Network


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

Hotspot


Dive into the research topics where Erin P. Hines is active.

Publication


Featured researches published by Erin P. Hines.


The Journal of Steroid Biochemistry and Molecular Biology | 2011

Endocrine disrupting properties of perfluorooctanoic acid

Sally S. White; Suzanne E. Fenton; Erin P. Hines

Perfluoroalkyl acids (PFAAs) have attracted attention in recent years for their environmental ubiquity, as well as their toxicity. Several PFAAs are found in human tissues globally, as humans are exposed on a daily basis through intake of contaminated food, water, and air, irrespective of proximity to industry. Perfluorooctanoic acid (PFOA) is a PFAA shown to be developmentally toxic in mice, with broad and varied health consequences that may include long-lasting effects in reproductive tissues and metabolic reprogramming. To date, the only demonstrated mode of action by which the health effects of PFOA are mediated is via the activation of the peroxisome proliferator-activated receptor alpha (PPARα). The endogenous roles for this receptor, as well as the adverse outcomes of activation by exogenous agents during development, are currently under investigation. Recent studies suggest that PFOA may alter steroid hormone production or act indirectly, via ovarian effects, as a novel means of endocrine disruption. Here we review the existing literature on the known health effects of PFOA in animal models, focusing on sensitive developmental periods. To complement this, we also present epidemiologic health data, with the caveat that these studies largely address only associations between adult exposures and outcomes, rarely focusing on endocrine-specific endpoints, susceptible subpopulations, or windows of sensitivity. Further research in these areas is needed.


Molecular and Cellular Endocrinology | 2009

Phenotypic dichotomy following developmental exposure to perfluorooctanoic acid (PFOA) in female CD-1 mice: low doses induce elevated serum leptin and insulin, and overweight in mid-life.

Erin P. Hines; Sally S. White; Jason P. Stanko; Eugene A. Gibbs-Flournoy; Christopher Lau; Suzanne E. Fenton

The synthetic surfactant, perfluorooctanoic acid (PFOA) is a proven developmental toxicant in mice, causing pregnancy loss, increased neonatal mortality, delayed eye opening, and abnormal mammary gland growth in animals exposed during fetal life. PFOA is found in the sera and tissues of wildlife and humans throughout the world, but is especially high in the sera of children compared to adults. These studies in CD-1 mice aim to determine the latent health effects of PFOA following: (1) an in utero exposure, (2) an in utero exposure followed by ovariectomy (ovx), or (3) exposure as an adult. Mice were exposed to 0, 0.01, 0.1, 0.3, 1, 3, or 5mg PFOA/kg BW for 17 days of pregnancy or as young adults. Body weight was reduced in the highest doses on postnatal day (PND) 1 and at weaning. However, the lowest exposures (0.01-0.3mg/kg) significantly increased body weight, and serum insulin and leptin (0.01-0.1mg/kg) in mid-life after developmental exposure. PFOA exposure combined with ovx caused no additional increase in mid-life body weight. At 18 months of age, the effects of in utero PFOA exposure on body weight were no longer detected. White adipose tissue and spleen weights were decreased at high doses of PFOA in intact developmentally exposed mice, and spleen weight was reduced in PFOA-exposed ovx mice. Brown adipose tissue weight was significantly increased in both ovx and intact mice at high PFOA doses. Liver weight was unaffected in late life by these exposure paradigms. Finally, there was no effect of adult exposure to PFOA on body weight. These studies demonstrate an important window of exposure for low-dose effects of PFOA on body weight gain, as well as leptin and insulin concentrations in mid-life, at a lowest observed effect level of 0.01mg PFOA/kg BW. The mode of action of these effects and its relevance to human health remain to be explored.


