National Academies Press: OpenBook

Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop (2018)

Chapter: Appendix C: Biographical Sketches of Workshop Speakers and Moderators

« Previous: Appendix B: Biographical Sketches of Planning Committee
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

Appendix C

Biographical Sketches of Workshop Speakers and Moderators

Tracy L. Bale is a professor of neuroscience in the School of Veterinary Medicine and in the Department of Psychiatry of the Perelman School of Medicine. She obtained her PhD from the University of Washington in the Department of Pharmacology and the Program in Neurobiology. She completed her postdoctoral training with Dr. Wylie Vale and the Salk Institute in La Jolla, California. Her research focuses on understanding the role of stress dysregulation in neurodevelopmental and neuropsychiatric diseases, and the sex differences that underlie disease vulnerability using mice as the model organism. Mechanistic examination includes studies on the contributions of the placenta, germ cells, and microbiome in epigenetic programming of the brain. Dr. Bale is the co-director of the Penn Center for the Study of Sex and Gender in Behavioral Health, which is funded by a National Institute of Mental Health (NIMH) and Office of Research on Women’s Health (ORWH) Specialized Centers of Interdisciplinary Research (SCOR) P50 grant, and is the director of research for the Building Interdisciplinary Research Careers in Women’s Health (BIRCWH) Faculty Scholars. She serves on many internal and external advisory committees, panels, and boards and is currently a reviewing editor at the Journal of Neuroscience and serves as chair of the Neuroendocrinology, Neuroimmunology, Rhythms and Sleep, Center Scientific Review (NNRS CSR) study section. She has been the recipient of several awards for her research in this area, including the career development award for early career achievement and promise by the Society for Neuroscience, the Richard E. Weitzman Memorial award as exceptionally promising investigator award by the Endocrine Society, the Medtronic Award from the Society for Women’s Health Research for outstanding research that has led to the improvement of women’s health, and most recently, the Daniel H. Efron Research Award from the American College of Neuropsychopharmacology.

Richard S. Blumberg trained in internal medicine (The New York Hospital, 1982), infectious diseases (Massachusetts General Hospital, 1986), and gastroenterology and hepatology (Brigham and Women’s Hospital, 1989). He is currently Senior Physician in Medicine and Gastroenterology at Brigham and Women’s Hospital where he holds the position of division chief of gastroenter-

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

ology, hepatology and endoscopy, is a professor of medicine at Harvard Medical School, co-director of the Harvard Digestive Diseases Center, and immediate past-director of the Brigham Research Institute. In addition, Dr. Blumberg serves on the Executive Advisory Committee of the Department of Medicine. He also currently serves on the Gastrointestinal Pathobiology Study Section at the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and has served as a member of the Immunology Sciences Study Section of the National Institute of Allergy and Infectious Diseases (NIAID), a member on the National Commission of Digestive Diseases of the NIDDK, scientific consultant to the Human Microbiome Project of the National Human Genome Research Institute (NHGRI), a member of the Vaccine Branch External Advisory Board of the National Cancer Institute (NCI), and a member of the Board of Scientific Councilors. Dr. Blumberg has served as the chair of the National Scientific Advisory Committee of the Crohn’s & Colitis of America (2002-2005) and was former president of the Society for Mucosal Immunology (2007-2009). Dr. Blumberg is an elected member of the American Association of Clinical Investigation and the Association of American Physicians and the recipient of a Merit award from the National Institutes of Health (NIH) (2005), the William Beaumont Prize from the American Gastroenterological Association (2012), the Distinguished Scientific Achievement Award from the Crohn’s and Colitis Foundation of America (2012), and Lifetime Achievement Award from the Society of Mucosal Immunology (2015). He has been an NIH-funded investigator since 1989 whose long-standing research programs focus on mucosal immunology and specifically directed the role of CD1d-NKT cells, the unfolded protein response, CEACAM1, and FcRn in immunobiology. Dr. Blumberg was the scientific founder of Syntonix Pharmaceuticals that developed long-acting therapeutic agents, which were recently approved by the U.S. Food and Drug Administration and European Medicines Agency for the treatment of hemophilia, and is the scientific founder of Syntimmune, Inc., which is developing inhibitors of FcRn that are currently being tested in phase 1 studies.

Robert Britton is a professor in the Department of Molecular Virology and Microbiology and is a member of the Alkek Center for Metagenomics and Microbiome Research at Baylor College of Medicine. He currently directs the Therapeutic Microbiology laboratory, which is focused on the use of microbes to prevent and treat human disease. Currently funded research projects in the laboratory range from the study of how traditional probiotic strains can ameliorate osteoporosis to how intestinal microbial communities resist invasion by the diarrheal pathogen Clostridium difficile. His laboratory has made several advances in the development of genetic and microbial growth platforms to aid in the understanding of how microbes promote health and disease. These include the development of precision genome engineering technologies for lactic acid bacteria and the development of human fecal minibioreactor arrays to study the function of microbial communities in a high-throughput manner. Dr. Britton received a BS in Biology from the University of Nebraska–Lincoln and a PhD in

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

cell and molecular biology from Baylor College of Medicine. After performing postdoctoral training at the Massachusetts Institute of Technology he started his own laboratory at Michigan State University. After rising to the rank of professor in 2014 he moved to his current position at the Baylor College of Medicine.

Patrice D. Cani is a professor and senior researcher from the Belgian Fund for Scientific Research, and a group leader at the Louvain Drug Research Institute from University College London (UCL), Brussels, Belgium. He is a dietitian, earned an MSc in nutrition, an MSc in health sciences, and a PhD in biomedical sciences (2005). His main research interests are the investigation of interactions between the gut microbiota, the host, and specific biological systems, such as the endocannabinoid system and the innate immune system in the context of obesity, type 2 diabetes, cardiometabolic disorders, and metabolic inflammation. In 2007, he published the discovery of the concept of metabolic endotoxemia. More recently, he discovered the beneficial role of the bacteria Akkermansia muciniphila on obesity and cardiometabolic risk factors. Dr. Cani is a Walloon Excellence in Lifesciences and Biotechnology (WELBIO) investigator and the recipient of prestigious grants: European Research Council (ERC) Starting Grant 2013 (ENIGMO), a PoC ERC grant 2016, the prize “Baillet Latour Grant for Medical Research 2015,” and the International Prize of Physiology Lucien Dautrebande (2016). He is the author/co-author of more than 195 scientific research publications.

Alexander Chervonsky is a professor in the Department of Pathology at The University of Chicago. He is also a chair of the Committee on Immunology at The University of Chicago. Dr. Chervonsky joined The University of Chicago in 2005 from The Jackson Laboratory in Bar Harbor, Maine, where he was a staff scientist. His research interests are in the broad area of immunology with an emphasis on autoimmunity. He has contributed to the understanding of the mechanisms of destruction of the target tissues by the autoimmune response in organ-specific autoimmunity, to the understanding of trafficking of effector T cells to the site of antigen expression, and to the role of innate immunity in regulation of autoimmunity. His later work has been focused on the role of commensal microbes (the microbiota) in the regulation of autoimmunity. He has published a seminal paper describing the connections between the microbiota, innate immunity mechanisms, and the development of autoimmunity. The continuation of this work has revealed the role of the microbiota in the sexual dimorphism of autoimmunity. In addition, Dr. Chervonsky’s laboratory has discovered a role of the inducible changes in glycosylation of the mammalian host’s tissues under conditions of microbial invasion as a protective mechanism decreasing microbial virulence and increasing the wellness of the host. Dr. Chervonsky holds an MD degree from the 1st Moscow Medical School, Moscow, Russia, and a PhD in immunology from the Cancer Research Center, Moscow, Russia. He completed his postdoctoral training at Yale University, New Haven, Connecticut.

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

Angela Douglas is the Daljit S. and Elaine Sarkaria Professor of Insect Physiology and Toxicology at Cornell University in Ithaca, New York. She joined Cornell University in August 2008 from The University of York, United Kingdom, where she had been a professor (personal chair). Dr. Douglas’s research experience concerns beneficial microorganisms in animals, with a particular interest in the nutritional role of microorganisms in insects and their use as a biomedical models for metabolic health. Her works include more than 200 refereed research publications and four academic books on beneficial microbes and animal physiology. In addition to her publications, Dr. Douglas has given many invited lectures and has a strong record of service activities. She has held leadership positions in scientific societies, served on scientific advisory boards, and received awards for her research. She holds a BA in zoology from Oxford University, United Kingdom and a PhD in microbiology from the University of Aberdeen, United Kingdom.

Jeremiah Faith received his PhD in bioinformatics and systems biology from Boston University. He did his postdoctoral training in the laboratory of Jeffrey Gordon at Washington University in St. Louis Medical School with a focus on the structure and function of the gut microbiome in healthy individuals. He is currently an assistant professor in the Immunology Institute and the Institute for Genomics and Multiscale Biology in the Icahn School of Medicine at Mount Sinai in New York. His research focuses on modeling the interactions between diet, gut microbes, and host physiology with an emphasis on inflammatory bowel disease. Ongoing research in the lab includes (1) quantifying the influence of diet and the gut microbiota on host health and disease, (2) identifying microbial strains that modulate host phenotypic variation, and (3) the stability of the human gut microbiota.

James Fox received his veterinary training at Colorado State University. He was an NIH postdoctoral fellow at Stanford University before accepting a position at the University of Colorado Medical Center as an assistant professor. He was later recruited to Massachusetts Institute of Technology and is currently a professor and the director of the Division of Comparative Medicine. He is also an adjunct professor at the Tufts University School of Veterinary Medicine. Dr. Fox has received numerous scientific awards, and was elected to the National Academy of Medicine in 2004. He has been the principal investigator of an NIH postdoctoral training grant for veterinarians for 28 years and has trained 80 veterinarians, physicians, and PhDs for careers in biomedical research. He also has an NIH training grant for veterinary students and has introduced more than 120 veterinary students to careers in biomedical research. He has been funded by NIH and NCI to study infectious diseases of the gastrointestinal tract for the past 40 years and has focused on the pathogenesis of Campylobacter spp. and Helicobacter spp. infection in humans and animals. Dr. Fox has a long-standing interest in studying the gastrointestinal microbiome and how it interfaces with

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

and influences the host’s immune response to gastrointestinal pathogens, particularly Helicobacter species. These studies are complemented by extensive experience with mouse models, including those maintained under gnotobiotic conditions. His laboratory developed the ferret as a model for both Campylobacter- and Helicobacter-associated disease as well as the first rodent model to study Helicobacter-associated gastric disease, including gastric cancer. Dr. Fox is considered an international authority on the epidemiology and pathogenesis of gastric and enterohepatic helicobacters in humans and animals. He is largely responsible for identifying, naming, and describing many of the diseases attributed to various Helicobacter species; most notably their association with hepatitis, liver tumors, inflammatory bowel disease, and colon cancer.

Craig Franklin attended the University of Missouri (MU), where he received his DVM and PhD. He is currently a professor of veterinary pathobiology at MU and directs the Mutant Mouse Resource and Research Center, an NIH-funded repository of genetically engineered mutant mice; the Comparative Medicine Program, a post-DVM laboratory animal medicine residency and advanced degree program; and the Veterinary Research Scholars Program, a summer research program for veterinary students. He also co-directs the new MU Metagenomics Laboratory and is a co-investigator for the Rat Resource and Research Center. He has more than 25 years of experience in rodent disease and diagnostics with an emphasis on infectious and intestinal diseases. His current research home, the Comparative Metagenomics Laboratory, studies environmental variables that may modulate rodent microbiota, the impact of differing microbiota on rodent model phenotypes (e.g., inflammatory and neoplastic diseases of the intestine), and methods to practically manipulate and control complex microbiota to optimize model reproducibility. He also performs collaborative studies involving numerous rodent and domestic species models of disease.

Wendy S. Garrett is a physician-scientist, and her basic science laboratory is located at the Harvard T.H. Chan School of Public Health. Dr. Garrett is a physician at Dana-Farber Cancer Institute and Brigham and Women’s Hospital. Her laboratory is focused on defining the dynamic interactions between the mucosal immune system and gut microbiota. The Garrett laboratory’s experimental questions are grounded in understanding how interactions between intestinal microbial communities and the immune system contribute to the development of inflammatory bowel disease and colorectal cancer. Dr. Garrett has recently received the following awards for her research: a Damon Runyon Foundation Fellowship, a Burroughs Wellcome Career in Medical Sciences Award, a V Foundation Scholar, a Cancer Research Institute Investigator Award, and a Searle Scholars Award.

Jeffrey Gordon is the Dr. Robert J. Glaser Distinguished University Professor at Washington University in St. Louis. He received his AB from Oberlin College and his MD from The University of Chicago. After completing his clinical training in internal medicine and gastroenterology, and a postdoctoral fellowship at

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

NIH, he joined the faculty at Washington University where he has spent his entire career, first as a member of the Departments of Medicine and Biological Chemistry, then as the head of the Department of Molecular Biology and Pharmacology, and for the past decade as the founding director of an interdepartmental Center for Genome Sciences and Systems Biology. Students in his lab have created new types of gnotobiotic animal models and developed new experimental and computational approaches for characterizing the assembly, dynamic operations, functional properties, and biological effects of human gut microbial communities. He has combined these models with human studies involving twins as well as members of birth cohorts living in low-, middle-, and high-income countries representing diverse geographic locations and cultural traditions. His group is focused on addressing the global health challenges of obesity and childhood undernutrition through new understanding of the interactions between diets and the gut microbiome and new ways of promoting healthy development of the gut community during the first several years of postnatal life. He has been the research mentor to more than 125 PhD and MD/PhD students and postdoctoral fellows. He is a member of the National Academy of Sciences, the American Academy of Arts and Sciences, the National Academy of Medicine, and the American Philosophical Society.

Karen Guillemin is the Alec and Kay Keith Professor in the Department of Biology and the Institute of Molecular Biology at the University of Oregon. She is also the founding director of the Microbial Ecology and Theory of Animals (META) Center for Systems Biology, an NIH-funded National Center for Systems Biology established in 2012. Guillemin received her bachelor’s degree in biochemical sciences from Harvard College in 1991 and her PhD from the Department of Biochemistry at Stanford University School of Medicine in 1998, where she worked with Dr. Mark Krasnow studying organ development in the model organism of the fruit fly. She continued her postdoctoral training at Stanford in the Department of Microbiology and Immunology, studying bacterial–host interactions with Dr. Stanley Falkow, studying the bacterial pathogen and carcinogen, Helicobacter pylori. In 2001 she joined the faculty of the University of Oregon, where she established an independent research program that combines her interests in animal development and bacterial–host interactions. Her research group has been instrumental in pioneering the use of gnotobiotic zebrafish to study how resident microbial communities assemble and modulate host biology.

Timothy Hand is an assistant professor of pediatrics and immunology and the chair of the Committee on Gnotobiotics at the University of Pittsburgh. Dr. Hand’s laboratory is within the R.K. Mellon Institute for Pediatric Research at the Children’s Hospital of Pittsburgh. Dr. Hand received his PhD from Yale University in immunology and followed these studies with a productive postdoctoral fellowship at NIH with Dr. Yasmine Belkaid. Dr. Hand’s research focuses on the interaction between the host immune system and the intestinal microbiota, with a particular focus on how this relationship is contextually shaped by diet

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

and infection. Current lab focuses include studies directly relevant to children, such as oral vaccination, cystic fibrosis, necrotizing enterocolitis, and inflammatory bowel disease.

Donald E. Ingber is the founding director of the Wyss Institute for Biologically Inspired Engineering at Harvard University, the Judah Folkman Professor of Vascular Biology at Harvard Medical School, and the Vascular Biology Program at Boston Children’s Hospital, and professor of bioengineering at the Harvard John A. Paulson School of Engineering and Applied Sciences. He received his BA, MA, MPhil, MD, and PhD from Yale University. Dr. Ingber is a pioneer in the field of biologically inspired engineering, and at the Wyss Institute he currently leads a multifaceted effort to develop breakthrough bioinspired technologies to advance health care and to improve sustainability. His work has led to major advances in mechanobiology, tumor angiogenesis, tissue engineering, systems biology, nanobiotechnology, and translational medicine. Through his work, Dr. Ingber has also helped to break down boundaries between science, art, and design. Dr. Ingber has authored more than 400 publications and 125 patents, founded four companies, and been a guest speaker at more than 450 events internationally. He is a member of the National Academy of Medicine, National Academy of Inventors, American Institute for Medical and Biological Engineering, and the American Academy of Arts and Sciences. He was named one of the Top 20 Translational Researchers worldwide in 2012 (Nature Biotechnology) and a Leading Global Thinker of 2015 (Foreign Policy magazine). He has received numerous other honors in a broad range of disciplines, including the Robert A. Pritzker Award and the Shu Chien Award from the Biomedical Engineering Society, the Rous Whipple Award from the American Society for Investigative Pathology, the Lifetime Achievement Award from the Society of In Vitro Biology, the Leading Edge Award from the Society of Toxicology, and the Department of Defense Breast Cancer Innovator Award. Some of Dr. Ingber’s most recently developed technologies include an anticoagulant surface coating for medical devices that replaces the need for dangerous blood-thinning drugs; a dialysis-like sepsis therapeutic device that clears blood of pathogens and inflammatory toxins; a shear stress–activated nanotherapeutic that targets clot-busting drugs to sites of vascular occlusion; and human organs-on-chips created with microchip manufacturing methods and lined by living human cells, which are being used to replace animal testing as a more accurate and affordable in vitro platform for drug development and personalized medicine. In 2015, Dr. Ingber’s organs-on-chips technology was named Design of the Year by the London Design Museum and was also acquired by the Museum of Modern Art (MoMA) in New York City for its permanent design collection.

Stephen Jameson is in the Center for Immunology and Department of Laboratory Medicine and Pathology at the University of Minnesota. He obtained his PhD in Cambridge, England, and postdoctoral training at Scripps Research

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

Institute (La Jolla) and University of Washington (Seattle). His research has focused on factors that regulate CD8 T cell development, homeostasis, and function. A major current area of interest is control and maintain of protective memory CD8 T cells, capable of rapid effector responses and efficient control of pathogens and tumors. These studies included application of novel techniques to isolate and characterize memory-like cells from the pre-immune CD8 T cell pool, building from expertise in the use of peptide/MHC tetramers. Recent work involved characterization of highly protective CD8 T cells, which can be generated by modification of vaccination techniques and novel studies involving mice with more natural exposure to normal environmental pathogens (“dirty mice”), which were shown to be a more faithful model of the adult human immune system. Other studies revolve around the KLF2 transcription factor, which was shown to regulate numerous aspects of lymphocyte trafficking, including key roles in directing the production of recirculating versus resident memory CD8+ T cells and the formation of follicular helper CD4+ T cells. Dr. Jameson plays an active role in graduate student and postdoctoral education and training. He has trained/is training 14 graduate students and 14 postdoctoral fellows, in addition to several undergraduate researchers and two research associates. He was the director of Graduate Studies for the Microbiology, Immunology and Cancer Biology (MICaB) PhD graduate program from 2013 to 2016 and is a member of the steering committee for the NIH-funded T32 Cancer Biology Training Grant.

Aldons (Jake) Lusis is professor of microbiology, human genetics and medicine at the University of California, Los Angeles (UCLA). He obtained his PhD in biophysics from Oregon State University and did postdoctoral work in molecular genetics and mouse genetics at Roswell Park Memorial Institute prior to joining the faculty of UCLA. Dr. Lusis’s lab studies naturally occurring genetic variations in mice and in humans to help understand interactions underlying complex cardiovascular and metabolic disorders. A major focus of the lab over the past decade has been to leverage common genetic variation in populations to integrate clinical traits with “intermediate” phenotypes obtained using high-throughput technologies, such as expression arrays, sequencing, or proteomics, an approach known as “systems genetics.” To facilitate this approach, they have developed a reference resource termed the Hybrid Mouse Diversity Panel that can be used to carry out whole genome association mapping.

Andrew J. Macpherson is a professor of medicine and the director of gastroenterology at the University Hospital of Bern, Switzerland. He studied biochemistry and medicine at Cambridge University and did his PhD on sugar-proton symport systems in the laboratory of Sir Hans Kornberg and Peter Henderson. His clinical medical studies and clinical specialty training in gastroenterology were in Cambridge and London. The results (of control experiments) during a project in London on immune-mediated damage to intestinal epithelial cells focused his interest on the way in which the mucosal immune system responds to commensal intestinal microbes. In 1997 he moved to work with Rolf Zinkernagel at the Insti-

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

tute of Experimental Immunology in Zürich. Between 2004 and 2008 he was the Farncombe Professor of Medicine and a Canada Research Chair holder at McMaster University in Hamilton. His work has shown that there are different pathways of induction of immunoglobulin A in the intestinal mucosa by commensal intestinal microbes, with and without help from T cells. He has also shown a compartmentalization between the mucosal and systemic responses to commensals, since mucosal immune responses are driven locally in the mucosal compartment by dendritic cells that have sampled commensals at the epithelial surface. More recently his lab has developed methods of reversible colonization of germ-free mice to allow intestinal colonization with commensals and mucosal immune priming to be experimentally uncoupled, to address mucosal immune memory, and the functional consequences of mucosal immune responses in host–microbial mutualism and the effects of maternal colonization on immune system development in early life.

Nancy A. Moran is the Leslie Surginer Endowed Professor at the University of Texas in the Department of Integrative Biology. She obtained a BA from The University of Texas in 1976 and a PhD (zoology) from the University of Michigan in 1982. From 1986 to 2010, she served on the faculty of the University of Arizona and from 2010 to 2013 she was a professor at Yale University. She has mentored more than 30 graduate students and postdoctoral researchers. Dr. Moran has been elected to membership in the U.S. National Academy of Sciences, the American Academy of Arts and Sciences, and the American Academy of Microbiology. She received the 2010 International Prize for Biology from the Japan Society for the Promotion of Science, the 2014 James Tiedje Award for lifetime contribution in microbial ecology, and the 2016 Lifetime Research contribution award in molecular evolution from the Society for Molecular Biology and Evolution. Dr. Moran studies the evolution of bacteria and insects, using genomic approaches, and has focused on the evolution of symbiotic bacteria that affect insect ecology.

Joseph T. Newsome is currently an associate professor in the Department of Pathology and the clinical director-Division of Laboratory Animal Resources, University of Pittsburgh. He received a BSc in microbiology in 1980, master’s in pathobiology in 1982 working with the research team of Dr. Richard Olson which developed the first vaccine for feline leukemia, and obtained a DVM in 1986 from The Ohio State University College of Veterinary Medicine. For the next 10 years he wore multiple hats at Georgetown University in Washington, DC, including facility manager, assistant professor of surgery and pathology, and clinical veterinarian overseeing surgery and radiological research support. In 1996 he became a diplomat of the American College of Laboratory Animal Medicine and concurrently completed a postdoctoral training program in experimental pathobiology, assisting in the development of the animal models for the team that eventually led to the current human papilloma virus vaccine. From 2000 to 2012 he was the University of Pittsburgh’s attending veterinarian. He is

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

the author or co-author of more than 65 articles and book chapters. During his career he has been the principal investigator (4) or co-investigator (6) on multiple National Center for Research Resources, National Institutes of Health–funded grants focused on renovations or new construction projects related to vivaria in multiple institutions. He is involved in national and industry-level organizations, such as the American College of Laboratory Animal Medicine (ACLAM), American Association for Laboratory Animal Science (AALAS), and American Veterinary Medical Association (AVMA), with leadership roles such as being a subcommittee chair for the ACLAM foundation since 2010, and being the vice chair of the Policies & Procedures Coordinating Committee (PPCC) of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC) International. His current focus and expertise are in management, biosecurity, biocontainment, facility design and operations, and cancer modeling, immunology, and virology.

Federico Rey is an assistant professor in bacteriology at the University of Wisconsin–Madison. His research program focuses on the human microbiome, with a special interest in how gut microbial metabolism impacts cardiometabolic disease. He earned his bachelor’s and master’s degrees in clinical chemistry from Universidad Nacional de Cordoba in Argentina. As an undergraduate he explored how free radicals modulate vascular tone and atherosclerosis. He did his doctoral work with Professor Caroline Harwood (University of Iowa), studying anaerobic microbial metabolism. He engineered photosynthetic bacteria for improved hydrogen production. After obtaining his PhD, he went on to do postdoctoral studies with Professor Jeffrey Gordon (Washington University in St. Louis), where he explored how human gut microbes interact with each other and their host. In this work, he revealed the metabolic niches of several key members of the community, as well as identifying how they contribute to host health and disease. In Madison, he is expanding on this work, looking at how cardiometabolic disease is impacted by the interplay between host-genetics microbial metabolism and diet.

Buck Samuel aims to comprehensively define the genetic pathways that mediate the influences of microbes on host health. He is an assistant professor in the Alkek Center for Metagenomics and Microbiome Research and Department of Molecular Virology and Microbiology at Baylor College of Medicine. He earned two bachelor’s degrees (magna cum laude with honors) from the University of Idaho and served in the Science Division at the U.S. Department of State in Paris, France. He completed his PhD at Washington University under the mentorship of Jeffrey Gordon, studying the foundations of how microbes shape host metabolism, and performed his postdoctoral developing Caenorhabditis elegans as a facile, high-throughput amenable gnotobiotic system under Gary Ruvkun at Harvard Medical School. Dr. Samuel has been recognized as a National Science Foundation (NSF) Graduate Research Fellow, NIH Ruth L. Kirschstein National Research Service Award (NRSA) Postdoctoral Fellow, and

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

Charles King Trust Postdoctoral Fellow. His interdisciplinary research brings to bear expertise in organismal biology and genomics to high-throughput molecular studies on how microbes and their products impact host physiology.

R. Balfour Sartor, distinguished professor of medicine, microbiology, and immunology; director of the University of North Carolina Multidisciplinary Inflammatory Bowel Disease Center; and director of the Broad Medical Research Program at the Crohn’s & Colitis Foundation of America (CCFA), is a board certified gastroenterologist specializing in inflammatory bowel disease (IBD) and a basic and translational scientist, running a large NIH- and foundation-funded laboratory. Research interests include pathogenesis of Crohn’s disease and ulcerative colitis, microbial/genetic/immunological interactions in the intestine, using gnotobiotic mice and rats to explore mechanisms of resident bacterial induction of chronic, immune-mediated inflammation versus homeostatic protective mucosal immune responses, and translating basic science knowledge to improve IBD diagnosis and treatment. Dr. Sartor has published more than 330 research articles and reviews and edited five books. Recent studies have emphasized commensal bacterial induction of regulatory T and B cells, particularly IL-10-mediated protection, and identification of novel resident protective bacterial species. Recent translational research searches for microbial, immunological, genomic, and genetic predictors of post-operative recurrence of Crohn’s disease and for biomarkers that predict the natural history of Crohn’s disease (aggressive disease requiring biologic therapies with complications leading to Crohn’s disease versus Endoline, easily treated course). He directs a large gnotobiotic unit and uses germ-free mice to investigate mechanisms by which resident microbiota initiate, perpetuate, and protect against chronic intestinal inflammation. He currently serves on the American Gastroenterological Association’s Microbiome Advisory Committee, and has previously been an American Gastroenterological Association (AGA) council member as chair of the Immunology, Microbiology and IBD Section. He now directs the Broad Medical Research Program at CCFA after previously serving as the CCFA’s chief medical advisor, national board member, chairman of the National Scientific Advisory Committee and chairman of the Senior Research and Training Award Review Committees. He has a passion for better understanding immunopathogenic mechanisms and host–microbial interactions of IBD, improving therapies, and training the next generation of scientists and clinicians.

Betty Theriault is an associate professor in the Department of Surgery and clinical veterinarian within the Animal Resources Center at The University of Chicago. She is a veterinarian with more than 30 years of experience working with animals in a variety of settings and across a broad spectrum of species. Dr. Theriault joined The University of Chicago in 2005 and in 2006 embarked on developing The University of Chicago’s Gnotobiotic Research Animal Facility (GRAF), which has experienced 10 years of steady growth and success. Initially developed to support the research of a few scientists at the university, the gnotobiotic program currently supports the work of 12 principal investiga-

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

tors directly as well as The University of Chicago’s Digestive Disease Research Core Center (DDRCC) Host Microbiome Core Gnotobiotic Mouse Component, for which she is co-director. Her work focuses on assisting researchers with developing animal models of human disease and adapting technologies to unique applications. She has been a primary or co-author of publications spanning a diverse range of models and species, including but not limited to, animal models of transplantation, ovarian cancer metastasis, food allergy, circadian rhythm, and obesity. She is the immediate past president of the Chicago branch of the American Association for Laboratory Animal Science and president elect for both the Association for Gnotobiotics, which is currently being revitalized, and the International Society for Gnotobiology. She has received a Distinguished Leader in Program Development and Distinguished Faculty Award from The University of Chicago, as well as the Bengt E. Gustafsson Award presented by the 17th International Society for Gnotobiology and 34th Society of Microbial Ecology and Disease Joint Congress held in Yokohama, Japan, in 2011. Dr. Theriault has presented lectures on topics of gnotobiology at scientific meetings locally, nationally, and internationally and has led workshops at national meetings held for the laboratory animal community. She received her DVM from the University of California, Davis, completed a small animal medicine and surgery internship at the University of Pennsylvania, and is a diplomate of the American College of Laboratory Animal Medicine.

Herbert “Skip” Virgin is the Edward Mallinckrodt Professor and chair of the Department of Pathology and Immunology at the Washington University School of Medicine in Saint Louis, Missouri. He received his AB, MD, and PhD from Harvard University, trained in internal medicine and infectious diseases, and performed postdoctoral studies with Dr. Bernard Fields. He is a member of the American Society for Clinical Investigation, the Association of American Physicians, the American Academy of Microbiology, and the National Academy of Sciences. He serves on the Board of Reviewing Editors of Science and the Editorial Board of Cell. The Virgin laboratory uses genetic, structural, computational, and sequencing methods to define mechanisms of viral pathogenesis and immunity in vivo, with many studies focusing on mouse models. They have identified the physiological role and molecular mechanisms of several RNA and DNA virus immune evasion molecules and studied immune effector mechanisms, including ISG15, interferon-γ, interferon-λ, cGAS, and autophagy. They discovered the first murine norovirus and developed the first culture system for a norovirus. Studying this virus, they showed that virus-plus-host-gene interactions define disease phenotypes. They have recently focused on “trans-kingdom” interactions within the metagenome and on interactions between microbial and viral components of the metagenome and host immunity. They found that persistent γ-herpesvirus infection can “complement” genetic immunodeficiency, and can symbiotically protect the host against bacterial infection. Recent studies also reveal that enteric helminth infection reactivates murine γ-herpesviruses from latency through host cytokine competition at a viral promoter and that

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

these cytokine effects are conserved in a human herpesvirus. They found that bacteria control persistent enteric norovirus infection in a manner dependent on interferon-λ, and identified interferon-λ-induced sterilizing innate immune responses to enteric viral infection. They have used metagenomics to identify constituents of the mammalian virome and to show that the enteric virome is altered in humans and macaques with lentivirus infection as well as in humans with inflammatory bowel disease. Together these studies identify trans-kingdom interactions within the metagenome as key contributors to mammalian biology.

Chriss J. Vowles co-manages a multi-investigator germ-free research laboratory at the University of Michigan in Ann Arbor. He began his career at the University of Michigan in 2003, working full time as a husbandry technician in the Unit for Laboratory Animal Medicine. In 2006, he discovered gnotobiotic technology. At that time, the Germ Free Laboratory was just starting. Mr. Vowles joined the research group on the ground floor and has been growing with it ever since. He has authored the first practical guide in the field of gnotobiotics titled Gnotobiotic Mouse Technology: An Illustrated Guide.

Gary D. Wu is the Ferdinand G. Weisbrod Professor of Medicine at the Perelman School of Medicine at the University of Pennsylvania, where he is the associate chief for research in the Division of Gastroenterology, the associate director of the Center for Molecular Studies in Digestive and Liver Disease, and the co-director of the University of Pennsylvania and Children’s Hospital of Philadelphia (PennCHOP) Microbiome Program. He was the inaugural director and chair of the Scientific Advisory Board for the American Gastroenterological Association Center for Gut Microbiome Research and Education and is an elected member of both the American Society for Clinical Investigation and the American Association of Physicians. Research programs in the Wu laboratory focus on the mutualistic interactions between the gut microbiota and its host, with a particular emphasis on metabolism, including nitrogen balance, intestinal oxygen regulation, and epithelial intermediary metabolism. Of particular interest is the effect of diet on the gut microbiome and its relationship to therapeutic responses associated with the use of defined formula diets in the treatment of Crohn’s disease. Insights gained from these projects will hopefully lead to the development of better diets for patients with IBD.

Vincent B. Young is an associate professor in the Department of Internal Medicine/Infectious Diseases Division and the Department of Microbiology and Immunology at the University of Michigan Medical School. He received his undergraduate degree from the Massachusetts Institute of Technology and received his MD and PhD from Stanford University. He completed his clinical training in internal medicine and infectious diseases at the Massachusetts General Hospital. He was previously on the faculty at Michigan State University prior to joining the University of Michigan in 2007. Dr. Young has a long-standing interest in understanding the pathogenesis of bacterial infections of the gastrointestinal

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×

tract and the role of the normal microbiota in human health and disease. Dr. Young led a Human Microbiome Project on the role of the microbiome in inflammatory bowel disease. He is also involved in projects that look at microbial communities in the lungs of patients with HIV infection and cystic fibrosis. Current research in the Young Lab includes a “team science” effort to understand the pathogenesis of Clostridium difficile infection by an integrated approach that combines clinical research, bacterial genomics, microbial ecology, and immunology/host response projects. He is also leading a group of investigators that is developing the use of stem cell–derived intestinal organoids as a novel alternative model system for the study of enteric disease agents.

Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 78
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 79
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 80
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 81
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 82
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 83
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 84
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 85
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 86
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 87
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 88
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 89
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 90
Suggested Citation:"Appendix C: Biographical Sketches of Workshop Speakers and Moderators." National Academies of Sciences, Engineering, and Medicine. 2018. Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop. Washington, DC: The National Academies Press. doi: 10.17226/24858.
×
Page 91
Next: Appendix D: Glossary »
Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop Get This Book
×
 Animal Models for Microbiome Research: Advancing Basic and Translational Science: Proceedings of a Workshop
Buy Paperback | $45.00 Buy Ebook | $36.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

The surface of the human body and its mucous membranes are heavily colonized by microorganisms. Our understanding of the contributions that complex microbial communities make to health and disease is advancing rapidly. Most microbiome research to date has focused on the mouse as a model organism for delineating the mechanisms that shape the assembly and dynamic operations of microbial communities. However, the mouse is not a perfect surrogate for studying different aspects of the microbiome and how it responds to various environmental and host stimuli, and as a result, researchers have been conducting microbiome studies in other animals.

To examine the different animal models researchers employ in microbiome studies and to better understand the strengths and weaknesses of each of these model organisms as they relate to human and nonhuman health and disease, the Roundtable on Science and Welfare in Laboratory Animal Use of the National Academies of Sciences, Engineering, and Medicine convened a workshop in December 2016. The workshop participants explored how to improve the depth and breadth of analysis of microbial communities using various model organisms, the challenges of standardization and biological variability that are inherent in gnotobiotic animal-based research, the predictability and translatability of preclinical studies to humans, and strategies for expanding the infrastructure and tools for conducting studies in these types of models. This publication summarizes the presentations and discussions from the workshop.

READ FREE ONLINE

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!