You are on page 1of 9

REVIEWS

Role of gut microbiota in atherosclerosis


Annika Lindskog Jonsson1 and Fredrik Bäckhed1,2
Abstract | Infections have been linked to the development of cardiovascular disease and
atherosclerosis. Findings from the past decade have identified microbial ecosystems residing
in different habitats of the human body that contribute to metabolic and cardiovascular-related
disorders. In this Review, we describe three pathways by which microbiota might affect
atherogenesis. First, local or distant infections might cause a harmful inflammatory response that
aggravates plaque development or triggers plaque rupture. Second, metabolism of cholesterol
and lipids by gut microbiota can affect the development of atherosclerotic plaques. Third,
diet and specific components that are metabolized by gut microbiota can have various effects
on atherosclerosis; for example, dietary fibre is beneficial, whereas the bacterial metabolite
trimethylamine-N‑oxide is considered harmful. Although specific bacterial taxa have been
associated with atherosclerosis, which is supported by increasing mechanistic evidence, several
questions remain to be answered to understand fully how the microbiota contributes to
atherosclerosis and cardiovascular disease. Such knowledge might pave the way for novel
diagnostics and therapeutics based on microbiota.

The human body hosts a vast number of micro­organisms studying the microbiota and its role in disease, it is impor-
including bacteria, viruses, protozoa, archaea, and fungi, tant to keep in mind Koch’s postulates, which describe
which constitute the commensal microbiota that mainly the criteria that should be fulfilled in order to deter-
resides in the gut. Although the number of bacterial mine a causative relationship between a microorganism
species varies between individuals, at higher taxo­nomic and a disease (BOX 1). Given that these postulates were
levels (such as phyla) the microbiota is similar between established in the late nineteenth century, they should
individuals. In contrast to other environments, the be adapted to incorporate the increased knowledge of
gut microbiota is dominated by two bacterial phyla, host–microorganism interaction, and should include
Bacteroidetes or Firmicutes, which constitute >90% of the altered microbiome rather than only one pathogenic
the taxa present in the human gut, with a lower abun- species. Different methods exist to study microbiota and
dance of Actinobacteria, Cyanobacteria, Fusobacteria, its effect on host metabolism, such as animal models and
Proteobacteria, and Verrumicrobia1,2 (FIG. 1). modern culture-independent techniques (including bac-
1
Wallenberg Laboratory Humans have co-evolved with these microorganisms terial 16S rRNA gene sequencing), which provide a huge
for Cardiovascular and in a mutualistic fashion. Whereas microorganisms benefit amount of data that need interpretation (BOX 2).
Metabolic Research,
from a constant supply of human substrates, the human Periodontitis and gut microbial metabolites have
Department of Molecular
and Clinical Medicine, hosts benefit from microbial activities such as production been shown to modulate CVD, although the under­
Institute of Medicine, of essential vitamins (such as vitamin B and K), diges- lying mechanisms still remain unclear. Atherosclerosis
University of Gothenburg, tion of carbohydrates, ‘education’ of the immune sys- is a complex disease involving both inflammatory and
Bruna Stråket 16, tem, and production of signalling molecules including metabolic pathways and might be modulated by the gut
41345 Gothenburg, Sweden.
2
Novo Nordisk Foundation
short-chain fatty acids and secondary bile acids. Bacterial microbiota. In this Review, we discuss the microbial-­
Center for Basic Metabolic genes (microbiome) in the gut outnumber human genes related processes that could affect the development of
Research, Section for by at least two orders of magnitude3. The microbiota can atherosclerosis, including the infection-induced inflam-
Metabolic Receptology and influence host physiology, but external factors such as matory response, altered host lipid metabolism, and the
Enteroendocrinology, Faculty
genetics, diet, pharmacological compounds, lifestyle, interaction between specific dietary components and
of Health Sciences, University
of Copenhagen, Copenhagen, and hygiene also affect the microbial composition4. microbial metabolites.
DK‑2200, Denmark. Despite these beneficial effects described above, stud-
Correspondence to F.B. ies in the past 10 years have demonstrated that the micro- Role of oral and gut microbiota in CVD
fredrik@wlab.gu.se biota is associated with several diseases including obesity The development of atherosclerotic plaques can be
doi:10.1038/nrcardio.2016.183 and type 2 diabetes mellitus as well as atherosclerosis and affected by a distant infection or by a direct infection
Published online 1 Dec 2016 cardiovascular disease (CVD) in humans. However, when of vessel wall cells. This mechanism is supported by

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points with this condition, the number of Lactobacillales and


the ratio of Firmicutes to Bacteroidetes increased com-
• Microorganisms that reside in the human body, the majority of which colonize the gut, pared with control, whereas the number of Bacteroidetes
might affect host physiology in various ways decreased16. These findings need to be corroborated
• Bacteria from the gut or the oral cavity might translocate to atherosclerotic plaques in independent cohorts, and mechanistic studies are
and could affect the development of atherosclerosis and cardiovascular disease required to determine whether the altered microbiota
• Microbial transplantations in mice influence diet-enhanced susceptibility to causes, or merely reflects, coronary artery disease.
atherosclerosis and thrombosis
• Dietary components can either alter the composition of gut microbiota or be processed Infection and inflammation
into metabolites that can delay or accelerate the development of atherosclerosis Infections can affect the development of atherosclero-
• Trimethylamine N‑oxide is a potentially harmful bacterial metabolite that influences sis via two predominant mechanisms: direct infection
cholesterol metabolism and thrombosis activity of vessel wall cells, which creates a site prone to lesion
• To determine the role of the (gut) microbiota in cardiovascular disease and development, or an indirect infection at a site distant
atherosclerosis, the underlying mechanisms need to be further elucidated from the atherosclerotic plaque, which activates the
immune system that in turn increases the systemic
inflammatory status.
the finding of bacterial DNA in the plaques5,6. Bacteria
observed in the atherosclerotic plaques are also found Plaque microbiota. The presence of bacterial DNA in
at other body sites, predominantly the oral cavity human atherosclerotic plaques has been demonstrated
and the gut, which might thus serve as reservoirs of with various techniques and in different patients. One
these potentially pathogenic microorganisms (FIG. 1). of the most common bacteria found in atherosclerotic
An established association exists between perio­dontal plaques is Chlamydia pneumoniae17,18, which causes
disease and CVD7 as well as between poor dental health respir­atory tract infection. Furthermore, C. pneumoniae
and acute myocardial infarction 8. In a large study has been detected in postmortem coronary arteries and
that included almost 12,000 participants, poor oral endarterectomy specimens of living patients, but this
hygiene was associated with an increased risk of fatal bacterium was not associated with histological assess-
and nonfatal CVD events (such as myocardial infarc- ment of plaque stability (or lesion severity) or clinical
tion, coronary artery bypass graft surgery, coronary parameters19–21. By contrast, C. pneumoniae correlates
angioplasty, stroke, and heart failure) and low-grade with cytotoxic CD8+ T cells in symptomatic plaques and
inflammation, as demonstrated by increased concentra- is associated with increased C‑reactive protein levels in
tions of C‑reactive protein and fibrinogen9. A study of serum22,23. In addition to C. pneumoniae, >50 bacterial
92 individ­uals demonstrated that patients with sympto- species were detected in atherosclerotic plaques, whereas
matic atherosclerosis (myocardial infarction or stroke) no bacteria were found in control tissues5.
had a higher abundance of Anaeroglobus in the oral Improved sequencing technology and bioinformatics
cavity (around 50% of patients) compared with healthy analyses of bacterial 16S rRNA genes have provided a
controls (around 25% of individ­uals)10. Several oral detailed view of the bacterial content of atherosclero­
bacteria detected in atherosclero­tic plaques, including tic plaques. Whereas the majority of bacterial DNA
Porphyromonas gingivalis and Aggregatibacter actino­ belongs to Firmicutes and Proteobacteria phyla 6,24
mycetemcomitans, led to increased lesions in animal (FIG. 1), bac­terial DNA from taxa belonging to poten-
models following oral or intravenous infection, which tially pathogenic families such as Helicobacteraceae and
could be explained by an increase in proatherogenic Neisseriaceae was shown to be more abundant in athero­
mediators11–13. Although the mechanism is unclear, sclerotic plaques of symptomatic patients than in those
these findings indicate that bacteria from the oral cavity of asymptomatic ones25. However, this finding needs
are associated with CVD and might trans­locate to the to be corroborated in larger studies. Given that athero-
vessel, where they might affect growth and stability of sclerotic plaques from asymptomatic patients could be
the atherosclerotic plaque. vulnerable and at risk of rupture, it would be interesting
In addition to the oral cavity, the gut is a likely to correlate plaque morphology with bacterial content.
source of microorganisms that could influence CVD. Although the presence of bacterial DNA in athero­
Using shotgun sequencing, faecal samples from sclerotic plaques is established, whether an infection
13  healthy individ­u als and 12 patients with symp- initiates or augments their development is uncertain.
tomatic atherosclero­s is (myocardial infarction or Taking into account Koch’s postulates, only a few infec-
cerebro­vascular events) were found to differ in several tious agents have been shown to be potentially associated
aspects. Indeed, the genus Collinsella was enriched in with atherosclerosis. Microorganisms such as A. actino­
patients with symptomatic atherosclerosis, whereas mycetemcomitans, C. pneumoniae, Helicobacter pylori,
Eubacterium and Roseburia were enriched in healthy and P. gingivalis might contribute to atherosclerosis by
controls14 . In the patients, the altered microbial com- increasing lesion areas in animal models26; however,
position was also associated with increased abundance these data need to be validated in humans.
of genes involved in inflammatory processes14. The
abundance of gut microorganisms such as Bacteroides, Infectious agents. Since the first half of the nineteenth
Clostridium, and Lactobacillales has been shown to pre- century, infections of either viral or bacterial origin
dict coronary artery disease15. Furthermore, in patients have been considered as a cause of atherosclerosis27.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

These infectious agents include bacteria such as C. pneu­ To test whether a causal relationship exists between
moniae and H. pylori, periodontal microorganisms, and bacterial infection and CVD, several randomized, placebo-­
viruses including cytomegalovirus, hepatitis viruses, controlled trials have investigated whether treatment
influenza viruses, and human i­ mmunodeficiency virus26. with antibiotics has beneficial effects (TABLE 1). Although
The potential association between acute infection these trials showed no benefit in patients with coronary
and cardiovascular events was investigated in a study artery disease31–36, negative outcomes in such studies are
that included >40,000 patients28. The risk of myo­cardial to some extent inconclusive as the treatment duration
infarction or stroke was found to be increased after might have been too short or, if pathogens initiated the
systemic infection of the respiratory tract and infec- disease process, the treatment might have been too late.
tion of the urinary tract. This effect was most prom- Furthermore, the antibiotics used in these studies do not
inent within the first days of infection, and the risk target all bacteria residing in the human body. Further
gradually decreased afterwards28. In a meta-analysis studies are thus required to investigate the involvement
including >10,000 patients, seroprevalence of C. pneu­ of bacteria in the initiation of the athero­sclerotic process;
moniae correlated with an increase in the levels of sev- however, obtaining such evidence might not be feasible
eral inflammatory markers, such as C‑reactive protein owing to the long experimental duration and the risk of
(measured by high-affinity assay), IL‑6, and fibrinogen, spreading antibiotic resistance.
compared with seronegative patients with atherosclero­ Long-term treatment with antibiotics might cause
sis29. By contrast, in a study involving 1,100 patients, adverse effects, given that their administration early
Escherichia coli-related gastroenteritis did not increase in life to mice alters commensal gut microbiota and
the risk of cardiovascular events (myocardial infarc- induces obesity and insulin resistance 37, which in
tion, stroke, or heart failure) compared with 11,000 humans can cause cardiovascular disease. Interestingly,
controls30. Thus, specific bacterial infections, followed antibiotic treatment also affects lipid and cholesterol
by activation of the immune system and increased lev- metabolism 37, but whether atherosclerosis is also
els of pro­atherogenic inflammatory molecules, might increased is unknown. Other studies have suggested that
contribute to plaque vulnerability and thrombus patients exposed to a large number of pathogens have
formation (FIG. 2). an increased risk of CVD27,38. This evidence could also
help to explain the lack of effect of antibiotics, given that
some bacteria are resistant, and viruses are not targeted
by antibiotic treatment39. Although no direct evidence
from large clinical studies indicates that bacteria cause
CVD, this h ­ ypothesis cannot be ruled out.
Firmicutes
• Gram-positive bacteria
• Most abundant phylum in the Toll-like receptors and atherosclerosis. Regardless of
gut and oral cavity, and also the infection site (local or systemic), the immune system
Proteobacteria present in atherosclerotic plaques
• Include the genera Anaeroglobus,
responds to microbial components, and several inflam-
• Gram-negative bacteria matory pathways are activated. One of these pathways
Clostridium, Eubacterium,
• Most abundant phylum Lactobacillales, and Roseburia
in atherosclerotic involves Toll-like receptors (TLRs). TLRs are a family
plaques • The ratio of Firmicutes to of highly conserved membrane pattern-recognition
Bacteroidetes in patients with
• Include the genera CAD is higher than in controls receptors, which have an important role in the immune
Chryseomonas and
Helicobacter host defence system that protects against invading
pathogens40,41. For example, stimulation with lipopoly-
Bacteroidetes saccharides (LPS) — potent endotoxins that are part of
• Gram-negative bacteria mainly the outer membrane of Gram-negative bacteria — leads
present in the gut, and to a lesser to the recruitment of adaptor proteins such as myeloid
extent, in the oral cavity and
Actinobacteria atherosclerotic plaques differentiation primary response protein MYD88 to the
• Gram-positive bacteria • Include the genus Bacteroides cytoplasmic domain of TLRs. The recruitment of adap-
• Found in atherosclerotic and the species Porphyromonas
plaques, the oral cavity, tor proteins triggers downstream signalling cascades that
gingivalis
and the gut • The number of Bacteroidetes lead to production of proinflammatory cytokines and
• Include the genus decreases in patients with CAD chemokines40. LPS induce a low-grade inflammatory
Collinsella compared with healthy individuals state and aggravate the progression of atherosclerosis42.
Moreover, saturated fatty acids also act as non-­microbial
ligands for TLR4. These lipids are associated with an
Figure 1 | Body sites of microbiota that influences atherosclerosis. Bacterial DNA altered microbiota, elevated plasma LPS levels, and
detected in atherosclerotic plaques might derive from bacteriaNature Reviews
residing | Cardiology
in the oral cavity metabolic disease42–44. Furthermore, genetic deletion
or the gut. The description of the dominant phylum in each body habitat is based on a of Tlr4 and/or Myd88 in Apoe−/− mice was associated
previous publication6. Firmicutes is the most abundant phyla in the gut and oral cavity,
with reduced macrophage inflammatory response and
whereas Proteobacteria is most abundant in plaques. The methods for microbiota
characterization and taxonomic profiling are described in BOX 2. Specific bacterial taxa aortic lesions45,46.
in atherosclerotic plaques are also present in the oral cavity or gut6, and altered microbial In agreement with the aforementioned animal stud-
composition in the gut is associated with symptomatic atherosclerosis14. Bacteria in ies, TLR4 expression is increased in human atherosclero­
plaques, oral cavity, or gut might initiate or augment atherosclerosis, but causal evidence tic lesions, primarily in macrophages47,48. However,
is lacking. CAD, coronary artery disease. a 2012 meta-analysis found no association between

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Asp299Gly, a known TLR4 polymorphism that dimin- Primary bile acids formed in the liver are conjugated to
ishes inflammatory responses49, and risk of athero- either taurine or glycine, whereas intestinal deconjuga-
sclerosis50. Although convincing animal data linking tion by bacterial bile salt hydrolase and further metab-
inflammatory signalling to atherosclerosis have been olism in the colon lead to the formation of secondary
obtained, evidence in humans remains scarce. bile acids. Germ-free mice have larger gallbladders and
higher levels of bile acids than those of conventionally
Microbial effects on lipid metabolism raised mice, including a differential bile acid profile
In addition to infections and bacteria in atherosclerotic characterized by increased prevalence of conjugated
plaques or other body sites, the gut microbiota also primary bile acids and absence of secondary bile acids58.
influences CVD through the regulation of host metabo- These data indicate the importance of gut microbial
lism, including cholesterol and lipid metabolism (FIG. 2). ­regulation of bile acid composition.
Indeed, gut microbiota is now thought to have a piv- Bile acids can act as signalling molecules that bind
otal role in low-grade inflammatory metabolic diseases to FXR, a nuclear receptor that regulates several physio­
such as obesity and diabetes51–53, which are associated logical processes such as glucose and lipid metabo-
with CVD54. lism57,59. In the liver, bile acid-mediated activation of
FXR regulates triglyceride metabolism, especially the
Microbiota-regulated plasma lipids in CVD. A few production of VLDL and de novo lipogenesis. However,
studies have investigated the gut microbiota and plasma FXR signalling in the small intestine has also been
lipids in relation to CVD. Patients with atherosclerosis demonstrated to regulate steatosis and obesity60–63. The
have altered lipid metabolism, and bacterial taxa in the relative effect of microbial-induced FXR signalling on
oral cavity and gut were found to correlate with plasma liver and small intestine needs to be clarified59.
cholesterol levels6, potentially serving as disease bio- Conflicting data exist on the effect of FXR on athero­
markers. In addition, bacterial richness (that is, n­ umber sclerosis development. Nevertheless, diet is an impor-
of bacterial taxa identified) and specific taxa were associ­ tant factor that needs to be taken into consideration.
ated with BMI and levels of triglycerides and HDL, FXR-deficient mice fed a chow diet have impaired HDL
whereas no such clear association was found with levels metabolism and reverse cholesterol transport, and, as a
of total cholesterol or LDL55. Compared with germ-free consequence, develop hypercholesterolaemia64. Other
mice, the levels of cholesterol and triglycerides in plasma studies that used FXR agonists in atherosclerosis-­
of conventionally raised mice are reduced, whereas they prone mice have also suggested a protective role of this
are increased in adipose tissue and liver56. These find- receptor in atherosclerosis65–67, and this effect could be
ings support a relationship between the composition weight-­dependent65. Accordingly, Apoe and Fxr double-­
of the gut microbiota and plasma levels of cholesterol knockout mice had a worse plasma lipid profile and
and lipids. larger athero­sclerotic lesions than Apoe−/− mice68. By con-
trast, deletion of Fxr in mice lacking apolipo­protein E
Bile acid signalling. The gut microbiota can affect lipid or the LDL receptor led to reduced aortic lesions and
and cholesterol metabolism through bile acids and the decreased plasma LDL cholesterol, suggesting a non-
bile acid receptor (also known as farnesoid X receptor, protective role for FXR69,70. In addition, bacteria not
FXR). Bile acids are important for absorption of dietary only influence bile acid signalling through FXR in the
lipids and fat-soluble vitamins. Bile acids are synthesized intestine and liver, but possibly also directly in the vas-
from cholesterol in the liver, stored in the gallbladder, culature, given that FXR has been shown to be expressed
and released into the intestine after ingestion of a meal57. in aortic vascular smooth muscle cells and endothelial
cells71–73. To date, the role of microbial regulation of bile
acids and FXR in athero­genesis is not fully understood,
Box 1 | Koch’s postulates and several Fxr-expressing tissues might be involved.
Koch’s postulates, developed in the late nineteenth century, describe four criteria Most current data are based on animal studies, and
that should be fulfilled in order to determine a causative relationship between a human data are scarce.
microorganism and a disease:
• The microorganism should be present in all cases of disease Diet and microbial metabolites
• The microorganism should not be found in healthy individuals The gut microbiota interacts with dietary components74,
• After isolation and propagation in culture, the microorganism should cause the and data obtained in the past 5 years suggest that micro-
disease in an infected organism bial metabolites can promote or prevent CVD (FIG. 2).
• The microorganism, reisolated from the infected organism, should be identified
as the original disease-causing agent Atherosclerosis-promoting diet. A large proportion of
Technical advances in microbiology have led to the understanding that a single humans have switched from a healthy diet to a Western
pathogen is not the only cause of a disease, as resistor microorganisms (that is, those diet rich in cholesterols, fats, and sugars. Several compo-
mediating resistance to disease) and microbial consortia are also important122. Thus, nents of the Western diet can promote inflammation75,
Koch’s postulates should be adapted to include a holistic view that takes into account and changes in dietary pattern have been linked to meta­
whole microbial communities and their interaction with pathogenic microorganisms. bolic diseases and atherogenesis54. Moreover, saturated
Furthermore, metabolic diseases are associated with altered microbial communities fats in the Western diet increase endotoxin activity
and with the interaction between microorganisms, so the contribution of individual in plasma compared with a healthy diet, which could
pathogens might be of secondary importance in these cases.
be attributed to a change in microbiota composition

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Methods to study the microbiota risk of CVD84–87. Accordingly, increased levels of TMAO
have been correlated with an increased risk of cardio-
The effect of bacteria on host metabolism can be studied using germ-free animals, vascular events88, prevalent CVD85,89,90, poor prognosis91,
usually mice, which are completely free of any germs in the gut and other body sites. and increased risk of death92. Moreover, TMAO levels
In contrast to conventionally raised animals, germ-free animals enable the investigation are higher in patients with chronic heart failure than in
of the microbiota’s effect on specific conditions. Germ-free animals can also be
healthy controls93.
colonized with specific bacteria to investigate microbial effects on host physiology,
and faecal transplantations are used to determine whether a trait is transmissible A study in mice published in 2015 revealed that a­ ortic
through an intact microbiota. An important consideration is that germ-free animals lesion size positively correlated with plasma TMAO but
have an immature immune system owing to the lack of microorganisms. An alternative inversely correlated with choline, and that the com-
model is the depletion of the gut microbiota by antibiotic treatment. This procedure bined variations of plasma metabolites accounted for
is methodologically easier than using germ-free animals, and it does not require up to 40% of the variation in lesion size94. Apoe−/− mice
germ-free facilities. However, antibiotics might cause adverse effects. treated with phosphatidylcholine metabolites resulted
Given that many bacteria in the gut have not yet been isolated and cultured, in increased aortic lesions, which was dependent on the
culture‑independent technologies are used to characterize the microbiota. These gut microbiota84. Microbial capacity to produce TMA
methodologies, which have revolutionized studies on microbial ecology, are generally seems to be important for atherosclerosis development;
based on microbial 16S rRNA gene sequencing to determine taxonomic profiles
indeed, the aortic lesions of Apoe−/− mice transplanted
including diversity and abundance. Shotgun metagenomic sequencing is used to
investigate the microbiome and provides information on functional capacity in addition with microbiota from high-TMAO-producing mouse
to taxonomy. RNA, proteins, and metabolites can be investigated using strains (C57BL/6J) are increased compared with those
metatranscriptomics, metaproteomics, and metabolomics, respectively, providing a of Apoe−/− mice transplanted with microbiota from
holistic view of host–microorganism interaction. However, it is essential to bear in mind low-TMAO-producing strains (NZW/LacJ)95. Human
that many studies to date have not demonstrated a causal relationship, but rather intestinal isolates have been screened to identify ­bacterial
associations, between bacteria and diseases. strains that can produce large amounts of TMAO96.
Nine bacterial strains with TMA-producing properties
in vitro led to accumulated plasma TMAO levels when
and/or increased translocation of microorganisms across transplanted into germ-free mice96. Taken together,
the intestinal b­ arrier52,76,77. In Ldlr−/− mice, Western diet these experimental studies in mice support the potential
induces changes in circulating metabolites and inflam- ­harmful effect of TMAO on the cardiovascular system.
matory markers, indicating an altered cholesterol To date, the mechanisms by which TMAO contrib-
homeo­stasis and oxidative stress, which could be partly utes to atherosclerosis are only partially understood
reversed by switching the mice to a fibre-rich chow diet78. and might involve multiple pathways. In choline-fed
Circulating LPS levels and aortic lesion sizes are also partly Apoe−/− mice, the number of macrophages in aortic
reduced through a microbial-­dependent mech­anism78,79, lesions as well as foam cell formation are reduced when
in agreement with the reduced TLR4‑dependent pro­ TMA prod­uction is suppressed by antibiotic treatment84.
inflammatory signalling discussed above. By contrast, By contrast, different concentrations of TMAO did
germ-free Apoe−/− mice fed a low-cholesterol diet had not affect foam cell formation in mouse macrophages
increased aortic lesion sizes compared with convention- in vitro97. Therefore, the role of TMAO in foam cell
ally raised mice, whereas no difference was found in mice ­formation needs to be clarified.
fed a high-cholesterol diet80. Accordingly, atherosclero- TMAO can also contribute to atherosclerosis by
sis development in germ-free Apoe−/− mice fed a Western inhibiting reverse cholesterol transport. However, the
diet was similar to that of conventionally raised mice81. mechanisms underlying such an effect are unclear, and
Thus, a diet-­dependent microbial mechanism might be might not involve changes in the expression of choles-
involved in the development of atherosclerosis but, on the terol transporters in macrophages85. TMAO was also
basis of the data discussed above, the microbiota does not shown to induce several atherosclerosis-­promoting
have a causal role in atherosclerosis. However, it cannot inflammatory proteins in vascular cells, including
be excluded that microbiota induces atherosclerosis and prostaglandin G/H synthase 2 (also known as cyclo­
CVD ­indirectly by causing chronic metabolic dysfunc- oxygenase 2), IL‑6, E‑selectin, and intra­cellular adhesion
tion or, as discussed below, by producing proatherogenic molecule 1 through activation of nuclear f­ actor‑­κB98.
molecules from d ­ ietary precursors82,83. In addition to atherosclerosis, TMAO might also
affect thrombosis as increased levels of TMAO are
TMAO. Dietary components are also involved in the associated with higher rates of thrombotic events and
atherogenic process. One such example is the proathero­ platelet activation99.
genic metabolite trimethylamine N‑oxide (TMAO) Sex might be an important factor to consider when
generated from microbial metabolism of phosphatidyl- studying TMAO synthesis, because expression of
choline, which is common in red meat, shellfish, and eggs dimethylaniline monooxygenase [N‑oxide-forming] 3
(FIG. 2). Phosphatidylcholine is metabolized by the gut (also known as flavin monooxygenase 3; FMO3) is
microbiota to generate trimethylamine (TMA), which is increased in human and mouse females compared
further processed in the liver by flavin mono­oxygenases, with males100. In agreement with these data, germ-free
leading to the production of TMAO. Plasma levels of female mice colonized with TMA-producing bacterial
phosphatidylcholine-related metabolites and other strains had host-related and sex-related higher levels of
TMAO-producing molecules, including l­ -carnitine and plasma TMAO and increased activity of hepatic FMO3
γ‑butyrobetaine, have been associated with an increased compared with male mice96. Knock down of Fmo3 in

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

different mouse strains results in decreased TMAO Pharmacological elimination or reduction of plasma
plasma levels, altered lipid and cholesterol metabolism, TMAO is a potential therapeutic approach to reduce
and reduced atherosclerotic lesion areas101–103. FMO3 athero­s clerotic cardiovascular events. However, the
is regulated by FXR, and injection of FXR ligands in resulting accumulation of TMA might induce trimethyl­
mice induces the expression of FMO3 and TMAO100, aminuria, also known as fish odour syndrome104. Thus,
thus potentially explaining the reduced atherosclerosis it would be preferable to target the microbiota and TMA
in Fxr-deficient Apoe−/− mice65–67. Furthermore, TMAO synthesis directly. Inhibition of choline trimethylamine-­
decreases the total pool of bile acid in chow-fed mice85. lyase, the microbial enzyme that converts choline to
Taken together, these data indicate that FMO3 is dif- TMA, was demonstrated to reduce TMAO plasma
ferentially regulated by sex, is a target of FXR, and is levels and aortic lesions in mice105, but data in humans
­important for lipid and cholesterol metabolism. are lacking. Similarly, the antiatherosclerotic molecule
resveratrol increases the abundance of bifidobacteria
and lactobacilli in Apoe−/− mice. This increase is associ-
ated with reduced TMAO plasma levels through inhib­
Microbiota ition of microbial TMA production, but the mechanisms
are unclear106.
Infection Metabolism Diet and microbial metabolites Although a number of studies have pointed to an
Invading Choline/carnitine Dietary fibre association between plasma levels of choline, TMAO,
bacteria Lipids
and CVD88–93, available data are still inconsistent. After
bariatric surgery, increased plasma levels of TMAO are
LPS observed despite an improved metabolic profile, which
could be explained by increased intestinal abundance
Choline
Cholesterol TMA-lyase Short-chain of Pseudomonas107. Furthermore, Apoe−/− mice express-
TMA
Macrophage fatty acids: ing human cholesteryl ester transfer protein, which is
• Acetate essential for reverse cholesterol transport, were used
• Butyrate
• Propionate to investigate the role of l-carnitine, which can also be
Proatherogenic FXR
↑ FMO3 TMAO metabolized to TMAO and promote atherosclerosis85.
cytokines
Cyp7a1 ↓ RCT Mice fed a Western diet supplemented with l-carnitine
Cyp27a1 ↑ Foam cells had increased TMAO plasma levels and decreased aortic
Platelet ↓ Systemic
Foam cell ↓ Bile inflammation lesion sizes, with no changes in plasma lipid profile97,
activation
acids ? thus suggesting a protective role for l-carnitine and
TMAO in atherosclerosis. Why l­ -carnitine feeding
gave rise to opposite phenotypes in two different lab-
oratories is currently unclear, although these studies
Plaque rupture used different animal models (mice fed a chow diet85
Atherosclerosis or a Western diet97).
Figure 2 | Microbiota-related pathways in atherosclerosis. Microbiota can affect
Nature Reviews
atherosclerosis by three different pathways. First, bacterial infection activates| Cardiology
the Dietary prevention of atherosclerosis. Diet modula-
immune system causing a harmful inflammatory response. Both local infections by tion with specific dietary components might also be
bacteria invading the atherosclerotic plaque and distant infection can lead to a used to prevent atherosclerosis. However, most studies
proatherogenic response. Infections lead to an increase in proinflammatory cytokines have prov­ided only association data, and mechanistic
and chemokines, which could be mediated by Toll-like receptor 4 expressed in evidence is lacking. Dietary fibre is thought to provide
macrophages. The proinflammatory response, regardless of bacterial infection site health benefits as fibre is considered to lower the risk
(vessel wall or distant site), might affect the progression of atherosclerosis and of CVD. In humans, this effect could be attributed to a
potentially also vulnerability of the plaque. Second, microbiota can influence decreased level of plasma LDL108. Moreover, in healthy
atherosclerosis through altered cholesterol metabolism. The microbiota alters the levels
people, an inverse correlation exists between dietary
of serum triglycerides and cholesterol, and bacterial taxa in the oral cavity and gut
correlate with serum cholesterol6. Gut bacteria are also important in modifying bile acids, fibre and CVD mortality109. However, a limited number
which function as signalling molecules through the farnesoid X receptor (FXR). of human studies have been performed in this field, and
Activation of FXR induces flavin monooxygenase 3 (FMO3) in the liver; this enzyme the effects of diet­ary fibre might be indirectly related to
converts trimethylamine (TMA) to trimethylamine N‑oxide (TMAO). Third, specific CVD through the regulation of glucose metabolism.
dietary  components and microbial metabolites can lead to the production of both Prebiotics are plant-derived fibres that improve the
beneficial and harmful molecules. These pathways might contribute to the development composition or function of the gut microbiota. Indeed,
of atherosclerotic plaques and might also augment the disease, causing plaque rupture Apoe−/− mice fed inulin, a prebiotic polysaccharide, have
and thrombosis. Increased serum levels of TMAO, a bacterial-derived metabolite from reduced cholesterol levels and atherosclerotic lesions
choline and carnitine, are associated with CVD. Some effects of TMAO are directly compared with control mice110. Probiotics are live bac-
related to atherogenesis, such as the inhibition of reverse cholesterol transport (RCT)
teria with health benefits. Lactobacilli are common pro­
and the increase in foam cell formation, and indirect metabolic effects such as the
inhibition of enzymes involved in bile acid synthesis (cholesterol 7α‑monooxygenase biotics that have been evaluated in CVD animal models
[Cyp7a1] and sterol 26‑hydroxylase, mitochondrial [Cyp27a1])85 might also affect plaque with various outcomes, probably owing to strain-specific
development. The effects of dietary fibre and the bacteria-induced formation of effects. For example, consumption of a specific strain
short-chain fatty acids such as acetate, butyrate, and propionate are not clear, but might of Lactobacillus plantarum had beneficial effects in a
involve an overall reduction in systemic inflammation. LPS, lipopolysaccharide. small number of patients, including increased intestinal

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Clinical trials on antibiotic treatment for cardiovascular disease


Antibiotic Number Groups compared End points Conclusion
(treatment of
duration) patients
Azithromycin 7,747 Patients with CAD, serum-positive to Death, nonfatal No significant risk
(12 weeks)33 Chlamydia pneumoniae, treated with reinfarction, coronary reduction
antibiotic or placebo revascularization, angina
Azithromycin 4,012 Patients with CAD, serum-positive Death from CAD, No benefit
(1 year)34 and serum-negative to C. pneumoniae, nonfatal MI, coronary
treated with antibiotic or placebo revascularization, angina
Gatifloxacin 4,162 Patients with CAD, serum-positive Death, MI, angina, No benefit
(~2 years)35 and serum-negative to C. pneumoniae, revascularization, stroke
treated with antibiotic or placebo
Clarithromycin 4,373 Patients with CAD, serum-positive Mortality, MI, angina All-cause mortality
(2 weeks)36 and serum-negative to was higher in the
C. pneumoniae, treated with antibiotic antibiotic group than
or placebo in the placebo group
as a result of higher
CAD mortality
CAD, coronary artery disease; MI, myocardial infarction.

bacterial diversity111, and decreased inflammatory risk development via activation of the immune system,
factors for atherosclerosis112. Furthermore, although alteration of cholesterol metabolism, and production of
some studies showed that lactobacilli reduce blood cho- bacterial metabolites (such as TMAO; FIG. 2). Together,
lesterol levels in mice and humans113–115, three different these processes might initiate, but more likely sustain
strains of Lactobacillus reuteri were shown to have no and aggrav­ate, an ongoing process of plaque growth
effect on cholesterol levels or atherosclerotic lesions in and rupture. Specific bacterial infections seem to be of
Apoe−/− mice116,117. minor importance in atherosclerosis progression and the
An example of a bacterium with beneficial effects presence of bacterial DNA in the athero­sclerotic plaque
on atherosclerosis is Akkermansia muciniphila, which might reflect instead a systemic infection and/or bacterial
is part of the normal microbiota and is important in uptake by macrophages. However, ­bacterial ­infections
protecting the intestinal barrier in diet-induced obe- might contribute to plaque rupture.
sity118–120. Apoe−/− mice fed a Western diet supplemented Whether CVD and atherosclerosis can be success-
with A. muciniphila have reduced atherosclerotic lesions, fully treated by targeting the microbiota is still not clear.
an effect that could be attributed to an improved intes- Long-term treatment with antibiotics is unfeasible and
tinal barrier and inflammatory profile121. To summar­ could cause severe adverse effects. Although adherence
ize, data from mouse studies are conflicting, possibly to strict diets is generally low, dietary interventions,
because of different bacterial strains used. Furthermore, for example with fibre, might be more practical in
different dietary sources might explain the discrepancies atherosclerosis prevention compared with antibiotics.
between various studies. Large studies in patients with Modulation of microbiota composition or the regulation
atherosclerosis are needed to determine any potential of pathways that produce proatherogenic metabolites,
beneficial effect of probiotics. The modification of bac- such as TMAO and other molecules, might also be novel
terial composition through specific dietary components, therapeutic strategies.
or regulation of microbial metabolite formation, might New insights into how atherosclerosis can be affected
have long-term effects on the development of athero- by commensal microbiota have been provided over the
sclerosis, whereas short-term treatments might not be past decade. However, the interaction between host and
clinically effective. microorganisms throughout the human lifetime is a
crucial factor in the slow progression of atherosclerosis.
Conclusions The challenge is to determine whether specific bacterial
Atherosclerosis is a complex disease, and character- taxa and/or metabolites are the cause or consequence
izing the underlying mechanisms is, therefore, diffi- of the disease. To answer this question and to deter-
cult. However, increasing evidence indicates that the mine the disease mechanisms, linking the microbiota
micro­biota has a role in this disease. The microbiota (or microbial communities) to specific functions is of
might influence atherosclerosis by promoting plaque critical importance.

1. Human Microbiome Project Consortium. Structure, 3. Li, J. et al. An integrated catalog of reference genes 5. Ott, S. J. et al. Detection of diverse bacterial signatures
function and diversity of the healthy human in the human gut microbiome. Nat. Biotechnol. 32, in atherosclerotic lesions of patients with coronary
microbiome. Nature 486, 207–214 (2012). 834–841 (2014). heart disease. Circulation 113, 929–937 (2006).
2. Qin, J. et al. A human gut microbial gene catalogue 4. Sommer, F. & Backhed, F. The gut microbiota 6. Koren, O. et al. Human oral, gut, and plaque
established by metagenomic sequencing. Nature 464, — masters of host development and physiology. microbiota in patients with atherosclerosis. Proc. Natl
59–65 (2010). Nat. Rev. Microbiol. 11, 227–238 (2013). Acad. Sci. USA 108 (Suppl. 1), 4592–4598 (2011).

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

7. Trevisan, M. & Dorn, J. The relationship between 30. Hizo-Abes, P. et al. Cardiovascular disease after 55. Fu, J. et al. The gut microbiome contributes to a
periodontal disease (PD) and cardiovascular disease Escherichia coli O157:H7 gastroenteritis. CMAJ 185, substantial proportion of the variation in blood lipids.
(CVD). Mediterr. J. Hematol. Infect. Dis. 2, e2010030 E70–E77 (2013). Circ. Res. 117, 817–824 (2015).
(2010). 31. Andraws, R., Berger, J. S. & Brown, D. L. Effects 56. Velagapudi, V. R. et al. The gut microbiota modulates
8. Mattila, K. J. et al. Association between dental health of antibiotic therapy on outcomes of patients host energy and lipid metabolism in mice. J. Lipid Res.
and acute myocardial infarction. BMJ 298, 779–781 with coronary artery disease: a meta-analysis of 51, 1101–1112 (2010).
(1989). randomized controlled trials. JAMA 293, 2641–2647 57. Lefebvre, P., Cariou, B., Lien, F., Kuipers, F.
9. de Oliveira, C., Watt, R. & Hamer, M. Toothbrushing, (2005). & Staels, B. Role of bile acids and bile acid receptors
inflammation, and risk of cardiovascular disease: 32. Grayston, J. T. Antibiotic treatment of atherosclerotic in metabolic regulation. Physiol. Rev. 89, 147–191
results from Scottish Health Survey. BMJ 340, c2451 cardiovascular disease. Circulation 107, 1228–1230 (2009).
(2010). (2003). 58. Sayin, S. I. et al. Gut microbiota regulates bile acid
10. Fak, F., Tremaroli, V., Bergstrom, G. & Backhed, F. 33. O’Connor, C. M. et al. Azithromycin for the secondary metabolism by reducing the levels of tauro-beta-
Oral microbiota in patients with atherosclerosis. prevention of coronary heart disease events: the muricholic acid, a naturally occurring FXR antagonist.
Atherosclerosis 243, 573–578 (2015). WIZARD study: a randomized controlled trial. JAMA Cell Metab. 17, 225–235 (2013).
11. Hayashi, C. et al. Pathogen-mediated inflammatory 290, 1459–1466 (2003). 59. Wahlstrom, A., Sayin, S. I., Marschall, H. U.
atherosclerosis is mediated in part via Toll-like 34. Grayston, J. T. et al. Azithromycin for the secondary & Backhed, F. Intestinal crosstalk between bile acids
receptor 2‑induced inflammatory responses. J. Innate prevention of coronary events. N. Engl. J. Med. 352, and microbiota and its impact on host metabolism.
Immun. 2, 334–343 (2010). 1637–1645 (2005). Cell Metab. 24, 41–50 (2016).
12. Hayashi, C. et al. Porphyromonas gingivalis accelerates 35. Cannon, C. P. et al. Antibiotic treatment of Chlamydia 60. Li, F. et al. Microbiome remodelling leads to inhibition
inflammatory atherosclerosis in the innominate artery pneumoniae after acute coronary syndrome. N. Engl. of intestinal farnesoid X receptor signalling and
of ApoE deficient mice. Atherosclerosis 215, 52–59 J. Med. 352, 1646–1654 (2005). decreased obesity. Nat. Commun. 4, 2384 (2013).
(2011). 36. Jespersen, C. M. et al. Randomised placebo controlled 61. Jiang, C. et al. Intestinal farnesoid X receptor signaling
13. Zhang, T. et al. Aggregatibacter actinomycetemcomitans multicentre trial to assess short term clarithromycin promotes nonalcoholic fatty liver disease. J. Clin.
accelerates atherosclerosis with an increase in for patients with stable coronary heart disease: Invest. 125, 386–402 (2015).
atherogenic factors in spontaneously hyperlipidemic CLARICOR trial. BMJ 332, 22–27 (2006). 62. Jiang, C. et al. Intestine-selective farnesoid X receptor
mice. FEMS Immunol. Med. Microbiol. 59, 143–151 37. Cho, I. et al. Antibiotics in early life alter the murine inhibition improves obesity-related metabolic
(2010). colonic microbiome and adiposity. Nature 488, dysfunction. Nat. Commun. 6, 10166 (2015).
14. Karlsson, F. H. et al. Symptomatic atherosclerosis 621–626 (2012). 63. Parseus, A. et al. Microbiota-induced obesity requires
is associated with an altered gut metagenome. 38. Elkind, M. S. et al. Infectious burden and carotid farnesoid X receptor. Gut http://dx.doi.org/10.1136/
Nat. Commun. 3, 1245 (2012). plaque thickness: the northern Manhattan study. gutjnl-2015-310283 (2016).
15. Emoto, T. et al. Characterization of gut microbiota Stroke 41, e117–e122 (2010). 64. Lambert, G. et al. The farnesoid X‑receptor is an
profiles in coronary artery disease patients using data 39. Epstein, S. E., Zhu, J., Najafi, A. H. & Burnett, M. S. essential regulator of cholesterol homeostasis. J. Biol.
mining analysis of terminal restriction fragment length Insights into the role of infection in atherogenesis Chem. 278, 2563–2570 (2003).
polymorphism: gut microbiota could be a diagnostic and in plaque rupture. Circulation 119, 3133–3141 65. Miyazaki-Anzai, S., Masuda, M., Levi, M.,
marker of coronary artery disease. Heart Vessels (2009). Keenan, A. L. & Miyazaki, M. Dual activation of the
http://dx.doi.org/10.1007/s00380-016-0841-y 40. Akira, S., Uematsu, S. & Takeuchi, O. Pathogen bile acid nuclear receptor FXR and G‑protein-coupled
(2016). recognition and innate immunity. Cell 124, 783–801 receptor TGR5 protects mice against atherosclerosis.
16. Emoto, T. et al. Analysis of gut microbiota in coronary (2006). PLoS ONE 9, e108270 (2014).
artery disease patients: a possible link between gut 41. Akira, S. & Takeda, K. Toll-like receptor signalling. 66. Hartman, H. B. et al. Activation of farnesoid X receptor
microbiota and coronary artery disease. J. Atheroscler. Nat. Rev. Immunol. 4, 499–511 (2004). prevents atherosclerotic lesion formation in LDLR−/−
Thromb. 23, 908–921 (2016). 42. Ostos, M. A., Recalde, D., Zakin, M. M. and apoE−/− mice. J. Lipid Res. 50, 1090–1100
17. Shor, A., Kuo, C. C. & Patton, D. L. Detection of & Scott‑Algara, D. Implication of natural killer T cells (2009).
Chlamydia pneumoniae in coronary arterial fatty in atherosclerosis development during a LPS-induced 67. Mencarelli, A., Renga, B., Distrutti, E. & Fiorucci, S.
streaks and atheromatous plaques. S. Afr. Med. J. 82, chronic inflammation. FEBS Lett. 519, 23–29 (2002). Antiatherosclerotic effect of farnesoid X receptor.
158–161 (1992). 43. Rocha, D. M., Caldas, A. P., Oliveira, L. L., Bressan, J. Am. J. Physiol. Heart Circ. Physiol. 296, H272–H281
18. Blasi, F. et al. Detection of Chlamydia pneumoniae but & Hermsdorff, H. H. Saturated fatty acids trigger (2009).
not Helicobacter pylori in atherosclerotic plaques of TLR4‑mediated inflammatory response. 68. Hanniman, E. A., Lambert, G., McCarthy, T. C.
aortic aneurysms. J. Clin. Microbiol. 34, 2766–2769 Atherosclerosis 244, 211–215 (2016). & Sinal, C. J. Loss of functional farnesoid X receptor
(1996). 44. Caesar, R., Tremaroli, V., Kovatcheva-Datchary, P., increases atherosclerotic lesions in apolipoprotein
19. Thomas, M. et al. Relation between direct detection Cani, P. D. & Backhed, F. Crosstalk between gut E‑deficient mice. J. Lipid Res. 46, 2595–2604 (2005).
of Chlamydia pneumoniae DNA in human coronary microbiota and dietary lipids aggravates WAT 69. Guo, G. L. et al. Effects of FXR in foam-cell formation
arteries at postmortem examination and histological inflammation through TLR signaling. Cell Metab. 22, and atherosclerosis development. Biochim. Biophys.
severity (Stary grading) of associated atherosclerotic 658–668 (2015). Acta 1761, 1401–1409 (2006).
plaque. Circulation 99, 2733–2736 (1999). 45. Bjorkbacka, H. et al. Reduced atherosclerosis in 70. Zhang, Y. et al. FXR deficiency causes reduced
20. Gibbs, R. G. et al. Chlamydia pneumoniae does not MyD88‑null mice links elevated serum cholesterol atherosclerosis in Ldlr−/− mice. Arterioscler. Thromb.
influence atherosclerotic plaque behavior in patients levels to activation of innate immunity signaling Vasc. Biol. 26, 2316–2321 (2006).
with established carotid artery stenosis. Stroke 31, pathways. Nat. Med. 10, 416–421 (2004). 71. Bishop-Bailey, D., Walsh, D. T. & Warner, T. D.
2930–2935 (2000). 46. Michelsen, K. S. et al. Lack of Toll-like receptor 4 Expression and activation of the farnesoid X receptor
21. Berger, M. et al. Chlamydia pneumoniae DNA in or myeloid differentiation factor 88 reduces in the vasculature. Proc. Natl Acad. Sci. USA 101,
non‑coronary atherosclerotic plaques and circulating atherosclerosis and alters plaque phenotype in 3668–3673 (2004).
leukocytes. J. Lab. Clin. Med. 136, 194–200 mice deficient in apolipoprotein E. Proc. Natl Acad. 72. Zhang, Q. et al. FXR-mediated regulation of
(2000). Sci. USA 101, 10679–10684 (2004).
 angiotensin type 2 receptor expression in vascular
22. Nadareishvili, Z. G. et al. Increased CD8+ T cells 47. Edfeldt, K., Swedenborg, J., Hansson, G. K. smooth muscle cells. Cardiovasc. Res. 77, 560–569
associated with Chlamydia pneumoniae in & Yan, Z. Q. Expression of Toll-like receptors in human (2008).
symptomatic carotid plaque. Stroke 32, 1966–1972 atherosclerotic lesions: a possible pathway for plaque 73. He, F. et al. Downregulation of endothelin‑1 by
(2001). activation. Circulation 105, 1158–1161 (2002). farnesoid X receptor in vascular endothelial cells.
23. Johnston, S. C. et al. C‑reactive protein levels and 48. Xu, X. H. et al. Toll-like receptor‑4 is expressed Circ. Res. 98, 192–199 (2006).
viable Chlamydia pneumoniae in carotid artery by macrophages in murine and human lipid-rich 74. Sonnenburg, J. & Bäckhed, F. Diet-microbiota
atherosclerosis. Stroke 32, 2748–2752 (2001). atherosclerotic plaques and upregulated by oxidized interactions as moderators of human metabolism.
24. Calandrini, C. A. et al. Microbial composition of LDL. Circulation 104, 3103–3108 (2001). Nature 535, 56–64 (2016).
atherosclerotic plaques. Oral Dis. 20, e128–e134 49. Arbour, N. C. et al. TLR4 mutations are associated 75. Tilg, H. & Moschen, A. R. Food, immunity, and
(2014). with endotoxin hyporesponsiveness in humans. the microbiome. Gastroenterology 148, 1107–1119
25. Mitra, S. et al. In silico analyses of metagenomes from Nat. Genet. 25, 187–191 (2000). (2015).
human atherosclerotic plaque samples. Microbiome 3, 50. Zhang, K. et al. Lack of association between TLR4 76. Pendyala, S., Walker, J. M. & Holt, P. R. A high-fat diet
38 (2015). Asp299Gly polymorphism and atherosclerosis: is associated with endotoxemia that originates from
26. Rosenfeld, M. E. & Campbell, L. A. Pathogens and evidence from meta-analysis. Thromb. Res. 130, the gut. Gastroenterology 142, 1100–1101.e2
atherosclerosis: update on the potential contribution e203–208 (2012). (2012).
of multiple infectious organisms to the pathogenesis 51. Backhed, F. et al. The gut microbiota as an 77. Erridge, C., Attina, T., Spickett, C. M. & Webb, D. J.
of atherosclerosis. Thromb. Haemost. 106, 858–867 environmental factor that regulates fat storage. A high-fat meal induces low-grade endotoxemia:
(2011). Proc. Natl Acad. Sci. USA 101, 15718–15723 (2004). evidence of a novel mechanism of postprandial
27. Epstein, S. E. et al. Infection and atherosclerosis: 52. Cani, P. D. et al. Metabolic endotoxemia initiates inflammation. Am. J. Clin. Nutr. 86, 1286–1292
potential roles of pathogen burden and molecular obesity and insulin resistance. Diabetes 56, (2007).
mimicry. Arterioscler Thromb. Vasc. Biol. 20, 1761–1772 (2007). 78. Li, D. et al. Metabonomic changes associated
1417–1420 (2000). 53. Karlsson, F. H. et al. Gut metagenome in European with atherosclerosis progression for LDLR−/− mice.
28. Smeeth, L. et al. Risk of myocardial infarction and women with normal, impaired and diabetic glucose J. Proteome Res. 14, 2237–2254 (2015).
stroke after acute infection or vaccination. N. Engl. control. Nature 498, 99–103 (2013). 79. Ghosh, S. S., Bie, J., Wang, J. & Ghosh, S. Oral
J. Med. 351, 2611–2618 (2004). 54. Yusuf, S., Reddy, S., Ounpuu, S. & Anand, S. Global supplementation with non-absorbable antibiotics
29. Filardo, S., Di Pietro, M., Farcomeni, A., Schiavoni, G. burden of cardiovascular diseases: part I: general or curcumin attenuates western diet-induced
& Sessa, R. Chlamydia pneumoniae-mediated considerations, the epidemiologic transition, risk atherosclerosis and glucose intolerance in LDLR−/−
inflammation in atherosclerosis: a meta-analysis. factors, and impact of urbanization. Circulation 104, mice—role of intestinal permeability and macrophage
Mediators Inflamm. 2015, 378658 (2015). 2746–2753 (2001). activation. PLoS ONE 9, e108577 (2014).

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

80. Stepankova, R. et al. Absence of microbiota (germ-free accumulation of the proatherogenic metabolite reuteri on the prevention of hypercholesterolemia
conditions) accelerates the atherosclerosis in trimethylamine-N‑oxide. mBio 6, e02481 (2015). in mice. J. Dairy Sci. 83, 401–403 (2000).
ApoE‑deficient mice fed standard low cholesterol diet. 97. Collins, H. L. et al. l‑carnitine intake and high 114. Andrade, S. & Borges, N. Effect of fermented milk
J. Atheroscler. Thromb. 17, 796–804 (2010). trimethylamine N‑oxide plasma levels correlate containing Lactobacillus acidophilus and
81. Wright, S. D. et al. Infectious agents are not necessary with low aortic lesions in ApoE−/− transgenic mice Bifidobacterium longum on plasma lipids of women
for murine atherogenesis. J. Exp. Med. 191, expressing CETP. Atherosclerosis 244, 29–37 (2016). with normal or moderately elevated cholesterol.
1437–1442 (2000). 98. Seldin, M. M. et al. Trimethylamine N‑oxide promotes J. Dairy Res. 76, 469–474 (2009).
82. O’Connor, A., Quizon, P. M., Albright, J. E., Lin, F. T. vascular inflammation through signaling of mitogen- 115. London, L. E. et al. Exopolysaccharide-producing
& Bennett, B. J. Responsiveness of cardiometabolic- activated protein kinase and nuclear factor-κB. J. Am. probiotic Lactobacilli reduce serum cholesterol and
related microbiota to diet is influenced by host Heart Assoc. 5, e002767 (2016). modify enteric microbiota in ApoE-deficient mice.
genetics. Mamm. Genome 25, 583–599 (2014). 99. Zhu, W. et al. Gut microbial metabolite TMAO J. Nutr. 144, 1956–1962 (2014).
83. Org, E. et al. Genetic and environmental control enhances platelet hyperreactivity and thrombosis risk. 116. Fak, F. & Backhed, F. Lactobacillus reuteri prevents
of host-gut microbiota interactions. Genome Res. 25, Cell 165, 111–124 (2016). diet-induced obesity, but not atherosclerosis, in a
1558–1569 (2015). 100. Bennett, B. J. et al. Trimethylamine-N‑oxide, strain dependent fashion in Apoe−/− mice. PLoS ONE
84. Wang, Z. et al. Gut flora metabolism of a metabolite associated with atherosclerosis, exhibits 7, e46837 (2012).
phosphatidylcholine promotes cardiovascular disease. complex genetic and dietary regulation. Cell Metab. 117. Portugal, L. R. et al. Effect of Lactobacillus delbrueckii
Nature 472, 57–63 (2011). 17, 49–60 (2013). on cholesterol metabolism in germ-free mice and on
85. Koeth, R. A. et al. Intestinal microbiota metabolism 101. Shih, D. M. et al. Flavin containing monooxygenase 3 atherogenesis in apolipoprotein E knock-out mice.
of l‑carnitine, a nutrient in red meat, promotes exerts broad effects on glucose and lipid metabolism Braz. J. Med. Biol. Res. 39, 629–635 (2006).
atherosclerosis. Nat. Med. 19, 576–585 (2013). and atherosclerosis. J. Lipid Res. 56, 22–37 (2015). 118. Collado, M. C., Derrien, M., Isolauri, E., de Vos, W. M.
86. Koeth, R. A. et al. γ-butyrobetaine is a proatherogenic 102. Warrier, M. et al. The TMAO-generating enzyme flavin & Salminen, S. Intestinal integrity and Akkermansia
intermediate in gut microbial metabolism of monooxygenase 3 is a central regulator of cholesterol muciniphila, a mucin-degrading member of the
l‑carnitine to TMAO. Cell Metab. 20, 799–812 balance. Cell Rep. 10, 326–338 (2015). intestinal microbiota present in infants, adults, and
(2014). 103. Miao, J. et al. Flavin-containing monooxygenase 3 the elderly. Appl. Environ. Microbiol. 73, 7767–7770
87. Skagen, K. et al. The carnitine–butyrobetaine– as a potential player in diabetes-associated (2007).
trimethylamine-N‑oxide pathway and its association atherosclerosis. Nat. Commun. 6, 6498 (2015). 119. Derrien, M., Collado, M. C., Ben-Amor, K.,
with cardiovascular mortality in patients with carotid 104. Cashman, J. R. et al. Biochemical and clinical aspects Salminen, S. & de Vos, W. M. The mucin degrader
atherosclerosis. Atherosclerosis 247, 64–69 (2016). of the human flavin-containing monooxygenase form 3 Akkermansia muciniphila is an abundant resident of
88. Tang, W. H. et al. Intestinal microbial metabolism of (FMO3) related to trimethylaminuria. Curr. Drug the human intestinal tract. Appl. Environ. Microbiol.
phosphatidylcholine and cardiovascular risk. N. Engl. Metab. 4, 151–170 (2003). 74, 1646–1648 (2008).
J. Med. 368, 1575–1584 (2013). 105. Wang, Z. N. et al. Non-lethal inhibition of gut microbial 120. Everard, A. et al. Cross-talk between Akkermansia
89. Wang, Z. et al. Prognostic value of choline and trimethylamine production for the treatment of muciniphila and intestinal epithelium controls
betaine depends on intestinal microbiota-generated atherosclerosis. Cell 163, 1585–1595 (2015). diet‑induced obesity. Proc. Natl Acad. Sci. USA 110,
metabolite trimethylamine-N‑oxide. Eur. Heart J. 35, 106. Chen, M. L. et al. Resveratrol attenuates 9066–9071 (2013).
904–910 (2014). trimethylamine-N‑Oxide (TMAO)-induced 121. Li, J., Lin, S., Vanhoutte, P. M., Woo, C. W. & Xu, A.
90. Mente, A. et al. The relationship between atherosclerosis by regulating TMAO synthesis and bile Akkermansia muciniphila protects against
trimethylamine-N‑oxide and prevalent cardiovascular acid metabolism via remodeling of the gut microbiota. atherosclerosis by preventing metabolic endotoxemia-
disease in a multiethnic population living in Canada. mBio 7, e02210-15 (2016). induced inflammation in Apoe−/− Mice. Circulation
Can. J. Cardiol. 31, 1189–1194 (2015). 107. Tremaroli, V. et al. Roux‑en‑Y gastric bypass and 133, 2434–2446 (2016).
91. Tang, W. H. et al. Intestinal microbiota-dependent vertical banded gastroplasty induce long-term 122. Byrd, A. L. & Segre, J. A. Infectious disease. Adapting
phosphatidylcholine metabolites, diastolic dysfunction, changes on the human gut microbiome contributing to Koch’s postulates. Science 351, 224–226 (2016).
and adverse clinical outcomes in chronic systolic heart fat mass regulation. Cell Metab. 22, 228–238 (2015).
failure. J. Card. Fail. 21, 91–96 (2015). 108. Slavin, J. Fiber and prebiotics: mechanisms and health Acknowledgements
92. Tang, W. H. et al. Prognostic value of elevated levels benefits. Nutrients 5, 1417–1435 (2013). The authors thank Anna Hallén, Walleneberg Laboratory at
of intestinal microbe-generated metabolite 109. Sahyoun, N. R., Jacques, P. F., Zhang, X. L., Juan, W. University of Gothenburg, Sweden, for assistance with figures
trimethylamine-N‑oxide in patients with heart failure: & McKeown, N. M. Whole-grain intake is inversely and artwork. The authors’ laboratory is supported by grants
refining the gut hypothesis. J. Am. Coll. Cardiol. 64, associated with the metabolic syndrome and from AFA-insurances, the Ragnar Söderberg’s Foundation,
1908–1914 (2014). mortality in older adults. Am. J. Clin. Nutr. 83, the Swedish Foundation for Strategic Research, the Swedish
93. Troseid, M. et al. Microbiota-dependent metabolite 124–131 (2006). Heart Lung Foundation, the Swedish Research Council,
trimethylamine-N‑oxide is associated with disease 110. Rault-Nania, M. H. et al. Inulin attenuates Torsten Söderberg’s, and LUA-ALF grants from Västra
severity and survival of patients with chronic heart atherosclerosis in apolipoprotein E‑deficient mice. Götalandsregionen. F.B. is a recipient of ERC consolidator
failure. J. Intern. Med. 277, 717–726 (2015). Br. J. Nutr. 96, 840–844 (2006). Grant 2013 (European Research Council, Consolidator grant
94. Bennett, B. J. et al. Genetic architecture of 111. Karlsson, C. et al. Probiotic therapy to men with 615362-METABASE).
atherosclerosis in mice: a systems genetics analysis incipient arteriosclerosis initiates increased bacterial
of common inbred strains. PLoS Genet. 11, e1005711 diversity in colon: a randomized controlled trial. Author contributions
(2015). Atherosclerosis 208, 228–233 (2010). Both authors researched data for the article, discussed its
95. Gregory, J. C. et al. Transmission of atherosclerosis 112. Naruszewicz, M., Johansson, M. L., Zapolska- content, and wrote, reviewed, and edited the manuscript
susceptibility with gut microbial transplantation. Downar, D. & Bukowska, H. Effect of Lactobacillus before submission.
J. Biol. Chem. 290, 5647–5660 (2015). plantarum 299v on cardiovascular disease risk factors
96. Romano, K. A., Vivas, E. I., Amador-Noguez, D. in smokers. Am. J. Clin. Nutr. 76, 1249–1255 (2002). Competing interests statement
& Rey, F. E. Intestinal microbiota composition 113. Taranto, M. P., Medici, M., Perdigon, G., Ruiz F.B. is founder and shareholder of Metabogen AB. A.L.J.
modulates choline bioavailability from diet and Holgado, A. P. & Valdez, G. F. Effect of Lactobacillus declares no competing interests.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like