Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John L. Butenhoff is active.

Publication


Featured researches published by John L. Butenhoff.


Environmental Health Perspectives | 2007

Half-Life of Serum Elimination of Perfluorooctanesulfonate, Perfluorohexanesulfonate, and Perfluorooctanoate in Retired Fluorochemical Production Workers

Geary W. Olsen; Jean M. Burris; David J. Ehresman; John W. Froehlich; Andrew M. Seacat; John L. Butenhoff; Larry R. Zobel

Background The presence of perfluorooctanesulfonate (PFOS), perfluorohexanesulfonate (PFHS), and perfluorooctanoate (PFOA) has been reported in humans and wildlife. Pharmacokinetic differences have been observed in laboratory animals. Objective The purpose of this observational study was to estimate the elimination half-life of PFOS, PFHS, and PFOA from human serum. Methods Twenty-six (24 male, 2 female) retired fluorochemical production workers, with no additional occupational exposure, had periodic blood samples collected over 5 years, with serum stored in plastic vials at −80°C. At the end of the study, we used HPLC-mass spectrometry to analyze the samples, with quantification based on the ion ratios for PFOS and PFHS and the internal standard 18O2-PFOS. For PFOA, quantitation was based on the internal standard 13C2-PFOA. Results The arithmetic mean initial serum concentrations were as follows: PFOS, 799 ng/mL (range, 145–3,490); PFHS, 290 ng/mL (range, 16–1,295); and PFOA, 691 ng/mL (range, 72–5,100). For each of the 26 subjects, the elimination appeared linear on a semi-log plot of concentration versus time; therefore, we used a first-order model for estimation. The arithmetic and geometric mean half-lives of serum elimination, respectively, were 5.4 years [95% confidence interval (CI), 3.9–6.9] and 4.8 years (95% CI, 4.0–5.8) for PFOS; 8.5 years (95% CI, 6.4–10.6) and 7.3 years (95% CI, 5.8–9.2) for PFHS; and 3.8 years (95% CI, 3.1–4.4) and 3.5 years (95% CI, 3.0–4.1) for PFOA. Conclusions Based on these data, humans appear to have a long half-life of serum elimination of PFOS, PFHS, and PFOA. Differences in species-specific pharmacokinetics may be due, in part, to a saturable renal resorption process.


Critical Reviews in Toxicology | 2004

The toxicology of perfluorooctanoate.

Gerald L. Kennedy; John L. Butenhoff; Geary W. Olsen; John C. O'Connor; Andrew M. Seacat; Roger G. Perkins; Lisa B. Biegel; Sandra R. Murphy; David G. Farrar

ABSTRACT PFOA is a peroxisome proliferator (PPAR agonist) and exerts morphological and biochemical effects characteristic of PPAR agonists. These effects include increased β-oxidation of fatty acids, increases in several cytochrome P-450 (CYP450)-mediated reactions, and inhibition of the secretion of very low-density lipoproteins and cholesterol from the liver. These effects on lipid metabolism and transport result in a reduction of cholesterol and triglycerides in serum and an accumulation of lipids in the liver. The triad of tumors observed (liver, Leydig cell, and pancreatic acinar-cell) is typical of many PPAR agonists and is believed to involve nongenotoxic mechanisms. The hepatocellular tumors observed in rats are likely to have been the result of the activation of the peroxisome proliferator activated receptor α (PPARα). The tumors observed in the testis (Leydig-cell) have been hypothesized to be associated with an increased level of serum estradiol in concert with testicular growth factors. The mechanism responsible for the acinarcell tumors of the pancreas in rats remains the subject of active investigation. The mechanism resulting in the hepatocellular tumors in rats (PPARα activation) is not likely to be relevant to humans. Similarly, the proposed mechanism for Leydig-cell tumor formation is of questionable relevance to humans. Acinar tumors of the pancreas are rare in humans, and the relevance of the these tumors, as found in rats, to humans is uncertain. Epidemiological investigations and medical surveillance of occupationally exposed workers have not found consistent associations between PFOA exposure and adverse health effects.


Toxicology | 2003

Sub-chronic dietary toxicity of potassium perfluorooctanesulfonate in rats.

Andrew M. Seacat; Peter J. Thomford; Kris J Hansen; Lisa A Clemen; Sandra R Eldridge; Cliff Elcombe; John L. Butenhoff

Perfluorooctanesulfonate (PFOS) is a widely disseminated persistent compound found at low (part-per-billion) concentrations in serum and liver samples from humans and fish-eating wildlife. This study investigated the hypotheses that early hepatocellular peroxisomal proliferation and hepatic cellular proliferation are factors in chronic liver response to dietary dosing, that lowering of serum total cholesterol is an early clinical measure of response to treatment, and that liver and serum PFOS concentrations are proportional to dose and cumulative dose after sub-chronic treatment. PFOS was administered in diet as the potassium salt at 0, 0.5, 2.0, 5.0, and 20 parts per million (ppm) to Sprague Dawley rats for 4 or 14 weeks. At 4 weeks, effects included decreased serum glucose and an equivocal (<twofold) increase in hepatic palmitoyl CoA oxidase (PCoAO) activity in 20 ppm dose-group males in one of two assay systems [corrected]. At 14 weeks, the 20 ppm males had increased liver weight, decreased serum cholesterol, increased non-segmented neutrophils, and increased ALT. Relative liver weights and urea nitrogen were increased in both sexes at 14 weeks. Hepatocytic hypertrophy and cytoplasmic vacuolation were observed in the 5 or 20 ppm male and the 20 ppm female dose groups. An increase in hepatic PCoAO activity was not observed at 14 weeks, and the average hepatocyte proliferation index was not increased, although, individual animals had mild increases. Serum and liver PFOS concentrations were proportional to dose and cumulative dose. Serum concentrations were generally higher in females than in males. The liver-to-serum PFOS ratios ranged from approximately 3:1 to 12:1. After 14 weeks, the no-observed-adverse effect level (NOAEL) in males and females was 5 ppm. The NOAEL corresponded to mean serum PFOS concentrations of 44 ppm (microg/ml) in males and 64 ppm in females and mean liver PFOS concentrations of 358 ppm in males and 370 ppm in females. Results for this study: (1) did not provide strong evidence for hepatocellular peroxisomal or cellular proliferation at the doses tested; (2) suggested that lowering of serum total cholesterol may not be the earliest clinically-measurable response to treatment in the rat; and (3) confirmed that serum and liver PFOS concentrations on repeated dosing are proportional to dose and cumulative dose.


Environmental Health Perspectives | 2005

Historical comparison of perfluorooctanesulfonate, perfluorooctanoate, and other fluorochemicals in human blood.

Geary W. Olsen; Han Yao Huang; Kathy J. Helzlsouer; Kristen J. Hansen; John L. Butenhoff; Jeffrey H. Mandel

The purpose of this investigation was to determine whether there has been a change in the human blood concentration of perfluorooctanesulfonate (PFOS), perfluorooctanoate (PFOA), and five other fluorochemicals since 1974. Blood samples were collected in 1974 (serum) and 1989 (plasma) from volunteer participants of a large community health study. The study included a total of 356 samples (178 from each time period). These samples were analyzed by high-pressure liquid chromatography/tandem mass spectrometry methods. The median 1974 and 1989 fluorochemical concentrations, respectively, were as follows: PFOS, 29.5 ng/mL vs. 34.7 ng/mL; PFOA, 2.3 ng/mL vs. 5.6 ng/mL; perfluorohexanesulfonate (PFHS), 1.6 ng/mL vs. 2.4 ng/mL; and N-ethyl perfluorooctanesulfonamidoacetate (PFOSAA), less than the lower limit of quantitation (LLOQ; 1.6 ng/mL, vs. 3.4 ng/mL). For N-methyl perfluorooctanesulfonamidoacetate (M570), perfluorooctanesulfonamide, and perfluorooctanesulfonamidoacetate, median serum concentrations in both years were less than the LLOQ values (1.0, 1.0, and 2.5 ng/mL, respectively). Statistical analysis of 58 paired samples indicated that serum concentrations of PFOS, PFOSAA, PFOA, PFHS, and M570 were significantly (p < 0.001) higher in 1989 than in 1974. The data from 1989 were then compared with geometric mean fluorochemical concentrations of serum samples collected in 2001 from 108 American Red Cross adult blood donors from the same region. Except for M570, there were no statistically significant (p < 0.05) geometric mean fluorochemical concentration differences between the 1989 and 2001 samples. In conclusion, based on this study population, PFOS and other serum fluorochemical concentrations have increased between 1974 and 1989. Comparison with other regional data collected in 2001 did not suggest a continued increase in concentrations since 1989.


Toxicology | 2002

Interactions of flurochemicals with rat liver fatty acid-binding protein

Deanna J. Luebker; Kris J Hansen; Nathan M. Bass; John L. Butenhoff; Andrew M. Seacat

Liver-fatty acid binding protein (L-FABP) is an abundant intracellular lipid-carrier protein. The hypothesis that perfluorooctanesulfonate (PFOS), perfluorooctanoate (PFOA), and certain related perfluorooctanesulfonamide-based fluorochemicals (PFOSAs) can interfere with the binding affinity of L-FABP for fatty acids was tested. The relative effectiveness of PFOA, PFOS, N-ethylperfluorooctanesulfonamide (N-EtFOSA), N-ethylperfluorooctanesulfonamido ethanol (N-EtFOSE), and of the strong peroxisome proliferator Wyeth-14 643 (WY) to inhibit 11-(5-dimethylaminonapthalenesulphonyl)-undecanoic acid (DAUDA) binding to-L-FABP was determined. The dissociation constant (Kd) of the DAUDA-L-FABP complex was 0.47 nM. PFOS exhibited the highest level of inhibition of DAUDA-L-FABP binding in the competitive binding assays, followed by N-EtFOSA, WY, and, with equal IC50s, N-EtFOSE and PFOA. The in vitro data presented in this study support the hypothesis that these fluorochemicals may interfere with the binding of fatty acids or other endogenous ligands to L-FABP. Furthermore, this work provides evidence to support the hypothesis that displacement of endogenous ligands from L-FABP may contribute to toxicity in rodents fed these fluorochemicals.


Environmental Health Perspectives | 2006

The Applicability of Biomonitoring Data for Perfluorooctanesulfonate to the Environmental Public Health Continuum

John L. Butenhoff; Geary W. Olsen; Andrea Pfahles-Hutchens

Perfluorooctanesulfonate and its salts (PFOS) are derived from perfluorooctanesulfonyl fluoride, the basic chemical building block for many sulfonyl-based fluorochemicals used as surfactants and for their repellent properties. PFOS is highly persistent in the environment and has a long serum elimination half-life in both animals and humans. PFOS has been detected globally in the environment and in blood serum in various populations throughout the world, with the majority of human sampling done in the United States and Japan. The mechanisms and pathways leading to the presence of PFOS in human blood are not well characterized but likely involve both direct exposures to PFOS or chemicals and materials that can degrade to PFOS, either in the environment or from industrial and commercial uses. In 2000 the 3M Company, a major manufacturer, announced a phaseout of PFOS-related materials. Animal studies indicate that PFOS is well absorbed orally and distributes mainly in blood serum and the liver. Several repeat-dose toxicology studies in animals consistently demonstrated that the liver is the primary target organ. In addition there is a steep dose response for mortality in sexually mature rats and primates as well as in neonatal rats and mice exposed in utero. Several biomonitoring research needs that have been identified on PFOS include additional data from general populations pertaining to other matrices besides blood; matched serum and urine samples from humans and research animals; and comparison of whole blood, serum, and plasma concentrations from the same individuals.


Environment International | 2011

A temporal trend study (1972-2008) of perfluorooctanesulfonate, perfluorohexanesulfonate, and perfluorooctanoate in pooled human milk samples from Stockholm, Sweden

Maria Sundström; David J. Ehresman; Anders Bignert; John L. Butenhoff; Geary W. Olsen; Shu-Ching Chang; Åke Bergman

The widespread presence of perfluorooctanesulfonate (PFOS), perfluorooctanoate (PFOA), and perfluorohexanesulfonate (PFHxS) in human general populations and their slow elimination profiles have led to renewed interest in understanding the potential human neonatal exposures of perfluoroalkyls (PFAs) from consumption of human milk. The objective of this study was to evaluate the concentrations of PFOS, PFHxS, and PFOA in pooled human milk samples obtained in Sweden between 1972 and 2008 (a period representing the most significant period of PFA production) and to see whether the time trend of these analytes parallels that indicated in human serum. Chemical analysis of PFOS, PFHxS, and PFOA was performed on pooled Swedish human milk samples from 1972 to 2008 after methodological refinements. The 20 samples which formed the 2007 pool were also analyzed individually to evaluate sample variations. Analyses were performed by HPLC-MS/MS. Due to the complexities of the human milk matrix and the requirement to accurately quantitate low pg/mL concentrations, meticulous attention must be paid to background contamination if accurate results are to be obtained. PFOS was the predominant analyte present in the pools and all three analytes showed statistically significant increasing trends from 1972 to 2000, with concentrations reaching a plateau in the 1990s. PFOA and PFOS showed statistically significant decreasing trends during 2001-2008. At the end of the study, in 2008, the measured concentrations of PFOS, PFHxS, and PFOA in pooled human milk were 75 pg/mL, 14 pg/mL, and 74 pg/mL, respectively. The temporal concentration trends of PFOS, PFHxS, and PFOA observed in human milk are parallel to those reported in the general population serum concentrations.


Journal of Children's Health | 2004

Quantitative Evaluation of Perfluorooctanesulfonate (PFOS) and Other Fluorochemicals in the Serum of Children

Geary W. Olsen; Timothy R. Church; Kristen J. Hansen; Jean M. Burris; John L. Butenhoff; Jeffrey H. Mandel; Larry R. Zobel

ABSTRACTPerfluorooctanesulfonyl fluoride (POSF)–based materials include surfactants, paper and packaging treatments, and surface (e.g., carpet, upholstery, textile) protectants. A metabolite, perfluorooctanesulfonate (PFOS, C8F17SO3−), has been identified in the serum and liver tissue of nonoccupationally exposed adults and wildlife. Results from several repeat-dose toxicological studies consistently demonstrate that the liver is the primary target organ with an apparent threshold for the toxic effects of PFOS that can be expressed in terms of cumulative dose or body burden. The purpose of this study was to characterize the distribution of PFOS and six other fluorochemicals in 598 serum samples obtained from a multi-center study of children (ages 2–12) diagnosed with group A streptococcal infections. Using high-pressure liquid chromatography tandem mass spectrometry methods, serum PFOS concentrations ranged from 6.7 ppb (ng/mL) to 515 ppb (geometric mean 37.5 ppb, 95% CI 36.0–39.1) with an estimate of the...


Reproductive Toxicology | 2009

Gestational and lactational exposure to potassium perfluorooctanesulfonate (K+PFOS) in rats: toxicokinetics, thyroid hormone status, and related gene expression.

Shu Ching Chang; David J. Ehresman; James A. Bjork; Kendall B. Wallace; George A. Parker; Donald G. Stump; John L. Butenhoff

Perfluorooctanesulfonate (PFOS), a persistent and accumulative compound, is widely distributed in humans and wildlife. Human exposure can occur early in development, as evidenced by the detection of PFOS in umbilical cord blood and breast milk. As part of a developmental neurotoxicology study for which developmental endpoints, including those related to the developing nervous system, have been reported separately, groups of 25 pregnant Sprague Dawley rats were given daily oral doses of either vehicle control or potassium PFOS (K(+)PFOS) at 0.1, 0.3, and 1.0mg/kg-d from gestation day (GD) 0 (day positive for mating) through postnatal day (PND) 20. An additional 10 pregnant females per treatment group were treated through GD 19 and sacrificed on GD 20 in order to obtain maternal and fetal serum and tissue samples at the end of gestation. The present paper reports the results of samples of serum, liver, brain, and thyroid glands taken at various times to evaluate: (1) serum, liver, and brain PFOS concentrations by LC-MS/MS to establish the relationship between PFOS concentrations and study outcomes; (2) serum thyrotropin (TSH) concentrations by RIA; (3) thyroid follicular cell proliferation index by Ki-67 immunohistochemical staining; (4) thyroid follicle epithelial cell height and colloidal area by histomorphometric analysis; (5) selected liver mRNA transcripts by quantitative RT-PCR. PFOS concentrations in dam and pup serum, liver, and brain increased across treatment groups in approximate proportion to the proportional increases in maternal K(+)PFOS dose, and sex differences in PFOS concentrations were not apparent in pups on PND 21. In pups from K(+)PFOS maternal dose groups on PND 72, serum PFOS had decreased to about 3 and 11% of PND 21 concentrations in males and females, respectively, and liver PFOS had decreased to about 17% of PND 21 concentrations in both sexes. Liver PFOS concentrations were approximately 0.6-0.8 times serum PFOS in GD 20 fetuses, and increased to about 2-4 times serum concentrations on PND 4 and 21. GD 20 fetal and PND 4 pup brain PFOS concentrations were approximately 33% of the corresponding serum concentrations, dropping to approximately 10% by PND 21, in contrast to dam brain PFOS concentrations, which were approximately 4-9% of serum PFOS concentrations. Compared to controls, Cyp2b2 mRNA was increased (2.8-fold) in the 1.0mg/kg-d treatment-group dams on GD 20. In male pups on PND 21, Cyp4A1, ACoA, and Cyp2b2 were increased 2.1-, 1.5-, and 1.8-fold, respectively, and Cyp7A1 was decreased 3.5-fold. Serum TSH and thyroid follicular morphology were not altered by K(+)PFOS treatment. The mean number of proliferating thyroid follicular cells was increased 2.1-fold over control in GD 20 female fetuses from 1.0mg/kg-d-treated dams, yet the highest individual count was similar to that of controls (116 versus 113 in controls).


Toxicology | 2011

Multiplicity of nuclear receptor activation by PFOA and PFOS in primary human and rodent hepatocytes.

James A. Bjork; John L. Butenhoff; Kendall B. Wallace

Perfluorooctanoate (PFOA) and perfluorooctanesulfonate (PFOS) are surface active fluorochemicals that, due to their exceptional stability to degradation, are persistent in the environment. Both PFOA and PFOS are eliminated slowly in humans, with geometric mean serum elimination half-lives estimated at 3.5 and 4.8 years, respectively. The biological activity of PFOA and PFOS in rodents is attributed primarily to transactivation of the nuclear receptor peroxisome proliferator activated receptor alpha (PPARA), which is an important regulator of lipid and carbohydrate metabolism. However, there are significant species-specific differences in the response to PFOA and PFOS exposure; non-rodent species, including humans, are refractory to several but not all of these effects. Many of the metabolic effects have been attributed to the activation of PPARA; however, recent studies using PPARα knockout mice demonstrate residual PPARA-independent effects, some of which may involve the activation of alternate nuclear receptors, including NR1I2 (PXR), NR1I3 (CAR), NR1H3 (LXRA), and NR1H4 (FXR). The objective of this investigation was to characterize the activation of multiple nuclear receptors and modulation of metabolic pathways associated with exposure to PFOA and PFOS, and to compare and contrast the effects between rat and human primary liver cells using quantitative reverse transcription PCR (RT-qPCR). Our results demonstrate that multiple nuclear receptors participate in the metabolic response to PFOA and PFOS exposure resulting in a substantial shift from carbohydrate metabolism to fatty acid oxidation and hepatic triglyceride accumulation in rat liver cells. This shift in intermediary metabolism was more pronounced for PFOA than PFOS. Furthermore, while there is some similarity in the activation of metabolic pathways between rat and humans, particularly in PPARA regulated responses; the changes in primary human cells were more subtle and possibly reflect an adaptive metabolic response rather than an overt metabolic regulation observed in rodents.

Collaboration


Dive into the John L. Butenhoff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher Lau

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George A. Parker

Charles River Laboratories

View shared research outputs
Top Co-Authors

Avatar

Jeffrey M. Peters

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie R. Thibodeaux

United States Environmental Protection Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge