Members

Distinguished McKnight University Professor

Department of Medicinal Chemistry

College of Pharmacy

The focus of our research is to investigate the structural basis for carcinogenic and anticancer activity of DNA- and protein-modifying agents. Synthetic methodologies are developed to prepare structurally modified nucleosides and amino acids representing carcinogen- and drug-induced DNA and protein adducts. The effects of nucleobase modifications on DNA structure and stability are determined by NMR spectroscopy, mass spectrometry, CD spectroscopy, and computer modeling of chemically altered DNA. Biological mass spectrometry techniques are employed to quantify the formation of DNA and protein adducts in vivo. These studies identify the molecular targets of exogenous and endogenous electrophiles and provide an insight into the origins of their biological activity.

Associate Professor

Department of Biochemistry, Molecular Biology & Biophysics

College of Biological Sciences

The Chen Lab at the University of Minnesota develops proteomics technologies to discover novel posttranslational modification pathways on histones and chromatin proteins. We also develop novel quantitative strategies to determine functionally significant signaling pathways and molecular mechanisms involved in epigenetic regulations. We are particularly interested in understanding how cell metabolism and cellular microenvironment regulate protein homeostasis and epigenetic gene expression through posttranslational modification pathways in the context of neuronal development and cancer.

Regents Professor and Department Head, Robert Vince Endowed Chair, McKnight Presidential Chair, and Director of Institute of Therapeutics Discovery and Development

Department of Medicinal Chemistry

College of Pharmacy

Professor Georg and her group have published over 240 scientific articles and are involved in the design, semisynthesis, total synthesis, and evaluation of biologically active agents. Current therapeutic areas include cancer, male and female non-hormonal contraception, cancer, and epilepsy. These projects require the development of synthetic methods, synthesis of natural products, and structure-activity studies aimed at improving the therapeutic efficacy of lead compounds, including natural products, and hits from high throughput screening. Interdisciplinary projects are a main focus in the Georg group, involving medicinal chemistry, biochemistry, screening, structure-based drug design, pharmacology, pharmaceutical chemistry, and reproductive biology. Active collaboration exists with several research groups at the University of Minnesota, and around the country. Professor Georg is the PI of an NICHD P50 center: “Contraceptive Discovery, Development and Behavioral Research Center” that involves six institutions. The Georg group is interested in discovering selective inhibitors for bromodomain proteins with a special emphasis on discovering selective inhibitors for BRDT for male contraception.

Associate Professor

Division of Biostatistics

School of Public Health

Dr. Guan's research focuses on statistical analysis and methodology development related to epigenome-wide association studies. His lab aims to identify novel epigenetic markers and mechanism underlying complex diseases and risk factors.

Associate Professor

Department of Pediatrics

Medical School

The Hallstrom laboratory is studying the cellular mechanisms controlling Rb/E2F induced apoptosis during normal proliferation and in cancer development. Normal cellular proliferation is tightly regulated by cell size, mitogenic stimulation, and the absence of signals that block proliferation. The retinoblastoma (Rb) protein is a pivotal regulator of entry into the cell cycle and, importantly, disruption of various components of this control pathway leads to deregulated proliferation that underlies the development of many forms of cancer. Rb regulation of cell cycle progression and tumorigenesis is dependent on its control of E2F transcription factor function, a family of proteins that control the expression of genes required for proliferation. Further work has highlighted the role of E2F proteins, particularly E2F1, in forming the link between the deregulation of Rb pathway activity and induction of p53-dependent apoptosis. E2F1 apoptosis induction is believed to serve a tumor-suppressive mechanism by eliminating cells that have sustained an oncogenic mutation which activates the Rb pathway.

Professor

Division of Epidemiology & Community Health

School of Public Health

Dr. Hammond Nelson's research program encompasses environmental exposures and genetic traits that increase cancer susceptibility and impact patient outcomes. Our current work is focused on understanding the role of inter-individual differences in immunity, as well as viral exposures, in cancer epidemiology. In addition, I am a co-leader of the Screening, Prevention, Etiology and Cancer Survivorship program in the Masonic Cancer Center.

Associate Professor

Veterinary Clinical Sciences

College of Veterinary Medicine

Research in the Kassie laboratory focuses on preclinical development of chemopreventive agents against lung cancer. Chemoprevention is a relatively new field of cancer research and seeks to reverse, suppress, prevent or delay the carcinogenic process either by blocking the development of early lesions or by inhibiting the progression to invasive cancer. Despite improvements in early detection, diagnosis and treatment of lung cancer, this disease is still the leading cause of cancer related mortality in the U.S. and worldwide. One potential approach to reduce lung cancer mortality is chemoprevention. Our laboratory follows a unique strategy to identify lung cancer chemopreventive agents having acceptable safety and efficacy profiles to warrant human clinical trials. Agents identified, based on published epidemiological or in vitro studies, as promising chemopreventive agents will be administered orally to mice, and efficacy confirmed and target organ concentrations at efficacious dose determined. Subsequently, in vitro studies will be performed, using concentrations achieved at target organ, to determine the potential mechanisms of the chemopreventive agent and identify biomarker of efficacy. This will be followed by further studies in animal models to corroborate the in vitro biomarker findings and assess the correlation between efficacy and change in biomarker level.

Associate Professor

Cancer Epigenetics & Experimental Therapeutics

The Hormel Institute

Dr. Liu’s research programs focus on a translational approach to investigate the causes and the roles of receptor tyrosine kinases and epigenetics in cancer pathogenesis and drug resistance under normal physiological or obese conditions. He has authored more than 100 publications.

Professor

Division of Epidemiology & Community Health

School of Public Health

I am an epidemiologist whose long-term research goal is to identify and characterize risk factors for cardiovascular disease, type 2 diabetes, and other chronic conditions of aging. During my career I have helped assemble large genetic epidemiologic cohorts of cardiovascular disease, investigated genetic determinants of proteins involved in inflammation, hemostasis, and cellular adhesion, researched novel risk factors for type 2 diabetes, including genetic and epigenetic determinants, and evaluated new methods and approaches in statistical genetics in collaboration with faculty in biostatistics.

Associate Professor, McKnight Presidential Fellow, and McKnight Land Grant Professor

Department of Chemistry

College of Science & Engineering

Epigenetic regulatory proteins control the process of heritable phenotypes beyond that which is encoded at the genomic level. Chemical probes for these proteins are in high demand for therapeutic regulation of disease and to understand new biology. Bromodomains are epigenetic protein modules that bind to acetyl groups on proteins including those of acetylated histones, helping to interpret or “read” the histone code. Since the first report of a nanomolar inhibitor of bromodomain Brd4 in 2010, 18 clinical trials have been initiated to test the efficacy of bromodomain inhibition in oncology. However, many other bromodomains lack specific chemical probes to validate their role in both health and disease. We have since applied our PrOF NMR method to three bromodomains, BrdT, BPTF, and Brd4, in several screening campaigns. As a new advance, we have started to study multiple bromodomains at once to develop selective inhibitors. See Bruker's online blog "The Resonance". Demonstrating our path, we recently discovered and synthesized the first small molecule inhibitor, which we named AU1, for BPTF to understand its role in regulating transcription. BPTF has recently been recognized as an oncogene in melanoma and colorectal cancer, and by providing a new chemical probe, we are establishing collaborative programs for studying this protein’s role in various cancers. A strong medicinal chemistry effort in our lab has now been established for improving on our leads for all three bromdomains.

Professor

Department of Anesthesiology

Medical School

My role in the proposed project is that of PI. I have been studying the pathobiology of chronic pain and

analgesic pharmacology for over 25 years since my PhD training in Neuroscience. As a post-doctoral trainee at

the Oregon Health and Sciences University, I was the first recipient of the John J. Bonica Post-Doctoral

Training Fellowship from the International Association for the Study of Pain. After working in both

biotechnology and academia, I joined the Faculty at McGill University in 2007 where I was promoted to

Associate Professor in 2013. I joined the Department of Anesthesiology, University of Minnesota, as a Full

Professor in June 2020. I have 7 patents, have received research funding from both NIH and the Canadian

Institutes for Health Research (CIHR), have co-authored over 80 manuscripts and was awarded the Early

Career Award from the American Pain Society. I have served as a peer reviewer for NIH, CIHR, NSF, Medical

Research Council (UK), as well as private and non-profit organizations. I have expertise in chronic pain,

neuronal plasticity, epigenetics, neuropharmacology, and immunohistochemistry and in human studies

involving participants with chronic pain.


Of greatest relevance to the UMN Epigenetics Consortium is my published and preliminary work on the role of

DNA Methylation in chronic pain. Of note, we have demonstrated that i) epigenetic control of the sparc gene in

human and rodent intervertebral discs is associated with chronic low back pain, ii) chronic pain following nerve

injury in rodents is associated with wide-spread and changes in DNA methylation in the frontal cortex and in T

cells, iii) neuropathic pain-related cortical changes in methylation can be reversed by SAMe and physical

exercise (both of these treatments attenuate behavioral signs of pain in animal models, iv) there is a DNA

methylation signature in the T cells of humans with chronic LBP vs. controls, v) DNA methylation is

reprogrammed in degenerating intervertebral discs obtained from back pain patients vs. controls and finally, vi)

prenatal stress is associated with altered DNA methylation machinery and increased pain responses following

nerve injury in subsequent adults.

Distinguished McKnight University Professor

Department of Pharmacology

Medical School

Dr. Wei's lab is interested in regulatory pathways and underlying mechanisms in diseases related to the endocrine system, innate immunity and brain/neuron functions. Key targets of investigation are: a) hormone (vitamin A) signaling pathways that involve nuclear receptors and coregulators (chromatin remodeling), and b) extra-nuclear signals for post-transcriptional events (cytoplasmic signalsome, exosome, RNA transport and translation). Major experimental systems include genetic mouse models (CRISPR-edited), as well as in vitro models using primary neuron, macrophage, adipocyte and embryonic stem cell. Integrated methodologies include molecular, biochemical, immunological and neurological techniques. The ultimate goal is to understand disease mechanism for therapeutics development. Recent emphases are placed on a) identifying new therapeutic targets of inflammatory diseases (macrophage malfunction) and neurological diseases (neuronal stress and motor neuron diseases related to neuromuscular junction defects), and b) developing small molecular therapeutics for these diseases.