Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review

Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, whic...

Full description

Autores:
Cano, Raquel
Pérez, José L.
Angarita Dávila, Lissé
Ortega, Ángel
Gómez, Yosselin
Valero-Cedeño, Nereida Josefina
Parra, Heliana
Manzano, Alexander
Díaz Albornoz, María P
Cano, Gabriel
Rojas-Quintero, Joselyn
Chacín, Maricarmen
Bermúdez, Valmore
Véliz Castro, Teresa Isabel
Tipo de recurso:
Fecha de publicación:
2021
Institución:
Universidad Simón Bolívar
Repositorio:
Repositorio Digital USB
Idioma:
eng
OAI Identifier:
oai:bonga.unisimon.edu.co:20.500.12442/8361
Acceso en línea:
https://hdl.handle.net/20.500.12442/8361
https://doi.org/10.3390/ijms22094807
Palabra clave:
Non-alcoholic fatty liver disease
Endocrine-disrupting chemicals
Liver disorder
Environmental pollutants
Exposure
Rights
openAccess
License
Attribution-NonCommercial-NoDerivatives 4.0 Internacional
id USIMONBOL2_dcf6a557cfc2f1d731b30ab59fd660cd
oai_identifier_str oai:bonga.unisimon.edu.co:20.500.12442/8361
network_acronym_str USIMONBOL2
network_name_str Repositorio Digital USB
repository_id_str
dc.title.eng.fl_str_mv Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
title Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
spellingShingle Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
Non-alcoholic fatty liver disease
Endocrine-disrupting chemicals
Liver disorder
Environmental pollutants
Exposure
title_short Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
title_full Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
title_fullStr Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
title_full_unstemmed Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
title_sort Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review
dc.creator.fl_str_mv Cano, Raquel
Pérez, José L.
Angarita Dávila, Lissé
Ortega, Ángel
Gómez, Yosselin
Valero-Cedeño, Nereida Josefina
Parra, Heliana
Manzano, Alexander
Díaz Albornoz, María P
Cano, Gabriel
Rojas-Quintero, Joselyn
Chacín, Maricarmen
Bermúdez, Valmore
Véliz Castro, Teresa Isabel
dc.contributor.author.none.fl_str_mv Cano, Raquel
Pérez, José L.
Angarita Dávila, Lissé
Ortega, Ángel
Gómez, Yosselin
Valero-Cedeño, Nereida Josefina
Parra, Heliana
Manzano, Alexander
Díaz Albornoz, María P
Cano, Gabriel
Rojas-Quintero, Joselyn
Chacín, Maricarmen
Bermúdez, Valmore
dc.contributor.author.spa.fl_str_mv Véliz Castro, Teresa Isabel
dc.subject.eng.fl_str_mv Non-alcoholic fatty liver disease
Endocrine-disrupting chemicals
Liver disorder
Environmental pollutants
Exposure
topic Non-alcoholic fatty liver disease
Endocrine-disrupting chemicals
Liver disorder
Environmental pollutants
Exposure
description Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, which may increase liver-related mortality. In light of this, numerous efforts have been carried out in recent years in order to clarify its pathogenesis and create new prevention strategies. Currently, the essential role of environmental pollutants in NAFLD development is recognized. Particularly, endocrine-disrupting chemicals (EDCs) have a notable influence. EDCs can be classified as natural (phytoestrogens, genistein, and coumestrol) or synthetic, and the latter ones can be further subdivided into industrial (dioxins, polychlorinated biphenyls, and alkylphenols), agricultural (pesticides, insecticides, herbicides, and fungicides), residential (phthalates, polybrominated biphenyls, and bisphenol A), and pharmaceutical (parabens). Several experimental models have proposed a mechanism involving this group of substances with the disruption of hepatic metabolism, which promotes NAFLD. These include an imbalance between lipid influx/efflux in the liver, mitochondrial dysfunction, liver inflammation, and epigenetic reprogramming. It can be concluded that exposure to EDCs might play a crucial role in NAFLD initiation and evolution. However, further investigations supporting these effects in humans are required.
publishDate 2021
dc.date.accessioned.none.fl_str_mv 2021-09-13T23:36:01Z
dc.date.available.none.fl_str_mv 2021-09-13T23:36:01Z
dc.date.issued.none.fl_str_mv 2021
dc.type.coarversion.fl_str_mv http://purl.org/coar/version/c_970fb48d4fbd8a85
dc.type.coar.fl_str_mv http://purl.org/coar/resource_type/c_2df8fbb1
dc.type.driver.eng.fl_str_mv info:eu-repo/semantics/article
dc.type.spa.spa.fl_str_mv Artículo científico
dc.identifier.citation.eng.fl_str_mv Cano, R., Pérez, J., Dávila, L., Ortega, Á., Gómez, Y., Valero-Cedeño, N., Parra, H., et al. (2021). Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. International Journal of Molecular Sciences, 22(9), 4807. MDPI AG. Retrieved from http://dx.doi.org/10.3390/ijms22094807
dc.identifier.issn.none.fl_str_mv 14220067
dc.identifier.uri.none.fl_str_mv https://hdl.handle.net/20.500.12442/8361
dc.identifier.doi.none.fl_str_mv https://doi.org/10.3390/ijms22094807
identifier_str_mv Cano, R., Pérez, J., Dávila, L., Ortega, Á., Gómez, Y., Valero-Cedeño, N., Parra, H., et al. (2021). Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. International Journal of Molecular Sciences, 22(9), 4807. MDPI AG. Retrieved from http://dx.doi.org/10.3390/ijms22094807
14220067
url https://hdl.handle.net/20.500.12442/8361
https://doi.org/10.3390/ijms22094807
dc.language.iso.eng.fl_str_mv eng
language eng
dc.rights.*.fl_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
dc.rights.coar.fl_str_mv http://purl.org/coar/access_right/c_abf2
dc.rights.uri.*.fl_str_mv http://creativecommons.org/licenses/by-nc-nd/4.0/
dc.rights.accessrights.eng.fl_str_mv info:eu-repo/semantics/openAccess
rights_invalid_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
http://creativecommons.org/licenses/by-nc-nd/4.0/
http://purl.org/coar/access_right/c_abf2
eu_rights_str_mv openAccess
dc.format.mimetype.spa.fl_str_mv pdf
dc.publisher.eng.fl_str_mv MDPI
dc.source.eng.fl_str_mv International Journal of Molecular Sciences
dc.source.none.fl_str_mv Vol. 22 N° 9 (2021)
institution Universidad Simón Bolívar
bitstream.url.fl_str_mv https://bonga.unisimon.edu.co/bitstreams/f353d629-8e7a-4221-8ab3-92bdb633fd8a/download
https://bonga.unisimon.edu.co/bitstreams/4f1b3b77-8694-427d-9739-1789a24b531a/download
https://bonga.unisimon.edu.co/bitstreams/e2846100-9e4e-4166-b6b2-a9551a5ff110/download
https://bonga.unisimon.edu.co/bitstreams/0d257ec2-b3ba-495b-b303-3edfbc34c9d9/download
https://bonga.unisimon.edu.co/bitstreams/ee67a104-8056-413e-8ac8-f5b2af24db40/download
https://bonga.unisimon.edu.co/bitstreams/15aff6e2-822f-4647-9342-c24237c6d93e/download
https://bonga.unisimon.edu.co/bitstreams/344f77dc-5621-4a51-8dae-3e45aca0ce73/download
bitstream.checksum.fl_str_mv 8be15eccdb0818e35814d743e4750d9f
4460e5956bc1d1639be9ae6146a50347
733bec43a0bf5ade4d97db708e29b185
f745f73c03f63dd2c6e2100352bb8040
6eabd5082166a1c934c6617c0e00125c
34c3592590f32b2068ce88aa1e914367
75bb663256615307af65e226c4cff211
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
MD5
MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Digital Universidad Simón Bolívar
repository.mail.fl_str_mv repositorio.digital@unisimon.edu.co
_version_ 1812100476746858496
spelling Cano, Raquel713e8685-60eb-4b70-a3fb-d1cd0aebf52aPérez, José L.8cd3b0aa-9887-4ba3-84f8-9abec29a23acAngarita Dávila, Lisséc25715e4-e216-42d3-b331-a3d5afcca897Ortega, Ángelb6a809bb-4d26-4e53-9419-e4eb9fb40a9bGómez, Yosselin3ab23cf1-5f39-4a9f-aeab-c3da987d96eaValero-Cedeño, Nereida Josefina6feed145-b4b4-4ad7-97dc-9ad497201280Parra, Heliana13603b25-99ce-42ac-a469-5b65a89ce794Manzano, Alexander0dd1f5fe-19cd-42e0-b698-34b17a178629Díaz Albornoz, María P49b42ebf-75fb-41d1-ad6c-662397597e8eCano, Gabriel488f5d93-5a7f-43e8-ba52-647d54c46111Rojas-Quintero, Joselyn1fcd6ac1-186e-465c-a5fd-3c25563275abChacín, Maricarmen5c3b3d7c-4444-47e2-b2be-11f08df10409Bermúdez, Valmore29f9aa18-16a4-4fd3-8ce5-ed94a0b8663aVéliz Castro, Teresa Isabel6202a901-93d2-4c86-a603-d97730f4dfd32021-09-13T23:36:01Z2021-09-13T23:36:01Z2021Cano, R., Pérez, J., Dávila, L., Ortega, Á., Gómez, Y., Valero-Cedeño, N., Parra, H., et al. (2021). Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. International Journal of Molecular Sciences, 22(9), 4807. MDPI AG. Retrieved from http://dx.doi.org/10.3390/ijms2209480714220067https://hdl.handle.net/20.500.12442/8361https://doi.org/10.3390/ijms22094807Non-alcoholic fatty liver disease (NAFLD) is considered the most common liver disorder, affecting around 25% of the population worldwide. It is a complex disease spectrum, closely linked with other conditions such as obesity, insulin resistance, type 2 diabetes mellitus, and metabolic syndrome, which may increase liver-related mortality. In light of this, numerous efforts have been carried out in recent years in order to clarify its pathogenesis and create new prevention strategies. Currently, the essential role of environmental pollutants in NAFLD development is recognized. Particularly, endocrine-disrupting chemicals (EDCs) have a notable influence. EDCs can be classified as natural (phytoestrogens, genistein, and coumestrol) or synthetic, and the latter ones can be further subdivided into industrial (dioxins, polychlorinated biphenyls, and alkylphenols), agricultural (pesticides, insecticides, herbicides, and fungicides), residential (phthalates, polybrominated biphenyls, and bisphenol A), and pharmaceutical (parabens). Several experimental models have proposed a mechanism involving this group of substances with the disruption of hepatic metabolism, which promotes NAFLD. These include an imbalance between lipid influx/efflux in the liver, mitochondrial dysfunction, liver inflammation, and epigenetic reprogramming. It can be concluded that exposure to EDCs might play a crucial role in NAFLD initiation and evolution. However, further investigations supporting these effects in humans are required.pdfengMDPIAttribution-NonCommercial-NoDerivatives 4.0 Internacionalhttp://creativecommons.org/licenses/by-nc-nd/4.0/info:eu-repo/semantics/openAccesshttp://purl.org/coar/access_right/c_abf2International Journal of Molecular SciencesVol. 22 N° 9 (2021)Non-alcoholic fatty liver diseaseEndocrine-disrupting chemicalsLiver disorderEnvironmental pollutantsExposureRole of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Reviewinfo:eu-repo/semantics/articleArtículo científicohttp://purl.org/coar/version/c_970fb48d4fbd8a85http://purl.org/coar/resource_type/c_2df8fbb1Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The Diagnosis and Management of Nonalcoholic Fatty Liver Disease: Practice Guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [CrossRef]Amini, M.; Ansari, I.; Yekesadat, S.; Vaseie, M.; Malekhoseyni, M. Response rate to the vaccination with hepatitis b vaccine among cardiovascular health staff in Tehran. Latinoam. Hipertens. 2020, 14, 562–567.Younossi, Z.; Tacke, F.; Arrese, M.; Chander Sharma, B.; Mostafa, I.; Bugianesi, E.; Wai-Sun Wong, V.; Yilmaz, Y.; George, J.; Fan, J.; et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: Hepatology. Hepatology 2019, 69, 2672–2682. [CrossRef] [PubMed]Bermudez, V.; Moisés, R.; Lisney, V.; Yettana, L.; Ana, C.; ali, U.; Guerra-Torres, X.; Colmenares, C.A.; Chacín, M.; Rojas Quintero, J.; et al. Pharmacologic Treatment of Obesity: Pitfalls and New Promises. Rev. Latinoam. Hipertens. 2008, 5, 137–147Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global Burden of NAFLD and NASH: Trends, Predictions, Risk Factors and Prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [CrossRef]Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global Epidemiology of Nonalcoholic Fatty Liver Disease-Meta-Analytic Assessment of Prevalence, Incidence, and Outcomes. Hepatology 2016, 64, 73–84. [CrossRef] [PubMed]Fang, Y.-L.; Chen, H.; Wang, C.-L.; Liang, L. Pathogenesis of Non-Alcoholic Fatty Liver Disease in Children and Adolescence: From “Two Hit Theory” to “Multiple Hit Model”. World J. Gastroenterol. 2018, 24, 2974–2983. [CrossRef]Carr, R.M.; Oranu, A.; Khungar, V. Nonalcoholic Fatty Liver Disease. Gastroenterol. Clin. N. Am. 2016, 45, 639–652. [CrossRef]Souki-Rincón, A.; Sandoval, M.; Sánchez, G.; Andrade, U.; García-Rondón, D.; Cano-Ponce, C.; Medina, M.; Almarza, J.; Urdaneta, Y.; González, C. Intake of saturated fatty acids and insulin sensitivity in obese young adults from Maracaibo. Rev. Latinoam. Hipertens. 2008, 3, 159–165Petta, S.; Gastaldelli, A.; Rebelos, E.; Bugianesi, E.; Messa, P.; Miele, L.; Svegliati-Baroni, G.; Valenti, L.; Bonino, F. Pathophysiology of Non Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2016, 17, 2082. [CrossRef]Masarone, M.; Rosato, V.; Dallio, M.; Gravina, A.G.; Aglitti, A.; Loguercio, C.; Federico, A.; Persico, M. Role of Oxidative Stress in Pathophysiology of Nonalcoholic Fatty Liver Disease. Oxid. Med. Cell. Longev. 2018, 2018, 1–14. [CrossRef]Arciello, M.; Gori, M.; Maggio, R.; Barbaro, B.; Tarocchi, M.; Galli, A.; Balsano, C. Environmental Pollution: A Tangible Risk for NAFLD Pathogenesis. Int. J. Mol. Sci. 2013, 14, 22052–22066. [CrossRef]Papalou, O.; Kandaraki, E.A.; Papadakis, G.; Diamanti-Kandarakis, E. Endocrine Disrupting Chemicals: An Occult Mediator of Metabolic Disease. Front. Endocrinol. 2019, 10, 112. [CrossRef]Bergman, Å.; Heindel, J.J.; Kasten, T.; Kidd, K.A.; Jobling, S.; Neira, M.; Zoeller, R.T.; Becher, G.; Bjerregaard, P.; Bornman, R.; et al. The Impact of Endocrine Disruption: A Consensus Statement on the State of the Science. Environ. Health Perspect. 2013, 121, A104–A106. [CrossRef]VoPham, T. Environmental Risk Factors for Liver Cancer and Nonalcoholic Fatty Liver Disease. Curr. Epidemiol. Rep. 2019, 6, 50–66. [CrossRef] [PubMed]Klaunig, J.E.; Li, X.; Wang, Z. Role of Xenobiotics in the Induction and Progression of Fatty Liver Disease. Toxicol. Res. 2018, 7, 664–680. [CrossRef] [PubMed]Wei, J.; Sun, X.; Chen, Y.; Li, Y.; Song, L.; Zhou, Z.; Xu, B.; Lin, Y.; Xu, S. Perinatal Exposure to Bisphenol A Exacerbates Nonalcoholic Steatohepatitis-like Phenotype in Male Rat Offspring Fed on a High-Fat Diet. J. Endocrinol. 2014, 222, 313–325. [CrossRef] [PubMed]Marty, M.S.; Carney, E.W.; Rowlands, J.C. Endocrine Disruption: Historical Perspectives and Its Impact on the Future of Toxicology Testing. Toxicol. Sci. Off. J. Soc. Toxicol. 2011, 120 (Suppl. 1), S93–S108. [CrossRef] [PubMed]Darbre, P.D. The History of Endocrine-Disrupting Chemicals. Curr. Opin. Endocr. Metab. Res. 2019, 7, 26–33. [CrossRef]Lee, D.H. Evidence of the Possible Harm of Endocrine-Disrupting Chemicals in Humans: Ongoing Debates and Key Issues. Endocrinol. Metab. Seoul Korea 2018, 33, 44–52. [CrossRef] [PubMed]Committee, E.S. Scientific Opinion on the Hazard Assessment of Endocrine Disruptors: Scientific Criteria for Identification of Endocrine Disruptors and Appropriateness of Existing Test Methods for Assessing Effects Mediated by These Substances on Human Health and the Environment. EFSA J. 2013, 11, 3132. [CrossRef]Slama, R.; Bourguignon, J.-P.; Demeneix, B.; Ivell, R.; Panzica, G.; Kortenkamp, A.; Zoeller, R.T. Scientific Issues Relevant to Setting Regulatory Criteria to Identify Endocrine-Disrupting Substances in the European Union. Environ. Health Perspect. 2016, 124, 1497–1503. [CrossRef]Zoeller, R.T.; Brown, T.R.; Doan, L.L.; Gore, A.C.; Skakkebaek, N.E.; Soto, A.M.; Woodruff, T.J.; Vom Saal, F.S. EndocrineDisrupting Chemicals and Public Health Protection: A Statement of Principles from The Endocrine Society. Endocrinology 2012, 153, 4097–4110. [CrossRef] [PubMed]Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. Executive Summary to EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015, 36, 593–602. [CrossRef]Yang, O.; Kim, H.L.; Weon, J.-I.; Seo, Y.R. Endocrine-Disrupting Chemicals: Review of Toxicological Mechanisms Using Molecular Pathway Analysis. J. Cancer Prev. 2015, 20, 12–24. [CrossRef] [PubMed]Frye, C.; Bo, E.; Calamandrei, G.; Calzà, L.; Dessì-Fulgheri, F.; Fernández, M.; Fusani, L.; Kah, O.; Kajta, M.; Le Page, Y.; et al. Endocrine Disrupters: A Review of Some Sources, Effects, and Mechanisms of Actions on Behaviour and Neuroendocrine Systems: EDCs-Sources, Effects & Mechanisms. J. Neuroendocrinol. 2012, 24, 144–159. [CrossRef]Barouki, R. Endocrine Disruptors: Revisiting Concepts and Dogma in Toxicology. Comptes Rendus Biol. 2017, 340, 410–413. [CrossRef] [PubMed]Yilmaz, B.; Terekeci, H.; Sandal, S.; Kelestimur, F. Endocrine Disrupting Chemicals: Exposure, Effects on Human Health, Mechanism of Action, Models for Testing and Strategies for Prevention. Rev. Endocr. Metab. Disord. 2020, 21, 127–147. [CrossRef]Diamanti-Kandarakis, E.; Palioura, E.; Kandarakis, S.A.; Koutsilieris, M. The Impact of Endocrine Disruptors on Endocrine Targets. Horm. Metab. Res. 2010, 42, 543–552. [CrossRef] [PubMed]Sargis, R.M.; Heindel, J.J.; Padmanabhan, V. Interventions to Address Environmental Metabolism-Disrupting Chemicals: Changing the Narrative to Empower Action to Restore Metabolic Health. Front. Endocrinol. 2019, 10, 33. [CrossRef] [PubMed]Kabir, E.R.; Rahman, M.S.; Rahman, I. A Review on Endocrine Disruptors and Their Possible Impacts on Human Health. Environ. Toxicol. Pharmacol. 2015, 40, 241–258. [CrossRef]Diamanti-Kandarakis, E.; Bourguignon, J.-P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. EndocrineDisrupting Chemicals: An Endocrine Society Scientific Statement. Endocr. Rev. 2009, 30, 293–342. [CrossRef] [PubMed]Lauretta, R.; Sansone, A.; Sansone, M.; Romanelli, F.; Appetecchia, M. Endocrine Disrupting Chemicals: Effects on Endocrine Glands. Front. Endocrinol. 2019, 10, 178. [CrossRef] [PubMed]Jalal, N.; Surendranath, A.R.; Pathak, J.L.; Yu, S.; Chung, C.Y. Bisphenol A (BPA) the Mighty and the Mutagenic. Toxicol. Rep. 2018, 5, 76–84. [CrossRef] [PubMed]Rudel, R.A.; Gray, J.M.; Engel, C.L.; Rawsthorne, T.W.; Dodson, R.E.; Ackerman, J.M.; Rizzo, J.; Nudelman, J.L.; Brody, J.G. Food Packaging and Bisphenol A and Bis(2-Ethyhexyl) Phthalate Exposure: Findings from a Dietary Intervention. Environ. Health Perspect. 2011, 119, 914–920. [CrossRef] [PubMed]Chen, Y.; Li, C.; Song, P.; Yan, B.; Yang, X.; Wu, Y.; Ma, P. Hepatic and Renal Tissue Damage in Balb/c Mice Exposed to Diisodecyl Phthalate: The Role of Oxidative Stress Pathways. Food Chem. Toxicol. 2019, 132, 110600. [CrossRef] [PubMed]Wang, C.; Yang, L.; Wang, S.; Zhang, Z.; Yu, Y.; Wang, M.; Cromie, M.; Gao, W.; Wang, S.-L. The Classic EDCs, Phthalate Esters and Organochlorines, in Relation to Abnormal Sperm Quality: A Systematic Review with Meta-Analysis. Sci. Rep. 2016, 6, 19982. [CrossRef]Qin, W.; Deng, T.; Cui, H.; Zhang, Q.; Liu, X.; Yang, X.; Chen, M. Exposure to Diisodecyl Phthalate Exacerbated Th2 and Th17-Mediated Asthma through Aggravating Oxidative Stress and the Activation of P38 MAPK. Food Chem. Toxicol. 2018, 114, 78–87. [CrossRef] [PubMed]Amaro, A.A.; Esposito, A.I.; Mirisola, V.; Mehilli, A.; Rosano, C.; Noonan, D.M.; Albini, A.; Pfeffer, U.; Angelini, G. Endocrine Disruptor Agent Nonyl Phenol Exerts An Estrogen-like Transcriptional Activity on Estrogen Receptor Positive Breast Cancer Cells. Curr. Med. Chem. 2014, 21, 630–640. [CrossRef]Kazemi, S.; Mousavi Kani, S.N.; Ghasemi-Kasman, M.; Aghapour, F.; Khorasani, H.; Moghadamnia, A.A. Nonylphenol Induces Liver Toxicity and Oxidative Stress in Rat. Biochem. Biophys. Res. Commun. 2016, 479, 17–21. [CrossRef]Monneret, C. What Is an Endocrine Disruptor? Comptes Rendus Biol. 2017, 340, 403–405. [CrossRef]Bonefeld-Jørgensen, E.C.; Ghisari, M.; Wielsøe, M.; Bjerregaard-Olesen, C.; Kjeldsen, L.S.; Long, M. Biomonitoring and HormoneDisrupting Effect Biomarkers of Persistent Organic Pollutants In Vitro and Ex Vivo. Basic Clin. Pharmacol. Toxicol. 2014, 115, 118–128. [CrossRef]Deierlein, A.L.; Rock, S.; Park, S. Persistent Endocrine-Disrupting Chemicals and Fatty Liver Disease. Curr. Environ. Health Rep. 2017, 4, 439–449. [CrossRef]Hung, H.; Katsoyiannis, A.A.; Guardans, R. Ten Years of Global Monitoring under the Stockholm Convention on Persistent Organic Pollutants (POPs): Trends, Sources and Transport Modelling. Environ. Pollut. 2016, 217, 1–3. [CrossRef] [PubMed]Magulova, K.; Priceputu, A. Global Monitoring Plan for Persistent Organic Pollutants (POPs) under the Stockholm Convention: Triggering, Streamlining and Catalyzing Global POPs Monitoring. Environ. Pollut. 2016, 217, 82–84. [CrossRef] [PubMed]Rahman, Z.; Singh, V.P. The Relative Impact of Toxic Heavy Metals (THMs) (Arsenic (As), Cadmium (Cd), Chromium (Cr)(VI), Mercury (Hg), and Lead (Pb)) on the Total Environment: An Overview. Environ. Monit. Assess. 2019, 191, 419. [CrossRef] [PubMed]Barouki, R. Endocrine disruptor compounds and new mechanisms of toxicity networks. Rev. Prat. 2018, 68, 1069–1074.Sawada, N. Association between Arsenic Intake and Cancer—From the Viewpoint of Epidemiological Study. Nippon Eiseigaku Zasshi 2018, 73, 265–268. [CrossRef] [PubMed]Kim, K.-H.; Jahan, S.A.; Kabir, E.; Brown, R.J.C. A Review of Airborne Polycyclic Aromatic Hydrocarbons (PAHs) and Their Human Health Effects. Environ. Int. 2013, 60, 71–80. [CrossRef]Alegbeleye, O.O.; Opeolu, B.O.; Jackson, V.A. Polycyclic Aromatic Hydrocarbons: A Critical Review of Environmental Occurrence and Bioremediation. Environ. Manag. 2017, 60, 758–783. [CrossRef]Lau, C. Perfluorinated Compounds. Exp. Suppl. 2012, 101, 47–86. [CrossRef]Chen, A.; Zhou, X.; Cheng, Y.; Tang, S.; Liu, M.; Wang, X. Design and Optimization of the Cocktail Assay for Rapid Assessment of the Activity of UGT Enzymes in Human and Rat Liver Microsomes. Toxicol. Lett. 2018, 295, 379–389. [CrossRef] [PubMed]Rowland, A.; Miners, J.O.; Mackenzie, P.I. The UDP-Glucuronosyltransferases: Their Role in Drug Metabolism and Detoxification. Int. J. Biochem. Cell Biol. 2013, 45, 1121–1132. [CrossRef] [PubMed]Bissig, K.-D.; Han, W.; Barzi, M.; Kovalchuk, N.; Ding, L.; Fan, X.; Pankowicz, F.P.; Zhang, Q.-Y.; Ding, X. P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab. Dispos. 2018, 46, 1734–1744. [CrossRef] [PubMed]Muncke, J. Exposure to Endocrine Disrupting Compounds via the Food Chain: Is Packaging a Relevant Source? Sci. Total Environ. 2009, 407, 4549–4559. [CrossRef]Gálvez-Ontiveros, Y.; Páez, S.; Monteagudo, C.; Rivas, A. Endocrine Disruptors in Food: Impact on Gut Microbiota and Metabolic Diseases. Nutrients 2020, 12, 1158. [CrossRef] [PubMed]Cano-Sancho, G.; Marchand, P.; Le Bizec, B.; Antignac, J.-P. The Challenging Use and Interpretation of Blood Biomarkers of Exposure Related to Lipophilic Endocrine Disrupting Chemicals in Environmental Health Studies. Mol. Cell. Endocrinol. 2020, 499, 110606. [CrossRef]Nishida, K.; Kobayashi, M.; Miyamoto, H.; Yoshikawa, N.; Fumoto, S.; Sasaki, H.; Nakamura, J. Relationship between Lipophilicity and Absorption from the Liver Surface of Paraben Derivatives and Antipyrine in Rats: Lipophilic Drug Absorption from Liver Surface. J. Pharm. Pharmacol. 2011, 63, 736–740. [CrossRef]Lonard, D.M.; O’malley, B.W. Nuclear Receptor Coregulators: Judges, Juries, and Executioners of Cellular Regulation. Mol. Cell 2007, 27, 691–700. [CrossRef]Cave, M.C.; Clair, H.B.; Hardesty, J.E.; Falkner, K.C.; Feng, W.; Clark, B.J.; Sidey, J.; Shi, H.; Aqel, B.A.; McClain, C.J.; et al. Nuclear Receptors and Nonalcoholic Fatty Liver Disease. Biochim. Biophys. Acta 2016, 1859, 1083–1099. [CrossRefCruz-Hurtado, M.; de López-González, M.L.; Mondragón, V.; Sierra-Santoyo, A. In Vitro Phase I Metabolism of Vinclozolin by Human Liver Microsomes. Xenobiotica 2019, 49, 895–904. [CrossRef] [PubMed]Carrão, D.B.; Habenchus, M.D.; de Albuquerque, N.C.P.; da Silva, R.M.; Lopes, N.P.; de Oliveira, A.R.M. In Vitro Inhibition of Human CYP2D6 by the Chiral Pesticide Fipronil and Its Metabolite Fipronil Sulfone: Prediction of Pesticide-Drug Interactions. Toxicol. Lett. 2019, 313, 196–204. [CrossRef] [PubMed]Sychev, D.; Ashraf, G.M.; Svistunov, A.; Maksimov, M.; Tarasov, V.; Chubarev, V.N.; Otdelenov, V.A.; Denisenko, N.P.; Barreto, G.E.; Aliev, G. The Cytochrome P450 Isoenzyme and Some New Opportunities for the Prediction of Negative Drug Interaction in Vivo. Drug Des. Devel. Ther. 2018, 12, 1147–1156. [CrossRef]Docea, A.O.; Vassilopoulou, L.; Fragou, D.; Arsene, A.L.; Fenga, C.; Kovatsi, L.; Petrakis, D.; Rakitskii, V.N.; Nosyrev, A.E.; Izotov, B.N.; et al. CYP Polymorphisms and Pathological Conditions Related to Chronic Exposure to Organochlorine Pesticides. Toxicol. Rep. 2017, 4, 335–341. [CrossRef]Lee, P.C.; Chakraborty Patra, S.; Stelloh, C.T.; Lee, W.; Struve, M. Interaction of Nonylphenol and Hepatic CYP1A in Rats. Biochem. Pharmacol. 1996, 52, 885–889. [CrossRef]Ademollo, N.; Patrolecco, L.; Rauseo, J.; Nielsen, J.; Corsolini, S. Bioaccumulation of Nonylphenols and Bisphenol A in the Greenland Shark Somniosus Microcephalus from the Greenland Seawaters. Microchem. J. 2018, 136, 106–112. [CrossRef]Lv, Y.-Z.; Yao, L.; Wang, L.; Liu, W.-R.; Zhao, J.-L.; He, L.-Y.; Ying, G.-G. Bioaccumulation, Metabolism, and Risk Assessment of Phenolic Endocrine Disrupting Chemicals in Specific Tissues of Wild Fish. Chemosphere 2019, 226, 607–615. [CrossRef] [PubMed]Benjamin, S.; Masai, E.; Kamimura, N.; Takahashi, K.; Anderson, R.C.; Faisal, P.A. Phthalates Impact Human Health: Epidemiological Evidences and Plausible Mechanism of Action. J. Hazard. Mater. 2017, 340, 360–383. [CrossRef] [PubMed]Praveena, S.M.; Teh, S.W.; Rajendran, R.K.; Kannan, N.; Lin, C.-C.; Abdullah, R.; Kumar, S. Recent Updates on Phthalate Exposure and Human Health: A Special Focus on Liver Toxicity and Stem Cell Regeneration. Environ. Sci. Pollut. Res. 2018, 25, 11333–11342. [CrossRef]Larsson, K.; Ljung Björklund, K.; Palm, B.; Wennberg, M.; Kaj, L.; Lindh, C.H.; Jönsson, B.A.G.; Berglund, M. Exposure Determinants of Phthalates, Parabens, Bisphenol A and Triclosan in Swedish Mothers and Their Children. Environ. Int. 2014, 73, 323–333. [CrossRef]Lee, Y.-M.; Kim, K.-S.; Jacobs, D.R.; Lee, D.-H. Persistent Organic Pollutants in Adipose Tissue Should Be Considered in Obesity Research. Obes. Rev. Off. J. Int. Assoc. Study Obes. 2017, 18, 129–139. [CrossRef]Shin, M.-Y.; Shin, C.; Choi, J.W.; Lee, J.; Lee, S.; Kim, S. Pharmacokinetic Profile of Propyl Paraben in Humans after Oral Administration. Environ. Int. 2019, 130, 104917. [CrossRef]Nicolucci, C.; Errico, S.; Federico, A.; Dallio, M.; Loguercio, C.; Diano, N. Human Exposure to Bisphenol A and Liver Health Status: Quantification of Urinary and Circulating Levels by LC–MS/MS. J. Pharm. Biomed. Anal. 2017, 140, 105–112. [CrossRef]Ulasoglu, C.; Enc, F.Y.; Kaya, E.; Yilmaz, Y. Characterization of Patients with Biopsy-Proven Non-Alcoholic Fatty Liver Disease and Normal Aminotransferase Levels. J. Gastrointestin. Liver Dis. 2019, 28, 427–431. [CrossRef]Kang, Y.; Park, S.; Kim, S.; Koh, H. Normal Serum Alanine Aminotransferase and Non-Alcoholic Fatty Liver Disease among Korean Adolescents: A Cross-Sectional Study Using Data from KNHANES 2010–2015. BMC Pediatr. 2018, 18, 215. [CrossRef] [PubMed]Kumar, J.; Lind, L.; Salihovic, S.; van Bavel, B.; Ingelsson, E.; Lind, P.M. Persistent Organic Pollutants and Liver Dysfunction Biomarkers in a Population-Based Human Sample of Men and Women. Environ. Res. 2014, 134, 251–256. [CrossRef] [PubMed]Barali´c, K.; Buha Djordjevic, A.; Živanˇcevi´c, K.; Antonijevi´c, E.; Andelkovi´c, M.; Javorac, D.; ¯ Curˇci´c, M.; Bulat, Z.; Antonijevi´c, B.; ´ Ðuki´c-Cosi´c, D. Toxic Effects of the Mixture of Phthalates and Bisphenol A-Subacute Oral Toxicity Study in Wistar Rats. ´ Int. J. Environ. Res. Public Health 2020, 17, 746. [CrossRef]Heindel, J.J.; Blumberg, B.; Cave, M.; Machtinger, R.; Mantovani, A.; Mendez, M.A.; Nadal, A.; Palanza, P.; Panzica, G.; Sargis, R.; et al. Metabolism Disrupting Chemicals and Metabolic Disorders. Reprod. Toxicol. Elmsford N 2017, 68, 3–33. [CrossRef] [PubMed]Lim, S.; Ahn, S.Y.; Song, I.C.; Chung, M.H.; Jang, H.C.; Park, K.S.; Lee, K.-U.; Pak, Y.K.; Lee, H.K. Chronic Exposure to the Herbicide, Atrazine, Causes Mitochondrial Dysfunction and Insulin Resistance. PLoS ONE 2009, 4, e5186. [CrossRef]Jin, Y.; Lin, X.; Miao, W.; Wu, T.; Shen, H.; Chen, S.; Li, Y.; Pan, Q.; Fu, Z. Chronic Exposure of Mice to Environmental Endocrine-Disrupting Chemicals Disturbs Their Energy Metabolism. Toxicol. Lett. 2014, 225, 392–400. [CrossRef]Al-Eryani, L.; Wahlang, B.; Falkner, K.C.; Guardiola, J.J.; Clair, H.B.; Prough, R.A.; Cave, M. Identification of Environmental Chemicals Associated with the Development of Toxicant-Associated Fatty Liver Disease in Rodents. Toxicol. Pathol. 2015, 43, 482–497. [CrossRef] [PubMed]Maradonna, F.; Carnevali, O. Lipid Metabolism Alteration by Endocrine Disruptors in Animal Models: An Overview. Front. Endocrinol. 2018, 9, 654. [CrossRef]Armstrong, L.E.; Guo, G.L. Understanding Environmental Contaminants’ Direct Effects on Non-Alcoholic Fatty Liver Disease Progression. Curr. Environ. Health Rep. 2019, 6, 95–104. [CrossRef]Kawano, Y.; Cohen, D.E. Mechanisms of Hepatic Triglyceride Accumulation in Non-Alcoholic Fatty Liver Disease. J. Gastroenterol. 2013, 48, 434–441. [CrossRef]Ipsen, D.H.; Lykkesfeldt, J.; Tveden-Nyborg, P. Molecular Mechanisms of Hepatic Lipid Accumulation in Non-Alcoholic Fatty Liver Disease. Cell. Mol. Life Sci. CMLS 2018, 75, 3313–3327. [CrossRef]Geisler, C.E.; Renquist, B.J. Hepatic Lipid Accumulation: Cause and Consequence of Dysregulated Glucoregulatory Hormones. J. Endocrinol. 2017, 234, R1–R21. [CrossRef]Polyzos, S.A.; Kountouras, J.; Deretzi, G.; Zavos, C.; Mantzoros, C.S. The Emerging Role of Endocrine Disruptors in Pathogenesis of Insulin Resistance: A Concept Implicating Nonalcoholic Fatty Liver Disease. Curr. Mol. Med. 2012, 12, 68–82. [CrossRef] [PubMed]Wahlang, B.; Hardesty, J.E.; Jin, J.; Falkner, K.C.; Cave, M.C. Polychlorinated Biphenyls and Nonalcoholic Fatty Liver Disease. Curr. Opin. Toxicol. 2019, 14, 21–28. [CrossRef]Shan, Q.; Chen, N.; Liu, W.; Qu, F.; Chen, A. Exposure to 2,3,30 ,4,40 ,5-Hexachlorobiphenyl Promotes Nonalcoholic Fatty Liver Disease Development in C57BL/6 Mice. Environ. Pollut. 2020, 263, 114563. [CrossRef] [PubMed]Wan, H.T.; Zhao, Y.G.; Wei, X.; Hui, K.Y.; Giesy, J.P.; Wong, C.K.C. PFOS-Induced Hepatic Steatosis, the Mechanistic Actions on β-Oxidation and Lipid Transport. Biochim. Biophys. Acta BBA Gen. Subj. 2012, 1820, 1092–1101. [CrossRef] [PubMed]Das, K.P.; Wood, C.R.; Lin, M.T.; Starkov, A.A.; Lau, C.; Wallace, K.B.; Corton, J.C.; Abbott, B.D. Perfluoroalkyl Acids-Induced Liver Steatosis: Effects on Genes Controlling Lipid Homeostasis. Toxicology 2017, 378, 37–52. [CrossRef]Yan, S.; Wang, J.; Dai, J. Activation of Sterol Regulatory Element-Binding Proteins in Mice Exposed to Perfluorooctanoic Acid for 28 Days. Arch. Toxicol. 2015, 89, 1569–1578. [CrossRef] [PubMed]Zhang, H.; He, J.; Li, N.; Gao, N.; Du, Q.; Chen, B.; Chen, F.; Shan, X.; Ding, Y.; Zhu, W.; et al. Lipid Accumulation Responses in the Liver of Rana Nigromaculata Induced by Perfluorooctanoic Acid (PFOA). Ecotoxicol. Environ. Saf. 2019, 167, 29–35. [CrossRef]Chen, H.; Zhang, W.; Rui, B.; Yang, S.; Xu, W.; Wei, W. Di(2-Ethylhexyl) Phthalate Exacerbates Non-Alcoholic Fatty Liver in Rats and Its Potential Mechanisms. Environ. Toxicol. Pharmacol. 2016, 42, 38–44. [CrossRef]Zhang, Y.; Wang, S.; Zhao, T.; Yang, L.; Guo, S.; Shi, Y.; Zhang, X.; Zhou, L.; Ye, L. Mono-2-Ethylhexyl Phthalate (MEHP) Promoted Lipid Accumulation via JAK2/STAT5 and Aggravated Oxidative Stress in BRL-3A Cells. Ecotoxicol. Environ. Saf. 2019, 184, 109611. [CrossRef]Bai, J.; He, Z.; Li, Y.; Jiang, X.; Yu, H.; Tan, Q. Mono-2-Ethylhexyl Phthalate Induces the Expression of Genes Involved in Fatty Acid Synthesis in HepG2 Cells. Environ. Toxicol. Pharmacol. 2019, 69, 104–111. [CrossRef]Dallio, M.; Diano, N.; Masarone, M.; Gravina, A.G.; Patanè, V.; Romeo, M.; Di Sarno, R.; Errico, S.; Nicolucci, C.; Abenavoli, L.; et al. Chemical Effect of Bisphenol A on Non-Alcoholic Fatty Liver Disease. Int. J. Environ. Res. Public Health 2019, 16, 3134. [CrossRef] [PubMed]Marmugi, A.; Ducheix, S.; Lasserre, F.; Polizzi, A.; Paris, A.; Priymenko, N.; Bertrand-Michel, J.; Pineau, T.; Guillou, H.; Martin, P.G.P.; et al. Low Doses of Bisphenol A Induce Gene Expression Related to Lipid Synthesis and Trigger Triglyceride Accumulation in Adult Mouse Liver. Hepatol. Baltim. Md 2012, 55, 395–407. [CrossRef] [PubMed]Martella, A.; Silvestri, C.; Maradonna, F.; Gioacchini, G.; Allarà, M.; Radaelli, G.; Overby, D.R.; Di Marzo, V.; Carnevali, O. Bisphenol A Induces Fatty Liver by an Endocannabinoid-Mediated Positive Feedback Loop. Endocrinology 2016, 157, 1751–1763. [CrossRef] [PubMed]Wei, Y.; Rector, R.S.; Thyfault, J.P.; Ibdah, J.A. Nonalcoholic Fatty Liver Disease and Mitochondrial Dysfunction. World J. Gastroenterol. 2008, 14, 193–199. [CrossRef] [PubMed]Einer, C.; Hohenester, S.; Wimmer, R.; Wottke, L.; Artmann, R.; Schulz, S.; Gosmann, C.; Simmons, A.; Leitzinger, C.; Eberhagen, C.; et al. Mitochondrial Adaptation in Steatotic Mice. Mitochondrion 2018, 40, 1–12. [CrossRef]Simões, I.C.M.; Fontes, A.; Pinton, P.; Zischka, H.; Wieckowski, M.R. Mitochondria in Non-Alcoholic Fatty Liver Disease. Int. J. Biochem. Cell Biol. 2018, 95, 93–99. [CrossRef] [PubMed]Mansouri, A.; Gattolliat, C.-H.; Asselah, T. Mitochondrial Dysfunction and Signaling in Chronic Liver Diseases. Gastroenterology 2018, 155, 629–647. [CrossRef] [PubMed]Marroqui, L.; Tudurí, E.; Alonso-Magdalena, P.; Quesada, I.; Nadal, Á.; Dos Santos, R.S. Mitochondria as Target of EndocrineDisrupting Chemicals: Implications for Type 2 Diabetes. J. Endocrinol. 2018, 239, R27–R45. [CrossRef]Lim, S.; Cho, Y.M.; Park, K.S.; Lee, H.K. Persistent Organic Pollutants, Mitochondrial Dysfunction, and Metabolic Syndrome. Ann. N. Y. Acad. Sci. 2010, 1201, 166–176. [CrossRef] [PubMed]Sagarkar, S.; Gandhi, D.; Devi, S.S.; Sakharkar, A.; Kapley, A. Atrazine Exposure Causes Mitochondrial Toxicity in Liver and Muscle Cell Lines. Indian J. Pharmacol. 2016, 48, 200–207. [CrossRef] [PubMed]Khan, S.; Beigh, S.; Chaudhari, B.P.; Sharma, S.; Aliul Hasan Abdi, S.; Ahmad, S.; Ahmad, F.; Parvez, S.; Raisuddin, S. Mitochondrial Dysfunction Induced by Bisphenol A Is a Factor of Its Hepatotoxicity in Rats. Environ. Toxicol. 2016, 31, 1922–1934. [CrossRef]Jiang, Y.; Xia, W.; Zhu, Y.; Li, X.; Wang, D.; Liu, J.; Chang, H.; Li, G.; Xu, B.; Chen, X.; et al. Mitochondrial Dysfunction in Early Life Resulted from Perinatal Bisphenol A Exposure Contributes to Hepatic Steatosis in Rat Offspring. Toxicol. Lett. 2014, 228, 85–92. [CrossRef]Yu, J.; Yang, X.; Yang, X.; Yang, M.; Wang, P.; Yang, Y.; Yang, J.; Li, W.; Xu, J. Nonylphenol Aggravates Non-Alcoholic Fatty Liver Disease in High Sucrose-High Fat Diet-Treated Rats. Sci. Rep. 2018, 8, 3232. [CrossRef] [PubMed]Kourouma, A.; Keita, H.; Duan, P.; Quan, C.; Bilivogui, K.K.; Qi, S.; Christiane, N.A.; Osamuyimen, A.; Yang, K. Effects of 4-Nonylphenol on Oxidant/Antioxidant Balance System Inducing Hepatic Steatosis in Male Rat. Toxicol. Rep. 2015, 2, 1423–1433. [CrossRef]He, X.; Gao, J.; Hou, H.; Qi, Z.; Chen, H.; Zhang, X.-X. Inhibition of Mitochondrial Fatty Acid Oxidation Contributes to Development of Nonalcoholic Fatty Liver Disease Induced by Environmental Cadmium Exposure. Environ. Sci. Technol. 2019, 53, 13992–14000. [CrossRef] [PubMed]Ding, R.-B.; Bao, J.; Deng, C.-X. Emerging Roles of SIRT1 in Fatty Liver Diseases. Int. J. Biol. Sci. 2017, 13, 852–867. [CrossRef] [PubMed]Banks, A.S.; Kon, N.; Knight, C.; Matsumoto, M.; Gutiérrez-Juárez, R.; Rossetti, L.; Gu, W.; Accili, D. SirT1 Gain of Function Increases Energy Efficiency and Prevents Diabetes in Mice. Cell Metab. 2008, 8, 333–341. [CrossRef] [PubMed]Wang, H.; Qiang, L.; Farmer, S.R. Identification of a Domain within Peroxisome Proliferator-Activated Receptor γ Regulating Expression of a Group of Genes Containing Fibroblast Growth Factor 21 That Are Selectively Repressed by SIRT1 in Adipocytes. Mol. Cell. Biol. 2008, 28, 188–200. [CrossRef]Milne, J.C.; Lambert, P.D.; Schenk, S.; Carney, D.P.; Smith, J.J.; Gagne, D.J.; Jin, L.; Boss, O.; Perni, R.B.; Vu, C.B.; et al. Small Molecule Activators of SIRT1 as Therapeutics for the Treatment of Type 2 Diabetes. Nature 2007, 450, 712–716. [CrossRef]Li, X.; Zhang, S.; Blander, G.; Tse, J.G.; Krieger, M.; Guarente, L. SIRT1 Deacetylates and Positively Regulates the Nuclear Receptor LXR. Mol. Cell 2007, 28, 91–106. [CrossRef] [PubMed]Wang, L.; Sun, M.; Cao, Y.; Ma, L.; Shen, Y.; Velikanova, A.A.; Li, X.; Sun, C.; Zhao, Y. MiR-34a Regulates Lipid Metabolism by Targeting SIRT1 in Non-Alcoholic Fatty Liver Disease with Iron Overload. Arch. Biochem. Biophys. 2020, 695, 108642. [CrossRef]Yoshizaki, T.; Milne, J.C.; Imamura, T.; Schenk, S.; Sonoda, N.; Babendure, J.L.; Lu, J.-C.; Smith, J.J.; Jirousek, M.R.; Olefsky, J.M. SIRT1 Exerts Anti-Inflammatory Effects and Improves Insulin Sensitivity in Adipocytes. Mol. Cell. Biol. 2009, 29, 1363–1374. [CrossRef]Yamazaki, Y.; Usui, I.; Kanatani, Y.; Matsuya, Y.; Tsuneyama, K.; Fujisaka, S.; Bukhari, A.; Suzuki, H.; Senda, S.; Imanishi, S.; et al. Treatment with SRT1720, a SIRT1 Activator, Ameliorates Fatty Liver with Reduced Expression of Lipogenic Enzymes in MSG Mice. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E1179–E1186. [CrossRef]Colak, Y.; Ozturk, O.; Senates, E.; Tuncer, I.; Yorulmaz, E.; Adali, G.; Doganay, L.; Enc, F.Y. SIRT1 as a Potential Therapeutic Target for Treatment of Nonalcoholic Fatty Liver Disease. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2011, 17, HY5–HY9. [CrossRef]Bai, P.; Cantó, C.; Oudart, H.; Brunyánszki, A.; Cen, Y.; Thomas, C.; Yamamoto, H.; Huber, A.; Kiss, B.; Houtkooper, R.H.; et al. PARP-1 Inhibition Increases Mitochondrial Metabolism through SIRT1 Activation. Cell Metab. 2011, 13, 461–468. [CrossRef]. Peverill, W.; Powell, L.W.; Skoien, R. Evolving Concepts in the Pathogenesis of NASH: Beyond Steatosis and Inflammation. Int. J. Mol. Sci. 2014, 15, 8591–8638. [CrossRef]Luedde, T.; Schwabe, R.F. NF-KB in the Liver–Linking Injury, Fibrosis and Hepatocellular Carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 108–118. [CrossRef] [PubMed]Huc, L.; Lemarié, A.; Guéraud, F.; Héliès-Toussaint, C. Low Concentrations of Bisphenol A Induce Lipid Accumulation Mediated by the Production of Reactive Oxygen Species in the Mitochondria of HepG2 Cells. Toxicol. Vitro Int. J. Publ. Assoc. BIBRA 2012, 26, 709–717. [CrossRef] [PubMed]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-KB/TNF-α-Mediated Pathway. Chemosphere 2018, 191, 1056–1064. [CrossRef] [PubMed]Deng, P.; Barney, J.; Petriello, M.C.; Morris, A.J.; Wahlang, B.; Hennig, B. Hepatic Metabolomics Reveals That Liver Injury Increases PCB 126-Induced Oxidative Stress and Metabolic Dysfunction. Chemosphere 2019, 217, 140–149. [CrossRef] [PubMed]Park, C.G.; Sung, B.; Ryu, C.S.; Kim, Y.J. Mono-(2-Ethylhexyl) Phthalate Induces Oxidative Stress and Lipid Accumulation in Zebrafish Liver Cells. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2020, 230, 108704. [CrossRef] [PubMed]Arrese, M.; Cabrera, D.; Kalergis, A.M.; Feldstein, A.E. Innate Immunity and Inflammation in NAFLD/NASH. Dig. Dis. Sci. 2016, 61, 1294–1303. [CrossRef]Schuster, S.; Cabrera, D.; Arrese, M.; Feldstein, A.E. Triggering and Resolution of Inflammation in NASH. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 349–364. [CrossRef]Alisi, A.; Carpino, G.; Oliveira, F.L.; Panera, N.; Nobili, V.; Gaudio, E. The Role of Tissue Macrophage-Mediated Inflammation on NAFLD Pathogenesis and Its Clinical Implications. Mediat. Inflamm. 2017, 2017, 8162421. [CrossRef] [PubMed]Katsarou, A.; Moustakas, I.I.; Pyrina, I.; Lembessis, P.; Koutsilieris, M.; Chatzigeorgiou, A. Metabolic Inflammation as an Instigator of Fibrosis during Non-Alcoholic Fatty Liver Disease. World J. Gastroenterol. 2020, 26, 1993–2011. [CrossRef]Meyer, S.K.; Probert, P.M.E.; Lakey, A.F.; Axon, A.R.; Leitch, A.C.; Williams, F.M.; Jowsey, P.A.; Blain, P.G.; Kass, G.E.N.; Wright, M.C. Hepatic Effects of Tartrazine (E 102) after Systemic Exposure Are Independent of Oestrogen Receptor Interactions in the Mouse. Toxicol. Lett. 2017, 273, 55–68. [CrossRef]Acaroz, U.; Ince, S.; Arslan-Acaroz, D.; Gurler, Z.; Demirel, H.H.; Kucukkurt, I.; Eryavuz, A.; Kara, R.; Varol, N.; Zhu, K. Bisphenol-A Induced Oxidative Stress, Inflammatory Gene Expression, and Metabolic and Histopathological Changes in Male Wistar Albino Rats: Protective Role of Boron. Toxicol. Res. 2019, 8, 262–269. [CrossRef]Lv, Q.; Gao, R.; Peng, C.; Yi, J.; Liu, L.; Yang, S.; Li, D.; Hu, J.; Luo, T.; Mei, M.; et al. Bisphenol A Promotes Hepatic Lipid Deposition Involving Kupffer Cells M1 Polarization in Male Mice. J. Endocrinol. 2017, 234, 143–154. [CrossRef]Han, R.; Zhang, F.; Wan, C.; Liu, L.; Zhong, Q.; Ding, W. Effect of Perfluorooctane Sulphonate-Induced Kupffer Cell Activation on Hepatocyte Proliferation through the NF-KB/TNF-α/IL-6-Dependent Pathway. Chemosphere 2018, 200, 283–294. [CrossRef] [PubMed]Zhao, Z.-B.; Ji, K.; Shen, X.-Y.; Zhang, W.-W.; Wang, R.; Xu, W.-P.; Wei, W. Di(2-Ethylhexyl) Phthalate Promotes Hepatic Fibrosis by Regulation of Oxidative Stress and Inflammation Responses in Rats. Environ. Toxicol. Pharmacol. 2019, 68, 109–119. [CrossRef] [PubMed]Gaitantzi, H.; Hakenberg, P.; Theobald, J.; Heinlein, H.; Cai, C.; Loff, S.; Wölfl, S.; Ebert, M.P.; Breitkopf-Heinlein, K.; Subotic, U. Di (2-Ethylhexyl) Phthalate and Its Role in Developing Cholestasis: An In Vitro Study on Different Liver Cell Types. J. Pediatr. Gastroenterol. Nutr. 2018, 66, e28–e35. [CrossRef] [PubMed]Lee, C.-Y.; Suk, F.-M.; Twu, Y.-C.; Liao, Y.-J. Long-Term Exposure to Low-Dose Di-(2-Ethylhexyl) Phthalate Impairs Cholesterol Metabolism in Hepatic Stellate Cells and Exacerbates Liver Librosis. Int. J. Environ. Res. Public Health 2020, 17, 3802. [CrossRef] [PubMed]Harvey, W.A.; Jurgensen, K.; Pu, X.; Lamb, C.L.; Cornell, K.A.; Clark, R.J.; Klocke, C.; Mitchell, K.A. Exposure to 2,3,7,8- Tetrachlorodibenzo-p-Dioxin (TCDD) Increases Human Hepatic Stellate Cell Activation. Toxicology 2016, 344–346, 26–33. [CrossRef]Doskey, C.M.; Fader, K.A.; Nault, R.; Lydic, T.; Matthews, J.; Potter, D.; Sharratt, B.; Williams, K.; Zacharewski, T. 2,3,7,8- Tetrachlorodibenzo-p-Dioxin (TCDD) Alters Hepatic Polyunsaturated Fatty Acid Metabolism and Eicosanoid Biosynthesis in Female Sprague-Dawley Rats. Toxicol. Appl. Pharmacol. 2020, 398, 115034. [CrossRef] [PubMed]Foulds, C.E.; Treviño, L.S.; York, B.; Walker, C.L. Endocrine-Disrupting Chemicals and Fatty Liver Disease. Nat. Rev. Endocrinol. 2017, 13, 445–457. [CrossRef] [PubMed]Walker, C.L. Minireview: Epigenomic Plasticity and Vulnerability to EDC Exposures. Mol. Endocrinol. Baltim. Md. 2016, 30, 848–855. [CrossRef] [PubMed]Treviño, L.S.; Dong, J.; Kaushal, A.; Katz, T.A.; Jangid, R.K.; Robertson, M.J.; Grimm, S.L.; Ambati, C.S.R.; Putluri, V.; Cox, A.R.; et al. Epigenome Environment Interactions Accelerate Epigenomic Aging and Unlock Metabolically Restricted Epigenetic Reprogramming in Adulthood. Nat. Commun. 2020, 11, 2316. [CrossRef]Skinner, M.K.; Manikkam, M.; Guerrero-Bosagna, C. Epigenetic Transgenerational Actions of Environmental Factors in Disease Etiology. Trends Endocrinol. Metab. TEM 2010, 21, 214–222. [CrossRef]Rissman, E.F.; Adli, M. Minireview: Transgenerational Epigenetic Inheritance: Focus on Endocrine Disrupting Compounds. Endocrinology 2014, 155, 2770–2780. [CrossRef] [PubMed]Skinner, M.K. What Is an Epigenetic Transgenerational Phenotype? F3 or F2. Reprod. Toxicol. Elmsford N 2008, 25, 2–6. [CrossRef]Patterson, T.A.; Twaddle, N.C.; Roegge, C.S.; Callicott, R.J.; Fisher, J.W.; Doerge, D.R. Concurrent Determination of Bisphenol A Pharmacokinetics in Maternal and Fetal Rhesus Monkeys. Toxicol. Appl. Pharmacol. 2013, 267, 41–48. [CrossRef]Dolinoy, D.C.; Huang, D.; Jirtle, R.L. Maternal Nutrient Supplementation Counteracts Bisphenol A-Induced DNA Hypomethylation in Early Development. Proc. Natl. Acad. Sci. USA 2007, 104, 13056–13061. [CrossRef] [PubMed]Doshi, T.; Mehta, S.S.; Dighe, V.; Balasinor, N.; Vanage, G. Hypermethylation of Estrogen Receptor Promoter Region in Adult Testis of Rats Exposed Neonatally to Bisphenol A. Toxicology 2011, 289, 74–82. [CrossRef]Zhu, L.; Liu, Y.; Xue, X.; Yuan, C.; Wang, Z. BPA’s Transgenerational Disturbance to Transcription of Ovarian Steroidogenic Genes in Rare Minnow Gobiocypris Rarus via DNA and Histone Methylation. Sci. Total Environ. 2021, 762, 143055. [CrossRef]Qin, T.; Zhang, X.; Guo, T.; Yang, T.; Gao, Y.; Hao, W.; Xiao, X. Epigenetic Alteration Shaped by the Environmental Chemical Bisphenol A. Front. Genet. 2020, 11, 618966. [CrossRef] [PubMed]Longo, M.; Zatterale, F.; Naderi, J.; Nigro, C.; Oriente, F.; Formisano, P.; Miele, C.; Beguinot, F. Low-Dose Bisphenol-A Promotes Epigenetic Changes at Pparγ Promoter in Adipose Precursor Cells. Nutrients 2020, 12, 3498. [CrossRef]McCabe, C.F.; Padmanabhan, V.; Dolinoy, D.C.; Domino, S.E.; Jones, T.R.; Bakulski, K.M.; Goodrich, J.M. Maternal Environmental Exposure to Bisphenols and Epigenome-Wide DNA Methylation in Infant Cord Blood. Environ. Epigenetics 2020, 6, dvaa021. [CrossRef]Cimmino, I.; Fiory, F.; Perruolo, G.; Miele, C.; Beguinot, F.; Formisano, P.; Oriente, F. Potential Mechanisms of Bisphenol A (BPA) Contributing to Human Disease. Int. J. Mol. Sci. 2020, 21, 5761. [CrossRef] [PubMed]Ma, Y.; Xia, W.; Wang, D.Q.; Wan, Y.J.; Xu, B.; Chen, X.; Li, Y.Y.; Xu, S.Q. Hepatic DNA Methylation Modifications in Early Development of Rats Resulting from Perinatal BPA Exposure Contribute to Insulin Resistance in Adulthood. Diabetologia 2013, 56, 2059–2067. [CrossRef]Ke, Z.-H.; Pan, J.-X.; Jin, L.-Y.; Xu, H.-Y.; Yu, T.-T.; Ullah, K.; Rahman, T.U.; Ren, J.; Cheng, Y.; Dong, X.-Y.; et al. Bisphenol A Exposure May Induce Hepatic Lipid Accumulation via Reprogramming the DNA Methylation Patterns of Genes Involved in Lipid Metabolism. Sci. Rep. 2016, 6, 31331. [CrossRef] [PubMed]Chen, H. Chronic Inorganic Arsenic Exposure Induces Hepatic Global and Individual Gene Hypomethylation: Implications for Arsenic Hepatocarcinogenesis. Carcinogenesis 2004, 25, 1779–1786. [CrossRef]Ditzel, E.J.; Nguyen, T.; Parker, P.; Camenisch, T.D. Effects of Arsenite Exposure during Fetal Development on Energy Metabolism and Susceptibility to Diet-Induced Fatty Liver Disease in Male Mice. Environ. Health Perspect. 2016, 124, 201–209. [CrossRef]Gu, H.; Liu, Y.; Wang, W.; Ding, L.; Teng, W.; Liu, L. In Utero Exposure to Di-(2-Ethylhexyl) Phthalate Induces Metabolic Disorder and Increases Fat Accumulation in Visceral Depots of C57BL/6J Mice Offspring. Exp. Ther. Med. 2016, 12, 3806–3812. [CrossRef]Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Plastics Derived Endocrine Disruptors (BPA, DEHP and DBP) Induce Epigenetic Transgenerational Inheritance of Obesity, Reproductive Disease and Sperm Epimutations. PLoS ONE 2013, 8, e55387. [CrossRef]King, S.E.; McBirney, M.; Beck, D.; Sadler-Riggleman, I.; Nilsson, E.; Skinner, M.K. Sperm Epimutation Biomarkers of Obesity and Pathologies Following DDT Induced Epigenetic Transgenerational Inheritance of Disease. Environ. Epigenetics 2019, 5, dvz008. [CrossRef]Strakovsky, R.S.; Wang, H.; Engeseth, N.J.; Flaws, J.A.; Helferich, W.G.; Pan, Y.-X.; Lezmi, S. Developmental Bisphenol A (BPA) Exposure Leads to Sex-Specific Modification of Hepatic Gene Expression and Epigenome at Birth That May Exacerbate High-Fat Diet-Induced Hepatic Steatosis. Toxicol. Appl. Pharmacol. 2015, 284, 101–112. [CrossRef]Maranghi, F.; Lorenzetti, S.; Tassinari, R.; Moracci, G.; Tassinari, V.; Marcoccia, D.; Di Virgilio, A.; Eusepi, A.; Romeo, A.; Magrelli, A.; et al. In Utero Exposure to Di-(2-Ethylhexyl) Phthalate Affects Liver Morphology and Metabolism in Post-Natal CD-1 Mice. Reprod. Toxicol. Elmsford N 2010, 29, 427–432. [CrossRef]Chamorro-García, R.; Sahu, M.; Abbey, R.J.; Laude, J.; Pham, N.; Blumberg, B. Transgenerational Inheritance of Increased Fat Depot Size, Stem Cell Reprogramming, and Hepatic Steatosis Elicited by Prenatal Exposure to the Obesogen Tributyltin in Mice. Environ. Health Perspect. 2013, 121, 359–366. [CrossRef] [PubMed]Zuo, Z.; Chen, S.; Wu, T.; Zhang, J.; Su, Y.; Chen, Y.; Wang, C. Tributyltin Causes Obesity and Hepatic Steatosis in Male Mice. Environ. Toxicol. 2011, 26, 79–85. [CrossRef]Boverhof, D.R.; Burgoon, L.D.; Tashiro, C.; Sharratt, B.; Chittim, B.; Harkema, J.R.; Mendrick, D.L.; Zacharewski, T.R. Comparative Toxicogenomic Analysis of the Hepatotoxic Effects of TCDD in Sprague Dawley Rats and C57BL/6 Mice. Toxicol. Sci. Off. J. Soc. Toxicol. 2006, 94, 398–416. [CrossRef]Tomaszewski, K.E.; Montgomery, C.A.; Melnick, R.L. Modulation of 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Toxicity in F344 Rats by Di(2-Ethylhexyl)Phthalate. Chem. Biol. Interact. 1988, 65, 205–222. [CrossRef]Jones, G.; Greig, J.B. Pathological Changes in the Liver of Mice given 2,3,7,8-Tetrachlorodibenzo-p-Dioxin. Experientia 1975, 31, 1315–1317. [CrossRef] [PubMed]Rodríguez-Alcalá, L.M.; Sá, C.; Pimentel, L.L.; Pestana, D.; Teixeira, D.; Faria, A.; Calhau, C.; Gomes, A. Endocrine Disruptor DDE Associated with a High-Fat Diet Enhances the Impairment of Liver Fatty Acid Composition in Rats. J. Agric. Food Chem. 2015, 63, 9341–9348. [CrossRef]Tan, X.; Xie, G.; Sun, X.; Li, Q.; Zhong, W.; Qiao, P.; Sun, X.; Jia, W.; Zhou, Z. High Fat Diet Feeding Exaggerates Perfluorooctanoic Acid-Induced Liver Injury in Mice via Modulating Multiple Metabolic Pathways. PLoS ONE 2013, 8, e61409. [CrossRef] [PubMed]Lv, Z.; Li, G.; Li, Y.; Ying, C.; Chen, J.; Chen, T.; Wei, J.; Lin, Y.; Jiang, Y.; Wang, Y.; et al. Glucose and Lipid Homeostasis in Adult Rat Is Impaired by Early-Life Exposure to Perfluorooctane Sulfonate. Environ. Toxicol. 2013, 28, 532–542. [CrossRef]Cave, M.; Appana, S.; Patel, M.; Falkner, K.C.; McClain, C.J.; Brock, G. Polychlorinated Biphenyls, Lead, and Mercury Are Associated with Liver Disease in American Adults: NHANES 2003-2004. Environ. Health Perspect. 2010, 118, 1735–1742. [CrossRef]Kim, M.-J.; Marchand, P.; Henegar, C.; Antignac, J.-P.; Alili, R.; Poitou, C.; Bouillot, J.-L.; Basdevant, A.; Le Bizec, B.; Barouki, R.; et al. Fate and Complex Pathogenic Effects of Dioxins and Polychlorinated Biphenyls in Obese Subjects before and after Drastic Weight Loss. Environ. Health Perspect. 2011, 119, 377–383. [CrossRef]Pazderova-Vejlupková, J.; Lukás, E.; Nemcova, M.; P ˘ ícková, J.; Jirásek, L. The Development and Prognosis of Chronic Intoxication by Tetrachlordibenzo-p-Dioxin in Men. Arch. Environ. Health 1981, 36, 5–11. [CrossRef]Lee, C.-C.; Yao, Y.-J.; Chen, H.-L.; Guo, Y.-L.; Su, H.-J. Fatty Liver and Hepatic Function for Residents with Markedly High Serum PCDD/Fs Levels in Taiwan. J. Toxicol. Environ. Health A 2006, 69, 367–380. [CrossRef] [PubMed]Mocarelli, P.; Marocchi, A.; Brambilla, P.; Gerthoux, P.; Young, D.S.; Mantel, N. Clinical Laboratory Manifestations of Exposure to Dioxin in Children. A Six-Year Study of the Effects of an Environmental Disaster near Seveso, Italy. JAMA 1986, 256, 2687–2695. [CrossRef]Lin, C.-Y.; Lin, L.-Y.; Chiang, C.-K.; Wang, W.-J.; Su, Y.-N.; Hung, K.-Y.; Chen, P.-C. Investigation of the Associations between LowDose Serum Perfluorinated Chemicals and Liver Enzymes in US Adults. Am. J. Gastroenterol. 2010, 105, 1354–1363. [CrossRef] [PubMed]Gleason, J.A.; Post, G.B.; Fagliano, J.A. Associations of Perfluorinated Chemical Serum Concentrations and Biomarkers of Liver Function and Uric Acid in the US Population (NHANES), 2007-2010. Environ. Res. 2015, 136, 8–14. [CrossRef] [PubMed]Jin, R.; McConnell, R.; Catherine, C.; Xu, S.; Walker, D.I.; Stratakis, N.; Jones, D.P.; Miller, G.W.; Peng, C.; Conti, D.V.; et al. Perfluoroalkyl Substances and Severity of Nonalcoholic Fatty Liver in Children: An Untargeted Metabolomics Approach. Environ. Int. 2020, 134, 105220. [CrossRef] [PubMed]Lang, I.A.; Galloway, T.S.; Scarlett, A.; Henley, W.E.; Depledge, M.; Wallace, R.B.; Melzer, D. Association of Urinary Bisphenol A Concentration with Medical Disorders and Laboratory Abnormalities in Adults. JAMA 2008, 300, 1303–1310. [CrossRef] [PubMed]Lee, M.-R.; Park, H.; Bae, S.; Lim, Y.-H.; Kim, J.H.; Cho, S.-H.; Hong, Y.-C. Urinary Bisphenol A Concentrations Are Associated with Abnormal Liver Function in the Elderly: A Repeated Panel Study. J. Epidemiol. Community Health 2014, 68, 312–317. [CrossRef] [PubMed]Tarantino, G.; Valentino, R.; Di Somma, C.; D’Esposito, V.; Passaretti, F.; Pizza, G.; Brancato, V.; Orio, F.; Formisano, P.; Colao, A.; et al. Bisphenol A in Polycystic Ovary Syndrome and Its Association with Liver-Spleen Axis. Clin. Endocrinol. 2013, 78, 447–453. [CrossRef] [PubMed]Khalil, N.; Ebert, J.R.; Wang, L.; Belcher, S.; Lee, M.; Czerwinski, S.A.; Kannan, K. Bisphenol A and Cardiometabolic Risk Factors in Obese Children. Sci. Total Environ. 2014, 470–471, 726–732. [CrossRef] [PubMed]Medic-Stojanoska, M.; Milosevic, N.; Milanovic, M.; Stojanoski, S.; Vukovic, B.; Icin, T.; Bajkin, I.; Stepanovic, K.; Sudji, J.; Milic, N. Can Phthalates Impair Liver Function? Endocr. Abstr. 2019. [CrossRef]Miloševi´c, N.; Mili´c, N.; Živanovi´c Bosi´c, D.; Bajkin, I.; Perˇci´c, I.; Abenavoli, L.; Medi´c Stojanoska, M. Potential Influence of the Phthalates on Normal Liver Function and Cardiometabolic Risk in Males. Environ. Monit. Assess. 2017, 190, 17. [CrossRef]Motamed, N.; Sohrabi, M.; Ajdarkosh, H.; Hemmasi, G.; Maadi, M.; Sayeedian, F.S.; Pirzad, R.; Abedi, K.; Aghapour, S.; Fallahnezhad, M.; et al. Fatty Liver Index vs. Waist Circumference for Predicting Non-Alcoholic Fatty Liver Disease. World J. Gastroenterol. 2016, 22, 3023–3030. [CrossRef]Lee, S.B.; Kim, M.K.; Kang, S.; Park, K.; Kim, J.H.; Baik, S.J.; Nam, J.S.; Ahn, C.W.; Park, J.S. Triglyceride Glucose Index Is Superior to the Homeostasis Model Assessment of Insulin Resistance for Predicting Nonalcoholic Fatty Liver Disease in Korean Adults. Endocrinol. Metab. Seoul Korea 2019, 34, 179–186. [CrossRef]Xu, C.; Ma, Z.; Wang, Y.; Liu, X.; Tao, L.; Zheng, D.; Guo, X.; Yang, X. Visceral Adiposity Index as a Predictor of NAFLD: A Prospective Study with 4-Year Follow-Up. Liver Int. Off. J. Int. Assoc. Study Liver 2018, 38, 2294–2300. [CrossRef]Özcabı, B.; Demirhan, S.; Akyol, M.; Öztürkmen Akay, H.; Güven, A. Lipid Accumulation Product Is a Predictor of Nonalcoholic Fatty Liver Disease in Childhood Obesity. Korean J. Pediatr. 2019, 62, 450–455. [CrossRef]Mansour-Ghanaei, R.; Mansour-Ghanaei, F.; Naghipour, M.; Joukar, F.; Atrkar-Roushan, Z.; Tabatabaii, M.; Ghorani, N. The Role of Anthropometric Indices in the Prediction of Non-Alcoholic Fatty Liver Disease in the PERSIAN Guilan Cohort Study (PGCS). J. Med. Life 2018, 11, 194–202. [CrossRef]Hatch, E.E.; Nelson, J.W.; Qureshi, M.M.; Weinberg, J.; Moore, L.L.; Singer, M.; Webster, T.F. Association of Urinary Phthalate Metabolite Concentrations with Body Mass Index and Waist Circumference: A Cross-Sectional Study of NHANES Data, 1999–2002. Environ. Health Glob. Access Sci. Source 2008, 7, 27. [CrossRef]Stahlhut, R.W.; van Wijngaarden, E.; Dye, T.D.; Cook, S.; Swan, S.H. Concentrations of Urinary Phthalate Metabolites Are Associated with Increased Waist Circumference and Insulin Resistance in Adult U.S. Males. Environ. Health Perspect. 2007, 115, 876–882. [CrossRef]Lee, D.-H.; Lee, I.-K.; Jin, S.-H.; Steffes, M.; Jacobs, D.R. Association between Serum Concentrations of Persistent Organic Pollutants and Insulin Resistance among Nondiabetic Adults: Results from the National Health and Nutrition Examination Survey 1999–2002. Diabetes Care 2007, 30, 622–628. [CrossRef] [PubMed]La Merrill, M.A.; Johnson, C.L.; Smith, M.T.; Kandula, N.R.; Macherone, A.; Pennell, K.D.; Kanaya, A.M. Exposure to Persistent Organic Pollutants (POPs) and Their Relationship to Hepatic Fat and Insulin Insensitivity among Asian Indian Immigrants in the United States. Environ. Sci. Technol. 2019, 53, 13906–13918. [CrossRef]Geiger, S.D.; Yao, P.; Vaughn, M.G.; Qian, Z. PFAS Exposure and Overweight/Obesity among Children in a Nationally Representative Sample. Chemosphere 2021, 268, 128852. [CrossRef]. Carter, D.; Dieterich, D.T.; Chang, C. Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis in Liver Transplantation. Clin. Liver Dis. 2018, 22, 213–227. [CrossRef] [PubMed]ORIGINALPDF.pdfPDF.pdfPDFapplication/pdf1301440https://bonga.unisimon.edu.co/bitstreams/f353d629-8e7a-4221-8ab3-92bdb633fd8a/download8be15eccdb0818e35814d743e4750d9fMD51CC-LICENSElicense_rdflicense_rdfapplication/rdf+xml; charset=utf-8805https://bonga.unisimon.edu.co/bitstreams/4f1b3b77-8694-427d-9739-1789a24b531a/download4460e5956bc1d1639be9ae6146a50347MD52LICENSElicense.txtlicense.txttext/plain; charset=utf-8381https://bonga.unisimon.edu.co/bitstreams/e2846100-9e4e-4166-b6b2-a9551a5ff110/download733bec43a0bf5ade4d97db708e29b185MD53TEXTRole-of-Endocrine-Dirupting-Chemical.pdf.txtRole-of-Endocrine-Dirupting-Chemical.pdf.txtExtracted texttext/plain109511https://bonga.unisimon.edu.co/bitstreams/0d257ec2-b3ba-495b-b303-3edfbc34c9d9/downloadf745f73c03f63dd2c6e2100352bb8040MD54PDF.pdf.txtPDF.pdf.txtExtracted texttext/plain100529https://bonga.unisimon.edu.co/bitstreams/ee67a104-8056-413e-8ac8-f5b2af24db40/download6eabd5082166a1c934c6617c0e00125cMD56THUMBNAILRole-of-Endocrine-Dirupting-Chemical.pdf.jpgRole-of-Endocrine-Dirupting-Chemical.pdf.jpgGenerated Thumbnailimage/jpeg23372https://bonga.unisimon.edu.co/bitstreams/15aff6e2-822f-4647-9342-c24237c6d93e/download34c3592590f32b2068ce88aa1e914367MD55PDF.pdf.jpgPDF.pdf.jpgGenerated Thumbnailimage/jpeg5808https://bonga.unisimon.edu.co/bitstreams/344f77dc-5621-4a51-8dae-3e45aca0ce73/download75bb663256615307af65e226c4cff211MD5720.500.12442/8361oai:bonga.unisimon.edu.co:20.500.12442/83612024-08-14 21:52:32.217http://creativecommons.org/licenses/by-nc-nd/4.0/Attribution-NonCommercial-NoDerivatives 4.0 Internacionalopen.accesshttps://bonga.unisimon.edu.coRepositorio Digital Universidad Simón Bolívarrepositorio.digital@unisimon.edu.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