Becken Et Al 2021 Genotypic and Phenotypic Diversity Among Human Isolates of Akkermansia Muciniphila
Becken Et Al 2021 Genotypic and Phenotypic Diversity Among Human Isolates of Akkermansia Muciniphila
Becken Et Al 2021 Genotypic and Phenotypic Diversity Among Human Isolates of Akkermansia Muciniphila
a Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
b Department of Pediatrics, Duke University Hospital, Durham, North Carolina, USA
c Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
Bradford Becken and Lauren Davey are co-first authors. Author order was determined alphabetically.
phila phylogroups, yet the prevalence of these phylogroups and how they vary in Editor Margaret J. McFall-Ngai, University of
Hawaii at Manoa
their ability to influence human health is largely unknown. In this study, we per-
Copyright © 2021 Becken et al. This is an
formed a genomic and phenotypic analysis of 71 A. muciniphila strains and identified open-access article distributed under the terms
phylogroup-specific traits such as oxygen tolerance, adherence, and sulfur acquisi- of the Creative Commons Attribution 4.0
International license.
tion that likely influence colonization of the GI tract and differentially impact meta-
Address correspondence to Raphael H.
bolic and immunological health. In humans, we observed that single Akkermansia Valdivia, [email protected].
phylogroups predominate at a given time but that the phylotype can switch in an This article is a direct contribution from
individual. This collection of strains provides the foundation for the functional char- Raphael H. Valdivia, a Fellow of the American
acterization of A. muciniphila phylogroup-specific effects on the multitude of host Academy of Microbiology, who arranged for
and secured reviews by Eric Martens, University
outcomes associated with Akkermansia colonization, including protection from obe- of Michigan, and Manuela Raffatellu, University
sity, diabetes, colitis, and neurological diseases, as well as enhanced responses to of California San Diego.
cancer immunotherapies. Received 30 March 2021
Accepted 5 April 2021
KEYWORDS Verrucomicrobia, comparative genomics, phylogroups, microbiome, Published 18 May 2021
mucin, assimilatory sulfur reduction (ASR), adolescent obesity, phylogenetic analysis
®
May/June 2021 Volume 12 Issue 3 e00478-21 mbio.asm.org 1
®
Becken et al.
RESULTS
The relative abundance of A. muciniphila in fecal samples is highly variable in a
cohort of children with obesity. To survey the diversity of A. muciniphila strains pres-
ent among healthy children and those with obesity, we used fecal samples that had
been collected before and after various interventions aimed at decreasing their body
mass index (BMI) (18). We selected 5 lean controls (Z-score adjusted BMI, or zBMI, –0.99
to 0.41) and 36 children with extreme obesity (zBMI, –1.63 to 3.18 . 95th percentile)
(Table S1). For healthy lean control children, a single stool sample was obtained at the
time of enrollment. For the majority of the cohort with obesity (35/36), we used sam-
ples collected at baseline and at the end of the study (6 months). We enriched for
mucolytic bacteria directly from frozen fecal material by serial passage in liquid me-
dium with gastric porcine mucin as the sole carbon and nitrogen source and then iso-
lated single colonies on mucin agar plates. Bacterial colonies were purified to homoge-
neity and typed by sequencing the V3-4 region of the 16S rRNA locus. Overall, we
cultured 71 strains of A. muciniphila from 35 children and 1 adult. In parallel, we per-
formed a 16S rRNA-based survey of the bacterial communities present in selected fecal
samples for which we had baseline and at least one additional visit (Fig. 1 and
Table S2). A phylum-level analysis indicated that the relative abundance of
Verrucomicrobia ranged from undetectable to 31% of total bacteria (Table S2).
A. muciniphila clinical isolates are phenotypically diverse. Previous analysis of
microbial metagenomes indicated that there is significant diversity among A. mucini-
phila strains (19). To begin to address if this genetic diversity correlates with traits of
relevance to the colonization and health of the human host, we performed a range of
phenotypic tests that have been associated with Akkermansia biology, including varia-
tions in (i) growth rates, (ii) the ability to form bacterial aggregates, (iii) adherence to
epithelial surfaces, (iv) the generation of short-chain fatty acids (SCFA) during mucin
fermentation, (v) sensitivity to oxidative stress, and (vi) activation of Toll-like receptors
(TLRs).
Growth rates. The growth rates among individual strains were monitored under
anaerobic conditions in a semidefined synthetic medium consisting of glucose, N-ace-
tylglucosamine, soy peptone, and threonine. A. muciniphila strains displayed doubling
times ranging from 1.2 to 13 h. These differences were more pronounced when gastric
porcine mucin was the sole carbon and nitrogen source, with doubling times ranging
from 0.3 to .20 h (Fig. 2A and Table S3).
Aggregation. Some A. muciniphila isolates, unlike the reference strain A. mucini-
phila MucT, readily sedimented when grown in liquid culture media (Fig. 2B). We meas-
ured the extent to which strains aggregated by monitoring the changes in the optical
density in samples obtained from the surface of culture tubes that had been grown in
mucin medium (Table S3).
Adherence to epithelial cells. We determined if there are strain-level differences
in A. muciniphila isolates for their ability to attach to intestinal epithelial cells. Binding
is a range of responses, with some strains being fairly tolerant to prolonged exposure
to ambient oxygen (;60% survival at 24 h), while others were extremely sensitive
(,0.01% survival at 12 h) (Table S3).
Activation of innate immune sensors. Human monocytes are activated by expo-
sure to A. muciniphila MucT (23), and TLR2-dependent recognition is required for signal
transduction events that strengthen barrier function in the GI tract (24). We first tested
to be determine the relevant TLRs involved in the recognition of A. muciniphila by stim-
ulating bone marrow-derived macrophages (BMDM) from wild-type C57BL/6J mice
and from tlr4, tlr2 tlr4, unc93b1, and tlr2 tlr4 unc93b1 knockout mice (25). Unc93b12/2
mice are defective for the transport of TLRs that sense nucleic acids as well as the
expression of TLR5, which recognizes flagellin, at the cell surface (26, 27). BMDM were
incubated with A. muciniphila MucT at a ratio of 0.5 or 5 bacteria per cell for 6 h, and
the secretion of the cytokine tumor necrosis factor alpha (TNF-a) and interleukin-6 (IL-
6) was assessed (Fig. 3A). At the lowest dose of Akkermansia, the response of BMDM
was exclusively dependent on TLR4. TLR2-dependent activation was only apparent
when bacterial loads were increased by 10-fold. Consistent with previous findings (23),
additional TLRs were not required for immune activation of mouse BMDMs.
We next used HEK-TLR reporter cell lines to test the ability of A. muciniphila isolates
to specifically activate TLR2 and TLR4. We used cell lines expressing TLR4 or TLR2 and
its coreceptors TLR1 and TLR6, which respond to known TLR2 ligands (28). A. mucini-
phila strains were grown in mucin or synthetic medium and incubated with reporter
cell lines expressing TLR2 or TLR4 for 16 h at a ratio of 1 or 5 bacteria per HEK-TLR cell,
FIG 4 A. muciniphila isolates from children belong to three different phylogroups. (A) Whole-genome comparison of 40 A. muciniphila strains. The average
nucleotide identity (ANI) was calculated at 96% identity using Anvi’o and PyANI. Genomes for previously published strains (16, 17) belonging to each
Akkermansia phylogroup were included as controls (blue), as well as the type-strain MucT (red). Complete genomes were assembled from PacBio reads.
Note that phylogroup AMI can be subdivided into two subtypes at 96% ANI threshold. (B) Circle phylogram of new A. muciniphila strains. The graph
displays the pangenome of 40 sequenced isolates, 3 reference genomes (gray), and the type strain MucT (red). The phylogram is clustered based on gene
frequency and displays gene cluster presence/absence for each genome. Selected phylogroup-specific gene groups are highlighted to show their
phylogroups AmII and AmIV were consistently larger than the AmI genomes (Fig. 4A
and B and Table S3).
We determined that several phenotypes segregated by phylogroup (Fig. 5). AmI
strains displayed rapid doubling times, while most members of AmII and AmIV grew
slowly (Fig. 5A). Strains also differed in their sensitivity to ambient oxygen, with strains of
AmII being resistant and those of AmIV very sensitive (Fig. 5B). Differential oxygen sensi-
tivity was also observed within phylogroup AmI, with AmIb strains being highly sensitive
to exposure to air, while AmIa strains displayed intermediate resistance. AmIV strains had
higher adherence to epithelial cells and displayed a greater propensity to aggregate
when grown in mucin medium (Fig. 5C and D). We also saw a small drop in the ratio of
acetate/propionate fermentation end products of mucin fermentation for AmII strains
(Fig. 5E), which we postulate is because they synthesize vitamin B12 (16) and hence gener-
ate more propionate as vitamin B12 in the growth medium becomes limiting.
For the stimulation of HEK-TLR reporter cell lines, AmII and AmIV strains were more
stimulatory for both TLR2 and TLR4 than AmI strains (Fig. 5F and G), but it is unclear if
this is simply a reflection of their enhanced binding properties to cell surfaces. AmI
strains displayed a broad range of activation of TLR reporter cell lines, and while there
was a trend for AmIb strains to be more stimulatory, particularly for TLR4, the differen-
ces did not reach statistical significance given the relatively low number of AmIa iso-
lates in our strain collection.
To identify genes that may contribute to these phenotypes, we analyzed the pange-
nome of our A. muciniphila strains and identified 4,982 total gene clusters, with 1,647
core gene clusters found in all genomes and 506 gene clusters found only in single
genomes (Fig. 4A). We found several phylogroup-specific gene groups that may con-
tribute to phenotypic variation (Fig. 4B and C and Table S3). For example, all members
of phylogroups AmII and AmIV were predicted to encode distinct capsule and exopoly-
saccharide genes that were absent in AmI strains. These included putative capsular
polysaccharide (CPS) biosynthesis proteins EpsC and EpsI, a CMP-N-acetylneuraminic
acid synthetase, and a capsule modifying enzyme, polysaccharide pyruvyl transferase
WcaK (29). The AmI isolates have a single Cps locus, while AmII and AmIV genomes
contain two additional loci that are largely conserved among these phylogroups and
map to similar regions in the chromosome. This suggests common capsule types are
present in the phylogroup AmII and IV strains. Phylogroup AmIV also codes for DltB, an
enzyme typically involved in modification of lipoteichoic acid in Gram-positive bacteria
but that can also modify lipopolysaccharides in some Gram-negative bacteria (30).
Conversely, members of phylogroup AmI had additional chemotaxis genes, cyto-
chrome c biosynthetic genes, and code for a quality control sensor protein for outer
membrane biogenesis, NlpE (31).
The phylogroups also displayed differences in iron acquisition systems. Anaerobic con-
ditions favor reduced ferrous iron (Fe21), and aerobic conditions favor the oxidized ferric
iron (Fe31) (32). Although all genomes had a ferrous iron transport system consisting of
FeoAB genes (Amuc_1088, Amuc_1089, and Amuc_1090 in MucT), phylogroups AmI and
AmII encoded additional mechanisms to acquire ferric iron. Members of AmI had multiple
enterochelin transporter gene groups, suggesting that they might use siderophores to
scavenge ferric iron. Members of phylogroup AmII also have predicted ferric iron trans-
porters, although these appear to be distinct from the gene groups in phylogroup AmI. In
contrast, phylogroup AmIV lacks canonical mechanisms for ferric iron acquisition.
Phylogroup AmIV is highly sensitive to ambient oxygen, and it is plausible that defects in
iron acquisition or the absence of the oxidative stress protection associated with sidero-
phores (33, 34) may contribute to this phenotype. Additional contributors to differences
in oxygen sensitivity include a LexA repressor, indicative of an SOS system, that is present
in all phylogroup AmI and AmII strains.
The A. muciniphila strains were predicted to encode approximately 27 glycoside hy-
drolase (GH) enzyme families, but they varied in the abundance of GH families among
the phylogroups, particularly in AmIV genomes (Fig. 3B). GH97 enzymes, which com-
prise glycoamylases such as Bacteroides thetaiotaomicron SusB (35), were detected in
all strains except for AmIV. Similarly, AmIV genomes had fewer GH110 enzymes, a
group of galactosidases capable of cleaving blood group B antigens (36). Conversely,
AmIV strains were enriched for GH29 and GH95 L-fucosidases, which could potentially
cleave the terminal fucose residues that decorate mucin and human milk oligosaccha-
FIG 6 AmII and AmIV phylogroups are defective for assimilatory sulfate reduction (ASR). (A) Distribution of ASR genes among A. muciniphila phylogroups.
ASR genes (top) from A. muciniphila MucT were used to search for homologs among other sequenced isolates. Blue squares indicate that a gene is present,
and gray squares indicate that a gene was not detected. There are two cysK homologs in MucT, Amuc_1301 and Amuc_2014. (B) Genomic context of ASR
genes in Akkermansia phylogroups. The majority of the ASR genes are clustered in a single locus in the AmI phylogroup, as represented in strains MucT and
Akk0500B. The AmI strain, Akk2670, lacked the entire ASR locus, although the flanking genes remained conserved in other AmI strains. The AmII strain
to generate cysteine. In the MucT strain, ASR genes are clustered in a single locus
(Amuc_1294 to Amuc_1301), with the exception of a CysJ homolog (Amuc_0631)
and a second cysteine synthase (Amuc_2014). The locus also included a potential
inner membrane sulfide permease (Amuc_1295), an ABC transporter-related ATP-
binding protein (Amuc_1296), and a substrate-binding protein (Amuc_1297) (Fig. 6A
and B).
AmII and AmIV strains lacked the ASR gene cluster found in MucT but retained dis-
tally encoded homologs for CysJ and the Amuc_2014 cysteine synthase (CysK-b)
(Fig. 6A). All AmI strains were predicted to perform ASR, except for the strain Akk2760
(Fig. 6A and Fig. S3A), which lacked the entire ASR locus (Fig. 6B). Since Akk2670 grew
very poorly in mucin medium, with a growth rate comparable to that of AmII and
AmIV isolates (Fig. 5A and Table S3), we hypothesized that the inability to generate
reduced sulfur may be a limiting factor for their growth on mucin in vitro. To test this,
representative strains were grown in mucin medium with or without the addition of
cysteine or sodium hydrosulfide as a source of H2S. While cysteine and H2S shortened
the lag time for the growth of the MucT strain, it did not affect the maximal biomass
achieved (Fig. 6C). In contrast, cysteine significantly enhanced the maximal growth of
the predicted ASR-deficient AmIa strain Akk2670, the AmII strain Akk2196, and the
AmIV strain Akk0496 (Fig. 6C). These findings suggest that A. muciniphila strains benefit
FIG 7 A. muciniphila phylogroups AmII and AmIV strains outcompete AmI in antibiotic pretreated
mice. (A to D) Mice were either untreated (A and C) or treated (B and D) with antibiotics (Ab) and
gavaged with a three-phylogroup strain mix containing an equal amount of phylogroups AmIa
(MucT), AmIb (Akk1683), and AmII (Akk0580) (A and B) or a four-phylogroup strain mix containing
phylogroups AmIa (MucT), AmIb (Akk1570), AmII (Akk0580), and AmIV (Akk0490) (C and D). The AmIa
strain identified in mice that had not been pretreated with Ab (A and C) represents the endogenous
mouse Akkermansia. Each point represents the average of three cages (n = 4 mice/cage for panels A
and B, and n = 2 mice/cage for panels C and D), and error bars represent the standard deviation. The
Am1a strain in panels A to C represent the endogenous mouse A. muciniphila strain found at the
Duke University vivarium.
from the addition of reduced sulfur when grown in mucin and that the growth of ASR-
deficient phylogroups is significantly enhanced by the addition of cysteine or H2S.
FIG 8 Evidence for single A. muciniphila phylogroup dominance in humans. A subset of stool samples from 14
patients (PID, patient ID) were selected for detection and quantification of specific phylogroups by qPCR.
Samples were selected for analysis if patients had provided multiple samples (n . 3) throughout a 6-month
period, and 16S rRNA community profiling indicated the presence of Akkermansia at at least one time point.
The size of the bubbles represents the relative abundance of Akkermansia as assessed with QIIME analysis.
Triangles indicate that no Akkermansia/Verrucomicrobia 16S rRNA sequences were detected. The frequency of
phylogroup types was assessed by qPCR with specific primers and is color coded. In all cases, the dominant
phylogroup in the GI tract, additional strains can be present and viable despite being
below the levels of detection by molecular methods.
In addition to evidence for the coexistence of minor clades, we observed major
changes in the overall abundance of Akkermansia phylogroups within the same
patient. In 11 of 14 of patients with multiple sampling over a 6-month period, either
the relative amount of Akkermansia fluctuated significantly (.100-fold) among sam-
ples or the identity of the phylogroups switched (Fig. 8 and Table S6). For instance,
three patients (PID141, PID181, and PID182) switched their predominant phylogroup
from an AmI strain to either AmII or AmIV strains by 6 months. In two patients that
underwent bariatric surgery (PID001, PID019), both converted from almost undetect-
able levels of Akkermansia at baseline to a phylogroup AmIV-dominant microbiota by
the 6-month time point. It is unclear if the emergence of new dominant phylogroups
represents blooms of preexisting low-abundance strains or new colonization events.
DISCUSSION
Obesity is a multifactorial disease influenced by host genetics, diet, behavior, and
the microbial ecosystems that populate the human GI tract. Diet remains a key driver
of microbial composition, and the phenotypes associated with these resident microbial
communities strongly influence their impact on the immunological and metabolic
health of their host (42). However, some bacterial species seem to play an oversized
role on the health of their hosts. For instance, A. muciniphila has emerged as a poten-
tial probiotic since its abundance in the GI tract positively correlates with decreased
incidence of metabolic disease, obesity, and diabetes (9, 10).
There is a growing recognition that there is a great diversity of Akkermansia strains
and species. A pangenomic analysis of Akkermansia genomes revealed four A. muciniphila
phylogroups (16), and an analysis of .1,000 Akkermansia genomes reconstructed from
metagenomic sequences from human samples across the world suggested the existence
of up to four new species (43, 44). While metabolic capacities of different Akkermansia
strains have been inferred based on genome annotations, experimental validation is
largely lacking because isolates of these new phylogroups have either not been collected
or not been characterized. A notable exception is a recent analysis of the vitamin B12 bio-
Bifidobacterium genus, cysteine auxotrophies are prominent, and at least in the case of
B. bifidum, cysteine auxotrophy cannot be rescued by supplementation of glutathione
or taurine (52). Finally, several bacterial pathogens are cysteine auxotrophs, and even
among species with complete ASR systems, clinical isolates have been observed to
spontaneously become cysteine auxotrophs (48). Thus, there may be selective pressure
for the loss of ASR genes in the presence of alternative reduced sulfur sources.
The loss of ASR in some A. muciniphila strains suggests that AmII and IV strains
could be net consumers of any microbiota-derived H2S, especially under conditions
where AmII and AmIV strains constitute a significant proportion of the entire micro-
biota (Fig. 8). If so, their localized detoxification of H2S may contribute to some of the
protection that has been ascribed to Akkermansia in the context of inflammatory
bowel disease (IBD) and Crohn’s disease (53–55). On the other hand, H2S derived from
the breakdown of cysteine by intestinal cystathionine b -synthase has anti-inflamma-
tory properties (56) and may stimulate the production of mucins (57). Under these cir-
cumstances, colonization by AmII and AmIV strains may be proinflammatory if they
decrease the effective concentration of H2S at epithelial surfaces.
The relative competitive advantage of AmII and AmIV strains in antibiotic-treated mice
was unexpected given the relative high prevalence of AmI strains in human populations
(16, 44). However, we noted that microbiota of specific-pathogen-free (SPF) mice, which
has an endogenous mouse AmI strain, provided colonization resistance, which led us to
ask if a similar phylogroup exclusion is observed in humans. In stool samples collected at
various time points after various interventions aimed at reducing obesity, we observed
dominance by a single phylogroup. In some instances, both 16S rRNA-based community
profiling and qPCR indicated that A. muciniphila was not detectable in baseline samples
yet appeared by 6 months (PID001 and PID009), and in others, the dominant phylogroup
disappeared within 3 months after baseline sample collection (PID033 and PID137), and
yet in other patient samples, a phylogroup would disappear and return (PID049) or be
replaced by a new phylogroup (PID141, PID181, and PID182). The abrupt disappearance
of Akkermansia has been previously documented in densely sampled individuals (58). At
this, stage we cannot distinguish between population crashes that are followed by repo-
Three biological replicates of this assay were performed for every strain, and each contained three
technical replicates.
Adherence to epithelial cells. HT29-MTX cells were seeded into 96-well plates at a density of
2.5 104 cells per well and grown for 7 days past confluence. Wells were washed twice with PBS and
incubated with 2.5 106 A. muciniphila cells in DMEM for 2 h at 37°C under anaerobic conditions. As a
control for nonspecific binding of Akkermansia, UltraCruz high binding enzyme-linked immunosorbent
assay (ELISA) (sc-204463) plates were precoated with 100 m l of 1% bovine serum albumin (BSA). Wells
with HT29-MTX cells or coated with BSA were washed twice with PBS to remove nonadherent bacteria.
Synthetic medium (100 m l) was added to each well, and plates were cultured for either 48 h or 96 h at
37°C under anaerobic conditions. HT29-MTX cell or BSA binding was assessed by measuring the optical
density at 600 nm after outgrowth in the assay wells and calculating the ratio of HT29-MTX coated OD:
BSA coated OD. Data are reported as the average and standard deviation of 3 technical replicates from 3
to 4 independent biological replicates. For microscopy, HT29-MTX cells were seeded into 24-well plates
with 12-mm round glass coverslips at a density of 1 105 cells per well and grown for 7 days past con-
fluence. Wells were washed twice with PBS and incubated with 1 106 A. muciniphila cells in 500 ml an-
aerobic-adapted DMEM for 2 h at 37°C under anaerobic conditions. Wells were washed twice, fixed with
3.7% formaldehyde in PBS for 30 min on ice, washed twice with PBS, and blocked overnight at 4°C in
blocking buffer (2% [wt/vol] BSA in PBS). Coverslips were incubated with a 1:50 dilution of anti-
Akkermansia polyclonal antibody followed by an incubation with goat anti-rabbit-488 (Invitrogen; cata-
log number A-11008) and Hoechst for 1 h at 25°C. After two washes, coverslips were mounted on slides
with Vectashield medium and imaged on a Nikon Eclipse Ti2 inverted microscope with 20 objective.
Measurement of short-chain fatty acids (SCFA). For each strain, 1 ml of culture supernatants from
strains grown in mucin medium was removed for SCFA analysis following the protocol of Holmes et al.
(69). In brief, the supernatant was centrifuged at 14,000 relative centrifugal force (rcf) for 5 min at 4°C to
pellet debris, and then 750 m l of supernatant was passed through a 0.22-mm spin column filter. The re-
sultant filtrate was then acidified to a pH of ,3 with 50 m l of 6N HCL and transferred to a glass autosam-
pler vial for analysis. Filtrates were analyzed on an Agilent 7890 gas chromatograph (GC) equipped with
a flame-ionization detector (FID) and an Agilent HP-FFAP free fatty-acid column (69). The concentrations
of acetate and propionate in the samples were determined using an 8-point standard curve (0.1 mM to
16 mM).
Sensitivity to ambient oxygen. Strains were grown from frozen stocks in 0.5 ml mucin (0.4%) me-
dium in deep 96-well plates to saturation. After subculturing in mucin medium for 5 h, serial dilutions of
each strain were spotted on BBL BHI agar (BD; catalog number 211065) plates supplemented with 0.4%
mucin. One plate was left in the anaerobic chamber, while the others were exposed to ambient O2 for
12, 18, or 24 h, before being returned to the chamber. The relative sensitivity to ambient oxygen was
determined by monitoring the ratio of CFU with and without exposure to ambient oxygen.
Bone marrow macrophage (BMM) stimulations. BMMs were obtained from 6- to 12-week-old
C57BL/6J mice of the following genotypes: wild-type, Tlr2–/– Tlr4–/– Unc93b13d/3d, Tlr2–/– Tlr4–/–, Tlr2–/–
Unc93b13d/3d, and Tlr4–/– (25). Bone marrow was dissociated through a 70-m m filter, treated with ACK
lysis buffer (Gibco; catalog numberA1049201), and differentiated for 6 days in DMEM complete medium
(DMEM supplemented with 10% [vol/vol] fetal bovine serum, L-glutamine, penicillin-streptomycin, so-
dium pyruvate, HEPES, and 2-mercaptoethanol) supplemented with 10% (vol/vol) of supernatants from
3T3-CSF cells, overproducing macrophage colony-stimulating factor. For stimulations, BMMs were plated
in DMEM complete medium supplemented with 10% (vol/vol) M-CSF and incubated with A. muciniphila
at the indicated multiplicity of “infection” (MOI), 1 m M CpG-B (InvivoGen; tlr1-1668-1), 500 ng/ml
Pam3CSK4 (InvivoGen; tlr1-pms), or 50 ng/ml lipopolysaccharide (LPS) (InvivoGen; tlr1-3pelps). For analy-
sis of secreted cytokines, the supernatant was collected 4 h after stimulation and analyzed with the BD
cytometric bead array mouse inflammation kit (BD Biosciences; catalog number 552364) according to
AmI, AmII, and AmIII were described in Guo et al. and retrieved from NCBI (17) (GenBank assembly
accession numbers GCA_002885425.1, GCA_002885025.1, GCA_002884975.1, GCA_002884915.1, and
GCA_002885515.1). The phylogroup AmIV representative genome CDI-150b was obtained from the JGI
IMG database (16). Based on the resulting analysis, each genome was assigned to a phylogroup using
the function anvi-import-misc-data, and phylogroup-specific gene functions were then obtained using
the command anvi-get-enriched-functions-per-pan-group to identify cluster of orthologous groups of
proteins (COGs). COGs present in all members of a given phylogroup, and absent in all other phy-
logroups, were considered to represent phylogroup-specific gene functions. The adjusted q value repre-
sents the false-discovery rate adjusted P value corrected for multiple testing as calculated by Anvi-o.
Finally, the genomes were analyzed for metabolic pathways in Anvi’o (https://merenlab.org/software/
anvio/help/main/programs/anvi-estimate-metabolism/). Each genome was annotated with the KEGG
KOfam database using the program anvi-run-kegg-kofams (78, 79). The annotated genomes were then
used as input to the program anvi-estimate-metabolism, with the flag –module-completion-threshold
set to identify pathways with a minimum of 50% completion in at least one isolate genome. The module
output was further analyzed with the command anvi-compute-functional-enrichment to test for phy-
logroup-specific enrichment. To visualize the data, heatmaps were generated using the pheatmap R
package (80), and genes were plotted with the gggenes R package (81) in ggplot2 (82). To search for
specific ASR genes among the isolates, custom BLAST databases were generated using the annotated
isolate genomes (83). Searches were conducted using the sequences for the ASR genes from A. mucini-
phila MucT as the query.
To identify glycoside hydrolase families in the sequenced strains, we used dbCAN2 version 2.0.11 to
annotate carbohydrate-active enzymes (84). DNA fasta files were used as input, and the annotation was
run using the standalone tool run_dbcan. The resulting annotation tables were analyzed to determine
the number of each type of glycoside hydrolase family per genome. Annotations detected with
Diamond and at least one additional method, Hotpep or Hmmer, were considered positive.
Mouse colonization and phylogroup competitions. To prepare the inoculum for competition
experiments, A. muciniphila cultures were standardized by optical density, combined using equal parts
of each phylogroup to be tested, and stored at 280°C in PBS containing 20% glycerol.
All mouse experiments were approved by Duke University’s Institutional Animal Care and Use
Committee. In vivo competition experiments were caried out using 6-week-old female C57BL/6J mice
obtained from Jackson Laboratories. A. muciniphila colonization was tested in mice both with and with-
out pretreatment with antibiotics (3 g/liter tetracycline suspended in distilled water with 10% sucrose
for 2 weeks). Following antibiotic treatment, clearance of residual mouse Akkermansia was determined
by PCR using Akkermansia-specific 16S rRNA primers (12).
For in vivo competition assays, mice were inoculated by intragastric gavage with a mixture contain-
ing 2.5 108 CFU of each phylogroup in a total volume of 140 m l. The three-phylogroup competition
experiment used a mixture of the strains MucT (AmIa), Akk1683 (AmIb), and Akk0580 (AmII). Two addi-
tional competition experiments were run, each using a combination of four clades. The first four-clade
competition used a mixture of strains MucT (AmIa), Akk1570 (AmIb), Akk0580 (AmII), and Akk0490
SUPPLEMENTAL MATERIAL
Supplemental material is available online only.
FIG S1, TIF file, 2.7 MB.
FIG S2, TIF file, 0.9 MB.
FIG S3, GIF file, 1.5 MB.
FIG S4, TIF file, 2.8 MB.
TABLE S1, XLSX file, 0.01 MB.
TABLE S2, XLSX file, 0.02 MB.
TABLE S3, XLSX file, 0.02 MB.
TABLE S4, XLSX file, 0.01 MB.
TABLE S5, XLSX file, 0.02 MB.
TABLE S6, XLSX file, 0.01 MB.
ACKNOWLEDGMENTS
We thank the Duke University School of Medicine for the use of the Microbiome
Core Facility, which provided fecal samples and 16S rRNA analysis support and the
REFERENCES
1. Fan Y, Pedersen O. 2021. Gut microbiota in human metabolic health and the intestinal microbiota present in infants, adults, and the elderly. Appl
disease. Nat Rev Microbiol 19:55–71. https://doi.org/10.1038/s41579-020 Environ Microbiol 73:7767–7770. https://doi.org/10.1128/AEM.01477-07.
-0433-9. 13. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y,
2. Allaire JM, Crowley SM, Law HT, Chang SY, Ko HJ, Vallance BA. 2018. The Derrien M, Muccioli GG, Delzenne NM, de Vos WM, Cani PD. 2013. Cross-
intestinal epithelium: central coordinator of mucosal immunity. Trends talk between Akkermansia muciniphila and intestinal epithelium controls
Immunol 39:677–696. https://doi.org/10.1016/j.it.2018.04.002. diet-induced obesity. Proc Natl Acad Sci U S A 110:9066–9071. https://doi
3. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, .org/10.1073/pnas.1219451110.
Arumugam M, Batto JM, Kennedy S, Leonard P, Li J, Burgdorf K, Grarup N, 14. Plovier H, Everard A, Druart C, Depommier C, Van Hul M, Geurts L, Chilloux J,
Jørgensen T, Brandslund I, Nielsen HB, Juncker AS, Bertalan M, Levenez F, Ottman N, Duparc T, Lichtenstein L, Myridakis A, Delzenne NM, Klievink J,
Pons N, Rasmussen S, Sunagawa S, Tap J, Tims S, Zoetendal EG, Brunak S, Bhattacharjee A, van der Ark KCH, Aalvink S, Martinez LO, Dumas M-E,
Clément K, Doré J, Kleerebezem M, Kristiansen K, Renault P, Sicheritz- Maiter D, Loumaye A, Hermans MP, Thissen J-P, Belzer C, de Vos WM, Cani
Ponten T, De Vos WM, Zucker JD, Raes J, Hansen T, Bork P, Wang J, Ehrlich PD. 2017. A purified membrane protein from Akkermansia muciniphila or
SD, Pedersen O, Guedon E, Delorme C, Layec S, Khaci G, Van De Guchte M, the pasteurized bacterium improves metabolism in obese and diabetic
Vandemeulebrouck G, Jamet A, Dervyn R, Sanchez N, MetaHIT consortium, mice. Nat Med 23:107–113. https://doi.org/10.1038/nm.4236.
et al. 2013. Richness of human gut microbiome correlates with metabolic 15. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S,
markers. Nature 500:541–546. https://doi.org/10.1038/nature12506. Falony G, Raes J, Maiter D, Delzenne NM, de Barsy M, Loumaye A,
4. Sonnenburg ED, Sonnenburg JL. 2019. The ancestral and industrialized Hermans MP, Thissen J-PP, de Vos WM, Cani PD. 2019. Supplementation
gut microbiota and implications for human health. Nat Rev Microbiol with Akkermansia muciniphila in overweight and obese human volun-
17:383–390. https://doi.org/10.1038/s41579-019-0191-8. teers: a proof-of-concept exploratory study. Nat Med 25:1096–1103.
https://doi.org/10.1038/s41591-019-0495-2.
23. Ottman N, Reunanen J, Meijerink M, Pietilä TE, Kainulainen V, Klievink J, 43. Guo X, Zhang J, Wu F, Zhang M, Yi M, Peng Y. 2016. Different subtype
Huuskonen L, Aalvink S, Skurnik M, Boeren S, Satokari R, Mercenier A, strains of Akkermansia muciniphila abundantly colonize in southern
Palva A, Smidt H, de Vos WM, Belzer C. 2017. Pili-like proteins of Akker- China. J Appl Microbiol 120:452–459. https://doi.org/10.1111/jam.13022.
mansia muciniphila modulate host immune responses and gut barrier 44. Lv Q, Li SH, Zhang Y, Wang YC, Peng Y, Zhang XX. 2020. A thousand meta-
function. PLoS One 12:e0173004. https://doi.org/10.1371/journal.pone genome-assembled genomes of Akkermansia reveal new phylogroups
.0173004. and geographical and functional variations in human gut. bioRxiv
24. Cario E, Gerken G, Podolsky DK. 2007. Toll-like receptor 2 controls muco- 2020.09.10.292292.
sal inflammation by regulating epithelial barrier function. Gastroenterol- 45. Jin MS, Kim SE, Heo JY, Lee ME, Kim HM, Paik SG, Lee H, Lee JO. 2007. Crys-
ogy 132:1359–1374. https://doi.org/10.1053/j.gastro.2007.02.056. tal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-
25. Sivick KE, Arpaia N, Reiner GL, Lee BL, Russell BR, Barton GM. 2014. Toll- acylated lipopeptide. Cell 130:1071–1082. https://doi.org/10.1016/j.cell
like receptor-deficient mice reveal how innate immune signaling influen- .2007.09.008.
ces Salmonella virulence strategies. Cell Host Microbe 15:203–213. 46. Kang JY, Nan X, Jin MS, Youn SJ, Ryu YH, Mah S, Han SH, Lee H, Paik SG,
https://doi.org/10.1016/j.chom.2014.01.013. Lee JO. 2009. Recognition of lipopeptide patterns by Toll-like receptor 2-
26. Huh JW, Shibata T, Hwang M, Kwon E-H, Jang MS, Fukui R, Kanno A, Jung Toll-like receptor 6 heterodimer. Immunity 31:873–884. https://doi.org/10
.1016/j.immuni.2009.09.018.
DJ, Jang MH, Miyake K, Kim YM. 2014. UNC93B1 is essential for the plasma
47. Bäckström M, Ambort D, Thomsson E, Johansson MEV, Hansson GC. 2013.
membrane localization and signaling of Toll-like receptor 5. Proc Natl
Increased understanding of the biochemistry and biosynthesis of MUC2
Acad Sci U S A 111:7072–7077. https://doi.org/10.1073/pnas.1322838111.
and other gel-forming mucins through the recombinant expression of
27. Majer O, Liu B, Woo BJ, Kreuk LSM, Van Dis E, Barton GM. 2019. Release
their protein domains. Mol Biotechnol 54:250–256. https://doi.org/10
from UNC93B1 reinforces the compartmentalized activation of select
.1007/s12033-012-9562-3.
TLRs. Nature 575:371–374. https://doi.org/10.1038/s41586-019-1611-7. 48. Lensmire JM, Hammer ND. 2019. Nutrient sulfur acquisition strategies
28. Oliveira-Nascimento L, Massari P, Wetzler LM. 2012. The role of TLR2 in
employed by bacterial pathogens. Curr Opin Microbiol 47:52–58. https://
infection and immunity. Front Immunol 3:1–17. https://doi.org/10.3389/ doi.org/10.1016/j.mib.2018.11.002.
fimmu.2012.00079. 49. Tripp HJ, Kitner JB, Schwalbach MS, Dacey JWH, Wilhelm LJ, Giovannoni
29. Pan YJ, Lin TL, Chen CT, Chen YY, Hsieh PF, Hsu CR, Wu MC, Wang JT. SJ. 2008. SAR11 marine bacteria require exogenous reduced sulphur for
2015. Genetic analysis of capsular polysaccharide synthesis gene clusters growth. Nature 452:741–744. https://doi.org/10.1038/nature06776.
in 79 capsular types of Klebsiella spp. Sci Rep 5:15573. https://doi.org/10 50. Seif Y, Choudhary KS, Hefner Y, Anand A, Yang L, Palsson BO. 2020. Meta-
.1038/srep15573. bolic and genetic basis for auxotrophies in Gram-negative species. Proc
30. Percy MG, Gründling A. 2014. Lipoteichoic acid synthesis and function in Natl Acad Sci U S A 117:6264–6273. https://doi.org/10.1073/pnas
Gram-positive bacteria. Annu Rev Microbiol 68:81–100. https://doi.org/10 .1910499117.
.1146/annurev-micro-091213-112949. 51. Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, Gaskins HR. 2012. Mi-
31. May KL, Lehman KM, Mitchell AM, Grabowicz M. 2019. A stress response crobial pathways in colonic sulfur metabolism and links with health and
monitoring lipoprotein trafficking to the outer membrane. mBio 10:1–14. disease. Front Physiol 3:1–11. https://doi.org/10.3389/fphys.2012.00448.
https://doi.org/10.1128/mBio.00618-19. 52. Ferrario C, Duranti S, Milani C, Mancabelli L, Lugli GA, Turroni F, Mangifesta
32. Lau CKY, Krewulak KD, Vogel HJ. 2016. Bacterial ferrous iron transport: the M, Viappiani A, Ossiprandi MC, van Sinderen D, Ventura M. 2015. Exploring
Feo system. FEMS Microbiol Rev 40:273–298. https://doi.org/10.1093/ amino acid auxotrophy in Bifidobacterium bifidum PRL2010. Front Microbiol
femsre/fuv049. 6:1–11. https://doi.org/10.3389/fmicb.2015.01331.
33. Achard MES, Chen KW, Sweet MJ, Watts RE, Schroder K, Schembri MA, 53. Earley H, Lennon G, Balfe Á, Coffey JC, Winter DC, O’Connell PR. 2019. The
McEwan AG. 2013. An antioxidant role for catecholate siderophores in abundance of Akkermansia muciniphila and its relationship with sulph-
Salmonella. Biochem J 454:543–549. https://doi.org/10.1042/BJ20121771. ated colonic mucins in health and ulcerative colitis. Sci Rep 9:15683.
34. Peralta DR, Adler C, Corbalán NS, Paz García EC, Pomares MF, Vincent PA. https://doi.org/10.1038/s41598-019-51878-3.
62. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, 72. Hunt M, De Silva N, Otto TD, Parkhill J, Keane JA, Harris SR. 2015. Circlator:
Alexander H, Alm EJ, Arumugam M, Asnicar F, Bai Y, Bisanz JE, Bittinger K, automated circularization of genome assemblies using long sequencing
Brejnrod A, Brislawn CJ, Brown CT, Callahan BJ, Caraballo-Rodríguez AM, reads. Genome Biol 16:1–10. https://doi.org/10.1186/s13059-015-0849-0.
Chase J, Cope EK, Da Silva R, Diener C, Dorrestein PC, Douglas GM, Durall 73. Brettin T, Davis JJ, Disz T, Edwards RA, Gerdes S, Olsen GJ, Olson R,
DM, Duvallet C, Edwardson CF, Ernst M, Estaki M, Fouquier J, Gauglitz JM, Overbeek R, Parrello B, Pusch GD, Shukla M, Thomason JA, Stevens R,
Gibbons SM, Gibson DL, Gonzalez A, Gorlick K, Guo J, Hillmann B, Holmes Vonstein V, Wattam AR, Xia F. 2015. RASTtk: a modular and extensible
S, Holste H, Huttenhower C, Huttley GA, Janssen S, Jarmusch AK, Jiang L, implementation of the RAST algorithm for building custom annotation
Kaehler BD, Bin Kang K, Keefe CR, Keim P, Kelley ST, Knights D, Koester I, pipelines and annotating batches of genomes. Sci Rep 5:8365. https://doi
et al. 2019. Reproducible, interactive, scalable and extensible microbiome .org/10.1038/srep08365.
data science using QIIME 2. Nat Biotechnol 37:852–857. https://doi.org/10 74. Eren AM, Esen OC, Quince C, Vineis JH, Morrison HG, Sogin ML, Delmont
.1038/s41587-019-0209-9. TO. 2015. Anvi’o: an advanced analysis and visualization platform for
63. Hamady M, Knight R. 2009. Microbial community profiling for human ’omics data. PeerJ 2015:1–29. https://doi.org/10.7717/peerj.1319.
microbiome projects: tools, techniques, and challenges. Genome Res 75. Delmont TO, Eren EM. 2018. Linking pangenomes and metagenomes: the
19:1141–1152. https://doi.org/10.1101/gr.085464.108. prochlorococcus metapangenome. PeerJ 6:e4320. doi:https://doi.org/10
64. Hamady M, Walker JJ, Harris JK, Gold NJ, Knight R. 2008. Error-correcting .7717/peerj.4320.
barcoded primers for pyrosequencing hundreds of samples in multiplex. 76. Hyatt D, Chen GL, LoCascio PF, Land ML, Larimer FW, Hauser LJ. 2010.
Prodigal: prokaryotic gene recognition and translation initiation site iden-
Nat Methods 5:235–237. https://doi.org/10.1038/nmeth.1184.
tification. BMC Bioinformatics 11:119. https://doi.org/10.1186/1471-2105
65. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP.
-11-119.
2016. DADA2: high-resolution sample inference from Illumina amplicon
77. Pritchard L, Glover RH, Humphris S, Elphinstone JG, Toth IK. 2016.
data. Nat Methods 13:581–583. https://doi.org/10.1038/nmeth.3869.
Genomics and taxonomy in diagnostics for food security: soft-rotting
66. Bokulich NA, Kaehler BD, Rideout JR, Dillon M, Bolyen E, Knight R, Huttley
enterobacterial plant pathogens. Anal Methods 8:12–24. https://doi.org/
GA, Gregory Caporaso J. 2018. Optimizing taxonomic classification of
10.1039/C5AY02550H.
marker-gene amplicon sequences with QIIME 2’s q2-feature-classifier plu-
78. Aramaki T, Blanc-Mathieu R, Endo H, Ohkubo K, Kanehisa M, Goto S,
gin. Microbiome 6:1–17. https://doi.org/10.1186/s40168-018-0470-z.
Ogata H. 2020. KofamKOALA: KEGG Ortholog assignment based on pro-
67. Yilmaz P, Parfrey LW, Yarza P, Gerken J, Pruesse E, Quast C, Schweer T,
file HMM and adaptive score threshold. Bioinformatics 36:2251–2252.
Peplies J, Ludwig W, Glöckner FO. 2014. The SILVA and “All-species Living https://doi.org/10.1093/bioinformatics/btz859.
Tree Project (LTP)” taxonomic frameworks. Nucleic Acids Res 42: 79. Muto A, Kotera M, Tokimatsu T, Nakagawa Z, Goto S, Kanehisa M. 2013.
D643–D648. https://doi.org/10.1093/nar/gkt1209. Modular architecture of metabolic pathways revealed by conserved
68. Sprouffske K, Wagner A. 2016. Growthcurver: an R package for obtaining sequences of reactions. J Chem Inf Model 53:613–622. https://doi.org/10
interpretable metrics from microbial growth curves. BMC Bioinformatics .1021/ci3005379.
17:17–20. https://doi.org/10.1186/s12859-016-1016-7. 80. Kolde R. 2013. pheatmap: pretty heatmaps. R package version 1.0.12.
69. Holmes ZC, Silverman JD, Dressman HK, Wei Z, Dallow EP, Armstrong SC, https://cran.r-project.org/web/packages/pheatmap/index.html.
Seed PC, Rawls JF, David LA. 2020. Short-chain fatty acid production by 81. Wilkins D. 2020. gggenes: draw gene arrow maps in “ggplot2.” R package ver-
gut microbiota from children with obesity differs according to prebiotic sion 0.4.1. https://cran.r-project.org/web/packages/gggenes/index.html.
choice and bacterial community composition. mBio 11:e00914-20. 82. Wickham H. 2009. ggplot2: elegant graphics for data analysis. Springer-
https://doi.org/10.1128/mBio.00914-20. Verlag, New York, NY.
70. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, 83. 2008. BLAST command line applications user manual [Internet]. National
Abecasis G, Durbin R, 1000 Genome Project Data Processing Subgroup. Center for Biotechnology Information, Bethesda (MD). https://www.ncbi
2009. The Sequence Alignment/Map format and SAMtools. Bioinformatics .nlm.nih.gov/books/NBK52640/.
25:2078–2079. https://doi.org/10.1093/bioinformatics/btp352. 84. Zhang H, Yohe T, Huang L, Entwistle S, Wu P, Yang Z, Busk PK, Xu Y, Yin Y.
71. Kolmogorov M, Yuan J, Lin Y, Pevzner PA. 2019. Assembly of long, error-