137
The I nstitute for R eproductive H ealth and R egenerative M edicine IRHRM Annual Report Calendar Year 2012 The University of Kansas Medical Center 3901 Rainbow Boulevard Kansas City, KS 66160

Annual Report - The University of Kansasirsurvey/hlc2015/Annual_Reports_KUMC... · This document represents the second annual report of the newly formed Institute for Reproductive

Embed Size (px)

Citation preview

The Institute for Reproductive Health and Regenerative Medicine

IRHRM

Annual ReportCalendar Year 2012

The University of Kansas Medical Center3901 Rainbow Boulevard

Kansas City, KS 66160

1

OVERVIEW: CY2012 AND FUTURE PLANS This document represents the second annual report of the newly formed Institute for Reproductive Health and Regenerative Medicine (IRHRM) at the University of Kansas Medical Center (KUMC). Executive Vice Chancellor Barbara Atkinson established the IRHRM during the fall of 2010 with the intent of coalescing the efforts of three smaller research units engaged in overlapping research missions. The Center for Reproductive Sciences, which had its origins in the 1960s under the leadership of Gilbert Greenwald, the research arm of the Department of Obstetrics and Gynecology, and the Institute of Maternal-Fetal Biology, a group with interests in developmental and regenerative biology, were brought under the umbrella of the IRHRM. The IRHRM is organized into three centers:

i) Center for Epigenetics and Stem Cell Biology (CESCB) ii) Center for Reproductive Sciences (CRS) iii) Center for Developmental Origins of Health and Adult Disease (CDOHAD)

The goal of the IRHRM is to facilitate investigator and especially multi-investigator research initiatives in basic, translational, and clinical research directed toward reproductive health and regenerative medicine. The institute is committed to enriching the scientific and intellectual environment of its membership and enhancing the infrastructure and resources available to facilitate these endeavors. Programs in faculty development, postdoctoral training, and graduate education are integrated into the institute and will be further developed. Generation of intellectual property and community outreach will also be emphasized. Through these efforts, the institute will become the premier research unit in reproductive health and regenerative medicine. Success will be measured in terms of the profile and impact of research performed by its scientists. The IRHRM currently consists of 76 researchers at KUMC (67), KU-Lawrence (5), Kansas State University (3), and UMKC (1) representing 26 different academic departments. We anticipate growing through the addition of investigators from KUMC, KU-Lawrence, and other Kansas City area institutions. Members participate in IRHRM efforts by performing outstanding research related to the Institute’s mission, pursuing programmatic efforts with colleagues, recommending and hosting visiting scientists for our seminar series, and participating in chalk talks and scientific interactions sponsored by the Institute. Activities 1) The IRHRM supports submission of grant applications from its membership. Our administrative staff facilitates interactions between the Investigator and the Research Institute/Sponsored Programs Administration staff, including all work in Cayuse (KUMC's online grant submission system). During CY2012, the IRHRM administrative staff has assisted our members with 23 grant submissions. The IRHRM distributes information about relevant grant opportunities and has also recently established an effort to provide scientific peer review and feedback prior to grant application submissions. 2) The IRHRM facilitates programmatic research efforts. This is accomplished in several ways including our monthly center-based chalk talks (see below) and through organizing meetings for investigators with common research interests. Our leadership and administrative staff also are available to guide these focus groups through the preparation of multi-investigator grant applications. These efforts have resulted in the formation of some research focus groups:

2

i) Stem cells and trophoblast lineage development (Soares, Paul, Rumi, Vivian, Krieg, Wolfe, Albertini)

ii) Molecular regulation of erythropoiesis (Peterson, Fields, Paul, Slawson, Vivian and Fontes) iii) Obesity, pregnancy, and postnatal outcomes (Carlson, Hull, Gustafson, Colombo, M. Petroff,

Wolfe)

These potential programmatic efforts have different trajectories. Some are close to submitting multi-investigator grant applications, while others are at very early stages of the process. 3) The IRHRM is involved in organizing a number of different events that support its membership. These include:

Institute for Reproductive Health and Regenerative Medicine Seminar Series. The IRHRM supports interactions with 10-12 visiting scientist per semester. Each visiting scientist presents a lecture and meets with faculty and trainees during their visit. Greenwald Symposium in Reproduction. This event is an annual day and a half symposium that was initiated in 2004 to honor Dr. Gilbert Greenwald. The symposium provides our faculty and trainees in the reproductive sciences with an opportunity to interact with outstanding scientists. Special Lectures. The IRHRM organizes two annual lectures. The Donald C. Johnson Lecture is focused on reproduction, whereas the James L. Voogt Lecture is focused on neuroendocrinology. Each lecture brings an outstanding scientist to KUMC to lecture and visit with our faculty and trainees. We anticipate adding a third special lecture focused on stem cell biology this next year, which is being established in honor of Dr. Ivan Damjanov. Monthly Center Chalk Talks. Our "Chalk Talks" consist of informal presentations and discussions of research ideas, preliminary data, and potential specific aims for future grant applications. All three of our Centers hold monthly Chalk Talks, and scheduling is flexible to meet the investigators' needs. 4) The IRHRM oversees and maintains equipment that is located on the 3rd floor of the Hemenway Building. The equipment includes centrifuges, microscopes, film developer, etc. 5) The IRHRM manages a Reagent Store, which provides convenient and discounted laboratory reagents and supplies for members of the IRHRM and KUMC research community. 6) The IRHRM also contributes to the communication of the research accomplishments of our membership. These efforts include a website, newsletter, and interactions with the public relations unit of KUMC. Objectives for the coming year 1) Grant submissions: Our administrative efforts in supporting the submission of grants will continue. We plan to increase the number of research proposals undergoing internal scientific review prior to submission. 2) Programmatic efforts: We will continue to facilitate multi-investigator research initiatives. As the programmatic research focus groups mature, we will help them identify seed funds to assist with their collaborative efforts and prepare them for eventual grant submission.

3

3) Our seminar series, faculty and trainee chalk talks, and scientific/social interactions will continue during the upcoming year. 4) Greenwald Symposium and special lectures: The 2012 Greenwald Symposium has been set for October 2012 and the speakers and venue determined. The Johnson and Voogt Lectures have been set for the spring of 2013 and the speakers have been confirmed. A new special lecture focused on stem cell biology in honor of Ivan Damjanov, Professor, Department of Pathology, KUMC, is in the planning stages. 5) The IRHRM will continue to manage the Reagent Store and oversee and maintain shared equipment. 7) The IRHRM will also continue to communicate the research accomplishments of our membership through our website and newsletter.

4

The Center for Epigenetics and Stem Cell Biology (CESCB) Center Director: Kenneth R. Peterson, PhD CESCB Overview

Members of the Center for Epigenetics and Stem Cell Biology investigate how cells become specialized in their function. This process is referred to as cell differentiation. Cell differentiation is a hallmark of embryogenesis. During embryonic development, cells increase in number, become specialized, and organize into tissues. Some examples of cell specialization include the formation of red blood cells, which transport oxygen; muscle cells, which produce movement; and neurons, which allow us to reason. The developmental fate of an undifferentiated cell, also referred to as a stem cell, is dictated by the cell's genetic program and its interactions with its environment. Acquisition of a specific cell fate is associated with the systematic modulation of regulatory processes controlling the function of genes and proteins. Abnormalities in cell differentiation cause birth defects and lead to adult disease. Understanding molecular mechanisms controlling cell differentiation will result in the development of new strategies for the treatment of disease. These approaches will include the generation of unique drug- and cell-based therapies. The applications of these new therapeutic tools will be numerous and include potential treatments for infertility and a diverse range of debilitative diseases, such as, cancer, diabetes, liver fibrosis, stroke, heart disease, vascular and blood diseases, Alzheimer's and Parkinson's diseases, and spinal cord injury, in addition to many others.

CESCB Chalk Talks: January to December, 2012

“Updates from the Transgenic Facility”, Melissa A. Larson, Ph.D., Molecular and Integrative Medicine, January 10, 2012 “Genome Sequencing Facility-Services and Strategies”, Clark Bloomer, B.S., Genome Sequencing Facility & Microarray Facility, February 14, 2012 “Functional Analysis of Histone Demthylase Expression in Hypoxic Cancer Cells”, Adam J. Krieg, Ph.D., Obstetrics and Gynecology, March 13, 2012 “Macrophages as Progressive Factors in Polycystic Kidney Disease”, Katherine Swenson Fields, Ph.D., Anatomy and Cell Biology, April 10, 2012 “Gradient Strategies and Umbilical Cord Wharton’s Jelly Cells for Osteochondral Tissue Engineering”, Michael Detamore, Ph.D., Chemical and Petroleum Engineering, The University of Kansas, May 8, 2012 “Early Molecular Mechanisms Underlying Human Ductal Carcinoma Progression to Invasive Breast Cancer”, Fariba Behbod, PharmD, Ph.D., Pathology and Laboratory Medicine, September 26, 2012 “Molecular Switching from DCIS to Invasive Carcinoma through CCR2 Chemokine Receptor Signaling”, Nikki Cheng, Ph.D., Pathology and Laboratory Medicine, October 17, 2012 “HBO in Umbilical Cord Blood Transplantation: What have we learned?”, Omar Aljitawi, M.D., Internal Medicine, November 21, 2012

5

The Center for Reproductive Sciences (CRS) Interim Center Director: Michael J. Soares, PhD CRS Overview In seeking new avenues for translational research, the Center for Reproductive Sciences retains a dual focus on the issues of population control and treatment of human infertility. Active basic and applied programs melding experts in the areas of molecular genetics, developmental and cellular biology synergize the use of various animal models with state-of-the-art technology resources to address human reproductive health problems. Amongst these, basic research programs in gonadal physiology, gamete maturation, fertilization, pre and peri-implantation development, reproductive tract disorders, and endocrine disruptors are collaboratively integrated to investigate disease states that impact humans. Genetic and epigenetic causes of birth defects, human ARTs, ovarian cancer, paternal and maternal forms of infertility, endometriosis and uterine fibroids all represent thematic focus groups upon which the Center is designed.

CRS Chalk Talks: January to December, 2012

“Transforming Growth Factor Beta (TGfβ) Signaling in the Male Excurrent System”, Fernando Pierucci-Alves D.V.M., Veterinary Medicine, Kansas State University, January 11, 2012 “Regulating Cellular Function by the O-GIcNAc Post-Translational Modification”, Chad Slawson, Ph.D., Biochemistry and Molecular Biology, February 6, 2012 “An Update on Endometriosis Research”, Warren B. Nothnick, Ph.D., H.C.L.D., Molecular and Integrative Physiology, March 5, 2012 “The Latest Chapter in Ovarian Germ Line Stem Cells”, David F. Albertini, Ph.D., Molecular and Integrative Physiology, April 24, 2012 “Estrogens Modulate Serotonin Receptor Signaling in Rat Hypothalmus: Synergy with SSRIs”, Nancy A. Muma, Ph.D., Pharmacology & Toxicology, The University of Kansas, June 19, 2012 “Cytochrome P45017A1 as a Drug Target for Metastatic Prostrate Cancer”, Emily E. Scott, Ph.D., Medicinal Chemistry, The University of Kansas, July 24, 2012 “The Life and Death of Oocytes: Apoptosis and DNA Repair are Critical Regulators of Oocyte Number and Quality”, Karla Hutt, Ph.D., Ovarian Biology, Prince Henry’s Institute of Medical Research, Melbourne, Australia, August 10, 2012 “Changes of Large-Scale Chromatin Configuration During Mammalian Oocyte Differentiation: Role of Intercellular Coupling and Intracellular Messages”, Alberto M. Luciano, Ph.D., Reproductive and Developmental Biology Laboratory, Health, Animal Science and Food Safety, University of Milan, August 17, 2012 “Histone H4 Acetylation Changes During In Vivo Versus In Vitro Maturation of Equine Oocytes”, Federica Franciosi, D.V.M., Ph.D., Reproductive and Developmental Biology Laboratory, Health, Animal Science and Food Safety, University of Milan, August 17, 2012

6

“Multi-Gene Target Immunotherapy for Prostate Cancer”, Dev Karan, Ph.D., Urology, September 5, 2012 “RNAseq: Seq’ing Biological Insights into Tumor Heterogeneity, Molecular Carcinogenesis, and Chemotherapy Resistance”, Jeremy Chien, Ph.D., Translational Genomics, Cancer Biology, October 3, 2012 “Estrogen Receptors and the Regulation of Glucose Metabolism”, Paige Geiger, Ph.D., Molecular and Integrative Physiology, November 7, 2012 “Clinical Characteristics of Prostate Cancer and the Case for Hsp90 as a Drug Target”, Jeffrey M. Holzbeierlein, M.D., December 5, 2012 “Molecular Targets for Dietary Intervention, New Insights into the Pathogenesis of Ovarian Cancer”, Dale Buchanan Hales, Ph.D., Physiology, Southern Illinois University, December 19, 2012

7

The Center for the Developmental Origins of Health and Adult Disease (CDOHAD) Center Director: Carl P. Weiner, MD CDOHAD Overview The quality of postnatal life has its origins in the womb. Scientists in the Center for the Developmental Origins of Health and Adult Disease seek to understand how maternal physiology and pathology impact fetal development and program postnatal health and disease. Pregnancy is a well conserved process and designed to ensure the survival of the species. A specialized and highly adaptive organ derived from the embryo called the placenta orchestrates pregnancy and creates the milieu in which the fetus develops. Failures in placental adaptations to the maternal environment lead to diseases of pregnancy, such as preeclampsia, intrauterine growth restriction, and pre-term birth. In utero insults have fundamental organizational effects on the developing fetus, which affect postnatal health and susceptibility to adult disease. Cardiovascular disease, obesity, and many cancers have their origins during fetal life. Consequently, the efforts of our researchers are key to improving the health and quality of life of our species.

CDOHAD Chalk Talks: January to December, 2012

“Regulation of Calmodulin Binding Peptide, PCP4 (PEP-19), in Human Myometrium: Implications on Calcium-Calmodulin Dependent Signal Transduction and Preterm Labor”, Clifford W. Mason, Ph.D., Obstetrics and Gynecology, February 21, 2012 “DHA and the Developing Human Brain”, Susan E. Carlson, Ph.D., Dietetics and Nutrition, March 20, 2012 “Biomagnetometry: Applications for Measures of Fetal Cardiac Autonomic Control and Neurobehaviors”, Kathleen M. Gustafson, Ph.D., Neurology, Hoglund Brain Imaging Center, April 17, 2012 “Linking Maternal Nutrition and Lifestyle to Fetal Growth”, Holly Hull,Ph.D., Dietetics and Nutrition, May 22, 2012 “A New Concept: The Novel Insight into Mechanisms of Hypoxic Fetal Brain Injury Mediated by Cofilin Modification”, Yafeng Dong, Ph.D., Obstetrics and Gynecology, September 12, 2012 “The Role of Mirn451 in the Pathogenesis of Endometriosis”, Warren B. Nothnick, Ph.D., H.C.L.D, Molecular and Integrative Physiology, October 10, 2012 “Painful Consequences of Early Life Stress or Injury”, Julie A. Carlsten Christianson, Ph.D., Anatomy and Cell Biology, November 14, 2012 “New Questions Coming from DHA Supplementation Trials in Pregnancy”, Kathleen M. Gustafson, Ph.D., Neurology, Hoglund Brain Imaging Center and Susan E. Carlson, Ph.D., Dietetics and Nutrition, December 12, 2012

8

EVENTS SEMINAR PROGRAM The Institute for Reproductive Health and Regenerative Medicine Seminar Series Established in Spring 2005 as the Research Seminar Series in Cancer and Developmental Biology, this seminar program's research emphasis and focus has evolved over time and reflects the interests of the membership in reproductive health and regenerative medicine. Distinguished scientists from across the nation present their work at KUMC and meet with faculty and trainees. The seminars are held at 8:30 am on Thursdays and are sponsored in part by the Peter T. Bohan Fund at the University of Kansas Medical Center. Below we have provided a full list of our Spring 2012 – Spring 2013 seminars. “Developmental Origins of Health and Disease (DOHAD) Hypothesis: Can Environmental Exposures in Early Life Determine Disease Susceptibility in Adulthood?” Cheryl Lyn Walker,

Ph.D., A.T.S., Fellow, Texas A&M, March 1, 2012 “Functional Organization of Chromatin Over Long Distances,” Ann Dean, National Institute of

Diabetes and Digestive and Kidney Diseases, National Institutes of Health, March 15, 2012 “Foxo Transcription Factors in the Maintenance and Differentiation of the Mammalian Germline,” Diego H. Castrillion, M.D., Ph.D., UT Southwestern Medical Center, March 22, 2012

“From Ovulation to Ovarian Cancer, a Surprising Journey,” Donald C. Johnson Lecture in

Reproduction, JoAnne S. Richards, Ph.D., Baylor, March 29, 2012 “The Etiology of Ovarian Cancer: Lessons Learned from Mouse Models,” Barbara Vanderhyden,

Ph.D., University of Ottawa, April 26, 2012 “New Approaches of Gene Discovery in Reproductive Endocrinology: Use of Human Disease Models,” Inaugural James L. Voogt Lecture in Neuroendocrinology, William F. Crowley, Jr., M.D.,

Harvard, May 3, 2012 “Dynamics and Importance of Epigenetic Patterning in Germ Cells and Embryos,” Jacquetta

Trasler, M.D., Ph.D., McGill University, May 17, 2012 “Placental sFlt-1 Production – a Friend or Foe?” Nihar R. Nayak, D.V.M., Ph.D., Stanford,

September 6, 2012 “Transcriptional Regulation of Embryonic and Trophoblast Stem Cell Self-Renewal and Differentiation,” Benjamin L. Kidder, Ph.D., National Heart, Lung and Blood Institute, National

Institutes of Health, September 13, 2012 “Transcriptional Regulation of ETV-2 During Cardiovascular Development in Vertebrates,”

Ramani Ramchandran, M.D., Ph.D., Medical College of Wisconsin, September 20, 2012 “Complex Interplay between Transposable Elements and Host Genes,” Dixie L. Mager, Ph.D.,

University of British Columbia, October 18, 2012

9

“Epigenetic Regulation of the Epithelial Phenotype in Trophoblast Stem Cells,” Gary L. Johnson,

Ph.D., University of North Carolina School of Medicine, November 8, 2012 “HIF Signaling Pathway in Development and Differentiation,” Ernestina Schipani, M.D., Ph.D.,

Indiana University, December 6, 2012 “Mechanisms of Fetomaternal Immune Tolerance,” Adrian Erlebacher, M.D., Ph.D., NYU Lagone

Medical Center, December 13, 2012 “Mitochondria as Signaling Organelles,” Navdeep S. Chandel, Ph.D., Northwestern, January 10, 2013 “Emerging Roles of Tcfap2c and Brg1 During Early Embryogenesis in the Mouse,” Jason G. Knott, Ph.D., Michigan State University, March 7, 2013 “Effect of Endocrine Disrupting Chemicals on the Mammalian Ovary,”Jodi A. Flaws, Ph.D., University of Illinois, March 28, 2013 “Basal Cells and Growth Control in the Breast,” Lindsay Hinck, Ph.D., University of California, April 18, 2013 “Maternal Matrices in Fertilization and Early Development,” Jurrien Dean, M.D., NIDDK, National Institutes of Health, May 2, 2013 “Pharmacoperones: A New Therapeutic Approach Un-Folding,” James L. Voogt Lecture in Neuroendocrinology, P. Michael Conn, Ph.D., Oregon Health & Science University, May 9, 2013 “The Endometrial Stromal Cell: The Molecular Evolution of a Major Evolutionary Novelty,” Donald C. Johnson Lecture in Reproduction, Günter P. Wagner, Ph.D., Yale University, May 16, 2013 “Liver Cell Transplantation,” Markus Grompe, M.D., Oregon Health & Science University, May 30, 2013

10

SPECIAL LECTURES

Annual Donald C. Johnson Lecture in Reproduction In honor of Dr. Johnson's research career, the reproductive biology group at the University of Kansas Medical Center hosts an annual lecture in the Spring, the Donald C. Johnson Lecture in Reproduction.

2012 Donald C. Johnson Lecture in Reproduction JoAnne S. Richards, PhD

Distinguished Professor of Molecular and Cellular Biology Baylor College of Medicine

“From Ovulation to Ovarian Cancer, a Surprising Journey” March 29, 2012

2013 Donald C. Johnson Lecture in Reproduction

Günter P. Wagner, PhD Alison Richard Professor of Ecology and Evolutionary Biology

Yale University May 16, 2013

2014 Donald C. Johnson Lecture in Reproduction

Richard L. Stouffer, PhD Senior Scientist and Head

Division of Reproductive & Developmental Sciences Oregon National Primate Research Center

Professor of Obstetrics & Gynecology Oregon Health & Science University

April 3, 2014

Annual James L. Voogt Lecture in Neuroendocrinology In honor of Dr. Voogt's research career, the IRHRM at the University of Kansas Medical Center hosts an annual lecture in the Spring, the James L. Voogt Lecture in Neuroendocrinology.

2012 Inaugural James L. Voogt Lecture in Neuroendocrinology

William F. Crowley, Jr., MD Daniel K. Podolsky Professor of Medicine

Harvard Medical School Director of Clinical Research

Massachusetts General Hospital Director, Harvard Reproductive Endocrine Science Center

“New Approaches of Gene Discovery in Reproductive Endocrinology: Use of Human Disease Models”

May 3, 2012

11

2013 James L. Voogt Lecture in Neuroendocrinology

P. Michael Conn, PhD Director of the Office of Research Advocacy, Senior Scientist in Reproductive Sciences &

Neuroscience Oregon National Primate Research Center

Professor of Physiology and Pharmacology, Cell Biology and Development, and OB/GYN Oregon Health & Science University

May 9, 2013

2014 James L. Voogt Lecture in Neuroendocrinology

Andrea C. Gore, PhD Gustavus & Louise Pfeiffer Professor of Pharmacology and Toxicology

College of Medicine The University of Texas at Austin

12

SYMPOSIUM Annual Gilbert S. Greenwald Symposium on Reproduction The reproductive biology group at the University of Kansas Medical Center hosts the annual Gilbert S. Greenwald Symposium on Reproduction in honor and as a memorial to the life and research career of Gilbert S. Greenwald, Ph.D. Professor Greenwald had an illustrious career as a Distinguished Pro-fessor at the Medical Center and as an internationally recognized reproductive biologist.

9th Annual Gilbert S. Greenwald Symposium on Reproduction October 11-12, 2012

R. Michael Roberts, PhD, Keynote Lecturer

Curator’s Professor, Animal Sciences, University of Missouri-Columbia "Trophoblast from Pluripotent Stem Cells: Can Induced Pluriopotent Cells Provide a Glimpse

into a Past Pregnancy?"

Francesco J. DeMayo, PhD (Plenary) Dan L. Duncan Professor & Gordon Cain Professor of Molecular and Cellular Biology

Baylor College of Medicine "Molecular Mechanisms Involved in Preganancy Establishment and Maintenance"

Courtney Griffin, PhD

Assistant Member, Cardiovascular Biology Research Program Oklahoma Medical Research Foundation

Adjunct Assistant Professor of Cell Biology University of Oklahoma Health Sciences Center

"Transcriptional Regulation of Vascular Development by Chromatin-Remodeling Complexes"

Kyle Orwig, PhD (Plenary) Associate Professor of Obstetrics, Gynecology & Reproductive Sciences,

and Molecular Genetics and Biochemistry University of Pittsburgh

"Translating Spermatogonial Stem Cell Transplantation to the Clinic"

Michael S. Bloom, PhD Assistant Professor of Environmental Health Sciences University of Albany (SUNY), School of Public Health

"Environmental and Assisted Reproduction: Do ‘Trace’ Exposures to Toxic Elements Interfere with IVF?"

Bruce D. Murphy, PhD (Plenary)

Director of the Center for Research in Animal Reproduction Veterinary Biomedicine, University of Montreal

"Liver-Receptor Homolog-1 Rules Reproductive Processes"

Fernando Pierucci-Alves, DVM Assistant Professor of Anatomy and Physiology

College of Veterinary Medicine, Kansas State University

13

"Cellular Signaling by Transforming Growth Factor Beta in the Male Excurrent System”

Yoel Sadovsky, MD (Plenary) Director, Magee-Womens Research Institute

Elsie Hilliard Hillman Chair of Women’s Health Research Professor of OB/GYN, Microbiology and Molecular Genetics and Clinical and Translational Science

Dept. of OB/GYN and Reproductive Sciences, University of Pittsburgh "Feto-placental Defense: A Macro Role for microRNAs"

Joan Riley, PhD

Assistant Professor of Obstetrics and Gynecology Washington University

"Uterine Natural Killer Cell Activation and Development"

10th Annual Gilbert S. Greenwald Symposium on Reproduction

October 17-18, 2013 Katherine F. Roby, PhD, Chair, Organizing Committee

Martin M. Matzuk, MD, PhD, Keynote Lecturer

Stuart A. Wallace Chair and Professor of Pathology and Immunology Baylor College of Medicine, Director of Clinical Chemistry

Ben Taub General Hospital “Genetic Manipulation of the Mouse for Translational Studies in Reproductive Medicine”

Frederick vom Saal, PhD, (Plenary)

Curator’s Professor, Division of Biological Sciences, University of Missouri-Columbia “Fetal Exposure to Bisphenol A: Adverse Effects on Reproductive and Metabolic Systems and

Why the FDA is Ignoring These Findings”

Mary Hunzicker-Dunn, PhD (Plenary) Edward R. Meyer Distinguished Professor

School of Molecular Biosciences, Washington State University “Tale of How FSH Hijacks Unexpected Signaling Pathways to Regulate Gene Expression in

Granulosa Cells”

Louis J. Muglia, MD, PhD (Plenary) Professor of Pediatrics, Obstetrics & Gynecology

University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center “Preventing Prematurity: Human Evolution, Genetics, and Birth Timing”

Kartik Shankar, PhD

Assistant Professor, Arkansas Children’s Nutrition Center Dept. of Pediatrics, University of Arkansas Medical Sciences

“Maternal Obesity and Developmental Programming: A Translational Perspective”

Derek Boerboom, DVM, PhD (Plenary) Associate Professor, Centre de Recherche en Reproduction Animale

Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire Université de Montréal

14

“WNT Signaling in Ovarian Follicle Development and Function”

Aileen Keating, PhD Assistant Professor, Department of Animal Science, Iowa State University

“Phosphatidylinositol-3 Kinsae Signaling Involvement in Ovarian Xenobiotic Metabolism”

Shoukhrat Mitalipov, PhD (Plenary) Senior Scientist, Division of Reproductive & Developmental Sciences

Oregon National Primate Research Center Oregon Health & Science University

“Reproductive and Reprogramming Strategies for Treatment of mtDNA Disease”

Ryan A. Cabot, PhD Associate Professor, Department of Animal Sciences, Purdue

“Histone Methylation and Embryo Development”

OUR RESEARCHERS

15

David F. Albertini, PhD

Professor

Omar Aljitawi, MD

Assistant Professor

Udayan Apte, Ph.D.

Assistant Professor

Fariba Behbod, PharmD, Ph.D.

Assistant Professor

Justin P. Blumenstiel, PhD

Assistant Professor

Kelly A. Bosak, Ph.D., APRN

Assistant Professor

Merlin G. Butler, M.D., Ph.D., F.F.A.C.M.G

Professor

Susan E. Carlson, Ph.D.

AJ Rice Professor of Nutrition

Nikki Cheng, Ph.D.

Assistant Professor

Jeremy Chien, PhD

Assistant Director, Translational Genomics

Julie A. Carlsten Christianson, Ph.D.

Assistant Professor

John Colombo, PhD

Professor

OUR RESEARCHERS

16

Ivan Damjanov, M.D., Ph.D.

Professor

Michael Detamore, Ph.D.

Professor

Animesh Dhar, Ph.D.

Associate Professor

Luciano DiTacchio, PhD

Assistant Professor

Yafeng Dong, Ph.D.

Research Associate Professor

Leigh M. Eck, M.D.

Associate Professor

Patrick E. Fields, Ph.D.

Assistant Professor

Timothy A. Fields, M.D., Ph.D.

Associate Professor

Katherine Swenson Fields, Ph.D.

Research Associate Professor

Sarah Finocchario Kessler, Ph.D., M.P.H.

Assistant Research Professor

Paige Geiger, PhD

Assistant Professor

Matthew C. Goering, PhD, HCLD

Director of Clinical Embryology, The Center for

Advanced Reproductive Medicine

OUR RESEARCHERS

17

Kathleen M. Gustafson, Ph.D. Research

Assistant Professor

Jeffrey M. Holzbeierlein, M.D.

John W. Weigel Endowed Associate Professor

Holly Hull, Ph.D.

Assistant Professor

Tomoo Iwakuma, M.D., Ph.D.

Associate Professor

Rajasingh Johnson, M.Phil., Ph.D., HCLD

Assistant Professor

Dev Karan, Ph.D.

Assistant Professor

Sarah L. Kieweg, Ph.D.

Assistant Professor

S. Samuel Kim, M.D., FACOG

Associate Professor

Gregory S. Kopf, Ph.D.

Associate Vice Chancellor for Research

Administration

Adam J. Krieg, Ph.D.

Assistant Professor

Sacha A. Krieg, M.D., Ph.D., FACOG

Assistant Professor

Melissa A. Larson, Ph.D.

Research Assistant Professor

OUR RESEARCHERS

18

Gene Lee, M.D.

Assistant Professor

Joan Lewis-Wambi, PhD

Assistant Professor

Benyi Li, Ph.D.

Associate Professor

Xiaogang Li, PhD

Associate Professor

Ann Manzardo, Ph.D., M.S.C.R.

Assistant Professor

Clifford W. Mason, Ph.D.

Research Assistant Professor

Nancy A. Muma, PhD

Professor and Chair

Ajay K. Nangia, M.B.B.S.

Associate Professor

Warren B. Nothnick, Ph.D., H.C.L.D.

Professor

Arindam Paul, PhD

Research Assistant Professor

Soumen Paul, Ph.D.

Associate Professor

Kenneth R. Peterson, Ph.D.

Professor and Director, CESCB

OUR RESEARCHERS

19

Brian K. Petroff, D.V.M., Ph.D.

Associate Professor

Fernando Pierucci-Alves, DVM

Research Assistant Professor

Evelyn A. Reynolds, M.D.

Assistant Professor

Katherine F. Roby, Ph.D.

Research Associate Professor

M.A. Karim Rumi, M.B.B.S., M.S., Ph.D.

Research Associate Professor

Irfan Saadi, Ph.D.

Assistant Professor

Bruce Schultz, PhD

Professor

Chad Slawson, Ph.D.

Assistant Professor

Peter G. Smith, Ph.D.

Professor

Michael J. Soares, Ph.D.

Director, IRHRM

Russell H. Swerdlow, M.D.

Professor

Paul F. Terranova, Ph.D.

Professor and Associate Vice Chancellor for

Research

OUR RESEARCHERS

20

Patricia A. Thomas, MD, MA, FACP, FASCP

Professor

J. Brantley Thrasher, M.D., F.A.C.S.

Professor and the William L. Valk Chair

Jay L. Vivian, Ph.D.

Research Associate Professor

Jinxi Wang, M.D., Ph.D.

Harrington Distinguished Professor

Carl P. Weiner, M.D., M.B.A.

Associate Director, IRHRM; Director, CDOHAD

Mark L. Weiss, Ph.D.

Professor

Michael W. Wolfe, Ph.D.

Associate Professor

Thomas M. Yankee, Pharm.D., Ph.D.

Associate Professor

Alan S. L. Yu, M.B., B.Chir.

Harry Statland and Solon Summerfield

Professor of Medicine

Xuan Zhang, PhD

Research Assistant Professor

Bao-Ting Zhu, PhD

Professor

Hao Zhu, PhD

Associate Professor

21

David F. Albertini, Ph.D, Professor Department of Molecular and Integrative Physiology Member, Center for Reproductive Sciences Our laboratory employs genetic, molecular and imaging strategies to study basic aspects of the process of reproduction that bear on human disease and its clinical management by stem cell therapy. The overall emphasis is on Women's Health in relation to causes of human infertility, ovarian cancer, and the deployment of Assisted Reproductive Technologies (ARTS) for improving egg and embryo quality in human and animal models. Three project areas are actively under study:

1. Fertility Preservation-ameliorating the loss of fertility experienced by women undergoing radiation or chemotherapy is the goal of this research. Using our long standing interest in signaling between the somatic and germ cell components of the ovarian follicle, we have initiated projects that focus on (1) understanding the mechanisms that underlie DNA damage and repair in oocytes following chemotherapy or radiation induced damage; and (2) implementation of cryopreservation strategies for oocytes and ovarian tissues that could subsequently be used for embryo production.

2. Cell Cycle Regulation In Oocytes and Embryos-how modifications in cell cycle checkpoint control ensure chromosome balance during meiosis in oocytes and mitosis in embryos is investigated by biochemical, live cell imaging, and pharmacological approaches that permit assessment of genomic integrity during cell cycle progression. This strategy is used to understand the effects of maternal aging and/or environmental chemicals (endocrine disruptors) on oocytes or embryos produced by ARTs (in vitro maturation, in vitro fertilization).

3. Stem Cell Biology- stem cells derived from adult, embryonic, or iPS sources hold great promise for providing new insights into the origins of human disease and strategies for treatments. Improvements in derivation and maintenance of stem cell lines are needed for the utility of genetically stable cells capable of realizing these promises. Our lab studies human and rat embryonic stem cells by focusing on culture conditions that assure genetic stability during proliferation and differentiation into neural progenitors. We are exploring the role of the Notch signaling pathway in an effort to understand regulation of apoptotic, autophagic, and necrotic pathways mediated by the microtubule cytoskeleton.

Editorial and Grant Reviews Editor-in Chief, Journal of Assisted Reproduction and Genetics Section Editor, 4th Edition Knobil “Physiology of Reproduction” Ad hoc reviewer, Nature, Science, PNAS Grant reviewer, NIH Seminars Presented 2012 - “Testing the genetic integrity of oocytes derived from stem cells,” Ovarian Club II, Prague 2012 – “Cell cycle control in human embryos-Why all the aneuploidy?” Ovarian Club II, Prague 2012 - “From within and without: how ovarian somatic cells influence oocyte quality during aging,” NIH-ASRM

Joint Workshop on the Ovarian Reserve, San Diego 2012 - “Overview of cryobiology in the field of fertility preservation,” Big Chill Symposium, American Society for

Reproductive Medicine, San Diego 2012 - “Follicle quality and chances to pregnancy: The Researcher’s Viewpoint,” Serono Symposium Honoring

Jacques Donnez, Brussels, Belgium

22

2012 - “Coordinating oogenesis and folliculogenesis,” Japanese Society for Fertility and Infertility, Osaka,

Japan 2012 - “Maintaining follicle integrity after cryopreservation and culture,” Symposium on Basic Science and

fertility Preservation, Asian South Pacific Initiative for Reproduction and Embryology (ASPIRE) 2nd Annual Meeting, Osaka, Japan

2012 - “Human oocyte cryopreservation-the science behind the technology,” Society for the Study of

Reproduction Annual Meeting, Penn. State University, Happy Valley PA 2012 - “Coordinating oogenesis and folliculogenesis to obtain high quality oocytes,” “Cell cycle checkpoint

control in oocytes and embryos,” and “Imaging strategies for gametes and embryos,” Visiting Professor Lectureship Series in Reproductive Physiology, University of Sassari, College of Veterinary Medicine, Sardinia, Italy

2012 - “Maintaining genome integrity in mammalian oocytes,” Dept. Anatomy and cell Biology, New York

medical College, Valhalla, NY 2012 - “Should elective oocyte cryopreservation be offered to young women,” and “The basic science of oocyte

cryopreservation,” New England Fertility Society, Portland ME 2012 - “Genome integrity in mammalian oocytes,” ESHRE Workshop on Oogenesis and Folliculogenesis,

Stresa Italy 2012 - ”New perspectives on genetic stability in mammalian oocytes,” Reproduction Medicine Associates,

Department of Obstetrics and Gynecology, Rutgers University, Morristown, NJ 2012 - “Empowering female germ cells with developmental potential,” Department of veterinary Science,

Cornell University, Ithaca, NY 2012 - “Oocyte stem cells-An update”, NYU Fertility Center, Department of Obstetrics and Gynecology, NYU

Medical Center, New York, NY 2012 - “Determinants of oocyte quality for fertility preservation-beyond the big chill,” The Oncofertility

Consortium Virtual Grand Rounds, Northwestern University, Chicago IL 2012 - “How do human oocytes maintain genetic stability?” Grand Rounds, The Center for Human

Reproduction, New York, NY Omar Aljitawi, M.D. Assistant Professor Department of Internal Medicine, Division of Hematology/Oncology Blood and Marrow Transplantation Member, Center for Epigenetics and Stem Cell Biology Dr. Aljitawi is interested in exploring the interaction of stem cells with their microenvironment and in utilizing this interaction in expanding umbilical cord blood stem cells, in improving umbilical cord blood homing post- transplant, and in developing a three dimensional leukemia and myeloma in vitro models for chemotherapy testing. Dr. Aljitawi also has been studying Wharton's jelly matrix as a scaffolding material for tissue regenerative applications like bone and cartilage regeneration.

23

Meetings Attended 2012 – “A 3-Dimensional Co-Culture Model to Investigate Adhesion-Mediated Drug Resistance in Multiple

Myeloma,” 54th ASH Annual Meeting, Atlanta, GA Committees KUMC Member, Cancer Center data and safety monitoring board (DSMB), Cancer Center protocol development and

monitoring committee (PRMC) Udayan Apte, Ph.D. Assistant Professor Department of Pharmacology, Toxicology and Therapeutics Member, Center for Epigenetics and Stem Cell Biology Dr. Udayan Apte’s research is focused on understanding the basic mechanisms of hepatocyte proliferation and applying them to develop novel therapies for acute liver failure and hepatocellular cancer. Liver is exposed to a number of drugs and toxic chemicals due to its anatomical and physiological role and is prone to drug-induced acute liver failure (ALF). ALF is a common and growing clinical problem, with liver transplantation as the only viable treatment option. Recent studies indicate that stimulating regeneration in the ALF patients may have immense therapeutic potential. However, the detailed mechanisms of liver regeneration following acute liver failure are unknown. Similarly, currently there are no reliable biomarkers to detect innate liver regeneration in ALF patients. Dr. Apte’s laboratory is investigating the mechanisms of liver regeneration and exploring novel biomarkers of liver regeneration following acute liver failure using acetaminophen overdose, the most common cause of ALF in the US, as a model system. Another aspect of liver regeneration that we understand very little about is the mechanisms of termination of liver regeneration. It is known that liver regrowth following surgical resection is tightly regulated and liver size is precisely maintained. However, the pathways that terminate liver regeneration and regulate liver size are not completely clear. Dr. Apte’s laboratory is exploring novel pathways involved in termination of liver regeneration. Dr. Apte is also testing the hypothesis that signaling pathways that terminate liver regeneration following PHX are dysfunctional during pathogenesis of hepatocellular carcinoma (HCC), the most common hepatic malignancy. The specific pathways under investigation in Dr. Apte’s laboratory include hepatocyte nuclear factor-4alpha

(HNF-4 ), Wnt/ -catenin signaling and the Hippo Kinase signaling pathway. They are also interested in identifying the role of epigenetic changes regulated by these pathways associated with hepatocyte proliferation. Meetings Attended 2012 – Experimental Biology, San Diego, CA 2012 – American Association for the Study of the Liver Diseases, Boston, MA 2012 – FASEB Summer Conferences in Liver Biology, Snowmass, CO

24

Committees Departmental Member, Seminar Committee Editorial and Grant Reviews Ad hoc reviewer, Toxicology, Toxicological Sciences, Toxicology and Applied Pharmacology, Liver

International, American Journal of Pathology, Hepatology Seminars Presented 2012 - “FXR and Bile Acids: Role in Liver Regeneration and Pathogenesis of Hepatocellular Carcinoma,”

Experimental Biology (ASIP section) meeting, San Diego, CA Trainees Chad Walesky – Graduate Student Bharat Bbushan – Graduate Student Joshua Cleveland – Summer Medical Student Ashu Agerwal – Summer Student Fariba Behbod, PharmD., Ph.D. Assistant Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology The research in our laboratory is focused on the understanding of molecular mechanism underlying human ductal carcinoma in situ progression to invasive disease. Meetings Attended 2012 – Mammary Gland Biology Gordon conference, Italy, Berga 2012 – San Antonio Breast Cancer Symposium, San Antonio, Texas 2012 – Daniel Medina Symposium, Galveston, Texas Committees KUMC Co-Leader, KUCC Cancer Prevention Program, Stem Cell Biology and Biomarker Focused Group

Member, KUMC MD/PhD Admission Committee

Editorial and Grant Reviews Guest Editor, Journal of Mammary Gland Biology and Neoplasia

Scientific Review Panel Member, SRB/NIEHS/NIH, NIH/NCI Career Development

25

Seminars Presented

2012 – “An In Vivo DCIS model,” 28th International Association for Breast Cancer Research Breakthrough

Breast Cancer Conference. Manchester, UK

2012 – “Role of miR146b during normal mammary gland development and breast cancer,” National Cancer Institute

Trainees Hanan Elsarraj – Graduate Student Shane Stecklein – MD/PhD Student Marcus Hook – Medical Student Whitney Michaels – Medical Student Justin P. Blumenstiel, Ph.D. Assistant Professor Department of Ecology and Evolutionary Biology-Univ. of Kansas Member, Center for Epigenetics and Stem Cell Biology Genome Evolution, Transposable Elements, RNA silencing, Epigenetics, and Molecular Evolution

My research is focused on understanding how genetic conflict shapes the evolution of systems of inheritance. Meiosis and sexual reproduction are prevalent across the tree of life, but they can be exploited by genetic parasites in ways that harm the host. I am particularly interested in understanding how this genetic conflict shapes the evolution of genetic and epigenetic systems. We are especially interested in answers to the following questions: How do RNA silencing mechanisms evolve in the face of varying transposable element content across species? How does the persistence of genetic conflict shape mechanisms of epigenetic gene control by small RNAs? What are the mechanisms underlying changes in the rate of recombination? Are these changes driven by natural selection or drift? How has conflict shaped the machinery of meiosis? To answer these questions, we work with different species within the Drosophila genus, including Drosophila melanogaster and Drosophila virilis. The lab uses a wide variety of approaches including cytogenetics, bioinformatics, molecular genetics and population genetics. We are especially interested in the evolutionary dynamics of transposable element control and gene regulation by piwi-interacting RNAs (piRNAs). Overall, we hope to integrate the experimental approach within a broader theoretical framework. Meetings Attended 2012 – “The epigenetics and evolution of TE control by piRNA: The significance of dose,” SMBE 2012, Dublin,

Ireland 2012 – “The epigenetics and evolution of TE control by piRNA: The significance of dose,” 3rd International

Conference on the Genomic Impact of Eukaryotic Transposable Elements, Pacific Grove, CA 2012 – “The role of piRNAs in genome stability and germline maintenance in Drosophila virilis,” Midwest

Drosophila Research Conference, Allerton, IL 2012 – “The epigenetics and evolution of TE control by piRNA: The significance of dose,” 53rd Annual

Drosophila Research Conference, Chicago, IL 2012 – “The role of piRNAs in genome stability and germline maintenance in Drosophila virilis,” 53rd Annual

Drosophila Research Conference, Chicago, IL

26

Committees KU Chair, EEB Seminar Committee Member, Genomics Search Committee, EEB Seminar Committee, Graduate Student Committee for Danny

Miller, Tori Pocisu and Sonia Hall National Co-Organizer, Midwest Drosophila Conference International Symposium Co-organizer, “RNA silencing and heterochromatin as genome defense: TE’s, Viruses and Selfish

Elements,” SMBE 2012, Annual Meeting of the Society for Molecular Biology and Evolution, Dublin, Ireland

Editorial and grant reviews Ad hoc reviewer, Molecular Biology and Evolution, Molecular Phylogenetics and Evolution, Journal of

Molecular Evolution, Evolution, PLoS Genetics, European Journal of Entomology, Oikos, Genetics, Proceedings of the National Academy of Sciences, Heredity, BMC Genetics, BMC Genomics, BMC Evolutionary Biology, BMC Bioinformatics, Genetics Research, Current Biology, Trends in Ecology and Evolution, Genome Biology and Evolution, PLoS ONE, G3

Panel Referee, NSF MCB Ad hoc reviewer, The Netherlands Organisation for Scientific Research, Technology Foundation STW, The

Netherlands Seminars Presented July 2012 – “Understanding meioitic decision making by whole genome sequencing,” American Genetic

Society 2012 Conference: Recombination: Molecular Mechanisms and Evolutionary Consequences, Durham, NC

Trainees Alex Erwin – Graduate Student Lucas Hemmer – Graduate Student Kendra Marr – Undergraduate Student Daniel Brown – Undergraduate Student Alisher Abdullayev – Undergraduate Student Kim Box – Undergraduate Student

Kelly A. Bosak, Ph.D, APRN Assistant Professor School of Nursing Member, Center for the Developmental Origins of Health and Adult Disease Dr. Bosak's research interests include the neurophysiology and epigenetics of health behaviors, and patient-oriented research methods, including meta-analysis and comparative effectiveness research. Dr. Bosak's

27

career goals are to support physical activity and other health behaviors to reduce cardiometabolic risk and prevent cardiovascular disease and diabetes, and associated chronic conditions. The ultimate goal of her research is translation of effective health behavior interventions to clinical practice. Meetings Attended 2012 – “Neuroimaging of Goal-Directed Behavior in Overweight Women: Preliminary Analysis,” University of

Kansas Student Research Forum, KUMC, Kansas City, KS 2012 – “Midlife Women’s Perceptions of Real-time Automated Feedback for Physical Activity,” University of

Kansas Student Research Forum, KUMC, Kansas City, KS Committees Departmental Member,Council on Collegiate Nursing Education (CCNE) Review, Research Committee, Strategic Planning

Implementation, Mentoring Task Force, Curriculum Committee National Member, Midwest Nursing Research Society (MNRS), Research Section Advisory Committee Editorial and Grant Reviews Reviewer, Western Journal of Nursing Research Seminars Presented 2012 – “An Intervention Delivering Automated Real-Time Feedback for Physical Activity,” Midwest Nursing

Research Society (MNRS), Dearborn, MI Trainees A. Hiatt – Graduate Student L. Morgan – Graduate Student A. Davis – Graduate Student D. Gillan – Graduate Student K. Russell – Graduate Student M. Grace – Graduate Student K. Gillenwater – Graduate Student L. Powell – Graduate Student A. Davis – Graduate Student Merlin G. Butler, M.D., Ph.D,, F.F.A.C.M.G. Director, Division of Research Professor of Psychiatry, Behavioral Sciences and Pediatrics Departments of Psychiatry & Behavioral Sciences and Pediatrics Member, Center for Epigenetics and Stem Cell Biology Genetics of obesity, autism and developmental disabilities; Prader-Willi syndrome. Under the direction of Dr. Butler, the primary focus of the research program is understanding the cause and diagnosis of Prader-Willi syndrome (PWS), as the clinical genetic model of obesity and genomic imprinting, and for genotype-phenotype correlations by utilizing an NIH funded rare disease center for genetics and natural history studies in

28

PWS and early onset morbid obesity. PWS is the most commonly recognized cause of life-threatening obesity in children generally due to errors in genomic imprinting usually a 15q11-q13 chromosome deletion of paternal origin. The 15q11-q13 region involves important genes for development of obesity, behavioral problems and autism. This research has led to the discovery of genomic imprinting and clinical differences in PWS subjects having either the larger typical type I or smaller type II chromosome 15q11-q13 deletion. Greater maladaptive and abnormal behavioral scores are seen in those PWS subjects with the larger type I deletion and candidate genes identified. Other obesity-related measures under study include body composition, energy balance, regional fat distribution, neuroimaging patterns and neuropeptides regulating eating behavior and comparison with PWS genetic subtypes. Furthermore, DNA, coding and non-coding RNA (microRNAs) studies are underway with targeted messenger RNAs from structural and regulatory genes involved in the pathogenesis of obesity, autism and neurodevelopment. Analyzing and comparing coding and non-coding RNA patterns in individuals with Prader-Willi syndrome and those with simple obesity will allow for identification of disturbed obesity-related gene network pathways leading to potential treatment modalities applicable to the general population. In collaboration with others, functional MRI scans and startle modulation responses in PWS and matched obese subjects using food picture stimulation paradigms during pre- and post-meal assessments are underway to better understand specific brain regions involved in eating behavior and satiation. More recently, studies are underway to examine induced pluripotent stem cells in PWS and to characterize their cell biology which is required to learn more about pathophysiology and to develop potential therapeutic interventions. Meetings Attended 2012 – “Apo-lipoprotein A1 gene polymorphisms predict cardio-metabolic risk in South Asian immigrants,”

World Heart Federation and World Congress of Cardiology Scientific Sessions, Dubai, United Arab Emirates

2012 – “X chromosome inactivation in blood and prefrontal cortex or women with alcoholism,” External Scientific Advisory Board Meeting, Institute of Reproductive Health and Regenerative Medicine (IRHRM), Kansas University Medical Center, Kansas City, Kansas

2012 – “Genome wide promoter methylation in prefrontal cortex of alcoholics and controls using NimbleGen DNA methylation 2.1M arrays,” External Scientific Advisory Board Meeting, Institute of Reproductive Health and Regenerative Medicine (IRHRM), Kansas University Medical Center, Kansas City, Kansas

2012 – “Gene expression patterns from brain samples in alcoholics,” American Psychiatric Association (APA) Annual Meeting, Philadelphia, Pennsylvania

2012 – “A risk algorithm for SIB, aggression and sterotyped behavior of infants and toddlers,” Annual Convention of the American Psychological Association, Orlando, Florida

2012 – “Maternal UPD2: A new genetic locus for Russell-Silver syndrome,” Fourth Congress of the European Academy of Paediatric Societies 2012, Istanbul,Turkey

2012 – “Plasma cytokine levels in children with autistic disorder and unrelated siblings,” 10th Edition of Planet XMap Europe-Luminex Symposium, Monte Carlo, Monaco

2012 – “Analysis of NIPA1 and NIPA2 in neuronal development and neurodegeneration using Zebrafish motor neurons as an experimental model system,” 2nd International Conference on Hyperphagia and 26th Annual Prader-Willi Syndrome Association (USA) Scientific Conference, Baton Rouge, LA

2012 – “Probing the genes for hyperphagia in rare obesity-related syndromes,” 2nd International Conference on Hyperphagia and 26th Annual Prader-Willi Syndrome Association (USA) Scientific Conference, Baton Rouge, LA

2012 – “Growth hormone receptor gene polymorphism and Prader-Willi syndrome,” 26th Annual Prader-Willi Syndrome Association (USA) Scientific Conference, Baton Rouge, LA

29

2012 – “Chromosomal microarray analysis of individuals with autism or learning deficits presenting for genetic

services,” 62th Annual Meeting of American Society of Human Genetics, San Francisco, CA

2012 – “Growth hormone receptor gene polymorphism and Prader-Willi syndrome,” 62th Annual Meeting of American Society of Human Genetics, San Francisco, CA

Committees Regional Governor Appointed Member, State of Kansas Newborn Genetics Screening Committee KUMC Member, IRHRM Executive Research Board International Member, Scientific Review Committee, NSW Tissue Resource Centre and Brain Bank Network, University of

Sydney, Australia

Editorial and grant reviews Editorial Board, Journal of Neurodevelopmental Disorders, Journal of Assisted Reproduction and Genetics,

World Journal of Medical Genetics, World Journal of Clinical Pediatrics, Hereditary Genetics, Journal Of Single Cell Protein, Journal of Bioscience and Medicine

Grant Reviewer, Frontiers Clinical Pilot and Collaborative Studies Funding Program, Canadian Institutes of

Health Research (CIHR), National Natural Science Foundation of China (NSFC), American Association for the Advancement of Science (AAAS)

Seminars Presented April 2012 - “Genetic Syndromes,” Psychiatry PGY3 &4 Residency Lectures, University of Kansas,

Kansas City, KS

August 2012 – “Research Activities in the KUMC Department of Psychiatry and Behavioral Sciences,”

University of Kansas, Dept. of Psychiatry and Behavioral Sciences (Psychiatry Residents), Kansas City,

KS

September 2012 - “Psychiatry Resident Research Activities and Forum,” Psychiatry Residents, University of

Kansas, Dept. of Psychiatry and Behavioral Sciences, Kansas City, KS

September 2012 - “Medical Genetics,” University of Kansas, Dept. of Psychiatry and Behavioral Sciences

(Psychology Graduate Students), Kansas City, KS

September 2012 – “Medical Genetics, Behavior and Autism,” Dept. of Psychiatry and Behavioral Sciences

(Child Psychiatry Fellows),University of Kansas, Kansas City, KS

October 2012 – “Dysmorphology and Genetic Disorders,” Dept. of Psychiatry and Behavioral Sciences,

University of Kansas, Kansas City, KS

30

October 2012 - “Principals of Medical Genetics and Application to Autism Spectrum Disorders,” 2nd year

medical student curriculum (Brain and Behavior section), University of Kansas, Kansas City, KS

November 2012 – “Analysis of Cytokine Levels in Children with Autism Spectrum Disorders,” Beyond the

Diagnosis: Autism Across the Life Span Meeting, University of Kansas, Overland Park, KS

December 2012 – “Genetics of Autism: An Update and Clinical Experience,” Dept. of Psychiatry and

Behavioral Sciences Grand Rounds, University of Kansas, Kansas City, KS

Academic Honors Scientific Organizer and Chairperson, 26th Annual Prader-Willi Syndrome Association (PWSA) Scientific Day

Conference, October 19-20, 2012, Baton Rouge, Louisiana Invited Participant, 1st World Genetics & Genomics Online Conference (Dr. Frank Ma, organizer), May 17-19,

2012 August 14-15, 2012 - Invited planning session panel participant, The 2nd International Conference on

Hyperphagia and 26th Annual Prader-Willi Syndrome Scientific Conference, Baton Rouge, Louisiana Marquis Who's Who in Medicine and Healthcare 2011-2012 (8th Edition) Marquis Who’s Who in Science and Engineering 2011-2012 (11th Edition) Selected to “The Best Doctors in America”, Best Doctors, Inc., Boston, MA., 2011-2012 edition Recognized by the Kansas City Business Journal (March 2012 issue) for being selected to “The Best Doctors

in America” list representing the Kansas City area physicians as the top 5% of physicians nationally Selected by “Consumers’ Research Council of America, Guide to America’s Top Physicians”, 2011-2012 Governor Appointed Member, State of Kansas Newborn Genetics Screening Committee, 2012- Appointed as HealthTap Medical Expert, HealthTap Medical Expert Network, 2012 Ivanhoe Medical News (Ivanhoe.com) entitled “Unraveling the Obesity Gene”, April 9, 2012 Newswise Medical News (Newswise.com) entitled “KU Researchers Find Further Evidence of Disturbed

Immune System in Autism”, April 16, 2012 Mentor for 2012 New Investigator for Prader-Willi Syndrome Research Award Recipient (Dr. Ann Manzardo)- National Institutes of Health Rare Disease Clinical Research Network (RDCRN) (Prader-Willi, Angelman, and

Rett syndromes)

Who's Who in North American Education 2012-2013 Edition

Scientific Chairperson and Organizer, 26th Annual Prader-Willi Syndrome Scientific Conference, Baton Rouge,

LA, October 20-21, 2012

Chairperson, Scientific Advisory Board, Prader-Willi Syndrome Association (USA), 2000-

Marquis Who’s Who in Science and Engineering, 2011-2012 (11th Edition)

31

Susan E. Carlson, Ph.D. AJ Rice Professor of Nutrition and Director Director, PhD Program in Medical Nutrition Science Director, KUMC Biomedical Interdisciplinary Research Careers in Women’s Health (BIRCWH) President, International Society for the Study of Fatty Acids and Lipids (ISSFAL) Member, Developmental Origins of Health and Disease (DOHAD) Department of Dietetics and Nutrition Member, Center for the Developmental Origins of Health and Adult Disease We perform intervention studies using docosahexaenoic acid (DHA) and arachidonic acid (AA) supplementation in pregnant women, infants and children that focus largely on pregnancy outcomes and infant/child developmental outcomes. My collaboration is with Dr. John Colombo at the University of Kansas, who is the current directory of the Lifespan Institute (KU and KUMC). Our current NICHD funding is to evaluate children from 2 to 6 years of age who were born to pregnant women provided 600 mg/day of docosahexaenoic acid (DHA), a nutritional source of long chain omega-3 fatty acids in a Phase III trial. The specific developmental outcomes we target are autonomic nervous system development, cognitive development and visual acuity development. In addition, we monitor infant/child growth, illness and food intake. Committees KUMC Chair, Faculty Affairs Research Committee (FARC) Member, Human Subjects Committee, Diversity Committee, School of Allied Health, Interdisciplinary Committee, School of Allied Health National Member, Institutional Biosafety Committee, University of Missouri Editorial and grant reviews Consulting Editor, American Journal of Clinical Nutrition Editorial Board, American Nutrition Society, American Pediatric Society, European Journal of Lipid Science

and Technology, International Society for Research on Human Milk and Lactation, Update on Pediatrics Charter Member and President, International Society for the Study of Fatty Acids and Lipids Ad hoc reviewer, Journal of Nutrition, European Journal of Clinical Nutrition, American Society for Parenteral

and Enteral Nutrition, Journal of Pediatric Gastroenterology and Nutrition, Lipids, Journal of Lipid

Research, Proceedings of the Society for Experimental Biology and Medicine, Journal of Pediatrics,

Pediatric Research, Pediatrics, Acta Paediatrica Scandinavica, National Academy of Sciences, New

England Journal of Medicine, New York Academy of Sciences, World Health Organization, National

Science Foundation, Journal of Comparative Physiology, Journal of Human Lactation, Nutritional

Neuroscience

Seminars Presented 2012 – “Pregnancy and LCPUFA,” Pediatric Society of Australia and New Zealand, Vancouver, BC 2012 – “Clinical overview of effects of dietary LCPUFA during the pernatal period on infant health,” 77th Nestle

Nutrition Institution Workshop, Panama

32

2012 – “Overview of effects of dietary LCPUFA during the perinatal period,” University of Cinncinati, Dept. of

Nutrition, Rosevear Lecture 2012 – “Nutrition and brain development,” Guangzhou and Beijing, China Trainees Deanna Mortimer – Graduate Student Claire Cody – Graduate Student Danielle Atwood - Graduate Student Lindsey Currie – Graduate Student Mallory Bratton – Graduate Student Nikki Cheng, Ph.D, Assistant Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology My laboratory is interested in investigating the functions of stromal fibroblasts in the tumor microenvironment during breast cancer progression. Fibroblasts are a major cellular component of the tumor microenvironment and influence cancer cell behavior directly and indirectly through secretion of soluble factors, including growth regulators and angiogenic factors. While genetic alterations in breast fibroblasts may exert pro-tumorigenic effects, little is known of the cellular and molecular signals that regulate fibroblast functions in the tumor microenvironment. Studies in my laboratory suggest that fibroblasts may interact with breast cancer cells to regulate cancer cell motility and invasion through chemokines signaling. Chemokines are a family of soluble proteins which signal through seven transmembrane G coupled receptors and regulate immune cell recruitment during inflammatory responses and defenses against foreign pathogens. Studies in our laboratory indicate that CCL2 and CXCL1 chemokine signaling may also regulate fibroblast interactions with other cell types in the microenvironment to promote tumor progression. Using multiple approaches including mouse models of cancer, molecular biology, biochemistry and cell culture systems, we are interested in:

Understanding the mechanisms through which chemokines regulate fibroblast : cancer cell interactions during cancer progression

Understanding the mechanisms through which chemokines regulate fibroblast mediated immune cell recruitment

Identifying the signaling pathways regulated by chemokine signaling in the breast cancer microenvironment

Identifying the regulatory mechanisms of chemokine expression Ultimately, we are interested in understanding the functions of stromal cells in the tumor microenvironment and the impact of the tumor microenvironment on metastatic spread. By identifying and understanding the molecular signals that create a tumor permissive environment, these studies may contribute to identifying new molecular targets for therapy and developing improved methods for diagnosing and treating metastatic breast cancer. Meetings Attended 2012 – AACR Annual Meeting, Chicago, IL 2012 – ESAB, Kansas City, KS

33

2012 – KUCC Research Symposium, Kansas City, KS Committees Departmental Member, Graduate Program Advisory Committee KUMC Member, KUCC Cancer Prevention Group Thesis committee member, Yong Luo, Jackie Peda, Jennifer Crowe, Hanan Elsarraj, An Zou Editorial and Grant Reviews Ad hoc Reviewer, Cancer Research, Cell Biochemistry and Function, FEBS letter, Molecular Cancer, Stem

Cells International Seminars Presented 2012 – “Functions of CCL2 Chemokine signaling in the breast tumor microenvironment,” KUCC Seminar,

KUMC

Trainees Wei-Bin Fang – Postdoctoral Fellow An Zou – Graduate Student Malinda Algaier – Rotation Student Jeremy Chien, Ph.D, Assistant Professor Assistant Director, Translational Genomics, University of Kansas Cancer Center Cancer Biology Member, Center for Reproductive Sciences

The primary focus of my research program is to understand the genetic basis of ovarian cancer and to exploit cancer specific-genetic defects for the diagnosis, screening, and therapeutic targeting of ovarian cancer. We are developing genetic biomarkers for the diagnosis and screening of ovarian cancer, and performing functional screens to identify therapeutic targets that are altered in ovarian cancer.

Meetings Attended 2012 –Pittsburgh Ovarian Cancer Symposium, Pittsburgh, PA Committees KUMC Member, UKCC Shared Resources Strategic Planning Committee, Faculty Search Committee, Biostatistics

34

Departmental Member, Faculty Search Committee Editorial and Grant Reviews Ad hoc reviewer, PLos One, Cancer Research, Scientific Reports Grant reviewer, Ovarian Cancer Research Fund, Institut National du Cancer (NCI, France) Seminars Presented 2012 – “Clinical Potentials of TCGA Gene Signatures in Ovarian Cancer Treatment,” Pittsburgh Ovarian

Cancer Symposium, Pittsburgh, PA Trainees Amanda Brinker, IGPBS rotation student Dandan Li, IGPBS rotation student Yen Hoang – Graduate Student Julie A. Carlsten Christianson, Ph.D, Assistant Professor Department of Anatomy and Cell Biology Member, Center for the Developmental Origins of Health and Adult Disease The focus of my research is the impact of early adverse events on pain processing during adulthood. Exposure to early life stress or trauma is a significant risk factor for developing several chronic pelvic pain disorders, including irritable bowel syndrome, interstitial cystitis, chronic prostatitis and vulvodynia. High comorbidity among these disorders contributes to a greater negative impact on quality of life and complicates already less-than-optimal treatment strategies. My laboratory uses two models of early life stress or trauma, including neonatal maternal separation and neonatal vaginal irritation, to investigate changes in stress and pain processing in adult mice. Both models generate enhanced pelvic pain perception, associated with dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis, which is responsible for responding to stress and the subsequent return to homeostasis. Current research is focused on identifying the specific ligands and receptors that are affected by these perturbations to determine whether they are appropriate therapeutic targets. By focusing on stress-induced changes in visceral sensitivity, we are gaining insight as to how best treat a specific subpopulation of patients suffering from chronic pelvic pain syndromes.

Meetings Attended 2012 – 14th World Congress on Pain, International Association for the Study of Pain, Milan, Italy 2012 – Neuroscience 2012, Society for Neuroscience, New Orleans, LA Committees KUMC Member, Women in Medicine and Science program committee, Kansas City chapter of the Society for Neuroscience Executive committee-KUMC

35

Departmental Seminar series coordinator Member, Graduate Studies Committee Editorial and Grant Reviews Ad hoc Reviewer – Free Radical Research, Neuroscience, Neuroscience Letters, Journal of Inflammation

Research Seminars Presented 2012 – “The painful consequences of early adverse events,” KUMC, Dept. of Molecular and Integrative

Physiology, Kansas City, KS 2012 – “The painful consequences of early life stress or injury,” Center for the Developmental Origins of Health

and Adult Disease Chalk Talk, Institute for Reproductive Health and Regenerative Medicine, Kansas City, KS

Trainees Angela Pierce – PhD student and Self Fellow scholar Briana Holt – Rotating IGPBS student John Colombo, Ph.D. Professor Director, Life Span Institute Department of Psychology – University of Kansas, Lawrence Member, Center for the Developmental Origins of Health and Adult Disease My research interests are in the developmental cognitive neuroscience of attention and learning, with a special focus on early individual differences in these areas and how they relate to the typical and atypical development of cognitive and intellectual function. Currently, I conduct a basic program of research on attention (i.e., the neural basis of learning and how it relates to learning), and its development from infancy to school age. We have conducted research on different attentional profiles in infancy and their predictive validity for intellectual and language outcomes in childhood. We are also currently exploring the use of behavioral and autonomic indices as biobehavioral markers for different developmental disabilities, including autism and ADHD. Finally, we have active programs of work on the degree to which early measures of attention and cognition can be used as outcomes for early intervention; most notably we have employed our expertise in measurement in the evaluation of the effects of prenatal and postnatal supplementation of nutritional compounds on cognitive development. Meetings Attended 2012 – International Conference on Infant Studies, Minneapolis, MN 2012 – Joint Statistical Meeting, San Diego, CA 2012 – International Society for Fatty Acids and Lipids, Vancouver, BC, Canada 2012 – Nestle Nutrition Institute Workshop on the Importance of Immunonutrition, Zurich, Switzerland

36

Editorial and Grant Reviews Associate Editor, Child Development Editorial Board/Consulting Editor, Infancy Reviewer, Infant Behavior and Development, Journal of Applied Developmental Psychology, Developmental

Disabilities Research Reviews Grant Reviewer, NIDCD Seminars Presented 2012 – “Nutrients and micronutrients in early cognitive development,” 2012 Regional Scientific Lecture Series,

Pediatric Society of Vietnam, Ho Chi Minh City, Vietnam 2012 – “Nutrition and stimulation in early behavioral development,” 2012 Continuing Medical Education Series,

Malaysian Pediatric Society, Bejaya Hills, Malaysia Trainees Lori Curtindale – PhD Student Leah Kapa – PhD Student Sara McElhaney – Graduate Student Megan Stratton Blossom – PhD Student Caitlin Brez – Postdoctoral Student Brenda Salley - Postdoctoral Student Ivan Damjanov, M.D., Ph.D. Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology Collaborative research providing histopathologic and immunohistochemical expertise Seminars Presented 2012 – “From teratoma to human embryonic stem cells,” Keynote speaker at European Congress of Pathology,

Prague, Czech Republic Animesh Dhar, Ph.D. Associate Professor Department of Cancer Biology Member, Center for Epigenetics and Stem Cell Biology Novel Therapeutic Strategies for Pancreatic Cancer: Pancreatic cancer is one of the most lethal

malignancies in humans. There is no effective conventional treatment available for the cure of patients with

pancreatic cancer, because chemotherapy and radiation therapy are largely ineffective. There are ancient

reports of saffron being used to treat various diseases, particularly cancer, by the Indian, Greek, and Chinese

cultures. This information motivated us to perform preliminary experiments evaluating the effect of crocetin

treatment on cultured pancreatic cancer cells. We demonstrated that crocetin treatment has a potent

antimitogenic effect as it inhibits pancreatic cancer cell proliferation and impairs cell cycle

37

distribution. Additional preliminary studies evaluating the effect of crocetin treatment of athymic (nude) mice

with pancreatic cancer xenografts demonstrated significant inhibition of tumor growth in the animals treated

with crocetin. Crocetin will be compared with FDA approved conventional chemotherapeutic agents used

regularly in pancreatic cancer patients. PI is developing a novel compound derived from saffron to better

understand the the mechanism responsible for the anti-tumorigenic effects of crocetin. PI is purifying novel

crocetin components from unfractionated commercially available crude crocetin that demonstrates more

potency than crude crocetin. This study design will develop novel potent crocetin component in the treatment

of pancreatic cancer using in vitro and in vivo models and we will understand the molecular mechanisms of

crocetin in relation to growth and apoptosis using novel purified crocetin alone.

Targeting of Epigenetics in pancreatic Cancer Stem Cells:

PI will be to developing a novel compounds targeted towards Jmjd1a, histone lysine demethylases, to better

understand the epigenetic mechanism in pancreatic cancer. Hypoxia is a major component of cancer for

regulation of cellular processes such as proliferation, apoptosis, angiogenesis, mortality, cell differentiation etc. The members of Jmjd family, 2-oxoglutarate (2OG)-dependent histone lysine demethylases (KDMs), are also overexpressed in a number of cancers and their inhibition suppresses cancer growth. Recent evidence suggested that hypoxia plays an important role in cancer stem cell formation; we would like to develop small molecule inhibitors of Jmjd1 and evaluate its efficacy as inhibitor of CSCs in pancreatic cancer. The laboratory of PI will take lead on developing direct an inhibitor of Jmjd1a using high-throughput screening. This study design will develop novel potent components in the treatment of pancreatic cancer using in vitro and in vivo models and we will understand the molecular mechanisms of those Jmjd1a inhibitors in relation to tumorigenesis in pancreatic cancer. Meetings Attended 2012 - AACR 103rd Annual Meeting, Chicago, IL

2012 - Second International on Perspectives of Cell Signaling and Molecular Medicine, Bose Institute, Kolkata, India

2012 - 99th Indian Science Congress Association, Bhubaneswar, India

2012 – International Conference of Molecular Medicine (MMC 2012), Bangkok, Thailand

Committees

KUMC

Member, KUMC Faculty Council representing Cancer Biology, Cancer Biology Faculty Recruitment Committee

Departmental

Member, Graduate Student Committee for Keke Pounds

Editorial and Grant Reviews

Ad hoc reviewer, Proteome Science, Biochem Biophys Acta

Grant reviewer, KUCC Pilot Project, Cancer Biology Section, MU Research Board

Ad hoc grant reviewer, NIH Tumor Progression and Metastasis (TPM) Study Section

38

Seminars Presented

2012 – “Cancer Stem Cells: Possible Target for Novel Therapeutic Approaches in Pancreatic Cancer,” KUCC

Seminar Series, Kansas City, KS

2012 – “Tumor Initiating Cells/Cancer Stem Cells: Natural Regulators and Potential Therapeutic Targets for

Pancreatic Cancer,” Second International on Perspectives of Cell Signaling and Molecular Medicine,

Bose Institute, Kolkata, India

2012 – “Cancer Stem Cells: Novel Targets for Prevention of Pancreatic Cancer,” Plenary Lecture, 99th Indian

Science Congress Association, Bhubaneswar, India

2012 – “Cancer Stem Cells: Novel Chemotherapeutic targets for Pancreatic Cancer,” International Conference

of Molecular Medicine (MMC 2012), Bangkok, Thailand

Academic Honors

Faculty Travel Award for AACR Meeting in Chicago, KUMC Research Institute

Trainees

Shamima Islam – Postdoctoral Fellow

Kanagaraj Palaniyandi – Postdoctoral Fellow

Michael S. Detamore, Ph.D, Professor Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence Member, Center for Epigenetics and Stem Cell Biology My general areas of interest include tissue engineering, biomaterials, stem cells, biomechanics and the temporomandibular joint (TMJ). My specific research interests are gradient-based scaffolds, interpenetrating network hydrogels, and umbilical cord mesenchymal stromal cells. Techniques employed in my laboratory include microsphere fabrication, electrospinning, colloidal gels, dense-phase CO2 sintering, and viral and non-viral gene delivery to mesenchymal stem cells. Applications include tissue engineering with TMJ tissues, knee cartilage and bone, cranium, and trachea.

Meetings Attended 2012 - “Raw materials and growth factor encapsulated 3d microsphere based gradient scaffolds for

osteochondral tissue engineering,” Society for Physical Regulation in Biology and Medicine, San Juan, Puerto Rico

2012 - “Approaching inner ear hair cell regeneration through non-viral gene delivery,” 9th Annual Capitol

Research Summit, Topeka, KS 2012 - “A Wharton’s jelly mesenchymal stromal cell derived 3D osteogenic scaffold does not result in

expansion of the CD34+ population of umbilical cord blood mononuclear cells,” International Cord Blood Symposium, San Francisco, CA

2012 - “Evaluation of apparent fracture toughness for articular cartilage and hydrogels in cartilage tissue

engineering,” Society For Biomaterials, Fall Symposium, New Orleans, LA

39

2012 - “Material composition and growth factor gradient scaffolds for tracheal defect repair,” Biomedical Engineering Society, Atlanta, GA

2012 - “Tuning the mechanical properties of chondroitin sulfate hydrogels independently of polymer

composition using oligo(ethylene glycol) diacrylates,” American Institute of Chemical Engineers, Pittsburgh, PA

Committees KU Member, Doctoral Education Work Group, University Committee, Chemical and Petroleum Engineering ABET

Committee, Engineering Library Committee, Graduate Engineering Association Advisory Committee

Director, Biomaterials and Tissue Engineering Track

Co-advisor, BMES Student Chapter

Chapter Advisor, KU Student Society for Stem Cell Research

Secretary, Chemical Engineering Department Advisory Board

Departmental Member, Thesis Committee (Joshua Johnson, Vidyashankara Iyer Gowrishankara, Karthik Ramachandran,

Amir Fakhari) National Member, American Society of TMJ Surgeons Program Committee Editorial and Grant Reviews Reviewer, Acta Biomaterialia, The Anatomical Record, Annals of Biomedical Engineering, Archives of Oral

Biology, Biomacromolecules, Biomaterials, Biotechnology and Bioengineering, Cellular and Molecular

Bioengineering, Chemical Engineering Education, Chemical Engineering Journal, Computer Methods

and Programs in Biomedicine, Cells Tissues Organs, European Journal of Oral Sciences, European

Journal of Pharmaceutics and Biopharmaceutics, Journal of Biomaterials Applications, Journal of

Biomechanical Engineering, Journal of Biomechanics, Journal of Biomedical Materials Research-Parts

A, B, Journal of Cellular and Molecular Medicine, Journal of Dental Research, Journal of Engineering in

Medicine, Journal of Orofacial Pain, Journal of Orthopaedic Research, Journal for Oto-Rhino-

Laryngology, Head and Neck Surgery, Journal of the Royal Society Interface, Journal of Tissue

Engineering and Regenerative Medicine, Nanomedicine, Orthopedic Reviews, Osteoarthritis and

Cartilage, Recent Patents on Regenerative Medicine, Stem Cells and Development, Tissue

Engineering, Parts A, B, C, Wound Repair and Regeneration

Grant Reviewer, Federal Advisory Committee, Department of Veterans Affairs, standing study section member;

NIH: MTE Study Section, standing study section member

Seminars Presented 2012 - “Interpenetrating networks and colloidal gels,” Society for Physical Regulation in Biology and Medicine

Annual Meeting, San Juan, Puerto Rico

40

February 7, 2012 - “Gradient strategies and umbilical cord Wharton's jelly cells for osteochondral tissue

engineering,” Rice University, Department of Bioengineering, Houston, TX 2012 - “Microsphere-based strategies for tissue regeneration in the TMJ,” American Association for Dental

Research Annual Meeting, Tampa, FL 2012 - “Gradient strategies and umbilical cord Wharton's jelly cells for osteochondral tissue engineering,”

University of Akron, Department of Chemical Engineering, Akron, OH 2012 - “Gradient strategies and umbilical cord Wharton's jelly cells for osteochondral tissue engineering,”

Harvard University, Khademhosseini Group, Cambridge, MA 2012 - “Curing Tracheal Stenosis,” University Research and Entrepreneurship Symposium (URES),

Cambridge, MA 2012 - “Current advances in tissue regeneration,” American Association of Oral and Maxillofacial Surgery:

Symposium on Stem Cell Technology and Tissue Engineering, San Diego, CA 2012 - “Gradient strategies and umbilical cord Wharton's jelly cells for osteochondral tissue engineering,”

Colorado School of Mines, Department of Chemical & Biological Engineering, Golden, CO Academic Honors Co-Chair, TMJ Bioengineering Conference (September 2012) Co-Chair, Society for Physical Regulation in Biology and Medicine Annual Meeting (January 2013)

Co-Chair, TMJ Bioengineering Conference (September 2012)

#11 Most Recently Read Article, Tissue Eng B, Renth et al., over 1-month period (November 2012) Trainees Neethu Mohan – Postdoctoral Student A.J. Mellott – PhD Student Lindsey Ott – PhD Student Amanda Renth – PhD Student Emily Mangus – PhD Student Vineet Gupta – PhD Student BanuPriya Sridharan – PhD Student Cate Wisdom – PhD Student Sushma Jadalannagari – PhD Student Amanda Sutherland – Graduate Student Brooke Lohman – Undergraduate Student Ashley Farris - Undergraduate Student Gelareh Samandi - Undergraduate Student Austin Smith - Undergraduate Student Deena Rennerfeldt - Undergraduate Student Daniel Tabakh - Undergraduate Student Shawn Briley - Undergraduate Student Bharath Krishnamoorthi - Undergraduate Student Megan Godsey - Undergraduate Student Cindy Vu - Undergraduate Student

41

Luciano DiTacchio, Ph.D. Assistant Professor Department of Pharmacology, Toxicology & Therapeutics Member, Center for Epigenetics and Stem Cell Biology Genomic and envrionmental factors in the development of obesity, metabolic syndrome and Type 2 diabetes

According to the Centers for Disease Control and Prevention, the rate of obesity in the United States has doubled since 1990. Today, over sixty percent of the U.S. population is overweight and 30% of adults are categorized as obese, while type II diabetes is the fastest growing non-communicable disease in the U.S. Thus, there is a compelling need to understand and the mechanisms that give rise to and underlie the pathologies associated with excess body weight.

The circadian system is an endogenous timing mechanism that is present across phyla and is responsible for synchronizing an organism's behavior and physiology to the most optimal time of day. In mammals, this system is based on a subcellular transcription-translation circuit which generates strong, ~24-hour oscillations in up to 20% of the transcriptome of any given organ. Thus, the impact of this system is far-reaching, exerting considerable influence and control over most, if not all, major organismal processes. Importantly, the circadian oscillator has emerged as a critical orchestrator of metabolism and energy homeostasis. Consistently, circadian dysfunction due to environmental factors such as those commonly found in modern lifestyles (jet lag, shift work, artificially-extended photoperiod) has been linked to a number of disease processes, including cancer, obesity, and metabolic syndrome.

The overarching goal of my laboratory is to understand how the genome and the environment interact with one another, and the role this interaction plays in human health in general, and in the development of obesity, metabolic syndrome and Type 2 diabetes in particular. Towards this end I aim to:

(1) dissect the genetic programs involved in the maintenance of energy homeostasis of adult, post-differentiated tissues,

(2) understand the molecular mechanisms of the circadian oscillator, and

(3) elucidate how the circadian oscillator and energy metabolism interact with one another and become dysregulated in obesity and disease.

Yafeng Dong, Ph.D, Research Associate Professor Director, Molecular Biology Core Facility Department of Obstetrics and Gynecology Member, Center for the Developmental Origins of Health and Adult Disease The problem of perinatal brain injury, in terms of the costs to society and to the affected individuals and their families, is extraordinary. The most common underlying cause of perinatal brain injury is hypoxia/ischemia. Intrauterine hypoxia and birth asphyxia induced brain damage are associated with increased perinatal mortality and long term sequelae of neurodevelopmental compromise, seizure disorders and cerebral palsy. The roles of ROS, Ca2+, NMDA receptors, excitatory amino acids, and apoptotic genes on fetal brain injury have been studied exclusively. These works have led to substantial conceptual agreement on a general outline of how fetal brain injury triggers and evolves to produce neuropathologic lesions and neurodevelopmental disabilities. However, the precise etiological factors responsible for the development of the majority of fetal hypoxic brain injury have not been identified.

42

Leigh M. Eck, M.D. Assistant Professor Department of Internal Medicine Member, Center for the Developmental Origins of Health and Adult Disease

Vitamin D and its impact on premenopausal bone health

Vitamin D in end stage liver disease

Meetings Attended 2012 – OSARM Results and Work in Progress, University of Kansas, Kansas City, KS 2012 – AACE 21st Annual Scientific and Clinical Congress, Philadelphia, PA 2012 – ENDO 2012, Houston, TX 2012 – American Society of Bone and Mineral Research/Endocrine Fellows Foundation Meeting, Minneapolis,

MN 2012 – American College of Physicians Kansas Chapter Meeting, Kansas City, KS Committees KUMC Member, Academic Society Directors Group, Residency Liaison Committee, Diabetes Care Committee,

Thyroid Tumor Board, Graduate Medical Education Committee, Academic and Professionalism Committee

Departmental Member, The Internal Medicine In-Training Examination Question Writing Committee, Medical Education

Committee, Grand Rounds Planning Committee, Board of Trustees National Member, American College of Physicians/AAIM Internal Medicine In-Training Question-Writing Editorial and Grant Reviews Reviewer, American Family Physician, Alliance for Academic Internal Medicine in The American Journal of

Medicine, Kansas Journal of Medicine Trainees Jennifer Fink – MD Student Laura Thomas – MD Student Collin Lovitt – MD Student Kamran Aghaie – MD Student

43

Patrick E. Fields, Ph.D. Associate Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology Recent work in my laboratory has focused on two major areas: the mechanisms of T cell activation and differentiation, and fundamental aspects of embryonic hematopoiesis. Regarding T cells, we are interested in both membrane-proximal and -distal (nuclear) events regulating gene expression involved in cell fate decisions during peripheral T cell differentiation. Of particular interest to the lab is the study of chromatin remodeling in the regulation of cytokine gene expression during peripheral T cell differentiation. We identified a locus control region (LCR), which regulates gene expression in the Th2 cytokine locus. LCRs are regulatory elements that are thought to control gene expression by regulating the accessibility of gene promoters to transcriptional machinery. We use mouse genetics (knockout and transgenic technology) as well as molecular biology and biochemical approaches to study the mechanism by which this LCR functions. These studies will facilitate our long-term goal, which is to understand normal T cell function at the molecular level. Another major area of research in the laboratory is focused on the role of chromatin remodeling in embryonic hematopoiesis. We have identified a crucial role for the epigenetic modifier, the DOT1L methyltransferase, in early blood development. To examine the function of DOT1L in hematopoiesis, we created a mutant mouse that lacks this enzyme. Mutant embryos are severely anemic, and die at mid-gestation. Yolk sac-derived, erythroid progenitors from these mice exhibit defective responses to erythropoietin as well as abnormal growth and reduced survival, in vitro. Interestingly, the effects on hematopoiesis are relatively erythroid-specific. These observations are indicative of a novel role for this enzyme in regulating growth and differentiation factor responses during hematopoiesis, as well as promoting normal erythroid development. Committees KUMC Member, Microarray Facility Committee, Transgenic Mouse Facility Committee Alternate Member, Faculty Council Committee, Graduate Program Advisory Committee IGPBS Graduate Student Admissions Committee Departmental Member, Pathology Graduate Program Advisory Committee, Website Development Committee, Graduate

Program Advisory Committee Member, Dissertation Committees for Todd Bradley, Yi Feng, Beth Dille, Damayanti Chakraborty, Jitu George,

Fang Tao, Caitlin Linscheid, Jacqueline Peda, Nehemiah Alvarez (Mentor), Amy Cantilina, Carrie Malcom (Mentor)

Editorial and Grant Reviews Ad hoc Reviewer - Proceedings of the National Academy of Sciences, USA (PNAS), Immunity,

Molecular and Cellular Biology (MCB), Immunology, Journal of Blood Research Editorial Board Member, Journal of Biological Chemistry Reviewer, NIH/NHLBI and NIH/NIGMS

44

Seminars Presented 2012 – “Role of Dot1L in Embryonic Hematopoiesis,” Hematopoiesis symposium sponsored by the NIDDK

Trainees Yi Feng – Graduate Student Nehemiah Alvarez – Graduate Student Carrie Malcom – Graduate Student Jessica Rossol-Alison - Postdoctoral Fellow Timothy A. Fields, M.D., Ph.D, Associate Professor Director, MD-PhD Physician-Scientist Training Program Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology Briefly, our lab is primarily focused on understanding factors that influence progression of polycystic kidney disease (PKD). In particular, we are interested in the influence of inflammation, especially macrophages, on PKD progression. We have recently shown that macrophages infiltrate human PKD kidneys and convert to a phenotype that is deleterious in PKD. Also, using mouse models, we have shown that these infiltrating cells promote disease progression. Our current work is focused on understanding the factors that promote recruitment of macrophages to diseased kidneys, the mechanism by which the PKD kidney microenvironment influences the phenotypic conversion of macrophages, and the specific mechanism(s) by which the macrophages promote disease. An understanding of these mechanisms could identify new targets for therapy in PKD. Other interests in the lab include understanding signaling mechanisms, particularly those controlled by the Wnt family of secreted molecules, that regulate differentiation and migration of progenitor cells and cancer cells. Meetings Attended

2012 - American Society of Nephrology 2012 Meeting, San Diego, CA

2012 - USCAP Annual Meeting 2012 (Renal Pathology Society Affiliated Meeting), Vancouver, BC

Committees KUMC Chair, Research Committee (Faculty Council) Member, Medical School Admissions Selection Committee, MD-PhD Admissions and Advisory Committee,

Executive Committee of the Faculty Council, Faculty Assembly Research Committee, Transplant Committee, Dissertation Committee for 8 current graduate students and one other who graduated last fall, Strategic Planning for LCME

National Member, Organizing Committee for Renal Pathology Society Annual Meeting

45

Editorial and Grant Reviews Ad hoc reviewer, Oncogene, J Histochem Cytochem

Academic Honors

Our paper in press in Kidney International (“Macrophages Promote Polycystic Kidney Disease Progression”) was selected by Faculty of 1000 for F1000Prime. It was recommended as being of special significance in its field (http://f1000.com/prime/717980903?bd=1&ui=27410).

Trainees Sally Salah – Graduate Student Jacqueline Peda – Graduate Student Paige Geiger, Ph.D. Associate Professor Department of Molecular and Integrative Physiology Member, Center for Reproductive Sciences My research focus also includes the role of estrogen receptors in glucose regulation. Evidence from both human and rodent studies demonstrates the ability of estrogens to modify glucose homeostasis. Premenopausal women have increased insulin sensitivity compared with age-matched men. Premenopausal women are also less likely to develop insulin resistance and have higher levels of GLUT4, the protein responsible for insulin-stimulated glucose uptake in skeletal muscle. In contrast, following menopause a significant decline in insulin sensitivity occurs along with a corresponding increase in fat mass. Estrogen replacement has been shown to ameliorate the increased risk for type 2 diabetes in postmenopausal women and improve whole body and skeletal muscle glucose metabolism. In animal models, insulin sensitivity and glucose metabolism are impaired following ovariectomy and estrogen replacement protects against insulin resistance. Further, aromatase knockout mice, which lack the ability to synthesize estrogen hormones, are insulin resistant. The primary estrogen receptors, ERα and ERβ, are products of two distinct genes. Increased adiposity occurs in humans and mice as a result of decreased ERα activation and mice with global knockout of ERα exhibit impaired glucose tolerance and skeletal muscle insulin resistance. Based on this evidence, the beneficial effects of estrogens on glucose metabolism are thought to be mediated by ERα. My research aims to 1) discover the signaling pathways mediating the beneficial effects of ERα stimulation on glucose uptake in skeletal muscle and 2) determine the ways in which ERα stimulation alters fatty acid handling in adipose tissue. Committees KUMC President, Women in Medicine and Science Member, Orr Academic Society Faculty Advisor, Landon Center on Aging and Department of Physical Therapy

and Rehabilitation Sciences Faculty Search Committee Departmental Member, Graduate Student Affairs Committee

46

National Member, American Physiological Society Integrative Biology of Exercise 2012 Meeting Planning Committee Editorial and Grant Reviews Editorial Board, American Journal of Physiology, Regulatory, Integrative and Comparative Physiology Ad hoc member, National Institutes of Aging, NIH, PPG study section review, NIH, Integrative Physiology of

Obesity (IPOD) study section Seminars Presented 2012 - “Targeting heat shock proteins in the prevention of insulin resistance,” The Impact of Heat Shock

Protein Expression on Muscle Metabolism, Exercise Capacity and Disease Prevention. Integrative Biology of Exercise meeting, Westminster, CO

2012 - “Targeting heat shock proteins in the prevention of insulin resistance,” Department of Pharmacology

and Toxicology Trainees Amanda Ward – Summer Undergraduate Student Kathleen White - Summer Undergraduate Student David Wilson - Summer Undergraduate Student Robert Rogers – Graduate Student Matthew C. Goering, Ph.D., HCLD Director, Clinical Embryology, The Center for Advanced Reproductive Medicine Assistant Professor Department of Obstetrics and Gynecology Member, Center for Reproductive Sciences Errors in meiotic chromosome segregation occur in as many as one in every four human oocytes, and the frequency and complexity of these errors increases dramatically as a woman ages. The gain or loss of a chromosome (aneuploidy) is a leading cause of infertility, pregnancy loss and birth defect. During meiosis, recombination tethers pairs of homologous chromosomes to one another through the formation of crossovers. These crossovers, together with the cohesin complex and associated proteins, ensure the proper alignment and segregation of chromosomes at the first meiotic division. In human females, the formation of crossovers occurs during oogenesis in the fetal ovary but does not resolve itself until some 20 to 40 years later when the oocytes are recruited for ovulation during monthly ovulatory cycles. The primary focus of our research is directed at understanding how ovarian physiology and pathophysiology impact the stability of these recombination intermediates and their associated proteins over time. Our long-term goal is to identify therapeutic targets or interventions that may preserve fertility and reduce the risk of pregnancy loss arising from aneuploidy. Seminars Presented 2012 – “Cohesin & Maternal Age-Related Non-Disjunction,” University of Alabama Birmingham School of

Medicine, Dept. of OB/GYN, Birmingham, AL

47

Kathleen M. Gustafson, R. EP T., Ph.D, Research Assistant Professor Department of Neurology Director, Fetal Biomagnetometry Laboratory, Hoglund Brain Imaging Center Member, Center for the Developmental Origins of Health and Adult Disease Since the 1980's, there has been increasing recognition that events that occur in utero have long-term implications for future health. Maternal nutrition, physical activity, psychological stress and social disparities have the potential to put the fetus at risk or "program" the offspring for obesity, insulin resistance, diabetes, cardiovascular disease and cancer. Our research is focused on the developmental origins of health and disease. To accomplish these studies, we use a dedicated fetal biomagnetometer to measure naturally occurring magnetic fields that surround bioelectric currents in the maternal and fetal bodies. There are only two dedicated fetal biomagnetometers in the United States. This device is housed at the Hoglund Brain Imaging Center on the Kansas University Medical Center campus. It allows for completely safe, non-invasive studies of women during their pregnancy. Of principal importance to our research is the magnetocardiogram (MCG), recorded simultaneously from mother and child. Using the MCG, we are able to determine fetal behavioral states and fetal movements including non-nutritive sucking and swallowing, hiccups and periodic fetal breathing. During these unique fetal activities, we have shown how the fetus regulates its heart rate and heart rate variability and how these activities differ when women exercise during pregnancy or take an omega-3 supplement. We now know that when women exercise during pregnancy, their fetus has greater ability to vary its heart rate which may give it an adaptive advantage. Development and maturation of fetal cardiac autonomic control not only gives us insight into cardiac regulation, but also brain development. The autonomic nervous system, in particular vagal regulation, has also been linked to basic cognitive components related to arousal and attention. We believe we have a unique opportunity to make significant contributions to the field of developmental origins. Meetings Attended 2012 – 10th Congress of the International Society for the Study of Fatty Acids and Lipids, Vancouver, BC,

Canada

2012 – International Society on Infant Studies, Minneapolis, MN, USA 2012 – “Physical Activity Interventions in Overweight and Obese Pregnant Women,” Featured Science

Session. 59th Annual Meeting and 3rd World Congress on Exercise is Medicine, San Francisco, CA Committees KUMC Member, PhD Advisory Committee – Shengqi Li, MS Advisory Committee – Megan Brinker Editorial and Grant Reviews Ad hoc Reviewer, Journal of Parenteral and Enteral Nutrition, Clinical Neurophysiology, Autonomic

Neuroscience: Basic and Clinical

Grant Reviewer, University of Kansas, Frontiers Seminars Presented 2012 – “Exploring the Fetal Origins Hypothesis Using Magnetocardiology,” International Perinatal Brain &

Behavior Network. New Orleans, LA

48

2012 – “The Role of Fetal Biomagneometry in Understanding Developmental Origins,” CDOHAD/IRHRM, Kansas City, KS

2012 – “Maternal Health and Implications for Fetal Programming: Strategies for Intervention,” DN 901,

Graduate Seminar Series, Nutrition & Neuroscience, KUMC. Kansas City, KS 2012 – “Applications and Principals of Fetal Biomagnetometry,” REHS 863 Pathobiology of Human Function,

Graduate Course. Kansas University Medical Center. Kansas City, KS Jeffrey M. Holzbeierlein, M.D. John W. Weigel Endowed Associate Professor Director of Urologic Oncology Department of Urology Member, Center for Reproductive Sciences Dr. Holzbeierlein specializes in the treatment of genitourinary malignancies including prostate, bladder, kidney, testicular, and penile cancers. His research interest includes the androgen receptor as a target of Hsp90 inhibitors in prostate cancer and clinically decreasing the morbidity associated with cystectomy. Committees KUMC Member, Executive Research Committee; Data Safety and Review Monitoring Board, Compliance Committee Cancer Committee, Pensions and Benefits Committee, LCME Task Force Committee, Radiation Oncology

Search Committee, Opthalmology Chair Search Committee, Nominating Committee for the Medical Staff

National Member, American Urological Association Practice Guidelines Committee, Public Relations and Media

Committee for the American Urological Association, Young Urologist’s Committee for the American Urological Association

Editorial and grant reviews Ad hoc reviewer, Journal of Urology, Practical Reviews in Urology, Cancer, Nature Cancer, Journal of Urologic Oncology Holly R. Hull, Ph.D, Assistant Professor Department of Dietetics and Nutrition Member, Center for the Developmental Origins of Health and Adult Disease Dr. Hull’s research agenda revolves around two themes: examining factors that influence maternal cardiometabolic health during pregnancy and exploring maternal factors that impact fetal development and infant growth and health. Current research ongoing in Dr. Hull’s laboratory examines the influence of in utero hyperglycemia and maternal obesity on fetal growth and offspring adiposity and early growth, a second study examines the impact of maternal body composition, inflammation and fat patterning on infant body composition and a final study is a physical activity intervention to encourage appropriate weight gain in pregnancy.

49

Meetings Attended 2012 - The Obesity Society Annual Meeting, San Antonio, TX Committees KUMC Member, SHP Research Committee, Dietetics and Nutrition PhD Advisory & Development Committee,

Dietetics and Nutrition Committee for the Nutrition Clinic

National Member, Dean’s Research Committee, OU Health Sciences Center; Statistical Sub-Committee, OU Health

Sciences Center; Student Grant Sub-Committee, OU Health Sciences Center; Faculty Board, OU Health Sciences Center; By-Laws, OU Health Sciences Center; Academic Misconduct, OU Health Sciences Center; Rehabilitation Sciences Doctoral Admissions Committee, OU Health Sciences Center; Nutritional Sciences Doctoral Admissions Committee, OU Health Sciences Center; PhD Advisory and Development; Nutrition Clinic

Chair, Tenure Track Faculty Committee, OU Health Sciences Center Editorial and Grant Reviews Ad hoc Reviewer - American Journal of Clinical Nutrition, American Journal of Obstetrics and Gynecology,

American Journal of Perinatology, American Journal of Preventative Medicine, International Journal of Behavior, Nutrition and Physical Activity, International Journal of Body Composition Research, International Journal of Obesity, Internal Journal of Pediatric Obesity, Journal of Women’s Health, Medicine and Sport in Science and Exercise, Obesity, Pediatric Research, Acta Paediatrica

Seminars Presented 2012 – “Development of a pilot grant to promote appropriate gestational weight gain in overweight women,”

Building Interdisciplinary Research Careers in Women’s Health meeting, University of Kansas Medical Center, Kansas City, KS

2012 – “Effectiveness of a real-time automated stimulus response intervention to promote healthy gestational

weight gain,” Faculty wide School of Health Professions meeting, University of Kansas Medical Center, Kansas City, KS

2012 - “Novel methods to prevent excessive gestational weight gain in overweight women,” Institute for

Reproductive Health & Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS Trainees Jessica Nunez – Graduate Student Renna Crawford – Graduate Student Sara Chandra – Graduate Student Shengqi Li – Graduate Student Debbie Obaden – Graduate Student Cheng Li – Graduate Student Emily Zans – Graduate Student Robyn Johnson – Graduate Student

50

Tomoo, Iwakuma M.D. Ph.D, Associate Professor Department of Cancer Biology Member, Center for the Epigenetics and Stem Cell Biology Dr. Iwakuma's primary research focuses on the field of Cancer Research, specifically on cancer progression in bone and soft tissue sarcoma. Over 50% of human cancer has mutations in the tumor suppressor p53 which regulates cell cycle progression, cell death, senescence, chromosome integrity, DNA repair, and metastasis. Therefore, understanding of the pathway involved in the regulation of p53 is essential for discovering novel cancer therapies. With special focus on the tumor suppressor p53 pathway, Dr. Iwakuma dissects the mechanism of cancer progression using genetically engineered mice, as well as tumor transplantation models, and applies disease models to translational research, to ultimately cure cancer.

Meetings Attended 2012 – “Downregulation of mutant p53 attenuates CSC-like properties of osteoscarcoma,” Cancer Center

Reserarch Symposium, KUMC, Kansas City, KS 2012 – “Downregulation of mutant p53 attenuates CSC-like properties of osteoscarcoma,” “A mechanism of

metastasis suppression by MDM2 Binding Protein,” KUMC Postdoc Research Day, Kansas City, KS

2012 – “Metastasis suppression by MDM2 Binding Protein through inhibition of ACTN4 function,” 2012 AACR,

Chicago, IL

2012 – “What is going on Dr. Iwakuma’s lab,” IGPBS Interaction at Physiology, Kansas City, KS

2012 – “Functional association of MDM2 Binding Protein with migratory and metastatic potential of

hepatocellular carcinoma,” “Roles of genes regulating sphere forming potential of osteosarcoma,” 2012

Cancer Center Research Symposium, KUMC, Kansas City, KS

Committees

KUMC

Chair, KU Cancer Center Educational Strategy Plan Committee

Departmental

Member, Faculty Search Committee

Editorial and Grant Reviews

Ad hoc reviewer, Cancer Research, Cancer Metastasis Review, British Journal of Cancer, The New England Journal of Medicine, Achieves of Dermatological Research, CELL STEM CELL, Cell Cycle

Seminars Presented

2012 – “A mechanism of metastasis suppression by MDM2 Binding Protein,” KU Cancer Center Seminar

Series, Kansas City, KS

2012 – “Mutant p53 gain of function in mouse models and its potential as a therapeutic target for cancer,”

UMKC, Dept. of Oral Biology Seminar Series, Kansas City, KS

51

2012 – “Mutant p53 gain of function in the stem cell-like properties of osteosarcoma,” The 1st Australian p53

workshop

2012 – “MDM2 Binding Protein as a novel metastasis suppressor,” Australia Viktoria Hospital Seminar Series

2012 - “Dissecting osteosarcoma genesis in mouse models,” UMKC, Dept. of Pharmacology annual AAPS-

PSGSA Student Chapter Annual Seminar Series

2012 – “MDM2 Binding Protein plays a crucial role in mitotic progression, chromosome stability, and tumor

progression,” KU Dept. of Molecular Bioscience Speaker Series, Lawrence, KS

Academic Honors

KUMC Student Research Forum, the Best Presentation in Stem Cells & Epigenetics Award (Swathi Iyer).

2012 Summer Student Research Training Program (Tarek Shaath/ Tomoo Iwakuma) "A Pilot Study of the

Role MDM2-MTBP Axis in Osteosarcoma Metastasis"

KUMC travel award: $258.00

KU Cancer Center Research Symposium, Poster Award: $500

Trainees

Qian Bi – Postdoctoral Narinder Sharma – Postdoctoral Dr. Aaron Althaus – Resident Tarek Shaath – Medical Summer Student John Weitlich – Medical Summer Student Debleena Dey - Postdoctoral

Rajasingh Johnson, M.Phil., Ph.D., HCLD (ABB) Assistant Professor Department of Internal Medicine, Division of Cardiovascular Diseases Member, Center for Epigenetics and Stem Cell Biology

Reprogramming of somatic cells to generate induced pluripotent (iPS cells) or multipotent stem cells and its therapeutic potential in regenerative medicines

Study the mechanisms of reprogramming by histone deacetylation and DNA methylation Use of embryonic and adult stem cells in cardiovascular and lung vascular repair and regeneration Defensive role of epigenetic modifiers during infection and inflammation.

Meetings Attended

2012 – American Heart Association Scientific Meeting, Los Angeles, CA

2012 – American Association of Bioanalysts Missouri chapter spring workshop, Lake of the Ozarks, MO

Committees KUMC Member, IACUC Committee, Faculty LCME self-study committee

52

Editorial and Grant Reviews Editorial Board Member, Advances in Life Sciences, Journal of Tissue Science & Engineering, Journal of

Regenerative Medicine and Tissue Engineering, Cell Science and Therapy Academic Editor, PIoS One Grant reviewer, National Grant Panel: AHA-Basic Cell-Regenerative Cell Biology 1, AHA-Western State

Affiliate Innovative Award; NIEHS-Environmental Stem Cells Research Seminars Presented 2012 – “Effects of Epigenetic Modifiers on LPS-induced eNos/Rho Signaling and Restoration of Adherens

Junction Integrity and Endothelial Barrier Protection,” “Reprogramming of Human Somatic Cells into Cardiomyocytes,” American Heart Association Scientific Conference, Los Angeles, CA

2012 – “Reprogramming of Somatic Cells and Therapy,” Grand Rounds & Seminar presentation, Regenerative

Medicine Center, University of Nebraska Medical Center, Omaha, NE Trainees Jayakumar Thangavel – Postdoctoral Amy Cantilena – Graduate Student Bliss O’Bryhim – Graduate Student Sarah Finocchario Kessler, Ph.D,, M.P.H. Adjunct Assistant Professor Department of Family Medicine Member, Center for Reproductive Sciences Her current research combines interests in reproductive health and HIV by exploring childbearing intentions among people living with HIV, HIV-provider communication about reproductive options, and options for safer conception among serodiscordant couples. She is exploring these issues among HIV-infected women and men in the U.S. (Baltimore), Brazil and Uganda where in collaboration with colleagues, she has documented a similar unmet need for comprehensive reproductive counseling to reduce transmission risks to partners and infants while helping individuals more safely realize their childbearing goals. Dr. Finocchario Kessler plans to work with KUMC fertility specialists to explore similar outcomes among couples receiving preconception counseling in the context of infertility, cancer or other chronic illnesses. Dr. Finocchario Kessler also works in Kenya and Malawi to pilot an intervention designed to improve Early Infant Diagnosis services for HIV-exposed newborns. The primary outcomes of this research include increased retention in care and early initiation of treatment among infants diagnoses HIV-positive. Meetings Attended 2012 - North America Primary Care Research Group Conference, New Orleans, LA

2012 - 140th APHA Annual Meeting, San Francisco, CA

2012 - XIX International AIDS Conference, Washington DC

53

Committees National Member, CDC Stakeholders Group to Eliminate Perinatal HIV Transmission in the U.S., CDC Expert Panel on

HIV and Preconception Care Dev Karan, Ph.D, Assistant Professor Department of Urology Member, Center for Reproductive Sciences

Prostate cancer biology

Cancer immunology immunotherapy

Inflammation

Prostate tumor microenvironment

Meetings Attended 2012 – Society for Immunotherapy of Cancer, North Bethesda, MD. Editorial and Grant Reviews Guest Editor, Current Cancer Therapy Reviews (ISSN: 1573-3947 and 1875-6301) 2012: Vol. 8; Issue 4 Editorial Board Member, Journal of Andrology, Immunotherapy: Future Medicine Ad hoc reviewer, Cancer Epidemiology, Journal of Proteomics and Genomics Research, Cancer Control:

Journal of the Moffitt Cancer Center, Research and Report in Urology, Cancer Medicine-open access, Cancer Letters

Grant reviewer, Prostate Cancer UK Research Awards, DOD: Prostate Cancer Research Panel, NIH-Cancer

Health Disparity (R01)

Seminars Presented 2012 – “Multi-gene target immunotherapy for prostate cancer,” IRHRM, Kansas City, KS 2012 – “Multi-gene target immunotherapy for prostate cancer,” University of Alabama, AL Sarah L. Kieweg, Ph.D. Assistant Professor Department of Mechanical Engineering, School of Engineering, University of Kansas, Lawrence Member, Center for the Developmental Origins of Health and Adult Disease Dr. Kieweg is an Assistant Professor in Mechanical Engineering, at the Lawrence campus of the University of Kansas. She has held a courtesy position in Obstetrics & Gynecology at the KU School of Medicine since 2007. Dr. Kieweg conducts research in non-Newtonian fluid mechanics with applications in biomechanics, primarily to improve the drug delivery of anti-HIV microbicides. Her research also has applications in women’s health including instrument design, soft tissue mechanics of the female pelvic floor, and the biomechanics of delivery.

54

Dr. Kieweg was an NIH K12 Building Interdisciplinary Research Careers in Women’s Health (BIRCWH) Scholar (2007 – 2011) at KUMC and is the PI of a 5-year NIH phased R21/R33 award, funded through the NIH Microbicide Innovation Program. As co-PI on a Major Research Instrumentation grant from the National Science Foundation, she is conducting high performance computational simulations of thin film flow of non-Newtonian fluids to enable the rational design of microbicide delivery vehicles. Other projects include the development of mathematical models of relevant transport phenomena to design nanomedicines for microbicide drug delivery. Additional funding includes a Kauffman Foundation/Institute for Advancing Medical Innovation proof-of-concept award for a device that will automatically vitrify reproductive cells and tissue to preserve fertility in cancer patients. Her IRHRM collaborators include Dr. David Albertini, Dr. S. Samuel Kim, and Dr. Carl Weiner. Meetings Attended June 2012 – American Society of Mechanical Engineers (ASME) Summer Bioengineering Conference,

Fajardo, Puerto Rico November 2012 – “Contact line instability of gravity-driven flow of power-law fluids,” American Physical Society

Division of Fluid Dynamics 2012 Meeting, San Diego, CA Committees KU Member, Thermofluids Core Research Lab Planning Committee Departmental Chair, ME Department Recruitment Committee Member, Scholarship Committee, ME Doctoral Qualifying Exam Committee National Chair, Student Paper Competition, 2013 ASME Summer Bioengineering Conference Member, Conference Organizing Committee, 2013 ASME Summer Bioengineering Conference, Fluids

Technical Committee, ASME Bioengineering Division, Education Committee, ASME Bioengineering Division

Editorial and grant reviews Ad Hoc Reviewer, Computers & Fluids,ASME Journal of Biomechanical Engineering Reviewer, NIH Peer Review Panel: NIAID Integrated Preclinical/Clinical Program for HIV Topical Microbicides Seminars Presented April 2012 – “Mathematical models of the fluid mechanics of non-Newtonian polymer solutions used to prevent

HIV transmission,” Applied and Computational Mathematics Seminar Series, Dept. of Mathematics, University of Kansas, Lawrence, KS

July 2012 – “Biomechanical approach to microbicide delivery systems and mathematical modeling of

nanoparticle transport,” Youan Research Group, Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City

55

Academic Honors 2012 University of Kansas Bellows Scholar Trainees Mark Pacy – Graduate Student Bin Hu – Graduate Student Thora Whitmore – Graduate Student Md. Rajib Anwar – Graduate Student Derek Taylor – Graduate Student Zouhair Talbi – Graduate Student James Fleenor – Graduate Student Alex Hodes – Undergraduate Student S. Samuel Kim, M.D,, FACOG Associate Professor Division Director, Reproductive Endocrinology and Infertility Director, Center for Advanced Reproductive Medicine KU Cancer Center Department of Obstetrics and Gynecology Member, Center for Reproductive Sciences Dr. S. Samuel Kim is an internationally renowned specialist in reproductive endocrinology and infertility. He has 20 years of experience in clinical reproductive medicine and surgery. Dr. Kim is also a highly-esteemed scientist whose reputation as a pioneer in ovarian tissue cryopreservation and transplantation has been recognized worldwide. He spearheaded the founding of the International Society for Fertility Preservation (ISFP), and currently serves ISFP as President. Dr. Kim is Division Director for Reproductive Endocrinology and Infertility at the University of Kansas and Medical Director for the Center for Advanced Reproductive Medicine. He received numerous scientific awards and has been lecturing around the world. In 2011, he published a comprehensive textbook, “Principals and Practice of Fertility Preservation”. Seminars Presented 2012 – “Fertility Preservation: practice guidelines,” International Conference and Hands-on Courses on Fertility

Preservation, Palermo, Italy 2012 – “The genomic and proteomic consequences of ovarian tissue cryopreservation,” ASRM 2012

Symposium, San Diego, CA

2012 – “Fertility Preservation: from endometriosis to ovarian tissue cryopreservation,” Brussels, Belgium

2012 – “Fertility Preservation: harmony of sciences and clinical practice,” ASPIRE 2012, Osaka, Japan

2012 – “Fertility Preservation: Practice Guidelines,” BCGiP-COGI Congress, Barcelona, Spain

2012 – “Aging, Ovarian Reserve, and Reproduction,” Kansas City Gynecological Society Meeting, Kansas City,

KS

2012 – 1)”Fertility Preservation in Cancer Patients;” 2) “Aging, Ovarian Reserve, Reproduction;” KU Wichita

campus

56

Committees KUMC Member, KU Faculty Council, OB/GYN Education Committee, Dept. Resident Research Committee, KUCC

Cancer Prevention Research Program Other Faculty Mentor, Wahl Society Editorial and Grant Reviews Editorial Board Member, Public Library of Science (PLoS ONE), Journal of Assisted Reproduction and

Genetics (JARG), Fertility and Sterility

Ad hoc Reviewer, Human Reproduction, Reproductive Biology and Endocrinology, Journal of Assisted Reproduction and Genetics (JARG), Journal of Clinical Endocrinology and Metabolism, Fertility and Sterility, Lancet Oncology, Nature Reviews Clinical Oncology, PLos ONE, US Obstetrics & Gynecology

Academic Honors Super Doctors (REI, KS) Gregory S. Kopf, Ph.D, Associate Vice Chancellor for Research Administration Executive Director, KUMC Research Institute Professor, Department of Molecular and Integrative Physiology Member, IRHRM Internal Advisory Board Member, Center for Reproductive Sciences My academic research interests have focused on invertebrate and mammalian fertilization, gametogenesis, and early events of egg activation. Much of this work was carried out as a faculty member in the Dept. of Obstetrics and Gynecology at the University of Pennsylvania School of Medicine. Prior to coming to KUMC, I was Assistant Vice President and Interim Vice President, Discovery at Wyeth Pharmaceuticals. In this capacity, I directed a drug discovery group responsible for the identification, validation and patenting of various targets for contraception and other disease areas in women's health (osteoporosis; reproductive disorders; urinary incontinence). My group was also involved in HTS assay development, compound testing, and lead optimization and I was also involved in various in- and out-licensing activities for Wyeth. Meetings Attended

2012 – Vanderbilt University Postdoctoral Research Symposium, Nashville, TN

Committees National Member, CTSA Regulatory Knowledge Key Function Committee, CTSA Drug Discovery & Development

Consortium Subcommittee, Board of Directors, Kansas BIO, Public Policy Subcommittee, Kansas BIO

Working Group for Planning Meeting on Contraception; Board on Children, Youth and Families at the Institute of Medicine and National Research Council of the National Academies, Multipurpose Technologies Mapping/Matrix/Pipeline Gap Analysis Working Group (NIAID; DAIDS; DMID)

57

Chairman, Long Acting Injectable Contraceptive (LA6+) Technical Advisory Group, FHI360

Advisor, UCSF Bixby Center for Global Reproductive Health Contraceptive Advocacy Project

KUMC Member, Board of Directors, Kansas University Medical Center Research Institute Member, Executive, Finance, Research, Nominating, Research Properties Subcommittees, Kansas University Medical Center Research Institute Other Member, Board of Directors, Vasogenix Pharmaceuticals Member, Board of Directors, Immunogenetix Member, Board of Directors, Kansas University Center for Technology Commercialization Member, Finance Subcommittee, Kansas University Center for Technology Commercialization

Editorial and Grant Reviews Advisory Board, Zoological Science Associate Editor, Zygote Seminars Presented 2012 – Keynote Speaker, Vanderbilt University Annual Postdoctoral Research Symposium Nashville, TN

Adam J. Krieg, Ph.D. Assistant Professor Department of Obstetrics and Gynecology Member, Center for Epigenetics and Stem Cell Biology The primary focus of our laboratory is the study of the transcriptional mechanisms activated in response to reduced cellular oxygen, or hypoxia. A significant proportion of hypoxic gene expression is mediated by the Hypoxia Inducible Factors (HIFs), transcription factors that induce the expression of genes important for anaerobic metabolism, blood vessel recruitment, cell motility, and stem cell maintenance. Of particular interest is the hypoxic expression of several histone demethylase genes by the HIFs. Since histone demethylases affect gene expression by modifying the chromatin of target genes, hypoxic regulation of this phenomenon creates an intriguing link between cellular microenvironment, HIF activation, and downstream cascades of gene expression that could prolong the initial cellular response to hypoxia. We are currently studying the functional consequences of hypoxic histone demethylase expression in the context of normal cell biology and in disease states ranging from cancer to intrauterine growth restriction. Meetings Attended 2012 – Keystone Symposia Conference: Advances in Hypoxic Signaling: From Bench to Bedside, Banff,

Alberta, Canada

58

Committees Member, 2013 Greenwald Symposium Planning Committee

Seminars Presented 2012 - “Hypoxic Regulation of Histone Demethylases Mediate Cellular Response to the Tumor

Microenvironment,” KU Cancer Center Cancer Biology Seminar, University of Kansas Medical Center, Kansas City, KS

2012 – “Hypoxic Regulation of Histone Demethylases Mediate Cellular Response to the Tumor Microenvironment,” KUMC Dept. of Biochemistry Seminar, University of Kansas Medical Center, Kansas City, KS

2012 – “Analysis of Histone Demethylase Activity in Hypoxic Cancer Cells,” (Research Presentation) Center for Epigenetics and Stem Cell Biology, KUMC Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS

2012 – “Differential gene expression in the decidua of recurrent pregnancy loss patients,” KUMC Institute for

Reproductive Health and Regenerative Medicine Annual Retreat, Brio Restaurant, Kansas City, MO

Trainees:

Lei Qiu - Graduate Student Yaqiong Wang – Graduate Student Kelsey Hampton – Graduate Student Ying Mu – Masters and Undergraduate Student Jacob New – Summer Student Judith Chapman - Postdoctoral Fellow Sacha A. Krieg, M.D., Ph.D., FACOG Assistant Professor Director of the Recurrent Pregnancy Loss Program Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology Member, Center for Reproductive Sciences Pregnancy loss is the most common complication of human pregnancy, impacting approximately 10-15% of all human conceptions. While for most fertile couples miscarriage is a sporadic event, approximately 1-5% of fertile couples suffer from recurrent pregnancy loss (RPL), having a profound impact on their fertility and emotional well-being. Although RPL has been attributed to several hematologic, anatomic, hormonal and genetic defects, more than 50% of cases remain classified as having unknown etiology. My research interests focus on this subgroup of patients, in particular patients who are at risk for having endometrial causes of early pregnancy loss. To date we have investigated decidual contributions to recurrent miscarriage via microarray analysis. We are beginning to further characterize dysregulated gene products both at a molecular level and in an in vitro model of trophoblastic invasion. Committees National Member, ASRM Scientific program and abstract review committee

59

Editorial and grant reviews Ad hoc reviewer, Reproductive Biology and Endocrinology, Gynecological Endocrinology, Journal of Assisted

Reproduction and Genetics, Human Reproduction Editorial board member, Medscape Journal of Medicine, Women’s Health, Obstetrics and Gynecology Seminars Presented May 2012 – “Infertility and Recurrent Pregnancy Loss,” Assurant Insurance April 2012 – “Recurrent Pregnancy Loss,” Kansas City Infertility Awareness April 2012 – “Aging and Fertility,” KUMC “Lunch and Learn” February 2012 – “Think you can’t get pregnant? Try again, study says,” Reuters Health News Interview January 2012 – “The Down and Dirty of Women’s Health,” Kansas City Professional women’s group Trainees Sarah Keller – Graduate Student Melissa A. Larson, Ph.D, Research Assistant Professor Department of Molecular and Integrative Physiology Technical Director, Transgenic and Gene-Targeting Institutional Facility Member, Center for Reproductive Sciences Dr. Larson serves as the Technical Director of the Transgenic and Gene-Targeting Facility, a shared, core, research support facility providing a centralized service for the production of transgenic and gene-targeted mice for the investigators of KUMC and the surrounding Kansas City research community. The facility provides services that include the generation of transgenic mice by pronuclear microinjection, generation of chimeric mice by blastocyst injection, targeting of embryonic stem cells, sperm and embryo cryopreservation and genotyping of mice. The facility provides consultation on new projects and training and demonstration in microinjection, embryo transfer surgeries and ES cell culture. The lab is also investigating new technologies that improve the production and maintenance of gene-modified mice, as well as introducing new services and technologies to our users. In addition, Dr. Larson's laboratory has conducted experiments to determine whether a novel recombinase, Dre, functions in mice. Demonstration of its action has provided another tool to manipulate the mouse genome in vivo. Meetings Attended 2012 – National Meeting for the American Association for Laboratory Animal Science, Minneapolis, MN Committees KUMC Member, Institutional Animal Care and Use Committee, Women in Medicine and Science Mentoring

Committee, Programmatic Sub-Committee of the IACUC Chair-elect, Women in Medicine and Science Mentoring Committee; Member-Executive Council

60

Editorial and grant reviews Ad hoc reviewer, Journal of Reproduction, Fertility and Development Seminars Presented 2012 – “Updates from the Transgenic Facility,” Center for Epigenetics and Stem Cell Biology, Institute for

Reproductive Health and Regenerative Medicine, Kansas City, KS 2012 – “Transgenic and Gene-Targeted Mice: Services of the Transgenic Facility,” Class Presentation for

Clinical Laboratory Sciences 730, Current Issues in Biotechnology, University of Kansas Medical Center, Kansas City, KS

2012 – “Making Transgenic and Gene-Targeted Mice and Other Fun Stuff We Can Do!” Kansas City branch of

the American Association for Laboratory Animal Science, Kansas City, KS Eugene Lee, M.D. Assistant Professor Department of Obstetrics and Gynecology Member, Center for the Developmental Origins of Health and Adult Disease Dr. Lee has recently completed his training in maternal-fetal medicine at the University of Colorado, and is pursuing a career in academic medicine under the WRHR training grant here at KUMC. The topic of his proposal is abnormal uterine contractility which manifests clinically as labor dystocia. The processes of cervical ripening, uterine activation, and electrophysiologic signaling and the coordination of them are topics of interest.

Joan Lewis-Wambi, Ph.D. Assistant Professor Department of Cancer Biology Member, Center for Epigenetics and Stem Cell Biology Despite the benefits of endocrine therapies such as tamoxifen and aromatase inhibitors in treating estrogen receptor alpha (ERa)-positive breast cancer, many tumors eventually become resistant. Identifying the underlying cellular and molecular mechanisms responsible for endocrine resistance remains a critical and immediate need. Our laboratory is interested in identifying novel pathways of endocrine-resistance in breast cancer and using that knowledge to help develop alternative treatment options for patients with endocrine resistant and metastatic disease. One of the projects in our lab involves studying the role of pigment epithelium-derived factor (PEDF) in the development of endocrine resistance in breast cancer. PEDF is a 50 kDa glycoprotein that belongs to the non-inhibitory serine protease inhibitor (SERPIN) superfamily but it does not inhibit proteases. PEDF was first discovered as a factor secreted by retinal pigment epithelial cells, but later found to be expressed in several tissues including brain, spinal cord, eye, plasma, bone, prostate, pancreas, heart and lung. PEDF is a potent antiangiogenic factor that is showing promise as a potential anticancer agent. It is considered a potential tumor suppressor because its expression is high in normal tissues but is loss or significantly reduced in various types of malignancies. Loss of PEDF expression in breast cancer tissue has been shown to be associated with disease progression and poor survival, however, the role of PEDF in antihormone resistance and its potential as a therapeutic target for preventing and/or reversing endocrine resistance in breast cancer is not known. Recently, my laboratory found that PEDF mRNA and protein levels were dramatically reduced in antihormone resistant breast cancer cells and that stable reexpression of PEDF in the resistant cells resensitized them to tamoxifen. In addition, tissue microarray studies of primary and recurrence tumors from patients (N=109) who initially responded to tamoxifen and subsequently failed, revealed that PEDF protein was reduced in ~52.8% of

61

recurrence tumors compared to primary tumors. Furthermore, we have found that recombinant PEDF is capable of inhibiting the growth of endocrine resistant breast cancer cells in athymic mice and that PEDF also inhibits the growth of ER-negative breast cancer cells. Based on these findings, we hypothesize that PEDF silencing is a novel mechanism for the development of endocrine-resistance in breast cancer and its expression influences the metastatic potential of ERa-expressing tumors and their ability to respond to antihormonal therapy. We plan to use lentiviruses to stably overexpress PEDF in endocrine resistant breast cancer cells to help address the following questions: how does loss of PEDF expression confer resistance to endocrine therapy? 2) How is PEDF expression regulated in breast cancer cells and what role (if any) does the estrogen receptor play in PEDF regulation? 3) What is the mechanism of action of PEDF in endocrine resistant breast cancer cells in vitro versus in vivo? Another area of research in our laboratory involves investigating the mechanism by which estrogen paradoxically induces apoptosis in endocrine resistant breast cancer cells. Specifically, our laboratory has developed two breast cancer cell lines called, MCF-7:5C and MCF-7:2A, which were clonally selected from parental MCF-7 human breast cancer cells following long-term (>1 year) estrogen deprivation. Unlike MCF-7 cells which require estrogen/estradiol to grow and whose growth is inhibited by tamoxifen and other antiestrogens, MCF-7:5C and MCF-7:2A cells are hormone independent and are resistant to tamoxifen and aromatase inhibitors and these cells undergo apoptosis in the presence of physiologic levels of 17b-estradiol (E2) in vitro and in vivo. Investigation into the mechanisms of E2-induced apoptosis in MCF-7:5C and MCF-7:2A breast cancer cells reveals that it is a mitochondrial mediated event involving the BCL-2 family proteins and endoplasmic reticulum stress. More recently, we have found evidence to suggest that the phospholipid scramblase 1 (PLSCR1) protein might be a novel mediator of estrogen-induced apoptosis in our endocrine resistant cells. PLSCR1 is a member of the PLSCR gene family that has been implicated in multiple cellular processes including movement of phospholipids, gene regulation, immuno-activation, and cell proliferation/apoptosis. PLSCR1 and other family members (PLSCR2, PLSCR3, PLSCR4, and PLSCR5) are highly induced by interferons (IFNs) such as INF-a, IFN-b, and to a lesser extent INF-g and their induction is associated with apoptosis. We have found that suppression of PLSCR1 using siRNA completely blocks the ability of the resistant cells to undergo apoptosis in the presence of E2 as well as other apoptosis-inducing agents. Furthermore, we have found that calcium mobilization is dramatically altered in these cells due to suppression of PLSCR1. Currently, we are investigating how PLSCR1 regulates E2-induced apoptosis in MCF-7:5C cells and what role (if any) the other family members (i.e. PLSCR2-PLSCR5) play in this process. We are also studying the clinical significance of PLSCR1 expression in invasive breast cancer and whether PLSCR1 protein has therapeutic benefits in other types of cancers such as triple negative and inflammatory breast cancer. Meetings Attended 2012 – American Association for Cancer Research (AACR), Chicago, IL Committees Departmental Member, Cancer Center Seminar Series Committee Editorial and Grant Reviews Editor, Journal of Breast Cancer Research Grant reviewer, California Breast Cancer Research Program (CBCRP), Etiology, Prevention & Progression

Section

62

Seminars Presented 2012 – “Stat-1 activation enhances phospholipid scramblase 1-mediated apoptosis in endocrine resistant

breast cancer cells,” Temple University, Philadelphia, PA 2012 – “The role of pigment epithelium derived factor (PEDF) in breast cancer progression and endocrine

resistance,” City College of New York (CCNY), Dept. of Pharmacology, New York City, NY 2012 – “Modeling Endocrine Resistance: Consequences of long-term estrogen deprivation therapy for breast

cancer and alternative treatment options for resistant disease,” The University of Kansas Medical Center, Kansas City, KS

2012 – “A novel therapeutic approach to treat endocrine resistant breast cancer,” University of Missouri, Dept.

of Pharmacology, Kansas City, KS Trainees Philipp Maximov – Graduate Student Rifat Jan – Graduate Student Charlene Brewer – Undergraduate Student Echi Onuoro – Undergraduate Student Leena Thomas – Undergraduate Student Benyi Li, Ph.D. Associate Professor Director of Basic Science Research Department of Urology Member, Center for Epigenetics and Stem Cell Biology Critical to the prevention and treatment of urologic cancers is basic science research. In the Urologic laboratory at Kansas University Medical Center we are investigating the various causes of cancer. Dr. Benyi Li, M.D., PhD. received training in molecular biology and prostate cancer research in China, Japan, and Baylor College of Medicine, Houston, TX. His interests focus on molecular pathways which cause prostate cancer to grow and metastasize. In addition, he has created an inducible Akt system to allow further study of the molecular mechanisms behind prostate cancer. Committees KUMC Member, International Affairs Committee, PhD Advisory Committee for Xing Zeng, Ruibao Chen and Jessica

Williams Editorial and Grant Reviews Ad hoc reviewer, Cancer Letter, Cancer Investigation, Nuclear Acid Therapy, Cancer Research, PLoS One,

Biometals Ad hoc grant reviewer, DoD Seminars Presented 2012 – “The role of p110beta kinase in AR signaling and Histone Modification,” Stower Institute for Medical

Research, Kansas City, MO

63

2012 – Conference Panel Moderator: “AUA abstract highlights,” World Chinese Urological Society Annual

Meeting, Atlanta, GA 2012 – “L-type Calcium channel in prostate cancer progression,” Annual meeting of Ningbo City Branch,

Chinese Urological Association, Ningbo, China 2012 – “Exosomes as novel therapeutic agents in human cancers,” Guangdong Medical College Hospital,

Zhanjiang, China 2012 – “Nanotech-based Tissue Specific Targeting of P13K/p110beta for Prostate Cancer Therapy,” KUCC

Monthly Seminar series, KUMC, Kansas City, KS 2012 – “L-type Calcium channel gene CACNA1D in AR signaling and prostate cancer progression,” The

Annual Meeting of Chinese Urological Association, Guangzhou, China 2012 – “Nanotech-based molecular targeting for prostate cancer management,” Dept. of Pathology, Zhejiang

University Shaoxin Hospital, Shaoxin, China Trainees Yan Huang – Visiting Fesearch Fellow Ruitao Zhang – Postdoctoral Fellow Yuzhe Tang – Visiting Research Fellow

Xiaogang Li, Ph.D. Associate Professor Department of Internal Medicine-Nephrology & Hypertension Member, Center for Epigenetics and Stem Cell Biology The primary focus of my research is to understand the molecular pathogenesis of autosomal dominant polycystic kidney diseases (ADPKD) and to translate these findings for ADPKD treatment. To this end, my lab has two emphasis areas of research: 1. TNF-alpha signaling and apoptosis signaling in ADPKD. In the past we found that a tumor necrosis factor-α-mediated pathway promoting cyst and treatment of Pkd2+/- mice with the TNF-α inhibitor etanercept prevented cyst formation. This study uncovered the connection among TNF-α signaling, polycystins and cystogenesis. (Published in Nature Medicine, 2008). Recently, we found that a second mitochondria-derived activator of caspase (Smac)-mimetic, induces TNFα-dependent cystic renal epithelial cell death specifically, leading to the removal of cystic epithelial cells from renal tissues, thus, preventing cyst formation. Our current study helps to clarify the role of apoptosis in the regulation of cyst size and in a larger sense may open a new approach to target renal cysts and prevent their endless expansion by the administration of Smac-mimetics, which encourages a paradigm shift from current efforts that focus on normalizing cell function in cystic epithelial cells to directly targeting these cells for removal. (Submitted to JASN). 2. Epigenetics and ADPKD. We have conducted extensive research on the function of histone deacetylases (HDACs) and histone methyltransferases (HMTs) in ADPKD. In the past, we found that: i) Polycystin-dependent fluid flow sensing targets histone deacetylase 5 to prevent the development of renal cysts (Xia et al., Development, 2010); ii) Inhibition of histone deacetylases targets the transcription regulator Id2 to attenuate cystic epithelial cell proliferation (Fan et al., Kidney International, 2012); iii) HDAC6 regulates epidermal growth factor receptor (EGFR) endocytic trafficking and degradation in renal epithelial cells (Liu et al., Plos One. 2013). We are now focusing on: i) Vitamin B3 prevents cyst formation through Sirt1 mediated cyst epithelial cell proliferation and apoptosis (Submitted to JCI after revision); ii) SIRT2 regulates ciliogenesis

64

and contributes to loss of polycystin-1 mediated abnormal centrosome amplification (Submitted to Nature Communication under review); iii) Aberrant histone and/or protein methylation, which is regulated by heat shock protein 90, and/or DNA methylation of CpG island containing promoters leads to permanent silencing of genes in both physiological and pathological contexts in cystic epithelial cells. Committees National Member, Research Committee of the American Society for Pediatrics Nephrology Editorial and Grant Reviews Ad hoc reviewer, International Journal of General Medicine, UpToDate Inc. Nephrology, PloS One, Kidney

International Editor, Frontiers in Renal and Epithelial Physiology Trainees Lucy X. Fan – Postdoctoral Julie X. Zhou – Postdoctoral Wei Liu – Postdoctoral Anne Manzardo, Ph.D., M.S.C.R. Assistant Professor Department of Psychiatry & Behaviorial Sciences Member, Center for Epigenetics and Stem Cell Biology Dr. Ann Manzardo is a Behavioral Pharmacologist who specializes in the field of addiction research. She has advanced training in biostatistics and expertise in the epidemiology of alcoholism. Dr. Manzardo has established a translational research program in the Department of Psychiatry and Behavioral Sciences through which she has directed funded research projects on alcoholism spanning multiple scientific disciplines: clinical, epidemiological and genetic. Her research has emphasized the study of gender disparate effects of inherited biological and neurodevlopmental risk factors for the development of alcoholism. And the role of thiamine deficiency in alcohol abuse behaviors. Dr. Manzardo has continued the tradition of collaborative study of the Danish Perinatal Cohort in the Department of Psychiatry with her studies of the influence of premature birth on the development of alcoholism. Dr. Manzardo has also recently expanded her research interests to include bioinformatics as it is applied to the genetics of alcoholism. Committees KUMC Member, Faculty Council, Human Subjects Committee Other Psychiatry Foundation Board of Trustees New South Wales Tissue Research Center Tissue Review Panel

65

Editorial and Grant Reviews Editorial Board, World Medical Laboratory Research, World Medical Imaging Research Grant Reviews New South Wales Tissue Research Center Tissue Review Panel Seminars Presented 2012 – “Nutritional Deficiencies in Alcoholism,” Lunar Society, Kansas City, KS 2012 – “Nutritional Deficiencies in Alcoholism,” Alcohol Medical Scholars Program Conference, Carmel, CA 2012 – “The Effect of Benfotiamine Treatment in Alcohol Dependence,” Department of Molecular & Integrative

Physiology, University of Kansas Medical Center, Kansas City, Kansas 2012 – “Cytokines in Autism,” Luminex Scientific Symposium, Monte Carlo, Monaco 2012 - “The Effect of Benfotiamine Treatment in Alcohol Dependence,” Psychiatry Grand Rounds, University of

Kansas Medical Center, Kansas City, Kansas 2012 - “The Effect of Benfotiamine Treatment in Alcohol Dependence,”Department of Pharmacology,

Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas Academic Honors 2012 New Investigator Award, NIH Rare Disease Clinical Research Network (RDCRN) for Prader-Willi,

Angelman and Retts syndrome Alcohol Medical Scholar Program, 10/2010 – 5/2012 Clifford W. Mason, Ph.D. Research Assistant Professor Department of Obstetrics and Gynecology Member, Center for the Developmental Origins of Health and Adult Disease The core of our research focuses on the pathopharmacology of the maternal-placental-fetal unit. Pregnant women often need to take medications to treat diseases including those induced during pregnancy. As a result, a major concern arising from the use of medications by pregnant women is the transfer of drugs across the placenta barrier. Our data indicate there are changes in drug transport proteins in placenta of women with normal and abnormal pregnancy. Changes in placental transporters could result in altered fetal drug exposure leading to drug toxicity. Our research addresses three core questions. First, how do pathophysiological responses to pregnancy disease affect placental drug transporters? Second, do pathological changes in transporter expression levels correlate with placental drug transfer, and therapeutic outcome? Finally, what are the regulatory pathways that drive transporter expression and can pharmacoresistance to drugs be overcome by targeted inhibition of proteins within these pathways? The results will help predict how pathophysiological responses to pregnancy diseases alter placental transfer and therapeutic efficacy or toxicity of drugs. We are also interested in understanding the mechanism(s) of myometrial activity as it pertains to labor. Specifically, we focus on molecular differences in pathways responsible for smooth muscle contraction during term and preterm labor.

66

Meetings Attended 2012 – Society for Gynecological Investigation, San Diego, CA Seminars Presented 2012 – “The Role of Purkinje Cell Protein 4 on Calcium Signaling and Smooth Muscle Contraction,” The Third

Affiliated Hospital, Sun Yat-sen University and Jinan University School of Medicine, China

Nancy A. Muma, Ph.D. Professor and Chair Department of Pharmacology and Toxicology-University of Kansas, Lawrence Member, Center for the Center for Reproductive Sciences Our research is directed toward an understanding of the mechanisms involved in neuropsychiatric and neurodegenerative disorders. Currently, we are examining the mechanisms regulating adaptations in serotonin receptor signaling as new targets for therapeutic intervention. Serotonin receptor signaling is altered by a number of drugs used to treat mood disorders such as depression and anxiety and psychiatric disorders including schizophrenia. For example, we recently found that a novel estrogen receptor system modifies serotonin receptor signaling and is a potential target for the treatment of depression and other mood disorders associated with the onset of menopause. Meetings Attended 2012 - “The role of GPR30 in the estradiol-induced desensitization of hypothalamic 5-HT1A receptor signaling,”

Winter Conference on Brain Research, Snowbird, Utah

2012 - “GPR30 plays a primary role in estradiol-mediated attenuation of 5-HT1A receptor signaling and in potentially accelerating the effects of SSRIs,” Serotonin Club Meeting, Montpelier, France

2012 – “Targeting Calmodulin to alter Transglutaminase in Huntington’s disease,” Gordon research conference

on Transglutaminases in Human disease Committees KU Member, School of Pharmacy Administrative Committee, School of Pharmacy Executive Committee, School of

Pharmacy Academic Misconduct Committee, University of Kansas Search Committee for Animal Care Veterinarian

Departmental

Member, Department of Pharmacology and Toxicology Faculty Search Committee

Editorial and Grant Reviews Editorial Board Member, Journal of Neuropathology and Experimental Neurology, Journal of Experimental

Pharmacology, Frontiers in Alzheimer’s Disease Ad hoc Reviewer, Psychoneuroendocrinology, International Journal of Neuropsychopharmacology,

Neuropsychopharmacology, Biochemical Pharmacology, Brain Pathology, Journal of

Neuroendocrinology

67

Seminars Presented

2012 - “Estrogens Modulate Serotonin Receptor Signaling in Rat Hypothalamus: Synergy with SSRIs,” The

Institute for Reproductive Health and Regenerative Medicine, University of Kansas School of Medicine

Trainees Zhen Mi – Graduate Student Carrie McAllister – Graduate Student Quan Li – Research Assistant Professor Paul Kimball – Research Associate Ajay K. Nangia, M.B., B.S. Associate Professor Clinical Director of Andrology Department of Urology Member, Center for Reproductive Sciences Dr. Nangia's interests in the field of urology are micro-surgical reconstruction including vasectomy reversal, male infertility and male sexual/reproductive dysfunction. He is actively involved with research in male contraception, as well as the study of vitamin D in sperm/testicular physiology. Meetings Attended 2012 – “Male and Female Public Opinion Regarding a Possible Male Contraceptive Pill,” ASRM National

Meeting, San Diego, CA 2012 – “Sertoli Cell Tumor of the Testis,” “33 year old male with suprapubic gun shot wound,” South Central

AUA Conference, Colorado Springs, CO Committees National Member, American Urological Association (AUA) (Public Media, Plenary Program Planning, Men’s Health

Initiative and Health Policy Committees), Society for the Study of Male Reproduction Nominating Committee, American Society of Reproductive Medicine Resident Education Committee, South Central Section of the American Urological Association (Health Policy and Resident Prize Paper Committees), Kansas Urological Society Program Planning Committee, American Board of Urology Written Examination Committee, Clerkship Advisory Committee, Dartmouth-Hitchcock Medical Center, Resident Prize Paper Committee, South Central Section of the AUA, Abstract Review Committee, American Society of Reproductive Medicine, 2013 ASA Program Planning Committee

KUMC

Member, Student Promotions Subcommittee, Election Committee, EMR Advisory Committee, Faculty Council,

Internet/Web Committee, Indian Creek Governance Committee, 2013 Greenwald Symposium Planning Committee

Departmental Member, Ethics Committee, Urology Staff Liason, Urology Resident Education Committee, Urology Residency

Committee

68

Editorial and grant reviews Editorial Board Member, Journal of Andrology, Journal of Assisted Reproductive Genetics, Journal of Men’s

Health Ad hoc Reviewer, Journal of Urology, Urology, Fertility and Sterility, Asian Journal of Andrology, Canadian

Journal of Urology, Journal of Endourology, Journal of Sexual Medicine, Human Reproduction, American Family Physician, British Medical Journal Online

Grant Reviewer, AUA Foundation Grant Review Committee Seminars Presented 2012 – “Pediatric Urological Diseases and Role in Adult Male Infertility,” Minnesota Urological Society,

Minneapolis, MN 2012 – “Office Evaluation for Male Infertility,” Indian Urological Association at the National AUA

2012 – “Forging Relationships with the CDC in a Cost-Constrained Environment,” American Urological

Foundation Research Forum 2012 – “Male Infertility – What All Family Planning Providers Need to Know,” National Reproduction

Conference – Family Planning and Title X Providers 2012 – “Male Infertility and Hypogonadism,” American Association of Clinical Endocrinologists National

Meeting 2012 – “Life After Prostate Cancer,” Wellness Program, University of Kansas Medical Center, Kansas City, KS 2012 – “Male Infertility,” Patient Infertility Awareness Group, Kansas City Infertility Association, Kansas City,

KS 2012 – “Male Reproduction and Men’s Health and Andropause,” Taking on the Challenges of Aging: Men’s

Health Overview, University of Kansas Medical Center, Kansas City, KS 2012 – “Men’s Preventative Health Lunch and Learn,” City of Lenexa Municipal Services, Lenexa, KS Academic Honors Marquis Who’s Who in America Award Kansas City Business Journal – Top Doctors in Kansas City Award Co-Program Chair, American Society of Andrology Annual Meeting Team Leader, 2013 AUA National Meeting Infertility Program Planning Committee Warren B. Nothnick, Ph.D., H.C.L.D. Professor Department of Molecular and Integrative Physiology Member, Center for Reproductive Sciences

69

The uterus is a vital organ for the successful propagation of all higher species. Understanding the molecular mechanisms that contribute to the development and subsequent function of the uterus are absolutely essential for successful reproduction to occur. It is well established that complex interactions among biological mediators dictate the normal pattern of uterine development and that disruption of these factors plays a causative role in uterine abnormalities, disease and infertility. Our research focuses on three major areas: 1) the role of microRNAs (miRNAs) in the pathophysiology of the female disease, endometriosis and the use of miRNA therapy in the treatment of this disease, 2) the role of miRNAs in uterine decidualization and 3) the identification and development of novel, estrogen-sparing targets for endometriosis treatment. Collectively, the research in my laboratory focuses on examining the mechanisms which regulate normal uterine development and function, identifying those factors which contribute to these mechanisms and understanding how alterations in these mechanisms lead to uterine diseases such as endometriosis and recurrent pregnancy loss/infertility. The goal of the research conducted in my laboratory is to better our understanding of the pathophysiology of these uterine diseases and in turn develop novel diagnostic/prognostic markers and therapeutic agents for their treatment. Meetings Attended 2012 - American Society for Reproductive Medicine, Endometriosis Special Interest Group, 68th Annual

Meeting of the American Society for Reproductive Medicine, San Diego, CA 2012 – The 9th Annual Gilbert S. Greenwald Symposium on Reproduction, Kansas City, KS Committees Departmental Member, Lacey Luense, Malinda Algaier, Wei-ting Hung, Nairita Roy (Doctoral candidates) KUMC Member, IRHRM 3rd Floor Enrichment Committee, Animal Care & Use Program Task Force, Executive Team,

Animal Research Protection Program

Chairman, Institutional Animal Care and Use Committee (IACUC) National Member, Abstract Review Committee, American Society for Reproductive Medicine, Endometriosis Special

Interest Group, 68th Annual Meeting of the American Society for Reproductive Medicine Editorial and grant reviews Editorial Board, Reproductive Biology and Endocrinology Member, Board of Reviewing Editors, Biology of Reproduction Ad hoc reviewer - American Journal of Obstetrics and Gynecology, Biology of Reproduction, Cancer,

Current Medicinal Chemistry, Current Opinion in Molecular Therapeutics, Domestic Animal Endocrinology, Endocrine Reviews, Endocrinology, Expert Reviews of Obstetrics and Gynecology, European Journal of Cell Biology, European Journal of Obstetrics and Gynecology and Reproductive Biology, Fertility and Sterility, Frontiers in Bioscience, Gynecologic and Obstetric Investigation, Human Reproduction, Journal of Assisted Reproduction and Genetics, Journal of Biological Chemistry, Journal of Clinical Endocrinology and Metabolism, Life Sciences, Molecular Endocrinology, Molecular Human Reproduction, Molecular Reproduction and Development, Obstetrics and Gynecology, Reproduction, Reproductive Biology and Endocrinology, Reproductive Sciences, Theriogenology, Trends in Molecular Medicine, Women’s Health in Primary Care

70

Ad hoc reviewer, NIH/HDM Initial Review Group, Nursing and Related Clinical Sciences (NRCS) Ad hoc reviewer, University of Nebraska Medical Center, Olson Center for Women’s Health Pilot Grant

reviewer Ad hoc reviewer, NIH/EMNR Initial Review Group, Integrative and Clinical Endocrinology and Reproduction

Study Section (ICER) Ad hoc reviewer, NIH/NICHD, Specialized Cooperative Centers Program in Reproduction Research Academic Honors Inaugural Member, College of CSR Reviewers, Center for Scientific Review, National Institutes of Health Ambassador, World Endometriosis Society Arindam Paul, Ph.D. Research Assistant Professor Department of Pathology and Laboratory Medicine Center for Epigenetics and Stem Cell Biology

My long-term research interest is on some key problems in the area of developmental biology and cancer biology described below.

Neuronal commitment of rat embryonic stem (ES) cells as well as induced pluripotent (iPS) cells. In this project, we are trying to develop serum free in vitro culture conditions to generate neural progenitors and terminally differentiated neural cell lines such as motor neurons, oligodendrocytes, astrocytes from rat ES and iPS cells.

Development of Novel Therapeutic Strategy to prevent breast cancer metastasis of targeting different signaling mechanism. We are evaluating small molecule compounds targeting different signaling pathways for regulation of epithelial mesenchymal transition, invasion and metastasis of triple negative breast cancer cells using both in vitro assay systems and in vivo conditions

Identification of novel therapeutic targets to prevent Invasive Breast Cancer development. We are pursuing genome wide screening approach to identify novel regulators at various stages of invasive breast cancer development under in vivo conditions. The long term goal for this project is to develop small molecule compounds for future therapeutic modality to prevent invasive breast cancer development. Soumen Paul, Ph.D. Associate Professor Department of Pathology and Laboratory Medicine Member, Centers for Epigenetics and Stem Cell Biology, and Reproductive Sciences

During mammalian embryogenesis, a whole organism is developed from a single fertilized egg. So, one of the fascinating questions in biology is "How do cells adopt different cell fates? Research in our laboratory focuses on defining molecular mechanisms that regulate tissue-specific gene expression to orchestrate developmental and physiological processes. We are asking how transcriptional mechanisms that involve, transcription factors/cofactors, distinct epigenetic marks, and other chromatin-associated factors regulate chromatin structure and thereby regulate gene expression during developmental, and physiological processes as well as during pathological conditions.

71

One of our research interests is to define molecular processes that control the genesis of early cell lineages, their self-renewal, differentiation, and function. The first lineage decision during mammalian development is the establishment of trophectoderm (TE) and inner cell mass (ICM) lineages. These differentiation events begin during pre-implantation development when blastomeres are fated towards TE and ICM. TE develops into parts of the placenta, while the ICM forms embryonic and some extra-embryonic structures. To understand this early lineage commitment, we are using embryonic stem (ES) and trophoblast stem (TS) cells as model systems. We are also using transgenic mouse models to test our hypotheses.

Another area of our research interest is to dissect mechanisms to understand the molecular regulation of blood vessel formation (vasculogenesis and angiogenesis) and vascular cell (endothelial cell) specification and function. Therefore, to begin to dissect regulatory mechanisms of blood vessel formation, we are defining the transcriptional regulation of key genes during early vascular development and adult angiogenesis.

We predict that our research will contribute towards development of progenitor cells or new tissues for regenerative therapeutics including vascular tissue engineering. Our efforts will also contribute towards therapeutics that will promote endogenous regeneration. In addition, we expect to establish new modes of anti-angiogenic therapy during pathological conditions. Committees KUMC Member, Greenwald Symposium Organizing Committee Editorial and grant reviews Reviewer, PloS Genetics, Stem Cells, Journal of Biological Chemistry, Journal of Cell Sciences, PloS One,

Journal of Molecular and Cellular Cardiology, Placenta, Molecular Reproduction and Development, Reproduction, Journal of Assisted Reproduction

Seminars Presented January 2012 – “Molecular Controls of First Mammalian Cell Lineage Commitment,” University of Missouri,

Kansas City, KS November 2012 – “Transcriptional Regulators in Trophoblast Development,” NICHD: Collaborative research

group on embryo implantation, Bethesda, MD December 2012 – “Protein Kinase C Signaling and Breast Cancer,” Institute for Reproductive Health and

Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS Trainees

Nairita Roy – Graduate Student Pratik Home – Postdoctoral Biswarup Saha – Postdoctoral Ganeshkumar Rajendran – Postdoctoral Avishek Ganguly – Postdoctoral Biraj Mahato-Postdoctoral

Kenneth R. Peterson, Ph.D. Professor and Vice Chair Department of Biochemistry and Molecular Biology

72

Director, Center for Epigenetics and Stem Cell Biology Red blood cells carry oxygen to tissues and organs throughout the body and ferry waste carbon dioxide from them to the lungs for exhalation. Hemoglobin is the molecule in red blood cells responsible for this transport

and is comprised of two -like globin chains, two -like globin chains and four heme molecules. Many diseases of red blood cells, termed hemoglobinopathies, have been described. Sickle cell disease (SCD) affects red cell shape and renders them ineffective; resulting in anemia along with attendant complications. SCD is gene- -

globin gene. A second disease of these cells, -thalassemia, also causes anemia. -thalassemias result from

an array of mutations in the -replacement of the mutant globin gene and help cure these disorders.

The human -globin locus consists of five functional -like globin genes, all of which serve as the -chain in the

hemoglobin molecule during different stages of development. The -globin gene is expressed in the primitive

yolk sac during the first six weeks of gestation; the G - and A -globin genes are transcribed in the fetal liver from

the sixth week to shortly after birth; and the -globin gene) is

expressed in bone marrow soon after birth for the duration of life. The globin and -globins are silenced in

the adult. Introducing an active fetal -globin gene in the adult by bone marrow transplantation to substitute for

a defective adult -globin gene is one goal of current gene therapy efforts. Realizing this goal requires understanding the molecular mechanisms that regulate globin gene switching. Our laboratory is focused on

the cis- and trans-control of human -like globin gene expression during development; that is, the identification and characterization of DNA elements and transcription factors regulating globin synthesis via interaction of the proteins with these sequences. A major regulatory motif of this class is the locus control region (LCR). The mechanisms by which LCRs function are largely unknown, but it is becoming clear that they are important

regulatory elements for developmental control of gene expression, not only for the -globin locus, but for other mammalian loci as well. Mechanisms underlying the developmental regulation of globin gene switching that are under analysis in the lab include: 1) the sequence determinants of LCR-globin gene interaction and their specificity, 2) the function of the LCR DNAse I-hypersensitive sites, 3) the physical structure of LCR-globin

gene contacts, 4) the role of chromatin domain boundary elements within the -globin locus, 5)

silencing - identification of both cis-acting silencer sequences and repressor proteins, and 6) activation of -globin gene expression - validation of putative, partially characterized protein activators, identification of novel transactivators, and testing of pharmacologic activators. Experimental systems involve analysis of transgenic

mice and cell lines produced with human -globin locus yeast artificial chromosomes ( -YACs) as transgenes, as well as the ancillary bacterial and yeast molecular biology procedures necessary to generate these mice

and cell lines. In addition, we have established unique cell lines from the bone marrow and fetal liver of our -YAC transgenic mouse lines using a novel system to enforce dimerization of growth signal transduction monomers into a functional molecule, resulting in multi-potential cell lines that proliferate, but do not differentiate. These will be used to select for novel hereditary persistence of fetal hemoglobin (HPFH)

mutations, fetal globin transactivator proteins and for screening small molecule inducers of -globin gene expression. A variety of cutting-edge molecular biology and biochemistry techniques are used to study cis-regulation, protein-DNA, and protein-protein interaction aspects of gene expression during development within these systems. Committees Departmental Member, Graduate Committee, Study Section, BMB KLSIC Space Advisory Committee Chairman, Admissions, Promotions and Tenure Committee

73

KUMC Chair, Institutional Human Stem Cell Research Oversight Committee Member, Dissertation/Thesis Committee-Elizabeth Dille, Subhash Naik, Yi Feng, Lu Chen, Valentine Agbor,

Shuai Lu, Mauricio Vargas Uribe, Rushi Trivedi, Julie Mitchell, Nairita Roy, Allen Chazelle, Zhen Zhang Member, Comprehensive Exam Committee-Julie Mitchell, Rushi Trivedi Associate member, KUCC, Risk Factors for Carcinogenesis Research Program, Executive Research Board,

Institute for Reproductive Health and Regenerative Medicine, Kidney Institute Internal Executive Advisory Committee, LCME Educational Resources Self-Study Committee, High Throughput Genomics Facility Advisory Committee, Advisory Board for Transgenic and Genetic Technologies Support Facility

Editorial and grant reviews Ad hoc reviewer – Blood; Blood Cells, Molecules and Disease, Journal of Molecular Biology, Journal of

Translational Medicine, PLoS One Guest Editor – Anemia, Sickle Cell Disease: Genetics, Cellular and Molecular Mechanisms and Therapies

special issue Ad hoc reviewer - Special Emphasis Panel/Scientific Review Group 2011/05 ZDK1 GRB-6 (M3) 1,

Hemoglobinopathies Program Projects Teleconference, NIDDK, 2011. Ad hoc member, NIH Molecular Genetics B (MGB) Study Section, 2012. International Reviewer, Portuguese Foundation for Science and Technology, 2012. Seminars Presented 2012 - “Fetal hemoglobin: regulation, activation and treatment for sickle cell anemia,” Department of

Biochemistry and Molecular Biology, University of Florida, Gainsville, FL. Trainees Flávia C. Costa – Postdoctoral Allen Chazelle – Graduate Student Nancy Stiles – Rotation Student Bennett Berning – Summer Research Medical Student Gaspar Maisonet – High School Student Julia Draper – Volunteer, Graduate of Rhodes College Tyler J. Stephenson – Summer Student

Brian K. Petroff, D.V.M., Ph.D. Associate Professor Division of Hematology/Oncology Department of Internal Medicine Member, Center for Reproductive Sciences Our research efforts have a dual focus 1) prevention of ovarian aging and chemotherapy and other ovarian toxicant induced infertility 2) prevention of breast and ovarian cancers through the characterization and antagonism of promising targets in human and animal chemoprevention trials. Early work showing ovarian

74

follicular loss in polluted environments (i.e. dioxin) mediated by the aryl hydrocarbon receptor was the underpinning for later work indicating that tamoxifen may protect against follicular loss from alkylating agent chemotherapy. I was recruited to the Department of Medicine, Division of Hematology/Oncology in 2004 to collaborate on translational aspects of early prevention trials in breast and ovarian cancer. This included development of the first model of nearly simultaneous ER+ breast and ovarian pre-cancer which would be invaluable in assessment of risk and mechanism of action biomarker modulation for Phase II human prevention trials. In this manner investigators would be able to preview the effects of an intervention on the ovary as well as breast in a model which is hormonally analogous to a late premenopausal woman. During the validation of this model with the Selective Estrogen Receptor Modulator (SERM), tamoxifen, it was noted that tamoxifen could protect against carcinogen (DMBA) induced ovarian follicle loss and hence aid in preserving fertility. The observations were repeated for cyclophosphamide. The breast and ovarian cancer model itself is being used in a multi-PI multi-project Komen Promise Grant awarded in 2010. I have been instrumental in overseeing the development of more advanced molecular techniques to characterize biomarker change in breast chemoprevention trials from the small amounts of material available from random peri-aerolar fine needle aspirations including proteomics and stem cell markers, lipidomics, hormone measurements and gene expression after and laser capture micro-dissection. I have held a leadership position in the developing University of Kansas Cancer Center as coleader of Cancer Prevention since 2008. Meetings Attended 2012 – “Autoimmune deficiency is associated with embryonic loss and generation of autoantibodies against the

uterus, placenta and embryo,” Annual Meeting of the Society for the Study of ReproductionState College, PA

2012 – “Reduction in Ki-67 in benign breast tissue of high risk premenopausal women with the SERM

acolbiphene,” Annual Meeting of the American Society of Clinical Oncology, Chicago, IL 2012 – “Effects of tamoxifen on markers of follicular health in rats treated with cancer chemotherapy,” “The

Autoimmune Regulator (AIRE) protects against infertility, reproductive tract inflammation and germ cell loss in male Balb/c mice,” Annual Greenwald Symposium, Institute for Reproduction Health and Regenerative Medicine, University of Kansas, Kansas City, KS

2012 – “Breast cancer risk biomarkers are associated with dietary intake and tissue content of n-3 polyuns

turated fatty acids,” Annual Meeting of the International Society for the Study of Fatty Acid Lipids, Vancouver, CA

Committees Departmental Member, Ad hoc research committee KUMC Member, Thesis committee, Pan Wang and Jonathan Fitzgerald, Institutional Animal Care and Use Committee,

Protocol Review and Monitoring Committee, KU Cancer Center, Shared Equipment Committee, KU Cancer Center, Leadership Council, KU Cancer Center, ACS Training Grant Advisory Board, KU Cancer Center

Editorial and Grant Reviews Ad Hoc Reviewer, Biology of Reproduction, Biomarkers, Biomarkers in Medicine, BMC Cancer, Breast Cancer:

Basic and Translational Research, Cancer Research,Carcinogenesis, Comparative Biochemistry and Physiology, Congenital Diseases and Environment, Current Medicinal Chemistry, Endocrine, Endocrinology, Fertility and Sterility, Journal of Assisted Reproduction and Genetics, Journal of

75

Endocrinology, Marine Drugs, Molecular and Cellular Probes, Reproduction,Reproductive Biology, Reproductive Biology and Endocrinology, Reproduction Nutrition and Development, Reproductive Toxicology, Toxicology, Toxicology and Applied Pharmacology, Toxicology Letters, Zoological Sciences

Editorial Board Member - Current Medicinal Chemistry North American Managing Editor - Reproductive Biology Grant reviewer, NICHD training grants Seminars Presented 2012 - “Effects of dioxins on female reproductive aging,” University of Illinois CVM 2012 - “Fetal origins of adult disease,” University of Illinois Trainees Pan Wang – Postdoctoral Jonathan Fitzgerald – Postdoctoral Fernando Pierucci-Alves, D.V.M. Research Assistant Professor Department of Anatomy & Physiology, Kansas State University Member, Center for Reproductive Sciences Sperm cells mature and acquire fertilizing capacity while transiting through the lumen of the male excurrent system, which is composed by the efferent ducts, epididymis and vas deferens. This ductal system is lined in its entirety by epithelial cells carrying out an array of secretory and absorptive mechanisms. These epithelial activities are required for successful spermatozoa maturation and fertility. Our research program is dedicated to understanding how growth factors, specifically transforming growth factor beta (TGFβ) regulate epithelial and sperm cell biology. By enhancing our knowledge of how this signaling pathway participates in the physiology of this organ system, we expect to form the basis for future diagnostics and treatment of infertility cases that remain primarily undiagnosed. Seminars Presented 2012 – “Cellular signaling by transforming growth factor beta in the male excurrent system - evaluating the

potential for high levels of signaling activity and possible impacts on reproductive function,” The 9th Annual Greenwald Symposium on Reproduction, KUMC Institute for Reproductive Health and Regenerative Medicine, Kansas City, KS

2012 – “Transforming growth factor beta (TGFβ) signaling in the male excurrent system - it occurs

physiologically and imbalances may impair human fertility,” Center for Reproductive Sciences Chalk Talk, KUMC Institute for Reproductive Health and Regenerative Medicine, Kansas City, KS

Evelyn A. Reynolds, M.D, Assistant Professor Division of Gynecologic Oncology Department of Obstetrics and Gynecology Member, Center for Reproductive Sciences

76

Evelyn A. Reynolds, M.D. is a specialist in gynecologic oncology and pelvic surgery. She completed her medical education and residency in obstetrics and gynecology at the University of Rochester School of Medicine in Rochester, NY. Following her residency, she completed a fellowship in pelvic surgery at Emory University in Atlanta, GA and held a faculty position at the same institution. She then joined the Mayo Clinic in Rochester, MN where she completed a fellowship program in gynecological oncology. Throughout her educational and professional career she has actively participated in cancer research. Dr. Reynolds has a particular interest in outcomes-based clinical research and the elimination of health disparities. Her current research involves the assessment of the treatment patterns of older women diagnosed with ovarian cancer. Seminars Presented 2012 – “Updates in the field of Gynecologic Oncology,” Missouri Society of the American College of

Osteopathic Family Physicians,” Winter Scientific Seminar, Independence, MO

Katherine F. Roby, Ph.D. Research Associate Professor Department of Anatomy and Cell Biology Member, Center for Reproductive Sciences The laboratory has two major areas of focus, ovarian biology and ovarian cancer. In regard to ovarian biology we are interested in understanding the cellular and molecular events controlling ovarian follicular development and ovulation. Specific interests focus on TNF, Src tyrosine kinase, and serum amyloid A. In regard to ovarian cancer, specific interests include defining the early molecular events associated with initiation of ovarian cancer, identification of targets for drug development, and the preclinical development of new therapies for the treatment of ovarian cancer. Ovarian cancer is primarily an intraperitoneal cancer and thus exhibits unique characteristics that can be exploited in treatment schemes. We have also extended our drug development/treatment studies to other cancers within the peritoneal cavity including disseminated colorectal cancer and mesothelioma. Committees KUMC Member, KUCC Biostatistics/Informatic Shared Resource (B/ISR) Advisory Committee, IRHRM Executive

Research Board, Institutional Animal Care and Use Committee, KU NanoScience Task Force Committee

Chair, Organizing Committee, Gilbert S. Greenwald Symposium on Reproduction and Regenerative Medicine Editorial and Grant Reviews Ad hoc reviewer, Biology of Reproduction; International Journal of Cancer; Reproductive Toxicology;;

Reproduction; Toxicological Sciences; Reproductive Biology & Endocrinology; Toxicology and Applied Pharmacology; Oncology Reviews; Reproductive Sciences; Journal of Endocrinology; American Journal of Physiology Endocrinology & Metabolism; International Journal of Nanomedicine; Journal of Ovarian Research; Current Cancer Drug Targets; Journal of Proteome Research; Immunological Investigations

Reviewer, Peer Reviewed Medical Research Program, Department of Defense, PRMRP, Nanomedicine for Drug Delivery Science

Reviewer, Department of Defense, Congressionally Directed Medical Research Programs Ovarian Cancer Research Program Review Panel Clinical and Experimental Therapeutics

77

Reviewer, Peer Reviewed Medical Research Program, Department of Defense, PRMRP, Nanomedicine for Drug Delivery Science

Reviewer, NIH, Population Sciences and Epidemiology: Chronic Disease Epidemiology and Genetics

Member, American Cancer Society Institutional Research Grant Committee M.A. Karim Rumi, M.B.B.S., M.S., Ph.D. Research Associate Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology SATB regulation of trophoblast stem-state. Research involves identifying mechanisms utilized by SATB homeodomain proteins to maintain the trophoblast stem state and inhibit trophoblast differentiation. Mutant SATB mouse models in conjunction with rodent and human trophoblast cell models are used in the analyses. A recently awarded NIH grant supports our mouse SATB model-related research. Regulation of the reproductive tract by sex steroid hormones. We are using zinc finger nuclease genome editing to generate rat knock-out models for disruptions in estrogen and progesterone receptor signaling. We are also utilizing the Brown Norway rat, an animal with progesterone resistance, to investigate the effects of sex steroid hormones on the uterus. Meetings Attended 2012 – “Fetal Programming and Environmental Exposures: Implications for Prenatal Care and Pre-Term Birth,”

The New York Academy of Sciences, NY, NY Editorial and Grant Reviews Ad hoc reviewer, Reproductive Biology (RB), Molecular Reproduction & Development (MRD), Case Reports in

Perinatal Medicine Grant reviewer, Oak Ridge Associated Universities (ORAU) Trainees George Bugarinovic – Undergraduate student – John Hopkins University Irfan Saadi, Ph.D. Assistant Professor Department of Anatomy and Cell Biology Member, Center for Epigenetics and Stem Cell Biology The focus of my research is to understand the pathogenetic mechanisms of craniofacial birth defects. Craniofacial malformations afflict about 5% of all infants born in the United States and comprise approximately one third of all birth defects. These anomalies result in significant medical, social and economic consequences. Orofacial clefts are one such common congenital facial defect that affects 1/800 live births. Cleft lip with or without cleft palate (CL/P) comprises the majority of orofacial clefts. The Center for Disease Control and Prevention (CDC) estimates that the lifetime cost for treating kids born each year with CL/P is over US$697 million. We have identified mutations in a novel cytoskeletal gene, SPECC1L, in patients with a severe manifestation of facial clefts that extend from the oral cavity to the eye - called oblique facial clefts (ObFC). Although less common, insights into the cellular and molecular mechanisms underlying ObFC will

78

directly impact our understanding of more common facial malformations, including cleft lip and hemifacial microsomia. Meetings Attended 2012 - American Society of Human Genetics Annual Meeting, San Francisco, CA 2012 - American Society of Cell Biology Annual Meeting, San Francisco, CA Seminars Presented 2012 - Molecular Genetics of Orofacial Clefting: New Perspectives and Initiatives. Pediatrics Grand Rounds,

Department of Pediatrics, The University of Kansas Medical Center, Kansas City, KS Editorial and Grant Reviews Reviewer, KUMC, FY13 Biomedical Research Training Grant Program Trainees Nathan Wilson –Graduate Student Kelly Stumpff – Medical Student Guerin Smith – Medical Student Syed K. Rafi – Visiting Senior Scientist Bruce Schultz, Ph.D. Professor Department of Pharmacology, Kansas State University Member, Center for Reproductive Sciences Research efforts are focused on understanding the physiological regulation of epithelial ion transport and barrier functions. Transepithelial movement of ions provides for electrolyte and fluid homeostasis and, in the case of milk, is necessary for production. Dysfunction of epithelial transport mechanisms, especially the anion channel CFTR, is associated with reproductive, pancreatic, renal, intestinal, and pulmonary disorders. In the laboratory, we strive to achieve a better understanding of epithelial physiology and to develop interventions that prevent or overcome such pathological conditions. Common mechanisms to accomplish ion transport are employed by a variety of epithelia. However, the cellular and subcellular location, along with regulatory apparatus, provides for unique combinations of mechanisms to support specific needs at each locale. Furthermore, a particular epithelium can modify its function depending upon the stage of tissue development or the endocrine state of the individual. In the laboratory, we are studying reproductive, renal, intestinal and mammary epithelia in order to understand their unique transport capabilities. These observations are particularly instructive for reproductive and mammary epithelia since relatively little is known regarding the mechanisms that they employ. We developed an in vitro system to study ion transport by epithelia lining the male reproductive tract. This system allows us to identify mechanisms of ion transport in this tissue along with the hormones and neurotransmitters that modulate such activity. This line of investigation is particularly important as we try to understand the causes of congenital bilateral absence of the vas deferens (CBAVD), a form of infertility that commonly affects cystic fibrosis patients. CBAVD has recently gained recognition as a 'mild' form of cystic fibrosis. The laboratory collaborates with Dr. John Tomich (Department of Biochemistry) in a project to develop synthetic channel forming peptides for the treatment of cystic fibrosis. Since the primary defect in cystic fibrosis

79

is the loss of an epithelial anion channel, we reasoned that providing such a conductance could reduce or preclude the effects of the disease. The production of milk defines mammals. Major components of milk include proteins, fats, carbohydrates, and minerals. These components are present in varying proportions, depending upon species. A major focus in the laboratory is to determine the mechanisms that can account for the concentrations of monovalent ions with a primary focus on Na+. Human milk has the lowest Na+ concentration of virtually all species. Thus, human mammary epithelial cell systems are the primary model for this line of investigation. Mastitis is an environmentally induced loss of epithelial integrity that affects a significant proportion of the human population, but has greatest impact on the dairy industry. An in vitro bovine mammary cell system is being employed in the laboratory to identify factors that lead from environmental insult to the loss of epithelial function. Finally, there is an ongoing collaboration with Dr. Ronette Gehring that focuses on the transport of xenobiotic compounds (environmental toxins, pharmaceuticals, etc.) across the mammary epithelium. This line of investigation seeks to identify mechanisms that can account for the active movement of these solutes into or from milk. We gratefully acknowledge ongoing or past support from the National Institutes of Health, the United States Department of Agriculture, and the Cystic Fibrosis Foundation. Meetings Attended

2012 - Society for the Study of Reproduction, State College, PA

2012 - “TGF-β1 impairs CFTR-mediated anion secretion across cultured porcine vas deferens epithelial monolayer via the p38 MAPK pathway,”North American Cystic Fibrosis Conference, Orlando, FL

Committees

KSU

Member, K-State 2025 Research Themes Committee, University Faculty Senate and Executive Committee, Provost’s Review Committee – Ralph Richardson CVM Dean, Veterinary Research Scholars Program Selection and Steering Committee ad hoc contributor, Safety Committee, Disaster Preparedness Special Committee, PhD Committee-Xiangming Li, Anatomy & Physiology Search Committee – A&P Director of Research Operations

Chair, Faculty Senate Caucus

Associate Department Head

National and International

Member, Society for the Study of Reproduction-Awards Committee; Publications Committee; Ethics Subcommittee Chair, Abstract Review for Male Reproductive Duct, Mastitis Research Workers (NE1048 Multistate Research Project)

PhD Examiner, Otago University, Dundeden, NZ Editorial and Grant Reviews Editorial Board, American Journal of Physiology – Cell Physiology Editorial Ad hoc, Endocrinology, Journal of Physiology, British Journal of Pharmacology, Human Reproduction,

Cell Biochemistry and Biophysics, Beneficial Microbes Grant Reviewer, KUMC Research Institute Internal Grants Program, Hong Kong Research Grants Council

80

Seminars Presented 2012 – “Epithelia Create the Fluid for Ebb and Flow of Sperm,” Center of Epithelial function in Health and

Disease Symposium. Manhattan, KS 2012 – “A Sperm’s-eye View of Epithelial Function,” SUNY at Buffalo Department of Physiology & Biophysics

Seminar Series. Buffalo, NY Trainees Qian Wang – Graduate Student Sheng Yi – Graduate Student Samuel Molina – Graduate Student Allan Prior – Graduate Student Jimmie Stewart – Undergraduate Student Jacob Hull – Undergraduate Student Tyler Dubek – Undergraduate Student Chad Slawson, Ph.D. Assistant Professor Department of Biochemistry and Molecular Biology Member, Center for Epigenetics and Stem Cell Biology Research Focus: To Understand the Regulation of the Post-Translational Modification O-GlcNAc During Growth and Development: O-GlcNAc is the addition of a single N-acetyl-glucosamine residue to serine/threonine residues of proteins found in the cytoplasm or nucleus (O-GlcNAcylation). Unlike extracellular glycosylation, the sugar residue is not elongated into complex oligosaccharides and is processed dynamically in response to cellular stimuli by a single O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA). O-GlcNAc is involved in many cellular processes such as nutrient sensing, stress response, transcription, translation, cell signaling, and cell cycle regulation. Currently, we are asking several questions to understand how O-GlcNAc regulates mitosis such as how is OGT targeted to specific structures at M phase as well as to specific substrates? What is the dynamics of O-GlcNAcylation throughout mitosis? What mitotic signaling pathways are regulated by O-GlcNAcylation? My laboratory uses a variety of techniques from cloning, western blotting, imaging, and mass spectroscopy in order to answer these questions. Committees Departmental Member, 2012 Heartland Undergraduate Biochemistry Forum Committee KUMC, Eva Selfridge Dissertation

Committee, Allen Chazelle Dissertation Committee, Mary Ashley Rimmer Dissertation Committee Medical School Member, Academic Academic and Professionalism Committee Member, Students Promotions and Special Programs Sub-committee University of Lausanne, Switzerland Tanja Bhuiyan, Dissertation Committee

81

Editorial Reviews Ad hoc Reviewer - Journal of Biological Chemistry Ad Hoc reviewer - Journal of Proteomics Research Seminars Presented 2012 - “O-GIcNAc Signaling Regulates Mitochondrial Function: Implications for Alzheimer’s Disease,” KUMC

Alzheimer’s Center Research Colloquium

2012 - “Novel Roles for O-GlcNAc Signaling in the Regulation of Mitosis and Gene Transcription,” University of

Lausanne, Switzerland, Center for Integrative Genomics. 2012 - “The O-GlcNAc Post-Translational Modification is a Key Regulator of Cellular Function,”

University of Kansas School of Medicine, Department of Pharmacology. Academic Honors Best poster in the DNA Replication, Recombination, Repair thematic session. “Identification of OGT

Interacting Proteins at M Phase,” Experimental Biology Meeting, KUMC

Trainees Zhen Zhang - IGPBS student Ee Phie Tan - IGPBS rotation student Sarah Caro – Masters in Biotechnology rotation student Anish Potnis - Summer Undergraduate Christopher Lanza - Summer Undergraduate Melody Chambers – Undergraduate student Peter G. Smith, Ph.D, Professor Co-Director, Kansas Intellectual and Developmental Disabilities Research Center Founding Director, Institute for Neurological Disorders Department of Molecular and Integrative Medicine Member, Center for Reproductive Sciences Nerves regulate function and structure of peripheral cells. Target cells in turn provide molecular signals that govern the quantity and type of innervation they receive. Our research examines this interplay between nerve and target and the factors that govern neuronal growth and degeneration. We are especially interested in how this relationship is affected by gonadal steroid hormones such as estrogen. Ongoing projects examine mechanisms and consequences of neuroplasticity in peripheral tissues including: reorganization of cardiac innervation following myocardial infarction, which may contribute to sudden cardiac death; estrogen-induced remodeling of innervation of the reproductive tract; mechanisms by which nerve projections are pruned under normal and pathophysiological conditions; and the role of estrogen in the etiology of female pain syndromes. Committees Departmental Member, Graduate Student Advisory Committee

82

KUMC Chair, cDNA Microarray Advisory Committee Member, Laboratory Animal Resources Advisory Committee, Research Committee, Research Institute,

Research Administration Steering Committee

Chair, Research Institute Research Committee Chair, Search Committee for Chair, Department of Microbiology, Molecular Genetics and Immunology Member and Team Leader, Search Committee for Executive Vice Chancellor and Interim Executive Dean National NIH - Ad Hoc Study Section service, Urologic and Kidney Development and Genitourinary Diseases;

Molecular, Cellular, and Developmental Neuroscience; Neurological, Aging and Musculoskeletal Epidemiology; Brain Disorders and Clinical Neuroscience; Distinguished Editorial Review Board for Specialized Centers on Research on Sex Differences; Continuous Submission Eligibility

Editorial and Grant Reviews Associate Editor - Autonomic Neuroscience: Basic and Clinical Ad hoc Reviewer, American Journal of Physiology, American Journal of Respiratory Cell and Molecular

Biology, Anatomical Record, Archives of Oral Biology, Autonomic Neuroscience: Basic and Clinical, Biology of Reproduction, Brain Research, British Journal of Pharmacology, British Journal of Nutrition, Cell and Tissue Research, Circulation, Circulation Research,Current Eye Research, Developmental Biology, DNA and Cell Biololgy, Endocrinology,Experimental Eye Research, Experimental Neurology, European Journal of Neuroscience, Histology and Histopathology, Hypertension, Investigative Ophthalmology and Visual Sciences,Journal of the Autonomic Nervous System, Journal of Comparative Neurology, Journal of Endocrinology, Journal of Histochemistry and Cytochemistry, Journal of Neuroscience, Journal of Neuroscience Methods, Journal of the Peripheral Nervous System, Journal of Urology, Journal of the Society for Gynecologic Investigation, Molecular and Cellular Biochemistry, Neuroscience, Neuroscience Letters, Pharmacological Research, PLOSOne, Psychobiology, Rejuvenation Research, Regulatory Peptides, Reproduction, Reproduction and Fertility, Synapse

Grant Reviewer - Pennsylvania Department of Health, The Grant Workshop, Fonds zur Forderung der

wissenschaftlichen Forschung (Austria Science Fund), Wellcome Trust, National Science Foundation, KUMC Center for Aging Research Review Committee, Feasibility Grants Competition, Claude Pepper Older Americans, Independence Center, Nathan Shock Center, & Alzheimer Disease Research Center, The Geriatrics Center, University of Michigan, KUMC School of Allied Health Research Committee, University of Calcutta School of Medicine, University of Vermont School of Medicine, Miami University, Oxford Ohio, KUMC Research Institute Lied Foundation, KUMRI Collaborative Research Grants (Chair), City University of New York, Oregon Health Sciences University, University of Missouri, Columbia

Seminars Presented 2012 – “Peripheral Nervous System Plasticity and Chronic Pain,” University of Tennessee – Memphis

83

Trainees Runa Chakrabarty – Senior Research Scientist Argenia Doss – Graduate Student Sarah Tague – Postdoctoral Fellow Aritra Battacherjee – Graduate Student Michael J. Soares, Ph.D. University Distinguished Professor Director, Institute for Reproductive Health and Regenerative Medicine Department of Pathology and Laboratory Medicine Member, Centers for Epigenetics and Stem Cell Biology, Reproductive Sciences, and Developmental Origins of Health and Adult Disease Our laboratory is interested in the regulation of cell differentiation, especially as related to trophoblast stem cells, and signaling pathways controlling their developmental fate. Our research efforts include investigating species-specific reproductive adaptations and signaling events involved in the establishment and maintenance of pregnancy; the prolactin gene family, intrauterine inflammatory and immune cells, uterine vasculature, decidual cells, and the invasive trophoblast cell lineage. These scientific pursuits have important implications regarding pregnancy-related diseases such as preeclampsia, intrauterine growth restriction, and pre-term birth. Our research also includes the establishment and characterization of mutant rat models. Genome editing strategies have been used to generate rats with mutations in key genes regulating estrogen signaling. These animal models represent new tools for biomedical scientists in a range of disciplines, including cancer biology, reproduction, women’s health, environmental health, metabolism, immunology, neurosciences, and cardiovascular biology. Meetings Attended

2012 – “Focal adhesion kinase regulates AP-1 factor expression and invasion in trophoblast cells,” Society for Gynecologic Investigation Annual Meeting, San Diego, California.

2012 – “Transcriptional hierarchy of SATB homeobox proteins in trophoblast stem cells,” New York Academy of Sciences, Fetal Programming and Environmental Exposures, New York.

2012 – “Placental endogenous retroviruses facilitate rapid evolution of core trophoblast regulatory network,” Annual Meeting of the Society for Molecular Biology and Evolution, Dublin, Ireland.

2012 – “OVO-like 1 is a regulator of human trophoblast differentiation,” 45th Annual Meeting of the Society for the Study of Reproduction, State College, Pennsylvania.

2012 – “FOSL1 regulation of the invasive trophoblast lineage: partners and gene targets,” 45th Annual Meeting of the Society for the Study of Reproduction, State College, Pennsylvania.

2012 – “Origin of a species-specific rheostat controlling testicular growth and steroidogenesis,” 45th Annual Meeting of the Society for the Study of Reproduction, State College, Pennsylvania.

2012 – “Role of hypoxia signaling in trophoblast cell lineage commitment,” FASEB, Snowmass, CO.

84

Committees KUMC Member, Research Institute Technology Transfer Advisory Committee, Executive Dean’s Basic Science

Planning Committee, High Throughput Genomics Facility Advisory Committee , Advisory Committee for the Huron Changing for Excellence Project, Executive Vice Chancellor Transition Team for Research Committee, Advisory Committee for the Genomics Core, Research Bridging Fund Organization Committee

Departmental Chair, Promotion and Tenure Committee National Member, Society for the Study of Reproduction Nominating Committee Editorial and grant reviews Board of Reviewing Editors, Biology of Reproduction Honorary Editorial Board, Reproductive Biology Insights Editorial Board, Placenta Ad hoc Reviewer, Biochimica Biophysica Acta, BMC Developmental Biology, BMC Genomics, Briefings in

Functional Genomics, Cell Research, Development, Developmental Biology, Developmental Dynamics, Endocrine Reviews, Endocrinology, European Journal of Biochemistry, Gene, Genesis, Human Reproduction,Journal of Biological Chemistry, Journal of Cell Science, Journal of Clinical Endocrinology & Metabolism, Journal of Clinical Investigation, Journal of Experimental Zoology, Journal of Reproductive Immunology, Mammalian Genome, Molecular Biology and Evolution, Molecular Carcinogenesis, Molecular & Cellular, Endocrinology, Molecular & Cellular Biology, Molecular Endocrinology, Molecular Pharmaceutics, Molecular Reproduction and Development, Nature Medicine, Nature Protocols, Pediatric Research, Physiological Genomics, Placenta, PLOS Biology, PLOS One, Proceedings of the National Academy of Sciences USA,Reproduction, Science, Systems Biology in Reproductive Medicine, Trends in Endocrinology and Metabolism

Reviewer – National Institutes of Health, NIAID Special Emphasis Panel; National Institutes of Health, NICHD

Special Emphasis Panel/Scientific Review Group Ad hoc Reviewer, Health Research Council of New Zealand, Natural Sciences and Engineering Research

Council of Canada (NSERC) Seminars Presented February 2012 - “Adaptations at the maternal-fetal interface,” Reproduction and Development Group, Dept. of

Physiology, University of Toronto, Ontario, Canada May 2012 - “Natural killer cell and genetic regulation of hemochorial placentation,” Joint International Congress

of the American Society for Reproductive Immunology and the European Society for Reproductive Immunology 2012, Hamburg, Germany

85

June 2012 - “Regulatory pathways controlling placentation,” Fetal Programming and Environmental Exposures: Implications for Prenatal Care and Pre-Term Birth,” The New York Academy of Sciences, New York, NY

Academic Honors External Scientific Advisory Board for the Center for the Center of Biomedical Research Excellence for

Perinatal Biology, Brown University, Providence, RI Internal Advisory Board, University of Kansas BIRCWH Faculty Development K12 program Internal Advisory Board, University of Kansas Women’s Reproductive Health Research Faculty Development

K12 program External Scientific Advisory Board, “Molecular and Cellular Controls of Placental Metabolism,” Magee

Women’s Research Institute, Pittsburgh, PA External Scientific Advisory Board, Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI Trainees Pengli Bu – Postdoctoral Stephen J. Renaud – Postdoctoral Kaiyu Kubota – Postdoctoral Pramod Dhakal – Postdoctoral Damayanti Chakraborty – Graduate Student George Bugarinovic – Undergraduate Student Anamita Ratri – Undergraduate Student Katherine Swenson-Fields, Ph.D. Research Associate Professor Department of Anatomy and Cell Biology Member, Center for Epigenetics & Stem Cell Biology The research focus of our lab, shared with Dr. Timothy Fields, is centered the role of the renal inflammatory environment, with a particular focus on macrophages, in polycystic kidney disease (PKD) progression. Macrophages normally infiltrate tissues in response to infection or injury where they act first to sterilize the environment and then to promote repair, cell proliferation and regeneration of damaged tissues, a function, which they effect by secreted factors. Following tissue repair, further infiltration of these cells no longer occurs and their numbers normally decline to that found in the pre-injured state. We have found large numbers of infiltrated macrophages in the kidneys of both human and mouse individuals with PKD, and have demonstrated that the presence of these cells contributes to cyst expansion and functional renal decline. In addition we have shown that macrophages promote the proliferation of PKD cyst cells when co-cultured in vitro, and that these pro-proliferative effects are mediated by soluble factor/s. These studies suggest that the normal functions of macrophages to transiently promote tissue regeneration following injury are continuously and, thus, pathologically present in PKD kidneys to promote cyst cell proliferation and resultant cyst expansion. Current studies are underway to identify the specific factors and signaling pathways that promote renal macrophage recruitment and the pro-proliferative cyst-expanding effects of these cells. This research will facilitate the achievement of our broader goals to develop therapeutic agents that block these macrophage processes in PKD that can be used clinically to slow progression of this disease.

86

Meetings Attended 2012 – “MCP-1 is the Primary Macrophage Recruitment Factor Produced by Human and Mouse Polycystic

Kidney Cells and Promotes Disease Progression in cpk Mice,” American Society of Nephrology, San Diego, CA

Academic Honors 2012 – Research Institute Travel Award Trainees Sally Salah – Graduate Student

Russell H. Swerdlow, M.D. Gene and Marge Sweeny Professor Departments of Neurology, Molecular & Integrative Physiology, Biochemistry & Molecular Biology Director, Alzheimer’s Disease Center Director, Neurodegenerative Disease Program Member, Center for Epigenetics and Stem Cell Biology Dr. Russell Swerdlow is a physician-scientist at the University of Kansas. He has studied Alzheimer's disease for approximately 25 years and is recognized for his contributions to the Alzheimer's disease research field. He directs the NIH-funded University of Kansas Alzheimer's Disease Center, serves as an attending physician at the Kansas University Medical Center's Memory Disorders Clinic, directs the Kansas University Medical Center's Neurodegenerative Disorders Program, and is a Professor in the Departments of Neurology, Molecular and Integrative Physiology, and Biochemistry and Molecular Biology at the University of Kansas School of Medicine. Dr. Swerdlow received his undergraduate and doctor of medicine degrees from New York University. He trained as a neurologist and Alzheimer's disease specialist at the University of Virginia, co-founded the University of Virginia's Memory Disorders Clinic, and participated in pivotal clinical trials for most FDA-approved Alzheimer's disease medications. Before leaving Virginia for Kansas in 2007, Dr. Swerdlow chaired the Alzheimer's Disease and Related Disorders Commission of the Commonwealth of Virginia. He is a recipient of an S. Weir Mitchell Award from the American Academy of Neurology, a Cotzias Award from the American Parkinson's Disease Foundation, and several grant awards from the National Institutes of Health. He has served as the Research Committee Chair of the CurePSP Foundation; is on the editorial board of several research journals including the Journal of Alzheimer's Disease; and frequently sits on NIH, Veteran's Administration, and non-profit research foundation study sections. In addition to his clinical duties, Dr. Swerdlow studies brain energy metabolism and the role brain energy metabolism plays in Alzheimer's disease and other neurodegenerative diseases. He was the first to propose using ketone bodies to improve brain energy metabolism in Alzheimer's disease patients, presaging the development of this now-utilized Alzheimer's disease treatment approach. His laboratory is actively working on new ways to manipulate brain energy metabolism. The goal of this work is to create new and effective treatments that will hopefully help people with Alzheimer's disease. Committees KUMC Vice-Chair, Research Committee Mentor, Nairita Roy, Dissertation Committee

87

Member, Dissertation Committee for Robert Rogers and, Kristen Watt Member, Shared Resources Subcommittee National Member, Dissertation Committee, Rushi Trivedi, Stowers Institute, KC External Advisory Board Member, Louisiana State University Institute for Dementia Research and Prevention,

Baton Rouge, LA External Advisory Board Member, Center of Excellence for Research in Complementary and Alternative

Medicine in Alzheimer’s Disease, Mount Sinai School of Medicine, NY External Advisory Board Member, Pennington Biomedical Research Center, Louisiana State University, Baton

Rouge, LA International Dissertation Examiner, Kathryn Cimdins, University of Melbourne, Australia Editorial and grant reviews Reviewer, The Journal of Mitochondrial Biology, The Open Pathology Journal, Mitochondrial Therapy,

Frontiers in Aging Neuroscience, International Journal of Clinical and Experimental Medicine, Biochimica et Biophysica Acta – Molecular Basis of Disease, Degenerative Neurological and Neuromuscular Disease, Metabolic Brain Disease, Neurology (Behavioral and Cognitive Neurology Section), Bioenergetics, American Journal of Neurodegenerative Disease

Senior Editor, Journal of Alzheimer’s Disease Grant Reviewer - Alzheimer’s Association, NIH: CDIN, MDCN, Several SEPs, University of Arizona

Alzheimer’s Disease Core Center Pilot Program, Alzheimer’s Research UK, Croucher Foundation, Thiel Foundation

Seminars Presented 2012 - “KU Alzheimer’s Disease Town Hall Meeting,” University of Kansas, Kansas City, KS 2012 - “Bioenergenics: A Possible Cause of AD and Potential Therapeutic Target,” First Annual University of

Kansas Alzheimer’s Disease Center Symposium, Kansas City, KS 2012 - “Bioenergetics: A Possible Cause of AD and Potential Therapeutic Target,” University of Kansas

Neurology Grand Rounds, Kansas City, KS 2012 - “The University of Kansas Alzheimer’s Disease Center,” “The University of Kansas Alzheimer’s Disease

Center” 2012 - “Manipulating Brain Bioenergetics,” University of Kansas Nutrition Sciences Graduate Program, Kansas

City, KS 2012 “Alzheimer’s Disease: The Big Picture,” 2012 University of Kansas Medical Center Resident and Post

Doc Research Day, Kansas City, KS

88

2012 - “Panel Discussion of Speakers,” University of Kansas 2012 Healthy Aging Symposium, Overland Park, KS

2012 - “My Journey as a Physician Scientist,” Annual MD-PhD Symposium, University of Kansas School of Medicine, Kansas City, KS

2012 - “The Spectrum of Neurodegenerative Dementias,” University of Kansas Neurology Grand Rounds,

Kansas City, KS 2012 - “Mitochondria in Sporadic Neurodegenerative Diseases,” Biophysical Society Annual Meeting, San

Diego, CA 2012 - “Mitochondrial Medicine for Alzheimer’s Disease,” Alzheimer’s Association “Let’s Get Real About Non-

Amyloid Targets” International Symposium, Washington DC 2012 - “Brain Bioenergetics and Aging: Therapeutic Strategies for Intervention,” 13th International Conference

on Alzheimer’s Drug Discovery. 2012 - “The Spectrum of Neurodegenerative Dementias,” University of Kansas Neurology Update Symposium,

Overland Park, KS 2012 – “Alzheimer’s Disease Research at the University of Kansas Alzheimer’s Disease Center,” Alzheimer’s

Association Heartland Chapter Symposium, Overland Park, KS Academic Honors 2012 – University of Kansas Scholarly Achievement Award 2012 – Listed as one of the most-referred to Neurologists in the KC metro area (435 South Survey) Trainees Lezi E - Graduate Student

Eva Selfridge – Graduate Student

Nairita Roy – Graduate Student

Diana Silva – Graduate Student

Andrea Nuckolls – Graduate Student

Isabella Wang - Graduate Student

Matthew Stroh - Graduate Student

Ee Phie Chen – Graduate Student

Rawan Albadareen – Neurology Resident

Eric Funk – Medical Student

Karthik Chellamathu – High School Student

J. Brantley Thrasher, M.D., F.A.C.S. Professor and the William L. Valk Chair Department of Urology Member, Center for Reproductive Sciences Dr. Thrasher's basic science research interest is in the area of prostate cancer and he is currently a co-investigator or consultant in NIH, Center for Disease Control, and Department of Defense funded research. His clinical research interests are in the area of prostate, bladder, and renal cancer, as well as

89

reconstruction, and he serves as the investigator on numerous investigator initiated, industry funded, and institutionally funded protocols. Committees National Member, Residency Review Committee for Urology KUMC

Member, EVC Leadership Advisory Group to Community Partnerships Initiative , Kansas State Budget Allocation Committee, Kansas Masonic Cancer Research Institute, Medical Disaster Committee, Medical Staff Executive Committee, Multidisciplinary Cancer Committee, KU Cancer Institute

Clinical Consultant, Scientific Committee for the Kansas IDeA Networks of Biomedical Research Excellence (K-INBRE) Course Director, Urology Grand Rounds Lecture Series

Editorial and Grant Reviews

Editorial, Practical Reviews in Urology, Annals in Urology, 5 Minute Urology Consult, American Family Physician, Cancer Prevention, Urologic Oncology: Seminars and Original Investigation, Urology Times Editorial Council, Journal of Urology

Ad hoc reviewer, Journal of Urology, Urology, Cancer, Prostate, Journal of Enourology

Seminars Presented

2012 – “Radical Retropubic Prostatectomy versus Robotic Assisted Laparoscopic Prostatectomy in High-Risk Prostate Cancer: Rates of 3-Year Biochemical Recurrence,” 91st Annual Meeting South Central Section Meeting, American Urological Association, Colorado Springs, ColoradO

2012 – “A Prospective Study of Erectile Function and Urinary Symptoms after Transrectal Ultrasound in Prostate Biopsy,” 91st Annual Meeting South Central Section Meeting, American Urological Association, Colorado Springs, CO

2012 – “Novel Natural Compound Alternol Activates the Apical Apoptosis Pathway in Prostate Cancer Cells,” 91st Annual Meeting South Central Section Meeting, American Urological Association, Colorado Springs, CO

2012 - “T Leon Howard Imaging Hour,” 91st Annual Meeting, South Central Section Meeting, American Urological Association, Colorado Springs, CO

2012 – “Transrectal Ultrasound and Biopsies: Are we Downplaying the Complications?” Georgia Urologic Association Annual Meeting, Sea Island, GA

2012 - “Chemoprevention for Prostate Cancer,” Georgia Urologic Association Annual Meeting, Sea Island, GA

2012 - “Chemoprevention for Prostate Cancer,” UNC Charlotte, Grand Rounds, Charlotte, NC

Academic Honors Kansas City’s Top Doctors, 435 South Magazine

90

American Association of GU Surgeons, Member Named Kansas City Super Doctor – Kansas City Magazine Jay L. Vivian, Ph.D. Assistant Professor Department of Pathology and Laboratory Medicine Member, Center for Epigenetics and Stem Cell Biology My research uses the mouse as a genetic, stem cell, and developmental system to study signaling during embryonic development. My group also makes substantial use of mouse embryonic stem cells for genetic engineering and as a model for regulation of gene expression and early embryonic differentiation. My group is interested in understanding the signaling pathways and genetic hierarchies that regulate gene expression and stem cell self-renewal in embryonic stem cells. My work utilizes mutant and transgenic mouse models for our studies. We also use and generate human induced pluripotent stem cells to both model human disease including congenital developmental disorders, and as a source for cellular therapies for spinal cord injury. Meetings Attended February 2012 – “TGF-beta-related signaling regulates stem cell heterogeneity,” Gordon Research

Conference, Galveston, TX November 2012 – “KUMC Transgenic and Gene Targeting Institutional Facility,” KU Cancer Center

Symposium, Kansas City, KS Committees KUMC Member, 2011 Greenwald Symposium Planning Committee; Member, KUCC Shared Resource Committee;

Member, Laboratory Animal Resource Center Advisory Committee; Member, Human Stem Cell Advisory Committee; Member, Ad Hoc Investigation Committee on Research Misconduct

Member, Graduate Student Advisory Committees for Kristen Watt and Nairita Roy Member, Comprehensive Exam Committees for Jonathan Fitzgerald and Shachi Bhatt Member, Dissertation Committees for Damayanti Chakraborty, Yi Feng and Shachi Bhatt Editorial and Grant Reviews Ad hoc Reviewer, Molecular and Cellular Proteomics, Journal of Assisted Preproduction and Genetics, Journal Editorial service Editorial Board Member - America Journal of Stem Cell Research, Cloning and Transgenesis Seminars Presented 2012 – “Updates from the Transgenic Facility,” KUMC Center for Epigenetics and Stem Cell Biology Chalk Talk

Meeting, Institute for Reproductive Health and Regenerative Medicine, Kansas City, KS 2012 – “TGF-beta-related signaling in embryonic stem cell maintenance: self-renewal as a dynamic and

regulated equilibrium,” KUCC Seminar Series, Kansas City, KS

91

2012 - “TGF-beta-related signaling in embryonic stem cell maintenance: self-renewal as a dynamic and

regulated equilibrium,” Genes and Development Graduate Program 20th Anniversary Symposium M.D. Anderson Cancer Center, Houston, TX

2012 – “Current technologies in mouse genetics and transgenesis,” KUMC Kidney Institute Sullivan Research

Conference, Kansas City, KS Trainees Jessica Copeland – Postdoctoral Fellow Katherine Burgess – Postdoctoral Fellow Catherine Schwartz – Postdoctoral Fellow Carrie Malcom – Rotation MD/PhD Student Jinxi Wang, M.D., Ph.D. Harrington Distinguished Professor Director, Harrington Laboratory for Molecular Orthopedics Department of Orthopedic Surgery Member, Center for Epigenetics and Stem Cell Biology The Harrington Laboratory for Molecular Orthopedics, which is primarily supported by NIH grants and the Mary Alice and Paul R. Harrington, M.D. Distinguished Professorship Endowment, was established in 2005. The laboratory is well equipped for conducting research involving biochemistry, cell biology, and molecular biology of skeletal tissues. Our major research interests are to study the regulatory mechanisms by which pluripotent mesenchymal stem cells differentiate into osteoblasts (bone-forming cells) or chondrocytes (cartilage-forming cells) and investigate the role of specific signaling pathways in bone/cartilage regeneration and diseases. Currently, research in our laboratory is focused on the following projects: (1) the role of bone sialoprotein (BSP) in osteoblast differentiation and bone regeneration, (2) molecular regulation of chondrocyte differentiation and articular cartilage regeneration, and (3) pathogenetic mechanisms and novel therapeutics for osteoarthritis. Meetings Attended 2012 – “Deficiency of Nfat1 transcription factor causes osteoarthritis through dysfunction of cells in multiple

joint tissues,” 2012 International Conference on Orthopedics and Rheumatology, Chicago, IL 2012 – “Role of Runx2 transcription factor in bone sialoprotein-mediated osteogenic differentiation,” 2012

Annual Student Research Forum, University of Kansas Medical Center 2012 – “Nfat1 deficiency causes osteoarthritic subchondral bone changes through pathological differentiation

of bone marrow stem cells,” 2012 Annual Student Research Forum, University of Kansas Medical Center. Sept. 14, 2012 – “The role of Nfat1 in the healing of osteochondral defects,” 2012 Annual Meeting of Kansas

State Orthopedic Society, Wichita, KS

2012 – “Transcription factor Nfat1 deficiency: A risk factor for progression of posttraumatic osteoarthritis in mice,” 58th Annual Meeting of the Orthopedic Research Society, San Francisco, CA

2012 – “Bone and muscle interactions during the progression of Nfat1 deficiency-mediated osteoarthritis,”

American Society for Bone and Mineral Research Topical Meeting: Bone and Skeletal Muscle Interactions, Kansas City, MO

92

2012 – “Contribution of the dura to bone sialoprotein-mediated cranial bone regeneration,” 2012 International Conference on Orthopedics and Rheumatology, Chicago, IL, USA

Committees Departmental Member, Research Committee KUMC Member, Institutional Animal Care and Use Committee (IACUC) National Member, New Investigator Mentoring Committee, the Orthopedic Research Society (ORS, USA) International Board Director, International Association for Biological and Medical Research (IABMR) Editorial and Grant Reviews Reviewer, Arthritis & Rheumatism, Molecular Endocrinology, Journal of Orthopaedic Surgery and Research Editorial Board, Journal of Bone and Mineral Research (JBMR) Grant Reviewer, Israel National Science Foundation, Netherlands Organization for Health Research and

Development, NIH, DOD Seminars Presented 2012 – “Grant applications for Medical Research: Opportunities and challenges,” Soochow University Medical

School, Suzhou, P.R. China 2012 – “NFAT signaling in Bone Marrow Stem Cells,” Stowers Institute for Medical Research, Kansas City, MO 2012 – “Posttraumatic osteoarthritis: A keynote lecture,” Invited by the Organizing Committee of the 2012

International Conference on Orthopedics and Rheumatology, Chicago, IL, USA Academic Honors

Organizing Committee Member and Scientific Advisor, 2012 International Conference on Orthopedics and

Rheumatology, Chicago, IL Session Chair, Regenerative and Molecular Orthopedics Session, 2012 International Conference on

Orthopedics and Rhematology, Chicago, IL

Carl P. Weiner, M.D., M.B.A. K.E. Krantz Professor and Chair Department of Obstetrics and Gynecology Associate Director, Institute for Reproductive Health and Regenerative Medicine Director, Center for Developmental Origins of Health and Adult Disease

93

Dr. Weiner's laboratory investigative interests focus on the regulation of uterine quiescence during pregnancy, impact of chronic fetal hypoxia, and the discovery, interpretation, and application of biomarkers for reproductive pathology. His laboratory has multiple firsts in the application of proteomics, genomics and transcriptomics to reproductive science. Dr. Weiner is a strong advocate of the strategic linking of clinical and basic research, and is the founder and President of Perinet Inc., a biomedical development company created to facilitate the development of his laboratory's findings. Meetings Attended 2012 - “Vimentin Suppression Enhances the Fetal Inflammatory Response in Endothelium During Chronic

Hypoxia,” “Purkinje Cell Protein 4 (PCP4) Repression in Human Myometrium During Preterm Labor,” Society for Maternal Fetal Medicine, Dallas, TX

Committees KUMC Member - Executive Committee, Executive Vice Chancellor’s Advisory Committee, BIRCWH Internal

Advisory Committee, WRHR Internal Advisory Committee, Medical Executive Committee, University of Kansas Hospital Strategic Plan Advisory Committee, University of Kansas Physicians Inc. Board of Directors

Departmental Member, Obstetrics and Gynecology Education Committee National Member, Blue Cross Blue Shield of Kansas City Medical Advisory Committee Editorial and Grant Reviews Reviewer, European Journal of Clinical Investigation, Placenta, Circulation, Cardiovascular Pharmacology,

Cardiovascular Research, Prenatal Diagnosis, Journal of Clinical Investigation, The Lancet, Prenatal and Neonatal Medicine, Human Reproduction, Lancet, Molecular Human Reproduction, Journal of Pharmacology and Therapeutics, New England Journal of Medicine, PLoS Medicine, Reproductive Sciences

Editorial Board, Fetal and Maternal Medicine Review Seminars Presented 2012 – “Noninvasive Prediction of Preterm Birth,” 28th Annual Conference on Obstetrics, Gynecology, Perinatal Medicine, Neonatology, and the Law. Costa Rica 2012 – “Chronic Fetal Hypoxia and Brain Injury: The fall of Dogma,” 28th Annual Conference on Obstetrics, Gynecology, Perinatal Medicine, Neonatology, and the Law. Costa Rica 2012 – “Improving Competencies for Obstetrical Emergencies by Simulation Training,” 28th Annual Conference on Obstetrics, Gynecology, Perinatal Medicine, Neonatology, and the Law. Costa Rica 2012 – “Cell free plasma RNA in pregnancy complications,” 11th World Congress in Fetal Medicine. Kos, Greece.

94

2012 – ““Cell free plasma RNA in pregnancy-The endocrine language of love and hate,” Dept. of OB/GYN, St. Louis University. St. Louis, MO Trainees Minghui Tai – MD Zhe Wang – MD Mark L. Weiss, Ph.D. Professor Department of Anatomy and Physiology – Kansas State University Adjunct Professor, KUMC, Dept. of Physiology Associate member, KUCC Associate Director, Terry C. Johnson Center for Basic Cancer Research Founding Fellow, Midwest Institute for Comparative Stem Cell Biology Center for Epigenetics & Stem Cell Biology Weiss’ research focus is on stem cell biotechnology and regenerative medicine. His lab successfully produced stem cell lines such as rat embryonic stem cells and induced pluripotent stem cells, and rat and human mesenchymal stromal cells derived from the umbilical cord matrix or from other tissues such as bone marrow. Weiss’ lab investigates promising cellular therapeutics for regenerative medicine. For example, mesenchymal stromal cells have been tested in a variety of rodent preclinical disease models including neurodegenerative diseases such as Parkinson’s disease, heart disease such as myocardial infarction, and cancer. Based upon the immune properties of Wharton’s jelly derived mesenchymal stromal cells (WJCs), Weiss’ lab tested them for treating graft versus host disease (GVHD). The first round of preclinical data indicated that rat WJCs prevent the development of GVHD. Now, Weiss’ group will determine if WJCs can treat on-going GVHD in their preclinical rodent model. Weiss’ lab focuses upon the mechanisms of pluripotency in stem cells. Weiss’ lab is producing new rat models of human disease using gene targeting in rat embryonic stem cells and reporter cell lines that report when a particular gene is activated during development. Meetings Attended 2012 - Second Midwest Conference on Stem Cell Biology and Therapy, Rochester, MI 2012 - ISCT, Seattle, WA Committees KSU Member, CVM College Graduate Advisory Committee, CVM College Research Committee, CVM Biosecurity

Committee, KSU Faculty Senate, KSU University Committee on Planning, Dept Head Search Committee

Editorial and Grant Reviews

Editorial board, The Open Stem Cell Journal

Editorial board, Recent Patents on Regenerative Medicine

Grant Reviewer, Polish Foundation for Science

95

Grant Reviewer, MIUR (Italian Ministry for Education, University and Research)

Grant Reviewer, Portuguese Foundation for Science and Technology

Editorial board, International Scholarly Research Network (ISRN) Transplantation

Editorial board, Journal of Regenerative Medicine and Tissue Engineering

Grant Reviewer, TEDCO (Maryland Stem Cell Research Fund) Seminars Presented

2012 – “Genetic engineering using rat pluripotent stem cells,” Wichita State University, Dept of Biological Sciences

2012 – “PiggyBac transposon mutagenesis in rat embryonic stem cells,” Second Midwest Conference on Stem Cell Biology and Therapy, Oakland University, Rochester, MI

2012 – “Genetic engineering using pluripotent stem cells,” Rockhurst University, Kansas City, MO

Trainees Joseph R. Smith – Undergraduate Student Phuoc Bui – Undergraduate Student Benjamin Ryba-White – Undergraduate Student Daniel Quach – Undergraduate Student Katrina Fox – DVM/MS John Hirt – Graduate Student Yelica Lopez – DVM/PhD Zongning (Adam) Miao – Visiting Scientist from China Pavan Rajanahalli – Postdoctoral Student John Hirt – DVM Yelica Lopez – DVM Zongning (Adam) Miao – Postdoctoral Student Pavan Rajanahalli – Postdoctoral Student Michael W. Wolfe, Ph.D. Associate Professor Research Integrity Officer, KUMC Department of Molecular and Integrative Physiology Member, Centers for the Developmental Origins of Health & Adult Disease, Reproductive Sciences Mammalian reproduction is regulated by a number of hormones produced at various locations: hypothalamus in the brain, gonadotropes within the anterior pituitary gland, the gonads and also by the placenta during pregnancy. The glycoprotein hormones, luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesized in pituitary gonadotropes and chorionic gonadotropin (CG) by the placenta, are essential to mammalian reproduction. Research in my laboratory is directed towards understanding the cellular and molecular mechanisms involved in regulating the genes encoding these hormones. One area of emphasis is on how gonadotropin-releasing hormone secretion by hypothalamic neurons is regulated and how it signals to pituitary gonadotropes to induce the expression of the genes for LH. A second area focuses on elucidating the events associated with the differentiation of placental trophoblast cells and their acquisition of expression of CG.

96

We use a variety of experimental approaches and models to examine cell differentiation and gonadotropin gene expression such as the study of DNA-protein and protein-protein interactions, DNA microarrays, promoter analysis, transgenic mice and primary cultures of human trophoblasts. Our overall goal is to identify the physiologically relevant molecular and cellular events responsible for regulating cell differentiation and expression of the gonadotropin subunit genes. This will provide a better understanding of how the reproductive system is normally regulated and ultimately, will provide clues as to how diseases, drugs and the environment impact reproductive success. Meetings Attended 2012 – 9th Annual Gilbert S. Greenwald Symposium Committees Departmental Director, Physiology Graduate Student Advisory Committee Member, Several Dissertation Committees KUMC Member - dissertation committees ( Pathology), Graduate Council, SOM IGPBS admissions and

advisory committees, SOM Elections committee, IACUC, IACUC programmatic sub-committee, research misconduct inquiry committee, Nominations Committee, Society for the Study of Reproduction, Phase Committee for SOM curriculum, Phase I remediation sub-committee

Chair, 9th Annual Gilbert S. Greenwald Symposium on Reproduction National Member, Society for the Study of Reproduction, By Laws Committee Member, Society for the Study of Reproduction, Nominations Committee . Thomas M. Yankee, Pharm.D., Ph.D, Associate Professor Department of Microbiology, Molecular Genetics, and Immunology Member, Center for Epigenetics and Stem Cell Biology T cell homeostasis is critical for maintaining the balance between immune competency, autoimmunity, and malignancy. To maintain a steady state number of T cells, we need to continuously produce new T cells to offset the number of T cells that die or differentiate. Our research is focused on the signaling pathways that regulate T cell development and activation. In particular, we study an adaptor protein called Gads and seek to understand the biochemical and biological functions of Gads. Gads consists of an N-terminal SH3 domain, an SH2 domain, a linker region, and a C-terminal SH3 domain. The SH2 and C-terminal SH3 domains bind LAT and SLP-76, respectively. The formation of the LAT/Gads/SLP-76 complex is required for TCR-mediated calcium mobilization. Whether Gads regulates other signaling pathways is currently unknown. In addition, the functions of the N-terminal SH3 domain and the linker region are unclear. Gads can also be phosphorylated, but the biological function of this phosphorylation is unclear. The biological functions of Gads can be divided into two areas: T cell development and T cell activation. T cell development is an ordered series of stages that culminates in the generation of a diverse T cell repertoire with

97

limited ability to recognize self-antigens. Gads is required for the two stages of T cell development that correspond to the stages at which the two chains of the T cell receptor are generated. Defects in these stages can lead to immune deficiency or autoimmune disease. The second area of interest is T cell activation. Although Gads appears to have the same biochemical function in CD4+ T cells and CD8+ T cells, the biological effects of Gads-deficiency are different in these populations. CD4+ T cells fail to survive without Gads while CD8+ T cells are only mildly impaired without Gads. Using an infection model, we showed that Gads is required for optimal expansion of CD8+ T cells, but not for the differentiation of CD8+ T cells into effector or memory cells. Meetings Attended 2012 - “An immunological assessment of B and T cells in patients with chronic graft vs host disease

undergoing extracorporal photopheresis (ECP),” American Society for Blood and Marrow Transplantation, San Diego, CA

2012 – “TCR signaling pathways regulating CD8+ T cell survival during the expansion phase,” 2nd International

Conference on Vaccines and Vaccination, Northbrook, IL 2012 –“Changes in Aiolos and Helios expression in T cell development,” Autumn Immunology Conference,

Chicago, IL Committees KUMC committees Chair, School of Medicine Research Committee Scientific Director, Flow Cytometry Core Laboratory Assessment Workgroup Chair, Phase I curriculum committee Vice Chair, Institutional Biological Safety Committee Alternate scientific member, IACUC Departmental Member, Graduate Affairs Committee, Promotion and Tenure Committee Editorial and Grant Reviews Editorial Board, World Journal of Immunology, ISRN Immunology Grant Reviewer, Study Section, Italian Ministry of Health Grant Reviewer, Study Section, Norman Hackerman Advanced Research Program Academic Honors KUMC Faculty Travel Award

98

Trainees John Szarjeko – Graduate Student Julie Mitchell - Graduate Student Ashraf Hassaballa - Postdoctoral Fellow Alan S. L. Yu, M.B., B.Chir. Harry Statland and Solon Summerfield Professor of Medicine, Director, Division of Nephrology and Hypertension and the Kidney Institute Department of Nephrology and Hypertension Member, Center for Epigenetics and Stem Cell Biology Renal tubule transport of salts, minerals and water Paracellular transport, and the role of tight junction proteins Disorders of mineral metabolism (calcium and magnesium) Claudins and paracellular transport A current focus of the laboratory is to understand the molecular and structural basis of paracellular epithelial transport and its regulation. Paracellular transport refers to transport in between cells. It is now well-recognized that paracellular transport is a major route for vectorial transport of solutes and water. The rate-limiting step in paracellular transport (the paracellular "barrier") is constituted by the tight junction, which is the most apical of the intercellular junctions. Tight junctions consist of large complexes of multiple different proteins. The claudins are a novel family of tight junction proteins that are postulated to form paracellular ion channels. If correct, claudins would likely be structurally and biophysically different from any known ion channels. There are at least 20 different claudin isoforms, raising the exciting possibility that isoform-specific expression may be responsible for the variability in paracellular permeability properties of different epithelial tissues. Investigation of claudin physiology promises to reveal novel insights into the pathogenesis of clinical renal diseases associated with disturbances of the paracellular barrier, such as oliguric acute tubular necrosis, ischemic allograft dysfunction, and certain forms of salt-sensitive hypertension, including pseudohypoaldosteronism, Type II (Gordon's syndrome). We are currently actively investigating the function of these proteins by overexpressing them in cell culture monolayers and performing electrophysiological and tracer flux measurements in Ussing chambers, and by site-directed mutagenesis of key residues in the putative pore-lining region. WNK kinases and renal tubule NaCl reabsorption WNK1 and WNK4 are serine-threonine kinases that regulate transcellular and possibly paracellular salt transport in the distal renal tubule. Mutations in these kinases cause pseudohypoaldosteronism, Type II (PHAII), which is characterized by salt-sensitive hypertension with hyperkalemia. WNKs seem to have broad regulatory roles in the distal tubule epithelium, but the mechanism underlying the pathogenesis of PHAII is still incompletely understood. We are currently exploring the substrates of WNK4 phosphorylation. In collaboration with Dr. Alicia McDonough in the Department of Cell and Neurobiology, we are investigating the role of angiotensin II and reactive oxygen species in the regulation of a key downstream effector of WNK kinases, the thiazide-sensitive NaCl cotransporter, NCC. Meetings Attended 2012 – “Structure-function studies of the claudin-2 ion pore,” Visiting professor, Leibniz Institute for Molecular

Pharmacology, Berlin, Germany 2012 – “Challenges in the diagnosis and management of hyponatremia,” Invited speaker, 2nd Oriental

Congress of Nephrology, Shanghai, China 2012 – “Probing the structure and function of claudin pores,” Invited speaker, Molecular Structure and Function

of the Apical Junctional Complex in Epithelia and Endothelia, Merida, Mexico

99

2012 – “Toward the Structure of Tight Junction Membrane Proteins: Human Claudins,” Center for Structure of Membrane Proteins Annual Meeting, UCSF, San Francisco CA

2012 – “Intensive Review of Internal Medicine Course,” Harvard Medical School. Guest Faculty, Boston, MA 2012 – “Structure-Function Studies of the Claudin Family of Tight Junction Proteins,” PSI:Biology Network

Meeting, NIH, Bethesda, MD Committees KUMC University of Kansas Medical Center EVC Transition Team, Research Committee National Member, NIGMS Protein Structure Initiative Biology Network Steering Committee Editorial and Grant Reviews Section Editor, Current Opinion in Nephrology and Hypertension Editorial Board, American Journal of Physiology: Renal Physiology, Frontiers in Renal and Epithelial

Physiology

Evaluation Board, Faculty of 1000 Medicine Cardiorenal Peer Review Committee, Peer Review Committee, American Heart Association Xuan Zhang, M.D., Ph.D. Senior Scientist Department of Cancer Biology Member, Center for Reproductive Sciences I am interested in the biology and therapy of women’s cancer. Specifically, my research has been focused on the roles of oncogenes such as RET, HGF, FoxM1 in the development and carcinogenesis of the ovary, uterus, and breast, as well as the potential of targeting these signaling pathways in cancer therapeutics. The objectives of my current research are to understand the interactions between p53 and FoxM1 in ovarian cancer, and to explore the role of FoxM1 in ovarian cancer drug resistance. By elucidating the regulation of FoxM1 and its clinical implication, I aim to help improve our understanding of ovarian cancer and advance the prevention and treatment of this lethal disease. Editorial and grant reviews Ad hoc reviewer, BMC Complementary and Alternative Medicine; Expert Review of Endocrinology and

Metabolism; Human Reproduction; International Journal of Biological Science; Molecular Human Reproduction; Reproduction; Reproductive Biology and Endocrinology; Reproductive Sciences

Bao-Ting Zhu, Ph.D. Professor Department of Pharmacology, Toxicology and Therapeutics Member, Center for Reproductive Sciences

100

• Enzymes involved in the multiple pathways of hepatic and extrahepatic estrogen metabolism, and factors that modulate the activity and levels of these metabolizing enzymes.

• Molecular mechanisms underlying the carcinogenic and anticancer actions of some endogenously-formed estrogen metabolites.

• Unique physiological actions (e.g., neuroprotection, neuro-endocrine modulation, immune modulation) exerted by bioactive endogenous estrogen metabolites, and the estrogen receptor-independent mechanism of their actions.

• Identification of novel cellular proteins that can modulate the biological functions of estrogen receptors and their ligands.

Committees Departmental Member, Promotion and Tenure Committee, Postdoctoral Development Committee Editorial and Grant Reviews Editorship/associate editorship, World Journal of Gastroenterology, Contributing Associate Editor-in-Chief

(since 2011) Editorial Board, Experimental and Therapeutic Medicine (since 1/2010) Editorial Board, Anti-cancer Agents in Medicinal Chemistry (Thematic Issue Guest Editor: 2012) Ad hoc Reviewer, Cancer Letters, Carcinogenesis, Chemical Research in Toxicology, Drug Metabolism and

Disposition, International Journal of Cancer, Journal of Endocrinology, Journal of Neurochemistry, Journal of Steroid Biochemistry and Molecular Biology, Nutrition and Cancer, Nutritional Biochemistry, Planta Medica, Steroids, World Journal of Gastroenterology

Seminars Presented 2012 – "Pharmacological and Molecular Study of Estrogen Actions,” Shenzhen University School of Medicine, Shenzhen, Guangdong, China 2012 – "Signaling Pathways Underlying Oxidative Neuronal Death and Protection by Natural Compounds,” KUMC and KU Joint Neuroscience Graduate Program Seminar Trainees Kazushi Okada – Postdoctoral Fellow Hao Zhu, Ph.D. Associate Professor Department of Clinical Laboratory Sciences Member, Center for Epigenetics and Stem Cell Biology As a biochemist and molecular biologist, I have a long-standing research interest in the structure and function of biologically important proteins and their roles in human diseases. My focus in the past twelve years has been on Ncb5or (NADH cytochrome b5 oxidoreductase). This is a novel redox enzyme associated with pathogenesis of lean diabetes. The human Ncb5or gene is linked to lean diabetes, and the Ncb5or knockout mice develop early onset lean diabetes by age 7 weeks due to beta-cell dysfunction and death. Our

101

recent findings show that Ncb5or deficiency in beta-cells leads to profound changes in lipid and iron metabolism, increased oxidative and ER stress, and lipotoxicity similar to that observed in animals with systemic lipid overload. Thus, our Ncb5or-null mouse represents a novel monogenic diabetes model. My lab is currently studying the role of Ncb5or in iron homeostasis and mitochondrial function and their relation to lipid metabolism in beta-cells and other cell types. Meetings Attended 2012 – The 9th Annual Great Plains Pediatric Endocrine Symposium, Kansas City, MO Committees KUMC Member, Promotion and Tenure Committee, Admissions Committee, Molecular Biotechnology Editorial and Grant Reviews Ad hoc reviewer, European Journal of Lipid Science and Technology, Lipids in Health and Disease, Chemico-

Biological Interactions, Journal of Clinical Endocrinology and Metabolism, Biochimie Grant Reviewer, Diabetes UK (The British Diabetic Association), RD Lawrence Fellowship Application,

Portuguese Foundation for Science and Technology, Exact Sciences and Engineering Seminars Presented 2012 – “Altered lipid and iron metabolism in monogenic Ncb5or diabetes,” The 9th Annual Great Plains Pediatric Endocrine Symposium, Kansas City, MO Trainees Jie Dai – Visiting Research Scholar Matthew Stroh – Graduate Student Haiping Wang – Visiting Research Scholar Keegan Zuk – MD Student Vivek Menon – High School Student IRHRM PUBLICATIONS – CY (2012) a. Manuscripts Published Cotichhio G, Albertini DF, and DeSantis L. (2012) Oogenesis Springer. Telfer EE and Albertini DF. (2012) The quest for human ovarian stem cells. Nature Medicine 18:353-354. Limback SD and Albertini DF. (2012) Imaging strategies for studying mammalian oogenesis. Oogenesis. Editors Coticchio G, Albertini DF, and DeSantis L. Ch.1. pp.3-18; Springer Publishing, N.Y. Guglielmo MC and Albertini DF. (2012) The structural basis for coordinating oogenesis and folliculogenesis. Oogenesis. Editors Coticchio G, Albertini DF, and DeSantis L. Ch.7. pp.63-74; Springer Publishing, N.Y. Albertini DF. (2012) Oocyte In Vitro Maturation: Formidable Obstacles on the Road to Fertility Preservation. Fertility Preservation, Eds. Seli E, and Agarwal A. Springer, London Ch. 10,pp121-128.

102

Li R and Albertini DF. (2012) The road to maturation: somatic cell interaction and self-organization of the mammalian oocyte. Nature Reviews Molecular Cell Biology 14:141-152. McGinnis LK, Limback SD, Albertini DF. (2012) Signaling modalities during oogenesis in mammals. Current Topics in Developmental Biology, 227-242. Aljitawi OS, Coates A, Zhang D, Ganguly S, Abhyankar S, Lin T, McGuirk JP. (2012) Umbilical cord graft versus leukemia effect induces remission without the price of graft versus host disease: the possible role of NK cells. Clin Transplant. 26(5):663-4. Aljitawi OS and McGuirk JP. (2012) Multiple myeloma preparative regimens for high-dose therapy and autologous transplantation: What’s new? Journal of Comparative Effectiveness Research, 57-70. Aljitawi, OS. (2012) Ex vivo expansion of umbilical cord blood: where are we? International Journal of Hematology. 95(4):371-9. Ganguly S, Divine CL, Aljitawi OS, Abhyankar S, McGuirk JP, Graves L. (2012) Prophylactic use of zoledronic acid to prevent early bone loss is safe and feasible in patients with acute myeloid leukemia undergoing allogeneic stem cell transplantation. Clinical Transplantation, 26:447–453. Williams T, Aljitawi OS, Moussa R, McHugh S, Dusing R, Abraha J, Yarlagadda SG. (2012) First case of donor transmitted non-leukemic promyelocytic sarcoma. Leuk Lymphoma. 53(12):2530-4. Aljitawi OS, Boone, Jamie M. (2012) Lymphoma Associated Hemophagocytic Lymphohistiocytosis. Blood 120:932. Aljitawi OS, Coats A, Zhang D, Ganguly S, Abhyankar S, Lin T, McGuirk JP. (2012) Umbilical cord graft-versus-leukemia effect induces remission without the price of graft-versus-host disease: the possible role of NK cells. Clinical Transplantation, 26: 663–664. Borude P, Edwards G, Walesky C, Li F, Ma X, Kong B, Guo GL, Apte U. (2012) Hepatocyte specific deletion of farnesoid X receptor delays, but does not inhibit liver regeneration after partial hepatectomy in mice. Hepatology. 56(6):2344-52. Ni HM, Boggess N, McGill MR, Lebofsky M, Borude P, Apte U, Jaeschke H, Ding WX. (2012) Liver Specific Loss of Atg5 Causes Persistent Activation of Nrf2 and Protects Against Acetaminophen-Induced Liver Injury. Toxicol Sci. 127:438-550. Septer S, Edwards G, Gunewardena S, Wolfe AW, Li H, Daniel J, Apte U. (2012) Yes associated protein is involved in proliferation and differentiation in postnatal liver development. Am J Physiol Gastrointest Liver Physiol. 302:G493–G503. Walesky C, Gunewardena S, Terwilliger EF, Edwards G, Borude P, Apte U. (2012) Hepatocyte-Specific Deletion of Hepatocyte Nuclear Factor 4 alpha in Adult Mice Results in Increased Hepatocyte Proliferation. Am J Physiol Gastrointest Liver Physiol. 304:G26-37. Elsarraj HS, Stecklein SR, Valdez K, Behbod F. (2012) Emerging Functions of microRNA-146a/b in Development and Breast Cancer : MicroRNA-146a/b in Development and Breast Cancer. J Mammary Gland Biol Neoplasia. 17(1):79-87.

Behbod F, Rosen JM. (2012) (Editorial): Mammary Gland Development and Breast Cancer; Connecting the dots by non-coding RNAs. J Mammary Gland Biology and Neoplasia. 17(1):1-2.

103

Borrego-Diaz, E, Terai, K, Lialyte, K, Wise, A, Esfandyari, T, Behbod, F, Mautner V.F., Spyra, M, Taylor, S, Parada, L.F., Upadhyaya, M, Farassati, F. (2012) Overactivation of Ras signaling Pathway in CD133+ Malignant peripheral nerve sheet tumor cells. Journal of Neuro-Oncology.108(3):423-34.

Scribner KC, Behbod F, Porter WW. (2012) Regulation of DCIS to invasive breast cancer progression by Singleminded-2s (SIM2s). Oncogene. doi: 10.1038/onc.2012.286. Yeh HW, Gajewski B, Mukhopadhyay P, and Behbod F. (2012) The Zero-truncated Poisson with Right Censoring: an Application to Translational Breast Cancer Research. Statistics in Biopharmaceutical Research. 4(3):252-263.

Stecklein SR, Kumaraswamy E, Behbod F, Wang W, Chaguturu V, Harlan-Williams LM, and Jensen RA. (2012) BRCA1 and HSP90 cooperate in homologous and non-homologous DNA double-strand-break repair and G2/M checkpoint activation. Proc Natl Acad Sci U S A. 109(34):13650-5. Li H, Duhachek-Muggy S, Qi Y, Hong Y, Behbod F, Zolkiewska A. (2012) An essential role of metalloprotease-disintegrin ADAM12 in triple negative breast cancer. Breast Cancer Research and Treatment. 135(3):759-69.

Valdez KE, Elsarraj H, Stecklein S, Behbod F. Targeting Breast Cancer Stem Cells. In Dittmar T, Mihich

E, Zänkereditor KS (Eds.) (2012) Role of Cancer Stem Cells in Cancer Biology and Therapy, pp. 258-283.

Miller DE, Takeo S, Nandanan K, Paulson A, Gogol MM, Noll AC, Walton KN, Gilliland WD, Li H, Staehling-Hampton K, Blumenstiel JP and Hawley RS. (2012) A whole chromosome analysis of recombination and gene conversion in Drosophila melanogaster. G3: Genes, Genomes, Genetics. 2(2):249-260.

Bosak KA. (2012) Managing metabolic syndrome in women. The Nurse Practitioner, 37 (8), 14-21.

Bosak KA. (2012) Managing metabolic syndrome: focus on physical activity. The Journal for Nurse Practitioners (JNP), 8(3), 206-211. Holsen LM, Savage CR, Martin JE, Bruce AS, Lepping RC, Ko E, Brooks WM, Butler MG, Zarcone JR, & Goldstein JM. (2012) Importance of prefrontal inhibitory circuitry in hunger and satiation: The case of Prader-Willi syndrome vs. simple obesity. Int. J. Obesity 36:638-647.

Youngs EL, Dasouki M & Butler MG. (2012) 14q32 deletion syndrome: A clinical report. Clin. Dysmorphol. 21(1):42-44.

Youngs EL, Henkhaus RS, Hellings JA & Butler, MG. (2012) 12-year-old boy with a 4q35.2 microdeletion and involvement of the MTRNA1A, FAT1 and F11 genes. Clin. Dysmorphol. 21:93-96.

Dodani S, Henkhaus RS, Dong L, & Butler MG. (2012) Apolipoprotein A-I gene polymorphisms predict cardio-metabolic risk in south Asian immigrants. Dis. Markers. 32:9-19.

Henkhaus R, Kim S-J, Kimonis VE, Gold J-A, Dykens EM, Driscoll DJ & Butler MG. (2012) Methylation –specific multiplex ligation-dependent probe amplification (MS-MLPA) and identification of deletion genetic subtypes in Prader-Willi syndrome. Genet. Test. Mol. Biomarkers. 16:178-186.

Uppala R, Nath SK, McElreavey K, Uppala R, Sun C, Hutchings D, Maiti AK, Newkirk HL, Sharp AJ, Everman DB, Murray J, Schwartz C, Antonarakis SE, & Butler MG. (2012) A 710 kb duplication on chromosome 1p31.3 causes autosomal dominant omphalocele. J. Med. Genet. 49:270-276.

Honea RA, Holsen LM, Lepping RJ, Perea R, Butler MG, Brooks WM & Savage CR. (2012) The neuroanatomy of genetic subtype differences in Prader-Willi syndrome. Am. J. Med. Genet. B. (Neuropsychiatr Genet).159B(2):243-253.

104

Youngs EL, Henkhaus R, Hellings JA & Butler MG. (2012) IL1RAPL1 gene deletion as a cause of X-linked mental retardation. Eur. J. Med. Genet. 55:32-36.

Manzardo A.M. Henkhaus R, Dhillon S & Butler MG. (2012) Plasma cytokine levels in children with autistic disorder and unrelated siblings. Int. J. Dev. Neuroscience. 30:121-127.

Rosenbloom ST & Butler MG. (2012) Development and implementation of electronic growth charts for infants with Prader-Willi syndrome. Am. J. Med. Genet. 158A(11):2743-49.

Butler MG, Youngs EL, Roberts JL & Hellings JA. (2012). Assessment and treatment in autism spectrum disorders: A focus on genetics and psychiatry. Autism Res. Treat. 2012:242537.

Manzardo AM, Hidaka B, Henkhaus R, Penick EC, Poje AB, & Butler MG. (2012) X chromosome inactivation in women with alcoholism. Alcohol. Clin. Exp. Res. 36(8):1325-1329.

Manzardo A, Henkhaus R & Butler MG. (2012) Global DNA promoter methylation in frontal cortex of alcoholics and controls. Gene. 498(1):5-12.

Rethmeyer J, Tan X, Manzardo A, Schroeder SR & Butler MG. (2012) Comparison of biological specimens and DNA collection methods for PCR amplification and microarray analysis. Clin. Chem. Lab. Med. 6:1-5

Mayo-Ortega L, Oyama-Ganiko R, Leblanc J, Schroede SR, Brady N, Butler MG, Reese RM, Richman DM, Peacock G, Foster J & Marquis J. (2012) Mass screening for severe problem behavior among infants and toddlers in Peru. J. Ment. Health Res. Intellect. Disabil. 5(3-4):246-259.

Scheimann AO, Butler MG, Miller JL, Lee PD, Stevenson DA, Heinemann J & Driscoll DJ. (2012) Letter to the editor: Long-term experience with duodenal switch in adolescents. Obesity Surgery, 22:517-518. Butler MG & Meaney FJ. (2012) Growth in Prader-Willi syndrome: Anthropometric patterns and analysis. In: Handbook of Growth and Growth Monitoring in Health and Disease, V.R. Preedy (ed.), Springer, New York, NY. Butler MG. (2012) Prader-Willi and Angelman Syndromes: Examples of Genomic Imprinting. In: Clinical Genomics: Practical Applications in Adult Patient Care, F. Alkuraya, M. Murray, M. Babyatsky, M. Giovanni, and D. Stewart (eds.), McGraw Hill Publishers: New York, NY. Butler MG. (2012) Genomic Imprinting Disorders in Obesity. In: Obesity- Epidemiology, Pathophysiology and Prevention, D. Bagchi and H. G. Preuss (eds.), CRC Press/Taylor & Francis Group, Boca Raton, FL. Cain S & Butler MG. (2012) The Ethics of Genetics in Child and Adolescent Psychiatry. In: Ethics Curriculum on the American Academy of Child & Adolescent Pyschiatry website (www.aacap.org). Cox JH, Doorlag D, Loker J, Loker C, & Butler MG. (2012). In: Nutritional Care for Infants and Toddlers with Prader-Willi Syndrome, L. Keder and D. Spencer (eds.), Prader-Willi Syndrome Association (USA), Sarasota, FL. Butler MG & Cassidy SB. (2012) Genetic Basis, Genetic Testing and Genetic Counseling for Prader-Willi Syndrome. In: Prader-Willi Syndrome, C. Hoybye (ed.), Nova Science Publishers, Inc., Hauppauge, NY. Butler MG. (2012) Prader-Willi Syndrome. In: Elsevier’s First Consult, M. Gilliam (ed.), Elsevier, Philadelphia, PA. Guo Y, Xu M, Deng B, Frontera JR, Kover KL, Aires D, Ding H, Carlson SE, Turk J, Wang W, Zhu H. (2012) Beta-cell injury in NcB5or-null mice is exacerbated by consumption of a high-fat diet. Eur J Lipid Sci Technol. 114(3):233-243.

105

Colombo J, Carlson SE. (2012) Is the measure the message: The BSID and nutritional interventions. Invited commentary. Pediatrics. 129:1166-7. He X, Khurana A, Maguire JL, Chien J, Shridhar V. (2012) HtrA1 sensitizes ovarian cancer cells to cisplatin-induced cytotoxicity by targeting XIAP for degradation. Int J Cancer. 130(5):1029-35. Fan JB, Chen J, April CS, Fisher JS, Klotzle B, Bibikova M, Kaper F, Ronaghi M, Linnarsson S, Ota T, Chien J, Laurent LC, Nisperos SV, Chen GY, Zhong JF. (2012) Highly parallel genome-wide expression analysis of single mammalian cells. PLoS ONE. 7(2):e30794. Fang WB, Jokar I, Dendukuri P, Cheng N. (2012) CCL2 derived from carcinoma associated fibroblasts

enhances mammary carcinoma cell motility and cell growth mediated by p42/44MAPK and Smad3 signaling

pathways. Journal of Biological Chemistry 287(43):36593-608.

Roberts JE, Hatton DD, Bailey D, Long ACJ, Anello V, & Colombo J. (2012) Visual attention and autistic

behavior in infants with fragile X syndrome. Journal of Autism and Developmental Disabilities. 42:937-946.

Brez CN, & Colombo J. (2012) Your eyes say “no,” but your heart says “yes.” Behavioral and

psychophysiological indices in infant quantitative processing. Infancy. 17:445-454.

Colombo J & Carlson SE. (2012) Is the measure the message? The BSID and nutritional interventions.

Pediatrics. 129,1166-1167.

Brez CC, Colombo J, & Cohen L. (2012) Infants' integration of featural and numerical information. Infant Behavior and Development. 35:705-710.

Aréchaga J, Damjanov I. (2012) Above the borderland between normal and neoplastic development. Int J Dev

Biol. 56(10-11-12):939-948.

Damjanov I. (2012) Medical students writing letters to their patients: teaching communication and empathy.

Acta Med Acad. 41(1):4-6.

Petersson F, Bulimbasic S, Hes O, Slavik P, Martínek P, Michal M, Gomolčáková B, Hora M, Damjanov I.

(2012) Biphasic alveolosquamoid renal carcinoma: a histomorphological, immunohistochemical, molecular

genetic, and ultrastructural study of a distinctive morphologic variant of renal cell carcinoma. Ann Diagn Pathol.

16(6):459-69.

Dormer NH, Singh M, Zhao L, Mohan N, Berkland CJ, Detamore MS. (2012) Osteochondral interface regeneration of the rabbit knee with macroscopic gradients of bioactive signals. Journal of Biomedical Materials Research Part A. 100(1):162-170. Ingavle G, Dormer NH, Gehrke SH, Detamore MS. (2012) Using chondroitin sulfate to improve the viability and biosynthesis of chondrocytes encapsulated in interpenetrating network (IPN) hydrogels of agarose and poly(ethylene glycol) diacrylate. Journal of Materials Science: Materials in Medicine. 23(1):157-170. Dormer NH, Qiu Y, Lydick AM, Allen ND, Mohan N, Berkland CJ, Detamore MS. (2012) Osteogenic differentiation of human bone marrow stromal cells in hydroxyapatite-loaded microsphere-based scaffolds. Tissue Engineering Part A. 18(7-8):757-767. Renth AN, Detamore MS. (2012) Leveraging ‘raw materials’ as building blocks and bioactive signals in regenerative medicine. Tissue Engineering Part B. 18(5):341-362.

106

Gutheil W, Reed G, Ray A, Anant S and Dhar A. (2012) Crocetin: a novel agent derived from saffron for

prevention and therapy for cancer. Current Pharmaceutical Biotechnology 13:173-179..

Dhar A, Fogt L, Subramaniam D and Anant S. (2012) Cancer Stem Cells: Novel target using dietary

components for prevention and treatment. Nutraceutical and Cancer ed Sarkar, F.S. (2012) P11-38.

Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, Evans RM. (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature, 485(7396):123-7.

DiTacchio L, Bowles J, Shin S, Lim DS, Koopman P and Janknecht R. (2012) Transcription Factors ER71/ETV2 and SOX9 Participate in a Positive Feedback Loop in Fetal and Adult Mouse Testis. J. Biol Chem. 287(28):23657 66. DiTacchio L, Vollmers C, Hatori M, Bushong E, Gill S, LaBlanc M, Fitzpatrick J, Ellisman M, and Panda S. (2012) Time-restricted feeding prevents adverse effects of a high-fat diet. Cell Metabolism, 15(6):848-60. Lowry BN, Rigler SK, McClain E, Kalender-Rich JL, Broxterman JT, Eck LM, Williamson TL, Simpson SQ, Lisbon DP, Vansaghi LM. (2012) Adaptation of ABIM/ACGME Developmental Milestones into an Internal Medicine Residency Curriculum at a University Medical Center - Examples of Rotational Curricula [Internet]. Association of American Medical Colleges: iCollaborative. Available from: https://www.aamc.org/icollaborative/meded/275086/resource271.html Hussain H, Eck LM. (2012) hCG Induced Hyperthyroidism Due to a Metastatic Germ Cell Tumor. Kansas Journal of Medicine. 5(2):58-61. Rockwell CE, Zhang M, Fields PE, and Klaassen CD. (2012).Th2 skewing by Activation of Nrf2 in CD4+ T Cells. J. Immunol. 188(4):1630-1637. Grantham JJ, Mulamalla S, Grantham CJ, Wallace DP, Cook LT, Wetzel LH, Fields TA, and Bae KT. (2012) Detected Renal Cysts Are Tips of the Iceberg in Adults with ADPKD. Clin J Amer Soc Nephrol 7(7):1087-93. Wang L, Ellis M, Gomez JA, Eisner W, Fennell W, Howell DN, Ruiz P, Fields TA, Howell DN, Spurney RF. (2012) Mechanisms of proteinuria induced by Rho GTPases. Kidney Int 81(11):1075-85, 2012. Spangenburg EE, Geiger PC, Leinward LA, Lowe DA. (2012) Novel Mechanistic Insights Into the Role of Female Sex Steroids in Regulating Physiological and Metabolic Function of Striated Muscle. Invited Reivew, Med Sci Sports Exerc , 44(9):1653-62. Touchberry CD, Gupte AA, Bomhoff GL, Graham ZA, Geiger PC, Gallagher PM. (2012) Hyperthermic preconditioning protects skeletal muscle from damage and alters hypertrophic signaling. Cell Stress Chaperones. 17(6):693-705.. Gustafson K M, May L E, Yeh H-W, Million S K, Allen JJB. (2012) The influence of maternal exercise on fetal cardiac autonomic control during breathing movements. Early Human Development. 88; 539-546. May LE, Suminski R, Yeh H-W, Langaker M Gustafson KM. (2012) Regular maternal exercise dose and fetal heart outcome. Medicine & Science in Sports & Exercise. 44; 1252-1258. Kibe R, Zhang S, Guo D, Marrero L, Tsien F, Rodriguez P, Khan S, Zieske A, Huang J, Li W, Durum SK, Iwakuma T, Cui Y. (2012) IL-7Rα deficiency in p53null mice exacerbates thymocyte telomere erosion and lymphomagenesis. Cell Death & Differentiation. 19(7):1139-51.

107

Agarwal N, Adhikari AS, Iyer SV, Hekmatdoost K, Welch DR, and Iwakuma T. (2012) MTBP suppresses cell migration and filopodia formation by inhibiting ACTN4. Oncogene. 32(4):462-70. Iwakuma T and Agarwal N. (2012) MDM2 Binding Protein, a novel metastasis suppressor. Cancer Metastasis Review. 31(3-4):633-40. Iyer SV and Iwakuma T. (2012) A novel link between the HER2-Akt and MDM2-p53 pathways via CSN6. News and View, Cell Cycle. 11(22). Johnson R. (2012). Reprogramming of Somatic Cells. Progress in Molecular Biology and Translational Science 111: 51-82. Dubey S, VanVeldhuizen P, and Karan D. (2012). Adenovirus-based immunotherapy for prostate cancer. Current Cancer Therapy Reviews. 8 (4): 264-270.

Hashmi M, Karan D, Phadke S, Saunthararajah Y, Kambhampat S, and VanVeldhuizen P. (2012). Prostate cancer immunotherapy: an evolving field. Current Cancer Therapy Reviews. 8(4): 274-282. Karan D and VanVeldhuizen P. (2012). Combination immunotherapy with prostate GVAX and ipilimumab: safety and toxicity. Immunotherapy. 4 (6): 577-580.

Karan D and Peter VanVeldhuizen P. (2012). Editorial: Hot topics in prostate cancer immunotherapy. Current Cancer Therapy Reviews. 8 (4): 237.

Karan D, Holzbeierlein JM, VanVeldhuizen P, and Thrasher JB. (2012). Cancer immunotherapy: a paradigm shift for prostate cancer treatment. Nature Reviews Urology. 9 (7): 376-385. Dubey S, VanVeldhuizen P, Holzbeierlein JM, Tawfik O, Thrasher JB and Karan D. (2012). Inflammation associated regulation of macrophage inhibitory cytokine (MIC-1) gene in prostate cancer. Oncology Letters 3 (5): 1166-1170. Finocchario-Kessler S, Bastos FI, Malta M, Anderson J, Goggin K, Sweat M, Dariotis J, Bertoni N, Kerrigan D. (2012) Discussing Childbearing with HIV-infected Women of Reproductive Age in Clinical Care: A Comparison of Brazil and the US. AIDS and Behavior. 16(1):99-107.

Finocchario Kessler S, Sweat M, Anderson J, Dariotis J, Jennings J, Keller J, Vyas A, Trent M. (2012)

Childbearing Motivations, Pregnancy Desires, and Perceived Partner Response to Pregnancy Among Urban

Female Youth; Does HIV Serostatus Make a Difference? AIDS Care 24(1):1-11.

Finocchario-Kessler S, Catley D, Thompson N, Bradley-Ewing A, Berkley-Patton J, Goggin K. (2012) Patient

Communication Tools to Enhance ART Adherence Counseling in Low and High Resource Settings. Patient

Education and Counseling. 89(1):163-70.

Finocchario Kessler, S., Mabachi, N., Dariotis, JK, Anderson, J., Goggin, K.., Sweat, M. (2012) “We weren’t

using condoms because we were trying to conceive”: The need for reproductive counseling for HIV+ women in

clinical care. AIDS Patient Care and STDs. 26(11):700-7.

JG Park, Ye Q, Singh V, Kieweg SL, Misra A, Spencer P. (2012) Synthesis and evaluation of novel dental

monomer with branched aromatic carboxylic acid group. Journal of Biomedical Materials Research: Part B:

Applied Biomaterials 100B: 569-576.

Osaka I, Hills JM, Kieweg SL, Shinogle HE, Moore DS, and Hefty PS. (2012) An automated image-based

108

method for rapid analysis of Chlamydia infection as a tool for screening antichlamydial agents. Antimicrobial

Agents and Chemotherapy 56(8): 4184-4188.

Hu B, Kieweg SL. (2012) The effect of surface tension on the gravity-driven thin film flow of Newtonian and

power-law fluids. Computers & Fluids, 64, pp. 83-90.

Kim SS. (2012) Assessment of long term endocrine function after transplantation of frozen-thawed human

ovarian tissue to the heterotopic site: 10 year longitudinal follow-up study. J Assist Reprod Genet 29(6):489-93.

Jadoul P, Kim SS. (2012) Fertility considerations in young women with hematological malignancies. J Assist

Reprod Genet 29(6):479-87.

Klemp J, Kim SS (2012) Fertility preservation in young women with cancer. J Assist Reprod Genet 29(6):469-

87.

Kim SS, ISFP (2012) Recommendations for Fertility Preservation in Patients with Lymphoma, Leukemia, and

Breast Cancer. J Assist Reprod Genet 29(6):465-68.

Krieg SA, Fan X, Hong Y, Sang QX, Giaccia A, Westphal LM, Lathi RB, Krieg AJ, Nayak NR. (2012) Global

alteration in gene expression profiles of deciduas from women with idiopathic recurrent pregnancy loss. Mol

Hum Reprod. 18(9):442-50.

Fan, X., Krieg, S.A.,Hwang, J.Y., Dhal, S.D., Kuo, C.J., Lasley, B.L., Brenner, R.M. and N.R. Nayak (2012)

Dynamic regulation of WNT7a expression in primate endometrium: implications for postmenstrual regeneration

and secretory transformation. Endocrinology 153 (3):1063-1069.

Home P, Saha B, Ray S, Dutta D, Gunewardena S, Yoo B, Pal A, Vivian JL, Larson M, Petroff M, Gallagher

PG, Schulz VP, White KL, Golos TG, Behr B, Paul S (2012) Altered subcellular localization of transcription

factor TEAD4 regulates first mammalian cell lineage commitment. Proc Natl Acad Sci USA 109(19):7362-7.

Staat BC, Galan HL, Harwood JE, Lee G, Marconi AM, Paolini CL, Cheung A, Battaglia FC. (2012) Transplacental supply of mannose and inositol in uncomplicated pregnancies using stable isotopes. J Clin Endocrinol Metab. 97:2497. Jan R, Brewer C, Huang M, Lewis-Wambi JS. (2012) Loss of PEDF expression: A novel mechanism for the development of endocrine resistance. Breast Cancer Res. 14:R146. Yang J, Xie SX, Huang Y, Ling M, Liu J, Ran Y, Wang Y, Thrasher JB, Berkland C, Li B. (2012) Prostate-targeted biodegradable nanoparticles loaded with androgen receptor 1 silencing constructs eradicate xenograft tumors in mice. Nanomedicine (Lond). 2012 May 14. [Epub ahead of print]. Zhao Y, Duan S, Zeng X, Liu C, Davies NM, Li B, Forrest ML. (2012) Prodrug strategy for PSMA-targeted delivery of TGX-221 to prostate cancer cells. Mol Pharm. 4;9(6):1705-16. Yao K, Youn H, Gao X, Huang B, Zhou F, Li B, Han H. (2012) Casein kinase 2 inhibition attenuates androgen receptor function and cell proliferation in prostate cancer cells. The Prostate. 15;72(13):1423-30. Cai Z, Li B, Li K, Zhao B. (2012) Down-Regulation of Amyloid-β Through AMPK Activation by Inhibitors of GSK-3β in SH-SY5Y and SH-SY5Y-AβPP695 Cells. J Alzheimers Dis. 29(1):89-98.

109

Chen P, Yu J, Chalmers B, Drisko J, Yang J, Li B, and Chen Q. (2012) Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy. Anticancer Drugs 23(4):437-44. Liu W, Fan LX, Zhou X, Sweeney WE Jr., Avner ED, Li X. (2012) HDAC6 regulates epidermal growth factor receptor (EGFR) endocytic trafficking and degradation in renal epithelial cells. PLoS One. 7(11):e49418. Fan LX, Li X, Magenheimer BS, Calvet JP, Li X. (2012) Inhibition of histone deacetylases targets the transcription regulator Id2 to attenuate cystic epithelial cell proliferation. Kidney Int. 81:76-85. Manzardo AM, Henkhaus R, Dhillon S, Butler MG. (2012) Reduction of Cytokine Levels in Children with

Classical Autism, International Journal of Developmental Neuroscience. (2): 121-7.

Manzardo AM, Henkhaus R, Butler MG. (2012) Genome-Wide Promoter Methylation in Frontal Cortex of

Alcoholics and Controls, Gene. 498(1): 5-12, 2012.

Manzardo AM, Henkhaus R, Hidakka B, Penick EC, Poje A, Butler MG. (2012) X Chromosome Inactivation in

Blood from Women with Alcoholism, Alcoholism Clinical and Experimental Research. 36(8): 1325-9.

Madarasz W, Manzardo AM, Mortensen EL, Penick EC, Knop J, Sorensen HJ, Becker U, Nickels EJ,

Gabrielli WF. (2012) 45-year Mortality Rate as a Function of the Number and Type of Psychiatric Diagnosis

Found in a Large Danish Birth Cohort, Canadian Journal of Psychiatry. 57(8):505-511.

Rethmeyer JA, Tan X, Manzardo AM, Butler MG. (2012) Comparison of biological specimens and DNA

collection methods for PCR amplification and microarray analysis, Clinical Chemistry and Laboratory Medicine.

6:105.

McAllister CE, Creech R, Kimball P, Muma NA, Li Q. (2012) GPR30 is necessary for estradiol-induced

desensitization of 5-HT1A receptor signaling in the paraventricular nucleus of the rat hypothalamus.

Psychoneuroendocrinology 37:1248-60.

Muma NA. (2012) Focus on RGS Proteins: Impact on the Treatment of Depression and Anxiety. International

Journal of Neuropsychopharmacology 25: 1-2.

Creech RD, Li Q, Carrasco GA, Van de Kar LD, Muma NA. (2012) Estradiol induces partial desensitization of

serotonin 1A receptor signaling in the paraventricular nucleus of the hypothalamus and alters expression and

interaction of RGSZ1and Gαz. Neuropharmacology, 62:2040-2049.

Anderson JE, Warner L, Jamieson DJ, Kissin DM, Nangia AK, Macaluso M. (2012) Contraceptive sterilization among married adults: National data on who chooses vasectomy and tubal sterilization. Contraception. 85:552-7. Zhang X, Healy C, Nothnick WB. (2012) Estrogen suppresses expression of the matrix metalloproteinase inhibitor reversion-inducing-cysteine-rich protein with Kazal motifs (RECK) within the mouse uterus. Endocrine. 42:97-106.

Nothnick WB. (2012) The role of microRNAs in the female reproductive tract. Reproduction. 143:559-576. Home P, Saha B, Ray S, Dutta D, Gunewardena S, Yoo B, Pal A, Vivian JL, Larson M, Petroff, M, Gallagher PG, Schulz V, Golos TG, Behr B, Paul S. (2012) Altered subcellular localization of transcription factor TEAD4 regulates first mammalian cell lineage commitment. Proc. Natl. Acad. Sci. USA 109(19):7362-67.

110

Pal A, Fontanilla D, Gopalakrishnan A, Chae YK, Markley JL, Ruoho AE. (2012) The sigma-1 receptor protects against cellular oxidative stress and activates antioxidant response elements. Eur J Pharmacol. 682(1-3):12-20. Home P, Saha B, Dutta D, Ray S, Pal A, Gunewardena S, Yoo B, Vivian J, Larson M, Petroff M, Gallagher P G, Schulz V, White KL, Golos T G, Behr B, and Paul S. (2012) Altered Subcellular Localization of Transcription Factor TEAD4 Regulates First Mammalian Cell Lineage Commitment. Proc. Natl. Acad. Sci. U.S.A. 109(19):7362-7. Hong J, He H, Bui P, Ryba-White B, Rumi MA, Soares MJ, Dutta D, Paul S, Kawamata M, Ochiya T, Ying QL, Rajanahalli P, Weiss ML. (2012) A focused microarray for screening rat embryonic stem cells. Stem Cells. Dev. 22:431-43.

Costa FC, Fedosyuk H, Neades R, Bravo de los Rios J, Barbas III CF, and Peterson KR. (2012) Induction of

fetal hemoglobin in vivo mediated by a synthetic -globin zinc finger activator. Anemia.

doi:10.1155/2012/507894.

Peterson KR, Fedosyuk H, and Harju-Baker S. (2012) LCR 5’ hypersensitive site specificity for globin gene

activation within the active chromatin hub. Nucleic Acids Res. 40:11256-11269.

Costa F C, Fedosyuk H, Chazelle AM, Neades RY, and Peterson KR. (2012. Mi2β is required for -globin

gene silencing: Temporal assembly of a GATA-1-FOG-1-Mi2 repressor complex in β -YAC transgenic mice.

PLoS Genet. 8:e1003155. doi:10.1371/journal.pgen.1003155.

Jasti S, Warren BD, McGinnis LK, Kinsey WH, Petroff BK, Petroff MG. (2012) The autoimmune regulator

prevents premature reproductive senescence in female mice. Biol Reprod. April, 12;86(4):110.

Franczak A, Zmijewska A, Kurowicka B, Wojciechowicz B, Petroff BK and Kotwica, G. (2012) The effect of

tumor necrosis factor alpha, interleukin 1 beta and interleukin 6 on endometrial prostaglandin synthesis,

metabolism and release in early-pregnant pigs. Theriogenology. 77:155-165.

Petroff BK. (2012) A case for combined breast and ovarian cancer prevention. Med. Hypoth. 79:549-551.

Pierucci-Alves F, Yi S and Schultz BD. (2012) Transforming growth factor beta 1 induces tight junction disruptions and loss of transepithelial resistance across porcine vas deferens epithelial cells. Biol Reprod 86: 36, 1-8. Zhang X, Samadi AK, Roby KF, Timmermann B, Cohen MS. (2012) Inhibition of cell growth and induction of apoptosis in ovarian carcinoma cell lines CaOV3 and SKOV3 by natural withanolide Withaferin A. Gynecol Oncol. 124(3):606-12. Dembinski JL, Wilson SM, Spaeth EL, Studeny M, Zompetta C, Samudio I, Roby KF, Andreeff M, Marini FC. (2012) Tumor stroma engraftment of gene-modified mesenchymal stem cells as anti-tumor therapy against ovarian cancer. Cytotherapy. 15:20-32 Pierucci-Alves F, Yi S, Schultz BD. (2012). Transforming growth factor beta 1 induces tight junction disruptions and loss of transepithelial resistance across porcine vas deferens epithelial cells. Biol Reprod. 86, 36.

111

Bukovnik U, Gao J, Cook GA, Shank LP, Seabra MB, Schultz BD, Iwamoto T, Chen J, Tomich JM. (2012). Structural and biophysical properties of a synthetic channel-forming peptide: Designing a clinically relevant anion selective pore. Biochim Biophys Acta. 1818,1039-1048. Al-Bataineh MM, Van Der Merwe D, Schultz BD, Gehring R. (2012). Molecular and functional identification of organic anion transporter isoforms in cultured bovine mammary epithelial cells (BME-UV). J Vet Pharmacol Ther. 35, 209-215. Ruan X, Crupper SS, Schultz BD, Robertson DC, Zhang W. (2012). Escherichia coli expressing EAST1 toxin did not cause an increase of cAMP or cGMP levels in cells, and no diarrhea in 5-day old gnotobiotic pigs. PLoS One. 7, e43203. Schultz BD. (2012). Airway epithelial cells: 'Bicarbonate in' not equal 'Bicarbonate out'. J Physiol. 590, 5263-5264. Griebling T, Liao Z, Smith PG. (2012) Systemic and topical hormone replacement therapies reduce vaginal

innervation density in post-menopausal women. Menopause 19:630-635.

Doss ALN and Smith PG. (2012) Nerve-Langerhans Cell Interactions in Diabetes and Aging, Histology and

Histopathology 27:1589-1598.

Asanoma K, Kubota K, Chakraborty D, Renaud SJ, Wake N, Fukushima K, Soares MJ, Rumi MA. (2012) SATB proteins regulate trophoblast stem cell renewal and differentiation. J Biol Chem. 287: 2257-68. Soares MJ, Chakraborty D, Renaud SJ, Kubota K, Bu P, Konno T, Rumi MA. (2012) Regulatory pathways controlling the endovascular invasive trophoblast cell lineage. J Reprod Dev. 58(3): 283-7. Chakraborty D, Rumi MA, Soares MJ. (2012) NK cells, hypoxia and trophoblast cell differentiation. Cell Cycle. 11(13):2427-30. Bhattacherjee A, Rumi MA, Staecker H, and Smith PG. (2012) Bone Morphogenetic Protein 4 mediates estrogen-regulated sensory axon plasticity in the adult female reproductive tract. J Neurosci. 33(3):1050-61a. Soares MJ, Chakraborty D, Rumi MA, Konno T, Renaud SJ. (2012) Rat placentation: an experimental model for investigating the hemochorial maternal-fetal interface. Placenta. 33: 233-243. Asanoma K, Kubota K, Chakraborty D, Renaud SJ, Wake N, Fukushima K, Soares MJ, Rumi MA. (2012) SATB proteins regulate trophoblast stem cell renewal and differentiation. J Biol Chem 287: 2257-68. Hong J, He H, Bui P, Ryba-White B, Rumi MA, Soares MJ, Dutta D, Paul S, Kawamata M, Ochiya T, Ying QL, Rajanahalli P, Weiss M. (2012) A focused microarray for screening rat embryonic stem cell lines. Stem Cells Dev. 22(3):431-43. Chuong EB, Rumi MA, Soares MJ, Baker JC. Endogenous retroviruses function as species-specific enhancer elements in the placenta. Nature Genetics 45:325-329. Kent LN, Ohboshi S, and Soares MJ. (2012) AKT1 and IGF2 regulate placentation and fetal and postnatal development. International Journal of Developmental Biology 56, 255-261. Hayakawa K, Nakanishi M, Ohgane J, Tanaka S, Takamori-Hirosawa M, Soares MJ, Yagi S, and Shiota K. (2012) Bridging sequence diversity and tissue-specific expression by DNA methylation in genes of the mouse prolactin superfamily. Mammalian Genome 23, 336-345.

112

Shankar K, Zhong Y, Kang P, Soares MJ, Badger TM, and Gomez-Acevedo H. (2012) RNA-seq analysis of the functional zones within the rat placentation site. Endocrinology 153, 1999-2011. Burns J, Honea RA, Vidoni ED, Hutfles L, Brooks WM, Swerdlow, RH. (2012) Insulin is differentially related to cognitive decline and atrophy in Alzheimer's disease and aging. BBA- Mol Basis Dis 1822:333-339. Swerdlow RH. (2012) Mitochondria and cell bioenergetics: Increasingly recognized components and a possible etiologic cause of Alzheimer’s disease. Antiox Redox Signal 16:1434-1455. Swerdlow RH. (2012) Alzheimer’s disease pathologic cascades: Who comes first, what drives what. Neurotoxicity Research 22:182-194. Vidoni ED, Honea RA, Billinger SA, Swerdlow RH, Burns JM. (2012) Cardiorespiratory Fitness is associated with atrophy in Alzheimer's and aging over two years. Neurobiol Aging 33:1624-1632.

Swerdlow RH. (2012) -apptists and tauists, it’s time for a sermon from the book of biogenesis. J Neurochem 120:347-349. Swerdlow RH, Jicha G. (2012) Alzheimer’s Disease: Can the exam predict the pathology? Neurology 78:374-375. Rademakers R, Baker, M, Nicholson AM, Rutherford NJ, Finch N, Soto-Ortolaza A, Lash J, Wider C, Wojtas A, DeJesus-Hernandez M, Adamson J, Kouri N, Sundal C, Shuster EA, Aasly J, MacKenzie J, Roeber S, Kretzschmar HA, Boeve BF, Knopman DS, Petersen RC, Cairns NJ, Ghetti B, Spina S, Garbern J, Tselis AC, Uitti R, Das P, Van Gerpen JA, Meschia JF, Levy S, Broderick DF, Graff-Radford N, Ross OA, Miller BB, Swerdlow RH, Dickson DW, Wszolek ZK. (2012) Mutations in the colony stimulating factor 1 receptor (CSF1R) cause hereditary diffuse leukoencephalopathy with spheroids. Nat Genet 44:200-205. Aires D, Rockwell G, Wang T, Frontera J, Wick J, Wang W, Tonkovic-Capin M, Lu J, E L, Zhu H, Swerdlow

RH. (2012) Potentiation of dieterary restriction-induced lifespan extension by polyphenols. Biochim Biophys

Acta 1822:522-526.

Swerdlow RH, Corder E. (2012) For Alzheimer’s GWAS, pulling needles from the haystack is just the first

step. Neurology 79:204-205.

Sundal C, Van Gerpen JA, Nicholson A, Wider C, Shuster E, Aasy JO, Spina S, Ghetti B, Roeber S, Garbern

J, Borjesson-Hanson AM, Tselis A, Swerdlow RH, Miller B, Shinsuke F, Heckman M, Uitti R, Josephs K,

Baker M, Andersen O, Rademakers R, Dickson D, Broderick D, Wszolek ZK. (2012) MRI characteristics and

scoring in HDLS due to CSF1R gene mutations. Neurology 279:566-574.

Lez E, Swerdlow RH. (2012) Mitochondria in neurodegeneration. Adv Exp Med Biol. 942:269-86.

Honea RA, Vidoni ED, Swerdlow RH, Burns JM. (2012) For the Alzheimer’s Disease Neuroimaging Initiative.

Maternal family history is associated with Alzheimer’s disease biomarkers. JAD 31:659-668.

Harris JL, Yeh HW, Choi IY, Lee P, Berman NE, Swerdlow RH, Craciunas SC, Brooks WM. (2012) Altered

neurochemical profile following traumatic brain injury: 1H-MRS biomarkers of pathological mechanisms.

JCBFM 32:2122-2134.

Swerdlow RH, Newell KL. (2012) “Untangling” the relationship between AD and DLB. Neurology 79:1938-1939.

113

Arduino DM, Estevex AR, Cortes L, Silva DFF, Oliveira CR, Swerdlow RH, Cardoso SM. (2012) Mitochondrial metabolisms in Parkinson’s disease impairs quality control autophagy by hampering mitcrotubule-dependent traffic. Hum Mol Genet 21:4680-4702. Martins-Branco D, Esteves AR, Santos D, Arduino DM, Swerdlow RH; Oliveira CR, Januario C, Cardoso SM. (2012) Ubiquitin Proteasome System in Parkinson Disease: a keeper or a witness? Exp Neurol 238:89-99. Swerdlow RH. (2012) Does Mitochondrial DNA Play a Role in Parkinson’s Disease? A Review of Cybrid and other Supportive Evidence. Antiox Redox Signal 16:950-964. Silva DF, Selfridge JE, Lu J, E L, Cardoso SM, Swerdlow, RH. (2012) Mitochondrial abnormalities in Alzheimer's disease: possible targets for therapeutic intervention. Adv Pharmacol 64:83-126.

Swerdlow, RH. (2012) Mitochondria in Neurodegeneration. In Choi LY and Gruetter R (eds): Neural

Metabolism In Vivo, Advances in Neurobiology 4: Springer, New York, 885-907.

Home P, Saha B, Ray S, Dutta D, Gunewardena S, Yoo B, Vivian JL, Larson M, Petroff M , Gallagher PG, Schulz V, White KL, Thaddeus G, Golos TG , Behr B, Paul S. (2012) Altered Subcellular Localization of Transcription Factor TEAD4 Regulates First Mammalian Cell Lineage Commitment. PNAS 109(19):7362-7. Weiner CP. (2012) New tools for an old quest. Am J Obstet Gynecol. doi:pii:S0002-9378(12)02263-6. 10.1016/j.ajog.(Epub ahead of print). Yankee TM. (2012) B-Cell Antigen Receptor. In: Lennarz WJ and Lane MD. (eds.) The Encyclopedia of Biological Chemistry. Vol. 3, pp. 161-164. Waltham, MA: Academic Press. Lee DH, Maunsbach AB, Riquier-Brison A, Nguyen MX, Fenton RA, Bachmann S, Yu AS, McDonough AA. Effects of ACE inhibition and AngII stimulation on renal Na-Cl cotransporter distribution, phosphorylation and membrane complex properties. Am J Physiol Cell Physiol, 304(2):C147-63. Engelund MB, Yu ASL, Li J, Madsen SS, Færgeman NJ, Tipsmark CK. (2012) Functional characterization and localization of a gill specific claudin isoform in Atlantic salmon. Am J Physiol Regul Integr Comp Physiol, 302:R300-11. Hanner F, Lam L, Nguyen MX, Yu AS. (2012) Peti-Peterdi J. Intrarenal localization of the plasma membrane ATP channel pannexin1. Am J Physiol Renal Physiol, 303(10):F1454-9. Zhang X, Timmermann B, Samadi AK, Cohen MS. (2012) Withaferin A induces proteasome-dependent degradation of breast cancer susceptibility gene 1 and heat shock factor 1 proteins in breast cancer cells. ISRN Biochemistry. Article ID 707586. Fukui M, Kang KS, Okada K and Zhu BT. (2013) EPA, an omega-3 fatty acid, induces apoptosis in human pancreatic cancer cells in vitro and in vivo: Role of ROS accumulation, caspase 8 activation, and autophagy induction. Journal of Cellular Biochemistry 114: 192-203.

Zhu BT. (2012) Natural compounds as cancer chemopreventive and chemotherapeutic agents: Insights gained

from mechanistic and pharmacologic studies. Anticancer Agents in Medicinal Chemistry 12:1-2.

Fu X, Wang P, Fukui M, Long C, Yin L, Choi HJ and Zhu BT. (2012) Pancreas-specific protein disulfide

isomerase (PDIp) is a major intracellular estrogen-storage protein that modulates the tissue levels of estrogen

in the pancreas. Biochemical Journal 447: 115-123.

114

Choi HJ and Zhu BT. (2012) Cyclin B1/Cdc2 up-regulation is required for the induction of mitotic prometaphase arrest in human breast cancer cells treated with 2-methoxyestradiol. Biochimica et Biophysica Acta (BBA) — Molecular Cell Research 1823: 1306-1315.

Fukui M and Zhu BT. (2012) Rapid generation of mitochondrial superoxide induces mitochondria-dependent

but caspase-independent hippocampal neuronal cell death that morphologically resembles necroptosis.

Toxicology and Applied Pharmacology 262: 156-166.

Guo Y, Xu M, Deng B, Frontera JR, Kover KL, Ding HL, Aires D, Carlson SE, Turk J, Wang WF, and Zhu H.

(2012) Beta-cell injury in Ncb5or-null mice is exacerbated by consumption of a high-fat diet”, European J. Lipid

Science and Technology, 114:233–243.

Aires D, Rockwell G, Wang T, Frontera J, Wick J, Wang WF, Tonkovic-Capin M, Lu J, E L, Zhu H, and

Swerdlow RH. (2012) Potentiation of dietary restriction-induced lifespan extension by polyphenols”, Biochim

Biophys Acta- Molecular Basis of Diseases, 1822:522-526.

Kang KS, Yamabe N, Wen Y, Fukui M and Zhu BT. (2013) Effects of tea catechins and bioflavonoids on

levodopa methylation and neurodegeneration. Brain Research 1497:1-14.

Fukui M, Kang KS, Okada K and Zhu BT. (2013) EPA, an omega-3 fatty acid, induces apoptosis in human

pancreatic cancer cells in vitro and in vivo: Role of ROS accumulation, caspase 8 activation, and autophagy

induction. Journal of Cellular Biochemistry 114:192-203.

b. Manuscripts in Press

Albertini DF and Olsen R. Effects of fertility preservation on oocyte genomic integrity. Oocyte Biology in Fertility Preservaion; Editor S S Kim; Chapter 4 Springer-Verlag, N.Y. Walesky C, Edwards G, Borude P, Gunewardena S, O’Neil M, Yoo B, and Apte U. Hepatocyte Nuclear Factor 4 alpha Deletion Promotes Diethylnitrosamine-induced Hepatocellular Carcinoma in Mice. Hepatology. Hays A, Apte U, and Hagenbuch B. Organic Anion Transporting Polypeptides Expressed in Pancreatic Cancer May Serve As Potential Diagnostic Markers and Therapeutic Targets for Early Stage Adenocarcinomas. Pharmaceutical Res.

Elsarraj HS, Hong Y, Valdez K, Carletti M, Salah SM, Taverna D, Prochasson P, Bharadwaj U, Tweardy DJ,

Christenson LK, and Behbod F. Novel Role of MicroRNA146b in promoting mammary alveolar progenitor cell

maintenance. Journal of Cell Science.

Stecklein S, Elsarraj H, Valdez K, Paul A, Behbod F. Breast cancer invasion and metastasis. In Malek A (Ed),

Experimental Metastasis: Modeling and Analysis.

Butler MG, Smith B, Lee J, Gibson C, Schmoll C, Moore W & Donnelly JE. Effects of growth hormone

treatment in adults with Prader-Willi syndrome. Growth Horm. IGF Res.

Anderson CJ, & Colombo J. Pupil and salivary indicators in autism spectrum disorder. Developmental Psychobiology. Salley BJ, Panneton R & Colombo J. Separable predictors of language outcome. Infancy.

115

Carlson SE, Colombo J, Gajewski B, Gustafson KM, Mundy D, Yeast J, Georgieff MK, Markley L, Kerling EH & Shaddy DJ. Docosahexaenoic acid supplementation and pregnancy outcomes. American Journal of Clinical Nutrition. Kapa LL, & Colombo J. Attentional control in early and later bilingual children. Cognitive Development. Gustafson KM, Carlson SE, Colombo J, Yeh H-W, Shaddy DJ, Li S, Kerling EH. Effects of docosahexaenoic acid supplementation during pregnancy on fetal heart rate and variability: A randomized clinical trial. Prostaglandins, Leukotrienes, and Essential Fatty Acids. Eck LM. Should Family Physicians Routinely Screen for Vitamin D Deficiency? Yes: Targeted Screening in At-Risk Populations Is Prudent. American Family Physician.

Kumru AM, Rouse M, Vansaghi LM, Eck LM. Hydroxychloroquine Associated Hyperinsulinemic Hypoglycemia. Kansas Journal of Medicine.

Zhang S, Gillihan R, He N, Fields TA, Liu S, Green T, and Stubbs JR. Dietary phosphate restriction

suppresses phosphaturia but fails to prevent FGF23 elevation in a mouse model of chronic kidney disease.

Kidney Int.

Sharma M, Magenheimer LK, Home T, Tamano KN, Singhal PC, Hyink DP, Klotman PE, Vanden Heuvel GB,

Fields TA. Inhibition of Notch pathway attenuates the progression of human immunodeficiency virus

associated nephropathy. Am J Physiol Renal Physiol.

Geiger PC and Gupte AA. The role of estrogen in the regulation of peripheral glucose dynamics. Integrative Biology of Women’s Health, Ed. EE Spangeburg. Springer. Steiner R, Dariotis JK, Finocchario-Kessler S. The time has come to engage HIV providers in conversations

with their reproductive age clients about fertility desires and intentions: A historical review of the HIV epidemic

in the United States.

Krieg SA, Fan X, Hong Y, Sang Q, Giaccia A, Westphal L, Lathi R, Krieg A. and Nayak NR. Global alteration

in gene expression profiles of deciduas from women with idiopathic recurrent pregnancy loss. Molecular

Human Reproduction.

Jan R, Brewer C, Smith A, King T, Simms P, Lewis-Wambi JS. Phospholipid Scramblase 1 mediates

estrogen-induced apoptosis in endocrine resistant breast cancer. Mol. Cancer Res.

Butler MG, Roberts J, Hayes J, Tan X, Manzardo AM. Growth Hormone Receptor Gene Polymorphism and Prader-Willi Syndrome, American Journal of Medical Genetics, Part A. Li Q, Sullivan NR, McAllister CE, Van de Kar LD, Muma NA. Estradiol accelerates the effects of fluoxetine on

serotonin 1A receptor signaling, Psychoneuroendocrinology.

Murray KS, James A, McGeady JB, Reed ML, Kuang WW, Nangia AK. The Effect of the New 2010 World Health Organization Criteria for Semen Analyses on Male Infertility. Fertil Steril.

Eisenberg ML, Lathi RB, Baker VL, Westphal LM, Milki AA, Nangia AK. The Frequency of Male Infertility in the USA. J Urol.

Smith, JS, Nangia AK. Epididymovasostomy: Tips and Tricks of the Trade. Editor Jay Sandlow. Publisher: Springer.

116

Stecklein SR, Elsarraj H, Valdez KE, Paul A, Behbod F. Modeling and Analysis of Breast Cancer Invasion and Metastasis. Experimental Metastasis: Modeling and Analysis, Malek, Springer.

Saha, B., Home, P., Ray, S., Pal, A., Larson, M., Rajendran, G., Behr, B., and Paul, S. EED and KDM6B

Coordinate First Mammalian Cell Lineage Commitment to Ensure Embryo Implantation, Mol Cell Biol.

Petroff BK, Phillips T, Kimler BF, Fabian CJ. Comparison of RNA endpoints in fixed vs. frozen benign human breast tissue harvested by ductal lavage or random periareolar fine needle aspiration (RPFNA). Reprod Biol. Al-kasspooles MF, Williamson SK, Henry D, Howell J, Niu F, Decedue CJ, Roby KF. Preclinical antitumor activity of a nanoparticulate SN38. Invest New Drugs.

Roby KF. Endocrine Disruptors. In: Ovarian Toxicology, 2nd Edition Hoyer PB (ed) New York, NY

E L, Lu J, Burns JM, Swerdlow RH. Effect of exercise on mouse liver and brain bioenergetic infrastructures.

Exp Physiol.

Selfridge JE, E Lezi, Lu J, Swerdlow RH. Role of mitochondrial homeostasis and dynamics in Alzheimer’s

disease. Neurobiol Dis.

Burns JM, Swerdlow RH. Backwaters and Rapids on the Amyloid River. Neurology.

Swerdlow RH, E L, Aires D, Lu J. Glycolysis-Respiration Relationships in a Neuroblastoma Cell Line. BBA-

General Subjects.

Dai Y, Zheng K, Clark J, Swerdlow RH, Pulst SM, Sutton JP, Shinobu LA, Simon DK. Rapamycin drives

selection against a pathogenic heteroplasmic mitochondrial DNA mutation. Autophagy.

Uysal U, Seremet S. Lamping JW, Adams JM, Liu DY, Swerdlow RH, Aires DJ. Consumption of polyphenol

plants may slow aging and associated diseases. Current Pharmaceutical Design.

Burns JM, Claassen D, Swerdlow RH. Brain Disorders: Memory. In Jones R, Aminoff M, Burns TM, Pomeroy

S (eds): Netter Neurology Atlas. Elsevier, Philadelphia.

Xiong J, Parker BL, and Yankee TM. IL-7 supports survival of TCRβ-expressing CD4(-) CD8(-) double negative thymocytes. Immunol. Jiang XR, Wang P, Smith CL and Zhu BT. Synthesis of novel estrogen receptor antagonists using metal-catalyzed coupling reactions and characterization of their biological activity. Journal of Medicinal Chemistry. Choi HJ and Zhu BT. Role of cyclin B1/Cdc2 in mediating Bcl-XL phosphorylation and apoptotic cell death following nocodazole-induced mitotic arrest. Molecular Carcinogenesis. c. Abstracts Aljitawi OS, Ganguly S, Abhyankar S, Wolfe K, Daniels K, Ferree M, Marks R, Kramer W, Pipkins JD, McGuirk JP. (2012) Phase IIa Open Label Randomized, Pharmacokinetic Comparative Cross-over Study of Melphalan HCL for Injection (Propylene Glycol Free) and Alkeran for Injection for Myeloablative Conditioning in Multiple Myeloma Patients Undergoing Autologous Transplantation. Biol Blood and Marrow Transplantation. 18: S257 (Abstract 143).

Abhyankar S, DeJarnette S, Aljitawi O, Ganguly S, Merkel D, McGuirk J. (2012) A Risk- Based Approach to Optimize Autologous Hematopoietic Stem Cell (HSC) Collection with the Use of Plerixafor. Bone Marrow Transplant. 47(4):483-7.

117

Kapil,. Bhalla, Fiskus W, Sharma S, Horrigan S, Mudunuru U, Reyes R, Abhyankar SH, McGuirk J, Aljitawi OS, Ganguly S. (2012) Anti-AML activity of a novel β-catenin antagonist BC2059. Chicago, ASCO.

Ganguly S, Loknath AK, Williams C, Divine C, Aljitawi O, Abhyankar S, McGuirk JP. (2012) BEAM and BEAC with or without Rituximab (R-BEAM or R-BEAC) are comparable and Effective Preparative Regimens for Patients with B-cell Lymphoma Undergoing Autologous Hematopoietic Stem Cell Transplantation. EBMT.

Ganguly N, Satelli D, Aljitawi O, McGuirk JP, Abhyankar S, Ganguly S. (2012) Extra-corporeal photopheresis in patients with extensive chronic graft-versus-host disease does not lower risk of infection in the early post ECP period in spite of its steroid sparing effect. EBMT.

Ganguly S, Amin M, Aljitawi O, Abhyankar S, McGuirk JP. (2012) Decitabine in relapsed acute myeloid leukemia after allogeneic bone marrow transplantation. EBMT, Annals of Hematology.

Fiskus W, Sharma S, Rao R, Balusu R, Venkannagari S, Hembruff S, Abhyankar S, McGuirk J, Aljitawi O, Ganguly S, Bearss D and Bhalla KN. (2012) Combined targeting of chromatin modifying enzymes LSD1, histone deacetylases (HDACs) and EZH2 has superior efficacy against human acute myeloid leukemia cells. Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research. Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1043.doi:1538- 445.AM2012-1043.

Aljitawi OS, Xiao Y, Eskew J, Parelkar N, Swink M, Radel J, McGuirk JP, Broxmeyer HE, Vielhauer G. (2012) Hyperbaric Oxygen Theraphy Might Alter Umbillical Cord Blood Engraftment Kinetics Post-Transplant. International Cord Blood Symposium.

Aljitawi OS, Xiao Y, Zhang D, Eskew J, Parelkar N, Swink M, Detamore M, Vielhauer G, Akel S. (2012) A Wharton’s Jelly Mesenchymal Stromal Cell Derived 3D Osteogenic Scaffold Does Not Result in Expansion of the CD34+ Population of Umbilical Cord Blood Mononuclear Cells. International Cord Blood Symposium.

Aljitawi OS, Ludlow A, Divine C, Lin T, Ganguly S, Abhyankar S, McGuirk JP. (2012) Recovery of Oral Mucositis Following Ablative Umbilical Cord Blood Transplantation is Independent of Neutrophil Recovery. International Cord Blood Symposium.

Pope C, Bhushan B, Walesky C, Edwards G, Borude P and Apte U. (2012) Mechanistic differences in liver regeneration after partial hepatectomy and regeneration after acetaminophen induced acute liver failure. Hepatology 56(4):1644. Apte U, Borude P, Jaeschke H, Lee WM (2012) Identification of novel biomarkers of liver regeneration following acetaminophen-induced acute liver failure. Hepatology 56(4):1676. Manley SM, Guo G, Apte U, and Ding WX. Modulation of autophagy by bile acids in hepatocytes and liver. FASEB J March 29, 2012 26:396.4 (oral presentation at 2012 Experimental Biology meeting). Ding WX, Ni HN, Apte U, and Jaeschke H. Liver Specific Knockout Atg5 Causes Persistent Activation of Nrf2 and Protects Against Acetaminophen-Induced Liver Injury. FASEB J March 29, 2012 26:396.3 (poster presentation at 2012 Experimental Biology meeting).

Walesky, C, Gunewardena S, Edwards G, Borude C, Apte U. (2012) Hepatocyte nuclear factor alpha (HNF4 ) knockdown stimulates pro-mitogenic gene expression in hepatocytes FASEB J 26:274.12 (oral presentation at 2012 Experimental Biology meeting). Bosak K. (2012) Abstract: Effects of real-time automated feedback to promote physical activity. Journal of Women’s Health, 20(10), 1389. ISSN: 1540-9996.

118

Roberts J, Hayes J, Dzidic N, Hovanes K, Dasouki M, Butler MG. (2012) Chromosomal microarray analysis of individuals with autism or learning deficits presenting for genetic services. (Abstract #3054). Presented at the 62nd Annual Meeting of The American Society of Human Genetics, November 9, 2012, Sans Francisco, California.

Butler MG, Roberts J, Hayes J, Tan X, Manzardo A. (2012) Growth hormone receptor gene polymorphism and Prader-Willi syndrome. (Abstract #3089). Presented at the 62nd Annual Meeting of The American Society of Human Genetics, November 7, 2012, San Francisco, California. Valdez KE, Fan F, Cusick T, Smith WP, Behbod F. (2012) Regulation of stem cells and their aberrant self-

renewal pathways by steroid hormones during ductal carcinoma in situ malignant progression. Gordon

Research Conference on Mammary Gland Biology, Lucca Italy.

Elsarraj HS, Hong Y, Valdez K, Carletti M, Salah SM, Raimo M, Taverna D, Prochasson P, Bharadwaj U,

Tweardy DJ, Christenson LK, and Behbod F. (2012) MicroRNA146b promotes mammary alveolar progenitor

cell maintenance through preferential regulation of STAT3. Gordon Research Conference on Mammary Gland

Biology, Lucca Italy.

Cheng, N. (2012) “Targeting the CCL2/CCR2 chemokine pathway in breast tumors through intra-tumoral delivery of calcium cross-linked TAT peptide: siRNA complexes,” AACR Annual Meeting, Chicago, IL. Cheng, N. (2012) “Targeting the CCL2/CCR2 chemokine pathway in breast tumors through intratumoral delivery of calcium cross linked TAT peptide:siRNA complexes,” ESAB, Kansas City, KS. Cheng, N. (2012) “Role of CXCL1 signaling in mammary tumor progression,” KUCC Research Symposium, Kansas City, KS.

Bibikova M, Humphray S, Grocock R, Peden J, Nielsen F, Chien J, Goode E, Ho V, April C, Munchel S, Cottrell

J, Tarabishy Y, Visscher V, Wieben E, Hartmann L, Kalli K, Viji S, Fan J-B, Heath G and Bentley D. (2012) Whole-genome and targeted sequencing analysis of early stage high-grade serous ovarian cancer. American Society of Human Genetics 2012, November 6-10.

Cicek MS, Koestler DC, Fridley BL, Armasu SM, Kalli KR, Larson MC, Winterhoff BJ, Chien J, Fan JB, Bibikova

M, Konecky G, Shridhar V, Cunningham JM, and Goode EL. (2012) Distinct DNA Methylation Signature in Ovarian Cancer Histological Subtypes. American Society of Human Genetics 2012, November 6-10.

Pierce AN, Christianson JA. (2012) Enhanced vaginal sensitivity and anxiety in adult mice that underwent neonatal stress and/or irritation. Program No. PH 410. 14th World Congress on Pain; Milan, Italy: International Association for the Study of Pain. Online.

Yang F-C, Tan T, Abdel Samad O, Liu Y, Christianson JA, Roberson D, Davis B, Ma Q. (2012) Runx1 selectively specifies the cutaneous pain pathway. Program No. 80.18. 2012 Neuroscience Meeting Planner. New Orleans, LA: Society for Neuroscience.

Pierce AN, Wang R, Ryals JM, Christianson JA. (2012) Enhanced vaginal sensitivity and anxiety in adult mice that underwent chronic neonatal stress. Program No. 473.01. 2012 Neuroscience Meeting Planner. New Orleans, LA: Society for Neuroscience. Online.

Christianson JA, Pierce AN, Wang R, Zhang Z, Ryals JM. (2012) Alterations in nociceptive processing following neonatal vaginal irritation and maternal separation. Program No. 473.02. 2012 Neuroscience Meeting Planner. New Orleans, LA: Society for Neuroscience. Online.

119

Rangarajan P, Subramaniam D, Anant S and Dhar, A. (2012) Targeting colon cancer stem cells using sulforopahne. AACR 103rd Annual Meeting: Abstract #5419. Oertel M, Rose C, Eck LM. (2012) Bisphosphonate-Associated Hip Fracture with Prodromal Symptomatology. Endocr Rev. Vol. 33 (03_MeetingAbstracts): SAT-370.

Rose C, Eck LM. (2012) Ectopic Cushing Syndrome: A Difficult Diagnosis. Endocr Rev. Vol. 33 (03_MeetingAbstracts): SAT-464. Fields KS, Vivian CJ, Wallace DP, and Fields TA. (2012) MCP-1 Is the Primary Macrophage Recruitment Factor Produced by Human and Mouse Polycystic Kidney Cells and Promotes Disease Progression in cpk Mice. American Society of Nephrology 2012 Meeting. San Diego, CA, Oct 30-Nov 4, 2012 (invited talk). Rogers RS, Beauoin MS, Wheatley JK, Wright DC, Geiger PC. (2012) Acute heat treatment alters adipose tissue fatty acid handling. Integrative Biology of Exercise, Westminster, CO. Wheatley, JL, White KS, and Geiger PC. (2012) Metabolic signaling in L6 muscle cells in response to heat treatment and resveratrol. Integrative Biology of Exercise, Westminster, CO. Dubey S and Karan D. 2012. Analysis of PSA-specific T cell response in PSA-transgenic mouse: a useful model to study prostate cancer immunotherapy. Journal of Immunotherapy 35 (9): 771. Dubey S, Van Veldhuizen, P, Thrasher JB and Karan D. 2012. Analysis of PSA-specific T cell response in PSA-transgenic mouse: a useful model to study prostate cancer immunotherapy. The University of Kansas Cancer Center Research Symposium, November 8, 2012. Abstract # 16: p.42. Hu B and Kieweg SL. (2012) Numerical study of epithelial deformation during vaginal application of a viscoelastic gel using a fluid-structure interaction model. American Society of Mechanical Engineers (ASME) Summer Bioengineering Conference, Fajardo, Puerto Rico. Osaka I, Kieweg SL, and Hefty PS. (2012) Alkylpolyamine DS-96 Prevents Chlamydia trachomatis Infection by Blocking the Attachment Process. American Society for Microbiology 2012 General Meeting, San Francisco,. Whitmore TW, Camarda KV, and Kieweg SL. (2012) Evaluation of Cellulose Ethers for Design of Polymeric Delivery Vehicles. 2012 Annual Meeting of the American Institute of Chemical Engineers (AICHE), Pittsburgh, PA. Hu B and Kieweg SL. (2012) Contact line instability of gravity-driven flow of power-law fluids. American Physical Society Division of Fluid Dynamics 2012 Meeting, San Diego, CA. Gustafson KM, Carlson SE, Yeh H-W, Colombo J, Li S, Shaddy DJ, Kerling EH. (2012) Docosahexaenoic acid supplementation During Pregnancy Results in Higher Fetal Heart Rate Variability at 36 weeks gestation. 10th Congress of the International Society for the Study of Fatty Acids and Lipids. May 26 – 30, Vancouver, British Columbia, Canada. May, L, Suminski, RS, Gustafson, KM, Yeh, H-W.(2012) Maternal continuous vs. Intermittent Exercise and the Fetal Heart. FASEB J 2012.

Thangavel J, Dawn, B and Johnson R. (2012). Effects of Epigenetic Modifiers on LPS-induced eNos/Rho

Signaling And Restoration of Adherens Junction Integrity and Endothelial Barrier Protection. Circulation.

2012;126:A11114.

Thangavel J, Dawn B, and Johnson R. (2012). Reprogramming of Human Somatic Cells Into Cardiomyocytes.

Circulation. 2012;126:A11107.

120

Kim SS. (2012) Assessment of long-term endocrine function after transplantation of frozen-thawed human

ovarian tissue to the heterotopic site: 10 year longitudinal follow-up study. Hum Reprod, 27 (suppl1): i117 (O-

298).

Kim SS, Collins L, He L, Dong Y, Kim DJ, Artigues A. (2012) Identification of altered protein expression after

cryopreservation of human ovarian tissue. Fertil Steril, 98 (3S);S89 (O-302).

Graham SM, Holzbeierlein JM, Yang J, Xie SX, Berkland C, Thrasher JB, Li B. (2012) Nanoparticle-based

prostate cancer cell-specific delivery of androgen receptor siRNA induces rapid tumor regression in nude mice.

Annual meeting of the AUA. May 20-25, 2012. Atlanta, GA.

Graham SM, Holzbeierlein JM, Thrasher JB, Li B. (2012) Calcium channel blocker modulates androgen

receptor-mediated gene expression and induces cytotoxicity in prostate cancer cells. Annual meeting of the

AUA. May 20-25, 2012. Atlanta, GA.

Brewer C, Jan R, Smith A, King T, Simms PJ, Lewis-Wambi JS (2012) STAT-1 activation enhances

phospholipid scramblase 1-mediated apoptosis in aromatase inhibitor resistant breast cancer cells.

Proceedings of the American Association for Cancer Research, Abstract #265, Chicago, IL.

Rosso F, Chen R, Huang Y, Thrasher JB, Li B. (2012) Novel natural compound alternol activates the apical

apoptosis pathway in prostate cancer cells. Annual meeting of the SCS AUA. Oct 24-27, 2012. Colorado

Springs, CO.

Zhang R and Li B. (2012) Long Noncoding RNA-mediated regulation of BCL-2 expression in Prostate cancers.

KUCC Annual Symposium, KUMC, Nov 8, 2012. Kansas City, KS.

Huang Y and Li B. (2012) Targeted Delivery of Nanomicelle-Encapsulated p110Beta-Specific Inhibitor for

Prostate Cancer Intervention. KUCC Annual Symposium, KUMC, Nov 8, 2012. Kansas City, KS

Manzardo AM, Hidaka B, Henkhaus R, Penick EC, Poje AB, Butler MG. (2012) X Chromosome Inactivation In Blood And Prefrontal Cortex Of Women With Alcoholism, #22, Institute for Reproductive Health and Regenerative Medicine 2012 Congress, April 19, 2012. Manzardo AM, Henkhaus R, Butler MG. (2012) Genome-Wide Promoter Methylation in Prefrontal Cortex of Alcoholics and Controls Using Nimblegen DNA Methylation 2.1m Arrays, #23, Institute for Reproductive Health and Regenerative Medicine 2012 Congress, April 19, 2012. Kanukuntla T, Manzardo AM, Penick EC, Poje A, Campbell J. (2012) What Predicts Neuropathic Pain in Severely Alcohol Dependent Subjects? American Psychiatric Association, 2012. Bhatia K, Manzardo AM, Butler MG. (2012) Gene Expression Patterns from Brain Samples in Alcoholics. Resident, Posdoc, Fellows Research Day, 2012. Bhatia K, Manzardo AM, Butler MG. (2012) Gene Expression Patterns from Brain Samples in Alcoholics. #35054 American Psychiatric Association, May 5, 2012. Karim S, Knop J, Penick EC, Jensen P, Nickel EC, Manzardo AM, Mortensen EL, Gabrielli WF. (2012) Do Premorbid Alcohol Challenge Responses Predict Alcohol Dependence by Age 40? American Psychiatric Association, 2012.

121

Butler MG, Roberts J, Hayes J, Tan X, Manzardo AM. (2012) Growth Hormone Receptor Gene Polymorphism and Prader-Willi Syndrome. American Society of Human Genetics, San Francisco, CA, November 8, 2012. Butler MG, Roberts J, Hayes J, Tan X, Manzardo AM. (2012) Growth Hormone Receptor Gene Polymorphism and Impact on Growth in Prader-Willi Syndrome. 26th Annual PWSA Scientific Day Conference, Baton Rouge, LA, October 19-20, 2012. Mason CW, Dong Y, Zhou H, Weiner CP. (2012) Expression, Localization, and Function of Purkinje Cell Protein 4 (PCP4) in Human Myometrium. Society for Gynecological Investigation, March 21-24, San Diego, CA. Mason CW, Dong Y, Buhimschi IA, Buhimschi C, Weiner CP. (2012) Purkinje Cell Protein 4 (PCP4) Repression in Human Myometrium During Preterm Labor. Society for Maternal Fetal Medicine, February 9-11, Dallas, TX. Warren BD, Jasti S, Petroff BK, Petroff MG. (2012) Autoimmune deficiency is associated with embryonic loss and generation of autoantibodies against the uterus, placenta and embryo. Annual Meeting of the Society for the Study of Reproduction, State College, PA. Kimler BF, Metheny T, Phillips T, Zalles C, Petroff BK, Fabian CJ. (2012) Reduction in Ki-67 in benign breast tissue of high risk premenopausal women with the SERM acolbiphene. Annual Meeting of the American Society of Clinical Oncology, Chicago, IL. Petroff BK. (2012) Effects of tamoxifen on markers of follicular health in rats treated with cancer chemotherapy. Annual Greenwald Symposium. Kansas City, KS. Warren, BD, Jasti S, Petroff BK, Petroff MG. (2012) The Autoimmune Regulator (AIRE) protects against

infertility, reproductive tract inflammation and germ cell loss in male Balb/c mice. Annual Greenwald

Symposium. Kansas City, KS.

Li S, Hidaka BH, Harvey KE, Sullivan DK, Klemp JR, Carlson SE, Petroff BK, Kimler BF, Fabian CJ. (2012)

Breast cancer risk biomarkers are associated with dietary intake and tissue content of n-3 polyuns turated fatty

acids. Annual Meeting of the International Society for the Study of Fatty Acid Lipids, Vancouver, CA.

Yi S, Pierucci-Alves F, and Schultz BD. (2012) TGF-β1 impairs CFTR-mediated anion secretion across

cultured porcine vas deferens epithelial monolayer via the p38 MAPK pathway. Ped Pulm Supp 35: 242.

Gupta V, Roby KF, Kern B, Hall T, Jakkaraj S, Chakrasali R, Georg GI, Broward M, Wood R, Weir S, Tash JS. (2012) KU-AS-272, a potential single-dose sterilant for cats and dogs shows safety and ability to block spermatogenesis in testis to sertoli cells only after a single subcutaneous injection in male rat. 45th Annual Meeting of the Society for the Study of Reproduction. State College, Pennsylvania. Holets LM, Gupta V, Roby KF, Tash JS. (2012) Spaceflight inhibits ovarian follicle development, induces down regulation of estrogen receptor alpha and alters metabolic pathways and gene expression in mice uterus. 45th Annual Meeting of the Society for the Study of Reproduction. State College, Pennsylvania. Roby KF, Nangia AK, Krieg SA, Hastings RC, Mijal RS. (2012) Glutathione S-transferase Polymorphisms and Mechanisms of Male Infertility. Frontiers: The Heartland Institute for Clinical and Translational Research 2012 Annual Symposium, Kansas City, Kansas. Rumi MAK, Kubota K, Ratri A, Chakraborty D, Bugarinovic G, Roby KF, Larson, MA, Vivian JL, Wolfe MW,

122

Soares MJ. (2012) Zinc Finger Nuclease Targeted Disruption of Estrogen Receptor Alpha Signaling in the Rat. 9th Annual Gilbert Greenwald Symposium on Reproduction. Kansas City, Kansas. Gupta V, Roby KF, Kern B, Hall T, Jakkaraj S, Chakrasali R, Georg GI, Broward M, Wood R, Weir S, Tash JS. (2012) KU-AS-272, a potential single-dose sterilant for cats and dogs shows safety and ability to block spermatogenesis in testis to sertoli cells only after a signle subcutaneous injection in male rat. 9th Annual Gilbert Greenwald Symposium on Reproduction. Kansas City, Kansas. Roby KF, Nangia AK, Krieg SA, Hastings RC, Mijal RS. (2012) Glutathione S-transferase polymorphisms and mechanisms of male infertility. 9th Annual Gilbert Greenwald Symposium on Reproduction, Kansas City, KS. Rumi MA, Chakraborty D, Asanoma K, Kubota K, Bu P, PhD, Renaud SJ, and Soares MJ. (2012) Transcriptional hierarchy of SATB homeobox proteins in trophoblast stem cells. Fetal Programming and Environmental Exposures: Implications for Prenatal Care and Pre-Term Birth. The New York Academy of Sciences, NY June 11-12, 2012. Renaud SJ, Rumi MA, and Soares MJ. (2012) Focal adhesion kinase regulates AP-1 factor expression and invasion in trophoblast cells. Reproductive Sciences, 2012; 19(3S): 387A (59th Annual Meeting of the Society for Gynecologic Investigation, San Diego, CA. March 2012). Chuong EB, Rumi MA, Soares MJ, Baker JC. (2012) Endogenous retroviruses facilitate rapid evolution of core trophoblast regulatory network. Society of Molecular Biology and Evolution, Dublin, Ireland, June 23-26 2012. Kubota K, Rumi MA, Kent LN, Soares MJ. (2012) FOSL1 regulation of the invasive trophoblast lineage: partners and gene targets. 45th Annual Meeting of the Society for the Study of Reproduction, State College, Pennsylvania. August 2012. Renaud SJ, Rumi MA, and Soares MJ. (2012) OVO-like 1 is a Regulator of human trophoblast differentiation. Society for the Study of Reproduction ACM, Penn State University, Happy Valley, PA Aug 2012. Chakraborty D, Rumi MAK, Krieg AJ, and Soares MJ. (2012) Role of hypoxia signaling in trophoblast cell lineage commitment. FASEB Meeting on Transcriptional Regulation, Snowmass, CO. Bu P, Alam SM, Yagi S, Shiota K, Kumar TR, Morohashi K, Vivian JL, Rumi MA and Soares MJ. (2012) Origin of a species-specific rheostat controlling testicular growth and steroidogenesis. 45th Annual Meeting of the Society for the Study of Reproduction, State College, Pennsylvania, August 2012. Wilson NR, Olm-Shipman AJ, Kosa E, Pitstick L, Stumpff KM, Smith G, Bjork BC, Czirok A, Saadi I. (2012) Deficiency of SPECC1L down-regulates PI3K-AKT signaling in the pathogenesis of Oblique Facial Clefts. American Society of Human Genetics Annual Meeting, San Francisco, CA. Rafi SK, Olm-Shipman AJ, Saadi I (2012) Unraveling the Molecular Mechanisms Underlying the Teratogenicity of Topamax (Topiramate). American Society of Human Genetics Annual Meeting, San Francisco, CA. Wilson NR, Olm-Shipman AJ, Kosa E, Pitstick L, Stumpff KM, Smith G, Bjork BC, Czirok A, Saadi I (2012) Cytoskeletal SPECC1L is a novel modulator of cell adhesion, cell motility, actin stability and PI3K-AKT signaling. American Society of Cell Biology Annual Meeting, San Francisco, CA. Gehring R, Malreddy P, Silver K, Wang F and Schultz B. (2012). In vitro culture alters the expression of xenobiotic transporters by mammary epithelial cells. FASEB J 26. Gehring LA., Malreddy PR, Schultz BD and Gehring R. (2012).In-vitro culture of bovine mammary epithelial cells alters the expression of breast cancer resistance protein (BCRP, ABCG2) and P-glycoprotein (P-gp, ABCB1). In: Merck-Merial NIH Veterinary Scholars Symposium.

123

Yi S, Pierucci-Alves F & Schultz BD. (2012). TGF-β1 impairs CFTR-mediated anion secretion across cultured porcine vas deferens epithelial monolayer via the p38 MAPK pathway. Ped Pulm. Supp 35, 242. Swenson-Fields KI, Vivian CJ, Wallace DP and Fields TA (2012). MCP-1 is the Primary Macrophage Recruitment Factor Produced by Human and Mouse Polycystic Kidney Cells and Promotes Disease Progression in cpk Mice. 2012 American Society of Nephrology Meeting, San Diego, CA. Peda J, Swenson-Fields, KI and Fields, TA. (2012) Macrophages in Polycystic Kidney Disease. KUMC Student Research Forum. Salah, SM, Swenson-Fields, KI and Fields, TA. (2012) The Role of Macrophages in Autosomal Dominant Polycystic Kidney Disease Progression. KUMC Student Research Forum. Swerdlow, RH. (2012) Mitochondria and Sporadic Neurodegenerative Disease. Biophysical Society 56th Annual Meeting Proceedings. Chavan H, Lu J, Swerdlow R, Krishnamurthy P. (2012) Effect of mitochondrial ATP binding cassette transporter B6 (ABCB6) in arsenic toxicity and mitochondrial function. Society for Toxicology 2012 Meeting Proceedings. E L, Billinger S, Swerdlow RH. (2012) The effects of exercise training on mouse liver and brain energetic metabolism: lactate and activation of monocarboxylate transporter 2 (MCT2). APTA Annual Meeting Proceedings. Sundal C,Van Gerpen J, Fujioka S, Aasly J, Wider C, Roeber S, Shuster E, Ghetti B, Garbern J, Tselis A, Swerdlow R, Rademakers R, Dickson D, Broderick D, Wszolek Z. (2012) Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS): observational scoring system for brain MRI. Journal NeuroImaging 22:101. Sundal C, Van Gerpen J, Fujioka S, Aasly J, Wider C, Roeber S, Shuster E, Ghetti B, Garbern J, Tselis A, Swerdlow R, Rademakers R, Dickson D, Broderick D, Wszolek Z. (2012) HDLS: Due to CSF1R gene mutation; Clinical characteristics. Neurology. Sundal C, Fujioka S, Van Gerpen J, Wider C, Aasly J, Roeber S, Shuster E, Ghetti B, Garbern J, Tselis A, Swerdlow R, Miller BB, Rademakers R, Dickson D, Broderick D, Wszolek Z. (2012) Observational study for MRI characteristics in HDLS with a known gene mutation on chromosome 5. Neurology. Newell K, Swerdlow R, Ghetti B. (2012) Niemann-Pick Disease Type C Associated with 2 mutations in the NPC1 Gene. 10th European Congress of Neuropathology 261.00. Harris J, Yeh HW, Choi IY, Lee P, Berman N, Swerdlow R, Craciunas S, Brooks W. (2012) The neurochemical profile of traumatic brain injury assessed with 1H-MRS at 9.4T, Neurotrauma Meeting Proceedings. Sundal S, Van Gerpen J, Nicholson A, Baker M, Wider C, Shuster E, Aasly J, Spina S, Ghetti B, Roeber S, Tselis A, Swerdlow R, Miller B, Fujioka S, Uitti R, Ross O, Rademakers R, Josephs K, Dickson D, Wszolek Z. (2012) Parkinsonism in Hereditary diffuse leukoencephalopathy with axonal spheroids due to CSF1R gene mutation. Movement Disorder Society's (MDS) 16th International Congress of Parkinson's Disease and Movement Disorders. Lu J, E L, Swerdlow R. (2012) Relationship between cholinergic signaing and cell bioenergetics. Alzheimer’s and Dementia 8:P579.

124

Honea R, Vidoni E, Swerdlow R, Burns J. (2012) Association of maternal family history of dementia with Alzheimer’s disease biomarkers. Alzheimer’s and Dementia 8:P677. Artigues A, Tan E, Villar MT, Lu J, Selfridge E, E L, Swerdlow RH, Slawson C. (2012) OGlcNAc signaling regulates mitochondrial function. Implications for Alzheimer’s disease. ASBMB-Post Translational Modifications Symposium. Swerdlow, RH. (2012) Brain bioenergetics and aging: Therapeutic strategies for intervention. 13th International Conference on Alzheimer’s Drug Discovery. Meeting Proceedings 31. Wang Y, Moore D, Katz J, Saperstein D, Walk D, Simpson, E, Genge A, Bertorini T, Fernandes J, Swenson A, Elman L, Swerdlow R, Dimachkie MM, McVey A, Herbelin L, Macchi Z, Barohn RJ. (2012) A multicenter screening trial of the safety and efficacy of rasagiline in people with ALS. Motor Neurone Disease Association 23rd International Symposium on ALS/MND. Rumi MA, Kubota K, Ratri A, Chakraborty D, Bugarinovic G, Roby KF, Larson MA, Vivian JL, Wolfe MW and Soares MJ. 2012. Zinc finger nuclease targeted disruption of estrogen receptor alpha signaling in the rat. 9th Annual Gilbert S. Greenwald Symposium on Reproduction, Kansas City, KS. Dubey S, VanVeldhuizen P, Holzbeierlein J, Tawfik O, Thrasher JB, Karan D. (2012) Inflammation-Associated Regulation of the Macrophage Inhibitory Cytokine (MIC-1) Gene in Prostate Cancer. Oncology Letters 3(5): 1166-1170.

Yang J, Xie S, Huang Y, Ling M, Liu J, Ran Y, Wang Y, Thrasher JB, Berklund C, Li B. (2012) Prostate-targeted biodegradable nanoparticles loaded with androgen receptor silencing constructs eradicate xenograft tumors in mice. Nanomedicine, Epub.

Burgess-Galvin KE, Travis ED, and Vivian JL. (2012) TGF-beta-related signaling regulates stem cell heterogeneity. Gordon Research Conference: Reprogramming, Galveston TX.

Wang J, Gardner B, Lu Q, Shaath M, Brandt K, Anderson H. (2012) Deficiency of NFAT1 transcription factor causes osteoarthritis with alterations in articular cartilage and subchondral bone in adult mice. Osteoarthritis and Cartilage. 20(Suppl 1):S60.

Lu Q, Furomoto B, Anderson HC, Wang J. (2012) Bone and muscle interations during the progression of Nfat1 deficiency-mediated osteoarthritis. J Bone Miner Res. 27(Suppl 1):S168.

Wang J, Lu Q, Shaath MK, Bernard J. (2012) Bone sialoprotein is essential for osteoblastic cell differentiation and maturation. J Bone Miner Res. 27(Suppl 1):S279.

Dalal J, Yankee TM, Hassaballa A, Ryan R, Chignola B, Sherman A, McGuirk J, Abhyankar S. (2012) An immunological assessment of B and T cells in patients with chronic graft vs host disease undergoing extracorporal photopheresis (ECP). American Society for Blood and Marrow Transplantation, San Diego, CA. Yankee TM. (2012) TCR signaling pathways regulating CD8+ T cell survival during the expansion phase. 2nd International Conference on Vaccines and Vaccination, Northbrook, IL. Mitchell JL, Szarejko J and Yankee TM. (2012) Changes in Aiolos and Helios expression in T cell development. Autumn Immunology Conference, Chicago, IL. Li J, Nakamura T, Zhuo M, Pei L, Yu ASL. (2012) Mapping the claudin-2 pore: comprehensive cysteine-scanning mutagenesis and thiol modification of claudin-2 first extracellular loop. Poster presentation. Experimental Biology Annual Meeting, CA.

125

McCusker EC, Vecchio AJ, Metzger LE, Yu ASL, Stroud RM. (2012) Expression, purification and characterization of tight junction claudins. Presentation, 4th NIH Roadmap meeting to Membrane Protein Structures and Complexes, San Francisco CA. RESEARCH SUPPORT D. Albertini: ESHE Fund - “Imaging Stem Cells,” Nov 30, 2011-Dec 1, 2012. Principal Investigator: D. Albertini. Total Costs: $16,000 NIH, Oregon Regional Primate Research Center, U54 Center Grant subproject (“Preventing ovarian DNA damage in a primate model for ovarian cancer”) , Principal Investigator M. Zelinski, D Albertini Co-PI; $65,000 MRC UK - “A human model for ovarian fertility preservation in cancer patients,” June 2012 – May 2014. Principal Investigator: E. Telfer. Direct Costs: $220,000. Biogenesi, Inc. Milan, Italy - “Optimizing Oocyte IVM in a Bovine Model,” September 2012 – October 2013, Principal Investigator: D Albertini. Total Costs: $20,000. O. Aljitawi: CORE SEED Grant – “Ex vivo expansion of umbilical cord blood using a unique bio-system,” 2009-2013. Principal Investigator: O Aljitawi. U. Apte: AASLD/ALF Liver Scholar Award – “Role of Wnt/beta-catenin signaling in liver regeneration after acetaminophen-induced acute liver failure,” July 2010 – June 2013. Principal Investigator: U. Apte. Total Costs: $225,000. F. Behbod: Department of Defense – “The use of Raman Tweezer Spectroscopy for the identification of mouse mammary tumor initiating cells,” 2011 –2013. Principal Investigator: F. Behbod. Total Costs: $99,000. NIH/NCI – “Role of Cancer Stem Cells in Malignant Progression of Human DCIS,” 2007 –2013. Principal Investigator: F. Behbod. Total Costs: $1,400,000. NIH/NCI – “Stem cells and their aberrant self-renewal pathways in DCIS malignant progression,” 2012 – 2015. Principal Investigator: K. Valdez. Total Costs: $378,000. DOD – “Role of BRCA1 in regulating TGFB1-Induced Epithelial-to-Mesenchymal Transition,” 2010 – 2012. Principal Investigator: S. Stecklein. Total Costs: $68,771.

J.P. Blumenstiel:

NSF – “The Epigenetics of RNA based silencing transposen control in Drosophila virilis,” 2010 – 2013.

Principal Investigator: J.P. Blumenstiel. Total Costs: $678,711.

126

K. Bosak:

University of Kansas Research Institute, - “Neuroimaging of Goal Directed Health Behavior in Overweight

Women,” May 2011- May 2013. Total Costs: $30,000.

NIH K12 (BIRCWH) Award – “Feasibility of an Intervention Delivering Automated Real-Time Feedback for

Physical,” December 1, 2012-present. $100,000/year.

M.G. Butler: NIH – “Rare Disease Clinical Research Center for New Therapies and New Diagnostics,” 2003 – 2014. Principal Investigator: M.G. Butler. Total Costs: $300,000. NIH – “Early Prevention of Aberrant Behavior In Neurodevelopmental Disorders in Peru,” 2010 – 2012. Principal Investigator: Total Costs: $40,834. Prader-Willi Syndrome Association (USA) (BIG Research Grant) – “Probing Genes for Hyperphagia in Rare Obesity-related Syndromes,” 2010-2012. Principal Investigator: M.G. Butler. Total Costs: $100,000. Prader-Willi Syndrome Association (USA) – “RDCRN Fellowship Support to Study Rare Diseases Including Prader-Willi Syndrome,” 2012. Principal Investigator: M.G. Butler. Total Costs: $25,000. Prader-Willi Syndrome Association (USA) – “A Pilot Study of Transcranial Direct Current Stimulation and Startle Modulation to Evaluate Hyperphagia in Prader-Willi Syndrome,” 2011-2013. Principal Investigator: M.G. Butler. Total Costs: $50,000. Prader-Willi Syndrome - KU Endowment funds generated by families for research on Prader-Willi syndrome, 2012. Investigator: M.G. Butler. Total Costs: $20,000.

Affymetrix, Inc., Santa Clara, CA, received funding by supplying DNA samples for development of new medical device or microarray, 2011-2012. Investigator: M.G. Butler. Total Costs: $28,062.

KUMC Clinical Pilot Research Grant Program (FY 2012) - “An Approach to Target X-Chromosome Genes in Females with Autism,” 2012. Investigator: M.G. Butler. Total Costs: $25,000.

NIH (R21) - “Fragile X Pre-mutation and Ovarian Insufficiency,” 2012. Co-Investigator: M.G. Butler. Total Costs: $25,800). National Health and Medical Research Council (NHMRC) of Australia - “The Function of Prader-Willi Syndrome Associated snoRNAs,” January 1, 2013 - December 31, 2017. Chief Investigator D: M.G. Butler. Total Costs: $5,000. Collaborative Research and Innovation Opportunities (CRIO) Canadian Program - “The Genetic Basis of Childhood Obesity,” 2012. Co-Investigator: M.G. Butler. Total Costs: $5,000.

S. Carlson: NIH/NICHD – “DHA and Pregnancy Outcome,” February 2012 – January 2017. Principal Investigator: S. Carlson. Total Costs: $1,612,037. N. Cheng: NIH – “Functions of TGF-beta and chemokine signaling in the breast cancer microenvironment.” September 1, 2010 – August 31, 2013. Principal Investigator: Nikki Cheng. Total Costs: $498,000.

127

J. Christianson: NIH – “Impact of early experience on vulvovaginal sensitivity in adult mouse,” July 2010 – June 2013. Principal Investigator: D. Abrahamson. Total Costs: 667,500. NIH - “Evaluation of sensory neuron plasticity following neonatal maternal separation,” April 1, 2010 – March 31, 2013. Principal Investigator: J. Carlsten Christianson. Total Costs: $148,500. J. Colombo:

NIH – “Training researchers in language impairments,” 2007 – 2012. Principal Investigator: M.L. Rice. Total Costs: $1,137,645. NIH – “Postdoctoral training in translational research on intellectual and developmental disabilities,” 2007 – 2012. Principal Investigator: K. Saunders. Total Costs: $1,059,652. NIH – “DHA supplementation and pregnancy outcome,” 2012 – 2017. Principal Investigator: J. Colombo and S.E. Carlson. Total Costs: $2,858,697. NIH – “Kansas Intellectual and Developmental Disabilities Research Center,” 2012 – 2017. Principal Investigator: J. Colombo. Total Costs: $8,109,647. Mead Johnson Nutrition – “Effect of Low Glycemic-Index Beverages on Toddler Cognition,” 2011 – 2012. Principal Investigator: J. Colombo. Total Costs: $193,575. M. Detamore: NIH/NIDCR – “Integrative colloidal gels for cranial defect repair,” 2012 – 2016. Principal Investigator: M. Detamore. Total Costs: $1,455,431. Coulter Foundation – “Microsphere-based gradient plugs for osteochondral regeneration,” 2011 – 2013. Principal Investigator: M. Detamore. Total Costs: $218,000. NIH/NIAMS – “Gradient-based strategy for osteochondral regeneration,” 2010 – 2015. Principal Investigator: M. Detamore. Total Costs: $1,347,542. NSF (DMR) – “CAREER - Gradients and umbilical cord stem cells in osteochondral tissue engineering,” 2009 – 2014. Principal Investigator: M. Detamore. Total Costs: $400,000. State of Kansas – “Umbilical cord matrix project,” 2008 – 2013. Principal Investigator: M. Detamore. Total Costs: $590,000. A. Dhar: NIH – “Pancreatic cancer: Crocetin as a novel therapeutic approach,” September 2011 – August 2014. Principal Investigator: A. Dhar. Total Costs: $800,301. Y. Dong: NIHCHD - “Chronic hypoxia mediated fetal brain injury is associated with a Rho/Rho-kinase,” 2010 - 2012. Principal Investigator: Y. Dong. Total Costs: $75,000.

128

P. Fields: NIH/NIDDK – “Role of the Histone Methyltransferase DOT1L in Embryonic Erythropoiesis,” April 2012 – March 2017. Principal Investigator: P. Fields. Total Costs: $1,087,500. NIH/NICHD – “Dissecting Uterine Progesterone Resistance,” July 2012 – June 2014. Principal Investigator: M. Soares. Total Costs: $188,750. NIH/NIDDK – “Training in polycystic kidney disease (PKD),” July 2012 – June 2014. Principal Investigator: J. Rossol-Alison. Total Costs: $105,812. T. Fields: Komen Investigator Award – “The Role of Heterotrimeric G proteins in Breast Cancer Metastasis,” June 2011 – May 2013. Principal Investigator: T. Fields. VA Merit Award – “Cytokine based murine focal glomerulosclerosis model: a prelude to novel therapy,” April 2011 – March 2015. Principal Investigator: V. Savin. Kansas City Area Life Sciences Institute-Patton Trust Research Award – “Renal Macrophages in ARPKD: Recruitment and Disease-Promoting Effects,” August 2012 – July 2013. Principal Investigator: T. Fields. Total Costs: $40,000. P. Geiger: NIA/NIAMS – “Targeting stress-mediated pathways in the treatment of muscle insulin resistance,” July 2010 – June 2015. Principal Investigator: P. Geiger. NIH/NINDS - “Painful Versus Insensate Diabetic Neuropathy,” January 2003 – December 2013. Principal Investigator: D. Wright. K. Gustafson: NICHD – “DNA Supplementation During Pregnancy: Cognitive Outcomes in Children ages 2-6 years,” December 2011 – December 2016. Principal Investigator: S. Carlson. Total Costs: $14,665. Mead Johnson Nutritionals – “State of the Art ERP Analysis of EEG Data from the DIAMOND Study,” September 2012 – August 2013. Principal Investigator: K.M. Gustafson. T. Iwakuma: ACS – “Uncovering the mechanisms of osteosarcoma metastasis suppression by MTBP,” July 2009 – June

2013. Principal Investigator: T. Iwakuma. Total Costs: $870,000.

R. Johnson: AHA – “Derivation of Novel Chemically Modified Multipotent Stem Cells from Endothelial Progenitor Cells in its Therapeutic Use,” January 2010 – December 2013. Principal Investigator: R. Johnson. Total Costs: $280,000. NIH – “Epigenetic modification of endothelial progenitor cells into multipotent cells,” September 2009 – June 2013. Principal Investigator: R. Johnson. Total Costs: $431,750. NIH – “Organ protection during cardiopulmonary arrest,” April 2010 – March 2013. Co-Principal Investigator: R. Johnson. Total Costs: 375,000.

129

NIH supported CTSA grant – “ Derivation of functional cardiomyocytes from human keratinocytes by using epigenetic modifiers,” March 2012 – February 2013. Principal Investigator: R. Johnson. Total Costs: 30,000. Dev Karan: NIH/NCI – “Modulation of NK cell activity by dietary product for prostate cancer prevention,” July 2012 – June 2014. Principal Investigator: D. Karan. Total Costs: $239,250. KUMC Research Institute, Clinical Pilot Program – “Assessment of immunotherapeutic response using novel bio-molecular approach,” June 2012 – May 2013. Principal Investigator: D. Karan. Total Costs: $25,000. S. Finocchario Kessler: NIH/NICHD – “Determinants of use of safer conception strategies among HIV clients in Uganda,” April 1, 2012 – March 31, 2016. Principal Investigator: S. Kessler. Total Costs: $1,607,810. S.L. Kieweg: NIH (NIAID) – “Biomechanical and Computational Molecular Design of Microbicide Delivery Systems,” September 2009 – August 2014. Principal Investigator: S.L. Kieweg. Total Costs: $1,717,881. Kauffman Foundation/The University of Kansas, Institute for Advancing Medical Innovation – “Automated Vitrification Device (2nd-Generation) and Closed Storage System,” November 2011 – May 2013. Principal Investigator: S.L. Kieweg. Total Costs: $143,207. NIH/NIAID – “Prevention of HIV/AIDS by Stimuli-Sensitive Nanomedicine for Microbicide Delivery,” August 2011 – July 2015. Subcontract Principal Investigator: S.L. Kieweg. PI: B.B. Youan.Total Costs: $251,960. NIH/NIDCR – “Integrative Colloidal Gels for Cranial Defect Repair,” March 2012 – February 2016. Principal Investigator: C. Berkland. Total Costs: $1,444,985. KU Center of Research, General Research Fund – “Assessment of Hand Posture and Pressures in Obstetricians Performing Mock Deliveries on a Mannequin,” July 2012 – June 2013. Principal Investigator: S.L. Kieweg. Total Costs: $8,651. NIH/NIDCR – “Proton Sponge Adhesives, Interfacial Milieu: Molecular Structure-Mechanics,” July 2011 – June 2016. Principal Investigators: P. Spencer and J. Laurence. Total Costs: $1,813,385. NSF (CNS) – “MRI: Acquisition of an Advanced Computational Infrastructure for Modeling Biological Systems,” August 2008 – July 2012. Principal Investigator: J. Evans. Total Costs: $300,000. S.S. Kim: Komen Foundation – “Flaxseed Lignan as a Prevention Strategy for Pre-Menopausal Women at High Risk for Development of Breast Cancer,” 2011 – 2012. Principal Investigator: C. Fabian. NIH - “The role of mi-R451 in endometriosis pathophysiology and treatment,” 2012. Principal Investigator: W.B. Nothnick. A.J. Krieg: NIH, KANSAS U COBRE: “Molecular Regulation of Cell Development and Differentiation,” July 2010-June 2012. Principal Investigator: D. Abrahamson. Total Costs: $294,000.

130

NIH – “Trophoblast Differentiation,” 2012 – 2017. Principal Investigator: M. J. Soares. Total Costs: $1,961,159. S.A. Krieg: NIH – “The role of miR-451 in endometriosis pathophysiology and treatment,” 2012- 2017. Principal Investigator: W.B. Nothnick. Total Costs: $1,218,244. M. Larson: COBRE – “Molecular Regulation of Cell Development and Differentiation,” Principal Investigator: D. Abrahamson. K-INBRE – “KUMC Transgenic Facility Procurement and Optimization of a PhiC31-Mediated Platform for Rapid In Vivo Trangenesis in the Mouse,” May 2012 – April 2013. Direct Costs: $36,921. J. Lewis-Wambi: DOD – “Loss of PEDF: A novel mechanism of endocrine resistance in breast cancer,” August 2012 – July 2014. Principal Investigator: J. Lewis-Wambi. Total Costs: $892,000. X. Li: Children’s Research Institute – “Molecular mechanisms of ADPKD,” August 2008 – July 2012. Principal Investigator: X. Li. Direct Costs: $650,000. NIH – “TNF-alpha signaling regulates cystic epithelial cell apoptosis and proliferation,” August 2009 – July 2012. Principal Investigator: X. Li. Direct Costs: $100,000. NIH – “ADPKD: Understanding the mechanisms and discovering the treatments,” August 2010 – July 2015. Principal Investigator: X. Li. Direct Costs: $1,100,000. C.W. Mason: NICHD – “Impact of Pregnancy Disease on the Regulation of Placenta Drug Transport,” 2012-2013. Principal Investigator: CW Mason. Total Costs: $90,000. N.A. Muma: NIH – “Mechanisms of 5-HT2A Receptor Desensitization,” July 2003 – June 2013. Principal Investigator: N.A. Muma. Total Costs: $1,640,000. NIH/NIMH – “Synergy Between SSRIs and Ovarian Hormones,” August 2005 – July 2012. Principal Investigator: N.A. Muma. Total Costs: $1,000,000. A.J. Nangia: CTSA – “Glutathione S-Transferase Polymorphisms and Mechanisms of Male-Infertility,” 2011 – 2012. Principal Investigator: R. Mijal. Direct Costs: $30,000. NICHD – “Male Contraceptive Clinical Trials Network,” 2012 – 2017. Principal Investigator: A.J. Nangia. Direct Costs: $1,916,000.

131

W.B. Nothnick: NIH – “The role of miR-451 in endometriosis pathophysiology and treatment,” March 2012 – January 2017. Principal Investigator: W.B. Nothnick. Direct Costs: 1,218,244. S. Paul: NIH – “GATA factor function in Trophoblast,” August 2010 – June 2015. Principal Investigator: S. Paul. Total Costs: $1,527,144. NIH – “Histone Chaperones in Angiogenesis,” February 2011 – January 2014. Principal Investigator: S. Paul. Total Costs: $412,500. NIH – “Protein Kinase C Signaling and Pluripotent Stem Cell,” August 2011 – July 2013. Principal Investigator: S. Paul. Total Costs: $412,500. Kansas University Endowment Association – “Spinal Cord Injury Research Program,” April 2011 – April 2016. Principal Investigator: P. Smith. Total Costs: 1,500,000. K. R. Peterson: NIH – “Transactivation of Fetal Hemoglobin,” September 1, 2008 – August 31, 2012. Principal Investigator: K. Peterson. Total Costs: $1,265,470. NIH - “Molecular Regulation of Cell Development and Differentiation,” September 27, 2007 – June 30, 2012. Principal Investigator: D. Abrahamson. Total Costs: $160,095. NIH - “Gamma Globin Induction: Molecular and Cellular Based Strategies,” September 1, 2008 – May 31, 2012. Principal Investigator: BS Pace. Total Costs: $330,750. NIH - “Nuclear Receptors in Liver Health and Disease,” July 2011 – June 2016. Principal Investigator: L. DiTacchio. Total Costs: $47,440. NIH – “HTS for HbF Inducers in Human Beta-globin YAC Transgenic Mice Bone Marrow Cells,” December 2012 – November 2013. Principal Investigator: K. Peterson. Brian K. Petroff: Komen Promise Program Grant – “Cancer Prevention with Flaxseed Lignan,” June 1 2010 – May 30, 2015. Co-Principal Investigators: B. Petroff and C.J. Fabian. Kansas City Area Life Sciences Institute – “Testing a Novel Benefit to Children from Altered Maternal Estrogen Signaling,” September 1, 2012 – August 31, 2013. Principal Investigator: B. Petroff. University of Kansas Cancer Center – Center grant establishing UKCC as a NCI-designated matrix cancer center and provides support for administrative and core facility for cancer research at KUMC. December 2012 – November 2017. Principal Investigator: R. Jensen. Lied Biomedical Sciences Grant – “Placental immunology and cancer risk,” June 2012 – May 2013. Co-Principal Investigators: B. Petroff and M. Petroff. Breast Cancer Research Foundation – “Biomarkers of Response to Prevention Strategies for Postmenopausal Women,” October 1, 2008 – September 30, 2013. Total Costs: $400,000. Co-Principal Investigators: C.J. Fabian and B. Petroff.

132

NIH/NCI – “Vitamin D for prevention of breast cancer in premenopausal women,” April 2009 – March 2014. Co- Principal Investigators: C.J. Fabian and B. Petroff. F. Pierucci-Alves: Johnson Center for Basic Cancer Research – “TGF-beta signaling in epithelia of the male excurrent system,” 2012 – 2013. Investigator: F. Pierucci-Alves.Total Costs: $20,000. K. F. Roby: KU Institute for Advancing Medical Innovation – “KU-AS-272 a sterilant in female and male animals: dose range finding, safety/efficacy in prepubertal, adult, and pregnant animals, and pharmacokinetic studies,” 2011-2013. Principal Investigator: KF Roby. Total Costs: $468,033. Mesothelioma Applied Research Foundation – “HSP90 targeted therapy for mesothelioma,” 2009-2012. Principal Investigator: KF Roby. Total Costs: $99,908. KUMC, CTSA – “Glutathione S-Tranferase Polymorphisms and Mechanisms of Male-Infertility,” 2011-2012. Principal Investigator: R.S. Mijal. Total Costs: 30,000. M.A. Rumi: NIH – “Role of SATB in placental development,” 2012 – 2014. Principal Investigator: M.A. Rumi. Total Costs: $150,000. NIH – “Dissecting Uterine Progesterone Resistance,” 2012 – 2014. Principal Investigator: M.J. Soares. Total Costs: $412,500. NIH – “Trophoblast Differentiation,” 2012 – 2017. Principal Investigator: M. J. Soares. Total Costs: $1,961,159. I. Saadi: NIH/NCRR – “Molecular Regulation of Cell Development and Differentiation,” September 2012 – June 2014. Principal Investigator: D. Abrahamson. Total Costs: $147,000. K-INBRE/CTSA Frontiers Pilot and Collaborative Studies Award – “High throughput discovery of craniofacial birth defect genes by exome sequencing. May 2012 – April 2013. Principal Investigator: M. Dasouki. Total Costs: $30,000. B. Schultz: NIH – “Neuroendocrine-modulated epithelial HCO3

- transport,” 2008 – 2013. Principal Investigator: B. Schultz. Direct Costs: $1,125,000. NIH – “Model synthetic channel assemblies,” 2010 – 2014. Principal Investigator: J.M. Tomich. Direct Costs: $850,000. C. Slawson: NIH – “Molecular Regulation of Cell Development and Differentiation,” 2011-2013. Principal Investigator: D. Abrahamson. Total Costs: $147,000. NIH/NIA – “Alzheimer’s Mitochondria Defects Alter O-GlcNAc Signaling,” July 1, 2011 to June 30 2012. Principal Investigator: R. Swerdlow. Total Costs:$30,000.

133

M. J. Soares NIH – “Decidual Cell Adaptations to Physiological Stressors,” April 2007 – March 2012. Principal Investigator: M.J. Soares. Total Costs: $1,556,250. NIH – “Trophoblast Differentiation,” May 2007 – July 2012. Principal Investigator: M.J. Soares. Total Costs: $1,556,250. NIH – “Genetics of Decidualization,” February 2010 – January 2012. Principal Investigator: M.J. Soares. Total Costs: $412,500. NIH – “Dissecting Uterine Progesterone Resistance,” April 2012 – March 2014. Principal Investigator: M.J. Soares. Total Costs: $412,500. NIH – “Trophoblast Differentiation,” July 2012 – April 2017. Principal Investigator: M.J. Soares. Total Costs: $1,961,159. K.I. Swenson-Fields: P&F Kidney Institute (Pilot Grant Award) – “Investigations into the abnormal, pro-proliferative expression of WNT5A in ADPKD cyst cells,” April 2012 – March 2013. Principal Investigator: K.I. Swenson-Fields. Total Costs: $35,000. Kansas City Area Life Sciences Institute-Patton Trust Research Award – “Renal Macrophages in ARPKD: Recruitment and Disease-Promoting Effects,” August 2012 – July 2013. Principal Investigator: T. Fields. Total Costs: $40,000. R. Swerdlow: NIH/NIA – “Kansas University Alzheimer’s Disease Core Center,” 2011-2016. Principal Investigator: R. Swerdlow. Total Costs: $6,364,433. PhotoThera, Inc. – “Near Infrared Laser Treatment’s Effect on Bioenergetic Fluxes and Mitochondrial Biogenesis,” 2012-2013. Principal Investigator: R. Swerdlow. Total Costs: $103,460. NIH/NINDS – “Oxaloacetate’s Brain Effects,” 2012-2014. Principal Investigator: R. Swerdlow. Total Costs: $151,000. NIH – “Aerobic Fitness in Slowing the Progression of Alzheimer’s Disease,” 2010-2013. Principal Investigator: J. Burns. Total Costs: $28,464. J.B. Thrasher: Astra Zeneca – “A Phase III, Randomized, Placebo-controlled, Double-blinded Study to Assess the Efficacy and Safety of Once-daily Orally Administered ZSD4054 10 mg in Non-metastatic Hormone Resistant Prostate Cancer Patients,” September 2007 - ongoing, Principal Investigator: J.B. Thrasher. Total Costs: $75,014. Amgen – “A Randomized, Double-Blind, Placebo-controlled Study to Evaluate AMG 162 in the Treatment of Bone Loss in Subjects Undergoing Androgen-Deprivation Therapy for Non-Metastatic Prostate Cancer,” May 2004-ongoing. Principal Investigator: J.B. Thrasher. Total Costs: $97,007. TAP Holdings - “CaPSURE: Cancer of the Prostate Strategic Urologic Research Endeavor,” August 2000-ongoing. Principal Investigator: J.B. Thrasher. Total Costs: $89,050.

134

UCOP/SWOG – “MVAC in Organ Confined Bladder Cancer/P53,” January 2001- ongoing. Investigator: Thrasher JB

J.L. Vivian: NIH – “MicroRNA regulation of Ovarian Function,” June 1, 2010 – May 31, 2015. Principal Investigator: L. Christiansen. Total Costs: $3,752. Spinal Cord Injury Project – “Spinal Cord Injury Project I: Human iPS cell differentiation models,” April 15, 2011 – April 15, 2015. Principal Investigator: P. Smith. Total Costs: $95,900. NIH/NIDDK – “Role of the Histone Methyltransferase DOT1L in Embryonic Erythropoiesis,” April 2012 – March 2017. Principal Investigator: P. Fields. Total Costs: $1,087,500. J. Wang: NIH/NIDCR – “Bone Sialoprotein in Osteogenesis and Bone Regeneration,” 2010 – 2015. Principal Investigator: J. Wang. Total Costs: $1,483,520. NIH/NIAMS - “Transcription Factor NFAT1 Deficiency and Osteoarthritis,” 2011 – 2016. Principal Investigator: J. Wang. Total Costs: $1,687,500. NIH/NIDCR – “Integrative Colloidal Gels for Cranial Defect Repair,” 2012 – 2016. Principal Investigator (Subcontract): J. Wang. Direct Costs: $1,035,000. DOD – “Deficiency of Transcription Factor NFAT1 and Posttraumatic Osteoarthritis,” 2012 – 2015. Principal Investigator: J. Wang. Total Costs: 1,125,000. C.P. Weiner: NIH – “Biomechanical & Computational Molecular Design of Microbicide Delivery Systems (R33),” 2009 – 2014. Principal Investigator: S. Kieweg. Total Costs: $76,006. NICHD – “KUMC Women’s Reproductive Health Research Career Development Program (K12),” 2010 – 2015. Principal Investigator: CP Weiner. Total Costs: $2,375,980. Vietnam Education Foundation – “Reducing Maternal Mortality in Vietnam,” 2011 – 2012. Principal Investigator: CP Weiner. Total Costs: $42,950. M.L. Weiss: Kansas Biosciences Authority - “Pancreatic cancer,” December 31, 2009 – December 31, 2012. Subcontract Principal Investigator: M. Weiss. Total Costs: $119,071. T. Yankee: American Cancer Society – “Survival, proliferation, and differentiation in early T cell development,” July 1, 2008 – June 30, 2012. Principal Investigator: T. Yankee. Total Costs: $720,000. NIH – “Novel Approaches for the Control of Microbial Pathogens,” September 1, 2012 – July 31, 2017. Principal Investigator: J. Lutkenhaus. Total Costs: $5,329,473. University of Kansas Cancer Center – “Alternative splicing of the Aiolos transcription factor promotes leukemogenesis,” December 1, 2011 – May 30, 2013. Principal Investigator: T. Yankee. Total Costs: $35,000.

135

KUMC Research Institute Lied Endowed Program – “The Gads adaptor protein regulates CD8+ T cell activation and proliferation,” April 1, 2010 – March 31, 2011. Principal Investigator: T. Yankee. Total Costs: $35,000. A.S.L. Yu: NIH – “Expression, function and regulation of renal claudins,” 2007-2012. Principal Investigator: A.S.L. Yu. Total Costs: $1,339,149. NIH - “Structure-function studies of tight junction membrane proteins,” 2010 – 2015. Principal Investigator: A.S.L. Yu. Total Costs: $2,299,338. H. Zhu: NIH – “Role of Ncb5or in insulin production,” January 2009 – June 2012. Principal Investigator: H. Zhu. KUMC School of Health Professions Research Fund Award – “Ncb5or-dependent iron hematostasis in beta-cell function and survival,” July 2012 – June 2013. Principal Investigator: H. Zhu.

The Institute for Reproductive Health and Regenerative Medicine

www.kumc.edu/irhrm