Clostridium difficile Colonizes Alternative Nutrient Niches during Infection across Distinct Murine Gut Microbiomes

Infection by the bacterium Clostridium difficile causes an inflammatory diarrheal disease which can become life threatening and has grown to be the most prevalent nosocomial infection. Susceptibility to C. difficile infection is strongly associated with previous antibiotic treatment, which disrupts the gut microbiota and reduces its ability to prevent colonization. In this study, we demonstrated that C. difficile altered pathogenesis between hosts pretreated with antibiotics from separate classes and exploited different nutrient sources across these environments. Our metabolite score calculation also provides a platform to study nutrient requirements of pathogens during an infection. Our results suggest that C. difficile colonization resistance is mediated by multiple groups of bacteria competing for several subsets of nutrients and could explain why total reintroduction of competitors through fecal microbial transplant currently is the most effective treatment for recurrent CDI. This work could ultimately contribute to the identification of targeted, context-dependent measures that prevent or reduce C. difficile colonization, including pre- and probiotic therapies. ABSTRACT Clostridium difficile is the largest single cause of hospital-acquired infection in the United States. A major risk factor for Clostridium difficile infection (CDI) is prior exposure to antibiotics, as they disrupt the gut bacterial community which protects from C. difficile colonization. Multiple antibiotic classes have been associated with CDI susceptibility, many leading to distinct community structures stemming from variation in bacterial targets of action. These community structures present separate metabolic challenges to C. difficile. Therefore, we hypothesized that the pathogen adapts its physiology to the nutrients within different gut environments. Utilizing an in vivo CDI model, we demonstrated that C. difficile highly colonized ceca of mice pretreated with any of three antibiotics from distinct classes. Levels of C. difficile spore formation and toxin activity varied between animals based on the antibiotic pretreatment. These physiologic processes in C. difficile are partially regulated by environmental nutrient concentrations. To investigate metabolic responses of the bacterium in vivo, we performed transcriptomic analysis of C. difficile from ceca of infected mice across pretreatments. This revealed heterogeneous expression in numerous catabolic pathways for diverse growth substrates. To assess which resources C. difficile exploited, we developed a genome-scale metabolic model with a transcriptome-enabled metabolite scoring algorithm integrating network architecture. This platform identified nutrients that C. difficile used preferentially between pretreatments, which were validated through untargeted mass spectrometry of each microbiome. Our results supported the hypothesis that C. difficile inhabits alternative nutrient niches across cecal microbiomes with increased preference for nitrogen-containing carbon sources, particularly Stickland fermentation substrates and host-derived glycans. IMPORTANCE Infection by the bacterium Clostridium difficile causes an inflammatory diarrheal disease which can become life threatening and has grown to be the most prevalent nosocomial infection. Susceptibility to C. difficile infection is strongly associated with previous antibiotic treatment, which disrupts the gut microbiota and reduces its ability to prevent colonization. In this study, we demonstrated that C. difficile altered pathogenesis between hosts pretreated with antibiotics from separate classes and exploited different nutrient sources across these environments. Our metabolite score calculation also provides a platform to study nutrient requirements of pathogens during an infection. Our results suggest that C. difficile colonization resistance is mediated by multiple groups of bacteria competing for several subsets of nutrients and could explain why total reintroduction of competitors through fecal microbial transplant currently is the most effective treatment for recurrent CDI. This work could ultimately contribute to the identification of targeted, context-dependent measures that prevent or reduce C. difficile colonization, including pre- and probiotic therapies.

[1]  P. D. Karp,et al.  The outcomes of pathway database computations depend on pathway ontology , 2006, Nucleic acids research.

[2]  W. Buckel,et al.  Effect of an Oxygen-Tolerant Bifurcating Butyryl Coenzyme A Dehydrogenase/Electron-Transferring Flavoprotein Complex from Clostridium difficile on Butyrate Production in Escherichia coli , 2013, Journal of bacteriology.

[3]  An-Ping Zeng,et al.  The Connectivity Structure, Giant Strong Component and Centrality of Metabolic Networks , 2003, Bioinform..

[4]  I. Martin-Verstraete,et al.  CcpA‐mediated repression of Clostridium difficile toxin gene expression , 2011, Molecular microbiology.

[5]  M. Monot,et al.  Deciphering Adaptation Strategies of the Epidemic Clostridium difficile 027 Strain during Infection through In Vivo Transcriptional Analysis , 2016, PloS one.

[6]  J. Weese,et al.  Evaluation of candidate reference genes in Clostridium difficile for gene expression normalization. , 2010, Anaerobe.

[7]  K. Wilson,et al.  Use of sodium taurocholate to enhance spore recovery on a medium selective for Clostridium difficile , 1982, Journal of clinical microbiology.

[8]  Joy Scaria,et al.  Microarray Identification of Clostridium difficile Core Components and Divergent Regions Associated with Host Origin , 2009, Journal of bacteriology.

[9]  B. Dupuy,et al.  Clostridium difficile toxin expression is inhibited by the novel regulator TcdC , 2007, Molecular microbiology.

[10]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[11]  D. Schomburg,et al.  Time-resolved amino acid uptake of Clostridium difficile 630Δerm and concomitant fermentation product and toxin formation , 2015, BMC Microbiology.

[12]  Isabelle Martin-Verstraete,et al.  The Regulatory Networks That Control Clostridium difficile Toxin Synthesis , 2016, Toxins.

[13]  Douglas G Bonett,et al.  Statistical inference for a linear function of medians: confidence intervals, hypothesis testing, and sample size requirements. , 2002, Psychological methods.

[14]  Julian Parkhill,et al.  The multidrug-resistant human pathogen Clostridium difficile has a highly mobile, mosaic genome , 2006, Nature Genetics.

[15]  Susumu Goto,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 2000, Nucleic Acids Res..

[16]  M. Grunberg‐Manago,et al.  Expression of both Bacillus subtilis threonyl-tRNA synthetase genes is autogenously regulated , 1994, Journal of bacteriology.

[17]  Sarah L. Westcott,et al.  Development of a Dual-Index Sequencing Strategy and Curation Pipeline for Analyzing Amplicon Sequence Data on the MiSeq Illumina Sequencing Platform , 2013, Applied and Environmental Microbiology.

[18]  Joachim Selbig,et al.  Mass-balanced randomization of metabolic networks , 2011, Bioinform..

[19]  K. Wilson,et al.  Role of competition for nutrients in suppression of Clostridium difficile by the colonic microflora , 1988, Infection and immunity.

[20]  J. Tiedje,et al.  Naïve Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy , 2007, Applied and Environmental Microbiology.

[21]  A. Sonenshein,et al.  Integration of metabolism and virulence in Clostridium difficile. , 2015, Research in Microbiology.

[22]  J. Nielsen,et al.  Uncovering transcriptional regulation of metabolism by using metabolic network topology. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[23]  P. Schloss,et al.  Antibiotic-Induced Alterations of the Murine Gut Microbiota and Subsequent Effects on Colonization Resistance against Clostridium difficile , 2015, mBio.

[24]  William T. Self,et al.  Analysis of Proline Reduction in the Nosocomial Pathogen Clostridium difficile , 2006, Journal of bacteriology.

[25]  A. Clarke,et al.  Assembly of the Chloroplast ATP-Dependent Clp Protease in Arabidopsis Is Regulated by the ClpT Accessory Proteins[C][W] , 2011, Plant Cell.

[26]  B. Weimer,et al.  Gut microbiota-produced succinate promotes C. difficile infection after antibiotic treatment or motility disturbance. , 2014, Cell host & microbe.

[27]  A. Sonenshein,et al.  Inhibiting the Initiation of Clostridium difficile Spore Germination using Analogs of Chenodeoxycholic Acid, a Bile Acid , 2010, Journal of bacteriology.

[28]  F. Schreiber,et al.  Centrality Analysis Methods for Biological Networks and Their Application to Gene Regulatory Networks , 2008, Gene regulation and systems biology.

[29]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[30]  Adrianne N. Edwards,et al.  CodY-Dependent Regulation of Sporulation in Clostridium difficile , 2016, Journal of bacteriology.

[31]  P. Dürre,et al.  Clostridium difficile Is an Autotrophic Bacterial Pathogen , 2013, PloS one.

[32]  A. Sonenshein,et al.  Integration of Metabolism and Virulence by Clostridium difficile CodY , 2010, Journal of bacteriology.

[33]  Casey M. Theriot,et al.  Dynamics and Establishment of Clostridium difficile Infection in the Murine Gastrointestinal Tract , 2014, Infection and Immunity.

[34]  D. Leffler,et al.  Clostridium difficile infection. , 2015, The New England journal of medicine.

[35]  P. Reeds,et al.  Nitrogen cycling in the gut. , 1998, Annual review of nutrition.

[36]  S. Nakamura,et al.  A defined growth medium for Clostridium difficile. , 1995, Microbiology.

[37]  Curtis J. Donskey,et al.  Metabolomics Analysis Identifies Intestinal Microbiota-Derived Biomarkers of Colonization Resistance in Clindamycin-Treated Mice , 2014, PloS one.

[38]  Lisa G Winston,et al.  Burden of Clostridium difficile Infection in the United States , 2015 .

[39]  Bruno Dupuy,et al.  Adaptive Strategies and Pathogenesis of Clostridium difficile from In Vivo Transcriptomics , 2013, Infection and Immunity.

[40]  Justin L Sonnenburg,et al.  A refined palate: bacterial consumption of host glycans in the gut. , 2013, Glycobiology.

[41]  Fernanda C. Lessa,et al.  Current Status of Clostridium difficile Infection Epidemiology , 2012, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[42]  K. Song,et al.  LuxS/autoinducer-2 quorum sensing molecule regulates transcriptional virulence gene expression in Clostridium difficile. , 2005, Biochemical and biophysical research communications.

[43]  S. Clare,et al.  The agr Locus Regulates Virulence and Colonization Genes in Clostridium difficile 027 , 2013, Journal of bacteriology.

[44]  C. Médigue,et al.  Reannotation of the genome sequence of Clostridium difficile strain 630. , 2011, Journal of medical microbiology.

[45]  J. Songer,et al.  Clostridium difficile: an important pathogen of food animals. , 2006, Anaerobe.

[46]  D. Rodionov,et al.  Global transcriptional control by glucose and carbon regulator CcpA in Clostridium difficile , 2012, Nucleic acids research.

[47]  V. Young,et al.  Persistence and Toxin Production by Clostridium difficile within Human Intestinal Organoids Result in Disruption of Epithelial Paracellular Barrier Function , 2014, Infection and Immunity.

[48]  L. Comstock,et al.  An Ecological Network of Polysaccharide Utilization among Human Intestinal Symbionts , 2014, Current Biology.

[49]  Bo Li,et al.  Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection , 2014, Nature Communications.

[50]  D. Schomburg,et al.  Closely related Campylobacter jejuni strains from different sources reveal a generalist rather than a specialist lifestyle , 2011, BMC Genomics.

[51]  Jeffrey D Goldsmith,et al.  A mouse model of Clostridium difficile-associated disease. , 2008, Gastroenterology.

[52]  D. Aronoff,et al.  Cefoperazone-treated mice as an experimental platform to assess differential virulence of Clostridium difficile strains , 2011, Gut microbes.

[53]  Hiroyuki Ogata,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 1999, Nucleic Acids Res..

[54]  R. Freter,et al.  Mechanisms That Control Bacterial Populations in Continuous-Flow Culture Models of Mouse Large Intestinal Flora , 1983, Infection and immunity.

[55]  E. Wingender,et al.  Topology of mammalian transcription networks. , 2005, Genome informatics. International Conference on Genome Informatics.

[56]  G. Pier,et al.  RNA Isolation of Pseudomonas aeruginosa Colonizing the Murine Gastrointestinal Tract , 2011, Journal of visualized experiments : JoVE.

[57]  R. Britton,et al.  Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. , 2014, Gastroenterology.

[58]  Christoph H Borchers,et al.  Effect of Antibiotic Treatment on the Intestinal Metabolome , 2011, Antimicrobial Agents and Chemotherapy.

[59]  J. Bond,et al.  Global Analysis of the Sporulation Pathway of Clostridium difficile , 2013, PLoS genetics.

[60]  W. Martin,et al.  Acetate:Succinate CoA-transferase in the Hydrogenosomes of Trichomonas vaginalis , 2008, Journal of Biological Chemistry.