Network


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

Hotspot


Dive into the research topics where Yu-Mei Tan is active.

Publication


Featured researches published by Yu-Mei Tan.


Science of The Total Environment | 2011

A biomonitoring framework to support exposure and risk assessments.

Jon R. Sobus; Yu-Mei Tan; Joachim D. Pleil; Linda Sheldon

BACKGROUND Biomonitoring is used in exposure and risk assessments to reduce uncertainties along the source-to-outcome continuum. Specifically, biomarkers can help identify exposure sources, routes, and distributions, and reflect kinetic and dynamic processes following exposure events. A variety of computational models now utilize biomarkers to better understand exposures at the population, individual, and sub-individual (target) levels. However, guidance is needed to clarify biomonitoring use given available measurements and models. OBJECTIVE This article presents a biomonitoring research framework designed to improve biomarker use and interpretation in support of exposure and risk assessments. DISCUSSION The biomonitoring research framework is based on a modified source-to-outcome continuum. Five tiers of biomonitoring analyses are included in the framework, beginning with simple cross-sectional and longitudinal analyses, and ending with complex analyses using various empirical and mechanistic models. Measurements and model requirements of each tier are given, as well as considerations to enhance analyses. Simple theoretical examples are also given to demonstrate applications of the framework for observational exposure studies. CONCLUSION This biomonitoring framework can be used as a guide for interpreting existing biomarker data, designing new studies to answer specific exposure- and risk-based questions, and integrating knowledge across scientific disciplines to better address human health risks.


Environment International | 2014

A proposal for assessing study quality: Biomonitoring, Environmental Epidemiology, and Short-lived Chemicals (BEES-C) instrument.

Judy S. LaKind; Jon R. Sobus; Michael Goodman; Dana Boyd Barr; Peter Fürst; Richard J. Albertini; Tye E. Arbuckle; Greet Schoeters; Yu-Mei Tan; Justin G. Teeguarden; Rogelio Tornero-Velez; Clifford P. Weisel

The quality of exposure assessment is a major determinant of the overall quality of any environmental epidemiology study. The use of biomonitoring as a tool for assessing exposure to ubiquitous chemicals with short physiologic half-lives began relatively recently. These chemicals present several challenges, including their presence in analytical laboratories and sampling equipment, difficulty in establishing temporal order in cross-sectional studies, short- and long-term variability in exposures and biomarker concentrations, and a paucity of information on the number of measurements required for proper exposure classification. To date, the scientific community has not developed a set of systematic guidelines for designing, implementing and interpreting studies of short-lived chemicals that use biomonitoring as the exposure metric or for evaluating the quality of this type of research for WOE assessments or for peer review of grants or publications. We describe key issues that affect epidemiology studies using biomonitoring data on short-lived chemicals and propose a systematic instrument – the Biomonitoring, Environmental Epidemiology, and Short-lived Chemicals (BEES-C) instrument – for evaluating the quality of research proposals and studies that incorporate biomonitoring data on short-lived chemicals. Quality criteria for three areas considered fundamental to the evaluation of epidemiology studies that include biological measurements of short-lived chemicals are described: 1) biomarker selection and measurement, 2) study design and execution, and 3) general epidemiological study design considerations. We recognize that the development of an evaluative tool such as BEES-C is neither simple nor non-controversial. We hope and anticipate that the instrument will initiate further discussion/debate on this topic.


Toxicological Sciences | 2016

Integration of Life-Stage Physiologically Based Pharmacokinetic Models with Adverse Outcome Pathways and Environmental Exposure Models to Screen for Environmental Hazards.

Hisham A. El-Masri; Nicole Kleinstreuer; Ronald N. Hines; Linda Adams; Tamara Tal; Kristin Isaacs; Barbara A. Wetmore; Yu-Mei Tan

A computational framework was developed to assist in screening and prioritizing chemicals based on their dosimetry, toxicity, and potential exposures. The overall strategy started with contextualizing chemical activity observed in high-throughput toxicity screening (HTS) by mapping these assays to biological events described in Adverse Outcome Pathways (AOPs). Next, in vitro to in vivo (IVIVE) extrapolation was used to convert an in vitro dose to an external exposure level, which was compared with potential exposure levels to derive an AOP-based margins of exposure (MOE). In this study, the framework was applied to estimate MOEs for chemicals that can potentially cause developmental toxicity following a putative AOP for fetal vasculogenesis/angiogenesis. A physiologically based pharmacokinetic (PBPK) model was developed to describe chemical disposition during pregnancy, fetal, neonatal, and infant to adulthood stages. Using this life-stage PBPK model, maternal exposures were estimated that would yield fetal blood levels equivalent to the chemical concentration that altered in vitro activity of selected HTS assays related to the most sensitive vasculogenesis/angiogenesis putative AOP. The resulting maternal exposure estimates were then compared with potential exposure levels using literature data or exposure models to derive AOP-based MOEs.


Toxicological Sciences | 2016

Estimating Margin of Exposure to Thyroid Peroxidase Inhibitors Using High-Throughput in vitro Data, High-Throughput Exposure Modeling, and Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling

Jeremy A. Leonard; Yu-Mei Tan; M.E. Gilbert; Kristin Isaacs; Hisham A. El-Masri

Some pharmaceuticals and environmental chemicals bind the thyroid peroxidase (TPO) enzyme and disrupt thyroid hormone production. The potential for TPO inhibition is a function of both the binding affinity and concentration of the chemical within the thyroid gland. The former can be determined through in vitro assays, and the latter is influenced by pharmacokinetic properties, along with environmental exposure levels. In this study, a physiologically based pharmacokinetic (PBPK) model was integrated with a pharmacodynamic (PD) model to establish internal doses capable of inhibiting TPO in relation to external exposure levels predicted through exposure modeling. The PBPK/PD model was evaluated using published serum or thyroid gland chemical concentrations or circulating thyroxine (T4) and triiodothyronine (T3) hormone levels measured in rats and humans. After evaluation, the model was used to estimate human equivalent intake doses resulting in reduction of T4 and T3 levels by 10% (ED10) for 6 chemicals of varying TPO-inhibiting potencies. These chemicals were methimazole, 6-propylthiouracil, resorcinol, benzophenone-2, 2-mercaptobenzothiazole, and triclosan. Margin of exposure values were estimated for these chemicals using the ED10 and predicted population exposure levels for females of child-bearing age. The modeling approach presented here revealed that examining hazard or exposure alone when prioritizing chemicals for risk assessment may be insufficient, and that consideration of pharmacokinetic properties is warranted. This approach also provides a mechanism for integrating in vitro data, pharmacokinetic properties, and exposure levels predicted through high-throughput means when interpreting adverse outcome pathways based on biological responses.


Regulatory Toxicology and Pharmacology | 2017

Investigating the state of physiologically based kinetic modelling practices and challenges associated with gaining regulatory acceptance of model applications

Alicia Paini; Jeremy A. Leonard; Tomas Kliment; Yu-Mei Tan; Andrew Worth

ABSTRACT Physiologically based kinetic (PBK) models are used widely throughout a number of working sectors, including academia and industry, to provide insight into the dosimetry related to observed adverse health effects in humans and other species. Use of these models has increased over the last several decades, especially in conjunction with emerging alternative methods to animal testing, such as in vitro studies and data‐driven in silico quantitative‐structure‐activity‐relationship (QSAR) predictions. Experimental information derived from these new approach methods can be used as input for model parameters and allows for increased confidence in models for chemicals that did not have in vivo data for model calibration. Despite significant advancements in good modelling practice (GMP) for model development and evaluation, there remains some reluctance among regulatory agencies to use such models during the risk assessment process. Here, the results of a survey disseminated to the modelling community are presented in order to inform the frequency of use and applications of PBK models in science and regulatory submission. Additionally, the survey was designed to identify a network of investigators involved in PBK modelling and knowledgeable of GMP so that they might be contacted in the future for peer review of PBK models, especially in regards to vetting the models to such a degree as to gain a greater acceptance for regulatory purposes. Graphical abstract Figure. No caption available. HighlightsPhysiologically Based kinetic (PBK) models are used widely in academia, industry, and government.Good modelling practice (GMP) for model development and evaluation continues to expand.Further guidance for establishing GMP is called for.There remains some reluctance among regulatory agencies to use PBK models.The next generation of PBK models could be developed using only data from in vitro and in silico methods.


Methods of Molecular Biology | 2012

Informing Mechanistic Toxicology with Computational Molecular Models

Michael R. Goldsmith; Shane D. Peterson; Daniel T. Chang; Thomas R. Transue; Rogelio Tornero-Velez; Yu-Mei Tan; Curtis C. Dary

Computational molecular models of chemicals interacting with biomolecular targets provides toxicologists a valuable, affordable, and sustainable source of in silico molecular level information that augments, enriches, and complements in vitro and in vivo efforts. From a molecular biophysical ansatz, we describe how 3D molecular modeling methods used to numerically evaluate the classical pair-wise potential at the chemical/biological interface can inform mechanism of action and the dose-response paradigm of modern toxicology. With an emphasis on molecular docking, 3D-QSAR and pharmacophore/toxicophore approaches, we demonstrate how these methods can be integrated with chemoinformatic and toxicogenomic efforts into a tiered computational toxicology workflow. We describe generalized protocols in which 3D computational molecular modeling is used to enhance our ability to predict and model the most relevant toxicokinetic, metabolic, and molecular toxicological endpoints, thereby accelerating the computational toxicology-driven basis of modern risk assessment while providing a starting point for rational sustainable molecular design.


Toxicological Sciences | 2018

Challenges Associated With Applying Physiologically Based Pharmacokinetic Modeling for Public Health Decision-Making

Yu-Mei Tan; Rachel Rogers Worley; Jeremy A. Leonard; Jeffrey W. Fisher

The development and application of physiologically based pharmacokinetic (PBPK) models in chemical toxicology have grown steadily since their emergence in the 1980s. However, critical evaluation of PBPK models to support public health decision-making across federal agencies has thus far occurred for only a few environmental chemicals. In order to encourage decision-makers to embrace the critical role of PBPK modeling in risk assessment, several important challenges require immediate attention from the modeling community. The objective of this contemporary review is to highlight 3 of these challenges, including: (1) difficulties in recruiting peer reviewers with appropriate modeling expertise and experience; (2) lack of confidence in PBPK models for which no tissue/plasma concentration data exist for model evaluation; and (3) lack of transferability across modeling platforms. Several recommendations for addressing these 3 issues are provided to initiate dialog among members of the PBPK modeling community, as these issues must be overcome for the field of PBPK modeling to advance and for PBPK models to be more routinely applied in support of public health decision-making.


Biomarkers in Toxicology | 2014

Biomarkers in computational toxicology

Yu-Mei Tan; Daniel T. Chang; Martin B. Phillips; Stephen W. Edwards; Christopher M. Grulke; Michael-Rock Goldsmith; Jon R. Sobus; Rory B. Conolly; Rogelio Tornero-Velez; Curtis C. Dary

Biomarkers are a means to evaluate chemical exposure and/or the subsequent impacts on toxicity pathways that lead to adverse health outcomes. Computational toxicology can integrate biomarker data with knowledge of exposure, chemistry, biology, pharmacokinetics, toxicology, and epidemiology to inform the linkages among exposure, susceptibility, and human health. This chapter provides an overview of four computational modeling approaches and their applications for interpreting biomarker data. Exposure models integrate the microenvironmental concentrations with human activity data to estimate intake doses. Dosimetry models incorporate mechanistic biological information to link intake doses to biomarkers. Biologically plausible models describe normal and xenobiotic-perturbed behaviors that can be distinguished using biomarkers. Cheminformatics-based models provide rapid assessments to inform future biomarker studies. Together, these modeling approaches allow for comprehensive investigations of biomarker data to link between exposures and disease.


Methods of Molecular Biology | 2012

Computational Toxicology: Application in Environmental Chemicals

Yu-Mei Tan; Rory B. Conolly; Daniel T. Chang; Rogelio Tornero-Velez; Michael R. Goldsmith; Shane D. Peterson; Curtis C. Dary

This chapter provides an overview of computational models that describe various aspects of the source-to-health effect continuum. Fate and transport models describe the release, transportation, and transformation of chemicals from sources of emission throughout the general environment. Exposure models integrate the microenvironmental concentrations with the amount of time an individual spends in these microenvironments to estimate the intensity, frequency, and duration of contact with environmental chemicals. Physiologically based pharmacokinetic (PBPK) models incorporate mechanistic biological information to predict chemical-specific absorption, distribution, metabolism, and excretion. Values of parameters in PBPK models can be measured in vitro, in vivo, or estimated using computational molecular modeling. Computational modeling is also used to predict the respiratory tract dosimetry of inhaled gases and particulates [computational fluid dynamics (CFD) models], to describe the normal and xenobiotic-perturbed behaviors of signaling pathways, and to analyze the growth kinetics of preneoplastic lesions and predict tumor incidence (clonal growth models).


Archive | 2018

Tiered Approaches to Incorporate the Adverse Outcome Pathway Framework into Chemical-Specific Risk-Based Decision Making

Jeremy A. Leonard; Shannon Bell; Noffisat Oki; Mark Nelms; Yu-Mei Tan; Stephen W. Edwards

The concept of Adverse Outcome Pathways (AOPs) arose as a means of addressing the challenges associated with establishing relationships between high-throughout (HT) in vitro dose response data and in vivo biological outcomes. However, AOP development has also been met with challenges of its own, such as the time, effort, and expertise necessary to achieve a scientifically sound construct able to support ecotoxicology and human health risk assessment. Thus, a staged development process has been developed to match the information content of an AOP with the decision context in which it will be used. This approach allows effort to be spent on detailed evidence evaluation and quantitative assessment of the dose-response characteristics for those AOPs where this level of confidence and precision is needed. In addition, through advances in computational analytical methodologies that integrate HT data (e.g., transcriptomic data) with traditional toxicology information spanning a broad chemical and biological space, computationally predicted AOPs can be rapidly generated to help accelerate the curation of AOPs. AOPs are chemical agnostic thereby allowing a single AOP to be coupled with in vitro dose-response information from a variety of chemicals. To predict an in vivo outcome, however, exposure and pharmacokinetic characteristics (i.e., absorption, metabolism, distribution, and elimination) must be considered. As with the staged development process for AOPs, it is possible to develop ADME predictions in a tiered manner such that lower tiers provide qualitative or semi-quantitative predictions when data is lacking, and higher tiers provide quantitative predictions with increasing confidence when data is abundant. Tiered approaches to AOP development and ADME predictions provide a mechanism for using AOPs, with chemical-specific exposure and pharmacokinetic considerations, for risk assessment both in data poor and data rich scenarios. They also provide a natural mechanism for identifying areas of research that would have the highest impact on risk-based decision making by highlighting AOPs and/or ADME predictions that are insufficient to address the decision context in which they could be used.

Collaboration


Dive into the Yu-Mei Tan's collaboration.

Top Co-Authors

Avatar

Jeremy A. Leonard

Oak Ridge Institute for Science and Education

View shared research outputs
Top Co-Authors

Avatar

Jon R. Sobus

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Rogelio Tornero-Velez

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Stephen W. Edwards

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Curtis C. Dary

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Daniel T. Chang

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Kristin Isaacs

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Justin G. Teeguarden

Pacific Northwest National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Martin B. Phillips

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar

Rory B. Conolly

United States Environmental Protection Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge