Gut Microbiom and Heart Disease

You might also like

You are on page 1of 49

The intestinal microbiota and cardiovascular disease

Themistoklis Katsimichas, MD, PhD,1,2 Alexios S. Antonopoulos, MD, PhD,1 Alexandros

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Katsimichas, MD, PhD,1 Tomohito Ohtani, MD, PhD,2 Yasushi Sakata, MD, PhD,2 Dimitris

Tousoulis, MD, PhD1

1 st
1 Cardiology Department, Athens Medical School, Hippokration General Hospital, Athens,

Greece.
2
Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine,

Suita, Osaka, Japan.

Category: Review.

Word count: 6,253 (main text), Figures: 5, Tables: 3, References: 107

Corresponding author:

Professor Dimitris Tousoulis, MD, PhD.

1st Cardiology Department, Athens Medical School, Hippokration General Hospital,

Vasilissis Sofias Avenue 114, PO 115 27, Athens, Greece.

Tel: (+30) 2132088099 Fax: (+30) 2132088676

Email: drtousoulis@hotmail.com

Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2019. For permissions please email:
journals.permissions@oup.com.

1
CVR-2019-0006-
R1

Abstract

The intestinal microbiota of human hosts is the community of microorganisms living in the

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


small and, mainly, the large intestine of humans. This microbial ecosystem has co-evolved with

humans across the millennia, has come to play an important interactive role in human

physiology, and has been aptly called our forgotten organ. Significant properties of the

microbiota benefiting its host include energy harvest from food sources indigestible by humans,

protection from pathogen colonization, and vitamin synthesis. Mounting evidence has linked

changes in the composition or metabolic profiles of the microbiota with human disease,

including disorders of the cardiovascular spectrum. Although cause and effect mechanisms are

as yet essentially unproven in the relevant literature, the established associations point to the

importance of the microbiota in the pathophysiology of cardiovascular disease. In this review,

we first summarize key information on the gut microbial communities and the elaborate tools

developed to analyze their structure and metabolic functions. Ecological terms are explained

and analytical techniques are simplified, to enhance the understanding of published studies.

Statistical methods used in microbial analysis are also described in simple terms. We then

present published literature on the association of the compositional and functional changes of

the microbiota in cardiovascular disease, including heart failure, hypertension, and

atherosclerosis. Each section of the review deals with the underlying pathophysiology of the

relevant associations, connecting the observational and mechanistic aspects. Finally, we

discuss the challenges that remain to be met before this field of research can generate

knowledge which can impact everyday clinical practice.

2
CVR-2019-0006-
R1

1. Introduction

The human intestinal microbiota is the community of microorganisms living in the human

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


intestinal tract, primarily the large intestine. This complex and diverse ecosystem comprises

symbiotic and mutualistic bacteria, fungi, viruses, and archaea, which colonize human hosts

from birth, have co-evolved with them across the millennia, and play important roles in several

host-related physiological functions.1.2 Such functions include the synthesis of important

vitamins, like vitamins K, B12, folate, and thiamine, contributed among others by species of

the genus Bifidobacterium, energy extraction from undigested fiber through bacterial

fermentation and production of the host-absorbable short-chain fatty acids propionate, acetate,

and butyrate, and metabolic processing of various non-dietary xenobiotic compounds, i.e.,

mainly hydrolysis and reduction by multiple bacterial species of non-human origin chemical

substances entering the gut through ingestion, either as pharmaceutical compounds or industrial

chemicals.1,2

Over 99% of the microbiota in the large intestine is composed of bacteria, mostly

anaerobic, with a total number of prokaryotic cells about equal to the total number of cells in

the human body.3-5 In recent years, this bacterial society of interdependent species has become

a center point of research, with multiple studies linking changes in gut microbiota synthesis

and diversity to a variety of human disorders.6

In this review, we focus on the associations of the intestinal microbiota with

cardiovascular disease (CVD). We first briefly overview important properties of the intestinal

microbiota and describe methods of microbial analysis. We go over the absolute basics of data

analysis to facilitate the understanding of study results. We then move to present key published

research in humans, connecting the microbiota with heart failure, hypertension, and

atherosclerosis (Table 1), and discuss remaining challenges to be met.

3
CVR-2019-0006-
R1

2. The microbial ecosystem of the human gut

The large intestine is a habitat for trillions of commensal or mutualistic microbes that form an

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


interdependent and interactive community of species termed the intestinal microbiota, or gut

flora. The microbiota plays a crucial role in many host-related functions, including but not

limited to the training and maturation of the host immune system, energy harvest through

fermentation of foods undigested by humans, vitamin synthesis, deconjugation of bile acids,

and, last but not least, resistance to colonization by or overgrowth of pathogenic

microorganisms (Figure 1).1,2,7

The composition of the intestinal microbiota is overwhelmingly dominated by

anaerobic bacteria and influenced by host age and genetics, geographic origin, and diet, among

others.3,4,8,9 Especially diet exerts considerable influence in the shaping of a bacterial

community. Human infants, relying on breast-fed milk for their dietary needs, have low

diversity microbiota dominated by various species of Bifidobacterium, which thrive in the

presence of human milk oligosaccharides.9 Older children and adults experience a growing

diversity of bacterial species, as breast feeding ceases and solid food is introduced into the diet.

Distinct dietary habits across different geographic regions may also explain variations in gut

bacterial community composition of diverse adult populations, e.g. between USA citizens and

rural Africans.9

In general, humans are immediately colonized at birth, the microbiota gradually

increases its diversity and, for the higher taxonomic levels, resembles an adult composition by

age 3, and it remains relatively stable until old age if no serious perturbation occurs, comprising

four major bacterial phyla (Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria) and

an average of 500 species per host.1,7,10-12 Although community synthesis is stable at the

phylum level and a core of bacterial genera is present in the majority of adult hosts, there is

huge variability at a subspecies level, to the point where a unique microbial synthesis can be

4
CVR-2019-0006-
R1

postulated for each individual.4,6 However, key microbial metabolic functions are present

throughout all species.13 That is, the most abundant metabolic pathways in terms of the number

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


of bacterial genes involved (e.g. vitamin biosynthesis, carbohydrate metabolism, or ATP

synthesis) are present in all bacteria and more or less equally prevalent among all gut microbial

communities, making the functional capacity of the intestinal microbiota much less variable

than its composition. Nevertheless, in terms of abundance, Bacteroides has been shown to be

the most abundant bacterial genus in the gut of western populations,13 while other important

genera include Prevotella and Ruminococcus.4,13

Bacteroides, an obligate anaerobic bacterial genus, makes up the greater portion of the

total microbial community and includes important and potentially infective species, such as

Bacteroides fragilis.3,13 Bacteria of this genus play crucial roles in fermenting plant fiber and

extracting energy usable by their host. Species of Prevotella are also well equipped to ferment

plant carbohydrates, and have been associated with fruit and vegetable-rich dietary patterns,14

colonizing the gut in an antagonistic struggle against species of Bacteroides. Members of

Ruminococcus, another anaerobic genus, also can ferment cellulose, pectin, and, starch,

contributing to the production of short-chain fatty acids.

Crucial to the field of intestinal microbial research are the potential ramifications that a

change in the composition or functions of the intestinal microbiota may bring about for its host.

Such departure from homeostasis has been termed “dysbiosis” and numerous lines of evidence

exist for compositional changes in a variety of human disorders, including CVD.6 A proposed

underlying mechanism posits that environmental triggers cause gut mucosal inflammation,

which in subjects predisposed to chronic disorders may lead to an increase in opportunistic

microbes and the transition to bacteria that, under certain physiological conditions, may

promote local or systemic pathogenetic functional changes in susceptible individuals.6

However, with few exceptions, current research generally remains far from establishing a cause

5
CVR-2019-0006-
R1

and effect relationship between changes in the composition and diversity of the microbiota and

human disease.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


3. Methods of microbial analysis

An in-depth coverage of all aspects of microbiome analysis is beyond the scope of this review.

A few important points should nevertheless be mentioned. After the collection of a sample and

appropriate extraction of bacterial DNA, the principal methods of analyzing the intestinal

microbial composition are either amplicon sequencing of the 16S ribosomal RNA (rRNA) gene,

or metagenomic processing of all the DNA strands recovered from the sample.15

Amplicon sequencing most commonly involves the identification and sequencing of

the 16S rRNA gene, the bacterial gene coding for the 16S rRNA component of the 30S small

subunit of the prokaryotic ribosome.16 This gene features highly preserved sequences present

in all bacteria, but also possesses multiple hypervariable regions showing differences between

different microorganisms. This unique feature makes it a prime target for both discovering all

bacteria in a sample and taxonomically differentiating between them.17 16S rRNA analysis is

a cheap, simple, and arguably accurate method of analysis, but yields a relatively low amount

of information, basically resulting in the identification and proportional calculation of bacterial

genera present in a sample.18

Metagenomic analysis, on the other hand, involves the direct, next-generation shotgun

sequencing of all the fragments of DNA recovered from all the organisms present in a sample.19

This is a much more expensive and complex method, but with a considerably higher yield of

functional information.4,19 Through appropriate techniques, bacterial species and strains

present in a sample can be identified and data on genes present in each bacterial organism can

be generated.19 This enables researchers to acquire information on important metabolic

6
CVR-2019-0006-
R1

pathways present in the bacterial community, thus highly facilitating a functional profiling of

the intestinal microbiota.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


4. Data analysis and visualization

The final output of the microbial composition analysis, generated through an appropriate

bioinformatics pipeline such as QIIME (Quantitative Insights Into Microbial Ecology),20 may

have the form of a typical high-dimensional spreadsheet (e.g. analyzed samples in rows and

the measured bacteria in hundreds of columns). Each cell will show a proportion, the percent

contribution of the specific bacterial taxon to the total of the bacterial community of a given

sample, called its relative abundance. For example, the relative abundance of Streptococcus in

sample 1 may be “α%”, meaning that α% of all prokaryotic cells present in sample 1 belong to

the genus of Streptococcus. This spreadsheet, and the preceding Operational Taxonomic Unit

(OTU) table [usually a non-human readable biological observation matrix (BIOM) file

constituting a part of the analysis pipeline], may form the basis for all subsequent analysis,20

which, at the very least, follows three basic pathways: the calculation of alpha diversity, the

calculation of beta diversity (both explained below), and a comparison of groups of subjects

by each bacterial taxon. It must be stressed that microbial relative abundance data are

multivariate (there is a large number of measured variables in each sample), that they almost

never follow a normal distribution, and that they are compositional in nature (they are

percentages that must total 100%). Extreme caution must thus be exercised on the choice of

statistical tests, as an inappropriate test will invariably lead to false conclusions.

Alpha diversity

Alpha diversity refers to specific characteristics of an ecological community, such as the

number of different species (richness) or how evenly the individual organisms are distributed

7
CVR-2019-0006-
R1

among the different species (evenness). Usually, diversity indices that incorporate and combine

this information, such as the Shannon index,21 can be calculated, averaged across subjects, and

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


compared between groups of subjects through a simple ANOVA or Student’s t-test. Generally

speaking, a lower alpha diversity can be thought of as a negative phenomenon in a given

ecosystem of interacting species. Although alpha diversity is a common tool of describing

ecological communities, collapsing the incredible bacterial variation to a single index is a

limited approach to comparing groups of samples.

Beta diversity, multivariate analysis, and predictive functional profiling

Beta diversity is a concept of direct comparison between different ecological communities. The

calculation of a measure of similarity, most commonly the Bray-Curtis similarity coefficient

or the UniFrac distance metric,22,23 makes possible the direct comparison of different bacterial

communities in different samples – e.g. “samples A and B are 65% similar based on their Bray-

Curtis similarity”. Multivariate permutational methods that can analyze the data en masse, such

as the non-parametric “analysis of similarities” (ANOSIM) and the semi-parametric

“permutational multivariate analysis of variance” (PERMANOVA),24,25 can then be applied to

decide whether the grouping of samples in, for example, “healthy controls” and “patients”

yields a statistically significant difference in the overall structure of the bacterial communities;

in other words, whether the “patient” samples are significantly more similar to each other than

they are to the “healthy control” samples. Importantly, these methods do not only constitute

tests for statistical significance, but also offer a measure of how big a difference between groups

really is, regardless of its statistical significance.

Of note, multivariate analysis is not restricted to comparing samples in terms of

bacterial relative abundance values, but can also be applied to gene content data, either

8
CVR-2019-0006-
R1

measured through next-generation sequencing or inferred by appropriate algorithms from 16S

rRNA data. A key such algorithm is PICRUSt, which stands for “Phylogenetic Investigation

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


of Communities by Reconstruction of Unobserved States”.26 The relative software is freely

available online and serves as a zero-cost, adequately accurate alternative of estimating

bacterial gene numbers present in a sample, to actually following the more accurate but

expensive shotgun sequencing pathway. In simple terms, PICRUSt works in two steps:26 On

the first step, it relies on a reference database to calculate gene numbers in bacteria with known,

sequenced genomes, and estimate gene numbers in bacteria without a known, sequenced

genome. This estimation is based on evolutionary models and phylogenetic principles, that

basically allow for prediction of gene content in a certain organism if the gene content of a

phylogenetically related organism is already known. On the second step, PICRUSt incorporates

the sample 16S rRNA data into the process (which bacteria are present in a sample and what

their abundance is), multiplies gene numbers from the first step by bacterial abundances from

the second step and estimates how many genes of each gene family exist in the sample. The

final output is a table of gene counts per sample per functional pathway, e.g. how many

bacterial genes related to methane metabolism are predicted to exist in sample A. This table

essentially offers a functional profile of the bacterial communities present in a researcher’s

samples. Applying multivariate analysis on such data thus allows for an extension of sample

comparison to functional (i.e. metabolic) dimensions, substantially improving research efforts

based on amplicon sequencing and allowing for a more spherical view of the bacterial

communities in question.

Ordination techniques

9
CVR-2019-0006-
R1

Similarities or differences between groups may already be apparent after appropriate

visualization of the data, usually through complex mathematical algorithms like non-metric

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


multidimensional scaling (MDS) and principal coordinates analysis (PCoA).27-29 These

ordination techniques are, in simple terms, tools that will take a spreadsheet of dozens of

samples and hundreds of bacteria and, through utilization of a measure of similarity and a series

of mathematical calculations, transform it into a scatterplot graph where samples that are more

similar to each other will be less distant to each other (Figure 2). Such a treatment of the data

enables the human eye to readily identify similarities and differences in the overall structure of

the bacterial communities of the analyzed samples.

Univariate analysis

A third route of analysis involves the one-by-one comparison of each microbial variable

between groups of subjects (e.g. the mean relative abundance of each bacterial genus), much

as these groups can be compared in terms of a mean body weight or the mean left ventricular

ejection fraction in common univariate analysis. As bacterial relative abundance data are

notoriously non-normally distributed, a non-parametric test must be used in order to achieve

meaningful results. Moreover, as this route will generate hundreds of individual comparisons

and, by chance alone, many false positive results, a correction for multiple comparisons must

also be made, usually by the method of false discovery rate adjustment developed by Benjamini

and Hochberg.30

5. The intestinal microbiota and cardiovascular disease

i. Gut microbiota and heart failure

Pathophysiological sequelae of heart failure, including a reduced cardiac output and peripheral

hypoxia, may alter gut function and compromise the intestinal epithelial barrier,31 allowing

10
CVR-2019-0006-
R1

increased leakage of microbial compounds such as lipopolysaccharide (LPS) into the

bloodstream and induction of low-grade systemic inflammation in the host (Figure 3, Table 2),

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


which has well established links with cardiac dysfunction and heart failure development (see

the section on atherosclerosis for a more detailed discussion of the association between the

microbiota and inflammation).32,33 However, whether gut microbial composition changes in

this context was until recently not known.

The link between heart failure and changes in the intestinal microbiota was already

hinted upon by Sandek et al., 34,35 who showed that heart failure patients had altered intestinal

function, bowel wall edema, and an overgrowth of microorganisms in examined biofilms. In

these studies, microorganisms were directly sampled from intestinal parts rather than only

collected from stool, which better reflects intestinal microbial communities. Interestingly, the

total amount of bacteria in the mucosal biofilms did not correlate with stool bacteria.35

The first study that showed changes in microbial community composition in the gut of

heart failure patients was published in 2015.36 Potentially pathogenic microbes, like species of

Shigella or Salmonella, were found in higher concentrations in stool samples of chronic heart

failure patients, as compared to healthy controls. Although this effort was flawed by neglecting

to correct for multiple comparisons and limited in its scope by being based on microbial

cultures instead of DNA analysis, it was the first step taken in the relative field.

Compositional changes were further examined by Luedde et al.,37 who were the first to

move beyond stool cultures and employ 16S rRNA amplicon sequencing. A general shift in

community composition and a depletion of several microbial genera was shown to characterize

heart failure, although alpha diversity was only nominally different between groups.

Subsequent studies have corroborated both the change in beta diversity and the fact that alpha

diversity is similar between heart failure patients and controls.38-40

11
CVR-2019-0006-
R1

A fundamental aspect of microbiota research is the examination of microbial metabolic

capacity. To this point, we have performed a predictive analysis of the metabolic gene content

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


of the microbiota using the PICRUSt algorithm, which unveiled a strong differentiation

between heart failure patients and controls, and numerous metabolic pathways, including

pathways in vitamin synthesis, depicting a change in the relative abundance of involved genes

between the groups.40 Although these data were suggestive only and did not constitute gene

expression results, metabolomics analyses have been performed and have established such a

differentiation.39

In fact, a specific bacterial metabolite, trimethylamine N-oxide (TMAO), which has

been implicated in atherosclerotic disease, has also been associated with heart failure.41 Stable

heart failure patients were shown to have higher fasting plasma levels of TMAO when

compared to healthy controls, and higher levels of TMAO within the heart failure group were

independently associated with a higher 5-year mortality risk.41 In this study, however, no

significant change of the intestinal microbiota was detected in the patients with a higher

mortality risk and no information was available on the use of antibiotics prior to sampling.

In summary, the composition of the intestinal microbiota in the context of human heart

failure has been mostly examined by a few small observational studies. An emerging pattern is

that alpha diversity is not significantly different in heart failure patients compared to controls,

meaning that the richness and distribution of the intestinal bacteria do not change in this context.

Although studies agree that the overall composition of the microbiota does change, the findings

on the changes in individual bacterial genera among the studies are not consistent. Some

evidence of functional alterations has also been provided,39,40 but this is only preliminary.

Contrary to evidence causally implicating gut bacteria with atherosclerotic disease, there is no

such evidence in heart failure.

12
CVR-2019-0006-
R1

ii. Gut microbiota and arterial hypertension

A link between the gut and hypertension has long been suggested, based on epidemiological

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


data associating salt and fiber intake with changes in blood pressure.42-48 Dietary sodium, a

potent factor in the pathogenesis of hypertension, makes first contact with the human organism

at the gut epithelium, where it is absorbed by enterocytes. The gut harbors its own renin-

angiotensin-aldosterone system, important for the uptake of sodium in the colon, which

interacts with gut bacteria and may be influenced by metabolites produced by the latter. Various

studies have shown that an increased sodium intake is associated with increases in blood

pressure.42-45 Moreover, meta-analyses have shown that an increased consumption of fiber,

which is primarily indigestible but can be fermented by gut bacteria as a source of short-chain

fatty acids, is associated with a lowering of both systolic and diastolic blood pressure in

hypertensive patients, as well as normotensives, to a lesser extent.47,48 Although the

mechanisms underlying this effect have not been solidly established, evidence implicating the

microbiota is accumulating (Figure 4). Unfortunately, research indicating a specific association

between compositional or functional alterations of the human intestinal microbiota and

hypertension has been limited, while some evidence has come from studies on rodents.49-51 A

mechanistic link between the intestinal microbiota and hypertension might involve quantitative

or qualitative changes in microbial metabolites, such as the short-chain fatty acid butyrate, that

could influence host inflammatory processes52 or sodium intake, and favor hypertension

development – the possible pathophysiological basis has been recently reviewed by Richards

et al.53 (Figure 4, Table 2, see also section 6 for sodium effects on the microbiota).

Yang et al.50 showed that hypertension in humans may be associated with changes in

the intestinal microbiota. Hypertensive patients reportedly had lower alpha diversity and a clear

visual separation of the hypertensive and control groups was shown in PCoA (based on the

weighted UniFrac distance metric). Although this study50 did not use healthy subjects as

13
CVR-2019-0006-
R1

controls, it set the foundation for further research in the field, which yielded similar findings.54

Interestingly, human hypertensive hosts might transfer a hypertensive profile to germ-free mice

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


receiving the hosts’ intestinal microbiota, pointing to as yet unexplained causative links

between the microbiota and hypertension (Figure 4).54 Yet another study comparing

hypertensive patients to healthy controls showed differentially enriched bacterial genera

between the groups and a visual separation in distance-based redundancy analysis, although

particulars about the statistical significance of multivariate analysis were not reported.55

In summary, the concept of microbial alterations in human hypertension has not yet

been fully addressed, as there are very few studies in the literature with small sample sizes.

Contrary to the heart failure field, evidence suggests that the alpha diversity of the intestinal

microbiota in hypertensive disease may be lower. Again, causative links between gut bacteria

and the development of hypertension have not been established yet.

iii. Gut microbiota and atherosclerosis

The association of intestinal bacteria with atherosclerosis has received considerable attention,

particularly since the reported link between bacterial metabolite TMAO and CVD. This

particular link (explained and referenced below) has drawn much scientific attention and stirred

cardiovascular research on gut microbiota. It should be mentioned that much evidence exists

linking the microbiota with obesity,56,57 an important risk factor for diabetes mellitus and

atherosclerosis.58,59 However, here we focus on studies directly addressing microbial changes

in human atherosclerotic subjects.

The pathophysiology underlying the links between the intestinal microbiota and

atherosclerosis has been recently reviewed by Jonsson and Bäckhed.60 The mechanisms may

include infection, inflammation, lipid metabolism, and, possibly, production of harmful

metabolites (Figure 5, Table 2).

14
CVR-2019-0006-
R1

Infection, regardless of site, leads to a pro-inflammatory profile that may affect plaque

stability.61 Microbial DNA has been recovered from atherosclerotic plaques, indicating a

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


potential link between the presence of symbiotic or pathogenic bacteria that may translocate

from the gut, the oral cavity, or other body locations to the plaque and the atherosclerotic

process.

Epidemiological evidence linking LPS with atherosclerosis exists for more than two

decades.62 Nonetheless, in trials where antibiotic use was tested, it did not show any

cardiovascular benefit.63 However, the interaction of the microbiota with the host immune

system is well established, and disruption of immune system homeostasis may influence CVD.

Human adaptive immunity has coevolved with gut bacteria, has matured in their presence and

has “learnt” to tolerate them.64 Maternal transfer of secretory IgA (sIgA) to the infant through

breast milk promotes regulatory immune networks favoring mutualism, since antigens bound

by sIgA are handled in a tolerogenic way.64 Gut associated lymphoid tissues (GALT) rely on

microbial signals to develop, as microbe-associated molecular patterns (MAMPs) are sensed

by pattern-recognition receptors, including Toll-like receptors (TLR), on epithelial and

dendritic cells, stimulating the maturation of cryptopatches.64 Under homeostatic conditions,

MAMPs lead to production of cytokines that promote tolerogenic macrophages and dendritic

cells. Innate immunity mechanisms, including secretion of antimicrobial peptides and tight

barrier function of the epithelium, mediate the confinement of bacteria in the lumen, but the

microbiota interacts with the host in regulating innate processes, as LPS and butyrate are

drivers of mucin production and maintenance of the mucus layer. Butyrate, in particular, has

strong anti-inflammatory properties through inhibition of nuclear factor kappa-light-chain-

enhancer of activated B cells (NF-κB),65 while another bacterial product, indole, increases

expression of genes that play a role in strengthening the intestinal barrier, as well as expression

of anti-inflammatory IL-10.66 Conceivably, a dysbiotic microbiota may interrupt the

15
CVR-2019-0006-
R1

aforementioned interactive pathways and initiate inflammatory responses that may affect

atherosclerosis and CVD risk. C-C chemokine receptor type 2 (CCR2) expressed in monocytes

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


is associated with arterial wall inflammation,67 and has been shown to both have a lower

expression after Roux-en-Y gastric bypass surgery in obese diabetic patients and negatively

correlate with an increase in Bifidobacterium.68 TLR4 signaling, induced by LPS, may mediate

myocardial inflammation and has been shown to be inhibited by adiponectin in rodents, 69 the

expression of which in Paneth cells is regulated by a strain of Lactobacillus in mice.70 Moreover,

advanced atherosclerotic plaques in apoE-/- mice have increased expressions of miR-10b, and

bacterial metabolite cyanidin-3-O-glucoside down-regulates miR-10b and leads to plaque

regression.71

Lastly, lipid metabolism influenced by the microbiota might lead to changes in blood

cholesterol levels, indirectly contributing to atherosclerosis, but a clear association between

changes in gut microbiota and shifts in LDL cholesterol levels has not yet been reported.72 Still,

certain microbial compounds, such as LPS, or metabolites, such as TMAO, may exert a

proatherogenic effect to the host (as explained below), while microbial substances are involved

in aspects of platelet activation, as depletion of gut flora resulted in reduction of platelet-

produced soluble P-selectin concentrations in a certain strain of mice.73 Such substances may

not include only LPS, but viral particles as well. Microbial compounds could activate platelets

by acting as ligands for TLR, as platelets, like macrophages or dendritic cells, express TLR on

their surface.

Koren et al.74 were the first to have examined the composition of the intestinal

microbiota in atherosclerotic patients. They assessed the microbiome recovered from both

atherosclerotic plaques and stool samples of patients with clinical atherosclerosis and found

that they were distinct from each other, i.e., plaque and intestinal bacterial communities are

different in composition. When they compared the gut microbiome of patients to non-

16
CVR-2019-0006-
R1

atherosclerotic controls they found no significant difference between them, although it is

unclear whether they performed a beta diversity analysis beyond the phylum level. In contrast,

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


other studies have found a significant difference in terms of beta diversity, or various individual

taxa (Table 1).75-78 More importantly, metabolic features of the microbiota differ between

atherosclerotic patients and controls.75,79 For example, the potential for butyrate synthesis by

the microbiota was shown to be lower in patients with atherosclerosis compared to healthy

subjects.79

TMAO and the TMAO paradox

Data on the bacterial metabolite TMAO and evidence that it may promote atherosclerosis were

first published in 2011 by Wang et al.80 The authors first isolated circulating plasma metabolites

that predict cardiovascular events from a cohort of human patients and controls and identified

TMAO and its precursor, choline, as linked to CVD risk. They next showed in mice that gut

flora presence is required for the formation of TMA from choline, before TMA is converted to

TMAO by enzymes in the liver. Choline itself is derived from dietary phosphatidylcholine

found in various foods, including red meat. The authors verified that TMAO levels predict

CVD risk in a large independent cohort of 1,876 subjects, even showing that TMAO levels are

proportionate to the number of stenotic coronary arteries. Finally, they proposed a mechanistic

link between TMAO and the atherosclerotic process through promotion of macrophage foam

cell formation, based on experiments on atherosclerotic prone mice fed with a choline enriched

diet.

Much work has followed on TMAO and its relation to CVD, with not always consistent

results. Tang et al.81 verified the predictive role of TMAO for CVD risk in a large cohort of

over 4,000 patients, on top of conventional risk factors. A recent meta-analysis supports these

results.82 In contrast, TMAO was comparatively decreased in the blood of over 300 Chinese

17
CVR-2019-0006-
R1

patients that had suffered an ischemic stroke or a transient ischemic attack, contrary to

expectations.78

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Beyond conflicting results, the TMAO paradox arises from the fact that the main dietary

source of free TMAO and the main food source capable of generating a high urinary excretion

of TMAO in humans is fish, which can yield significantly bigger amounts of absorbable TMAO

than choline contained in meat or eggs.83,84 Consumption of fish is widely accepted to be

beneficial for cardiovascular health.85-87 Not all fish have the same amounts of TMAO and

generalized epidemiological evidence might miss this distinction. However, this counter

argument should probably be taken into serious consideration. It has been suggested that

TMAO may simply be a confounding factor of a different causal relation, namely changes in

the composition of TMA-producing gut bacteria.88 TMA producers are generally of low

abundance and have been identified as Clostridium XIVa strains, Eubacterium species strain

AB3007, and Escherichia coli, although many more are expected to exist.89

To summarize, much evidence exists on the association of the intestinal microbiota with

atherosclerosis and ischemic heart disease. Studies in human subjects have yielded some

conflicting results, but a general pattern of differences in beta diversity between patients and

controls has nevertheless emerged. Importantly, evidence exists about specific microbial

metabolic products that are associated with increased CVD risk. However, data are again

conflicting and more research is therefore necessary.

6. Microbial associations with dietary sodium and carbohydrate intake, and probiotic use

in cardiovascular disease

Dietary habits play important roles in gut microbial composition, as has already been

mentioned. Two key dietary elements affecting the microbiota that merit further attention are

dietary sodium intake and carbohydrate consumption. A high salt intake has been shown to

18
CVR-2019-0006-
R1

negatively affect species of Lactobacillus in the human gut, and supplementation of

Lactobacillus murinus in mice (a species not found in humans) prevented the development of

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


hypertension by inhibiting salt-sensitive generation of TH17 cells.90 This finding is in line with

the small reduction of systolic blood pressure seen with use of Lactobacillus plantarum in

humans.91 Evidence also suggests that a high salt intake alters gut microbial composition in

mice, disrupts the epithelial barrier by affecting tight junction protein levels, and promotes

bacterial translocation from the gut to the kidney.92 Changes in microbial composition have

been linked to kidney injury, which may become the basis of a later rise in blood pressure.92 It

has been suggested that the mechanism underlying high salt intake-induced changes in

microbial composition is an increase in intestinal epithelial Na+/H+ exchange, since the latter

has been found to affect gut bacterial composition.93 Although these data are intriguing, they

are based on animal models and caution must be exercised when extrapolating to human

subjects.

Carbohydrate consumption is a major factor affecting the microbiota, as dietary fiber is

a key energy source for gut bacteria. In the context of CVD, evidence from mice harboring

synthetic human gut microbiota suggests that a diet low on fiber, which may be typical for

western populations, can result in gut mucus layer degradation and pathogen susceptibility, as

some of the microbiota turns to glycoproteins secreted by its host to use as energy source. 94

Conceivably, such a pro-inflammatory state can have consequences promoting the

atherosclerotic process. Human studies have also supported the role of a high fiber intake in

reducing cardiovascular risk,95 and it can be suggested that the underlying mechanism resides

partly in gut microbial metabolism and the production of anti-inflammatory short-chain fatty

acids. Conversely, a diet rich in refined carbohydrates, also typical in the western world, has

long been suggested to increase the risk of CVD.96

19
CVR-2019-0006-
R1

The use of probiotics has been explored as a method to modulate intestinal microbiota

properties in the context of CVD and other diseases. Probiotics are ingestible microorganisms

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


that reach the intestinal lumen, where they exert mainly functional effects – i.e., they generally

do not affect the composition and diversity of the resident microbiota, especially in healthy

subjects,97,98 although some evidence suggests that gut colonization by probiotics is possible in

a person- and strain-specific manner,99 and that a probiotic effect on the host gut microbiome

may be seen in susceptible individuals.99 To date, beneficial effects have been acknowledged

for some bacterial species, when these are ingested in adequate amounts.100 There is also

evidence of strain-specific benefits for certain disorders, mostly of the gastrointestinal

tract.97,100 However, most health claims for probiotics rely on poor evidence and the notion that

probiotic use can correct dysbiosis is not well supported for the majority of the examined

products.101 There is some evidence that probiotic use can confer short-lived benefits in healthy

humans, but certainly more research is required to explore any long-term effects.102 Such

research is made imperative given evidence that probiotics may adversely affect gut microbiota

recovery after experimental antibiotic use in healthy humans.103 Some probiotic strains may

take advantage of gut microbial depletion following broad spectrum antibiotic use, to

effectively colonize the intestinal mucosa. Once such a colonization takes place and while

probiotic use is continued, the new resident bacteria may antagonize and resist a

reestablishment of the homeostatic bacterial composition and diversity in multiple ways and

with conceivable negative implications for the host, in the same way that some of them are

expected to antagonize pathogen colonization in the context of preventing antibiotic-associated

diarrhea.

More specifically, there is limited data available for the use of probiotics in CVD. One

study reported that consumption of preparations containing Saccharomyces boulardii may

increase the left ventricular ejection fraction in heart failure patients.104 A larger trial is

20
CVR-2019-0006-
R1

underway to verify this result.105 Further, a recent meta-analysis of 9 randomized, placebo-

controlled human trials showed that probiotic consumption has a weak lowering effect on both

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


systolic and diastolic blood pressure, reducing systolic pressure by a mean 3.56mmHg and

diastolic pressure by a mean 2.38mmHg.91 However, the included studies and the effect on

blood pressure were relatively small, and more evidence is probably required.

Lastly, probiotic administration has also been utilized to counter the possible

proatherogenic effects of the intestinal microbiota, with some promising results.106 Potential

mechanisms include the induction of bile acid deconjugation, which will increase their

excretion and force the host to use up more cholesterol to balance the effect.106 While additional

evidence is required, consumption of Lactobacillus acidophilus may have a greater effect in

lowering cholesterol than other probiotic preparations.107

Although the research mentioned above points to potentially beneficial effects of

probiotics in CVD, caution must be exercised when contemplating probiotic use in critically

ill hospitalized patients. In particular, patients with a compromised gut epithelial barrier may

theoretically suffer a translocation of ingestible microbes from the gut to other body areas,

whereas patients with central venous catheters might infrequently suffer fungemia from

Saccharomyces boulardii.108

7. Remaining challenges in microbial research

Despite some promising evidence on the role of intestinal microbiota in cardiovascular

diagnostics and therapeutics, major challenges in the field must be overcome, before an impact

at the clinical level is felt.

Such challenges may relate to technical issues that hinder the generalizability of study

results. Although a common set of methodological tools is broadly used, a consensus on how

to conduct a microbiome study, from study design and sample collection to data analysis and

21
CVR-2019-0006-
R1

interpretation, is still missing. As a result, differences in methodology may render different

studies incomparable. For example, some studies differ from others on which hypervariable

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


regions of the 16S rRNA gene they have targeted for sequencing. Such a difference will

generate variation in the identification and proportional calculation of the assessed

microorganisms because of technical issues alone.

Moreover, studies involving heterogeneous patient populations may be biased by

confounders such as the microbial variation in the context of different disease states (e.g.

stable/decompensated heart failure), disease aetiology (e.g. ischemic vs. non-ischemic heart

failure), as well as differences due to the geographic origin of subjects, rendering comparisons

difficult at best. Confounding factors such as diet and antibiotic use must also be thoroughly

addressed before including subjects in a study. Often, variation in medication, age, and dietary

habits will affect results and complicate a conclusion. Especially regarding medication,

research suggests that widely prescribed drugs other than antibiotics, such as metformin and

proton pump inhibitors,109,110 are associated with alterations in gut microbial composition.

However, research regarding the direct impact of medication to gut microbial communities is

generally limited and faces various experimental challenges.111 There are virtually no studies

indexed in PubMed showing an association, causal or otherwise, of cardiovascular medication,

such as β-blockers, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers,

diuretics, and mineralocorticoid receptor antagonists, with changes in gut microbial

composition in humans.

Further, findings on the relative abundance of specific microorganisms are usually

inconsistent between studies, as can be seen in Table 1. This could be attributed either to the

study design and sample size, or to background characteristics of subjects. Alarmingly, it could

also be attributed to chance. However, this fact may nevertheless support the notion that the

real finding of major importance is the overall shift in bacterial community composition seen

22
CVR-2019-0006-
R1

in the context of CVD and not the changes in individual genera or species per se. Importantly,

changes in the relative abundance do not necessarily suggest a change in the actual abundance

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


of a given microorganism, since the rise in the relative abundance of a certain bacterium may

be the result of the actual decrease of another. This factor complicates both the outcome and

interpretation of study findings, as well as the comparison between studies. Employment of

direct quantitative PCR measurement of the bacterial genera of interest could provide more

reliable data on this issue.

Another important point relates to the statistical analysis of the data. The nature of

microbial data requires special attention and rigorous statistical methodology, which is

generally different from the common analytical tools used in the bulk of clinical cardiovascular

research.

Furthermore, prospective studies in human patients with serial sampling and reports on

the change of microbial communities as a function of time (e.g. before and after an

intervention) are absent from the relative literature. Such studies are urgently needed, to

elucidate critical questions on cause and causation.

Finally, it should also be stressed that the compositional differences of gut microbial

communities between humans and rodents make animal study findings not directly translatable

to humans.112

8. Conclusions

Ample evidence has highlighted the important functions of the gut microbiota that benefit

human hosts, as well as its compositional and functional alterations associated with heart

failure, atherosclerotic disease, and hypertension. Although associations between gut

microbiota and CVD have been reported, the existing evidence includes mostly cross-sectional

studies, with small sample sizes and inconsistent results. The overarching pathophysiological

23
CVR-2019-0006-
R1

link between gut bacteria and CVD seems to be the induction of inflammation in the host via

structural or functional alterations of the microbiota. However, whether such changes in the

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


gut microbiota are causally associated with CVD, are influenced by CVD, or are simple

bystanders, remains to be proved. Future research is therefore warranted to incorporate the

wealth of information on gut microbiota into clinical decision pathways and personalized

medicine.

24
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019

25
Conflict of interest: None declared.
CVR-2019-0006-
R1
CVR-2019-0006-
R1

References
1. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on
human health: An integrative view. Cell 2012;148:1258–1270.
2. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


gut microbiota metabolic interactions. Science 2012;336:1262–1267.
3. Sears CL. A dynamic partnership: Celebrating our gut flora. Anaerobe 2005;11:247–
251.
4. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons
N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang
H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier
D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu
H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H, Wang J, Brunak
S, Doré J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J; MetaHIT
Consortium, Bork P, Ehrlich SD, Wang J. A human gut microbial gene catalogue
established by metagenomic sequencing. Nature 2010;464:59–65.
5. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of
bacterial to host cells in humans. Cell 2016;164:337–340.
6. Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl
J Med 2016;375:2369–2379.
7. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003;361:512–519.
8. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet
on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice.
Sci Transl Med 2009;1:6ra14.
9. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et
al. Human gut microbiome viewed across age and geopraphy. Nature 2012;486:222–
227.
10. Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, et al.
The long-term stability of the human gut microbiota. Science 2013;341:1237439.
11. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent
LT, Ley RE. Succession of microbial consortia in
the developing infant gut microbiome. Proc Natl Acad Sci USA 2011;108 Suppl
1:4578–4585.
12. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill
SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora.
Science 2005;308:1635–1638.
13. Human Microbiome Project Consortium. Structure, function and diversity of
the healthy human microbiome. Nature 2012;486:207–214.
14. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M,
Knights D, Walters WA, Knight R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel
L, Li H, Bushman FD, Lewis JD. Linking long-term dietary patterns with gut microbial
enterotypes. Science 2011;334:105–8.
15. Goodrich JK, Di Rienzi SC, Poole AC, Koren O, Walters WA, Caporaso JG, Knight R,
Ley RE. Conducting a microbiome study. Cell 2014;158:250–262.
16. Kurland CG. Structure and function of the bacterial ribosome. Annu Rev Biochem
1977;46:173–200.
17. Clarridge JE 3rd. Impact of 16S rRNA gene sequence analysis for identification of
bacteria on clinical microbiology and infectious diseases. Clin Microbiol Rev
2004;17:840–862.

26
CVR-2019-0006-
R1

18. Tyler AD, Smith MI, Silverberg MS. Analyzing the human microbiome: a "how to"
guide for physicians. Am J Gastroenterol 2014;109:983–993.
19. Thomas T, Gilbert J, Meyer F. Metagenomics - a guide from sampling to data analysis.
Microb Inform Exp 2012;2:3.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


20. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello
EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, Huttley GA, Kelley ST, Knights
D, Koenig JE, Ley RE, Lozupone CA, McDonald D, Muegge BD, Pirrung M, Reeder
J, Sevinsky JR, Turnbaugh PJ, Walters WA, Widmann J, Yatsunenko T, Zaneveld
J, Knight R. QIIME allows analysis of high-throughput community sequencing data.
Nature Methods 2010;7:335–336.
21. Shannon CE. A mathematical theory of communication. Bell Syst Tech J 1948;27:379–
423.
22. Bray JR, Curtis JT. An ordination of upland forest communities of southern Wisconsin.
Ecol Monogr 1957;27:325–349.
23. Lozupone C, Knight R. UniFrac: A new phylogenetic method for comparing microbial
communities. Appl Environ Microbiol 2005;71:8228–8235.
24. Clarke KR. Non-parametric multivariate analyses of changes in community structure.
Austral Ecol 1993;18:117–143.
25. Anderson MJ. A new method for non-parametric multivariate analysis of variance.
Austral Ecol 2001;26:32–46.
26. Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente
JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C. Predictive
functional profiling of microbial communities using 16S rRNA marker gene sequences.
Nat Biotechnol 2013;31:814–821.
27. Shepard, R.N. The analysis of proximities: multidimensional scaling with an unknown
distance function. Psychometrika 1962;27:125–140.
28. Kruskal, J.B. Multidimensional scaling by optimizing goodness of fit to a nonmetric
hypothesis. Psychometrika 1964;29:1–27.
29. Gower, J.C. Some distance properties of latent root and vector methods used in
multivariate analysis. Biometrika 1966;53:325–328.
30. Benjamini Y, Hochberg Y. Controlling the false discovery rate: A practical and
powerful approach to multiple testing. J R Statist Soc Ser B 57 1995;1:289–300.
31. Rogler G, Rosano G. The heart and the gut. Eur Heart J 2014;35:426–430.
32. Sun XQ, Abbate A, Bogaard HJ. Role of cardiac inflammation in right ventricular
failure. Cardiovasc Res 2017;113:1441–1452.
33. Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac
remodelling. Cardiovasc Res 2017;113:1538–1550.
34. Sandek A, Bauditz J, Swidsinski A, Buhner S, Weber-Eibel J, von Haehling
S, Schroedl W, Karhausen T, Doehner W, Rauchhaus M, Poole-Wilson P, Volk
HD, Lochs H, Anker SD. Altered intestinal function in patients
with chronic heart failure. J Am Coll Cardiol 2007;50:1561–1569.
35. Sandek A, Swidsinski A, Schroedl W, Watson A, Valentova M, Herrmann
R, Scherbakov N, Cramer L, Rauchhaus M, Grosse-Herrenthey A, Krueger M, von
Haehling S, Doehner W, Anker SD, Bauditz J.
Intestinal blood flow in patients with chronic heart failure:
a link with bacterial growth, gastrointestinal symptoms, and cachexia. J Am Coll
Cardiol 2014;64:1092–1102.
36. Pasini E, Aquilani R, Testa C, Baiardi P, Angioletti S, Boschi F, Verri M, Dioguardi F.
Pathogenic gut flora in patients with heart failure. JACC Heart Fail 2016;4:220–227.

27
CVR-2019-0006-
R1

37. Luedde M, Winkler T, Heinsen FA, Rühlemann MC, Spehlmann ME, Bajrovic A, Lieb
W, Franke A, Ott SJ, Frey N. Heart
failure is associated with depletion of core intestinal microbiota.
ESC Heart Fail 2017;4:282–290.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


38. Kamo T, Akazawa H, Suda W, Saga-Kamo A, Shimizu Y, Yagi H, Liu Q, Nomura
S, Naito AT, Takeda N, Harada M, Toko H, Kumagai H, Ikeda Y, Takimoto E, Suzuki
JI, Honda K, Morita H, Hattori M, Komuro I.
Dysbiosis and compositional alterations with aging in
the gut microbiota of patients with heart failure. PLoS One 2017;12:e0174099.
39. Cui X, Ye L, Li J, Jin L, Wang W, Li S, Bao M, Wu S, Li L, Geng B, Zhou X, Zhang
J, Cai J.
Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic
heart failure patients. Sci Rep 2018;8:635.
40. Katsimichas T, Ohtani T, Motooka D, Tsukamoto Y, Kioka H, Nakamoto K, Konishi
S, Chimura M, Sengoku K, Miyawaki H, Sakaguchi T, Okumura R, Theofilis K, Iida
T, Takeda K, Nakamura S, Sakata Y. Non-ischemic heart failure with reduced ejection
fraction is associated with altered intestinal microbiota. Circ J 2018;82:1640–1650.
41. Tang WH, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, Wu Y, Hazen SL.
Prognostic value of elevated levels of intestinal microbe-
generated metabolite trimethylamine-N-oxide in patients with heart failure: refining
the gut hypothesis. J Am Coll Cardiol 2014;64:1908–1914.
42. Meneely GR, Dahl LK. Electrolytes in hypertension: the effects of sodium chloride.
The evidence from animal and human studies. Med Clin North Am 1961;45:271–283.
43. Intersalt Cooperative Research Group. Intersalt: an international study of electrolyte
excretion and blood pressure. Results for 24 hour urinary sodium and potassium
excretion. BMJ 1988;297:319–328.
44. Mente A, O’Donnell MJ, Rangarajan S, McQueen MJ, Poirier P, Wielgosz A, Morrison
H, Li W, Wang X, Di C, Mony P, Devanath A, Rosengren A, Oguz A, Zatonska K,
Yusufali AH, Lopez-Jaramillo P, Avezum A, Ismail N, Lanas F, Puoane T, Diaz R,
Kelishadi R, Iqbal R, Yusuf R, Chifamba J, Khatib R, Teo K, Yusuf S PURE
Investigators. Association of urinary sodium and potassium excretion with blood
pressure. N Engl J Med 2014;371:601–611.
45. Takase H, Sugiura T, Kimura G, Ohte N, Dohi Y. Dietary sodium consumption
predicts future blood pressure and incident hypertension in the Japanese normotensive
general population. J Am Heart Assoc 2015;4:e001959.
46. Angela Wright, P G Burstyn, M J Gibney. Dietary fiber and blood pressure. Br Med
J 1979;2:1541–1543.
47. Streppel MT, Arends LR, van 't Veer P, Grobbee DE, Geleijnse JM. Dietary fiber and
blood pressure: a meta-analysis of randomized placebo-controlled trials. Arch Intern
Med 2005;165:150–156.
48. Whelton SP, Hyre AD, Pedersen B, Yi Y, Whelton PK, He J. Effect of dietary fiber
intake on blood pressure: a meta-analysis of randomized, controlled clinical trials. J
Hypertens 2005;23:475–481.
49. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, Vijay-
Kumar M, Pennathur S, Joe B. Evidence for a link between
gut microbiota and hypertension in the Dahl rat. Physiol Genomics 2015;47:187–197.
50. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong
M, Qi Y, Zubcevic J, Sahay B, Pepine CJ, Raizada MK, Mohamadzadeh M. Gut
dysbiosis is linked to hypertension. Hypertension 2015;65:1331–1340.

28
CVR-2019-0006-
R1

51. Adnan S, Nelson JW, Ajami NJ, Venna VR, Petrosino JF, Bryan RM Jr, Durgan DJ.
Alterations in the gut microbiota can elicit hypertension in rats. Physiol
Genomics 2017;49:96–104.
52. Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


dysfunctional adipose tissue. Cardiovasc Res 2017;113:1009–1023.
53. Richards EM, Pepine CJ, Raizada MK, Kim S. The gut, its microbiome,
and hypertension. Curr Hypertens Rep 2017;19:36.
54. Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, Geng B, Zhang
W, Weldon R, Auguste K, Yang L, Liu X, Chen L, Yang X, Zhu B, Cai J. Gut
microbiota dysbiosis contributes to the development of hypertension.
Microbiome 2017;5:14.
55. Yan Q, Gu Y, Li X, Yang W, Jia L, Chen C, Han X, Huang Y, Zhao L, Li P, Fang Z,
Zhou J, Guan X, Ding Y, Wang S, Khan M, Xin Y, Li S, Ma Y. Alterations of the gut
microbiome in hypertension. Front Cell Infect Microbiol 2017;7:381.
56. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-
associated gut microbiome with increased capacity for energy harvest.
Nature 2006;444:1027–1031.
57. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML,
Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon
JI. A core gut microbiome in obese and lean twins. Nature 2009;457:480–484.
58. Antonopoulos AS, Margaritis M, Coutinho P, Shirodaria C, Psarros C, Herdman L,
Sanna F, De Silva R, Petrou M, Sayeed R, Krasopoulos G, Lee R, Digby J, Reilly S,
Bakogiannis C, Tousoulis D, Kessler B, Casadei B, Channon KM, Antoniades C.
Adiponectin as a link between type 2 diabetes and vascular NADPH oxidase activity in
the human arterial wall: the regulatory role of perivascular adipose tissue. Diabetes
2015;64:2207–2219.
59. Antonopoulos AS, Tousoulis D. The molecular mechanisms of obesity paradox.
Cardiovasc Res 2017;113:1074–1086.
60. Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev
Cardiol 2017;14:79–87.
61. Antoniades C, Cunnington C, Antonopoulos A, Neville M, Margaritis M,
Demosthenous M, Bendall J, Hale A, Cerrato R, Tousoulis D, Bakogiannis C, Marinou
K, Toutouza M, Vlachopoulos C, Leeson P, Stefanadis C, Karpe F, Channon KM.
Induction of vascular GTP-cyclohydrolase I and endogenous tetrahydrobiopterin
synthesis protect against inflammation-induced endothelial dysfunction in human
atherosclerosis. Circulation 2011;124:1860–1870.
62. Wiedermann CJ, Kiechl S, Dunzendorfer S, Schratzberger P, Egger G, Oberhollenzer
F, Willeit J. Association of endotoxemia with carotid atherosclerosis and
cardiovascular disease: prospective results from the Bruneck Study. J Am Coll
Cardiol 1999;34:1975–1981.
63. Taylor-Robinson D, Boman J. The failure of antibiotics to prevent heart attacks. BMJ
2005;331:361–2.
64. Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the
intestinal microbiota and immune system. Nature 2012;489:231–41.
65. Segain JP, Raingeard de la Blétière D, Bourreille A, Leray V, Gervois N, Rosales C,
Ferrier L, Bonnet C, Blottière HM, Galmiche JP. Butyrate inhibits inflammatory
responses through NFkappaB inhibition: implications for Crohn's disease. Gut
2000;47:397–403.

29
CVR-2019-0006-
R1

66. Bansal T, Alaniz RC, Wood TK, Jayaraman A. The bacterial signal indole increases
epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc
Natl Acad Sci U S A 2010;107:228–33.
67. Verweij SL, Duivenvoorden R, Stiekema LCA, Nurmohamed NS, van der Valk FM,

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Versloot M, Verberne HJ, Stroes ESG, Nahrendorf M, Bekkering S, Bernelot Moens
SJ. CCR2 expression on circulating monocytes is associated with arterial wall
inflammation assessed by 18F-FDG PET/CT in patients at risk for cardiovascular
disease. Cardiovasc Res 2018;114:468–475.
68. Chen H, Qian L, Lv Q, Yu J, Wu W, Qian H. Change in gut microbiota is correlated
with alterations in the surface molecule expression of monocytes after Roux-en-Y
gastric bypass surgery in obese type 2 diabetic patients. Am J Transl Res 2017;9:1243–
1254.
69. Jenke A, Wilk S, Poller W, Eriksson U, Valaperti A, Rauch BH, Stroux A, Liu P,
Schultheiss HP, Scheibenbogen C, Skurk C. Adiponectin protects against Toll-like
receptor 4-mediated cardiac inflammation and injury. Cardiovasc Res 2013;99:422–31.
70. Su X, Yan H, Huang Y, Yun H, Zeng B, Wang E, Liu Y, Zhang Y, Liu F, Che Y, Zhang
Z, Yang R.Expression of FABP, adipsin and adiponectin in Paneth cells is modulated
by gut Lactobacillus. Sci Rep 2015;5:18588.
71. Wang D, Wang W, Lin W, Yang W, Zhang P, Chen M, Ding D, Liu C, Zheng J, Ling
W. Apoptotic cell induction of miR-10b in macrophages contributes to advanced
atherosclerosis progression in ApoE-/- mice. Cardiovasc Res 2018;114:1794–1805.
72. Herbert KE, Erridge C. Regulation of low-density lipoprotein cholesterol by intestinal
inflammation and the acute phase response. Cardiovasc Res 2018;114:226–232.
73. Anselmo A, Riva F, Gentile S, Soldani C, Barbagallo M, Mazzon C, Feruglio F,
Polentarutti N, Somma P, Carullo P, Angelini C, Bacci M, Mendolicchio GL, Voza A,
Nebuloni M, Mantovani A, Garlanda C. Expression and function of IL-1R8
(TIR8/SIGIRR): a regulatory member of the IL-1 receptor family in platelets.
Cardiovasc Res 2016;111:373–384.
74. Koren O, Spor A, Felin J, Fåk F, Stombaugh J, Tremaroli V, Behre CJ, Knight
R, Fagerberg B, Ley RE, Bäckhed F. Human oral, gut, and plaque microbiota in
patients with atherosclerosis. Proc Natl Acad Sci USA 2011;108 Suppl 1:4592–4598.
75. Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, Bäckhed
F, Nielsen J. Symptomatic atherosclerosis is associated with an altered gut metagenome.
Nat Commun 2012;3:1245.
76. Emoto T, Yamashita T, Sasaki N, Hirota Y, Hayashi T, So A, Kasahara K, Yodoi
K, Matsumoto T, Mizoguchi T, Ogawa W, Hirata K. Analysis of gut
microbiota in coronary artery disease patients: a possible link between gut
microbiota and coronary artery disease. J AtherosclerThromb 2016;23:908–921.
77. Emoto T, Yamashita T, Kobayashi T, Sasaki N, Hirota Y, Hayashi T, So A, Kasahara
K, Yodoi K, Matsumoto T, Mizoguchi T, Ogawa W, Hirata KI.
Characterization of gut microbiota profiles in coronary artery disease patients using d
ata mininganalysis of terminal restriction fragment length polymorphism: gut microbi
ota could be a diagnostic marker of coronary artery disease. Heart
Vessels 2017;32:39–46.
78. Yin J, Liao SX, He Y, Wang S, Xia GH, Liu FT, Zhu JJ, You C, Chen Q, Zhou L, Pan
SY, Zhou HW. Dysbiosis of gut microbiota with reduced trimethylamine-N-Oxide
Level in patients with large-artery atherosclerotic stroke or transient ischemic attack. J
Am Heart Assoc 2015;4.pii:e002699.

30
CVR-2019-0006-
R1

79. Jie Z, Xia H, Zhong SL, Feng Q, Li S, Liang S, Zhong H, Liu Z, Gao Y, Zhao
H, Zhang D, Su Z, Fang Z, Lan Z, Li J, Xiao L, Li J, Li R, Li X, Li F, Ren H, Huang
Y, Peng Y, Li G, Wen B, Dong B, Chen JY, Geng QS, Zhang ZW, Yang H, Wang
J, Wang J, Zhang X, Madsen L, Brix S, Ning G, Xu X, Liu X, Hou Y, Jia H, He

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


K, Kristiansen K. The gut microbiome in atherosclerotic cardiovascular disease. Nat
Commun 2017;8:845.
80. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt
EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato
JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes
cardiovascular disease. Nature 2011;472:57–63.
81. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL.
Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl
J Med 2013;368:1575–1584.
82. Schiattarella GG, Sannino A, Toscano E, Giugliano G, Gargiulo G, Franzone
A, Trimarco B, Esposito G, Perrino C. Gut microbe-generated metabolite
trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and
dose-response meta-analysis. Eur Heart J 2017;38:2948–2956.
83. Zhang AQ, Mitchell SC, Smith RL. Dietary precursors of trimethylamine in man: a
pilot study. Food Chem Toxicol 1999;37:515–520.
84. Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, Sutter
JL, Caudill MA. Trimethylamine-N-oxide (TMAO) response to animal source foods
varies among healthy young men and is influenced by their gut microbiota composition:
A randomized controlled trial. Mol Nutr Food Res 2017;61.
85. He K, Song Y, Daviglus ML, Liu K, Van Horn L, Dyer AR, Greenland P. Accumulated
evidence on fish consumption and coronary heart disease mortality: a meta-analysis of
cohort studies. Circulation 2004;109:2705–2711.
86. Mozaffarian D, Appel LJ, Van Horn L. Components of a cardioprotective diet: new
insights. Circulation 2011;123:2870–2891.
87. Zheng J, Huang T, Yu Y, Hu X, Yang B, Li D. Fish consumption and CHD mortality:
an updated meta-analysis of seventeen cohort studies. Public Health
Nutr 2012;15:725–737.
88. Landfald B, Valeur J, Berstad A, Raa J. Microbial trimethylamine-N-oxide as a disease
marker: something fishy? Microb Ecol Health Dis 2017;28:1327309.
89. Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing
bacteria of the human gut microbiota. Microbiome 2017;5:54
90. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, Haase S,
Mähler A, Balogh A, Markó L, Vvedenskaya O, Kleiner FH, Tsvetkov D, Klug L,
Costea PI, Sunagawa S, Maier L, Rakova N, Schatz V, Neubert P, Frätzer C, Krannich
A, Gollasch M, Grohme DA, Côrte-Real BF, Gerlach RG, Basic M, Typas A, Wu C,
Titze JM, Jantsch J, Boschmann M Dechend R, Kleinewietfeld M1, Kempa S3, Bork
P, Linker RA, Alm EJ, Müller DN1. Salt-responsive gut commensal modulates TH17
axis and disease. Nature 2017;551:585–589.
91. Khalesi S, Sun J, Buys N, Jayasinghe R. Effect of probiotics on blood pressure: a
systematic review and meta-analysis of randomized, controlled trials.
Hypertension 2014;64:897–903.
92. Hu J, Luo H, Wang J, Tang W, Lu J, Wu S, Xiong Z, Yang G, Chen Z, Lan T, Zhou H,
Nie J, Jiang Y, Chen P. Enteric dysbiosis-linked gut barrier disruption triggers early
renal injury induced by chronic high salt feeding in mice. Exp Mol Med 2017;49:e370.

31
CVR-2019-0006-
R1

93. Wang C, Huang Z, Yu K, Ding R, Ye K, Dai C1, Xu X1, Zhou G, Li C. High-salt diet
has a certain impact on protein digestion and gut microbiota: a sequencing and
proteome combined study. Front Microbiol 2017;8:1838.
94. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA,

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck
TS, Núñez G, Martens EC. A dietary fiber-deprived gut microbiota degrades the colonic
mucus barrier and enhances pathogen susceptibility. Cell 2016;167:1339–1353.
95. Threapleton DE, Greenwood DC, Evans CE, Cleghorn CL, Nykjaer C, Woodhead C,
Cade JE, Gale CP, Burley VJ. Dietary fibre intake and risk of cardiovascular disease:
systematic review and meta-analysis. BMJ 2013;347:f6879.
96. Liu S, Willett WC, Stampfer MJ, Hu FB, Franz M, Sampson L, Hennekens CH, Manson
JE. A prospective study of dietary glycemic load, carbohydrate intake, and risk of
coronary heart disease in US women. Am J Clin Nutr 2000;71:1455–61.
97. Sánchez B, Delgado S, Blanco-Míguez A, Lourenço A, Gueimonde M, Margolles A.
Probiotics, gut microbiota, and their influence on host health and disease. Mol Nutr
Food Res 2017;61.
98. Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH, Pedersen O. Alterations in
fecal microbiota composition by probiotic supplementation in healthy adults: a
systematic review of randomized controlled trials. Genome Med 2016;8:52.
99. Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, Kotler
E, Zur M, Regev-Lehavi D, Brik RB, Federici S, Cohen Y, Linevsky R, Rothschild D,
Moor AE, Ben-Moshe S, Harmelin A, Itzkovitz S, Maharshak N, Shibolet O, Shapiro
H, Pevsner-Fischer M, Sharon I, Halpern Z, Segal E, Elinav E. Personalized gut
mucosal colonization resistance to empiric probiotics is associated with unique host and
microbiome features. Cell 2018;174:1388–1405.
100. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani
RB, Flint HJ, Salminen S, Calder PC, Sanders ME. Expert consensus document. The
International Scientific Association for Probiotics and Prebiotics consensus statement
on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol
2014;11:506–514.
101. McFarland LV. Use of probiotics to correct dysbiosis of normal microbiota
following disease or disruptive events: a systematic review. BMJ Open 2014;4:e005047.
102. Khalesi S, Bellissimo N, Vandelanotte C, Williams S, Stanley D, Irwin C. A
review of probiotic supplementation in healthy adults: helpful or hype? Eur J Clin Nutr
2018;doi: 10.1038/s41430-018-0135-9.
103. Suez J, Zmora N, Zilberman-Schapira G, Mor U, Dori-Bachash M, Bashiardes
S, Zur M, Regev-Lehavi D, Ben-Zeev Brik R, Federici S, Horn M, Cohen Y, Moor AE,
Zeevi D, Korem T, Kotler E, Harmelin A, Itzkovitz S, Maharshak N, Shibolet O,
Pevsner-Fischer M, Shapiro H, Sharon I, Halpern Z, Segal E, Elinav E. Post-antibiotic
gut mucosal microbiome reconstitution is impaired by probiotics and improved by
autologous FMT. Cell 2018;174:1406–1423.
104. Costanza AC, Moscavitch SD, Faria Neto HC, Mesquita ET. Probiotic therapy
with Saccharomyces boulardii for heart failure patients: A randomized, double-blind,
placebo-controlled pilot trial. Int J Cardiol 2015;179:348–350.
105. Mayerhofer CCK1, Awoyemi AO, Moscavitch SD, Lappegård KT, Hov JR,
Aukrust P, Hovland A, Lorenzo A, Halvorsen S, Seljeflot I, Gullestad L, Trøseid M,
Broch K. Design of the GutHeart-targeting gut microbiota to treat heart failure-trial: a
Phase II, randomized clinical trial. ESC Heart Fail 2018;5:977–984.

32
CVR-2019-0006-
R1

106. Thushara RM, Gangadaran S, Solati Z, Moghadasian MH. Cardiovascular


benefits of probiotics: a review of experimental and clinical studies. Food Funct
2016;7:632–642.
107. Rondanelli M, Faliva MA, Perna S, Giacosa A, Peroni G, Castellazzi AM.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Using probiotics in clinical practice: Where are we now? A review of existing meta-
analyses. Gut Microbes 2017;8:521–543.
108. McFarland LV. Systematic review and meta-analysis of Saccharomyces
boulardii in adult patients. World J Gastroenterol 2010;16:2202–2222.
109. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S,
Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, Arumugam M, Kristiansen
K, Voigt AY, Vestergaard H, Hercog R, Costea PI, Kultima JR, Li J, Jørgensen T,
Levenez F, Dore J6; MetaHIT consortium, Nielsen HB, Brunak S, Raes J, Hansen T,
Wang J, Ehrlich SD, Bork P, Pedersen O. Disentangling type 2 diabetes and metformin
treatment signatures in the human gut microbiota. Nature 2015;528:262–266.
110. Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, Tigchelaar EF,
Jankipersadsing SA, Cenit MC, Harmsen HJ, Dijkstra G, Franke L, Xavier RJ, Jonkers
D, Wijmenga C, Weersma RK, Zhernakova A. Proton pump inhibitors affect the gut
microbiome. Gut 2016;65:740–8.
111. Maier L, Typas A. Systematically investigating the impact of medication on the
gut microbiome. Curr Opin Microbiol 2017;39:128–135.
112. Nguyen TL, Vieira-Silva S, Liston A, Raes J. How informative is the mouse for
human gut microbiota research? Dis Model Mech 2015;8:1–16.

33
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
CVR-2019-0006-R1

Table 1. Summary of studies on the association of the intestinal microbiota with cardiovascular disease.

Authors Subjects Study design Analysis Main findings Suggested mechanisms


Heart failure
Sandek et Caucasians; Germans by Cross- FISH of biopsy Higher mean density in An abnormal mucus barrier allows
al.34 inference. 22 patients, 22 sectional films mucus and higher adherence bacteria to penetrate the mucus and reach
controls to the mucosa for bacteria in the mucosa in patients
patient samples
Sandek et Race and ethnicity not Cross- FISH of biopsy Concentrations and Alterations of the local mucosal
al.35 stated; Germans by sectional films; FISH and proportions of bacteria similar environment caused by reduced
inference. 65 patients, 25 epifluorescence in stool samples of patients mesenteric blood flow and endothelial
controls microscopy of and controls. Higher dysfunction favor the growth of bacteria
stool samples concentration of mostly
anaerobic bacteria in biofilms
of patients
Pasini et Race and ethnicity not Cross- Stool cultures Higher concentrations of 4 Changes in fluid balance affecting
al.36 stated; Italians by sectional genera of pathogenic bacteria gastrointestinal motility and transit time
inference. 60 patients, 20 (Shigella, Salmonella, favor stasis and bacterial overgrowth in
healthy controls Campylobacter, Yersinia) and patients. Acid-suppressing therapy,
1 genus of fungi (Candida) in nutrient deprivation, and hypoxemia also
patient samples suggested as factors
Luedde et Race and ethnicity not Cross- Amplicon Significant difference in beta Diet, fluid imbalance, bowel dysmotility,
al.37 stated; Germans by sectional sequencing (16S diversity between patients and and hypoxia may favor changes of
inference. 20 patients, 20 rRNA analysis) controls. A few bacterial bacterial composition in patients
controls genera significantly decreased
in patients (Blautia,
Collinsella, unclassified
Erysipelotrichaceae,
unclassified
Ruminococcaceae)
Kamo et Race and ethnicity not Cross- Amplicon Alpha diversity similar in HF-related epithelial dysfunction or
al.38 stated; Japanese by sectional sequencing (16S patients and controls. confounders, such as acid-suppressing
inference. 22 patients, 12 rRNA analysis) Differences in beta diversity medication, induce changes in bacterial
healthy controls between groups. Two composition. Age-related confounders
bacterial genera (Clostridium may explain age-related differences;
and Dorea) less abundant in currently uncertain how much HF
patients. Differences in beta influences such differences
diversity, phyla, and few
bacterial genera between

34
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
CVR-2019-0006-R1

younger (<60) and older


(>60) patients
Cui et al.39 Chinese; 53 patients, 31 Cross- Metagenomics 86 bacterial genera Bacterial dysbiosis contributes to host
controls sectional and metabolomics differentially enriched inflammation, thus aggravating HF status.
analysis between groups. Differences Increased TMAO leading to renal
extending to metabolic dysfunction may also aggravate HF
profiles. Genes for TMAO
synthesis enriched in patients
Katsimichas Japanese; 28 patients, 19 Cross- Amplicon Groups differing in beta but Gut endothelial dysfunction in patients
et al.40 healthy controls sectional sequencing (16S not alpha diversity. Few may be responsible for bacterial dysbiosis
rRNA analysis) bacterial genera significantly
different between groups
(Streptococcus, Veillonella,
SMB53). Metabolic profiles
significantly different, various
gene families differentially
enriched or depleted in
patients
Tang et al.41 Race and ethnicity not Prospective Liquid chromato- Fasting plasma TMAO levels Overt or subclinical atherosclerosis
stated; Americans by patient graphy/mass higher in patients. Adjusted influenced by proatherogenic TMAO may
inference. 720 patients, cohort/Cross- spectrometry for higher TMAO levels aggravate HF status
300 healthy controls sectional TMAO associated with increased
comparison measurement mortality risk
to healthy
controls
Hypertension
Yang et Race and ethnicity not Cross- Amplicon Alpha diversity lower in Gut dysbiosis-related host inflammation
al.50 stated; Americans by sectional sequencing (16S patients. Groups also different contributes to the establishment and
inference. 7 patients, 10 rRNA analysis) in beta diversity maintenance of high blood pressure
controls.
Li et al.54 Race and ethnicity not Cross- Metagenomics Alpha and beta diversity Host inflammation induced by bacterial
stated; Chinese by sectional and metabolomics different between healthy dysbiosis contributes to hypertension
inference. 99 hypertensive analysis controls and prehypertensive/ pathophysiology
patients, 56 pre- hypertensive subjects. Fecal
hypertensive controls, 41 transplantation of
healthy controls. hypertensive subject
microbiota resulted in higher
blood pressure in mice

35
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
CVR-2019-0006-R1

Yan et al.55 Race and ethnicity not Cross- Metagenomics Beta diversity visualization None provided
stated; Chinese by sectional analysis different between healthy
inference. 60 hypertensive controls and hypertensive
patients, 60 healthy subjects. Klebsiella,
controls Clostridium, Streptococcus,
Parabacteroides, Eggerthella,
and Salmonella enriched in
patients, Faecalibacterium,
Roseburia, and Synergistetes
enriched in controls.
Lipopolysaccharide
biosynthesis and TMA
production genes enriched in
patients, SCFA genes
depleted
Atherosclerosis
Koren et Caucasians; Swedes by Cross- Amplicon Plaque and gut microbiomes Bacteria phagocytosed by macrophages in
al.74 inference. 15 patients, 15 sectional sequencing (16S different in beta diversity; the gut migrate to atherosclerotic plaques
healthy controls rRNA analysis) several bacterial phylotypes
common between
atherosclerotic plaques and
gut microbiota within the
same individual. Patient and
control groups similar in
terms of gut microbiota

Karlsson et Caucasians; Swedes by Cross- Metagenomics Patients and controls different Anti-inflammatory properties depleted in
al.75 inference. 12 patients, 13 sectional analysis in beta diversity. A few the gut microbiome of patients may
controls genera significantly different influence atherosclerotic procedure
in relative abundance between
groups (Collinsella,
Roseburia, Eubacterium).
Metabolic profiles also
different between groups,
based on KEGG orthologs
Emoto et Race and ethnicity not Cross- Terminal- The order Lactobacillales was Gut microbiota affects CAD through
al.76 stated; Japanese by sectional restriction increased in patients, the immune system-related mechanisms
inference. 39 patients, 30 fragment length phylum Bacteroidetes was
non-CAD controls, 50 polymorphism decreased
healthy controls analysis of

36
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
CVR-2019-0006-R1

bacterial DNA
from stool
samples
Yin et al.78 Race and ethnicity not Cross- Amplicon Alpha diversity greater in Stroke itself or stroke treatment reduces
stated; Chinese by sectional sequencing (16S patients, beta diversity TMAO levels by unknown pathways
inference. 322 patients, rRNA analysis) adequately separated two
231 controls for gut microbiota subgroups of 47 age- and sex-
and liquid matched pairs of patients and
chromato- controls. TMAO levels lower
graphy/mass in patients than in controls
spectrometry for
TMAO
measurement
Jie et al.79 Chinese; 218 patients, 187 Cross- Metagenomics Groups distinct in beta Patient gut microbiota linked to
healthy controls sectional analysis diversity, similar in alpha inflammatory profiles that may influence
diversity. Several species atherosclerosis
differentially abundant
between groups. Metabolic
profiles also different
between groups, based on
KEGG orthologs
Wang et Race and ethnicity not Observational Liquid chromato- TMAO predicts CVD risk in TMAO influences macrophage foam cell
al.80 stated; Multiracial by for human graphy/mass humans; TMAO promotes formation
inference. For TMAO- subjects/Int- spectrometry and atherosclerosis in
based CVD prediction: erventional other metabolo- atherosclerosis-prone rodents
cohort of 1,876 patients for rodents mics techniques
for TMAO
measurement
Tang et al.81 Race and ethnicity not Prospective Liquid chromato- TMAO levels predict TMAO enhances the accumulation of
stated; Americans by patient cohort graphy/mass increased CVD risk cholesterol in macrophages and that of
inference. 4,007 patients for main spectrometry for independently of other risk foam cells in arterial walls.
for main prospective study study leg TMAO factors
measurement
FISH: fluorescence in situ hybridization, rRNA: ribosomal RNA, HF: heart failure, TMAO: trimethylamine N-oxide, SCFA: short-chain fatty
acid, KEGG: Kyoto Encyclopedia of Genes and Genomes, CAD: coronary artery disease, CVD: cardiovascular disease.

37
CVR-2019-0006-
R1

Table 2. Pathophysiological mechanisms linking the intestinal microbiota with

cardiovascular disease.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Disorder Pathophysiological mechanisms Key molecules
Heart failure • Reduced cardiac output may cause gut epithelial dysfunction • Cytokines (e.g. IL-1, TNF)
• Gut epithelial dysfunction may alter gut epithelial-microbial • Short-chain fatty acids
interactions, leading to dysbiosis (acetate, butyrate,
• A compromised intestinal epithelial barrier allows leakage of propionate)
bacterial LPS into the bloodstream, causing systemic inflammation • LPS
and aggravating heart failure status

Hypertension •Bacterial short-chain fatty acids affect immune and epithelial • Short-chain fatty acids
functions important in modulation of blood pressure (acetate, butyrate,
•Toxic bacterial byproducts may affect blood pressure physiology propionate)
•Microbiota-related host systemic inflammation may affect blood • TMAO
pressure levels •Tryptophan
•Enteric nervous system influenced by dysbiosis may affect blood •GABA
pressure by contributing to autonomic dysregulation •Aldosterone
•Dysbiosis modulating the gut renin-angiotensin-aldosterone •ACE2
system may affect blood pressure levels

Atherosclerosis • Bacterial infections activate inflammatory responses • Cytokines


• Bacterial compounds (LPS) leaking into the host's bloodstream • Toll-like receptors
cause systemic inflammation • Short-chain fatty acids
• Bacterial metabolites may be involved in proatherogenic (acetate, butyrate,
pathways. TMAO may inhibit reverse cholesterol transport and propionate)
increase foam cell formation • Bile acids
• Reduced bacterial short-chain fatty acid production may favor • LPS
host systemic inflammation • TMAO

LPS: lipopolysaccharide, IL-1: interleukin-1, TNF: tumor necrosis factor, TMAO: trimethylamine N-oxide,
GABA: gamma-aminobutyric acid, ACE2: angiotensin-converting enzyme 2.

38
CVR-2019-0006-
R1

Table 3. Glossary of terms.

Term Description

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Amplicon sequencing The analysis involving the identification and PCR amplification of a certain genetic
sequence, e.g. sequences of the 16S ribosomal RNA gene of the prokaryotic organisms
recovered from a stool sample.
Commensalism Non-harmful co-existence of two organisms in which only one of the two gains benefit.
Dysbiosis A change in microbial composition relative to the healthy state or homeostasis.
KEGG Kyoto Encyclopedia of Genes and Genomes. A database for genes and related gene
products involved in various cellular functions, among others. It is accessible at
www.kegg.jp.
Metabolomics The identification and measurement of the metabolites (products or intermediates of
cellular metabolic processes) found in a given sample.
Metagenomics The analysis involving the direct identification of all microorganisms and their genomes
in a given sample, without the need for cultures.
Microbiome A collective term for all the genomes of all the microorganisms in a microbial community
or sample.
Microbiota The community of microbes living in a given ecological site.
Mutualism Mutually beneficial relationship between host and hosted organisms.
Next generation Modern DNA sequencing techniques, as opposed to earlier efforts (Sanger sequencing),
sequencing that greatly reduce the time and cost required to sequence entire genomes.
Operational Roughly speaking, an alternative term for a taxon in microbiome studies, usually referring
taxonomic unit to species.
(OTU)
Probiotic The official WHO definition is “live microorganisms which when administered in
adequate amounts confer a health benefit on the host”.
Relative abundance The proportional abundance of an organism relative to the total abundance of all (similar)
organisms in a given ecosystem.
Taxon A taxonomic level for an organism, e.g. genus or family.

39
CVR-2019-0006-
R1

Figure legends

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


Figure 1. Key host-related functions of the intestinal microbiota. The figure shows six

important functions of the intestinal microbiota that benefit its human host, as well as examples of

bacteria associated with these functions and factors influencing gut microbial composition. Many

more such functions have been described in the literature.

Figure 2. Visualization and analysis of microbial data. The visualization of microbial data may

include stacked bar charts, scatterplot graphs, shade plots, and network graphs. On the upper left

(A), a stacked bar chart depicts a hypothetical classification of the microbiota among two groups

of samples on a certain taxonomic level. Each vertical composite bar represents a sample from a

subject, each particular colored bar within the composite bar represents a taxon (e.g. a bacterial

genus), and the height of each colored bar reflects that taxon’s relative abundance. On the upper

right (B), a 3D scatterplot graph depicts a hypothetical output of non-metric multidimensional

scaling (MDS) analysis for two different groups of samples. The positioning of the samples reflects

the ranking of their similarity based on the metric used for analysis – e.g. the Bray-Curtis similarity

coefficient. The important feature is not sample positioning per se, but rather the distances between

the samples and their position relative to each other. In this hypothetical case, the samples tend to

cluster close to other samples of the same color and a clear partitioning in three-dimensional space

is apparent. Hence, the visualized data clearly suggest that the “average” microbial composition

of the red samples is considerably different from the “average” microbial composition of the green

samples. This visual difference would next be tested in formal statistical analysis, e.g. by

PERMANOVA.

40
CVR-2019-0006-
R1

On the lower left (C), a shade plot (heatmap) depicts the relative abundance of 8 bacterial genera

for two groups of samples (10 samples for each group). In this hypothetical example, relative

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


abundances are reflected by the colored shading, which ranges from white (0%) to black (60%, the

highest relative abundance seen in these data). The differences are readily apparent between the

groups. Bacterial genera 1 to 4 are scarcely present in group 2, while the opposite is true for

bacterial genera 5 to 8. Again, such a group separation, although clear-cut, would next be tested

by an appropriate statistical method. On the lower right (D), an interaction network of bacterial

genera (nodes) and their correlations (lines) is presented. Such a visualization is generally an

advanced way of depicting the sample data, based on complex statistical software that should take

into account the compositionality of relative abundance values. Comparing networks of different

groups of samples may reveal different patterns of associations, which may be caused by differing

properties of the microbiota in each group. A discussion of networks and their applications can be

found in the supplementary text of our original study (reference 40).

41
CVR-2019-0006-
R1

Figure 3. The heart-gut axis in heart failure. The pathophysiology of heart failure may lead to

increased leakage of gut microbial compounds into the bloodstream and cause low grade systemic

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019


inflammation, which may aggravate heart failure status and complete a vicious cycle.

Accumulating evidence also suggests that the composition of the intestinal microbiota is altered in

the context of heart failure. LPS: lipopolysaccharide.

Figure 4. Evidence linking the intestinal microbiota with arterial hypertension. In an

interesting experiment by Li et al.54 (A), the intestinal microbiota of human donors with or without

hypertension was transplanted to germ-free mice, i.e. mice manipulated so that no microbes would

exist in their intestinal tracts. Ten weeks after transplantation, blood pressure was measured by the

tail cuff method and it was shown that the mice that had received the microbiota of hypertensive

subjects had significantly higher blood pressure than the mice that had received the microbiota of

the control subject. As this experiment involved a very small number of donors (2 hypertensives

and 1 normotensive), more evidence is required to solidify these findings. Part B of the figure

shows a schematic representation of how a healthy microbiota may modulate blood pressure and

reduce the risk of hypertension under a fiber rich diet. HTN: hypertension, SCFA: short-chain fatty

acids, BP: blood pressure.

Figure 5. Microbial links to atherosclerosis. The association of the intestinal microbiota with

atherosclerosis is based on infection, changes in lipid metabolism, induction of inflammatory

pathways, and production of bacterial metabolites that may exert proatherogenic effects. Overall,

inflammation caused by bacterial products or compounds may characterize the pathway linking

the microbiota with the atherosclerotic process. LPS: lipopolysaccharide, LBP: lipopolysaccharide

42
CVR-2019-0006-
R1

binding protein, CD14: cluster of differentiation 14, TLR: Toll-like receptor, NF-κB: nuclear

factor kappa-light-chain-enhancer of activated B cells, TMAO: trimethylamine N-oxide.

Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019

43
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Figure 1
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Figure 2
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Figure 3
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Figure 4
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Figure 5
Downloaded from https://academic.oup.com/cardiovascres/advance-article-abstract/doi/10.1093/cvr/cvz135/5510547 by Bethel University user on 05 June 2019
Graphical Abstract

You might also like