Network


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

Hotspot


Dive into the research topics where Cynthia J. Wolf is active.

Publication


Featured researches published by Cynthia J. Wolf.


Reproductive Toxicology | 2009

Developmental toxicity of perfluorooctane sulfonate (PFOS) is not dependent on expression of peroxisome proliferator activated receptor-alpha (PPARα) in the mouse

Barbara D. Abbott; Cynthia J. Wolf; Kaberi P. Das; Robert D. Zehr; Judith E. Schmid; Andrew B. Lindstrom; Mark J. Strynar; Christopher Lau

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are members of a family of perfluorinated compounds. Both are environmentally persistent and found in the serum of wildlife and humans. PFOS and PFOA are developmentally toxic in laboratory rodents. Exposure to these chemicals in utero delays development and reduces postnatal survival and growth. Exposure to PFOS on the last 4 days of gestation in the rat is sufficient to reduce neonatal survival. PFOS and PFOA are weak agonists of peroxisome proliferator activated receptor-alpha (PPAR alpha). The reduced postnatal survival of neonatal mice exposed to PFOA was recently shown to depend on expression of PPAR alpha. This study used PPAR alpha knockout (KO) and 129S1/SvlmJ wild type (WT) mice to determine if PPAR alpha expression is required for the developmental toxicity of PFOS. After mating overnight, the next day was designated gestation day (GD) 0. WT females were weighed and dosed orally from GD15 to 18 with 0.5% Tween-20, 4.5, 6.5, 8.5, or 10.5mg PFOS/kg/day. KO females were dosed with 0.5% Tween-20, 8.5 or 10.5mg PFOS/kg/day. Dams and pups were observed daily and pups were weighed on postnatal day (PND) 1 and PND15. Eye opening was recorded from PND12 to 15. Dams and pups were killed on PND15, body and liver weights recorded, and serum collected. PFOS did not affect maternal weight gain or body or liver weights of the dams on PND15. Neonatal survival (PND1-15) was significantly reduced by PFOS in both WT and KO litters at all doses. WT and KO pup birth weight and weight gain from PND1 to 15 were not significantly affected by PFOS exposure. Relative liver weight of WT and KO pups was significantly increased by the 10.5mg/kg dose. Eye opening of PFOS-exposed pups was slightly delayed in WT and KO on PND13 or 14, respectively. Because results in WT and KO were comparable, it is concluded that PFOS-induced neonatal lethality and delayed eye opening are not dependent on activation of PPAR alpha.


Ppar Research | 2010

Developmental Effects of Perfluorononanoic Acid in the Mouse Are Dependent on Peroxisome Proliferator-Activated Receptor-Alpha

Cynthia J. Wolf; Robert D. Zehr; Judy Schmid; Christopher Lau; Barbara D. Abbott

Perfluorononanoic acid (PFNA) is one of the perfluoroalkyl acids found in the environment and in tissues of humans and wildlife. Prenatal exposure to PFNA negatively impacts survival and development of mice and activates the mouse and human peroxisome proliferator-activated receptor-alpha (PPARα). In the current study, we used PPARα knockout (KO) and 129S1/SvlmJ wild-type (WT) mice to investigate the role of PPARα in mediating PFNA-induced in vivo effects. Pregnant KO and WT mice were dosed orally with water (vehicle control: 10u2009ml/kg), 0.83, 1.1, 1.5, or 2u2009mg/kg PFNA on gestational days (GDs) 1–18 (day of sperm plug = GD 0). Maternal weight gain, implantation, litter size, and pup weight at birth were unaffected in either strain. PFNA exposure reduced the number of live pups at birth and survival of offspring to weaning in the 1.1 and 2u2009mg/kg groups in WT. Eye opening was delayed (mean delay 2.1 days) and pup weight at weaning was reduced in WT pups at 2u2009mg/kg. These developmental endpoints were not affected in the KO. Relative liver weight was increased in a dose-dependent manner in dams and pups of the WT strain at all dose levels but only slightly increased in the highest dose group in the KO strain. In summary, PFNA altered liver weight of dams and pups, pup survival, body weight, and development in the WT, while only inducing a slight increase in relative liver weight of dams and pups at 2u2009mg/kg in KO mice. These results suggest that PPARα is an essential mediator of PFNA-induced developmental toxicity in the mouse.


Toxicology | 2013

Evaluation of perfluoroalkyl acid activity using primary mouse and human hepatocytes.

Mitchell B. Rosen; Kaberi P. Das; Carmen R. Wood; Cynthia J. Wolf; Barbara D. Abbott; Christopher Lau

While perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) have been studied at length, less is known about the biological activity of other perfluoroalkyl acids (PFAAs) detected in the environment. Using a transient transfection assay developed in COS-1 cells, our group has previously evaluated a variety of PFAAs for activity associated with activation of peroxisome proliferator-activated receptor alpha (PPARα). Here we use primary heptatocytes to further assess the biological activity of a similar group of PFAAs using custom designed Taqman Low Density Arrays. Primary mouse and human hepatoyctes were cultured for 48h in the presence of varying concentrations of 12 different PFAAs or Wy14,643, a known activator of PPARα. Total RNA was collected and the expression of 48 mouse or human genes evaluated. Gene selection was based on either in-house liver microarray data (mouse) or published data using primary hepatocytes (human). Gene expression in primary mouse hepatocytes was more restricted than expected. Genes typically regulated in whole tissue by PPARα agonists were not altered in mouse cells including Acox1, Me1, Acaa1a, Hmgcs1, and Slc27a1. Cyp2b10, a gene regulated by the constitutive androstane receptor and a transcript normally up-regulated by in vivo exposure to PFAAs, was also unchanged in cultured mouse hepatocytes. Cyp4a14, Ehhadh, Pdk4, Cpt1b, and Fabp1 were regulated as expected in mouse cells. A larger group of genes were differentially expressed in human primary hepatocytes, however, little consistency was observed across compounds with respect to which genes produced a significant dose response making the determination of relative biological activity difficult. This likely reflects weaker activation of PPARα in human versus rodent cells as well as variation among individual cell donors. Unlike mouse cells, CYP2B6 was up-regulated in human hepatocytes by a number of PFAAs as was PPARδ. Rankings were conducted on the limited dataset. In mouse hepatocytes, the pattern was similar to that previously observed in the COS-1 reporter cell assay. With the exception of PFHxA, longer chain PFAA carboxylates were the most active. The pattern was similar in human hepatocytes, although PFDA and PFOS showed higher activity than previously observed while PFOA showed somewhat less activity. These data reflect inherent challenges in using primary hepatocytes to predict toxicological response.


Toxicology | 2013

Testing for departures from additivity in mixtures of perfluoroalkyl acids (PFAAs)

Caroline Carr; Andrew M. Watkins; Cynthia J. Wolf; Barbara D. Abbott; Christopher Lau; Chris Gennings

This study is a follow-up to a paper by Carr et al. that determined a design structure to optimally test for departures from additivity in a fixed ratio mixture of four perfluoroalkyl acids (PFAAs) using an in vitro transiently-transfected COS-1 PPARα reporter model with a mixing ratio that is based on average serum levels in NHANES subjects. Availability of information regarding potential for additivity of PFAAs in mixtures is critically important for risk assessors who are concerned with the ability of the compounds to affect human health and impact ecological systems. It is clear that exposures are not to single compounds, but to mixtures of the PFAAs. This paper presents the results from the data collected using the design from Carr et al. along with subsequent analyses that were performed to classify the relationships among mixtures of PFAAs. A non-linear logistic additivity model was employed to predict relative luciferase units (RLU), an indicator of PPARα activation. The results indicated a less than additive relationship among the four PFAAs. To determine if the possible antagonism is from the competition among or between carboxylates and sulfonates, four different binary mixtures were also studied. There was a less than additive relationship in all four binary mixtures. These findings are generally similar to two other reports of interfering interactions between PFAAs in mixtures. The most conservative interpretation for our data would be an assumption of additivity (and lack of a greater than additive interaction), with a potential for antagonistic interactions.


Archive | 2002

Emerging Issues Related to Endocrine Disrupting Chemicals and Environmental Androgens and Antiandrogens

L. Earl GrayJr.; Christy Lambright; Louise Parks; Rochelle W. Tyl; Edward F. Orlando; J Louis GuilletteJr.; Cynthia J. Wolf; John C. Seely; Tsai-Yin Chang; Vickie S. Wilson; Andrew K. Hotchkiss; Joseph Ostby

Disclaimer. The research described in this article has been reviewed by the National Health and Environmental Effects Research Laboratory, U. S. Environmental Protection Agency, and approved for publication. Approval does not signify that the contents necessarily reflect the views and policies of the Agency nor does mention of trade names or commercial products constitute endorsement or recommendation for use.


Human Reproduction Update | 2001

Effects of environmental antiandrogens on reproductive development in experimental animals

L. E. Gray; Joseph Ostby; Johnathan Furr; Cynthia J. Wolf; Christy Lambright; Louise Parks; D.N.R. Veeramachaneni; Vickie S. Wilson; M. Price; Andrew K. Hotchkiss; Edward F. Orlando; Louis J. Guillette


Toxicology | 2006

Urinary biomarkers of di-isononyl phthalate in rats

Manori J. Silva; Kayoko Kato; Cynthia J. Wolf; Ella Samandar; Sathya S. Silva; Earl L. Gray; Larry L. Needham; Antonia M. Calafat


Toxicology | 2005

Urinary metabolites of di-n-octyl phthalate in rats

Manori J. Silva; Kayoko Kato; Earl L. Gray; Cynthia J. Wolf; Larry L. Needham; Antonia M. Calafat


Toxicology | 2007

Urinary metabolites of diisodecyl phthalate in rats.

Kayoko Kato; Manori J. Silva; Cynthia J. Wolf; L. Earl Gray; Larry L. Needham; Antonia M. Calafat


APMIS. Acta pathologica, microbiologica et immunologica scandinavica. Supplementum | 2001

Effects of environmental antiandrogens on reproductive development in experimental animals. Discussion

L. E. Gray; J. Ostby; Johnathan Furr; Cynthia J. Wolf; Christy S. Lambright; L. Parks; D. N. Veeramachaneni; Vickie S. Wilson; M. Price; Andrew K. Hotchkiss; E. Orlando; L. Guillette; John Ashby; Earl Gray; Melvin Grumbach; Charles Sultan; Ian Morris; Finn Bro-Rasmussen

Collaboration


Dive into the Cynthia J. Wolf's collaboration.

Top Co-Authors

Avatar

Barbara D. Abbott

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Christopher Lau

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonia M. Calafat

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Kayoko Kato

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Larry L. Needham

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Manori J. Silva

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Earl L. Gray

United States Environmental Protection Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge