Human genome-microbiome interaction: metagenomics frontiers for the aetiopathology of autoimmune diseases

Similar documents
Mini-Symposium MICROBIOTA. Free. Meet the speaker. 14. November :30 19:00 Bohnenkamp Haus. Everyone welcome. Sponsored by:

METAGENOMICS. Aina Maria Mas Calafell Genomics

METABOLOMICS: A Revolution in Omics

Companion Diagnostics in Autoimmune Disorders: Improving Outcomes Through Personalized Medicine

Topic: Genome-Environment Interactions in Inflammatory Skin Disease

Human Microbiome Project: First Map of the World Within Us. Hsin-Jung Joyce Wu "Microbiota and man: the story about us

The Human Microbiome: A New Paradigm? John Huss Department of Philosophy The University of Akron

PROVIDING ADAPTED TECHNOLOGICAL INNOVATION SOLUTIONS

Microbiota and What the Clinical Gastroenterologist Needs to Know

CONSIDERING THE MICROBIOME AS PART OF FUTURE MEDICINE AND NUTRITION STRATEGIES: Challenges and proposed answers

Genetics and Inflammatory Bowel Disease

THE HUMAN MICROBIOME: RECENT DISCOVERIES AND APPLICATIONS TO MEDICINE

Jianguo (Jeff) Xia, Assistant Professor McGill University, Quebec Canada June 26, 2017

- OMICS IN PERSONALISED MEDICINE

Next G eneration Generation Microbial Microbial Genomics : The H uman Human Microbiome P roject Project George Weinstock

Human-microbe mutualism: stability and resilience in health and disease

Lecture 8: Predicting metagenomic composition from 16S survey data

Our other genome - the foundation for new approaches in medicine. S. Dusko Ehrlich, INRA, MetaHIT coordinator Paris, June 4, 2010

EPIB 668 Genetic association studies. Aurélie LABBE - Winter 2011

Lecture 8: Predicting and analyzing metagenomic composition from 16S survey data

Promise and Pitfalls of MicrobiomeModulating Methods for Inflammatory

Shantelle Claassen-Weitz Division of Medical Microbiology Department of Pathology

2. Materials and Methods

Functional profiling of metagenomic short reads: How complex are complex microbial communities?

The Five Key Elements of a Successful Metabolomics Study

Microbial Metabolism Systems Microbiology

Our website:

Carl Woese. Used 16S rrna to developed a method to Identify any bacterium, and discovered a novel domain of life

An introduction into 16S rrna gene sequencing analysis. Stefan Boers

Conducting Microbiome study, a How to guide

Exploring the Genetic Basis of Congenital Heart Defects

UNITING AGRICULTURE AND MEDICINE. Director, Nebraska Food for Health Center

Statistical Methods for Network Analysis of Biological Data

Discovery of Bacterial Effectors in the Human Microbiome and Isolation of a GPCR G2A/GPR132 Agonist

Frumkin, 2e Part 1: Methods and Paradigms. Chapter 6: Genetics and Environmental Health

Sequencing the Astronaut Microbiome. Hernán Lorenzi PhD. July 17 th, 2014 The J. Craig Venter Institute

Course Descriptions. BIOL: Biology. MICB: Microbiology. [1]

Metagenomics of the Human Intestinal Tract

21.5 The "Omics" Revolution Has Created a New Era of Biological Research

Introducing a Highly Integrated Approach to Translational Research: Biomarker Data Management, Data Integration, and Collaboration

GREG GIBSON SPENCER V. MUSE

HLA and other tales: The different perspectives of Celiac Disease Gutierrez Achury, Henry Javier

Metagenomics Computational Genomics

Genetics and Bioinformatics

Customized Phage Therapies To Eradicate Harmful Bacteria In Chronic Diseases. Europe Microbiome Congress London, 14 Nov., 2018

So, just exactly when will genetics revolutionise medicine? - Part 1

Pharmacogenetics of Drug-Induced Side Effects

Carl Woese. Used 16S rrna to develop a method to Identify any bacterium, and discovered a novel domain of life

A new vision for AMR innovation to support medical care

Infectious Disease Omics

Microbiota: Agents for Health and Disease Dr. B. Brett Finlay

Cross Haplotype Sharing Statistic: Haplotype length based method for whole genome association testing

BTRY 7210: Topics in Quantitative Genomics and Genetics

Personal Genomics Platform White Paper Last Updated November 15, Executive Summary

From Genotype to Phenotype

Human Microbiome Project: A Community Resource. Lita M. Proctor, Ph.D. Coordinator, Human Microbiome Project NHGRI/NIH

Computational Workflows for Genome-Wide Association Study: I

Capabilities & Services

Course Agenda. Day One

Genome-Wide Association Studies. Ryan Collins, Gerissa Fowler, Sean Gamberg, Josselyn Hudasek & Victoria Mackey

Infectious Diseases Institute Faculty Hires See below for research interests, areas of expertise and contact information

ngs metagenomics target variation amplicon bioinformatics diagnostics dna trio indel high-throughput gene structural variation ChIP-seq mendelian

Introduction to Bioinformatics

Microbiomics I August 24th, Introduction. Robert Kraaij, PhD Erasmus MC, Internal Medicine

This place covers: Methods or systems for genetic or protein-related data processing in computational molecular biology.

From genome-wide association studies to disease relationships. Liqing Zhang Department of Computer Science Virginia Tech

SAS Microarray Solution for the Analysis of Microarray Data. Susanne Schwenke, Schering AG Dr. Richardus Vonk, Schering AG

Machine learning applications in genomics: practical issues & challenges. Yuzhen Ye School of Informatics and Computing, Indiana University

ST 591: Introduction to Quantitative Genomics Syllabus

Biomedical Big Data and Precision Medicine

Next Generation Sequencing

ROAD TO STATISTICAL BIOINFORMATICS CHALLENGE 1: MULTIPLE-COMPARISONS ISSUE

Microbially Mediated Plant Salt Tolerance and Microbiome based Solutions for Saline Agriculture

Ecological genomics and molecular adaptation: state of the Union and some research goals for the near future.

Clarity Genomics. Company Profile

MICROBIOTA: FROM MICROBIOME TO FUNCTIONS. Atelier «Outils et modèles d étude du microbiote Intestinal» LYONBIOPÔLE/CENS 17 Novembere 2014

Multi-SNP Models for Fine-Mapping Studies: Application to an. Kallikrein Region and Prostate Cancer

Session 3 Lecture 1 Dynamics of the GIT Microbiome: Microbial Darwinism

BTRY 7210: Topics in Quantitative Genomics and Genetics

leading the way in research & development

ICH Topic E16 Genomic Biomarkers Related to Drug Response: Context, Structure and Format of Qualification Submissions. Step 3

Introducing QIAseq. Accelerate your NGS performance through Sample to Insight solutions. Sample to Insight

The Human Microbiome Project : Implications for Probiotics

DNA METHYLATION RESEARCH TOOLS

Methods for comparing multiple microbial communities. james robert white, October 1 st, 2007

elin grundberg JULY 2018 JK - PRESENTATION- 11 MINS [MIXED RESPONDENTS] [Other comments:]

1 Biomarkers COPYRIGHTED MATERIAL. and bioinformatics. 1.1 Bioinformatics, translational research and personalized medicine

AIT - Austrian Institute of Technology

Introducing the Department of Life Sciences

May Microbiome Modulation. Assaf Oron, CBO Biomed Conference Tel Aviv, May 2017

Pharmacogenetics: A SNPshot of the Future. Ani Khondkaryan Genomics, Bioinformatics, and Medicine Spring 2001

Food Safety (Bio-)Informatics

Etienne Ruppé AP HP, Hôpital Bichat Claude Bernard UMR 1137 IAME

Grand Challenges in Computational Biology

Deciphering the Genes for Resilience

Predicting prokaryotic incubation times from genomic features Maeva Fincker - Final report

Microbiota Cutaneo, Salute e Bellezza

Axiom Biobank Genotyping Solution

Precision Medicine in Sepsis

Linking Genetic Variation to Important Phenotypes: SNPs, CNVs, GWAS, and eqtls

Transcription:

REVIEW Gundogdu and Nalbantoglu, Microbial Genomics 2017;3 DOI 10.1099/mgen.0.000112 Human genome-microbiome interaction: metagenomics frontiers for the aetiopathology of autoimmune diseases Aycan Gundogdu 1,2, * and Ufuk Nalbantoglu 2,3 Abstract A short while ago, the human genome and microbiome were analysed simultaneously for the first time as a multi-omic approach. The analyses of heterogeneous population cohorts showed that microbiome components were associated with human genome variations. In-depth analysis of these results reveals that the majority of those relationships are between immune pathways and autoimmune disease-associated microbiome components. Thus, it can be hypothesized that autoimmunity may be associated with homeostatic disequilibrium of the human-microbiome interactome. Further analysis of human genome human microbiome relationships in disease contexts with tailored systems biology approaches may yield insights into disease pathogenesis and prognosis. INTRODUCTION Two ground-breaking projects in the early genomic era changed the landscape of health sciences: the Human Genome Project (1990 2003) [1 3] and the Human Microbiome Project (2008 2012) [4, 5]. The early promise of the Human Genome Project was to be able to annotate the genetic variations associated with human diseases, based on comparisons with the published reference genomes. The scientific community has gradually diverged from that view given the findings of genome-wide association studies (GWAS), in which genetic variations have been investigated in large cohorts [6]. Genetic variations, even when considered in epistatic, multi-gene models, are unable to account for complex diseases. Indeed, an extensive GWAS, which employed relatively large cohorts, could explain no more than 3 % of metabolic disease cases using predictive models of 30 gene loci that had been inferred to be associated with the corresponding disease [7]. The new perspective on chronic diseases, with the exception of Mendelian diseases with relatively large effect sizes, is that many are driven by complex and systemslevel disorders. Accordingly, multiple genes, as well as non-genetic or non-epigenetic environmental factors, play crucial roles [8]. As certain complex diseases emerge as chronic impairments, where impairments of the immune system and the microbiome converge to create homeostatic imbalance, hologenomic interactions must be investigated. MICROBIOME AS QUANTITATIVE TRAIT Pioneering studies that jointly considered the human genome and microbiome reported that the presence or abundance of certain microbiome components is associated with the structural variations in the human genome [9, 10]. For instance, Blekhman et al. gleaned human genome contamination in the Human Microbiome Project data and detected specific host genetic variations in the context of each individual microbiome using bioinformatics approaches [9]. Their report was the first GWAS on human genome microbiome associations. Strikingly, they found that those variations associated with changes in the microbiome were mostly observed in the loci previously associated with autoimmune diseases. It has been postulated that autoimmune diseases develop after exposure to environmental triggers in patients with a genetic predisposition. Immune system pathway genes, such as HLA,CRF4, PTPN22, TBX21, STAT4, IRF4,IRF5, CD247, BANK1, BLK, IRAK1, FOXP3 and TNFAIP3 are commonly associated with several autoimmune diseases (e.g. rheumatoid arthritis, Coeliac disease, type I diabetes, psoriasis, Crohn s disease and systemic sclerosis) [11, 12]. Many autoimmune diseases, which involve systemic disorders, could be classified as pleiotropic disorders. Moreover, recent host genome microbiome association studies revealed that these genes were among those associated with microbiome composition. According to a study conducted by Volkmann et al., which considered the gut microbiota of 17 systemic sclerosis Received 27 October 2016; Accepted 31 March 2017 Author affiliations: 1 Erciyes University School of Medicine, Turkey; 2 Genome and Stem Cell Center (GenKok), Erciyes University, Turkey; 3 Department of Computer Engineering, Erciyes University, Turkey. *Correspondence: Aycan Gundogdu, agundogdu@erciyes.edu.tr Keywords: genome-microbiome interaction; metagenomics; autoimmune disease. Abbreviations: GWAS, genome-wide association studies; MGWAS, metagenome-wide association studies. Data statement: All supporting data, code and protocols have been provided within the article or through supplementary data files. 000112 ã 2017 The Authors This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. IP: 37.44.197.233 1

patients and an equal number of healthy controls in order to reveal potential pathobionts, the differentiation of the gut microbiota associated with the disease shows significant similarities with the gut dysbiosis of Crohn s disease [13]. This observation implies that autoimmune diseases may share characteristic host-microbiome variations. Furthermore, the pleiotropic nature of autoimmune diseases (i.e. a single variation may be associated with multiple phenotypes) supports the view that the autoimmunity reactome has certain properties which should be investigated at the systems level, in a multi-omic investigation of human genome microbiome metagenomes. THE HOMEOSTASIS OF A SUPERORGANISM The human microbiome corresponds to a complicated ecosystem; it is a large population that forms a symbiotic super-organism in combination with the human organism. As there are 10-fold more microbial cells than Homo sapiens cells in the human body, the human microbiome, which contains millions of genes, has a far greater proteome-coding and metabolome-producing potential than the human genome, which harbours only around 20 000 genes [14]. Environmental factors shape the microbiome composition, including the mode of delivery at birth and the inherited maternal microbiome [15]; diet [16, 17]; living space [18]; social interactions [19]; and exposure to xenobiotics, pathogens and parasitic organisms [20, 21]. In addition, the interaction of the gut microbiome with its host directly shapes the composition [22]. The currently illuminated part of this interaction system mainly corresponds to the immunity metabolisms. Recently revealed interactions between the microbiome and the immune system show that they exist in a delicate homeostasis. Tipping of the equilibrium by perturbations stemming from either, or both, of the symbiotic partners might result in disturbances of processes related to inflammation, autoimmunity, metabolism, neurodegeneration and the development and progression of cancer [23 25]. Gut microbiome dysbiosis might lead to a decrease in the activity of critical members or metabolic processes, resulting in autoimmune disease (with the microbiome being the primary cause of pathogenesis). Conversely, the primary driver of the aetiopathology of immune disorders may be the inability to distinguish between commensal and pathogenic components of the microbiome, induced by environmental triggers in genetically predisposed individuals (host genetics are the primary cause of pathogenesis). Although the pathogenesis of a large proportion of autoimmune diseases is currently unclear, the involvement of the human microbiome in both scenarios above raises an important question: can the gut microbiome be used as a diagnostic biomarker or a therapeutic target for the prevention and treatment of autoimmune diseases? The human genome is static and, except for specific modifications (e.g. genome editing), cannot be manipulated over the lifetime of an individual. The human microbiome, on the other hand, is the plastic other genome of the human super-organism that could be shaped or even reassembled by probiotics, IMPACT STATEMENT Recent studies on human genome microbiome interactions (metagenome-wide association studies, MGWAS) showed that the presence or abundance of certain microbiome components is associated with structural variations in the human genome. Here, with a focused look, we relate these recent findings to a relevant human disease class. A key observation in these studies is that differences in the microbiome composition are mostly associated with variations in human genome that have previously been associated with autoimmune diseases. This article aims to draw attention to the necessity for further studies that have been carefully designed to reveal the diseases that are related to microbiome dysbiosis, as well as the factors contributing to dysbiosis. Metagenome-wide association studies may enable the development of microbiome-based diagnostics and therapies. antibiotics, diet, vaccination and transplantation techniques. This potential makes prospective microbiome-targeted therapies attractive. The promise of this approach has been observed when the manipulation of commensal bacteria caused remission of rheumatoid arthritis in mice [26]. Similarly, a healthy microbiome prevented multiple sclerosis in mice, while germ-free mice in the same conditions developed the disease [27]. Further studies should be conducted to reveal the nature of the disease-related dysbiosis and the factors affecting it, in order to enable the development of diagnostics and therapies based on the microbiome. A COMPLEX INTERACTOME REQUIRES SOPHISTICATED ANALYSIS While the findings of the microbiome-gwas studies shed light on the association between human immunogenetics and the human microbiome, they are early discovery attempts in the genome microbiome multi-omic field, which requires further development. The current paradigm is built on a naive approach, which models the microbiome as a quantitative trait. This is mainly through the quantification of the microbial community via a or b diversity metrics or evaluation of the relative abundance of an operational taxonomic unit. However, in the context of the novel idea of the super-organism, or the second genome, the microbiome should be treated as an extension of the human genome rather than a quantitative trait. Previous 16S rrna gene sequencing studies were designed to taxonomically profile the microbiome. In order to explore its functional diversity and construct metabolic models within a microbiome, a shotgun metagenomics approach is required. Sequencing the genetic content in this manner would allow functionally classified genes to be grouped and gene networks to be constructed in order to form human genome microbiome interaction networks. Early versions of the related metagenomic unit co-occurence networks, such as the metahit catalogue [28], are already available. A systems IP: 37.44.197.233 2

Dysbiotic metagenome units directly associated with the human genome Dysbiotic metagenome units indirectly associated with the human genome log10(p) 5.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0 0.5 0.0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19202122 Genetic variations associated with the disease Fig. 1. A systems biology approach to MGWAS. Fusion gene-disease interaction networks are compiled from the GWAS Catalog and the metagenomic unit (e.g. gene, ontology group, metabolic pathway or operational taxonomic unit) co-occurrence network built using metagenome data. Feature selection by machine learning wrappers suggests associations based on variations in the metagenomic units to infer direct and indirect associations between genomic variations and the microbiome in the context of the disease. biology model, which integrates metagenomic gene networks with the disease associations map in the GWAS Catalog [29], has the potential to suggest direct and indirect genetic variation microbiome associations in the context of diseases. Fig. 1 depicts a model constructed from this perspective. Moreover, hologenome theory suggests that a host co-evolves with its microbiome. From this point of view, strain-level divergences that alter the function of certain microbial species could be expected. The study of strain-level differentiation, which is also interesting in the context of diseases, can only be resolved by employing shotgun metagenomics. Recent advances in metagenomics informatics enable careful taxonomic profiling at the strain level [30 32]. Parallel to the current paradigm for microbiome analytics, GWAS that associate genomic variations with quantitative traits depend on conventional statistical approaches and single gene/single-nucleotide polymorphism -quantitative trait correlation estimations. However, the majority of diseases and other attribute phenotypes are epistatic, meaning that the overall phenotype is the net effect of multiple genes with small effect sizes. Unfortunately, the conventional GWAS approach has weak sensitivity and is able to detect only genetic variations with large effect sizes [33]. The introduction of machine learning and data mining applications, in which multiple gene variations are associated with phenotypes, would allow epistatic inferences to be made with greater statistical accuracy, according to retrospective studies [6]. By assuming that either individual microbiome elements or the community diversity are quantitative traits, the current GWAS methodology to determine genome microbiome associations ignores the potentially epistatic nature of the symbiosis. Both of the recent studies conducted by Blekhman et al. [9] and Davenport et al. [10] followed the conventional one gene-one quantitative trait GWAS approach. However, human genome microbiome relationships are thought to be both epistatic and pleiotropic [34]. On the other hand, since the sampling and sequencing of metagenomes require substantial effort and costly procedures, current cohorts are orders of magnitude smaller than the operational ranges of GWAS. Thus, the observed associations likely appeared due to their large effect sizes. Importantly, many genetic associations may be ignored by underpowered and insensitive analyses. In order to mitigate this problem, enrichment via combining associated variances on the same pathways is suggested. This approach was observed to provide further associations [9]. Nevertheless, the improved approach still follows the GWAS paradigm, in which very low P-values are needed before associations can be proposed. As an alternative, predictive models supported by machine learning concepts can be proposed to statistically quantify genome microbiome associations. Feature selection with learning machines has enabled detection of cross-validated disease biomarkers at the current metagenome-sampling cohort size [35]. Once features comprised of groups of single nucleotide polymorphisms and bags of metagenomic genes are selected, predictive models (e.g. IP: 37.44.197.233 3

Support Vector Machines [36], Random Forest [37]) can be constructed to perform cross-validation tests that quantify the significance of the associations. For the aforementioned reasons, it is indispensable to build future projections on metagenome-wide association studies (MGWAS) bioinformatics, assessing multiple gene multiple phenotype associations in a machine learning context. It is important to consider that, while very sensitive and powerful enough to infer associations from a relatively small number of instances (e.g. the current cohort size of metagenome projects), machine learning approaches are vulnerable to overfitting and suggesting false positives, especially in the context of a large number of variables, as in the case of MGWAS [38]. Therefore, careful bioinformatics strategies should be developed in order to make sensitive, yet sufficiently specific, MGWAS analyses to reveal further human genome microbiome interactions. It should be noted that the top-down approach of systems biology via omic technologies only captures snapshots of a complex living system. While this in silico support is a valuable hypothesis proposal paradigm, the results (e.g. detected biomarkers or suggested biological models) should always be subjected to in vivo validations. For example, the precise microbiome targets suggested by the multi-omic studies should be tested in knockout animal experiments, as the early phase of metagenomic drug discovery studies. Moreover, independent autoimmune disease cohorts should be enrolled for the validation of the discovered, non-invasive diagnostic biomarkers. In the future, it will be possible to conduct multi-omic studies with larger cohorts as sequencing technologies provide lower cost and higher throughput solutions. This will generate more insightful data, with enhanced generalization ability and statistical power. Given that, focussing on longitudinal study designs may provide further insights, not only because they would capture temporal changes along with disease prognosis, but also because metagenomic units associated with human genetics might become more detectable. Recent twin studies have reported that the taxa that are controlled by host genetics tend to be temporally more stable taxa that were not associated with host genetics [39]. Consideration of the temporally low-variance metagenomic units could add another dimension to the computational prediction of genome variation microbiome associations. CONCLUSION Based on the recent literature, it is plausible to infer that the associations between variations in human immune system pathway genes and certain human microbiome taxa might imply the interplay of human genome microbiome in the pathogenesis of autoimmune diseases. Thus, multi-omic studies, in which the microbiome and the human genome are sequenced jointly, involving autoimmune patients and corresponding healthy cohorts will dramatically improve our understanding of complex diseases. To this end, machine learning-based systems biology may be useful, given that the current metagenomic cohorts, ranging from a few hundred to a few thousand subjects [10], are statistically underpowered to conduct conventional GWAS, which confines the discovery of associations to only highly specific mechanisms. Funding information The authors received no specific grant from any funding agency. Conflicts of interest The authors declare that there are no conflicts of interest. Ethical statement No human/animal experiment is involved in this study. References 1. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC et al. Initial sequencing and analysis of the human genome. Nature 2001;409: 860 921. 2. Lander ES. Initial impact of the sequencing of the human genome. Nature 2011;470:187 197. 3. Venter JC, Smith HO, Adams MD. The sequence of the human genome. Clin Chem 2015;61:1207 1208. 4. Human Microbiome Project Consortium. A framework for human microbiome research. Nature 2012;486:215 221. 5. Peterson J, Garges S, Giovanni M, McInnes P, Wang L et al. The NIH Human Microbiome project. Genome Res 2009;19:2317 2323. 6. Gibson G. Hints of hidden heritability in GWAS. Nat Genet 2010;42: 558 560. 7. Lu Y, Loos RJ. Obesity genomics: assessing the transferability of susceptibility loci across diverse populations. Genome Med 2013;5: 55. 8. Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA et al. Finding the missing heritability of complex diseases. Nature 2009; 461:747 753. 9. Blekhman R, Goodrich JK, Huang K, Sun Q, Bukowski R et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol 2015;16:1 12. 10. Davenport ER. Elucidating the role of the host genome in shaping microbiome composition. Gut Microbes 2016;7:178 184. 11. Gourh P, Agarwal SK, Divecha D, Assassi S, Paz G et al. Polymorphisms in TBX21 and STAT4 increase the risk of systemic sclerosis: evidence of possible gene-gene interaction and alterations in Th1/Th2 cytokines. Arthritis Rheum 2009;60:3794 3806. 12. Radstake TR, Gorlova O, Rueda B, Martin JE, Alizadeh BZ et al. Genome-wide association study of systemic sclerosis identifies CD247 as a new susceptibility locus. Nat Genet 2010;42:426 429. 13. Volkmann E, Chang Y, Barroso N, Furst DE, Clements P et al. Systemic sclerosis is associated with a unique colonic microbial consortium. Arthritis Rheum 2016;74:151. 14. Goodacre R. Metabolomics of a superorganism. J Nutr 2007;137: S259 S266. 15. van den Abbeele P, Gerard P, Rabot S, Bruneau A, El Aidy S et al. Arabinoxylans and inulin differentially modulate the mucosal and luminal gut microbiota and mucin-degradation in humanized rats. Environ Microbiol 2011;13:2667 2680. 16. Zhang C, Zhang M, Wang S, Han R, Cao Y et al. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J 2010;4:232 241. 17. Hacquard S, Garrido-Oter R, Gonzalez A, Spaepen S, Ackermann G et al. Microbiota and host nutrition across plant and animal kingdoms. Cell Host Microbe 2015;17:603 616. 18. Lax S, Smith DP, Hampton-Marcell J, Owens SM, Handley KM et al. Longitudinal analysis of microbial interaction between humans and the indoor environment. Science 2014;345:1048 1052. 19. Tung J, Barreiro LB, Burns MB, Grenier J-C, Lynch J et al. Social networks predict gut microbiome composition in wild baboons. Elife 2015;4:e05224. IP: 37.44.197.2334

20. Maurice CF, Haiser HJ, Turnbaugh PJ. Xenobiotics shape the physiology and gene expression of the active human gut microbiome. Cell 2013;152:39 50. 21. Hoffmann C, Hill DA, Minkah N, Kirn T, Troy A et al. Communitywide response of the gut microbiota to enteropathogenic Citrobacter rodentium infection revealed by deep sequencing. Infect Immun 2009;77:4668 4678. 22. Li E, Hamm CM, Gulati AS, Sartor RB, Chen H et al. Inflammatory bowel diseases phenotype, C. difficile and NOD2 genotype are associated with shifts in human ileum associated microbial composition. PLoS One 2012;7:e26284. 23. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121 141. 24. Ehrlich SD. The human gut microbiome impacts health and disease. C R Biol 2016;339:319 323. 25. Qin J, Li Y, Cai Z, Li S, Zhu J et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012;490: 55 60. 26. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity2010;32:815 827. 27. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 2011;108:22. 28. Li J, Jia H, Cai X, Zhong H, Feng Q et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol 2014;32:834 841. 29. Welter D, Macarthur J, Morales J, Burdett T, Hall P et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res 2014;42:D1001 D1006. 30. Greenblum S, Carr R, Borenstein E. Extensive strain-level copynumber variation across human gut microbiome species. Cell 2015;160:583 594. 31. Nayfach S, Rodriguez-Mueller B, Garud N, Pollard KS. An integrated metagenomics pipeline for strain profiling reveals novel patterns of bacterial transmission and biogeography. Genome Res 2016;26:1612 1625. 32. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G et al. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods 2015;12:902 903. 33. Bush WS, Moore JH. Chapter 11: Genome-wide association studies. PLoS Comput Biol 2012;8:e1002822. 34. Benson AK. Host genetic architecture and the landscape of microbiome composition: humans weigh in. Genome Biol 2015;16:203. 35. Cai L, Wu H, Li D, Zhou K, Zou F. Type 2 diabetes biomarkers of human gut microbiota selected via iterative sure independent screening method. PLoS One 2015;10:e0140827. 36. Wang Z, Montana G. The graph-guided group lasso for genomewide association studies. In: Suykens JAK, Signoretto M and Argyriou A (editors). Regularization, Optimization, Kernels, and Support Vector Machines. CRC Press; 2014. pp. 131 157. 37. Botta V, Louppe G, Geurts P, Wehenkel L. Exploiting SNP correlations within random forest for genome-wide association studies. PLoS One 2014;9:e93379. 38. Xing EP, Jordan MI, Karp RM. Feature Selection for High- Dimensional Genomic Microarray. Data in ICML, vol. 1; 2001. pp. 601 608. 39. Goodrich JK, Davenport ER, Beaumont M, Jackson MA, Knight R et al. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 2016;19:731 743. Five reasons to publish your next article with a Microbiology Society journal 1. The Microbiology Society is a not-for-profit organization. 2. We offer fast and rigorous peer review average time to first decision is 4 6 weeks. 3. Our journals have a global readership with subscriptions held in research institutions around the world. 4. 80% of our authors rate our submission process as excellent or very good. 5. Your article will be published on an interactive journal platform with advanced metrics. Find out more and submit your article at microbiologyresearch.org. IP: 37.44.197.233 5