessment.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, SE-14186 Stockholm, Sweden; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, SE-14186 Stockholm, Sweden.
| | - Julie Boberg
- National Food Institute, Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Hanna K L Johansson
- National Food Institute, Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, SE-14186 Stockholm, Sweden; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, SE-14186 Stockholm, Sweden
| | - Sofie Christiansen
- National Food Institute, Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, SE-14186 Stockholm, Sweden; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, SE-14186 Stockholm, Sweden
| |
Collapse
|
20
|
Silva ABP, Carreiró F, Ramos F, Sanches-Silva A. The role of endocrine disruptors in female infertility. Mol Biol Rep 2023; 50:7069-7088. [PMID: 37402067 PMCID: PMC10374778 DOI: 10.1007/s11033-023-08583-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 06/06/2023] [Indexed: 07/05/2023]
Abstract
INTRODUCTION According to the World Health Organization, infertility is a public health problem that affects around 48 million couples and 186 million individuals worldwide. Endocrine disruptors are one of the causes that raise more concern, given that it is a problem that has evolved with the progress of society. Many chemicals are used by food industry, entering food chain, and directly affecting human health. Endocrine disruptors have the capacity of interfering with the normal hormonal action, metabolism, and biosynthesis, which can lead to a variation of the normal hormonal homeostasis. Some of these endocrine disruptors are highly associated with diseases that are positively correlated with female infertility, such as polycystic ovary syndrome, endometriosis, irregular menstrual cycle and also disturbances on processes as steroidogenesis and development of the ovarian follicles. RESULTS The present literature review covers various aspects of the possible relationship between endocrine disruptors and female infertility. Bisphenol A and its metabolites, phthalates, dioxins, organochlorine, and organophosphate compounds are groups of chemicals considered to have the capacity to disrupt endocrine activity and herein addressed. The results reported in in vivo studies and in clinical trials addressing endocrine disruptors and female infertility were discussed as well as their possible mechanism of action. CONCLUSIONS Large, double-blind, placebo-controlled randomized clinical trials are needed to better understand the mechanisms of action of endocrine disruptors in female infertility, as well as the doses and frequency of exposure responsible for it.
Collapse
Affiliation(s)
- Ana Beatriz P Silva
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Filipa Carreiró
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- National Institute of Agrarian and Veterinary Research (INIAV), Rua dos Lagidos, Lugar da Madalena, Vairão, 4485-655, Vila do Conde, Portugal
| | - Fernando Ramos
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Ana Sanches-Silva
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
- National Institute of Agrarian and Veterinary Research (INIAV), Rua dos Lagidos, Lugar da Madalena, Vairão, 4485-655, Vila do Conde, Portugal.
- Center for Study in Animal Science (CECA)-ICETA, University of Porto, Praça Gomes Teixeira, 14 Apartado, 55142-401, Porto, Portugal.
- Associate Laboratory for Animal and Veterinary Sciences (Al4AnimalS), 1300-477, Lisbon, Portugal.
| |
Collapse
|
21
|
Yi H, Wu H, Zhu W, Lin Q, Zhao X, Lin R, Luo Y, Wu L, Lin D. Phthalate exposure and risk of ovarian dysfunction in endometriosis: human and animal data. Front Cell Dev Biol 2023; 11:1154923. [PMID: 37560165 PMCID: PMC10407402 DOI: 10.3389/fcell.2023.1154923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 07/13/2023] [Indexed: 08/11/2023] Open
Abstract
Objective: We aimed to explore the correlations between and possible mechanisms of common environmental endocrine disruptors, phthalates, and ovarian dysfunction in endometriosis. Methods: Subjects were included in the case group (n = 107) who were diagnosed with endometriosis by postoperative pathology in Fujian Maternal and Child Hospital from February 2018 to February 2021, and the women who were excluded from endometriosis by surgery were as the control group (n = 70). The demographic information of the subjects were evaluated by questionnaire, and the clinical characteristics were evaluated by medical records and 3-year follow-up results. Gas chromatography‒mass spectrometry was used to quantify 10 metabolites of phthalates, including dimethyl ortho-phthalate (DMP), mono-n-methyl phthalate (MMP), dioctyl ortho-phthalate (DEP), mono-ethyl phthalate (MEP), di-n-butyl ortho-phthalate (DBP), mono-butyl phthalate (MBP), benzylbutyl phthalate (BBzP), mono-benzyl; phthalate (MBzP), diethylhexyl phthalate (DEHP) and mono-ethylhexyl phthalate (MEHP), in the urine samples of the subjects. Furthermore, a total of 54 SD rats were exposed to DEHP 0, 5, 50, 100, 250, 500, 1,000, 2000, and 3,000 mg/kg/day for 2 weeks. The SD rats' body weight, oestrus cycle changes, and serum anti-mullerian hormone (AMH) levels were evaluated. After sacrifice, the mass index of the rat uterus and bilateral ovaries were calculated. Finally, bioinformatics analysis of rat ovarian tissues was performed to explore the possible mechanism. SPSS 24.0 (IBM, United