Assessing the human gut microbiota in metabolic diseases

Fredrik Karlsson, Valentina Tremaroli, Jens Nielsen, Fredrik Bäckhed, Fredrik Karlsson, Valentina Tremaroli, Jens Nielsen, Fredrik Bäckhed

Abstract

Recent findings have demonstrated that the gut microbiome complements our human genome with at least 100-fold more genes. In contrast to our Homo sapiens-derived genes, the microbiome is much more plastic, and its composition changes with age and diet, among other factors. An altered gut microbiota has been associated with several diseases, including obesity and diabetes, but the mechanisms involved remain elusive. Here we discuss factors that affect the gut microbiome, how the gut microbiome may contribute to metabolic diseases, and how to study the gut microbiome. Next-generation sequencing and development of software packages have led to the development of large-scale sequencing efforts to catalog the human microbiome. Furthermore, the use of genetically engineered gnotobiotic mouse models may increase our understanding of mechanisms by which the gut microbiome modulates host metabolism. A combination of classical microbiology, sequencing, and animal experiments may provide further insights into how the gut microbiota affect host metabolism and physiology.

Figures

FIG. 1.
FIG. 1.
Methods for studying the microbiota. Traditionally, microbial communities have been characterized by culturing on specific plates, but this is only amenable to the culturable fraction of the members (20–50% [84,86]) and has limited resolution. Culture-independent methods based on characterization of the 16S rRNA genes have been developed and also provide information for organisms that cannot be cultured. Shotgun sequencing of the whole genome provides information about the functional and metabolic potential of the community.
FIG. 2.
FIG. 2.
In the bioinformatic pipeline for analysis of whole metagenome shotgun sequences, sequences are subjected to quality control by removing uncertain base calls and contaminant sequences. Alignment of sequence reads to reference genomes is used for calculating species abundance. De novo assembly is used to identify genes not present in public databases. Genes can be functionally annotated and mapped onto metabolic networks such as in KEGG. Abundance of genes and species are compared among groups, and associations with disease can be tested.
FIG. 3.
FIG. 3.
Germ-free mice can be used to study the effect of a gut microbiota on its host. Colonization of germ-free mice with human microbiota from different donors can test if there are functional differences between communities. Colonization of germ-free mice also allows investigation of the interaction between the microbiota and specific diets. Synthetic microbiota are defined communities with known species composition and provide a controlled environment for testing the interaction of microbes with diet and host.

References

    1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science 2009;326:1694–1697
    1. Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science 2005;307:1915–1920
    1. O’Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Rep 2006;7:688–693
    1. Qin J, Li R, Raes J, et al. MetaHIT Consortium A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010;464:59–65
    1. Sommer F, Bäckhed F. The gut microbiota—masters of host development and physiology. Nat Rev Microbiol 2013;11:227–238
    1. Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature 2012;489:242–249
    1. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 2005;102:11070–11075
    1. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science 2005;308:1635–1638
    1. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222–227
    1. Reyes A, Haynes M, Hanson N, et al. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 2010;466:334–338
    1. Wall R, Ross RP, Ryan CA, et al. Role of gut microbiota in early infant development. Clin Med Pediatr 2009;3:45–54
    1. Koenig JE, Spor A, Scalfone N, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci U S A 2011;108(Suppl. 1):4578–4585
    1. Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012;488:178–184
    1. Ley RE, Hamady M, Lozupone C, et al. Evolution of mammals and their gut microbes. Science 2008;320:1647–1651
    1. Muegge BD, Kuczynski J, Knights D, et al. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science 2011;332:970–974
    1. Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013;19:576–585
    1. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–108
    1. De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A 2010;107:14691–14696
    1. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature 2006;444:1022–1023
    1. Duncan SH, Lobley GE, Holtrop G, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes (Lond) 2008;32:1720–1724
    1. Schwiertz A, Taras D, Schäfer K, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010;18:190–195
    1. Kalliomäki M, Collado MC, Salminen S, Isolauri E. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr 2008;87:534–538
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature 2009;457:480–484
    1. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006;444:1027–1031
    1. Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008;3:213–223
    1. Bäckhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004;101:15718–15723
    1. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 2007;104:979–984
    1. Larsen N, Vogensen FK, van den Berg FW, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010;5:e9085.
    1. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56:1761–1772
    1. Creely SJ, McTernan PG, Kusminski CM, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab 2007;292:E740–E747
    1. Caesar R, Reigstad CS, Bäckhed HK, et al. Gut-derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut 2012;61:1701–1707
    1. Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012;490:55–60
    1. Karlsson FH, Tremaroli V, Nookaew I, et al. Gut metagenome in European women with normal, impaired and diabeteic glucose control. Nature 2013;498:99–103
    1. Sjöström L, Lindroos AK, Peltonen M, et al. Swedish Obese Subjects Study Scientific Group Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med 2004;351:2683–2693
    1. Sjöström L, Narbro K, Sjöström CD, et al. Swedish Obese Subjects Study Effects of bariatric surgery on mortality in Swedish obese subjects. N Engl J Med 2007;357:741–752
    1. Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes 2010;59:3049–3057
    1. Zhang H, DiBaise JK, Zuccolo A, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A 2009;106:2365–2370
    1. Li JV, Ashrafian H, Bueter M, et al. Metabolic surgery profoundly influences gut microbial-host metabolic cross-talk. Gut 2011;60:1214–1223
    1. Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med 2013;5:178ra141
    1. Vrieze A, Van Nood E, Holleman F, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012;143:913–916.e7
    1. Harrison LC, Honeyman MC, Morahan G, et al. Type 1 diabetes: lessons for other autoimmune diseases? J Autoimmun 2008;31:306–310
    1. Brown CT, Davis-Richardson AG, Giongo A, et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 2011;6:e25792.
    1. Kriegel MA, Sefik E, Hill JA, Wu HJ, Benoist C, Mathis D. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci U S A 2011;108:11548–11553
    1. Oresic M, Simell S, Sysi-Aho M, et al. Dysregulation of lipid and amino acid metabolism precedes islet autoimmunity in children who later progress to type 1 diabetes. J Exp Med 2008;205:2975–2984
    1. Koren O, Spor A, Felin J, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci U S A 2011;108(Suppl. 1):4592–4598
    1. Karlsson FH, Fåk F, Nookaew I, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun 2012;3:1245.
    1. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011;472:57–63
    1. Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013;368:1575–1584
    1. Woese CR. Bacterial evolution. Microbiol Rev 1987;51:221–271
    1. Clarridge JE., 3rd Impact of 16S rRNA gene sequence analysis for identification of bacteria on clinical microbiology and infectious diseases. Clin Microbiol Rev 2004;17:840–862
    1. Tzeneva VA, Heilig HG, van Vliet WA, Akkermans AD, de Vos WM, Smidt H. 16S rRNA targeted DGGE fingerprinting of microbial communities. Methods Mol Biol 2008;410:335–349
    1. Li F, Hullar MA, Lampe JW. Optimization of terminal restriction fragment polymorphism (TRFLP) analysis of human gut microbiota. J Microbiol Methods 2007;68:303–311
    1. Rajilić-Stojanović M, Heilig HG, Molenaar D, et al. Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults. Environ Microbiol 2009;11:1736–1751
    1. Sogin ML, Morrison HG, Huber JA, et al. Microbial diversity in the deep sea and the underexplored “rare biosphere.” Proc Natl Acad Sci U S A 2006;103:12115–12120
    1. Andersson AF, Lindberg M, Jakobsson H, Bäckhed F, Nyrén P, Engstrand L. Comparative analysis of human gut microbiota by barcoded pyrosequencing. PLoS ONE 2008;3:e2836.
    1. Caporaso JG, Lauber CL, Walters WA, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A 2011;108(Suppl. 1):4516–4522
    1. Liu Z, DeSantis TZ, Andersen GL, Knight R. Accurate taxonomy assignments from 16S rRNA sequences produced by highly parallel pyrosequencers. Nucleic Acids Res 2008;36:e120.
    1. Liu Z, Lozupone C, Hamady M, Bushman FD, Knight R. Short pyrosequencing reads suffice for accurate microbial community analysis. Nucleic Acids Res 2007;35:e120.
    1. Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods 2010;7:335–336
    1. Schloss PD, Westcott SL, Ryabin T, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 2009;75:7537–7541
    1. DeSantis TZ, Hugenholtz P, Larsen N, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol 2006;72:5069–5072
    1. Pruesse E, Quast C, Knittel K, et al. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res 2007;35:7188–7196
    1. Patil KR, Haider P, Pope PB, et al. Taxonomic metagenome sequence assignment with structured output models. Nat Methods 2011;8:191–192
    1. Huson DH, Auch AF, Qi J, Schuster SC. MEGAN analysis of metagenomic data. Genome Res 2007;17:377–386
    1. Segata N, Waldron L, Ballarini A, Narasimhan V, Jousson O, Huttenhower C. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods 2012;9:811–814
    1. Martin J, Sykes S, Young S, et al. Optimizing read mapping to reference genomes to determine composition and species prevalence in microbial communities. PLoS ONE 2012;7:e36427.
    1. Li R, Zhu H, Ruan J, et al. De novo assembly of human genomes with massively parallel short read sequencing. Genome Res 2010;20:265–272
    1. Zerbino DR, Birney E. Velvet: algorithms for de novo short read assembly using de Bruijn graphs. Genome Res 2008;18:821–829
    1. Namiki T, Hachiya T, Tanaka H, Sakakibara Y. MetaVelvet: an extension of Velvet assembler to de novo metagenome assembly from short sequence reads. Nucleic Acids Res 2012;40:e155.
    1. Kultima JR, Sunagawa S, Li J, et al. MOCAT: a metagenomics assembly and gene prediction toolkit. PLoS ONE 2012;7:e47656.
    1. Kanehisa M, Goto S, Furumichi M, Tanabe M, Hirakawa M. KEGG for representation and analysis of molecular networks involving diseases and drugs. Nucleic Acids Res 2010;38(Database issue):D355–D360
    1. Tatusov RL, Fedorova ND, Jackson JD, et al. The COG database: an updated version includes eukaryotes. BMC Bioinformatics 2003;4:41.
    1. Punta M, Coggill PC, Eberhardt RY, et al. The Pfam protein families database. Nucleic Acids Res 2012;40(Database issue):D290–D301
    1. Arumugam M, Harrington ED, Foerstner KU, Raes J, Bork P. SmashCommunity: a metagenomic annotation and analysis tool. Bioinformatics 2010;26:2977–2978
    1. Abubucker S, Segata N, Goll J, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLOS Comput Biol 2012;8:e1002358.
    1. Sanli K, Karlsson FH, Nookaew I, Nielsen J. FANTOM: Functional and taxonomic analysis of metagenomes. BMC Bioinformatics 2013;14:38.
    1. Seshadri R, Kravitz SA, Smarr L, Gilna P, Frazier M. CAMERA: a community resource for metagenomics. PLoS Biol 2007;5:e75.
    1. Markowitz VM, Ivanova NN, Szeto E, et al. IMG/M: a data management and analysis system for metagenomes. Nucleic Acids Res 2008;36:D534–D538
    1. Meyer F, Paarmann D, D’Souza M, et al. The metagenomics RAST server - a public resource for the automatic phylogenetic and functional analysis of metagenomes. BMC Bioinformatics 2008;9:386.
    1. Mardinoglu A, Gatto F, Nielsen J. Genome-scale modeling of human metabolism—a systems biology approach. Biotechnol J 24 April 2013 [Epub ahead of print]
    1. Bordbar A, Lewis NE, Schellenberger J, Palsson BO, Jamshidi N. Insight into human alveolar macrophage and M. tuberculosis interactions via metabolic reconstructions. Mol Syst Biol 2010;6:422.
    1. Karlsson FH, Nookaew I, Petranovic D, Nielsen J. Prospects for systems biology and modeling of the gut microbiome. Trends Biotechnol 2011;29:251–258
    1. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature 2011;474:327–336
    1. Goodman AL, Kallstrom G, Faith JJ, et al. Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice. Proc Natl Acad Sci U S A 2011;108:6252–6257
    1. Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 2012;150:470–480
    1. Zoetendal EG, Vaughan EE, de Vos WM. A microbial world within us. Mol Microbiol 2006;59:1639–1650

Source: PubMed

3
Suscribir