1
|
Coe ST, Chakraborty S, Faheem M, Kupradit K, Bhandari RK. A second hit by PFOS exposure exacerbated developmental defects in medaka embryos with a history of ancestral BPA exposure. CHEMOSPHERE 2024; 362:142796. [PMID: 38972462 PMCID: PMC11309894 DOI: 10.1016/j.chemosphere.2024.142796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/09/2024]
Abstract
Bisphenol-A (BPA), a known endocrine-disrupting chemical (EDC) in plastics and resins, has been found to induce heritable health effects in fish and mammals, affecting directly exposed individuals and indirectly their progenies in subsequent generations. It is not clearly understood if subsequent generations of the BPA-exposed ancestors have increased sensitivity to the second hit by the chemicals of emerging concern. To understand this, the present study examined the effects of developmental exposure to perfluorooctanesulfonic acid (PFOS), which has been a global contaminant recently, in embryos whose ancestors were exposed to BPA. Two lineages of medaka (Oryzias latipes) were established: 1) the BPA lineage in which the F0 generation was exposed to 10 μg/L BPA during early development and 2) the control lineage with no BPA exposure in the F0 generation. These lineages were raised up to the F4 generation without further exposure. The embryos of the F4 generation were exposed to PFOS at 0, 0.002, 0.02, 0.2, 2, and 20 mg/L concentrations. Early developmental defects resulting in mortality, delayed hatching, teratogenic phenotypes, and altered gene expression were examined in both lineages. The expression level of genes encoding DNA methyltransferases and genes responsible for oxidative stress defense were determined. Following environmentally relevant PFOS exposure, organisms with a history of BPA exposure displayed significant changes in all categories of developmental defects mentioned above, including increased expression of genes related to oxidative stress, compared to individuals without BPA exposure. The present study provides initial evidence that a history of ancestral BPA exposure can alter sensitivity to developmental disorders following the second hit by PFOS exposure. The variable of ancestral BPA exposure could be considered in mechanistic, medical, and regulatory toxicology, and can also be applied to holistic environmental equity research.
Collapse
Affiliation(s)
- Seraiah T Coe
- Department of Biology, University of North Carolina at Greensboro, NC, 27412, USA
| | - Sourav Chakraborty
- Department of Biology, University of North Carolina at Greensboro, NC, 27412, USA; Division of Biological Sciences, University of Missouri Columbia, MO, 65211, USA
| | - Mehwish Faheem
- Division of Biological Sciences, University of Missouri Columbia, MO, 65211, USA
| | - Karabuning Kupradit
- Department of Biology, University of North Carolina at Greensboro, NC, 27412, USA
| | - Ramji K Bhandari
- Division of Biological Sciences, University of Missouri Columbia, MO, 65211, USA.
| |
Collapse
|
2
|
Marin M, Annunziato KM, Tompach MC, Liang W, Zahn SM, Li S, Doherty J, Lee J, Clark JM, Park Y, Timme-Laragy AR. Maternal PFOS exposure affects offspring development in Nrf2-dependent and independent ways in zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 271:106923. [PMID: 38669778 PMCID: PMC11177596 DOI: 10.1016/j.aquatox.2024.106923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Perfluorooctanesulfonic acid (PFOS) is a ubiquitous legacy environmental contaminant detected broadly in human samples and water supplies. PFOS can cross the placenta and has been detected in cord blood and breastmilk samples, underscoring the importance of understanding the impacts of maternal PFOS exposure during early development. This study aimed to investigate the effects of a preconception exposure to PFOS on developmental endpoints in offspring, as well as examine the role of the transcription factor Nuclear factor erythroid-2-related factor (Nrf2a) in mediating these effects. This transcription factor regulates the expression of several genes that protect cells against oxidative stress including during embryonic development. Adult female zebrafish were exposed to 0.02, 0.08 or 0.14 mg/L PFOS for 1 week (duration of one cycle of oocyte maturation) and then paired with unexposed males from Nrf2a mutant or wildtype strains. Embryos were collected for two weeks or until completion of 5 breeding events. PFOS was maternally transferred to offspring independent of genotype throughout all breeding events in a dose-dependent manner, ranging from 2.77 to 23.72 ng/embryo in Nrf2a wildtype and 2.40 to 15.80 ng/embryo in Nrf2a mutants. Although embryo viability at collection was not impacted by maternal PFOS exposure, developmental effects related to nutrient uptake, growth and pancreatic β-cell morphology were observed and differed based on genotype. Triglyceride levels were increased in Nrf2a wildtype eggs from the highest PFOS group. In Nrf2a wildtype larvae there was a decrease in yolk sac uptake while in Nrf2a mutants there was an increase. Additionally, there was a significant decrease in pancreatic β-cell (islet) area in wildtype larvae from the 0.14 mg/L PFOS accompanied by an increase in the prevalence of abnormal islet morphologies compared to controls. Abnormal morphology was also observed in the 0.02 and 0.08 mg/L PFOS groups. Interestingly, in Nrf2a mutants there was a significant increase in the pancreatic β-cell area in the 0.02 and 0.08 mg/L PFOS groups and no changes in the prevalence of abnormal islet morphologies. These results suggest that the regulation of processes like nutrient consumption, growth and pancreatic β-cell development are at least partially modulated by the presence of a functional Nrf2a transcriptomic response. Overall, preconception exposure to environmental pollutants, such as PFOS, may impact the maturing oocyte and cause subtle changes that can ultimately impact offspring health and development.
Collapse
Affiliation(s)
- Marjorie Marin
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA; Biotechnology Training Program, University of Massachusetts, Amherst, MA, USA
| | - Kate M Annunziato
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Madeline C Tompach
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA; Biotechnology Training Program, University of Massachusetts, Amherst, MA, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Wenle Liang
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Sarah M Zahn
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Sida Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Jeffery Doherty
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - Jonghwa Lee
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - John M Clark
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, MA, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
3
|
Yi W, Shi J, Wang L, Wang D, Wang Y, Song J, Xin L, Jiang F. Maternal PFOS exposure in mice induces hepatic lipid accumulation and inflammation in adult female offspring: Involvement of microbiome-gut-liver axis and autophagy. JOURNAL OF HAZARDOUS MATERIALS 2024; 470:134177. [PMID: 38565010 DOI: 10.1016/j.jhazmat.2024.134177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Perfluorooctane sulfonates (PFOS) are the persistent organic pollutants. In the present study, 0, 0.3, or 3-mg/kg PFOS were administered to pregnant mice from GD 11 to GD 18. The histopathology of liver and intestine, serum and hepatic lipid levels, lipid metabolism related genes, and gut microbiota were examined in adult female offspring. The results suggested that maternal PFOS exposure increased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and induced F4/80+ macrophage infiltration in adult female offspring, in addition to the elevation of TNF-α and IL-1β mRNA levels in low-dose and high-dose groups, respectively. Furthermore, maternal exposure to PFOS increased serum triglyceride (TG) and hepatic total cholesterol (TC) levels, which was associated with the alteration of the process of fatty acid transport and β-oxidation, TG synthesis and transport, cholesterol synthesis and excretion in the liver. The AMPK/mTOR/autophagy signaling was also inhibited in the liver of adult female offspring. Moreover, changes in gut microbiota were also related to lipid metabolism, especially for the Desulfovibrio, Ligilactobacillus, Enterorhabdus, HT002 and Peptococcaceae_unclassified. Additionally, maternal exposure to PFOS decreased mRNA expressions of the tight junction protein and AB+ goblet cells in the colon, while increasing the overproduction of lipopolysaccharides (LPS) and F4/80+ macrophage infiltration. Collectively, maternal PFOS exposure induced liver lipid accumulation and inflammation, which strongly correlated with the disruption of the gut-liver axis and autophagy in adult female offspring, highlighting the persistent adverse effects in offspring exposed to PFOS.
Collapse
Affiliation(s)
- Wenjie Yi
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Junwen Shi
- Suzhou Industrial Park Center for Disease Control and Prevention, Suzhou, Jiangsu, China
| | - Liying Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Dongxuan Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Yiting Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Jingwen Song
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Lili Xin
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China.
| | - Fei Jiang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China; School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Zhang M, Chen Y, Sun S, Zhang S, Yuan L, Xu Y, Li X, Chen G, Wei X, Liu C. Ketogenic diet alleviates β-cell dedifferentiation but aggravates hepatic lipid accumulation in db/db mice. Nutrition 2024; 119:112284. [PMID: 38118383 DOI: 10.1016/j.nut.2023.112284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/08/2023] [Accepted: 10/25/2023] [Indexed: 12/22/2023]
Abstract
OBJECTIVE The aim of this study was to explore the effect of the ketogenic diet (KD) on β-cell dedifferentiation and hepatic lipid accumulation in db/db mice. METHODS After a 3-wk habituation, male db/db mice ages 8 wk were assigned into one of three groups: normal diet (ND), KD, and 75% calorie restriction (CR) group. Free access to a standard diet, a KD, and 75% of a standard diet, respectively, were given to each group. Additionally, sex-matched 8-wk-old C57BL/6 mice were used to construct a control (C) group. After a 4-wk dietary intervention, mouse body weight, fasting blood glucose (FBG), blood lipids, fasting insulin (FINS), glucose tolerance, and β-hydroxybutyric acid level were measured. The morphologies of the islet and liver were observed by hematoxylin and eosin staining. Positive expressions of β-cell-specific transcription factors in mouse islets were determined by double immunofluorescence staining. The size and number of lipid droplets in mouse liver were examined by Oil Red O staining. Real-time quantitative reverse transcription polymerase chain reaction detected relative levels of adipogenesis-associated and lipolysis-associated genes in mouse liver. Additionally, expressions of CD36 protein in the mouse liver were determined by immunohistochemical staining and Western blot. RESULTS After a 4-wk dietary intervention, FBG, FINS, and glucose area under the curve in the KD group became significantly lower than in the ND group (all P < 0.05). Regular morphology of mouse islets was observed in the KD group, with an increased number of islet cells. The KD significantly reversed the decrease in β-cell number, disarrangement of β-cells, decline of β/α-cell ratio, and downregulation of β-cell-specific transcription factors in db/db mice. Serum levels of triacylglycerol, total cholesterol, and low-density lipoprotein cholesterol were comparable between the ND and KD groups. In contrast, serum triacylglycerol levels were significantly lower in the CR group than in the ND group (P < 0.05). Vacuolar degeneration and lipid accumulation in the liver were more prominent in the KD group than in the ND and CR groups. The mRNA levels of Pparα and Acox1 in the KD group were lower than those in the ND group, although no significant differences were detected. Relative levels of Cd36 and inflammatory genes in the mouse liver were significantly higher in the KD group than in the ND group (all P < 0.05). CONCLUSION The KD significantly reduced FBG and FINS and improved glucose tolerance in db/db mice by upregulating β-cell-specific transcription factors and reversing β-cell dedifferentiation. However, the KD also induced hepatic lipid accumulation and aggravated inflammatory response in the liver of db/db mice.
Collapse
Affiliation(s)
- Mengxiao Zhang
- Department of Geriatrics, Yancheng TCM Hospital Affiliated with Nanjing University of Chinese Medicine, Yancheng TCM Hospital, Yancheng, China
| | - Yu Chen
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuoshuo Sun
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaohong Zhang
- Department of Geriatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Li Yuan
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yijiao Xu
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingjia Li
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guofang Chen
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xiao Wei
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Chao Liu
- Endocrinology Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Zhang R, Yu G, Luo T, Zeng X, Sun Y, Huang B, Liu Y, Zhang J. Transcriptomic and metabolomic profile changes in the liver of Sprague Dawley rat offspring after maternal PFOS exposure during gestation and lactation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115862. [PMID: 38157801 DOI: 10.1016/j.ecoenv.2023.115862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
Epidemiological and experimental research has indicated an association between perfluorooctane sulfonate (PFOS) exposure and liver disease. However, the potential hepatotoxic effects and mechanisms of low-level prenatal PFOS exposure in offspring remain ambiguous. The objective of this research was to examine the alterations in liver transcriptomic and metabolomic profiles in offspring rats at postnatal day (PND) 30 following gestational and lactational exposure to PFOS (from gestational day 1 to 20 and PND 1 to 21). Pregnant Sprague-Dawley rats were separated into a control group (3% starch gel solution, oral gavage) and a PFOS exposure group (0.03 mg/kg body weight per day, oral gavage). Histopathological changes in liver sections were observed by hematoxylin and eosin staining. Biochemical analysis was conducted to evaluate changes in glucose and lipid metabolism. Transcriptomic and metabolomic analyses were utilized to identify significant genes and metabolites associated with alterations of liver glucose and lipid metabolism through an integrated multi-omics analysis. No significant differences were found in the measured biochemical parameters. In total, 167 significant differentially expressed genes (DEGs) related to processes such as steroid biosynthesis, PPAR signaling pathway, and fat digestion and absorption were identified in offspring rats in the PFOS exposure group. Ninety-five altered metabolites were exhibited in the PFOS exposure group, such as heptaethylene glycol, lysoPE (0:0/18:0), lucidenic acid K, and p-Cresol sulfate. DEGs associated with steroid biosynthesis, PPAR signaling pathway, fat digestion and absorption were significantly upregulated in the PFOS exposure group (P < 0.05). The analysis of correlations indicated that there was a significant inverse correlation between all identified differential metabolites and the levels of fasting blood glucose, high-density lipoprotein, and triglycerides in the PFOS exposure group (P < 0.05). Our findings demystify that early-life PFOS exposure can lead to alterations in transcriptomic and metabolomic profiles in the offspring's liver, which provided mechanistic insights into the potential hepatotoxicity and developmental toxicity associated with environmentally relevant levels of PFOS exposure.
Collapse
Affiliation(s)
- Ruiyuan Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Guoqi Yu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Singapore
| | - Tingyu Luo
- School of Public Health, Guilin Medical University, 541001 Guilin, Guangxi, China
| | - Xiaojing Zeng
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Yan Sun
- School of Public Health, Guilin Medical University, 541001 Guilin, Guangxi, China
| | - Bo Huang
- School of Public Health, Guilin Medical University, 541001 Guilin, Guangxi, China
| | - Yongjie Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai Academy of Environment Sciences, 200233, Shanghai, China.
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
6
|
Ismail T, Lee HK, Lee H, Kim Y, Kim E, Lee JY, Kim KB, Ryu HY, Cho DH, Kwon TK, Park TJ, Kwon T, Lee HS. Early life exposure to perfluorooctanesulfonate (PFOS) impacts vital biological processes in Xenopus laevis: Integrated morphometric and transcriptomic analyses. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115820. [PMID: 38103469 DOI: 10.1016/j.ecoenv.2023.115820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Perfluorooctanesulfonate (PFOS) is a ubiquitous environmental pollutant associated with increasing health concerns and environmental hazards. Toxicological analyses of PFOS exposure are hampered by large interspecies variations and limited studies on the mechanistic details of PFOS-induced toxicity. We investigated the effects of PFOS exposure on Xenopus laevis embryos based on the reported developmental effects in zebrafish. X. laevis was selected to further our understanding of interspecies variation in response to PFOS, and we built upon previous studies by including transcriptomics and an assessment of ciliogenic effects. Midblastula-stage X. laevis embryos were exposed to PFOS using the frog embryo teratogenesis assay Xenopus (FETAX). Results showed teratogenic effects of PFOS in a time- and dose-dependent manner. The morphological abnormalities of skeleton deformities, a small head, and a miscoiled gut were associated with changes in gene expression evidenced by whole-mount in situ hybridization and transcriptomics. The transcriptomic profile of PFOS-exposed embryos indicated the perturbation in the expression of genes associated with cell death, and downregulation in adenosine triphosphate (ATP) biosynthesis. Moreover, we observed the effects of PFOS exposure on cilia development as a reduction in the number of multiciliated cells and changes in the directionality and velocity of the cilia-driven flow. Collectively, these data broaden the molecular understanding of PFOS-induced developmental effects, whereby ciliary dysfunction and disrupted ATP synthesis are implicated as the probable modes of action of embryotoxicity. Furthermore, our findings present a new challenge to understand the links between PFOS-induced developmental toxicity and vital biological processes.
Collapse
Affiliation(s)
- Tayaba Ismail
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyun-Kyung Lee
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hongchan Lee
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Youni Kim
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eunjeong Kim
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jun-Yeong Lee
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kee-Beom Kim
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hong-Yeoul Ryu
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dong-Hyung Cho
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Tae Joo Park
- Department of Biological Sciences, College of Information-Bio Convergence, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Taejoon Kwon
- Department of Biomedical Engineering, College of Information-Bio Convergence, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Hyun-Shik Lee
- KNU LAMP Research Center, KNU, Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
7
|
Ma R, Liu Q, Liu Z, Sun X, Jiang X, Hou J, Zhang Y, Wu Y, Cheng M, Dong Z. H19/Mir-130b-3p/Cyp4a14 potentiate the effect of praziquantel on liver in the treatment of Schistosoma japonicum infection. Acta Trop 2023; 247:107012. [PMID: 37659685 DOI: 10.1016/j.actatropica.2023.107012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Schistosomiasis is a prevalent infectious disease caused by the parasitic trematodes of the genus Schistosoma. Praziquantel (PZQ), a safe and affordable drug, is the recommended oral treatment for schistosomiasis. The main pathologic manifestation of schistosomiasis is liver injury. However, the role and interactions of various RNA molecules in the effect of PZQ on the liver after S. japonicum infection have not been elucidated. RESULTS In this study, C57BL/6 mice were randomly divided into the control group, infection group, and PZQ treatment group. Total RNA was extracted from the livers of the mice. High-throughput whole transcriptome sequencing was performed to detect the RNA expression profiles in the three groups. A co-expression gene-interaction network was established based on the significant differentially expressed genes in the PZQ treatment group; messenger RNA (mRNA) Cyp4a14 was identified as a critical hub gene. Furthermore, competitive endogenous RNA networks were constructed by predicting the specific binding relations between mRNA and long noncoding (lnc) RNA and between lncRNA and microRNA (miRNA) of Cyp4a14, suggesting the involvement of the H19/miR-130b-3p/Cyp4a14 regulatory axis. Dual luciferase reporter assay result proved the specific binding of miR-130b-3p with Cyp4a14 3'UTR. CONCLUSIONS Our findings indicate the involvement of the H19/miR-130b-3p/Cyp4a14 axis in the effect of PZQ on the liver after S. japonicum infection. Moreover, the expression of mRNA Cyp4a14 could be regulated by the bonding of miR-130b-3p with 3'UTR of Cyp4a14. The findings of this study could provide a novel perspective to understand the host response to PZQ against S. japonicum in the future.
Collapse
Affiliation(s)
- Rui Ma
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Qiang Liu
- Department of Anesthesia, Binzhou Medical University Hospital, Binzhou, Shandong, 256600, China
| | - Zimo Liu
- Electrocardiogram Room, Yantai Yuhuangding Hospital, Yantai, Shandong, 264000, China
| | - Xu Sun
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Xinze Jiang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Jiangshan Hou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Yumei Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Yulong Wu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| | - Mei Cheng
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| | - Zhouyan Dong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| |
Collapse
|
8
|
Carberry CK, Bangma J, Koval L, Keshava D, Hartwell HJ, Sokolsky M, Fry RC, Rager JE. Extracellular Vesicles altered by a Per- and Polyfluoroalkyl Substance Mixture: In Vitro Dose-Dependent Release, Chemical Content, and MicroRNA Signatures involved in Liver Health. Toxicol Sci 2023; 197:kfad108. [PMID: 37851381 PMCID: PMC10823775 DOI: 10.1093/toxsci/kfad108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) have emerged as high priority contaminants due to their ubiquity and pervasiveness in the environment. Numerous PFAS co-occur across sources of drinking water, including areas of North Carolina (NC) with some detected concentrations above the Environmental Protection Agency's health advisory levels. While evidence demonstrates PFAS exposure induces harmful effects in the liver, the involvement of extracellular vesicles (EVs) as potential mediators of these effects has yet to be evaluated. This study set out to evaluate the hypothesis that PFAS mixtures induce dose-dependent release of EVs from liver cells, with exposures causing differential loading of microRNAs (miRNAs) and PFAS chemical signatures. To test this hypothesis, a defined PFAS mixture was prioritized utilizing data collected by the NC PFAS Testing Network. This mixture contained three substances, PFOS, PFOA, and PFHxA, selected based upon co-occurrence patterns and the inclusion of both short-chain (PFHxA) and long-chain (PFOA and PFOS) substances. HepG2 liver cells were exposed to equimolar PFAS, and secreted EVs were isolated from conditioned media and characterized for count and molecular content. Exposures induced a dose-dependent release of EVs carrying miRNAs that were differentially loaded upon exposure. These altered miRNA signatures were predicted to target mRNA pathways involved in hepatic fibrosis and cancer. Chemical concentrations of PFOS, PFOA, and PFHxA were also detected in both parent HepG2 cells and their released EVs, specifically within a 15-fold range after normalizing for protein content. This study therefore established EVs as novel biological responders and measurable endpoints for evaluating PFAS-induced toxicity.
Collapse
Affiliation(s)
- Celeste K Carberry
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jacqueline Bangma
- Center for Environmental Measurement and Modeling, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Lauren Koval
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Deepak Keshava
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hadley J Hartwell
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marina Sokolsky
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Rebecca C Fry
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- School of Medicine, Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Julia E Rager
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- School of Medicine, Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
9
|
Lykkebo CA, Mortensen MS, Davidsen N, Bahl MI, Ramhøj L, Granby K, Svingen T, Licht TR. Antibiotic induced restructuring of the gut microbiota does not affect oral uptake and accumulation of perfluorooctane sulfonic acid (PFOS) in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 334:122179. [PMID: 37454717 DOI: 10.1016/j.envpol.2023.122179] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Perfluorooctane sulfonic acid (PFOS) is a manmade legacy compound belonging to the group of persistent per- and polyfluorinated substances (PFAS). While many adverse health effects of PFOS have been identified, knowledge about its effect on the intestinal microbiota is scarce. The microbial community inhabiting the gut of mammals plays an important role in health, for instance by affecting the uptake, excretion, and bioavailability of some xenobiotic toxicants. Here, we investigated (i) the effect of vancomycin-mediated microbiota modulation on the uptake of PFOS in adult Sprague-Dawley rats, and (ii) the effects of PFOS exposure on the rat microbiota composition. Four groups of twelve rats were exposed daily for 7 days with either 3 mg/kg PFOS plus 8 mg/kg vancomycin, only PFOS, only vancomycin, or a corn oil control. Vancomycin-induced modulation of the gut microbiota composition did not affect uptake of branched and linear PFOS over a period of 7 days, measured in serum samples. 16S rRNA amplicon sequencing of faecal and intestinal samples revealed that vancomycin treatment lowered microbial alpha-diversity, while PFOS increased the microbial diversity in vancomycin-treated as well as in non-antibiotic treated animals, possibly because an observed decrease in the Enterobacteriaceae abundance allows other microbial species to propagate. Colonic short-chain fatty acids were significantly lower in vancomycin-treated animals but remained unaffected by PFOS. Our results suggest that PFOS exposure may disturb the intestinal microbiota, but that antibiotic-induced modulation of the intestinal ecosystem does not affect systemic uptake of PFOS in rats.
Collapse
Affiliation(s)
- Claus Asger Lykkebo
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark.
| | | | - Nichlas Davidsen
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark
| | - Louise Ramhøj
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark
| | - Kit Granby
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kgs, Lyngby, DK-2800, Denmark.
| |
Collapse
|
10
|
Bernal K, Touma C, Erradhouani C, Boronat-Belda T, Gaillard L, Al Kassir S, Le Mentec H, Martin-Chouly C, Podechard N, Lagadic-Gossmann D, Langouet S, Brion F, Knoll-Gellida A, Babin PJ, Sovadinova I, Babica P, Andreau K, Barouki R, Vondracek J, Alonso-Magdalena P, Blanc E, Kim MJ, Coumoul X. Combinatorial pathway disruption is a powerful approach to delineate metabolic impacts of endocrine disruptors. FEBS Lett 2022; 596:3107-3123. [PMID: 35957500 DOI: 10.1002/1873-3468.14465] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023]
Abstract
The prevalence of metabolic diseases, such as obesity, diabetes, metabolic syndrome and chronic liver diseases among others, has been rising for several years. Epidemiology and mechanistic (in vivo, in vitro and in silico) toxicology have recently provided compelling evidence implicating the chemical environment in the pathogenesis of these diseases. In this review, we will describe the biological processes that contribute to the development of metabolic diseases targeted by metabolic disruptors, and will propose an integrated pathophysiological vision of their effects on several organs. With regard to these pathomechanisms, we will discuss the needs, and the stakes of evolving the testing and assessment of endocrine disruptors to improve the prevention and management of metabolic diseases that have become a global epidemic since the end of last century.
Collapse
Affiliation(s)
- Kévin Bernal
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Charbel Touma
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Chedi Erradhouani
- Université Paris Cité, France.,Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Talía Boronat-Belda
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Lucas Gaillard
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Sara Al Kassir
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Hélène Le Mentec
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Corinne Martin-Chouly
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Normand Podechard
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Dominique Lagadic-Gossmann
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Sophie Langouet
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - François Brion
- Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Anja Knoll-Gellida
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Patrick J Babin
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Iva Sovadinova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karine Andreau
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Robert Barouki
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Jan Vondracek
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Etienne Blanc
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Min Ji Kim
- INSERM UMR-S 1124, Paris, France.,Université Sorbonne Paris Nord, Bobigny, France
| | - Xavier Coumoul
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| |
Collapse
|
11
|
Wang P, Liu D, Yan S, Cui J, Liang Y, Ren S. Adverse Effects of Perfluorooctane Sulfonate on the Liver and Relevant Mechanisms. TOXICS 2022; 10:toxics10050265. [PMID: 35622678 PMCID: PMC9144769 DOI: 10.3390/toxics10050265] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent, widely present organic pollutant. PFOS can enter the human body through drinking water, ingestion of food, contact with utensils containing PFOS, and occupational exposure to PFOS, and can have adverse effects on human health. Increasing research shows that the liver is the major target of PFOS, and that PFOS can damage liver tissue and disrupt its function; however, the exact mechanisms remain unclear. In this study, we reviewed the adverse effects of PFOS on liver tissue and cells, as well as on liver function, to provide a reference for subsequent studies related to the toxicity of PFOS and liver injury caused by PFOS.
Collapse
|
12
|
Wan C, Gu T, Ling J, Qin Y, Luo J, Sun L, Hua L, Zhao J, Jiang S. Perfluorooctane sulfonate aggravates CCl4-induced hepatic fibrosis via HMGB1/TLR4/Smad signaling. ENVIRONMENTAL TOXICOLOGY 2022; 37:983-994. [PMID: 34990082 DOI: 10.1002/tox.23458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a widespread environmental pollutant and may cause a variety of adverse health effects. The hepatotoxicity of PFOS has attracted particular attention, given the fact that the liver has one of the highest PFOS accumulations among human tissues. In this study, we revealed that subchronic PFOS exposure may exacerbate carbon tetrachloride (CCl4 )-induced liver fibrosis in animal models. Administration with 1 mg/kg PFOS every other day for 56 days dramatically enhanced CCl4 -mediated liver injury and hepatic stellate cell (HSC) activation. Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. PFOS exposure induced the translocation of HMGB1 from the nucleus into the cytoplasm of hepatocytes and cultured BRL-3A cells at a starting concentration of 50 μM. This process is accompanied with concurrent flux of calcium, suggesting a link between calcium signaling and HMGB1 release following PFOS exposure. Finally, we showed that PFOS-exposed conditional medium (PFOS-CM) of hepatocytes may induce the translocation of Smad2/3 in HSCs in a TLR4-dependent manner. Taken together, subchronic PFOS exposure might play a pro-fibrotic role via a HMGB1/TLR4-dependent Smad signaling in HSCs. Our findings for the first time uncovered an involvement of PFOS exposure in liver fibrosis via HMGB1/TLR4/Smad signaling.
Collapse
Affiliation(s)
- Chunhua Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Tianye Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Junyi Ling
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Yi Qin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- Haimen District Center for Disease Control and Prevention, Haimen, Nantong, People's Republic of China
| | - Jiashan Luo
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Lingli Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Lu Hua
- Department of Oncology, Taizhou People's Hospital, Taizhou, China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
13
|
Costello E, Rock S, Stratakis N, Eckel SP, Walker DI, Valvi D, Cserbik D, Jenkins T, Xanthakos SA, Kohli R, Sisley S, Vasiliou V, La Merrill MA, Rosen H, Conti DV, McConnell R, Chatzi L. Exposure to per- and Polyfluoroalkyl Substances and Markers of Liver Injury: A Systematic Review and Meta-Analysis. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:46001. [PMID: 35475652 PMCID: PMC9044977 DOI: 10.1289/ehp10092] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Experimental evidence indicates that exposure to certain pollutants is associated with liver damage. Per- and polyfluoroalkyl substances (PFAS) are persistent synthetic chemicals widely used in industry and consumer products and bioaccumulate in food webs and human tissues, such as the liver. OBJECTIVE The objective of this study was to conduct a systematic review of the literature and meta-analysis evaluating PFAS exposure and evidence of liver injury from rodent and epidemiological studies. METHODS PubMed and Embase were searched for all studies from earliest available indexing year through 1 December 2021 using keywords corresponding to PFAS exposure and liver injury. For data synthesis, results were limited to studies in humans and rodents assessing the following indicators of liver injury: serum alanine aminotransferase (ALT), nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, or steatosis. For human studies, at least three observational studies per PFAS were used to conduct a weighted z-score meta-analysis to determine the direction and significance of associations. For rodent studies, data were synthesized to qualitatively summarize the direction and significance of effect. RESULTS Our search yielded 85 rodent studies and 24 epidemiological studies, primarily of people from the United States. Studies focused primarily on legacy PFAS: perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), perfluorononanoic acid (PFNA), and perfluorohexanesulfonic acid. Meta-analyses of human studies revealed that higher ALT levels were associated with exposure to PFOA (z-score= 6.20, p<0.001), PFOS (z-score= 3.55, p<0.001), and PFNA (z-score= 2.27, p=0.023). PFOA exposure was also associated with higher aspartate aminotransferase and gamma-glutamyl transferase levels in humans. In rodents, PFAS exposures consistently resulted in higher ALT levels and steatosis. CONCLUSION There is consistent evidence for PFAS hepatotoxicity from rodent studies, supported by associations of PFAS and markers of liver function in observational human studies. This review identifies a need for additional research evaluating next-generation PFAS, mixtures, and early life exposures. https://doi.org/10.1289/EHP10092.
Collapse
Affiliation(s)
- Elizabeth Costello
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sarah Rock
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nikos Stratakis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sandrah P. Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Douglas I. Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dora Cserbik
- Barcelona Institute for Global Health, Barcelona, Spain
| | - Todd Jenkins
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stavra A. Xanthakos
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, California, USA
| | - Hugo Rosen
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Leda Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
14
|
Aghaei Z, Steeves KL, Jobst KJ, Cahill LS. The impact of perfluoroalkyl substances on pregnancy, birth outcomes and offspring development: A review of data from mouse models1. Biol Reprod 2021; 106:397-407. [PMID: 34875017 DOI: 10.1093/biolre/ioab223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Per- and polyfluoroalkyl substances (PFASs) such as perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are persistent in the environment and bioaccumulate in wildlife and humans, potentially causing adverse health effects at all stages of life. Studies from human pregnancy have shown that exposure to these contaminants are associated with placental dysfunction and fetal growth restriction; however, studies in humans are confounded by genetic and environmental factors. Here, we synthesize the available results from mouse models of pregnancy to show the causal effects of prenatal exposure to PFOA and PFOS on placental and fetal development and on neurocognitive function and metabolic disorders in offspring. We also propose gaps in the present knowledge and provide suggestions for future research studies.
Collapse
Affiliation(s)
- Zahra Aghaei
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Katherine L Steeves
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Karl J Jobst
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Lindsay S Cahill
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
15
|
Attenuation of Perfluorooctane Sulfonate-Induced Steatohepatitis by Grape Seed Proanthocyanidin Extract in Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2020:8818160. [PMID: 33457418 PMCID: PMC7787751 DOI: 10.1155/2020/8818160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 11/30/2020] [Indexed: 01/09/2023]
Abstract
Perfluorooctane sulfonate (PFOS), an environmentally persistent pollutant, has been revealed to elicit hepatic toxicity. In the current study, we investigated the protective role of grape seed proanthocyanidin extract (GSPE) against PFOS-caused steatohepatitis in mice. Animals were exposed intragastrically to PFOS (10 mg/kg/day), GSPE (150 mg/kg/day), or their combination. After 21 days of treatment, mice exposed to PFOS exhibited steatosis, oxidative stress, and inflammation in the liver. Nevertheless, simultaneous administration of GSPE resumed the declined serum hepatic enzyme activities and histological abnormalities in PFOS-exposed mice. Furthermore, GSPE supplementation reduced the contents of triglyceride (TG) and total cholesterol (TC) and expression of lipid metabolism-associated genes CD36 and fatty acid-binding protein 4 (FABP4) in the liver of mice treated with PFOS. Moreover, GSPE suppressed the generation of lipid peroxidative product malondialdehyde and restored the activity of superoxide dismutase in the liver of PFOS-exposed mice. In addition, GSPE repressed the PFOS-induced hepatic overproduction of proinflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Our results demonstrate that GSPE attenuates PFOS-caused steatohepatitis in mice by regulating lipid metabolism, oxidative stress, and inflammatory response.
Collapse
|
16
|
Liang H, Yang M, Zeng C, Wu W, Zhao L, Wang Y. Perfluorooctane sulfonate exerts inflammatory bowel disease-like intestinal injury in rats. PeerJ 2021; 9:e10644. [PMID: 33510972 PMCID: PMC7798615 DOI: 10.7717/peerj.10644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/03/2020] [Indexed: 12/27/2022] Open
Abstract
Background Perfluorooctane sulfonate (PFOS), a type of perfluorinated compounds (PFCs), can induce various organ toxicity, including hepatomegaly, immunotoxicity, and gut microbiota disorder. PFCs have been associated with inflammatory bowel disease (IBD). Yet, whether PFOS exposure causes IBD-like disorder and the underlying mechanism remains undefined. Here, we investigated the influence of PFOS exposure on the development of IBD-like disorder in rats. Methods Sprague-Dawley rats were intraperitoneally injected with PFOS (1 or 10 mg/kg) or normal saline (NS) every other day for 15 days. Body weight, serum concentrations of serum amyloid A (SAA) and high sensitivity C reactive protein (hsCRP) were measured. Pathological assessments of villi height and crypt depth in the proximal duodenum and jejunum were performed using H&E staining. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to assay cell apoptosis in the jejunum. The infiltration of inflammatory cells and cytokines in the jejunum were detected by immunohistochemistry analysis. Results PFOS (10 mg/kg) significantly increased the body weight, SAA and hsCRP, whereas no significant differences were observed in PFOS 1 mg/kg group of rats. The villi height and crypt depth in the proximal duodenum and jejunum were significantly reduced upon PFOS exposure. PFOS induced higher histopathological score in intestinal tissues compared to NS. Notably, TUNEL-positive cells were significantly higher in the jejunum upon PFOS exposure. Further, neutrophil and macrophage accumulated, and inflammatory cytokines infiltration were also remarkably increased in rats exposed to PFOS. Conclusion PFOS induces IBD-like phenotypes in rats, with associated inflammatory infiltration to intestinal.
Collapse
Affiliation(s)
- Hai Liang
- Department of Pharmacy, The People's Hospital of Bozhou, Bozhou, Anhui Province, China
| | - Miao Yang
- Department of Neurology, The People's Hospital of Bozhou, Bozhou, Anhui Province, China
| | - Cheng Zeng
- Department of Pharmacy, The People's Hospital of Bozhou, Bozhou, Anhui Province, China
| | - Wei Wu
- Department of Pharmacy, The People's Hospital of Bozhou, Bozhou, Anhui Province, China
| | - Liying Zhao
- Department of Pharmacy, Deqing People's Hospital, Huzhou, Zhejiang Province, China
| | - Yu Wang
- Department of Pharmacy, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
17
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Ceccatelli S, Cravedi J, Halldorsson TI, Haug LS, Johansson N, Knutsen HK, Rose M, Roudot A, Van Loveren H, Vollmer G, Mackay K, Riolo F, Schwerdtle T. Risk to human health related to the presence of perfluoroalkyl substances in food. EFSA J 2020; 18:e06223. [PMID: 32994824 PMCID: PMC7507523 DOI: 10.2903/j.efsa.2020.6223] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluoroalkyl substances (PFASs) in food. Based on several similar effects in animals, toxicokinetics and observed concentrations in human blood, the CONTAM Panel decided to perform the assessment for the sum of four PFASs: PFOA, PFNA, PFHxS and PFOS. These made up half of the lower bound (LB) exposure to those PFASs with available occurrence data, the remaining contribution being primarily from PFASs with short half-lives. Equal potencies were assumed for the four PFASs included in the assessment. The mean LB exposure in adolescents and adult age groups ranged from 3 to 22, the 95th percentile from 9 to 70 ng/kg body weight (bw) per week. Toddlers and 'other children' showed a twofold higher exposure. Upper bound exposure was 4- to 49-fold higher than LB levels, but the latter were considered more reliable. 'Fish meat', 'Fruit and fruit products' and 'Eggs and egg products' contributed most to the exposure. Based on available studies in animals and humans, effects on the immune system were considered the most critical for the risk assessment. From a human study, a lowest BMDL 10 of 17.5 ng/mL for the sum of the four PFASs in serum was identified for 1-year-old children. Using PBPK modelling, this serum level of 17.5 ng/mL in children was estimated to correspond to long-term maternal exposure of 0.63 ng/kg bw per day. Since accumulation over time is important, a tolerable weekly intake (TWI) of 4.4 ng/kg bw per week was established. This TWI also protects against other potential adverse effects observed in humans. Based on the estimated LB exposure, but also reported serum levels, the CONTAM Panel concluded that parts of the European population exceed this TWI, which is of concern.
Collapse
|
18
|
Lind PM, Lind L. Are Persistent Organic Pollutants Linked to Lipid Abnormalities, Atherosclerosis and Cardiovascular Disease? A Review. J Lipid Atheroscler 2020; 9:334-348. [PMID: 33024729 PMCID: PMC7521972 DOI: 10.12997/jla.2020.9.3.334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 01/06/2023] Open
Abstract
The term persistent organic pollutants (POPs) denotes chemicals with known or suspected adverse health effects in animals or humans and with chemical properties that make them accumulate in the environment, including animals or humans. Lipid-soluble POPs, like dioxins, polychlorinated biphenyls (PCBs) and organochlorine pesticides are transported by lipoproteins and accumulate in adipose tissue. High levels of these compounds in the circulation have been associated with elevated cholesterol and triglycerides in cross-sectional studies and with an increase in mainly low-density lipoprotein cholesterol in a longitudinal study. Also, non-lipid-soluble POPs, such as perfluoroalkyl substances (PFASs) compounds have been associated with increased total cholesterol levels. Carotid artery atherosclerosis has been related to elevated levels of mainly highly chlorinated PCBs and to highly fluorinated PFASs, but in this case only in women. Both cross-sectional and prospective studies have shown dioxins, PCBs, as well as PFASs to be linked to cardiovascular disease (CVD) and mortality. In conclusion, as highlighted in this review, several lines of evidence support the view that POPs of different chemical classes could be linked to lipid abnormalities, carotid atherosclerosis and overt CVD like myocardial infarction and stroke.
Collapse
Affiliation(s)
- P Monica Lind
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
Endocrine Disruptors in Water and Their Effects on the Reproductive System. Int J Mol Sci 2020; 21:ijms21061929. [PMID: 32178293 PMCID: PMC7139484 DOI: 10.3390/ijms21061929] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Anthropogenic contaminants in water can impose risks to reproductive health. Most of these compounds are known to be endocrine disrupting chemicals (EDCs). EDCs can impact the endocrine system and subsequently impair the development and fertility of non-human animals and humans. The source of chemical contamination in water is diverse, originating from byproducts formed during water disinfection processes, release from industry and livestock activity, or therapeutic drugs released into sewage. This review discusses the occurrence of EDCs in water such as disinfection byproducts, fluorinated compounds, bisphenol A, phthalates, pesticides, and estrogens, and it outlines their adverse reproductive effects in non-human animals and humans.
Collapse
|
20
|
Clinical characteristics of colorectal cancer patients and anti-neoplasm activity of genistein. Biomed Pharmacother 2020; 124:109835. [PMID: 31958764 DOI: 10.1016/j.biopha.2020.109835] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/29/2019] [Accepted: 01/06/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Epidemiologically, the disease incidence of colorectal cancer (CRC) ranks the third among all malignant tumors, and its mortality is the second following lung cancer. If unmanaged, CRC will develop fatal invasiveness and metastasis. However, existing chemotherapy is limitedly effective to treat metastatic CRC. Genistein, a functional phytoestrogen, is found with potent pharmacological activity against cancer cells. Therefore, this study was designed to characterize the clinical signatures of human CRC and to conduct anti-CRC experiments using genistein. METHODS Briefly, the plasma, tumor, non-tumor samples of CRC patients were harvested for biological experiments, followed by analysis of clinical data. A pharmacological study in vitro of genistein for treating CRC cells was conducted accordingly. RESULTS In diagnostic data, molecular tumor biomarkers in CRC patients were detected in plasma samples, consistent with pathological and imaging diagnoses of CRC. Notably, carcinomatous expressions of miR-95, serum glucocorticoid kinase 1 (SGK1), B-cell lymphoma-2 (Bcl-2), extracellular regulated protein kinase 1 (Erk1) in human CRC were notably elevated when compared to those in non-tumor controls. In pharmacological experiments using cell culture model, genistein-treated CRC cells resulted in reduced cellular viability, elevated lactate dehydrogenase (LDH) content, increased apoptotic cells and TdT mediated dUTP nick end labeling (TUNEL)-positive cells following a dose-dependent manner. Interestingly, down-regulated expressions of endogenous miR-95, SGK1, Bcl-2, Erk1 were observed after genistein treatments in a dose-dependent way. CONCLUSIONS Collectively, the current clinical data indicate pathological markers of miR-95, SGK1, Erk1 in human CRC cases, and further experimental findings reveal that anti-CRC pharmacological mechanism using genistein was implicated in suppression of cellular miR-95, SGK1, Erk1 expressions. Together, genistein may be a promising bioactive compound for treating CRC.
Collapse
|