An Insight Into Multifunctional Tool Box .99681 Compressed

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;

RMM-D-21-00103

An insight into multifunctional tool box: Akkermansia


muciniphila provides dynamic benefits to the
human gut
Downloaded from http://journals.lww.com/revmedmicrobiol by V0M3eMr08MG7uRhx2HJZ0/FHjJYEWcIOKA7zR1AWeDHfGf5yKxZxvFbgPdPAymQYqUVf28aAof2OieOrNJ7Ke767CO7ANLoyv4VWSvJDvYhuKWNPjJce2LUKpF3/pbTsgTdCO2e1Dk6zC93TzBle4MOqzXzeJXqvZ1RpoAfi80QR2ZzcD30kEzfPUhZjd/d/zkAPKn/SDBQ= on 02/01/2022

Kartikeya Tiwari

Akkermansia muciniphila is a mucin degrader that plays a major role in the human gut.
Multiple studies have shown that the level of abundance of this organism in the human
body is inversely related to diseases, such as diabetes, obesity and inflammatory bowel
disease. This organism has abilities to regulate the intestinal permeability, gut barrier
and inflammatory responses in various metabolic disorders by activating various
transcription factors and enhance the expression of receptors (TLRs, NLRs). Pangenome
of A. muciniphila provides comprehensive detail and role of encoding genes in
mitigation of metabolic dysbiosis. The present review discusses and summarizes the
role of A. muciniphila in diabetes mellitus type 2, obesity, nonalcoholic fatty liver
disease and other metabolic disorders.
Copyright ß 2022 Wolters Kluwer Health, Inc. All rights reserved.

Reviews and Research in Medical Microbiology 2022, 33:000–000

Keywords: Akkermansia muciniphila, gut barrier, intestinal epithelium,


probiotics, toll-like-receptors

Introduction processes [4]. This organism belongs to phylum Verruco-


microbia (Fig. 1), and is discovered in the human gut [2]. It
Our gastrointestinal tract hosts an environment for the is known as mucin-degrading bacterium as it uses solely a
various kinds of microorganisms, which is involved in source of carbon and nitrogen elements [2]. This organism
many metabolic, nutritional, physiological and immuno- helps in maintaining a healthy protective barrier by
logical functions. In the past decade, it has been evidently degrading the external mucus layer, which leads to
proved that gut microbiota has an important role in continuous renovation of mucosa and prevents entrance
determining the well being of our health. One of the of enteropathogens [5]. Therefore, because of its unique
organisms in the colonic mucosa-associated microbiota is characteristics, it has paved more scientific research focused
Akkermansia muciniphila (AKM), which colonizes a on this organism’s roles in down-regulating metabolic
considerable part of the human population and make up disorders and strengthening the gut barrier (Fig. 2). AKM is
1–4% of the fecal microbiota of normal adults [1]. AKM is affected by nutrients in the mucus layer located nearby to
a Gram-negative, oval shaped, nonspore former, aero- the intestinal epithelia [6]. Thus, the abundance of AKM is
tolerant, anaerobic bacterium [2,3]. This organism was negatively related to several diseases [1]. The population of
discovered in 2004 serendipitously at Wageningen AKM is reduced in metabolic disorders, such as obesity and
University of Netherlands, when the research group was type 2 diabetes mellitus (T2DM) (Fig. 3).
searching for a new mucin-degrading microbe in human
faeces [2]. According to European Research Council, Recent developments on this bacterium compiled by
initially A. muciniphila was identified and isolated by Zhang et al. in 2021 [7] described the potential of AKM in
Professor Willem deVos from Wageningen University, and inflammatory bowel disease and summarized the research
thereafter in 2007, Professor Patrice Daniel Cani findings with specific key points: AKM shows anti-
highlighted the behavior of this bacterium in metabolic inflammatory potential in inflammatory bowel disease;

Department of Microbiology, International Medical School, Management and Science University, Shah Alam, Selangor, Malaysia.
Correspondence to Kartikeya Tiwari, Professor, Department of Microbiology, International Medical School, Management and
Science University, Shah Alam, Selangor 40100, Malaysia.
E-mail: drkartikeyaa2zmicrobiology@gmail.com
Received: 11 September 2021; revised: 21 October 2021; accepted: 2 December 2021

DOI:10.1097/MRM.0000000000000305

ISSN 2770-3150 Copyright Q 2022 Wolters Kluwer Health, Inc. All rights reserved. 1
Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

2 Reviews and Research in Medical Microbiology 2022, Vol 33 No 00

Fig. 1. Taxonomic description of Akkermansia muciniphila bacterium.

the colitogenicity of AKM is context-dependent. Another important mechanism of A. muciniphila in mucin


Similarly, Zou and Chen [8] in 2020 described the use degradation, is the secretion of sulfate [9] by using sulfatase
of AKM against multiple diseases in their review and enzyme [1]. This produced sulfate is utilized by sulfate-
discussed the possible ways of engineering the AKM reducing bacteria and converted into sulfide [10]. The
bacterium, so that the complete benefits can be toxicity of hydrogen sulphide is minimized by the action of
accomplished. A. muciniphila through this unique mechanism [11].

Symbiotic interaction of Akkermansia


muciniphila in the human gut Role of Akkermansia muciniphila in type 2
diabetes mellitus
A. muciniphila is a part of the gut, which hosts a complex
and diverse microbiota. AKM interactions with other gut A metagenome-wide association analysis of intestinal
bacteria has been explained in several studies. Mucin microbiota in patients with T2DM suggests intestinal
degradation by A. muciniphila produces various com- dysbiosis and altered gut microbiota composition [12].
pounds, such as 1, 2-propanediol, propionate and acetate. Multiple researches demonstrated that A. muciniphila
The acetate produced is further utilized by Anaerostipes balances energy homeostasis by affecting gut barriers, which
caccae, Eubacterium hallii and Faecalibacterium prausnitzii is important for metabolic endotoxemia and metabolic
bacteria. The combined effect of A. muciniphila and these disorders. These findings raise the potential in future medical
organisms showed enhanced butyrate production. In the fields for targeting gut microbiota in order to down-regulate
cascade reactions, the cofactor pseudo-vitamin B12 is metabolic diseases and benefit human health [13].
produced by E. hallii organisms. This cofactor pseudo-
vitamin B12 is utilized by A. muciniphila, plays a role in the T2DM is a very common endocrine and metabolic
conversion of succinate to propionate [9]. disease [14]. Multiple molecular mechanisms of AKM are

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

Akkermansia muciniphila provides benefits to the human gut Tiwari 3

Fig. 2. Akkermansia muciniphila and other probiotic bacteria interaction with intestinal epithelial barrier. Mucus secreted by
goblet cells, which protect the external intestinal layer from invading pathogens. The beneficial bacteria at the external surface of
the intestinal epithelial layer breaks down mucus and dietary fibers into SCFA, which provides energy to intestinal cells. MAMP of
probiotics recognized by PRRs, such as NLRs and TLRs and regulate IEC pathways, such as peroxisome proliferator-activated
receptor gamma (PPAR-g) and mitogen-activated protein kinases (MAPK) to initiate protection to intestinal epithelial layer. IEC,
intestinal epithelial cells; MAMP, microorganism-associated molecular pattern; NLRs, NOD-like receptors; PRRs, pattern
recognition receptors, SCFA, short-chain fatty acid; TLRs, toll-like receptors.

believed to contribute to down-regulating the metabolic muscle protects from ageing-related insulin resistance
diseases and T2DM. Some of the potential mechanisms [14]. Dagdeviren and his co-workers observed Lactobacil-
are modulating inflammation, disruption of dietary lus, Bacteroides and A. muciniphila have been found to
constituents, interruption of gut permeability, glucose suppress tumour necrosis factor (TNF) in order to inhibit
and lipid metabolism, insulin tolerance and overall energy inflammation [1,16].
homeostasis in the mammalian host [15].
The immune system plays a significant role in balancing
T2DM is associated with higher levels of pro-inflamma- between commensal and pathogenic bacteria. The
tory cytokines, chemokines and inflammatory proteins. immune system of the healthy body not only eradicates
The harmful bacterial colonization and dysbiosis leads to harmful bacteria but also helps in colonization of
metabolic endotoxemia and low-grade inflammatory beneficial bacteria. This homeostasis is established by
conditions. For example, induction of interleukin-10 by the activation of pattern recognition receptors (TLRs,
A. muciniphila plays a role in improvement of glucose NLRs). AKM induces the cytokine production through
metabolism as over-expression of this cytokine in the activation of Toll-like receptors 2 and 4, which will

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

4 Reviews and Research in Medical Microbiology 2022, Vol 33 No 00

permeability and enhances tight junction expression. This


expression is enhanced by AMPK activation in the gut
epithelium. The gut integrity is enhanced by Amuc_1100
modulation-based expression of occludin and tight
junction protein-1 [18]. The main factor behind this
modulation is up-regulated mRNA expression for the
synthesis of various types of tight junction of proteins [19].

AKM increases fatty acid oxidation, energy expenditure and


reduces fatty acid synthesis to cut down body fat mass. This
specific modulated pathway is to reduce obesity and T2DM
[18]. This organism increases the production of endocan-
nabinoid receptors agonists (Fig. 4), such as 2-arachido-
noylglycerol, 2-palmitoylglycerol, 2-oleoylglycerol to
enhance the anti-inflammatory effects, metabolic endotox-
emia and the stimulation of gut peptide (GLP-1 and GLP-
2), which helps in glucose homeostasis, respectively [20].

Latest study conducted by Depommier et al. [21] in 2019


found significant improvement in overweight and obese
human volunteers when supplemented with AKM for 3
months (Fig. 5). This study suggested the use of AKM in
pasteurized form instead of live form AKM to improve
the insulin sensitivity and significant fall in insulinemia.

Fig. 3. Akkermansia muciniphila deficiency in various con-


ditions, such as diabetes and obesity lead to intestinal dys-
Interaction of Akkermansia muciniphila
biosis, which results in increased gut permeability. with lipid metabolism in obesity and
Weakened gut barrier allows translocation of bacterial com- related diseases
ponents, such as LPS into circulation and promotes metabolic
endotoxemia. This inflammation results in further organ injury The relationship between abundance levels of A.
and increases complications, such as NAFLD, insulin resis- muciniphila is inversely proportional to the body weight
tance and hyperlipidemia. AKM breaks down nondigestible of animals and humans [22]. The research work conducted
diet components and degrades mucin in the intestinal mucous by Schneeberger and co-workers demonstrated that the
layer into energy source to cells. The metabolite (SCFA- modulated gene expression in white adipose tissue in high-
acetate, butyrate, propionate) and components of AmEVs, fat diet (HFD) induced obesity in mice [23]. This leads to
recognized by PRRs (TLRs, NLRs) activates various transcrip- profound changes in key genes implicated in inflammation,
tion factors (AMPK, FFARs) in regulating host metabolism and fatty acid oxidation and lipogenesis [23].
strengthen gut barrier, and modulate intestinal permeability
by enhancing gene expression of gut tight junction proteins. Higher abundance level of A. muciniphila was found in
AKM, Akkermansia muciniphila; AmEVs, A. muciniphila- fecal samples from healthy individuals compared with
derived extracellular vesicle; AMPK, adenosine monopho-
obesity and T2DM patients. However, diet intervention
sphate-activated protein kinase; FFAR, free fatty acid recep-
was strongly believed to progressively improve the
tors; LPS, lipopolysaccharides; NAFLD, nonalcoholic fatty
abundance level of AKM [23]. Previous studies reported
liver disease; NLRs, NOD-like receptors; PRRs, pattern rec-
ognition receptors; SCFA, short-chain fatty acids; TLRs, toll-
that A. muciniphila convert the dietary fiber into acetate,
like receptors. butyrate and propionate by degrading the mucin
protective layer in the colon as their carbon and energy
source. Furthermore, these short-chain fatty acid
regulate bacterial recognition, and they can also optimize (SCFAs) metabolites have been widely discussed
the host metabolism and improve intestinal barrier (Fig. 3) to have an important role in our gut system as
function (Table 1). it has beneficial impact on lipid and glucose metabolism as
well as gut permeability [22]. Li and co-workers found
Another feature of human T2DM is increased gut out in their research about an improvement in body
permeability, this translocates intestinal microbial products weight and fat mass in HFD mice after introducing
into the blood and causes metabolic endotoxemia [17]. live and heat-killed A. muciniphila. They also discovered
Extracellular vesicle-coated A. muciniphila modulates gut the suppression of intercellular adhesion 1 (ICAM-1)

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

Akkermansia muciniphila provides benefits to the human gut Tiwari 5

Table 1. Interaction of Akkermansia muciniphila within the gut of humans.

Receptors/co-receptors Role and action of Akkermansia muciniphila Study model Disorders/diseases References

PPARg SCFA lowers PPARg expression and this Male C57Bl/6J mice Obesity [11]
stimulates the UCP2-AMPK signaling
pathway. This shifts the lipid synthesis to
lipid oxidation and aids in lipid
dysregulation.
FFAR/GPR SCFA stimulates FFAR on L-cells of colon, Male C57BL6 mice Obesity and Diabetes [12]
releases GLP-1 and PYY. These hormones
aid in lipids dysregulation and glucose
metabolism.
Tight junction proteins Amuc 1100 increases mRNA expression of C57BL (Apoe-/-) mice - [13]
tight junction proteins (occludin, claudin 3).
Consequently, this improves gut barrier and
modulates intestinal permeability.
ICAM-1, MCP-1 and TNFa mRNA expressions of ICAM-1, MCP-1 and C57BL (Apoe-/-) mice Atherosclerosis [13]
TNFa are inhibited with the supplement of
AKM. This prevents adhesion of
macrophage onto endothelium and
subsequent transmigration into intima.
Gut TLR2/4 Amuc 1100 activates TLR2/4, which further Mice T2DM [14]
induces cytokine production. This
mechanism mediates immune homeostasis
in intestinal mucosa and improves gut
barrier.
Hepatic TLR4 Bacterial components (LPS) bind to hepatic Mice NAFLD [15,16]
TLR4 and activate inflammatory cascade.
AKM prevents inflammatory response by
suppressing SERBP gene and IL-6
expression.

AMPK, adenosine monophosphate-activated protein kinase; FFAR, free fatty acid receptor; GLP-1, glucagon like peptide 1; GPR, G-protein
receptor; ICAM, 1-intracellular adhesion molecule 1; IL-6, interleukin; LPS, lipopolysaccharides; MCP-1, monocyte chemoattractant protein 1;
NAFLD, nonalcoholic fatty liver disease; PPARg, peroxisome proliferator-activated receptor gamma; PYY, peptide tyrosine tyrosine; T2DM, type 2
diabetes mellitus; TLRs, toll-like receptors; TNFa, tumor necrosis factor alpha; UCP2, uncoupling protein 2.

(Table 1), monocyte chemoattractant protein-1 (MCP-1) obesity, diabetes, liver diseases and ulcerative colitis.
and TNF-a [11,13]. There are various causes for hepatic disorders, such as
fatty liver disease, chronic alcohol intake and viral
The metabolites (SCFAs) produced in the colon can infections [25]. Prolong HFD intake induces dysbiosis,
stimulate free fatty acid receptor (FFAR 2/3) located on which leads to invasion of bacterial products from gut into
enteroendocrine L-cells in the colon (Fig. 4), which will systemic circulation and transported to the liver [30]. The
release the anorexigenic gut hormones, for example, bacterial products stimulate lipogenesis and inflammatory
glucagon-like peptide (GLP)-1 and peptide tyrosine response, which results in nonalcoholic fatty liver disease
tyrosine (PYY) [11,12]. Canfora and Blaak in 2017 (NAFLD) [26,27]. A study conducted in 2013, reported
reported that the acetate binds and activates the GPR- alteration in microbial population of gut with consump-
enriched cell and other ligand receptors that inhibit the tion of HFD, which further leads to hyperglycemia and
secretion of ghrelin. This results in less feeling of hunger fatty liver [33]. The study conducted by Spencer et al. in
[23,24]. Oral administration of acetate in HFD mice, 2013 reported that HFD is also linked to fatty liver disease
SCFA have been shown to decrease the expression of [28]. TLR activation is the frontline in host immune
peroxisome proliferator-activated receptor gamma defense as it stimulates production of pro-inflammatory
(PPARg) in intestinal epithelial organoids and this leads cytokines and chemokines against invading pathogens.
to increased expression of mitochondrial uncoupling However, prolong TLR stimulation may cause harm to
protein 2 and increased AMP-to-ATP ratio via AMPK the host [29,30]. TLRs are among the PRRs, which
signaling pathway. Consequently, this provides a route to recognize bacterial and viral components [31]. This
an increase in energy expenditure, a change from lipid higher expression of TLRs is mainly associated with
synthesis to lipid oxidation [11]. NAFLD [15,32]. TLR4 is the well known receptor for
LPS (cell wall component of Gram-negative) bacteria.
Liver has high tolerance to TLR activation and signaling
Roles of Akkermansia muciniphila in [39]. This is because symbiont microbiota is able to
nonalcoholic fatty liver disease inhibit this inflammatory response by interfering in TLR-
dependent nuclear factor kappa B (NF-kB) transcription
Multiple studies had been done regarding intestinal [33,34]. TLR signaling is suppressed in the normal liver.
dysbiosis relationships with various diseases, such as However, pathogenic condition of liver lowers liver

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

6 Reviews and Research in Medical Microbiology 2022, Vol 33 No 00

Fig. 4. The Akkermansia muciniphila degradation of mucin and indigestible diet components in short-chain fatty acids activates
various metabolic pathways. This organism is determined to interact directly with several lipid metabolic substances, including
changing the endotoxin level, short-chain fatty acid production and increased fatty acid oxidation. The molecular mechanism of
AKM on host metabolism is represented. AKM, Akkermansia muciniphila; SCFA, short chain fatty acids.

tolerance because of disruption of gut microbiota Role of Akkermansia muciniphila in future


composition, increases TLR expressions (Table 1) [35]. with medical and dietary intervention
Therefore, we can conclude that dysbiosis induce
metabolic and inflammatory responses play a major role Probiotics are live beneficial bacteria that are naturally
in pathogenesis of NAFLD. produced by the fermentation process in foods like
yogurt, kimchi, and others. The most common probiotic
Gut microbes and TLRs are potential targets in the found in yogurt and fermented products is Lactobacillus
treatment of NAFLD. Kim and co-workers in 2020 followed by Bifidobacterium. Prebiotic fibers are the
investigated that AKM-treated mice have more beneficial nondigestible part of foods like bananas, garlic, onion,
gut microbiota compared with non-AKM-treated mice. apples skin, beans, asparagus and many others that enter
AKM improves gut diversity and gut homeostasis [5,36]. the small intestine in an undigested form and are only
AKM treatment reduces invasion and translocation of fermented in the large colon. This fermentation process
pathogenic bacteria by increasing gut microbiota diversity feeds beneficial bacteria in order to increase its number in
and strengthening gut barriers. This mechanism reduces our gut for better health and to reduce disease risk.
bacterial components in circulation and prevents activa-
tion of hepatic TLRs. This suppresses possible pro- AKM can be used as a probiotic (Fig. 2) when consumed
inflammatory component production, down-regulates in various forms. The abundance of A. muciniphila in the
lipogenesis and further prevents the development of human gut can be promoted by dietary intervention and
NAFLD [16]. beneficial interaction with drugs. Many research has been

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

Akkermansia muciniphila provides benefits to the human gut Tiwari 7

AKM, pasteurized AKM (70 0C, 30 min) exerts more


effects on HFD-induced obesity, glucose tolerance and
insulin resistance. Treatment with the pasteurized AKM
aids in reducing adipocyte diameter and enhances the
number of intestinal goblet cells [51,52].

Apart from dietary intervention, some drugs have been


proven to promote the abundance of AKM in the human
gut. For instance, discoveries have shown that metformin
significantly increases the population of AKM up to 20%
of the bacterial population, which was correlated with its
ability to improve glucose tolerance [53,54]. AKM
produces SCFA, such as acetic acid from mucin and
numerously supplies vitality to goblet cells that produce
mucin, which contributes to an anti-inflammatory
impact and antidiabetic activity. Thus, AKM-positive
interaction with metformin has resulted in enhanced
glucose tolerance [55,56]. Therefore, this organism has
shown tremendous potential to be used as a supplement in
the treatment of metabolic diseases [57,58].
Fig. 5. Summarized flow sheet of Akkermansia muciniphila
supplements in overweight and obese human volunteers Becken et al. [59] in 2021 categorized all strains of AKM
[21]. (Fig. 6) and divided into four phylogroups by 16S rRNA
sequencing to get complete details and gene expression.
The pangenome of AKM strains contain 4982 gene
done to prove the increase of A. muciniphila abundance clusters (Fig. 7). The significant findings of their work
level in association with calorie restriction and consump- were interpreted pointwise and mentioned that the strains
tion of dietary products containing polyphenol (cranber- of all members of AKM-II and AKM-IV phylogroups.
ries extract, black tea or red wine grape) and
fructooligosaccharides (FOS) (soybeans, asparagus, shal- (1) Contains distinct capsule and exopolysaccharide genes.
lots and red onions). Dietary polyphenol are natural (2) Contains putative capsular polysaccharide (CPS)
antioxidants that elevate A. muciniphila level by protecting biosynthesis protein EPsC and Epsl.
against attacks of free oxygen radicals [37,38]. (3) Contains CMP-N-acetyl neuraminic acid synthetase.
(4) Contains capsule-modifying enzyme.
Roopchand and co-workers found in their research study (5) Contains polysaccharide pyruvyl transferase WcaK.
that dietary supplementation of cranberry extract and
grape polyphenol increased A. muciniphila abundance in
HFD mice faeces [39,40]. Everard and co-workers found
out that significantly increased abundance A. muciniphila
upon FOS administration for 5 weeks in ob/ob mice
[41–43]. Dao and co-workers reported in their research
that an increase of A. muciniphila was observed in the
lower abundance group after 12 weeks of calorie
restriction period along with an improvement in total
and low-density lipoprotein (LDL) cholesterol levels as
well as waist circumference [44,45].

The unique characteristics and promising outcome by


dietary intervention makes this organism more profound
as an upcoming supplement in the medical field. Earlier
this organism was thought to be pathogenic because of its
process of adhesion and degradation of intestinal mucus
layer [46,47]. However, this organism restricts itself only
up to the outer mucosal layer preventing it from reaching
the inner layer during mucin degradation [48]. Therefore,
this organism is not pathogenic, in fact it prevents
colonization of pathogens because of its strong attach- Fig. 6. Sample collection, factors and results of pediatric
ment to the mucus layer [49,50]. In comparison with live obesity microbiome and metabolome study [50].

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

8 Reviews and Research in Medical Microbiology 2022, Vol 33 No 00

Conclusion
A. muciniphila is a bacterium that does wonders in the
human body through its abilities to modulate the
intestinal permeability, gut barrier and inflammatory
response. This organism can benefit patients with
diabetes, obesity and NAFLD. In a nutshell, this organism
can be a key role player to fight against diabetes and
obesity. However, further studies should be done to
determine the effects of A. muciniphila in vivo as well as its
safety in humans, probably in upcoming future encoding
genes of AKM could be tailored by latest technologies,
such as CRISPR/cas system to obtain target-based
outcome in patients with metabolic disorders, T2DM
and obesity.

Acknowledgements
Fig. 7. Akkermansia muciniphila pangenome, phylogroups Conflicts of interest
categories, gene clusters and significant findings in latest There are no conflicts of interest.
study [50].

(6) Contains Cps locus and two conserved additional loci


then AKM-I. References
(7) Phylogroup AKM-IV codes DltB, an enzyme typically
involved in modification of lipoteichoic acid in Gram- 1. Ottman N, Reunanen J, Meijerink M, Pietila TE, Kainulainen V,
positive bacteria and also modify lipopolysaccharides in Klievink J, et al. Pili-like proteins of Akkermansia muciniphila
modulate host immune responses and gut barrier function.
some Gram-negative bacteria. PLoS One 2017; 12:e0173004.
(8) AKM-I contains additional chemotaxis genes, cyto- 2. Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia
municiphila gen. nov., sp. nov., a human intestinal mucin-
chrome C biosynthetic genes and code for quality degrading bacterium. Int J Syst Evol Microbiol 2004;
control sensor protein for outer membrane 54:1469–1476.
biogenesis NlpE. 3. Macchione IG, Lopetuso LR, Ianiro G, Napol M, Gibiino G,
Rizzatt G, et al. Akkermansia muciniphila: key player in meta-
(9) All phylogroups contains FeOAB genes, AKM-I and bolic and gastrointestinal disorders. Eur Rev Med Pharmacol Sci
AKM-II encoded additional mechanisms, such as 2019; 23:8075–8083.
AKM-I contains multiple enterochelin transporter 4. Akkermansia, a friendly bacterium who cares. (2020) ERC:
European Research Council. Available at: https://erc.euro-
gene groups, use siderophores to scavenge ferric iron. pa.eu/projects-figures/stories/akkermansia-friendly-bacterium-
AKM-II contains distinct gene groups for ferric iron who-cares. [Accessed 13 July 2021]
transporters than AKM-I. AKM-IV phylogroup lacks 5. Lukovac S, Belzer C, Pellis FG. Differential modulation by
Akkermansia muciniphila and Faecalibacterium prausnitzii of
in canonical mechanisms for ferric iron acquisition. host peripheral lipid metabolism and histone acetylation in
(10) AKM-I and AKM-II phylogroups contains LexA mouse gut organoids. M Bio 2014; 5:1–10.
repressor, indicates an SOS system for differences in 6. Belkaid Y, Hand TW. Role of the microbiota in immunity and
inflammation. Cell 2014; 157:121–141.
oxygen sensitivity. 7. Zhang T, Ji X, Lu G, Zhang F. The potential of Akkermansia
(11) All phylogroups encode 27 glycoside hydrolase muciniphila in inflammatory bowel disease. Appl Microbiol
enzyme families but varied in abundance of GH Biotechnol 2021; 105 (14–15):5785–5794.
8. Zou Y, Chen T. Engineered Akkermansia muciniphila: a
families. AKM-IV phylogroup does not contain GH promising agent against diseases. Exp Ther Med 2020;
97 enzymes, which constitute glycoamylases, such as 20:285.
9. Belzer C, Chia LW, Aalvink S, Chamlagain B, Piironen V, Knol J,
Bacteroides thetaiotaomicron Sus B. AKM-IV phy- et al. Microbial metabolic networks at the mucus layer led to
logroup also had fewer GH 110 enzymes, which is a diet-independent butyrate and vitamin B12 production by
galactosidases enzyme group capable of cleaving blood intestinal symbionts. M Bio 2017; 8:1–14.
10. Derrien M. (2007) Mucin utilisation and host interactions of
group B antigens. Moreover, strains of AKM-IV the novel intestinal microbe Akkermansia muciniphila. Vol.
phylogroup were enriched for GH 29 and GH 95 L- Dissertati Narcisinfo.
fucosidases, responsible for cleaving the terminal 11. Willis CL, Cummings JH, Neale G, Gibson GR. In vitro effects
of mucin fermentation on the growth of human colonic sul-
fucose residues that decorate mucin and human phate-reducing bacteria. Anaerobe 1996; 2:117–122.
milk oligosaccharides. 12. Ottman N, Geerlings SY, Aalvink S, de Vos WM, Belzer C.
(12) Phylogroups AKM-II and AKM-IV are deficient for Action and function of Akkermansia muciniphila in micro-
biome ecology, health and disease. Best Pract Res Clin Gastro-
reductive sulfur assimilation. enterol 2017; 31:637–642.

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

Akkermansia muciniphila provides benefits to the human gut Tiwari 9

13. Wang J, Qin J, Li Y, Cai Z, Li S, Zhu J, et al. A metagenome-wide 35. Dhillon N, Walsh L, Krüger B, Ward SC, Godbold JH, Radwan
association study of gut microbiota in type 2 diabetes. Nature M, et al. A single nucleotide polymorphism of Toll-like receptor
2012; 490:55–60. 4 identifies the risk of developing graft failure after liver
14. Cani PD, Possemiers S, Van De Wiele T, Guiot Y, Everard A, transplantation. J Hepatol 2010; 53:67–72.
Rottier O, et al. Changes in gut microbiota control inflam- 36. Miyake Y, Yamamoto K. Role of gut microbiota in liver dis-
mation in obese mice through a mechanism involving GLP-2- eases. Hepatol Res 2013; 43:139–146.
driven improvement of gut permeability. Gut 2009; 37. Zhang G, Ghosh S. Toll-like receptor-mediated NF-kB activa-
58:1091–1103. tion: a phylogenetically conserved paradigm in innate immu-
15. Kim S, Lee Y, Kim Y, Seo Y, Lee H, Ha J, et al. Akkermansia nity. J Clin Invest 2001; 107:13–19.
muciniphila prevents fatty liver disease, decreases serum tri- 38. Besten DG, Bleeker A, Gerding A, van Eunen K, Havinga R, van
glycerides, and maintains gut homeostasis. Appl Environ Micro- Dijk TH, et al. Short chain fatty acids protect against high-fat
biol 2020; 86:e03004-19. diet-induced obesity via a PPARg- dependent switch from
16. Hossain P, Kawar B, El Nahas M. Obesity and diabetes in the lipogenesis to fat oxidation. Diabetes 2015; 64:2398–2408.
developing world - a growing challenge. N Engl J Med 2007; 39. Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S,
356:213–215. Cartenı̀ M, et al. Gut-liver axis: the impact of gut microbiota on
17. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, nonalcoholic fatty liver disease. Nutr Metab Cardiovasc Dis
Gordon JI. An obesity-associated gut microbiome with 2012; 22:471–476.
increased capacity for energy harvest. Nature 2006; 40. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA,
444:1027–1031. Hanyaloglu AC, et al. The short chain fatty acid propionate
18. Dagdeviren S, Jung DY, Friedline RH, Noh HL, Kim JH, Patel PR, stimulates GLP-1 and PYY secretion via free fatty acid receptor
et al. IL-10 prevents aging-associated inflammation and insulin 2 in rodents. Int J Obest 2014; 39:424–429.
resistance in skeletal muscle. FASEB J 2017; 31:701–710. 41. Derrien M, Van Baarlen P, Hooiveld G, Norin E, Müller M, de
19. Cani PD, Bibiloni R, Knauf C, Neyrinck AM, Delzenne NM. Vos WM. Modulation of mucosal immune response, tolerance,
Changes in gut microbiota control metabolic diet–induced and proliferation in mice colonized by the mucin-degrader
obesity and diabetes in mice. Diabetes 2008; 57:1470–1481. Akkermansia muciniphila. Front Microbiol 2011; 2:166.
20. Chelakkot C, Choi Y, Kim DK, Park HT, Ghim J, Kwon Y, et al. 42. Zhou K. Strategies to promote abundance of Akkermansia
Akkermansia muciniphila-derived extracellular vesicles influ- muciniphila, an emerging-probiotics in the gut, evidence from
ence gut permeability through the regulation of tight junctions. dietary intervention studies. J Funct Foods 2017; 33:194–201.
Exp Mol Med 2018; 50:e450. 43. Roopchand D, Carmody R, Kuhn P, Moskal K, Rojas-Silva P,
21. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Turnbaugh P, Raskin I. Dietary polyphenols promote growth of
Vieira-Silva S, et al. Supplementation with Akkermansia mu- the gut bacterium Akkermansia muciniphila and attenuate high fat
ciniphila in overweight and obese human volunteers: a proof- diet-induced metabolic syndrome. Diabetes 2015; 64:2847–2858.
of-concept exploratory study. Nat Med 2019; 25:1096–1103. 44. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG,
22. Carpino G, Del Ben M, Pastori D, Carnevale R, Baratta F, Overi D, Neyrinck AM, et al. Responses of gut microbiota and glucose
et al. Increased liver localization of lipopolysaccharides in hu- and lipid metabolism to prebiotics in genetic obese and diet-
man and experimental NAFLD. Hepatology 2020; 72:470–485. induced leptin-resistant mice. Diabetes 2011; 60:2775–2786.
23. Choi Y, Kwon Y, Kim DK, Jeon J, Jang SC, Wang T, et al. 45. Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia
Gut microbe-derived extracellular vesicles induce insulin re- muciniphila protects against atherosclerosis by preventing
sistance, thereby impairing glucose metabolism in skeletal metabolic endotoxemia-induced inflammation in ApoeS/S
muscle. Sci Rep 2015; 5:15878. Mice. Circulation 2016; 133:2434–2446.
24. Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, 46. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels
Delzenne NM, Cani PD. The endocannabinoid system links gut LB, et al. Cross-talk between Akkermansia muciniphila and
microbiota to adipogenesis. Mol Syst Biol 2010; 6:392. intestinal epithelium controls diet-induced obesity. Proc Natl
25. Xu Y, Wang N, Tan HY, Li S, Zhang C, Feng Y. Function of Acad Sci USA 2013; 110:9066–9071.
Akkermansia muciniphila in obesity: interactions with lipid 47. Plovier H, Everard A, Druart C, Depommier C, Van Hul M,
metabolism, immune response and gut systems. Front Microbiol Geurts L, et al. A purified membrane protein from Akkermansia
2020; 11:219. muciniphila or the pasteurized bacterium improves metabo-
26. Canfora EE, Blaak EE. Acetate: a diet-derived key metabolite in lism in obese and diabetic mice. Nat Med 2017; 23:107–113.
energy metabolism: good or bad in context of obesity and 48. Reid DT, Eller LK, Nettleton JE, Reimer RA. Postnatal prebiotic
glucose homeostasis? Curr Opin Clin Nutr Metab Care 2017; fibre intake mitigates some detrimental metabolic outcomes of
20:477–483. early overnutrition in rats. Eur J Nutr 2016; 55:2399–2409.
27. Wu W, Lv L, Shi D, Ye J, Fang D, Guo F, et al. Protective effect 49. Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E,
of Akkermansia muciniphila against immune-mediated liver Verger EO, et al. Akkermansia muciniphila and improved
injury in a mouse model. Front Microbiol 2017; 8:1–13. metabolic health during a dietary intervention in obesity:
28. Dai X, Wang B. Role of gut barrier function in the pathogenesis Relationship with gut microbiome richness and ecology. Gut
of nonalcoholic fatty liver disease. Gastroenterol Res Pract 2016; 65:426–436.
2015; 2015:287348. 50. Schneeberger M, Everard A, G omez-Valades AG, Matamoros S,
29. Aron-Wisnewsky J, Gaborit B, Dutour A, et al. Gut microbiota Ramı́rez S, Delzenne NM, et al. Akkermansia muciniphila
and nonalcoholic fatty liver disease. Eur Soc Clin Microbiol inversely correlates with the onset of inflammation, altered
Infect Dis 2013; 25:338–348. adipose tissue metabolism and metabolic disorders during
30. Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, obesity in mice. Sci Rep 2015; 5:16643.
et al. Intestinal microbiota determines development of non- 51. Derrien M, van Passel MWJ, van de Bovenkamp JHB, Schipper
alcoholic fatty liver disease in mice. Gut 2013; 62:1787–1794. RG, de Vos WM, Dekker J. Mucin-bacterial interactions in the
31. Spencer MD, Hamp TJ, Reid RW, Fischer LM, Steven H, Fodor human oral cavity and digestive tract. Gut Microbes 2010;
AA. Association between composition of the human gastro- 1:254–268.
intestinal microbiome and development of fatty liver with 52. Bashiardes S, Shapiro H, Rozin S, Shibolet O, Elinav E. Nonalco-
choline deficiency. Gastroenterology 2012; 140:976–986. holic fatty liver and the gut microbiota. Mol Metab 2016; 5:782–
32. Inokuchi S, Tsukamoto H, Park E, Liu ZX, Brenner DA, Seki E. 794.
Toll-like receptor 4 mediates alcohol-induced steatohepatitis 53. G omez-Gallego C, Pohl S, Salminen S, De Vos WM, Kneifel W.
through bone marrow-derived and endogenous liver cells in Akkermansia muciniphila: a novel functional microbe with
mice. Bone 2014; 23:1509–1518. probiotic properties. Benef Microbes 2016; 7:571–584.
33. Kawai T, Akira S. The role of pattern-recognition receptors in 54. Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Mannerås-
innate immunity: Update on toll-like receptors. Nat Immunol Holm L, et al. Metformin alters the gut microbiome of indivi-
2010; 11:373–384. duals with treatment-naive type 2 diabetes, contributing to the
34. Rivera CA, Adegboyega P, van Roijen N, Tagallicud A, Allman therapeutic effects of the drug. Nat Med 2017; 23:850–858.
M, Wallace M. Toll-like receptor-4 signaling and Kupffer cells 55. Belzer C, De Vos WM. Microbes inside from diversity to
play pivotal roles in the pathogenesis of nonalcoholic steato- function: the case of Akkermansia muciniphila. ISME J 2012;
hepatitis. Bone 2012; 23:571–579. 6:1449–1458.

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
CE: Tripti; RMM-D-21-00103; Total nos of Pages: 10;
RMM-D-21-00103

10 Reviews and Research in Medical Microbiology 2022, Vol 33 No 00

56. Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, 58. Zhou JC, Zhang XW. Akkermansia muciniphila: a promising
Bae JW. An increase in the Akkermansia spp. population target for the therapy of metabolic syndrome and related
induced by metformin treatment improves glucose home- diseases. Chin J Nat Med 2019; 17:835–841.
ostasis in diet-induced obese mice. Gut 2014; 63:727– 59. Becken B, Davey L, Middleton DR, Mueller KD, Sharma A,
735. Holmes ZC, et al. Genotypic and phenotypic diversity among
57. Anhê FF, Marette A. A microbial protein that alleviates meta- human isolates of Akkermansia muciniphila. mBio 2021;
bolic syndrome. Nat Med 2017; 23:11–12. 12:e00478–e521.

Copyright © 2022 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

You might also like