Environmental Health Perspectives | 2009

Concentrations of Phthalate Metabolites in Milk, Urine, Saliva, and Serum of Lactating North Carolina Women

Erin P. Hines; Antonia M. Calafat; Manori J. Silva; Pauline Mendola; Suzanne E. Fenton

Background Phthalates are ubiquitous in the environment, but concentrations in multiple media from breast-feeding U.S. women have not been evaluated. Objectives The objective of this study was to accurately measure and compare the concentrations of oxidative monoester phthalate metabolites in milk and surrogate fluids (serum, saliva, and urine) of 33 lactating North Carolina women. Methods We analyzed serum, saliva, urine, and milk for the oxidative phthalate metabolites mono(3-carboxypropyl) phthalate, mono(2-ethyl-5-carboxypentyl) phthalate (MECPP), mono(2-ethyl-5-hydroxyhexyl) phthalate, and mono(2-ethyl-5-oxohexyl) phthalate using isotope-dilution high-performance liquid chromatography tandem mass spectroscopy. Because only urine lacks esterases, we analyzed it for the hydrolytic phthalate monoesters. Results We detected phthalate metabolites in few milk (< 10%) and saliva samples. MECPP was detected in > 80% of serum samples, but other metabolites were less common (3–22%). Seven of the 10 urinary metabolites were detectable in ≥ 85% of samples. Monoethyl phthalate had the highest mean concentration in urine. Metabolite concentrations differed by body fluid (urine > serum > milk and saliva). Questionnaire data suggest that frequent nail polish use, immunoglobulin A, and fasting serum glucose and triglyceride levels were increased among women with higher concentrations of urinary and/or serum phthalate metabolites; motor vehicle age was inversely correlated with certain urinary phthalate concentrations. Conclusions Our data suggest that phthalate metabolites are most frequently detected in urine of lactating women and are less often detected in serum, milk, or saliva. Urinary phthalate concentrations reflect maternal exposure and do not represent the concentrations of oxidative metabolites in other body fluids, especially milk.


Reproductive Toxicology | 2009

Polyfluoroalkyl chemicals in the serum and milk of breastfeeding women.

Ondine S. von Ehrenstein; Suzanne E. Fenton; Kayoko Kato; Zsuzsanna Kuklenyik; Antonia M. Calafat; Erin P. Hines

Polyfluoroalkyl chemicals (PFCs) comprise a group of man-made organic compounds, some of which are persistent contaminants with developmental toxicity shown in laboratory animals. There is a paucity of human perinatal exposure data. The US EPA conducted a pilot study (Methods Advancement for Milk Analysis) including 34 breastfeeding women in North Carolina. Milk and serum samples were collected at 2-7 weeks and 3-4 months postpartum; 9 PFCs were assessed in milk and 7 in serum. Perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), and perfluorohexane sulfonic acid (PFHxS) were found in nearly 100% of the serum samples. PFOS and PFOA were found at the highest concentrations. PFCs were below the limit of quantification in most milk samples. Serum concentrations of PFOS, PFOA and PFHxS were lower (p<0.01) at the second visit compared to the first visit. Living in North Carolina 10 years or longer was related to elevated PFOS, PFOA and PFNA (p<or=0.03). These pilot data support the need to further explore perinatal PFC exposures and potentially related health effects, as planned in the upcoming National Childrens Study which provided the framework for this investigation.


Reproductive Toxicology | 2015

Concentrations of environmental phenols and parabens in milk, urine and serum of lactating North Carolina women

Erin P. Hines; Pauline Mendola; Ondine S. von Ehrenstein; Xiaoyun Ye; Antonia M. Calafat; Suzanne E. Fenton

Phenols and parabens show some evidence for endocrine disruption in laboratory animals. The goal of the Methods Advancement for Milk Analysis (MAMA) Study was to develop or adapt methods to measure parabens (methyl, ethyl, butyl, propyl) and phenols (bisphenol A (BPA), 2,4- and 2,5-dichlorophenol, benzophenone-3, triclosan) in urine, milk and serum twice during lactation, to compare concentrations across matrices and with endogenous biomarkers among 34 North Carolina women. These non-persistent chemicals were detected in most urine samples (53-100%) and less frequently in milk or serum; concentrations differed by matrix. Although urinary parabens, triclosan and dichlorophenols concentrations correlated significantly at two time points, those of BPA and benzophenone-3 did not, suggesting considerable variability in those exposures. These pilot data suggest that nursing mothers are exposed to phenols and parabens; urine is the best measurement matrix; and correlations between chemical and endogenous immune-related biomarkers merit further investigation.


Reproductive Toxicology | 2009

Effects of perfluorooctanoic acid on mouse mammary gland development and differentiation resulting from cross-foster and restricted gestational exposures ☆

Sally S. White; Kayoko Kato; Lily T. Jia; Brian J. Basden; Antonia M. Calafat; Erin P. Hines; Jason P. Stanko; Cynthia J. Wolf; Barbara D. Abbott; Suzanne E. Fenton

The adverse consequences of developmental exposures to perfluorooctanoic acid (PFOA) are established in mice, and include impaired development of the mammary gland (MG). However, the relationships between timing or route of exposure, and consequences in the MG have not been characterized. To address the effects of these variables on the onset and persistence of MG effects in female offspring, timed pregnant CD-1 dams received PFOA by oral gavage over various gestational durations. Cross-fostering studies identified the 5mg/kg dose, under either lactational- or intrauterine-only exposures, to delay MG development as early as postnatal day (PND) 1, persisting beyond PND 63. Intrauterine exposure during the final days of pregnancy caused adverse MG developmental effects similar to that of extended gestational exposures. These studies confirm a window of MG sensitivity in late fetal and early neonatal life, and demonstrate developmental PFOA exposure results in early and persistent MG effects, suggesting permanent consequences.


Environmental Health Perspectives | 2011

Gestational and chronic low-dose PFOA exposures and mammary gland growth and differentiation in three generations of CD-1 mice.

Sally S. White; Jason P. Stanko; Kayoko Kato; Antonia M. Calafat; Erin P. Hines; Suzanne E. Fenton

Background: Prenatal exposure to perfluorooctanoic acid (PFOA), a ubiquitous industrial surfactant, has been reported to delay mammary gland development in female mouse offspring (F1) and the treated lactating dam (P0) after gestational treatments at 3 and 5 mg PFOA/kg/day. Objective: We investigated the consequences of gestational and chronic PFOA exposure on F1 lactational function and subsequent development of F2 offspring. Methods: We treated P0 dams with 0, 1, or 5 mg PFOA/kg/day on gestation days 1–17. In addition, a second group of P0 dams treated with 0 or 1 mg/kg/day during gestation and their F1 and F2 offspring received continuous PFOA exposure (5 ppb) in drinking water. Resulting adult F1 females were bred to generate F2 offspring, whose development was monitored over postnatal days (PNDs) 1–63. F1 gland function was assessed on PND10 by timed-lactation experiments. Mammary tissue was isolated from P0, F1, and F2 females throughout the study and histologically assessed for age-appropriate development. Results: PFOA-exposed F1 dams exhibited diminished lactational morphology, although F1 maternal behavior and F2 offspring body weights were not significantly affected by P0 treatment. In addition to reduced gland development in F1 females under all exposures, F2 females with chronic low-dose drinking-water exposures exhibited visibly slowed mammary gland differentiation from weaning onward. F2 females derived from 5 mg/kg PFOA-treated P0 dams displayed gland morphology similar to F2 chronic water exposure groups on PNDs 22–63. Conclusions: Gestational PFOA exposure induced delays in mammary gland development and/or lactational differentiation across three generations. Chronic, low-dose PFOA exposure in drinking water was also sufficient to alter mammary morphological development in mice, at concentrations approximating those found in contaminated human water supplies.


Reproductive Toxicology | 2009

Analysis of PFOA in dosed CD-1 mice. Part 2: Disposition of PFOA in tissues and fluids from pregnant and lactating mice and their pups.

Suzanne E. Fenton; Jessica L. Reiner; Shoji F. Nakayama; Amy D. Delinsky; Jason P. Stanko; Erin P. Hines; Sally S. White; Andrew B. Lindstrom; Mark J. Strynar; Syrago-Styliani E. Petropoulou

Previous studies in mice with multiple gestational exposures to perfluorooctanoic acid (PFOA) demonstrate numerous dose dependent growth and developmental effects which appeared to worsen if offspring exposed in utero nursed from PFOA-exposed dams. To evaluate the disposition of PFOA in the pregnant and lactating dam and her offspring, time-pregnant CD-1 mice received a single 0, 0.1, 1, or 5mg PFOA/kg BW dose (n=25/dose group) by gavage on gestation day 17. Maternal and pup fluids and tissues were collected over time. Pups exhibited significantly higher serum PFOA concentrations than their respective dams, and their body burden increased after birth until at least postnatal day 8, regardless of dose. The distribution of milk:serum PFOA varied by dose and time, but was typically in excess of 0.20. These data suggest that milk is a substantial PFOA exposure route in mice and should be considered in risk assessment modeling designs for this compound.


Reproductive Toxicology | 2012

Histopathologic changes in the uterus, cervix and vagina of immature CD-1 mice exposed to low doses of perfluorooctanoic acid (PFOA) in a uterotrophic assay.

Darlene Dixon; Casey E. Reed; Alicia B. Moore; Eugene A. Gibbs-Flournoy; Erin P. Hines; Elizabeth A. Wallace; Jason P. Stanko; Yi Lu; Wendy N. Jefferson; Retha R. Newbold; Suzanne E. Fenton

The estrogenic and antiestrogenic potential of perfluorooctanoic acid (PFOA) was assessed using an immature mouse uterotrophic assay and by histologic evaluation of the uterus, cervix and vagina following treatment. Female offspring of CD-1 dams were weaned at 18days old and assigned to groups of equal weight, and received 0, 0.01, 0.1, or 1mg PFOA/kg BW/d by gavage with or without 17-β estradiol (E(2), 500μg/kg/d) from PND 18-20 (n=8/treatment/block). At 24h after the third dose (PND 21), uteri were removed and weighed. Absolute and relative uterine weights were significantly increased in the 0.01mg/kg PFOA only group. Characteristic estrogenic changes were present in all E(2)-treated mice; however, they were minimally visible in the 0.01 PFOA only mice. These data suggest that at a low dose PFOA produces minimal histopathologic changes in the reproductive tract of immature female mice, and does not antagonize the histopathologic effects of E(2).


Critical Reviews in Toxicology | 2014

Improving the risk assessment of lipophilic persistent environmental chemicals in breast milk

Geniece McCollum Lehmann; Marc-André Verner; Bryan Luukinen; Cara Henning; Sue Anne Assimon; Judy S. LaKind; Eva D. McLanahan; Linda Phillips; Matthew H. Davis; Christina M. Powers; Erin P. Hines; Sami Haddad; Matthew P. Longnecker; Michael T. Poulsen; David G. Farrer; Satori A. Marchitti; Yu Mei Tan; Jeffrey C. Swartout; Sharon K. Sagiv; Clement Welsh; Jerry L. Campbell; Warren G. Foster; Raymond S. H. Yang; Suzanne E. Fenton; Rogelio Tornero-Velez; Bettina M. Francis; John B. Barnett; Hisham A. El-Masri; Jane Ellen Simmons

Abstract Lipophilic persistent environmental chemicals (LPECs) have the potential to accumulate within a womans body lipids over the course of many years prior to pregnancy, to partition into human milk, and to transfer to infants upon breastfeeding. As a result of this accumulation and partitioning, a breastfeeding infants intake of these LPECs may be much greater than his/her mothers average daily exposure. Because the developmental period sets the stage for lifelong health, it is important to be able to accurately assess chemical exposures in early life. In many cases, current human health risk assessment methods do not account for differences between maternal and infant exposures to LPECs or for lifestage-specific effects of exposure to these chemicals. Because of their persistence and accumulation in body lipids and partitioning into breast milk, LPECs present unique challenges for each component of the human health risk assessment process, including hazard identification, dose–response assessment, and exposure assessment. Specific biological modeling approaches are available to support both dose–response and exposure assessment for lactational exposures to LPECs. Yet, lack of data limits the application of these approaches. The goal of this review is to outline the available approaches and to identify key issues that, if addressed, could improve efforts to apply these approaches to risk assessment of lactational exposure to these chemicals.

Collaboration


Dive into the Erin P. Hines's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sally S. White

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Antonia M. Calafat

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Jennifer Richmond-Bryant

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Ellen Kirrane

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Lisa Vinikoor-Imler

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

David Svendsgaard

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Dennis Kotchmar

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge