Oral probiotic combination of Lactobacillus and Bifidobacterium alters the gastrointestinal microbiota during antibiotic treatment for Clostridium difficile infection

T J De Wolfe, S Eggers, A K Barker, A E Kates, K A Dill-McFarland, G Suen, N Safdar, T J De Wolfe, S Eggers, A K Barker, A E Kates, K A Dill-McFarland, G Suen, N Safdar

Abstract

Perturbations in the gastrointestinal microbiome caused by antibiotics are a major risk factor for Clostridium difficile infection (CDI). Probiotics are often recommended to mitigate CDI symptoms; however, there exists only limited evidence showing probiotic efficacy for CDI. Here, we examined changes to the GI microbiota in a study population where probiotic treatment was associated with significantly reduced duration of CDI diarrhea. Subjects being treated with standard of care antibiotics for a primary episode of CDI were randomized to probiotic treatment or placebo for 4 weeks. Probiotic treatment consisted of a daily multi-strain capsule (Lactobacillus acidophilus NCFM, ATCC 700396; Lactobacillus paracasei Lpc-37, ATCC SD5275; Bifidobacterium lactis Bi-07, ATCC SC5220; Bifidobacterium lactis B1-04, ATCC SD5219) containing 1.7 x 1010 CFUs. Stool was collected and analyzed using 16S rRNA sequencing. Microbiome analysis revealed apparent taxonomic differences between treatments and timepoints. Subjects administered probiotics had reduced Verrucomicrobiaceae at week 8 compared to controls. Bacteroides were significantly reduced between weeks 0 to 4 in probiotic treated subjects. Ruminococcus (family Lachnospiraceae), tended to be more abundant at week 8 than week 4 within the placebo group and at week 8 than week 0 within the probiotic group. Similar to these results, previous studies have associated these taxa with probiotic use and with mitigation of CDI symptoms. Compositional prediction of microbial community function revealed that subjects in the placebo group had microbiomes enriched with the iron complex transport system, while probiotic treated subjects had microbiomes enriched with the antibiotic transport system. Results indicate that probiotic use may impact the microbiome function in the face of a CDI; yet, more sensitive methods with higher resolution are warranted to better elucidate the roles associated with these changes. Continuing studies are needed to better understand probiotic effects on microbiome structure and function and the resulting impacts on CDI.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Microbial α-diversity as measured by…
Fig 1. Microbial α-diversity as measured by the Shannon diversity index.
The Shannon diversity index, which is a heterogeneity measure that combines richness and evenness components of microbial diversity does not significantly differ when subjects take probiotics.
Fig 2. Taxa were found to be…
Fig 2. Taxa were found to be differentially abundant depending on treatment and time point.
(A) Subjects in the probiotic group had a significantly lower abundance of the bacterial family Verrucomicrobiaceae at week 8 than placebo treated subjects; (B) Members of the bacterial genus Bacteroides were significantly reduced in abundance between weeks 0 to 4 in probiotic treated subjects; (C) Ruminococcus (family Lachnospiraceae) tended to be more abundant at week 8 than week 4 within the placebo group and at week 8 than week 0 within the probiotic group; * 0.05 < p < 0.1 and **p ≤ 0.05.
Fig 3. Predicted functional profiling of the…
Fig 3. Predicted functional profiling of the GI microbiome in placebo or probiotic treated subjects undergoing CDI.
Profiling of KEGG modules was based on 16S rRNA marker gene sequences from week 4 using PICRUSt. The α parameter for pairwise tests was set to 0.05 for class normality and the threshold on the logarithmic score of LDA analysis was set to 2.0. Modules that differed significantly in abundance between the treatment groups are displayed with the respective LDA score.

References

    1. Miller BA, Chen LF, Sexton DJ, Anderson DJ. Comparison of the burdens of hospital-onset, healthcare facility-associated Clostridium difficile infection and of healthcare-associated infection due to methicillin-resistant Staphylococcus aureus in community hospitals. Infect Control Hosp Epidemiol. 2011;32: 387–390. 10.1086/659156
    1. McDonald LC, Owings M, Jernigan DB. Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996–2003. Emerg Infect Dis. 2006;12: 409–415. 10.3201/eid1203.051064
    1. Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, et al. Burden of Clostridium difficile infection in the United States. N Engl J Med. 2015;372: 825–834. 10.1056/NEJMoa1408913
    1. Centers for Disease Control and Prevention. Antibiotic resistance threats in the United States, 2013 [Internet]. 2013. pp. 1–114.
    1. Department of Veterans Affairs. VHA Directive 1031: Antimicrobial Stewardship Programs. 2014;0: 1–9.
    1. Butler M, Olson A, Drekonja D, Shaukat A, Schwehr N, Shippee N, et al. Early diagnosis, prevention, and treatment of Clostridium difficile: update. Rockville (MD): Agency for Healthcare Research and Quality (US); 2016.
    1. Britton RA, Young VB. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology. 2014;146: 1547–1553. 10.1053/j.gastro.2014.01.059
    1. Sebaihia M, Wren BW, Mullany P, Fairweather NF, Minton N, Stabler R, et al. The multidrug-resistant human pathogen Clostridium difficile has a highly mobile, mosaic genome. Nat Genet. 2006;38: 779–786. 10.1038/ng1830
    1. Mathur H, Rea MC, Cotter PD, Ross RP, Hill C. The potential for emerging therapeutic options for Clostridium difficile infection. Gut Microbes. 2014;5: 696–710. 10.4161/19490976.2014.983768
    1. Olson MM, Shanholtzer CJ, Lee JT, Gerding DN. Ten years of prospective Clostridium difficile-associated disease surveillance and treatment at the Minneapolis VA medical center, 1982–1991. Infect Control Hosp Epidemiol. 1994;15: 371–381. 10.2307/30145589
    1. Miller M. The fascination with probiotics for Clostridium difficile infection: lack of evidence for prophylactic or therapeutic efficacy. Anaerobe. Elsevier Ltd; 2009;15: 281–284. 10.1016/j.anaerobe.2009.08.005
    1. Barker AK, Duster M, Valentine S, Hess T, Archbald-Pannone L, Guerrant R, et al. A randomized controlled trial of probiotics for Clostridium difficile infection in adults (PICO). Journal of Antimicrobial Chemotherapy. 2017;72: 1–4.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Applied and Environmental Microbiology. 2013;79: 5112–5120. 10.1128/AEM.01043-13
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Applied and Environmental Microbiology. 2009;75: 7537–7541. 10.1128/AEM.01541-09
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, et al. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Research. 2007;35: 7188–7196. 10.1093/nar/gkm864
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Applied and Environmental Microbiology. 2006;72: 5069–5072. 10.1128/AEM.03006-05
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian Classifier for Rapid Assignment of rRNA Sequences into the New Bacterial Taxonomy. Applied and Environmental Microbiology. 2007;73: 5261–5267. 10.1128/AEM.00062-07
    1. R Core Team. R: a language and environment for statistical computing [Internet]. Vienna; 2017.
    1. Oksanen J, Blanchet FG, Friendly M, Kindt R, Legendre P, McGlinn D, et al. vegan: community ecology package. 2017;0: 1–292.
    1. Noguchi K, Gel YR, Brunner E, 2012. nparLD: an R software package for the nonparametric analysis of longitudinal data in factorial experiments. Journal of Statistical Software.
    1. Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Oxford University Press; 2000.
    1. Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Research. 2016;44: D457–D462. 10.1093/nar/gkv1070
    1. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Research. 2017;45: D353–D361. 10.1093/nar/gkw1092
    1. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. Nature Publishing Group; 2013;31: 814–821. 10.1038/nbt.2676
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. Eisen JA, editor. PLoS Comput Biol. 2012;8: e1002358–17. 10.1371/journal.pcbi.1002358
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. BioMed Central Ltd; 2011;12: R60 10.1186/gb-2011-12-6-r60
    1. Goldenberg JZ, Ma S, Saxton JD. Probiotics for the prevention of Clostridium difficile‐associated diarrhea in adults and children. Cochrane Database of Systematic Reviews. 2013. 10.1002/14651858.CD006095.pub3
    1. Sanders ME. Probiotics and microbiota composition. BMC Medicine. BMC Medicine; 2016;14: 1–3.
    1. Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH, Pedersen O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials. Genome Med. Genome Medicine; 2016;8: 1–11.
    1. Cammarota G, Ianiro G, Bibbò S, Gasbarrini A. Gut microbiota modulation: probiotics, antibiotics or fecal microbiota transplantation? Intern Emerg Med. 2014;9: 365–373. 10.1007/s11739-014-1069-4
    1. Tannock GW, Munro K, Harmsen HJ, Welling GW, Smart J, Gopal PK. Analysis of the fecal microflora of human subjects consuming a probiotic product containing Lactobacillus rhamnosus DR20. Applied and Environmental Microbiology. 2000;66: 2578–2588.
    1. Grehan MJ, Borody TJ, Leis SM, Campbell J, Mitchell H, Wettstein A. Durable alteration of the colonic microbiota by the administration of donor fecal flora. J Clin Gastroenterol. 2010;44: 551–561. 10.1097/MCG.0b013e3181e5d06b
    1. Bassis CM, Theriot CM, Young VB. Alteration of the Murine Gastrointestinal Microbiota by Tigecycline Leads to Increased Susceptibility to Clostridium difficile Infection. Antimicrob Agents Chemother. 2014;58: 2767–2774. 10.1128/AAC.02262-13
    1. Weingarden AR, Chen C, Bobr A, Yao D, Lu Y, Nelson VM, et al. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficileinfection. AJP: Gastrointestinal and Liver Physiology. 2014;306: G310–G319. 10.1152/ajpgi.00282.2013
    1. Engelbrektson A, Korzenik JR, Pittler A, Sanders ME, Klaenhammer TR, Leyer G, et al. Probiotics to minimize the disruption of faecal microbiota in healthy subjects undergoing antibiotic therapy. Journal of Medical Microbiology. 2009;58: 663–670. 10.1099/jmm.0.47615-0
    1. Rolfe RD, Helebian S, Finegold SM. Bacterial Interference Between Clostridium-Difficile and Normal Fecal Flora. J Infect Dis. 1981;143: 470–475.
    1. Abujamel T, Cadnum JL, Jury LA, Sunkesula VCK, Kundrapu S, Jump RL, et al. Defining the vulnerable period for re-establishment of Clostridium difficile colonization after treatment of C. difficile infection with oral vancomycin or metronidazole. Paredes-Sabja D, editor. PLoS ONE. 2013;8: e76269 10.1371/journal.pone.0076269
    1. Dabard J, Bridonneau C, Phillipe C, Anglade P, Molle D, Nardi M, et al. Ruminococcin A, a new lantibiotic produced by a Ruminococcus gnavus strain isolated from human feces. Applied and Environmental Microbiology. American Society for Microbiology (ASM); 2001;67: 4111–4118. 10.1128/AEM.67.9.4111-4118.2001
    1. Antharam VC, Li EC, Ishmael A, Sharma A, Mai V, Rand KH, et al. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J Clin Microbiol. 2013;51: 2884–2892. 10.1128/JCM.00845-13
    1. Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, Daigneault MC, et al. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: “RePOOPulating” the gut. Microbiome. 2013;1: 1–1.
    1. McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Goodman AL, et al. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci Transl Med. 2011;3: 1–26. 10.1126/scitranslmed.3002701
    1. Brown JS, Holden DW. Iron acquisition by Gram-positive bacterial pathogens. Microbes and Infection. 2002;4: 1149–1156. 10.1016/S1286-4579(02)01640-4
    1. Mendez C, Salas JA. The role of ABC transporters in antibiotic-producing organisms: drug secretion and resistance mechanisms. Res Microbiol. 2001;152: 341–350. 10.1016/S0923-2508(01)01205-0
    1. Wilson DN. Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat Rev Micro. Nature Publishing Group; 2014;12: 35–48. 10.1038/nrmicro3155
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. Nature Publishing Group; 2014;505: 559–563. 10.1038/nature12820
    1. Jackson MA, Verdi S, Maxan M-E, Shin CM, Zierer J, Bowyer RCE, et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat Comms. Springer US; 2018; 1–8. 10.1038/s41467-018-05184-7
    1. Louie TJ, Cannon K, Byrne B, Emery J, Ward L, Eyben M, et al. Fidaxomicin Preserves the Intestinal Microbiome During and After Treatment of Clostridium difficile Infection (CDI) and Reduces Both Toxin Reexpression and Recurrence of CDI. Clinical Infectious Diseases. 2012;55: S132–S142. 10.1093/cid/cis338
    1. Schubert AM, Sinani H, Schloss PD. Antibiotic-induced alterations of the murine gut microbiota and subsequent effects on colonization resistance against Clostridium difficile. mBio. 2015;6: e00974–15–10. 10.1128/mBio.00974-15

Source: PubMed

3
Abonnieren