Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter August 29, 2013

Developments and insights into the analysis of the human microbiome

Entwicklungen und Erkenntnisse in der Analyse des humanen Mikrobioms

  • Ovidiu Rücker EMAIL logo , Alexandra Dangel and Hanns-Georg Klein
From the journal LaboratoriumsMedizin

Abstract

The intense research focused on the human microbiome during the last years has shed some light on this mostly uncharacterized part of the human body. The constantly improving sequencing technologies have additionally eased the process of collecting a large amount of genome data from metagenomics samples. Using these methods, large studies with sufficient number of subjects have started to reveal the implications of our microbiome in health and disease. Here, we present a review on the last developments of sequencing technology together with an overview on the findings in this fast-evolving field of science.

Zusammenfassung

In den letzten Jahren haben sich immer mehr wissenschaftliche Untersuchungen auf das humane Mikrobiom konzentriert, um diesen noch meist unbekannten Teil des menschlichen Körpers intensiver zu durchleuchten. Die konstante Verbesserung der verfügbaren Sequenziertechnologien hat weiterhin die Verfügbarkeit von großen Mengen an genomischen Daten deutlich erleichtert. Hiermit konnten große Studien mit einer genügend großen Anzahl an Probanden beginnen zu beschreiben, welche Auswirkungen unser Mikrobiom nicht nur in Verbindung mit Krankheiten sondern auch auf unseren gesunden Körper hat. In diesem Übersichtsartikel beschreiben wir die neuesten Entwicklungen in den Sequenziertechnologien zusammen mit einer Darstellung neuester Erkenntnisse aus diesem Forschungsbereich, welcher sich immer schneller entwickelt.

Rezensierte Publikation:

Klein H.-G.


Introduction

Studies of the human microbiome using extracted DNA can be divided in two main categories; the use of the entire DNA for sequencing is often referred to as shotgun metagenomics and offers a random representation of genomic sequences. Alternatively, one can focus on one or few marker genes and sequence only these amplified genomic regions. For this so-called targeted amplicon approach, the ribosomal small subunit RNA (16S rRNA) gene has emerged as the most used marker. This marker has the advantage of having highly variable regions interrupted by more or less conserved regions (Figure 1A), which allow the design of conserved primers. The present article is aimed at giving a short overview on recent studies on metagenomics and new developments in next-generation sequencing (NGS) technology.

Figure 1 Schematic view of the 16S rRNA gene together with an example of long amplicon sequence data achieved using the GS-FLX+ technology.(A) The schematic view of the 16S rRNA displays the variable regions (V1–9) in orange, and the bars indicate a 100-bp span. The length and approximate position of the long amplicons are indicated with bars. (B) The distribution of the achieved read lengths for each amplicon is plotted against the relative number of reads. The read length achieved using GS-FLX/Junior (indicated in light blue) is compared with the read length achieved when using the GS-FLX+ technology when sequencing two different long amplicons. The exact length of the amplicons is indicated in the graph.
Figure 1

Schematic view of the 16S rRNA gene together with an example of long amplicon sequence data achieved using the GS-FLX+ technology.

(A) The schematic view of the 16S rRNA displays the variable regions (V1–9) in orange, and the bars indicate a 100-bp span. The length and approximate position of the long amplicons are indicated with bars. (B) The distribution of the achieved read lengths for each amplicon is plotted against the relative number of reads. The read length achieved using GS-FLX/Junior (indicated in light blue) is compared with the read length achieved when using the GS-FLX+ technology when sequencing two different long amplicons. The exact length of the amplicons is indicated in the graph.

Developments in NGS methods

Before the use of NGS methods, amplicons were usually cloned and individually sequenced using the Sanger method, which can still be regarded as the gold standard. However, the development of new sequencing technologies had a big impact leading to the extensive usage of the Roche/454 and the Illumina technologies. Excellent reviews on this technological leap forward are already available [1, 2].

Pyrosequencing was the first NGS method widely adopted for metagenomics studies [3]. This method, which was developed by 454 Technologies (now part of the Roche group), offers a throughput of about 1 million reads with read lengths of 400–500 bp. A higher throughput of hundreds of millions of reads can be achieved when applying the sequencing-by-synthesis technology developed by Illumina/Solexa. This technology has often been used mainly for the advantage of high throughput in shotgun sequencing, and the limitations resulting from short read lengths of 150 bp are widely accepted [4]. Recently, Illumina has enabled paired end reads of 250 bp to be sequenced on the smaller MiSeq platform (Illumina, San Diego, USA), which now also allows the sequencing of 400- to 500-bp amplicons. Another recent development is the upgrade of the Roche/454 FLX-Titanium technology to the FLX+ technology (Branford, USA), which enables reads of more than 1 kb in length for shotgun sequencing. We have assessed the usability of this method also for long amplicon sequencing showing that long reads of over 900 bp can be achieved (Figure 1B, data not published). Using this technology, one can combine the advantages, which long reads bring about regarding bioinformatic analysis with the high throughput produced by NGS. Another sequencing technology, the Ion Torrent semiconductor technology from Life Technologies (Carlsbad, USA) has also been used for metagenomic analyses [5], but only recently, its read length increased to 400 bp. The single molecule sequencing technology developed by Pacific Biosciences (Menlo Park, USA) has been used for the identification of bacterial isolates [6] enabling sequencing reads of more than 3 kb, but its low accuracy of 85% and lower throughput are hindering its usability for metagenomic samples.

Despite its high adoption, there are some drawbacks preventing the 16S amplicon sequencing from becoming the method of choice for all metagenomic analyses. The design of primers, which are able to amplify all known species remains challenging [7], therefore, optimal primers have to be carefully chosen and tested. Mosaicism through horizontal gene transfer may not be reported by the 16S region [8], and whole-genome sequencing is still necessary in such cases. Additionally, similarity between 16S sequences of two species can be nearly 100%, resulting in misinterpretation/missing information. Shotgun sequencing can overcome these problems, and functional gene grouping can even reveal additional information [9]. Nevertheless, sequencing highly diverse samples like those of the gut microbiome, besides still being expensive, results in large amounts of data and, therefore, still cannot be the method of choice for fast routine analyses. The analysis of the huge amount of data emerging from shotgun or amplicon metagenomic studies remains challenging. More information on recent solutions for this issue can be found in the reviews of Thomas et al. and Kuczinski et al. [1, 10], which describe most of the common bioinformatic methods used to gain insight into this vast and complex amount of data.

Microbial diversity of healthy humans

The human body is the habitat of numerous microbes, which occupy different niches. Beginning with our birth, the colonization of our body starts and is subject to continuous change during a lifetime. Various factors are shaping the microbial composition, but numerous analyses have revealed the four bacterial phyla Actinobacteria, Firmicutes, Proteobacteria, and Bacteriodetes as being the most abundant [11, 12]. The variation in the microbiota compared between individuals and also within one individual is impressively large as it has been determined by large sequencing projects like the Human Microbiome Project [12] and Metagenomics of the Human Intestinal Tract (MetaHIT) [13]. Given the exponential growth of data and knowledge acquirement, which was recently achieved, only some general aspects of the healthy human microbiome can be presented in the following sections.

The gut microbiome consists of about 1014 bacterial cells, thus, being the biggest community of the human microbiome. This amount of cells, which is 10 times the number of human cells in the body, also harbors a combined gene pool, which is two orders of magnitude greater than the human gene set [12]. It is, therefore, legitimate to regard it as an additional organ with vast metabolic and biochemical abilities. The implication of these bacteria in several aspects of host biology, with effects, which are sometimes essential, are being increasingly studied and reviewed [14].

Although the initial inoculum transmitted from the mother during and after birth has a big impact on the initial microbiota of the child, recent studies have shown that its gut microbiome is no more similar to that of its mother than to that of its biological father. Moreover, the microbiomes of genetically unrelated but cohabiting mothers and fathers were significantly more similar to one another than to members of different families, which indicates that the constant environmental exposure is considerably shaping the gut community [15].

It is not surprising that most of the gut microbiota are anaerobic bacteria, most of which are belonging to the phyla Firmicutes and Bacteroidetes. The study of samples from different locations all over the world lead to the discovery of three enterotypes, but the factors driving the emergence of these different clusters are still unknown [16]. The approximately 500–1000 species inhabit niches shaped by different gradients found in the gastrointestinal tract, starting with the challenging stomach habitat exhibiting the lowest and ending with the colon having the highest abundance and diversity. Metabolically, some species facilitate the uptake of otherwise indigestible polysaccharides and produce essential vitamins. Additionally, they are required for the development and differentiation of the host’s intestinal epithelium [14]. Unlike the microbial taxa divergence, microbiome metabolic pathways are ubiquitous among individuals and body habitats. The most abundant among them are part of the essential pathways for host-associated microbial life, like ATP synthesis and the ribosome machinery. Only a few pathways are highly variable among subjects indicating a high degree of host-microbe and microbe-microbe interactions. Nevertheless, between 78% and 86% of genes annotated from assemblies could not be assigned a metabolic function and probably encode much of the uncharacterized metabolism of the metagenome [12]. Thus, many of these newly described genes and pathways remain to be explored in future metatranscriptomic studies.

The skin also harbors a very diverse microbiome, given the fact that as an ecosystem, it provides very variable niches within short distances. The characterization of 20 distinct sites revealed 19 bacterial phyla being present, with different species dominating the distinct sites [17]. For example, propionibacteria species and staphylococci species predominated in sebaceous sites, while corynebacteria species predominated in most sites, although staphylococci species were also represented. Recent investigations of the skin have also revealed the wide dispersion of fungal communities, which have not been described yet and, therefore, showed that microbial diversity is not limited to bacteria. Interestingly, most of the sites were dominated by fungi of the genus Malassezia, while three foot sites showed high fungal diversity [18]. The skin protects humans from invasion by pathogenic microorganisms, and therefore, such characterizations are of high interest and might provide insight into the delicate balance between skin health and disease.

The human microbiome consists of, and is also influenced by, viruses, most of them being bacteriophages but comprising also a substantial number of eukaryotic viruses. While lacking conserved genes, which could be used for the classification of all viruses, insights in the human virome were facilitated by the development of newer high-throughput sequencing technologies. Among other important discoveries, such studies could reveal that bacteriophages encode antibiotic resistance genes and other genes associated with bacterial metabolic pathways, showing that viruses serve as reservoirs for mobile genetic elements in bacteria [19].

Applications in medical microbiology

Today’s medical microbiology faces important tasks in clinical diagnostics, such as characterization and treatment of pathogens, as well as in epidemiology and public health, like the detection and tracking of outbreaks and the identification of new potential sources of infection. Many of the common standard techniques have been developed decades ago and are very specialized and species specific. They are often associated with a substantial delay and require a high degree of expertise of the microbiologist. Furthermore, they strongly rely on the cultivability of microorganisms. As the majority of bacterial species on earth is difficult to grow or does not grow at all in in vitro cultures, reliable, culture-independent alternatives for pathogen identification could facilitate and fasten the diagnostic process especially for culture-negative patient samples [20]. NGS-based metagenomic methods, which are already widely used in ecological microbiology for the community analysis of various environmental samples, e.g., from marine, soil, plant, or animal-associated origin [21–23] are entering the field of medical microbiology. Owing to limited read lengths, which have been obtained in the first years of NGS analyses, poor gene annotation and overestimates of bacterial richness and abundances were some of the technical problems. But over the last years, read lengths were extensively elongated, and the impression from the data became less distorted. Thus, these analyses can serve as highly valuable tools for diagnostic community assessment and are about to revolutionize the field of medical microbiology and its different application areas [24].

Diagnostics and disease management

Generally, the detection of shifts in the community composition of the human microbiome can help clinicians in terms of diagnosis, risk assessment, prognosis, and maybe also therapy. One example is the monitoring of the oral microbiome, which is essential in maintaining health. Shifts in this personalized community allow pathogens to manifest and cause disease. Severe forms of oral disease may result in systemic disease at different body sites. Detection of such shifts using metagenomic studies can go hand in hand with conservative microbiological techniques and lead to more effective therapeutic and diagnostic techniques [25].

As discussed above, especially the human gut microbiome represents a complex and dynamic community of microorganisms, which has a big influence on human health. Changes in its composition in favor of one or another group of bacteria can be an important hint for diagnosis and treatment of certain diseases such as antibiotic-associated diarrhea, Crohn’s disease, ulcerative colitis, obesity, and pouchitis [26, 27]. Based on recent evidence, implications for the influence of human gut microbiota on metabolic processes, such as nutrient acquisition, energy harvest, and a myriad of host metabolic pathways, have been made [28].

The interaction between the human microbiome and its host in the human gut seems to have not only direct consequences on health or illness but also on modulation of the human immune system. Lakhardi et al. studied interactions between intestinal epithelial cells and gut bacteria in Crohn’s disease patients [29]. They developed a functional metagenomics approach to study bacteria-driven NF-kB regulation, which is an important factor in the development of inflammatory bowel diseases (IBDs). It was furthermore shown that genetic polymorphisms, which are related to IBD phenotype and Crohn’s disease and are involved in host immunity (NOD2 genotype) are associated with shifts in relative frequencies of certain bacteria such as actinobacteria and Bacteroides. It was thereby concluded that the effects of genetic factors on IBD are partly mediated by shifts in the enteric flora, besides environmental covariates such as smoking [30].

It is even argued that with more knowledge about the human gut microbiome, this community could be used not only as a diagnostic but even as a therapeutical target. This will lead to the development of drugs, which alter community composition of human-residing bacteria to enhance health-promoting functions [31]. First studies have already been published, which report durable and, therefore, highly promising results in treatment of Clostridium difficile infections by easy-to-perform microbiome transformations from healthy donors to patients [32, 33].

Chronic wounds affect millions of people and account for an estimated $6 to $15 billion annually in health care costs in the US alone [34]. As molecular testing is more sensitive than culturing, NGS-based 16S sequencing can be an expedient alternative or addition for the identification of bacteria in chronic wounds. As pointed out before, 16S sequencing does not always deliver the same results as culturing methods. But if not only qualitative results but also relative abundances of 16S sequencing analyses are considered, and sequencing depth is high enough, both high and low abundant bacteria can be detected, and a significantly higher portion of anaerobes and slow-growing or dormant bacteria can be detected. In sum, the presence of bacteria can be elucidated regardless of whether or not they can be cultured. Thereby, a more complete picture of the community of the often polymicrobial infection can be drawn than with conservative techniques only. These results can help clinicians in treatment decisions and lead to an improvement of patient outcomes [35]. However, many of the associations postulated, so far, between special microbial compositions and, for example, disease risks could not be proven by independent studies. Therefore, it is suggested that bigger cohort numbers should be used in the future. Additionally, not only characterization of bacteria but also further functional aspects, such as metabolites of microbial origin, should be taken more into account to find significant causalities between the microbial community and disease risks [36].

Resistance and outbreak tracking

The problem of growing resistances in infectious bacteria is another important issue that today’s clinical microbiologists are highly aware of. Despite this, little is known about diversity, distribution, and origins of resistance genes. Using metagenomic approaches, environmental resistance reservoirs, for example, wastewater or animal manure can be elucidated [31]. Additionally, new methods are evolving fast. The publication of Belda-Ferre et al. [37] is an example for this: they developed a technique based on the comparison of the gene pool of a whole microbial population to a specific pathogenic strain. Thereby, metagenomic islands could be detected, defined as regions with extremely limited or no coverage. These represent highly variable regions where virulence genes are often located. Using this comparative technique, the authors anticipate that the number of genes to be cloned or mutated for investigation of genetic virulence determinants will be strikingly reduced, and the process of identifying and characterizing virulence factors will be simplified and accelerated.

Another important topic to which metagenomic sequencing approaches are contributing is the analysis and tracking of pathogen outbreaks. Rasko and coworkers analyzed the Escherichia coli strains responsible for the outbreak of the hemolytic uremic syndrome in Germany in 2011 [6]. Using third-generation single-molecule sequencing on the Pacific Biosciences platform and a subsequent comparative assembly approach, they could obtain phylogenetic information of the strains’ origin and also the accumulated virulence potential acquired by horizontal gene transfer [6]. Baker et al. even built models for typhoid fever transmission using a combination of geographical case clustering on a “Google Earth” basis with high-resolution sequencing. Such results can give important hints for transmission ways and elucidate transmission sources, such as public water reservoirs [38]. Thereby, appropriate precautionary measures could be taken by public health authorities in the future.

Future perspectives and conclusions

Regarding further future perspectives, metagenomic studies looking even beyond bacterial populations might become highly beneficial instruments for the analysis of other possible pathogens or health-influencing populations of the human body. In this context, viral metagenomics is a strongly evolving field. NGS can serve as a diagnostic technique for the identification of viruses and the characterization of the viral community in and on the human body [39]. Comparing the viromes of children with unexplained fever to afebriles by deep sequencing revealed an association of the amount of viral reads as well as viral diversity with the occurrence of fever. With such knowledge, adapted treatment recommendations beyond the default antibiotic therapy, which is often applied if a clear diagnosis is absent, can be made. In the mentioned study, advantages of both long read lengths and a high-sequencing depth were elucidated [40]. As viruses not only directly affect human health, but prokaryotic viruses also influence the bacterial community of the human body, the investigation of the phage community is beginning to gain researchers’ interest, too [12, 41]. However, the application of viral metagenomics in the medical field is still in its infancy [42].

Usage of metagenomic studies for the identification of fungi in humans, especially on the human skin, is also conceivable, although the current interest is only at a fraction of that for the bacterial microbiome. For fungal identification, internal transcribed spacer (especially ITS 2) regions are preferred targets, as they are composed of conserved and highly variable regions, similarly to 16S rRNA in bacteria [43]. Recently, fungal communities from different skin sites were analyzed by ITS 1 sequencing, as discussed above. The experience of this study demonstrated that especially physiologic attributes and topography have influence on bacterial and fungal communities on the human body surface [18]. But at the moment, fungal metagenomics is still at its infancy in the context of diagnosis and medical microbiology and is still very rarely used in human samples in comparison to bacteria-associated metagenomic studies.

Many of the metagenomic NGS-based studies in medical microbiology still only contribute to retrospective analyses. To include metagenomics in an effective way in a routine lab, some obstacles still need to be overcome. Reasonable limitations come from the costs. Although costs decreased for most NGS platforms in the last years, as competition on the market is increasing, still a high amount of money has to be spent per sequencing run [44, 45]. Also, the degree of standardization is still far from the needs of a clinical routine lab. Many steps between sample collection, DNA extraction [46], amplification [47], subsequent library preparation and even data analysis can lead to a high variability of results. To avoid this and to complement existing initiatives in data standards, there are first efforts to propose standards for sequencing library preparation, e.g., with regard to replicate experimental design to achieve data comparability [48], or by the use of standard materials [49] as well as by standardization of bioinformatic data analysis [50].

Furthermore, the time consumption and complexity of laboratory and bioinformatic techniques used for obtaining and analyzing data, which are still high at the moment, need to be reduced to make NGS-based metagenomics approaches accessible for routine labs. But as techniques are evolving fast, and in parallel, knowledge of the human microbiome is extensively growing, metagenomic studies will soon become a part of clinical routine, and thereby, the very conservative field of medical microbiology will undergo considerable evolutionary changes.


Correspondence: Dr. Ovidiu Rücker, IMGM Laboratories GmbH, Lochhamer Str. 29, 82152 Martinsried, Germany, Tel.: +49 89 89557840, Fax: +49 89 89557841, E-Mail:

Conflict of interest statement

Authors’ conflict of interest disclosure: The authors stated that there are no conflicts of interest regarding the publication of this article.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

References

1. Thomas T, Gilbert J, Meyer F. Metagenomics – a guide from sampling to data analysis. Microb Inform Exp 2012;2:3.10.1186/2042-5783-2-3Search in Google Scholar PubMed PubMed Central

2. Metzker ML. Sequencing technologies – the next generation. Nat Rev Genet 2010;11:31–46.10.1038/nrg2626Search in Google Scholar PubMed

3. Sogin ML, Morrison HG, Huber JA, Welch DM, Huse SM, Neal PR, et al. Microbial diversity in the deep sea and the underexplored “rare biosphere.” PNAS 2006;103:12115–20.10.1073/pnas.0605127103Search in Google Scholar PubMed PubMed Central

4. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci USA 2011;108:4516–22.10.1073/pnas.1000080107Search in Google Scholar PubMed PubMed Central

5. Whiteley AS, Jenkins S, Waite I, Kresoje N, Payne H, Mullan B, et al. Microbial 16S rRNA Ion Tag and community metagenome sequencing using the Ion Torrent (PGM) Platform. J Microbiol Methods 2012;91:80–8.10.1016/j.mimet.2012.07.008Search in Google Scholar PubMed

6. Rasko DA, Webster DR, Sahl JW, Bashir A, Boisen N, Scheutz F, et al. Origins of the E. coli strain causing an outbreak of hemolytic-uremic syndrome in Germany. N Engl J Med 2011;365:709–17.10.1056/NEJMoa1106920Search in Google Scholar PubMed PubMed Central

7. Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucl Acids Res 2013;41:e1.10.1093/nar/gks808Search in Google Scholar PubMed PubMed Central

8. Altermann E. Tracing lifestyle adaptation in prokaryotic genomes. Front Microbiol 2012;3:48.10.3389/fmicb.2012.00048Search in Google Scholar PubMed PubMed Central

9. Harris SR, Clarke IN, Seth-Smith HMB, Solomon AW, Cutcliffe LT, Marsh P, et al. Whole genome analysis of diverse Chlamydia trachomatis strains identifies phylogenetic relationships masked by current clinical typing. Nat Genet 2012;44:413–S1.10.1038/ng.2214Search in Google Scholar PubMed PubMed Central

10. Kuczynski J, Lauber CL, Walters WA, Parfrey LW, Clemente JC, Gevers D, et al. Experimental and analytical tools for studying the human microbiome. Nat Rev Genet 2011;13:47–58.10.1038/nrg3129Search in Google Scholar PubMed PubMed Central

11. 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;326:1694–7.10.1126/science.1177486Search in Google Scholar PubMed PubMed Central

12. Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207–14.10.1038/nature11234Search in Google Scholar PubMed PubMed Central

13. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010;464:59–65.10.1038/nature08821Search in Google Scholar PubMed PubMed Central

14. Sommer F, Bäckhed F. The gut microbiota – masters of host development and physiology. Nat Rev Micro 2013;11:227–38.10.1038/nrmicro2974Search in Google Scholar PubMed

15. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222–7.10.1038/nature11053Search in Google Scholar PubMed PubMed Central

16. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature 2011;473:174–80.10.1038/nature09944Search in Google Scholar PubMed PubMed Central

17. Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, et al. Topographical and temporal diversity of the human skin microbiome. Science 2009;324:1190–2.10.1126/science.1171700Search in Google Scholar PubMed PubMed Central

18. Findley K, Oh J, Yang J, Conlan S, Deming C, Meyer JA, et al. Topographic diversity of fungal and bacterial communities in human skin. Nature [Internet] 2013 [cited 2013 May 27]; advance online publication. Available from: http://www.nature.com/nature/journal/vaop/ncurrent/full/nature12171.html.Search in Google Scholar

19. Wylie KM, Weinstock GM, Storch GA. Emerging view of the human virome. Transl Res 2012;160:283–90.10.1016/j.trsl.2012.03.006Search in Google Scholar PubMed PubMed Central

20. Didelot X, Bowden R, Wilson DJ, Peto TEA, Crook DW. Transforming clinical microbiology with bacterial genome sequencing. Nat Rev Genet 2012;13:601–12.10.1038/nrg3226Search in Google Scholar PubMed PubMed Central

21. Buttigieg PL, Hankeln W, Kostadinov I, Kottmann R, Yilmaz P, Duhaime MB, et al. Ecogenomic perspectives on domains of unknown function: correlation-based exploration of marine metagenomes. PLoS One 2013;8:e50869.10.1371/journal.pone.0050869Search in Google Scholar PubMed PubMed Central

22. Hirsch PR, Mauchline TH. Who’s who in the plant root microbiome? Nat Biotechnol 2012;30:961–2.10.1038/nbt.2387Search in Google Scholar PubMed

23. Fouts DE, Szpakowski S, Purushe J, Torralba M, Waterman RC, MacNeil MD, et al. Next generation sequencing to define prokaryotic and fungal diversity in the bovine rumen. PLoS One 2012;7:e48289.10.1371/journal.pone.0048289Search in Google Scholar PubMed PubMed Central

24. Temperton B, Giovannoni SJ. Metagenomics: microbial diversity through a scratched lens. Curr Opin Microbiol 2012;15:605–12.10.1016/j.mib.2012.07.001Search in Google Scholar PubMed

25. Zarco M, Vess T, Ginsburg G. The oral microbiome in health and disease and the potential impact on personalized dental medicine. Oral Dis 2012;18:109–20.10.1111/j.1601-0825.2011.01851.xSearch in Google Scholar PubMed

26. Blaser MJ, Falkow S. What are the consequences of the disappearing human microbiota? Nat Rev Micro 2009;7:887–94.10.1038/nrmicro2245Search in Google Scholar PubMed

27. Frank DN, Pace NR. Gastrointestinal microbiology enters the metagenomics era. Curr Opin Gastroenterol 2008;24:4–10.10.1097/MOG.0b013e3282f2b0e8Search in Google Scholar PubMed

28. Devaraj S, Hemarajata P, Versalovic J. The human gut microbiome and body metabolism: implications for obesity and diabetes. Clin Chem 2013;59:617–28.10.1373/clinchem.2012.187617Search in Google Scholar PubMed PubMed Central

29. Lakhdari O, Cultrone A, Tap J, Gloux K, Bernard F, Ehrlich SD, et al. Functional metagenomics: a high throughput screening method to decipher microbiota-driven NF-κB modulation in the human gut. PLoS One 2010;5:e13092.10.1371/journal.pone.0013092Search in Google Scholar PubMed PubMed Central

30. Li E, Hamm CM, Gulati AS, Sartor RB, Chen H, Wu X, 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:e26284.10.1371/journal.pone.0026284Search in Google Scholar PubMed PubMed Central

31. Schmieder R, Edwards R. Insights into antibiotic resistance through metagenomic approaches. Future Microbiol 2012;7:73–89.10.2217/fmb.11.135Search in Google Scholar PubMed

32. Bakken JS, Borody T, Brandt LJ, Brill JV, Demarco DC, Franzos MA, et al. Treating clostridium difficile infection with fecal microbiota transplantation. Clin Gastroenterol Hepatol 2011;9:1044–9.10.1016/j.cgh.2011.08.014Search in Google Scholar PubMed PubMed Central

33. Brandt LJ, Aroniadis OC, Mellow M, Kanatzar A, Kelly C, Park T, et al. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am J Gastroenterol 2012;107:1079–87.10.1038/ajg.2012.60Search in Google Scholar PubMed

34. Markova A, Mostow EN. US skin disease assessment: ulcer and wound care. Dermatol Clin 2012;30:107–111, ix.10.1016/j.det.2011.08.005Search in Google Scholar PubMed

35. Rhoads DD, Cox SB, Rees EJ, Sun Y, Wolcott RD. Clinical identification of bacteria in human chronic wound infections: culturing vs. 16S ribosomal DNA sequencing. BMC Infect Dis 2012;12:321.10.1186/1471-2334-12-321Search in Google Scholar PubMed PubMed Central

36. Holmes E, Li JV, Marchesi JR, Nicholson JK. Gut microbiota composition and activity in relation to host metabolic phenotype and disease risk. Cell Metab 2012;16:559–64.10.1016/j.cmet.2012.10.007Search in Google Scholar PubMed

37. Belda-Ferre P, Cabrera-Rubio R, Moya A, Mira A. Mining virulence genes using metagenomics. PLoS One 2011;6:e24975.10.1371/journal.pone.0024975Search in Google Scholar PubMed PubMed Central

38. Baker S, Holt KE, Clements ACA, Karkey A, Arjyal A, Boni MF, et al. Combined high-resolution genotyping and geospatial analysis reveals modes of endemic urban typhoid fever transmission. Open Biol 2011;1:110008.10.1098/rsob.110008Search in Google Scholar PubMed PubMed Central

39. Law J, Jovel J, Patterson J, Ford G, O’keefe S, Wang W, et al. Identification of hepatotropic viruses from plasma using deep sequencing: a next generation diagnostic tool. PLoS One 2013;8:e60595.10.1371/journal.pone.0060595Search in Google Scholar PubMed PubMed Central

40. Wylie KM, Mihindukulasuriya KA, Sodergren E, Weinstock GM, Storch GA. Sequence analysis of the human virome in febrile and afebrile children. PLoS One 2012;7:e27735.10.1371/journal.pone.0027735Search in Google Scholar PubMed PubMed Central

41. Reyes A, Semenkovich NP, Whiteson K, Rohwer F, Gordon JI. Going viral: next generation sequencing applied to human gut phage populations. Nat Rev Microbiol 2012;10:607–17.10.1038/nrmicro2853Search in Google Scholar PubMed PubMed Central

42. Delwart E. A Roadmap to the Human Virome. PLoS Pathog 2013;9(2):e1003146. doi:10.1371/journal.ppat.1003146.10.1371/journal.ppat.1003146Search in Google Scholar PubMed PubMed Central

43. Merget B, Koetschan C, Hackl T, Forster F, Dandekar T, Muller T, et al. The ITS2 database. J Vis Exp 2012.10.3791/3806Search in Google Scholar PubMed PubMed Central

44. Liu L, Li Y, Li S, Hu N, He Y, Pong R, et al. Comparison of next-generation sequencing systems. J Biomed Biotechnol 2012;2012:251364.10.1155/2012/251364Search in Google Scholar PubMed PubMed Central

45. Quail MA, Smith M, Coupland P, Otto TD, Harris SR, Connor TR, et al. A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers. BMC Genomics 2012;13:341.10.1186/1471-2164-13-341Search in Google Scholar PubMed PubMed Central

46. Willner D, Daly J, Whiley D, Grimwood K, Wainwright CE, Hugenholtz P. Comparison of DNA extraction methods for microbial community profiling with an application to pediatric bronchoalveolar lavage samples. PLoS One 2012;7:e34605.10.1371/journal.pone.0034605Search in Google Scholar PubMed PubMed Central

47. Frank JA, Reich CI, Sharma S, Weisbaum JS, Wilson BA, Olsen GJ. Critical evaluation of two primers commonly used for amplification of bacterial 16S rRNA genes. Appl Environ Microbiol 2008;74:2461–70.10.1128/AEM.02272-07Search in Google Scholar PubMed PubMed Central

48. Knight R, Jansson J, Field D, Fierer N, Desai N, Fuhrman JA, et al. Unlocking the potential of metagenomics through replicated experimental design. Nat Biotechnol 2012;30:513–20.10.1038/nbt.2235Search in Google Scholar PubMed PubMed Central

49. Huggett JF, Laver T, Tamisak S, Nixon G, O’Sullivan DM, Elaswarapu R, et al. Considerations for the development and application of control materials to improve metagenomic microbial community profiling. Accred Qual Assur 2013;18:77–83.10.1007/s00769-012-0941-zSearch in Google Scholar

50. Raes J, Foerstner KU, Bork P. Get the most out of your metagenome: computational analysis of environmental sequence data. Curr Opin Microbiol 2007;10:490–8.10.1016/j.mib.2007.09.001Search in Google Scholar PubMed

Erhalten: 2013-6-5
Angenommen: 2013-7-22
Online erschienen: 2013-8-29
Erschienen im Druck: 2013-11-1

©2013 by Walter de Gruyter Berlin Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 24.4.2024 from https://www.degruyter.com/document/doi/10.1515/labmed-2013-0018/html
Scroll to top button