You are on page 1of 6

Clinica Chimica Acta 507 (2020) 236–241

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/cca

Review

Butyrate-producing bacteria and the gut-heart axis in atherosclerosis T


a,1 a,1 b a a,⁎ c,⁎
Wujun Chen , Shun Zhang , Jianfeng Wu , Ting Ye , Shuai Wang , Pan Wang ,

Dongming Xingd,a,
a
Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
b
Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, The Second Affiliated Hospital of
University of South China, Hengyang, Hunan 421001, China
c
Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and
Peking Union Medical College, Beijing 100021, China
d
School of Life Sciences, Tsinghua University, Beijing 100084, China

A R T I C LE I N FO A B S T R A C T

Keywords: The gut microbiota plays an important role in controlling atherosclerosis progression to support the link between
Butyrate the gut and coronary heart disease. Recent studies have shown that an imbalance in the gut-heart axis due to the
TMAO gut microbiota plays an important role in atherosclerosis progression. The gut microbiota promotes the devel-
LPS opment of atherosclerosis by producing intermediate metabolites, including TMAO, LPS, PAGln and reducing
PAGln
butyrate. TMAO and PAGln might be potential biomarkers of coronary heart disease. Many studies have shown
Gut-heart axis
that butyrate-producing bacteria prevent atherosclerosis progression by producing butyrate and maintaining the
Atherosclerosis
bacterial balance, the intestinal barrier function and the expression of various genes, including those encoding
lipids and those related to immunity, inflammation, differentiation, apoptosis, phagocytosis and efferocytosis.
This review focuses on recent advances in our understanding of the interplay between butyrate-producing
bacteria and the gut-heart axis in atherosclerosis.

1. Introduction formation of foam cells. Ox-LDL also regulates initial monocyte re-
cruitment and the inflammatory response, including the expression of
Atherosclerosis is a major factor in coronary heart disease (CHD), interleukin-7 (IL-7), annexin A1 (ANXA1), and IL-15. Therefore, ox-LDL
which is characterized by the formation of fat-laden plaques in large plays a key role in foam cell formation and the inflammatory response
and medium-sized vessels, and is considered a chronic inflammatory [3].
disease of the artery wall. The development of atherosclerosis is asso- The gut microbiota in the gastrointestinal tract, which comprises
ciated with the recruitment of monocytes and their differentiation into trillions of bacteria, is a complex community with metabolic activities
macrophages, and the resulting macrophages take up lipids to become that contributes to education of the immune response by providing
foam cells that affect the inflammatory response. The processes of foam essential nutrients and the maintenance of human health by protecting
cell formation and chronic inflammation are regulated by a cholesterol against pathogens [4]. In addition, accumulating evidence suggests that
imbalance and the expression of inflammatory cytokines (e.g., IL-10 the gut microbiota plays an important role in controlling atherosclerosis
and IL-1β) [1,2], and the cholesterol balance is correlated with cho- progression to support the link between the gut and CHD [5,6]. Bacteria
lesterol synthesis, uptake, esterification, and efflux. An uncontrolled with the capacity to produce butyrate, trimethylamine-N-oxide
uptake of oxidized low-density lipoprotein (ox-LDL) by oxidized low- (TMAO), endotoxin (LPS) and phenylacetyl glutamine (PAGln) have
density lipoprotein receptor 1 (LOX-1), SRA, and CD36, excessive been found to play roles in the gut-heart axis in atherosclerosis (Fig. 1).
cholesterol esterification by acetyl-CoA acetyltransferase 1 (ACAT1) Many studies have shown that a reduction in butyrate in the gut pro-
and/or impaired cholesterol efflux by ATP-binding cassette transporter motes local inflammation and foam cell formation, aggravates dysbiosis
A1 (ABCA1) and ABCG1 result in the accumulation of cholesterol esters and contributes to impairments in the gut barrier function, and the
stored as cytoplasmic lipid droplets and subsequently trigger the latter exacerbates the release and invasion of bacterial toxins such as


Corresponding authors at: Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong
266071, China and School of Life Sciences, Tsinghua University, Beijing 100084, China.
E-mail addresses: sdwftcmws@163.com (S. Wang), wangpan@cicams.ac.cn (P. Wang), xdm_tsinghua@163.com (D. Xing).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.cca.2020.04.037
Received 16 March 2020; Received in revised form 29 April 2020; Accepted 29 April 2020
Available online 04 May 2020
0009-8981/ © 2020 Elsevier B.V. All rights reserved.
W. Chen, et al. Clinica Chimica Acta 507 (2020) 236–241

Fig 1. Role of microbial-derived substances in gut-heart axis crosstalk during atherosclerosis progression.

LPS and TMAO and thereby further fuels local and systemic in- Red meat has markedly higher levels of choline, phosphatidylcho-
flammation [7,8]. This review focuses on recent advances in our un- line and L-carnitine, which are precursors of trimethylamine (TMA).
derstanding of the interplay between butyrate-producing bacteria and Thus, eating a high quantity of red meat promotes the expression of
the gut-heart axis in atherosclerosis. TMA precursors by gut microbiota and directly contributes to athero-
sclerosis pathology. TMA is oxidized by FMO3 in the liver to TMAO
[13], and high TMAO levels were found to be associated with an in-
2. Microbial-derived PAGln, LPS and TMAO impair the balance of
creased risk of incident major adverse cardiovascular events (MACEs) in
the gut-heart axis
a cohort of 4007 patients who underwent coronary angiography and
were followed-up for 3 years [14]. In another study, high concentra-
The gut microbiota regulate human physiology through the release
tions of TMAO and its precursors were associated with increased risks of
of different classes of metabolites. An imbalance in the gut microbiota
MACEs and all-cause mortality in 19,256 patients, independent of tra-
produces many harmful substances, including TMAO, LPS, PAGln, in-
ditional risk factors [15]. Another study with 26,167 patients found a
doxyl sulfate (IS), and p-cresylsulfate (PCS), and reduces many bene-
positive dose-dependent association among the TMAO plasma levels,
ficial substances, including short-chain fatty acids (SCFAs), particularly
cardiovascular risk and mortality [16]. TMAO contributes to athero-
butyrate [9,10]. As a result, the intestinal barrier is damaged, and large
sclerosis development by interfering with cholesterol transport, en-
amounts of toxic substances, such as PAGln, LPS and TMAO, enter the
dothelial and smooth muscle cell activation, foam cell formation,
blood and thereby influence known risk factors for atherosclerosis, such
myocardial and inflammatory responses and platelet aggregation
as platelet invasiveness and thrombosis, foam cell formation, the in-
[17–20]. Taken together, the results reveal that microbial-derived
flammatory response, and oxidative stress. PAGln has been found to be
TMAO, LPS and PAGln play a key role in gut-heart axis crosstalk during
associated with cardiovascular disease and death in an independent
atherosclerosis progression.
cohort (n = 4,000 subjects). The majority of ingested phenylalanine,
which is the precursor for PAGln, is absorbed in the small intestines, but
unabsorbed phenylalanine is metabolized to phenylpyruvic acid and 3. Butyrate-producing bacteria maintain balance of the gut-heart
subsequently phenylacetic acid by the gut microbiota in the large in- axis
testines. Following absorption into the portal system, phenylacetic acid
is readily metabolized in the liver to produce PAGln, and PAGln in- A reduction in dietary fiber results in a reduction in the bacterial
creases platelet responsiveness and thrombosis through G-protein cou- production of butyrate. Butyrate is the major source of energy to the
pled receptors, including α2A, α2B, and β2-adrenergic receptors [10]. colonic mucosa needed for the maintenance of human gut health and is
These results suggest that PAGln is a potential biomarker of cardio- an important regulator of the bacterial balance, the intestinal barrier
vascular disease. In addition, the LPS levels in 516 patients aged function and the expression of various genes, including those encoding
50–79 years were measured, and the results showed that the risk of lipids and those related to immunity, inflammation, differentiation,
incident atherosclerosis was increased three fold in patients with serum apoptosis, phagocytosis and efferocytosis. Butyrate enters colonic epi-
LPS levels beyond the 90th percentile [11]. High LPS levels are asso- thelial cells via solute carrier (SLC) transporters and then induces the
ciated with obesity, metabolic syndrome, diabetes and CHD events, as upregulation of tight junctions and the maintenance of gut barrier
determined through a 10-year follow-up study of 2452 patients, and function. These functions inhibit permeability, and thus, fewer mole-
these findings are independent of other established risk factors [12]. cules of LPS and other endotoxins, including TMAO, reach the general
Thus, high serum LPS activity due to an imbalance in the gut microbiota circulation and induce systemic inflammation, which could regulate
is associated with the development of atherosclerosis. macrophages and contribute to the formation of atherosclerotic plaques

237
W. Chen, et al. Clinica Chimica Acta 507 (2020) 236–241

Table 1 expression of SP1 by inhibiting HDACs, and these effects damage the
Butyrate-producing bacteria in the human gut. structure of the complex [29,30]. HDAC3 decreases ABCA1 expression
Bacterial species/group Clostridial cluster by reducing the LXRα promoter level [31]. SP1 binds to the promoter of
miR-27a and promotes the transcription and expression of miR-27a
Clostridium acetobutylicum I [32,33]. Previous studies conducted in our and other laboratories have
Clostridium butyricum I
demonstrated that ABCA1 is a target gene of miR-27a [34–38]. A
Clostridium saccharobutylicum I
Clostridium tyrobutyricum I
bioinformatics analysis using GeneCards showed that HDAC1, HDAC2
Anaerotruncus colihominis IV and HDAC3 exert potential effects in promoting the transcriptional
Butyricicoccus pullicaecorum IV regulation of miR-27a. Thus, butyrate increases ABCA1 expression by
Clostridium orbiscidens IV inhibiting the HDAC3/SP1-miR-27a pathway. Butyrate also induces
Faecalibacterium prausnitzii IV
macrophage phagocytosis and efferocytosis by increasing ANXA1 ex-
Papillibacter cinnamivorans IV
Subdoligranulum variabile IV pression [39–41], whereas miR-27a reduces ANXA1 expression [42].
Caldocellum saccharolyticum x However, the mechanism through which butyrate and miR-27a affect
Megasphaera elsdenii IX ANXA1 is unclear. Interestingly, an important recent study showed that
Eubacterium cylindroides XVI
ABCA1 is involved in ANXA1 expression and secretion [43,44]. In
Anaerostipes butyraticus XIVa
Anaerostipes caccae XIVa
macrophages and endocrine cells, ANXA1 secretion requires the action
Anaerostipes hadrus XIVa of ABCA1 [45]. ANXA1, which acts as an anti-inflammatory protein, is
Anaerostipes rhamnosivorans XIVa localized in macrophages and endothelial cells in human coronary
Butyrivibrio crossotus XIVa atherosclerotic plaques [46–48]. Previous studies conducted in our and
Butyrivibrio fibrisolvens XIVa
other laboratories demonstrated that ABCA1 combined with apoA-I
Butyrivibrio proteoclasticus XIVa
Coprococcus catus XIVa plays a critical role in cholesterol efflux and anti-inflammatory activity
Coprococcus comes XIVa [35,49,50]. Thus, further studies on ABCA1 are likely to shed important
Coprococcus eutactus XIVa light on potential therapeutic targets for cholesterol efflux and ANXA1-
Clostridium hathewayi XIVa mediated anti-inflammatory effects. The available evidence demon-
Clostridium indolis XIVa
Clostridium nexile XIVa
strates that butyrate increases ANXA1 expression by regulating the
Clostridium symbiosum XIVa HDAC3/SP1-miR-27a-ABCA1 pathway, and it should be noted that SP1
Eubacterium hallii XIVa is a target gene of miR-27a [51], which suggests that HDAC3/SP1-miR-
Eubacterium limosum XIVa 27a could form a negative feedback loop. In addition, the effects of
Eubacterium ramulus XIVa
butyrate on cells might be mediated by G protein-coupled receptors
Eubacterium rectale XIVa
Eubacterium ruminantium XIVa (GPRs), including GRP41, GPR43, and GPR109A. Interestingly,
Eubacterium ventriosum XIVa GPR109a differs from GPR41 and GPR43 in that it is activated by longer
Roseburia cecicola XIVa SCFAs, particularly butyrate, and is also activated by niacin [52,53].
Roseburia faecis XIVa GPR109a activation stimulates ABCA1 expression in macrophages and
Roseburia hominis XIVa
Roseburia intestinalis XIVa
then promotes apoA-I-mediated cholesterol efflux [54–56], which
Roseburia inulinivorans XIVa suggests that butyrate enhances ABCA1 expression by increasing
Ruminococcus gnavus XIVa GPR109A expression. Butyrate also increases the expression of peroxi-
Ruminococcus obeum XIVa some proliferator-activated receptor-gamma (PPARγ), which increases
SS3/4, GM2/1 XIVa
ABCA1 expression [57]. However, additional studies are needed to
Shuttleworthia satelles XIVa
SSC/2, SS2/1 XIVa elucidate the mechanism underlying the relationship between butyrate
and ABCA1.
Many butyrate-producing bacteria have been identified by culture-
[21,22]. Butyrate is mainly produced in the gut, and the concentration based approaches, DNA sequence analysis and sequence-based detec-
of butyrate in the blood passing from the intestinal tract to the liver, tion methods (Table 1) [52,58,59]. Butyrate-producing bacteria play a
heart, and lungs mimics the production rate in the large intestine. In- key role in human health, and these bacteria, including Roseburia in-
creased gut production of butyrate further increases the circulating testinalis, Butyrivibriocrossotus and Faecalibacterium cf. prausnitzii, are
level of butyrate [23]. Compared with fasted human subjects, nonfasted relatively depleted in atherosclerotic cardiovascular disease (ACVD)
subjects exhibit increased absolute and relative concentrations of bu- and type 2 diabetes (T2D) samples. The butyrate-producing bacterium
tyrate in portal blood [24]. Butyrate has been shown to regulate reverse Roseburia intestinalis is inversely correlated with atherosclerotic lesion
cholesterol transport by stimulating apoA-IV-containing lipoprotein development in mice, and the addition of Roseburia intestinalis in
secretion. Butyrate treatment also reduces cholesterol absorption by combination with a high-fiber diet reduces the sizes of atherosclerotic
increasing the transcriptional activity of liver X receptor (LXR), ATP- plaques in the aorta. Exogenous butyrate administered via the diet also
binding cassette transporters G5 and G8 (ABCG5 and G8) expression reduces the plaque sizes in apoE-/- and LDLR-/- mice [60]. Roseburia
and reducing Niemann-Pick C1-Like 1 (NPC1L1) in Caco-2 cells [25]. intestinalis improves the gut barrier function by increasing the butyrate
Butyrate influences lipoprotein assembly and secretion by altering the levels and then decreases the permeability, circulating LPS, systemic
membrane fluidity, the posttranscriptional modification of apolipo- inflammation (reduced proinflammatory cytokine expression in pla-
protein (apo) B 48 and 100, modulation of microsomal triglyceride (TG) ques), and macrophage infiltration into plaques. Akkermansia mucini-
transfer protein (MTP), the expression of sterol enzyme-encoding genes, phila also produces butyrate and decreases the size of plaques through
and lipoprotein secretion [26]. Butyrate increases ABCA1 expression in reductions in the intestinal permeability, foam cell formation, circu-
macrophages by regulating the Sp1 pathway but not LXRα [27]. lating LPS levels and inflammation in apoE-/- mice [61–64]. Faecali-
However, the mechanism through which butyrate affects SP1 and SP1 bacterium prausnitzii is an anti-inflammation-associated bacterium that
on ABCA1 is unclear. Interestingly, SP1 is often compounded with produces butyrate. Treatment with atorvastatin increases the abun-
HDACs and HATs to achieve the acetylation-mediated regulation of dance of Faecalibacterium prausnitzii in 27 hypercholesterolemic pa-
genes. The HDACs that form protein complexes with SP1 are mainly tients compared with that found in 15 untreated hypercholesterolemic
HDAC1, HDAC2 and HDAC3, and the HATs are mainly CBP/p300 and patients [65]. Clostridium butyricum promotes a hepatic antioxidant
PCAF [28]. Notably, butyrate is an inhibitor of HDACs and inhibits the status and decreases the levels of serum lipids and proinflammatory
cytokines by producing butyrate to enhance PPARγ. Clostridium

238
W. Chen, et al. Clinica Chimica Acta 507 (2020) 236–241

Table 2
Main metabolites produced by the gut microbiota.
Metabolites Substrate Synthesis Reference

PAGln Phenylalanine Phenylalanine by gut microbiota → Phenylacetic acid (PAA) 10


Liver: PAA conjugation to the amino acid glutamine (Gln) of Gln PA-transferase and PA-CoA-ligase → PAGln
(major product in humans) or PAGly (major product in mice)
LPS A structural component of the microbiota itself 11, 12
TMAO Choline, phosphatidyl choline, L- Choline, phosphatidyl choline, and L-carnitine by gut microbiota → TMA 13–16
carnitine in red meat Liver: oxidation of TMA by FMO3 → TMAO
Butyrate Dietary fiber Dietary fiber fermentation by the gut microbiota → Butyrate 52, 58, 59

butyricum increases the abundance of the gut microbiota and particu- LXR Liver X receptor Cholesterol efflux
ABCG5 ATP-binding cassette transporters Cholesterol efflux
larly enriches butyrate-producing bacteria [66,67]. However, the role
G5
of Faecalibacterium prausnitzii and Clostridium butyricum in athero- ABCG8 ATP-binding cassette transporters Cholesterol efflux
sclerosis is unclear. The above-mentioned results indicate that butyrate G8
plays a key role in reducing atherosclerosis development and main- NPC1L1 Niemann-Pick C1-Like 1 Cholesterol uptake
taining the gut-heart axis balance via butyrate-producing bacteria. apoB Apolipoprotein B Chylomicron assembly and se-
cretion
MTP Microsomal triglyceride transfer Chylomicron assembly and se-
4. Summary protein cretion
HDAC Histone deacetylase Histone acetylation
Atherosclerosis is known as the main cause of mortality and mor- CBP/p300 CREB-binding protein Histone acetylation
PCAF p300/CBP-associated factor Histone acetylation
bidity in patients with coronary heart disease. Recent evidence shows
ApoA-I Apolipoprotein A-I Cholesterol efflux
that an imbalance in the gut-heart axis due to the gut microbiota plays ApoA-IV Apolipoprotein A-IV Cholesterol efflux
an important role in atherosclerosis progression. The gut microbiota GPRs G protein-coupled receptors Butyrate receptors
promotes the development of atherosclerosis by producing intermediate PPARγ Peroxisome proliferator-activated Cholesterol efflux
metabolites, including TMAO, LPS and PAGln, and reducing butyrate receptor-gamma

(Table 2). Butyrate-producing bacteria prevent atherosclerosis by pro-


ducing butyrate and maintaining the bacterial balance, the intestinal 6. Statement of ethics
barrier function and the expression of various genes, including those
that encode lipids and those related to immunity, inflammation, dif- The authors have no ethical conflicts to disclose.
ferentiation, apoptosis, phagocytosis and efferocytosis. Previous studies
have shown that butyrate-producing bacteria belonging to Clostridium Declaration of Competing Interest
cluster XIVa harbor the gene needed to metabolize precursors, in-
cluding choline, phosphatidylcholine and L-carnitine, into TMA and
The authors declare that they have no known competing financial
TMAO. These bacteria might be harmful if the diet (such as a red meat-
interests or personal relationships that could have appeared to influ-
based diet) is rich in choline, phosphatidylcholine, or carnitine. How-
ence the work reported in this paper.
ever, further studies in this research area must be performed. Butyrate-
producing bacteria might offer opportunities for manipulating nutrition
Acknowledgments
and opens up interesting new prospects related to the conservative
treatment of CHD, but extensive validation is needed. Future studies
We thank colleagues in Dr. Dongming Xing and Jianfeng Wu’s la-
should assess the role of more butyrate-producing bacteria in the
boratory for the technical help provided and the stimulating discussions
treatment of CHD.
during this investigation.
5. List of abbreviations
Funding

Abbreviation Full name Main function The authors are grateful for the financial support provided by the
Qingdao Major Scientific and Technological Project for Distinguished
CHD Coronary heart disease Scholars (20170103), the Laoshan Major Scientific and Technological
IL-1β Interleukin 1β Inflammatory response
IL-10 Interleukin 1β Inflammatory response
Project for Distinguished Scholars (20181030), the China Postdoctoral
ox-LDL Oxidized low-density lipoprotein Foam cell formation and in- Science Foundation (2019M652331, 2018M642619), the Qingdao
flammatory response Postdoctoral Application Project (RZ1900013298, 2018121236,
LOX-1 Low-density lipoprotein receptor Cholesterol uptake 2018121238), the Department of Health of Shandong Province
1
(2018WS068), and the Department of Health of Hunan Province
SRA Cholesterol uptake
CD36 Cholesterol uptake (20201533).
ACAT1 Acetyl-CoA acetyltransferase 1 Cholesterol esterification
ABCA1 ATP-binding cassette transporter Cholesterol efflux References
A1
ABCG1 ATP-binding cassette transporter Cholesterol efflux
[1] W. Chen, Y. Wu, Q. Lu, S. Wang, D. Xing, Endogenous ApoA-I expression in mac-
G1
rophages: a potential target for protection against atherosclerosis, Clin. Chim. Acta
ANXA1 Annexin A1 Inflammatory response
505 (2020) 55–59.
TMAO Trimethylamine-N-oxide Foam cell formation and pla- [2] A.V. Poznyak, W.K. Wu, A.A. Melnichenko, R. Wetzker, V. Sukhorukov,
telet reactivity A.M. Markin, V.A. Khotina, A.N. Orekhov, Signaling pathways and key genes in-
LPS Endotoxin Inflammatory response volved in regulation of foam cell formation in atherosclerosis, Cells 9 (3) (2020).
PAGln Phenylacetyl glutamine Platelet reactivity [3] A.N. Orekhov, Y. Oishi, N.G. Nikiforov, A.V. Zhelankin, L. Dubrovsky, I.A. Sobenin,
FMO3 Flavin-containing monooxygenase Oxidation A. Kel, D. Stelmashenko, V.J. Makeev, K. Foxx, X. Jin, H.S. Kruth, M. Bukrinsky,
3 Modified LDL particles activate inflammatory pathways in monocyte-derived
SLC Solute carrier Transporter macrophages: transcriptome analysis, Curr. Pharm. Des. 24 (26) (2018) 3143–3151.

239
W. Chen, et al. Clinica Chimica Acta 507 (2020) 236–241

[4] W. Chen, S. Wang, Y. Wu, X. Shen, Z. Guo, Q. Li, D. Xing, Immunogenic cell death: a Nat. Commun. 10 (1) (2019) 5719.
link between gut microbiota and anticancer effects, Microb. Pathog. 141 (2020) [29] J.R. Davie, Inhibition of histone deacetylase activity by butyrate, J. Nutr. 133 (7
103983. Suppl) (2003) 2485S–2493S.
[5] E. Sanchez-Rodriguez, A. Egea-Zorrilla, J. Plaza-Diaz, J. Aragon-Vela, S. Munoz- [30] A.J. Wilson, D.S. Byun, N. Popova, L.B. Murray, K. L'Italien, Y. Sowa, D. Arango,
Quezada, L. Tercedor-Sanchez, F. Abadia-Molina, The gut microbiota and its im- A. Velcich, L.H. Augenlicht, J.M. Mariadason, Histone deacetylase 3 (HDAC3) and
plication in the development of atherosclerosis and related cardiovascular diseases, other class I HDACs regulate colon cell maturation and p21 expression and are
Nutrients 12 (3) (2020). deregulated in human colon cancer, J. Biol. Chem. 281 (19) (2006) 13548–13558.
[6] M. Troseid, G.O. Andersen, K. Broch, J.R. Hov, The gut microbiome in coronary [31] W. Huang, J. Zhou, G. Zhang, Y. Zhang, H. Wang, Decreased H3K9 acetylation level
artery disease and heart failure: Current knowledge and future directions, of LXRalpha mediated dexamethasone-induced placental cholesterol transport
EBioMedicine 52 (2020) 102649. dysfunction, Biochim. Biophys. Acta, Mol. Cell. Biol. Lipids 1864 (12) (2019)
[7] M.D. Pieczynska, Y. Yang, S. Petrykowski, O.K. Horbanczuk, A.G. Atanasov, 158524.
J.O. Horbanczuk, Gut microbiota and its metabolites in atherosclerosis develop- [32] B.Y. Kang, K.K. Park, D.E. Green, K.M. Bijli, C.D. Searles, R.L. Sutliff, C.M. Hart,
ment, Molecules 25 (3) (2020). Hypoxia mediates mutual repression between microRNA-27a and PPARgamma in
[8] Y. Zhu, Q. Li, H. Jiang, Gut microbiota in atherosclerosis: focus on trimethylamine the pulmonary vasculature, PLoS ONE 8 (11) (2013) e79503.
N-oxide, APMIS (2020). [33] S. Min, L. Li, M. Zhang, Y. Zhang, X. Liang, Y. Xie, Q. He, Y. Li, J. Sun, Q. Liu,
[9] C. Cosola, M.T. Rocchetti, A. Cupisti, L. Gesualdo, Microbiota metabolites: pivotal X. Jiang, Y. Zhang, Y. Che, R. Yang, TGF-beta-associated miR-27a inhibits dendritic
players of cardiovascular damage in chronic kidney disease, Pharmacol. Res. 130 cell-mediated differentiation of Th1 and Th17 cells by TAB3, p38 MAPK, MAP2K4
(2018) 132–142. and MAP2K7, Genes Immun. 13 (8) (2012) 621–631.
[10] Ina Nemet, Prasenjit Prasad Saha, Nilaksh Gupta, Weifei Zhu, Kymberleigh [34] W.J. Chen, K. Yin, G.J. Zhao, Y.C. Fu, C.K. Tang, The magic and mystery of
A. Romano, Sarah M. Skye, Tomas Cajka, Maradumane L. Mohan, Lin Li, microRNA-27 in atherosclerosis, Atherosclerosis 222 (2) (2012) 314–323.
Yuping Wu, Masanori Funabashi, Amanda E. Ramer-Tait, Sathyamangla [35] M. Zhang, J.F. Wu, W.J. Chen, S.L. Tang, Z.C. Mo, Y.Y. Tang, Y. Li, J.L. Wang,
Venkata Naga Prasad, Oliver Fiehn, Federico E. Rey, W.H. Wilson Tang, Michael X.Y. Liu, J. Peng, K. Chen, P.P. He, Y.C. Lv, X.P. Ouyang, F. Yao, D.P. Tang,
A. Fischbach, Joseph A. DiDonato, Stanley L. Hazen, A cardiovascular disease- F.S. Cayabyab, D.W. Zhang, X.L. Zheng, G.P. Tian, C.K. Tang, MicroRNA-27a/b
linked gut microbial metabolite acts via adrenergic receptors, Cell 180 (5) (2020) regulates cellular cholesterol efflux, influx and esterification/hydrolysis in THP-1
862–877.e22, https://doi.org/10.1016/j.cell.2020.02.016. macrophages, Atherosclerosis 234 (1) (2014) 54–64.
[11] E. Forkosh, Y. Ilan, The heart-gut axis: new target for atherosclerosis and congestive [36] Y. Yao, Y. Xu, W. Wang, J. Zhang, Q. Li, Glucagon-like peptide-1 improves beta-cell
heart failure therapy, Open Heart 6 (1) (2019) e000993. dysfunction by suppressing the miR-27a-induced downregulation of ATP-binding
[12] K.A. Kallio, K.A. Hatonen, M. Lehto, V. Salomaa, S. Mannisto, P.J. Pussinen, cassette transporter A1, Biomed. Pharmacother. 96 (2017) 497–502.
Endotoxemia, nutrition, and cardiometabolic disorders, Acta Diabetol. 52 (2) [37] T. Shirasaki, M. Honda, T. Shimakami, R. Horii, T. Yamashita, Y. Sakai, A. Sakai,
(2015) 395–404. H. Okada, R. Watanabe, S. Murakami, M. Yi, S.M. Lemon, S. Kaneko, MicroRNA-27a
[13] Z. Wang, A.B. Roberts, J.A. Buffa, B.S. Levison, W. Zhu, E. Org, X. Gu, Y. Huang, regulates lipid metabolism and inhibits hepatitis C virus replication in human he-
M. Zamanian-Daryoush, M.K. Culley, A.J. DiDonato, X. Fu, J.E. Hazen, D. Krajcik, patoma cells, J. Virol. 87 (9) (2013) 5270–5286.
J.A. DiDonato, A.J. Lusis, S.L. Hazen, Non-lethal inhibition of gut microbial tri- [38] W. Mo, J. Zhang, X. Li, D. Meng, Y. Gao, S. Yang, X. Wan, C. Zhou, F. Guo, Y. Huang,
methylamine production for the treatment of atherosclerosis, Cell 163 (7) (2015) S. Amente, E.V. Avvedimento, Y. Xie, Y. Li, Identification of novel AR-targeted
1585–1595. microRNAs mediating androgen signalling through critical pathways to regulate
[14] W.H. Tang, Z. Wang, B.S. Levison, R.A. Koeth, E.B. Britt, X. Fu, Y. Wu, S.L. Hazen, cell viability in prostate cancer, PLoS ONE 8 (2) (2013) e56592.
Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk, N. [39] T. Montero-Melendez, J. Dalli, M. Perretti, Gene expression signature-based ap-
Engl. J. Med. 368 (17) (2013) 1575–1584. proach identifies a pro-resolving mechanism of action for histone deacetylase in-
[15] Y. Heianza, W. Ma, J.E. Manson, K.M. Rexrode, L. Qi, Gut microbiota metabolites hibitors, Cell Death Differ. 20 (4) (2013) 567–575.
and risk of major adverse cardiovascular disease events and death: a systematic [40] D. Mu, Z. Gao, H. Guo, G. Zhou, B. Sun, Sodium butyrate induces growth inhibition
review and meta-analysis of prospective studies, J. Am. Heart Assoc. 6 (7) (2017). and apoptosis in human prostate cancer DU145 cells by up-regulation of the ex-
[16] G.G. Schiattarella, A. Sannino, E. Toscano, G. Giugliano, G. Gargiulo, A. Franzone, pression of annexin A1, PLoS ONE 8 (9) (2013) e74922.
B. Trimarco, G. Esposito, C. Perrino, Gut microbe-generated metabolite trimethy- [41] J. Shin, I.S. Song, J.H. Pak, S.W. Jang, Upregulation of annexin A1 expression by
lamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose- butyrate in human melanoma cells induces invasion by inhibiting E-cadherin ex-
response meta-analysis, Eur. Heart J. 38 (39) (2017) 2948–2956. pression, Tumour Biol. 37 (11) (2016) 14577–14584.
[17] S. Harikrishnan, Diet, the gut microbiome and heart failure, Card. Fail. Rev. 5 (2) [42] T. Colangelo, G. Polcaro, P. Ziccardi, B. Pucci, L. Muccillo, M. Galgani, A. Fucci,
(2019) 119–122. M.R. Milone, A. Budillon, M. Santopaolo, C. Votino, M. Pancione, A. Piepoli,
[18] P. Aldana-Hernandez, K.A. Leonard, Y.Y. Zhao, J.M. Curtis, C.J. Field, R.L. Jacobs, G. Mazzoccoli, M. Binaschi, M. Bigioni, C.A. Maggi, M. Fassan, C. Laudanna,
Dietary choline or trimethylamine N-oxide supplementation does not influence G. Matarese, L. Sabatino, V. Colantuoni, Proteomic screening identifies calreticulin
atherosclerosis development in Ldlr-/- and Apoe-/- male mice, J. Nutr. 150 (2) as a miR-27a direct target repressing MHC class I cell surface exposure in colorectal
(2020) 249–255. cancer, Cell Death Dis. 7 (2016) e2120.
[19] Z. He, W. Hao, E. Kwek, L. Lei, J. Liu, H. Zhu, K.Y. Ma, Y. Zhao, H.M. Ho, W.S. He, [43] L. Li, L. Xu, W. Chen, X. Li, Q. Xia, L. Zheng, Q. Duan, H. Zhang, Y. Zhao, Reduced
Z.Y. Chen, Fish oil is more potent than flaxseed oil in modulating gut microbiota Annexin A1 secretion by ABCA1 causes retinal inflammation and ganglion cell
and reducing trimethylamine-N-oxide-exacerbated atherogenesis, J. Agric. Food apoptosis in a murine glaucoma model, Front. Cell. Neurosci. 12 (2018) 347.
Chem. 67 (49) (2019) 13635–13647. [44] S. Omer, D. Meredith, J.F. Morris, H.C. Christian, Evidence for the role of adenosine
[20] A. Mohammadi, A.G. Najar, M.M. Yaghoobi, Y. Jahani, Z. Vahabzadeh, 5'-triphosphate-binding cassette (ABC)-A1 in the externalization of annexin 1 from
Trimethylamine-N-oxide treatment induces changes in the ATP-binding cassette pituitary folliculostellate cells and ABCA1-transfected cell models, Endocrinology
transporter A1 and scavenger receptor A1 in murine macrophage J774A.1 cells, 147 (7) (2006) 3219–3227.
Inflammation 39 (1) (2016) 393–404. [45] S. Seidel, H. Neymeyer, T. Kahl, T. Roschel, K. Mutig, R. Flower, J. Schnermann,
[21] M. Bastin, F. Andreelli, The gut microbiota and diabetic cardiomyopathy in hu- S. Bachmann, A. Paliege, Annexin A1 modulates macula densa function by in-
mans, Diabetes Metab. (2019). hibiting cyclooxygenase 2, Am. J. Physiol. Renal Physiol. 303 (6) (2012)
[22] K.S. Fluitman, M. Wijdeveld, M. Nieuwdorp, I.J. RG, Potential of butyrate to in- F845–F854.
fluence food intake in mice and men, Gut 67(7) (2018) 1203–1204. [46] J.D.S. Parisi, M.P. Correa, C.D. Gil, Lack of endogenous Annexin A1 increases mast
[23] S.J. Bultman, Bacterial butyrate prevents atherosclerosis, Nat. Microbiol. 3 (12) cell activation and exacerbates experimental atopic dermatitis, Cells 8 (1) (2019).
(2018) 1332–1333. [47] E.Y. Senchenkova, J. Ansari, F. Becker, S.A. Vital, Z. Al-Yafeai, E.M. Sparkenbaugh,
[24] Z. Jie, H. Xia, S.L. Zhong, Q. Feng, S. Li, S. Liang, H. Zhong, Z. Liu, Y. Gao, H. Zhao, R. Pawlinski, K.Y. Stokes, J.L. Carroll, A.M. Dragoi, C.X. Qin, R.H. Ritchie, H. Sun,
D. Zhang, Z. Su, Z. Fang, Z. Lan, J. Li, L. Xiao, J. Li, R. Li, X. Li, F. Li, H. Ren, H.H. Cuellar-Saenz, M.R. Rubinstein, Y.W. Han, A.W. Orr, M. Perretti,
Y. Huang, Y. Peng, G. Li, B. Wen, B. Dong, J.Y. Chen, Q.S. Geng, Z.W. Zhang, D.N. Granger, F.N.E. Gavins, Novel role for the AnxA1-Fpr2/ALX signaling axis as a
H. Yang, J. Wang, J. Wang, X. Zhang, L. Madsen, S. Brix, G. Ning, X. Xu, X. Liu, key regulator of platelet function to promote resolution of inflammation,
Y. Hou, H. Jia, K. He, K. Kristiansen, The gut microbiome in atherosclerotic car- Circulation 140 (4) (2019) 319–335.
diovascular disease, Nat. Commun. 8 (1) (2017) 845. [48] E. Pessolano, R. Belvedere, V. Bizzarro, P. Franco, I. Marco, F. Petrella, A. Porta,
[25] W. Xiong, X. Zhao, L. Villacorta, O. Rom, M.T. Garcia-Barrio, Y. Guo, Y. Fan, T. Zhu, A. Tosco, L. Parente, M. Perretti, A. Petrella, Annexin A1 contained in extracellular
J. Zhang, R. Zeng, Y.E. Chen, Z. Jiang, L. Chang, Brown adipocyte-specific vesicles promotes the activation of keratinocytes by mesoglycan effects: an auto-
PPARgamma (Peroxisome Proliferator-Activated Receptor gamma) deletion impairs crine loop through FPRs, Cells 8 (7) (2019).
perivascular adipose tissue development and enhances atherosclerosis in mice, [49] S.L. Tang, W.J. Chen, K. Yin, G.J. Zhao, Z.C. Mo, Y.C. Lv, X.P. Ouyang, X.H. Yu,
Arterioscler. Thromb. Vasc. Biol. 38 (8) (2018) 1738–1747. H.J. Kuang, Z.S. Jiang, Y.C. Fu, C.K. Tang, PAPP-A negatively regulates ABCA1,
[26] H. Nazih, F. Nazih-Sanderson, M. Krempf, J. Michel Huvelin, S. Mercier, J. Marie ABCG1 and SR-B1 expression by inhibiting LXRalpha through the IGF-I-mediated
Bard, Butyrate stimulates ApoA-IV-containing lipoprotein secretion in differ- signaling pathway, Atherosclerosis 222 (2) (2012) 344–354.
entiated Caco-2 cells: role in cholesterol efflux, J. Cell. Biochem. 83 (2) (2001) [50] N.L. Price, N. Rotllan, X. Zhang, A. Canfran-Duque, T. Nottoli, Y. Suarez,
230–238. C. Fernandez-Hernando, Specific disruption of Abca1 targeting largely mimics the
[27] Y. Du, X. Li, C. Su, M. Xi, X. Zhang, Z. Jiang, L. Wang, B. Hong, Butyrate protects effects of miR-33 knockout on macrophage cholesterol efflux and atherosclerotic
against high-fat diet-induced atherosclerosis via up-regulating ABCA1 expression in plaque development, Circ. Res. 124 (6) (2019) 874–880.
apolipoprotein E-deficiency mice, Br. J. Pharmacol. (2019). [51] R. Kasiappan, I. Jutooru, K. Mohankumar, K. Karki, A. Lacey, S. Safe, Reactive
[28] H. Wang, H. Cai, X. Wang, M. Zhang, B. Liu, Z. Chen, T. Yang, J. Fang, Y. Zhang, Oxygen Species (ROS)-inducing triterpenoid inhibits rhabdomyosarcoma cell and
W. Liu, J. Han, Q. Guo, H. Zhang, H. Wang, G. Xia, C. Wang, HDAC3 maintains tumor growth through targeting Sp transcription factors, Mol. Cancer Res. 17 (3)
oocyte meiosis arrest by repressing amphiregulin expression before the LH surge, (2019) 794–805.

240
W. Chen, et al. Clinica Chimica Acta 507 (2020) 236–241

[52] X. Fu, Z. Liu, C. Zhu, H. Mou, Q. Kong, Nondigestible carbohydrates, butyrate, and Roseburia intestinalis and diet modulate atherogenesis in a murine model, Nat.
butyrate-producing bacteria, Crit. Rev. Food Sci. Nutr. 59 (sup1) (2019) Microbiol. 3 (12) (2018) 1461–1471.
S130–S152. [61] C. Depommier, A. Everard, C. Druart, H. Plovier, M. Van Hul, S. Vieira-Silva,
[53] A. Bedford, J. Gong, Implications of butyrate and its derivatives for gut health and G. Falony, J. Raes, D. Maiter, N.M. Delzenne, M. de Barsy, A. Loumaye,
animal production, Anim. Nutr. 4 (2) (2018) 151–159. M.P. Hermans, J.P. Thissen, W.M. de Vos, P.D. Cani, Supplementation with
[54] J.T. Chai, J.E. Digby, R.P. Choudhury, GPR109A and vascular inflammation, Curr. Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-
Atheroscler. Rep. 15 (5) (2013) 325. concept exploratory study, Nat. Med. 25 (7) (2019) 1096–1103.
[55] I. Gaidarov, X. Chen, T. Anthony, D. Maciejewski-Lenoir, C. Liaw, D.J. Unett, [62] J. Li, S. Lin, P.M. Vanhoutte, C.W. Woo, A. Xu, Akkermansia Muciniphila protects
Differential tissue and ligand-dependent signaling of GPR109A receptor: implica- against atherosclerosis by preventing metabolic endotoxemia-induced inflamma-
tions for anti-atherosclerotic therapeutic potential, Cell. Signal. 25 (10) (2013) tion in Apoe-/- mice, Circulation 133 (24) (2016) 2434–2446.
2003–2016. [63] S. Lukovac, C. Belzer, L. Pellis, B.J. Keijser, W.M. de Vos, R.C. Montijn, G. Roeselers,
[56] G. Tumurkhuu, J. Dagvadorj, R.A. Porritt, T.R. Crother, K. Shimada, E.J. Tarling, Differential modulation by Akkermansia muciniphila and Faecalibacterium praus-
E. Erbay, M. Arditi, S. Chen, Chlamydia pneumoniae Hijacks a Host autoregulatory nitzii of host peripheral lipid metabolism and histone acetylation in mouse gut
IL-1beta loop to drive foam cell formation and accelerate atherosclerosis, Cell organoids, mBio 5 (4) (2014).
Metab. 28 (3) (2018) 432-448 e4. [64] S. Verhoog, P.E. Taneri, Z.M. Roa Diaz, P. Marques-Vidal, J.P. Troup, L. Bally,
[57] M.X. Byndloss, E.E. Olsan, F. Rivera-Chavez, C.R. Tiffany, S.A. Cevallos, O.H. Franco, M. Glisic, T. Muka, Dietary factors and modulation of bacteria strains
K.L. Lokken, T.P. Torres, A.J. Byndloss, F. Faber, Y. Gao, Y. Litvak, C.A. Lopez, of Akkermansia muciniphila and Faecalibacterium prausnitzii: A systematic review,
G. Xu, E. Napoli, C. Giulivi, R.M. Tsolis, A. Revzin, C.B. Lebrilla, A.J. Baumler, Nutrients 11 (7) (2019).
Microbiota-activated PPAR-gamma signaling inhibits dysbiotic Enterobacteriaceae [65] T.J. Khan, Y.M. Ahmed, M.A. Zamzami, A.M. Siddiqui, I. Khan, O.A.S. Baothman,
expansion, Science 357 (6351) (2017) 570–575. M.G. Mehanna, A. Kuerban, M. Kaleemuddin, M. Yasir, Atorvastatin treatment
[58] P. Louis, H.J. Flint, Diversity, metabolism and microbial ecology of butyrate-pro- modulates the gut microbiota of the hypercholesterolemic patients, OMICS 22 (2)
ducing bacteria from the human large intestine, FEMS Microbiol. Lett. 294 (1) (2018) 154–163.
(2009) 1–8. [66] L. Jia, D. Li, N. Feng, M. Shamoon, Z. Sun, L. Ding, H. Zhang, W. Chen, J. Sun,
[59] P. Louis, P. Young, G. Holtrop, H.J. Flint, Diversity of human colonic butyrate- Y.Q. Chen, Anti-diabetic effects of Clostridium butyricum CGMCC0313.1 through
producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase promoting the growth of gut butyrate-producing bacteria in Type 2 diabetic mice,
gene, Environ. Microbiol. 12 (2) (2010) 304–314. Sci. Rep. 7 (1) (2017) 7046.
[60] K. Kasahara, K.A. Krautkramer, E. Org, K.A. Romano, R.L. Kerby, E.I. Vivas, [67] H. Weng, K. Endo, J. Li, N. Kito, N. Iwai, Induction of peroxisomes by butyrate-
M. Mehrabian, J.M. Denu, F. Backhed, A.J. Lusis, F.E. Rey, Interactions between producing probiotics, PLoS ONE 10 (2) (2015) e0117851.

241

You might also like