Sie sind auf Seite 1von 8

Review

Biodiversity and functional genomics


in the human microbiome
Xochitl C. Morgan1, Nicola Segata1, and Curtis Huttenhower1,2
1
Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
2
Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA

Over the course of our lives, humans are colonized by a From Van Leeuwenhoek to high-throughput
tremendous diversity of commensal microbes, which sequencing: enabling metaomic studies of the human
comprise the human microbiome. The collective genetic microbiome
potential (metagenome) of the human microbiome is Although human-associated microbes were among the first
orders of magnitude more than the human genome, and investigated by microscopy, most are difficult or impossible
it profoundly affects human health and disease in ways to culture under convenient laboratory conditions. The
we are only beginning to understand. Advances in com- vast majority of microbial taxa were uncultured and thus
puting and high-throughput sequencing have enabled minimally studied until the development of DNA-based
population-level surveys such as MetaHIT and the re- techniques. The earliest high-throughput technique for
cently released Human Microbiome Project, detailed microbial ecology (and still the most common) is to se-
investigations of the microbiome in human disease, quence the highly conserved 16S rRNA gene. Also known
and mechanistic studies employing gnotobiotic model as 16S gene (or amplicon) profiling, this provides a rapid
organisms. The resulting knowledge of human micro- and very cost-effective survey of the bacteria present with-
biome composition, function, and range of variation in a community [11], but it is rarely informative with
across multiple body sites has begun to assemble a rich regard to non-bacterial microbes and offers little functional
picture of commensal hostmicrobe and microbemi- information. Historically, this gene was amplified with
crobe interactions as well as their roles in human health
and disease and their potential as diagnostic and thera-
peutic tools. Glossary
16S rRNA gene: the small subunit of the bacterial and archaeal ribosome; the
Understanding our microbial majority DNA sequence of this gene is the most commonly used taxonomic marker for
microbial communities. Other markers include hsp65 and rpoB, which encode
Humans consist only of our own somatic cells until birth, a highly-conserved heat shock protein and the b subunit of RNA polymerase,
but over the first several years of life, our bodies, includ- respectively.
ing the skin surface, oral cavity, and gut, are colonized by a Diversity: the organismal diversity within a sample. Quantification metrics
may emphasize richness (total number of organisms in sample) and/or
an enormous variety of bacteria, archaea, fungi, and evenness (whether they are evenly distributed, in contrast to some more
viruses, which form a community collectively known abundant than others).
b Diversity: the organismal diversity shared between two or more commu-
as the human microbiome or microbiota (see Glossary)
nities, for example, from two or more different people sampled at the same
[13]. Our microbiome contains tenfold as many cells as body site. Can be measured either taxonomically (counting shared microbes)
the rest of our bodies, and orders of magnitude more or phylogenetically (quantifying shared phylogenetic branches).
Clade: a group of organisms descended from a common ancestor, correspond-
genes than the human genome [1,4]. Normally these ing to a single branch on the tree of life; often used to refer to higher-level taxa
microbes are commensal or mutualists, helping to digest or phylogenetically related groups of microbes.
our food and maintain our immune systems. Although Commensal bacteria/microbes: members of a host-associated microbial
community that are present in a typical state independent of disease, and
the human microbiome has long been understood to that are considered to be beneficial, harmless, or non-pathogenic under
influence human health and disease, low-cost high- ordinary circumstances.
throughput sequencing has only recently given us the Gnotobiotic: a host animal containing a defined set of microorganisms, either
synthetically implanted or transferred from another host; often used to refer to
experimental tools to investigate the breadth of its in- model organisms with microbiota humanized by fecal transfer or gavage.
volvement. This has resulted in an explosion of recent Kochs postulates: a set of criteria used to determine if a microorganism is the
work illuminating the role of the microbiome in condi- causative agent of a particular disease, including presence in disease, absence
in health, isolation, induction of disease upon introduction, and subsequent
tions ranging from obesity [5,6] to inflammatory bowel reisolation.
disease (IBD) [7,8] to kidney stones [9]. Large-scale Metagenome/metatranscriptome: the total genomic DNA or RNA transcripts
surveys such as the Metagenomics of the Human Intes- of all organisms within a community.
Microbiome: the total microbial community, including biomolecules, within a
tinal Tract (MetaHIT) consortium [4] and the recent defined environment such as the human body or individual body-site habitats.
Human Microbiome Project (HMP) [1,10] have shown Microbiota: the total collection of microbial organisms within a community,
here generally referring to the resident microbes of the human body.
that these are only the beginning of a richer functional
Operational taxonomic unit (OTU): a cluster of organisms similar at a defined
understanding of the human microbiome. sequence identity threshold (e.g., 95%) within some region (e.g., the whole 16S
gene), used in the absence of a named species, genus, etc.
Taxon: the microbes belonging to a taxonomic category, typically of low level
Corresponding author: Huttenhower, C. (chuttenh@hsph.harvard.edu). such as genus or species.
Keywords: Human Microbiome Project; microbiota; metagenomics.

0168-9525/$ see front matter 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tig.2012.09.005 Trends in Genetics, January 2013, Vol. 29, No. 1 51
Review Trends in Genetics January 2013, Vol. 29, No. 1

universal bacterial primers, and the individual sequences sequences each, corresponding to between 400 and 4000
were cloned and determined using Sanger sequencing. taxa (this range of species counts reflects largely bioinfor-
More recent 16S sequencing techniques are very high- matic rather than biological differences between studies).
throughput, generating up to millions of reads per sample, These investigations showed that gut a diversity was much
but with typically very short read lengths [12]. higher than most previous estimates and that b diversity
Metagenomic studies, in which the DNA of an entire was high both between subjects as well as between mucosal
community is sequenced, provide a much broader and more and stool samples from the same subject [17]. This, com-
complex picture of microbial communities by simulta- bined with preliminary associations between altered
neously cataloging community organisms (including the microbiota and disease, highlighted the need for much
non-bacterial component), genes, and genomes (Figure 1). larger-scale studies to determine the range of microbial
The earliest large-scale metagenomic studies began soon variation and function in the human gut across entire
after the turn of the century [13,14], but the expense of populations.
clone library construction and Sanger sequencing made
metagenomic human microbiome studies unfeasible for Large-scale human microbiome studies
most groups. The advent of high-throughput sequencing The first population-scale metagenomic study of the hu-
made much larger-scale metagenomic efforts, including man microbiome was conducted by the MetaHIT consor-
MetaHIT [4] and the HMP [1,10], economically feasible. tium [4]. Stool sequenced from 124 Spanish and Danish
Here we review what these and recent disease and model- subjects were estimated to contain 1150 common bacterial
organism studies have shown us about the composition and species in the human gut with a collective 3.3 million
genetic capabilities of the human microbiome and discuss genes. However, only 75 of these organisms and 294 000
the future of the field of human microbiome research. genes were shared by more than half of subjects, even
within a population with limited genetic and environmen-
Early human microbiome studies tal diversity. In addition to essential housekeeping genes,
The largest early efforts at measuring the composition of common gut bacteria were enriched for pathways that
the human microbiome were 16S cloning-based studies of metabolized complex sugars from the host diet and intes-
the gut that included fecal and mucosal samples from up to tinal lining, as well as for adhesion and vitamin and
a dozen individuals [15,16]. These resulted in 1020 000 xenobiotic processing. The abundance of archaeal and

Current techniques for human microbiome proling Emerging methods


Nasal
Oral Human microbiome sample Human microbiome sample
Skin
Gastrointesnal
Urogenital
Major ecological
areas within the
microbiome
Extract DNA
Which organisms are What are the funcons of How does the community
present? the community? respond to its environment?
Amplify 16S rRNA gene Extract RNA, protein, and small molecules
Sequence total DNA
by PCR and sequence Metaproteomics

Metametabolomics
Metatranscriptomics

OTU1
Bin similar sequences
OTU2
into OTUs
OTU3

Compare sequences to Compare sequences BLAST Mass Metabolite


Compare OTUs to Reverse
databases reference genomes to databases transcribe spectroscopy proling

GATTACA
Abundance

Abundance

Key:
GATTACA Regulaon
Flux
GATTTCA Interacon
GATTTCA
Funcons
OTU

Idenfy OTUs in sample and Phylogenec view of Idenfy microbial sequences, variants, and Idenfy genes, pathways, and Idenfy interacons between expressed genes,
relave frequencies community composion polymorphisms in sample relave frequencies in sample proteins, and metabolic products in sample

TRENDS in Genetics

Figure 1. Overview of bioinformatic methods for functional metagenomics. Microbial community samples typically contain many species of bacteria and other
microorganisms, here indicated by different colors. After total DNA has been extracted, the composition of the community is determined by amplifying and sequencing the
16S rRNA gene. Highly similar sequences are grouped into operational taxonomic units (OTUs), and these OTUs are labeled by comparison with databases of recognized
organisms. OTUs can then be analyzed in terms of presence/absence, abundance, or phylogenetic diversity. To determine biomolecular and metabolic functions present in
the community, the total metagenomic DNA may be sequenced and compared with function-oriented databases. Alternatively, sequenced community DNA can be
compared to reference genomes. This allows identification of microbial sequence variants and polymorphisms, as well as providing an alternative means for detecting the
presence and abundance of specific organisms. In the future, the emerging fields of metatranscriptomics, metaproteomics, and metametabolomics will help to build an
integrated picture of gene expression and interactions in microbial communities.

52
Review Trends in Genetics January 2013, Vol. 29, No. 1

fungal components was very low (<1%) although, as in all identifying a microbiome-wide count of over 10 million
such studies, this is influenced by sample collection and total unique microbial genes, the HMP allowed detection
DNA extraction [4]. of between-body-site microbial associations.
The HMP was by far the largest human microbiome
study to date, a five-year effort that determined the range Diversity in the human microbiome
of microbial diversity in disease-free individuals by se- Estimating the richness of species in a community is
quencing over 5000 samples from approximately 250 bioinformatically challenging. Thus, the total human
healthy volunteers in two U.S. cities. Each subject was microbiome assessed by the HMP contained between
sampled at 15 or 18 body sites (nine oral, four skin, one 3500 and 35 000 species-level operational taxonomic units
nasal, one stool, three vaginal), and roughly half of subjects (OTUs) depending on parameter choices [1]. These, how-
were sampled at up to two additional time-points. Most ever, confidently spanned roughly 600 genera and covered
samples were profiled using 16S amplicon sequencing to 90% of the phylogenetic range of microbes expected in this
determine community composition; approximately 15% population [1,18]. Several signature bacterial genera were
were subjected to metagenomic sequencing to determine observed across nearly all individuals and represented the
community function [1,10]. In addition to expanding the plurality (and often majority) of particular body-site com-
MetaHIT gut gene catalog by nearly 1.8 million genes and munities (Figure 2), but specific taxa were highly variable

A map of Propionibacterium acnes


lives on the
Many Corynebacterium
species characterize
skin and dierent body sites:
diversity nose of most C. matrucho
people the plaque
in the human Streptococcus dominates C. accolens
the oral cavity with the nose
microbiome S. mis > 75% in the cheek C. croppenstedi
the skin
Lactobacillus species
(L. gasseri, L. jensenii,
L. crispatus, L. iners)
are predominant
but mutually
exclusive in Several Prevotella species are
the vagina present in the gastrointesnal
s Coryn
to coccu ebact
erium tract. P. copri is present in
Strep 19% of the subjects and
Ac
nom Bi dominates the intesnal
yce dob
s ac ora when present
ter
Staphylococcus s ium
lu
ci l
epidermidis ob
a
Po

colonizes ct
La
rp
hy

external
ro
mo

body sites
us

na
occ

s P
loc

rev
p hy

ote

Bacteroides
Sta

lla

is the most
abundant
occus

genus in
Bacteroides
Ruminoc

the gut
of almost
all healthy
Clostridium

subjects
bacter
Campylo

Campylobacter includes
opportunisc
Key: pathogens,
Commensal but members
microbes live in the
An

Potenal oral cavies


ae

a
eri

of most
roc

pathogens
iss
oc

healthy
Ne
cu

The four most people in


s

r
abundant phyla te the cohort
Se ac
len ob
Acnobacteria om Vei in et
o n l l on Ac
as ell
Bacteroidetes Fus a us
richia phil
oba
Esche mo
Firmicutes cte
rium Hae
Proteobacteria
Stool
Low abundance phyla Cheek
Plaque
Chloroexi Spirochaetes Tongue
Cyanobacteria Synergistetes Nose E. coli is present in the gut of
Euryarchaeota Tenericutes Vagina
Fusobacteria Thermi Skin the majority of healthy subjects
Lensphaerae Verrucomicrobia but at very low abundance

TRENDS in Genetics

Figure 2. A map of diversity in the human microbiome. The human microbiome is dominated by four phyla: Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria.
In the center is a phylogenetic tree of organisms abundant in the human microbiome. Commensal microbes are indicated by circles, and potential pathogens are indicated
by stars. The middle ring corresponds to body sites at which the various taxa are abundant and is color-coded by site [e.g., Ruminococcus (blue) is found mostly in the gut,
whereas Lactobacillus (purple) is found mostly in the vagina]. The bar heights on the outside of the circle are proportional to taxa abundance at the body site of greatest
prevalence [e.g., Streptococcus mitis (yellow) dominates the inside of the cheek, whereas the gut is abundant in a variety of Bacteroides]. The intensity of external colors
corresponds to species prevalence in each body site. Data from the Human Microbiome Project [10] and MetaPhlan [75]; microscopy images from BacMap [76].

53
Review Trends in Genetics January 2013, Vol. 29, No. 1

and almost never universal [1]. The gastrointestinal tract, elbows and behind the ear) was low both within and
including both the oral cavity and stool, was the most between subjects, perhaps due to exposure and frequent
diverse microbiome among the sampled body habitats, disruption [1]. The human body community of lowest a
estimated to contain several thousand OTUs correspond- diversity was that of the vagina; most women were domi-
ing to approximately 150 genera [19]. The well-studied gut nated by one of four single species of Lactobacillus [20]. The
microbiome exemplified inter-individual variability; the vaginal microbiome becomes even less diverse during
Bacteroidetes and Firmicutes phyla dominated as pregnancy [21], and it is emerging as a useful human-
expected, but abundance ranged from >90% Bacteroidetes associated model community due to its uniqueness relative
to >90% Firmicutes in different subjects. Only some half- to other primates [22] and highly structured temporal
dozen gut taxa were near-universal (present in 95% of fluctuations [23,24].
subjects) [20]. The HMP discovered and validated several novel taxa,
All body areas examined by the HMP dramatically mostly at the genus level, but hinted at a possible novel
differed not only in composition but also in ecological family within the order Clostridiales [25]. Novel OTUs
organization (Box 1). The oral microbiome was essentially were generally of low abundance (<2%), a level comparable
as diverse as the gut but, by contrast, more of this diversity to that of Escherichia coli in healthy subjects [1], but were
was shared between individuals. Signature clades also carried across multiple subjects and multiple visits. The
varied strikingly even between very similar oral surfaces novel taxa were mainly from the largely uncharacterized
(e.g., mucosa, saliva, and plaque); abundant genera includ- Barnesiella genus, but also included novel Dorea, Oscilli-
ed Streptococcus, Haemophilus, Veillonella, Actinomyces, bacter, and Desulfovibrio genera, which are associated
and Fusobacterium. Skin diversity (sampled at the inner broadly with colon cancer, dietary shifts, and opportunistic
infections, respectively [25]. Low-abundance community
Box 1. Class discovery in the microbiome members can affect health even at low numbers [26]
and, in the worst case, under specific circumstances, can
The human body comprises a set of unique ecosystems with a
broad range of configurations. At one extreme, the low-diversity overgrow to the detriment of host health (e.g., Clostridium
vaginal microbiome typically occurs in one of five discrete types: difficile). Further study may reveal important roles for
four are each dominated by a different signature species of these novel taxa. However, because the microbial variation
Lactobacillus [25], and the fifth is characterized by the absence of between individuals was greater than that of samples from
Lactobacillus. At the other extreme, the highly diverse human
mouth microbiome has a complex core of prevalent but variably
the same subject at different points in time [1], these low-
abundant taxa [20]. In communities such as the vagina, it is easy to abundance microbes at the very least contribute to a
perform class discovery by community clustering or typing; uniquely personalized human microbiome when consid-
organizing the complexity of other human body habitats in this ered in terms of microbial presence and abundance.
manner has been the subject of much recent work.
In the gut in particular it has been hypothesized that the
microbiome forms enterotypes, or discrete, stable clusters of similar
Beyond the baseline: microbial variation in children, the
microbiome configurations [68]. Like that of the mouth, the gut elderly, and global populations
microbiome is highly diverse; a combination of environmental Human microbiome composition appears to be particularly
effects, early exposures, and long-term diet determine the overall dependent on age and geography, although both are con-
community profile. Short-term dietary changes induce more modest
founded by a variety of dietary, developmental, environmen-
and transient effects [44] realized primarily at the transcriptional level
[44]. The existence of human gut enterotypes and their associations tal, and genetic factors. The HMP included only American
with factors such as genetics or diet are much less straightforward adults between the ages of 18 and 40, and MetaHIT extend-
than vaginal ecotypes, and are an area of active research. ed this somewhat with Spanish and Danish individuals up
A recent study of individuals from six nations initially proposed to age 70. The infant microbiome is of particular interest
three gut microbiome types, dominated by Prevotella, Ruminococcus,
with respect to acquisition, immune training, and extreme
and Bacteroides [68]. Subsequent studies [1,44] also observed a
distinct Prevotella type, but suggested that the Bacteroides and dietary changes over the first few years of life [27,28]. Some
Ruminococcus types instead represented two ends of a single of the earliest studies of microbiome structure and function
continuum between abundant Bacteroidetes and abundant Firmi- indicated that immediate acquisition occurs from the envi-
cutes. An associated controlled-feeding study associated the Pre- ronment and is dependent on mode of delivery [28], with
votella enterotype with a high-carbohydrate diet, and Bacteroides
with higher animal fat and protein consumption. Short-term enforced
stabilization to a more adult-like community taking place
dietary shifts from high-fat and high-protein to high-carbohydrate over two to three years [27]. Functionally, the shift from the
(and vice versa) somewhat modified organismal abundances, but newborn to adult-like gut microbiome includes enrichment
rarely changed the presence or absence of specific gut microbes in for anerobic fermentation, complex carbohydrate degrada-
particular subjects [43].
tion (e.g., glycosyl hydrolases), and depletion of simple sugar
The largest currently published study (528 Americans, Malawians,
and Amerindians) found that diverse Prevotella OTUs, distinct from
(lactose/galactose/sucrose) transport and metabolism pro-
the proposed Prevotella enterotype, were highly predictive in teins [27]. Recent studies of the microbiota of the elderly
distinguishing between the populations because the genus was found that frailty is correlated with an overall decrease in
more prevalent in Amerindians and Malawians (who ate maize- microbial diversity, with overall increases in Bacteroides
based diets) than in Americans, who typically had higher levels of
and decreases in Firmicutes that corresponded to a decrease
Bacteroides. Although Bacteroides and Prevotella were almost
mutually exclusive in most subjects, a considerable minority of in glutarate and the anti-inflammatory short-chain fatty
subjects harbored substantial populations of both genera, raising acid butyrate [29].
the intriguing possibility that even the Prevotella and Bacteroides Another recent comprehensive study addressed several
enterotypes may not represent wholly discrete energy minima in of these questions simultaneously by surveying the stool
gut community configurations [30].
microbiomes of a highly diverse population that included
54
Review Trends in Genetics January 2013, Vol. 29, No. 1

314 Americans, 114 Malawians, and 100 Amerindians. genomes, each habitat thus seems to have a core metagen-
Roughly two-thirds of the subjects were under the age of ome present in most hosts, in addition to a pan-metagenome
17, whereas the remaining third were between the ages of of more flexible auxiliary genes carried by the community of
18 and 70 [30]. In all three populations of this study, inter- each habitat.
subject variability was higher in children than in adults,
infant microbiomes were dominated by Bifidobacteria, and Gnotobiotics and model systems for the human
microbiomes reached an adult-like configuration by age microbiome
three. Microbial diversity increased with age in all popula- Much of our knowledge of microbiome influence in human
tions, but both Malawian and Amerindian adults achieved health comes from gnotobiotic systems, which predate the
higher diversity than American adults. The Malawian and field of metagenomics [34]. Gnotobiotics are animals (typi-
Amerindian microbiomes were also more similar to one cally mice or zebrafish) in which normal host microbiota
another than to the U.S. microbiome, in both adults and has been replaced by a defined set of microbes, allowing
children [30]. studies of how microbial colonization influences host
When the authors examined functional gene families and health and development or other processes of interest.
enzyme classes within these global gut metagenomes, no Gnotobiotic animals are born under germ-free conditions
enzyme classes were wholly unique to adults or babies, and and subsequently colonized (typically by gavage) with a
the total number of enzyme classes was similar in both single species [35], a defined set of species such as the
groups. However, the adults had many more uncharacter- eight-species Schaedler flora, or entire gut communities
ized enzymes than babies, likely reflecting the age-related from mice or humans with phenotypes of interest [34,36].
increase in diversity. The infant microbiome had higher Gnotobiotic studies have shown that the gut microbiota
levels of folate-producing enzymes than that of adults, likely affects the host in many ways, including fertility, the
reflecting the ability of adults to obtain folate from diet. development of the heart, lungs, and liver [34], and even
Conversely, biosynthetic capacity for the vitamins cobala- brain gene expression and behavior. For example, a recent
min, thiamine, and biotin increased with age. Urease genes study found that germ-free mice were less anxious and
were more frequently observed in Malawian and Amerindi- more active than mice with a normal gut microbiota, but
an infant microbiomes, possibly corresponding to greater these effects were largely reversed by exposure to gut
probability of dietary nitrogen deficiency. Malawian and microbiota at an early age [37]. Three areas of particularly
Amerindian adult microbiomes were enriched in glutamate intense focus are the heavy influence of the microbiome on
synthase relative to the American microbiomes, the latter nutrition [34] and obesity [5], immune system development
instead carrying an increased capacity for degradation of [35,3840], and inflammatory bowel disease [7,8,41].
glutamine and other amino acids [30]. Interestingly, this
functional difference has also been observed when compar- The gut microbiome: diet and obesity
ing the gut microbiota of herbivores and carnivores [31]. The earliest attempts at raising germ-free animals dem-
onstrated the vital contribution of the gut microbiota to
Habitat-specific functionality as a principle of human nutrition because the animals rapidly succumbed to vita-
microbiome organization min deficiencies and required extensive supplementation
Several human microbiome studies have reported the [34]. When germ-free mice were colonized with gut bacteria
metagenomic distribution of pathways within each body from conventionally raised adult mice, their food intake
habitat to be much more consistent among individuals decreased, but their body fat increased due to increased
than are microbial abundances [1,4,6,32]. In each case, monosaccharide availability from food signaling the liver
microbial abundances within the same habitat (gut, oral to increase fat storage [5]. Studies of mice genetically
cavity, etc.) varied tremendously among subjects, but two predisposed to obesity found that they had more Firmi-
types of core pathways were stable in abundance. The cutes and fewer Bacteroidetes than their genetically-lean
first, corresponding to a more-or-less universal set of siblings, and when a group of germ-free mice were colo-
housekeeping genes such as those reported by MetaHIT nized with microbiota from obese mice, they gained more
[4], was present in every habitat and represents processes weight than those colonized with microbiota from lean
necessary for human-associated microbial life, such as the mice [5]. Furthermore, mice colonized with human gut
ribosomal machinery, ATP synthesis, and glycolysis. microbiota showed large and rapid shifts in gut micro-
More surprisingly, specific sets of site-specific gene func- biome composition in response to diet; these were accom-
tion were maintained within each habitat regardless of the panied by stable changes in microbial taxa abundance and
taxa present. The stool microbiome was particularly abun- gene expression [42].
dant in genes related to complex carbohydrate degradation The results of human obesity studies have proven more
despite highly variable Bacteroidetes:Firmicutes ratios; hy- variable than the mouse studies; Bacteroides have been
drogen sulfide production and methionine degradation were reported by various groups to increase, decrease, and not
also site-specific to the gut at low abundance [1]. Individual change during weight loss [5]. Other controlled-feeding
profiles of carbohydrate-active enzymes within each body studies suggest that short-term dietary changes can modi-
habitat were also very similar by site. The oral cavity fy taxon abundances [43] or transcriptional levels [44], but
microbiome, for example, was optimized for simple sugar seldom change the presence or absence of specific taxa [43].
metabolism and particularly for dextran, whereas the vagi- Thus, it has initially been difficult to interpret the extreme
nal microbiome was optimized for glycogen and peptidogly- changes often seen in gnotobiotic animals relative to the
can degradation [33]. Much like individual bacterial more subtle behavior in the human gut.
55
Review Trends in Genetics January 2013, Vol. 29, No. 1

A study comparing the gut microbiota of normal weight, Table 1. Conditions recently associated with changes in the
obese, and post-gastric-bypass subjects noted that obese human microbiome
individuals were more likely to harbor both Prevotella and Disease Microbiome-related findings Refs
methanogenic archaea [45]. The authors proposed that Asthma Large increase in airway microbial [69]
methanogens removed hydrogen produced by Prevotella, diversity; Comamonadaceae,
Sphingomonadaceae, Oxalobacteraceae
allowing Prevotella to produce short-chain fatty acids more increases correlate with bronchial
efficiently, and these were absorbed by the host, contrib- hyperresponsiveness
uting to host obesity [45]. This proposal is supported by Atopic dermatitis Decreased microbial diversity, increased [70]
evidence from gnotobiotic mouse studies; mice co-colonized Staphylococcus aureus; therapy restored
with Bacteroides thetaiotaomicron (which, similarly to Pre- commensals
votella, ferments carbohydrates into short-chain fatty acids Colorectal cancer Tumors were enriched in typical gut [71]
microbes relative to surrounding healthy
and produces hydrogen) and the methanogen Methanobre- tissue; healthy tissue enriched in
vibacter smithii had higher numbers of total gut bacteria, Citrobacter, Cronobacter, Salmonella,
higher acetate levels in the intestinal lumen and blood, and and Shigella
more body fat than monocolonized mice or mice co-cultured Kidney stones Lack of Oxalobacter formigenes [9,72]
with B. thetaiotaomicron and the sulfate-reducing bacteria increases urinary oxalate levels and risk
of kidney stones. Supplementation may
Desulfovibrio piger [46].
be beneficial; results mixed
Periodontitis Shift from Actinomyces and [73]
Host immune interactions in the gut and body-wide Streptococcus to Haemophilus and
In addition to nutrient absorption, the gut microbiome also Selenomonas
greatly affects immune development. The immune systems Psoriasis Decrease in commensal Staphylococcus [74]
of germ-free mice are strikingly abnormal, with smaller and Propionibacterium and increase in
Proteobacteria
lymph nodes, lower serum immunoglobin levels, and lower
levels of leukocytes than conventially raised mice [34].
Germ-free mice are particularly deficient in CD4+ T cells
[47,48], but colonization with Bacteroides fragilis or even
exposure to its polysaccharide restores CD4+ cells to nor- disease if they were kept germ-free [50] or pre-emptively
mal levels [47]. Monocolonization of germ-free mice with treated with antibiotics [41]. Furthermore, fostering or co-
segmented filamentous bacteria (SFB), a mouse-commen- caging healthy mice with IBD-predisposed mice was suffi-
sal Clostridia, simultaneously stimulated many kinds of T- cient to cause IBD in the healthy mice; this correlated with
cells, including Th1, Th2, Th17, and Treg [35]. This im- the transfer of the Enterobacteriaceae species Klebsiella
mune activation is not universally beneficial; SFB also pneumonia and Proteus mirabilis from the IBD mice to the
triggers autoimmune arthritis in genetically predisposed healthy mice [41]. Intriguingly, this colitis was attenuated
mice in a Th17-dependent manner, but they do not became by the administration of probiotic fermented milk contain-
arthritic if raised in germ-free conditions [49]. ing Bifidobacterium spp. [51].
Gut/immune system interactions are not limited to The relationships between human gut microbiota and
CD4+ cells; natural killer (NK) cells accumulate in large IBD are less well understood, but it is widely accepted that
numbers in the lungs and colons of germ-free mice, making human IBD is not caused by a specific pathogen, but
them much more susceptible to experimental asthma and instead by an immune system imbalance with respect to
colitis. This NK accumulation cannot be reversed by ex- normal gut bacteria. Metagenomic studies of IBD in the
posing mice to germs later in development but is pre- human gut have consistently shown that IBD is associated
vented by very early exposure to normal mouse gut flora with gut dysbiosis [8] and reduced microbial diversity [4]. A
[38]. The continuum between early immune development, lack of F. prausnitzii was associated with high postopera-
ongoing training, and acute or localized inflammation is tive recurrence of Crohns disease, and F. prausnitzii sup-
still being explored. Some specific microbes, primarily plementation attenuated experimental colitis in mice [7],
Faecalibacterium prausnitzii, have been suggested to but no human IBD F. prausnitzii supplementation studies
act as suppressors of inflammation via short-chain fatty have been reported to date. Many groups have observed
acid production and stimulation of the anti-inflammatory decreases in members of anti-inflammatory Clostridia
cytokine IL-10 [7]. from clades IV and XIVa, and increases in E. coli [8],
but it has not yet been determined whether these micro-
The microbiome in disease: studies from IBD biome changes are a cause of IBD in humans, or are merely
In addition to its roles in obesity and immune response, it is caused by IBD. Two probiotics, E. coli strain Nissle (iso-
increasingly evident that the microbiome influences the lated from an exceptionally gastroenteritis-resistant
development of many other diseases (Table 1). IBD is only World War 1 soldier) and VSL#3 (eight strains of Bifido-
one example of a human disease that apparently does not bacterium, Lactobacillus, and Streptococcus) have been
follow Kochs postulates, but is intimately connected to shown to be effective treatments for maintaining and
host microbiome. It has long been known that IBD is both inducing remission in ulcerative colitis [52], but the un-
etiologically and microbially distinct, and that antibiotics, derlying molecular mechanisms are not yet well under-
particularly rifampicin, are sometimes beneficial in its stood. Further probiotic research may shed light on how
treatment [50]. Rodent studies showed that rats and mice short- or long-term ecosystem therapy may restore bal-
genetically predisposed to IBD would not develop the ance to dysbiosis.
56
Review Trends in Genetics January 2013, Vol. 29, No. 1

Future directions in human microbiome research shown that the human microbiome is highly variable be-
In addition to understanding the basic ecological principles tween individuals, and its genetic capacity is far greater
of human microbiome organization and its life-long epide- than that of the human genome. Bacterial community func-
miology, two major areas of microbiota function have recent- tion is highly conserved between corresponding body sites in
ly begun to be explored. The first is diagnostic: does human hosts, whereas community gene transcription is
microbiota composition or genetic function predict human much more variable; these all affect health in many ways
disease onset, progression, or outcome? The past decade has we are just beginning to understand. The most productive
seen great strides in assessing disease history and predict- future research will integrate the metagenome, metatran-
ing risk using high-dimensional gene expression and genetic scriptome, metaproteome, and metametabolome to model
biomarkers; the next decade is likely to see the same with entire ecosystems.
respect to the microbiome [53,54]. The second area in which
the microbiome has tremendous potential is in therapeutic Acknowledgments
intervention; human genetic defects are now detectable but This work was funded by NIH grant 1R01HG005969-01 to C.H.
difficult to modify, whereas the microbiome is both measur-
able and plastic. Targeted narrow-spectrum antibiotics [55], References
1 Human Microbiome Project Consortium (2012) Structure, function and
drugmicrobe interactions [56], probiotics [44,51,52], and diversity of the healthy human microbiome. Nature 486, 207214
dietary interventions [57] all show promise for beneficial 2 Peterson, J. et al. (2009) The NIH Human Microbiome Project. Genome
microbiome modulation. Remarkably, the roles and mecha- Res. 19, 23172323
nisms by which host genetics, early life events, geographical 3 Reyes, A. et al. (2010) Viruses in the faecal microbiota of monozygotic
twins and their mothers. Nature 466, 334338
location, or transmission of the microbiota might operate all
4 Qin, J. et al. (2010) A human gut microbial gene catalogue established
represent largely open questions. by metagenomic sequencing. Nature 464, 5965
The technologies available to realize these research 5 Ley, R.E. (2010) Obesity and the human microbiome. Curr. Opin.
goals continue to develop at a remarkable pace, and are Gastroenterol. 26, 511
rapidly approaching diagnostic utility. Sanger sequencing 6 Turnbaugh, P.J. et al. (2009) A core gut microbiome in obese and lean
twins. Nature 457, 480484
has been replaced by Roche 454, and this is in turn giving
7 Sokol, H. et al. (2009) Low counts of Faecalibacterium prausnitzii in
way to ultra-high-throughput Illumina sequencing [12,58], colitis microbiota. Inflamm. Bowel Dis. 15, 11831189
with longer reads for microbial isolate genomes and com- 8 Joossens, M. et al. (2011) Dysbiosis of the faecal microbiota in patients
munity metagenomes on the horizon [59,60]. Metagenomes with Crohns disease and their unaffected relatives. Gut 60, 631637
measure only what a community is capable of doing, but 9 Kaufman, D.W. et al. (2008) Oxalobacter formigenes may reduce the risk
of calcium oxalate kidney stones. J. Am. Soc. Nephrol. 19, 11971203
developing functional assays will better assess how it
10 Human Microbiome Project Consortium (2012) A framework for
dynamically responds to its environment. Metatranscrip- human microbiome research. Nature 486, 215221
tomics presents additional technical challenges such as 11 Tringe, S.G. and Hugenholtz, P. (2008) A renaissance for the
depletion of ribosomal RNA, but has already been used to pioneering 16S rRNA gene. Curr. Opin. Microbiol. 11, 442446
describe marine [61] and fecal [62] communities. Metapro- 12 Lazarevic, V. et al. (2009) Metagenomic study of the oral microbiota
by Illumina high-throughput sequencing. J. Microbiol. Methods 79,
teomics, or the quantification of community peptides, has 266271
likewise been steadily developing to describe, for example, 13 Tyson, G.W. et al. (2004) Community structure and metabolism
the gut microbiota [63,64]. Metametabolomics, or the through reconstruction of microbial genomes from the environment.
quantification of community small-molecule metabolites, Nature 428, 3743
14 Venter, J.C. et al. (2004) Environmental genome shotgun sequencing of
has been used to compare differences in host blood plasma
the Sargasso Sea. Science 304, 6674
between germ-free and conventionally raised mice [65] and 15 Gill, S.R. et al. (2006) Metagenomic analysis of the human distal gut
to compare fecal samples from healthy individuals and microbiome. Science 312, 13551359
those with Crohns disease [66]. The immense computa- 16 Ley, R.E. et al. (2006) Microbial ecology: human gut microbes
tional challenge of assembling these vast and diverse associated with obesity. Nature 444, 10221023
17 Eckburg, P.B. et al. (2005) Diversity of the human intestinal microbial
metaomic data to describe entire ecosystems, identifying
flora. Science 308, 16351638
how the genes interact with one another within and be- 18 Fodor, A. and Earl, A. (2012) Most wanted taxa from the human
tween bacteria to produce metabolites, and quantifying microbiome. PLoS ONE 7, e41294
and understanding how these processes change over time 19 Li, K. et al. (2012) Analyses of the microbial diversity across the human
and in response to the environment, is the goal of the microbiome. PLoS ONE 7, e32118
20 Huse, S.M. et al. (2012) A core human microbiome as viewed through
emerging field of molecular ecosystems biology [67]. Inte- 16S rRNA sequence clusters. PLoS ONE 7, e34242
grating these new technologies with model organisms and 21 Ratner, A.J. et al. (2012) A metagenomic approach to characterization of
culture-based approaches will help us to unravel our rela- the vaginal microbiome signature in pregnancy. PLoS ONE 7, e36466
tionship with our microbial majority. 22 Rivera, A.J. et al. (2011) Differences between the normal vaginal
bacterial community of baboons and that of humans. Am. J.
Primatol. 73, 119126
Concluding remarks 23 Gajer, P. et al. (2012) Temporal dynamics of the human vaginal
It is an exciting time for work on human-associated micro- microbiota. Sci. Transl. Med. 4, 132ra52
bial communities, both for basic biological discovery and for 24 Ravel, J. et al. (2011) Vaginal microbiome of reproductive-age women.
translational research. Experimental and computational Proc. Natl. Acad. Sci. U.S.A. 108 (Suppl. 1), 46804687
25 Wylie, K.M. et al. (2012) Novel bacterial taxa in the human microbiome.
tools are at the cusp of widespread affordability and avail-
PLoS ONE 7, e35294
ability, driven both by large-scale projects such as the HMP 26 Hajishengallis, G. et al. (2011) Low-abundance biofilm species
and by the democratization of high-throughput genomics orchestrates inflammatory periodontal disease through the
and bioinformatics. Large-scale metagenomic studies have commensal microbiota and complement. Cell Host Microbe 10, 497506

57
Review Trends in Genetics January 2013, Vol. 29, No. 1

27 Koenig, J.E. et al. (2011) Succession of microbial consortia in the 52 Jonkers, D. et al. (2012) Probiotics in the management of inflammatory
developing infant gut microbiome. Proc. Natl. Acad. Sci. U.S.A. 108 bowel disease: a systematic review of intervention studies in adult
(Suppl. 1), 45784585 patients. Drugs 72, 803823
28 Dominguez-Bello, M.G. et al. (2010) Delivery mode shapes the 53 Oh, P.L. et al. (2012) Characterization of the ileal microbiota in
acquisition and structure of the initial microbiota across multiple rejecting and nonrejecting recipients of small bowel transplants.
body habitats in newborns. Proc. Natl. Acad. Sci. U.S.A. 107, Am. J. Transplant. 12, 753762
1197111975 54 Farrell, J.J. et al. (2012) Variations of oral microbiota are associated
29 Claesson, M.J. et al. (2012) Gut microbiota composition correlates with with pancreatic diseases including pancreatic cancer. Gut 61, 582588
diet and health in the elderly. Nature 488, 178184 55 Louie, T.J. et al. (2011) Fidaxomicin versus vancomycin for Clostridium
30 Yatsunenko, T. et al. (2012) Human gut microbiome viewed across age difficile infection. N. Engl. J. Med. 364, 422431
and geography. Nature 486, 222227 56 Okuda, H. et al. (1998) A possible mechanism of eighteen patient
31 Ley, R.E. et al. (2008) Evolution of mammals and their gut microbes. deaths caused by interactions of sorivudine, a new antiviral drug,
Science 320, 16471651 with oral 5-fluorouracil prodrugs. J. Pharmacol. Exp. Ther. 287,
32 Burke, C. et al. (2011) Bacterial community assembly based on 791799
functional genes rather than species. Proc. Natl. Acad. Sci. U.S.A. 57 Spencer, M.D. et al. (2011) Association between composition of the
108, 1428814293 human gastrointestinal microbiome and development of fatty liver
33 Cantarel, B.L. et al. (2012) Complex carbohydrate utilization by the with choline deficiency. Gastroenterology 140, 976986
healthy human microbiome. PLoS ONE 7, e28742 58 Bentley, D.R. et al. (2008) Accurate whole human genome sequencing
34 Rahija, R.J. (2007) Gnotobiotics, In The Mouse in Biomedical Research using reversible terminator chemistry. Nature 456, 5359
(2nd edn) (James, G.F. et al., eds), pp. 217233, Academic Press 59 Eid, J. et al. (2009) Real-time DNA sequencing from single polymerase
35 Gaboriau-Routhiau, V.R. et al. (2009) The key role of segmented molecules. Science 323, 133138
filamentous bacteria in the coordinated maturation of gut helper T 60 Rothberg, J.M. et al. (2011) An integrated semiconductor device
cell responses. Immunity 31, 677689 enabling non-optical genome sequencing. Nature 475, 348352
36 Faith, J.J. et al. (2010) Creating and characterizing communities of 61 Gilbert, J.A. et al. (2008) Detection of large numbers of novel sequences
human gut microbes in gnotobiotic mice. ISME J. 4, 10941098 in the metatranscriptomes of complex marine microbial communities.
37 Heijtz, R.D. et al. (2011) Normal gut microbiota modulates brain PLoS ONE 3, e3042
development and behavior. Proc. Natl. Acad. Sci. U.S.A. 108, 30473052 62 Booijink, C.C. et al. (2010) Metatranscriptome analysis of the human
38 Olszak, T. et al. (2012) Microbial exposure during early life has fecal microbiota reveals subject-specific expression profiles, with genes
persistent effects on natural killer T cell function. Science 336, 489493 encoding proteins involved in carbohydrate metabolism being
39 Ivanov, I.I. et al. (2008) Specific microbiota direct the differentiation of dominantly expressed. Appl. Environ. Microbiol. 76, 55335540
IL-17-producing T-helper cells in the mucosa of the small intestine. 63 Li, X. et al. (2011) A metaproteomic approach to study human
Cell Host Microbe 4, 337349 microbial ecosystems at the mucosal luminal interface. PLoS ONE
40 Atarashi, K. et al. (2008) ATP drives lamina propria TH17 cell 6, e26542
differentiation. Nature 455, 808812 64 Verberkmoes, N.C. et al. (2009) Shotgun metaproteomics of the human
41 Garrett, W.S. et al. (2010) Enterobacteriaceae act in concert with the distal gut microbiota. ISME J. 3, 179189
gut microbiota to induce spontaneous and maternally transmitted 65 Wikoff, W.R. et al. (2009) Metabolomics analysis reveals large effects of
colitis. Cell Host Microbe 8, 292300 gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci.
42 Turnbaugh, P.J. et al. (2009) The effect of diet on the human gut U.S.A. 106, 36983703
microbiome: a metagenomic analysis in humanized gnotobiotic mice. 66 Jansson, J. et al. (2009) Metabolomics reveals metabolic biomarkers of
Sci. Transl. Med. 1, 6ra14 Crohns disease. PLoS ONE 4, e6386
43 Wu, G.D. et al. (2011) Linking long-term dietary patterns with gut 67 Raes, J. and Bork, P. (2008) Molecular eco-systems biology: towards an
microbial enterotypes. Science 334, 105108 understanding of community function. Nat. Rev. Microbiol. 6, 693699
44 McNulty, N.P. et al. (2011) The impact of a consortium of fermented 68 Arumugam, M. et al. (2011) Enterotypes of the human gut microbiome.
milk strains on the gut microbiome of gnotobiotic mice and monozygotic Nature 473, 174180
twins. Sci. Transl. Med. 3, 106ra106 69 Huang, Y.J. et al. (2011) Airway microbiota and bronchial
45 Zhang, H. et al. (2009) Human gut microbiota in obesity and after hyperresponsiveness in patients with suboptimally controlled
gastric bypass. Proc. Natl. Acad. Sci. U.S.A. 106, 23652370 asthma. J. Allergy Clin. Immunol. 127, 372381
46 Samuel, B.S. and Gordon, J.I. (2006) A humanized gnotobiotic mouse 70 Kong, H.H. et al. (2012) Temporal shifts in the skin microbiome
model of host-archaeal-bacterial mutualism. Proc. Natl. Acad. Sci. associated with disease flares and treatment in children with atopic
U.S.A. 103, 1001110016 dermatitis. Genome Res. 22, 850859
47 Mazmanian, S.K. et al. (2005) An immunomodulatory molecule of 71 Marchesi, J.R. et al. (2011) Towards the human colorectal cancer
symbiotic bacteria directs maturation of the host immune system. microbiome. PLoS ONE 6, e20447
Cell 122, 107118 72 Hoppe, B. et al. (2011) Efficacy and safety of Oxalobacter formigenes to
48 Macpherson, A.J. et al. (2001) IgA responses in the intestinal mucosa reduce urinary oxalate in primary hyperoxaluria. Nephrol. Dial.
against pathogenic and non-pathogenic microorganisms. Microbes Transplant. 26, 36093615
Infect. 3, 10211035 73 Liu, B. et al. (2012) Deep sequencing of the oral microbiome reveals
49 Wu, H-J. et al. (2010) Gut-residing segmented filamentous bacteria drive signatures of periodontal disease. PLoS ONE 7, e37919
autoimmune arthritis via T helper 17 cells. Immunity 32, 815827 74 Fahlen, A. et al. (2012) Comparison of bacterial microbiota in skin
50 Khan, K.J. et al. (2011) Antibiotic therapy in inflammatory bowel biopsies from normal and psoriatic skin. Arch. Dermatol. Res. 304,
disease: a systematic review and meta-analysis. Am. J. 1522
Gastroenterol. 106, 661673 75 Segata, N. et al. (2012) Metagenomic microbial community profiling
51 Veiga, P. et al. (2010) Bifidobacterium animalis subsp. lactis fermented using unique clade-specific marker genes. Nat. Methods 9, 811814
milk product reduces inflammation by altering a niche for colitogenic 76 Stothard, P. et al. (2005) BacMap: an interactive picture atlas of
microbes. Proc. Natl. Acad. Sci. U.S.A. 107, 1813218137 annotated bacterial genomes. Nucleic Acids Res. 33, D317D320

58

Das könnte Ihnen auch gefallen