Our understanding of human physiology and disease susceptibility is dramatically changing. Emerging research over the past decade revealed our bodies integrate the biology of our human cells and genome with the trillions of bacteria that live within and on us - a significant component of how susceptible we are to disease appears to be derived from resident microbial communities.
Defining the molecular participants and pathways that link human commensal to physiologic and disease processes is a critical ongoing need. Defining new relationships governing the natural history of human disease offers new prevention and treatment approaches.
Cleveland Clinic formed the Center for Microbiome & Human Health (CMHH) in 2017 to be on the forefront of this emerging area of research. The goal of the CMHH is to aid clinical, translational and basic researchers in performing microbiome-related research. The CMHH aims to enable investigators to improve our understanding of the mechanisms by which the human microbiome impacts human health and disease, and to become the recognized world leader in the development of microbiome-targeted therapeutics.
Philip P. Ahern, D. Phil.
Assistant Staff, Cardiovascular & Metabolic Sciences
Research in my laboratory is focused on understanding host-microbiota interactions in the development and resolution of inflammatory bowel disease. Specifically, we are interested in understanding:
(1) What members of the microbiota are responsible for the initiation and perpetuation of chronic intestinal inflammation?
(2) What pathways promote the resolution of bacterially-triggered intestinal inflammation?
(3) How do gut microbes regulate intestinal CD4+ T cell fate in health and disease?
Our overarching goal is to enhance our understanding of the role played by host-microbiota interactions in chronic intestinal inflammation, and to leverage this knowledge to develop new and improved therapeutic strategies for the treatment of IBD.
Daniel Blankenberg, PhD
Assistant Staff, Genomic Medicine Institute
Our laboratory develops tools and approaches to generate and harness the rich content of high-throughput omics datasets. Through our work on the Galaxy project, we enable and perform accessible, reproducible, and transparent computational biomedical research. We are currently building tools and resources for data-intensive collaborative analysis and visualization of microbiome datasets.
J. Mark Brown, PhD
Director of Research, Center for Microbiome & Human Health
My laboratory is focused on understanding mechanisms by which gut microbial nutrient metabolism produces hormone-like small molecules that drive host disease pathogenesis. Our major efforts now are identifying novel gut microbe-derived metabolites that play a causative role in obesity, type 2 diabetes, non-alcoholic and alcoholic liver disease, and liver cancer. These ongoing efforts use animal and cell models to identify the host receptor systems necessary for sensing gut microbial metabolites, as well as understanding the microbial enzymes responsible for the production of disease-associated metabolites.
Our ultimate goal is to identify disease mitigating therapeutic strategies targeting the gut microbial endocrine organ using a compliment of probiotic, small molecule, and bacterial gene therapy approaches.
Jianguo Cheng, MD, PhD
Staff, Neurosciences
Our lab is focused on developing new therapeutic approaches for chronic pain and opioid tolerance.
Jan Claesen, PhD
Assistant Staff, Cardiovascular & Metabolic Sciences
The Claesen group aims to functionally characterize molecular mechanisms that control bacterial interspecies and microbe-host interactions in the human microbiome. Bacteria use small molecule chemicals to mediate these interactions and the genetic information required for their production is typically encoded in one physical location of the bacterial chromosome, in biosynthetic gene clusters (BGCs). Using in silico techniques, we identified several widespread families of BGCs that we are now characterizing experimentally, prioritizing on the BGCs predicted to be involved in modulation of community composition or interaction with the host immune system. Our research will contribute to a better mechanistic understanding of the microbes that live in our gut, leading to the discovery of druggable small molecules, new targets for antibacterial therapy and beneficial bacterial strains that can be employed for intervention therapies. The areas of expertise in the Claesen group include microbiology, bacterial genetics and synthetic biology, small molecule biosynthesis and biochemistry.
Gail Cresci, PhD
Staff, Pediatric Gastroenterology; Inflammation & Immunity
My laboratory centers on clinical situations in which gut dysbiosis occurs with investigations targetingalterations in metabolic byproducts (post-biotics) of the gut microbiota. Taking a nutritional preventative and therapeutic approach, my ultimate goal is to discover and develop therapies to maintain or restore digestive and gut health that is altered during gut dysbiosis and associated pathologies.
Our recent work investigates mouse models of antibiotic and ethanol-induced gut dysbiosis and associated gut-liver injury. Here we have investigated repletion of both depleted commensal bacteria and butyrate as a means to mitigate gut-liver injury. Developing data investigates a targeted approach to positively affect gut luminal butyrate levels and prevent injury through provision of designer synbiotics.
One of our major goals is to identify and characterize the microbial and metabolomic signature associated with the progression of pathologies including alcoholic and non-alcoholic fatty liver disease, chemotherapy-induced mucositis, as well as clinical outcomes and organ integrity in liver and intestinal organ transplant patients.
Srinivasan Dasarathy MD
Staff, Inflammation & Immunity
My laboratory is focused on understanding the mechanisms of skeletal muscle proteostasis in chronic diseases. We and others have shown that ammonia perturbs skeletal muscle proteostasis. Over the past decade we have dissected the adaptive and maladaptive cellular responses to ammonia induced stress. Ammonia is a cytotoxic metabolite that is generated by amino acid metabolism in the body as well as from gut bacterial catabolism of urea and proteins. The best known clinical consequence of hyperammonemia is encephalopathy. Interestingly, the major therapeutic strategy to prevent and reverse hepatic encephalopathy is to decrease gut bacterial ammoniagenesis. Gut dysbiosis has been reported to worsen fatty liver and alcoholic liver disease via perturbations in metabolic pathways all of which are of interest to our research program in my laboratory, we have a great interest in developing methods to identify and alter the gut microbiome.
Curtis J. Donskey, MD
Infectious Diseases Section, Cleveland VA Medical Center
The goal of my laboratory is to develop improved strategies to prevent infections due to Clostridium difficile and other healthcare-associated pathogens that colonize the intestinal tract. One of our primary efforts focuses on understanding the impact of antibiotic therapy on colonization by pathogens. We use mouse models of intestinal colonization and conduct translational studies in patients. Some of our current interests include identification of biomarkers of colonization resistance, characterizing the effects of different antibiotics on pathogen colonization, and evaluation of novel approaches to eliminate pathogens or prevent adverse effects of antibiotics (eg, administration of beta-lactamases that eliminate antibiotics from the intestinal tract).
Mohammed Dwidar, PhD
Director, Microbial Culturing & Engineering Core
The Microbial Culturing & Engineering Core provides services for culturing and genetic engineering of bacteria in concordance with other mechanistic studies related to the microbiome.
Haytham Elgharably, MD
Associate Staff Surgeon, Thoracic & Cardiovascular Surgery
My research interest is microbial virulence including biofilm formation in infective endocarditis, in particular staphylococcus aureus infection.
Charis Eng MD, PhD
Department Chair, Genomic Medicine Institute
Our research identifies and characterizes genes (e.g., PTEN, SDH) that cause susceptibility to inherited cancer syndromes, determines their role in sporadic carcinogenesis and performs molecular epidemiologic analyses as they relate to clinical care in the realm of precision healthcare, with a significant focus on understanding the mechanistic switch between cancer predisposition and autism spectrum disorder in those carrying PTEN mutations.
Claudio Fiocchi, MD
Staff, Inflammation & Immunity
Our broad interest is the pathogenesis of Crohn’s disease and ulcerative colitis, the two main forms of inflammatory bowel disease (IBD). Any microbial factor or abnormalities associated with IBD are, in principle, of interest to us.
Neetu Gupta, PhD
Associate Staff, Inflammation & Immunity
My lab investigates mechanisms of lymphocyte activation in adaptive immunity, autoimmunity and cancer in order to identify novel therapeutic targets and develop better vaccines. Specifically, we are interested in evaluating the impact of the microbiome and metabolome on immune cell activation in the context of novel cancer immunotherapies with the goal of altering the gut flora to improve clinical outcomes. To achieve our goals we employ primary patient specimens, mouse models, and multi-omics approaches supported by bioinformatics.
Adeline (Lynn) Hajjar DVM, PhD
Associate Staff, Cardiovascular & Metabolic Sciences
My lab uses germfree mouse models to study how the host selects for microbiota that in turn affect the immune response. By performing fecal-microbiota transplants (FMT) into our germfree cystic fibrosis (CF) mouse model compared to non-CF mice we have evidence that the CF gut selects for unique microbiota and that these in turn alter the balance of Th17/Treg immune cells. We aim to define the mechanism and consequences of this selection, to determine whether and how to alter the selection, and ultimately to develop new treatment approaches aimed at manipulating the microbiota to lessen the consequences of pathogenic inflammation in CF.
Stanley Hazen, MD, PhD
Director, Center for Microbiome & Human Health
My laboratory is focused on understand mechanisms responsible for the development and propagation of cardiovascular disease. Our major efforts now are targeting how gut microbiota contribute to this process. Serving as a filter for our largest environmental exposure, what we eat, our gut microbiome generates myriad metabolites that have potential to impact physiological processes, and disease susceptibility. Numerous projects in our laboratory explore different aspects of gut microbial participation in cardiovascular disease susceptibility and pathogenesis. Projects range from discovery efforts aimed at identifying microbial participants in atherosclerosis, thrombosis, and heart failure risks, as well as metabolic diseases like chronic kidney disease, and components of metabolic syndrome. Development of therapeutic interventions that target identified microbial participants in disease processes are also a major focus.
Christopher Hine, PhD
Assistant Staff, Cardiovascular & Metabolic Sciences
The interactions between diet, metabolism, and aging are the focus of my laboratory. As aging and aging related diseases are major driving forces for death and disability, understanding the physiological and molecular mechanisms behind them provides avenues for safe and effective treatments. Specifically, we examine how diet and aging regulate the production of the gaseous signaling molecule hydrogen sulfide inside of our cells, tissues, and gut microbe inhabitants and how this change in hydrogen sulfide production affects our health and wellbeing. Having a better understanding of how microbiome produced hydrogen sulfide relates to dysfunctional and diseased states will allow for targeted nutritional and pharmacological interventions to harness this powerful and potent gas.
Michael Kattan, PhD
Department Chair, Quantitative Health Sciences
My interest is in the development, validation, and use of prediction models. I have developed several such models in cancer and released them as freely available software, from rcalc.ccf.org.
Hyun Jung Kim, PhD
Assistant Staff, Inflammation & Immunity
Our lab explores the biomimetic microphysiological system to uncover fundamental mechanisms of host-microbiome crosstalk in health and disease.
Justin Lathia, PhD
Staff, Cardiovascular & Metabolic Sciences
My laboratory seeks to understand the mechanistic links between the gut microbiome and the most common and lethal adult brain cancer, glioblastoma (GBM). We have investigated the effect of high fat diet on the microbiome and its relationship to disease progression of GBM. We also explored sex-specific changes in the composition of the gut microbial community during the course of cancer progression and its potential correlation with anti-tumor immune response. In both cases, we identified strains of bacteria enriched in animal models of GBM. Relative abundance of these bacterial communities were determined by host sex and specific to tumor type. As we expand our research program, we are specifically interested in identifying the set of bacteria-derived metabolites and macromolecules that traverse the gut-brain axis to modify the function of tumor cells as well as immune cell populations infiltrating the tumor microenvironment. This fundamental understanding will open avenues for novel therapeutic development in GBM.
Feng Lin, PhD
Staff, Department of Inflammation and Immunity
My laboratory is interested in studying the interactions between the innate immune system and gut microbiota. We have been analyzing gut microbiota and their metabolites in animals sufficient or deficient in certain complement components or receptors. Our ultimate goals are to understand the mechanism by which the innate immune system regulates the gut microbiota and to use the insights to develop new reagents for the modification of gut microbiome.
Phoebe Lin, MD, PhD
Staff, Ophthalmic Research
Dr. Lin is a clinician-scientist trained in vitreoretinal surgery, ocular inflammatory disease, pharmacology and ocular immunology. Her lab focuses on discovering novel pathogenic pathways in diseases of the eye that involve ocular inflammation, with the purpose of devising new therapies that might either improve or prevent vision loss. Specifically, her lab utilizes preclinical models of uveitis (an inflammatory condition of the middle lining of the eye) to address several pathways: leukocyte migration from the intestinal tract during autoimmune uveitis, immune regulation through intestinal Tregs as a way to re-establish immune homeostasis and prevent autoimmune uveitis, and the role of the mammalian commensal intestinal microbiota in intestinal and systemic immunity affecting the development and mitigation of ocular inflammation. Dr. Lin's lab is also investigating the impact of an intestinal dysbiosis that occurs in age-related macular degeneration, a disease that is caused by disruptions in innate immunity and fatty acid metabolism.
Ignacio Mata, PhD
Assistant Staff, Genomic Medicine Institute
I work on the genetics of Parkinson’s disease, with a especial interest in underrepresented populations. I lead a large consortium (35 centers in 12 different countries) in Latin America and the Caribbean to understand the genetic and environmental factors that may be associated to disease development and progression. I have been interested in studying aspects related to the microbiome, and be able to compare patients in different countries/environments. I understand this would be very interesting but also very challenging. I just wrote a small grant with some colleagues in Chile to start a pilot project to see how we could do this in LATAM.
Keith McCrae, MD
Staff, Taussig Cancer Institute; Cardiovascular & Metabolic Sciences
Our laboratory has longstanding interest in multiple areas broadly related to vascular function, thrombosis, extracellular vesicles and the role of the vasculature in cancer biology. While we have not performed in depth research in the microbiome, we are currently initiating studies to examine the roles of microbial metabolites in the development of cancer associated thrombosis. A major interest of the lab is the antiphospholipid syndrome, an immune-mediated thrombotic disease and we plan to explore whether microbial metabolites influence the development of antiphospholipid antibodies and their propensity to cause thrombotic events.
Christine McDonald, PhD
Associate Staff, Inflammation & Immunity
Dr. McDonald investigates anti-microbial defense mechanisms in the gastrointestinal tract with a focus on how dietary and genetic factors alter these protective responses to result in increased susceptibility to infection or development of chronic inflammatory diseases, such as Crohn's disease. She has found that common dietary additives found in Western diets dramatically alter both the composition and function of the normal intestinal microbiota and impair anti-microbial defenses of the intestine. These studies may ultimately lead to the use of beneficial dietary interventions as innovative and safe means to prevent intestinal disease or reduce disease severity in individuals with established intestinal inflammation.
Thomas McIntyre PhD
Director of Project Development, Center for Microbiome & Human Healt
Our lab studies how cells respond to external stimuli, specifically in the vascular system.
Jeannette Messer, DVM, PhD
Assistant Staff, Inflammation & Immunity
The Messer Lab studies the role of gut microbes in chronic inflammatory diseases. Our focus is on identifying disease-causing changes in microbial behavior across the gut microbial community. In particular, we study how virulence is activated in normally commensal gut bacteria and how that virulence contributes to inflammatory bowel diseases, metabolic diseases, and cancers. We have recently discovered a novel mechanism of gut antibacterial defense that specifically targets bacteria expressing virulence, inactivates virulence mechanisms, and turns off expression of that virulence. More importantly, this antibacterial defense system is compromised in active inflammatory bowel diseases. We are now working to exploit this innate immune mechanism to identify microbes and microbial behaviors that contribute to damage during inflammatory bowel diseases and develop strategies to target these bacteria and their disease-causing behavior therapeutically.
Aaron Miller, PhD
Assistant Staff, Cardiovascular & Metabolic Sciences
The Stones Translational Research Laboratory is focused on understanding the interactions between the microbiome and urinary stone disease (USD), along with the development of bacteriotherapies designed to inhibit recurrent USD. We approach these goals using multi-omics experiments and bioinformatics in three broad arms:
Laura Nagy, PhD
Staff, Inflammation & Immunity
My laboratory is interested in understanding the pathophysiology of alcoholic and non-alcoholic liver disease, with a particular emphasis on the contributions of gut-immune-liver interactions in disease progression. Both chronic, heavy alcohol consumption and obesity/metabolic syndrome have strong effects on the gut microbiome and dysbiosis is thought to contribute to disease progression and severity. Our long term goal is to design therapeutic interventions that positively impact gut microbial health to prevent and treat metabolic liver diseases.
Sathyamangla Prasad, PhD
Staff, Cardiovascular & Metabolic Sciences
My lab is investigating the interactions between the microbiome and immune homeostasis, with the long term goal to design bacteriotherapies to alleviate autoimmune diseases (including Psoriasis, Lupus and Sjögren's Syndrome). To achieve these goals, our approaches include multi-omics experiments, bioinformatics and genetic manipulation.
Ofer Reizes, PhD
Staff, Cardiovascular & Metabolic Sciences
My laboratory focuses on women’s cancers with a specific interest in overcoming chemoresistance to therapeutics agents in gynecologic cancers. Uterine and ovarian cancer are the first and second highest diagnosed gynecologic malignancies with ovarian cancer patients exhibit the poorest overall survival. Our studies focus on platinum resistance mechanisms. Emerging studies indicate that the gut microbiome modulates the response of multiple cancers to immune checkpoint inhibitors and chemotherapeutics including platinum. Given the need to identify strategies to overcome chemotherapeutic resistance in gynecologic cancers, our studies focus on the gut and vaginal microbiomes as source of biomarkers and targets to sensitize gynecologic cancers to mainstay therapeutic regimens.
Daniel Rhoads, MD
Section Head of Microbiology, Cleveland Clinic
I am interested in chronic, multispecies biofilm infections, and the role of bacteriophages in microbial systems.
Florian Rieder, MD
Staff, Inflammation & Immunity; Gastroenterology, Hepatology & Nutrition
The inflammatory bowel disease (IBD) course is highly heterogenous. Intestinal fibrosis causing clinically apparent stricture formation is a common feature of both entities of IBD, Crohn’s disease and Ulcerative colitis and leads to a significantly impaired quality of life in affected patients, intestinal obstruction as well as need for surgical intervention.
Our group focusses on the discovery of novel mechanisms of intestinal fibrogenesis, with a particular emphasis on microbiome host interactions driving stricture formation. For this purpose, we are using primary human cells, tissues and organ culture systems as well as novel animal models of intestinal fibrosis.
Naseer Sangwan, PhD
Director, Microbial Sequencing Core
The Microbial Sequencing & Analytics Resource facility provides investigators with a single point of contact for study design, power calculations, automated nucleic acid (DNA/RNA) extraction, sequencing library preparation and integrative microbiome informatics and analysis of the sequencing data.
Stephanie Schmit, PhD
Associate Staff, Genomic Medicine Institute
I’m a genetic and molecular epidemiologist with a research focus on colorectal cancer. My interest in this area focuses on investigating the role of the microbiome in shaping the strength and diversity of immune responses in the tumor microenvironment of colorectal cancer and its precursors. Also, I have a specific interest in future work on butyrate-producing bacteria and F. nucleatum.
Natalie Silver, MD
Associate Staff, Center for Immunotherapy & Precision Immuno-Oncology; Otolaryngology
Our lab is exploring novel ways to reprogram the tumor immune microenvironment to develop therapeutics for head and neck cancers.
Apollo Stacy, PhD
Assistant Staff, Cardiovascular & Metabolic Sciences
Led by Dr. Apollo Stacy, the Apollo Lab aims to decipher microbiota-host metabolic communication in the oral cavity, specifically in the settings of gum disease and oral malignancies.
Thaddeus Stappenbeck, MD, PhD
Chair, Inflammation & Immunity
Our lab studies the factors that impact inflammation and wound repair and cause inflammatory bowel disease.
Olga Stenina, PhD
Staff, Cardiovascular & Metabolic Sciences
The focus of our microbiome-related research is on the diabetes-related changes in gut microbiome and the effects of these changes on inflammation and cancer growth. Obesity, Western diet, insulin resistance, and type 2 diabetes are associated with increased local and systemic inflammation and increased risk of several cancers. Association studies in patients and the analysis of antibiotics use suggested that gut microbiome directly affects these diabetes-related changes, but the cause for alterations in microbiome and the mediators of diabetes-associated inflammation and cancer growth remain unknown. Microbial growth critically depends on gut secretome, which can either promote or prevent it, or selectively affect distinct species. Our work is focused on two secreted extracellular matrix proteins, thrombospondin-1 (TSP1) and thrombospondin-4 (TSP4). Both TSP1 and TSP4 are changed in diabetic tissues, regulate microbiome, and are implicated in regulation of inflammation and cancer growth. The goal of our work is to identify new therapeutic targets to normalize microbiome in diabetes in order to reduce inflammation and to prevent diabetes-associated cancer growth.
Hoonkyo Suh, PhD
Associate Staff, Neurosciences
My potential interest is to investigate the role of microbiome in brain functions. In particular, I would like to study the function of microbiome in induction and progression of alcohol dependence and its related pathological conditions, as well as neuropsychological responses such as autism, anxiety, and depression.
W. H. Wilson Tang, MD
Staff, Cardiovascular Medicine
As a practicing heart failure/transplant cardiologist, my primary research interest is to investigate how the heart interacts with other organs in the setting of heart failure and renal dysfunction (particularly with the kidneys, lungs, and gastrointestinal systems). My NIH-funded translational research projects focus on understanding the mechanisms through which gut microbiome and its dietary metabolites contribute to disease progression in heart failure, specific cardiomyopathies, cardio-renal diseases. This includes mechanistic animal models and human studies. Since 2015, I also lead the Center for Clinical Genomics, with one of the tasks aiming to promote scientific investigations in targeting clinical microbiome towards therapeutic applications for various diseases.
Zeneng Wang, PhD
Staff Scientist, Cardiovascular & Metabolic Sciences
My laboratory is focused on understanding mechanisms by which gut microbial metabolites are involved in metabolism pathways linked to atherosclerotic cardiovascular disease using mass spectrometry based metabolomics approach. Our major efforts now are identifying novel gut microbe-derived metabolites by comparing difference in metabolome between conventionized mice and germ free mice, once the gut microbiota metabolites are identified, we will used a larger human cohort to test their clinical relevance in cardiovascular disease prevalence and prospective risk for major adverse cardiac events. Then we will demonstrate the biosynthesis of the gut microbiota metabolites and figure out the bacterium species involved. Our ultimate goal is to identify gut microbiota metabolite pathway linked to atherosclerotic cardiovascular disease and develop gut microbiota enzymatic inhibitors to attenuate atherosclerosis.
James C. Witten, MD
Xianfang Wu, PhD
Assistant Staff, Cancer Biology
We are developing a human pluripotent stem cell-derived model to connect intestinal and liver organs using microfluidic device and use them to understand relationship between gut microbiome and liver disease.
Jennifer Yu, MD, PhD
Staff, Department of Radiation Oncology; Staff, Department of Cancer Biology
We aim to understand the role of the microbiome in modulating therapeutic responses, particularly radiation, in primary and metastatatic brain tumors. We also aim to assess the impact of the microbiome in modulating long-term toxicities of radiation, such as neurocognitive deficits.
Jianjun Zhao, MD, PhD
Assistant Professor, Cancer Biology
Multiple myeloma (MM) is a malignancy of plasma cells. There is an increasing evidence that the gut microbiome plays an important role on its host’s adaptive and innate immunity and the bone marrow niche. We are interested in the potential mechanisms by which the microbiome may influence the initiation of MM, MM patients’ responses to therapy, and toxicities of MM patients with autologous transplant.
Weifei Zhu, PhD
Staff Scientist, Cardiovascular & Metabolic Sciences
Research in my laboratory is focused on the role of gut microbes on cerebral vascular diseases. Employing preclinical models, multidisciplinary and innovative approaches, we aim to identify the contribution of specific gut microbiota driven pathways to the development of heightened stroke risks, and adverse functional outcomes following stroke. We are working to provide preclinical evidence that drugging the microbiome may serve as a novel strategy to prevent and treat cerebrovascular diseases. Specifically, we are interested in:
The Center for Microbiome & Human Health provides and manages core facilities available to clinicians and scientists across Cleveland Clinic. These core facilities allow investigators to leverage sequencing-based platforms to analyze microbial community structure in clinical samples; culture and genetically engineer human commensal bacteria for functional studies; perform germ-free and microbial transplantation studies in mouse models; and analyze the microbe-derived metabolome in biological samples from humans or animal models. Additionally, the Center supports the large and growing biobanking of materials with integrated patient medical history necessary for clinical and translational studies.
The Proteomics & Metabolomics Core provides a wide range of metabolomics mass spectrometry services to investigators within Cleveland Clinic.
Learn MoreThe Gnotobiotics facility enables experimental use of germfree and gnotobiotic mice colonized exclusively with investigator-established microbiota.
The Microbial Sequencing & Analytics Resource facility provides investigators with a single point of contact for study design, power calculations, automated nucleic acid (DNA/RNA) extraction, sequencing library preparation and integrative microbiome informatics and analysis of the sequencing data.
Learn MoreThe Microbial Culturing & Engineering Core provides services for culturing and genetic engineering of bacteria in concordance with other mechanistic studies related to the microbiome.
Learn MoreOur members are actively engaged in both basic science and clinical investigation surrounding the mechanisms by which diverse microbiomes impact human health. Several recent publications from Center members can be found using the link to PubMed below.
View PublicationsDisruption of the gut microbiota confers cisplatin resistance in epithelial ovarian cancer.
Chambers LM, Esakov Rhoades EL, Bharti R, Braley C, Tewari S, Trestan L, Alali Z, Bayik D, Lathia JD, Sangwan N, Bazeley P, Joehlin-Price AS, Wang Z, Dutta S, Dwidar M, Hajjar A, Ahern PP, Claesen J, Rose P, Vargas R, Brown JM, Michener CM, Reizes O. Cancer Res. 2022 Oct 7:CAN-22-0455. doi: 10.1158/0008-5472.CAN-22-0455. Online ahead of print. PMID: 36206317
Gut microbe-targeted choline trimethylamine lyase inhibition improves obesity via rewiring of host circadian rhythms.
Schugar RC, Gliniak CM, Osborn LJ, Massey W, Sangwan N, Horak A, Banerjee R, Orabi D, Helsley RN, Brown AL, Burrows A, Finney C, Fung KK, Allen FM, Ferguson D, Gromovsky AD, Neumann C, Cook K, McMillan A, Buffa JA, Anderson JT, Mehrabian M, Goudarzi M, Willard B, Mak TD, Armstrong AR, Swanson G, Keshavarzian A, Garcia-Garcia JC, Wang Z, Lusis AJ, Hazen SL, Brown JM. Elife. 2022 Jan 24;11:e63998. doi: 10.7554/eLife.63998. PMID: 35072627 Free PMC
Local barriers configure systemic communications between the host and microbiota.
Lu Q, Stappenbeck TS. Science. 2022 May 27;376(6596):950-955. doi: 10.1126/science.abo2366. Epub 2022 May 26. PMID: 35617395 Review.
The microbial gbu gene cluster links cardiovascular disease risk associated with red meat consumption to microbiota L-carnitine catabolism.
Buffa JA, Romano KA, Copeland MF, Cody DB, Zhu W, Galvez R, Fu X, Ward K, Ferrell M, Dai HJ, Skye S, Hu P, Li L, Parlov M, McMillan A, Wei X, Nemet I, Koeth RA, Li XS, Wang Z, Sangwan N, Hajjar AM, Dwidar M, Weeks TL, Bergeron N, Krauss RM, Tang WHW, Rey FE, DiDonato JA, Gogonea V, Gerberick GF, Garcia-Garcia JC, Hazen SL. Nat Microbiol. 2022 Jan;7(1):73-86. doi: 10.1038/s41564-021-01010-x. Epub 2021 Dec 23. PMID: 34949826 Free PMC article.
Fusobacterium is enriched in oral cancer and promotes induction of programmed death-ligand 1 (PD-L1).
Michikawa C, Gopalakrishnan V, Harrandah AM, Karpinets TV, Garg RR, Chu RA, Park YP, Chukkapallia SS, Yadlapalli N, Erikson-Carter KC, Gleber-Netto FO, Sayour E, Progulske-Fox A, Chan EKL, Wu X, Zhang J, Jobin C, Wargo JA, Pickering CR, Myers JN, Silver N. Neoplasia. 2022 Sep;31:100813. doi: 10.1016/j.neo.2022.100813. Epub 2022 Jul 11. PMID: 35834946
Targeting the Gut Microbiotaand Host Immunity with a Bacilli-Species Probiotic during Antibiotic Exposure in Mice.
Shapiro D, Kapourchali FR, Santilli A, Han Y, Cresci GAM. Microorganisms. 2022 Jun 8;10(6):1178. doi: 10.3390/microorganisms10061178.