Network


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

Hotspot


Dive into the research topics where Barbara A. Wetmore is active.

Publication


Featured researches published by Barbara A. Wetmore.


Toxicological Sciences | 2012

Integration of Dosimetry, Exposure and High-Throughput Screening Data in Chemical Toxicity Assessment

Barbara A. Wetmore; John F. Wambaugh; Stephen S. Ferguson; Mark A. Sochaski; Daniel M. Rotroff; Kimberly Freeman; Harvey J. Clewell; David J. Dix; Melvin E. Andersen; Keith A. Houck; Brittany Allen; Richard S. Judson; Reetu R. Singh; Robert J. Kavlock; Ann M. Richard; Russell S. Thomas

High-throughput in vitro toxicity screening can provide an efficient way to identify potential biological targets for chemicals. However, relying on nominal assay concentrations may misrepresent potential in vivo effects of these chemicals due to differences in bioavailability, clearance, and exposure. Hepatic metabolic clearance and plasma protein binding were experimentally measured for 239 ToxCast Phase I chemicals. The experimental data were used in a population-based in vitro-to-in vivo extrapolation model to estimate the daily human oral dose, called the oral equivalent dose, necessary to produce steady-state in vivo blood concentrations equivalent to in vitro AC(50) (concentration at 50% of maximum activity) or lowest effective concentration values across more than 500 in vitro assays. The estimated steady-state oral equivalent doses associated with the in vitro assays were compared with chronic aggregate human oral exposure estimates to assess whether in vitro bioactivity would be expected at the dose-equivalent level of human exposure. A total of 18 (9.9%) chemicals for which human oral exposure estimates were available had oral equivalent doses at levels equal to or less than the highest estimated U.S. population exposures. Ranking the chemicals by nominal assay concentrations would have resulted in different chemicals being prioritized. The in vitro assay endpoints with oral equivalent doses lower than the human exposure estimates included cell growth kinetics, cytokine and cytochrome P450 expression, and cytochrome P450 inhibition. The incorporation of dosimetry and exposure provide necessary context for interpretation of in vitro toxicity screening data and are important considerations in determining chemical testing priorities.


Toxicological Sciences | 2010

Incorporating Human Dosimetry and Exposure into High-Throughput In Vitro Toxicity Screening

Daniel M. Rotroff; Barbara A. Wetmore; David J. Dix; Stephen S. Ferguson; Harvey J. Clewell; Keith A. Houck; Edward L. LeCluyse; Melvin E. Andersen; Richard S. Judson; Cornelia M. Smith; Mark A. Sochaski; Robert J. Kavlock; Frank Boellmann; Matthew T. Martin; David M. Reif; John F. Wambaugh; Russell S. Thomas

Many chemicals in commerce today have undergone limited or no safety testing. To reduce the number of untested chemicals and prioritize limited testing resources, several governmental programs are using high-throughput in vitro screens for assessing chemical effects across multiple cellular pathways. In this study, metabolic clearance and plasma protein binding were experimentally measured for 35 ToxCast phase I chemicals. The experimental data were used to parameterize a population-based in vitro-to-in vivo extrapolation model for estimating the human oral equivalent dose necessary to produce a steady-state in vivo concentration equivalent to in vitro AC(50) (concentration at 50% of maximum activity) and LEC (lowest effective concentration) values from the ToxCast data. For 23 of the 35 chemicals, the range of oral equivalent doses for up to 398 ToxCast assays was compared with chronic aggregate human oral exposure estimates in order to assess whether significant in vitro bioactivity occurred within the range of maximum expected human oral exposure. Only 2 of the 35 chemicals, triclosan and pyrithiobac-sodium, had overlapping oral equivalent doses and estimated human oral exposures. Ranking by the potencies of the AC(50) and LEC values, these two chemicals would not have been at the top of a prioritization list. Integrating both dosimetry and human exposure information with the high-throughput toxicity screening efforts provides a better basis for making informed decisions on chemical testing priorities and regulatory attention. Importantly, these tools are necessary to move beyond hazard rankings to estimates of possible in vivo responses based on in vitro screens.


Chemical Research in Toxicology | 2011

Estimating toxicity-related biological pathway altering doses for high-throughput chemical risk assessment.

Richard S. Judson; Robert J. Kavlock; R. Woodrow Setzer; Elaine A. Cohen Hubal; Matthew T. Martin; Thomas B. Knudsen; Keith A. Houck; Russell S. Thomas; Barbara A. Wetmore; David J. Dix

We describe a framework for estimating the human dose at which a chemical significantly alters a biological pathway in vivo, making use of in vitro assay data and an in vitro-derived pharmacokinetic model, coupled with estimates of population variability and uncertainty. The quantity we calculate, the biological pathway altering dose (BPAD), is analogous to current risk assessment metrics in that it combines dose-response data with analysis of uncertainty and population variability to arrive at conservative exposure limits. The analogy is closest when perturbation of a pathway is a key event in the mode of action (MOA) leading to a specified adverse outcome. Because BPADs are derived from relatively inexpensive, high-throughput screening (HTS) in vitro data, this approach can be applied to high-throughput risk assessments (HTRA) for thousands of data-poor environmental chemicals. We envisage the first step of HTRA to be an assessment of in vitro concentration-response relationships across biologically important pathways to derive biological pathway altering concentrations (BPAC). Pharmacokinetic (PK) modeling is then used to estimate the in vivo doses required to achieve the BPACs in the blood at steady state. Uncertainty and variability are incorporated in both the BPAC and the PK parameters and then combined to yield a probability distribution for the dose required to perturb the critical pathway. We finally define the BPADL as the lower confidence bound of this pathway-altering dose. This perspective outlines a framework for using HTRA to estimate BPAD values; provides examples of the use of this approach, including a comparison of BPAD values with published dose-response data from in vivo studies; and discusses challenges and alternative formulations.


Toxicological Sciences | 2013

Incorporating new technologies into toxicity testing and risk assessment: moving from 21st century vision to a data-driven framework.

Russell S. Thomas; Martin A. Philbert; Scott S. Auerbach; Barbara A. Wetmore; Michael J. DeVito; Ila Cote; J. Craig Rowlands; Maurice Whelan; Sean M. Hays; Melvin E. Andersen; M. E. (Bette) Meek; Lawrence W. Reiter; Jason C. Lambert; Harvey J. Clewell; Martin L. Stephens; Q. Jay Zhao; Scott C. Wesselkamper; Lynn Flowers; Edward W. Carney; Timothy P. Pastoor; Dan D. Petersen; Carole L. Yauk; Andy Nong

Based on existing data and previous work, a series of studies is proposed as a basis toward a pragmatic early step in transforming toxicity testing. These studies were assembled into a data-driven framework that invokes successive tiers of testing with margin of exposure (MOE) as the primary metric. The first tier of the framework integrates data from high-throughput in vitro assays, in vitro-to-in vivo extrapolation (IVIVE) pharmacokinetic modeling, and exposure modeling. The in vitro assays are used to separate chemicals based on their relative selectivity in interacting with biological targets and identify the concentration at which these interactions occur. The IVIVE modeling converts in vitro concentrations into external dose for calculation of the point of departure (POD) and comparisons to human exposure estimates to yield a MOE. The second tier involves short-term in vivo studies, expanded pharmacokinetic evaluations, and refined human exposure estimates. The results from the second tier studies provide more accurate estimates of the POD and the MOE. The third tier contains the traditional animal studies currently used to assess chemical safety. In each tier, the POD for selective chemicals is based primarily on endpoints associated with a proposed mode of action, whereas the POD for nonselective chemicals is based on potential biological perturbation. Based on the MOE, a significant percentage of chemicals evaluated in the first 2 tiers could be eliminated from further testing. The framework provides a risk-based and animal-sparing approach to evaluate chemical safety, drawing broadly from previous experience but incorporating technological advances to increase efficiency.


Hepatology | 2010

Quantitative analyses and transcriptomic profiling of circulating messenger RNAs as biomarkers of rat liver injury

Barbara A. Wetmore; Dominique Brees; Reetu R. Singh; Paul B. Watkins; Melvin E. Andersen; James Loy; Russell S. Thomas

Serum aminotransferases have been the clinical standard for evaluating liver injury for the past 50‐60 years. These tissue enzymes lack specificity, also tracking injury to other tissues. New technologies assessing tissue‐specific messenger RNA (mRNA) release into blood should provide greater specificity and permit indirect assessment of gene expression status of injured tissue. To evaluate the potential of circulating mRNAs as biomarkers of liver injury, rats were treated either with hepatotoxic doses of D‐(+)‐galactosamine (DGAL) or acetaminophen (APAP) or a myotoxic dose of bupivacaine HCl (BPVC). Plasma, serum, and liver samples were obtained from each rat. Serum alanine aminotransferase and aspartate aminotransferase were increased by all three compounds, whereas circulating liver‐specific mRNAs were only increased by the hepatotoxicants. With APAP, liver‐specific mRNAs were significantly increased in plasma at doses that had no effect on serum aminotransferases or liver histopathology. Characterization of the circulating mRNAs by sucrose density gradient centrifugation revealed that the liver‐specific mRNAs were associated with both necrotic debris and microvesicles. DGAL treatment also induced a shift in the size of plasma microvesicles, consistent with active release of microvesicles following liver injury. Finally, gene expression microarray analysis of the plasma following DGAL and APAP treatment revealed chemical‐specific profiles. Conclusion: The comparative analysis of circulating liver mRNAs with traditional serum transaminases and histopathology indicated that the circulating liver mRNAs were more specific and more sensitive biomarkers of liver injury. Further, the possibility of identifying chemical‐specific transcriptional profiles from circulating mRNAs could open a range of possibilities for identifying the etiology of drug/chemical‐induced liver injury. HEPATOLOGY 2010


Toxicological Sciences | 2013

Relative Impact of Incorporating Pharmacokinetics on Predicting In Vivo Hazard and Mode of Action From High-Throughput In Vitro Toxicity Assays

Barbara A. Wetmore; John F. Wambaugh; Stephen S. Ferguson; Lili Li; Harvey J. Clewell; Richard S. Judson; Kimberly Freeman; Wenjun Bao; Mark A. Sochaski; Tzu-Ming Chu; Michael B. Black; Eric Healy; Brittany Allen; Melvin E. Andersen; Russell D. Wolfinger; Russell S. Thomas

The use of high-throughput in vitro assays has been proposed to play a significant role in the future of toxicity testing. In this study, rat hepatic metabolic clearance and plasma protein binding were measured for 59 ToxCast phase I chemicals. Computational in vitro-to-in vivo extrapolation was used to estimate the daily dose in a rat, called the oral equivalent dose, which would result in steady-state in vivo blood concentrations equivalent to the AC 50 or lowest effective concentration (LEC) across more than 600 ToxCast phase I in vitro assays. Statistical classification analysis was performed using either oral equivalent doses or unadjusted AC 50 /LEC values for the in vitro assays to predict the in vivo effects of the 59 chemicals. Adjusting the in vitro assays for pharmacokinetics did not improve the ability to predict in vivo effects as either a discrete (yes or no) response or a low effect level (LEL) on a continuous dose scale. Interestingly, a comparison of the in vitro assay with the lowest oral equivalent dose with the in vivo endpoint with the lowest LEL suggested that the lowest oral equivalent dose may provide a conservative estimate of the point of departure for a chemical in a dose-response assessment. Furthermore, comparing the oral equivalent doses for the in vitro assays with the in vivo dose range that resulted in adverse effects identified more coincident in vitro assays across chemicals than expected by chance, suggesting that the approach may also be used to identify potential molecular initiating events leading to adversity.


Toxicological Sciences | 2015

Incorporating High-Throughput Exposure Predictions with Dosimetry-Adjusted In Vitro Bioactivity to Inform Chemical Toxicity Testing

Barbara A. Wetmore; John F. Wambaugh; Brittany Allen; Stephen S. Ferguson; Mark A. Sochaski; R. Woodrow Setzer; Keith A. Houck; Cory L. Strope; Katherine E. Cantwell; Richard S. Judson; Edward L. LeCluyse; Harvey J. Clewell; Russell S. Thomas; Melvin E. Andersen

We previously integrated dosimetry and exposure with high-throughput screening (HTS) to enhance the utility of ToxCast HTS data by translating in vitro bioactivity concentrations to oral equivalent doses (OEDs) required to achieve these levels internally. These OEDs were compared against regulatory exposure estimates, providing an activity-to-exposure ratio (AER) useful for a risk-based ranking strategy. As ToxCast efforts expand (ie, Phase II) beyond food-use pesticides toward a wider chemical domain that lacks exposure and toxicity information, prediction tools become increasingly important. In this study, in vitro hepatic clearance and plasma protein binding were measured to estimate OEDs for a subset of Phase II chemicals. OEDs were compared against high-throughput (HT) exposure predictions generated using probabilistic modeling and Bayesian approaches generated by the U.S. Environmental Protection Agency (EPA) ExpoCast program. This approach incorporated chemical-specific use and national production volume data with biomonitoring data to inform the exposure predictions. This HT exposure modeling approach provided predictions for all Phase II chemicals assessed in this study whereas estimates from regulatory sources were available for only 7% of chemicals. Of the 163 chemicals assessed in this study, 3 or 13 chemicals possessed AERs < 1 or < 100, respectively. Diverse bioactivities across a range of assays and concentrations were also noted across the wider chemical space surveyed. The availability of HT exposure estimation and bioactivity screening tools provides an opportunity to incorporate a risk-based strategy for use in testing prioritization.


Journal of Pharmacology and Experimental Therapeutics | 2006

Alterations in the Rat Serum Proteome During Liver Injury from Acetaminophen Exposure

B. Alex Merrick; Maribel Bruno; Jennifer H. Madenspacher; Barbara A. Wetmore; Julie F. Foley; Rembert Pieper; Ming Zhao; Anthony J. Makusky; Andrew M. McGrath; Jeff Zhou; John L. Taylor; Kenneth B. Tomer

Changes in the serum proteome were identified during early, fulminant, and recovery phases of liver injury from acetaminophen in the rat. Male F344 rats received a single, noninjury dose or a high, injury-producing dose of acetaminophen for evaluation at 6 to 120 h. Two-dimensional gel electrophoresis of immunodepleted serum separated approximately 800 stained proteins per sample from which differentially expressed proteins were identified by mass spectrometry. Serum alanine aminotransferase/aspartate aminotransferase levels and histopathology revealed the greatest liver damage at 24 and 48 h after high-dose acetaminophen corresponding to the time of greatest serum protein alterations. After 24 h, 68 serum proteins were significantly altered of which 23 proteins were increased by >5-fold and 20 proteins were newly present compared with controls. Only minimal changes in serum proteins were noted at the low dose without any histopathology. Of the 54 total protein isoforms identified by mass spectrometry, gene ontology processes for 38 unique serum proteins revealed involvement of acute phase response, coagulation, protein degradation, intermediary metabolism, and various carrier proteins. Elevated serum tumor necrosis factor-α from 24 to 48 h suggested a mild inflammatory response accompanied by increased antioxidant capability demonstrated by increased serum catalase activity. Antibody array and enzyme-linked immunosorbent assay analyses also showed elevation in the chemokine monocyte chemoattractant protein-1 and the metalloprotease inhibitor tissue inhibitor of metalloproteinases-1 during this same period of liver injury. This study demonstrates that serum proteome alterations probably reflect both liver damage and a concerted, complex response of the body for organ repair and recovery during acute hepatic injury.


Toxicology | 2015

Quantitative in vitro-to-in vivo extrapolation in a high-throughput environment

Barbara A. Wetmore

High-throughput in vitro toxicity screening provides an efficient way to identify potential biological targets for environmental and industrial chemicals while conserving limited testing resources. However, reliance on the nominal chemical concentrations in these in vitro assays as an indicator of bioactivity may misrepresent potential in vivo effects of these chemicals due to differences in clearance, protein binding, bioavailability, and other pharmacokinetic factors. Development of high-throughput in vitro hepatic clearance and protein binding assays and refinement of quantitative in vitro-to-in vivo extrapolation (QIVIVE) methods have provided key tools to predict xenobiotic steady state pharmacokinetics. Using a process known as reverse dosimetry, knowledge of the chemical steady state behavior can be incorporated with HTS data to determine the external in vivo oral exposure needed to achieve internal blood concentrations equivalent to those eliciting bioactivity in the assays. These daily oral doses, known as oral equivalents, can be compared to chronic human exposure estimates to assess whether in vitro bioactivity would be expected at the dose-equivalent level of human exposure. This review will describe the use of QIVIVE methods in a high-throughput environment and the promise they hold in shaping chemical testing priorities and, potentially, high-throughput risk assessment strategies.


Chemico-Biological Interactions | 2008

Genotoxicity of intermittent co-exposure to benzene and toluene in male CD-1 mice.

Barbara A. Wetmore; Melanie F. Struve; Pu Gao; Sheela Sharma; Neil Allison; Kay C. Roberts; Daniel J. Letinski; Mark J. Nicolich; Michael G. Bird; David C. Dorman

Benzene is an important industrial chemical. At certain levels, benzene has been found to produce aplastic anemia, pancytopenia, myeloblastic anemia and genotoxic effects in humans. Metabolism by cytochrome P450 monooxygenases and myeloperoxidase to hydroquinone, phenol, and other metabolites contributes to benzene toxicity. Other xenobiotic substrates for cytochrome P450 can alter benzene metabolism. At high concentrations, toluene has been shown to inhibit benzene metabolism and benzene-induced toxicities. The present study investigated the genotoxicity of exposure to benzene and toluene at lower and intermittent co-exposures. Mice were exposed via whole-body inhalation for 6h/day for 8 days (over a 15-day time period) to air, 50 ppm benzene, 100 ppm toluene, 50 ppm benzene and 50 ppm toluene, or 50 ppm benzene and 100 ppm toluene. Mice exposed to 50 ppm benzene exhibited an increased frequency (2.4-fold) of micronucleated polychromatic erythrocytes (PCE) and increased levels of urinary metabolites (t,t-muconic acid, hydroquinone, and s-phenylmercapturic acid) vs. air-exposed controls. Benzene co-exposure with 100 ppm toluene resulted in similar urinary metabolite levels but a 3.7-fold increase in frequency of micronucleated PCE. Benzene co-exposure with 50 ppm toluene resulted in a similar elevation of micronuclei frequency as with 100 ppm toluene which did not differ significantly from 50 ppm benzene exposure alone. Both co-exposures - 50 ppm benzene with 50 or 100 ppm toluene - resulted in significantly elevated CYP2E1 activities that did not occur following benzene or toluene exposure alone. Whole blood glutathione (GSH) levels were similarly decreased following exposure to 50 ppm benzene and/or 100 ppm toluene, while co-exposure to 50 ppm benzene and 100 ppm toluene significantly decreased GSSG levels and increased the GSH/GSSG ratio. The higher frequency of micronucleated PCE following benzene and toluene co-exposure when compared with mice exposed to benzene or toluene alone suggests that, at the doses used in this study, toluene can enhance benzene-induced clastogenic or aneugenic bone marrow injury. These findings exemplify the importance of studying the effects of binary chemical interactions in animals exposed to lower exposure concentrations of benzene and toluene on benzene metabolism and clastogenicity. The relevance of these data on interactions for humans exposed at low benzene concentrations can be best assessed only when the mechanism of interaction is understood at a quantitative level and incorporated within a biologically based modeling framework.

Collaboration


Dive into the Barbara A. Wetmore's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John F. Wambaugh

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard S. Judson

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keith A. Houck

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

David J. Dix

United States Environmental Protection Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge