Application of the Key Characteristics of Carcinogens to Per and Polyfluoroalkyl Substances
Abstract
:1. Introduction
2. Existing Research on PFAS Exposure and Cancer Risk
3. Literature Search for PFAS and Key Characteristics of Carcinogens
4. Strength of Evidence Assessment
5. Analysis of Individual Key Characteristics
5.1. Is Electrophilic or Can Be Metabolically Activated
5.2. Is Genotoxic
5.3. Alters DNA Repair or Causes Genomic Instability
5.4. Induces Epigenetic Alterations
5.5. Induces Oxidative Stress
5.6. Induces Chronic Inflammation
5.7. Is Immunosuppressive
5.8. Modulates Receptor-Mediated Effects
5.9. Causes Immortalization
5.10. Alters Cell Proliferation, Cell Death or Nutrient Supply
6. Discussion
7. Conclusions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- International Agency for Research on Cancer. Some Chemicals Used as Solvents and in Polymer Manufacture IARC Monographs on the Evaluation of Carcinogenic Risks to Humans Volume 110; International Agency for Research on Cancer: Lyon, France, 2016. [Google Scholar]
- Environmental Protection Agency. Drinking Water Health Advisory for Perfluorooctanoic Acid (PFOA); EPA Document Number: 822-R-16-005; US Environmental Protection Agency Office of Water (4304T) Health and Ecological Criteria Division: Washington, DC, USA, 2016.
- Environmental Protection Agency. Drinking Water Health Advisory for Perfluorooctane Sulfonate (PFOS); EPA Document Number: 822-R-16-004; US Environmental Protection Agency Office of Water (4304T) Health and Ecological Criteria Division: Washington, DC, USA, 2016.
- Environmental Protection Agency. Human Health Toxicity Values for Hexafluoropropylene Oxide (HFPO) Dimer Acid and Its Ammonium Salt (CASRN 13252-13-6 and CASRN 62037-80-3) Also Known as “GenX Chemicals”; EPA Document Number: 823-P-18-001; US Environmental Protection Agency Office of Water (4304T) Health and Ecological Criteria Division: Washington, DC, USA, 2018.
- Guyton, K.Z.; Rusyn, I.; Chiu, W.A.; Corpet, D.E.; van den Berg, M.; Ross, M.K.; Christiani, D.C.; Beland, F.A.; Smith, M.T. Application of the key characteristics of carcinogens in cancer hazard identification. Carcinogenesis 2018, 39, 614–622. [Google Scholar] [CrossRef] [PubMed]
- Smith, M.T.; Guyton, K.Z.; Gibbons, C.F.; Fritz, J.M.; Portier, C.J.; Rusyn, I.; DeMarini, D.M.; Caldwell, J.C.; Kavlock, R.J.; Lambert, P.F.; et al. Key characteristics of carcinogens as a basis for organizing data on mechanisms of carcinogenesis. Environ. Health Perspect. 2016, 124, 713–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atwood, S.T.; Lunn, R.M.; Garner, S.C.; Jahnke, G.D. New Perspectives for Cancer Hazard Evaluation by the Report on Carcinogens: A Case Study Using Read-Across Methods in the Evaluation of Haloacetic Acids Found as Water Disinfection By-Products. Environ. Health Perspect. 2019, 127, 125003. [Google Scholar] [CrossRef] [PubMed]
- International Agency for Research on Cancer. IARC Monographs on the Identification of Carcinogenic Hazard to Humans Preamble; International Agency for Research on Cancer: Lyon, France, 2019. [Google Scholar]
- Environmental Protection Agency. Systematic Review Protocol for the Hexavalent Chromium IRIS Assessment (Preliminary Assessment Materials); US Environmental Protection Agency: Washington, DC, USA, 2019.
- Office of Environmental Health Hazard Assessment. Evidence on the Carcinogenicity of Gentian Violet; California Environmental Protection Agency: Sacramento, CA, USA, 2019.
- IARC; Monographs Priorities Group. Advisory Group recommendations on priorities for the IARC Monographs. Lancet Oncol. 2019, 20, 763–764. [Google Scholar] [CrossRef]
- Olsen, G.W.; Mair, D.C.; Lange, C.C.; Harrington, L.M.; Church, T.R.; Goldberg, C.L.; Herron, R.M.; Hanna, H.; Nobiletti, J.B.; Rios, J.A.; et al. Per-and polyfluoralkyl substances (PFAS) in American Red Cross adult blood donors, 2000–2015. Environ. Res. 2017, 157, 87–95. [Google Scholar] [CrossRef] [PubMed]
- Hurley, S.; Goldberg, D.; Wang, M.; Parks, J.S.; Petreas, M.; Bernstein, L.; Anton-Culver, H.; Nelson, D.O.; Reynolds, P. Time trends in per- and polyfluoroalkyl substances (PFASs) in California women: Declining serum levels, 2011–2015. Environ. Sci. Technol. 2018, 52, 277–287. [Google Scholar] [CrossRef]
- Yu, C.H.; Riker, C.D.; Lu, S.E.; Fan, Z.T. Biomonitoring of emerging contaminants, perfluoroalkyl and polyfluoroalkyl substances (PFAS), in New Jersey adults in 2016–2018. Int. J. Hyg. Environ. Health 2020, 223, 34–44. [Google Scholar] [CrossRef]
- Hu, X.C.; Dassuncao, C.; Zhang, X.; Grandjean, P.; Weihe, P.; Webster, G.M.; Nielsen, F.; Sunderland, E.M. Can profiles of poly-and perfluoroalkyl substances (PFASs) in human serum provide information on major exposure sources? Environ. Health 2018, 17, 11. [Google Scholar] [CrossRef] [Green Version]
- Morck, T.A.; Nielsen, F.; Nielsen, J.K.; Siersma, V.D.; Grandjean, P.; Knudsen, L.E. PFAS concentrations in plasma samples from Danish school children and their mothers. Chemosphere 2015, 129, 203–209. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention. Fourth Report on Human Exposure to Environmental Chemicals, Updated Tables, (January 2019); US Department of Health and Human Services, Centers for Disease Control and Prevention: Atlanta, GA, USA, 2019.
- Calafat, A.M.; Kato, K.; Hubbard, K.; Jia, T.; Botelho, J.C.; Wong, L.Y. Legacy and alternative per- and polyfluoroalkyl substances in the U.S. general population: Paired serum-urine data from the 2013–2014 National Health and Nutrition Examination Survey. Environ. Int. 2019, 131, 105048. [Google Scholar] [CrossRef]
- Sunderland, E.M.; Hu, X.C.; Dassuncao, C.; Tokranov, A.K.; Wagner, C.C.; Allen, J.G. A review of the pathways of human exposure to poly- and perfluoroalkyl substances (PFASs) and present understanding of health effects. J. Exp. Sci. Environ. Epidemiol. 2019, 29, 131–147. [Google Scholar] [CrossRef] [Green Version]
- Boronow, K.E.; Brody, J.G.; Schaider, L.A.; Peaslee, G.F.; Havas, L.; Cohn, B.A. Serum concentrations of PFASs and exposure-related behaviors in African American and non-Hispanic white women. J. Exp. Sci. Environ. Epidemiol. 2019, 29, 206–217. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.C.; Andrews, D.Q.; Lindstrom, A.B.; Bruton, T.A.; Schaider, L.A.; Grandjean, P.; Lohmann, R.; Carigan, C.C.; Blum, A.; Balan, S.A.; et al. Detection of poly- and perfluoroalkyl substances (PFASs) in U.S. drinking water linked to industrial sites, military fire training areas, and wastewater treatment plants. Environ. Sci. Technol. Lett. 2016, 3, 344–350. [Google Scholar] [CrossRef] [PubMed]
- Emmett, E.A.; Shofer, F.S.; Zhang, H.; GFreeman, D.; Desai, C.; Shaw, L.M. Community exposure to perfluorooctanoate: Relationships between serum concentrations and exposure source. J. Occup. Environ. Med. 2006, 48, 759–770. [Google Scholar] [CrossRef] [PubMed]
- Ingelido, A.M.; Abballe, A.; Gemma, S.; Dellatte, E.; Iacovella, N.; De Angelis, G.; Zampaglioni, F.; Marra, V.; Miniero, R.; Valentini, S.; et al. Biomonitoring of perfluorinated compounds in adults exposed to contaminated drinking water in the Veneto Region, Italy. Environ. Int. 2018, 110, 149–159. [Google Scholar] [CrossRef] [PubMed]
- Jackson, E.; Shoemaker, R.; Larian, N.; Cassis, L. Adipose tissue as a site of toxin accumulation. Compr. Physiol. 2017, 7, 1085–1135. [Google Scholar] [PubMed] [Green Version]
- Kamendulis, L.M.; Wu, Q.; Sandusky, G.E.; Hocevar, B.A. Perfluorooctanoic acid exposure triggers oxidative stress in the mouse pancreas. Toxicol. Rep. 2014, 1, 513–521. [Google Scholar] [CrossRef] [Green Version]
- Perez, F.; Nadal, M.; Navarro-Ortega, A.; Fabrega, F.; Domingo, J.L.; Barcelo, D.; Farré, M. Accumulation of perfluoroalkyl substances in human tissues. Environ. Int. 2013, 59, 354–362. [Google Scholar] [CrossRef]
- Cariou, R.; Veyrand, B.; Yamada, A.; Berrebi, A.; Zalko, D.; Durand, S.; Pollono, C.; Marchand, P.; Leblanc, J.C.; Antignac, J.P.; et al. Perfluoroalkyl acid (PFAA) levels and profiles in breast milk, maternal and cord serum of French women and their newborns. Environ. Int. 2015, 84, 71–81. [Google Scholar] [CrossRef]
- Mamsen, L.S.; Bjorvang, R.D.; Mucs, D.; Vinnars, M.T.; Papadogiannakis, N.; Lindh, C.H.; Andersen, C.Y.; Damdimopoulou, P. Concentrations of perfluoroalkyl substances (PFASs) in human embryonic and fetal organs from first, second, and third trimester pregnancies. Environ. Int. 2019, 124, 482–492. [Google Scholar] [CrossRef] [PubMed]
- McCord, J.; Strynar, M. Identification of per- and polyfluoroalkyl substances in the Cape Fear River by high resolution mass spectrometry and nontargeted screening. Environ. Sci. Technol. 2019, 53, 4717–4727. [Google Scholar] [CrossRef] [PubMed]
- Chang, E.T.; Adami, H.-O.; Boffetta, P.; Cole, P.; Starr, T.B.; Mandel, J.S. A critical review of perfluorooctanoate and perfluorooctanesulfonate exposure and cancer risk in humans. Crit. Rev. Toxicol. 2014, 44, 1–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodgers, K.M.; Udesky, J.O.; Rudel, R.A.; Brody, J.G. Environmental chemicals and breast cancer: An updated review of epidemiological literature informed by biological mechanisms. Environ. Res. 2018, 160, 152–182. [Google Scholar] [CrossRef]
- Stanifer, J.W.; Stapleton, H.M.; Souma, T.; Wittmer, A.; Zhao, X.; Boulware, L.E. Perfluorinated chemicals as emerging environmental threats to kidney health: A scoping review. Clin. J. Am. Soc. Nephrol. 2018, 13, 1479–1492. [Google Scholar] [CrossRef] [Green Version]
- Gilliland, F.D.; Mandel, J.S. Mortality among employees of a perfluorooctanoic acid production plant. J. Occup. Med. 1993, 35, 950–954. [Google Scholar] [CrossRef] [PubMed]
- Raleigh, K.K.; Alexander, B.H.; Olsen, G.W.; Ramachandran, G.; Morey, S.Z.; Church, T.R.; Logan, P.W.; Scott, L.L.; Allen, E.M. Mortality and cancer incidence in ammonium perfluorooctanoate production workers. Occup. Environ. Med. 2014, 71, 500–506. [Google Scholar] [CrossRef] [Green Version]
- Lundin, J.I.; Alexander, B.H.; Olsen, G.W.; Church, T.R. Ammonium perfluooctanoate production and occupational mortality. Epidemiology 2009, 20, 921–928. [Google Scholar] [CrossRef]
- Girardi, P.; Merler, E. A mortality study on male subjects exposed to polyfluoroalkyl acids with high internal dose of perfluorooctanoic acid. Environ. Res. 2019, 179, 108743. [Google Scholar] [CrossRef]
- Barry, V.; Winquist, A.; Steenland, K. Perfluorooctanoic acid (PFOA) exposures and incident cancers among adults living near a chemical plant. Environ. Health Perspect. 2013, 121, 1313–1318. [Google Scholar] [CrossRef] [Green Version]
- Eriksen, K.T.; Sorensen, M.; McLaughlin, J.K.; Lipworth, L.; Tjonneland, A.; Overvad, K.; Raaschou-Nielsen, O. Perfluooctanoate and perfluooctanesulfanate plasma levels and risk of cancer in the general Danish population. J. Natl. Cancer Inst. 2009, 101, 605–609. [Google Scholar] [CrossRef]
- Bonefeld-Jorgensen, E.C.; Long, M.; Fredslund, S.O.; Bossi, R.; Olsen, J. Breast cancer risk after exposure to perfluorinated compounds in Danish women: A case-control study nested in the Danish National Birth Cohort. Cancer Causes Control 2014, 25, 1439–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonefeld-Jorgensen, E.C.; Long, M.; Bossi, R.; Ayotte, P.; Asmund, G.; Kruger, T.; Ghisari, M.; Mulvad, G.; Kern, P.; Nzulumiki, P.; et al. Perfluorinated compounds are related to breast cancer risk in Greenlandic Inuit: A case control study. Environ. Health 2011, 10, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- National Toxicology Program. NTP technical report on the toxicology and carcinogenesis studies of perfluorooctanoic acid (CAS no. 335-67-1) administered in feed to Sprague Dawley (Hsd:Sprague Dawley® SD®) rats. In Research Triangle; Technical Report 598; National Toxicology Program: Park, NC, USA, 2019. [Google Scholar]
- Butenhoff, J.L.; Kennedy, G.L.; Chang, S.C., Jr.; Olsen, G.W. Chronic dietary toxicity and carcinogenicity study with ammonium perfluorooctanoate in Sprague-Dawley rats. Toxicology 2012, 298, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Butenhoff, J.L.; Chang, S.C.; Olsen, G.W.; Thomford, P.J. Chronic dietary toxicity and carcinogenicity study with potassium perfluorooctanesulfonate in Sprague Dawley rats. Toxicology 2012, 293, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Klaunig, J.E.; Shinohara, M.; Iwai, H.; Chengelis, C.P.; Kirkpatrick, J.B.; Wang, Z.; Bruner, R.H. Evaluation of the chronic toxicity and carcinogenicity of perfluorohexanoic acid (PFHxA) in Sprague-Dawley rats. Toxicol. Pathol. 2015, 43, 209–220. [Google Scholar] [CrossRef] [PubMed]
- Caverly Rae, J.M.; Craig, L.; Slone, T.W.; Frame, S.R.; Buxton, L.W.; Kennedy, G.L. Evaluation of chronic toxicity and carcinogenicity of ammonium 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate in Sprague-Dawley rats. Toxicol. Rep. 2015, 2, 939–949. [Google Scholar] [CrossRef] [Green Version]
- Gomis, M.I.; Vestergren, R.; Borg, D.; Cousins, I.T. Comparing the toxic potency in vivo of long-chain perfluoroalkyl acids and fluorinated alternatives. Environ. Int. 2018, 113, 1–9. [Google Scholar] [CrossRef]
- Agency for Toxic Substances and Disease Registry (ATSDR). Toxicological Profile for Perfluoroalkyls. (Draft for Public Comment); US Department of Health and Human Services, Public Health Service: Atlanta, GA, USA, 2018.
- Environmental Protection Agency. Human Health Toxicity Values for Perfluorobutane Sulfonic Acid (CASRN 375-73-5) and Related Compound Potassium Perfluorobutane Sulfonate (CASRN 29420-49-3); EPA Document Number: 823-R-18-307; US Environmental Protection Agency Office of Research and Development (8101R) National Center for Environmental Assessment: Washington, DC, USA, 2018.
- Li, Y.; Fletcher, T.; Mucs, D.; Scott, J.M.; Lindh, C.H.; Tallving, P.; Jakobsson, K. Half-lives of PFOS, PFHxS and PFOA after end of exposure to contaminated drinking water. Occup. Environ. Med. 2018, 75, 46–51. [Google Scholar] [CrossRef] [Green Version]
- Olsen, G.W.; Burris, J.M.; Ehresman, D.J.; Froehlich, J.W.; Seacat, A.M.; Butenhoff, J.L.; Zobel, L.R. Half-life of serum elimination of perfluorooctanesulfonate, perfluorohexanesulfonate, and perfluorooctanoate in retired fluorochemical production workers. Environ. Health Perspect. 2007, 115, 1298–1305. [Google Scholar] [CrossRef]
- Olsen, G.W.; Chang, S.-C.; Noker, P.E.; Gorman, G.S.; Ehresman, D.J.; Lieder, P.H.; Butenhoff, J.L. A comparison of the pharmacokinetics of perfluorobutanesulfonate (PFBS) in rats, monkeys, and humans. Toxicology 2009, 256, 65–74. [Google Scholar] [CrossRef]
- Gannon, S.A.; Fasano, W.J.; Mawn, M.P.; Nabb, D.L.; Buck, R.C.; Buxton, L.W.; Jepson, G.W.; Frame, S.R. Absorption, distribution, metabolism, excretion, and kinetics of 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)propanoic acid ammonium salt following a single dose in rat, mouse, and cynomolgus monkey. Toxicology 2016, 340, 1–9. [Google Scholar] [CrossRef] [PubMed]
- D’Eon, J.C.; Crozier, P.W.; Furdui, V.I.; Reiner, E.J.; Libelo, E.L.; Mabury, S.A. Observation of a commercial fluorinated material, the polyfluoroalkyl phosphoric acid diesters, in human sera, wastewater treatment plant sludge, and paper fibers. Environ. Sci. Technol. 2009, 43, 4589–4594. [Google Scholar] [CrossRef] [PubMed]
- Butt, C.M.; Muir, D.C.G.; Mabury, S.A. Biotransformation pathways of fluorotelomer-based polyfluoroalkyl substances: A review. Environ. Toxicol. Chem. 2014, 33, 243–267. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.C.; Robinson, V.G.; Waidyanatha, S.; Dzierlenga, A.L.; DeVito, M.J.; Eifrid, M.A.; Gibbs, S.T.; Blystone, C.R. Toxicokinetics of 8:2 fluorotelomer alcohol (8:2-FTOH) in male and female Hsd:Sprague Dawley SD rats after intravenous and gavage administration. Toxicol. Rep. 2019, 6, 924–932. [Google Scholar] [CrossRef]
- D’Eon, J.C.; Mabury, S.A. Exploring indirect sources of human exposure to perfluoroalkyl carboxylates (PFCAs): Evaluating uptake, elimination, and biotransformation of polyfluoroalkyl phosphate esters (PAPs) in the rat. Environ. Health Perspect. 2011, 119, 344–350. [Google Scholar] [CrossRef]
- Kabadi, S.V.; Fisher, J.; Aungst, J.; Rice, P. Internal exposure-based pharmacokinetic evaluation of potential for biopersistence of 6:2 fluorotelomer alcohol (FTOH) and its metabolites. Food Chem. Toxicol. 2018, 112, 375–382. [Google Scholar] [CrossRef]
- Cui, L.; Liao, C.Y.; Zhou, Q.F.; Xia, T.M.; Yun, Z.J.; Jiang, G.B. Excretion of PFOA and PFOS in male rats during a subchronic exposure. Arch. Environ. Contam Toxicol. 2010, 58, 205–213. [Google Scholar] [CrossRef]
- Crebelli, R.; Caiola, S.; Conti, L.; Cordelli, E.; De Luca, G.; Dellatte, E.; Eleuteri, P.; Iacovella, N.; Leopardi, P.; Marcon, F.; et al. Can sustained exposure to PFAS trigger a genotoxic response? A comprehensive genotoxicity assessment in mice after subacute oral administration of PFOA and PFBA. Regul Toxicol. Pharmacol. 2019, 106, 169–177. [Google Scholar] [CrossRef]
- Emerce, E.; Cetin, O. Genotoxicity assessment of perfluoroalkyl substances on human sperm. Toxicol. Ind. Health 2018. [Google Scholar] [CrossRef]
- National Toxicology Program. NTP technical report on the toxicity studies of perfluoroalkyl carboxylates (perfluorohexanoic acid, perfluorooctanoic acid, perfluorononanoic acid, and perfluorodecanoic acid) administered by gavage to Sprague Dawley (Hsd:Sprague Dawley SD) rats. In Research Triangle; Toxicity Report 97; National Toxicology Program: Park, NC, USA, 2019. [Google Scholar]
- National Toxicology Program. NTP Technical Report on the Toxicity Studies of Perfluoroalkyl Sulfonates (Perfluorobutane Sulfonic Acid, Perfluorohexane Sulfonate Potassium Salt, and Perfluorooctane Sulfonic Acid) Administered by Gavage to Sprague Dawley (Hsd:Sprague Dawley SD) Rats; Toxicity Report 96; National Toxicology Program Public Health Service, U.S. Department of Health and Human Services: Research Triangle Park, NC, USA, 2019.
- Luch, A.; Frey, F.C.; Meier, R.; Fei, J.; Naegeli, H. Low-dose formaldehyde delays DNA damage recognition and DNA excision repair in human cells. PLoS ONE 2014, 9, e94149. [Google Scholar] [CrossRef] [Green Version]
- Martin, E.M.; Fry, R.C. Environmental influences on the epigenome: Exposure-associated DNA methylation in human populations. Annu. Rev. Public Health 2018, 39, 309–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bouras, E.; Karakioulaki, M.; Bougioukas, K.I.; Aivaliotis, M.; Tzimagiorgis, G.; Chourdakis, M. Gene promoter methylation and cancer: An umbrella review. Gene 2019, 710, 333–340. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Huang, Q.; Zeng, F.; Li, Q.; He, Z.; Chen, W.; Zhu, W.; Zhang, B. The prognostic value of global DNA hypomethylation in cancer: A meta-analysis. PLoS ONE 2014, 9, e106290. [Google Scholar] [CrossRef]
- Fardi, M.; Solali, S.; Farshdousti Hagh, M. Epigenetic mechanisms as a new approach in cancer treatment: An updated review. Genes Dis. 2018, 5, 304–311. [Google Scholar] [CrossRef]
- Herceg, A.; Lambert, M.P.; van Veldhoven, K.; Demetriou, C.; Vineis, P.; Smith, M.T.; Straif, K.; Wild, C.P. Towards incorporating epigenetic mechanisms into carcinogen identification and evaluation. Carcinogenesis 2013, 34, 1955–1967. [Google Scholar] [CrossRef] [Green Version]
- Guerrero-Preston, R.; Goldman, L.R.; Brebi-Mieville, P.; Ili-Gangas, C.; Lebron, C.; Witter, F.R.; Apelber, B.J.; Hernández-Roystacher, M.; Jaffe, A.; Halden, R.U.; et al. Global DNA hypomethylation is associated with in utero exposure to cotinine and perfluorinated alkyl compounds. Epigenetics 2010, 5, 539–546. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.Y.; Chen, P.C.; Lien, P.C.; Liao, Y.P. Prenatal perfluorooctyl sulfonate exposure and Alu DNA hypomethylation in cord blood. Int. J. Environ. Res. Public Health 2018, 15, 1066. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, S.; Azumi, K.; Goudarzi, H.; Araki, A.; Miyashita, C.; Kobayashi, S.; Itoh, S.; Sasaki, S.; Ishizuka, M.; Nakazawa, H.; et al. Effects of prenatal perfluoroalkyl acid exposure on cord blood IGF2/H19 methylation and ponderal index: The Hokkaido Study. J. Exp. Sci. Environ. Epidemiol. 2017, 27, 251–259. [Google Scholar] [CrossRef] [Green Version]
- Miura, R.; Araki, A.; Miyashita, C.; Kobayashi, S.; Kobayashi, S.; Wang, S.L.; Chen, C.H.; Miyake, K.; Ishizuka, Y.; Iwasaki, Y.; et al. An epigenome-wide study of cord blood DNA methylations in relation to prenatal perfluoroalkyl substance exposure: The Hokkaido study. Environ. Int. 2018, 115, 21–28. [Google Scholar] [CrossRef]
- Kingsley, S.L.; Kelsey, K.T.; Butler, R.; Chen, A.; Eliot, M.N.; Romano, M.E.; Houseman, A.; Koestler, D.C.; Lanphear, B.P.; Yolton, K.; et al. Maternal serum PFOA concentration and DNA methylation in cord blood: A pilot study. Environ. Res. 2017, 158, 174–178. [Google Scholar] [CrossRef]
- Leung, Y.K.; Ouyang, B.; Niu, L.; ZXie, C.; Ying, J.; Medvedovic, M.; Chen, A.; Weihe, P.; Valvi, D.; Grandjean, P.; et al. Identification of sex-specific DNA methylation changes driven by specific chemicals in cord blood in a Faroese birth cohort. Epigenetics 2018, 13, 290–300. [Google Scholar] [CrossRef]
- Leter, G.; Consales, C.; Eleuteri, P.; Uccelli, R.; Specht, I.O.; Toft, G.; Moccia, T.; Budillon, A.; Jönsson, B.A.; Lindh, C.H.; et al. Exposure to perfluoroalkyl substances and sperm DNA global methylation in Arctic and European populations. Environ. Mol. Mutagen. 2014, 55, 591–600. [Google Scholar] [CrossRef]
- Wan, Y.J.; Li, Y.Y.; Xia, W.; Chen, J.; Lv, Z.Q.; Zeng, H.C.; Zhang, L.; Yang, W.J.; Chen, T.; Lin, Y.; et al. Alterations in tumor biomarker GSTP gene methylation patterns induced by prenatal exposure to PFOS. Toxicology 2010, 274, 57–64. [Google Scholar] [CrossRef]
- Tian, M.; Peng, S.; Martin, F.L.; Zhang, J.; Liu, L.; Wang, Z.; Dong, S.; Shen, H. Perfluorooctanoic acid induces gene promoter hypermethylation glutathione-S-transferase Pi in human liver L02 cells. Toxicology 2012, 296, 48–55. [Google Scholar] [CrossRef]
- Ma, Y.; Yang, J.; Wan, Y.; Peng, Y.; Ding, S.; Li, Y.; Xu, B.; Chen, X.; Xia, W.; Ke, Y.; et al. Low-level perfluorooctanoic acid enhances 3T3-L1 preadipocyte differentiation via altering peroxisome proliferator activated receptor gamma expression and its promoter DNA methylation. J. Appl. Toxicol. 2018, 38, 398–407. [Google Scholar] [CrossRef]
- Bastos Sales, L.; Kamstra, J.H.; Cenijn, P.H.; van Rijt, L.S.; Hamers, T.; Legler, J. Effects of endocrine disrupting chemicals on in vitro global DNA methylation and adipocyte differentiation. Toxicol. In Vitro 2013, 27, 1634–1643. [Google Scholar] [CrossRef]
- van den Dungen, M.W.; Murk, A.J.; Kok, D.E.; Steegenga, W.T. Persistent organic pollutants alter DNA methylation during human adipocyte differentiation. Toxicol. In Vitro 2017, 40, 79–87. [Google Scholar] [CrossRef]
- Watkins, D.J.; Wellenius, G.A.; Butler, R.A.; Bartell, S.M.; Fletcher, T.; Kelsey, K.T. Associations between serum perfluroalkyl acids and LINE-1 DNA methylation. Environ. Int. 2014, 63, 71–76. [Google Scholar] [CrossRef] [Green Version]
- Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [Green Version]
- Bundy, J.G.; Davey, M.P.; Viant, M.R. Environmental metabolomics: A critical review and future perspectives. Metabolomics 2009, 5, 3–21. [Google Scholar] [CrossRef]
- Wang, X.; Liu, L.; Zhang, W.; Zhang, J.; Du, X.; Huang, Q.; Tian, M.; Shen, H. Serum metabolome biomarkers associate low-level environmental perfluorinated compound exposure with oxidative/nitrosative stress in humans. Environ. Pollut. 2017, 229, 168–176. [Google Scholar] [CrossRef]
- Eriksen, K.T.; Raaschou-Nielsen, O.; Sørensen, M.; Roursgaard, M.; Loft, S.; Møller, P. Genotoxic potential of the perfluorinated chemicals PFOA, PFOS, PFBS, PFNA and PFHxA in human HepG2 cells. Mutat. Res. 2010, 700, 39–43. [Google Scholar] [CrossRef]
- Yao, X.; Zhong, L. Genotoxic risk and oxidative DNA damage in HepG2 cells exposed to perfluooctanoic acid. Mutat. Res. 2005, 587, 38–44. [Google Scholar] [CrossRef]
- Rigden, M.; Pelletier, G.; Poon, R.; Zhu, J.; Auray-Blais, C.; Gagnon, R.; Kubwabo, C.; Kosarac, I.; Lalonde, K.; Cakmak, S.; et al. Assessment of urinary metabolite excretion after rat acute exposure to perfluorooctanoic acid and other peroxisomal proliferators. Arch. Environ. Contam. Toxicol. 2015, 68, 148–158. [Google Scholar] [CrossRef]
- Panaretakis, T.; Shabalina, I.G.; Grandér, D.; Shoshan, M.C.; DePierre, J.W. Reactive oxygen species and mitochondria mediate the induction of apoptosis in human hepatoma HepG2 cells by the rodent peroxisome proliferator and hepatocarcinogen, perfluorooctanoic acid. Toxicol. Appl. Pharmacol. 2001, 173, 56–64. [Google Scholar] [CrossRef]
- Singh, T.S.; Lee, S.; Kim, H.H.; Choi, J.K.; Kim, S.H. Perfluorooctanoic acid induces mast cell-mediated allergic inflammation by the release of histamine and inflammatory mediators. Toxicol. Lett. 2012, 210, 64–70. [Google Scholar] [CrossRef]
- Liu, W.; Yang, B.; Wu, L.; Zou, W.; Pan, X.; Zou, T.; Liu, F.; Xia, L.; Wang, X.; Zhang, D. Involvement of NRF2 in perfluorooctanoic acid-induced testicular damage in male mice. Biol. Reprod. 2015, 93, 41. [Google Scholar] [CrossRef]
- Suh, K.S.; Choi, E.M.; Kim, Y.J.; Hong, S.M.; Park, S.Y.; Rhee, S.Y.; Oh, S.; Kin, S.W.; Pak, Y.K.; Choe, W.; et al. Perfluorooctanoic acid induces oxidative damage and mitochondrial dysfunction in pancreatic β-cells. Mol. Med. Rep. 2017, 15, 3871–3878. [Google Scholar] [CrossRef] [Green Version]
- Yuan, Y.; Ge, S.; Lv, Z.; Wu, M.; Kuang, H.; Yang, B.; Yang, J.; Wu, L.; Zou, W.; Zhang, D. Attenuation of perfluorooctanoic acid-induced testicular oxidative stress and apoptosis by quercetin in mice. RSC Adv. 2017, 7, 45045–45052. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Arellano, P.; Lopez-Arellano, K.; Luna, J.; Flores, D.; Jimenez-Salazar, J.; Gavia, G.; Teteltitla, N.; Rodríguez, J.J.; Domínguez, A.; Casas, E.; et al. Perfluorooctanoic acid disrupts gap junction intercellular communication and induces reactive oxygen species formation and apoptosis in mouse ovaries. Environ. Toxicol. 2019, 34, 92–98. [Google Scholar] [CrossRef] [Green Version]
- Han, R.; Hu, M.; Zhong, Q.; Wan, C.; Liu, L.; Li, F.; Zhang, F.; Ding, W. Perfluorooctane sulphonate induces oxidative hepatic damage via mitochondria-dependent and NF-κB/TNF-α-mediated pathway. Chemosphere 2018, 191, 1056–1064. [Google Scholar] [CrossRef]
- Khansari, M.R.; Yousefsani, B.S.; Kobarfard, F.; Faizi, M.; Pourahmad, J. In vitro toxicity of perfluorooctane sulfonate on rat liver hepatocytes: Probability of distructive binding to CYP 2E1 and involvement of cellular proteolysis. Environ. Sci. Pollut. Res. Int. 2017, 24, 23382–23388. [Google Scholar] [CrossRef]
- Zeng, Z.; Song, B.; Xiao, R.; Zeng, G.; Gong, J.; Chen, M.; Xu, P.; Zhang, P.; Shen, M.; Yi, H. Assessing the human health risks of perfluorooctane sulfonate by in vivo and in vitro studies. Environ. Int. 2019, 126, 598–610. [Google Scholar] [CrossRef]
- Singh, S.; Singh, S.K. Chronic exposure to perfluorononanoic acid impairs spermatogenesis, steroidogenesis and fertility in male mice. J. Appl. Toxicol. 2019, 39, 420–431. [Google Scholar] [CrossRef]
- Costantini, D.; Blevin, P.; Herzke, D.; Moe, B.; Bustnes, J.O.; Chastel, O. Higher plasma oxidative damage and lower plasma antioxidant defences in an Arctic seabird exposed to longer perfluoroalkyl acids. Environ. Res. 2019, 168, 278–285. [Google Scholar] [CrossRef]
- Pan, X.; Qin, P.; Liu, R.; Yu, W.; Dong, X. Effects of carbon chain length on the perfluoroalkyl acids-induced oxidative stress of erythrocytes in vitro. J. Agric. Food Chem. 2018, 66, 6414–6420. [Google Scholar] [CrossRef]
- Wielsoe, M.; Long, M.; Ghisari, M.; Bonefeld-Jorgensen, E.C. Perfluoroalkylated substances (PFAS) affect oxidative stress biomarkers in vitro. Chemosphere 2015, 129, 239–245. [Google Scholar] [CrossRef]
- Slotkin, T.A.; MacKillop, E.A.; Melnick, R.L.; Thayer, K.A.; Seidler, F.J. Developmental neurotoxicity of perfluorinated chemicals modeled in vitro. Environ. Health Perspect. 2008, 116, 716–722. [Google Scholar] [CrossRef] [Green Version]
- Thompson, P.A.; Khatami, M.; Baglole, C.J.; Sun, J.; Harris, S.A.; Moon, E.Y.; Al-Mulla, F.; Al-Temaimi, R.; Brown, D.G.; Colacci, A.; et al. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 2015, 36 (Suppl. 1), S232–S253. [Google Scholar] [CrossRef] [Green Version]
- Chang, E.T.; Adami, H.O.; Boffetta, P.; Wedner, H.J.; Mandel, J.S. A critical review of perfluorooctanoate and perfluorooctanesulfonate exposure and immunological health conditions in humans. Crit. Rev. Toxicol. 2016, 46, 279–331. [Google Scholar] [CrossRef] [PubMed]
- Steenland, K.; Kugathasan, S.; Barr, D.B. PFOA and ulcerative colitis. Environ. Res. 2018, 165, 317–321. [Google Scholar] [CrossRef] [PubMed]
- Steenland, K.; Zhao, L.; Winquist, A.; Parks, C. Ulcerative colitis and perfluorooctanoic acid (PFOA) in a highly exposed population of community residents and workers in the mid-Ohio valley. Environ. Health Perspect. 2013, 121, 900–905. [Google Scholar] [CrossRef] [PubMed]
- Innes, K.E.; Ducatman, A.M.; Luster, M.I.; Shankar, A. Association of osteoarthritis with serum levels of the environmental contaminants perfluorooctanoate and perfluorooctane sulfonate in a large Appalachian population. Am. J. Epidemiol. 2011, 174, 440–450. [Google Scholar] [CrossRef]
- Uhl, S.A.; James-Todd, T.; Bell, M.L. Association of osteoarthritis with perfluorooctanoate and perfluorooctane sulfonate in NHANES 2003–2008. Environ. Health Perspect. 2013, 121, 447–452. [Google Scholar] [CrossRef]
- Steenland, K.; Zhao, L.; Winquist, A. A cohort incidence study of workders exposed to perfluorooctanoic acid (PFOA). Occup. Environ. Med. 2015, 72, 373–380. [Google Scholar] [CrossRef]
- Sun, Q.; Zong, G.; Valvi, D.; Nielsen, F.; Coull, B.; Grandjean, P. Plasma concentrations of perfluoroalkyl substances and risk of type 2 diabetes: A prospective investigation among U.S. women. Environ. Health Perspect. 2018, 126, 037001. [Google Scholar] [CrossRef] [Green Version]
- Li, D. Diabetes and pancreatic cancer. Mol. Carcinog. 2012, 51, 64–74. [Google Scholar] [CrossRef] [Green Version]
- Cardenas, A.; Hivert, M.F.; Gold, D.R.; Hauser, R.; Kleinman, K.P.; Lin, P.D.; Fleisch, A.F.; Calafat, A.M.; Ye, X.; Webster, T.F.; et al. Associations of perfluoroalkyl and polyfluoroalkyl substances with incident diabetes and microvascular disease. Diabetes Care 2019, 42, 1824–1832. [Google Scholar] [CrossRef]
- Donat-Vargas, C.; Bergdahl, I.A.; Tornevi, A.; Wennberg, M.; Sommar, J.; Kiviranta, H.; Koponen, J.; Rolandsson, O.; Akesson, A. Perfluoroalkyl substances and risk of type II diabetes: A prospective nested case-control study. Environ. Int. 2019, 123, 390–398. [Google Scholar] [CrossRef]
- Lee, J.K.; Kim, S.H. Correlation between mast cell-mediated allergic inflammation and length of perfluorinated compounds. J. Toxicol. Environ. Health A 2018, 81, 302–313. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.K.; Lee, S.; Baiek, M.C.; Lee, B.H.; Lee, H.S.; Kwon, T.K.; Park, P.H.; Shin, T.Y.; Khang, D.; Kin, S.H. Association between perfluorooctanoic acid exposure and degranulation of mast cells in allergic inflammation. J. Appl. Toxicol. 2017, 37, 554–562. [Google Scholar] [CrossRef] [PubMed]
- Son, H.Y.; Lee, S.; Tak, E.N.; Cho, H.S.; Shin, H.I.; Kim, S.H.; Yang, J.H. Perfluorooctanoic acid alters T lymphocyte phenotypes and cytokine expression in mice. Environ. Toxicol. 2009, 24, 580–588. [Google Scholar] [CrossRef] [PubMed]
- Gimenez-Bastida, J.A.; Surma, M.; Zielinski, H. In vitro evaluation of the cytotoxicity and modulation of mechanisms associated with inflammation induced by perfluorooctanesulfonate and perfluorooctanoic acid in human colon myofibroblasts CCD-18Co. Toxicol. In Vitro 2015, 29, 1683–1691. [Google Scholar] [CrossRef]
- Zhou, X.; Dong, T.; Fan, Z.; Peng, Y.; Zhou, R.; Wang, X.; Song, N.; Han, M.; Fan, B.; Jia, J.; et al. Perfluorodecanoic acid stimulates NLRP3 inflammasome assembly in gastric cells. Sci. Rep. 2017, 7, 45468. [Google Scholar] [CrossRef] [Green Version]
- Sorli, J.B.; Lag, M.; Ekeren, L.; Perez-Gil, J.; Haug, L.S.; Da Silva, E.; Matrod, M.N.; Gutzkow, K.B.; Lindeman, B. Per- and polyfluoroalkyl substances (PFASs) modify lung surfactant function and pro-inflammatory responses in human bronchial epithelial cells. Toxicol. In Vitro 2019. [Google Scholar] [CrossRef]
- Wang, J.; Pan, Y.; Cui, Q.; Yao, B.; Wang, J.; Dai, J. Penetration of PFASs across the blood cerebrospinal fluid barrier and its determinants in humans. Environ. Sci. Technol. 2018, 52, 13553–13561. [Google Scholar] [CrossRef]
- DeWitt, J.M.; Blossom, S.J.; Schaider, L.A. Exposure to per- and polyfluoroalkyl substances leads to immunotoxicity: Epidemiological and toxicological evidence. J. Exp. Sci. Environ. Epidemiol. 2019, 29, 148–156. [Google Scholar] [CrossRef]
- Ladics, G.S. The sheep erythrocyte T-dependent antibody response (TDAR). Methods Mol. Biol. 2018, 1803, 83–94. [Google Scholar]
- Grandjean, P.E.W.A.; Budtz-Jorgensen, E.; Nielsen, F.; Molbak, K.; Weihe, P.; Heilmann, C. Serum vaccine antibody concentrations in children exposed to perfluorinated compounds. JAMA 2012, 307, 391–397. [Google Scholar] [CrossRef] [Green Version]
- Mogensen, U.B.; Grandjean, P.; Heilmann, C.; Nielsen, F.; Weihe, P.; Budtz-Jorgensen, E. Structural equation modeling of immunotoxicity associated with exposure to perfluorinated alkylates. Environ. Health. 2015, 14, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Granum, B.; Haug, L.S.; Namork, E.; Stolevik, S.B.; Thomsen, C.; Aaberge, I.S.; van Loveren, H.; Lovik, M.; Nygaard, U.C. Pre-natal exposure to perfluoroalkyl substances may be associated with altered vaccine antibody levels and immune-related health outcomes in early childhood. J. Immunotoxicol. 2013, 10, 373–379. [Google Scholar] [CrossRef] [PubMed]
- Looker, C.; Luster, M.I.; Calafat, A.M.; Johnson, V.J.; Burleson, G.R.; Burleson, F.G.; Fletcher, T. Influenza vaccine response in adults exposed to perfluorooctanoate and perfluorooctanesulfonate. Toxicol. Sci. 2014, 138, 76–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stein, C.R.; McGovern, K.J.; Pajak, A.M.; Maglione, P.J.; Wolff, M.S. Perfluoroalkyl and polyfluoroalkyl substances and indicators of immune function in children aged 12–19 y: National Health and Nutrition Examination Survey. Pediatr. Res. 2016, 79, 348–357. [Google Scholar] [CrossRef] [Green Version]
- Dong, G.-H.; Tung, K.-Y.; Tsai, C.-H.; Liu, M.-M.; Wang, D.; Liu, W.; Jin, Y.H.; Hsieh, W.S.; Lee, Y.L.; Chen, P.C. Serum polyfluoroalkyl concentrations, asthma outcomes, and immunological markers in a case-control study of Taiwanese children. Environ. Health Perspect. 2013, 121, 507–513. [Google Scholar] [CrossRef] [Green Version]
- Timmermann, C.A.; Budtz-Jorgensen, E.; Jensen, T.K.; Osuna, C.E.; Petersen, M.S.; Steuerwald, U.; Nielsen, F.; Poulsen, L.K.; Weihe, P.; Grandjean, P. Association between perfluoroalkyl substance exposure and asthma and allergic disease in children as modified by MMR vaccination. J. Immunotoxicol. 2017, 14, 39–49. [Google Scholar] [CrossRef]
- Dewitt, J.C.; Copeland, C.B.; Strynar, M.J.; Luebke, R.W. Perfluorooctanoic acid-induced immunomodulation in adult C57BL/6J or C57BL/6N female mice. Environ. Health Perspect. 2008, 116, 644–650. [Google Scholar] [CrossRef] [Green Version]
- Vetvicka, V.; Vetvickova, J. Reversal of perfluorooctanesulfonate-induced immunotoxicity by a glucan-resveratrol-vitamin C combination. J. Orient Pharm. Exp. Med. 2013, 13, 77–84. [Google Scholar] [CrossRef]
- Yang, Q.; Xie, Y.; Depierre, J.W. Effects of peroxisome proliferators on the thymus and spleen of mice. Clin. Exp. Immunol. 2000, 122, 219–226. [Google Scholar] [CrossRef]
- Frawley, R.P.; Smith, M.T.; Cesta, M.F.; Hayes-Bouknight, S.; Blystone, C.; Kissling, G.E.; Harris, S.; Germolec, D. Immunotoxic and hepatotoxic effects of perfluoro-n-decanoic acid (PFDA) on female Harlan Sprague-Dawley rats and B6C3F1/N mice when administered by oral gavage. J. Immunotoxicol. 2018, 15, 41–52. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Kong, B.; He, B.; Wei, L.; Zhu, J.; Jin, Y.; Shan, Y.; Wang, W.; Pan, C.; Fu, Z. 8:2 Flurotelomer alcohol causes immunotoxicity and liver injury in adult male C57BL/6 mice. Environ. Toxicol. 2019, 34, 141–149. [Google Scholar] [PubMed]
- Brieger, A.; Bienefeld, N.; Hasan, R.; Goerlich, R.; Haase, H. Impact of perfluorooctanesulfonate and perfluorooctanoic acid on human peripheral leukocytes. Toxicol. In Vitro 2011, 25, 960–968. [Google Scholar] [CrossRef] [PubMed]
- Corsini, E.; Avogadro, A.; Galbiati, V.; dell’Agli, M.; Marinovich, M.; Galli, C.L.; Germolec, D.R. In vitro evaluation of the immunotoxic potential of perfluorinated compounds (PFCs). Toxicol. Appl. Pharmacol. 2011, 250, 108–116. [Google Scholar] [CrossRef] [PubMed]
- Corsini, E.; Sangiovanni, E.; Avogadro, A.; Galbiati, V.; Viviani, B.; Marinovich, M.; Galli, C.L.; Dell’Agli, M.; Germolec, D.R. In vitro characterization of the immunotoxic potential of several perfluorinated compounds (PFCs). Toxicol. Appl. Pharmacol. 2012, 258, 248–255. [Google Scholar] [CrossRef]
- Fairley, K.J.; Purdy, R.; Kearns, S.; Andersen, S.E.; Meade, B. Exposure to the immunosuppressant, perfluorooctanoic acid, enhances the murine IgE and airway hyperactivity response to ovalbumin. Toxicol. Sci. 2007, 97, 375–383. [Google Scholar] [CrossRef] [Green Version]
- Ryu, M.H.; Jha, A.; Ojo, O.O.; Mahood, T.H.; Basu, S.; Detillieux, K.A.; Nikoobakht, N.; Wong, C.S.; Loewen, M.; Becker, A.B.; et al. Chronic exposure to perfluorinated compounds: Impact on airway hyperresponsiveness and inflammation. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, L765–L774. [Google Scholar] [CrossRef] [Green Version]
- Dong, G.-H.; Liu, M.-M.; Wang, D.; Zheng, L.; Liang, Z.-F.; Jin, Y.-H. Sub-chronic effect of perfluorooctanesulfonate (PFOS) on the balance of type 1 and type 2 cytokine in adult C57BL6 mice. Arch. Toxicol. 2011, 85, 1235–1244. [Google Scholar] [CrossRef]
- National Toxicology Program. NTP Monograph on Immunotoxicity Associated with Exposure to Perfluorooctanoic Acid of Perfluorooctane Sulfonate; Office of Health Assessment and Translation Divisions of the National Toxicology Program, National Institute of Environmental Health Sciences, U.S. Department of Health and Human Services: Research Triangle Park, NC, USA, 2016.
- Arzuaga, X.; Smith, M.T.; Gibbons, C.F.; Skakkebaek, N.E.; Yost, E.E.; Beverly, B.E.J.; Hotchkiss, A.K.; Hauser, R.; Pagani, R.L.; Shrader, S.M.; et al. Proposed key characteristics of male reproductive toxicants as an approach for organizing and evaluating mechanistic evidence in human health hazard assessments. Environ. Health Perspect. 2019, 127, 65001. [Google Scholar] [CrossRef]
- Luderer, U.; Eskenazi, B.; Hauser, R.; Korach, K.S.; McHale, C.M.; Moran, F.; Rieswijk, L.; Solomon, G.; Udagawa, O.; Zhang, L.; et al. Proposed key characteristics of female reproductive toxicants as an approach for organizing and evaluating mechanistic data in hazard assessment. Environ. Health Perspect. 2019, 127, 75001. [Google Scholar] [CrossRef] [Green Version]
- Hankinson, S.E.; Eliassen, A.H. Endogenous estrogen, testosterone and progesterone levels in relation to breast cancer risk. J. Steroid Biochem. Mol. Biol. 2007, 106, 24–30. [Google Scholar] [CrossRef] [Green Version]
- Hardell, L.; van Bavel, B.; Lindstrom, G.; Carlberg, M.; Dreifaldt, A.C.; Wijkstrom, H.; Starkhammar, H.; Eriksson, M.; Hallquist, A.; Kolmert, T. Increased concentrations of polychlorinated biphenyls, hexachlorobenzene and chlordanes in mothers of men with testicular cancer. Environ. Health Perspect. 2003, 111, 930–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hilakivi-Clarke, L.; de Assis, S. Fetal origins of breast cancer. Trends Endocrinol. Metab. 2006, 17, 340–348. [Google Scholar] [CrossRef] [PubMed]
- Skakkebaek, N.E.; Rajpert-De Meyts, E.; Buck Louis, G.M.; Toppari, J.; Andersson, A.M.; Eisenberg, M.L.; Jensen, T.K.; Jorgensen, N.; Swan, S.H.; Sapra, K.J.; et al. Male reproductive disorders and fertility trends: Influences of environment and genetic susceptibility. Physiol. Rev. 2016, 96, 55–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thankamony, A.; Pasterski, V.; Ong, K.K.; Acerini, C.L.; Hughes, I.A. Anogenital distance as a marker of androgen exposure in humans. Andrology 2016, 4, 616–625. [Google Scholar] [CrossRef] [Green Version]
- Guyton, K.Z.; Chiu, W.A.; Bateson, T.F.; Jinot, J.; Scott, C.S.; Brown, R.C.; Caldwell, J.C. A reexamination of the PPAR-alpha activation mode of action as a basis for assessing human cancer risks of environmental contaminants. Environ. Health Perspect. 2009, 117, 1664–1672. [Google Scholar] [CrossRef]
- Schugg, T.T.; Janesick, A.; Blumberg, B.; Heindel, J.J. Endocrine disrupting chemicals and disease susceptibility. J. Steroid Biochem. Mol. Biol. 2011, 127, 204–215. [Google Scholar] [CrossRef] [Green Version]
- Winkens, K.; Vestergren, R.; Berger, U.; Cousins, I.T. Early life exposure to per- and polyfluoroalkyl substance (PFASs): A critical review. Emerg. Contam. 2017, 3, 55–68. [Google Scholar] [CrossRef]
- Bach, C.C.; Vested, A.; Jorgensen, K.T.; Bonde, J.P.; Henriksen, T.B.; Toft, G. Perfluoroalkyl and polyfluoroalkyl substances and measures of human fertility: A systematic review. Crit. Rev. Toxicol. 2016, 46, 735–755. [Google Scholar] [CrossRef]
- Vested, A.; Ramlau-Hansen, C.H.; Olsen, S.F.; Bonde, J.P.; Kristensen, S.L.; Halldorsson, T.I.; Becher, G.; Haug, L.S.; Ernst, E.H.; Toft, G. Associations of in utero exposure to perfluorinated alkyl acids with human semen quality and reproductive hormones in adult men. Environ. Health Perspect. 2013, 121, 453–458. [Google Scholar] [CrossRef] [Green Version]
- Itoh, S.; Araki, A.; Mitsui, T.; Miyashita, C.; Goudarzi, H.; Sasaki, S.; Cho, K.; Nakazawa, H.; Iwasaki, Y.; Shinohara, N.; et al. Association of perfluoroalkyl substances exposure in utero with reproductive hormone levels in cord blood in the Hokkaido Study on Environment and Children’s Health. Environ. Int. 2016, 94, 51–59. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Espinosa, M.J.; Mondal, D.; Armstrong, B.G.; Eskenazi, B.; Fletcher, T. Perfluoroalkyl substances, sex hormones, and insulin-like growth factor-1 at 6–9 years of age: A cross-sectional analysis within the C8 Health Project. Environ. Health Perspect. 2016, 124, 1269–1275. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.S.; Lin, C.Y.; Lin, C.C.; Chen, M.H.; Hsu, S.H.; Chien, K.L.; Sung, F.C.; Chen, P.C.; Su, T.C. Association between perfluoroalkyl substances and reproductive hormones in adolescents and young adults. Int. J. Hyg. Environ. Health 2015, 218, 437–443. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Hu, L.W.; Qian, Z.M.; Geiger, S.D.; Parrish, K.L.; Dharmage, S.C.; Campbell, B.; Roponen, M.; Jalava, P.; Hirvonen, M.R.; et al. Interaction effects of polyfluoroalkyl substances and sex steroid hormones on asthma among children. Sci. Rep. 2017, 7, 899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Nisio, A.; Sabovic, I.; Valente, U.; Tescari, S.; Rocca, M.S.; Guidolin, D.; Dall’Acqua, S.; Acquasaliente, L.; Pozzi, N.; Plebani, M.; et al. Endocrine Disruption of Androgenic Activity by Perfluoroalkyl Substances: Clinical and Experimental Evidence. J. Clin. Endocrinol. Metab. 2019, 104, 1259–1271. [Google Scholar] [CrossRef] [Green Version]
- Lind, D.V.; Priskorn, L.; Lassen, T.H.; Nielsen, F.; Kyhl, H.B.; Kristensen, D.M.; Christesen, H.T.; Jorgensen, J.S.; Grandjean, P.; Jensen, T.K. Prenatal exposure to perfluoroalkyl substances and anogenital distance at 3 months of age in a Danish mother-child cohort. Reprod. Toxicol. 2017, 68, 200–206. [Google Scholar] [CrossRef] [Green Version]
- Tian, Y.; Liang, H.; Miao, M.; yang, F.; Ji, H.; Cao, W.; Liu, X.; Zhang, X.; Chen, A.; Xiao, H.; et al. Maternal plasma concentrations of perfluoroalkyl and polyfluoroalkyl substances during pregnancy and anogenital distance in male infants. Hum. Reprod. 2019, 34, 1356–1368. [Google Scholar] [CrossRef]
- Liew, Z.; Goudarzi, H.; Oulhote, Y. Developmental exposures to perfluoralkyl substances (PFASs): An update of associated health outcomes. Curr. Environ. Health Rep. 2018, 5, 1–19. [Google Scholar] [CrossRef]
- Melzer, D.; Rice, N.; Depledge, M.H.; Henley, W.E.; Galloway, T.S. Association between serum perfluorooctanoic acid (PFOA) and thyroid disease in the U.S. National Health and Nutrition Examination Survey. Environ. Health Perspect. 2010, 118, 686–692. [Google Scholar] [CrossRef] [Green Version]
- Ballesteros, V.; Costa, O.; Iniguez, C.; Fletcher, T.; Ballester, F.; Lopez-Espinosa, M.J. Exposure to perfluoroalkyl substances and thyroid function in pregnant women and children: A systematic review of epidemiologic studies. Environ. Int. 2017, 99, 15–28. [Google Scholar] [CrossRef] [Green Version]
- Lau, C.C.; Anitole, K.; Hodes, C.; Lai, D.; Pfahles-Hutchens, A.; Seed, J. Perfluoroalkyl acids: A review of monitoring and toxicological findings. Toxicol. Sci. 2007, 99, 366–394. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Lu, Y.; Luo, B.; Yan, S.; Guo, X.; Dai, J. Proteomic analysis of mouse testis reveals perfluorooctanoic acid-induced reproductive dysfunction via direct disturbance of testicular steroidogenic machinery. J. Proteome Res. 2014, 13, 3370–3385. [Google Scholar] [CrossRef] [PubMed]
- Song, P.; Li, D.; Wang, X.; Zhong, X. Effects of perfluorooctanoic acid exposure during pregnancy on the reproduction and development of male offspring mice. Andrologia 2018, 50, e13059. [Google Scholar] [CrossRef] [PubMed]
- Conley, J.M.; Lambright, C.S.; Evans, N.; Strynar, M.J.; McCord, J.; McIntyre, B.S.; Travlos, G.S.; Cardon, M.C.; Medlock-Kakaley, E.; Hartig, P.C.; et al. Adverse maternal, fetal, and postnatal effects of hexafluoropropylene oxide dimer acid (GenX) from oral gestational exposure in Sprague-Dawley rats. Environ. Health Perspect. 2019, 127, 37008. [Google Scholar] [CrossRef] [PubMed]
- Ramhoj, L.; Hass, U.; Boberg, J.; Scholze, M.; Christensen, S.; Nielsen, F.; Axelstad, M. Perfluorohexane sulfonate (PFHxS) and a mixture of endocrine disrupters reduce thyroxine levels and cause antiandrogenic effects in rats. Toxicol. Sci. 2018, 163, 579–591. [Google Scholar] [CrossRef] [Green Version]
- Tucker, D.K.; Macon, M.B.; Strynar, M.J.; Dagnino, S.; Andersen, E.; Fenton, S.E. The mammary gland is a sensitive pubertal target in CD-1 and C57Bl/6 mice following perinatal perfluorooctanoic acid (PFOA) exposure. Reprod. Toxicol. 2015, 54, 26–36. [Google Scholar] [CrossRef] [Green Version]
- White, S.S.; Calafat, A.M.; Kuklenyik, Z.; Villanueva, L.; Zehr, R.D.; Helfant, L.; Strynar, M.J.; Lindstrom, A.B.; Thibodeaux, J.R.; Wood, C.; et al. Gestational PFOA exposure of mice is associated with altered mammary gland development in dams and female offspring. Toxicol. Sci. 2007, 96, 133–144. [Google Scholar] [CrossRef]
- Cope, H.; Blake, B.; Fenton, S.E. Effects of PFOA and GenX on Early Developmental Outcomes and Puberty in CD-1 Mice. In Proceedings of the Society of Toxicology 58th Annual Meeting, Baltimore, MD, USA, 10–14 March 2019. [Google Scholar]
- Cohn, B.A.; La Merrill, M.A.; Krigbaum, N.Y.; Wang, M.; Park, J.S.; Petreas, M.; Yeh, G.; Hovey, R.C.; Zimmermann, L.; Cirillo, P.M. In utero exposure to poly and perfluoroalkyl substances (PFASs) and subsequent breast cancer. Reprod. Toxicol. 2019. [Google Scholar] [CrossRef]
- Dixon, D.; Reed, C.E.; Moore, A.B.; Gibbs-Flournoy, E.A.; Hines, E.P.; Wallace, E.A.; Stanko, J.P.; Lu, Y.; Jefferson, W.N.; Newbold, R.R.; et al. Histopathologic changes in the uterus, cervix and vagina of immature CD-1 mice exposed to low doses of perfluorooctanoic acid (PFOA) in a uterotrophic asay. Reprod. Toxicol. 2012, 33, 506–512. [Google Scholar] [CrossRef] [Green Version]
- Yao, P.L.; Ehresman, D.J.; Rae, J.M.; Chang, S.C.; Frame, S.R.; Butenhoff, J.L.; Kennedy, G.L.; Peters, J.M. Comparative in vivo and in vitro analysis of possible estrogenic effects of perfluorooctanoic acid. Toxicology 2014, 326, 62–73. [Google Scholar] [CrossRef]
- Qiu, Z.; Qu, K.; Luan, F.; Liu, Y.; Zhu, Y.; Yuan, Y.; Li, H.; Zhang, H.; Hai, Y.; Zhao, C. Binding specificities of estrogen receptor with perfluorinated compounds: A cross species comparison. Environ. Int. 2020, 134, 105284. [Google Scholar] [CrossRef]
- Chiu, W.A.; Guyton, K.Z.; Martin, M.T.; Reif, D.M.; Rusyn, I. Use of high-throughput in vitro toxicity screening data in cancer hazard evaluations by IARC Monograph Working Groups. ALTEX 2018, 35, 51–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosen, M.B.; Das, K.P.; Rooney, J.; Abbott, B.D.; Lau, C.; Corton, J.C. PPARα-independent transcriptional targets of perfluoroalkyl acids revealed by transcript profiling. Toxicology 2017, 387, 95–107. [Google Scholar] [CrossRef] [PubMed]
- Xin, Y.; Ren, X.M.; Wan, B.; Guo, L.H. Comparative in vitro and in vivo evaluation of the estrogenic effect of hexafluoropropylene oxide homologues. Environ. Sci. Technol. 2019, 53, 8371–8380. [Google Scholar] [CrossRef] [PubMed]
- Rosenmai, A.K.; Ahrens, L.; le Godec, T.; Lundgvist, J.; Oskarsson, A. Relationship between peroxisome proliferator-activated receptor alpha activity and cellular concentration of 14 perfluoroalkyl substances in HepG2 cells. J. Appl. Toxicol. 2018, 38, 219–226. [Google Scholar] [CrossRef]
- Wolf, C.J.; Schmid, J.E.; Lau, C.; Abbott, B.D. Activation of mouse and human peroxisome proliferator-activated receptor-alpha (PPARa) by perfluoroalkyl acids (PFAAs): Further investigation of C4–C12 compounds. Reprod. Toxicol. 2012, 33, 546–551. [Google Scholar] [CrossRef]
- Kjeldsen, L.S.; Bonefeld-Jorgensen, E.C. Perfluorinated compounds affect the function of sex hormone receptors. Environ. Sci. Pollut. Res. Int. 2013, 20, 8031–8044. [Google Scholar] [CrossRef]
- Behr, A.C.; Lichtenstein, D.; Braeuning, A.; Lampen, A.; Buhrke, T. Perfluoroalkylated substances (PFAS) affect neither estrogen and androgen receptor activity nor steroidogenesis in human cells in vitro. Toxicol. Lett. 2018, 291, 51–60. [Google Scholar] [CrossRef]
- Du, G.; Hu, J.; Huang, H.; Qin, Y.; Han, X.; Wu, D.; Song, L.; Xia, Y.; Wang, X. Perfluorooctane sulfonate (PFOS) affects hormone receptor activity, steroidogenesis, and expression of endocrine-related genes in vitro and in vivo. Environ. Toxicol. Chem. 2013, 32, 353–360. [Google Scholar] [CrossRef]
- Rosenmai, A.K.; Nielsen, F.K.; Pedersen, M.; Hadrup, N.; Trier, X.; Christensen, J.H.; Vinggaard, A.M. Fluorochemicals used in food packaging inhibit male sex hormone synthesis. Toxicol. Appl. Pharmacol. 2013, 266, 132–142. [Google Scholar] [CrossRef] [Green Version]
- Rosenmai, A.K.; Taxvig, C.; Svingen, T.; Trier, X.; van Vugt-Lussenburg, B.M.; Pedersen, M.; Lsne, L.; Jegou, B.; Vinggaard, A.M. Fluorinated alkyl substances and technical mixtures used in food paper-packaging exhibit endocrine-related activity in vitro. Andrology 2016, 4, 662–672. [Google Scholar] [CrossRef] [Green Version]
- Gao, K.; Zhuang, T.; Liu, X.; Fu, J.; Zhang, J.; Fu, J.; Wang, L.; Zhang, A.; Liang, Y.; Song, M.; et al. Prenatal exposure to per- and polyfluoroalkyl substances (PFASs) and association between the placental transfer efficiencies and dissociation constant of serum proteins-PFAS complexes. Environ. Sci. Technol. 2019, 53, 6529–6538. [Google Scholar] [CrossRef]
- Ng, C.A.; Hungerbuehler, K. Exploring the use of molecular docking to identify bioaccumulative perfluorinated alkyl acids (PFAAs). Environ. Sci. Technol. 2015, 49, 12306–12314. [Google Scholar] [CrossRef] [Green Version]
- Kar, S.; Sepulveda, M.S.; Roy, K.; Leszczynski, J. Endocrine-disrupting activity of per- and polyfluoroalkyl substances: Exploring combined approaches of ligand and structure based modeling. Chemosphere 2017, 184, 514–523. [Google Scholar] [CrossRef]
- Ren, X.M.; Qin, W.P.; Cao, L.Y.; Zhang, J.; Yang, Y.; Wan, B.; Guo, L.H. Binding interactions of perfluoroalkyl substances with thyroid hormone transport proteins and potential toxicological implications. Toxicology 2016, 366, 32–42. [Google Scholar] [CrossRef]
- Feng, X.; Cao, X.; Zhao, S.; Wang, X.; Hua, X.; Chen, L.; Chen, L. Exposure of pregnant mice to perfluorobutanesulfonate causes hypothyroxinemia and developmental abnormalities in female offspring. Toxicol. Sci. 2017, 155, 409–419. [Google Scholar] [CrossRef]
- Martin, M.T.; Brennan, R.J.; Hu, W.; Ayanoglu, E.; Lau, C.; Ren, H.; Wood, C.R.; Corton, J.C.; Kablock, R.J.; Dix, D.L. Toxicogenomic study of triazole fungicides and perfluoroalkyl acids in rat livers predicts toxicity and categorizes chemicals based on mechanisms of toxicity. Toxicol.Sci. 2007, 97, 595–613. [Google Scholar] [CrossRef]
- Thibodeaux, J.R.; Hanson, R.G.; Rogers, J.M.; Grey, B.E.; Barbee, B.D.; Richards, J.H.; Butenhoff, J.L.; Stevenson, L.A.; Lau, C. Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. I: Maternal and prenatal evaluations. Toxicol. Sci. 2003, 74, 369–381. [Google Scholar] [CrossRef]
- Selano, J.; Richardson, V.; Washington, J.; Mazur, C. Characterization of non-radiolabeled Thyroxine (T4) uptake in cryopreserved rat hepatocyte suspensions: Pharmacokinetic implications for PFOA and PFOS chemical exposure. Toxicol. In Vitro 2019, 58, 230–238. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Martens, D.S.; Nawrot, T.S. Air pollution stress and the aging phenotype: The telomere connection. Curr. Environ. Health Rep. 2016, 3, 258–269. [Google Scholar] [CrossRef]
- Perdiz, D.; Oziol, L.; Pous, C. Early mitochondrial fragmentation is a potential in vitro biomarker of environmental stress. Chemosphere 2019, 223, 577–587. [Google Scholar] [CrossRef]
- Liu, H.; Chen, Q.; Lei, L.; Zhou, W.; Huang, L.; Zhang, J.; Chen, D. Prenatal exposure to perfluoroalkyl and polyfluoroalkyl substances affects leukocyte telomere length in female newborns. Environ. Pollut. 2018, 235, 446–452. [Google Scholar] [CrossRef]
- Vriens, A.; Nawrot, T.S.; Janssen, B.G.; Baeyens, W.; Bruckers, L.; Covaci, A.; De Craemer, S.; De Henauw, S.; Den Hond, E.; Loots, I.; et al. Exposure to environmental pollutants and their association with biomarkers of aging: A multipollutant approach. Environ. Sci. Technol. 2019, 53, 5966–5976. [Google Scholar] [CrossRef]
- Blevin, P.; Angelier, F.; Tartu, S.; Bustamante, P.; Herzke, D.; Moe, B.; Bech, C.; Gabrielsen, G.W.; Bustnes, J.O.; Chastel, O. Perfluorinated substances and telomeres in an Arctic seabird: Cross-sectional and longitudinal approaches. Environ. Pollut. 2017, 230, 360–367. [Google Scholar] [CrossRef]
- Otto, T.; Sicinski, P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 2017, 17, 93–115. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez, T.L.; Rae, J.M.; Colacino, J.A. Implication of environmental estrogens on breast cancer treatment and progression. Toxicology 2019, 421, 41–48. [Google Scholar] [CrossRef]
- Pierozan, P.; Jerneren, F.; Karlsson, O. Perfluorooctanoic acid (PFOA) exposure promotes proliferation, migration and invasion potential in human breast epithelial cells. Arch. Toxicol. 2018, 92, 1729–1739. [Google Scholar] [CrossRef] [Green Version]
- Pierozan, P.; Karlsson, O. PFOS induces proliferation, cell-cycle progression, and malignant phenotype in human breast epithelial cells. Arch. Toxicol. 2018, 92, 705–716. [Google Scholar] [CrossRef] [Green Version]
- Dairkee, S.H.; Luciani-Torres, G.; Moore, D.H.; Jaffee, I.M.; Goodson, W.H.R. A ternary mixture of common chemicals perturbs benign human breast epithelial cells more than the same chemicals do individually. Toxicol. Sci. 2018, 165, 131–144. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Liu, X.; Li, F.; Wang, Y.; Xu, Y.; Zhang, M.; Zhang, X.; Ying, X.; Zhang, X. Perfluorooctanoic acid induces human Ishikawa endometrial cancer cell migration and invasion through activation of ERK/mTOR signaling. Oncotarget 2016, 7, 66558–66568. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Bao, C.; Ma, Z.; Xu, B.; Ying, X.; Liu, X.; Zhang, X. Perfluorooctanoic acid stimulates ovarian cancer cell migration, invasion via ERK/NF-κB/MMP-2/-9 pathway. Toxicol. Lett. 2018, 294, 44–50. [Google Scholar] [CrossRef]
- Gogola, J.; Hoffmann, M.; Ptak, A. Persistent endocrine-disrupting chemicals found in human follicular fluid stimulate the proliferation of granulosa tumor spheroids via GPR30 and IGF1R but not via the classic estrogen receptors. Chemosphere 2019, 217, 100–110. [Google Scholar] [CrossRef]
- Cui, R.; Zhang, H.; Guo, X.; Cui, Q.; Wang, J.; Dai, J. Proteomic analysis of cell proliferation in a human hepatic cell line (HL-7702) induced by perfluorooctane sulfonate using iTRAQ. J. Hazard. Mater. 2015, 299, 361–370. [Google Scholar] [CrossRef]
- Zhang, H.; Cui, R.; Guo, X.; Hu, J.; Dai, J. Low dose perfluorooctanoate exposure promotes cell proliferation in a human non-tumor liver cell line. J. Hazard. Mater. 2016, 313, 18–28. [Google Scholar] [CrossRef]
- Maras, M.; Vanparys, C.; Muylle, F.; Robbens, J.; Berger, U.; Barber, J.L.; Blust, R.; De Coen, W. Estrogen-like properties of fluorotelomer alcohols as revealed by mcf-7 breast cancer cell proliferation. Environ. Health Perspect. 2006, 114, 100–105. [Google Scholar] [CrossRef] [Green Version]
- Krewski, D.; Bird, M.; Al-Zoughool, M.; Birkett, N.; Billard, M.; Milton, B.; Rice, J.M.; Grosse, Y.; Cogliano, V.J.; Hill, M.A.; et al. Key characteristics of 86 agents known to cause cancer in humans. J. Toxicol. Environ. Health B Crit. Rev. 2019, 22, 244–263. [Google Scholar] [CrossRef]
- Miller, M.F.; Goodson, W.H.; Manjili, M.H.; Kleinstreuer, N.; Bisson, W.H.; Lowe, L. Low-Dose Mixture Hypothesis of Carcinogenesis Workshop: Scientific Underpinnings and Research Recommendations. Environ. Health Perspect. 2017, 125, 163–169. [Google Scholar] [CrossRef] [Green Version]
- Goodson, W.H.; Lowe, L., 3rd; Carpenter, D.O.; Gilbertson, M.; Manaf Ali, A.; Lopez de Cerain Salsamendi, A.; Lasfar, A.; Carnero, A.; Azqueta, A.; Amedei, A.; et al. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: The challenge ahead. Carcinogenesis 2015, 36 (Suppl. 1), S254–S296. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
Key Characteristics | Examples of Relevant Evidence |
---|---|
1—Is electrophilic or can be metabolically activated | Parent compound or metabolite with an electrophilic structure (e.g., epoxide, quinone, etc.), formation of DNA and protein adducts |
2—Is genotoxic | DNA damage (DNA strand breaks, DNA-protein cross-links, unscheduled DNA synthesis), intercalation, gene mutations, cytogenetic changes (e.g., chromosome aberrations, micronuclei) |
3—Alters DNA repair or causes genomic instability | Alterations of DNA replication or repair (e.g., topoisomerase II, base-excision or double-strand break repair |
4—Induces epigenetic alterations | DNA methylation, histone modification, microRNAs |
5—Induces oxidative stress | Oxygen radicals, oxidative stress, oxidative damage to macromolecules (e.g., DNA, lipids) |
6—Induces chronic inflammation | Elevated white blood cells, myeloperoxidase activity, altered cytokine and/or chemokine production |
7—Is immunosuppressive | Decreased immunosurveillance, immune system dysfunction |
8—Modulates receptor-mediated effects | Receptor in/activation (e.g., ER, PPAR, AhR) or modulation of endogenous ligands (including hormones) |
9—Causes immortalization | Inhibition of senescence, cell transformation |
10—Alters cell proliferation, cell death or nutrient supply | Increased proliferation, decreased apoptosis, changes in growth factors, energetics and signaling pathways related to cellular replication or cell-cycle control, angiogenesis |
Chemical Groups and Abbreviations | a Study Findings from Lines of Evidence for KC 1—Is Electrophilic or Can Be Metabolically Activated | |
---|---|---|
Epidemiological | Animal Bioassay | |
PFOA | No assocaition: reviewed in [2]; reviewed in [47]; [49]; [50] | No Association: reviewed in [2]; reviewed in [47]; [58] |
Long-chain PFAS b | ||
PFOS | No association: reviewed in [3]; [49]; [50]; reviewed [47] | No Association: reviewed in [3]; [58]; reviewed in [47] |
PFHxS | No association: [49]; [50]; reviewed [47] | No association: reviewed in [47] |
PFNA | No association: reviewed [47] | No association: reviewed in [47] |
PFDA | No association: reviewed in [47] | No association: reviewed in [47] |
PFUnA | No association: reviewed in [47] | |
PFDoA | No association: reviewed in [47] | |
PFOSA | No association: reviewed in [47] | |
8:2 FTOH | * Association: [55] | |
8:2 diPAP | * Association: [56] | |
10:2 diPAP | * Assocaition: [56] | |
Short-chain PFAS c | ||
PFBS | No association: [51] | No association: reviewed in [47] |
PFHxA | No association: reviewed in [47] | |
PFBA | No association: reviewed in [47] | |
PFHpA | No association: reviewed in [47] | |
GenX (HFPO-DA); PMOH | No association: [52] | |
6:2 FTOH | * Association: [57] | |
4:2 diPAP | * Assocaition: [53] | * Association: [56] |
6:2 diPAP | * Association: [53] | * Association: [56] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 4—Induces Epigenetic Alterations | ||
---|---|---|---|
Epidemiological | Animal Bioassay | In Vitro | |
PFOA | Association: [70]; [72]; [73]; [74] No association: [71]; [82]; [76] | Association: [78] | |
Long-chain PFAS a,b | |||
PFOS | Association: [71]; [73]; [82] No association: [70]; [76] | Association: [77] | Association: [79]; [81] No association: [80] |
PFHxS | No association: [82]; [76] | ||
PFNA | No association: [71]; [82]; [76] | ||
PFUnA | No association: [71] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 5—Induces Oxidative Stress | ||
---|---|---|---|
Epidemiological | Animal Bioassay | In Vitro | |
PFOA | Association: [85] | Association: [88]; [91]; [25]; [92] | Association: [86]; [87]; [89]; [90]; [93]; [94]; [100]; [101]; [102] |
Long-chain PFAS a | |||
PFOS | Association: [85] | Association: [95]; reviewed in [32]; reviewed in [97]; [99] | Association: [86]; [96]; reviewed in [32]; reviewed in [97]; [100]; [101]; [102] |
PFHxS | Association: [85] | Association: [101] | |
PFNA | Association: [85] | Association: [98]; [99] | Association: [101] |
PFDA | Association: [85] | Association: [99] | Association: [101] |
PFUnA | Association: [85] | Association: [99] | Association: [101] |
PFDoA | Association: [99] | Association: [101] | |
PFTrDA | Association: [99] | ||
PFTeDA | Association: [99] | ||
PFOSA | Association: [101] | ||
Short-chain PFAS b | |||
PFBS | Association: [102] No association: [86] | ||
PFHxA | No association: [86] | ||
PFPeA | Association: [100] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 6—Induces Chronic Inflammation | ||
---|---|---|---|
Epidemiological | Animal Bioassay | In Vitro | |
PFOA | Association: [105]; [106]; [107]; [108]; [110]; [112] No association: [109] | Association: [115]; [116] | Association: [90]; [117] |
Long-chain PFAS a | |||
PFOS | Association: [110] No association: [107]; [113] | Association: [114] | Association: [117]; [119] |
PFHxS | No association: [119] | ||
PFDA | Association: [114]; [118] | Association: [118] | |
PFUnA | Association: [114] | ||
8:2 FTOH | No association: [119] | ||
Short-chain PFAS b | |||
PFBS | No association: [119] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 7—Is Immunosuppressive | ||
---|---|---|---|
Epidemiological | Animal Bioassay | In Vitro | |
PFOA | Association: [123]; [125]; [126]; [127] | Association: reviewed in [141]; [132]; [130] | Association: [135]; [136] |
Long-chain PFAS a | |||
PFOS | Association: [123]; [125]; [127] | Association: reviewed in [141]; [131]; [132] | Association: [135]; [136] |
PFHxS | Association; [125]; [127] | ||
PFNA | Association: [125] | ||
PFDA | Association: [133] | Association: [137] | |
PFOSA | Association: [137] | ||
8:2 FTOH | Association: [134] | Association: [134]; [137] | |
Short-chain PFAS b | |||
PFBS | Association: [137] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 8—Modulates Receptor-Mediated Effects | ||
---|---|---|---|
Epidemiological | Animal Biosassay | In Vitro | |
PFOA | Association: [153]; [154]; [155]; [156]; [157] | Association: reviewed in [164]; [91]; [165]; [166]; [169]; [170]; [173]; [175]; [191]; [61] No association: [174] | Association: [176]; [158]; [175]; [181]; [188]; [189]; [193]; [179]; [180] No association: [182] |
Long-chain PFAS a | |||
PFOS | Association: [154]; [155]; [156]; [157]; [160]; reviwed in [163] No Association: [153] | Association: [191]; [192] | Association: [177]; [175]; [181]; [188]; [189]; [193]; [179]; [180] No association: [182] |
PFHxS | Association: [159]; reviwed in [163] | No association: [168] | Association: [177]; [175]; [181]; [188]; [189]; [179]; [180] No association: [182] |
PFNA | Association: [155]; [157]; [159]; reviewed in [163] | Association: [98] | Association: [177]; [181]; [188]; [189]; [179]; [180] |
PFDA | Association: [157]; [159]; [160] | Association: [181]; [188]; [189]; [179] No Association: [180] | |
PFUnA | Association: [156]; [160] | Association: [188]; [189]; [180] No association: [179] | |
PFDoA | Association: [188]; [189]; [179] No association: [180] | ||
PFTrDA | Association: [189] | ||
PFTeDA | Association: [188]; [189]; [179] | ||
8:2 FTOH | Association: [184]; [185] No association: [188] | ||
8:2 monoPAP | Association: [184]; [185] | ||
8:2 diPAP | Association: [184] | ||
8:2 triPAP | Association: [184] | ||
10:2 diPAP | Association: [184] | ||
Short-chain PFAS b | |||
PFBS | Association: [190] | Association: [175]; [188]; [189]; [179]; [180] No association: [182] | |
PFHxA | Association: [61] | Association: [175]; [188]; [189]; [179]; [180] No association: [182] | |
PFBA | Association: [175]; [189]; [179]; [180] No association: [182] | ||
PFPeA | Association: [179]; [180] | ||
PFHpA | Association: [188]; [189]; [179]; [180] | ||
GenX (HFPO-DA); PMOH | Association: [167]; [171] | Association: [178] No association: [182] | |
PMPP/ADONA | No association: [182] | ||
4:2 FTOH | Association: [175]; [185] | ||
6:2 FTOH | Association: [175]; [185] No association: [188] |
Chemical Groups and Abbreviations | Study Findings from Lines of Evidence for KC 10—Alters Cell Proliferation, Cell Death or Nutrient Supply | ||
---|---|---|---|
Epidemiological | Animal Biosassay | In Vitro | |
PFOA | Association: [61] | Association: [202]; [204]; [205]; [207]; [209] | |
Long-chain PFAS a | |||
PFOS | Association: [203]; [207]; [208] | ||
PFHxS | Association: [62] | ||
PFNA | Association: [61] | ||
8:2 FTOH | Association: [210] | ||
Short-chain PFAS b | |||
PFBS | Association: [62] | ||
PFHxA | Association: [61] | ||
6:2 FTOH | Association: [210] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Temkin, A.M.; Hocevar, B.A.; Andrews, D.Q.; Naidenko, O.V.; Kamendulis, L.M. Application of the Key Characteristics of Carcinogens to Per and Polyfluoroalkyl Substances. Int. J. Environ. Res. Public Health 2020, 17, 1668. https://doi.org/10.3390/ijerph17051668
Temkin AM, Hocevar BA, Andrews DQ, Naidenko OV, Kamendulis LM. Application of the Key Characteristics of Carcinogens to Per and Polyfluoroalkyl Substances. International Journal of Environmental Research and Public Health. 2020; 17(5):1668. https://doi.org/10.3390/ijerph17051668
Chicago/Turabian StyleTemkin, Alexis M., Barbara A. Hocevar, David Q. Andrews, Olga V. Naidenko, and Lisa M. Kamendulis. 2020. "Application of the Key Characteristics of Carcinogens to Per and Polyfluoroalkyl Substances" International Journal of Environmental Research and Public Health 17, no. 5: 1668. https://doi.org/10.3390/ijerph17051668