States) was used for data analysis. p-value <0.05 was considered statistically significant. Results: The human urinary levels of DMP (p < 0.001), MMP (p = 0.001), DEP (p = 0.003), MEP (p = 0.002), DBP (p = 0.041), MBP (p < 0.001), BBzP (p = 0.009), DEHP (p < 0.001), and MEHP (p < 0.001) were significantly higher in women with endometriosis than in controls. Notably, DEHP was a significant risk factor for endometriosis (OR: 11.0, 95% CI: 5.4-22.6). The area under the ROC curve increased when multiple phthalates were diagnosed jointly, reaching 0.974 as the highest value, which was helpful for the diagnosis of endometriosis. In vivo experiments showed that after DEHP exposure in rats, the mass index of the ovary and uterus decreased in a dose-dependent manner; the oestrus cycle of SD rats was irregularly prolonged and disordered; and the serum AMH level was negatively correlated with the DEHP exposure dose (Rho = -0.8, p < 0.001). Bioinformatics analysis of rat ovarian tissues showed that seven genes involved in the steroid biosynthesis pathway were upregulated and may play a negative role in ovarian function. Conclusion: Exposure to phthalates, especially DEHP, is associated with the occurrence of endometriosis and affects women's reproductive prognosis and ovarian function. The steroid biosynthesis pathway may be related to ovarian dysfunction. The detection of phthalate in urine may become a new biological target for the diagnosis of endometriosis.
Collapse
Affiliation(s)
- Huan Yi
- National Key Gynecology Clinical Specialty Construction Unit of China, Fujian Provincial Key Gynecology Clinical Specialty, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Huamin Wu
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Fuzhou, Fuzhou, Fujian, China
| | - Wenbin Zhu
- National Key Gynecology Clinical Specialty Construction Unit of China, Fujian Provincial Key Gynecology Clinical Specialty, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Qi Lin
- Fujian Health College, Health Management Department, Fuzhou, Fujian, China
| | - Xiaoyan Zhao
- National Key Gynecology Clinical Specialty Construction Unit of China, Fujian Provincial Key Gynecology Clinical Specialty, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Rong Lin
- National Key Gynecology Clinical Specialty Construction Unit of China, Fujian Provincial Key Gynecology Clinical Specialty, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Luo
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Fuzhou, Fuzhou, Fujian, China
| | - Lixiang Wu
- National Key Gynecology Clinical Specialty Construction Unit of China, Fujian Provincial Key Gynecology Clinical Specialty, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Danmei Lin
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Fuzhou, Fuzhou, Fujian, China
| |
Collapse
|
22
|
Cinzori ME, Pacyga DC, Babayev E, Duncan FE, Li Z, Williams PL, Flaws JA, Strakovsky RS. Ovarian volume partially explains associations of phthalate biomarkers with anti-Müllerian hormone and estradiol in midlife women. ENVIRONMENT INTERNATIONAL 2023; 172:107771. [PMID: 36724714 PMCID: PMC10012419 DOI: 10.1016/j.envint.2023.107771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/23/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND/OBJECTIVES Women are ubiquitously exposed to endocrine disruptors, including phthalates. Ovarian follicles undergoing folliculogenesis (indirectly measured by ovarian volume) produce anti-Müllerian hormone (AMH) and estradiol (E2). We evaluated associations of phthalates with ovarian volume to assess whether this explained prior positive associations of phthalates with AMH and E2. METHODS Women ages 45-54 years (n = 614) had transvaginal ultrasounds of right/left ovaries to calculate mean ovarian volume. Women provided up-to-four urine and blood samples for quantifying AMH (first serum sample), E2 (all serum samples), and nine phthalate metabolites (from pooled urine, representing six parent phthalates). Multivariable linear or logistic regression models (for individual phthalate biomarkers), as well as weighted quantile sum (WQS) regression (for mixture analyses) evaluated associations of phthalate biomarkers with ovarian volume. Using cross-sectional mediation analysis, we assessed whether associations of phthalates with ovarian volume partially explained those of phthalates with AMH or E2. RESULTS Most women were non-Hispanic White (68%) and pre-menopausal (67%) with higher urinary phthalate metabolite concentrations than U.S. women. In single-pollutant models, 10% increases in mono(3-carboxypropyl) phthalate (MCPP) and monobenzyl phthalate (MBzP) were associated with 0.44% (95% CI: -0.02%, 0.91%) and 0.62% (95% CI: 0.02%, 1.23%) larger ovarian volumes, respectively. As a cumulative mixture, 10% increases in the phthalate mixture were associated with 2.89% larger ovarian volume (95%CI: 0.27, 5.59) with MCPP (35%) and MBzP (41%) identified as major contributors. Higher ovarian volume due to a 10% increase in MBzP (indirect effect OR: 1.004; 95% CI: 1.00, 1.01) explained 16% of the positive association between MBzP and higher AMH, whereas higher ovarian volume due to a 10% increase in MCPP (indirect effect %Δ: 0.11; 95% CI: -0.01, 0.22) explained 23% of the positive association between MCPP and E2. CONCLUSION In this cross-sectional study, phthalates were associated with increased ovarian volume, with implications for midlife hormone production.
Collapse
Affiliation(s)
- Maria E Cinzori
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Zhong Li
- Roy J. Carver Biotechnology Center, University of Illinois, Urbana-Champaign, IL 61801, United States
| | - Paige L Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61802, United States
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
23
|
Li T, Vazakidou P, Leonards PEG, Damdimopoulos A, Panagiotou EM, Arnelo C, Jansson K, Pettersson K, Papaikonomou K, van Duursen M, Damdimopoulou P. Identification of biomarkers and outcomes of endocrine disruption in human ovarian cortex using In Vitro Models. Toxicology 2023; 485:153425. [PMID: 36621641 DOI: 10.1016/j.tox.2023.153425] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are raising concerns about adverse effects on fertility in women. However, there is a lack of information regarding mechanisms and effects in humans. Our study aims to identify mechanisms of endocrine disruption using two EDCs, diethylstilbestrol (DES) and ketoconazole (KTZ)1. Human ovarian cortical tissue obtained from Caesarean section patients was exposed to 10-9 M - 10-5 M KTZ and 10-10 M - 10-6 M DES in vitro for 6 days. Follicle survival and growth were studied via histology analysis and liquid-chromatography-mass spectrometry-based steroid quantification. RNA-sequencing was performed on COV434, KGN, and primary ovarian cells that were exposed for 24 h. Significantly lower unilaminar follicle densities were observed in DES 10-10 M group, whereas low KTZ exposure reduced secondary follicle density. KTZ 10-5 M reduced levels of pregnenolone and progesterone. RNA-sequencing revealed that 445 and 233 differentially expressed genes (false discovery rate < 0.1) altogether in DES and KTZ exposed groups. Gene set variation analysis showed that both chemicals modulated pathways that are important for folliculogenesis and steroidogenesis. We selected stearoyl-CoA desaturase (SCD) and 7-dehydrocholesterol reductase (DHCR7) for further validation. Up-regulation of both genes in response to KTZ was confirmed by qPCR and in situ RNA hybridization. Further validation with immunofluorescence focused on the expression of SCD in growing follicles in exposed ovarian tissue. In conclusion, SCD may serve as a potential novel human-relevant biomarker of EDC exposure and effects on ovaries.
Collapse
Affiliation(s)
- Tianyi Li
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| | - Paraskevi Vazakidou
- Department Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands.
| | - Pim E G Leonards
- Department Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands.
| | - Anastasios Damdimopoulos
- Bioinformatics and Expression Analysis Core Facility, Department of Biosciences and Nutrition, Karolinska Institute, 14186 Stockholm, Sweden.
| | - Eleftheria Maria Panagiotou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| | - Catarina Arnelo
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| | - Kerstin Jansson
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| | - Karin Pettersson
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| | - Kiriaki Papaikonomou
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| | - Majorie van Duursen
- Department Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands.
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden.
| |
Collapse
|
24
|
Milankov A, Milanović M, Milošević N, Sudji J, Pejaković S, Milić N, Bjelica A, Medić Stojanoska M. The effects of phthalate exposure on metabolic parameters in polycystic ovary syndrome. Clin Chim Acta 2023; 540:117225. [PMID: 36627009 DOI: 10.1016/j.cca.2023.117225] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
BACKGROUND-AIM Phthalates are known as endocrine disrupting chemicals which are present in wide-range of products. The objective of the study was to investigate whether phthalate exposure may attribute to the metabolic syndrome development in women with polycystic ovary syndrome (PCOS). METHOD The cross-sectional study involved 60 women in reproductive age with confirmed PCOS. Anthropometric and biochemical measurements were examined together with detected levels of ten phthalate metabolites measured by GC-MS in morning urine samples. RESULTS In this study at least one phthalate metabolite was detected in 51.7% of samples. Total phthalate metabolites urine concentrations were positively associated with BMI, waist circumference, waist-to-height-ratio (WtHR), leptin serum levels as well as lipid accumulation product (LAP) and visceral adiposity index (VAI). Mono-methyl-phthalate (MMP) levels was significantly correlated with WtHR, LAP and VAI. Additionally, total phthalate metabolites levels were significantly linked with fasting plasma glucose and HOMA index, whereas MMP concentrations were associated with fasting plasma glucose and insulin levels. Total cholesterol (TC) level was statistically significantly higher among PCOS women with detected phthalate metabolites compared to those without phthalates. The sum of all phthalates was correlated with LDL and triglyceride levels as well as TC/HDL. MMP concentrations were linked positively with TC, LDL and triglyceride levels as well as with TC/HDL. It is noteworthy that MMP concentrations were positively associated with testosterone serum levels while the total phthalate metabolites concentrations were also linked but with moderate significance. CONCLUSIONS The increased phthalate metabolites concentrations may interfere with obesity, glucose and lipid impairment in PCOS women. Additionally, testosterone serum levels can be disrupted by MMP.
Collapse
Affiliation(s)
- Andrijana Milankov
- University of Novi Sad, Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Maja Milanović
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Novi Sad, Serbia.
| | - Nataša Milošević
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Novi Sad, Serbia
| | - Jan Sudji
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Novi Sad, Serbia
| | - Slađana Pejaković
- University of Novi Sad, Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Nataša Milić
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Novi Sad, Serbia
| | - Artur Bjelica
- University of Novi Sad, Faculty of Medicine, Department of Obstetrics and Gynecology, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Milica Medić Stojanoska
- University of Novi Sad, Faculty of Medicine, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Vojvodina, Novi Sad, Serbia
| |
Collapse
|
25
|
Bellavia A, Zou R, Björvang RD, Roos K, Sjunnesson Y, Hallberg I, Holte J, Pikki A, Lenters V, Portengen L, Koekkoek J, Lamoree M, Van Duursen M, Vermeulen R, Salumets A, Velthut-Meikas A, Damdimopoulou P. Association between chemical mixtures and female fertility in women undergoing assisted reproduction in Sweden and Estonia. ENVIRONMENTAL RESEARCH 2023; 216:114447. [PMID: 36181890 PMCID: PMC9729501 DOI: 10.1016/j.envres.2022.114447] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/07/2022] [Accepted: 09/25/2022] [Indexed: 05/07/2023]
Abstract
OBJECTIVE Women of reproductive age are exposed to ubiquitous chemicals such as phthalates, parabens, and per- and polyfluoroalkyl substances (PFAS), which have potential endocrine disrupting properties and might affect fertility. Our objective was to investigate associations between potential endocrine-disrupting chemicals (EDCs) and female fertility in two cohorts of women attending fertility clinics. METHODS In a total population of 333 women in Sweden and Estonia, we studied the associations between chemicals and female fertility, evaluating ovarian sensitivity index (OSI) as an indicator of ovarian response, as well as clinical pregnancy and live birth from fresh and frozen embryo transfers. We measured 59 chemicals in follicular fluid samples and detected 3 phthalate metabolites, di-2-ethylhexyl phthalate (DEHP) metabolites, 1 paraben, and 6 PFAS in >90% of the women. Associations were evaluated using multivariable-adjusted linear or logistic regression, categorizing EDCs into quartiles of their distributions, as well as with Bayesian Kernel Machine Regression. RESULTS We observed statistically significant lower OSI at higher concentrations of the sum of DEHP metabolites in the Swedish cohort (Q4 vs Q1, β = -0.21, 95% CI: -0.38, -0.05) and methylparaben in the Estonian cohort (Q3 vs Q1, β = -0.22, 95% CI: -0.44, -0.01). Signals of potential associations were also observed at higher concentrations of PFUnDA in both the combined population (Q2 vs. Q1, β = -0.16, 95% CI -0.31, -0.02) and the Estonian population (Q2 vs. Q1, β = -0.27, 95% CI -0.45, -0.08), and for PFOA in the Estonian population (Q4 vs. Q1, β = -0.31, 95% CI -0.61, -0.01). Associations of chemicals with clinical pregnancy and live birth presented wide confidence intervals. CONCLUSIONS Within a large chemical mixture, we observed significant inverse associations levels of DEHP metabolites and methylparaben, and possibly PFUnDA and PFOA, with OSI, suggesting that these chemicals may contribute to altered ovarian function and infertility in women.
Collapse
Affiliation(s)
- Andrea Bellavia
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Runyu Zou
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Richelle D Björvang
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Kristine Roos
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia; Nova Vita Clinic AS, Tallinn, Estonia
| | - Ylva Sjunnesson
- Department of Clinical Sciences, Division of Reproduction, The Center for Reproductive Biology in Uppsala, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ida Hallberg
- Department of Clinical Sciences, Division of Reproduction, The Center for Reproductive Biology in Uppsala, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jan Holte
- Carl von Linnékliniken, Uppsala, Sweden; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Anne Pikki
- Carl von Linnékliniken, Uppsala, Sweden; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Virissa Lenters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lützen Portengen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jacco Koekkoek
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marja Lamoree
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Majorie Van Duursen
- Amsterdam Institute for Life and Environment, Section Environment and Health, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Andres Salumets
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Competence Center on Health Technologies, Tartu, Estonia; Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
26
|
Redox Status, Estrogen and Progesterone Production by Swine Granulosa Cells Are Impaired by Triclosan. Animals (Basel) 2022; 12:ani12243559. [PMID: 36552479 PMCID: PMC9774123 DOI: 10.3390/ani12243559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Triclosan is a chlorinated biphenolic with a broad spectrum of antiseptic activities used in cosmetics and hygiene products. Continuous exposure can lead to absorption and bioaccumulation of this substance with harmful health effects. In fact, previous studies have shown that Triclosan acts as an endocrine-disrupting chemical on reproductive organs, with consequent negative effects on reproductive physiology. Therefore, to assess potential adverse impacts on fertility, we tested Triclosan on swine granulosa cells, a model of endocrine reproductive cells. We examined its effects on the main features of granulosa cell functions such as cell growth (BrdU incorporation and ATP production) and steroidogenesis (17-β estradiol and progesterone secretion). Moreover, since oxidant−antioxidant balance plays a pivotal role in follicular function, redox status markers (superoxide, hydrogen peroxide and nitric oxide production, enzymatic and non-enzymatic scavenging activity) were studied. Our results show that Triclosan significantly inhibits cell growth (p < 0.001), steroidogenesis (p < 0.001), superoxide and nitric oxide production (p < 0.001), while it increases (p < 0.05) enzymatic defense systems. Collectively, these data suggest a disruption of the main granulosa cell functions, i.e., proliferation and hormone production, as well as an imbalance in redox status. On these bases, we can speculate that Triclosan would impair granulosa cell functions, thus exerting negative effects on reproductive function. Further studies are needed to explore lower Triclosan concentrations and to unravel its mechanisms of action at gene level.
Collapse
|
27
|
Land KL, Miller FG, Fugate AC, Hannon PR. The effects of endocrine-disrupting chemicals on ovarian- and ovulation-related fertility outcomes. Mol Reprod Dev 2022; 89:608-631. [PMID: 36580349 PMCID: PMC10100123 DOI: 10.1002/mrd.23652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/21/2022]
Abstract
Exposure to endocrine-disrupting chemicals (EDCs) is unavoidable, which represents a public health concern given the ability of EDCs to target the ovary. However, there is a large gap in the knowledge about the impact of EDCs on ovarian function, including the process of ovulation. Defects in ovulation are the leading cause of infertility in women, and EDC exposures are contributing to the prevalence of infertility. Thus, investigating the effects of EDCs on the ovary and ovulation is an emerging area for research and is the focus of this review. The effects of EDCs on gametogenesis, uterine function, embryonic development, and other aspects of fertility are not addressed to focus on ovarian- and ovulation-related fertility issues. Herein, findings from epidemiological and basic science studies are summarized for several EDCs, including phthalates, bisphenols, per- and poly-fluoroalkyl substances, flame retardants, parabens, and triclosan. Epidemiological literature suggests that exposure is associated with impaired fecundity and in vitro fertilization outcomes (decreased egg yield, pregnancies, and births), while basic science literature reports altered ovarian follicle and corpora lutea numbers, altered hormone levels, and impaired ovulatory processes. Future directions include identification of the mechanisms by which EDCs disrupt ovulation leading to infertility, especially in women.
Collapse
Affiliation(s)
- Katie L. Land
- Department of Obstetrics & Gynecology, College of MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Frances G. Miller
- Department of Obstetrics & Gynecology, College of MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Ava C. Fugate
- Department of Obstetrics & Gynecology, College of MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Patrick R. Hannon
- Department of Obstetrics & Gynecology, College of MedicineUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
28
|
Beyer A, Schorgg P, Karavasiloglou N, Sarwar S, Rohrmann S, Bärnighausen T, Cassidy A, Connolly L, Kühn T. Urinary phthalate concentrations and mortality risk: A population-based study. ENVIRONMENTAL RESEARCH 2022; 214:113927. [PMID: 35868575 DOI: 10.1016/j.envres.2022.113927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/22/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
Phthalates are widely used as plasticizers. Laboratory-based mechanistic and epidemiological studies suggest that phthalates are detrimental to human health. Here, we present prospective analyses on phthalate exposure and all-cause, as well as cause-specific, mortality from the National Health and Nutrition Examination Survey (NHANES), a population-based cohort. Between 1999 and 2018, urinary concentrations of 12 phthalate metabolites were measured by high-performance liquid chromatography-electrospray ionization tandem mass spectrometry in spot urine samples of 10,881 adults aged 40-85 years, of which 2382 died over a median duration of 8.9 years after sample provision. Multivariable Cox regression analyses adjusted for a wide range of lifestyle factors and comorbidities showed that higher concentrations of mono-benzyl phthalate (MBzP) and Mono-n-butyl phthalate (MnBP) were associated with increased mortality. The hazard ratios for participants in the highest quartiles of MBzP and MnBP concentrations were at 1.27 [95% confidence interval: 1.08, 1.49; p linear trend = 0.002] and 1.35 [1.13, 1.62; p linear trend = 0.005). These findings reinforce the need for monitoring of phthalate exposure in relation to health outcomes.
Collapse
Affiliation(s)
- Anika Beyer
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg, Germany; Medical Faculty of the University of Heidelberg, Heidelberg, Germany
| | - Paula Schorgg
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg, Germany; Medical Faculty of the University of Heidelberg, Heidelberg, Germany
| | - Nena Karavasiloglou
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Switzerland
| | - Sneha Sarwar
- Institute of Nutrition and Food Science, University of Dhaka, Bangladesh
| | - Sabine Rohrmann
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics and Prevention Institute (EBPI), University of Zurich, Switzerland
| | - Till Bärnighausen
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg, Germany; Harvard Center for Population and Development Studies, Cambridge, MA, USA; Africa Health Research Institute, Somkhele and Durban, South Africa
| | - Aedin Cassidy
- The Institute for Global Food Security, Queen's University Belfast, UK
| | - Lisa Connolly
- The Institute for Global Food Security, Queen's University Belfast, UK
| | - Tilman Kühn
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg, Germany; The Institute for Global Food Security, Queen's University Belfast, UK.
| |
Collapse
|
29
|
Sol CM, Gaylord A, Santos S, Jaddoe VWV, Felix JF, Trasande L. Fetal exposure to phthalates and bisphenols and DNA methylation at birth: the Generation R Study. Clin Epigenetics 2022; 14:125. [PMID: 36217170 PMCID: PMC9552446 DOI: 10.1186/s13148-022-01345-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Phthalates and bisphenols are non-persistent endocrine disrupting chemicals that are ubiquitously present in our environment and may have long-lasting health effects following fetal exposure. A potential mechanism underlying these exposure-outcome relationships is differential DNA methylation. Our objective was to examine the associations of maternal phthalate and bisphenol concentrations during pregnancy with DNA methylation in cord blood using a chemical mixtures approach. METHODS This study was embedded in a prospective birth cohort study in the Netherlands and included 306 participants. We measured urine phthalates and bisphenols concentrations in the first, second and third trimester. Cord blood DNA methylation in their children was processed using the Illumina Infinium HumanMethylation450 BeadChip using an epigenome-wide association approach. Using quantile g-computation, we examined the association of increasing all mixture components by one quartile with cord blood DNA methylation. RESULTS We did not find evidence for statistically significant associations of a maternal mixture of phthalates and bisphenols during any of the trimesters of pregnancy with DNA methylation in cord blood (all p values > 4.01 * 10-8). However, we identified one suggestive association (p value < 1.0 * 10-6) of the first trimester maternal mixture of phthalates and bisphenols and three suggestive associations of the second trimester maternal mixture of phthalates and bisphenols with DNA methylation in cord blood. CONCLUSIONS Although we did not identify genome-wide significant results, we identified some suggestive associations of exposure to a maternal mixture of phthalates and bisphenols in the first and second trimester with DNA methylation in cord blood that need further exploration in larger study samples.
Collapse
Affiliation(s)
- Chalana M. Sol
- grid.5645.2000000040459992XThe Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Pediatrics, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Abigail Gaylord
- grid.137628.90000 0004 1936 8753Department of Population Health, New York University School of Medicine, 403 East 34th Street, Room 115, New York City, NY 10016 USA
| | - Susana Santos
- grid.5645.2000000040459992XThe Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Pediatrics, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W. V. Jaddoe
- grid.5645.2000000040459992XThe Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Pediatrics, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F. Felix
- grid.5645.2000000040459992XThe Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Pediatrics, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Leonardo Trasande
- Department of Population Health, New York University School of Medicine, 403 East 34th Street, Room 115, New York City, NY, 10016, USA. .,Department of Pediatrics, New York University School of Medicine, 403 East 34th Street, Room 115, New York City, NY, 10016, USA. .,Department of Environmental Medicine, New York University School of Medicine, 403 East 34th Street, Room 115, New York City, NY, 10016, USA. .,New York Wagner School of Public Service, New York City, NY, 10016, USA. .,New York University Global Institute of Public Health, New York City, NY, 10016, USA.
| |
Collapse
|
30
|
Gao L, Zhang C, Yu S, Liu S, Wang G, Lan H, Zheng X, Li S. Glycine ameliorates MBP-induced meiotic abnormalities and apoptosis by regulating mitochondrial-endoplasmic reticulum interactions in porcine oocytes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 309:119756. [PMID: 35839969 DOI: 10.1016/j.envpol.2022.119756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/16/2022] [Accepted: 07/08/2022] [Indexed: 05/14/2023]
Abstract
Monobutyl phthalate (MBP) is the main metabolite of dibutyl phthalate (DBP) in vivo. MBP has a stable structure, can continuously accumulate in living organisms, and has the potentially to harm animal and human reproductive function. In the ovarian follicle microenvironment, MBP may lead to defects in follicular development and steroid production, abnormal meiotic maturation, impaired ovarian function and other reproductive deficits. In this study, SMART-seq was used to investigate the effects of MBP exposure on the in vitro maturation (IVM) and development of porcine oocytes. The results showed that differentially expressed genes after MBP exposure were enriched in the biological processes cytoskeleton, cell apoptosis, endoplasmic reticulum (ER) and mitochondria. Glycine (Gly) improved the developmental potential of porcine oocytes by regulating mitochondrial and ER function. The effect of Gly in protecting oocytes against MBP-induced damage was studied. The results showed that the addition of Gly significantly decreased the rate of MBP-induced spindle abnormalities, decreased the frequency of MBP-induced mitochondria-associated ER membrane (MAM) interactions, and downregulated the protein and gene expression of the linkage molecules Mitofusin 1 (MFN1) and Mitofusin 2 (MFN2) in the MAM. Additionally, treatment with Gly restored the distribution of the 1,4,5-triphosphate receptor 1 (IP3R1) and voltage-dependent anion channel 1 (VDAC1), further decreasing the intracellular free calcium concentration ([Ca2+]i) levels and mitochondrial Ca2+ ([Ca2+]m) , increasing the ER Ca2+ ([Ca2+]ER) levels, and thus significantly increasing the ER levels and mitochondrial membrane potential (ΔΨ m). Gly also decreased the levels of reactive oxygen species (ROS) and increased the levels of Glutathione (GSH), oocyte apoptosis-related indicators (Caspase-3 activity and Annexin V) and oocyte apoptosis-related genes (BAX, Caspase 3 and AIFM1). Our results suggest that Gly can ameliorate microtubule cytoskeleton abnormalities and improve oocyte maturation by reducing the defective mitochondrial-ER interactions caused by MBP exposure in vitro.
Collapse
Affiliation(s)
- Lepeng Gao
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Chang Zhang
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Sicong Yu
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Shuang Liu
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Guoxia Wang
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Hainan Lan
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Xin Zheng
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Suo Li
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China.
| |
Collapse
|
31
|
Mixed Contaminants: Occurrence, Interactions, Toxicity, Detection, and Remediation. Molecules 2022; 27:molecules27082577. [PMID: 35458775 PMCID: PMC9029723 DOI: 10.3390/molecules27082577] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/18/2022] Open
Abstract
The ever-increasing rate of pollution has attracted considerable interest in research. Several anthropogenic activities have diminished soil, air, and water quality and have led to complex chemical pollutants. This review aims to provide a clear idea about the latest and most prevalent pollutants such as heavy metals, PAHs, pesticides, hydrocarbons, and pharmaceuticals—their occurrence in various complex mixtures and how several environmental factors influence their interaction. The mechanism adopted by these contaminants to form the complex mixtures leading to the rise of a new class of contaminants, and thus resulting in severe threats to human health and the environment, has also been exhibited. Additionally, this review provides an in-depth idea of various in vivo, in vitro, and trending biomarkers used for risk assessment and identifies the occurrence of mixed contaminants even at very minute concentrations. Much importance has been given to remediation technologies to understand our current position in handling these contaminants and how the technologies can be improved. This paper aims to create awareness among readers about the most ubiquitous contaminants and how simple ways can be adopted to tackle the same.
Collapse
|
32
|
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: 0.7] [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]
|
33
|
Jadhao M, Chen CL, Liu W, Deshmukh D, Liao WT, Chen JYF, Urade R, Tsai EM, Hsu SK, Wang LF, Chiu CC. Endoglin Modulates TGFβR2 Induced VEGF and Proinflammatory Cytokine Axis Mediated Angiogenesis in Prolonged DEHP-Exposed Breast Cancer Cells. Biomedicines 2022; 10:biomedicines10020417. [PMID: 35203627 PMCID: PMC8962291 DOI: 10.3390/biomedicines10020417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis is the process of vascular network development and plays a crucial role in cancer growth, progression, and metastasis. Phthalates are a class of environmental pollutants that have detrimental effects on human health and are reported to increase cancer risk. However, the interplay between phthalate exposure and angiogenesis has not been investigated thoroughly. In this study, we investigated the effect of prolonged di (2-ethylhexyl) phthalate (DEHP) treatment on the angiogenic potential of triple-negative breast cancer. MDA-MB-231 cells were exposed to physiological concentrations of DEHP for more than three months. Prolonged DEHP exposure induced angiogenesis in breast cancer cells. Endoglin (ENG)/CD105 is a membrane glycoprotein and an auxiliary receptor of the TGFβ receptor complex. In endothelial cells, ENG is highly expressed and it is a prerequisite for developmental angiogenesis. A literature review highlights endoglin as a well-known mesenchymal stem cell marker responsible for vascular development and angiogenesis. NGS analysis showed that endoglin overexpression in DEHP-exposed MDA-MB-231 cells correlated with tumor development and growth. An in vivo zebrafish xenograft assay showed that VEGFA induced sprouting of the subintestinal vein (SIV) in embryos injected with DEHP-exposed cells. Endoglin knockdown reduced SIV sprouting and VEGFA expression in zebrafish embryos. An in vitro HUVEC tube formation assay showed that endoglin depletion reversed DEHP-induced VEGF-mediated HUVEC tube formation in coculture. DEHP-induced endoglin activated TGFβ/SMAD3/VEGF and MAPK/p38 signaling in MDA-MB-231 cells. A cytokine angiogenesis antibody array showed induced expression of the inflammatory cytokines IL1α, IL1β, IL6, and IL8, along with GMCSF and VEGF. Endoglin knockdown reversed DEHP-induced activation of the TGFβ/SMAD3/VEGF signaling axis, MAPK/p38 signaling, and cytokine regulation, limiting angiogenesis potential both in vivo and in vitro. Targeting endoglin might serve as a potential alternative treatment to control angiogenesis, leading to metastasis and limiting cancer progression.
Collapse
Affiliation(s)
- Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.J.); (D.D.)
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; (C.-L.C.); (R.U.)
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.L.); (W.-T.L.); (J.Y.-F.C.); (S.-K.H.)
| | - Dhanashri Deshmukh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.J.); (D.D.)
| | - Wei-Ting Liao
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.L.); (W.-T.L.); (J.Y.-F.C.); (S.-K.H.)
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.L.); (W.-T.L.); (J.Y.-F.C.); (S.-K.H.)
| | - Ritesh Urade
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; (C.-L.C.); (R.U.)
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.L.); (W.-T.L.); (J.Y.-F.C.); (S.-K.H.)
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.J.); (D.D.)
- Correspondence: (L.-F.W.); (C.-C.C.); Tel.: +886-67-312-1101 (ext. 2217) (L.-F.W.); +886-67-312-1101 (ext. 2368) (C.-C.C.); Fax: +886-67-312-5339 (L.-F.W.)
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; (C.-L.C.); (R.U.)
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.L.); (W.-T.L.); (J.Y.-F.C.); (S.-K.H.)
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (L.-F.W.); (C.-C.C.); Tel.: +886-67-312-1101 (ext. 2217) (L.-F.W.); +886-67-312-1101 (ext. 2368) (C.-C.C.); Fax: +886-67-312-5339 (L.-F.W.)
| |
Collapse
|