Clostridium difficile Alters the Structure and Metabolism of Distinct Cecal Microbiomes during Initial Infection To Promote Sustained Colonization

Clostridium difficile has become the most common single cause of hospital-acquired infection over the last decade in the United States. Colonization resistance to the nosocomial pathogen is primarily provided by the gut microbiota, which is also involved in clearing the infection as the community recovers from perturbation. As distinct antibiotics are associated with different risk levels for CDI, we utilized a mouse model of infection with 3 separate antibiotic pretreatment regimens to generate alternative gut microbiomes that each allowed for C. difficile colonization but varied in clearance rate. To assess community-level dynamics, we implemented an integrative multi-omics approach that revealed that infection significantly changed many aspects of the gut community. The degree to which the community changed was inversely correlated with clearance during the first 6 days of infection, suggesting that C. difficile differentially modifies the gut environment to promote persistence. This is the first time that metagenome-enabled metatranscriptomics have been employed to study the behavior of a host-associated microbiota in response to an infection. Our results allow for a previously unseen understanding of the ecology associated with C. difficile infection and provide the groundwork for identification of context-specific probiotic therapies. ABSTRACT Susceptibility to Clostridium difficile infection (CDI) is primarily associated with previous exposure to antibiotics, which compromise the structure and function of the gut bacterial community. Specific antibiotic classes correlate more strongly with recurrent or persistent C. difficile infection. As such, we utilized a mouse model of infection to explore the effect of distinct antibiotic classes on the impact that infection has on community-level transcription and metabolic signatures shortly following pathogen colonization and how those changes may associate with persistence of C. difficile. Untargeted metabolomic analysis revealed that C. difficile infection had significantly larger impacts on the metabolic environment across cefoperazone- and streptomycin-pretreated mice, which became persistently colonized compared to clindamycin-pretreated mice, where infection quickly became undetectable. Through metagenome-enabled metatranscriptomics, we observed that transcripts for genes associated with carbon and energy acquisition were greatly reduced in infected animals, suggesting that those niches were instead occupied by C. difficile. Furthermore, the largest changes in transcription were seen in the least abundant species, indicating that C. difficile may “attack the loser” in gut environments where sustained infection occurs more readily. Overall, our results suggest that C. difficile is able to restructure the nutrient-niche landscape in the gut to promote persistent infection. IMPORTANCE Clostridium difficile has become the most common single cause of hospital-acquired infection over the last decade in the United States. Colonization resistance to the nosocomial pathogen is primarily provided by the gut microbiota, which is also involved in clearing the infection as the community recovers from perturbation. As distinct antibiotics are associated with different risk levels for CDI, we utilized a mouse model of infection with 3 separate antibiotic pretreatment regimens to generate alternative gut microbiomes that each allowed for C. difficile colonization but varied in clearance rate. To assess community-level dynamics, we implemented an integrative multi-omics approach that revealed that infection significantly changed many aspects of the gut community. The degree to which the community changed was inversely correlated with clearance during the first 6 days of infection, suggesting that C. difficile differentially modifies the gut environment to promote persistence. This is the first time that metagenome-enabled metatranscriptomics have been employed to study the behavior of a host-associated microbiota in response to an infection. Our results allow for a previously unseen understanding of the ecology associated with C. difficile infection and provide the groundwork for identification of context-specific probiotic therapies.

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

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

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

[4]  Patrick D. Schloss,et al.  Clostridium difficile Colonizes Alternative Nutrient Niches during Infection across Distinct Murine Gut Microbiomes , 2017, mSystems.

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

[6]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[7]  Chao Xie,et al.  Fast and sensitive protein alignment using DIAMOND , 2014, Nature Methods.

[8]  C. Huttenhower,et al.  Relating the metatranscriptome and metagenome of the human gut , 2014, Proceedings of the National Academy of Sciences.

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

[10]  Vincent B. Young,et al.  Suppression of Clostridium difficile in the Gastrointestinal Tracts of Germfree Mice Inoculated with a Murine Isolate from the Family Lachnospiraceae , 2012, Infection and Immunity.

[11]  S. Sørensen,et al.  Antagonism correlates with metabolic similarity in diverse bacteria , 2017, Proceedings of the National Academy of Sciences.

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

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

[14]  Taane G. Clark,et al.  Targeted Restoration of the Intestinal Microbiota with a Simple, Defined Bacteriotherapy Resolves Relapsing Clostridium difficile Disease in Mice , 2012, PLoS pathogens.

[15]  A. Moya,et al.  Structural and functional changes in the gut microbiota associated to Clostridium difficile infection , 2014, Front. Microbiol..

[16]  E. Fikrig,et al.  Pathogen-mediated manipulation of arthropod microbiota to promote infection , 2017, Proceedings of the National Academy of Sciences.

[17]  K. Turner,et al.  Metatranscriptomics of the Human Oral Microbiome during Health and Disease , 2014, mBio.

[18]  Jo Handelsman,et al.  Conditionally Rare Taxa Disproportionately Contribute to Temporal Changes in Microbial Diversity , 2014, mBio.

[19]  Anne-Laure Boulesteix,et al.  AUC-RF: A New Strategy for Genomic Profiling with Random Forest , 2011, Human Heredity.

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

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

[22]  R. Freter The fatal enteric cholera infection in the guinea pig, achieved by inhibition of normal enteric flora. , 1955, The Journal of infectious diseases.

[23]  Hilary G. Morrison,et al.  Reproducible Community Dynamics of the Gastrointestinal Microbiota following Antibiotic Perturbation , 2009, Infection and Immunity.

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

[25]  G. Gloor,et al.  Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘RePOOPulating’ the gut , 2013, Microbiome.

[26]  Leo Breiman,et al.  Random Forests , 2001, Machine Learning.

[27]  E. Vollaard,et al.  Colonization resistance , 1994, Antimicrobial Agents and Chemotherapy.

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

[29]  J. Silva,et al.  Antibiotic-associated colitis: effects of antibiotics on Clostridium difficile and the disease in hamsters. , 1979, Reviews of infectious diseases.

[30]  Kunihiko Sadakane,et al.  MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph , 2014, Bioinform..

[31]  G. Dick,et al.  Metabolic flexibility of enigmatic SAR324 revealed through metagenomics and metatranscriptomics. , 2014, Environmental Microbiology.

[32]  Bernard Henrissat,et al.  Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla , 2009, Proceedings of the National Academy of Sciences.

[33]  Philipp Berens,et al.  CircStat: AMATLABToolbox for Circular Statistics , 2009, Journal of Statistical Software.

[34]  Xiaowei Zhang,et al.  Where less may be more: how the rare biosphere pulls ecosystems strings , 2017, The ISME Journal.

[35]  G E Bignardi,et al.  Risk factors for Clostridium difficile infection. , 1998, The Journal of hospital infection.

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

[37]  David N. Fisman,et al.  Meta-Analysis of Antibiotics and the Risk of Community-Associated Clostridium difficile Infection , 2013, Antimicrobial Agents and Chemotherapy.

[38]  Guoyao Wu,et al.  Amino acid metabolism in intestinal bacteria and its potential implications for mammalian reproduction. , 2015, Molecular human reproduction.

[39]  David A. Spratt,et al.  Same Exposure but Two Radically Different Responses to Antibiotics: Resilience of the Salivary Microbiome versus Long-Term Microbial Shifts in Feces , 2015, mBio.

[40]  Toni Giorgino,et al.  Computing and Visualizing Dynamic Time Warping Alignments in R: The dtw Package , 2009 .

[41]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[42]  Chris Sander,et al.  Precision microbiome restoration of bile acid-mediated resistance to Clostridium difficile , 2014, Nature.

[43]  A. Viale,et al.  Profound Alterations of Intestinal Microbiota following a Single Dose of Clindamycin Results in Sustained Susceptibility to Clostridium difficile-Induced Colitis , 2011, Infection and Immunity.

[44]  D. Gerding,et al.  Colonization for the Prevention of Clostridium difficile Disease in Hamsters , 2002 .

[45]  R. Britton,et al.  Interaction between the intestinal microbiota and host in Clostridium difficile colonization resistance. , 2012, Trends in microbiology.

[46]  C. Theriot,et al.  Shifts in the Gut Metabolome and Clostridium difficile Transcriptome throughout Colonization and Infection in a Mouse Model , 2018, mSphere.

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

[48]  M. Ferrer,et al.  Clostridium difficile heterogeneously impacts intestinal community architecture but drives stable metabolome responses , 2015, The ISME Journal.

[49]  M. Stevenson,et al.  Antibiotics and hospital-acquired Clostridium difficile-associated diarrhoea: a systematic review. , 2003, The Journal of antimicrobial chemotherapy.