M-LEEAD Logo

Pilot Projects: Funded

More about Pilot Projects

If your pilot project appears below you can update your project here (intranet, for U-M personnel only).

December 2023 (Short Term Seed Projects)

“Assessment of Fluorene-9-bisphenol Toxicity in Mouse Embryoid Bodies Through RNA-Seq”
Primary Investigator: Besa Xhabija
Abstract
The substitution of bisphenol A (BPA) with alternatives like Fluorene-9-bisphenol (BHPF) in consumer products has raised new safety concerns. Previous RT-PCR analyses from our laboratory have confirmed the adverse effects of BHPF on the differentiation of mouse embryoid bodies, disrupting crucial gene expressions integral for cellular development. This project is poised to explore the developmental toxicity of BHPF in depth using embryonic stem cell models. Our approach includes comprehensive RNA-Seq analyses at decisive developmental phases—days 2, 4, 6, and 8—post-exposure to BHPF, mirroring concentrations detected in human blood. This investigation will facilitate a direct comparison between the transcriptomic influences of BHPF and BPA, providing insight into their endocrine-disrupting effects and relative safety as BPA substitutes. The study's findings will be instrumental in addressing the gaps in current knowledge, with the ultimate goal of collecting data for a high-impact publication. Additionally, these insights will serve the broader objective of informing public health policy. Our endeavor will also involve the Community Engagement Core to disseminate our findings effectively, ensuring that communities, particularly those most vulnerable, are educated about the potential risks of environmental chemicals. This commitment to translating research into practice underscores the potential of this project to significantly influence public health initiatives and regulatory frameworks.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Early life heavy metal mixtures and adolescent health in Mexico City”
Primary Investigator: Sara Stein
Abstract
Mental health problems, such as depression, anxiety, and post-traumatic stress disorder, are increasing worldwide and multiple early life risk factors likely co-occur. Heavy metals (e.g., lead, arsenic, cadmium) have been associated with worsening mental health, though far less research has considered mixture exposure in sensitive early life periods in relation to mental health or physiological pathways linking these exposures and outcomes. Emerging literature suggests that heavy metals may dysregulate physiological stress pathways such as the hypothalamic-pituitary-adrenal axis resulting in physiological adaptations, such as increasing oxidative stress and allostatic load. Over time, these adaptations have been linked to worsening mental health. Therefore, heavy metals could influence mental health through dysregulation of physiological pathways as children age, thus increasing risk for mental health problems in young adulthood. Yet, the influence of heavy metal mixtures on physiological stress biomarkers, such as oxidative stress and allostatic load, especially in sensitive stress physiology developmental periods such as adolescence, is less studied. The Early Life Exposures in Mexico to Environmental Toxicants (ELEMENT) cohort is uniquely positioned to address these research gaps, given length of follow up spanning from birth to adolescence and stored biospecimens available for analysis. Among 536 participants followed since birth who are now approaching their 30s, we will leverage archived data from the prenatal period (e.g., heavy metals), and biomarkers of physiological stress, both previously assayed and stored biospecimens, from adolescence. This proposal aims to characterize heavy metal mixture effects on physiological stress biomarkers including 1) oxidative stress and 2) allostatic load. This proposal will provide pilot data for an R21 proposal which will collect mental health data from young adult participants and examine physiological stress biomarkers as mediators between prenatal heavy metal mixtures and young adult PTSD, anxiety, and depression. Mental health problems are increasing worldwide; understanding the joint impact of heavy metals on physiological stress and mental health will provide critical knowledge of those at greatest risk for worse outcomes as well as mechanistic pathways that could be targeted through interventions to prevent the development of mental health problems.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Identifying the Early Life Epigenetic Response to in utero Arsenic Exposure: piRNA Expression and DNA Methylation in Male Mouse Germline Tissues”
Primary Investigator: Mathia Colwell, Dana Dolinoy
Abstract
Inorganic arsenic (iAs) is one of the most common toxic exposures to impact humanity worldwide, with over 140 million people exposed to iAs via contaminated drinking water. Exposure to iAs during pregnancy disrupts normal DNA methylation patterns in developing offspring and leads to the onset of adult diseases, with the potential to be transmitted to multiple generations through germline exposures. While previous reports of prenatal iAs exposure identify effects on DNA methylation, they did not uncover the mechanism leading to this change or characterize the effects on the regulation of DNA methylation in germline tissues. It is unclear if piRNAs, small non-coding RNAs that protect the germline stability, respond to early-life exposures by interacting with the epigenome. While it is biologically plausible that iAs exposure affects piRNA expression in germline tissues, leading to dysregulation of DNA methylation, the characterization between piRNAs and DNA methylation in germline tissues has yet to be uncovered at an early developmental stage in response to a in utero exposure. Our central hypothesis is that in utero iAs exposure increases postnatal germline piRNA transcription, where genic piRNAs are associated with regions of dysregulated DNA methylation, as a mechanism to preserve the epigenome within the germline and germ cells. In this study, we will use testes tissue from 3 week old mice, exposed to a human-relevant doses of iAs (10 and 245 ppb) during fetal development. In Aim 1 we will determine the transcriptional response of piRNAs to in utero iAs exposure on still-developing postnatal germline tissues. In Aim 2, we will compare the dose-specific effects of prenatal iAs exposure on DNA methylation in the testes and identify the correlation between differential DNA methylation and piRNA expression. Collectively, these experiments will provide insight into how early-life exposures impact the interaction between the epigenome and piRNAome in germline tissues focusing on the still-developing gonads and will advance the role of piRNAs in the field of pediatric exposome research.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

May 2023 (Traditional Pilot Projects)

“Epigenetic reprogramming by Pb exposure during mouse in utero development”
Primary Investigator: Bambarendage Perera
Abstract
The objective of this study is to determine epigenetic reprogramming events that impact genomic imprinting upon in utero exposure of mice to human-relevant levels of lead (Pb). Pb is an infamous environmental exposure to human populations in the US and around the world, due in part to its neurotoxic effects. Pb exposure during early development has been linked to adverse health outcomes later in life. Preliminary data indicates that perinatal and in utero Pb exposure alters the DNA methylation (DNAm) of imprinted genes (IGs),and increases placenta size, respectively. However, the molecular mechanisms by which Pb exposure alters genomic imprinting during early development remain largely unknown. IGs are epigenetically regulated in a parent-of-origin specific manner with their mono-allelic expression driving critical periods of development, particularly mid-gestation. Known mechanisms of genomic imprinting include long non-coding RNA (lncRNA),which program allele-specific epigenetic regulation of imprinting control regions. Although dysfunctional genomic imprinting is implicated in several human diseases, the mechanisms leading to toxicant-induced imprinting dysregulation by this mechanism remain poorly understood. Using a mouse model, this study seeks to profile thein utero mRNA, lncRNA, and DNAm changes that impact genomic imprinting due to altered epigenetic reprogramming between the mother and offspring. Thus, female animals will be exposed to a human-relevant dose of Pb via drinking water, two weeks prior to mating through 13-14days post-conception. The proposed pilot study will test the following Aims:1)Determine genomic imprinting mechanisms associated with in utero Pb exposure in the mouse hypothalamus during pregnancy,and2)Assess fetal sex-specific mechanisms of Pb exposure on placental genomic imprinting. Pb-exposed animals will be compared against controls to investigate potential molecular mechanisms by characterizing sex-, tissue-, and developmental stage-specific IG dysregulation. As a result, this study will reveal Pb-associated mechanisms that inform toxic epigenetic reprogramming. The M-LEEaD Center provides the ideal environment to conduct the proposed research in collaboration with Core facilities and a multidisciplinary team of experts outlined herein. The proposed study will address fundamental knowledge gaps of genomic imprinting in the field to inform potential interventions to diseases induced by Pb exposure.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Impact of human milk metal exposure on maternal health and infant growth outcomes”
Primary Investigator: Lindsay Ellsworth
Abstract
Heavy metal exposure poses a significant health concern, particularly in early childhood. Infants are uniquely susceptible to environmental toxicants due to the rapid organ growth and development that occurs in the first months of life. The developmental programming influences of metal intake during this critical period have implications on childhood growth and neurodevelopment as well as long-term metabolic disease risk. Research is largely nonexistent evaluating associations of metals with early offspring growth beyond lead and mercury exposures. Metal exposure through human milk is key in understanding exposure risks as human milk is the recommended primary source of nutrition in infancy; however, significant knowledge gaps remain in the comprehensive evaluation of human milk environmental toxicant burden and associations with maternal and infant health outcomes. We will determine the levels of 10heavymetals in milk samples collected through two longitudinal mother-infant cohorts from different regions. Archived data and biospecimens will be analyzed from the ELEMENT (n=65) and IMAGE (n=35) cohorts. ELEMENT is a longitudinal cohort of dyads from Mexico City with expansive maternal and infant phenotyping with milk samples. The IMAGE mother-infant dyads from southeast Michigan contains detailed maternal metabolic health information with biospecimens and infant growth measures. The ELEMENT and IMAGE cohorts offer a unique opportunity to study two populations with potentially different levels of exposures. Milk samples will be analyzed at two time points including an early time in lactation as milk is transitioning to a mature phase (<1 month post-partum) and a second later timepoint tof mature milk (2 to 4 months post-partum).Milk will be analyzed for lead, arsenic, mercury, cadmium, copper, chromium, selenium, manganese, zinc, and nickel by the NSF International Laboratories in Ann Arbor using microwave digestion and inductively coupled plasma mass spectrometry for analysis. Maternal health factors (age, pre-pregnancy body mass index, smoking status) and pregnancy history will be evaluated to determine factors associated with milk metal levels. Infant growth trajectory based on Z-score for weight, length, head circumference, body mass index and weight-for-length from birth to 6 months will be calculated to determine associations with milk metal levels using multivariate mixed linear modeling.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Integrated Spatial Metals and Transcriptomics Analysis to Understand Triple Negative Breast Cancer Disparities”
Primary Investigator: Justin Colacino
Abstract
The goal of this project is to develop methods for the joint high-resolution spatial analysis of metals concentrations and RNA expression in aggressive triple negative breast cancers to understand how metals alter developmental, inflammatory, and cancer associated processes and promote cancer disparities. There are profound racial disparities in heavy metal exposures in the US. The mechanisms by which heavy metals promote breast cancer formation and progression remain poorly understood in large part due to challenges inaccurate quantification of human metals exposure. For example, there is little known about how metals distribution in breast tissue may impact molecular profiles and function relevant to cancer, such as inflammation and dysregulated stem cell pathways. This line of investigation is critical in designing clinical preventative and treatment measures against aggressive breast cancers in minority communities disproportionately exposed to toxic metals from the environment. This study will leverage an ongoing NIGMSR01, in which we are funded to perform spatial transcriptomic analyses of breast tissues from diverse donors. By layering on spatial metals profiles to RNA measures, we will map how metals distribute in human breast cancers, quantify how these distributions vary by tumor factors, and test how local concentrations of metals in a tissue dysregulate breast cancer associated processes in a cell type specific and region-specific manner. Our overarching hypothesis is that high local concentrations of metals with documented exposure disparities will be associated with dysregulation of breast cancer biological processes including altered “stemness” and inflammatory signaling. Aim 1 will optimize methods for the high-resolution quantification of metals in human breast cancers. Aim 2 will develop methods to integrate spatial metals exposure data with spatial transcriptomics. This project brings together a diverse study team, including scientists new to the field of environmental health, to develop these cutting-edge approaches. If successful, these studies will help to define new biomarkers of risk for breast cancer and define novel pathways by which environmental exposures promote breast cancer disparities. We anticipate this pilot project will provide support for a large multiple PI NIEHS R01 submission In June 2024.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Per- and polyfluoroalkyl substances &amp; T cell epigenetics in lupus”
Primary Investigator: Emily Somers
Abstract
Systemic lupus erythematosus (SLE, or lupus) is a prototypic and serious autoimmune disease of largely unknown etiology that disproportionately affects females and people from certain racial and ethnic groups, including Black, American Indian/Alaska Native, and Arab American persons. Collectively, autoimmune diseases including lupus rank among the leading causes of death for females. There is a critical need to identify environmental exposures that may contribute to lupus and its progression. Per-and polyfluoroalkyl substances (PFAS)are toxicants with extensive distribution and persistent effects in the environment, for which chronic, low-dose exposures are common. Accumulating data support immunotoxic potential of PFAS. This pilot project will characterize PFAS exposure levels in adult females with lupus, utilizing the population-based and extensively-phenotyped Michigan Lupus Epidemiology & Surveillance (MILES)Cohort. A leading paradigm for lupus etiology and potential mechanism by which exposures exert immunotoxic effects involves epigenetic alterations to lymphocyte DNA that increase the propensity for lupus development and flares. This project will examine relationships between PFAS exposures and T cell epigenetics (DNA methylation).We hypothesize that PFAS are associated with alterations in lymphocyte DNA methylation patterns of key genes/pathways involved in immune regulation among persons with lupus. In this pilot project, we will characterize PFAS exposures in lupus and explore associations with DNA methylation in CD4+ T lymphocytes among adult females with SLE from the general population in southeastern Michigan, and compare distributions according to race. We will then examine associations between serum PFAS concentrations and DNA methylation in CD4+ T lymphocytes, using a targeted-approach to interrogate methylation at loci with known impact in lupus, and a discovery epigenome-wide approach to identify additional loci in CD4+ T cell DNA that associate with PFAS concentrations and may impact disease expression. This pilot will generate preliminary data to support anR01-levelproposal for in-depth investigation of the role of PFAS in development and progression of autoimmunity and lupus, within a toxicant-epigenetic framework.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

September 2022 (Traditional Pilot Projects)

“Addressing Inequities in Emergency Response in Environmental Justice Communities in Detroit, MI”
Primary Investigator: Amy Schulz
Abstract
The overarching objective of the proposed Addressing Inequities in Emergency Response in Environmental Justice Communities in Detroit, Michigan, is to create a community and scientifically informed emergency response system that is responsive to the needs of environmental justice communities in Detroit, Michigan. Detroit communities experience high concentrations of hazardous land uses and industrial activities in close proximity to residential neighborhoods, schools and health care facilities. Hazardous land uses are concentrated in areas with large Black and Latinx populations, who live daily with the associated risk of spills, explosions, and hazardous emissions. Community leaders, environmental justice advocates and residents have long expressed concern about inadequate, untimely and unclear emergency notifications and warnings associated with these exposures. The proposed project was collaboratively developed by community leaders and researchers through a series of conversations, precipitated by an emergent situation in southwest Detroit. It seeks to address long-standing community concerns about deficiencies and inequities in emergency response and communications. To reach our overarching objective, we propose the following aims: Aim 1) Develop a draft set of recommendations or best practices for addressing emergency preparedness as an environmental justice issue. These recommendations will draw on: a) existing recommendations on environmental justice and emergency preparedness, b) analysis of public facing information on emergency preparedness and response on local government websites, and c) focused group discussions with leaders from environmental justice communities in Detroit and nearby to identify challenges and strategic opportunities for improvement; Aim 2) Refine and finalize the draft recommendations developed in Aim 1, working with leaders from heavily burdened communities, residents and environmental justice advocates in Detroit. The final recommendations will have as am central goal ensuring responsiveness to environmental justice communities; and Aim 3) Disseminate recommendations to relevant audiences, to include: Detroit Sustainability Office, Detroit Green Task Force, State police, Detroit’s Climate Equity Committee, and state legislators. The resulting recommendations will recognize emergency preparedness and emergency response as an environmental justice issue with existing inequities in the risk of emergencies associated with infrastructure. Actionable recommendations for emergency preparedness will recognize disproportionate risk and associated need for additional/amplified emergency response strategies.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Evaluation of Responses to Chlorinated Hydrocarbons in Primary Placenta Tissue Using Multi-omics Approaches”
Primary Investigator: SEAN HARRIS
Abstract
Adverse pregnancy outcomes such as fetal growth restriction and preterm birth are significant public healthproblems with potential life-long impacts for affected newborns. Exposure to the pollutants trichloroethylene (TCE) and/or perchloroethylene (PERC) are common and associated with elevated risk of fetal growth restriction and preterm birth. As the placenta plays essential roles in maintaining fetal growth and timing of birth, understanding how TCE and PERC impact the placenta is critical to understanding their effects on pregnancy outcomes. Studies in our lab showed that the glutathione-conjugation pathway metabolites of TCE (S-(1,2-dichlorovinyl)-L-cysteine; DCVC) and PERC (S-(1,2-dichlorovinyl)-L-cysteine; TCVC) activate apoptosis and stress response pathways in placental cell lines in vitro. However, the effects of DCVC and TCVC on intact placental tissues have not been studied. Moreover, the key molecular pathways underpinning DCVC/TCVC placental toxicity are unknown. Additionally, responses in the placenta likely differ by fetal sex and it is unknown to what extent fetal sex mediates DCVC or TCVC toxicity. In this study, we will use an in vitro tissue culture model of full-term human placental explants to identify sex-specific transcriptomic and proteomic responses to DCVC and TCVC exposure. Use of placental tissue explants will provide advantages over cell lines because explants more closely replicate tissue in situ, improving biologic interpretability of our findings. We will treat tissue explants derived from freshly obtained male (n=4) and female (n=4) placentae with DCVC or TCVC at two human relevant concentrations (10 or 20μM). Following exposure, we will perform sex- stratified transcriptomic analyses (RNA-sequencing), proteomic analyses (data-independent acquisition mass spectrometry), and integrated multi-omics analyses on the data. As impacts on the proteome will reflect changes in placental phenotype, this study will identify key molecular pathways underpinning DCVC/TCVC placental toxicity. Thus, this study will help to identify fetal sex-specific molecular targets for nutritional or behavioral interventions aimed at reducing the adverse impact of TCE/PERC exposures on the placenta. In addition, this project will generate exciting new preliminary data for an early-stage investigator (Dr. Harris) which will be used to support future NIH grant proposals aimed at understanding environmental pathways to adverse pregnancy outcomes.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Exposure to metals, longitudinal cognitive decline, and Alzheimer&acirc;€™s Disease biomarkers from the mid-to-late life: the Study of Women&acirc;€™s Health Across the Nation”
Primary Investigator: Xin Wang
Abstract
The rising prevalence of Alzheimer's Disease and Related Dementias (ADRD) is a major public health and clinical challenge. ADRD is currently incurable, and identification of modifiable risk factors of ADRD in midlife many decades before clinical detection, is an effective way to prevent later-life onset. The disappointing result of ADRD prevention efforts in late life also suggest that there is a promising opportunity to target at-risk individuals in midlife to slow the aging process before debilitating brain decline has been cemented. Environmental pollutants such as metals are of particular interest due to their known neurotoxic effects. Exposure to metals has been linked to neurodevelopmental disorders in children; however, studies examining metals and cognitive decline and ADRD from mid-to-late life, are extremely limited. Furthermore, the mechanisms underlying the associations between metal exposures and cognitive decline/ADRD remain poorly understood. The goal of the proposed study is to utilize repository urine and blood samples and existing data from the Study of Women’s Health Across the Nation (SWAN) - an ongoing, multi-racial/ethnic, community- based, prospective cohort study of women from mid-to-late life – in order to examine the associations between metals (including lead, cadmium, arsenic, mercury, selenium, and manganese) and cognitive decline trajectories from 2003 to 2017. In Aim 1 we will examine the relationships of metals and metal mixtures with longitudinal trajectories of cognitive function. In Aim 2, we will explore the associations of metals and metal mixtures with ADRD biomarkers (including Aβ42, Aβ40, p-tau-231, and total tau) and assess if ADRD biomarkers mediate associations between metals and cognitive decline trajectories. The expected outcomes of this project are to identify metal exposures as modifiable risk factors for cognitive decline and ADRD; to elucidate the pathophysiology of cognitive decline and ADRD; and to explore midlife as a critical life stage for ADRD prevention. The proposed study will provide initial support for a future large-scale epidemiologic study of the impact of exposure to metals on cognitive decline and ADRD incidence. Ultimately, these findings will inform future clinical (e.g., metals surveillance in adults, nutrition) and policy (e.g., safe homes, occupational protections) interventions to prevent cognitive decline and ADRD.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Sex-Specific Effects of Developmental Lead Exposure on the Kmt2d Complex and Cardiac Function in Adulthood”
Primary Investigator: Laurie Svoboda
Abstract
Sex and age play critical roles in the etiology of cardiovascular diseases (CVDs), but how they interact with developmental environment to influence cardiovascular health is poorly understood. The heavy metal lead (Pb) is a widespread environmental contaminant linked to several CVDs, but the effects of developmental Pb exposure on sex-specific CVD risk later in life are unknown. Our preliminary data demonstrate that Pb alters action potential kinetics in human cardiomyocytes, and leads to oxidative stress in weanling mouse hearts. Pb may interfere with normal cardiovascular development and function through effects on the epigenome. Work from our lab has demonstrated that developmental Pb exposure alters cardiac DNA methylation in a sex-specific manner, but effects on other epigenetic mechanisms have not been investigated. The Kmt2d/Kdm6a complex plays a critical role in normal cardiac development through regulation of histone modifications, and its function is altered in the context of several cardiovascular diseases. Our preliminary data demonstrate that both developmental Pb exposure and age alter transcript expression of this complex in a sex-specific manner, but the implications this has for disease are unknown. Using an established mouse model of developmental exposures, we will investigate the sex-specific effects of Pb exposure during gestation and lactation on cardiac function in adulthood, long after cessation of exposure. We hypothesize that developmental exposure to Pb will have sex- specific, adverse effects on cardiac function in adulthood that are mediated, at least in part, by dysregulation of the Kmt2d/Kdm6a complex. We will use non-invasive tail cuff, echocardiography and histology to measure cardiac structure and function, as well as ChIP-seq, qRT-PCR and western blot to assess the effects of Pb exposure on the function of the Kmt2d/Kdm6a complex and its downstream targets. How environmental exposures interact with age and sex to influence the risk of CVD is an area of public health that has received little attention. Data generated under this proposal will be utilized to develop a collaborative R01 application expanding upon these findings to investigate how Pb interacts with aging and other stressors to impact the risk of CVDs across the life course.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

Traditional Pilots, Summer 2020

“Cardiovascular health effects from exposure to electronic cigarette aerosol”
Primary Investigator: Abhijit Ghosh
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Community Voice in Environmental Health Priorities”
Primary Investigator: Susan Goold
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Environmental Cadmium and Influenza-related Mortality in NHANES: An Environment-Infectious Disease Interaction Study with Implications for Strategies for Reducing COVID-19-related Morbidity and Mortality”
Primary Investigator: Sung Park
Abstract
We propose research that has the potential to lead to a low-cost, off-the-shelf treatment strategy for significantly reducing morbidity and mortality associated with COVID-19-associated pulmonary disease. Previous research has shown that environmental cadmium exposure is widespread in the U.S. (from exposure to cigarette smoke as well as dietary sources). Cadmium is poorly excreted, accumulates in the body, triggers pulmonary inflammation, is associated with decrements in respiratory function, and has been shown to greatly enhance lung injury by respiratory syncytial virus. In this pilot project, we propose to conduct a rigorous analysis of existing data from the National Health and Nutrition Examination Survey (NHANES) to test the hypothesis that urinary and/or blood levels of cadmium - an excellent biomarker of environmental cadmium exposure - predicts higher mortality from influenza and/or pneumonia. Our team already has over 20 years of experience in environmental epidemiology research on metals, including research on cadmium using NHANES data. Pilot support would enable us to examine and appropriately extract the right data sets for this project and conduct a rigorous and deep analysis testing the hypothesis while controlling for a number of potential confounders and examining the data for potential effect modification by age, sex, smoking status, and diets that boost levels of glutathione. Results (which we believe we can generate quickly) favoring the hypothesis would provide strong evidence supporting follow-up research. This could potentially include the conduct of randomized trials in COVID-19 patients of n-acetyl cysteine, a well-known low-cost, off-the-shelf dietary supplement that is a precursor of reduced glutathione and that has been shown to reduce the lung&apos;s inflammatory response to cadmium, as reflected by cytokine expression.
Accomplishments
Two news articles 1) "New study links cadmium to more severe flu, pneumonia infections" https://news.umich.edu/new-study-links-cadmium-to-more-severe-flu-pneumonia-infections/ 2) "Chemical found in cigarettes could increase severity of COVID-19, new study finds" https://www.news-medical.net/news/20201216/Chemical-found-in-cigarettes-could-increase-severity-of-COVID-19-new-study-finds.aspx
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Exploring associations between gestational phthalate exposure, cord blood metabolome, and early measures of neurodevelopment”
Primary Investigator: Deborah Watkins
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Exploring the roles of manganese on neurodevelopment”
Primary Investigator: Young Ah Seo
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Exposome target prediction with coupled matrix-matrix completion”
Primary Investigator: Maureen Sartor
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Extracellular Vesicles as Biomarkers and Mediators of Toxicant Impacts on the Placenta”
Primary Investigator: Kelly Bakulski
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

November 2019 (Short Term Seed Projects)

“Effect of developmental phthalate exposure on metabolic and epigenetic programming in the heart ”
Primary Investigator: Laurie Kathleen Svoboda
Abstract
Phthalate exposures clearly play a role in the pathogenesis of cardiovascular diseases, which are a significant cause of morbidity and mortality around the world. However, surprisingly little is known about the mechanisms by which phthalates induce cardiovascular dysfunction, particularly those exposures that occur during early development. This proposal will address several critical unanswered questions regarding the effects of developmental phthalate exposure on human cardiac health: Does developmental DEHP exposure interfere with the normal programming of DNA methylation during development? Are there sex differences in the effect of DEHP on DNA methylation in offspring hearts? Are these changes mediated by alterations in the metabolites that govern cellular differentiation? The work outlined in this proposal is therefore highly relevant to the mission of the M-LEEaD center, to understand how environmental exposures contribute to the etiology of chronic diseases.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Elucidating mechanisms of cellular cadmium toxicity”
Primary Investigator: David Benner Lombard
Abstract
Cadmium (Cd) is a toxic heavy metal, and represents a major pollutant world-wide. Cd exerts bone, renal, reproductive, neurodevelopmental, and pulmonary toxicities, and increases cancer risk. No mechanism-based treatments exist for Cd toxicity, representing an important unmet need in toxicology and public health. Cd binds to chemical moieties on proteins to interfere with their function, injuring cells via pleiotropic mechanisms. A large literature documents the ability of Cd exposure to alter DNA methylation. A few recent reports have linked Cd exposure to altered levels of histone modifications, including reduced acetylation. Via small molecule screening, we identified an ability of histone deacetylase inhibitors (HDACi) and Bromodomain and Extra-Terminal motif inhibitors (BETi) to suppress cell death occurring in response to Cd treatment. Both classes of agents target histone acetylation, suggesting that this post-translational modification represents a functionally important target of Cd in cells. Reduced mitochondrial respiration and altered cellular metabolite profiles induced by Cd are also rescued by these classes of drugs. These findings suggest that cellular Cd toxicity involves an interplay between reduced histone acetylation and mitochondrial function. Some of the genes most affected by Cd treatment, and reduced histone acetylation, may be involved in mitochondrial function. Conversely, mitochondria are a driver of histone acetylation. Thus, impaired mitochondrial function in response to Cd treatment may induce reduced histone acetylation, in turn worsening mitochondrial function, in a vicious cycle. We will test this model, by identifying the important gene and histone targets of Cd-driven epigenetic dysfunction in cells. These studies will provide a strong foundation for an R01 application to NIEHS, and provide new mechanistic insight into the basis for Cd cellular toxicity.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Geospatial Analysis of California Wildland Fire Smoke Exposures during Pregnancy”
Primary Investigator: Patricia D Koman
Abstract
Wildfires are a growing source of fine particle air pollution, with over a third of U.S. homes adjacent to wildfire-prone areas. Fine particles in the air may increase adverse birth outcomes such as pre-term birth. However, there little information to guide public health officials and clinicians about risks to pregnant women from rapidly increasing wildfire pollution. Our long-term research aim is to quantify the extent to which air pollution, particularly from wildfires, increases hypertensive disorders of pregnancy, which will improve our understanding of the contribution of environmental exposures toward the etiology of chronic diseases like cardiovascular disease. We will ultimately link health data from about 6 million births across the entire state of California (2008-2018) to maps of air pollution from computer models distinguishing fine particles from wildfires from this proposal. Our immediate next step is to utilize MLEED IHSC core services and update to current years of modeled air quality (2014-18) and to obtain air quality monitored data from routine compliance networks and special studies of wildfire to improve model performance. To obtain fire-specific estimates to prepare for proposals for epidemiologic studies, we will process the updated CMAQ-derived PM2.5 concentrations to create analysis-ready exposure metrics, including identification of smoke-wave-day indicator, wildfire smoke intensity levels, and total non-fire pollutant concentration. We will also obtain air quality monitoring measurements from special studies and routine in situ surveillance networks to assess and improve model performance. Previous validation studies of CMAQ model performance have assessed PM2.5 prediction in populated California urban area. We propose to use similar techniques. Outdoor air pollution is a leading cause of death and disease, accounting for 3 million premature deaths per year globally, yet relatively little is known about the specific source of wildfire. By studying pregnancy, researchers may observe the development of diseases in a short time frame during sensitive life stages that have a major impact on children and their mothers; pregnancy outcomes are also related to developing disease decades later. We can learn how air pollution affects health and ultimately prevent adverse outcomes through prevention and public health policy.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Impact of prenatal exposure to native and environmental steroids on adipose tissue non-coding RNA expression”
Primary Investigator: Vasantha Padmanabhan
Abstract
Developmental exposure to endocrine disruptor bisphenol A (BPA) is associated with metabolic defects during adulthood. In sheep, prenatal BPA treatment induced metabolic defects include insulin resistance and adipocyte defects during early adulthood. Although BPA is a known obesogen that influences adipocyte differentiation, the mechanism through which prenatal BPA exposure influences adipocyte defects are not known. One possible mechanism through which BPA could cause adipose disruption is through epigenetic changes mediated altered gene transcription. A major epigenetic mechanism induced by steroidal endocrine disruptors like BPA is changes in expression of small non-coding RNA like microRNAs which modulate the stability and the translation of mRNA. Therefore, this proposal tests the hypothesis that prenatal BPA induces depot-specific differential expression of miRNAs that specifically target genes that regulate adipogenesis, adipocyte differentiation and function in the female sheep. To test this the robustness of RNAseq technology is utilized. The results of this study will shed light on the changes brought about by early BPA exposure in the development of adipose tissue defects associated with chronic metabolic diseases and provide opportunities to develop interventional strategies. These aims are inline with the core mission of M-LEEaD of understanding the contribution of environmental exposures in development of chronic diseases including metabolic disorders.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Using transcriptomics to understand how trichloroethylene exposure contributes to adverse birth outcomes ”
Primary Investigator: Rita Loch-Caruso
Abstract
The contribution of environmental contaminant exposure to adverse pregnancy outcomes is a significant public health concern. Trichloroethylene (TCE) is a common contaminant found in the majority of U.S. Superfund sites. Although TCE exposure during pregnancy is associated with increased risk of adverse pregnancy outcomes, significant gaps exist in our understanding of biological mechanisms underlying TCE&acirc;??s adverse effects on pregnancy. Disrupted placental function and infection of gestational tissues are known risks to pregnancy. Prior work in our laboratory using in vitro approaches showed that a TCE metabolite, dichlorovinyl cysteine (DCVC), activates cellular redox, cytokine, prostaglandin, and apoptosis responses in a human placental cell line, and strongly suppresses pathogen-stimulated innate immune response in extraplacental membranes. Although placental macrophages provide early defense against infection of the gestational compartment, no reports to date have assessed DCVC suppression of macrophage immune response to pathogens. The proposed work would use transcriptomics to provide an unbiased in-depth characterization of TCE-stimulated responses in placenta and macrophages. Using next generation sequencing (RNAseq) of stored samples, we will identify genes differentially expressed in placenta from rats with and without TCE exposure in pregnancy, and in macrophages with and without DCVC exposure in vitro. Analysis will also be performed to identify treatment-related enriched pathways. To accomplish our objectives, we will work with the M-LEEaD Omics and Bioinformatics Core (OBIC) to access services of the University of Michigan DNA Sequencing Core for the RNAseq analysis, and we will augment data analysis with statistical support through the M-LEEaD Integrated Health Sciences Core as needed. We anticipate that the RNAseq approach will allow us to identify new targets and pathways impacted by TCE, as well as new insights into how TCE exposure contributes to the complex pathway interactions that underpin adverse birth outcomes. Findings from these experiments will provide new information to public health officials and clinicians for improved communication and regulation regarding health risks from exposure to this compound during pregnancy.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

May 2019 (Traditional Pilot Projects)

“Analyzing community response and environmental health literacy in the context of the 1973 PBB Contamination in Michigan”
Primary Investigator: Amy J Schulz
Abstract
The Michigan Chemical Company (later named Velsicol) closed its doors in 1978 and left behind a toxic legacy of three superfund sites and a residential neighborhood that are still contaminated 40 years later. In 1973, the chemical company was also responsible for the largest agricultural disaster in U.S. history. They shipped brominated flame-retardants (PBB) instead of a nutritional supplement to be mixed into livestock feed. Millions of Michigan residents consumed farm products containing PBB. <br /><br /> The proposed pilot project, Analyzing community response and environmental health literacy in the context of the 1973 PBB contamination in Michigan, involves a collaboration between researchers involved with the Environmental Health Science Core Centers (EHSCC’s) at the University of Michigan (M-LEEaD) and Emory (HERCULES), and at Central Michigan University (CMU) in response to the priorities identified by Michigan residents seeking to document and understand this toxic legacy. This proposed pilot project will provide preliminary data for a larger grant proposal designed to substantially extend our understanding of environmental health literacy (EHL), a relatively new field with critical implications for the work of the EHSCC’s and their Community Engagement Cores (CECs), responsible for contributing to our knowledge of environmental health communication. It builds on an oral history project developed by an oral historian at Central Michigan University with community feedback, and preliminary analyses conducted by researchers at M-LEEaD, HERCULES and CMU to begin to identify themes and code categories relevant to understanding EHL in five oral history transcripts. In this pilot project proposal, we intend to: <br /><br /> Aim 1: Analyze processes through which individuals affected by the PBB contamination developed knowledge and took action to protect and promote health, through analysis of 15-20 oral histories;<br /> Aim 2: Consider implications of findings for translation of information about large scale chemical contaminations with affected communities and elected and administrative decision makers through a scientific convening;<br /> Aim 3: Convene members of the affected communities and discuss opportunities for translation and action (e.g., creation of registries of affected individuals to facilitate dissemination of scientific findings, especially health effects for the affected population).
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Duplex sequencing for ultra-sensitive detection of environmentally-induced genome variants”
Primary Investigator: Thomas E Wilson
Abstract
Our genome is a major target through which environmental exposures can impact human health. Toxicants and other factors can influence the genome functionally, via epigenetics, and structurally, via time-honored mutational mechanisms. Indeed, recent investigations have revealed that our genomes are never genetically stable, but accrue substantial new mutations throughout life, every time any cell divides. Our new appreciation for this extent of gonadal and somatic mosaicism has exposed two major areas of interest. First, there is a need to explore how organismal, cellular and genomic contexts influence mutation acquisition in studies that are powered with sufficient cellular and genetic resolution. Second, somatic mutations represent a massive sphere of potential influence of genotoxicants that could impact human development and health more than we have realized (especially in concert with epigenetic mechanisms), but sensitive methods are once again required to understand these effects.<br/><br/> We will address these needs by developing one new DNA sequencing technology and making it routinely available at Michigan. Duplex sequencing is a logical approach to next generation sequencing in which high accuracy and exceptional sensitivity are achieved by demanding that both input DNA strands are analyzed and agree on a variant call. We will apply duplex sequencing to structural variant (SV) detection, a novel application for the approach, using control materials that are uniquely available to our research groups.<br/><br/> The technical goals of this project are only one part of a larger effort developing at Michigan to study somatic mosaicism. One of many ways this pilot will leverage M-LEEaD funding is by coordinating with other support to develop parallel and complementary technologies for mutation monitoring. The long term goal of our growing team is to apply a suite of technologies in research programs whose projects will address biological systems that build on specific expertise at Michigan, and ultimately the impact of environmental factors on mutation acquisition in those systems.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Effects of methyl paraben exposure on adipose inflammation, stem cells, and breast cancer metastasis”
Primary Investigator: Justin Adam Colacino
Abstract
According to the American Cancer Society, there will be an estimated 42,270 breast cancer associated deaths in 2019, which are typically due to metastasis. Relative to breast cancer incidence, comparatively little is known about environmental risk factors for metastasis. Obesity is a major risk factor for both breast cancer incidence and mortality. A potential mechanism linking obesity and breast cancer risk is increased adipose tissue inflammation and inflammatory cytokine secretion. Our lab has shown that inflammatory factors secreted from obese adipose tissue modulates breast epithelial stem cell self-renewal in vitro, a process thought to be important early in carcinogenesis. Parabens, a class of endocrine disrupting chemicals (EDCs) commonly used as preservatives in cosmetics, induce adipogenesis in vitro and are under investigation as potential breast carcinogens. Methylparaben (MPB) is ubiquitously detectable in human breast tissue, breast milk, and urine samples. MPB exposure also increases body weight, total white adipose tissue weight (WAT), leptin mRNA expression, and increases glandular tissue and mammary tissue gene expression of genes related to DNA repair and cell cycle regulation in rodent models. While MPB exposure can induce an obesity phenotype and alter mammary gland development, its effects on breast cancer initiation and progression remain to be elucidated. Here, we will test the hypothesis that MPB exposure induces adipose inflammation and accelerates mammary tumor formation and dissemination in MMTV-Her2 transgenic mice, which are genetically susceptible to breast cancer, through alterations of stem cell biology. We will compare the effects of MPB to high fat diet (HFD) to assess similarities and differences in the effects of these exposures. The results of this study will provide novel insights into the mechanisms by which environmental factors promote breast cancer metastasis, and will be key preliminary data to support an NIEHS R01 application to assess environmental impacts on adipose, breast stem cells, and breast cancer metastases.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Migrant and Seasonal Farmworkers Health and Labor: Building a Foundation for Community-Based Participatory Research in Michigan”
Primary Investigator: Alexis J Handal
Abstract
The overarching goal of the project is to conduct a community engaged qualitative pilot study to bridge our understanding between environmental and occupational health exposures and relate this understanding to broader social determinants such as labor exploitation and forced labor in migrant and seasonal farmworkers in Michigan. This innovative research is critical because labor exploitation and forced labor are intrinsically related to the physical (e.g., heat) and toxicant (e.g., pesticide)exposures that workers may experience in the environment and work place. Labor violation standards can contribute to undermining lawful labor conditions and enable more extreme forms of exploitation, hence contributing to the existence of forced labor and its impacts on health. We have initiated collaborations with well-established community organizations that work directly with migrant and seasonal farmworkers in the state: Farmworker Legal Services of Michigan; the Michigan Immigrant Rights Center; the Community Engagement Liaison for Hispanic/Latinx Community for the Michigan Department of Civil Rights; and the Director of Office of Migrant Affairs and Chair of the Michigan Interagency Migrant Services Committee. We will conduct five focus group discussions with female and male migrant and seasonal farmworkers who have been employed in agriculture within the last 24 months. We will also conduct in-depth interviews (~15) with key stakeholders that actively participate in the Michigan Interagency Migrant Services Committee which includes non-profit, public and private agencies, growers, farmworkers and concerned citizens. The results of the study will provide information on actions that may hinder or facilitate service provision and environmental health interventions for migrant and seasonal farmworkers in the State of Michigan. The impact of this pilot study will be to lay groundwork for developing collaborations and using the results to inform the development of a larger mixed-methods environmental health research study with a community-based participatory framework. Our long-term goal will be to use this research to inform programs and policies to improve the environment and public health and better address the needs of migrant and seasonal farmworkers in the state of Michigan, who play an essential role in providing healthy and fresh produce for consumers.
Accomplishments
The Michigan Farmworker Project Website: https://sph.umich.edu/covid/epidemiology/michigan-farmworker.html "Essential but Expendable? Protecting Farm Workers During COVID-19" a Q&A with Alexis Handal and Lisbeth Iglesias-Rios: https://sph.umich.edu/news/2020posts/protecting-farm-workers-from-covid-19.html
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Phthalate exposure and epigenetic programming, differentiation and function of human cardiac myocytes”
Primary Investigator: Laurie Svoboda
Abstract
Cardiovascular diseases pose a significant public health burden worldwide, and it is increasingly clear that environmental exposures play an important role in their pathogenesis. In particular, exposures during critical windows of development, including pregnancy and lactation, may lead to an increased risk of cardiovascular diseases later in life. Although the mechanisms underlying this environment-induced programming of disease risk are not fully understood, rewiring of the normal epigenetic processes underlying development likely plays a role. Normal development is characterized by profound, dynamic programming of DNA methylation, an epigenetic mark that is vital for establishment of tissue-specific gene expression patterns and maintenance of genome stability. Proper establishment of DNA methylation patterning is critical for normal cardiovascular development, and this modification is altered in the context of diseases and environmental exposures. Phthalates are chemicals used in a variety of applications, including food packaging, cosmetics, and medical devices, resulting in ubiquitous human exposure. Importantly, pre- and postnatal phthalate exposures have been linked to a number of adverse cardiovascular outcomes, including cardiac arrhythmias, hypertension, and coronary heart disease. In spite of this, the molecular mechanisms underlying the deleterious effects of phthalates on cardiovascular health are poorly understood. In particular, the effects of phthalate exposures on the human cardiac epigenome, and the implications they may have for cardiac function and disease, have not been investigated. Using a novel, human induced pluripotent stem cell model, the purpose of this proposal is to test the hypothesis that exposure to phthalates interferes with cardiac differentiation by altering normal DNA methylation patterning. We will further test the hypothesis that these effects on differentiation are linked to impaired cardiac myocyte function. These studies will identify potential molecular links between early phthalate exposures and adverse cardiovascular outcomes. Data generated as part of this proposal will open the door to future funding applications aimed at understanding the effects of phthalate exposures on human cardiovascular development, with the aim of identifying novel opportunities for therapeutic and behavioral interventions.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“The cumulative health burdens of air pollution and heat in a changing climate under different local energy mix scenarios”
Primary Investigator: Carina Gronlund
Abstract
In SE Michigan, air pollution from power plants, particularly coal power plants, causes substantial adverse cardiovascular and respiratory health effects with concomitant monetary health costs. Furthermore, the health benefits and costs are likely not distributed equally across the energy continuum--from energy production to use--such that higher costs from air pollution relative to benefits, such as protection from extreme heat, may be born by people of color, of low socioeconomic status, and of foreign birth. In a community-academic collaboration, we propose first to perform a health impact assessment to evaluate the health costs averted in a scenario for decommissioning coal plants to be proposed by DTE Energy alongside alternative energy mix scenarios. We will then estimate the spatial variation in health impacts from air pollution from electricity generation from all sources in SE Michigan--coal and natural gas plants--and overlay these spatially varying estimates with spatially varying estimates of extreme heat health effects. Consolidating the extreme heat and air pollution health effects may reveal greater disparities than the effects of each individually. Air pollution exposure estimates will be based on a combination of existing estimates and derived using the AERMOD dispersion model. Health and monetary impacts will be estimated using BenMAP. Extreme heat health effects will be estimated for present and future climates using spatially varying outdoor extreme heat estimates with an existing model predicting central air conditioning at the parcel level, which reduces indoor extreme heat exposure. Heat-associated mortality, hospitalization, emergency department, and preterm birth estimates will be derived using existing concentration-response associations from the literature and the present and future exposure estimates. Finally, we will present findings to several stakeholder groups attended regularly by the investigators, and we will develop recommendations to be published with the research findings in a report to be posted on the M-LEEaD website. The proposed project was developed in response to priorities identified by a community partner member of the M-LEEaD Stakeholder Advisory Board, and engages M-LEEaD Center researchers in scientific research that addresses those priorities, in keeping with M-LEEaD Center and CEC goals and objectives.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

January 2019 (Rapid Response Projects)

“Characterization of human saliva cell heterogeneity by single cell RNA-sequencing”
Primary Investigator: Kelly Bakulski
Abstract
Significance. Saliva is a commonly used, non-invasive source of biological samples for environmental epidemiology and clinical research. It contains cells from the cheek, tongue, palate, mucosal glands, and immune system with a high-level of inter-individual and across time variability. Some cell types in saliva (epithelial cells and leukocytes) have been characterized through flow cytometry, though the range of potential cell types has not yet been profiled (e.g. keratinocytes, acinar cells, fibroblasts). Environmental exposures are commonly associated with altered gene expression in bulk tissue samples. Cellular differentiation is regulated by transcriptional signals. Apparent associations between environmental exposures and gene expression, however, can be confounded by altered cell type proportions. To adjust for the range of cell populations in saliva in environmental epidemiology studies, we must first understand the possible cell types that are found in saliva. In this study, we propose to screen a pooled saliva sample with single cell RNA sequencing to discover saliva cell types with unique gene expression profiles. &bull; Approach. We will collect and pool fresh saliva from 10 healthy convenience participants. We will submit 5,000 saliva cells for barcoding and cDNA library construction on the Chromium (10X Genomics) and sequencing on the NovaSeq (Illumina) at the University of Michigan DNA Sequencing Core. Single cell RNA-sequencing reads will be processed, deconvoluted, and aligned with Cell Ranger and filtered with Seurat. We will use principal components analysis to cluster similar cells. We will identify genes that most distinguish each of the cell clusters and validate with immunofluorescence. We will consult with the M-LEEaD Omics and BioInformatics Core for data processing. &bull; Outcome. We will produce the first unbiased map of saliva gene expression on the single cell level. Understanding cell types and their proportions in saliva will provide essential information for environmental epidemiology studies. We expect to observe differences in immune-epithelial cell proportions in individuals exposed to toxicants. Our results will specifically apply to the Fragile Families and Child Wellbeing Study (FFCWS) and Center for Oral Health Research in Appalachia (COHRA) childhood cohorts with well characterized saliva biospecimens and linked environmental exposures/health outcomes data.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Developmental impact of prenatal BPA on adipose tissue transcriptome”
Primary Investigator: Vasantha Padmanabhan
Abstract
Developmental exposure to endocrine disruptor bisphenol A (BPA) is associated with metabolic defects during adulthood. In sheep, prenatal BPA treatment causes metabolic defects such as insulin resistance and adipocyte hypertrophy during early adulthood. Although BPA is a known obesogen that influences adipocyte differentiation, the mechanism through which prenatal BPA exposure influences adipocyte defects are not known. The process of adipogenesis, adipose tissue proliferation, differentiation and function are intricately regulated by transcriptional changes. As gestational exposure to endocrine disruptors can program disruptions in transcriptome leading to dysfunction and pathology, this proposal aims to utilize the robustness of RNA sequencing technology to elucidate and determine the mechanistic pathways involved in gestational BPA- induced adipocyte defects. The results of this study will shed light on the changes brought about by early BPA exposure in the development of adipose tissue defects associated with chronic metabolic diseases and provide opportunities to develop interventional strategies.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Effects of gestational and perinatal phthalate exposure on the male and female cardiac transcriptome”
Primary Investigator: Laurie Svoboda
Abstract
Cardiovascular diseases are a major cause of morbidity and mortality world-wide, and accumulating evidence suggests an important role for early-life exposures to toxicants, including phthalates, in their etiology. As ubiquitous exposure to phthalates remains a public health concern in both industrialized and developing nations, a better understanding of how they contribute to cardiovascular diseases is urgently needed. Although early and later-life phthalate exposures have been linked to adverse cardiovascular outcomes in human and animal models, the mechanisms underlying these effects are currently unclear. In particular, how phthalate exposures during pregnancy may interfere with normal developmental programming of the cardiovascular system, and the implications this may have for long-term disease risk, is unknown. Notably, recent studies suggest that the effects of phthalates, like other endocrine disrupting chemicals, are sex-specific. However, the sex-specific effects of developmental phthalate exposures on the heart and long-term cardiovascular health have not been investigated. Using heart tissue generated from a well-established mouse model of developmental toxicant exposures, the purpose of this proposal is to test the hypothesis that exposure to the commonly used phthalate, di-2- ethylhexyl phthalate (DEHP) in utero and during lactation leads to sex-specific
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Evaluation of transcriptomic response in first-trimester human placental villous explants exposed in vitro to the trichloroethylene metabolite S-(1, 2-dichlorovinyl)-L-cysteine (DCVC) ”
Primary Investigator: Rita Loch-Caruso
Abstract
The contribution of environmental toxicant exposure to pregnancy outcomes such as low birth weight, preterm birth and preeclampsia is a significant public health concern. Trichloroethylene, a toxicant commonly found in the environment, has been linked to increased risk of one or more of these outcomes in epidemiology studies. However, the specific mechanisms by which this compound contributes to adverse birth outcomes is still largely unclear. Transcriptomics is a powerful approach which offers the ability to analyze biological tissues for changes in global gene expression. For these studies, we will utilize first-trimester human placental villous explant cultures exposed to the TCE metabolite DCVC for 12 h. The proposed study will utilize transcriptomics in order to elucidate mechanisms of TCE toxicity in the placenta and to validate findings in a placental cell line. The results of these studies will provide important new insight into the mechanisms by which TCE contribute to adverse pregnancy outcomes.
Accomplishments
The experimental phase of this project has been completed. First-trimester placental chorionic villi explants were exposed to media alone (control) or 20 µM DCVC for 12 h. RNA for each sample was isolated for next-generation RNA sequencing (RNAseq). Sequencing was performed by the University of Michigan DNA Sequencing Core through the M-LEEaD Omics & Bioinformatics Core. The data analysis phase of this project is also complete. Both differential gene expression and gene set enrichment analyses were conducted. The results show that 20 µM DCVC exposure for 12 h caused significant up-regulation of TNFα signaling via the NFKB, p53 pathway and the unfolded protein response (FDR<0.05). These results confirm the findings of a previous study in which placental HTR-8/SVneo cells were exposed to 20 µM DCVC for 12 h. The next phase of this study is to perform follow-up experiments and to publish the results.
Policy or Public Health Impacts
Ultimately, completion of this study will provide new insights that will inform public health officials and clinicians to allow for more effective communication regarding health risks from exposure to trichloroethylene during pregnancy.
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
The results of this project were included as preliminary data for a NIEHS Superfund Research Center P42 project grant submitted by Dr. Rita-Loch Caruso in collaboration with Wayne State University investigators. Scored, not funded.

July 2018 (Traditional Pilot Projects)

“Catalyzing collaboration on air pollution and adverse birth outcomes in Michigan”
Primary Investigator: Marie O'Neill
Abstract
The overarching goal of the project is to lay a foundation for acquiring multi-year external grant funding to support innovative, community-engaged research on environmental determinants of adverse birth outcomes. We will invite input from potential collaborators from the state health department, University of Michigan, Michigan State University, members of the community and the COEC Science Advisory Board on a research agenda and needed variables for this work. To start, we will generate estimates of air pollution exposure using geospatial techniques, link these to mothers’ residential locations from geocoded birth registry data for the state of Michigan (2000-2013), and conduct preliminary analyses. These air pollution estimates may be linked in the future to pregnancy outcome data collected for the Environmental Influences on Child Health Outcomes (ECHO) project, which includes birth outcomes from three Michigan-based cohorts, as well a prospective, larger, Michigan-wide sample of birth outcomes including over 1,000 pregnancies. The ECHO Michigan cohorts will enable evaluation of mechanisms by which air pollution and other exposures may contribute to adverse birth outcomes and provide key variables useful for epidemiologic research which are not collected by the state birth registry. We will also develop novel exposure estimates that go beyond traditional estimates by accounting for human mobility, yet without using personal exposure monitors, which can pose a considerable participant burden. We will construct such estimates by mining social media data, building on new research endeavors and the expertise of mathematician Daniel Romero. We plan to design the research and share findings with fellow academics, community partners and public health officials, engaging the Community and Outreach and Engagement Core (COEC) of the M-LEEaD center and other stakeholders in an effort to ensure research is relevant to designing interventions to improve maternal and newborn environmental health.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Cell type reference for saliva DNA methylation studies in children for environmental epidemiology”
Primary Investigator: Colter Mitchell
Abstract
A key focus of health and health inequality research is discerning mechanisms between childhood environmental exposures (pollution, dietary, chronic social stressors, toxicant, etc) and child and adult health outcomes. Childhood exposures are often some of the largest predictors of many behaviors and diseases. One particularly noteworthy biological mechanism linking childhood exposures to health is DNA methylation (DNAm)— an epigenetic modification of the genome that adds a methyl group to the Cytosine, but does not change underlying nucleotide sequence. Methods for collecting and analyzing DNAm have become rapidily more accessible over the last two decades and there is strong evidence that many childhood exposures and environments influence DNAm. However, for the studies that use salivary DNAm, there is significant concern that cell type may be confounding the relationship between DNAm, environment, and health. Public health prevention or clinical intervention would rely on an understanding of which mechanism is in action. This proposal fills these gaps by comparing DNAm across saliva cell types and generating the first deconvolution of saliva cell type-specific DNAm patterns. In this proposal we have three aims:<br /><br /> AIM 1. Identify DNA methylation signatures of salivary cell types<br /> AIM 2. Adapt a deconvolution algorithm for estimating relative cell proportions from composite tissue DNA methylation measures for the saliva<br /> Aim 3. Compare cellular distribution methods (data driven, leukocyte-only, and new saliva reference panel) in the Fragile Families and Child Wellbeing Study (n=2000 x 2 ages (9 and 15)). We will examine the extent to which different exposures are affected by the cell distribution estimates.<br /><br /> To conduct this study we have assembled an experienced team with collaboration history and expertise in salivary sample collection, epigenetics, cell sorting, statistical analyses, environmental and social exposures, and R-package production to ensure this work is carried out successfully and to pursue rigorously whether salivary cell types contribute to saliva DNAm heterogeneity. Notably, this team includes two epigenetic researchers (Bakulski and Colacino) who are established exposure and M-LEEaD scientists and two epigenetic researchers (Mitchell and Ware) who just beginning to integrate exposure data into their biosocial research.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Feminine Care Products and Exposure to Volatile Organic Compounds (VOCs)”
Primary Investigator: Sung Kyun Park
Abstract
Feminine care products, such as menstrual pads and tampons, are used widely by women worldwide. These products are intended for use on vaginal and vulvar tissues, which are more permeable than exposed skin and therefore have higher uptake and are potentially more vulnerable to exposure to toxic chemicals. They are marketed and sold with little to no data assuring the ingredients they contain are safe, and there are no government requirements to disclose all ingredients and manufacturing byproducts to consumers. Potential chemical exposure, especially of volatile organic compounds (VOCs), through use of feminine care products has been a public health concern. Although there is considerable concern and fear regarding toxic VOC exposure from menstrual pads and tampons, little is known about potential exposure risks of VOC through use of feminine care products. The widespread use of these products, the VOCs known to be present, the unique characteristic of the vaginal and vulvar tissues, the potential for health disparities among various groups of women through culturally-determined use of these products, and the large research gaps regarding exposure and effects, make this an emerging and significant health issue and research priority. Here, we propose a pilot study to achieve two specific aims: Identify VOCs in feminine care products by type (pads, tampons, douches, powders), brand (products with the top market share), and country (US, Korea, China); and Examine associations between patterns of the use of feminine care products (type, frequencies) and concentrations of VOCs in urine samples collected before, during and after menstruation. The expected outcomes of this project are to determine the emerging of VOCs including suspected and unexpected VOCs and potential differences in VOC concentration and composition among different products; and to provide the evidence that women may be at higher risk of VOCs exposure and related health outcomes due to the unavoidable use of feminine care products during the reproductive age. Ultimately, our research results will inform the public, regulators and policy makers about toxic chemicals contained in these products, and help reduce or avoid exposures and their resulting health impacts.
Accomplishments
Conference presentation: Lin N, Ding N, Park SK, Batterman S. Exposure to Volatile Organic Compounds (VOCs) in Feminine Care Products. ISES-ISIAQ 2019. Kaunas, Lithuania, August 18-22, 2019. Our main findings are: 1. Non-Hispanic black women had higher whole blood concentrations of 1,4-dichlorobenzene (1,4-DCB), a possible human carcinogen, and more likely to report frequent douche use. 2. We also found a positive relationship between frequency of vaginal douching and 1,4-DCB concentrations, indicating douche as a potential source of this VOC and might account for racial/ethnic differences among reproductive-aged women. 3. Use of feminine powder might also lead to increased whole blood concentrations of ethylbenzene. 4. Our findings suggest that douching or use of feminine powder be discouraged, especially during pregnancy. Manufacturers should disclose all ingredients, and screening tests to evaluate chemicals in feminine hygiene products should be considered.
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“In utero lead exposure and the heart: Potential intersection between metabolism and DNA methylation”
Primary Investigator: Laurie Svoboda
Abstract
Cardiovascular diseases are a major cause of morbidity and mortality world-wide, and accumulating evidence suggests an important role for early-life toxicant exposures, including lead (Pb), in their etiology. As developmental Pb exposure remains a public health concern in both industrialized and developing nations, a better understanding of how Pb contributes to cardiovascular diseases is urgently needed. The mechanisms by which Pb and other toxicant exposures influence the long-term risk of cardiovascular diseases are not fully understood; however, developmental reprogramming of DNA methylation and hydroxymethylation likely plays a role. Despite known impacts of Pb exposure on heart function, the effects of Pb on the cardiac transcriptome and epigenome have not been investigated. Recent studies in cancer and stem cell biology demonstrate that epigenetic changes are closely coupled to the metabolic state of the cell, enabling cells to detect, and rapidly respond to, environmental cues. However, surprisingly little is known about how toxicant exposures, including Pb, influence the dynamic interplay between cellular metabolism and the epigenome. Using tissue generated from a well-established mouse model of developmental toxicant exposures, the purpose of this proposal is to test the hypothesis that exposure to Pb <em>in utero</em> and during lactation leads to changes in the transcriptome of the heart, particularly in environmentally labile genes critical for normal cardiac development and physiology. We will further test the hypothesis that Pb-induced transcriptomic and epigenomic changes are linked to alterations in cellular metabolism. The findings from this study will provide critical mechanistic insight into Pb’s deleterious effects on the heart, and identify potential opportunities for nutritional and pharmacologic intervention. Moreover, it will provide the key preliminary data necessary for us to seek further funding to explore this exciting area of investigation.
Accomplishments
We have conducted transcriptomic analysis of hearts from male and female mice exposed to control or leaded water at three separate time points during the animal’s life span: at weaning, in young adulthood, and in later adulthood. Additionally, we have performed enhanced reduced representation bisulfite sequencing of hearts from control or lead-exposed animals in young adulthood to look at reprogramming of DNA methylation by lead exposure. Finally, we have analyzed levels of several metabolites that are key for epigenetic programming in the hearts of control and lead-exposed animals at weaning. Quality control and analysis of the data are in progress.
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
This work will provide critical preliminary data for applications to the NIEHS (Transition to Independent Environmental Health Research Career Award K01) and the American Heart Association (Career Development Award), which will be submitted in October, 2019.
“One-Carbon Metabolism and Chemical Toxicokinetics in the Developing Embryo”
Primary Investigator: Karin Veltman
Abstract
Human and animal studies have implicated impaired one-carbon (C1) metabolism as a potential mechanism through which prenatal exposure to environmental factors (incl. toxicants and nutrients) cause epigenetic modifications (specifically modified DNA methylation patterns). These modifications to the embryonic or fetal genome can result in an adverse phenotype in later life. Yet, it remains poorly understood how environmental exposures perturb C1 metabolism and how these perturbations impact DNA methylation patterns. This is partly due to the complexity of the C1 pathway. Mathematical, systems biology based models are well-suited to capture this complexity and the consequences of network perturbations on C1 metabolites and DNA methylation, but quantitative approaches are scarce.<br /><br /> Our long-term research goal is to develop a mechanistic, system-biology based model that can be used to explain and predict the impact of environmental factors (specifically toxicants and nutrient deficiency) on DNA methylation patterns in the organogenesis-stage mammalian embryo. A lack of data currently hampers our ability to develop such a model and in this pilot project we seek to fill this data gap using well-established whole embryo culture (WEC) techniques. Specifically, we seek to i) characterize the ontogeny of key C1 network components (metabolites & cofactors) under unperturbed conditions, and ii) to develop and validate experimental methods that can accurately quantitate the toxicokinetics (absorption, distribution, metabolism, excretion rates) of mono-ethylhexyl-phthalate (MEHP) in the developing embryo. The acquired data will be used to develop an empirically-based mass-balance model for C1 metabolism and to evaluate the applicability of a published model for C1 metabolism in the adult liver to the embryo.<br /><br /> This pilot project will provide the analytical techniques, preliminary data and first model prototype needed for a R01 application. It furthermore addresses two Funding Objectives of this RFA: First, dr. Veltman is an early career investigator (less than 10 years beyond her doctoral degree) and this proposal provides initial support for her own research line. Second, this proposal provides the basis for initiating a new research direction in EHS that combines mechanistic modeling and experimental research to define, explain and predict the impacts of environmental exposures on C1 metabolism and epigenetic modifications during early embryogenesis.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Toward discovering stabilizers of cellular manganese levels”
Primary Investigator: Young Ah Seo
Abstract
Manganese (Mn) is an essential nutrient required for normal growth and development. However, exposure to elevated Mn concentrations can cause irreversible neuronal injury in both children and adults. The essential yet neurotoxic nature of Mn appears as a biphasic dose-response curve that reflects its neurodevelopmental health outcomes in children. Children are particularly vulnerable to the neurotoxic effects of environmental Mn exposure as their brains are undergoing a dynamic growth and development process. Neurotoxic injury to the developing brain is closely linked to human neurological disorders, but we do not know the genetic basis for the apparent greater sensitivity of children.<br /><br /> Environmental exposure studies have extensively focused on the downstream effects of Mn toxicity on brain function and development. This has hindered research into how the alteration of cellular Mn levels initially occurs. Little is known about how cells maintain Mn levels within an optimal range when extracellular Mn levels increase or decrease. Our preliminary studies have shown that mRNA expression of <em>SLC39A8</em>, encoding a newly-identified Mn transporter, increases when extracellular Mn levels decrease, while expression is downregulated with excess extracellular Mn. These results indicate the existence of homeostatic mechanisms by which cells sense extracellular Mn levels, signal the nucleus, and alter the expression of this key Mn transporter gene.<br /><br /> Capitalizing on our preliminary observations, we seek to identify genes that are essential for cellular Mn homeostasis. We plan to employ unbiased CRISPR/Cas9 genetic screens to uncover the upstream machineries that maintain optimal cellular Mn levels in response to environmental Mn changes. We propose to generate cell lines that report SLC39A8 expression using CRISPR/Cas9–mediated GFP tagging of endogenous SLC39A8. Such genetic screening strategies have yet to be explored in the field of environmental toxicant exposure. If successful, this pilot project will generate the first Mn-responsive gene reporter system in human cells. Future studies will perform CRISPR/Cas9 screens in human cells and primary neurons to uncover the key cellular players in Mn homeostasis and developmental toxicity. This pilot project will provide the preliminary results required for the submission of a future NIEHS R01 proposal.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

January 2018 (Targeted Pilot Projects)

“Developing Effective Training Approaches to Reduce Chemical Exposures Among Nail Salon Workers ”
Primary Investigator: Marie-Anne Rosemberg
Abstract
Nail salons workers are exposed to harmful chemical, biological, and ergonomic hazards. Furthermore, most workers in this industry (especially in the State of Michigan) are women of Vietnamese descent, with limited English proficiency and low literacy. Over the past two years, we have been growing an effort through the Michigan Health Nail Salon Cooperative with the goal of promoting a healthier nail salon workforce through advocacy, education and research. Information on the hazards of nail salon work is available on the internet and in print. However, our focus group meetings with nail salon workers within the community indicate that the information is not being transmitted effectively to the workers. The nail salon workers expressed the need for and importance of a culturally sensitive interactive online training approach. This community-participatory project aims to address this expressed need. We propose to collaborate with local nail salon owners and workers to augment our nascent efforts by developing online video-enhanced training materials and to evaluate the effectiveness of the materials. We will focus on chemical exposures and safety measures. Results will be used to refine our approach (as needed) and to support a larger proposal that expands the scope to include module on other key topics (e.g. ergonomics, biological exposures, labor practices). Ultimately, our goal is to not only promote good practices among nail salon owners but also increase client awareness. Our vision includes creating a certification program for “safe nail salons” with a medallion for qualified salons to display, letting customers know that their nail salon is “safe” and thereby of “higher quality”. For this proposed project our aims are to 1) design and develop a multi-part, video-enhanced, on-line training module focusing on best practices for reducing exposures to hazardous chemicals encountered by nail salon workers, and 2) pilot and evaluate our training module/approach with 2-3 salons and at least 6 nail technicians in Ann Arbor and/or Ypsilanti).
Accomplishments
Podium presentation at : The 11th International Occupational Hygiene Association (IOHA): International Scientific Conference. Washington DC, USA. September 25th, 2018
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Evaluating the Association between Biomarkers of Parabens and Phenol Exposure and Recurrence in ER-Positive Breast Cancer Patients”
Primary Investigator: Justin Colacino
Abstract
While many ER-positive (ER+) breast cancer patients respond well to treatments that target the synthesis of estrogen in vivo, such as aromatase inhibitors (AIs), many patients still experience a recurrence for unknown reasons. A possible mechanism of resistance to antiestrogen therapy for patients on AIs may be exposure to environmental estrogen mimicking chemicals, or xenoestrogens. It is not well understood what effect estrogen mimicking compounds have in ER+ breast cancer patients taking antiestrogen therapy or whether xenoestrogen exposure could contribute as possible mechanism of resistance for these patients. Given the ubiquitous exposure of the public to xenoestrogens like BPA and parabens, it is possible that xenoestrogen exposure could explain, in part, why adjuvant chemotherapy fails in patients taking AIs. The long-term goal of this study is to evaluate the association between urinary paraben and phenol concentrations and a patient&#8217;s risk for recurrence. Our hypothesis is that ER+ breast cancer patients who have recurred will have significantly higher concentrations of urinary xenoestrogens compared to breast cancer patients who have not recurred. We will test this hypothesis by comparing xenoestrogen concentrations in banked urine samples from ER+ breast cancer patients on anti-estrogen therapy who have not recurred (n=25), and urine samples collected from ER+ breast cancer patients who have experienced a recurrence (n=15). Urinary paraben and phenol concentrations will be quantified by liquid chromatography-mass spectrometry analysis at NSF International using an established protocol. We anticipate that these results will provide key preliminary data to support a larger clinical study to assess the role between xenoestrogen exposures and progression of ER+ breast cancer. The larger study will be prospective and will enroll non-metastatic patients taking anti-estrogen therapy who are at a high-risk for recurrence in the adjuvant setting. The larger sample size and prospective design would provide clear evidence as to whether or not exposure to parabens, or other estrogenic compounds such as phenols, is a risk-factor for breast cancer recurrence among patients on anti-estrogen therapy.
Accomplishments
These analyses were successfully completed, however, during the molecular analyses we realized that urine samples form the ER+ breast cancers with metastatic disease were preserved in a different way than the samples from the ER+ breast cancer patients without metastatic disease. We did not observe any significant differences in estrogenic chemical exposures between groups. However, the background literature review conducted by Dr. Gonzalez was developed into an article published in Toxicology, which outlines the broader hypotheses of the study.
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Geospatial Analysis of Air Pollution Exposures During Pregnancy: Ambient Air Pollution and Wildland Fire Smoke Modeling ”
Primary Investigator: Patricia Koman
Abstract
Fine particulate air pollution (PM2.5) exposures during pregnancy have been positively associated with adverse birth outcomes and increased pregnancy-induced hypertensive disorders. Air pollution is a preventable source of cardiovascular disease during vulnerable lifestages. Little information exists, however, about exposures and health effects associated with an important growing contributor to air pollution of wildland fire smoke, especially related to exposures during pregnancy. We hypothesize that increased exposure to air pollution during pregnancy will be associated with higher odds of adverse maternal and birth outcomes. The purpose of this short-term pilot is two-fold: to perform exposure assessments utilizing air quality modeling data and to engage with government representatives who translate data to communities impacted by wildland fires and other air pollution. We propose to create an exposure assignment methodology and to create coding that will match geospatial modeled air quality outputs with residential geocoded addresses of participants (e.g., using zipcodes or latitude and longitude by date). Because we plan to use hypothetical addresses for our coding queries as proof of concept, no IRB approval is needed. Our proposed modeling domain is California (years 2008-2012), which has been mapped with the U.S. Environmental Protection Agency (EPA) state-of-the-art atmospheric model called Community Multi-scale Air Quality (CMAQ v5.0.1/5.0.2) at 12 km grid cell resolution. We also propose innovative new methods such as create smokewave indicators to account for exposures specific to wildland fire smoke. Our project includes input from forest ecologists, geospatial analysts, epidemiologists, biostatisticians, and public health practitioners. This publicly engaged translational research project partners with California Department of Public Health and adds new partners from the U.S. EPA Office of Research and Development. We propose a new engagement activity to present our initial results for partner feedback to inform their work translating health data to impacted communities and vulnerable groups.
Accomplishments
We obtained, cleaned and performed descriptive statistics including additional exposure assessments utilizing air quality modeling data. We engaged with California Department of Public Health and US Environmental Protection Agency Office of Research and Development representatives who translate data to communities impacted by wildland fires and other air pollution. We presented our initial results and we worked together to submit two grant proposals. We completed two future grant proposals to continue this research (Graham Sustainability Institute, Koman (PI), and American Heart Association, French (PI)). We made progress toward creating smokewave indicators and procedures for matching air quality and administrative health records. Our manuscript was accepted for publication and we presented several posters at national conferences..
Policy or Public Health Impacts
We made progress on research that will fill an urgent gap in information about how wildland fire smoke affects maternal health and birth outcomes. With improved understanding, better mitigation and public warning systems can be created to improve public health.
Publications
  • Koman, P.D., M. Billmire, K.R. Baker, F. J. Anderson, B. Thelen, S. Hoshiko, R. de Majo, N.H. French. Mapping modeled exposure to wildland fire smoke for human health studies in California. (accepted June 1, 2019, Atmosphere)
  • Koman, P.D., M. Billmire, N.H. French. Understanding health equity and population risk implications of exposure to air pollution from wildland fires and hypertensive disorders of pregnancy in California APHA ID 413874, American Public Health Association Annual Meeting, San Diego, CA, Nov 4, 2018
  • French, N.H., K. Anthony, K. Baker, M. Billmire, S. Hoshiko, J. Hutchinson, J. Johnson, P.D. Koman, B. Koziol, V. Limaye, J. McCarty, R. C. Owen, C. Reid, B. Thelen, S. Wu. Approaches and challenges to connecting exposure to air pollution from wildland fire emissions to health outcomes. University of Michigan Environmental Statistics Day, Ann Arbor, MI, March 14, 2018
  • Koman, P.D., NH French, M Billmire, S Hoshiko, BJ Thelen. Modeling Wildland Fire Smoke Exposure for Quantifying Human Health Associations, University of Michigan MUSE conference for February 14-15, 2018
Grant applications and grant awards resulting from Pilot Project
We completed two future grant proposals (Graham Sustainability Institute, Koman (PI), and American Heart Association, French (PI)).
“Impact of exposome on maternal and fetal steroidal milieu”
Primary Investigator: Vasantha Padmanabhan
Abstract
Humans are exposed to a heterogeneous mixture of endocrine-disrupting chemicals (EDCs) with pregnant women being particularly vulnerable. Emerging evidence suggests that these EDCs alter metabolic homeostasis during pregnancy and affect fetal growth and initiate a set of physiologic responses in the fetus that predisposes the offspring to obesity and other chronic diseases later in life. However, the pathways through which this mixture of chemicals additively or synergistically impacts offspring health in humans are mostly unknown. One pathway that can alter the fetal developmental trajectory involves steroidal hormones, which are powerful programming agents as evident by their role in sexual differentiation during fetal life. Our research program is focused on understanding the effects of EDC mixtures on the maternal metabolic state across pregnancy and their impact on birth weight, a risk factor that influences the health of the offspring across the life course. This pilot proposal will use maternal blood samples collected during first trimester and delivery and cord blood at delivery to determine the relationship between environmental exposures and maternal and fetal steroidal changes, and birth outcomes. We have already characterized a wide spectrum of chemical exposures (phthalates, phenols, and metals) during early pregnancy and fetal life as well as maternal and newborn inflammasome, metabolome, and epigenome (CHEAR support). In this pilot proposal, we propose to characterize the steroidal hormone profile in the same set of samples/subjects to determine how exposure to a heterogeneous mixture of EDCs influences maternal and fetal steroidal milieu and how changes in steroid hormones relate to changes in inflammasome, metabolome, epigenome, and birth outcomes. We hypothesize that maternal exposures to EDC mixtures disrupt maternal and fetal steroidal profile that underlie the changes in maternal inflammasome, metabolome and epigenome and birth outcomes. The proposed study will help understand the maternal pathways through which EDCs impact fetal growth. The results and insights gained in this study will form the foundation for future research studying the effects of heterogeneous exposures to EDCs on the intrauterine environment and health outcomes that have their origins in fetal development.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“MI-Environment: Heat Stress Vulnerability Rapid Response”
Primary Investigator: Patricia D Koman
Abstract
The National Academies of Science report points to the need for innovative methodologies and tools to consider cumulative environmental exposures in policy (NAS 2009). California has been a leader in cumulative risk frameworks and has implemented a cumulative exposure index in the EnviroScreen tool, which serves as a model for our endeavor (Cal EPA 2014, Morello Frosch et al. 2011). Northern states such as Michigan have a need to characterize climate change related exposures and vulnerabilities, especially regarding heat stress for which the population, infrastructure, and practices may not be well adapted. Our previous research led to an innovative data product -- a heat stress Geographic Information System (GIS) layer of a larger cumulative exposure visualization tool: http://spatial.mtri.org/static/greatlakeswaterquality/romoGIS/. We assembled underlying data and created climate change vulnerability indices and maps that describe the location and relative magnitude of heat stress vulnerability across the state of Michigan at the census tract level. Our aim is to characterize the extent and impact of cumulative exposures to environmental and social vulnerabilities in the State of Michigan at the census tract level. This proposal focuses on publishing a manuscript related to these vulnerabilities and susceptibilities to heat stress, and the use of a Integrated Health Sciences Core (IHSC) geospatial analyst. In addition, using IHSC consultation and services, we will perform statistical analyses to examine to what extent racial disparities in exposures and/or susceptibilities exist using techniques and prepare maps and graphics for a manuscript (Schulz et al. 2016). Our MI-Environment visualization tools can help identify and reduce environmental hazards for our state&#8217;s most vulnerable communities. If funded, our project will create a manuscript for an innovative, translational instrument to bring scientific data in service to the public in a meaningful, engaged and scientifically rigorous manner. Our long-term goal is to engage the public in citizen science to use local knowledge to validate geospatial locations and to add additional environmental and social vulnerability factor layers (as part of future funding). Our long-term project will foster population studies to understand of susceptibility across the life span to chronic diseases resulting from environmental factors and to promote health.
Accomplishments
We completed additional statistical analyses and we successfully prepared a manuscript now under review for publication in a peer reviewed journal.
Policy or Public Health Impacts
Coming soon!
Publications
  • Koman, P.D., F. Romo, P. Swinton, G. Mentz, R, de Majo, N. Sampson, M.J. Battaglia, K. Hill-Knott, G.O. Williams, M.S. O’Neill, A.J. Schulz. MI-Environment: Geospatial Patterns and Inequality of Relative Heat Stress Vulnerability in Michigan (Manuscript under review)
  • Sand, S.L. , Koman, P.D., Sampson, N. , Gronlund, C., Romo, F., Reyes, A.G. 5 , Gray, C., Williams, G.O., Schulz, A.J., Use of a Science Cafe to Share and Discuss Climate and Health Research Findings, Partnerships for Environmental Public Health (PEPH) Meeting, Poster Presentation, 2018 National Institute of Environmental Health Sciences, Research Triangle Park, NC, December 13, 2018
  • Koman, P.D., F. Romo, C. Gray, N. Sampson, S. Landfried, M. Battaglia, K Hill-Knott, N.H. French, M.S. O’Neill, A.J. Schulz. MI-Environment Geospatial Analysis: Promoting Climate-Related Health within Michigan Vulnerable Communities. Planetary Health and GeoHealth Annual Meeting, Poster Presentation, Boston, MA, April 29, 2017
  • Koman, P.D., F. Romo, C. Gray, N. Sampson, S. Landfried, M. Battaglia, M.S. O’Neill, A.J. Schulz. MI-Environment: Promoting Climate-Related Health within Michigan Vulnerable Communities. University of Michigan Environmental Epidemiology research seminar, Ann Arbor, MI, February 14, 2017
  • Koman, P.D., F. Romo, C. Gray, N. Sampson, S. Landfried, M. Battaglia, R Goodspeed, M.S. O’Neill, A.J. Schulz. MI-Environment: Heat Stress Vulnerability in Michigan Communities. Michigan University-wide Sustainability & Environment (MUSE), oral presentation, Ann Arbor, MI, February 9, 2017
  • Koman, P.D., F. Romo, C. Gray, N. Sampson, S. Landfried, M. Battaglia, M.S. O’Neill, A.J. Schulz. MI-Environment: Climate and Health in Michigan. National Institute for Environmental Health Sciences (NIEHS) 50th Anniversary Research conference, Poster Presentation, Durham, NC, December 8, 2016
  • Koman, P.D., F. Romo, C. Gray, N. Sampson, S. Landfried, M. Battaglia, M.S. O’Neill, A.J. Schulz. MI-Environment: Promoting Climate-Related Health within Michigan Vulnerable Communities. American Public Health Association, Denver, CO, October 31, 2016
  • Koman, P.D., F. Romo, S. Landfried, M O’Neill. MI-Environment: Heat Vulnerability Mapping Interactive Tool Michigan Lifestage Environmental Exposure & Disease Center pilot, Science Cafe, Detroit, MI, March 24, 2016
  • Koman, P.D., invited speaker, Climate Change and MI-Environment Heat Vulnerability Index: Opportunities for Research and Partnerships, University of Michigan, NIEHS Community Outreach & Engagement Core, Michigan Lifestage Environmental Exposure & Disease Center, Faculty and partners mobile meeting, Detroit, MI, November 13, 2015
  • Koman, P.D., K. Huber, F. Romo, S. Landfried, R. Goodspeed, M O’Neill. MI-Environment: Heat Vulnerability Mapping. NIEHS Community Outreach & Engagement Core, Michigan Lifestage Environmental Exposure & Disease Center, Stakeholder Advisory Board Meeting, Detroit, MI, July 22, 2015
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Neighborhood Noise Prediction Models for Epidemiology Studies of Chronic Health Across the Life-Course: Which Ones Work?”
Primary Investigator: Sara Adar
Abstract
Neighborhood noise has been linked to chronic health problems across the life-course ranging from disturbed cognition and sleep in children to hypertension and cardiovascular disease in adults and dementia in seniors. As a result, neighborhood noise has been identified as a major contributor to disease in Europe by the World Health Organization. However, virtually no work has been done to characterize the population burden and health disparities in the US largely due to a lack of an established methodology to assess noise. With a growing interest in this exposure by community groups and funding agencies, there have been a few groups (including ours) that have generated either empirical or deterministic estimates for neighborhood noise on the national or local scale. These models have not been externally validated to date with the exception of a recent survey by our group that showed that one new national model validates very poorly (i.e., R2~0-0.3). As a result there is a critical need to assess how well new spatiotemporal models being employed in the US actually estimate noise, especially in complex urban landscapes, before applying them in epidemiology studies. The aim of this proposed project, which forms new collaborations between an epidemiologist (Dr. Adar), noise expert (Dr. Seto), and an exposure scientist (Dr. Seto), will systematically compare and validate four different exposure assessment techniques now being deployed in the US for noise ascertainment. The result of this research will be the identification of the most robust method for predicting noise in a large US urban area. As such this pilot project is expected to create an important citation for planning noise epidemiology research in the US. In addition, this research will also specifically support a new R01 proposal by our group on neighborhood noise as one risk factor for cognitive decline and dementia in a large prospective cohort study.
Accomplishments
Two field campaigns were successfully completed in Seattle, collecting over 400 samples of noise across the city. We have constructed our land-use regression model for the city and compared to other exposure models as planned. Importantly, our work showed some critical limitations with some of the existing national noise models. Preliminary results of these analyses were presented at the Conference for the International Societies for Exposure Science and Environmental Epidemiology in August of 2018 and we are currently working on a publication regarding our findings.
Policy or Public Health Impacts
This pilot study was informative regarding the usefulness of existing noise models for public health research.
Publications
  • Noise exposure assessment in North America: Past Challenges and Future Opportunities Abstract presented at the Joint Conference of the International Societies for Exposure Science and Environmental Epidemiology
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Single cell mapping of the human placenta for identifying environmentally sensitive cell populations”
Primary Investigator: Kelly Bakulski
Abstract
Improvements in maternal and infant health are vital to creating a healthy future. The United States has the highest infant mortality rate and percentage of preterm births among high-income countries. Major contributing factors include placental insufficiency and toxicant exposures. Since the placenta provides the interface between the mother and fetus and regulates nutrients, waste, and oxygen, changes in placental function affect fetal and long-term health. The placenta is a transient, rapidly developing organ comprised of a mixture of heterogeneous cell types, including Hofbauer cells, endothelial cells, fibroblasts, leukocytes, extravillous trophoblasts (EVT), cytotrophoblasts (CT), and syncytiotrophoblasts (ST). A few placental cell types have been gene expression profiled, however analysis of thousands of cells in aggregate conceals functional variability among cells. In order to understand placental environmental sensitivity, we must be able to identify cell types at risk. Current bulk placental tissue environmental epidemiology is hampered by lack of consideration of cellular heterogeneity. We are unsure of the number and proportion of distinct cell types in the placenta, or their gene expression characteristics. Indeed, cells have different levels of xenobiotic metabolizing capacity, which directly influences sensitivity to environmental toxicants. To achieve an unparalleled understanding of placental composition, biology, and function, we propose to generate a cellular RNA atlas of the human placenta, using single cell RNA-sequencing (scRNA-seq) analysis. ScRNA-seq will uncover cell type specific biological functions, which will provide the first unbiased survey of functional heterogeneity in placenta, serving as a biological reference for placental toxicological models and environmental birth cohorts. We will compare these unbiased findings to our more traditional sorted cell results. Understanding links between cellular differentiation state and placental transcription in response to toxicants may offer mechanistic insight into fetal programming and adult disease.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Single Cell Transcriptomics to Map the Effects of Developmental Lead Exposure on the Hippocampus”
Primary Investigator: Kelly Bakulski, Justin Colacino
Abstract
Lead exposure is ubiquitous and CDC estimates that 500,000 or 2.5% of US children under the age of 5 currently have blood lead levels above the reference level for demonstrated neurological deficits (5 μg/dL). Lead is responsible for approximately 1% of the global burden of disease, primarily through permanent effects on childhood intelligence and behavior. The hippocampus is a key brain region involved in learning, memory, and response to stimuli. Both epidemiology and toxicology studies have noted changes in hippocampal function and bulk tissue molecular characteristics. Recent spatial transcriptomics work highlights the highly heterogeneous nature of the hippocampus. Toxicants can act on the hippocampus by altering gene expression and most current work investigates bulk brain tissue or composite regions, but gene regulation is tissue and cell type specific. Study of the hippocampus’ sensitivity to environmental factors, on the level of individual cells, is needed to understand biological determinants of neurodevelopment. In archived tissue from a longitudinal study of mice perinatally exposed to lead or control diet, at 5-months we test for differences in the hippocampus at the single cell level. We will perform single cell RNA-sequencing analysis on 26,000 cells from the hippocampus by lead exposure to 1) test for genome-wide gene expression differences by exposure across all cells; 2) compare cell population and subpopulation proportions by exposure; and 3) test for gene expression differences within cell populations by exposure. We will determine whether the toxic effects of lead are uniform across hippocampal cells or specific to particular sensitive cell types. This proposal is a stepping stone to future work linking cell population specific neurological changes to behavioral changes, characterizing the molecular basis by which lead influences cognition and behavior. Understanding links between cellular differentiation state and hippocampal transcription with toxicant exposure may offer mechanistic insights into the neurodevelopmental effects of lead exposure and can highlight potential interventions.
Accomplishments
This experiment was conducted and the data were generated. Preliminary analyses of these data were used to support an unsuccessful NIEHS R21 application, where one of the major critiques was that we had not published enough single cell papers. As of June 2019, we are close to getting the manuscript describing the results of these experiments finalized for submission.
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Grant application - "Single Cell Transcriptomics to Map the Effects of Developmental Lead Exposure on the Hippocampus" NIEHS R21, Not funded
“Targeted metabolomics analysis of amniotic fluid to identify new TCE actions in a rat model of fetal growth restriction”
Primary Investigator: Rita Loch-Caruso
Abstract
Exposure to trichloroethylene (TCE), a common environmental contaminant, has been associated with preterm birth and decreased fetal weight in epidemiological studies [1-3]. Moreover, toxicology studies from our laboratory found placental oxidative DNA damage, epigenetic alterations, and changes in gene expression in a rat model of fetal growth restriction due to TCE exposure during pregnancy [4]. Although the latter study illuminates possible molecular mechanisms by which TCE may act to produce adverse pregnancy outcomes, further studies are needed to elucidate the role of the placenta in TCE-induced fetal growth restriction. The proposed project will use targeted metabolomics approaches to analyze amniotic fluid samples already collected from pregnant Wistar rats exposed to TCE. The rats were exposed to 480 mg TCE/kg/day, a dose that is relevant to human occupational exposures and induces oxidative stress in rats [5-6]. Using the vanilla wafer method developed by Seegal et al. [7], the rats were administered either the wafer alone (control) or a wafer with TCE on a daily basis from gestational day (GD) 6 through GD16, with the day of copulation designated GD0. Prior to TCE exposure, the rats were trained to eat the wafers without TCE on GD3 to GD5, which ensured that the wafers were eaten within a few minutes of presentation. Amniotic fluid samples were snap frozen in liquid nitrogen and are currently stored at -80 &Acirc;&deg;C. To allow analysis for sex as a biological variable, we will analyze samples from 4 male fetuses and 4 female fetuses in each treatment group, with samples from one male and one female fetus per litter (8 litters, 16 samples total). These samples will be analyzed at the University of Michigan Metabolomics Core by TCA (tricarboxylic acid)-plus targeted and short chain fatty acid targeted platforms. Because our prior studies show that a TCE metabolite induces oxidative stress and disrupts mitochondrial function in placental cells in vitro [8,9], metabolites detected by these metabolomics platforms will provide insight into whether TCE exposure disrupts cellular respiration in vivo with possible discovery of pathways that contribute to adverse pregnancy outcomes.
Accomplishments
Trichloroethylene (TCE) is an environmental contaminant shown to be associated with low birth weight and small for gestational age in human epidemiological studies, decreased fetal weight in Wistar rats, and impaired mitochondrial function in placental cells. Building upon this information, the present study conducted tricarboxylic acid (TCA) Plus and short chain fatty acid (SCFA) targeted metabolomics analysis on amniotic fluid samples to identify changes in metabolites induced by exposure to TCE. Rats were exposed to 480 mg TCE/kg/day from gestational day (GD) 6 to GD 16 in a food treat, and amniotic fluid samples were collected on GD 16. We observed statistically significant changes in several metabolite concentrations, some of which were specific for fetal sex. In general, our results indicate impacts to metabolites directly or indirectly involved in energy supply in the amniotic fluid as part of the response to TCE exposure, with different effects on amniotic fluid metabolites in male and female fetuses.
Policy or Public Health Impacts
This work advances understanding of molecular mechanisms by which trichloroethylene (TCE) exposure may increase risk for adverse pregnancy outcomes. As such, this knowledge may help inform public health approaches for reducing the occurrence of adverse pregnancy outcomes from environmental contaminants.
Publications
  • Su AL, Harris SM, Elkin, ER, Loch-Caruso R. Trichloroethylene Stimulates Metabolomic Changes in the Amniotic Fluid of a Timed-Pregnant Wistar Rat Model of Fetal Growth Restriction. In: 2019 Annual Meeting Abstract Supplement, Society of Toxicology, 2019. Abstract no. 1511. (poster presentation).
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Toxicant Impacts on Exosomal Cell-Cell Communication between Immune and Placental Cells”
Primary Investigator: Rita Loch-Caruso
Abstract
Toxicant insult to the placenta can adversely impact health of the offspring and mother. However, advances in understanding how toxicants disrupt human placental development and function have been slowed, in part, due to limited access to the placenta during human pregnancy. The proposed work on placental function focuses on exosomes, nanosized extracellular vesicles secreted from living cells. Exosomes are released by various cell types including placental and immune cells. Because their content can include signaling molecules such as RNA, lipid mediators, hormones and proteins, exosomes represent a mechanism of cell-cell communication. Placental-specific exosomes have been isolated from maternal blood and shown to modulate immune cell function. Likewise, exosomes isolated from macrophages have been shown to modify placental cells. We hypothesize that toxicants modify exosomal cell-cell communication between placental and immune cells. For this short-term project, we will conduct experiments with immune (K562) and placental (BeWo) cell lines exposed to mono-(2-ethylhexyl) phthalate (MEHP), a bioactive metabolite of bis(2-ethylhexyl) phthalate (DEHP). DEHP is a plasticizer previously associated with preterm birth, and MEHP has been shown by us to activate prostaglandin, oxidative stress, and pro-inflammatory cytokine pathways in placental cells. The human BeWo cell line will be used as a cell culture model for placental syncytiotrophoblasts, the placental cells in direct contact with maternal blood, and the human K562 cell line will be used as an immune cell model. The proposed work has two aims: 1) Isolate and characterize exosome release from immune (K562) and syncytiotrophoblast (BeWo) cells without and with toxicant exposure, and 2) Assess MEHP-induced disruption of exosomal cell-cell communication between immune (K562) and syncytiotrophoblast (BeWo) cells. The data generated will provide validation for future NIH grant applications of the expertise of our research team to isolate, characterize, and utilize exosomes to study placental cell function, while yielding novel information on toxicant impacts on exosomal cell-cell communication between immune and placental cells. In the long term, this project will lead to new insights into the feasibility of using liquid biopsy materials to inform, interpret, and test relationships between environmental exposures and placental health.
Accomplishments
Exosomes are secreted nanosized extracellular vesicles (EVs). Our initial experiments investigated exosomal cell-cell communication between placental and immune cells in media swap experiments with immune (K562) and placental (BeWo) cell lines, with and without exposure to mono-(2-ethylhexyl) phthalate (MEHP), a bioactive metabolite of bis(2-ethylhexyl) phthalate (DEHP). We measured global gene expression changes in K-562 cells after treatment with media conditioned with secreted factors from a placental cell line (BeWo). We observed a robust set of gene expression changes in K562 cells exposed to BeWo-conditioned media versus K562 cells exposed to non-conditioned media (controls). In contrast, there was a relatively lower number of gene expression changes in K562 cells exposed to conditioned media from BeWo cells treated with 10 µM MEHP versus K-562 cells exposed to conditioned media from BeWo cells not exposed to MEHP. The media swap experimental results are consistent with placental BeWo cell secretion of soluble factors that modify gene expression in immune K562 cells. We are conducting follow-up experiments to identify key signaling molecules driving these gene expression changes. In additional experiments, we are investigating if the changes we observed in immune cells are driven by EVs/exosomes. We successfully isolated nanosized EVs from K562 cells and observed a concentration-dependent increase in total number of exosomes in cells treated with tert-butyl hydroperoxide, a positive control toxicant that generates reactive oxygen species. We had low yield in our attempts to isolate EVs from BeWo cells: we are working on enhancements to our technique and study design to improve our EV yield. Future experiments will determine whether phthalates elicit similar effects on the number and/or contents of exosomes released by K-562 and BeWo cells.
Policy or Public Health Impacts
This project will lead to new insights into the feasibility of using liquid biopsy materials to inform, interpret, and test relationships between environmental exposures and placental health as well as inform the broader etiology of placental disorders. Ultimately, these findings will have the potential to develop circulating markers of placental health leading to earlier detection and improved treatment for placental disorders.
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Sean Harris (PI) and Rita Loch-Caruso (Mentor). NIH/NIEHS, “Identifying Molecular Mechanisms of Pathogen-Toxicant Interactions in Extraplacental Membrances: Implications for Preterm Birth.” Under review. Sean Harris (PI) and Rita Loch-Caruso (Mentor). Burroughs Wellcome Trust, “Identifying Molecular Mechanisms of Pathogen-Toxicant Interactions in Extraplacental Membranes: Implications for Preterm Birth.” Not funded.

May 2017

“Health Risks to Vulnerable Populations from Exposures to Airborne Asbestos from the Demolition of Abandoned Homes in Detroit”
Primary Investigator: Richard Neitzel
Abstract
Abandoned residential dwellings (ARDs) represent social, physical, and economic hazards to communities, and especially to people in vulnerable lifestages, such as children and the elderly. Demolition of ARDs is essential to protect public health, but is complicated by the need to remove regulated asbestos-containing material prior to demolition. Working in close partnership with the City of Detroit, we propose to evaluate the benefit to public health from asbestos abatement before the demolition of ARDs. Specifically, we propose: • To conduct a systematic analysis of airborne asbestos data collected in Detroit. The City has already demolished >10,000 homes. Asbestos in the majority of these homes was abated predemolition, but a fraction was demolished unabated under emergency protocols. We will compare airborne asbestos concentrations during the demolition of 300 abated and 300 unabated homes. • To measure airborne asbestos using a traditional method, as well as a more advanced method The standard technique used by the demolition contractors, Phase Contrast Microscopy (PCM), has been criticized as it cannot distinguish among types of asbestos fibers. We will directly measure airborne asbestos concentrations during the demolition of 10 abated and 10 unabated homes using PCM as well as transmission electron microscopy with an energy dispersive X-ray system analyzer (TEM/EDX), which can more definitively identify and quantify asbestos fibers. • To calculate the lifetime risk of asbestos-related disease due to demolition activities. We will use the existing measurements and our new measurements to estimate demolition-related asbestos exposures under both abated and non-abated conditions. Using these exposures, we will estimate the incremental lifetime risk of asbestos-related disease among vulnerable individuals, as well as the community as a whole, using dose-response models. By quantifying the benefit of asbestos removal in ARDs prior to demolition to the health of vulnerable populations and communities, this research will have far-reaching implications, not only for the City of Detroit, but also for other cities struggling to revitalize their urban core through blight eradication. The proposed research would tangibly strengthen UM efforts to be a supportive partner to the City of Detroit, and would support a number of M-LEEad funding objectives.
Accomplishments
The Detroit abandoned residential dwelling demolition project was suspended during our project period in order to investigate possible contractor violations of regulations pertaining to the treatment and disposal of asbestos. Following the restart of the program, we were able to collect air samples from 25 homes that were demolished under the emergency demolition program. These structures were too dangerous to enter, and so were demolished without abatement of asbestos-containing materials (ACM), but with the application of water suppression during demolition. We were also able to sample 1 additional home which had undergone asbestos abatement. For each of these homes we were able to collect a variety of information, including the age, size, and location of the home, weather conditions during the demolition, and the total cost of demolition for the home. We collected a total of 101 air samples that were analyzed using phased contrast microscopy (PCM), the traditional analysis for air sampling, but a technique with known issues regarding accuracy. We analyzed 46 of these samples with Transmission Electron Microscopy, as well, to take advantage of the much greater accuracy associated with this method, as well as the ability to speciate the types of asbestos measured. We have nearly completed the analysis of this dataset; our analyses to date indicate that 53% of the PCM samples showed asbestos concentrations that exceeded the limit of detection. However, TEM analysis indicated the presence of only two asbestos fibers among the 46 samples analyzed by TEM, indicating that virtually all of the PCM samples that exceeded the limit of detection had detected non-asbestos materials. We are in the process of preparing a manuscript summarizing these results. In addition to collecting air samples, we were also able to randomly sample the demolition records for over 600 Detroit homes that have been or are slated to be demolished. Using this information, we have compiled a dataset of ACM in these homes, along with other pertinent information, e.g., age, size, and location of the home, as well as the estimated cost of asbestos abatement and demolition for each home. We are in the process of analyzing these data in order to estimate the total burden of asbestos present in homes being demolished as part of Detroit's demolition program; these analyses will be documented in a second manuscript. We will combine this information with the air sampling results to estimate the risk of lung cancer and mesothelioma in the community based on the emissions we've measured. Finally, we are preparing a third related manuscript. This manuscript will review the existing literature on asbestos emissions from residential demolition program.
Policy or Public Health Impacts
No policy or public health impacts to date, though we do have an excellent working relationship with the Landbank Authority of Detroit.
Publications
Grant applications and grant awards resulting from Pilot Project
We submitted a preproposal to the Department of Housing and Urban Development in May of 2017, but the preproposal was not selected for a full proposal.
“Increasing Climate Resiliency in Detroit through the Co-Analysis of Survey Data ”
Primary Investigator: Larissa Larsen
Abstract
With climate change, heatwaves are becoming more frequent, intense, and long lasting. Research shows that the health burden of heat exposure is borne unequally, with the elderly and the very young, racial/ethnic minorities, urban populations, and the socioeconomically disadvantaged being disproportionately affected. Detroit is one U.S. city where health disparities related to heat exposure have been well documented, and concern about heat and other climate related exposures, including homes flooding after extreme precipitation, has been expressed by Detroit residents. This pilot project will use a community-based participatory research (CBPR) approach to conduct new environmental health research on a key priority identified by the Stakeholder Advisory Committee to the Center’s Community Outreach and Engagement Core (COEC): research and action on preparing for climate change and minimizing its effects on health in Detroit. With guidance from the COEC, we propose to analyze existing data obtained through funding from the National Science Foundation (NSF) and the Detroit Urban Research Center (Detroit URC) to address new research questions on how housing features and access/use of energy efficiency and weatherization services may be linked with reported heatrelated health outcomes among 100 Detroit residents. This project brings investigators with training in urban planning, energy efficiency, and policy administration into a novel environmental health translational research effort with strong community engagement. The community-academic partnership includes five representatives from community groups in Detroit who, along with investigators Larsen and O’Neill, are on the steering committee of the CBPR partnership called “Heatwaves, Housing, and Health: Increasing Climate Resiliency in Detroit” (HHH). The HHH steering committee will provide guidance, consistent with CBPR principles, to the team to co-analyze and co-interpret existing survey data related to heatwaves and health in vulnerable Detroit communities. In addition, COEC member Chris Coombe will evaluate the partnership processes and outcomes of the project and make recommendations for improvement. We hope this project will result in new high quality research of relevance to Detroit communities that informs actions to increase resilience to climate change. Additionally, we expect our findings to provide support for future grant applications to support ongoing efforts in this domain.
Accomplishments
Academic partners conducted four internal workshops with project steering committee to provide direct opportunities for community-based organizations (CBOs) to engage in data analysis and co-interpretation of existing datasets focused on extreme heat and health in vulnerable Detroit communities “Exploring Heat & Health in Detroit &Developing a Common Analysis Agenda” Outcome(s): discussed what is known about heat & health in the literature; identified research questions of relevance to community partners “Identifying Action Opportunities” (9.21.17) Outcome(s): identified policy and other action opportunities that could benefit from our research; identified potential audience(s) for research findings “Overview of Directed Acyclic Graph (DAG) Co-Analysis Tool” (10.16.17) Outcome: introduced community partners to basic epidemiological concepts; continued discussion on key findings and began prioritizing action opportunities Engaging in Directed Acyclic Graph (DAG) Co-Analysis Tool”(11.9.17) Outcome: consensus on community driven research questions and DAG representing community-identified variables to be addressed Collaboratively developed a Directed Acyclic Graph (DAG) with CBO partners using survey data from 103 Detroit residents to investigate the hypothesis that the odds of reporting heat related illness (HRI) would be greater among respondents with central air-conditioning (AC) or window-unit AC versus no AC and among those with poorer self-rated health. Academic partners selected the initial confounders and, after instruction on the use of DAGs, the HHH steering committee discussed alternate DAGs and confounders to enhance the epidemiologic analysis of community-specific social and health vulnerabilities including housing and neighborhood characteristics, utility cost concerns, and age. Created an Action Opportunity Table to align research findings with existing community-defined issues and prioritize action. Collaboratively developed and conducted three focus groups to provide opportunities for direct engagement with prior study participants to disseminate research findings directly to study participants, elicit feedback on the most useful dissemination materials and approaches for community residents (including redesign of the Individual Climate Report [ICR] sent in the summer of 2017), and engage in additional dialogue on broader community environmental health issues and discuss recommended actions to protect the health of vulnerable communities as it relates to extreme heat, energy efficiency, healthy housing, and the reach and effectiveness of community-based resources and services A second evaluation of partnership process and outcomes was conducted in July 2018
Policy or Public Health Impacts
Our partnership met with director of the new Office of Sustainability in November 2017. This meeting introduced our partnership to the Office of Sustainability and facilitated discussion on potential new research questions or research products to support sustainability efforts in Detroit communities. In the summer of 2018, two undergraduate students from our project supported survey data analysis and development of town halls presentations as part of the Detroit Sustainability Action Agenda (DSAA).
Publications
Grant applications and grant awards resulting from Pilot Project
Ginsberg Center Impact Grant; Expanding the Impact of the Heatwaves, Housing, and Health Partnership; Oct 2018-Sept 2019
“Placental Epigenomics for Environmental Health Research”
Primary Investigator: Jaclyn Goodrich
Abstract
Mounting evidence suggests a role for the placenta in epigenetic reprogramming in response to maternal exposure. Consistent with this and the Developmental Origins of Health and Disease hypothesis, the placenta’s ability to adapt to varying maternal conditions is likely accomplished through epigenetic changes across the tissue. Many studies have connected placental epigenetic signatures to specific maternal disease phenotypes (e.g. obesity, gestational diabetes) and infant outcomes. While current practice utilizes whole placental tissue samples for epigenetic studies, we propose the use of a cell sorting technique that we are currently developing. It is necessary and important to develop a deconvolution algorithm for placenta tissue to apply to epigenomic analyses primarily because of its heterogeneous nature, but also because of the propensity of specific cell types to contribute more substantially to the functional significance of whole-tissue epigenomic signatures. In this pilot proposal, we will profile DNA methylation at >850,000 CpG sites in seven sorted placental cell types and develop this algorithm. We will then profile DNA methylation of 40 placenta samples that were collected from the University of Michigan Women’s Hospital as part of the Michigan Mother-Infant Pairs (MMIP) cohort. With the MMIP cohort data, which includes measurements of first trimester urinary maternal bisphenol A (BPA) and phthalate metabolites, we will identify differentially methylated CpG sites, and we will apply the cell type estimation algorithm to this analysis to more accurately evaluate the extent to which these exposures influence the placental epigenome. This proposal offers us the opportunity to combine cutting edge techniques to contribute to the burgeoning field of placental epigenomics and to establish a foundation in our understanding of maternal exposure, placental function and offspring health.
Accomplishments
The protocol for cell-sorting placenta has been optimized, and cells have been sorted from several freshly collected placenta. DNA has been isolated from 40 placenta samples of the MMIP cohort. DNA from placenta was bisulfite converted and submitted to the DNA Sequencing Core where epigenome-wide DNA methylation analysis via the Illumina Infinium MethylationEPIC platform was completed. Data quality control was completed, and 36 samples passed quality control checks. Statistical analysis to identify exposure-associated differentially methylated CpG sites and genes is nearly complete with manuscript in preparation. DNA from sorted cells will be submitted to the DNA Sequencing Core for epigenome-wide DNA methylation analysis soon.
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Several grant applications have been submitted including aspects of this work by Co-Investigators on this pilot project. Dr. Bakulski submitted a R21 to further profile placental cell types. Dr. Padmanabhan in collaboration with Drs. Goodrich, Dolinoy, and Bakulski submitted a R01 to study racial disparities in birth outcomes with included plans for mechanistic analyses (placenta epigenomics). Neither grant was funded, but re-submissions are in progress. Dr. Goodrich submitted an NIEHS Outstanding New Environmental Scientist (ONES) R01 application in February 2019, with Dr. Padmanabhan as a co-investigator, to expand this work in a larger sample size.

January 2017

“Alteration of epigenomic reprogramming of normal human breast stem cells by cadmium ”
Primary Investigator: Justin Colacino
Co-Investigator: Lada Koneva (Research Fellow, Department of Computational Medicine and Bioinformatics). Dr. Koneva will be responsible for the integrated analysis of the transcriptomic and epigenomic data generated i
Abstract
Throughout puberty and pregnancy, the breast undergoes extensive growth and remodeling driven by proliferating and differentiating stem cells. These periods are termed “windows of susceptibility” because carcinogen exposure during these times disproportionately increases later life cancer risk. Recently, the number of stem cells in a tissue has been strongly associated with cancer risk. Exposures during windows of susceptibility could influence breast cancer risk by the increasing the number of stem cells or by modifying stem cell epigenetic reprogramming during development. A major issue in assessing the carcinogenic potential of environmental exposures in human populations, however, is that the relevant exposures for cancer initiation and promotion likely happen decades before the tumor clinically manifests. Reflective of this, there are a number of known human carcinogens, including the toxic heavy metal cadmium, that have suggestive, but still inconclusive, epidemiological evidence of breast carcinogenicity. Thus, further research using novel methods is still required to establish a mechanistic link between many environmental exposures, including cadmium, and breast carcinogenesis. We have recently developed methods for the three-dimensional tissue culture of primary human breast organoids from a single breast stem cell. The formation of these organoids recapitulates the complex branching morphogenesis that occurs during breast development. We found that low dose exposure to cadmium significantly inhibited the formation of mammary branching structures in three-dimensional culture, demonstrating functional effects of cadmium on breast stem cell differentiation. The goal of this project is to understand the molecular mechanisms by which cadmium alters breast development. Our hypothesis is that cadmium exposure dysregulates epigenetic reprogramming during breast stem cell differentiation, leading to a more stem-like transcriptional profile. To test this hypothesis, the aim of this proposal is to determine the epigenetic effects of cadmium exposure during normal breast stem cell differentiation. Utilizing the M-LEEaD Omics and Bioinformatics Core Facility, we will determine the epigenomic, by enhanced reduced representation bisulfite sequencing, and transcriptomic, by RNA sequencing, effects of cadmium exposure during breast stem cell proliferation and differentiation in three-dimensional culture. These results will provide essential supporting data for a high impact publication and future NIEHS R01 grant proposal.
Accomplishments
This project was successfully completed, the data were published in Toxicological Sciences, and the preliminary data were used in Dr. Colacino's successful ONES application.
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
NIEHS ONES R01 ES028802 - Awarded 1/1/18
“Climate and cardiopulmonary health: Does excess weight enhance risk? ”
Primary Investigator: Patricia Koman
Co-Investigator: 1) Ricardo de Majo, geospatial analyst, Health Behavior Health Education, acquire and analyze data 2) Dr. Marie O&apos;Neill, Associate Professor, Epidemiology and Environmental Health Sciences, Integ
Abstract
Despite the growing prevalence of obesity and overweight worldwide, and increases in global temperatures due to climate change, evidence is lacking about the extent to which obesity or overweight status increases risk of cardiopulmonary disease from temperature exposure. Understanding links between environmental exposures and health informs efforts to prevent heart and lung disease and to increase health and longevity of the U.S. populace. This pilot project will be a first step to generating new knowledge about excess weight being related to susceptibility to the environmental exposure of high temperature. In this pilot we will acquire and link available temperature and humidity measurements from government sources to participants of the Coronary Artery Risk Development In Young Adults Study (CARDIA), using residential history. Starting in 1985, 5,115 black and white adults aged 18 to 30 years and with no previous history of cardiovascular disease were recruited from the general population in four U.S. cities. CARDIA participants have higher body mass indices (BMIs) than contemporary national samples. With the acquired data from this proposal, I will use linear mixed models to test the hypotheses that higher exposure to temperature will be associated with diminished pulmonary function and/or impaired cardiac function. I propose to measure health outcomes from records taken during clinical exams from 1985- 2010. I predict these associations will persist when controlling for other potential confounding factors at the individual level, including: age, race, sex, education, smoking status, asthma, season, study center, air pollution exposure, and calendar year. I will also determine the extent to which obesity and overweight status, indicated by BMI or waist circumference, contributes to susceptibility to the effects of temperature exposure. The role of excess weight is rarely examined in the climate change and temperature literature, in part due to limited data on this variable in administrative datasets. Adding temperature exposures to the CARDIA cohort will allow me to identify potential links among these environmental factors, early markers of pulmonary disease and susceptibility to the negative health effects of temperature by excess weight status, with important implications for environmental health policy and disease prevention.
Accomplishments
-Downloaded temperature data -Completed SAS programming for matching temperature data to CARDIA participants - Executed programming (several overnight runs) - Matched temperature exposure with CARDIA participants
Policy or Public Health Impacts
- Research will lead to publications that will allow us to test hypotheses about the linkages between temperature and cardiopulmonary outcomes in the CARDIA cohort.
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
N/A
“Evaluation of Placental Responses to Environmental Toxicants Using Metabolomic and Transcriptomic Approaches”
Primary Investigator: Rita Loch-Caruso
Co-Investigator: Sean Harris, Postdoctoral Fellow, Environmental Health Sciences Elana Elkin, Graduate Student, Environmental Health Sciences Anthony Su, Graduate Student, Environmental Health Sciences
Abstract
The contribution of environmental toxicant exposure to pregnancy outcomes such as low birth weight, preterm birth and preeclampsia is a significant public health concern. Diethylhexyl phthalate (DEHP) and trichloroethylene exposure, two toxicants commonly found in the environment, has been linked to increased risk of one or more of these outcomes in epidemiology studies. However, the specific mechanisms by which these compounds contribute to adverse birth outcomes is still largely unclear. Transcriptomics and metabolomics are powerful approaches which offer the ability to analyze biological tissues for changes in global gene expression and metabolite levels, respectively. The proposed studies will utilize transcriptomics and metabolomics in order to elucidate mechanisms of DEHP and TCE toxicity in the placenta. For these studies, we will utilize tissues from exposed mice as well as cell lines which model placental cell types (HTR-8, BeWo) that are critical in placental development and the maintenance of pregnancy. The results of these studies will provide important new insight into the mechanisms by which DEHP and TCE contribute to adverse pregnancy outcomes. These insights will inform public health officials and clinicians and allow for more effective communication regarding the risks of exposure to these compounds during pregnancy.
Accomplishments
Samples from mouse placenta and human placental cell lines exposed to the toxicants have undergone transcriptomic and metabolomic analyses. RNA-seq analysis of mRNA expression revealed 102 genes with significantly changed expression in placentas of mice exposed to DEHP. Several of these genes play significant roles in placental function and development. Analysis of short RNA transcripts (e.g., miRNA) identified approximately 80 miRNAs with significant changes in DEHP-exposed mouse placentas. Further analyses of the placental RNA data are underway. Metabolomics experiments with HTR-8/SVneo cells (human extravillous trophoblast cell line) revealed that exposure to the TCE metabolite S-1,2(dichlorovinyl)-L-cysteine (DCVC) for 6 h or 12 h significantly increased over 20 metabolites associated with cellular energy metabolism pathways in the absence of significant changes in ATP concentrations. These results suggest that the short-term DCVC exposure caused metabolic perturbations necessitating adaptations in macronutrient and energy metabolism pathway utilization in order to maintain adequate ATP levels. In targeted metabolomics experiments using the tricarboxylic acid (TCA) plus platform with BeWo cells (human villous trophoblast cell line) exposed to 10 and 20 µM DCVC, results indicate that forskolin-stimulated syncytialization and DCVC co-treatment during forskolin-stimulated syncytialization modified metabolites in important energy metabolism pathways that have established implications to pregnancy. In addition, RNA isolated from BeWo cells exposed to 0, 10, and 20 µM DCVC was submitted to the University of Michigan DNA Sequencing Core through the M-LEEaD Omics and Bioinformatics Core for PCR-based microarrays focusing on oxidative stress and NF-κB signaling.
Policy or Public Health Impacts
Ultimately, results from these studies will help inform public health officials and clinicians regarding risks of exposure to these compounds during pregnancy.
Publications
  • Elkin E, Bridges D, Loch-Caruso R. Exposure to the trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine causes compensatory changes to macronutrient utilization and energy metabolism in placental HTR-8/SVneo cells. Submitted, under review.
  • Su AL, Harris SM, and Loch-Caruso R. The trichloroethylene (TCE) metabolite S-(1,2-dichlorovinyl)-L-cysteine (DCVC) alters oxidative stress and apoptotic responses during syncytialization in a human placental cell model (BeWo). Society of Toxicology 57th Annual Meeting, San Antonio, TX, March 13, 2018. Abstract no. 1779. Poster presentation.
  • Elkin E, Bridges D, Loch-Caruso R. Poster presentation. The trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine causes an early energy metabolic shift followed by mitochondrial dysfunction in a first-trimester extravillous trophoblast cell line. Presented at the Annual Meeting of the Society of Toxicology, Baltimore, MD, March 10-14, 2019. In: 2019 Annual Meeting Abstract Supplement, Society of Toxicology, 2019. Abstract no. 1510. Poster presentation.
  • Su AL, Karnovsky A, Loch-Caruso R. The trichloroethylene metabolite S-(1,2-dichlorovinyl)-L-cysteine stimulates metabolomic changes in human placental trophoblast BeWo cells undergoing syncytialization. Presented at the Annual Meeting of the Superfund Research Program, Seattle, WA, November 18-20, 2019. Poster presentation.
  • Elkin ER, Bakulski KM, Colacino JA, Bridges D, Ruden D, Armant DR, Loch-Caruso R. Transcriptional Profiling of the tTrichloroethylene Metabolite S-(1,2-Dichlorovinyl)-L-Cysteine Revealed Activation of the EIF2α/ATF4 Integrated Stress Response in the HTR-8/SVneo Trophoblast Cell Line. Invited presentation at the Annual Meeting of the Superfund Research Program, Seattle, WA, November 18-20, 2019. Oral presentation.
Grant applications and grant awards resulting from Pilot Project
Sean Harris (PI) and Rita Loch-Caruso (Mentor). NIH/NIEHS F32, "Using Transcriptomics and Metabolomics to Identify Mechanisms and Biomarkers of Phthalate Placental Toxicity." Not funded. NIH/NIEHS P42 ES017198. (Akram Alshawabkeh, Center PI; Rita Loch-Caruso, Project #2 PI). Toxicant Activation of Pathways of Preterm Birth in Gestational Tissue (Project 2). Scored, awaiting funding decision. NIH/NIEHS P42 ES030991. (Carol Miller and Melissa Runge-Morris, Center MPIs; Rita Loch-Caruso and Kelly Bakulski, Project B2 MPIs). The Placenta in Early Pregnancy as an Integrator of Environmental Exposure with Implications for Maternal, Fetal and Child Health (Project B2). Scored, not funded.
“Impact of exposome on maternal and fetal inflammasome”
Primary Investigator: Vasantha Padmanabhan
Co-Investigator: Charles Burant, MD, PhD Robert C. and Veronica Atkins Professor of Metabolism Professor of Internal Medicine (Metabolome) Jaclyn Goodrich, PhD Research Assistant Professor of Environmental Health Scie
Abstract
Humans are exposed to a heterogeneous mixture of endocrine-disrupting chemicals (EDCs) that can impact the health of the mother and the fetus during pregnancy. Emerging evidence suggests that these EDCs alter metabolic homeostasis during pregnancy and affect fetal growth and initiate a set of physiologic responses in the fetus that predisposes the offspring to obesity and other chronic diseases later in life. However, the pathways through which this mixture of chemicals additively or synergistically impacts offspring health in humans are mostly unknown. Our research program is focused on understanding the effects of EDC mixtures on the maternal metabolic state across pregnancy and their impact on birth weight, a risk factor that influences the health of the offspring across the life course. This pilot proposal will use maternal blood samples collected during first trimester and delivery and cord blood at delivery to determine the relationship between environmental exposures and maternal and fetal inflammatory status, and birth outcomes. We have already characterized a wide spectrum of chemical exposures (phthalates, phenols, and metals) during early pregnancy and fetal life as well as maternal and newborn metabolome, lipidome, and epigenome (CHEAR support). In this pilot proposal, we propose to characterize an array of inflammatory markers in the same set of samples/subjects to determine how exposure to a heterogeneous mixture of EDCs influences maternal and fetal inflammatory status and how changes in inflammatory status relate to changes in metabolome, epigenome and birth outcomes. We hypothesize exposure to EDC mixtures during pregnancy leads to disruptions in maternal and fetal inflammatory cascade, and increases in pro-inflammatory and/or decreases in anti-inflammatory markers are associated with reduced birth weight and accompanying changes in the metabolome and epigenome. The proposed study will aid in understanding the maternal pathways through which EDCs impact fetal growth. The results and insights gained in this study will form the foundation for future research studying the effects of heterogeneous exposures to EDCs on the intrauterine environment and health outcomes that have their origins in fetal development.
Accomplishments
• The inflammatory marker measures have all been completed. Data were analyzed to relate inflammatory markers to endocrine disrupting chemical exposure biomarkers and birth outcomes. Statistical analysis relating inflammasome data to metabolomics is in process. • An abstract was accepted as a poster presentation for the Society for Reproductive Investigation (SRI) Meeting 2018 (see attached).
Policy or Public Health Impacts
Insights gained in this study will form the basis for studying the effects of heterogeneous exposures to EDCs on the intrauterine environment and health outcomes that have their origins in fetal development.
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Particulate Matter Induced DNA Methylomic Changes in Bronchial Epithelial Cells”
Primary Investigator: Steven Huang
Co-Investigator: Maureen Sartor Associate Professor of Computational Medicine and Bioinformatics, Medical School and Associate Professor of Biostatistics, School of Public Health Omics and Bioinformatics Core Lead and
Abstract
With increasing global industrialization, air pollution remains a major public health challenge in the future. Particulate matter (PM) is a principal pollutant that contributes to the development of many diseases, including asthma, COPD, cancer, and heart disease. Many of the health effects of PM have been shown to occur with low levels of exposure and involve the development of chronic diseases that do not manifest until later in life, suggesting that they exert epigenetic changes. Bronchial epithelial cells (BEC) line the airways and are the first cells of the lung in direct contact with PM. Although studies have demonstrated DNA methylation changes in blood samples of individuals exposed to PM, the ability of PM to directly cause DNA methylation changes in BECs has not been reported. In previous experiments, we have treated BEAS-2B cells, a BEC cell line, with PM2.5 obtained from Beijing, China and observed gene expression and DNA methylation changes for several candidate genes. The goal of this proposal is to utilize next-generation sequencing to identify in an unbiased fashion genome-wide DNA methylomic and gene expression changes in BECs exposed to PM2.5. We hypothesize that exposure of cells to PM2.5 results in global changes to the DNA methylome that affect the expression of many genes beyond those previously examined. We propose to submit samples for RNA-Seq and reduced representation bisulfite sequencing and utilize the M-LEEaD Omics and Bioinformatics Core to assist in analysis. Findings from these studies will elucidate the broader extent to which PM2.5 alters the epigenetic landscape of BECs, identify novel genes and pathways in BECs altered by PM that may be important to the development of chronic lung disease, and serve as the necessary starting point for future studies that investigate the mechanisms by which PM2.5 signal to cause DNA methylation changes.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Understanding the combined effects of breastmilk PCB exposure and maternal metabolic disease on infant growth outcomes ”
Primary Investigator: Brigid Ellen Gregg
Co-Investigator: Lindsay Ellsworth-Fellow in Neonataology, Department of Pediatrics Role: will be carrying out proposed sample extractions and analyses
Abstract
Exposure of infants to endocrine disrupting chemicals including polychlorinated biphenyls (PCBs) may influence long-term childhood disease risk through multi-factorial developmental programming. Research has shown adverse neuro-developmental outcomes, alterations in childhood growth patterns, and thyroid disorders associated with exposure to PCBs both prenatally in the developing fetus and postnatally through breast milk. Maternal breast milk is the recommend primary source of nutrition for infants, however the complex composition is altered by maternal health and environmental exposures. PCBs accumulate in maternal adipose tissue over a lifetime and have been associated with the development of maternal metabolic disease including obesity and gestational diabetes as well as the generation of breast milk containing PCBs. The cumulative effects of PCB breast milk exposure and maternal metabolic disease state on developmental programming for childhood disease has not been well described. PCBs have also been shown to deregulate microRNA expression, which may provide an important epigenetic biomarker. We will evaluate the association between maternal breast milk and urine PCB levels in states of maternal metabolic diseases with infant growth from birth to 2 years of age. We are recruiting mother-baby pairs from the University of Michigan with collection of breast milk and urine samples from lactating mothers at 1-2 days, 2 weeks, and 2 months post-partum. Breast milk and urine will be analyzed for PCB congeners (138, 153, 180) using gas chromatography with electron capture detection. Mammary epithelial cells from breast milk will be analyzed for microRNA content through qRT-PCR. Infant weight, length, BMI and head circumference will be measured over the first 2 years of life. We will analyze for an interaction between maternal metabolic disease, PCB levels, microRNA biomarkers and infant growth over 2 years. Current recruitment includes 34 mothers.
Accomplishments
Our work "Postnatal lactational exposure to polychlorinated biphenyls with elevated maternal body mass index" by Lindsay Ellsworth, Stephanie Liu, Harlan McCaffery, Vasantha Padmanabhan and Brigid Gregg has been presented at Pediatric Academic Society and University of Michigan Pediatric Research Symposium in spring 2019.
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

January 2016

“Are sex differences in effects of early obesogen exposure on metabolic outcomes in adolescence mediated by genome-wide DNA methylation?”
Primary Investigator: Karen Peterson
Co-Investigator: Jaclyn Goodrich, Wei Perng, Peter Song
Abstract
Metabolic syndrome affects up to 25% of US adults and 30% of obese adolescents. The prevalence continues to rise, fueled in part by trends in child obesity1. The literature on developmental origins and life course epidemiology describes sensitive periods for obesity risk2,3, but these frameworks are not systematically applied to investigations of how and why environmental exposures may perturb metabolic homeostasis during key developmental periods posited by EPA lifestage framework. Through a recently awarded Administrative Supplement (P01 ES02284403S1): High-Dimensional epigenomic and metabolomic responses to metal and EDC exposure, we now have the opportunity to examine the effect of repeated exposures to metals and EDCs in pregnancy and adolescence on novel, high-dimensional markers of metabolic homeostasis, e.g. shotgun lipidomics, in a subsample of 40 girls only from the Early Life Exposures in Mexico to ENvironmental Toxicants Project (ELEMENT) cohort, and the potential for mediation by DNA methylation using a genome-wide platform. Building on the unique availability of repeated measures of exposure in ELEMENT cohort, we propose to evaluate DNA methylation at >850,000 CpG sites throughout the genome in cord blood leukocytes from a subsample of 32 boys. The specific aims of the pilot project are to 1) identify regions of the genome that are differentially methylated by prenatal exposures using repeat exposure biomarker data for metals and EDCs during pregnancy among newborn boys and 2) examine whether differential methylation of the neonatal genome mediates the association of exposures to metals and EDCs during the sensitive in utero developmental period with high-dimensional biomarkers, e.g., untargeted metabolomics in peripuberty.
Accomplishments
DNA from cord blood leukocytes was bisulfite converted for 79 ELEMENT girls and boys. These children were followed-up and provided blood samples in early (in 2011 and 2012) and late adolescence (in 2016). DNA was also isolated and bisulfite converted from these samples to prepare for epigenome-wide analysis of DNA methylation at three critical developmental periods in boys and girls. Through collaboration with the OBIC and the UM DNA Sequencing Core, 34 matching samples from girls at birth and early adolescence were submitted for DNA methylation analysis via the Illumina Infinium MethylationEPIC BeadChip in 2016. Data pre-processing and quality control checks are complete. Following the award of a NIEHS CHEAR project (project PI: Perng), we submitted 252 additional ELEMENT DNA samples from three time points (both sexes; birth, early and late adolescence) for DNA methylation analysis via MethylationEPIC. Data pre-processing and quality control checks are complete. Data is now available for 163 unique ELEMENT children at 1-3 time points per child. Specifically, 62 boys and girls have data at all three time points, 35 have data at birth and early OR late adolescence, and 66 have data solely at late adolescence. We also completed untargeted metabolomics analyses on all children at early and/or late adolescence. Analyses are currently underway to determine sex differences and similarities with regards to: 1) relationships between early life exposures and DNA methylation (at birth and change over time as children age), 2) relationships between early life exposures and metabolomics, and 3) interplay between the epigenome and metabolome.
Policy or Public Health Impacts
Pending manuscript writing and dissemination (2017-2018)
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
We submitted four grant applications to expand and continue our work on sex differences in toxicant-induced perturbation of the epigenome and the impact on adolescent health. We submitted a proposal to NIEHS and ORWH for the program ‘Administrative Supplements for Research on Sex/Gender Differences’ (PA-16-066, PI: Peterson). Dr. Perng submitted a proposal to the NIH Common Fund RFA focused on metabolomics data (RFA-RM-15-021). These proposals were not successful. Dr. Peterson submitted a proposal to extend the life of the ELEMENT cohort and enable continued research on intergenerational epigenetics, and it was awarded in Fall 2017 (R24 ES028502). Dr. Perng also was awarded funding from the NIEHS CHEAR program to increase our sample size for multi-developmental period epigenomics and metabolomics in ELEMENT.
“Creating fine spatial resolution air pollution and temperature data for linking to Michigan birth outcomes”
Primary Investigator: Marie O’Neill
Co-Investigator: Amy Schulz, Carina Gronlund, Veronica Berrocal, Ricardo de Majo
Abstract
This pilot project proposes to generate a data set of estimated air pollution concentrations, specifically ozone and PM2.5, at a finely resolved spatial scale (census tract) that can be analyzed in conjunction with birth outcome data from the state of Michigan for the years 2000 to 2013. This birth outcome dataset has a range of demographic and clinical risk factors for birth outcomes, as well as geo-coordinates for the residential addresses of the mothers. Additionally, we propose to link this dataset with a finely spatially resolved temperature dataset. In Michigan, birth registry data from the Detroit area (1990-2001) have been evaluated and associations found between air pollution exposures (linked by zip code of maternal residence) and both small size for gestational age and preterm birth, with associations differing by pollutant and exposure window. We have since expanded that effort to link the birth registry data (again at zipcode level) to ambient temperature and humidity measurements and conducted a case-crossover analysis of apparent temperature and preterm birth, in collaboration with epidemiologists at the Michigan Department of Community Health (unpublished data). However, neither of these efforts was able to make the environmental linkages at the residential level, and were limited to the Detroit area, versus the state of Michigan. Thus, the new question we will address is: is it possible to generate a finer scale exposure dataset and link it to geocodes from this birth outcome dataset for the state of Michigan? This pilot funding will support the development of a database that combines temperature and air pollution datasets. These datasets can ultimately be linked with the geocoded birth outcomes data and other environmental and social variables. These data can be used to address questions such as: are exposures to ambient air pollutants during pregnancy associated with adverse birth outcomes in Michigan? Do these associations vary by trimester or proximity to emissions sources? Are birth outcomes associate with extreme temperature exposures during pregnancy, and do these associations vary by housing or neighborhood characteristics, including vegetation? Do impacts of air pollution/heat on birth outcomes differ for various population subgroups?
Accomplishments
Work on the temperature data is ongoing because it involves complex modeling of combined satellite imagery and temperature measures, and but we expect to have the dataset finalized in 2017.
Policy or Public Health Impacts
n/a
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
An R01 grant proposal, Research to Action: Improving Neighborhood Environments and Birth Outcomes (RAINEBO), with O'Neill and Schulz as Multi-PIs, was submitted to NIEHS. The proposal will be reviewed Feb. 2017.
“Epigenetic modifications and altered gene expression induced by source-specific air born fine particles in human lung cells”
Primary Investigator: Andrew Ault
Co-Investigator: Ying-Hsuan Lin
Abstract
It is well established that exposure to ambient fine particulate matter (PM2.5; airborne particles less than 2.5 micrometers in diameter) is associated with a broad range of adverse health effects, such as cardiopulmonary disease and metabolic syndrome (1, 2). In particular, PM2.5 has also been identified as a cancer-causing agent in humans with sufficient scientific evidence, showing an increasing risk of lung cancer with increasing levels of PM2.5 exposure (3). Organic aerosols in the atmosphere constitute a large fraction (20-90%) of ambient PM2.5 mass worldwide (4). Depending on the emission sources and formation mechanisms, organic aerosols possess distinct chemical and physical properties that ultimately lead to their differential impact on human health. Owing to the fact that organic aerosols are composed of a complex mixture of numerous individual chemical compounds, many of which are unidentified, the organic carbon (OC) constituents have to date been treated collectively as a hazardous component in PM2.5, while health effects induced by exposure to specific organic aerosol compounds through inhalation remain largely unknown. The questions we will focus on examining relate to the effects of source-specific organic aerosol constituents on differential epigenetic modifications and gene expression in human epithelial lung cells (BEAS-2B). We have cellular total RNA samples collected from exposure to source-specific organic aerosols generated in the laboratory settings. This M-LEEaD project will support the gene expression analysis and small non-coding RNA profiling to test the hypothesis that epigenetic modifications induced by exposure to organic aerosol constituents play a central role in altered gene expression.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“External proficiency testing of heavy metal assessment for epidemiologic studies in the Study of Women’s Health Across the Nation (SWAN)”
Primary Investigator: Sung Kyun Park
Co-Investigator: Stuart Batterman
Abstract
Exposure assessment is a critical part in environmental epidemiologic research. Good quality exposure data can be obtained with best practices including validated and high-quality analytical methods, qualified laboratory personnel, and strict quality assurance (QA)/quality control (QC). QC and proper laboratory techniques prevent erroneous conclusions, ensuring validity (accuracy) and reliability (precision) of exposure data. For example, for heavy metal assessment, QC includes analysis of reference materials before, during and after every analytical run, calibration standards, procedural blanks, duplicate samples, and spiked samples. Another important step to ensure the quality of exposure data is ‘external proficiency testing’. Proficiency testing enables a laboratory to achieve its validity (accuracy) compared to other laboratories measuring the same chemical. Although QC procedures are routinely conducted in the Exposure Assessment Core’s Trace Metals Laboratory, M-LEEaD Trace Metals Laboratory does not participate in external proficiency testing programs consistently and thus, external verification results are currently unavailable. Having such results will not only enhance the credibility of the Trace Metals Laboratory but improve the quality of exposure data and eventually improve environmental epidemiologic research conducted in collaboration with M-LEEaD Center’s Exposure Assessment Core. Therefore, our new question is to conduct external proficiency testing and inter-laboratory comparisons for heavy metal assessment. The testing results will enable us to ensure and increase validity and reliability of heavy metal exposure data provided by the Center’s Trace Metals Laboratory.
Accomplishments
Heavy metal exposure assessment data from an external laboratory were obtained. Inter-laboratory comparisons will be conducted as our next step.
Policy or Public Health Impacts
Given the lack of external proficiency testing result for heavy metal exposure assessment, the proposed work is critical for potential future environmental epidemiologic studies in SWAN and other study cohorts. Having external verification results will not only enhance the credibility of the Trace Metals Laboratory but improve the quality of exposure data and eventually improve environmental epidemiologic research conducted in collaboration with M-LEEaD Center’s Exposure Assessment Core.
Publications
Grant applications and grant awards resulting from Pilot Project
Two R01 grants have been awarded. R01 ES026578 (Park) 08/01/16 – 04/30/21 NIH/NIEHS “Exposure to Multipollutants and Obesity, Type-2 Diabetes and Metabolic Syndrome” R01 ES026964 (Park) 08/01/16 – 07/31/21 NIH/NIEHS “A Longitudinal Study of Endocrine Disruptor Mixtures and Reproductive Aging”
“Measurements of PBDE Levels in a population-based sample of lupus patients and controls in Southeastern Michigan”
Primary Investigator: Emily Somers
Co-Investigator: Siobán D. Harlow, Stuart Batterman
Abstract
Lupus is a complex and serious autoimmune disease of unknown etiology that, like most other rheumatic and autoimmune diseases, predominantly affects females. A model has emerged whereby genetic and environmental factors are thought to converge with estrogens and the hormonal milieu to influence lupus. There is a great need for investigation of endocrine disruptors in this context. Brominated flame retardants are increasingly recognized to have endocrine disrupting capacity, and in animal studies have been linked to immunotoxicity based on a range of immunologic parameters, even at low exposure levels. Their widespread exposure and persistence in the environment, along with strong signals of immunotoxicity, compel investigation of their effects on the human immune system at exposure levels encountered in the general population. The proposed pilot project will analyze baseline plasma specimens from the Michigan Lupus Epidemiology & Surveilance (MILES) Program Biobank, for 20 brominated flame retardants, including diphenyl ethers (PBDEs) and tetrabromobisphenol. The pilot budget will support analysis of approximately 100 plasma samples. We will statistically sample 50 cases and 50 controls from female MILES Cohort participants; lupus cases will be first stratified by disease activity to ensure that a range of activity is represented. We plan to examine the distributions of each compound in the study population, and compare these between lupus cases and controls. For compounds detected in the study population, we will assess associations between various sociodemographic factors, and clinical/laboratory features of SLE. We will also have renal markers available for cases and controls alike, which will be examined within each group in association with the target compounds. Further, within the lupus group, we will examine the association between these compounds and lupus disease activity, measured by a validated index. Risk factors for lupus activity are important to recognize, as disease flares represent immune activation and increase the risk of progression to target organ damage.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“MI-Environment: Promoting climate-related health within Michigan’s vulnerable communities”
Primary Investigator: Natalie Sampson
Co-Investigator: Trish Koman
Abstract
Climate change is an important public health issue globally, and several researchers have called for the integration of climate and health data to better inform climate change mitigation and adaptation planning decisions. With the mass proliferation of climate adaptation tools and outreach materials (e.g., www.cakex.org), local decision makers need the best available, locally relevant information to translate this environmental science to action. An overarching goal of this project will be to develop and refine multiple opportunities for various partners and diverse stakeholders to access and engage with climate-related health promotion and decision-making materials, including MI-Environment, through varied media. MI-Environment, an existing Geographic Information System visualization tool, depicts several metrics, including ensemble climate modeling projections. MI-Environment assembles readily available data into maps to allow the public to understand location and relative magnitude of climate vulnerability on selected metrics across the state. Preliminary results indicate many census tracts within Detroit and Southeast Michigan are disproportionately vulnerable to heat stress exposures. Underlying our dissemination efforts, we pursue several research questions through MI-Environment: What geographic areas are most vulnerable to climate-related environmental exposures? What factors (e.g., housing, stage of life) contribute to this vulnerability? What is the location or extent of racial or socioeconomic disparities? Working with the COEC, we seek to refine MI-Environment through feasibility testing, develop dissemination materials that reflect its preliminary findings, and work with diverse stakeholders to identify interventions that use these findings and materials to improve health, particularly for children. We propose two aims; 1) to test the feasibility and usability of the MI-Environment visualizations in partnership with the Community Outreach & Engagement Core (COEC) and in consultation with the Biostatistical Core and 2) to engage decision makers and residents of Detroit and Southeast Michigan in bi-directional dialogue about climate change and its associated health risks across the life course, with a particular focus on vulnerable populations such as infants and children, in order to exchange information, access local knowledge, and support local efforts to reduce and mitigate adverse effects of climate change.
Accomplishments
On March 24, 2016, we held a Science Café hosted by Detroit Hispanic Development Corporation (DHDC). The event was attended by 25 participants with representatives from academic, government, and community organizations, including Michigan Department of Health and Human Services, Michigan Environmental Justice Coalition, and the Michigan Department of Environmental Quality, among others. In preparation for the Science Café through a partnership between the COEC and Detroit Climate Action Collaborative, the team developed two new fact sheets. At the Science Café, we conducted preliminary feasibility testing of the MI-Environment tool. After three lightning talks and refreshments, participants engaged in an interactive session in DHDC’s computer lab offering extensive feedback on the tool and its use. Based on this feedback and statistical consultation with M-LEEaD, we refined MI-Environment. This pilot project also supported a training opportunity for three students.
Policy or Public Health Impacts
The MI-Environment visualization tool can help communities prepare for climate change and resolve inequities by identifying census tracts with the most vulnerable residents and highest potential exposures.
Publications
Grant applications and grant awards resulting from Pilot Project
University of Michigan, Graham Institute, Rapid Response pilot: MI-Environment Heat Stress Vulnerability in Michigan $2,500 Koman, Principal Investigator (2018)

January 2015

“Addressing Public Health Issues within the Detroit Blight Elimination Program: Transforming Health Data into Action and Policy”
Primary Investigator: Eden Wells, MD, MPH, FACPM
Co-Investigator: Regina Royan, Doug Strane
Abstract
Systemic lupus erythematosus (SLE) is a chronic relapsing autoimmune disease that predominantly affectswomen. The disease is associated with significant morbidity and is sometimes fatal. Genetic and environmentalfactors play a role in the etiology of this disease, and significant health disparities have been found in the riskand progression of lupus. Several environmental factors have been associated with lupus. Mercury is aubiquitous and persistent environmental toxicant known to have immunotoxic effects. Furthermore, mercuryhas been associated with increased risk of autoimmunity, and increased risk of SLE has been associated withdental professionals exposed to inorganic mercury. Defective CD4+ T cell gene expression due tohypomethylation of DNA may contribute to the pathogenesis of SLE. This proposal aims to investigate the roleof methylmercury (organic) in contributing to the epigenetic changes seen in CD4+ T cells in lupus. This will betested with 2 specific aims. Aim 1 will measure hair mercury, a biomarker of exposure to organic mercury(methylmercury; MeHg) in a population‐based cohort of lupus patients, and examine the association betweenMeHg and lupus disease activity. Aim 2 will characterize gene‐specific methylation of CD4+ T cell DNA in thepromoter regions of 3 immune‐relevant genes (CD70, KIR, and perforin) in a population‐based cohort of lupuspatients, and examine the effects of race and MeHg. Finally, Aim 3 will be to work with the CommunityOutreach and Engagement Core (COEC) to identify and pursue community engagement opportunities in order topromote retention of participants in the longitudinal Michigan Lupus Epidemiology & Surveillance (MILES)Cohort & Biobank, a population‐based lupus program in Southeastern Michigan, which will serve as thesubstrate for the proposed research.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“In Vivo Monitoring of Toxicant-Induced Placental Aging in Mice”
Primary Investigator: Rita Loch-Caruso, PhD; Steven Domino, MD, PhD
Co-Investigator: Kelly Hogan, PhD; Mark Chames, MD
Abstract
A position paper published in 2013 by the American College of Obstetricians and Gynecologists (ACOG) states that there is sufficient evidence to link toxicant exposures to untoward reproductive health effects across the life course (ACOG Committee Opinion No. 575). This ACOG statement is the first of its kind to encourage the translation of toxicological data into obstetric and gynecologic practice, and calls for research into mechanisms of toxicity that undermine pregnancy and early development. Cellular senescence is emerging as a recognized mechanism of toxicity. In the placenta, cellular senescence was recently identified as a process required for maintenance of the integrity of the maternal-fetal interface. Our studies will quantify toxicant-induced accelerated senescence in vivo in the placenta of a transgenic mouse model capable of expressing a bioluminescent protein upon induction of senescence marker p16ink4a. Visualization of bioluminescence in mouse placentae using in vivo imaging is an innovative way to study reproductive toxicants. By focusing on placenta, an organ unique to pregnancy, this proposal complements the mission of the Michigan Center on Lifestage Exposures and Disease. Similarly, by addressing life stage vulnerability and mechanisms of disease in a susceptible population, the proposal is responsive to the 2012-2017 Strategic Plan of the National Institute of Environmental Health Sciences (NIEHS) (Theme 1 and Goals 1 and 2). In addition, the novel application of a p16ink4a transgenic mouse model to visualize cellular senescence in placenta in vivo addresses the NIEHS Strategic Plan call for new exposure sensor tools and new models for systems biology research (Theme 2 and Goal 3). Moreover, the proposed work has the potential to make novel contributions to the evidence necessary for translating environmental health into the practice of obstetrics and gynecology.
Accomplishments
Our research team gained experience working with bioluminescence imaging of gestational tissues of mice. We successfully troubleshooted visualization problems for bioluminescence measurement. We successfully utilized a new low-stress technique for oral exposure of pregnant mice to di(2-ethylhexyl) phthlate (DEHP). Our results fail to support the hypothesis of DEHP-induced accelerated senescence of the placenta. Nonetheless, we showed that the DEHP exposure significantly decreased fetal weights compared with vehicle-exposed controls. We collected various tissues for subsequent study.
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Methylmercury and gene‐specific DNA methylation of CD4+ T cells in lupus”
Primary Investigator: Emily Somers, PhD, ScM
Abstract
Systemic lupus erythematosus (SLE) is a chronic relapsing autoimmune disease that predominantly affectswomen. The disease is associated with significant morbidity and is sometimes fatal. Genetic and environmentalfactors play a role in the etiology of this disease, and significant health disparities have been found in the riskand progression of lupus. Several environmental factors have been associated with lupus. Mercury is aubiquitous and persistent environmental toxicant known to have immunotoxic effects. Furthermore, mercuryhas been associated with increased risk of autoimmunity, and increased risk of SLE has been associated withdental professionals exposed to inorganic mercury. Defective CD4+ T cell gene expression due tohypomethylation of DNA may contribute to the pathogenesis of SLE. This proposal aims to investigate the roleof methylmercury (organic) in contributing to the epigenetic changes seen in CD4+ T cells in lupus. This will betested with 2 specific aims. Aim 1 will measure hair mercury, a biomarker of exposure to organic mercury(methylmercury; MeHg) in a population‐based cohort of lupus patients, and examine the association betweenMeHg and lupus disease activity. Aim 2 will characterize gene‐specific methylation of CD4+ T cell DNA in thepromoter regions of 3 immune‐relevant genes (CD70, KIR, and perforin) in a population‐based cohort of lupuspatients, and examine the effects of race and MeHg. Finally, Aim 3 will be to work with the CommunityOutreach and Engagement Core (COEC) to identify and pursue community engagement opportunities in order topromote retention of participants in the longitudinal Michigan Lupus Epidemiology & Surveillance (MILES)Cohort & Biobank, a population‐based lupus program in Southeastern Michigan, which will serve as thesubstrate for the proposed research.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Transcriptional and Post-Transcriptional Effects of Mutations and Environmental Factors Associated with Autism”
Primary Investigator: Mats Ljungman, PhD
Co-Investigator: Donna Martin, MD
Abstract
Autistic spectrum disorder (ASD) is a neuronal development disorder linked to genetic (~50%) and environmental (~50%) risk factors (Gaugler et al. 2014; Lyall et al. 2014; Rossignol et al. 2014; Sandin et al. 2014). The genetics of ASD is very complex and it is thought to involve up to 1000 genes (Willsey et al. 2013). Many ASD risk-contributing mutations, including ARID1B, CHD8, DYRK1A and SETD2, have known functions in the regulation of gene expression (O'Roak et al. 2012a; O'Roak et al. 2012b; Willsey et al. 2013; Liu et al. 2014). These genes belong to the top de novo ASD risk-contributing mutations and these mutations lead to loss of function of one allele resulting in haploinsufficiency. There is very little known about the effect of these mutations on global regulation of gene expression. In addition to the strong genetic link, exposure to specific environmental toxins such as air pollution, heavy metals and pesticides during vulnerable stages of life, such as embryogenesis and early development, have shown a very strong connection with ASD manifestation (Rossignol et al. 2014). A comprehensive view of the effects of these metals on transcriptional and post-transcriptional regulation in human cells is presently not available. In this pilot study, we will explore the functional consequences of the ASD-specific mutations in the ARID1B, CHD8, DYRK1A and SETD2 genes on transcription and RNA stability using the Bru-seq and the BruChase-seq techniques developed in our lab (Paulsen et al. 2013; Paulsen et al. 2014). We will also compare these signatures with signatures obtained by environmental toxins, here limited to the heavy metals mercury, lead and cadmium. Bru-seq and BruChase-seq provide novel information about rates of synthesis and turnover of both coding and non-coding RNA (Paulsen et al. 2013; Paulsen et al. 2014). The results will provide a comprehensive assessment of the effects of these ASD-specific mutations on transcriptional and post-transcriptional events. Furthermore, signatures obtained from exposure of cells to the heavy metals mercury, lead and cadmium will inform on the potential similarities of mechanism of action with the autism-specific mutations. We will use lymphocyte cell lines derived from ASD patients and from their unaffected siblings obtained from the Simons Foundation (SFARI) (Fischbach and Lord 2010) to monitor the effect of these mutations on transcription regulation. Furthermore, we will use the normal sibling lymphocytes for experiments assessing signatures following exposure to the heavy metals and then compare these signatures to the signatures obtained from the matched autistic patient. These studies will provide very comprehensive and novel signatures of the transcriptional and post-transcriptional effects of autism-specific mutations and the environmental risk factors mercury, lead and cadmium. These signatures will be very informative on their own but if we find commonalities between these signatures, it may lead to a more unified model of mechanisms responsible for the development of autism and promote the development of new therapeutic interventions. We will utilize the Environmental Genomics & Epigenomics Core for assistance with deep sequencing involving Bru-seq and will employ the Bioinformatics Core for assistance with data analysis.
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!

January 2014

“A Pilot Study for Multiple Pollutant Exposures and Ovarian Aging”
Primary Investigator: Sung Kyun Park, PhD
Co-Investigator: Sioban Harlow, PhD; Bhramar Mukherjee, PhD; Stuart Batterman, PhD
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Two R01 grants have been awarded. R01 ES026578 (Park) 08/01/16 – 04/30/21 NIH/NIEHS “Exposure to Multipollutants and Obesity, Type-2 Diabetes and Metabolic Syndrome” R01 ES026964 (Park) 08/01/16 – 07/31/21 NIH/NIEHS “A Longitudinal Study of Endocrine Disruptor Mixtures and Reproductive Aging”
“Epigenetic Effects of Prenatal Environmental Exposures”
Primary Investigator: Margit Burmeister, PhD; Betsy Lozoff, MD
Co-Investigator: Dana Dolinoy, PhD; John Meeker, PhD, Maureen Sartor, PhD; Peter Song, PhD; Jie Shao, MD
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“The Influence of Prostaglandin E2 and Transforming Growth Factor-ß1 on DNA Methylation Patterns in Idiopathic Pulmonary Fibrosis”
Primary Investigator: Steven Huang, PhD
Co-Investigator: Maureen Sartor, PhD
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Tissue Specific Epigenetic Alterations and Birth Outcomes Following Perinatal Bispehnol A (BPA) Exposure”
Primary Investigator: Vasantha Padmanabhan
Co-Investigator: Dana Dolinoy, PhD
Abstract
Coming soon!
Accomplishments
January 2019 Developmental impact of prenatal BPA on adipose tissue transcriptome” Sequencing is complete and data is getting analyzed. Manuscript under preparation January 2018 “Impact of exposome on maternal and fetal steroidal milieu” All steroid measures are complete. Data getting analyzed for preparing a manuscript January 2017 “Impact of exposome on maternal and fetal inflammasome ” Primary Investigator: Vasantha Padmanabhan January 2014 “Tissue Specific Epigenetic Alterations and Birth Outcomes Following Perinatal Bispehnol A (BPA) Exposure”
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
NIH/NIEHS UG3 OD023285-01 Prenatal exposures and child health outcomes: a statewide study (PI: Barone, Copeland, Elliott, Paneth, Ruden). Role: co-investigator, Total: $28,232,679, 09/01/16-08/31/23. R01 ES 030374 Transgenerational consequences of pre-conceptional and in utero exposure to real-life chemical mixtures on fertility and metabolic health. (PI: Padmanabhan/Evans/Sinclair). Total requested: $2,915,338, 09/01/19-08/31/24, Role: Administrative PI (12% effort). Reviewed June 2019. Score: 6th percentile.

November 2012

“Epigenetics and Mercury Exposure in the American Dental Association Cohort”
Primary Investigator: Niladri Basu, PhD; Dana Dolinoy, PhD
Co-Investigator: Al Franzblau, PhD
Abstract
Mercury is a potent toxic substance of concern occupationally and to public health. New studies show mercury-associated changes to cardiovascular function, though findings are inconsistent, underlying mechanisms (e.g. oxidative stress, inflammation) are unexplored, and epigenetic influences are just becoming apparent. Epigenetic modifications (e.g., DNA methylation) influence gene expression and have recently emerged as potential mechanisms of toxicity for various substances including methylmercury, though the influence of inorganic mercury or methylmercury on genome-wide DNA methylation remains unknown. This research hypothesizes that inorganic mercury (via dental amalgams) and methylmercury (via fish consumption) are associated with altered DNA methylation patterns in humans, and that this may increase understanding of mercury-associated cardiovascular effects.
Accomplishments
We recruited 442 dental professionals at the American Dental Association (ADA) Annual Session in October 2012. We measured blood pressure and collected samples (blood, hair, urine, saliva) for mercury measurement, epigenetic analyses, and cardiovascular biomarkers (e.g., lipoproteins, CRP, measured blood pressure) from participants. Mercury was quantified in blood (mean ± SD: 5.06 ± 3.96 µg/L, n= 434) and hair (0.96 ± 0.98 µg/g, n=424) with funds from the P30 Center and also in urine (1.87 ± 1.76 µg/L, n= 606 ) by collaborators at the ADA. Funds were used to interrogate epigenomic (genome-wide CpG methylation; via Illumina Infinium HumanMethylation450 BeadChip) variation on 48 participants using blood leukocyte DNA. Preliminary statistical analysis reveals CpG sites differentially methylated by blood and hair mercury levels. The top five differentially methylated regions will be followed-up in all samples (n=442 blood leukocyte DNA, n=235 matched saliva DNA) via quantitative bisulfite Pyrosequencing. This study will be one of the first epidemiological trials to relate mercury exposure to epigenetic effects and a panel of cardiovascular risk factors, with the ultimate aim of improving understanding of mercury’s cardiovascular risk.
Policy or Public Health Impacts
This study will be one of the first to relate mercury exposure to epigenetic effects in an epidemiological cohort. It will be the first to examine DNA methylation changes across the entire genome with respect to mercury exposure (both inorganic mercury and methylmercury). This study will further interrogate DNA methylation change as a mediator of relationships between mercury exposure cardiovascular health outcomes with the ultimate aim of improving understanding of mercury’s cardiovascular risk.
Publications
Grant applications and grant awards resulting from Pilot Project
Pending: K99/R00 grant application to NIEHS planned for February cycle. This grant application aims to expand beyond the P30-funded pilot work in the ADA cohort. Specifically, the investigators aim to determine correlations between two mercury species and cardiometabolic risk factors in combination with potential mediators (e.g., DNA methylation patterns) and modifiers (e.g., polyunsaturated fatty acids). Dr. Goodrich will be the PI of this mentored grant application, and Dr. Dolinoy will be her primary mentor.
“Genome-wide DNA methylation changes in Behçet’s disease monocytes”
Primary Investigator: Amr Sawalha, PhD
Abstract
Behçet’s disease (BD) is a multifactorial systemic vasculitis with significant morbidity and mortality. The pathogenesis of BD is poorly understood. In addition to the strong genetic association with HLA-B51, using a GWAS approach, we and others have recently uncovered novel genetic susceptibility loci for BD. There are also a number of known environmental triggers that contribute to the disease etiology. However, the genetic susceptibility loci for BD only explain a small proportion of its heritability, and genetic-environmental interaction in BD has not been studied. Our monocyte microarray expression data from patients with BD highlight response to infection and the interferon pathway in genes overexpressed in BD compared to controls. We hypothesize that DNA methylation changes that induce chromatin configuration favorable for interferon-regulated gene expression is important in the pathogenesis of BD. We further propose that identifying epigenetic changes in BD might explain some of the missing heritability of the disease and pave the way to identify novel pathogenic mechanisms and therapeutic targets for the disease. These hypotheses will be addressed by characterizing the DNA methylome in monocytes from patients with BD compared to controls. Epigenetic studies have never been previously explored in BD.
Accomplishments
Through funding from this pilot project, we have completed the first epigenome-wide study in BD. We examined genome-wide DNA methylation in monocytes and CD4+ T cells from a set of16 untreated male BD patients and age, sex, and ethnicity-matched controls. Additional samples were collected from 12 of the same BD patients after treatment and disease remission. Genome-wide DNA methylation patterns were assessed using the IlluminaMethylation450 array which includes over 485,000 individual CpG sites across the genome. We identified 383 differentially methylated CpG sites between BD patients and controls in monocytes and 125 differentially methylated CpG sites in CD4+ T cells. Bioinformatics analysis revealed a pattern of aberrant DNA methylation among genes that regulate cytoskeletal dynamics suggesting that aberrant DNA methylation of multiple classes of structural and regulatory proteins of the cytoskeleton might contribute to the pathogenesis of BD. Further, DNA methylation changes associated with treatment act to restore methylation differences observed between patients and controls. Indeed, among CpG sites differentially methylated before and after disease remission, there was almost exclusive reversal of the direction of aberrant methylation observed between patients and healthy controls. Taken together, our study provides strong evidence that epigenetic modification of cytoskeletal dynamics underlies the pathogenesis and therapeutic response in BD.
Policy or Public Health Impacts
This project is providing new data on epigenetic modifications in patient’s Behçet’s disease (BD), a multifactorial systemic vasculitis with significant morbidity and mortality. The data generated by this project provided insight into novel pathogenic mechanisms in BD.
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Mother and Child Environmental (Mace) Cohort – Epigenetic Changes Associated With Ambient Pollution”
Primary Investigator: Stuart Batterman, PhD
Co-Investigator: P. Reddy, PhD; N. Naidoo, PhD
Abstract
Coming soon!
Accomplishments
Coming soon!
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“The interplay of heavy metals and gut microbiome on epigenetic modification”
Primary Investigator: Chuanwu Xi, PhD; Dana Dolinoy, PhD
Co-Investigator: Christopher Faulk, PhD; Jianfeng Wu, PhD
Abstract
Chronic exposure to heavy metals even at low concentration levels can interfere with normal cellular functions leading to a wide variety of adverse health outcomes. Among heavy metals, lead is the most important as an environmental pollutant, because it is found all over the world and damages the growth of children and the health of adults. Most previous studies mainly focus on direct effect of heavy metals on cellular functions and diseases development without looking at the indirect effect mediated via gut microbiome. It is very recently suggested that interaction between heavy metals and gut microbiome are complex and the entire outcome from these interactions can affect human health and diseases development. In order to investigate these interactions, we took advantage of an ongoing project in Dr. Dolinoy’s group on epigenetic changes induced by exposure to physiologically relevant levels of lead and to study the interaction between lead and gut microbiota.
Accomplishments
Our initial data suggest that gut microbiota changed dramatically in mice offspring who were exposed perinatally to water containing 50 ppm of lead acetate (peak maternal blood lead levels [BLL] ~25 ug/dl). The short term goal of this project is to further characterize the gut microbial populations and functions in mice exposed to different concentrations of lead acetate (peak maternal BLL of ~ 3, 10, and 25 ug/dL) at different stage of life (1 month and 10 months of age) compared to mice without exposure to lead using both culture based method and DNA high throughput sequencing techniques. Statistical tools will be used to correlate potential links between key bacterial populations with markers of epigenetic changes as well as life-course metabolic phenotyping, including energy expenditure, activity patterns, body composition, and adiponectin/leptin hormone levels. The long term goal of this project will be elucidate the complex interaction between lead and human gut microbiota and its impact on health outcomes.
Policy or Public Health Impacts
This project will provide new data on interactions between lead and gut microbiome, with potential new insight into how these interactions can affect human health and diseases development, with a focus on obesity.
Publications
Coming soon!
Grant applications and grant awards resulting from Pilot Project
Chuanwu Xi and Dana Dolinoy (MPI) Pending NIH/NIEHS “Interplay of Lead and Gut Microbiome on Epigenetic Changes and Obesity” The overall objective of this project is to elucidate the interplay of perinatal lead exposure and gut microbiome on epigenetic changes and obesity in adulthood.

February 2012

“Effect of in utero BPA exposure on allergen-induced pulmonary inflammation in adult mice”
Primary Investigator: Peter Mancuso, PhD
Co-Investigator: Edmund O’Brien, PhD
Abstract
Bisphenol A (BPA), a monomer of polycarbonate plastics and epoxide resin, has been implicated in asthma pathogenesis when exposure occurs to the developing fetus. However, few studies have examined the effect of in utero and early-life BPA exposure on asthma pathogenesis in adulthood.
Accomplishments
Using an allergen-induced model of asthma, we examined whether perinatal BPA exposure alters inflammatory responses in adult offspring. BALB/c dams were randomly assigned to a diet containing 50 ng, 50 µg, and 50 mg of BPA/kg diet or a BPA-free control diet. Dams remained on the assigned diet throughout gestation and lactation until postnatal day 21 when offspring were weaned onto the BPA-free diet. Twelve-week-old offspring were sensitized to ovalbumin with alum intraperitoneally and subsequently challenged with aerosolized ovalbumin to induce an allergic inflammatory response. We observed that serum anti-ovalbumin IgE levels were elevated in the offspring exposed to the 50 µg and 50 mg doses of BPA compared with animals on the control diet. We also found an increase in the production of IL-13 and IFN-γ from ovalbumin stimulated splenocytes obtained from mice fed all diets containing BPA. Lymphocyte counts in the bronchoalveolar lavage fluid were increased in animals exposed to the 50 ng dose of BPA which was associated with and increased RANTES production. However, offspring exposed to 50 ng or 50 mg doses of BPA exhibited decreased macrophage, neutrophil, and eosinophil bronchoalveolar lavage fluid counts and decreased TNF-α, IFN-, IL-4, IL-13, cysteinyl leukotrienes, and prostaglandin D2. While we did not find that perinatal BPA exposure worsens the pulmonary inflammatory response to ovalbumin challenge in adult mice, our data suggest that early life BPA exposure may exacerbate the allergic response in adulthood by elevating systemic IgE levels.
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“In Utero Lead Exposure and Neuron-specific Epigenetic Change Associated with Life-Course Neurobehavioral Phenotypes”
Primary Investigator: Dana Dolinoy, PhD
Co-Investigator: Christopher Faulk, PhD; Howard Hu, MD, ScD; Karen E. Peterson, ScD; Brisa N. Sanchez, PhD
Abstract
Recent human post-mortem evidence indicates that frontal cortex neuronal cells have distinct DNA methylation signatures compared to non-neurons. In addition, the methylation landscape of neuronal cells varies depending on neuronal activity, based on in vivo research in adult mice. This evidence suggests that the methylome of neuronal cells may be plastic throughout the lifespan and it may be particularly sensitive to environmental conditions such as lead (Pb) exposure. Previous epigenetic epidemiology research in the brain has studied mixed cell types in whole tissue samples, but it is now clear that epigenetic analyses that target specific cell types may be more informative, particularly since neurons are known to represent approximately only a fraction of cells in CNS tissue.
Accomplishments
We are conducting a novel study of neuron-specific epigenetic signatures associated with Pb exposure in a mouse model. Using post-mortem frontal cortex tissues from 10-month old mice exposed in utero to Pb and control water, we have optimized the use of Fluorescence Assisted Cell Sorting (FACS) to separate neuronal nuclei from non-neuronal nuclei in 9 control samples and 18 Pb exposed samples. We have performed a whole-genome tiling array (Nimblegen) on DNA from neuronal nuclei from control and Pb-exposed mice. Next we will assay for differential DNA methylation in the neuronal cell population compared to non-neuronal cells in the promoter regions of four candidate genes: Adam17, Bace1, Aph1a, and Drd1a, as well as globally. This mouse work will lay the foundation for future epigenetic epidemiology studies in Alzheimer’s disease cases versus control samples, as well as ongoing studies in epigenetics, life course obesity phenotyping, and metabolic syndrome risk.
Policy or Public Health Impacts
The developmental origins of health and disease literature has emphasized perinatal environmental exposures and chronic disease in adulthood, but has failed to fully integrate life-course analysis with epigenetic approaches that carefully consider cell-type specificity. Using a well-established gestational lead exposure mouse model, this project is examining the relationship of in utero lead exposure to neuronal frontal cortex DNA methylation in four key genes involved in Alzheimer’s Disease-like pathologies and activity, as well as global methylation levels. By integrating analysis of lead exposure, frontal cortex neuronal DNA methylation patterns, and activity/energy endpoints, data from this project will illuminate mechanisms by which early environmental exposures are linked to later disease and may shed light on potential preventive approaches to reduce long-term disease risk.
Publications
Grant applications and grant awards resulting from Pilot Project
P01 ES022844 (Center PI: Peterson: Project 3 PI: Dolinoy) 04/01/13-03/31/18 NIH NIEHS/EPA “Lifecourse Exposures & Diet Epigenetics, Maturation & Metabolic Syndrome” Goals: This proposed Center seeks to understand the mechanisms by which exposures to mixtures of EDCs (bisphenol A (BPA), phthalate metabolites, lead, and cadmium) in utero and during the pubertal transition affect physical growth and maturation and lead to alterations in metabolic homeostasis, oxidative stress and contribute to risk of metabolic syndrome. R21 grant submitted for RFA: Research Linking Environmental Exposure to Neurodegenerative Disease
“Prenatal Bisphenol-A Exposure and Overeating on Metabolic Homeostasis”
Primary Investigator: Almudena Veiga-Lopez, DVM, PhD
Co-Investigator: Vasantha Padmanabhan, PhD
Abstract
Epidemiological studies point to the impact of increased BPA exposure on adult pathologies, such as metabolic syndrome. Prenatal BPA induces insulin resistance and inhibits adiponectin release in rodents. Ongoing studies show that BPA induces hyperinsulinemia in pregnant sheep, a risk factor for metabolic disease in offspring. Obesity is associated with a pro-inflammatory state and development of insulin resistance. We hypothesized that 1) prenatal BPA induces insulin resistance that correlates with changes in adiponectin and inflammatory markers in adipose tissue and 2) postnatal obesity exacerbates these effects. Pregnant sheep were treated with 0.5mg of BPA/kg BW/day (30-90 of gestation, s.c.), in corn oil. Controls received vehicle only (C). Free BPA levels averaged 0.43±0.09 and 2.62±0.52ng/ml in umbilical cord blood of C and BPA-treated [n=6/group] day 60 fetuses, within range reported in human cord blood. At ~1.5 mo of age, about half of the C and prenatal BPA-treated female offspring were overfed (OF) to achieve BW (kg) of ~25% over the maintenance-fed group (C: 71.9±2; n=11, BPA: 71.3±3; n=9, COF: 88.7±4; n=11, and BPAOF: 88.8±2; n=12; at 15 mo of age). Intravenous glucose tolerance tests (IVGTT) were performed at 15 mo of age. Subcutaneous (SC) and visceral adipose tissues were collected at 22 mo of age for assessing expression of adiponectin and the macrophage marker, CD68 by qPCR. ANOVA followed by Bonferroni posthoc test revealed that both OF groups were hyperinsulinemic vs. maintenance-fed groups (P<0.05). Other IVGTT parameters did not differ amongst groups. Adiponectin was reduced (P<0.05) and CD68 increased (P<0.05) in the visceral depot of both OF groups relative to matching maintenance-fed groups and CD68 expression lower in BPAOF compared to COF (P=0.057). In the SC depot, prenatal BPA increased CD68 expression relative to the C group (P=0.02). Impact of postnatal weight gain on adiponectin was restricted to the BPA group (BPAOF vs. BPA; P=0.02). In conclusion, prenatal BPA had no effect on insulin homeostasis. Hyperinsulinemia seen in OF females may stem from reduced adiponectin and increased CD68 expression in the visceral fat depot. The selective elevation in CD68 in the SC, but not visceral depot of BPA females, and the differential impact of weight gain on CD68 expression in the visceral depot suggests that prenatal BPA and diet may interact to modulate inflammatory mechanisms within the two adipose tissue depots, differentially.
Accomplishments
This research developed under the realm of the P30 pilot project was presented at the Annual Endocrine Society Meeting (2013) and was selected for platform presentation, press conference, and received special media attention in Annual Research Summaries Book.
Policy or Public Health Impacts
This study is contributing to our understanding of relationships between prenatal exposure to bisphenol-A (BPA), at levels seen in human maternal circulation, and the development of a sequence of responses leading to insulin resistance and type 2 diabetes, including perturbation of insulin homeostasis, fatty acid metabolism and the inflammatory cascade.
Publications
Grant applications and grant awards resulting from Pilot Project
Coming soon!
“Probing Trichloroethylene Embryotoxicity in the Mouse: Searching for Mechanistic Fidelity between Gene Expression and DNA Methylation using a Genome-Wide Expression and Tiling Array Approach”
Primary Investigator: Dana Dolinoy, PhD and Craig Harris, PhD
Co-Investigator: Bhramar Mukherjee, PhD; Jung Hei (Julie) Kim, PhD; Karilyn E. Sant, MPH
Abstract
Exposures to environmental chemicals during the prenatal stages of development have been causally linked to agent-initiated changes in susceptibility to diseases and adverse health outcomes observed later in life. A majority of these studies have focused on insults initiated during the fetal stages of development. We propose that initiation earlier in development, during the postimplantation organogenesis stages, may have similar or even greater impact on subsequent susceptibilities. Cell growth, differentiation, and the creation tissues and organs during the organogenesis period are controlled through epigenetic mechanisms that set the patterns of gene expression responsible for determination of cell fate, form, and function. Preliminary data suggests that exposure to chemicals which induce oxidative stress also alter the epigenetic patterns of global DNA methylation. Virtually nothing is currently known about the specific molecular targets, regulatory pathways, or modes of action that may be impacted by chemical agents during this critical phase of development. In order to identify relevant targets for further study and characterization we propose a pilot study to utilize global array techniques to survey changes in DNA methylation and gene expression and to compare these outcomes to determine whether common affected pathways and molecular targets can be identified. The project design will utilize organogenesis-stage mouse conceptuses grown in whole embryo culture (WEC) and exposed directly to the environmental contaminant, trichloroethylene (TCE), or one of its primary metabolites (TCA,DCVC). We hypothesize that: Primary metabolites of TCE elicit oxidative stress in the organogenesis-stage mouse conceptus resulting in alterations in DNA methylation and a subsequent shift in the pattern and extent of gene expression within common targets and pathways. The first specific aim will identify and characterize the embryotoxicity of TCE and its primary metabolites and will be used to optimize, dose, exposure times, and toxicity outcomes for subsequent experiments. TCE-induced oxidative stress will also be characterized using quantitative HPLC methods to determine redox potentials (Eh) for glutathione and cysteine redox couples. The second aim will be used to characterize the effects of TCE exposure on genome-wide DNA methylation (NimbleGen DNA methylation tiling array) and genome-wide gene expression (Affymetrix mouse genome 430 2.0-gene array). The results from these array platforms will be analyzed using best practice data mining, cluster analysis, and bioinformatics techniques to assess the overall fidelity between DNA methylation and gene expression. Where common pathways or molecular targets are identified, the potential for redox misregulation of specific elements will also be examined.
Accomplishments
The aims outlined in this proposal were modified to use the phthalate MEHP as the test compound due to some technical difficulties with the use of TCE in WEC. All of the animal exposures and tissue collections are complete. Combining resources with our Grand Challenges Explorations (GCE) grant from the Bill and Melinda Gates Foundation with the P30 pilot funds we were able to complete a gene expression array analysis (Affymetrix), DNA methylation tiling array (Nimblegen), histone methylation analysis, global DNA methylation (LUMA), Redox profiles, histiotrophic nutrition analysis, substrate utilization in one-carbon metabolism, embryonic growth, and anatomical defect characterization. Due to the large volume of data generated using these high throughput methods, our current efforts are focused on creating the bioinformatics procedures necessary to integrate and properly interpret the results of these studies.
Policy or Public Health Impacts
Coming soon!
Publications
Grant applications and grant awards resulting from Pilot Project
Focus on Histiotrophic Nutrition: Models and Mechanisms for Understanding How Diet and Environment Affect Growth and Development during Organogenesis. Agency: Bill and Melinda Gates Foundation - Grand Challenges Explorations (GCE) PI: Craig Harris Co-I: Dana C Dolinoy, Maureen Sartor
“The role of environmental toxicants in the dysregulation of microRNAs leading to amyotrophic lateral sclerosis”
Primary Investigator: Raymond Yung, MB, ChB and Eva L. Feldman MD, PhD
Co-Investigator: Brian Christopher Callaghan, MD; Claudia Figueroa-Romero, PhD; Junguk Hur, PhD
Abstract
Amyotrophic lateral sclerosis (ALS) is a terminal disease involving the progressive degeneration of motor neurons. Most cases are sporadic (sALS) with unknown causes suggesting that the etiology of sALS may not be limited to the genotype of patients, but may be influenced by exposure to environmental factors. Alterations in epigenetic modifications are likely to play a role in disease onset and progression in ALS, as aberrant epigenetic patterns may be acquired throughout life. The aim of this study was to identify epigenetic marks associated with sALS.
Accomplishments
We are examining both DNA methylation and microRNA changes in ALS. Our published work (see citation below) we observed alterations in global methylation (5mC) and hydroxymethylation (5HmC) in postmortem sALS spinal. Loci-specific differentially methylated and expressed genes in sALS spinal cord were identified by genome-wide 5mC and expression profiling using high-throughput microarrays. Concordant direction, hyper- or hypo-5mC with parallel changes in gene expression (under- or over-expression), was observed in 112 genes highly associated with biological functions related to immune and inflammation response. Integration of methylomics and transcriptomics data successfully revealed methylation changes in sALS spinal cord. In a second study related to the pilot project, we performed miRNA array profiling studies on the same postmortem sALS spinal cord tissue. We have identified correlations between differentially expressed genes (DEGs) and miRNAs with altered expression. Gene and miRNA expression was confirmed by qPCR. Experimental validation of miRNA-DEG pairs is in progress. In a third study, we are characterizing an exposure-ranked cohort of sALS and control subjects to identify sALS-related miRNAs and environmental/occupational exposures in fibroblast and/or whole blood. We anticipate that this will result in two publications in the coming months.
Policy or Public Health Impacts
This study represents an initial identification of epigenetic regulatory mechanisms in spontaneous amyotrophic lateral sclerosis which may improve our understanding of spontaneous amyotrophic lateral sclerosis pathogenesis for the identification of biomarkers and new therapeutic targets.
Publications
Grant applications and grant awards resulting from Pilot Project
Research Linking Environmental Exposure to Neurodegenerative Disease (R21) RF-ES-13-007 “miRNA dysregulation and environmental exposures in sporadic ALS” LOI submitted: October 15, 2013 Grant submitted: November 1, 2013