October 10.1152/ajpgi.00261.2020 A novel taurine-respiring murine gut bacterium contributes to colonization resistance against enteropathogens Huimin Ye 10 11 12 4 9 Sabrina Borusak 10 11 12 2 9 Claudia Eberl 10 11 12 8 9 Buck T. Hanson 0 10 11 12 4 6 9 Benjamin Zwirzitz 0 1 10 11 12 4 6 9 Craig W. 10 11 12 9 Herbold 10 11 12 4 9 Petra Pjevac 10 11 12 4 7 9 Bela Hausmann 10 11 12 3 7 9 Bärbel Stecher 10 11 12 5 8 9 David Schleheck 10 11 12 2 9 Alexander Loy alexander.loy@univie.ac.at 10 11 12 4 7 9 Tulln 10 11 12 9 Austria 10 11 12 9 Austrian Competence Centre for Feed and Food Quality, Safety and Innovation FFoQSI GmbH Current address of BZ: Institute of Food Science, University of Natural Resources and Life Sciences Department of Biology, University of Konstanz , Konstanz , Germany Department of Laboratory Medicine, Medical University of Vienna , Vienna , Austria Division of Microbial Ecology, Centre for Microbiology and Environmental Systems Science German Center for Infection Research (DZIF), partner site Ludwig Maximilian University Munich Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna Max-von-Pettenkofer Institute, Ludwig Maximilian University Munich , Munich , Germany Medicine , Vienna , Austria Munich , Germany University of Vienna , Vienna , Austria Vienna , Austria 2022 5 2022 3 44

#SB and CE contributed equally to this work.

-

Taurine-respiring gut bacteria produce H2S with ambivalent impact on host health. We report the isolation and genomic-ecophysiological characterization of the first taurine-respiring mouse gut bacterium. Taurinivorans muris represents a new widespread species with protective capacity against pathogens and differs from the human gut sulfidogen Bilophila wadsworthia in its sulfur metabolism and host distribution. Despite alternative physiologies, taurine respiration was the main in vivo lifestyle of T. muris independent of mouse diet and genotype. In gnotobiotic mice, T. muris selectively enhanced the activity of a sulfur metabolism gene-encoding prophage and provided slightly increased colonization resistance against Salmonella Typhimurium, which showed reduced expression of galactonate catabolism genes. We identified T. muris as the dominant sulfidogen of a mouse microbiota that conferred H2S-mediated protection against Klebsiella pneumoniae in a previous study. Together, we revealed the realized physiological niche of a key murine gut sulfidogen and its impact on pathogen and phage gene expression.

One sentence summary

Our work identified and characterized a new core member of the murine gut microbiota, revealed sulfidogenic taurine respiration as its predominant in vivo lifestyle, and emphasizes its protective function in pathogen colonization. metabolic gene, virus intestinal microbiome, synthetic microbiota, sulfur metabolism, taurine, Salmonella, Klebsiella, auxiliary 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69

Introduction

Hydrogen sulfide (H2S) is an intestinal metabolite with pleiotropic effects, particularly on the gut mucosa 1,2. H2S can have a detrimental impact on the intestinal epithelium by chemically disrupting the mucus barrier 3, causing DNA damage 4, and impairing energy generation in colonocytes through inhibition of cytochrome c oxidase and beta-oxidation of short-chain fatty acids 5,6. In contrast, low micromolar concentrations of H2S are anti-inflammatory and contribute to mucosal homeostasis and repair 7,8. Furthermore, H2S acts as a gaseous transmitter, a mitochondrial energy source, and an antioxidant in cellular redox processes, and thus its impact on mammalian physiology and health reaches beyond the gastrointestinal tract 9. For example, colonic luminal H2S can promote somatic pain in mice 10 and contribute to regulating blood pressure 11,12. The multiple (patho)physiological functions of H2S in various organs and tissues are dependent on its concentration and the health status of the host, but possibly also on the source of H2S. Mammalian cells can produce H2S from cysteine via several known pathways 13. In contrast to these endogenous sources, H2S-releasing drugs and H2S-producing intestinal microorganisms are considered exogenous sources. Compared to the colonic epithelium, sulfidogenic bacteria, which either metabolize organic sulfur compounds (e.g. cysteine) or anaerobically respire organic (e.g. taurine) or inorganic (e.g. sulfate, sulfite, tetrathionate) sulfur compounds in the gut, have a higher H2S-producing capacity and are thus potentially harmful to their hosts 1,2,14. Indeed, the abundance and activity of sulfidogenic gut bacteria were associated with intestinal diseases such as inflammatory bowel disease and colon cancer in various studies 15317 and many gut pathogens, such as Salmonella enterica and Chlostridioides difficile, are also sulfidogenic 18,19. Excess bacterial H2S production combined with a reduced capacity of the inflamed mucosa to metabolize H2S is one of many mechanisms by which the gut microbiome can contribute to disease 20. Yet, the manifold endogenous and microbial factors and processes that regulate intestinal H2S homeostasis are insufficiently understood. 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92

A major substrate of sulfidogenic bacteria in the gut is the organosulfonate taurine (2aminoethanesulfonate) that derives directly from meat- or seafood-rich diets and is also liberated by microbial bile salt hydrolases (BSHs) from endogenously produced taurocholic bile acids 21. Bilophila wadsworthia is the most prominent taurine-utilizing bacterium in the human gut. Diets that contain high quantities of meat, dairy products or fats can be associated with the outgrowth of B. wadsworthia in the gut 22,23. Consumption of high-fat food triggers taurocholic bile acid production and increases the taurine: glycine ratio in the bile acid pool 23. In mouse models, higher abundances of B. wadsworthia can promote colitis and systemic inflammation 23,24 and aggravate metabolic dysfunctions 25.

B. wadsworthia metabolizes taurine via the two intermediates sulfoacetaldehyde and isethionate (2hydroxyethanesulfonate) to sulfite 26 and the sulfite is utilized as electron acceptor for energy , conservation and reduced to H2S via the DsrAB-DsrC dissimilatory sulfite reductase system 27. The highly oxygen-sensitive isethionate sulfite-lyase system IslAB catalyzes the abstraction of sulfite (desulfonation) of isethionate 26,28. Alternative taurine degradation pathways in other bacteria involve direct desulfonation of sulfoacetaldehyde by the oxygen-insensitive, thiamine-diphosphate-dependent sulfoacetaldehyde acetyltransferase Xsc 29331. Xsc is employed in taurine utilization as carbon and energy source in a wide range of aerobic bacteria 30,32, as well as for anaerobic taurine fermentation by Desulfonispora thiosulfatigenes 31.

Here, we isolated the first taurine-respiring and H2S-producing bacterium from the murine intestinal tract and elucidated its fundamental and in vivo realized nutrient niche. Strain LT0009 represents a new Desulfovibrionaceae genus, termed Taurinivorans muris gen. nov., sp. nov., and differs from its human counterpart B. wadsworthia by using the Xsc pathway for taurine degradation and its distribution across 93 94 different animal hosts. We further provide insights into the protective role of this newly identified species of the core mouse microbiome against pathogen colonization. 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118

Materials and methods Supplementary Information provides further details on the methods described below. Isolation of strain LT0009 and growth experiments

Intestinal content of wild-type C57BL/6 mice was used as inoculum for the enrichment cultures. A modified Desulfovibrio medium was used for the isolation of strain LT0009 with taurine as electron acceptor and lactate and pyruvate as electron donors and for further growth experiments. Consumption of taurine and lactate and production of H2S and short-chain fatty acids (SCFA) were measured as previously reported 33.

Microscopy

Gram staining of the LT0009 isolate was performed u sing a Gram-staining kit according to the manufacturer9s instruction (Sigma Aldrich, 77730-1KT-F) and its cellular morphology was imaged with a scanning electron microscope (JSM-IT300, JOEL). A new probe was designed, tested, and applied for fluorescence in situ hybridization (FISH)-based microscopy of the genus Taurinivorans (Supplementary

Genome sequencing and comparative sequence analyses

The complete genome of strain LT0009 was determined by combined short- (Illumina) and long-read (Nanopore) sequencing. The automated annotation of the genome was manually curated for genes of interest, focusing on energy metabolism. Phylogenomic analyses comprised treeing with 43 concatenated marker protein sequences and calculation of average amino acid identities (AAI) and whole-genome average nucleotide identities (gANI). Additional phylogenetic trees were calculated with LT0009 using sequences of the 16S rRNA gene and selected sulfur metabolism proteins or genes. Source information of 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 16S rRNA gene reference sequences was manually compiled from the NCBI SRA entries (Supplementary as electron acceptor and lactate/pyruvate as electron donor were determined and compared.

Analyses of publicly available 16S rRNA sequence data

The occurrence and prevalence of Taurinivorans muris- and Bilophila wadsworthia-related 16S rRNA gene sequences were analyzed across 123,723 amplicon datasets from gut samples, including 81,501 with host information. Further information on mouse studies with at least 20 samples that were positive for B. wadsworthia was manually compiled from the NCBI SRA entries or the corresponding publications (Supplementary Table 3).

The identity of Desulfovibrionaceae 16S rRNA gene sequences from amplicon sequencing data of wildR mice, which showed increased representation of Deltaproteobacteria and colonization resistance against the enteropathogen Klebsiella pneumoniae in a previous study 34, was re-analyzed.

Gnotobiotic mouse experiments

The animal experiment was approved by the local authorities (Regierung von Oberbayern; ROB-55.22532.Vet_02-20-84). Twelve germ-free C57BL/6 mice w ere stably colonized with the 12-strain OligoMouse-Microbiota (OMM12) community 35. OMM12 mice (n=6) were orally (50 µl) and rectally (100 µl) inoculated with the LT0009 strain. The control OMM 12 mouse group (n=6) was treated with the same volume of sterile 1x phosphate-buffered saline. After 10 days, the mice were infected with the human enteric pathogen Salmonella enterica serovar Typhimurium (avirulent S. enterica Tm strain M2702; 5×107 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 c.f.u.) and sacrificed two days post infection (p. i.). Fecal microbiota composition was determined by strain-specific qPCR assays as previously described 35, including a newly developed assay for strain LT0009. Abundance of viable S. enterica Tm in feces and cecal content was determined by plating. Fecal samples of three mice from each group on day two p.i. were selected for metatranscriptome sequencing (JMF project JMF-2104-01) and analyses.

LT0009-centric gut metatranscriptome analyses of laboratory mice

Cecal and fecal metatranscriptomes from a high-glucose diet experiment in mice (HG study, Hanson et al., unpublished) (JMP project JMF-2101-5) were analyzed for LT0009 gene expression. Mouse experiments were conducted following protocols approved by Austrian law (BMWF-66.006/ 0032-WF/V/3b/2014). Additionally, mouse gut metatranscriptomes from a previous study were analyzed for LT0009 gene expression (Plin2 study) 36. Sequence data (PRJNA379425) derived from eight-we ek-old C57BL/6 wild-type and Perilipin2-null (Plin2) mice fed high-fat/low-carbohydrate or low-fat/high-carbohydrate diets.

Strain and data availability

Strain LT0009 has been deposited in the German Coll ection of Microorganisms and Cell Cultures (DSMZ) as DSM 111569 and the Japan Collection of Microorga nisms (JCM) as JCM 34262. The genome and the 16S rRNA gene sequence of T. muris LT0009 are available at NCBI GenBank under accessi on numbers CP065938 and MW258 658, respectively .

Sequencing data of the LT0009 pure culture transcri ptome (JMF-2012-1) and the mouse gut metatranscriptomes from the HG study (JMF-2101-05) and the gnotobiotic study (JMF-2104-01) were deposited to the Sequence Read Archive (SRA) under BioProject accession PRJNA867178. 165

Results and Discussion

Desulfovibrionaceae The first taurine-respiring bacterium isolated from the murine gut represents a new genus of the family Strain LT0009 was enriched from mouse gut contents (cecum and colon) using an anoxic, non-reducing, modified Desulfovibrio medium with L-lactate and pyruvate as electron donors (and carbon source) and taurine as the sulfite donor for sulfite respiration. Its isolation was achieved by several transfers in liquid medium, purification by streaking on ferric-iron supplemented agar plates, indicating sulfide production by black FeS formation and picking of black colonies, and by additional purification using dilution-toextinction in liquid medium. Strain LT0009 produced sulfide and acetate during taurine degradation. We sequenced and reconstructed the complete LT0009 gen ome, which has a size of 2.2 Mbp, a G+C content of 43.6%, and is free of contamination as assessed by CheckM. The genome comprises 2,059 proteincoding genes, 56 tRNA genes, 4 rRNA operons (with 5S, 16S, and 23S rRNA genes), 4 pseudogenes, and 6 miscellaneous RNA genes.

LT0009 formed a monophyletic, genus-level (>94.5% s imilarity) lineage with other 16S rRNA gene sequences from the gut of mice and other hosts. It has <92% 16S rRNA gene sequence identity to the closest related isolates Marseille-P3669 and Mailhella massiliensis Marseille-P3199 T, two strains isolated from human stool (Fig. 1a). Phylogenomic treeing and an AAI of <60% to other described species strongly suggested that LT0009 represents the type strain of a novel genus in the family Desulfovibrionaceae of the phylum Desulfobacterota 37 for which we propose the name Taurinivorans muris (Fig. 1b, Supplementary Fig. 2, Supplementary Information). The previously described mouse gut MAGs UBA8003 and extra-SRR4116659.59 have >98% ANI and AAI to LT 0009 and thus would also belong to the species T. muris 38340. Furthermore, the mouse gut MAGs extra-SRR4116662.45 and single-China-D-Fe10-120416.2 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 showed <80% AAI/ANI to strain LT0009 and 84% AAI an d 82% ANI to each other, which indicates that each of these two MAGs would likely represent a separate species in the novel genus Taurinivorans. Notably, the genome of T. muris LT0009 with only 2.2 Mbp in size is considerably sm aller than that of other freeliving bacteria of the Desulfovibrio-Bilophila-Lawsonia-Mailhella-lineage (Fig. 1b). Only the obligate intracellular intestinal pathogen Lawsonia intracellularis has a smaller genome at 1.5-1.7 Mbp 41,42. The Gram-staining of T. muris LT0009 was negative. FISH imaging of the LT0009 pu re culture with the newly-developed genus-specific 16S rRNA-targeted probe TAU1151 showed cells with a conspicuous spiral-shaped morphology and considerably varying lengths (1.7 to 28 µm) (Fig. 1c). SEM imaging furthe r indicated that LT0009 cells have multiple polar fla gella and are thus motile (Fig. 1c).

Complete utilization of taurine as electron acceptor in modified Desulfovibrio liquid medium with electron donors lactate and pyruvate in excess, resulted in production of nearly quantitative amounts of H2S (Fig. 1d). Strain LT0009 in pure culture did not grow in absence of 1,4-naphthoquinone and yeast extract, indicating an absolute requirement of menaquinone (vitamin K2) and other essential growth factors, respectively. Both growth rate and final growth yields were increased when taurine was provided at 20 and 40 mmol/l concentration in comparison to 10 mmol/l, while growth was inhibited at concentrations g60 mmol/l taurine (Supplementary Fig. 3a). Strain LT0009 grew with a lower growth rate and final grow th yield when pyruvate was omitted as additional electron donor (Supplementary Fig. 3b). Strain LT0009 grew equally well at a pH range of 6 to 8.5 (Supple mentary Fig. 3c) and temperatures between 27-32°C, but with reduced final growth yield at 37 and 42°C (Supplementary Fig. 3d). No colony formation was observed on agar plates under aerobic conditions, suggesting a strict anaerobic lifestyle of T. muris LT0009. 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236

Sulfur and energy metabolism of T. muris LT0009

Based on a genome-inferred metabolism prediction of strain LT0009 (Fig. 2a, Supplementary Table 6), we tested its growth with substrates that could serve as energy and sulfur sources in the gut. Fermentative growth with only pyruvate or only taurine, i.e., as both electron donor and acceptor, was not observed. In addition to pyruvate and lactate, LT0009 also us ed formate as electron donor for growth under taurinerespiring conditions, albeit with an extended lag phase and a lower growth yield. For the alternative electron acceptors tested, LT0009 used 3-sulfolacta te and thiosulfate in combination with lactate and pyruvate, but did not grow with 2,3-dihydroxypropane-1-sulfonate (DHPS), isethionate, cysteate, and not with inorganic sulfate or sulfite (Fig. 2b).

The metabolic pathways used for growth by respiration with taurine, sulfolactate or thiosulfate and with lactate/pyruvate as electron donors were further analyzed by differential transcriptomics and proteomics. This demonstrated that taurine is degraded via the Tpa-Xsc pathway and the produced sulfite respired via the DsrAB-DsrC pathway (Fig. 2c,d). Pyruvate-dependent taurine transaminase Tpa catalyzes initial conversion of taurine to alanine and sulfoacetaldehyde 31. Oxidative deamination of alanine to pyruvate is catalyzed by alanine dehydrogenase Ald (Fig. 2a,c,d). Lack of sarD and islAB and an inability to grow with isethionate showed that LT0009 does not have the ta urine degradation pathway of B. wadsworthia 26. Instead, sulfoacetaldehyde is directly desulfonated to acetyl-phosphate and sulfite by thiaminediphosphate-dependent sulfoacetaldehyde acetyltransferase Xsc 29331. The acetyl-phosphate is then converted to acetate and ATP by acetate kinase AckA. Strain LT0009 seems to lack candidate genes for the TauABC taurine transporter 43. While homologs of tauABC are encoded in the genome, the individual genes do not form a gene cluster like in Escherichia coli 44 and were not expressed during growth on taurine (Supplementary Table 7). Instead, the LT0009 genome encodes three copies of gene sets for tripartite ATP-independent periplasmic (TRAP) transporter 45 that are co-encoded in the taurine 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 degradation gene cluster and were expressed during growth with taurine, thus are most likely involved in taurine import, including DctPQM1 (with fused DctQM1) (TAUVO_v1_1026 and 1027) and DctPQM3 (TAUVO_v1_1467-1469) (Fig. 2c,d). (2S)-3-sulfolactate is degraded by LT0009 via the S lcC-ComC-SuyAB pathway as shown by differential expression of these enzymes in cells grown with racemic sulfolactate (Fig. 2c, d, Supplementary Table 7). The dehydrogenases (S)-sulfolactate dehydrogenase SlcC and (R)-sulfolactate oxidoreductase ComC isomerize (2S)-3-sulfolactate to (2R)-3-sulfolactate via 3-sulfopyruvate. (2R)-3-sulfolactate is desulfonated to pyruvate and sulfite by sulfo-lyase SuyAB. The neighboring gene clusters slcHFG-slcC-comC and hpsNdctPQM-suyAB were both significantly upregulated in the transcriptome of sulfolactate-grown cells (Fig. 2d). The DctPQM2 (with fused DctQM2) (TAUVO_v1_1434 and 1435) TRAP transporter and the SlcGFH tripartite tricarboxylate transporters (TTT) 45,46 are putative sulfolactate importers. The gene cluster further includes a homolog to hpsN, encoding sulfopropanediol-3-dehydrogenase 47. This enzyme converts (R)-DHPS to (R)-sulfolactate during aerobic catabolism of DHPS by diverse bacteria in soils 47 and the ocean 48. However, LT0009 did not grow with racemic DHPS wh en tested (Fig. 2b). The hpsN gene was transcribed in LT0009 with taurine, sulfolactate, a nd thiosulfate treatments, but the HpsN protein was not detected (Supplementary Table 7). LT0009 did not gr ow with cysteate as electron acceptor under the conditions we used, although it encodes a homolog of L-cysteate sulfo-lyase CuyA that desulfonates Lcysteate to pyruvate, ammonium, and sulfite 49,50 (Fig. 2b). The cuyA gene was transcribed in the presence of taurine, sulfolactate, and thiosulfate. Furthermore, CuyA was significantly higher expressed in LT0009 with taurine (P<0.001) compared with sulfolactate and thiosulfate (Supplementary Table 7), yet its physiological role in LT0009 remains unclear. 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282

Strain LT0009 respired thiosulfate, such as B. wadsworthia strain RZATAU 51, but lacks genes for PhsABC thiosulfate reductase 52 and thiosulfate reductase from Desulfovibrio (EC 1.8.2.5) 53, which both (i) disproportionate thiosulfate to sulfide and sulfite and (ii) are present in the human B. wadsworthia strains ATCC 49260, 4_1_30, and 3_1_6. LT0009 has a gene fo r a homolog of rhodanese-like, sulfur-trafficking protein SbdP (TAUVO_v1_1430) (Supplementary Fig. 4d) 54. Rhodaneses (EC 2.8.1.1.) can function as thiosulfate sulfurtransferase and produce sulfite 55. Homologs of SbdP are broadly distributed in members of the

Desulfovibrio-Bilophila-Mailhella-Taurinivorans

clade and other reduction and energy conservation by the Dsr sulfite reductase system and (ii) transfer the second sulfur atom from thiosulfate to an unknown acceptor protein/enzyme. A candidate sulfur-accepting protein is encoded by a dsrE-like gene in LT0009 (TAUVO_v1_1364) (Supplementary Fig. 4b). High expression of rhodanese-like sulfur transferases, a DsrE-like protein, and DsrAB sulfite reductase was reported for thiosulfate-respiring Desulfurella amilsii 56. However, the functions of the SbdP-sulfur transferase and the DsrE-like protein in the thiosulfate metabolism of LT0009 remain unconfirmed. First, these proteins ar e not homologous to the highly expressed D. amilsii proteins. Second, comparative transcriptome and proteome analyses were inconclusive as only the transcription of the dsrE-like gene was upregulated in

LT0009 grown with thiosulfate (Fig. 2d, Supplementa ry Table 7).

Additional genes homologous to known sulfur metabolism genes whose functions in LT0009 remain enigmatic include sudAB, which encode sulfide dehydrogenase for reduction of sulfur or polysulfide to H2S 57, and dsrEFH, which are involved in sulfur atom transfer in sulfur oxidizers (Supplementary Fig. 4c) 58. 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306

LT0009 encodes an incomplete pathway for dissimilat ory sulfate reduction. While homologs of genes for the sulfate transporter SulP 59 and adenylyl-sulfate reductase AprAB were present, absence of genes for sulfate adenylyltransferase Sat and the electron-transferring QmoAB complex (Supplementary Table 6) was consistent with the inability of LT0009 to resp ire sulfate. Although externally supplied sulfite did not support growth, the DsrAB-DsrC dissimilatory sulfite reductase system was highly expressed in cells grown with taurine, sulfolactate, or thiosulfate (Fig. 2a, d, Supplementary Table 7). This suggests that intracellularly produced sulfite is respired to sulfide via the DsrAB-DsrC system, which includes transfer of electrons from the oxidation of electron donors via the membrane quinone pool and the DsrMKJOP complex (Fig. 2a) 60.

Genome reconstruction of LT0009 suggested the poten tial to utilize lactate, pyruvate, and H2 as electron donors (Supplementary Information). We experimentally confirmed that lactate and pyruvate but also formate are used as electron donors for taurine respiration (Fig. 2b).

Notably, T. muris employs similar electron acceptors and donors as B. wadsworthia, yet they differ in the metabolic pathways to use them.

Distinct distribution patterns of Taurinivorans muris and Bilophila wadsworthia suggest different host preferences T. muris is the first taurine-utilizing, sulfidogenic isolate from the mouse gut. B. wadsworthia was repeatedly reported as a taurine-degrading member of the murine intestine based on molecular surveys 23,25,61. We performed a meta-analysis to compare the presence and relative abundance of B. wadsworthia-related and T. muris-related sequences across thousands of 16S rRNA gene amplicon datasets from the intestinal tract of diverse hosts. T. muris-related 16S rRNA gene sequences were most 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 often detected in the mouse gut, i.e. in 14.4% of all mouse amplicon datasets, but also present in the datasets from multiple other hosts (shrimp, pig, rat, chicken, fish, cow, humans, termites, and other insects) (Fig. 3a, Supplementary Table 8). In compa rison, B. wadsworthia-related sequences are most widespread in the human gut, i.e. in 30.7% of human gut amplicon datasets, but are also prevalent in pig (15.8%), chicken (13.7%), and rat (9.8%) and occasi onally detected in other hosts (Fig. 3a). We also identified B. wadsworthia-related sequences in 7.5% of mouse datasets. Notably, T. muris- and B. wadsworthia-related sequences co-occurred only in 28 mouse data sets, which suggests competitive exclusion possibly due to competition for taurine. Furthermore, we found that 82% of the B. wadsworthiapositive samples are from mice that were 8humanized9 by receiving human feces transplants or human strain consortia 62365, which suggests a much lower prevalence of B. wadsworthia strains that are indigenous in mice. T. muris-related sequences represented >5% of the total community in 2.8% of mouse gut datasets (Fig. 3a). Such very high relative 16S rRNA gene abundances were more often observed in mice on high-fat diets 66,67, but sporadically also in mice on standard chow and other diets (Fig. 3b) 68. Overall, T. muris is considerably more abundant and prevalent in the mouse gut microbiome than B. wadsworthia. Notably, a mouse native B. wadsworthia strain has not yet been isolated. Our phylogenomic analysis of all de-replicated, high-quality Desulfovibrionaceae MAGs from the integrated mouse gut metagenome catalog (iMGMC) 69 revealed two MAGs form a well-supported monophyletic group with B. wadsworthia strains (Fig. 1b). Mouse MAG iMGMC-189 has a minim um ANI of 82% and AAI of 79% to B. wadsworthia, which suggests it represents the population genome of a new, murine Bilophila species. Mouse MAG extra-SRR7761328.52 is more distantly rel ated and has a minimum ANI of 78% and AAI of 65% to B. wadsworthia. Both mouse MAGs encode the taurine degradation pathway (tpa-sarD-islAB) of B. wadsworthia (Supplementary Fig. 2), while the pathway for sulfolactate degradation (slcC-comC-suyAB) is absent in MAG extra-SRR7761328.52 .

In general, genes for utilization of diverse organosulfonates are widely and patchily distributed in the Desulfovibrio-Bilophila-Mailhella-Taurinivorans clade (Supplementary Fig. 2) 33. Other mouse Desulfovibrionaceae that encode the capability for taurine respiration include (i) the Desulfovibrioaffiliated MAGs extra-SRR7533634.94 and iMGMC-585 w ith the tpa-xsc pathway and (ii) the Mailhellarelated MAG extra-SRR7691169.24 with the tpa-sarD-islAB pathway (Supplementary Fig. 2).

Taurine degradation is the main in vivo realized nutritional niche of Taurinivorans muris

We next performed metatranscriptome analysis and re-analyzed published metatranscriptome datasets of gut samples from different mouse models to reveal the metabolic pathways that are most expressed by T. muris in its murine host.

In our gnotobiotic model, strain LT0009 or a mock c ontrol were added to germ-free mice stably colonized with the synthetic OMM12 community (Fig. 4a). Strain-specific qPCR assays showed that ten OMM12 strains and strain LT0009 colonized the mice (Fig. 4b). Con sistent with previous studies, strains A. muris KB18 and B. longum subsp. animalis YL2 were not detected 35,70. Colonization of LT0009 did not affect the abundan ce of other strains, which indicated that LT0009 occup ied a free niche in the intestinal tract of this gnotobiotic mouse model. The taurine metabolism (tpa, ald, xsc) and sulfite reduction (dsrAB, dsrC) genes were in the top 5% expression rank of all LT0009 genes (Fig. 4c ). In contrast, gene expression of the putative thiosulfate transferase (sbdP) ranked at 17% and of sulfolactate degradation (suyAB, slcC, comC) ranked from 62% to 88% of all LT0009 genes (Fig. 4c). T. muris LT0009 thus largely occupied the vacant taurinenutrient niche in the intestinal tract of OMM12 mice. 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377

Metatranscriptome analysis of intestinal samples from conventional laboratory mice on various diets (e.g. high-glucose; high-fat/low-carbohydrate; low-fat/high-carbohydrate) and with different genetic backgrounds (wildtype; plin2) also showed that taurine degradation and sulfite respiration were within the top 5% of expressed LT0009 genes (Supplementary Fig.5). Collectively, this demonstrated that taurine is the predominant electron acceptor for energy conservation of T. muris in the murine intestinal tract. Free taurine in the murine intestine largely derives from microbial deconjugation of host-derived taurocholic bile acids 71,72. LT0009 does not encode genes for bile salt hydrol ase (BSH) and is thus likely dependent on other gut microbiota members for liberation of taurine from taurocholic bile acids. BSH genes are encoded across diverse bacterial taxa in the human and mouse gut 71,73,74. In agreement with previous studies of bile acid transformations in the OMM12 model 75,76, we identified BSH genes in seven OMM12 strains (Supplementary Table 5). The expression of BSH genes in these strains did not change significantly with the presence of LT0009. Yet, BSH gene transcription increased in E. clostridioformis YL32, E. faecalis KB1, B. caecimuris I48, and M. intestinale YL27, and decreased in L. reuteri I49 (Supplementary Table 5). Bile acid deconjugation by some of these OMM12 strains has been confirmed in vitro 76. Specifically, B. caecimuris I48, B. animalis YL2, E. faecalis KB1, and M. intestinale YL27 were tested positive for deconjugation of taurine-conjugated deoxycholic acid, while E. clostridioformis YL32 was either tested negative or inhibited by addition of the bile acids. The down-regulation of the BSH gene in L. reuteri I49 is consistent with the in vitro deconjugation capacity of this strain for glycine-conjugated deoxycholic acid but not for taurine-conjugated deoxycholic acid 76 and the generally lower abundance of glycineconjugated bile acids in rodents 72. We hypothesize that taurine degradation by LT0009 could provide a selective feedback mechanism on expression of BSHs for taurocholic bile acid deconjugation in the OMM12 model. 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401

Thiosulfate is presumably a constantly present electron acceptor for microbial respiration in the mammalian gut as it is generated by mitochondrial H2S oxidation in the gut epithelium 1. The sulfide oxidation pathway is mainly located apically in the crypts of human colonic tissue at the interface to the gut microbiota 77. The amount of thiosulfate supplied into the gut lumen will depend on epithelial H2S metabolism 1,77. The expression level of the putative SbdP thiosulfate transferase of T. muris ranked relatively high with 15-24% of all LT0009 genes acr oss all mouse gut samples (Fig. 4c, Supplementary Fig. 5). However, the function of this protein remains unconfirmed as its expression was not differentially upregulated in the thiosulfate-metabolizing T. muris pure culture (Fig. 2a,d, Supplementary Table 7). In vivo taurine respiration, and potentially thiosulfate respiration, are likely fueled by pyruvate, H2, and lactate as electron donors, as expression of genes for their oxidation ranked at 5-7% (por), 1.5-9.1% (hybAC), and 2.5-31% (lutABC, lutP) of all LT0009 genes across all mouse gut metatran scriptomes, respectively (Fig. 4c, Supplementary Fig. 5).

Taurinivorans muris LT0009 slightly increased colonization resistance against S. enterica and activated a sulfur metabolism gene-encoding prophage in a gnotobiotic mouse model The human enteropathogen S. enterica Tm can invade and colonize the intestinal tract by utilizing various substrates for respiratory growth that are available at different infection stages 78. The gnotobiotic OMM12 mouse model provides intermediate colonization resistance against S. enterica Tm 35 and is widely used as a model system of modifiable strain composition for investigating causal involvement of cultivated mouse microbiota members in diverse host diseases and phenotypes 79. Yet, a bacterial isolate from the mouse gut with proven dissimilatory sulfidogenic capacity was not available until now 80. T. muris has fundamental physiological features that could on the one hand contribute to colonization resistance against S. enterica Tm by direct competition for pyruvate 81, lactate 82, H2 83, formate 84, and host-derived 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 thiosulfate 84,85. On the other hand, T. muris could also promote S. enterica Tm expansion during inflammation by fueling tetrathionate production through enhanced intestinal sulfur metabolism 18,86. Furthermore, expansion of sulfidogenic Deltaproteobacteria commensals and the tpa-xsc-dsr pathway in the metagenome fueled by host-derived taurine was shown to increase protection against the enteropathogen Klebsiella pneumoniae in mice; with sulfide-mediated inhibition of aerobic respiration by pathogens being proposed as a generic protective mechanism 34. Notably, our re-analysis of the 16S rRNA gene amplicon data from the wildR mouse model of this study identified T. muris as the dominant deltaproteobacterium (Desulfobacterota) of the protective community (Supplementary Fig. 6). Given that taurine respiration via the sulfidogenic tpa-xsc-dsr pathway is the main energy niche of T. muris in the mouse gut (Fig. 4c, Supplementary Fig. 5), enhanced resistance in the wildR mouse model was thus likely primarily due to the activity of T. muris.

Here, we investigated the impact of T. muris LT0009 during the initial niche invasion of S. enterica Tm using an avirulent, non-colitogenic strain 87. Compared to OMM12 mice without LT0009, mice colonized with the OMM12 and LT0009 had a slightly reduced load of S. enterica Tm at 48 p.i. that was significant in feces but not in cecum (Fig. 4d). Comparative metatranscriptome analysis did not provide evidence for a mechanism of direct interaction as only six S. enterica Tm genes were differentially expressed, i.e. significantly downregulated, in the presence of strain LT0009 (Fig. 4e, Supplementary Table 9). Three of the six genes are involved in transport and metabolism of galactonate (D-galactonate transporter DgoT, D-galactonate dehydratase DgoD, 2-dehydro-3-deoxy-6-phosphogalactonate aldolase DgoA), which is produced by some bacteria as an intermediate in D-galactose metabolism and is also present in mammalian tissue and body secretions 88. D-galactonate catabolism capability was suggested as a distinguishing genetic feature of intestinal Salmonella strains compared to extraintestinal serovars, with serovars Typhi, Paratyphi A, Agona, and Infantis lacking genes for utilizing D-galactonate as a sole carbon 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 source 89. The putative D-galactonate transporter DgoT in Salmonella enterica serovar Choleraesuis was identified as a virulence determinant in pigs 90. The OMM12 strains and LT0009 do not encode the DgoTDAKR galactonate pathway. The significance of galactonate for S. enterica Tm gut colonization and competition remains to be elucidated.

Colonization of T. muris LT0009 in the gnotobiotic mice had variable impact on the differential gene expression pattern of the OMM12 members (Fig. 4e, Supplementary Fig. 7 and Table 9 ). While gene expression was not significantly affected in L. reuteri I49, E. clostridioformis YL32 was most affected with 84 differentially expressed genes (55 up-regulated and 29 down-regulated) (Fig. 4e). Most of the significantly up-regulated genes (n=41) in E. clostridioformis YL32 are clustered in a large genomic region (I5Q83_10075-10390) that encoded various phage gene homologs and was identified as a prophage using PHASTER (Fig. 4f). This prophage, named YL32-pp-2.059, Saumur, is among a set of thirteen previously identified prophages of the OMM12 consortium that represent novel viruses, were induced under various in vitro and/or in vivo conditions, and constitute the temporally stable viral community of OMM12 mice 91. We show that colonization of LT0009 in the OMM 12 mouse model selectively enhanced the transcriptional activity of the E. clostridioformis YL32 prophage Saumur, which carries a gene (I5Q83_1 0375) for phosphoadenosine-phosphosulfate reductase (CysH) that functions in the assimilatory sulfate reduction pathway of many bacteria (Fig. 4f). Various organosulfur auxiliary metabolic genes, particularly cysH, are widespread in environmental and human-associated viromes, which suggests viruses augment sulfur metabolic processes in these environments, including the gut 92. Addition of physiologically relevant concentrations of sulfide to a Lactococcus lactis strain culture resulted in increased production of viable particles of its phage P087 92. We thus hypothesize that T. muris LT0009 not only impacts intestinal sulfur homeostasis via its sulfur metabolism but also by H2S-mediated activation of the sulfur metabolism geneexpressing phage Saumur in E. clostridioformis YL32. If activation of prophage Saumur contributes to protection from S. enterica Tm remains subject of further study. 495 represents the new genus/species Taurinivorans muris in the family Desulfovibrionaceae. a. 16S rRNA gene tree and FISH probe coverage. Maximum likelihood branch supports (1000 resamplings) equal to or greater than 95% and 80% are indicated by black and grey circles, respectively. The scale bar indicates 0.1 estimated substitutions per residue. Accession numbers are shown in parentheses. Strain LT0009 is show n in bold and the type strains are marked with a superscript 8T9. The sequence sources are indicated with different colors (Supplementary Table 2). Sequences were assigned to Taurinivorans and Taurinivorans muris based on the genus-level similarity cutoff of 94.5 % and species-level similarity cutoff of 98.7% 96, respectively. The perfect-match coverage of probes TAU1151 for Taurinivorans and MAIL1151 for Mailhella is indicated. b. Phylogenomic tree. Ultrafast bootstrap support values equal to or greater than 95% and 80% for the maximum likelihood tree are ind icated with black and grey circles, respectively. Accession numbers are shown in parentheses. Strain LT0009 is shown in bold. Strains with complete genomes (genome size is indicated) are marked with a star. Genomes were assigned to Taurinivorans based on the genus-level AAI cut-off value of 63.4% 97. The scale bar indicates 0.1 estimated substitutions per residue. c. Morphology of LT0009 cells in pure culture. SEM: S canning electron microscopy images of cells of varying lengths. White arrows indicate the flagella. FISH: Cells hybridized with Cy3-labeled probe TAU1151 and Fluos-labeled probe EUB338mix and counterstained by DAPI. d. Growth of strain LT0009 in modified Desulfovibrio medium confirmed complete utilization of taurine as electron acceptor concomitant with nearly stoichiometric production of sulfide. Electron donors L-lactate and pyruvate were provided in excess, and their utilization also contributed to acetate formation. Pyruvate in the medium and ammonia released from deamination of taurine were not quantified in this 518 519 experiment. Lines represent averages of measures in triplicate cultures. Error bars represent one standard deviation. a. Cell cartoon of the central sulfur and energy metabolism of LT0009 as determined by genome, transcriptome, and proteome analyses. Genes/proteins detected in the transcriptome and proteome of LT0009 grown with taurine, sulfolactate or thiosulf ate as electron acceptor are shown by colored circles and squares, respectively. Circle size indicates gene transcription level normalized as TPM. Proteins of all transcribed genes that are shown were also detected in the proteome, with the exception of AprAB, TauA (TAU_v1_0027, TAU_v1_1344), TauB, TauC, TauE, DctMQ2, SlcG, DsrEFH, AscA, two [FeFe] hydrogenases (TAU_v1_1126, TAU_v1_1901), cytochrome c and Sdh. Protein complexes (e.g. Rnf, SlcFGH, DctPMQ2, Atp) are not shown with transcriptome and proteome data because one or more of the complex units were not expressed. The gene annotations are listed in Supplementary Table 6. b. Anaerobic growth tests of strain LT0009 with various substrates. Upper pan el: All substrates were added at 10 mmol/l concentration, except acetate (20 mmol/l), which was added as carbon source together with H2. Lower panel: The different sulfur compounds were added at 10 mmol/l concentration together with pyruvate, lactate, and 1,4-naphthoquinone. OD600: optical density at 600 nm. c. Organization of sulfur metabolism genes in the LT0009 genome. Numbers show the RefSeq locus tag with the prefix TAUVO_v1. d. Comparative transcriptome and proteome analysis of LT0009 grown with lactate and taurine, sulfolactate or thiosulfate as electron acceptor; each in triplicate culture. Numbers following protein names refer to RefSeq locus tag numbers (prefix TAUVO_v1). Protein expression was normalized to DsrC for each growth condition. Bars represent averages of triplicate measures with error bars representing one standard deviation. Asterisk indicates significant (p<0.05) differences in gene transcription/protein expression 542 543 compared to growth with taurine. TCA, tricarboxylic acid cycle; WL, Wood-Ljungdahl pathway; PEP, phosphoenolpyruvate; DHPS, 2,3-dihydroxypropane-1-sulfonate; TPM, transcripts per million. a. Occurence and prevalence of Taurinivorans muris- and Bilophila wadsworthia-related sequences in 16S rRNA gene amplicon datasets of human and animal guts. T. muris- and B. wadsworthia-like sequences at 97% similarity cut-off are expressed as percentages of positive samples in each host (the numbers of samples used for the analysis are shown in parenthesis) and different colors indicate percentages of samples positive for T. muris and B. wadsworthia at different relative abundance ranges. Hosts with less than 20 amplicon samples are not shown. T. muris- and B. wadsworthia-related sequences co-occur in only 28 mouse gut samples as shown by the Venn diag ram. b. Visualization of Taurinivorans in a colon tissue section of a mouse fed a polysaccharide- and fiber-deficient diet 98 by FISH. TAU1151-Cy3-labeled Taurinivorans cells appear in pink and the remaining bacterial cells and tissue in blue due to DAPI-staining.

The dashed line indicates the border between epithelial cells and gut lumen.

resistance against Salmonella enterica in a gnotobiotic mouse model. a. Schematic outline of the gnotobiotic mouse experiment. Mice stably colonized with the 12-strain OligoMouse-Microbiota (OMM12) were inoculated with T. muris LT0009 (n=6) or sterile phosphate-buffered saline (PBS) as control (n=6) and, after 10 days, orally and rectally infected with S. enterica Tmavir M2702. Mice were sacrificed two days post infection (p.i.). Fecal samples were used for strain-specific 16S rRNA gene-targeted quantitative PCR (qPCR). Fecal and cecal samples at 24 h and 48 h p.i. were used for ana lysis of colony forming units (CFU) of S. enterica Tm. Fecal samples at 48 h p.i. were used for metatranscriptomics (RNAseq). b. Absolute abundances (16S rRNA gene copy numbers per gram feces) of 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 each OMM12 strain and strain LT0009 on day 10 in feces of mic e with LT0009 and the PBS-control mice. Small horizontal lines indicate median values. Gray horizontal lines indicate the detection limit of each strain-specific qPCR assay. c. Ranked relative transcript abundance of LT0009 gen es in OMM12 mice fecal metatranscriptomes. Each point is the mean relative abundance of a gene and error bars correspond to the 95% confidence interval of the mean (n = 3). Th e total number of transcribed LT0009 genes is shown (n=1884). Genes for taurine ( tpa, xsc, ald), sulfite (dsrAB, dsrC), sulfolactate (suyAB, sclC, comC) thiosulfate (sbdP, dsrE), pyruvate (por), lactate (lutABC), and hydrogen (hybA, hybC) metabolism are shown in different colors. Sulfur metabolism genes are further highlighted in bold font. Vertical dashed lines delineate the top 1%, 5%, and 10% expression rank of all protein-coding genes in the LT0009 genome (n=2059). d. CFU of S. enterica Tm at 24 h and 48 h p.i. in the feces and at 48 h p.i. in the cecal content. Small horizontal lines indicate median values. The dotted horizontal line shows the CFU detection limit. The asterisk indicates significant differences (p<0.05; ANOVA using Kruskal-Wallis and Dunn9s Multiple Comparison test) between S. enterica Tmavir CFU in mice with LT0009 and the PBS-control mice. ns, not significant. e. Volcano plots of differential gene transcription of S. enterica Tmavir M2702 and E. clostridioformis YL32 in OMM12 mice with and without LT0009. The x-axis shows log -fold-change in transcription and the y-axis shows the negative logarithm10-transformed adjusted p values. Blue dots show significantly down-regulated genes (adjusted p-value <0.05, log2 fold change <-1) in mice with LT0009 and are labeled with locus tag numbers. Up-r egulated E. clostridioformis YL32 prophage genes in I5Q83_10075-10390 are highlighted in bold. f. Structure of the activated prophage gene cluster of E. clostridioformis YL32 and phylogeny of its encoded phosphoadenosine-phosphosulfate reductase (CysH). Virus- and bacteria-encoded sequences are shown in red and black, respectively. The maximum likelihood CysH tree is midpoint rooted. Ultrafast bootstrap support values equal to or greater than 95% and 80% for the maximum likelihood tree are indicated with black and grey circles, respectively. The identity of the 61 genes in the prophage region (I5Q83_10075-10390) of E. clostridioformis YL32 as predicted by PHASTER 590 591 592 593 594 91. Genes encoding hypothetical proteins are in black and annotated genes are in grey. Numbers indicate the locus tag. PLP, phage-like protein; Sha, tail shaft; Pla, plate protein; Coa, coat protein; Pro, protease; Por, portal protein; Ter, terminase; Fib, fiber protein. g. Cartoon illustrating the impact of T. muris LT0009 on S. enterica Tmavir M2702 colonization resistance in the OMM12 mouse model. Created with

Biorender.com.

muris mainly utilizes taurine as the main electron acceptor for anaerobic respiration in the gut but is also capable of thiosulfate and sulfolactate respiration. Pyruvate, lactate, and likely hydrogen are the main electron donors of T. muris, while formate could also be used. Taurine is cleaved from host-derived taurocholic bile acids by other gut bacteria via bile salt hydrolase (BSH). Thiosulfate derives from mitochondrial oxidation of H2S in the gut epithelium. T. muris produces H2S from taurine via pyruvatedependent taurine transaminase (Tpa), sulfoacetaldehyde (SA) acetyltransferase (Xsc), and dissimilatory sulfite reductase (DsrAB). H2S can have various effects on the gut microbiota and host health. For example, excess H2S can impair mucus integrity 3. H2S can enhance resistance against enteropathogens by directly inhibiting enzymes in aerobically respiring Klebsiella pneumoniae 34. H2S could further impact microbial interactions and intestinal metabolism by activating phages and expression of their auxiliary metabolic genes, such as those involved in sulfur metabolism (S-AMG) 92. Created with Biorender.com. 611

Blachier, F. et al. Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am. J. Physiol. Gastrointest. Liver Physiol. (2020) doi:10.1152/ajpgi.00261.2020. Blachier, F., Beaumont, M. & Kim, E. Cysteine-derived hydrogen sulfide and gut health: a matter of

endogenous or bacterial origin. Curr. Opin. Clin. Nutr. Metab. Care 22, 68375 (2019).

Ijssennagger, N. et al. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation

by opening the mucus barrier in colon. Proc. Natl. Acad. Sci. U. S. A. 112, 10038310043 (2015).

Attene-Ramos, M. S., Wagner, E. D., Rex Gaskins, H. & Plewa, M. J. Hydrogen Sulfide Induces Direct Radical-Associated DNA Damage. Molecular Cancer Research vol. 5 4553459 Preprint at https://doi.org/10.1158/1541-7786.mcr-06-0439 (2007 ). 5. Babidge, W., Millard, S. & Roediger, W. Sulfides impair short chain fatty acid beta-oxidation at acyl CoA dehydrogenase level in colonocytes: implications for ulcerative colitis. Mol. Cell. Biochem. 181, Kabil, O. & Banerjee, R. Redox biochemistry of hydrogen sulfide. J. Biol. Chem. 285, 21903321907 Wallace, J. L., Motta, J.-P. & Buret, A. G. Hydrogen sulfide: an agent of stability at the microbiome

mucosa interface. Am. J. Physiol. Gastrointest. Liver Physiol. 314, G1433G149 (2018).

Linden, D. R. Hydrogen sulfide signaling in the gastrointestinal tract. Antioxid. Redox Signal. 20, 8183 Goubern, M., Andriamihaja, M., Nübel, T., Blachi er, F. & Bouillaud, F. Sulfide, the first inorganic

substrate for human cells. FASEB J. 21, 169931706 (2007). 10.

Matsunami, M. et al. Luminal hydrogen sulfide plays a pronociceptive role in mouse colon. Gut vol.

58 7513761 Preprint at https://doi.org/10.1136/gut. 2007.144543 (2009). 11. Huc, T. et al. Colonic hydrogen sulfide produces portal hypertension and systemic hypotension in

Metwaly, A. et al. Integrated microbiota and metabolite profiles link Crohn9s disease to sulfur 16. Yazici, C. et al. Race-dependent association of sulfidogenic bacteria with colorectal cancer. Gut 66, 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 mammalian cells. Nat. Commun. 4, 1366 (2013).

Dis. Sci. 62, 224132257 (2017). metabolism. Nat. Commun. 11, 4322 (2020). 467, 4263429 (2010). channel. Nature 483, 494-497 (2012).

disease. Nat. Commun. 7, 13419 (2016).

Winter, S. E. et al. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature

19. Bryan K. Czyzewski, D.-N. W. Identification and characterization of a bacterial hydrosulfide ion 20.

Mottawea, W. et al. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn9s 21. Ridlon, J. M., Wolf, P. G. & Rex Gaskins, H. Taurocholic acid metabolism by gut microbes and colon cancer. Gut Microbes vol. 7 2013215 Preprint at https://doi.org/10.1080 /19490976.2016.1150414 (2016). 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 563 (2014).

Natividad, J. M. et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions

26. Peck, S. C. et al. A glycyl radical enzyme enables hydrogen sulfide production by the human intestinal bacterium Bilophila wadsworthia. Proceedings of the National Academy of Sciences vol. 116 31713 3176 Preprint at https://doi.org/10.1073/pnas.18156 61116 (2019). 27. Santos, A. A. et al. A protein trisulfide couples dissimilatory sulfate reduction to energy conservation.

Science 350, 154131545 (2015).

bacteria. Nat. Commun. 10, 1609 (2019). 28. Xing, M. et al. Radical-mediated C-S bond cleavage in C2 sulfonate degradation by anaerobic 29. Fernández-Calleja, J. M. S. et al. Non-invasive continuous real-time in vivo analysis of microbial hydrogen production shows adaptation to fermentable carbohydrates in mice. Sci. Rep. 8, 15351 30. Ruff, J., Denger, K. & Cook, A. M. Sulphoacetaldehyde acetyltransferase yields acetyl phosphate: purification from Alcaligenes defragrans and gene clusters in taurine degradation. Biochem. J 369, (2018).

2753285 (2003). 31. Denger, K., Ruff, J., Rein, U. & Cook, A. M. Sulphoacetaldehyde sulpho-lyase (EC 4.4.1.12) from Desulfonispora thiosulfatigenes: purification, properties and primary sequence. Biochemical Journal vol. 357 5813586 Preprint at https://doi.org/10.104 2/bj3570581 (2001). 32.

Denger, K., Ruff, J., Schleheck, D. & Cook, A. M. Rhodococcus opacus expresses the xsc gene to utilize

taurine as a carbon source or as a nitrogen source but not as a sulfur source. Microbiology 150,

Hanson, B. T. et al. Sulfoquinovose is a select nutrient of prominent bacteria and a source of 185931867 (2004).

hydrogen sulfide in the human gut. ISME J. 1313 (2021).

Waite, D. W. et al. Proposal to reclassify the proteobacterial classes Deltaproteobacteria and Oligoflexia, and the phylum Thermodesulfobacteria into four phyla reflecting major functional

capabilities. International Journal of Systematic and Evolutionary Microbiology Preprint at 38. Richter, M. & Rosselló-Móra, R. Shifting the ge nomic gold standard for the prokaryotic species definition. Proc. Natl. Acad. Sci. U. S. A. 106, 19126319131 (2009). 39. Varghese, N. J. et al. Microbial species delineation using whole genome sequences. Nucleic Acids Res. 43, 676136771 (2015).

187, 625836264 (2005). 40. Konstantinidis, K. T. & Tiedje, J. M. Towards a genome-based taxonomy for prokaryotes. J. Bacteriol. 41. Sait, M. et al. Genome Sequence of Lawsonia intracellularis Strain N343, Isolated from a Sow with

Hemorrhagic Proliferative Enteropathy. Genome Announcements

vol.

1 at 42. Pusterla, N. & Gebhart, C. Lawsonia intracellularis infection and proliferative enteropathy in foals.

Vet. Microbiol. 167, 34341 (2013).

TauA dictates ligand specificity for the alkanesulfonate ABC importer TauABC. Biochem. J 476, 36493

3660 (2019).

Transporters and Tripartite Tricarboxylate Transporters (TTT): From Uptake to Pathogenicity. Front.

Cell. Infect. Microbiol. 8, 33 (2018). 46. Denger, K. & Cook, A. M. Racemase activity effected by two dehydrogenases in sulfolactate degradation by Chromohalobacter salexigens: purification of (S)-sulfolactate dehydrogenase.

Microbiology 156, 9673974 (2010). 47. Mayer, J. et al. 2,3-Dihydroxypropane-1-sulfonate degraded by Cupriavidus pinatubonensis JMP134: purification of dihydroxypropanesulfonate 3-dehydrogenase. Microbiology 156, 155631564 (2010). 48. Durham, B. P. et al. Cryptic carbon and sulfur cycling between surface ocean plankton. Proc. Natl.

Acad. Sci. U. S. A. 112, 4533457 (2015). phosphate-coupled desulphonative enzyme purified from Silicibacter pomeroyi DSS-3(T). Biochem. J 394, 6573664 (2006). 50. Denger, K. et al. Bifurcated degradative pathway of 3-sulfolactate in Roseovarius nubinhibens ISM via sulfoacetaldehyde acetyltransferase and (S)-cysteate sulfolyase. J. Bacteriol. 191, 564835656 51. Laue, H., Denger, K. & Cook, A. M. Taurine reduction in anaerobic respiration of Bilophila wadsworthia RZATAU. Applied and environmental microbiology vol. 63 201632021 Preprint at phs operon in Salmonella typhimurium and the contribution of thiosulfate reduction to anaerobic 54. Aussignargues, C. et al. Rhodanese functions as sulfur supplier for key enzymes in sulfur energy

IUBMB Life 59, 51359 (2007).

proteomics. Environ. Microbiol. 21, 2093225 (2019). 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 56. Florentino, A. P. et al. Insight into the sulfur metabolism of Desulfurella amilsii by differential 57. Hagen, W. R. et al. Novel structure and redox chemistry of the prosthetic groups of the iron-sulfur flavoprotein sulfide dehydrogenase from Pyrococcus furiosus; evidence for a [2Fe-2S] cluster with Asp(Cys)3 ligands. J. Biol. Inorg. Chem. 5, 5273534 (2000). 58. Stockdreher, Y. et al. New proteins involved in sulfur trafficking in the cytoplasm of Allochromatium vinosum. J. Biol. Chem. 289, 12390312403 (2014). 59. Marietou, A., Røy, H., Jørgensen, B. B. & Kjeld sen, K. U. Sulfate Transporters in Dissimilatory Sulfate

Reducing Microorganisms: A Comparative Genomics Analysis. Front. Microbiol. 9, 309 (2018).

60. Pires, R. H. et al. Characterization of theDesulfovibrio desulfuricansATCC 27774 DsrMKJOP ComplexA Membrane-Bound Redox Complex Involved in the Sulfate Respiratory Pathway. Biochemistry vol. 45 2493262 Preprint at https://doi.org/10.1021/bi05152 65 (2006). 61. Kieser, S., Zdobnov, E. M. & Trajkovski, M. Comprehensive mouse microbiota genome catalog reveals microbial factors. Elife 8, (2019).

Sci. 11, 10 (2019).

Nature 529, 2123215 (2016).

Cell Rep. 16, 2269 (2016).

major difference to its human counterpart. PLoS Comput. Biol. 18, e1009947 (2022). 62. Contijoch, E. J. et al. Gut microbiota density influences host physiology and is shaped by host and 63. Li, B. et al. Oral bacteria colonize and compete with gut microbiota in gnotobiotic mice. Int. J. Oral 64. Sonnenburg, E. D. et al. Diet-induced extinctions in the gut microbiota compound over generations.

Griffin, N. W. et al. Prior Dietary Practices and Connections to a Human Gut Microbial Metacommunity Alter Responses to Diet Interventions. Cell Host Microbe 21, 84396 (2017).

66.

Mavilio, M. et al. A Role for Timp3 in Microbiota-Driven Hepatic Steatosis and Metabolic Dysfunction.

67. Lu, Y. et al. Short Chain Fatty Acids Prevent High-fat-diet-induced Obesity in Mice by Regulating G

Protein-coupled Receptors and Gut Microbiota. Sci. Rep. 6, 37589 (2016). 68. Li, B. et al. Microbiota Depletion Impairs Thermogenesis of Brown Adipose Tissue and Browning of White Adipose Tissue. Cell Rep. 26, 272032737.e5 (2019). Microbiome. Cell Rep. 30, 290932922.e6 (2020).

Different Animal Facilities. Front. Microbiol. 10, 2999 (2019). 69. Lesker, T. R. et al. An Integrated Metagenome Catalog Reveals New Insights into the Murine Gut 70. Eberl, C. et al. Reproducible Colonization of Germ-Free Mice With the Oligo-Mouse-Microbiota in 71. Ridlon, J. M. et al. The 9 lifestyle9 of bile acid 7³-dehydroxylating bacteria: comparative genomics, metatranscriptomic, and bile acid metabolomics analysis of a defined microbial community in gnotobiotic mice. Gut Microbes 11, 3813404 (2020). 72. Swann, J. R. et al. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc. Natl. Acad. Sci. U. S. A. 108 Suppl 1, 452334530 (2011). 73. Song, Z. et al. Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. Microbiome 7, 9 (2019). metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc. Natl. Acad. Sci. U. S. A. 105, 13580313585 (2008).

Res. 61, 145031463 (2020).

gnotobiotic mice. Gut Microbes 13, 1321 (2021).

Marion, S. et al. Biogeography of microbial bile acid transformations along the murine gut. J. Lipid 76. Streidl, T. et al. The gut bacterium produces secondary bile acids and influences liver physiology in 77. Libiad, M. et al. Hydrogen sulfide perturbs mitochondrial bioenergetics and triggers metabolic reprogramming in colon cells. J. Biol. Chem. 294, 12077312090 (2019). 78. Ferreyra, J. A., Ng, K. M. & Sonnenburg, J. L. The Enteric Two-Step: nutritional strategies of bacterial pathogens within the gut. Cell. Microbiol. 16, 99331003 (2014). 79. Stecher, B. Establishing causality in Salmonell a-microbiota-host interaction: The use of gnotobiotic mouse models and synthetic microbial communities. Int. J. Med. Microbiol. 311, 151484 (2021). 80. Lagkouvardos, I. et al. The Mouse Intestinal Bacterial Collection (miBC) provides host-specific insight into cultured diversity and functional potential of the gut microbiota. Nat Microbiol 1, 16131 (2016). 81.

Noel, G. et al. Enterotoxigenic Escherichia coli is phagocytosed by macrophages underlying villus-like

intestinal epithelial cells: modeling ex vivo innate immune defenses of the human gut. Gut Microbes 00300 Preprint at https://doi.org/10.1080/19490976. 2017.1398871 (2017). 82.

Gillis, C. C. et al. Dysbiosis-Associated Change in Host Metabolism Generates Lactate to Support Salmonella Growth. Cell Host Microbe 23, 570 (2018).

ecosystem. Cell Host Microbe 14, 6413651 (2013). 83.

Maier, L. et al. Microbiota-derived hydrogen fuels Salmonella typhimurium invasion of the gut 84. Stoffels, L., Krehenbrink, M., Berks, B. C. & U nden, G. Thiosulfate reduction in Salmonella enterica is

(2021). 86. Shen, X. et al. Microbial regulation of host hydrogen sulfide bioavailability and metabolism. Free

Radical Biology and Medicine

Preprint at 87. Stecher, B. et al. Salmonella enterica serovar typhimurium exploits inflammation to compete with the intestinal microbiota. PLoS Biol. 5, 217732189 (2007).

by the Transcriptional Regulator DgoR in. J. Bacteriol. 201, (2019). 88. Singh, B. et al. Molecular and Functional Insights into the Regulation of d-Galactonate Metabolism 89. Seif, Y. et al. Genome-scale metabolic reconstructions of multiple Salmonella strains reveal serovarspecific metabolic traits. Nat. Commun. 9, 3771 (2018). 90. Ku, Y.-W., McDonough, S. P., Palaniappan, R. U. M., Chang, C.-F. & Chang, Y.-F. Novel attenuated Salmonella enterica serovar Choleraesuis strains as live vaccine candidates generated by signaturetagged mutagenesis. Infect. Immun. 73, 819438203 (2005). 91. Lamy-Besnier, Q. et al. Chromosome folding and prophage activation reveal gut-specific genome dynamics of bacteria in the

OMM12 consortium. bioRxiv 2022.05.18.492453 (2022) 92. Kieft, K. et al. Virusassociated organosulfur metabolism in human and environmental systems. Cell Rep. 36, 109471 (2021). 93.

Wolf, P. G. et al. Diversity and distribution of sulfur metabolic genes in the human gut microbiome and their association with colorectal cancer. Microbiome 10, 64 (2022). 94. Beresford-Jones, B. S. et al. The Mouse Gastrointestinal Bacteria Catalogue enables translation between the mouse and human gut microbiotas via functional mapping. Cell Host Microbe 30, 1243

immune responses. Science 365, (2019). 95. Rosshart, S. P. et al. Laboratory mice born to wild mice have natural microbiota and model human 96. Yarza, P. et al. Uniting the classification of cultured and uncultured bacteria and archaea using 16S rRNA gene sequences. Nat. Rev. Microbiol. 12, 6353645 (2014). 97. Park, M.-J. et al. Establishment of Genome Based Criteria for Classification of the Family Desulfovibrionaceae and Proposal of Two Novel Genera, gen. nov. and gen. nov. Front. Microbiol. 13, 738205 (2022). 98. Riva, A. et al. A fiber-deprived diet disturbs the fine-scale spatial architecture of the murine colon microbiome. Nat. Commun. 10, 4366 (2019). (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is available under aCC-BY-NC 4.0 International license.

Figure 1 b (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is available under aCC-BY-NC 4.0 International license.

Figure 5

9. 12 . Donertas Ayaz , B. & Zubcevic , J. Gut microbiota and neuroinflammation in pathogenesis of hypertension: A potential role for hydrogen sulfide . Pharmacol. Res . 153 , 104677 ( 2020 ). 13 . Shibuya , N. et al. A novel pathway for the production of hydrogen sulfide from D-cysteine in 14 . Barton , L. L. , Ritz , N. L. , Fauque , G. D. & Lin , H. C. Sulfur Cycling and the Intestinal Microbiome . Dig. 17. Rowan , F. E. , Docherty , N. G. , Coffey , J. C. & O9Connell , P. R. Sulphate-reducing bacteria and hydrogen sulphide in the aetiology of ulcerative colitis . Br. J. Surg . 96 , 1513158 ( 2009 ). 18. 22 . David , L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome . Nature 505 , 5593 23 . Devkota, S. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il102/2 mice . Nature vol. 487 1043108 Preprint at https://doi.org/10.10 38/nature11225 ( 2012 ). 24 . Feng , Z. et al. A human stool-derived strain caused systemic inflammation in specific-pathogen-free mice . Gut Pathog . 9 , 59 ( 2017 ). in mice. Nat. Commun . 9 , 2802 ( 2018 ). 25. 34. Stacy , A. et al. Infection trains the host for microbiota-enhanced resistance to pathogens . Cell 184 , 35 . Brugiroux , S. et al. Genome-guided design of a defined mouse microbiota that confers colonization resistance against Salmonella enterica serovar Typhimurium . Nat Microbiol 2 , 16215 ( 2016 ). 36 . Xiong , X. et al. Perilipin-2 modulates dietary fat-induced microbial global gene expression profiles in the mouse intestine . Microbiome 5 , 117 ( 2017 ). 37. 43. Qu , F. , ElOmari , K. , Wagner , A. , De Simone , A. & Beis , K. Desolvation of the substrate-binding protein 44 . Eichhorn , E., van der Ploeg, J. R. & Leisinger , T. Deletion analysis of the Escherichia coli taurine and alkanesulfonate transport systems . J. Bacteriol . 182 , 268732695 ( 2000 ). 45 . Rosa , L. T. , Bianconi , M. E. , Thomas , G. H. & Kelly , D. J. Tripartite ATP-Independent Periplasmic (TRAP) 49 . Denger , K. , Smits , T. H. M. & Cook , A. M. L-cys teate sulpho-lyase, a widespread pyridoxal 59- 52 . Heinzinger, N. K. , Fujimoto , S. Y. , Clark , M. A. , Moreno , M. S. & Barrett , E. L. Sequence analysis of the energy metabolism . J. Bacteriol . 177 , 281332820 ( 1995 ). 53 . Aketagawa , J. , Kobayashi , K. & Ishimoto , M. Purification and properties of thiosulfate reductase from Desulfovibrio vulgaris , Miyazaki F. J. Biochem . 97 , 102531032 ( 1985 ). 55. Cipollone , R. , Ascenzi , P. & Visca , P. Common themes and variations in the rhodanese superfamily . metabolism. J. Biol . Chem . 287 , 19936319948 ( 2012 ). 74. Jones , B. V. , Begley , M. , Hill , C. , Gahan , C. G. M. & Marchesi , J. R. Functional and comparative driven by the proton motive force . J. Bacteriol . 194 , 4753485 ( 2012 ). 85. Rogers , A. W. L. , Tsolis , R. M. & Bäumler , A. J . versus the Microbiome . Microbiol. Mol. Biol. Rev . 85 ,