ArticlePDF Available

An Expanded Genetic Code Enables Trimethylamine Metabolism in Human Gut Bacteria

American Society for Microbiology
mSystems®
Authors:

Abstract and Figures

Links between trimethylamine- N -oxide (TMAO) and cardiovascular disease (CVD) have focused attention on mechanisms by which animal-based diets have negative health consequences. In a meta-analysis of data from foundational gut microbiome studies, we found evidence that specialized bacteria have and express a metabolic pathway that circumvents TMAO production and is often misannotated because it relies on genetic code expansion. This naturally occurring mechanism for TMAO attenuation is negatively correlated with CVD. Ultimately, these findings point to new avenues of research that could increase microbiome-informed understanding of human health and hint at potential biomedical applications in which specialized bacteria are used to curtail CVD development.
Content may be subject to copyright.
An Expanded Genetic Code Enables Trimethylamine
Metabolism in Human Gut Bacteria
Veronika Kivenson,
a
Stephen J. Giovannoni
a
a
Department of Microbiology, Oregon State University, Corvallis, Oregon, USA
ABSTRACT Cardiovascular disease (CVD) has been linked to animal-based diets,
which are a major source of trimethylamine (TMA), a precursor of the proathero-
genic compound trimethylamine-N-oxide (TMAO). Human gut bacteria in the genus
Bilophila have genomic signatures for genetic code expansion that could enable
them to metabolize both TMA and its precursors without production of TMAO. We
uncovered evidence that the Bilophila demethylation pathway is actively transcribed
in gut microbiomes and that animal-based diets cause Bilophila to rapidly increase
in abundance. CVD occurrence and Bilophila abundance in humans were signifi-
cantly negatively correlated. These data lead us to propose that Bilophila, which is
commonly regarded as a pathobiont, may play a role in mitigating cardiovascular
disease. Human gut microbiomes have been shown to affect the development of a
myriad of disease states, but mechanistic connections between diet, health, and mi-
crobiota have been challenging to establish. The hypothesis that Bilophila reduces
cardiovascular disease by circumventing TMAO production offers a clearly defined
mechanism with a potential human health impact, but investigations of Bilophila cell
biology and ecology will be needed to fully evaluate these ideas.
IMPORTANCE Links between trimethylamine-N-oxide (TMAO) and cardiovascular dis-
ease (CVD) have focused attention on mechanisms by which animal-based diets
have negative health consequences. In a meta-analysis of data from foundational
gut microbiome studies, we found evidence that specialized bacteria have and ex-
press a metabolic pathway that circumvents TMAO production and is often misan-
notated because it relies on genetic code expansion. This naturally occurring mecha-
nism for TMAO attenuation is negatively correlated with CVD. Ultimately, these
findings point to new avenues of research that could increase microbiome-informed
understanding of human health and hint at potential biomedical applications in
which specialized bacteria are used to curtail CVD development.
KEYWORDS cardiovascular disease, microbiome, molecular genetics
The human gut microbiome is increasingly recognized for its influential role in
health. The gut microbiome has been implicated in cardiovascular disease (CVD),
the leading cause of death in developed countries (1). Metabolic end products asso-
ciated with the consumption of animal products have been shown to promote CVD;
dietary sources of proatherogenic compounds include choline, phosphatidylcholine,
and L-carnitine (2–11). Gut microorganisms convert these compounds to trimethyl-
amine (TMA), which in turn is converted to trimethylamine-N-oxide (TMAO) through the
action of host hepatic flavin monooxygenase 3 (12, 13). TMAO is a causative agent of
CVD pathogenesis; therefore, elucidating pathways relevant to this compound is
central to understanding human health (10, 14).
The canonical view of CVD and TMAO involves the action of the gut microbiota in
transforming precursor compounds from a range of animal-based dietary sources to
TMA, which can then be converted to TMAO in the liver (Fig. 1A). While TMAO is
Citation Kivenson V, Giovannoni SJ. 2020. An
expanded genetic code enables
trimethylamine metabolism in human gut
bacteria. mSystems 5:e00413-20. https://doi
.org/10.1128/mSystems.00413-20.
Editor Robert G. Beiko, Dalhousie University
Copyright © 2020 Kivenson and Giovannoni.
This is an open-access article distributed under
the terms of the Creative Commons Attribution
4.0 International license.
Address correspondence to Veronika Kivenson,
kivensov@oregonstate.edu.
Received 18 May 2020
Accepted 24 September 2020
Published
RESEARCH ARTICLE
Molecular Biology and Physiology
crossm
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 1
27 October 2020
frequently referred to as the sole breakdown product of TMA, a subset of methano-
genic archaea in the gut have the ability to utilize TMA by an alternative pathway (15,
16). This pathway is enabled by genetic code expansion (GCE), via insertion of the 22nd
amino acid, pyrrolysine (Pyl), in place of a TAG amber codon at conserved sites of the
tri-, di-, and monomethylamine methyltransferase genes, with methane as the end
product (17–19). TMA metabolism that relies on proteins that use an expanded code
has also been described in some bacteria from environmental settings, including
symbionts of gutless marine worms (20), as well as the Firmicutes bacterium Acetoha-
lobium arabaticum isolated from a Crimean lagoon (21). Despite its potential impor-
tance, bacterial metabolism of TMA remains largely unexplored in the gut microbiome.
ADeltaproteobacterium commonly found in the human gut, Bilophila wadsworthia,
also has genes necessary for encoding pyrrolysine (22). First identified in an appendi-
citis infection in 1989, this “bile-loving,” taurine-degrading bacterium is commonly
referred to as a pathobiont associated with abscesses (23, 24). Additionally, Bilophila is
able to produce hydrogen sulfide, and these bacteria may be linked to inflammatory
bowel disease, colorectal cancer, and systematic inflammation (24–28). Despite its
classification as a pathobiont, Bilophila is also commonly present in healthy human
microbiomes (29, 30). There is uncertainty about GCE and TMA metabolic pathway
characteristics in these organisms; notably, whether the pyrrolysine pathway is func-
tional in trans and whether the pathway is expressed. The function of this pathway is
questioned (21) because of organizational differences in the pyrrolysyl-tRNA synthetase
gene (required for encoding Pyl): in archaea, this synthetase is encoded by a single gene,
while in Bilophila, the N- and C-terminal domains are encoded by two distinct genes.
Second, in previous reports of Pyl pathways, the trimethylamine methyltransferase and
Pyl genes are commonly adjacent to one another (21, 22), while this is not the case in
Bilophila. Third, the TMA methyltransferase protein is prematurely truncated at the TAG
codon in public Bilophila data sets available on the NCBI and JGI-IMG webservers.
Despite the ubiquity of this bacterium, and its potential importance in modulating
TMAO levels (and thus CVD risk), these ambiguities remain unresolved, and studies of
the human gut microbiome make no mention of the possibility of a bacterial pathway
for TMA utilization. In this study, we investigate this alternative pathway of trimethyl-
amine metabolism in Bilophila, the change in abundance of this taxon in response to
an animal-based diet, and the correlation between Bilophila abundance and CVD
pathology (Fig. 1B).
FIG 1 (A) Canonical view of TMA: this compound is converted to TMAO in the liver via a host hepatic flavin monooxygenase 3, leading
to increased risk of cardiovascular disease. (B) A new view of TMA metabolism proposed in this study: the specialized gut bacteria,
Bilophila, increase in abundance and use genetic code expansion to augment metabolism, thereby reducing the amount of TMA available
for conversion to TMAO. TMA, trimethylamine; FMO-3, flavin monooxygenase 3; TMAO, trimethylamine-N-oxide; CVD, cardiovascular
disease; DMA, dimethylamine.
Kivenson and Giovannoni
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 2
RESULTS
To explore the potential for Bilophila TMA metabolism, we retranslated the protein-
coding sequences from publicly available Bilophila genomes (Table S1) using a custom
translation table with the TAG codon as a readthrough rather than a stop codon (see
Materials and Methods). We found an in-frame TAG codon at a conserved position in
the TMA methyltransferase, corresponding to the pyrrolysine residue of this protein in
archaeal methanogens (17) and bacteria (20, 21)(Fig. 2 and Table S2). All genes known
to have functions specific to pyrrolysine production were also identified in the Bilophila
genomes: PylB, PylC, and PylD and the pyrrolysyl-tRNA synthetase N and C termini, as
well as the Pyl-specific tRNA, with the corresponding CUA anticodon (Fig. 2; Fig. S1).
One exception was that Bilophila wadsworthia ATCC 49260 is missing one component:
a Pyl tRNA was not located. In addition to the Pyl-containing TMA methyltransferase,
accessory genes for the TMA methyltransferase pathway, including ramA (the activating
gene) and the TMA methyltransferase corrinoid protein, are encoded in all of the
Bilophila genomes (Fig. 2 and Table S3). The dimethylamine methyltransferase gene is
not encoded in any of the genomes, and a gene similar to monomethylamine meth-
yltransferase, while present, does not have the conserved in-frame TAG codon that is
a hallmark of this gene. In summary, metabolic inference from genomic data
indicates that Bilophila have the potential to convert TMA to dimethylamine (DMA)
in the human gut.
FIG 2 Mechanistic overview of genetic code expansion augmented metabolism performed by Bilophila in the human gut. Bcct
transporters, betaine/carnitine/choline transporter family proteins; CutD, choline trimethylamine lyase activating enzyme; CutC, choline
trimethylamine lyase; TMA, trimethylamine; DMA, dimethylamine; PylB, 3-methylornithine synthase; PylC, 3-methylornithine-l-lysine ligase;
PylD, 3-methylornithyl-N
6
-L-lysine dehydrogenase; Pyl-tRNA synthetase, pyrrolysyl-tRNA synthetase; Pyl tRNA, pyrrolysine tRNA; ramA,
methylamine methyltransferase corrinoid protein reductive activase; TMA corrinoid, methyltransferase cognate corrinoid protein; SelD,
selenide, water dikinase; SelA, L-seryl-tRNA(Sec) selenium transferase; SelB, selenocysteine-specific translation elongation factor;
Sec-tRNA, selenocysteine tRNA; SECIS Element, selenocysteine insertion sequence element; TMAO, trimethylamine-N-oxide; FMO-3,
flavin monooxygenase 3.
TMA Metabolism in Human Gut Bacteria
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 3
The TMA-utilizing pathway we observed may benefit Bilophila by enabling them to
use TMA directly and also TMA produced metabolically within the cell from two
precursor compounds, choline and glycine betaine (Fig. 2). Briefly, Bilophila genomes
encode the glycyl radical choline-TMA lyase and its associated activating protein (CutC
and CutD, respectively), which convert choline to TMA (31). Glycine betaine (sources of
which includes choline [32] and possibly carnitine [33]) can be converted to TMA via the
selenocysteine (Sec)-containing glycine betaine reductase (GRD) pathway (34). The GRD
pathway and the full set of machinery required for the noncanonical amino acid
selenocysteine, encoded with a repurposed UGA stop codon, are also present in the
Bilophila genomes (Table S3). Multiple copies of proteins belonging to the betaine/
carnitine/choline family of transporters are also encoded in the Bilophila genomes. We
conclude that the genomic data indicate that Bilophila has the ability to use choline and
glycine betaine, converting these compounds to TMA, and subsequently to DMA. In
doing so, these bacteria may deplete precursor compounds and TMA that would
otherwise be available for host hepatic processes, thereby reducing or circumventing
production of TMAO (Fig. 2).
Next, we asked whether the pyrrolysine pathway is functional and whether GCE-
enabled TMA metabolism is active in Bilophila from the gut environment. To do so, we
reexamined data sets from a recent human gut metatranscriptomic study (35) and a
mouse model system study (36) (Table S1). Applying a minimum threshold of 98%
amino acid sequence identity with annotated Bilophila proteins, we identified expres-
sion of the TMA methyltransferase and pyrrolysine machinery proteins in both human
fecal and mouse cecum samples (Fig. 2 and Tables S4 and S5). The fraction of TMA
consumed via this bacterial metabolic process in the human gut microbiome remains
uncertain, but expression data support the conclusion that this metabolic process is
active.
DISCUSSION
This survey of published microbiome sequence data uncovered evidence that
bacteria in the genus Bilophila use genetic code expansion in the human gut to
produce a TMA methyltransferase. We hypothesized that this mechanism could be used
to compete with other TMA-utilizing processes, potentially decreasing the production
of TMAO from the proatherogenic precursor trimethylamine. To explore this hypoth-
esis, we reexamined additional publicly available data (Table S1) to determine whether
this naturally occurring mechanism for TMAO attenuation is correlated with CVD. In a
recent study describing the gut microbiome in atherosclerotic cardiovascular disease,
Jie et al. (29) report that Bilophila is one of the 20 most abundant genera in the samples
examined for this project. Their data also show that the abundance of Bilophila is
significantly enriched in the microbiomes of individuals in the healthy/control group
(n187) compared to the CVD group (n218). Second, in a study describing a rapid
diet-induced change in the human gut microbiome, David et al. (37) reveal that
Bilophila significantly increase in abundance in response to an animal-based diet
compared to a plant-based diet. Finally, in a study detailing the transmission of
atherosclerosis susceptibility via gut microbial transplanation, Gregory et al. (38) show
that mice with certain taxa have increased TMAO levels and develop atherosclerotic
lesions and postulate that this is a microbiome-dependent, transmissible trait. Bilophila
is one of only six taxa that are significantly enriched in both the cecal and fecal
microbiome of the healthy group compared to the mice that developed atherosclerotic
lesions. The observations we report may challenge the widely held idea that members
of this taxon act exclusively as pathobionts; their role in the microbiome and human
health may be context dependent, and their potential to mitigate the impacts of animal
products on CVD warrants further study.
MATERIALS AND METHODS
Microbiome data selection and accession numbers. Genomic, metagenomic, and metatranscrip-
tomic data sets used for this study were accessed on 15 January 2020 from publicly available sequencing
projects (see Table S1 in the supplemental material). The Bilophila genomes analyzed are reference
Kivenson and Giovannoni
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 4
genomes from the Human Microbiome Project (39) as well as the type strain for this genus, from the
Refseq database (40). All of the available genomes from the genus Bilophila were examined. BioProject
accession no. PRJEB33885 was excluded because it includes a duplicate of a previously published
genome (included in accession no. PRJNA41963). Paired metagenomic-metatranscriptomic data were
accessed from a large cohort study of adult men (35), and additional metatranscriptomic data were used
from a mouse model system (36). Metatranscriptomic and metagenomic read data were obtained using
the SRA-toolkit (https://github.com/ncbi/sra-tools) v2.9.1 data via the fastq-dump option.
Genomic analysis and implementation of a custom protein translation table. Predicted proteins
in each genome were initially identified using Prodigal (42) v.2.6.3 using single genome mode, and
functional annotation was determined using Hmmer (43) v.3.1, with the hmmscan option (1E10 cutoff),
with top hits only, against the Pfam (44) database v.31 and Tigrfam (45) database v.15.0.
For genomes in which enzymes were identified for the synthesis of pyrrolysine, we applied an
alternate protein prediction procedure that reassigned TAG codons from stop to readthrough. To
accomplish this task, we modified the source code for Prodigal by adding a custom translation table that
has TAG readthrough and retains all three canonical bacterial start codons. Proteins with in-frame stop
codons in the relevant genes were then manually inspected to determine whether the region containing
and following the stop codon was conserved in comparisons to homologues containing pyrrolysine at
a similar position (see Fig. S2 in the supplemental material). The modified code and documentation for
the modified translation table are freely available at https://github.com/VeronikaKivenson/Prodigal.
Protein searches for the full-length TMA methyltransferase amino acid sequence from Bilophila were
performed on the NCBI and JGI-IMG webservers on 1 July 2020. For searching and identifying putative
selenocysteine-containing proteins, the bSECIS (46) webserver was used and results were inspected and
compared with previously identified selenoproteins. Protein sequence alignment was performed using
Muscle (47), with Geneious 2020.1 used for visualization of the alignments. The Aragorn (48) web server
was used to locate the tRNA sequences from each genome and to determine the secondary structure of
Sec- and Pyl-specific tRNAs, as well as their corresponding anticodon sequences. tRNAscan-SE (49) v.1.23
was also used to search for the tRNAs for Sec. Chemdraw and Biorender software were used to create
figures.
Metatranscriptomic data analysis. Preliminary mapping of metatranscriptomic sequence data to
Bilophila genomes was done using Bowtie2 (50) v2.3.4.1. This approach identified samples in which these
Bilophila bacteria were present and active at detectable levels. Next, metatranscriptomic reads were
coassembled using SRA data from select samples belonging to each BioProject (Table S4) using Megahit
(51) v.1.1.1, with default parameters. Functional annotation and identification of stop codon readthrough
were performed as described earlier. In addition to Prodigal, FragGeneScan was used to identify partial
genes (52). The computational biology data processing and analysis workflow were completed using the
Extreme Science and Engineering Discovery Environment (XSEDE) Bridges resource at the Pittsburgh
Supercomputing Center (53, 54).
SUPPLEMENTAL MATERIAL
Supplemental material is available online only.
FIG S1, PDF file, 0.2 MB.
FIG S2, PDF file, 0.1 MB.
TABLE S1, PDF file, 0.04 MB.
TABLE S2, PDF file, 0.04 MB.
TABLE S3, PDF file, 0.03 MB.
TABLE S4, PDF file, 0.1 MB.
TABLE S5, PDF file, 0.05 MB.
ACKNOWLEDGMENTS
This work was supported by a grant from the Simons Foundation (SFARI 649176 to
V.K.). This work used the Extreme Science and Engineering Discovery Environment
(XSEDE), which is supported by National Science Foundation grant number ACI-
1548562. Specifically, it used the Bridges system, which is supported by NSF award
number ACI-1445606, at the Pittsburgh Supercomputing Center (PSC), through alloca-
tion TG-DEB170007.
Any opinions, findings, and conclusions or recommendations expressed in this
material are those of the author(s) and do not necessarily reflect the views of the
National Science Foundation.
We thank Grace Deitzler and Maude David for helpful discussion of the gut micro-
biome data sets.
We declare that we have no competing interests.
The project was conceived by V.K. and S.J.G. V.K. performed the bioinformatic data
analysis, and interpretation was performed by V.K. and S.J.G. V.K. and S.J.G. wrote the
manuscript.
TMA Metabolism in Human Gut Bacteria
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 5
REFERENCES
1. Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson
AP, Chamberlain AM, Chang AR, Cheng S, Das SR, Delling FN, Djousse L,
Elkind MSV, Ferguson JF, Fornage M, Jordan LC, Khan SS, Kissela BM,
Knutson KL, Kwan TW, Lackland DT, Lewis TT, Lichtman JH, Longenecker
CT, Loop MS, Lutsey PL, Martin SS, Matsushita K, Moran AE, Mussolino
ME, O’Flaherty M, Pandey A, Perak AM, Rosamond WD, Roth GA, Samp-
son UKA, Satou GM, Schroeder EB, Shah SH, Spartano NL, Stokes A,
Tirschwell DL, Tsao CW, Turakhia MP, VanWagner LB, Wilkins JT, Wong
SS, Virani SS, American Heart Association Council on Epidemiology and
Prevention Statistics Committee and Stroke Statistics Committee. 2019.
Heart disease and stroke statistics—2019 update: a report from the
American Heart Association. Circulation 139:e56 e528. https://doi.org/
10.1161/CIR.0000000000000659.
2. Ufnal M, Zadlo A, Ostaszewski R. 2015. TMAO: a small molecule of great
expectations. Nutrition 31:1317–1323. https://doi.org/10.1016/j.nut.2015
.05.006.
3. Falony G, Vieira-Silva S, Raes J. 2015. Microbiology meets big data: the
case of gut microbiota-derived trimethylamine. Annu Rev Microbiol
69:305–321. https://doi.org/10.1146/annurev-micro-091014-104422.
4. Janeiro MH, Ramírez MJ, Milagro FI, Martínez JA, Solas M. 2018. Impli-
cation of trimethylamine N-oxide (TMAO) in disease: potential biomarker
or new therapeutic target. Nutrients 10:1398. https://doi.org/10.3390/
nu10101398.
5. Velasquez MT, Ramezani A, Manal A, Raj DS. 2016. Trimethylamine N-oxide:
the good, the bad and the unknown. Toxins (Basel) 8:326. https://doi.org/
10.3390/toxins8110326.
6. Tang WHW, Hazen SL. 2014. The contributory role of gut microbiota in
cardiovascular disease. J Clin Invest 124:4204 4211. https://doi.org/10
.1172/JCI72331.
7. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE,
Britt EB, Fu X, Chung Y-M, Wu Y, Schauer P, Smith JD, Allayee H, Tang
WHW, DiDonato JA, Lusis AJ, Hazen SL. 2011. Gut flora metabolism of
phosphatidylcholine promotes cardiovascular disease. Nature 472:
57– 63. https://doi.org/10.1038/nature09922.
8. Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen
SL. 2013. Intestinal microbial metabolism of phosphatidylcholine and
cardiovascular risk. N Engl J Med 368:1575–1584. https://doi.org/10
.1056/NEJMoa1109400.
9. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X,
Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier
M, Brown JM, Krauss RM, Tang WHW, Bushman FD, Lusis AJ, Hazen SL.
2013. Intestinal microbiota metabolism of L-carnitine, a nutrient in red
meat, promotes atherosclerosis. Nat Med 19:576 –585. https://doi.org/10
.1038/nm.3145.
10. Brown JM, Hazen SL. 2018. Microbial modulation of cardiovascular disease.
Nat Rev Microbiol 16:171–181. https://doi.org/10.1038/nrmicro.2017.149.
11. Chittim CL, Martínez del Campo A, Balskus EP. 2019. Gut bacterial phos-
pholipase Ds support disease-associated metabolism by generating choline.
Nat Microbiol 4:155–163. https://doi.org/10.1038/s41564-018-0294-4.
12. Lang D, Yeung C, Peter R, Ibarra C, Gasser R, Itagaki K, Philpot R, Rettie
A. 1998. Isoform specificity of trimethylamine N-oxygenation by human
flavin-containing monooxygenase (FMO) and P450 enzymes: selective
catalysis by FMO3. Biochem Pharmacol 56:1005–1012. https://doi.org/
10.1016/s0006-2952(98)00218-4.
13. Zhang AQ, Mitchell SC, Smith RL. 1999. Dietary precursors of trimethyl-
amine in man: a pilot study. Food Chem Toxicol 37:515–520. https://doi
.org/10.1016/s0278-6915(99)00028-9.
14. Gilbert JA, Quinn RA, Debelius J, Xu ZZ, Morton J, Garg N, Jansson JK,
Dorrestein PC, Knight R. 2016. Microbiome-wide association studies link
dynamic microbial consortia to disease. Nature 535:94 –103. https://doi
.org/10.1038/nature18850.
15. Borrel G, McCann A, Deane J, Neto MC, Lynch DB, Brugère J-F, O’Toole
PW. 2017. Genomics and metagenomics of trimethylamine-utilizing Ar-
chaea in the human gut microbiome. ISME J 11:2059 –2074. https://doi
.org/10.1038/ismej.2017.72.
16. Brugère J-F, Borrel G, Gaci N, Tottey W, O’Toole PW, Malpuech-Brugère
C. 2014. Archaebiotics: proposed therapeutic use of archaea to prevent
trimethylaminuria and cardiovascular disease. Gut Microbes 5:5–10.
https://doi.org/10.4161/gmic.26749.
17. Paul L, Ferguson DJ, Krzycki JA. 2000. The trimethylamine methyltrans-
ferase gene and multiple dimethylamine methyltransferase genes of
Methanosarcina barkeri contain in-frame and read-through amber
codons. J Bacteriol 182:2520 –2529. https://doi.org/10.1128/jb.182.9
.2520-2529.2000.
18. Srinivasan G, James CM, Krzycki JA. 2002. Pyrrolysine encoded by UAG in
Archaea: charging of a UAG-decoding specialized tRNA. Science 296:
1459 –1462. https://doi.org/10.1126/science.1069588.
19. Borrel G, Gaci N, Peyret P, O’Toole PW, Gribaldo S, Brugère J-F. 2014. Unique
characteristics of the pyrrolysine system in the 7th order of methanogens:
implications for the evolution of a genetic code expansion cassette. Archaea
2014:374146. https://doi.org/10.1155/2014/374146.
20. Zhang Y, Gladyshev VN. 2007. High content of proteins containing 21st
and 22nd amino acids, selenocysteine and pyrrolysine, in a symbiotic
deltaproteobacterium of gutless worm Olavius algarvensis. Nucleic Acids
Res 35:4952– 4963. https://doi.org/10.1093/nar/gkm514.
21. Prat L, Heinemann IU, Aerni HR, Rinehart J, O’Donoghue P, Söll D. 2012.
Carbon source-dependent expansion of the genetic code in bacteria.
Proc Natl Acad SciUSA109:21070 –21075. https://doi.org/10.1073/pnas
.1218613110.
22. Gaston MA, Jiang R, Krzycki JA. 2011. Functional context, biosynthesis,
and genetic encoding of pyrrolysine. Curr Opin Microbiol 14:342–349.
https://doi.org/10.1016/j.mib.2011.04.001.
23. Laue H, Denger K, Cook AM. 1997. Taurine reduction in anaerobic
respiration of Bilophila wadsworthia RZATAU. Appl Environ Microbiol
63:2016 –2021. https://doi.org/10.1128/AEM.63.5.2016-2021.1997.
24. Baron EJ, Summanen P, Downes J, Roberts MC, Wexler H, Finegold SM.
1989. Bilophila wadsworthia, gen. nov. and sp. nov., a unique gram-
negative anaerobic rod recovered from appendicitis specimens and
human faeces. J Gen Microbiol 135:3405–3411. https://doi.org/10.1099/
00221287-135-12-3405.
25. Peck SC, Denger K, Burrichter A, Irwin SM, Balskus EP, Schleheck D. 2019.
A glycyl radical enzyme enables hydrogen sulfide production by the
human intestinal bacterium Bilophila wadsworthia. Proc Natl Acad Sci
U S A 116:3171–3176. https://doi.org/10.1073/pnas.1815661116.
26. Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T,
Watanabe H, Masuda K, Nishimoto Y, Kubo M, Hosoda F, Rokutan H,
Matsumoto M, Takamaru H, Yamada M, Matsuda T, Iwasaki M, Yamaji T,
Yachida T, Soga T, Kurokawa K, Toyoda A, Ogura Y, Hayashi T, Hat-
akeyama M, Nakagama H, Saito Y, Fukuda S, Shibata T, Yamada T. 2019.
Metagenomic and metabolomic analyses reveal distinct stage-specific
phenotypes of the gut microbiota in colorectal cancer. Nat Med 25:
968 –976. https://doi.org/10.1038/s41591-019-0458-7.
27. Feng Z, Long W, Hao B, Ding D, Ma X, Zhao L, Pang X. 2017. A human
stool-derived Bilophila wadsworthia strain caused systemic inflamma-
tion in specific-pathogen-free mice. Gut Pathog 9:59. [CrossRef] https://
doi.org/10.1186/s13099-017-0208-7.
28. Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A,
Antonopoulos DA, Jabri B, Chang EB. 2012. Dietary-fat-induced tauro-
cholic acid promotes pathobiont expansion and colitis in Il10/mice.
Nature 487:104 –108. https://doi.org/10.1038/nature11225.
29. Jie Z, Xia H, Zhong S-L, Feng Q, Li S, Liang S, Zhong H, Liu Z, Gao Y, Zhao
H, Zhang D, Su Z, Fang Z, Lan Z, Li J, Xiao L, Li J, Li R, Li X, Li F, Ren H,
Huang Y, Peng Y, Li G, Wen B, Dong B, Chen J-Y, Geng Q-S, Zhang Z-W,
Yang H, Wang J, Wang J, Zhang X, Madsen L, Brix S, Ning G, Xu X, Liu X,
Hou Y, Jia H, He K, Kristiansen K. 2017. The gut microbiome in athero-
sclerotic cardiovascular disease. Nat Commun 8:845. https://doi.org/10
.1038/s41467-017-00900-1.
30. Nava GM, Carbonero F, Croix JA, Greenberg E, Gaskins HR. 2012. Abun-
dance and diversity of mucosa-associated hydrogenotrophic microbes
in the healthy human colon. ISME J 6:57–70. https://doi.org/10.1038/
ismej.2011.90.
31. Craciun S, Balskus EP. 2012. Microbial conversion of choline to trimeth-
ylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A
109:21307–21312. https://doi.org/10.1073/pnas.1215689109.
32. Andresen PA, Kaasen I, Styrvold OB, Boulnois G, Strøm AR. 1988. Molec-
ular cloning, physical mapping and expression of the bet genes gov-
erning the osmoregulatory choline-glycine betaine pathway of Esche-
richia coli. J Gen Microbiol 134:1737–1746. https://doi.org/10.1099/
00221287-134-6-1737.
33. Lindstedt G, Lindstedt S. 1961. On the biosynthesis and degradation of
carnitine. Biochem Biophys Res Commun 6:319 –323. https://doi.org/10
.1016/0006-291x(61)90137-1.
Kivenson and Giovannoni
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 6
34. Andreesen JR. 1994. Glycine metabolism in anaerobes. Antonie Van
Leeuwenhoek 66:223–237. https://doi.org/10.1007/BF00871641.
35. Abu-Ali GS, Mehta RS, Lloyd-Price J, Mallick H, Branck T, Ivey KL, Drew
DA, DuLong C, Rimm E, Izard J, Chan AT, Huttenhower C. 2018. Meta-
transcriptome of human faecal microbial communities in a cohort of
adult men. Nat Microbiol 3:356 –366. https://doi.org/10.1038/s41564-017
-0084-4.
36. Natividad JM, Lamas B, Pham HP, Michel M-L, Rainteau D, Bridonneau C,
da Costa G, van Hylckama Vlieg J, Sovran B, Chamignon C, Planchais J,
Richard ML, Langella P, Veiga P, Sokol H. 2018. Bilophila wadsworthia
aggravates high fat diet induced metabolic dysfunctions in mice. Nat
Commun 9:2802. https://doi.org/10.1038/s41467-018-05249-7.
37. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE,
Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ,
Turnbaugh PJ. 2014. Diet rapidly and reproducibly alters the human gut
microbiome. Nature 505:559 –563. https://doi.org/10.1038/nature12820.
38. Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, Wagner MA,
Bennett BJ, Li L, DiDonato JA, Lusis AJ, Hazen SL. 2015. Transmission of
atherosclerosis susceptibility with gut microbial transplantation. J Biol
Chem 290:5647–5660. https://doi.org/10.1074/jbc.M114.618249.
39. Human Microbiome Jumpstart Reference Strains Consortium, Nelson KE,
Weinstock GM, Highlander SK, Worley KC, Creasy HH, Wortman JR, Rusch
DB, Mitreva M, Sodergren E, Chinwalla AT, Feldgarden M, Gevers D, Haas
BJ, Madupu R, Ward DV, Birren BW, Gibbs RA, Methe B, Petrosino JF,
Strausberg RL, Sutton GG, White OR, Wilson RK, Durkin S, Giglio MG,
Gujja S, Howarth C, Kodira CD, Kyrpides N, Mehta T, Muzny DM, Pearson
M, Pepin K, Pati A, Qin X, Yandava C, Zeng Q, Zhang L, Berlin AM, Chen
L, Hepburn TA, Johnson J, McCorrison J, Miller J, Minx P, Nusbaum C,
Russ C, Sykes SM, Tomlinson CM, Young S, et al. 2010. A catalog of
reference genomes from the human microbiome. Science 328:994 –999.
https://doi.org/10.1126/science.1183605.
40. Tatusova T, Ciufo S, Fedorov B, O’Neill K, Tolstoy I. 2014. RefSeq microbial
genomes database: new representation and annotation strategy. Nu-
cleic Acids Res 42:D553–D559. https://doi.org/10.1093/nar/gkt1274.
41. Reference deleted.
42. Hyatt D, Chen G-L, LoCascio PF, Land ML, Larimer FW, Hauser LJ. 2010.
Prodigal: prokaryotic gene recognition and translation initiation site
identification. BMC Bioinformatics 11:119. https://doi.org/10.1186/1471
-2105-11-119.
43. Finn RD, Clements J, Eddy SR. 2011. HMMER web server: interactive
sequence similarity searching. Nucleic Acids Res 39:W29 –W37. https://
doi.org/10.1093/nar/gkr367.
44. Bateman A, Coin L, Durbin R, Finn RD, Hollich V, GriffithsJones S, Khanna
A, Marshall M, Moxon S, Sonnhammer EL, Studholme DJ, Yeats C, Eddy
SR. 2004. The Pfam protein families database. Nucleic Acids Res 32:
D138 –D141. https://doi.org/10.1093/nar/gkh121.
45. Haft DH, Selengut JD, White O. 2003. The TIGRFAMs database of protein
families. Nucleic Acids Res 31:371–373. https://doi.org/10.1093/nar/
gkg128.
46. Zhang Y, Gladyshev VN. 2005. An algorithm for identification of bacterial
selenocysteine insertion sequence elements and selenoprotein genes.
Bioinformatics 21:2580 –2589. https://doi.org/10.1093/bioinformatics/
bti400.
47. Edgar RC. 2004. MUSCLE: multiple sequence alignment with high accu-
racy and high throughput. Nucleic Acids Res 32:1792–1797. https://doi
.org/10.1093/nar/gkh340.
48. Laslett D, Canback B. 2004. ARAGORN, a program to detect tRNA genes
and tmRNA genes in nucleotide sequences. Nucleic Acids Res 32:11–16.
https://doi.org/10.1093/nar/gkh152.
49. Lowe TM, Eddy SR. 1997. tRNAscan-SE: a program for improved detec-
tion of transfer RNA genes in genomic sequence. Nucleic Acids Res
25:955–964. https://doi.org/10.1093/nar/25.5.955.
50. Langmead B, Salzberg SL. 2012. Fast gapped-read alignment with Bow-
tie 2. Nat Methods 9:357–359. https://doi.org/10.1038/nmeth.1923.
51. Li D, Liu C-M, Luo R, Sadakane K, Lam T-W. 2015. MEGAHIT: an ultra-fast
single-node solution for large and complex metagenomics assembly via
succinct de Bruijn graph. Bioinformatics 31:1674 –1676. https://doi.org/
10.1093/bioinformatics/btv033.
52. Rho M, Tang H, Ye Y. 2010. FragGeneScan: predicting genes in short and
error-prone reads. Nucleic Acids Res 38:e191. https://doi.org/10.1093/
nar/gkq747.
53. Towns J, Cockerill T, Dahan M, Foster I, Gaither K, Grimshaw A, Hazle-
wood V, Lathrop S, Lifka D, Peterson GD, Roskies R, Scott JR, Wilkins-
Diehr N. 2014. XSEDE: accelerating scientific discovery. Comput Sci Eng
16:62–74. https://doi.org/10.1109/MCSE.2014.80.
54. Nystrom NA, Levine MJ, Roskies RZ, Scott JR. 2015. Bridges: a uniquely
flexible HPC resource for new communities and data analytics, p 1– 8. In
Proceedings of the 2015 XSEDE Conference: Scientific Advancements
Enabled by Enhanced Cyberinfrastructure. Association for Computing
Machinery, New York, NY.
TMA Metabolism in Human Gut Bacteria
September/October 2020 Volume 5 Issue 5 e00413-20 msystems.asm.org 7
... Within the MAGICdb, the proatherogenic and non-TMA gene richness was nearly equivalent (1,597 and 1,434, respectively) with cutC (choline TMA lyase) and mtxB (non-pyrrolysine methyltransferase) being the most dominant types sampled, respectively (Fig. 3C). Considering the unique genes only, MAGICdb sampled up to 12-fold more genes compared to prior reports (22,24,(34)(35)(36)(37). This expansion of MA gene diversity was attributed to the vast number of genomes collected from nonreference-based gut metagenome samples, rather than only relying on genomes from cultivated microorgan isms like most prior analyses. ...
... Forty percent of these sequen ces were in cluster 1, which was composed exclusively of pyrrolysine-containing genes for directly utilizing TMA (mttB). Prior knowledge of this TMA-utilizing metabolism was limited to a study focused on available genomes within the Bilophila and a study of six draft methanogen genomes (24,36). In comparison, MAGICdb contains 1,071 mttB genes assigned to Bilophila from multiple species and 61 methanogen genomes that span 3 genera, including one uncultivated genus UBA71 (Fig. 3A). ...
Article
Full-text available
Despite the promise of the gut microbiome to predict human health, few studies expose the molecular-scale processes underpinning such forecasts. We mined over 200,000 gut-derived genomes from cultivated and uncultivated microbial lineages to inventory the gut microorganisms and their gene content that control trimethylamine-induced cardiovascular disease. We assigned an atherosclerotic profile to the 6,341 microbial genomes that encoded metabolisms associated with heart disease, creating the Methylated Amine Gene Inventory of Catabolism database (MAGICdb). From microbiome gene expression data sets, we demonstrate that MAGICdb enhanced the recovery of disease-relevant genes and identified the most active microorganisms, unveiling future therapeutic targets. From the feces of healthy and diseased subjects, we show that MAGICdb predicted cardiovascular disease status as effectively as traditional lipid blood tests. This functional microbiome catalog is a public, exploitable resource, designed to enable a new era of microbiota-based therapeutics and diagnostics. IMPORTANCE One of the most-cited examples of the gut microbiome modulating human disease is the microbial metabolism of quaternary amines from protein-rich foods. By-products of this microbial processing promote atherosclerotic heart disease, a leading cause of human mortality globally. Our research addresses current knowledge gaps in our understanding of this microbial metabolism by holistically inventorying the microorganisms and expressed genes catalyzing critical atherosclerosis-promoting and -ameliorating reactions in the human gut. This led to the creation of an open-access resource, the Methylated Amine Gene Inventory of Catabolism database, the first systematic inventory of gut methylated amine metabolism. More importantly, using this resource we deliver here, we show for the first time that these gut microbial genes can predict human disease, paving the way for microbiota-inspired diagnostics and interventions.
... The demethylation of TMA in the gut by methanogens has been suggested as a possible route to lower the body burden of TMAO 16 . Enzymes for TMA demethylation are encoded in the genomes of intestinal bacteria as well 17,18 . These developments have added additional impetus toward further understanding of the metabolism of TMA by methanogenic Archaea. ...
... The dimer is further stabilized by interactions provided by an N-terminal β-strand (β2, residues [17][18][19]. This β-strand lies near the twofold axis between the dimers and forms a two stranded βsheet with its twofold-related partner. ...
Article
Full-text available
The 22nd genetically encoded amino acid, pyrrolysine, plays a unique role in the key step in the growth of methanogens on mono-, di-, and tri-methylamines by activating the methyl group of these substrates for transfer to a corrinoid cofactor. Previous crystal structures of the Methanosarcina barkeri monomethylamine methyltransferase elucidated the structure of pyrrolysine and provide insight into its role in monomethylamine activation. Herein, we report the second structure of a pyrrolysine-containing protein, the M. barkeri trimethylamine methyltransferase MttB, and its structure bound to sulfite, a substrate analog of trimethylamine. We also report the structure of MttB in complex with its cognate corrinoid protein MttC, which specifically receives the methyl group from the pyrrolysine-activated trimethylamine substrate during methanogenesis. Together these structures provide key insights into the role of pyrrolysine in methyl group transfer from trimethylamine to the corrinoid cofactor in MttC. Structures of Methanosarcina barkeri trimethylamine methyltransferase (MttB) with its substrates reveal the role of pyrrolysine in methyl group transfer from trimethylamine to the corrinoid cofactor in MttC.
... Holdemania may exert important effects on metabolic status in PCOS patients through the bene cial in uence of dietary ber and PUFAs, but further studies are needed to prove this assumption. Although Bilophila has been shown to mitigate cardiovascular disease by metabolizing both trimethylamine and its precursors without the production of trimethylamine-N-oxide [39], no evidence has been reported for the positive effect of Bilophila on PCOS, and additional in-depth studies are needed to explore the underlying mechanism involved. ...
Preprint
Full-text available
Background Previous studies have reported an association between gut microbial dysbiosis and ovarian diseases, however, it is not clear whether a causal association exists. Methods Two-sample Mendelian randomization (MR) analysis was performed to genetically predict the causal effects of the gut microbiota on polycystic ovary syndrome (PCOS), premature ovarian failure (POF), ovarian endometriosis, and malignant and benign ovarian neoplasms. The inverse variance weighted (IVW) method was used as the primary statistical method. A series of sensitivity analyses, including weighted median, MR-Egger, simple mode, weighted mode methods, MR pleiotropy residual sum and outlier (MR-PRESSO) and leave-one-out analysis, were also conducted to assess the robustness of the MR analysis results. Reverse MR analysis was implemented to explore whether ovarian diseases have any causal impact on the bacterial genera. Additionally, the Cochran’s Q test was used to evaluate heterogeneity among instrumental variables. Results IVW analysis revealed that several bacteria were associated with decreased risk of PCOS, POF, ovarian endometriosis, and benign and malignant ovarian neoplasm. Moreover, several bacteria were the causes of increased risks for POF, ovarian endometriosis, and benign and malignant ovarian neoplasm, respectively. Reverse MR analysis did not reveal a significant causal effect of these ovarian diseases on the gut microbiota. These findings were robust according to extensive sensitivity analyses. Conclusion Our results provide genetic evidence to support the causal relationship between specific gut microbiota taxa and ovarian diseases; thus, the gut microbiota should be considered a preventative strategy for ovarian diseases.
... Bilophila has been shown to induce inflammation and metabolic disorders by restoring and transforming sulfites into hydrogen sulfide (David et al., 2014;Xing et al., 2019;Zhao et al., 2022). Kivenson V. el al found Bilophila have genomic signatures for genetic code expansion that could enable them to metabolize both trimethylamine (TMA) and its precursors without production of trimethylamine N-oxide (TMAO), which result in bile acid metabolism disorder (Kivenson and Giovannoni, 2020). Bilophila and levels of TBA and LDL increased simultaneously on 7th day and sustained throughout hyperuricemia stages, indicating that Bilophila may be involved in bile acid metabolism in hyperuricemia. ...
Article
Full-text available
Introduction The intricate interplay between gut microbiota and hyperuricemia remains a subject of growing interest. However, existing studies only provided snapshots of the gut microbiome at single time points, the temporal dynamics of gut microbiota alterations during hyperuricemia progression and the intricate interplay between the gut barrier and microbiota remain underexplored. Our investigation revealed compelling insights into the dynamic changes in both gut microbiota and intestinal barrier function throughout the course of hyperuricemia. Methods The hyperuricemia mice (HY) were given intragastric administration of adenine and potassium oxalate. Gut microbiota was analyzed by 16S rRNA sequencing at 3, 7, 14, and 21 days after the start of the modeling process. Intestinal permeability as well as LPS, TNF-α, and IL-1β levels were measured at 3, 7, 14, and 21 days. Results We discovered that shifts in microbial community composition occur prior to the onset of hyperuricemia, key bacterial Bacteroidaceae, Bacteroides, and Blautia exhibited reduced levels, potentially fueling microbial dysbiosis as the disease progresses. During the course of hyperuricemia, the dynamic fluctuations in both uric acid levels and intestinal barrier function was accompanied with the depletion of key beneficial bacteria, including Prevotellaceae, Muribaculum, Parabacteroides, Akkermansia, and Bacteroides, and coincided with an increase in pathogenic bacteria such as Oscillibacter and Ruminiclostridium. This microbial community shift likely contributed to elevated lipopolysaccharide (LPS) and pro-inflammatory cytokine levels, ultimately promoting metabolic inflammation. The decline of Burkholderiaceae and Parasutterella was inversely related to uric acid levels, Conversely, key families Ruminococcaceae, Family_XIII, genera Anaeroplasma exhibited positive correlations with uric acid levels. Akkermansiaceae and Bacteroidaceae demonstrating negative correlations, while LPS-containing microbiota such as Desulfovibrio and Enterorhabdus exhibited positive correlations with intestinal permeability. Conclusion In summary, this study offers a dynamic perspective on the complex interplay between gut microbiota, uric acid levels, and intestinal barrier function during hyperuricemia progression. Our study suggested that Ruminiclostridium, Bacteroides, Akkermansiaceae, Bilophila, Burkholderiaceae and Parasutterella were the key bacteria that play vital rols in the progress of hyperuricemia and compromised intestinal barrier, which provide a potential avenue for therapeutic interventions in hyperuricemia.
... Compared with the blank group, the abundance of Bilophila in the bee pollen SDF group increased. Bilophila was reported to be associated with the gut dysbiosis of pancolitis in patients with ulcerative colitis [53], which is also commonly found in the gut flora of healthy humans and may alleviate cardiovascular disease in the host [54]. Blautia and Bifidobacterium were wildly reported to have probiotic characteristics, such as biological transformation, the regulation of host health and metabolic syndrome alleviation [55,56]. ...
Article
Full-text available
In this study, soluble dietary fibers (SDFs) were extracted from rape bee pollen using four methods including acid extraction (AC), alkali extraction (AL), cellulase extraction (CL) and complex enzyme extraction (CE). The effects of different extraction methods on the structure of SDFs and in vitro fermentation characteristics were further investigated. The results showed that the four extraction methods significantly affected the monosaccharide composition molar ratio, molecular weight, surface microstructure and phenolic compounds content, but showed little effect on the typical functional groups and crystal structure. In addition, all SDFs decreased the Firmicutes/Bacteroidota ratio, promoted the growth of beneficial bacteria such as Bacteroides, Parabacteroides and Phascolarctobacterium, inhibited the growth of pathogenic bacteria such as Escherichia-Shigella, and increased the total short-chain fatty acids (SCFAs) concentrations by 1.63–2.45 times, suggesting that the bee pollen SDFs had a positive regulation on gut microbiota. Notably, the SDF obtained by CE exhibited the largest molecular weight, a relatively loose structure, higher extraction yield and phenolic compounds content and the highest SCFA concentration. Overall, our results indicated that CE was an appropriate extraction method of high-quality bee pollen SDF.
Article
Full-text available
Background Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. Methods FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2 . Results Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia , a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium . Metabolic changes in FGR offspring were associated with persistent gut microbial changes. Conclusion FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. Impact Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia , Bilophilia and Bifidobacteria . The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Article
Full-text available
ABSTRACT: Broccoli extract mainly contains polyphenols and glucosinolates (GSLs). GSLs can be hydrolyzed by gut microorganisms into isothiocyanates (ITCs) and other active substances. These substances have anticancer, antiinflammatory, antimicrobial, and atherosclerosis-reducing functions. In this study, a high concentration (2000 μmol/L GSLs and 24 μmol/L polyphenols) and a low concentration (83 μmol/L GSLs and 1 μmol/L polyphenols)of broccoli extract were prepared. Gut microorganisms from fresh human feces were cultured to simulate the gut environment in vitro. The GSL content decreased and the types and content of ITCs increased with broccoli extract hydrolysis through cyclic condensation and gas chromatography−mass spectrometry (GC-MS) analyses. Broccoli extract signifcantly increased probiotics and inhibited harmful bacteria through 16S rDNA sequencing. Based on phylum level analysis, Firmicutes and Lachnospiraceae increased signifcantly (P < 0.05). At the genus level, both high- and low-concentration groups signifcantly inhibited Escherichia and increased Bilophila and Alistipes (P < 0.05). The high-concentration group signifcantly increased Bifidobacterium (P < 0.05). The broccoli extract improved the richness of gut microorganisms and regulated their structure. The GSL hydrolysis was signifcantly correlated with Bilophila, Lachnospiraceae, Alistipes, Bifidobacterium, Escherichia, and Streptococcus (P < 0.05). These study fndings provide a theoretical foundation for further exploring a probiotic mechanism of broccoli extract in the intestine.
Article
Full-text available
In most cases of sporadic colorectal cancers, tumorigenesis is a multistep process, involving genomic alterations in parallel with morphologic changes. In addition, accumulating evidence suggests that the human gut microbiome is linked to the development of colorectal cancer. Here we performed fecal metagenomic and metabolomic studies on samples from a large cohort of 616 participants who underwent colonoscopy to assess taxonomic and functional characteristics of gut microbiota and metabolites. Microbiome and metabolome shifts were apparent in cases of multiple polypoid adenomas and intramucosal carcinomas, in addition to more advanced lesions. We found two distinct patterns of microbiome elevations. First, the relative abundance of Fusobacterium nucleatum spp. was significantly (P < 0.005) elevated continuously from intramucosal carcinoma to more advanced stages. Second, Atopobium parvulum and Actinomyces odontolyticus, which co-occurred in intramucosal carcinomas, were significantly (P < 0.005) increased only in multiple polypoid adenomas and/or intramucosal carcinomas. Metabolome analyses showed that branched-chain amino acids and phenylalanine were significantly (P < 0.005) increased in intramucosal carcinomas and bile acids, including deoxycholate, were significantly (P < 0.005) elevated in multiple polypoid adenomas and/or intramucosal carcinomas. We identified metagenomic and metabolomic markers to discriminate cases of intramucosal carcinoma from the healthy controls. Our large-cohort multi-omics data indicate that shifts in the microbiome and metabolome occur from the very early stages of the development of colorectal cancer, which is of possible etiological and diagnostic importance.
Article
Full-text available
Significance This paper describes a pathway for anaerobic bacterial metabolism of taurine (2-aminoethanesulfonate), an abundant substrate in the human intestinal microbiota, by the intestinal bacterium and opportunistic pathogen, Bilophila wadsworthia . This metabolism converts taurine to the toxic metabolite hydrogen sulfide (H 2 S), an activity associated with inflammatory bowel disease and colorectal cancer. A critical enzyme in this pathway is isethionate sulfite-lyase, a member of the glycyl radical enzyme family. This enzyme catalyzes a novel, radical-based C-S bond-cleavage reaction to convert isethionate (2-hydroxyethanesulfonate) to sulfite and acetaldehyde. This discovery improves our understanding of H 2 S production in the human body and may also offer new approaches for controlling intestinal H 2 S production and B. wadsworthia infections.
Article
Full-text available
The essential nutrient choline is metabolized by gut bacteria to the disease-associated metabolite trimethylamine (TMA). However, most of the choline obtained via the diet and present in the human body is incorporated into larger metabolites, including the lipid phosphatidylcholine (PC). Here, we report that many choline-utilizing gut microorganisms can hydrolyse PC using a phospholipase D (PLD) enzyme and further convert the released choline to TMA. Genetic and in vitro characterization of the PLD from Escherichia coli MS 200-1 showed this enzyme is essential for bacterial hydrolysis of PC and prefers this substrate. PLDs are also found in gut bacterial isolates that are unable to convert choline to TMA, suggesting that additional members of the gut microbiota may influence access to this substrate. Unexpectedly, this PLD is only distantly related to characterized PLDs from pathogenic bacteria, suggesting a distinct evolutionary history. Together, these results reveal a previously underappreciated role for gut microorganisms in phospholipid metabolism and a potential target for inhibiting TMA production. © 2018, The Author(s), under exclusive licence to Springer Nature Limited.
Article
Full-text available
Trimethylamine N-oxide (TMAO) is a molecule generated from choline, betaine, and carnitine via gut microbial metabolism. The plasma level of TMAO is determined by several factors including diet, gut microbial flora, drug administration and liver flavin monooxygenase activity. In humans, recent clinical studies evidence a positive correlation between elevated plasma levels of TMAO and an increased risk for major adverse cardiovascular events. A direct correlation between increased TMAO levels and neurological disorders has been also hypothesized. Several therapeutic strategies are being explored to reduce TMAO levels, including use of oral broad spectrum antibiotics, promoting the growth of bacteria that use TMAO as substrate and the development of target-specific molecules. Despite the accumulating evidence, it is questioned whether TMAO is the mediator of a bystander in the disease process. Thus, it is important to undertake studies to establish the role of TMAO in human health and disease. In this article, we reviewed dietary sources and metabolic pathways of TMAO, as well as screened the studies suggesting possible involvement of TMAO in the etiology of cardiovascular and neurological disorders, underlying the importance of TMAO mediating inflammatory processes. Finally, the potential utility of TMAO as therapeutic target is also analyzed.
Article
Full-text available
Dietary lipids favor the growth of the pathobiont Bilophila wadsworthia, but the relevance of this expansion in metabolic syndrome pathogenesis is poorly understood. Here, we showed that B. wadsworthia synergizes with high fat diet (HFD) to promote higher inflammation, intestinal barrier dysfunction and bile acid dysmetabolism, leading to higher glucose dysmetabolism and hepatic steatosis. Host-microbiota transcriptomics analysis reveal pathways, particularly butanoate metabolism, which may underlie the metabolic effects mediated by B. wadsworthia. Pharmacological suppression of B. wadsworthia-associated inflammation demonstrate the bacterium's intrinsic capacity to induce a negative impact on glycemic control and hepatic function. Administration of the probiotic Lactobacillus rhamnosus CNCM I-3690 limits B. wadsworthia-induced immune and metabolic impairment by limiting its expansion, reducing inflammation and reinforcing intestinal barrier. Our results suggest a new avenue for interventions against western diet-driven inflammatory and metabolic diseases.
Article
Full-text available
The gut microbiome is intimately related to human health, but it is not yet known which functional activities are driven by specific microorganisms' ecological configurations or transcription. We report a large-scale investigation of 372 human faecal metatranscriptomes and 929 metagenomes from a subset of 308 men in the Health Professionals Follow-Up Study. We identified a metatranscriptomic 'core' universally transcribed over time and across participants, often by different microorganisms. In contrast to the housekeeping functions enriched in this core, a 'variable' metatranscriptome included specialized pathways that were differentially expressed both across participants and among microorganisms. Finally, longitudinal metagenomic profiles allowed ecological interaction network reconstruction, which remained stable over the six-month timespan, as did strain tracking within and between participants. These results provide an initial characterization of human faecal microbial ecology into core, subject-specific, microorganism-specific and temporally variable transcription, and they differentiate metagenomically versus metatranscriptomically informative aspects of the human faecal microbiome.
Article
Full-text available
Background Bilophila wadsworthia is a major member of sulfidogenic bacteria in human gut, it was originally recovered from different clinical specimens of intra-abdominal infections and recently was reported potentially linked to different chronic metabolic disorders. However, there is still insufficient understanding on its detailed function and mechanism to date. Methods A B. wadsworthia strain was isolated from fresh feces of a latent autoimmune diabetes in adults patient and we investigated its pathogenicity by oral administration to specific-pathogen-free mice. Tissue samples and serum were collected after sacrifice. Stool samples were collected at different time points to profile the gut microbiota. Results Bilophila wadsworthia infection resulted in the reduction of body weight and fat mass, apparent hepatosplenomegaly and elevated serum inflammatory factors, including serum amyloid A and interleukin-6, while without significant change of the overall gut microbiota structure. Conclusions These results demonstrated that higher amount of B. wadsworthia caused systemic inflammatory response in SPF mice, which adds new evidence to the pathogenicity of this bacterium and implied its potential role to the chronic inflammation related metabolic diseases like diabetes.
Article
Full-text available
The gut microbiota has been linked to cardiovascular diseases. However, the composition and functional capacity of the gut microbiome in relation to cardiovascular diseases have not been systematically examined. Here, we perform a metagenome-wide association study on stools from 218 individuals with atherosclerotic cardiovascular disease (ACVD) and 187 healthy controls. The ACVD gut microbiome deviates from the healthy status by increased abundance of Enterobacteriaceae and Streptococcus spp. and, functionally, in the potential for metabolism or transport of several molecules important for cardiovascular health. Although drug treatment represents a confounding factor, ACVD status, and not current drug use, is the major distinguishing feature in this cohort. We identify common themes by comparison with gut microbiome data associated with other cardiometabolic diseases (obesity and type 2 diabetes), with liver cirrhosis, and rheumatoid arthritis. Our data represent a comprehensive resource for further investigations on the role of the gut microbiome in promoting or preventing ACVD as well as other related diseases.
Article
Microbial communities are associated with the human host, primarily in the intestinal tract, where they affect host metabolism and contribute to the pathogenesis of cardiovascular disease. The susceptibility to atherosclerosis and thrombosis can be transmitted via gut microbial transplantation in mouse models. Microbial-associated molecular patterns are sensed by host pattern recognition receptors and affect cardiovascular disease pathogenesis. Microbial metabolism of common dietary nutrients produces both anti-atherogenic and pro-atherogenic metabolites that engage distinct host receptor systems and affect cardiovascular disease pathogenesis. Plasma levels of the gut microbial metabolite trimethylamine-N-oxide are associated with incident development of cardiovascular disease and its adverse outcomes in humans. Bacterially derived short-chain fatty acids (acetate, propionate and butyrate) can engage host receptor systems and potentially affect cardiovascular pathogenesis. Bacterially derived secondary bile acids can modulate dietary fat absorption and signal through cell-surface and nuclear hormone receptors, potentially affecting cardiovascular disease pathogenesis. Gut microorganism-targeted therapeutic strategies hold promise for the prevention and/or treatment of cardiovascular disease.