Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Justine W. Debelius is active.

Publication


Featured researches published by Justine W. Debelius.


Cell | 2016

Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease

Timothy R. Sampson; Justine W. Debelius; Taren Thron; Stefan Janssen; Gauri G. Shastri; Zehra Esra Ilhan; Collin Challis; Catherine E. Schretter; Sandra Rocha; Viviana Gradinaru; Marie-Françoise Chesselet; Ali Keshavarzian; Kathleen M. Shannon; Rosa Krajmalnik-Brown; Pernilla Wittung-Stafshede; Rob Knight; Sarkis K. Mazmanian

The intestinal microbiota influence neurodevelopment, modulate behavior, and contribute to neurological disorders. However, a functional link between gut bacteria and neurodegenerative diseases remains unexplored. Synucleinopathies are characterized by aggregation of the protein α-synuclein (αSyn), often resulting in motor dysfunction as exemplified by Parkinsons disease (PD). Using mice that overexpress αSyn, we report herein that gut microbiota are required for motor deficits, microglia activation, and αSyn pathology. Antibiotic treatment ameliorates, while microbial re-colonization promotes, pathophysiology in adult animals, suggesting that postnatal signaling between the gut and the brain modulates disease. Indeed, oral administration of specific microbial metabolites to germ-free mice promotes neuroinflammation and motor symptoms. Remarkably, colonization of αSyn-overexpressing mice with microbiota from PD-affected patients enhances physical impairments compared to microbiota transplants from healthy human donors. These findings reveal that gut bacteria regulate movement disorders in mice and suggest that alterations in the human microbiome represent a risk factor for PD.


Cell Metabolism | 2014

Specialized Metabolites from the Microbiome in Health and Disease

Gil Sharon; Neha Garg; Justine W. Debelius; Rob Knight; Pieter C. Dorrestein; Sarkis K. Mazmanian

The microbiota, and the genes that comprise its microbiome, play key roles in human health. Host-microbe interactions affect immunity, metabolism, development, and behavior, and dysbiosis of gut bacteria contributes to disease. Despite advances in correlating changes in the microbiota with various conditions, specific mechanisms of host-microbiota signaling remain largely elusive. We discuss the synthesis of microbial metabolites, their absorption, and potential physiological effects on the host. We propose that the effects of specialized metabolites may explain present knowledge gaps in linking the gut microbiota to biological host mechanisms during initial colonization, and in health and disease.


Nature | 2016

Microbiome-wide association studies link dynamic microbial consortia to disease

Jack A. Gilbert; Robert A. Quinn; Justine W. Debelius; Zhenjiang Zech Xu; James T. Morton; Neha Garg; Janet K. Jansson; Pieter C. Dorrestein; Rob Knight

Rapid advances in DNA sequencing, metabolomics, proteomics and computational tools are dramatically increasing access to the microbiome and identification of its links with disease. In particular, time-series studies and multiple molecular perspectives are facilitating microbiome-wide association studies, which are analogous to genome-wide association studies. Early findings point to actionable outcomes of microbiome-wide association studies, although their clinical application has yet to be approved. An appreciation of the complexity of interactions among the microbiome and the hosts diet, chemistry and health, as well as determining the frequency of observations that are needed to capture and integrate this dynamic interface, is paramount for developing precision diagnostics and therapies that are based on the microbiome.


PLOS ONE | 2014

HLA-B27 and human β2-microglobulin affect the gut microbiota of transgenic rats

Phoebe Lin; Mary Bach; Mark Asquith; Aaron Y. Lee; Lakshmi Akileswaran; Patrick Stauffer; Sean Davin; Yuzhen Pan; Eric D. Cambronne; Martha L. Dorris; Justine W. Debelius; Christian L. Lauber; Gail Ackermann; Yoshiki Vazquez Baeza; Tejpal Gill; Rob Knight; Robert A. Colbert; Joel D. Taurog; Russell N. Van Gelder; James T. Rosenbaum

The HLA-B27 gene is a major risk factor for clinical diseases including ankylosing spondylitis, acute anterior uveitis, reactive arthritis, and psoriatic arthritis, but its mechanism of risk enhancement is not completely understood. The gut microbiome has recently been shown to influence several HLA-linked diseases. However, the role of HLA-B27 in shaping the gut microbiome has not been previously investigated. In this study, we characterize the differences in the gut microbiota mediated by the presence of the HLA-B27 gene. We identified differences in the cecal microbiota of Lewis rats transgenic for HLA-B27 and human β2-microglobulin (hβ2m), compared with wild-type Lewis rats, using biome representational in situ karyotyping (BRISK) and 16S rRNA gene sequencing. 16S sequencing revealed significant differences between transgenic animals and wild type animals by principal coordinates analysis. Further analysis of the data set revealed an increase in Prevotella spp. and a decrease in Rikenellaceae relative abundance in the transgenic animals compared to the wild type animals. By BRISK analysis, species-specific differences included an increase in Bacteroides vulgatus abundance in HLA-B27/hβ2m and hβ2m compared to wild type rats. The finding that HLA-B27 is associated with altered cecal microbiota has not been shown before and can potentially provide a better understanding of the clinical diseases associated with this gene.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models

Egle Cekanaviciute; Bryan B. Yoo; Tessel F. Runia; Justine W. Debelius; Sneha Singh; Charlotte A. Nelson; Rachel Kanner; Yadira Bencosme; Yun Kyung Lee; Stephen L. Hauser; Elizabeth Crabtree-Hartman; Ilana Katz Sand; Mar Gacias; Yungjiao Zhu; Patrizia Casaccia; Bruce Cree; Rob Knight; Sarkis K. Mazmanian; Sergio E. Baranzini

Significance We have experimentally investigated the immunoregulatory effects of human gut microbiota in multiple sclerosis (MS). We have identified specific bacteria that are associated with MS and demonstrated that these bacteria regulate T lymphocyte-mediated adaptive immune responses and contribute to the proinflammatory environment in vitro and in vivo. Thus, our results expand the knowledge of the microbial regulation of immunity and may provide a basis for the development of microbiome-based therapeutics in autoimmune diseases. The gut microbiota regulates T cell functions throughout the body. We hypothesized that intestinal bacteria impact the pathogenesis of multiple sclerosis (MS), an autoimmune disorder of the CNS and thus analyzed the microbiomes of 71 MS patients not undergoing treatment and 71 healthy controls. Although no major shifts in microbial community structure were found, we identified specific bacterial taxa that were significantly associated with MS. Akkermansia muciniphila and Acinetobacter calcoaceticus, both increased in MS patients, induced proinflammatory responses in human peripheral blood mononuclear cells and in monocolonized mice. In contrast, Parabacteroides distasonis, which was reduced in MS patients, stimulated antiinflammatory IL-10–expressing human CD4+CD25+ T cells and IL-10+FoxP3+ Tregs in mice. Finally, microbiota transplants from MS patients into germ-free mice resulted in more severe symptoms of experimental autoimmune encephalomyelitis and reduced proportions of IL-10+ Tregs compared with mice “humanized” with microbiota from healthy controls. This study identifies specific human gut bacteria that regulate adaptive autoimmune responses, suggesting therapeutic targeting of the microbiota as a treatment for MS.


Movement Disorders | 2017

Parkinson's disease and Parkinson's disease medications have distinct signatures of the gut microbiome

Erin M. Hill-Burns; Justine W. Debelius; James T. Morton; William T. Wissemann; Matthew R. Lewis; Zachary D. Wallen; Shyamal D. Peddada; Stewart A. Factor; Eric Molho; Cyrus P. Zabetian; Rob Knight; Haydeh Payami

There is mounting evidence for a connection between the gut and Parkinsons disease (PD). Dysbiosis of gut microbiota could explain several features of PD.


Fems Microbiology Reviews | 2015

Microbial endocrinology: the interplay between the microbiota and the endocrine system

Hadar Neuman; Justine W. Debelius; Rob Knight; Omry Koren

The new field of microbiome research studies the microbes within multicellular hosts and the many effects of these microbes on the hosts health and well-being. We now know that microbes influence metabolism, immunity and even behavior. Essential questions, which are just starting to be answered, are what are the mechanisms by which these bacteria affect specific host characteristics. One important but understudied mechanism appears to involve hormones. Although the precise pathways of microbiota-hormonal signaling have not yet been deciphered, specific changes in hormone levels correlate with the presence of the gut microbiota. The microbiota produces and secretes hormones, responds to host hormones and regulates expression levels of host hormones. Here, we summarize the links between the endocrine system and the gut microbiota. We categorize these interactions by the different functions of the hormones, including those affecting behavior, sexual attraction, appetite and metabolism, gender and immunity. Future research in this area will reveal additional connections, and elucidate the pathways and consequences of bacterial interactions with the host endocrine system.


Genome Biology | 2016

Heritable components of the human fecal microbiome are associated with visceral fat

Michelle Beaumont; Julia K. Goodrich; Matthew A. Jackson; Idil Yet; Emily R. Davenport; Sara Vieira-Silva; Justine W. Debelius; Tess Pallister; Massimo Mangino; Jeroen Raes; Rob Knight; Andrew G. Clark; Ruth E. Ley; Tim D. Spector; Jordana T. Bell

BackgroundVariation in the human fecal microbiota has previously been associated with body mass index (BMI). Although obesity is a global health burden, the accumulation of abdominal visceral fat is the specific cardio-metabolic disease risk factor. Here, we explore links between the fecal microbiota and abdominal adiposity using body composition as measured by dual-energy X-ray absorptiometry in a large sample of twins from the TwinsUK cohort, comparing fecal 16S rRNA diversity profiles with six adiposity measures.ResultsWe profile six adiposity measures in 3666 twins and estimate their heritability, finding novel evidence for strong genetic effects underlying visceral fat and android/gynoid ratio. We confirm the association of lower diversity of the fecal microbiome with obesity and adiposity measures, and then compare the association between fecal microbial composition and the adiposity phenotypes in a discovery subsample of twins. We identify associations between the relative abundances of fecal microbial operational taxonomic units (OTUs) and abdominal adiposity measures. Most of these results involve visceral fat associations, with the strongest associations between visceral fat and Oscillospira members. Using BMI as a surrogate phenotype, we pursue replication in independent samples from three population-based cohorts including American Gut, Flemish Gut Flora Project and the extended TwinsUK cohort. Meta-analyses across the replication samples indicate that 8 OTUs replicate at a stringent threshold across all cohorts, while 49 OTUs achieve nominal significance in at least one replication sample. Heritability analysis of the adiposity-associated microbial OTUs prompted us to assess host genetic-microbe interactions at obesity-associated human candidate loci. We observe significant associations of adiposity-OTU abundances with host genetic variants in the FHIT, TDRG1 and ELAVL4 genes, suggesting a potential role for host genes to mediate the link between the fecal microbiome and obesity.ConclusionsOur results provide novel insights into the role of the fecal microbiota in cardio-metabolic disease with clear potential for prevention and novel therapies.


Annual Review of Genomics and Human Genetics | 2017

The Microbiome and Human Biology

Rob Knight; Chris Callewaert; Clarisse Marotz; Embriette R. Hyde; Justine W. Debelius; Daniel McDonald; Mitchell L. Sogin

Over the past few years, microbiome research has dramatically reshaped our understanding of human biology. New insights range from an enhanced understanding of how microbes mediate digestion and disease processes (e.g., in inflammatory bowel disease) to surprising associations with Parkinsons disease, autism, and depression. In this review, we describe how new generations of sequencing technology, analytical advances coupled to new software capabilities, and the integration of animal model data have led to these new discoveries. We also discuss the prospects for integrating studies of the microbiome, metabolome, and immune system, with the goal of elucidating mechanisms that govern their interactions. This systems-level understanding will change how we think about ourselves as organisms.


Genome Biology | 2016

Tiny microbes, enormous impacts: what matters in gut microbiome studies?

Justine W. Debelius; Se Jin Song; Yoshiki Vázquez-Baeza; Zhenjiang Zech Xu; Antonio González; Rob Knight

Many factors affect the microbiomes of humans, mice, and other mammals, but substantial challenges remain in determining which of these factors are of practical importance. Considering the relative effect sizes of both biological and technical covariates can help improve study design and the quality of biological conclusions. Care must be taken to avoid technical bias that can lead to incorrect biological conclusions. The presentation of quantitative effect sizes in addition to P values will improve our ability to perform meta-analysis and to evaluate potentially relevant biological effects. A better consideration of effect size and statistical power will lead to more robust biological conclusions in microbiome studies.

Collaboration


Dive into the Justine W. Debelius's collaboration.

Top Co-Authors

Avatar

Rob Knight

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarkis K. Mazmanian

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Bruce Cree

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrizia Casaccia

City University of New York

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge