Faculty Research

 

Sally WenzelSally Wenzel - Department Chair

Our laboratory has focused on understanding both the subtypes of human asthma and the combination of environmental and genetic factors which drive them. Research has focused on the role of airway epithelial cells in human disease, given the position of these cells to link the environment with the host.  Work encompasses use of large epidemiologic/research databases, including those both local and national, to define patient characteristics and relationships to environmental triggers, but also to include studies of specific human immunology, both in human samples and model systems. Current pathways of interest include those related to environmental and innate lung oxidative stress, as well as their intersections with inflammation, mucins and cell death pathways.

Wenzel was chosen as the American Thoracic Society Amberson Lecturer in 2021. The Amberson Lecture recognizes exemplary professionalism, collegiality and citizenship through mentorship and leadership in the ATS community. The Amberson Lecturer is an individual with a career of major lifetime contributions to clinical or basic pulmonary research and/or clinical practice. The lecture is given in honor of James Burns Amberson, an international authority on chest disease and tuberculosis.

Firoz Abdoel Wahid

My professional journey has been defined by a fervant commitment to understanding and mitigating the impacts of environmental factors on population health, particularly within vulnerable groups, such as pregnant women and children. Originating from Suriname, I bring a unique perspective to my population health research, which centers on the intricate interplay between chemical exposures (such as pesticides, heavy metals and PFAS), climate change, as well as food security and safety. A cornerstone of my research endeavors revolves around the profound impact of nutrition on neurodevelopment, particularly among children prenatally exposed to metals like mercury and lead in Suriname. Looking ahead, my research trajectory is resolutely focused on the dynamic and pressing challenges posed by climate change, both locally and globally. I am deeply invested in deciphering the multifaceted impacts of climate change on health, such as the combined effect of heat exposure and pesticide exposure on kidney health. 

In essence, my work strives to bridge the gap between environmental health research and actionable solutions that promote better health outcomes. 
 

 

Aaron Barchowsky

Aaron Barchowsky

The primary focus of current research is investigating the cellular and molecular mechanisms underlying human blood vessel and lung diseases caused by environmental exposures to metals and chronic changes in redox status. In vivo and cell cultured-based studies focus on the molecular pathology and etiology of vascular disease caused by chronic exposure to low levels of arsenic in drinking water. The cell signaling pathways that mediate arsenic stimulated pathogenic phenotypic changes in endothelial cells are being investigated. Additional studies examine the molecular signaling mechanisms mediating gene induction and silencing in airway epithelial cells exposed to chromium. The objective of these studies is to identify the pathways through which inhaled chromium aggravates lung injury from infections and exposure to other metals.

Nesta Bortey-Sam

Nesta Bortey-Sam

My research focuses on understanding many aspects of environmental toxicology, including both the fate, bioaccumulation, phase I & II metabolism, excretion, and effects of toxic substances such as metals. Other research areas include isotope ratios and epidemiological studies investigating the association between exposure to Pb, As, and PAHs, and occurrences of chronic kidney disease and respiratory outcomes.

Hannah Covert

I provide scientific and administrative leadership for environmental health research and capacity-building projects. While diverse in topic, these projects are often grounded in community-academic partnerships that seek to advance resilience in communities facing environmental health threats and long-standing health disparities. My subject matter expertise is in community-based participatory research, environmental health literacy, climate change impacts on health, community health worker programs, and community resilience. I am skilled in evaluation, program management, and qualitative research methodologies. 

Haider Dar

Haider Dar

My research is focused on understanding/deciphering deciphering the mechanisms of lipid peroxidation triggered programmed cell death pathways- ferroptosis/necroptosis and their regulation in the context of diseases/injury imposed on tissues like lungs, gut, and brain cortex by different insults- environmental, chemical, physical or biological. Decoding these mechanisms will lead to identification of new drug intervention targets and are crucial for the development of specific drugs and diagnostic procedures. At present the focus is to explore ferroptosis in the context of host-pathogen interaction and using Pseudomonas aeruginosa as a model, investigate the pathogen induced theft-ferroptosis as a virulence mechanism particularly in immune compromised patients within hospital environment, in cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD).

Berthony Deslouches

Berthony Deslouches

The primary goal of my laboratory is to develop peptide-based antimicrobial therapeutics against multidrug-resistant bacteria. Antibiotic resistance constitutes a global health crisis, which threatens to reverse many advances in the field of medicine. In that regard, cationic antimicrobial peptides (AMPs) are a class of antimicrobial agents that are very promising therapeutics against multidrug-resistant (MDR) bacteria-related infections because of their ability to directly disrupt bacterial membranes and their lower propensity to invoke selection of resistance compared to conventional antibiotics. However, cationic peptides present several challenges related to their susceptibility to protease digestion and to their lack of activity in certain types of biological environments (e.g., divalent cations, blood). While sequence optimization or de novo engineering can help overcome some of these limitations, AMP design is currently done mainly by trial and error based on the principle of cationic amphipathicity. Thus, the future of this promising class of therapeutics depends on the ability to design AMPs for specific applications by dissecting the AMP functional motifs to uncouple their dual properties of antimicrobial activity and host toxicity. In my laboratory, we are addressing this challenge through iterative structure-function correlations to minimize peptide length required for optimal antimicrobial activities and negligible toxicity to mammalian cells, which will result in high antimicrobial selectivity index. Data are usually streamlined to select candidates that display systemic efficacy with a high therapeutic index in small animal infection models.

Peter Di

Yuanpu Di

My research aims to investigate the effects of environmental exposure on the host. We are particularly interested in infection and immunity in the lung and the associated pathophysiological response during injury, repair, and regeneration. My lab's primary research focuses on the cellular and molecular actions of environmental or occupational exposures to toxic chemicals and microorganisms that underlie the pathogenesis of chronic human lung diseases. The three main areas of my lab research are 1) elucidating antimicrobial resistance (AMR) mechanisms and developing novel antibiotics to overcome AMR; 2) studying the pathogenesis of environmental exposure and host immunity in human diseases, including asthma, CF, and COPD; and 3) investigating inflammation-associated tissue injury and remodeling and lung tumorigenesis. We research the normal and pathogenic interactions between the body's cells concerning the environment by harnessing the knowledge of lung biology, immunology, cellular and molecular biology, and state-of-the-art technologies to develop and integrate systematic and innovative approaches. 

Jim Fabisiak

James Fabisiak

My overall research mission is dedicated to the investigation of cellular mechanisms by which various environmental agents, particularly those that affect the lung, perturb cell physiology, and, thus, contribute to organ dysfunction during toxicity. Only by understanding the cellular and molecular mechanisms of toxin action can effective chemopreventive and therapeutic strategies be designed. Of primary current interest is the role of oxidative stress, not only as a mediator of cellular damage, but also as a physiologic signaling mechanism that can dictate numerous cellular responses.

Nicholas Fitz

Nicholas Fitz

Neurodegenerative diseases are complex multifactorial diseases with identified genetic determinants along with environmental influences and life-style choices. My research is focused on understating how the exposome and associated genetic factors interact to modify normal brain development, healthy brain aging and the molecular pathogenesis of neurodegeneration. APOE and TREM2 are major risk factors for Alzheimer’s disease so I am particularly interested in uncovering their role in glial function, lipid and cholesterol transport, neuroinflammation and associated pathologies. I use broad approaches with primary glial culture, transgenic mouse models, and Alzheimer’s disease patient samples and data, -omics including CHIPseq, RNAseq, single cell RNAseq, spatial RNAseq, lipidomics, in vivo microdialysis, and complex imaging.

Peng Gao

Our research is centered around understanding the impact of environmental contaminants and xenobiotics on human health, aiming to illuminate the origins of chronic and idiopathic diseases. We use cutting-edge mass spectrometry and next/third-generation sequencing techniques to analyze the exposome profiles in environmental and biological samples as well as their biological consequences in humans. Our grand vision encapsulates the goal of an all-encompassing integration of exposomics with multi-omics profiles, which aims to offer a more comprehensive and holistic perspective on the manifold ways the exposome influences human health. As an analytical chemist and exposure scientist, my expertise straddles both environmental and biomedical sciences. This multifaceted background equips us to conduct groundbreaking research in transdisciplinary fields, including but not limited to, environmental health sciences, environmental chemistry, toxicology, analytical chemistry, and metagenomics. By merging these areas, we strive to unlock novel insights and fuel innovation in the field of human health and disease.

Valerian Kagan

Valerian Kagan

Kagan’s laboratory and Center for Free Radical and Antioxidant Health has been studying redox mechanisms of physiological processes in cells and tissues as well as their aberrant changes caused by exposure to environmental factors and disease conditions. The major focus of this work is on phospholipids and their role in signaling. The Lab has developed highly sensitive and specific LC-MS based protocols for the detection, identification and quantitative analysis of oxidatively modified lipids.  With this technological advancement, the major efforts are directed towards understanding and deciphering the signaling language of peroxidized phospholipids. Among the most advanced areas of research are studies of phospholipid signals in programs of regulated death such as apoptosis, necroptosis and ferroptosis. This work resulted in the discovery of new mechanisms of cell death in acute brain injury, acute radiation syndrome, pulmonary diseases (including ARDS, asthma and bacterial pathogen/host interactions in the lung), organ transplantation. Another aspect of the current work is related to decoding of mechanisms of lipid reprogramming of innate immune cells in tumor microenvironment leading to immunosuppression of myeloid cells in cancer.  Unearthing of new enzymatic mechanisms of redox phospholipid signaling leads to the design and development of new therapeutic modalities. This work is also going on in the Lab.

Oleksandr Kapralov

I am studying the role of lipid peroxidation in the control of cell death with focus on understanding of the role of enzymatic lipid peroxidation in regulation of ferroptosis and apoptosis.

 

Rada Koldamova

Our laboratory uses broad approaches to dissect regulatory networks and to explore the role of lipid-associated genes and proteins in molecular pathogenesis of Alzheimer’s disease.

 

Iliya Lefterov

Iliya Lefterov

Current projects relate to genetically modified mouse models of Alzheimer’s disease (AD) and cholesterol metabolism. A particular focus is on liver X receptors (LXR). Their regulatory function in the brain in health and disease is being approached using complex transgenic mouse models of altered lipid metabolism. Behavioral phenotyping and histopathology are used to reveal clues of LXR-controlled regulatory networks in the brain. Age-dependent and disease-related changes in immediate early genes (IEG) response to environmental factors is the second major research theme. Molecular, pharmacological, and genetic approaches; gene profiling; and chromatin immunoprecipitation followed by massive parallel sequencing (ChIP-seq) in intact animal models of AD are being used to assess IEG-controlled signaling pathways. AD pathogenesis in those models is assessed in the context of gene-environment interactions genome-wide using high-throughput genomic and epigenetic tools, diet, and dietary manipulations.


George LeikaufGeorge Leikauf

I am investigating the functional genomics of acute lung injury, asthma, and chronic obstructive pulmonary disease. Molecular mechanisms by which air pollutants exacerbate or cause lung diseases are being studied by various strategies including genetic linkage and microarray analyses and transgenic/gene-targeted murine systems. A major research interest is uncovering the genetic basis of increased susceptibility to pulmonary epithelial injury and repair. In addition, recent studies are examining transcriptional regulation of molecular targets (e.g., surfactant proteins) altered by exposure to ozone, aldehydes, and particulate matter.

Maureen Lichtveld - Pitt Public Health Dean 

As a physician scientist and member of the National Academy of Medicine (NAM), I have over 35 years of experience in environmental public health and health disparities research. My research focuses on environmentally-induced disease, health disparities, community-based participatory research, climate and health, community resilience, environmental health policy, disaster preparedness, and public health systems. My track record is in environmental epidemiology studies with a special emphasis on persistent environmental health threats affecting health disparate communities and disaster management. For example, I serve as PI on a project which linked implementation research and research training grants designed to identify exposures to a complex mixture of developmental neurotoxicants including mercury, arsenic, lead, and selected pesticides through comprehensive dietary, environmental, and health risk assessments, and biomarker monitoring in 1200 mother-child dyads. I have extensive experience in the development, implementation and monitoring, including recruitment and retention of large environmental epidemiological cohorts (EEC). 

Tina Ndoh

My research focuses on understanding and illuminating environmental health disparities and policy mechanisms that can mitigate impacts. I have an emphasis on cumulative impacts of environmental stressors and focus my research on the two core tenets of environmental justice- meaningful engagement and fair treatment. I am currently researching air quality impacts in areas with higher environmental justice burdens and the efficacy of public engagement in the development of national air quality standards. I also study and promote engagement efforts that build environmental health literacy.

Patty Opresko

Patricia Opresko

My research focuses on telomeres at chromosome ends, which profoundly influence genome stability, life span and human health. When chromosomes lose their telomere caps the cells can no longer divide, impairing regeneration and driving degenerative diseases during aging. Loss of telomeric caps in pre-cancerous cells causes genetic alterations that hasten cancer progression. Telomeres shorten with age, but genetic and environmental factors including oxidative stress and inflammation can damage telomeres and accelerate shortening and dysfunction. My lab is investigating how these factors increase telomere loss, how damaged telomeres affect health, and how telomere damage may be prevented or repaired. We use cellular and biochemical approaches and a chemoptogenetic tool that selectively damages telomeres in living cells and model organisms. Ultimately, we hope to develop new strategies that preserve telomeres in healthy cells and delay aging-related diseases including cancer, or that conversely deplete telomeres in cancer cells to stop their proliferation.

Linda Pearce

I am conducting studies of the interactions of reactive oxygen, nitrogen, and radiation with mitochondria, particularly using microelectrodes and magnetic circular dichroism spectroscopy.

James Peterson

James Peterson

My research focuses on (1) amelioration of acute cyanide toxicity, (2) the cytotoxic effects of nitric-oxide-derived oxidants, (3) ionizing radiation-induced mechanisms of cell death, and (4) application of magneto-optical spectroscopy to the study of biological systems.

Alison Sanders

 

Wan-yee Tang

 

Alison Sanders

Alison Sanders

Our lab examines how environmental exposures during susceptible periods of life (perinatal to adolescence to pregnancy to postpartum) can impact kidney development and function that predict chronic disease. Our research uses novel methods to examine complex chemical (e.g., metals, air pollution, fluoride) and non-chemical (e.g., heat, stress, sleep) risk factors for kidney dysfunction among susceptible populations including pregnant women, children, agricultural workers as well as diverse populations with chronic kidney disease.

Winnie Tang

Wan-yee Tang

My current research focuses on deciphering how environmental stressors (chemicals and non-chemicals) and dietary factors can epigenetically alter gene activity in a life-time manner, leading to complex diseases. These epigenetic changes can persist after the exposure has stopped to cause long-lasting effects on development, metabolism, and health, sometimes even in subsequent generations. My long-term goal is to apply innovative and promising epigenetic approaches to understand the underlying mechanisms by which epigenetic changes may contribute to common diseases. Our findings may lead to the development of improved preventive measures and therapeutic strategies to reduce the burden of chronic diseases. Also, translating our scientific findings into human may provide proper disease management and lifetime recommendations to the public. Current funding/projects: 1) Explore the role of DNA hydroxymethylation in asthma pathogenesis; and 2) Understand the impact of early-life exposure to inorganic arsenic (in drinking water) on later-life asthma risk.

Vladimir Tyurin

Vladimir Tyurin

My goal is to elucidate the molecular mechanisms through which lipid metabolites regulate cellular membrane structures as well as membrane-bound complexes, particularly under conditions of oxidative/nitrosative stress.

 

Yulia Tyurina

Yulia Tyurina

My primary research is concerned with the role of free radical reactions and, more specifically, the role of lipid peroxidation in apoptosis.

 

 

Jinming Zhao

Jinming Zhao

My research interests are focused on the translational study of asthma using primary human epithelial cells as a model, particularly the role of T2/15LO1/autophagy/ferroptosis as related to asthma pathogenesis. My research interests also include the effects of respiratory virus infection in human airway such as Influenza, Rhinoviruses, and SARS-CoV-2. We are currently working on a NIH-funded project to investigate the 15LO1-dependent ferropsutotic mitochondria damage and cell death, and have published a series of key publications in prestigious journals such as Cell, PNAS and JCI.