You are on page 1of 9

European Review for Medical and Pharmacological Sciences 2019; 23: 8075-8083

Akkermansia muciniphila: key player in


metabolic and gastrointestinal disorders
I.G. MACCHIONE1,2, L.R. LOPETUSO1,2, G. IANIRO1,2, M. NAPOLI1,2,
G. GIBIINO1,2, G. RIZZATTI1,2, V. PETITO1, A. GASBARRINI1,2, F. SCALDAFERRI1,2

Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Rome, Italy
1

UOC Medicina Interna e Gastroenterologia, Area Medicina Interna, Gastroenterologia ed


2

Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e


Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy

Abstract. – OBJECTIVE: Gut microbiota has as the “gut microbiota”, has been estimated to
a key role in host metabolic regulation and im- exceed 1014 microorganisms, which encompasses
mune response, and its dysbiosis represents ~10 times more bacterial cells than the number
one of the main causes of gastrointestinal dis-
eases. In this scenario, Akkermansia muciniph-
of human cells and over 100 times the amount
ila is a crucial player in keeping the integrity of of genomic content (microbiome) as the human
the gastrointestinal tract. genome1,2.
MATERIALS AND METHODS: This review fo- Latest evidence supports the role of gut micro-
cuses on the correlation between gut microbio- biota in several host functions, and this dysbiotic
ta and intestinal homeostasis, primarily explor- condition has been linked to cardiovascular dis-
ing A. muciniphila and its involvement in the de-
velopment of metabolic disorders and gastroin- ease, neurodegenerative conditions, psychiatric
testinal diseases. disorders, and autoimmune conditions3-6.
RESULTS: Akkermansia muciniphila belongs to Early studies7,8 sought to identify the normal
the Verrucomicrobia phylum, and it colonizes the set of microbes that colonizes healthy people,
mucus layer in the gastrointestinal tract, repre- by culture and characterization of physiological
senting 1 to 4% of the fecal microbiota. It stimu- properties. The introduction of strictly anaerobic
lates mucosal microbial networks, and it improves
intestinal barrier function, providing crucial host techniques in the 1970s allowed the recovery of
immunological responses. Several studies have more than 300 bacterial species.
demonstrated the possible involvement of A. mu- Then, above all, the introduction of 16SrRNA
ciniphila in the development of intestinal and met- profiling studies provided invaluable insights into
abolic disorders. Indeed, adipose and glucose the taxonomic composition of the gut microbio-
metabolisms are influenced by A. muciniphila,
and its levels inversely correlate to inflammatory
ta, which in turn has facilitated the inference of
conditions, such as inflammatory bowel disease, broad evolutionary patterns. However, species
obesity, and diabetes. Conversely, its therapeutic diversity may only present the top of the iceberg
administration decreases their development. of microbial complexity in the gut9-12. Recent
CONCLUSIONS: A. muciniphila exerts a key studies applying methods that go beyond the typ-
role in the maintenance of intestinal health and ical 16SrRNA profiling approach have provided
in host metabolic modulation. Future studies
could open new horizons towards its potential compelling evidence that the bacterial species of
therapeutic applications in gastrointestinal and the gut microbiota are composed of a multitude of
extra-intestinal diseases. strains, which are likely to influence gut microbi-
Key Words: ota functions.
Akkermansia muciniphila, Gut microbiota, Gastro- Despite variability in species composition, the
intestinal diseases, Metabolic diseases, Obesity, IBD, mammalian gut microbiota is dominated by rela-
Colitis. tively few bacterial phyla, including Firmicutes,
Bacteroidetes, Proteobacteria, Actinobacteria,
Introduction and Verrucomicrobia. Each phylum is generally
classified in a hierarchical manner into sub-phy-
The collection of bacteria, Archaea and Eu- la, classes, orders, families, genera, and finally
karya colonising the GI tract, usually known species and strains. Some of the earliest efforts to

Corresponding Author: Franco Scaldaferri, MD; e-mail: franco.scaldaferri@policlinicogemelli.it 8075


I.G. Macchione, L.R. Lopetuso, G. Ianiro, M. Napoli, G. Gibiino, et al.

sequence 16SrRNA genes directly from samples survival when exposed to atmospheric oxygen, so
showed that 85-95% of bacterial abundance cor- that it can be defined as an aerotolerant anaerobic
responding to known species could be attributed bacterium.
to three bacterial groups related to Bacteroides, A. muciniphila encodes for 567 secreted pro-
Clostridium cluster XIVa, and Clostridium clus- teins, such as sugar hydrolase, sialidase, and
ter IV13,14. sulfatase, which are involved in mucin utilization.
Among these, Verrucomicrobia belongs to the It is thus considered a mucin-degrading bacteri-
Planctomycetes-Verrucomicrobia-Chlamydiae um31. In fact, intestinal mucins, the highly gly-
bacterial superphylum15. The first microbe of this cosylated proteins of the epithelial mucus layer,
phylum, Akkermansia muciniphila, was original- represent the main source of carbon and nitrogen
ly isolated from a fecal sample from a healthy for this species16 that can be found in high num-
female in a specific medium that contained pu- bers in mucosal biopsy specimens of the human
rified mucins, sole carbon source, suggesting colon32. Moreover, A. muciniphila has been found
specific metabolic properties located in the inter- to have a role not only in mucins biodegradation,
face between the luminal bacteria and the host16. but also in stimulating mucin production33,34. Be-
Since then, many functions were hypothesized to cause of its influence on mucin metabolism and
support the role of Verrucomicrobia in the mod- consequently on the thickness of the mucus layer,
ulation of metabolism in the human host. In this a crucial role of A. muciniphila in gut barrier
review, we will summarize the current evidence function is easy to be hypothesized. Indeed, it has
on Verrucomicrobia, gut pathophysiology, and been demonstrated that A. muciniphila is depleted
metabolic disorders. in patients affected by inflammatory bowel dis-
ease (IBD), in which a reduction in barrier func-
The Verrucomicrobia Phylum tions has been largely proved35,36. A. muciniphila
Verrucomicrobia are Gram-negative bacteria reduction has been shown in ulcerative colitis
belonging to the Planctomycetes, Verrucomi- (UC) and Crohn’s disease (CD), both in clinically
crobia, Chlamydiae (PVC) superphylum, that is active disease and during remission35,36. A study
a group of six bacterial phyla: Planctomycetes, of 2011 also demonstrated an inverse correlation
Verrucomicrobia, Chlamydiae, Lentisphaerae, between A. muciniphila levels and the severity of
Poribacteria, and OP317,18. The bacteria from acute appendicitis37.
this superphylum were previously suggested Moreover, Reunanen et al30 examined the ef-
to have a compartmentalized cell plan with a fects of A. muciniphila on in vitro colonic cells
cytoplasmic membrane as the outermost mem- lines Caco2 and HT-29, highlighting the capacity
brane, and an intracytoplasmic membrane con- of this bacteria to adhere to the epithelium and
taining a condensed nucleoid and ribosomes19. to strengthen the intestinal barrier. The same
However, more recently it has been suggested study showed that A. muciniphila can induce
that these bacteria have an outer and an inner a weak pro-inflammatory activity stimulating
membrane (IM) with possible invaginations enterocytes production of interleukin 8 (IL-8) at
of the IM inside the cytoplasm, representing a cell concentrations 100-fold higher than those of
variation and not an exception of the Gram-neg- E. coli. Thus, this bacterium does not activate a
ative cell plan 20. strong inflammatory cascade in the epithelium
Verrucomicrobia phylum is involved in but seems to be able to keep the mucosa-asso-
soil-based environments21, in aquatic environ- ciated immune system alerted at an appropriate
ments22,23 and, not secondarily, is associated with level. These results are in line with other in vivo
eukaryotic species24-27. studies that link A. muciniphila to a non-inflamed
Akkermansia muciniphila belongs to this phy- mucosa35,36 and suggest important host-bacteria
lum, a bacterium that shares only very little simi- interactions involved in the maintenance of host
larity with Verrucomicrobia genomes, indicating immune response.
how this phylum includes a vast bio-diversity of Moreover, A. muciniphila has a crucial role in
species28. metabolic homeostasis. Studies on animals38-40
A. muciniphila colonizes the mucus layer of the showed a lower abundance of A. muciniphila
human gastrointestinal (GI) tract (12) and consti- in mice fed with high-fat diet (HFD), obese
tutes 1 to 4% of the fecal microbiota29. It is con- mice, and with type 2 diabetes-like symptoms.
sidered to be an anaerobic bacterium, although These results were confirmed by clinical trials
a more recent study30 showed that it has a good that reported a negative correlation between A.

8076
Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders

muciniphila and dietary fat intake, serum total diet for one year, when compared to lean, nor-
cholesterol, and LDL 41. A possible role of A. mal-weight individuals at baseline. However, the
muciniphila in preventing metabolic disorders ratio returned to normal in those individuals who
was supposed by a study42 on HFD fed mice, had successful and sustained weight loss.
in which a daily administration of 108 CFU of Jumpertz et al52 reported differences in the
this strain for four weeks improved metabol- fecal microbiota in lean and obese individuals
ic profile, prevented weight gain, and reduced during diets, that varied in caloric content (2400
LPS endotoxemia related to metabolic disorders. kcal/day vs. 3400 kcal/day), showing that an al-
Overall, the effects of A. muciniphila on metab- tered nutrient loading induced rapid changes in
olism represent a field of great interest that may the gut bacterial community.
lead to possible future therapeutic applications A high-fat diet also has an important role in
in such disorders. the change of gut microbiota; indeed, Western
diet induces an important intestinal dysbiosis,
Gut Microbiota and Obesity especially the proliferation of gram-negative bac-
Obesity is a major risk factor for the develop- teria, such as Enterobacteriaceae. This condition
ment of common diseases, such as type 2 diabe- promotes local inflammation, and it increases
tes, metabolic syndrome, and cardiovascular dis- intestinal permeability53.
ease, which are leading causes of morbidity and Examination of gut microbiota has been ex-
mortality in developed nations with an estimated tended to children, but the results are heteroge-
cost exceeding hundreds of billion euro in the last neous. However, it is now quite definite that it is
years and an estimated 250,000 deaths per year43. involved in triggering an innate immune response
Both environmental and genetic factors con- in early life, resulting in the maturation of gut-as-
tribute to obesity and T2D development. Many sociated lymphoid tissue and developing adaptive
researches emphasize the role of the gut micro- immunity54.
biota as critical metabolism modifier and recent Some studies reported an increased level of
studies44-46 showed its role in the development Firmicutes and an increased Firmicutes/Bacte-
of obesity and progression to T2D. On one side, roidetes ratio in obese individuals55, while others
obesity determines a significant reduction of mi- showed increased levels of Bacteroidetes and an
crobial variety and T2D causes dysbiosis with a increased Bacteroidetes/Firmicutes ratio56.
reduction in butyrate-producing bacteria levels A report from Egypt further demonstrated dif-
on the other. In this connection, it was observed ferent results57, suggesting that the composition of
that Roseburia and Faecalibacterium prausnitzii gut microbiota may depend on the environment,
were less abundant in the feces of T2D patients particularly in children.
compared to controls47,48. Obese phenotype seems to be transmissible, as
Human gut microbiota exerts some key-func- shown further58. It was observed that in the mu-
tions, such as digestion of otherwise indigestible rine models, the germ-free mice remain signifi-
substrates, production of specific vitamins im- cantly leaner than the conventional raised mice,
possible to synthesize via human enzymatic re- despite a significantly higher food intake59.
actions, modification of host metabolism through Vice versa, colonization of GF mice with cae-
the production of certain metabolites as short- cum-derived microbiota of conventional mice
chain fatty acids (SCFA), or via translocation of (“conventionalization”) demonstrated a signifi-
endotoxins49. cant increase in total body fat content without
One of them, known as the gram-negative bac- changes in the caloric intake60. This finding was
terial wall compound lipopolysaccharide (LPS), later reproduced using inoculation in GF mice
is an innate immune regulator capable of acti- with feces of obese human subjects showing iden-
vating macrophages in both adipose tissue and tical effects61.
peripheral blood50. Some investigations confirm
that the translocation of specific intestinal bac- Akkermansia Muciniphila and Obesity:
teria plays an important role in metabolism and Evidence from Mouse Models
development of obesity and insulin resistance. Some of the previous studies62 have found a
Ley et al51 reported a higher ratio of Firmicutes/ link between caloric intake and the abundance of
Bacteroidetes and a lower number of Bacteroide- Akkermansia muciniphila.
tes in 12 obese adult men and women randomly Administration of prebiotics in high fat diet-fed
assigned to either a low-fat or low-carbohydrate mice abolished the metabolic endotoxemia that

8077
I.G. Macchione, L.R. Lopetuso, G. Ianiro, M. Napoli, G. Gibiino, et al.

characterizes the impaired metabolic state pres- evidence of inverse correlation between these
ent in obese subjects, reduced the total fat mass two variables66. Specifically, it was observed a
and the body weight. Of note, these results were reduction in numbers of Bifidobacterium and A.
significantly and inversely correlated with A. mu- muciniphila and an increase in numbers of Staph-
ciniphila abundance63. ylococcus, Enterobacteriaceae, and Escherichia
Schneeberger et al64 reported that the levels of Coli in overweight pregnant compared with nor-
A. muciniphila in mice are inversely associated mal-weight pregnant women.
with inflammatory markers, lipid synthesis, and Their gut microbiota composition was an-
several plasma markers of insulin resistance, alysed by quantitative real-time PCR. In the
cardiovascular risk, and adiposity. The greatest whole population, the increased total bacteria and
impact on body weight gain and adiposity in mice Staphylococcus numbers were correlated to in-
occurs after 6 consecutive weeks of HFD admin- creased plasma cholesterol levels, where instead
istration. the increased Bacteroides numbers were linked to
There was a significative inverse correlation increased HDL-cholesterol and folic acid levels,
between A. muciniphila levels and inflammatory and reduced TAG levels67.
markers, all circulating parameters (i.e., insulin, Karlsson et al68 evaluated the gut microbio-
glucose, triglycerides, and leptin) and positive ta of twenty overweight or obese children and
correlation with 118 out of 13 genes involved in twenty normal weight control children of 4-5
fatty oxidation, synthesis, and browning, while years of age. Interestingly, the levels of A. mu-
Bifidobacterium spp. abundance was significantly ciniphila were significantly reduced in obese/
and negatively correlated with 3 inflammatory overweight children, whereas the concentration
markers and leptin, and positively correlated with of the gram-negative Enterobacteriaceae family
only 2 genes involved in fatty oxidation. was significantly higher in the same group. The
Therefore, these data indicate that HFD affects concentration of Bifidobacterium was inversely
specific gut bacteria, and highlight that the abun- correlated to alanine aminotransferase (ALT) in
dance of A. muciniphila progressively declines obese/overweight children.
with prolonged dietary treatment in mice, and Since diabetes and overweight have been re-
that this effect is exacerbated upon HFD. It is lated to increased gut permeability and low-
also demonstrated that this bacterium declines grade inflammation, LPS-induced endotoxemia
before the onset of metabolic alterations, thereby has been suggested as one of the causative agents
suggesting a causative implication in the disease of metabolic disorders related to obesity. The in
progression64. vitro observation that A. muciniphila fortifies
Moreover, the gut concentration of A. muciniph- epithelial barrier function could provide a work-
ila is increased and decreased respectively in mice ing hypothesis to rationalize the in vivo findings
fed with fish oil-enriched diet and lard-enriched connecting decreased fecal A. muciniphila levels
diet, and this effect is associated with better control with diabetes and obesity. This could reveal one
of the gut barrier function and lower adipose tissue possible mechanism behind the protective effect
inflammation, a phenomenon that can be trans- of the bacterium against high-fat-diet-induced
ferred to germ-free recipient mice65. LPS endotoxemia in obese mice30.
Likewise, other authors33-42 demonstrated that At this time, no study relationship between
gut barrier dysfunction, the weight and fat gain in A. muciniphila and the markers of hypothalamic
mice fed with HFD can be reduced with simulta- control of food intake has been evaluated.
neous administration of A. muciniphila. In accordance with all this evidence, we can
Finally, there is also a link between age and suppose that in humans A. muciniphila influences
A. muciniphila in mice, since the intestinal levels the response to caloric restriction diet in terms of
of this bacterium are lower in older mice. HFD improvement of inflammatory markers, insulin
heavily influence adipose tissue profile and intes- resistance, and glycemia.
tinal microbiota in a way that mimicked aging64.
Akkermansia Muciniphila and
A. Muciniphila and Obesity: Evidence Intestinal Barrier Modulation
from Human Studies Mucus is an essential part of maintaining the
Emerging studies evaluated the relationship health of the gastrointestinal tract and its func-
between the gut abundance of Akkermansia mu- tional integrity. It represents both an efficient bar-
ciniphila and body weight in human. There is rier that protects the epithelial surface, preventing

8078
Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders

particles or extraneous agent penetration, and tory bowel diseases (mainly in ulcerative colitis)
an excellent lubricant for metabolic processes69. and metabolic disorders, which suggest that it
The mucus layer offers substrates for bacterial may have potential anti-inflammatory proper-
growth, adhesion, and protection. ties73. Specifically, recent trials demonstrated that
The resident microbiota also represents an es- A. muciniphila increased the endogenous pro-
sential resource to ensure an effective gut barrier; duction of specific bioactive lipids, especially
it provides to obtain nutrients, it processes neces- Glucagon-like peptide 2 (GLP-2), involved in gut
sary molecules for mucosal integrity, and it com- barrier regulation77.
petes with pathogens to gain space70. Conversely, Next research challenge will be to analyze
gut microbiota composition has been shown to the molecular mechanism of actions involved in
influence mucus barrier properties. Higher levels the metabolic and immunological A. muciniphila
of specific bacterial species are able in the inflam- properties in order to find potential therapeutic
matory conditions to increase the permeability applications.
of the mucus layer and thus decrease the barrier
function71. Therefore, its presence influences in- Akkermansia Muciniphila and
testinal colonization by attracting bacteria that Gastrointestinal Diseases
have the ability to survive and proliferate within The relation between mucin and bacteria var-
the mucus layer29. ies depending on the microbiota, and several
Akkermansia muciniphila is known to colonize studies16 have reported a potential involvement
the mucosal layer of the human intestine where of mucin-degrading bacteria in the pathogenesis
it triggers both hosts metabolic and immune re- of intestinal diseases. The levels of Akkermansia
sponses. It was found being particularly effective muciniphila have been shown to be inversely cor-
in increasing mucus thickness and increasing related with several disorders, such as inflamma-
gut barrier function. Its metabolic and mucolytic tory bowel diseases (IBD), obesity, diabetes, and
activity make A. muciniphila a key species in appendicitis, but few information exists about its
the mucus layer, stimulating beneficial mucosal immunological mechanisms of action76.
microbial networks. Specifically, the production In IBD, the abundance of A. muciniphila was
of short-chain fatty acids is beneficial to the host found to be decreased75. Indeed, several stud-
and microbiota members72. It was demonstrat- ies76 have demonstrated its depletion in the fecal
ed that A. muciniphila colonizes the intestinal microbiota of ulcerative colitis patients, and low
tract in early life and develops within a year to levels of A. muciniphila have also been found in
a level close to that observed in healthy adults29. biopsies of intestinal mucosa from IBD-patients
In the colon of a healthy human, A. muciniphila in comparison to healthy controls.
is present in high levels with an abundance of Also, individuals with acute appendicitis were
approximately 3%37. Its specialization in mucin found to have low levels of A. muciniphila, and
degradation makes it a key organism at the muco- the amount of A. muciniphila was inversely relat-
sal interface between the lumen and host cells73. ed to the severity of appendicitis37.
Thanks to A. muciniphila, intestinal mucin is Because of its ability to degrade mucins, it was
degraded into mainly propionic and acetic acid supposed that A. muciniphila could contribute
that become the substrate for F. prausnitzii, one to intestinal inflammation because low levels of
of the main producers of butyrate in the intestine. mucin allow the pathogen to access the mucosa.
This leads to the inhibition of inflammation in the To support this, trials with SIHUMI mice report-
gastrointestinal tract and to the prevention of the ed that the intestinal inflammation induced by
increased intestinal permeability74. Salmonella typhimurium was exacerbated by A.
A. muciniphila also has an active role in stimu- muciniphila, suggesting that this was based on its
lation and regulation of host immunity, improving ability to interfere with host mucus formation and
intestinal barrier function, and probably provid- production77.
ing competitive exclusion at the host-microbe However, several trials have demonstrated that
interface28,75. It is positively correlated with a lean A. muciniphila controls fat storage, adipose tissue
phenotype, reduced body weight gain, increate metabolism, and glucose homeostasis42. It is wide-
metabolic responses, and with the restoration of ly considered as a novel potential candidate to im-
gut barrier function by modulation of mucus layer prove metabolic disorders associated with obesity,
thickness76. Notably, low levels of A. muciniphila diabetes, liver diseases, and cardiometabolic dis-
have been observed in patients with inflamma- orders. Indeed, its administration has been shown

8079
I.G. Macchione, L.R. Lopetuso, G. Ianiro, M. Napoli, G. Gibiino, et al.

to reduce the development of such diseases78. ponent has a specific role in metabolic function,
This outline, therefore, the key role of A. mu- and immune modulation and dysbiosis condition
ciniphila in GI and extra GI diseases and how im- is related to the development of gastrointestinal
portant it is to keep investigating its mechanisms impairment. In this background, a crucial role is
in order to find many other potential therapeutic played by Akkermansia muciniphila, a mucin-de-
approaches. grading bacterium, which revealed important
metabolic and immunological properties. Several
Therapeutic Role of Akkermansia studies have demonstrated how it is centrally
Muciniphila for the Treatment of involved in controlling of fat storage and glucose
Metabolic Disorders homeostasis; its abundance is inversely related to
The examination of Akkermansia muciniphila inflammatory conditions, such as dyslipidaemia,
beneficial properties in metabolic regulation and obesity, inflammatory bowel disease (IBD), and
immune response led to better investigate its type 2 diabetes.
potential therapeutic effects in the treatment of In accordance with these observations, other
certain diseases and metabolic disorders. detailed studies would be needed to deepen A.
Notably, A. muciniphila seems to have an ac- muciniphila activity, in order to improve its use
tive role in amine butyrate production and propi- in the therapeutic application for metabolic and
onate extracellular pool output. It is also crucial gastrointestinal disorders.
in hydrogen sulfide production79, that may have
anti-inflammatory, muscular relaxing, and anti-
oxidant properties80.
Conflict of Interest
Prebiotic feeding strongly increases the pres- The Authors declare that they have no conflict of interests.
ence of A. muciniphila and it improves metabolic
disorders. All trials in which animals were treated
with A. muciniphila showed that it reduces body
weight and fat-mass gain, hepatic steatosis, in- References
flammation, cholesterol levels, and atherosclero-
sis; improves insulin sensitivity and restores gut 1) Neish AS. Microbes in gastrointestinal health and
barrier function by influencing different factors, disease. Gastroenterol 2009; 136: 65-80.
like mucus-layer thickness, tight-junction pro- 2) Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh
teins, antimicrobial peptides, and immunity81. PJ, Samuel BS, Gordon JI, Relman DA. Fraser -L igget
CM, Nelson KE. Metagenomic analysis of the hu-
A. muciniphila especially acts with a specific man distal gut microbiome. Science 2006; 312:
protein called Amuc_1100, which is involved 1355-1359.
in immunomodulatory aspects81. Recently, Ott- 3) A hmadmehrabi S, Tang WHW. Gut microbiome and
man et al76 have reported that the administration its role in cardiovascular diseases. Curr Opin Car-
of the purified recombinant Amuc_1100 protein diol 2017; 32: 761-766.
improved glucose tolerance and decreased body 4) Quigley EMM. Microbiota-brain-gut axis and neu-
weight and fat mass gain in mice fed on high-fat rodegenerative diseases. Curr Neurol Neurosci
Rep 2017; 17: 94.
diet in comparison to untreated mice.
In this scenario, the essential role of A. muciniph- 5) Rios AC, M aurya PK, Pedrini M, Zeni -Graiff M, A se-
vedo E, M ansur RB, Wieck A, Grassi -Oliveira R, Mc-
ila in intestinal health maintenance, especially in lntyre RS, H ayashi MAF, Brietzke E. Microbiota ab-
immunomodulation of metabolic disorders is clear. normalities and the therapeutic potential of pro-
Consequently, future research will need to enhance biotics in the treatment of mood disorders. Rev
the key-role of this microorganism and its proteins Neurosci 2017; 28: 739-749.
in metabolic regulation and host immune modula- 6) M aeda Y, Takeda K. Role of gut microbiota in rheu-
tion, in order to improve its therapeutic application matoid arthritis. J Clin Med 2017; 6: 60.
in gastrointestinal diseases. 7) R ajilić-Stojanović M, de Vos WM. The first 1000
cultured species of the human gastrointestinal
microbiota. FEMS Microbiol Rev 2014; 38: 996-
1047.
Conclusions 8) Turnbaugh PJ, L ey RE, Hamady M, Fraser -L iggett CM,
K night R, Gordon JI. The human microbiome proj-
Gut microbiota is an active element to ensure ect. Nature 2007; 449: 804-810.
mucosal integrity, and its balance is necessary 9) Faith JJ, Guruge JL, Charbonneau M, Subramanian
for intestinal health maintenance. Every com- S, Seedorf H, Goodman AL, Clemente JC, K night R,

8080
Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders

Heath AC, L eibel RL, Rosenbaum M, Gordon JI. The nutrient loading on bacterioplankton communi-
long-term stability of the human gut microbiota. ty composition in lake mesocosms. Microb Ecol
Science 2013; 341: 1237439. 2006; 51: 137-146.
10) Schloissnig S, A rumugam M, Sunagawa S, Mitreva M, 24) Petroni G, Spring S, Schleifer K-H, Verni F, Rosati G.
Tap J, Zhu A, Waller A, Mende DR, Kultima JR, M ar - Defensive extrusive ectosymbionts of Euplotid-
tin J, Kota K, Sunyaev R, Weinstock GM, Bork P. Ge- ium (Ciliophora) that contain microtubule-like
nomic variation landscape of the human gut mi- structures are bacteria related to Verrucomicro-
crobiome. Nature 2013; 493: 45-50. bia. Proc Natl Acad Sci U S A 2000; 97: 1813-
11) Greenblum S, C arr R, Borenstein E. Extensive 1817.
strain-level copy-number variation across human 25) Vandekerckhove TTM, Coomans A, Cornelis K, Baert
gut microbiome species. Cell 2015; 160: 583-594. P, Gillis M. Use of the Verrucomicrobia-specific
12) Zhu A, Sunagawa S, Mende DR, Bork P. Inter-individ- probe EUB338-III and fluorescent in situ hybrid-
ual differences in the gene content of human gut ization for detection of “Candidatus xiphinemato-
bacterial species. Genome Biol 2015; 16: 82. bacter” cells in nematode hosts. Appl Environ Mi-
crobiol 2002; 68: 3121-3125.
13) Suau A, Bonnet R, Sutren M, Godon JJ, Gibson GR,
Collins MD, Doré J. Direct analysis of genes en- 26) Shinzato N, Muramatsu M, M atsui T, Watanabe Y.
coding 16S rRNA from complex communities re- Molecular phylogenetic diversity of the bacterial
veals many novel molecular species within the community in the gut of the termite Coptotermes
human gut. Appl Environ Microbiol 1999; 65: formosanus. Biosci Biotechnol Biochem 2005;
4799-4807. 69: 1145-1155.
14) Hold GL, Pryde SE, Russell VJ, Furrie E, Flint HJ. 27) Wang M, A hrné S, Jeppsson B, Molin G. Compari-
Assessment of microbial diversity in human co- son of bacterial diversity along the human intesti-
lonic samples by 16S rDNA sequence analysis. nal tract by direct cloning and sequencing of 16S
FEMS Microbiol Ecol 2002; 39: 33-39. rRNA genes. FEMS Microbiol Ecol 2005; 54: 219-
15) van Niftrik L, Devos DP. Editorial: Planctomyce-
231.
tes-Verrucomicrobia-Chlamydiae Bacterial Su- 28) de Vos WM. Microbe profile: Akkermansia mu-
perphylum: new model organisms for evolution- ciniphila: a conserved intestinal symbiont that
ary cell biology. Front Microbiol 2017; 8: 1458. acts as the gatekeeper of our mucosa. Microbiol
16) Derrien M, Vaughan EE, Plugge CM, de Vos WM. 2017; 163: 646-648.
Akkermansia municiphila gen. nov., sp. nov., a 29) Collado MC, Derrien M, Isolauri E, de Vos WM,
human intestinal mucin-degrading bacterium. Int Salminen S. Intestinal integrity and Akkermansia
J Syst Evol Microbiol 2004; 54 (Pt 5): 1469-1476. muciniphila, a mucin-degrading member of the
17) Wagner M, Horn M. The Planctomycetes, Verru- intestinal microbiota present in infants, adults,
comicrobia, Chlamydiae and sister phyla com- and the elderly. Appl Environ Microbiol 2007; 73:
prise a superphylum with biotechnological and 7767-7770.
medical relevance. Curr Opin Biotechnol 2006; 30) Reunanen J, K ainulainen V, Huuskonen L, Ottman N,
17: 241-249. Belzer C, Huhtinen H, de Vos W, Satokari R. Akker-
18) Gupta RS, Bhandari V, Naushad HS. Molecular sig- mansia muciniphila adheres to enterocytes and
natures for the pvc clade (Planctomycetes, Ver- strengthens the integrity of the epithelial cell lay-
rucomicrobia, Chlamydiae, and Lentisphaerae) er. Appl Environ Microbiol 2015; 81: 3655-3662.
of bacteria provide insights into their evolutionary 31) van Passel MWJ, K ant R, Zoetendal EG, Plugge CM,
relationships. Front Microbiol 2012; 3: 327. Derrien M, M alfatti SA, Chain PSG, Woyke T, Palva
19) L ee KC, Webb RI, Janssen PH, Sangwan P, Romeo T, A, de Vos WM, Smidt H. The genome of Akkerman-
Staley JT, Fuerst JA. Phylum Verrucomicrobia rep- sia muciniphila, a dedicated intestinal mucin de-
resentatives share a compartmentalized cell plan grader, and its use in exploring intestinal metage-
with members of bacterial phylum Planctomyce- nomes. PLoS One 2011; 6: e16876.
tes. BMC Microbiol 2009; 9: 5. 32) Lyra A, Forssten S, Rolny P, Wettergren Y, L ahtinen
20) Devos DP. PVC bacteria: variation of, but not ex- SJ, Salli K, Cedgård L, Odin E, Gustavsson B, Ouwe-
ception to, the Gram-negative cell plan. Trends hand A. Comparison of bacterial quantities in left
Microbiol 2014; 22: 14-20. and right colon biopsies and faeces. World J Gas-
troenterol 2012; 18: 4404-4411.
21) Sangwan P, Kovac S, Davis KER, Sait M, Janssen PH.
Detection and cultivation of soil verrucomicrobia. 33) Shin NR, L ee JC, L ee HY, K im MS, Whon TW, L ee MS,
Appl Environ Microbiol 2005; 71: 8402-8410. Bae JW. An increase in the Akkermansia spp. pop-
ulation induced by metformin treatment improves
22) M artiny AC, A lbrechtsen HJ, A rvin E, Molin S. Iden-
glucose homeostasis in diet-induced obese mice.
tification of bacteria in biofilm and bulk water sam-
Gut 2014; 63: 727-735.
ples from a nonchlorinated model drinking water
distribution system: detection of a large nitrite-ox- 34) L ee H, Ko G. Effect of metformin on metabolic im-
idizing population associated with Nitrospira spp. provement and gut microbiota. Appl Environ Mi-
Appl Environ Microbiol 2005; 71: 8611-8617. crobiol 2014; 80: 5935-5943.
23) Haukka K, Kolmonen E, Hyder R, Hietala J, Vakkilain - 35) Png CW, L indén SK, Gilshenan KS, Zoetendal EG,
en K, K airesalo T, H aario H., Sivonen K. Effect of McSweeney CS, Sly LI, McGuckin MA, Florin TH.

8081
I.G. Macchione, L.R. Lopetuso, G. Ianiro, M. Napoli, G. Gibiino, et al.

Mucolytic bacteria with increased prevalence in 46) Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren
IBD mucosa augment in vitro utilization of mucin O, Blekhman R, Beaumont M, Van Treuren W, K night
by other bacteria. Am J Gastroenterol 2010; 105: R, Bell JT, Spector TD, Clark AG, L ey RE. Human
2420-2428. genetics shape the gut microbiome. Cell 2014;
36) R ajilić-Stojanović M, Shanahan F, Guarner F, de Vos 159: 789-799.
WM. Phylogenetic analysis of dysbiosis in ulcer- 47) Montandon SA, Jornayvaz FR. Effects of antidiabet-
ative colitis during remission. Inflamm Bowel Dis ic drugs on gut microbiota composition. Genes
2013; 19: 481-488. Basel 2017; 8: 250.
37) Swidsinski A, Dörffel Y, Loening -Baucke V, Theissig 48) Aydin O, Nieuwdorp M, Gerdes V. The gut microbi-
F, Rückert JC, Ismail M, R au WA, G aschler D, Wei - ome as a target for the treatment of type 2 diabe-
zenegger M, Kühn S, Schilling J, Dörffel WV. Acute tes. Curr Diab Rep 2018; 18: 55.
appendicitis is characterised by local invasion 49) G ao Z, Yin J, Zhang J, Ward RE, M artin RJ, L efevre
with Fusobacterium nucleatum/necrophorum. Gut M, Cefalu WT, Ye J. Butyrate improves insulin sen-
2011; 60: 34-40. sitivity and increases energy expenditure in mice.
38) K iilerich P, Myrmel LS, Fjære E, Hao Q, Hugenholtz Diabetes 2009; 58: 1509-1517.
F, Sonne SB, Derrien M, Pedersen LM, Petersen RK, 50) C ani PD, Neyrinck AM, Fava F, K nauf C, Burcelin
Mortensen A, L icht TR, Rømer MU, Vogel UB, Waag - RG, Tuohy KM, Gibson GR, Delzenne NM. Selec-
bø LJ, Giallourou N, Feng Q, X iao L, L iu C, L iasert tive increases of bifidobacteria in gut microflo-
B, K leerebezem M, Wang J, M adsen L, K ristiansen K. ra improve high-fat-diet-induced diabetes in mice
Effect of a long-term high-protein diet on survival, through a mechanism associated with endotox-
obesity development, and gut microbiota in mice. aemia. Diabetologia 2007; 50: 2374-2383.
Am J Physiol Endocrinol Metab 2016; 310: 886-
899. 51) L ey RE, Turnbaugh PJ, K lein S, Gordon JI. Microbi-
al ecology: human gut microbes associated with
39) Nobel YR, Cox LM, K irigin FF, Bokulich NA, Yaman - obesity. Nature 2006; 444: 1022-1023.
ishi S, Teitler I, Chung J, Sohn J, Barber CM, Gold -
farb DS, R aju K, A bubucker S, Zhou Y, Ruiz VE, L i
52) Jumpertz R, L e DS, Turnbaugh PJ, Trinidad C, Bogar -
dus C, Gordon JI, K rakoff J. Energy-balance stud-
H, Mitreva M, A lekseyenko AV, Weinstock GM, So -
dergren E, Blaser MJ. Metabolic and metagenom-
ies reveal associations between gut microbes,
ic outcomes from early-life pulsed antibiotic treat- caloric load, and nutrient absorption in humans.
ment. Nat Commun 2015; 6: 7486. Am J Clin Nutr 2011; 94: 58-65.
40) Cox LM, Yamanishi S, Sohn J, A lekseyenko A V, L eung 53) Bibbò S, Ianiro G, Giorgio V, Scaldaferri F, M asucci L,
JM, Cho I, K im SG, L i H, G ao Z, M ahana D, Z árate G asbarrini A, C ammarota G. The role of diet on gut
Rodriguez JG, Rogers AB, Robin N, Loke P, Blaser microbiota composition. Eur Rev Med Pharmacol
MJ. Altering the intestinal microbiota during a crit- Sci 2016; 20: 4742-4749.
ical developmental window has lasting metabolic 54) M angiola F, Nicoletti A, G asbarrini A, Ponziani FR.
consequences. Cell 2014; 158: 705-721. Gut microbiota and aging. Eur Rev Med Pharma-
41) Brahe LK, L e Chatelier E, Prifti E, Pons N, K ennedy S, col Sci 2018; 22: 7404-7413.
Hansen T, Pedersen O, A strup A, Ehrlich SD, L arsen 55) Xu P, Li M, Zhang J, Zhang T. Correlation of intestinal
LH. Specific gut microbiota features and metabol- microbiota with overweight and obesity in Kazakh
ic markers in postmenopausal women with obesi- school children. BMC Microbiol 2012; 12: 283.
ty. Nutr Diabetes 2015; 5: e159. 56) Zhu L, Baker SS, Gill C, L iu W, A lkhouri R, Baker
42) Everard A, Belzer C, Geurts L, Ouwerkerk JP, Dru - RD, Gill SR. Characterization of gut microbiomes
art C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, in nonalcoholic steatohepatitis (NASH) patients:
Delzenne NM, de Vos WM, C ani PD. Cross-talk be- a connection between endogenous alcohol and
tween Akkermansia muciniphila and intestinal ep- NASH. Hepatology 2013; 57: 601-609.
ithelium controls diet-induced obesity. Proc Natl 57) Ismail NA, R agab SH, ElBaky AA, Shoeib ARS, A lho -
Acad Sci U S A 2013; 110: 9066-9071. sary Y, Fekry D. Frequency of Firmicutes and Bac-
43) Cecchini M, Sassi F, L auer JA, L ee YY, Guajardo -Bar - teroidetes in gut microbiota in obese and normal
ron V, Chisholm D. Tackling of unhealthy diets, weight Egyptian children and adults. Arch Med
physical inactivity, and obesity: health effects and Sci 2011; 7: 501-507.
cost-effectiveness. Lancet 2010; 376: 1775-1784. 58) A mar J, Serino M, L ange C, Chabo C, Iacovoni J,
44) Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Mondot S, L epage P, K lopp C, M ariette J, Bouchez O,
K eilbaugh SA, Bewtra M, K nights D, Walters WA, Perez L, Courtney M, M arren M, K lopp P, L antieri O,
K night R, Sinha R, Gilroy E, Gupta K, Baldassano R, Doré J, Charles M, Balkau B, Burcelin R; D.E.S.I.R.
Nessel L, Li H, Bushman FD, L ewis JD. Linking long- Study Group. Involvement of tissue bacteria in the
term dietary patterns with gut microbial entero- onset of diabetes in humans: evidence for a con-
types. Science 2011; 334: 105-108. cept. Diabetologia 2011; 54: 3055-3061.
45) David LA, M aurice CF, C armody RN, Gootenberg DB, 59) Backhed F, Ding H, Wang T, Hooper L V., Koh GY,
Button JE, Wolfe BE, L ing AV, Devlin AS, Varma Y, Nagy A, Semenkovich CF, Gordon GI. The gut mi-
Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh crobiota as an environmental factor that regulates
PJ. Diet rapidly and reproducibly alters the human fat storage. Proc Natl Acad Sci U S A 2004; 101:
gut microbiome. Nature 2014; 505: 559-563. 15718-15723.

8082
Akkermansia muciniphila: key player in metabolic and gastrointestinal disorders

60) Turnbaugh PJ, L ey RE, M ahowald MA, M agrini V, 70) Lopetuso LR, Scaldaferri F, Bruno G, Petito V, Fran -
M ardis ER, Gordon JI. An obesity-associated gut ceschi F, G asbarrini A. The therapeutic managment
microbiome with increased capacity for energy of gut barrier leaking: the emerging role for mu-
harvest. Nature 2006; 444: 1027–1031. cosal barrier protectors. Eur Rev Med Pharmacol
61) Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Sci 2015; 19: 1068-1076.
K au AL, Griffin NW, Lombard V, Henrissat B, Bain JR, 71) Ottman N, Davids M, Suarez-Diez M, Boeren S,
Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Schaap PJ, M artins dos Santos VAP, Smidt H, Belzer
Hayashi DK, Ly le BJ, M artini MC, Ursell LK, Clem - C, de Vos WM. Genomescale model and omics
ente JC, Van Treuren W, Walters WA, K night R, New - analysis of metabolic capacities of Akkermansia
gard CB, Heath AC, Gordon J. Gut microbiota from muciniphila reveal a preferential mucin-degrading
twins discordant for obesity modulate metabolism lifestyle. Appl Environ Microbiol 2017; 83: e01014-
in mice. Science 2013; 341: 1241214. 01017
62) Remely M, Hippe B, Geretschlaeger I, Stegmayer S, 72) Ottman N, Geerlings SY, A alvink S, de Vos WM, Bel-
Hoefinger I, Haslberger A. Increased gut microbi- zer C. Action and function of Akkermansia mu-
ota diversity and abundance of Faecalibacterium ciniphila in microbiome ecology, health and dis-
prausnitzii and Akkermansia after fasting: a pilot ease. Best Pract Res Clin Gastroenterol 2017; 31:
study. Wien Klin Wochenschr 2015; 127: 394-398. 637-642.
63) Everard A, M atamoros S, Geurts L, Delzenne NM, 73) Derrien M, Belzer C, de Vos WM. Akkermansia mu-
C ani PD. Saccharomyces boulardii administration ciniphila and its role in regulating host functions.
changes gut microbiota and reduces hepatic ste- Microb Pathog 2017; 106: 171-181.
atosis, low-grade inflammation, and fat mass in
74) Szachta P, Bartnicka A, G alecka M. Microbiota – a
obese and type 2 diabetic db/db mice. MBio 2014;
key to healing the gastrointestinal tract?. Pomer-
5: e01011-01014.
anian J life Sci 2016; 62: 21-24.
64) Schneeberger M, Everard A, Gómez-Valadés AG, M at-
75) Geerlings S, Kostopoulos I, de Vos W, Belzer C. Ak-
amoros S, R amírez S, Delzenne NM, R amon G, Clar -
kermansia muciniphila in the human gastrointesti-
et M, C ani PD. Akkermansia muciniphila inverse-
nal tract: when, where, and how? Microorganisms
ly correlates with the onset of inflammation, al-
2018; 6. pii: E75.
tered adipose tissue metabolism and metabolic
disorders during obesity in mice. Sci Rep 2015; 5: 76) Ottman N, Reunanen J, Meijerink M, Pietila TE, K a-
16643. inulainen V, K lievink J, Huuskon A alvink S, Skurnik

65) C aesar R, Tremaroli V, Kovatcheva-Datchary P, C ani M, Boeren S, Satokari R, Mercenier A, Palvi A, Smidt
PD, Bäckhed F. Crosstalk between gut microbiota H, de Vos WM, Belzer C. Pili-like proteins of Ak-
and dietary lipids aggravates WAT inflammation kermansia muciniphila modulate host immune
through TLR signaling. Cell Metab 2015; 22: 658- responses and gut barrier function. PLoS One
668 2017; 12: e0173004.
66) Everard A, L azarevic V, Derrien M, Girard M, Muc- 77) G anesh BP, K lopfleisch R, Loh G, Bl aut M. Com-
cioli GM, Neyrinck AM, Possemiers S, Van Holle A,
mensal Akkermansia muciniphila exacerbates
François P, de Vos WM, Delzenne NM, Schrenzel J, gut inflammation in Salmonella typhimurium-in-
C ani PD. Responses of gut microbiota and glu- fected gnotobiotic mice. PLoS One 2013; 8:
cose and lipid metabolism to prebiotics in genetic e74963.
obese and diet-induced leptin-resistant mice. Di- 78) C ani PD, de Vos WM. Next-generation beneficial
abetes 2011; 60: 2775-2786. microbes: the case of Akkermansia muciniphila.
67) Santacruz A, Collado MC, G arcía-Valdés L, Segura Front Microbiol 2017; 8: 1765.
MT, M arítn -L agos JA, A njos T, M artí -Romero M, Lo - 79) Rosario D, Benfeitas R, Bidkhori G, Zhang C, Uhlen
pez RM, Florido J, C ampoy C, SA nz Y. Gut microbi- M, Shoaie S, M ardinoglu A. Understanding the
ota composition is associated with body weight, representative gut microbiota dysbiosis in met-
weight gain and biochemical parameters in preg- formin-treated Type 2 diabetes patients using ge-
nant women. Br J Nutr 2010; 104: 83-92. nome-scale metabolic modeling. Front Physiol
68) K arlsson CLJ, Önnerfält J, Xu J, Molin G, A hrné 2018; 9: 775.
S, Thorngren -Jerneck K. The microbiota of the gut 80) A l K hodor S, Reichert B, Shatat IF. The microbiome
in preschool children with normal and excessive and blood pressure: can microbes regulate our
body weight. Obesity 2012; 20: 2257-2261. blood pressure?. Front Pediatr 2017; 5: 138.
69) Cone RA. Barrier properties of mucus. Adv Drug 81) C ani PD. Human gut microbiome: hopes, threats
Deliv Rev 2009; 61: 75-85. and promises. Gut 2018; 67: 1716-1725.

8083

You might also like