Please wait a minute...
Frontiers of Environmental Science & Engineering

ISSN 2095-2201

ISSN 2095-221X(Online)

CN 10-1013/X

Postal Subscription Code 80-973

2018 Impact Factor: 3.883

Front. Environ. Sci. Eng.    2023, Vol. 17 Issue (3) : 28    https://doi.org/10.1007/s11783-023-1628-x
RESEARCH ARTICLE
Untargeted metabolomic analysis of pregnant women exposure to perfluorooctanoic acid at different degrees
Kaige Yang1, Zhouyi Zhang1, Kangdie Hu1, Bo Peng1, Weiwei Wang1, Hong Liang3, Chao Yan1, Mingyuan Wu1,2(), Yan Wang1()
1. School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
2. Engineering Research Center of Cell and Therapeutic Antibody (Ministry of Education), School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
3. NHC Key Laboratory of Reproduction Regulation, Shanghai Institute of Planned Parenthood, Shanghai 200237, China
 Download: PDF(8788 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

● Metabolome can distinguish pregnant women exposure to PFOA at different degrees.

● Metabolome can reveal the metabolic changes of pregnant women exposure to PFOA.

● PFOA exposure degrees could affect the GSH metabolism of pregnant women.

● PFOA exposure degrees could change the microbiota metabolism of pregnant women.

Perfluorooctanoic acid (PFOA) is a novel type of persistent synthetic organic pollutant, and its exposure on pregnant women can cause some adverse effects, such as pregnancy-induced hypertension, gestational diabetes mellitus, and preeclampsia. Therefore, understanding the metabolic changes caused by PFOA exposure is of great significance to protect pregnant women from its adverse effects. In this study, the metabolomes from the urine samples of pregnant women exposure to PFOA at different degrees were analyzed by GC-MS and LC-MS. The samples in different groups were distinguished and the differential metabolites were screened based on the VIP value, FC, and P-value of each comparison group through multivariate statistical analysis. The pathways related to differential metabolites were searched to reveal the effects of PFOA exposure on metabolic changes in pregnant women at different degrees. Finally, the ROC of differential metabolites was performed, and the differential metabolites with large area under the curve (AUC) values were selected and compared to identify the mutually differential metabolites. Meanwhile, these metabolites were fitted with a multivariable to explore if they could be used to distinguish different groups. The quantitative comparison of mutually differential metabolites revealed that the levels of L-cysteine, glycine, and 5-aminovaleric acid were positively correlated with the degree of PFOA exposure, indicating that different degrees of PFOA exposure could affect the synthesis or degradation of GSH and change the metabolism of oral or intestinal microbiota. Additionally, they may cause oxidative stress and abnormal fat metabolism in pregnant women.

Keywords Perfluorooctanoic acid      Exposure      Pregnant women      Metabolomic      GSH      Microbiota metabolism     
Corresponding Author(s): Mingyuan Wu,Yan Wang   
Issue Date: 18 November 2022
 Cite this article:   
Kaige Yang,Zhouyi Zhang,Kangdie Hu, et al. Untargeted metabolomic analysis of pregnant women exposure to perfluorooctanoic acid at different degrees[J]. Front. Environ. Sci. Eng., 2023, 17(3): 28.
 URL:  
https://academic.hep.com.cn/fese/EN/10.1007/s11783-023-1628-x
https://academic.hep.com.cn/fese/EN/Y2023/V17/I3/28
Fig.1  Multivariate statistical analysis of the metabolomic data of pregnant women’s urine samples in the QC, Exp-L, Exp-M, and Exp-H groups acquired by GC-MS and LC-MS. The PCA score plots of metabolomic data of pregnant women’s urine samples in the QC, Exp-L, Exp-M, and Exp-H groups acquired by (a) GC-MS and (b) LC-MS. The OPLS-DA score plots of metabolomic data of (c) Exp-H vs. Exp-M, (d) Exp-M vs. Exp-L, and (e) Exp-H vs. Exp-L groups acquired by GC-MS. The OPLS-DA score plots of metabolomic data of (f) Exp-H vs. Exp-M, (g) Exp-M vs. Exp-L, and (h) Exp-H vs. Exp-L groups acquired by LC-MS.
Fig.2  Volcano plots of metabolites in the urine samples of pregnant women in the Exp-H, Exp-M, and Exp-L groups acquired by GC-MS and LC-MS. The volcano plots of metabolites in the urine samples of (a) Exp-H vs. Exp-M, (b) Exp-M vs. Exp-L, and (c) Exp-H vs. Exp-L groups acquired by GC-MS. The volcano plots of metabolites in the urine samples in (d) Exp-H vs. Exp-M, (e) Exp-M vs. Exp-L, and (f) Exp-H vs. Exp-L groups acquired by LC-MS.
Fig.3  Bubble diagrams of the KEGG pathways of differential metabolites in Exp-H, Exp-M, and Exp-L groups acquired by GC-MS and LC-MS. Bubble diagrams of the KEGG pathways of differential metabolites observed in (a) Exp-H vs. Exp-M, (b) Exp-M vs. Exp-L, and (c) Exp-H vs. Exp-L by GC-MS. Bubble diagrams of the KEGG pathways of differential metabolites detected in (d) Exp-H vs. Exp-M, (e) Exp-M vs. Exp-L, and (f) Exp-H vs. Exp-L by LC-MS.
Fig.4  Box plots of L-cysteine, dehydroabietic acid, glycine, and 5-aminovaleric acid contents in pregnant women’s urine samples in the Exp-L, Exp-M, and Exp-H groups: (a) L-cysteine and (b) dehydroabietic acid analyzed by GC-MS; and (c) glycine and (d) 5-aminovaleric acid analyzed by LC-MS. *p < 0.05, ** p < 0.01, *** p < 0.001.
Fig.5  ROC of fitted multivariable from the comparison pairs Exp-H vs. Exp-M, Exp-M vs. Exp-L, and Exp-H vs. Exp-L. ROC of the multivariable was fitted with the data on L-cysteine and dehydroabietic acid acquired by GC-MS from the comparison pairs (a) Exp-H vs. Exp-M, (b) Exp-M vs. Exp-L, and (c) Exp-H vs. Exp-L. ROC of multivariable fitted using the data on glycine and 5-aminovaleric acid acquired by LC-MS from the comparison pairs (d) Exp-H vs. Exp-M, (e) Exp-M vs. Exp-L, and (f) Exp-H vs. Exp-L.
1 T L Alderete, R Jin, D I Walker, D Valvi, Z Chen, D P Jones, C Peng, F D Gilliland, K Berhane, D V Conti, M I Goran, L Chatzi. (2019). Perfluoroalkyl substances, metabolomic profiling, and alterations in glucose homeostasis among overweight and obese Hispanic children: a proof-of-concept analysis. Environment International, 126 : 445– 453
https://doi.org/10.1016/j.envint.2019.02.047 pmid: 30844580
2 H Arazi, E Eghbali, K Suzuki. (2021). Creatine supplementation, physical exercise and oxidative stress markers: a review of the mechanisms and effectiveness. Nutrients, 13( 3): 869
https://doi.org/10.3390/nu13030869 pmid: 33800880
3 G Asantewaa, I S Harris. (2021). Glutathione and its precursors in cancer. Current Opinion in Biotechnology, 68 : 292– 299
https://doi.org/10.1016/j.copbio.2021.03.001 pmid: 33819793
4 R Avanasi, H M Shin, V M Vieira, S M Bartell. (2016). Variability and epistemic uncertainty in water ingestion rates and pharmacokinetic parameters, and impact on the association between perfluorooctanoate and preeclampsia in the C8 Health Project population. Environmental Research, 146 : 299– 307
https://doi.org/10.1016/j.envres.2016.01.011 pmid: 26796985
5 P E Baldwin, J R Cain, R Fletcher, K Jones, N Warren ( 2007). Dehydroabietic acid as a biomarker for exposure to colophony. Occupational Medicine (Oxford, England), 57( 5): 362– 366
https://doi.org/10.1093/occmed/kqm034 pmid: 17548866
6 A C Behr, A Kwiatkowski, M Ståhlman, F F Schmidt, C Luckert, A Braeuning, T Buhrke. (2020). Impairment of bile acid metabolism by perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) in human HepaRG hepatoma cells. Archives of Toxicology, 94( 5): 1673– 1686
https://doi.org/10.1007/s00204-020-02732-3 pmid: 32253466
7 V Bisht, K Nash, Y Xu, P Agarwal, S Bosch, G V Gkoutos, A Acharjee. (2021). Integration of the microbiome, metabolome and transcriptomics data identified novel metabolic pathway regulation in colorectal cancer. International Journal of Molecular Sciences, 22( 11): 5763
https://doi.org/10.3390/ijms22115763 pmid: 34071236
8 Rosso M Cialiè, F Stilo, S Squara, E Liberto, S Mai, C Mele, P Marzullo, G Aimaretti, S E Reichenbach, M Collino, C Bicchi, C Cordero. (2021). Exploring extra dimensions to capture saliva metabolite fingerprints from metabolically healthy and unhealthy obese patients by comprehensive two-dimensional gas chromatography featuring Tandem Ionization mass spectrometry. Analytical and Bioanalytical Chemistry, 413( 2): 403– 418
https://doi.org/10.1007/s00216-020-03008-6 pmid: 33140127
9 R Cluett, S M Seshasayee, L B Rokoff, S L Rifas-Shiman, X Ye, A M Calafat, D R Gold, B Coull, C M Gordon, C J Rosen, E Oken, S K Sagiv, A F Fleisch. (2019). Per- and polyfluoroalkyl substance plasma concentrations and bone mineral density in midchildhood: a cross-sectional study (Project Viva, United States). Environmental Health Perspectives, 127( 8): 087006
https://doi.org/10.1289/EHP4918 pmid: 31433236
10 X Feng, X Chen, Y Yang, L Yang, Y Zhu, G Shan, L Zhu, S Zhang. (2021). External and internal human exposure to PFOA and HFPOs around a mega fluorochemical industrial park, China: differences and implications. Environment International, 157 : 106824
https://doi.org/10.1016/j.envint.2021.106824 pmid: 34411760
11 S E Fenton, A Ducatman, A Boobis, J C DeWitt, C Lau, C Ng, J S Smith, S M Roberts. (2021). Per- and polyfluoroalkyl substance toxicity and human health review: current state of knowledge and strategies for informing future research. Environmental Toxicology and Chemistry, 40( 3): 606– 630
https://doi.org/10.1002/etc.4890 pmid: 33017053
12 K Y Gebreab, M N H Eeza, T Bai, Z Zuberi, J Matysik, K E O'shea, A Alia, J P Berry ( 2020). Comparative toxicometabolomics of perfluorooctanoic acid (PFOA) and next-generation perfluoroalkyl substances. Environmental Pollution, 265(Pt A): 114928
13 I Gyllenhammar, J P Benskin, O Sandblom, U Berger, L Ahrens, S Lignell, K Wiberg, A Glynn. (2018). Perfluoroalkyl acids (PFAAs) in serum from 2–4-month-old infants: influence of maternal serum concentration, gestational age, breast-feeding, and contaminated drinking water. Environmental Science & Technology, 52( 12): 7101– 7110
https://doi.org/10.1021/acs.est.8b00770 pmid: 29758986
14 T I Halldorsson, D Rytter, L S Haug, B H Bech, I Danielsen, G Becher, T B Henriksen, S F Olsen. (2012). Prenatal exposure to perfluorooctanoate and risk of overweight at 20 years of age: a prospective cohort study. Environmental Health Perspectives, 120( 5): 668– 673
https://doi.org/10.1289/ehp.1104034 pmid: 22306490
15 N Kudo, Y Kawashima. (2003). Toxicity and toxicokinetics of perfluorooctanoic acid in humans and animals. Journal of Toxicological Sciences, 28( 2): 49– 57
https://doi.org/10.2131/jts.28.49 pmid: 12820537
16 J Lam, E Koustas, P Sutton, P I Johnson, D S Atchley, S Sen, K A Robinson, D A Axelrad, T J Woodruff. (2014). The Navigation Guide−evidence-based medicine meets environmental health: integration of animal and human evidence for PFOA effects on fetal growth. Environmental Health Perspectives, 122( 10): 1040– 1051
https://doi.org/10.1289/ehp.1307923 pmid: 24968389
17 P D Lin, A Cardenas, R Hauser, D R Gold, K P Kleinman, M F Hivert, A F Fleisch, A M Calafat, T F Webster, E S Horton, E Oken. (2019). Per- and polyfluoroalkyl substances and blood lipid levels in pre-diabetic adults-longitudinal analysis of the diabetes prevention program outcomes study. Environment International, 129 : 343– 353
https://doi.org/10.1016/j.envint.2019.05.027 pmid: 31150976
18 D Lv, Y Gu, M Guo, P Hou, Y Li, R Wu. (2019). Perfluorooctanoic acid exposure induces apoptosis in SMMC-7721 hepatocellular cancer cells. Environmental Pollution, 247 : 509– 514
https://doi.org/10.1016/j.envpol.2019.01.035 pmid: 30703684
19 A Podder, A H M A Sadmani, D Reinhart, N B Chang, R Goel. (2021). Per and poly-fluoroalkyl substances (PFAS) as a contaminant of emerging concern in surface water: A transboundary review of their occurrences and toxicity effects. Journal of Hazardous Materials, 419 : 126361
https://doi.org/10.1016/j.jhazmat.2021.126361 pmid: 34157464
20 X Qiu, J Ma, P Li, Z Geng, C Sun, D Wang, W Xu. (2020). Development of indirect competitive ELISA for determination of dehydroabietic acid in duck skin and comparison with the HPLC method. Poultry Science, 99( 6): 3280– 3285
https://doi.org/10.1016/j.psj.2020.01.031 pmid: 32475463
21 S Salihovic, T Fall, A Ganna, C D Broeckling, J E Prenni, T Hyötyläinen, A Kärrman, P M Lind, E Ingelsson, L Lind. (2019). Identification of metabolic profiles associated with human exposure to perfluoroalkyl substances. Journal of Exposure Science & Environmental Epidemiology, 29( 2): 196– 205
https://doi.org/10.1038/s41370-018-0060-y pmid: 30185940
22 D A Savitz, C R Stein, S M Bartell, B Elston, J Gong, H M Shin, G A Wellenius ( 2012). Perfluorooctanoic acid exposure and pregnancy outcome in a highly exposed community. Epidemiology (Cambridge, Mass.), 23( 3): 386– 392
https://doi.org/10.1097/EDE.0b013e31824cb93b pmid: 22370857
23 K Steenland, T Fletcher, C R Stein, S M Bartell, L Darrow, M J Lopez-Espinosa, P Barry Ryan, D A Savitz. (2020). Review: evolution of evidence on PFOA and health following the assessments of the C8 Science Panel. Environment International, 145 : 106125
https://doi.org/10.1016/j.envint.2020.106125 pmid: 32950793
24 B N VanNoy, J Lam, A R Zota. (2018). Breastfeeding as a predictor of serum concentrations of per- and polyfluorinated alkyl substances in reproductive-aged women and young children: a rapid systematic review. Current Environmental Health Reports, 5( 2): 213– 224
https://doi.org/10.1007/s40572-018-0194-z pmid: 29737463
25 J Wang, J Zhang, Y Fan, Z Li, C Tao, W Yan, R Niu, Y Huang, Q Xu, X Wang, Q Xu, L Han, C Lu. (2022). Association between per- and polyfluoroalkyl substances and risk of gestational diabetes mellitus. International Journal of Hygiene and Environmental Health, 240 : 113904
https://doi.org/10.1016/j.ijheh.2021.113904 pmid: 34915280
26 S Wikström, P I Lin, C H Lindh, H Shu, C G Bornehag. (2020). Maternal serum levels of perfluoroalkyl substances in early pregnancy and offspring birth weight. Pediatric Research, 87( 6): 1093– 1099
https://doi.org/10.1038/s41390-019-0720-1 pmid: 31835271
27 T Xu Y Guo J Lu J Shan L Lin W Qian W Chen J Wang X Lv M Ke, et al.. (2021). Untargeted serum metabolomics and potential biomarkers for Sjogren’s syndrome. Clinical and Experimental Rheumatology, 39 133 (Suppl 6): 23– 29
28 W Zeng, K E Huang, Y Luo, D X Li, W Chen, X Q Yu, X H Ke. (2020). Nontargeted urine metabolomics analysis of the protective and therapeutic effects of Citri Reticulatae Chachiensis Pericarpium on high-fat feed-induced hyperlipidemia in rats. Biomedical Chromatography, 34( 4): e4795
https://doi.org/10.1002/bmc.4795 pmid: 31967660
29 H Zhang, J He, N Li, N Gao, Q Du, B Chen, F Chen, X Shan, Y Ding, W Zhu, Y Wu, J Tang, X Jia. (2019). Lipid accumulation responses in the liver of Rana nigromaculata induced by perfluorooctanoic acid (PFOA). Ecotoxicology and Environmental Safety, 167 : 29– 35
https://doi.org/10.1016/j.ecoenv.2018.09.120 pmid: 30292973
30 Y Zhang, S Beesoon, L Zhu, J W Martin. (2013). Biomonitoring of perfluoroalkyl acids in human urine and estimates of biological half-life. Environmental Science & Technology, 47( 18): 10619– 10627
https://doi.org/10.1021/es401905e pmid: 23980546
31 Y Zhang, L Zhang, J Bao, L Liu, X Wang. (2021). Perfluorooctanoic acid exposure in early pregnancy induces oxidative stress in mice uterus and liver. Environmental Science and Pollution Research International, 28( 46): 66355– 66365
https://doi.org/10.1007/s11356-021-15453-6 pmid: 34331232
[1] Ling Qi, Zhige Tian, Nan Jiang, Fangyuan Zheng, Yuchen Zhao, Yishuo Geng, Xiaoli Duan. Collaborative control of fine particles and ozone required in China for health benefit[J]. Front. Environ. Sci. Eng., 2023, 17(8): 92-.
[2] Yuhan Zhao, Xiaoping Kang, Xue Tian, Lulu Liu, Zemeng Zhao, Lili Luo, Lixin Tao, Xiangtong Liu, Xiaonan Wang, Xiuhua Guo, Juan Xia, Yanxia Luo. Long-term exposure to air pollution and cerebrovascular disease: findings from Beijing Health Management Cohort study[J]. Front. Environ. Sci. Eng., 2023, 17(7): 84-.
[3] Jianxun Yang, Qi Gao, Miaomiao Liu, John S. Ji, Jun Bi. Same stimuli, different responses: a pilot study assessing air pollution visibility impacts on emotional well-being in a controlled environment[J]. Front. Environ. Sci. Eng., 2023, 17(2): 20-.
[4] Haoduo Zhao, Min Liu, Junjie Yang, Yuyang Chen, Mingliang Fang. Time-dependent metabolomics uncover dynamic metabolic adaptions in MCF-7 cells exposed to bisphenol A[J]. Front. Environ. Sci. Eng., 2023, 17(1): 4-.
[5] Xianjun Tan, Zhenying Jiang, Yuxiong Huang. Photo-induced surface frustrated Lewis pairs for promoted photocatalytic decomposition of perfluorooctanoic acid[J]. Front. Environ. Sci. Eng., 2023, 17(1): 3-.
[6] Mengtian Li, Ge Song, Ruiping Liu, Xia Huang, Huijuan Liu. Inactivation and risk control of pathogenic microorganisms in municipal sludge treatment: A review[J]. Front. Environ. Sci. Eng., 2022, 16(6): 70-.
[7] Jiuli Yang, Mingyang Liu, Qu Cheng, Lingyue Yang, Xiaohui Sun, Haidong Kan, Yang Liu, Michelle L. Bell, Rohini Dasan, Huiwang Gao, Xiaohong Yao, Yang Gao. Investigating the impact of air pollution on AMI and COPD hospital admissions in the coastal city of Qingdao, China[J]. Front. Environ. Sci. Eng., 2022, 16(5): 56-.
[8] Elizabeth Eastman, Kelly A. Stevens, Cesunica Ivey, Haofei Yu. On the potential of iPhone significant location data to characterize individual mobility for air pollution health studies[J]. Front. Environ. Sci. Eng., 2022, 16(5): 63-.
[9] Aifang Gao, Junyi Wang, Jianfei Luo, Aiguo Li, Kaiyu Chen, Pengfei Wang, Yiyi Wang, Jingyi Li, Jianlin Hu, Hongliang Zhang. Temporal variation of PM2.5-associated health effects in Shijiazhuang, Hebei[J]. Front. Environ. Sci. Eng., 2021, 15(5): 82-.
[10] Weichuan Qiao, Rong Li, Tianhao Tang, Achuo Anitta Zuh. Removal, distribution and plant uptake of perfluorooctane sulfonate (PFOS) in a simulated constructed wetland system[J]. Front. Environ. Sci. Eng., 2021, 15(2): 20-.
[11] Yingdan Zhang, Na Liu, Wei Wang, Jianteng Sun, Lizhong Zhu. Photosynthesis and related metabolic mechanism of promoted rice (Oryza sativa L.) growth by TiO2 nanoparticles[J]. Front. Environ. Sci. Eng., 2020, 14(6): 103-.
[12] Cong Liu, Yinping Zhang. Relations between indoor and outdoor PM2.5 and constituent concentrations[J]. Front. Environ. Sci. Eng., 2019, 13(1): 5-.
[13] Weiqi Luo, Yanping Ji, Lu Qu, Zhi Dang, Yingying Xie, Chengfang Yang, Xueqin Tao, Jianmin Zhou, Guining Lu. Effects of eggshell addition on calcium-deficient acid soils contaminated with heavy metals[J]. Front. Environ. Sci. Eng., 2018, 12(3): 4-.
[14] Ling-Li Li, Yin-Hua Cui, Jie-Jie Chen, Han-Qing Yu. Roles of glutathione and L-cysteine in the biomimetic green synthesis of CdSe quantum dots[J]. Front. Environ. Sci. Eng., 2017, 11(6): 7-.
[15] Jingjing PENG, Hong LI, Jianqiang SU, Qiufang ZHANG, Junpeng RUI, Chao CAI. Response of bacterial communities to short-term pyrene exposure in red soil[J]. Front Envir Sci Eng, 2013, 7(4): 559-567.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed