Previous Article in Journal
Chromium Selectively Accumulates in the Rat Hippocampus after 90 Days of Exposure to Cr(VI) in Drinking Water and Induces Age- and Sex-Dependent Metal Dyshomeostasis
Previous Article in Special Issue
Segmentation of Renal Thyroid Follicle Colloid in Common Carp: Insights into Perfluorooctanoic Acid-Induced Morphometric Alterations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Adverse Outcomes Following Exposure to Perfluorooctanesulfonamide (PFOSA) in Larval Zebrafish (Danio rerio): A Neurotoxic and Behavioral Perspective

1
Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
2
Department of Chemistry, Federal University of Lavras (UFLA), Minas Gerais, Lavras 37203-202, Brazil
3
Instituto de Ciencias Ambientais, Quimicas e Farmaceuticas, Universidade Federal de São Paulo, Campus Diadema, Diadema 09972-270, Brazil
4
Animal Health Research Institute, Agriculture Research Centre, Giza 3751254, Egypt
5
UF Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL 32611, USA
*
Author to whom correspondence should be addressed.
Toxics 2024, 12(10), 723; https://doi.org/10.3390/toxics12100723
Submission received: 17 August 2024 / Revised: 23 September 2024 / Accepted: 26 September 2024 / Published: 4 October 2024

Abstract

:
Toxicity mechanisms of per- and polyfluoroalkyl substances (PFASs), a chemical class present in diverse ecosystems, as well as many of their precursors, have been increasingly characterized in aquatic species. Perfluorooctanesulfonamide (PFOSA, C8H2F17NO2S) is a common precursor of perfluorooctane sulfonic acid (PFOS), a long-chain PFAS. Here, we assessed sub-lethal endpoints related to development, oxidative stress, transcript levels, and distance moved in zebrafish embryos and larvae following continuous exposure to PFOSA beginning at 6 h post-fertilization (hpf). PFOSA decreased survival in fish treated with 1 µg/L PFOSA; however, the effect was modest relative to the controls (difference of 10%). Exposure up to 10 µg/L PFOSA did not affect hatch rate, nor did it induce ROS in 7-day-old larvae fish. The activity of larval fish treated with 100 µg/L PFOSA was reduced relative to the solvent control. Transcripts related to oxidative stress response and apoptosis were measured and BCL2-associated X, apoptosis regulator (bax), cytochrome c, somatic (cycs), catalase (cat), superoxide dismutase 2 (sod2) were induced with high concentrations of PFOSA. Genes related to neurotoxicity were also measured and transcript levels of acetylcholinesterase (ache), elav-like RNA binding protein 3 (elavl3), growth-associated protein 43 (gap43), synapsin II (syn2a), and tubulin 3 (tubb3) were all increased in larval fish with higher PFOSA exposure. These data improve our understanding of the potential sub-lethal toxicity of PFOSA in fish species.

1. Introduction

Since the 1930s, per- and polyfluoroalkyl substances (PFASs) have been used in a variety of consumer and commercial goods, including textiles, surfactants, and food packaging materials. These compounds comprise short or long carbon chains, where short-chain PFAS are considered less harmful than long-chain PFAS [1]. According to the Danish Environmental Protection Agency, because long-chain PFAS persist in the environment, their production usage has been gradually phased out since the early 2000s [2]. Perfluorooctanesulfonamide (PFOSA, C8H2F17NO2S) is a common precursor of perfluorooctane sulfonic acid (PFOS), a long-chain PFAS [3], and it is a synthetic compound used to produce non-stick, waterproof, and stain-repellent coatings [4]. Most studies to date focus on the presence and toxicity of other commonly used PFAS, such as PFOS or perfluorooctanoic acid (PFOA) [5,6,7]. Currently, toxicity data regarding the precursor PFOSA are lacking.
There are a few studies that report on the environmental presence of PFOSA. Konwick et al. (2008) found that PFOSA ranged from 162 to 283 ng/L in the Conasauga River, Georgia, United States [8]. Additionally, other studies reported the following ranges for PFOSA: 0.09–20,000 µg/kg in surface soil, 0.07–2500 µg/kg in subsurface soil, 15 µg/L in surface water, and 12 µg/L in groundwater across various testing sites worldwide [9,10]. In regards to PFOSA within fish tissues, Fair et al. (2019) measured different PFAS within edible fish species from South Carolina, United States [11]. In whole fish, the average relative percent of PFOSA in mullet, spot, croaker, red drum, and seatrout was 2.04, 3.81, 3.64, 3.12, and 4.66%, respectively, whereas, in fillets, the same species had an average relative percent of 1.44, 2.03, 0.70, 2.18, 4.65, and 1.79%, respectively. PFOSA was also found to range between 0.105 and 16.4 ng/mL in serum across various fish species, including crucian carp (Carassius auratus), tilapia (Oreochromis niloticus), common carp (Cyprinus carpio), and leather catfish (Clarias lazera) [12]. However, the mechanisms of uptake, metabolism, and toxicity of PFOSA are relatively unknown for aquatic species. One study reports that the half-life of PFOSA in rainbow trout (Oncorhynchus mykiss) is 6.0 ± 0.4 days following a 30-day dietary exposure to 10 µg/g wet weight PFOSA and a 30-day depurination period [13]. Thus, PFOSA is measurable in fish tissues and may pose a health risk to both aquatic/terrestrial animals and humans.
According to studies, adverse morphological and physiological effects in aquatic organisms are potential consequences associated with the environmental presence of long-chain PFAS [14,15]. For example, studies show that PFOSA exerts cardiotoxicity in zebrafish (Danio rerio). Exposure to 0.1–100 µg/L PFOSA has been reported to reduce cardiac output, heart rate, stroke volume and reduce cardiac vasoconstriction-related genes [16]. PFOSA has also been reported to significantly increase sinus venosus and bulbus arteriosus distances at 10 and 100 µg/L [16,17]. Other studies report that exposure to PFOSA can induce hepatic and renal toxicity in zebrafish [18,19]; however, limited studies investigate the neurotoxic potential of PFOSA in developing fish. Consequently, the objectives of this study were to evaluate the neurotoxicity potential of PFOSA. To achieve this, we measured developmental endpoints, reactive oxygen species (ROS), locomotor behavior, and genes related to oxidative damage response, apoptosis, and neurotoxicity as indicators of central nervous system damage. We hypothesized that neurotoxicity endpoints would reflect dose response increased in PFOSA exposure, suggesting adverse effects on the nervous system.

2. Materials and Methods

2.1. Chemical Preparation

Perfluorooctanesulfonamide (PFOSA, (1,1,2,2,3,3,4,4,5,5,6,6,7,7,8,8,8-heptadecafluorooctane-1-sulfonamide Perfluorooctane sulfonamide) (CAS Number: 754-91-6, purity > 95%) was purchased from Fisher Scientific (Thermo Scientific Chemicals, Waltham, MA, USA, Cat# AC459640010). PFOSA stock solutions were prepared in dimethyl sulfoxide (DMSO, dimethyl sulfoxide, CAS 67-68-5, Sigma-Aldrich, Inc., St. Louis, MO, USA) and added to embryo rearing media (ERM) containing the zebrafish embryos. The ERM recipe can be located in Westerfield [20]. Exposure solutions were prepared daily to yield final nominal environmentally relevant concentrations of 0.1, 1, 10, and 100 µg/L PFOSA with a final concentration <0.1% v/v DMSO in experimental treatments.

2.2. Maintenance and Egg Production of Zebrafish

Zebrafish (AB x Tübingen, Danio rerio) husbandry has been described previously [21,22] and rearing and staging of zebrafish embryos followed that described previously [23]. The Supplemental Methods section contain full details on zebrafish husbandry and water quality. The University of Florida Institutional Animal Care and Use Committee approved all experiments (UF_IACUC202300000140).

2.3. PFOSA Exposure Regime

Fertilized and normally developing eggs were selected at ∼6 h post-fertilization (hpf) and were assigned into experimental groups in random fashion [ERM, 0.1% DMSO, or one dose of 0.1, 1, 10 or 100 µg/L PFOSA]. Four independent experiments were conducted. There were 5 to 6 replicate glass beakers for each treatment group containing between 20 and 30 embryos and 10 mL of ERM per beaker. Once chemicals were added, the beakers were placed inside an incubator at 27 ± 1 °C. Mortality, deformities, hatch times and images using an EVOS™ FL Auto Imaging System (ThermoFisher Scientific, USA) were collected daily. Deformity assessments included the presence of spinal lordosis and edema (yolk sack/pericardial) was noted each day. Water was renewed each day with a 90% water change using freshly made stocks of PFOSA.

2.4. Reactive Oxygen Species

Following fertilization in the morning, embryos at 6 h post fertilization were collected and treated as per above for 7 days in the ERM with designated concentrations of PFOSA. Fertilized embryos were assigned into sterile 25 mL glass beakers containing the designated concentration of ERM, 0.1% DMSO and 0.1, 1 and 10 µg/L PFOSA (n = 5 beakers of 10 fish each/treatment) in a 10 mL volume. The Supplemental Methods section contains full details on the analysis of ROS levels in zebrafish larvae.

2.5. Visual Motor Response Test

The visual motor response test was conducted following our published methods [24]. Fish were exposed continuously for 7 days with PFOSA as described above and assessed for locomotor activity behavior at a temperature of 27 ± 1 °C and a photoperiod of 14:10 h using the DanioVision instrument (Noldus, Wageningen, The Netherlands). The Supplemental Methods section contains full details on visual motor response tests.

2.6. Real-Time PCR

The Supplemental Methods section contains full details on real-time PCR assays. Briefly, zebrafish larvae at 6 hpf were exposed to either ERM, 0.1% DMSO or 0.1, 1, or 10 µg/L PFOSA. Each beaker contained 10–15 embryos and exposure conditions were maintained as mentioned above. Following the 7-day exposure period, larvae were pooled within a beaker, subjected to liquid nitrogen, and placed at −80 °C for RNA extraction. Sample sizes ranged from 4 to 6 beakers per group for gene expression analysis. The primers used in this study were obtained from published literature [25,26,27,28,29,30,31,32,33,34] and all targets measured in this study are listed in Supplemental Table S1. Two housekeeping genes (ribosomal subunit 18, rps18, and beta actin, b-actin) were used to normalize expression levels of all target genes. Normalized expression was calculated using CFX Manager™ software (v3.1) (baseline subtracted) and the Cq method.

2.7. Statistical Analysis

All data were compared to the solvent control (DMSO group). A log-rank test (Mantel–Cox) was employed to evaluate survival data. Data for hatch times were evaluated using a One-Way ANOVA at each time point. Levels of ROS and relative mRNA levels were first log(10) transformed following a Shapiro–Wilk test for normality. Group mean differences were then tested using a One-Way ANOVA (Dunnett’s multiple comparisons test). A simple linear regression was also conducted on the gene expression data to determine whether expression varied with concentration. Because there was no difference in expression between the ERM and DMSO group, these two experimental groups were combined for the regression as a “control” or baseline group. For the VMR, distance moved for larval fish in each treatment across the three independent experiments were binned into a single graph, but each individual run is shown in Supplemental Figure. The distance moved for fish in the DMSO group was normalized to a value of 1, and all treatments were compared relative to this group. A Kruskal–Wallis test followed by Dunn’s multiple comparisons test was employed to analyze discrete temporal units (light and dark periods) as the data were not normally distributed. All data are presented as mean ± S.D. Significance of difference was determined using a threshold of p < 0.05. Statistics and graphing were performed using GraphPad V9.3 (La Jolla, CA, USA).

3. Results

3.1. Survival, Hatch Rate, and Deformity

After zebrafish were exposed to several concentrations of PFOSA for a period of 7 days, intriguingly only fish treated with 1 µg/L PFOSA exhibited significantly decreased survival [chi square = 88.68, df = 5, p value < 0.0001] (Figure 1) relative to the DMSO control group. Survival was approximately 10% lower for some eggs treated with PFOSA and these responses were most notable in the first 48 h of exposure. The hatch rate increased in fish treated with 0.1 and 10 µg/L PFOSA as all embryos were hatched at 2 dpf compared to the other treatment groups (p < 0.05) (Figure 2). There were no significant deformities observed; however, the few zebrafish that did have deformities (less than 3%) had caudal tail malformations.

3.2. Reactive Oxygen Species

We assessed the effects of PFOSA on ROS induction in larval zebrafish at 7 dpf. PFOSA exposure did not significantly affect ROS levels (F (4, 19) = 0.14, p = 0.96) (Figure 3).

3.3. Visual Motor Response Test

Three independent trials were conducted, and data for the relative distance travelled (log transformed and relative to the DMSO control standardized to a value of 1) were combined for each experiment. Zebrafish larvae exposed to 100 µg/L exhibited reduced activity in Dark Period 2 only (number of groups = 6, Kruskal–Wallis statistic = 13.60; p value = 0.018) (Figure 4, panel C). Individual VMR trials can be found in Supplementary Figure, Supplemental Figure S1.

3.4. Expression Analysis of Transcripts

The effects of PFOSA on mRNA steady state levels were measured in larval fish. Regression analysis revealed that the apoptosis-related transcript (bax) on oxidative stress-related transcripts (cat, sod2) increased with increasing concentrations of PFOSA and the statistical information and R2 of the linear regressions are shown in Figure 5. Bax mRNA levels were elevated in zebrafish from the 1 µg/L PFOSA treatment compared to the DMSO control group (F (4, 21) = 4.160, p = 0.0123) (Figure 6A).
Regarding neurotoxicity-related genes, transcript levels of elavl3 were significantly elevated in zebrafish treated with 1 and 10 µg/L PFOSA (F (4, 23) = 4.802, p = 0.0058) (Figure 6B). No other transcript was significantly different when comparing group means to the mean of the DMSO control (p > 0.05); however, regression analysis revealed that many neurotoxicity transcripts showed a concentration independent response with PFOSA, and several transcripts were increased with higher exposure concentrations (Figure 7). The statistical information and R2 of the simple linear regressions are provided in the figures.

4. Discussion

We observed that PFOSA affected the survival of zebrafish treated with some concentrations of PFOSA 1 µg/L (10% decline at this concentration relative to the DMSO control). Dasgupta et al. (2020) exposed zebrafish embryos to 0.78–50 µM (389–24,900 µg/L) PFOSA for up to 72 hpf and 100% mortality was observed in all treated embryos by 72 hpf [18]. Regarding abnormalities, we observed only a few deformities (less than 2–3 percent) across all treatment groups; several studies examining toxicity effects of PFOSA report significant deformities, which are likely due to much higher treatment concentrations. It was also reported that all treated embryos exhibiting concentration- and duration-dependent abnormalities, as well as developmental delays, at 24 hpf. For instance, embryos exposed to PFOSA starting at 0.75 hpf exhibited stronger concentration-dependent delays in epiboly compared to embryos exposed at 4 or 5 hpf. Truong et al. (2022) exposed dechorionated zebrafish embryos to 0.015–100 µM (7.4–49,900 µg/L) of various PFAS, including PFOSA from 6 to 120 hpf [35]. PFAS were ranked on potency based on morphological effects (i.e., pericardial and yolk sac edema, brain and eye malformation) in which PFOSA was ranked second highest. Various studies also report cardiac abnormalities, including heart elongation and reduced cardiac output, heart rate, and stroke volume, in zebrafish exposed to concentrations in the range of 0.01–100 µg/L [16,17]. In our case with survival, 1 µg/L may have been too low a concentration to sufficiently activate defense mechanisms to PFOSA exposure, leading to lower overall survival in developing fish while higher concentrations of PFOSA may elicit a stronger defense response to mitigate toxicity, leading to higher survival. Such dose-dependent responses have been observed for other chemical exposures in zebrafish [36,37]. This hypothesis is supported by the increase in anti-oxidant defense enzymes with higher concentrations of PFOSA. Nevertheless, survival remained relatively high (80–90%) for fish exposed up to 100 µg/L PFOSA, suggesting that the chemical, spanning environmental concentrations, is not overtly toxic up to 100 µg/L.
The amount of ROS is often indicative of the amount of oxidative stress in cells, and an excess of ROS can contribute to damage at the molecular level. Limited studies in the literature examine the impact PFOSA has on organisms. In our study, we did not observe any increase in ROS in zebrafish treated with PFOSA; however, there was a concentration-dependent increase in both cat and sod2 expression. Mitochondrial dysfunction has been thought to contribute to the progression of neurodegenerative disorders and the presence of ROS is one clear indicator of dysfunction as antioxidant systems are implemented to counteract oxidative stress. This supports our oxidative stress-related gene responses (PFOSA-induced increase in cat and sod2 mRNA levels), which could have mitigated any change in ROS levels in the larval zebrafish. Similar results have also been observed in rodent models where oxidative stress-related genes, like cat, were significantly increased to counteract damage by PFOA-induced lipid peroxidation in mouse brain and liver tissues [38]. ROS can also trigger apoptosis to mediate inflammation. Though PFOSA was found to only significantly upregulate two apoptosis-related genes (bax and cycs) in our study, another study reports increased apoptotic cells in the brain and upregulated bcl-2, caspase3, and p53 zebrafish exposed to PFOS [39]. Bax is a pro-apoptotic factor in the Bcl-2 family, signaling mitochondria and cell death, while cytochrome c is an intrinsic apoptotic signal activating downstream caspase enzymes. Other studies investigating PFOSA report mixed results for antioxidant gene expression and proteins. Olufsen and Arukwe (2015) exposed Atlantic salmon (Salmo salar) hepatocytes to 25 or 50 µM (12,400–24,900 µg/L) PFOSA for 24 or 48 h and analyzed catalase, glutathione peroxidase, glucocorticoid receptor, and glutathione S-transferase mRNA levels, which were not significantly impacted [40]. Another study also exposed Atlantic salmon hepatocytes to 2, 20, or 50 µM (998, 9900, or 24,900 µg/L) PFOSA for 12 or 24 h [3]. No significant changes to gpx mRNA levels were found, but cat mRNA levels were significantly increased by 20 and 50 µM PFOSA following 24 h of exposure, suggesting that antioxidant defense mechanisms were activated. Differences among studies may occur due to the type of model used to investigate PFOSA toxicity (e.g., cells versus larvae). Taken together, there is evidence that PFOSA initiates an antioxidant defense and any elevation in ROS may lead to higher levels of apoptosis in larval zebrafish.
We hypothesized that PFOSA would induce neurotoxicity in the form of behavioral changes and altered expression of genes related to neurotoxicity. Indeed, we observed hypoactivity at 100 µg/L, which corresponded to the highest expression levels of several neurotoxicity biomarkers. These responses indicate some form of neurological impairment [41]. Chemical neurotoxins cause damage to, or death of, cells in the nervous system, disrupting neuronal pathways linked to neurodegenerative illnesses and other neurodevelopmental issues (i.e., Parkinson’s disease and schizophrenia). For instance, zebrafish exposed to PFOS had transcriptome changes linked to disturbance of the neuromuscular system [42] and zebrafish exposed to perfluorononanoic acid (PFNA) showed evidence of altered neuroinflammatory pathways [43]. To our knowledge, this is the first study to examine the neurotoxicity mechanism in zebrafish exposed to PFOSA. We observed hypoactivity effects on locomotor activity in larval zebrafish, suggesting neurotoxicity or motor deficits with PFOSA exposure. Fish treated with 100 µg/L PFOSA showed reduced activity in Dark Period 2 of the combined VMR. Truong et al. (2022) exposed dechorionated zebrafish embryos to 0.015–100 µM PFOSA from 6 to 120 hpf and found that PFOSA induced both a refractory and an excitatory phase of hyperactivity [35]. Consistent with our observation, after exposing zebrafish embryos to 1 or 10 µg/L PFOSA for 120 h, Liu et al. (2022) observed reductions in the total distance moved, average swimming velocity, and maximum acceleration in fish treated with 1 µg/L PFOSA [17]. Our results also revealed that PFOSA alters the expression of neurotoxicity-related genes, as notable effects were observed in elavl3, and positive associations were detected between PFOSA concentration and expression levels (e.g., ache, elavl3, gap43, syn2a, and tubb3). Elavl3 is expressed in different nervous system structures and is known to regulate neurogenesis [44]. Additionally, ache is involved in cholinergic functioning and syn2a is involved in dopamine and serotonin release. PFAS exposure has previously been shown to alter these transcripts; PFOS, the metabolic product of PFOSA, was reported to decrease ache expression [45] and perfluorododecanoate (PFDoA) decreased mRNA levels of elavl3, gap43, and syn2a [46]. Here, we report an elevation in the expression of elavl3 and many other neurotoxic-related transcripts with PFOSA exposure, and this may reflect a compensatory response to impaired neurogenesis and neurotransmitter release. Conversely, different types of PFAS may elicit unique responses in the CNS in relation to gene expression patterns. Regardless, there is evidence from the molecular response that PFOSA alters genes related to neuronal integrity and structure, suggesting the potential for neurotoxicity in developing larval fish. Thus, early developmental exposures to PFOSA may have long-lasting detrimental effects on the adult brain and this should be further investigated.

5. Conclusions

In summary, very few studies have been carried out on PFOSA and there is little information on PFOSA’s environmental presence and accumulation in aquatic species. PFOSA did not significantly impact the prevalence of malformations or reactive oxygen species generated in larval fish; however, PFOSA did affect locomotor activity and transcripts related to oxidative damage response, apoptosis, and neurotoxicity. Further mechanistic studies in zebrafish are warranted to further address PFOSA neurotoxicity in the CNS. This study contributes to our knowledge regarding the relative toxicity of PFAS on fish to assist future risk assessment methodologies of these concerning, persistent environmental pollutants.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/toxics12100723/s1: Figure S1: Independent VMR experiments; Table S1: Primers used for qPCR analysis.

Author Contributions

Conceptualization, C.J.M.; Writing—review and editing, A.S., C.D.E., C.E., E.I., E.M.A.V., I.K., L.A., M.K., M.L.R., N.D. and C.J.M.; Investigation, A.S., C.D.E., C.E., E.I., E.M.A.V., I.K., L.A., M.K., M.L.R. and N.D.; writing—original draft preparation, N.D. and E.I.; validation, A.S. and E.M.A.V.; supervision, C.J.M.; funding acquisition, C.J.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the University of Florida and the College of Veterinary Medicine (UF IACUC202300000140). E.M.A.V was supported by FAPESP (Fundação de Amparo à Pesquisa do Estado de São Paulo). I.K. would like to thank the Coordination for the Improvement of Higher Education Personnel (CAPES—code 001, Brazil) for scholarship support.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board Institutional Animal Care and Use Committee of University of Florida (UFIACUC202300000140).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data will be made available upon request.

Acknowledgments

We thank Kari Buchanan and the Animal Care Services at the University of Florida for their assistance with zebrafish husbandry and technical support.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Solan, M.E.; Lavado, R. The use of in vitro methods in assessing human health risks associated with short-chain perfluoroalkyl and polyfluoroalkyl substances (PFAS). J. Appl. Toxicol. 2022, 42, 1298–1309. [Google Scholar] [CrossRef] [PubMed]
  2. Jensen, A.A.; Warming, M. Short-chain polyfluoroalkyl substances (PFAS); The Danish Environmental Protection Agency: Odense, Denmark, 2015. [Google Scholar]
  3. Wågbø, A.M.; Cangialosi, M.V.; Cicero, N.; Letcher, R.J.; Arukwe, A. Perfluorooctane sulfonamide-mediated modulation of hepatocellular lipid homeostasis and oxidative stress responses in Atlantic salmon hepatocytes. Chem. Res. Toxicol. 2012, 25, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
  4. EWG. Perfluorooctanesulfonamide (PFOSA). Available online: https://www.ewg.org/tapwater/contaminant.php?contamcode=E314 (accessed on 27 July 2024).
  5. Gust, K.A.; Mylroie, J.E.; Kimble, A.N.; Wilbanks, M.S.; Steward, C.S.; Chapman, K.A.; Jensen, K.M.; Kennedy, A.J.; Krupa, P.M.; Waisner, S.A. Survival, Growth, and Reproduction Responses in a Three-Generation Exposure of the Zebrafish (Danio rerio) to Perfluorooctane Sulfonate. Environ. Toxicol. Chem. 2024, 43, 115–131. [Google Scholar] [CrossRef] [PubMed]
  6. Ma, T.; Wu, P.; Wang, L.; Li, Q.; Li, X.; Luo, Y. Toxicity of per-and polyfluoroalkyl substances to aquatic vertebrates. Front. Environ. Sci. 2023, 11, 1101100. [Google Scholar] [CrossRef]
  7. Razak, M.R.; Aris, A.Z.; Zainuddin, A.H.; Yusoff, F.M.; Yusof, Z.N.B.; Kim, S.D.; Kim, K.W. Acute toxicity and risk assessment of perfluorooctanoic acid (PFOA) and perfluorooctanesulfonate (PFOS) in tropical cladocerans Moina micrura. Chemosphere 2023, 313, 137377. [Google Scholar] [CrossRef] [PubMed]
  8. Konwick, B.J.; Tomy, G.T.; Ismail, N.; Peterson, J.T.; Fauver, R.J.; Higginbotham, D.; Fisk, A.T. Concentrations and patterns of perfluoroalkyl acids in Georgia, USA surface waters near and distant to a major use source. Environ. Toxicol. Chem. Int. J. 2008, 27, 2011–2018. [Google Scholar] [CrossRef]
  9. Anderson, R.H.; Long, G.C.; Porter, R.C.; Anderson, J.K. Occurrence of select perfluoroalkyl substances at US Air Force aqueous film-forming foam release sites other than fire training areas: Field validation of critical fate and transport properties. In Perfluoroalkyl Substances in the Environment; CRC Press: Boca Raton, FL, USA, 2018; pp. 353–372. [Google Scholar]
  10. Brusseau, M.L.; Anderson, R.H.; Guo, B. PFAS concentrations in soils: Background levels versus contaminated sites. Sci. Total Environ. 2020, 740, 140017. [Google Scholar] [CrossRef]
  11. Fair, P.A.; Wolf, B.; White, N.D.; Arnott, S.A.; Kannan, K.; Karthikraj, R.; Vena, J.E. Perfluoroalkyl substances (PFASs) in edible fish species from Charleston Harbor and tributaries, South Carolina, United States: Exposure and risk assessment. Environ. Res. 2019, 171, 266–277. [Google Scholar] [CrossRef]
  12. Li, X.; Yeung, L.W.Y.; Xu, M.; Taniyasu, S.; Lam, P.K.; Yamashita, N.; Dai, J. Perfluorooctane sulfonate (PFOS) and other fluorochemicals in fish blood collected near the outfall of wastewater treatment plant (WWTP) in Beijing. Environ. Pollut. 2008, 156, 1298–1303. [Google Scholar] [CrossRef]
  13. Brandsma, S.H.; Smithwick, M.; Solomon, K.; Small, J.; de Boer, J.; Muir, D.C. Dietary exposure of rainbow trout to 8:2 and 10:2 fluorotelomer alcohols and perfluorooctanesulfonamide: Uptake, transformation and elimination. Chemosphere 2011, 82, 253–258. [Google Scholar] [CrossRef]
  14. Hagenaars, A.; Vergauwen, L.; De Coen, W.; Knapen, D. Structure–activity relationship assessment of four perfluorinated chemicals using a prolonged zebrafish early life stage test. Chemosphere 2011, 82, 764–772. [Google Scholar] [CrossRef] [PubMed]
  15. Wasel, O.; Thompson, K.M.; Freeman, J.L. Assessment of unique behavioral, morphological, and molecular alterations in the comparative developmental toxicity profiles of PFOA, PFHxA, and PFBA using the zebrafish model system. Environ. Int. 2022, 170, 107642. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, H.; Qiu, W.; Yang, X.; Chen, F.; Chen, J.; Tang, L.; Zhong, H.; Magnuson, J.T.; Zheng, C.; Xu, E.G. Perfluorooctane sulfonamide (PFOSA) induces cardiotoxicity via aryl hydrocarbon receptor activation in zebrafish. Environ. Sci. Technol. 2022, 56, 8438–8448. [Google Scholar] [CrossRef] [PubMed]
  17. Liu, X.; Liu, S.; Qiu, W.; Magnuson, J.T.; Liu, Z.; Yang, G.; Chen, H.; Li, Y.; Xu, X.; Zheng, C. Cardiotoxicity of PFOA, PFOS, and PFOSA in early life stage zebrafish: Molecular changes to behavioral-level response. Sustain. Horiz. 2022, 3, 100027. [Google Scholar] [CrossRef]
  18. Dasgupta, S.; Reddam, A.; Liu, Z.; Liu, J.; Volz, D.C. High-content screening in zebrafish identifies perfluorooctanesulfonamide as a potent developmental toxicant. Environ. Pollut. 2020, 256, 113550. [Google Scholar] [CrossRef]
  19. Xuan, R.; Qiu, X.; Wang, J.; Liu, S.; Magnuson, J.T.; Xu, B.; Qiu, W.; Zheng, C. Hepatotoxic response of perfluorooctane sulfonamide (PFOSA) in early life stage zebrafish (Danio rerio) is greater than perfluorooctane sulfonate (PFOS). J. Hazard. Mater. 2024, 461, 132552. [Google Scholar] [CrossRef]
  20. Westerfield, M. The Zebrafish Book; A Guide for the Laboratory Use of Zebrafish (Danio Rerio); University of Oregon Press: Eugene, OR, USA, 2007. [Google Scholar]
  21. Cao, F.; Souders, C.L., II; Li, P.; Pang, S.; Liang, X.; Qiu, L.; Martyniuk, C.J. Developmental neurotoxicity of maneb: Notochord defects, mitochondrial dysfunction and hypoactivity in zebrafish (Danio rerio) embryos and larvae. Ecotoxicol. Environ. Saf. 2019, 170, 227–237. [Google Scholar] [CrossRef]
  22. Perez-Rodriguez, V.; Souders, C.L., II; Tischuk, C.; Martyniuk, C.J. Tebuconazole reduces basal oxidative respiration and promotes anxiolytic responses and hypoactivity in early-staged zebrafish (Danio rerio). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2019, 217, 87–97. [Google Scholar] [CrossRef]
  23. Kimmel, C.B.; Ballard, W.W.; Kimmel, S.R.; Ullmann, B.; Schilling, T.F. Stages of embryonic development of the zebrafish. Dev. Dyn. 1995, 203, 253–310. [Google Scholar] [CrossRef]
  24. Liang, X.; Adamovsky, O.; Souders, C.L., II; Martyniuk, C.J. Biological effects of the benzotriazole ultraviolet stabilizers UV-234 and UV-320 in early-staged zebrafish (Danio rerio). Environ. Pollut. 2019, 245, 272–281. [Google Scholar] [CrossRef]
  25. Dong, M.; Wang, J.; Liu, Y.; He, Q.; Sun, H.; Xu, Z.; Hong, H.; Lin, H.; Gao, P. 3-bromocarbazole-induced developmental neurotoxicity and effect mechanisms in zebrafish. ACS EST Water 2023, 3, 2471–2480. [Google Scholar] [CrossRef]
  26. Fang, C.; Di, S.; Yu, Y.; Qi, P.; Wang, X.; Jin, Y. 6PPD induced cardiac dysfunction in zebrafish associated with mitochondrial damage and inhibition of autophagy processes. J. Hazard. Mater. 2024, 471, 134357. [Google Scholar] [CrossRef] [PubMed]
  27. Guo, Y.; Fu, Y.; Sun, W. 50 Hz Magnetic Field Exposure Inhibited Spontaneous Movement of Zebrafish Larvae through ROS-Mediated syn2a Expression. Int. J. Mol. Sci. 2023, 24, 7576. [Google Scholar] [CrossRef] [PubMed]
  28. Hahn, M.E.; McArthur, A.G.; Karchner, S.I.; Franks, D.G.; Jenny, M.J.; Timme-Laragy, A.R.; Stegeman, J.J.; Woodin, B.R.; Cipriano, M.J.; Linney, E. The transcriptional response to oxidative stress during vertebrate development: Effects of tert-butylhydroquinone and 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin. PLoS ONE 2014, 9, e113158. [Google Scholar] [CrossRef] [PubMed]
  29. Jiang, F.; Liu, J.; Zeng, X.; Yu, L.; Liu, C.; Wang, J. Tris (2-butoxyethyl) phosphate affects motor behavior and axonal growth in zebrafish (Danio rerio) larvae. Aquat. Toxicol. 2018, 198, 215–223. [Google Scholar] [CrossRef]
  30. McCurley, A.T.; Callard, G.V. Characterization of housekeeping genes in zebrafish: Male-female differences and effects of tissue type, developmental stage and chemical treatment. BMC Mol. Biol. 2008, 9, 102. [Google Scholar] [CrossRef]
  31. Sarkar, S.; Mukherjee, S.; Chattopadhyay, A.; Bhattacharya, S. Low dose of arsenic trioxide triggers oxidative stress in zebrafish brain: Expression of antioxidant genes. Ecotoxicol. Environ. Saf. 2014, 107, 1–8. [Google Scholar] [CrossRef]
  32. Wang, X.H.; Souders, C.L., II; Zhao, Y.H.; Martyniuk, C.J. Paraquat affects mitochondrial bioenergetics, dopamine system expression, and locomotor activity in zebrafish (Danio rerio). Chemosphere 2018, 191, 106–117. [Google Scholar] [CrossRef]
  33. Wu, Q.; Yan, W.; Liu, C.; Li, L.; Yu, L.; Zhao, S.; Li, G. Microcystin-LR exposure induces developmental neurotoxicity in zebrafish embryo. Environ. Pollut. 2016, 213, 793–800. [Google Scholar] [CrossRef]
  34. Yang, Q.; Deng, P.; Xing, D.; Liu, H.; Shi, F.; Hu, L.; Zou, X.; Nie, H.; Zuo, J.; Zhuang, Z. Developmental neurotoxicity of difenoconazole in zebrafish embryos. Toxics 2023, 11, 353. [Google Scholar] [CrossRef]
  35. Truong, L.; Rericha, Y.; Thunga, P.; Marvel, S.; Wallis, D.; Simonich, M.T.; Field, J.A.; Cao, D.; Reif, D.M.; Tanguay, R.L. Systematic developmental toxicity assessment of a structurally diverse library of PFAS in zebrafish. J. Hazard. Mater. 2022, 431, 128615. [Google Scholar] [CrossRef] [PubMed]
  36. English, C.D.; Kazi, K.J.; Konig, I.; Ivantsova, E.; Souders, C.L., II; Martyniuk, C.J. Exposure to the antineoplastic ifosfamide alters molecular pathways related to cardiovascular function, increases heart rate, and induces hyperactivity in zebrafish (Danio rerio). Environ. Toxicol. Pharmacol. 2024, 107, 104427. [Google Scholar] [CrossRef] [PubMed]
  37. Ivantsova, E.; Huang, M.; Wengrovitz, A.S.; Souders, C.L., II; Martyniuk, C.J. Molecular and behavioral assessment in larval zebrafish (Danio rerio) following exposure to environmentally relevant levels of the antineoplastic cyclophosphamide. Environ. Toxicol. Pharmacol. 2022, 90, 103809. [Google Scholar] [CrossRef] [PubMed]
  38. Endirlik, B.Ü.; Eken, A.; Canpınar, H.; Öztürk, F.; Gürbay, A. Perfluorooctanoic acid affects mouse brain and liver tissue through oxidative stress. Arh. Za Hig. Rada I Toksikol. 2022, 73, 148–156. [Google Scholar] [CrossRef]
  39. Mahapatra, A.; Gupta, P.; Suman, A.; Ray, S.S.; Malafaia, G.; Singh, R.K. Unraveling the mechanisms of perfluorooctanesulfonic acid-induced dopaminergic neurotoxicity and microglial activation in developing zebrafish. Sci. Total Environ. 2023, 887, 164030. [Google Scholar] [CrossRef]
  40. Olufsen, M.; Arukwe, A. Endocrine, biotransformation, and oxidative stress responses in salmon hepatocytes exposed to chemically induced hypoxia and perfluorooctane sulfonamide (PFOSA), given singly or in combination. Environ. Sci. Pollut. Res. 2015, 22, 17350–17366. [Google Scholar] [CrossRef]
  41. Selderslaghs, I.W.; Hooyberghs, J.; De Coen, W.; Witters, H.E. Locomotor activity in zebrafish embryos: A new method to assess developmental neurotoxicity. Neurotoxicol. Teratol. 2010, 32, 460–471. [Google Scholar] [CrossRef]
  42. Wang, X.; Shi, X.; Zheng, S.; Zhang, Q.; Peng, J.; Tan, W.; Wu, K. Perfluorooctane sulfonic acid (PFOS) exposures interfere with behaviors and transcription of genes on nervous and muscle system in zebrafish embryos. Sci. Total Environ. 2022, 848, 157816. [Google Scholar] [CrossRef]
  43. Liu, S.; Qiu, W.; Li, R.; Chen, B.; Wu, X.; Magnuson, J.T.; Xu, B.; Luo, S.; Xu, E.G.; Zheng, C. Perfluorononanoic acid induces neurotoxicity via synaptogenesis signaling in zebrafish. Environ. Sci. Technol. 2023, 57, 3783–3793. [Google Scholar] [CrossRef]
  44. Ogawa, Y.; Kakumoto, K.; Yoshida, T.; Kuwako, K.I.; Miyazaki, T.; Yamaguchi, J.; Konno, A.; Hata, J.; Uchiyama, Y.; Hirai, H.; et al. Elavl3 is essential for the maintenance of Purkinje neuron axons. Sci. Rep. 2018, 8, 2722. [Google Scholar] [CrossRef]
  45. Starnes, H.M.; Rock, K.D.; Jackson, T.W.; Belcher, S.M. A critical review and meta-analysis of impacts of per-and polyfluorinated substances on the brain and behavior. Front. Toxicol. 2022, 4, 881584. [Google Scholar] [CrossRef] [PubMed]
  46. Foguth, R.; Sepúlveda, M.S.; Cannon, J. Per-and polyfluoroalkyl substances (PFAS) neurotoxicity in sentinel and non-traditional laboratory model systems: Potential utility in predicting adverse outcomes in human health. Toxics 2020, 8, 42. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Percent of surviving zebrafish following exposure to one of either ERM, 0.1% DMSO or 0.1, 1, 10, or 100 µg/L PFOSA over 7 days. Error bars (S.E.M.) are small and within symbols in some cases.
Figure 1. Percent of surviving zebrafish following exposure to one of either ERM, 0.1% DMSO or 0.1, 1, 10, or 100 µg/L PFOSA over 7 days. Error bars (S.E.M.) are small and within symbols in some cases.
Toxics 12 00723 g001
Figure 2. Total percent of hatched zebrafish embryos/larvae following exposure to one of either ERM, 0.1% DMSO or 0.1, 1, 10, or 100 µg/L PFOSA over 7 days.
Figure 2. Total percent of hatched zebrafish embryos/larvae following exposure to one of either ERM, 0.1% DMSO or 0.1, 1, 10, or 100 µg/L PFOSA over 7 days.
Toxics 12 00723 g002
Figure 3. Normalized reactive oxygen species (to µg/mL media/protein). Each circle represents a biological replicate; the mean (±S.D.) is represented by the horizontal line (One-Way ANOVA and Dunnett’s multiple comparisons test; n = 5/treatment). ns = not significant.
Figure 3. Normalized reactive oxygen species (to µg/mL media/protein). Each circle represents a biological replicate; the mean (±S.D.) is represented by the horizontal line (One-Way ANOVA and Dunnett’s multiple comparisons test; n = 5/treatment). ns = not significant.
Toxics 12 00723 g003
Figure 4. The distance moved in each of the light and dark zones (10 min bins) of 7-day zebrafish larvae exposed to 0.1% DMSO, ERM or 0.1, 1, 10, or 100 µg/L PFOSA. Graphs are the combined output from three independent VMR runs. Columns depict mean (±S.D.) (Kruskal–Wallis test and Dunn’s multiple comparisons test; n = 8–12 fish/treatment/run). Asterisk indicates difference at ** p < 0.01.
Figure 4. The distance moved in each of the light and dark zones (10 min bins) of 7-day zebrafish larvae exposed to 0.1% DMSO, ERM or 0.1, 1, 10, or 100 µg/L PFOSA. Graphs are the combined output from three independent VMR runs. Columns depict mean (±S.D.) (Kruskal–Wallis test and Dunn’s multiple comparisons test; n = 8–12 fish/treatment/run). Asterisk indicates difference at ** p < 0.01.
Toxics 12 00723 g004
Figure 5. Linear regression for relative expression of (A) bax, (B) cat, and (C) sod2 in 7-day old zebrafish. The DMSO and ERM group were combined (“Cntl”). Each circle indicates a biological replicate or beaker of pooled fish (n = 4 to 6). The solid line indicates the relationship between expression and the region between the two outermost dotted lines is the 95% confidence interval of the X intercept.
Figure 5. Linear regression for relative expression of (A) bax, (B) cat, and (C) sod2 in 7-day old zebrafish. The DMSO and ERM group were combined (“Cntl”). Each circle indicates a biological replicate or beaker of pooled fish (n = 4 to 6). The solid line indicates the relationship between expression and the region between the two outermost dotted lines is the 95% confidence interval of the X intercept.
Toxics 12 00723 g005
Figure 6. Relative expression of (A) bax and (B) elavl3 in 7-day old larval zebrafish exposed to 0.1% DMSO, ERM or 0.1, 1, or 10 µg/L PFOSA. Each circle is a beaker of pooled fish (biological replicate), and the mean (±S.D.) is indicated by the horizontal line (One-Way ANOVA, Dunnett’s multiple comparisons test; n = 4 to 6). Asterisk indicates * p < 0.05 or ** p < 0.01.
Figure 6. Relative expression of (A) bax and (B) elavl3 in 7-day old larval zebrafish exposed to 0.1% DMSO, ERM or 0.1, 1, or 10 µg/L PFOSA. Each circle is a beaker of pooled fish (biological replicate), and the mean (±S.D.) is indicated by the horizontal line (One-Way ANOVA, Dunnett’s multiple comparisons test; n = 4 to 6). Asterisk indicates * p < 0.05 or ** p < 0.01.
Toxics 12 00723 g006
Figure 7. Linear regression for relative expression of (A) ache, (B) elavl3, (C) gap43, (D) syn2a, and (E) tubulin (tubb3) in 7-day old zebrafish. The DMSO and ERM group were combined (“Cntl”). Each circle indicates a biological replicate or beaker of pooled fish (n = 4 to 6). The solid line indicates the relationship between expression and the region between the two outermost dotted lines is the 95% confidence interval of the X intercept.
Figure 7. Linear regression for relative expression of (A) ache, (B) elavl3, (C) gap43, (D) syn2a, and (E) tubulin (tubb3) in 7-day old zebrafish. The DMSO and ERM group were combined (“Cntl”). Each circle indicates a biological replicate or beaker of pooled fish (n = 4 to 6). The solid line indicates the relationship between expression and the region between the two outermost dotted lines is the 95% confidence interval of the X intercept.
Toxics 12 00723 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

David, N.; Ivantsova, E.; Konig, I.; English, C.D.; Avidan, L.; Kreychman, M.; Rivera, M.L.; Escobar, C.; Valle, E.M.A.; Sultan, A.; et al. Adverse Outcomes Following Exposure to Perfluorooctanesulfonamide (PFOSA) in Larval Zebrafish (Danio rerio): A Neurotoxic and Behavioral Perspective. Toxics 2024, 12, 723. https://doi.org/10.3390/toxics12100723

AMA Style

David N, Ivantsova E, Konig I, English CD, Avidan L, Kreychman M, Rivera ML, Escobar C, Valle EMA, Sultan A, et al. Adverse Outcomes Following Exposure to Perfluorooctanesulfonamide (PFOSA) in Larval Zebrafish (Danio rerio): A Neurotoxic and Behavioral Perspective. Toxics. 2024; 12(10):723. https://doi.org/10.3390/toxics12100723

Chicago/Turabian Style

David, Nikita, Emma Ivantsova, Isaac Konig, Cole D. English, Lev Avidan, Mark Kreychman, Mario L. Rivera, Camilo Escobar, Eliana Maira Agostini Valle, Amany Sultan, and et al. 2024. "Adverse Outcomes Following Exposure to Perfluorooctanesulfonamide (PFOSA) in Larval Zebrafish (Danio rerio): A Neurotoxic and Behavioral Perspective" Toxics 12, no. 10: 723. https://doi.org/10.3390/toxics12100723

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop