The Clinical Drug Ebselen Attenuates Inflammation and Promotes Microbiome Recovery in Mice after Antibiotic Treatment for CDI

Megan Garland, Andrew J Hryckowian, Martina Tholen, Kristina Oresic Bender, William W Van Treuren, Sebastian Loscher, Justin L Sonnenburg, Matthew Bogyo, Megan Garland, Andrew J Hryckowian, Martina Tholen, Kristina Oresic Bender, William W Van Treuren, Sebastian Loscher, Justin L Sonnenburg, Matthew Bogyo

Abstract

Clostridium difficile infection (CDI) is an enteric bacterial disease that is increasing in prevalence worldwide. C. difficile capitalizes on gut inflammation and microbiome dysbiosis to establish infection, with symptoms ranging from watery diarrhea to toxic megacolon. We reported that the safe-in-human clinical drug ebselen (ClinicalTrials.gov: NCT03013400, NCT01452607, NCT00762671, and NCT02603081) has biochemical, cell-based, and in vivo efficacy against the toxins of C. difficile. Here, we show that ebselen treatment reduces recurrence rates and decreases colitis in a hamster model of relapsing CDI. Furthermore, ebselen treatment does not alter microbiome diversity and promotes recovery back to that of healthy controls after antibiotic-induced dysbiosis in healthy and C. difficile-infected mice. This increased microbiome recovery upon ebselen treatment correlates with a decrease in host-derived inflammatory markers, suggesting that the anti-inflammatory properties of ebselen, combined with its anti-toxin function, help to mitigate the major clinical challenges of CDI, including recurrence, microbial dysbiosis, and colitis.

Conflict of interest statement

DECLARATION OF INTERESTS M.B. is a co-founder, chair of the Scientific Advisory Board and shareholder of Facile Therapeutics, which is a company working to develop ebselen for treatment of CDI. The authors declare a patent application WO2015/200358Al that covers the use of ebselen for treatment of C. difficile infections.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Ebselen Reduces Inflammation and Recurrence in a Hamster Model of Relapsing CDI (A) Schematic of hamster models. Top: hamster model of CDI. Golden Syrian hamsters were randomly divided into four treatment groups: vehicle 1 (0.9% NaCl; n = 5), vehicle 2 (6.7% DMSO, 1% Tween 80 in PBS; n = 4), ebselen (100 mg/kg; n = 10), or vancomycin (10 mg/kg; n = 10). Dysbiosis was induced with clindamycin on day −1 prior to challenge with BI/NAP1 027 ribotype C. difficile (BAA-1805) spores on day 0. Vancomycin was dosed twice daily via oral gavage starting on day 0 for 5 days. Ebselen was dosed twice daily via oral gavage starting on day 0 for 10 days. Hamsters were observed for 28 days post-infection. Bottom: hamster model of relapsing CDI. Hamsters were randomly divided into four treatment groups of vehicle (n = 10), ebselen (100 mg/kg; n = 10), vancomycin (10 mg/kg; n = 10), or vancomycin and ebselen (10 mg/kg and 100 mg/kg respectively; n = 10). Dysbiosis was induced with clindamycin on day −1 prior to challenge with BI/NAP1 027 ribotype C. difficile (VA20) spores on day 0. Vancomycin was dosed once daily via oral gavage starting on day 0 for 5 days. Ebselen was dosed twice daily via oral gavage starting on day 0 for the duration of the study. Hamsters were observed for 21 days post-infection. (B) A Kaplan-Meier curve of a hamster model of CDI. (C) Representative histological images for the hamster model of CDI. The cecum and colon were harvested from animals at the time of humane sacrifice, death, or on day 28 for surviving animals. Samples were formalin fixed and paraffin embedded for H&E staining and histopathology. (D) Histopathology quantification from cecum and colon samples from hamsters from (B) treated with vehicle (0.9% NaCl; n = 8), vancomycin (n = 20), vehicle (6.7% DMSO, 1% Tween 80 in PBS; n = 10), or ebselen (n = 20). Slides were scored blinded by a board-certified veterinary pathologist for epithelial damage, vascular congestion and hemorrhage, and inflammation, with statistical analysis by unpaired t test. ∗p 

Figure 2

Ebselen Does Not Alter the…

Figure 2

Ebselen Does Not Alter the GI Microbiome (A) Conventional Swiss Webster mice were…

Figure 2
Ebselen Does Not Alter the GI Microbiome (A) Conventional Swiss Webster mice were randomly divided into four groups and treated with vehicle (6.7% DMSO, 1% Tween 80 in PBS; n = 4), ebselen (100 mg/kg; n = 4), vancomycin (100 mg/kg; n = 4), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively; n = 4). Animals were dosed once daily via oral gavage for five treatments starting on day 0 after sampling. Fecal samples were collected daily for the 5-day experiment. (B) A principal-component analysis (PCA) plot of weighted UniFrac distances between microbiota of individual mice on day 5. Daily fecal samples were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between microbiota at day 5. Quantification is shown for intra-group measurements (side-by-side spheres, all intra-group comparisons NS; n = 14 for each) and inter-group comparisons (indicated by spheres below; n = 16 for each). Statistics were measured via one-way ANOVA with multiple comparisons; ∗∗∗∗p 

Figure 3

Ebselen Promotes Microbiome Recovery after…

Figure 3

Ebselen Promotes Microbiome Recovery after Antibiotic Treatment (A) Schematic of experiment. Conventional Swiss…

Figure 3
Ebselen Promotes Microbiome Recovery after Antibiotic Treatment (A) Schematic of experiment. Conventional Swiss Webster mice were randomly divided into four groups treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS; n = 6), ebselen (100 mg/kg; n = 6), vancomycin (100 mg/kg; n = 6), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively; n = 6). Treatments were dosed once daily via oral gavage for five treatments (treatment phase) starting on day 0 after sampling. (B) Fecal samples (n = 4 for vehicle; n = 6 for all others) were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. Alpha diversity of GI microbial ASVs was measured over the treatment and recovery phases via Shannon diversity index. (C) Statistical analysis of microbial diversity shown in (B) at day 28 (n = 4 for vehicle and n = 6 for all others): statistical analysis by one-way ANOVA. ∗∗p 

Figure 4

Ebselen Attenuates Host-Derived Inflammation and…

Figure 4

Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of…

Figure 4
Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of CDI (A) Schematic of a mouse model of CDI. Dysbiosis was induced in conventional Swiss Webster mice with an antibiotic cocktail in drinking water (kanamycin 0.4 mg/mL, gentamicin 0.035 mg/mL, colistin 850 U/mL, metronidazole [0.215 mg/mL], and vancomycin [0.045 mg/mL]) for 3 days, beginning 6 days before inoculation (gray box, pre-treatment). Mice were switched to regular water for 2 days and then treated with 1 mg of clindamycin via oral gavage on day −1. On day 0, mice were randomly divided into an uninfected control group (n = 5) or four treatment groups (n = 7 for each) treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS), ebselen (100 mg/kg), vancomycin (100 mg/kg), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively). Mice were orally challenged with approximately 108 CFUs of C. difficile strain 630 (treatment groups) or PBS control. Treatments were dosed once daily via oral gavage for five treatments (acute infection and treatment phase) starting on day 0 after sampling. Fecal samples were collected for 28 days post-infection. (B) PCA plot of weighted UniFrac distances of GI microbiota at day 28. Fecal samples from each animal were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between vehicle/vancomycin (n = 42), vehicle/ebselen and vancomycin (n = 42), vancomycin/ebslen and vancomycin (n = 42), and intra groups (n = 21 each) at day 28. Colored spheres indicate groups from which distances were measured. Analysis of comparison between weighted UniFrac distances were made via one-way ANOVA, ∗∗p 
Similar articles
Cited by
References
    1. Lessa F.C., Mu Y., Bamberg W.M., Beldavs Z.G., Dumyati G.K., Dunn J.R., Farley M.M., Holzbauer S.M., Meek J.I., Phipps E.C. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 2015;372:825–834. - PubMed
    1. Desai M.S., Seekatz A.M., Koropatkin N.M., Kamada N., Hickey C.A., Wolter M., Pudlo N.A., Kitamoto S., Terrapon N., Muller A. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell. 2016;167:1339–1353.e21. - PMC - PubMed
    1. Gerding D.N., Kelly C.P., Rahav G., Lee C., Dubberke E.R., Kumar P.N., Yachyshyn B., Kao D., Eves K., Ellison M.C. Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis. 2018;67:649–656. - PMC - PubMed
    1. Hryckowian A.J., Pruss K.M., Sonnenburg J.L. The emerging metabolic view of Clostridium difficile pathogenesis. Curr. Opin. Microbiol. 2017;35:42–47. - PMC - PubMed
    1. Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–215. - PMC - PubMed
Show all 45 references
Publication types
MeSH terms
Associated data
LinkOut - more resources
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2
Figure 2
Ebselen Does Not Alter the GI Microbiome (A) Conventional Swiss Webster mice were randomly divided into four groups and treated with vehicle (6.7% DMSO, 1% Tween 80 in PBS; n = 4), ebselen (100 mg/kg; n = 4), vancomycin (100 mg/kg; n = 4), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively; n = 4). Animals were dosed once daily via oral gavage for five treatments starting on day 0 after sampling. Fecal samples were collected daily for the 5-day experiment. (B) A principal-component analysis (PCA) plot of weighted UniFrac distances between microbiota of individual mice on day 5. Daily fecal samples were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between microbiota at day 5. Quantification is shown for intra-group measurements (side-by-side spheres, all intra-group comparisons NS; n = 14 for each) and inter-group comparisons (indicated by spheres below; n = 16 for each). Statistics were measured via one-way ANOVA with multiple comparisons; ∗∗∗∗p 

Figure 3

Ebselen Promotes Microbiome Recovery after…

Figure 3

Ebselen Promotes Microbiome Recovery after Antibiotic Treatment (A) Schematic of experiment. Conventional Swiss…

Figure 3
Ebselen Promotes Microbiome Recovery after Antibiotic Treatment (A) Schematic of experiment. Conventional Swiss Webster mice were randomly divided into four groups treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS; n = 6), ebselen (100 mg/kg; n = 6), vancomycin (100 mg/kg; n = 6), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively; n = 6). Treatments were dosed once daily via oral gavage for five treatments (treatment phase) starting on day 0 after sampling. (B) Fecal samples (n = 4 for vehicle; n = 6 for all others) were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. Alpha diversity of GI microbial ASVs was measured over the treatment and recovery phases via Shannon diversity index. (C) Statistical analysis of microbial diversity shown in (B) at day 28 (n = 4 for vehicle and n = 6 for all others): statistical analysis by one-way ANOVA. ∗∗p 

Figure 4

Ebselen Attenuates Host-Derived Inflammation and…

Figure 4

Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of…

Figure 4
Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of CDI (A) Schematic of a mouse model of CDI. Dysbiosis was induced in conventional Swiss Webster mice with an antibiotic cocktail in drinking water (kanamycin 0.4 mg/mL, gentamicin 0.035 mg/mL, colistin 850 U/mL, metronidazole [0.215 mg/mL], and vancomycin [0.045 mg/mL]) for 3 days, beginning 6 days before inoculation (gray box, pre-treatment). Mice were switched to regular water for 2 days and then treated with 1 mg of clindamycin via oral gavage on day −1. On day 0, mice were randomly divided into an uninfected control group (n = 5) or four treatment groups (n = 7 for each) treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS), ebselen (100 mg/kg), vancomycin (100 mg/kg), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively). Mice were orally challenged with approximately 108 CFUs of C. difficile strain 630 (treatment groups) or PBS control. Treatments were dosed once daily via oral gavage for five treatments (acute infection and treatment phase) starting on day 0 after sampling. Fecal samples were collected for 28 days post-infection. (B) PCA plot of weighted UniFrac distances of GI microbiota at day 28. Fecal samples from each animal were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between vehicle/vancomycin (n = 42), vehicle/ebselen and vancomycin (n = 42), vancomycin/ebslen and vancomycin (n = 42), and intra groups (n = 21 each) at day 28. Colored spheres indicate groups from which distances were measured. Analysis of comparison between weighted UniFrac distances were made via one-way ANOVA, ∗∗p 
Similar articles
Cited by
References
    1. Lessa F.C., Mu Y., Bamberg W.M., Beldavs Z.G., Dumyati G.K., Dunn J.R., Farley M.M., Holzbauer S.M., Meek J.I., Phipps E.C. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 2015;372:825–834. - PubMed
    1. Desai M.S., Seekatz A.M., Koropatkin N.M., Kamada N., Hickey C.A., Wolter M., Pudlo N.A., Kitamoto S., Terrapon N., Muller A. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell. 2016;167:1339–1353.e21. - PMC - PubMed
    1. Gerding D.N., Kelly C.P., Rahav G., Lee C., Dubberke E.R., Kumar P.N., Yachyshyn B., Kao D., Eves K., Ellison M.C. Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis. 2018;67:649–656. - PMC - PubMed
    1. Hryckowian A.J., Pruss K.M., Sonnenburg J.L. The emerging metabolic view of Clostridium difficile pathogenesis. Curr. Opin. Microbiol. 2017;35:42–47. - PMC - PubMed
    1. Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–215. - PMC - PubMed
Show all 45 references
Publication types
MeSH terms
Associated data
LinkOut - more resources
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3
Figure 3
Ebselen Promotes Microbiome Recovery after Antibiotic Treatment (A) Schematic of experiment. Conventional Swiss Webster mice were randomly divided into four groups treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS; n = 6), ebselen (100 mg/kg; n = 6), vancomycin (100 mg/kg; n = 6), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively; n = 6). Treatments were dosed once daily via oral gavage for five treatments (treatment phase) starting on day 0 after sampling. (B) Fecal samples (n = 4 for vehicle; n = 6 for all others) were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. Alpha diversity of GI microbial ASVs was measured over the treatment and recovery phases via Shannon diversity index. (C) Statistical analysis of microbial diversity shown in (B) at day 28 (n = 4 for vehicle and n = 6 for all others): statistical analysis by one-way ANOVA. ∗∗p 

Figure 4

Ebselen Attenuates Host-Derived Inflammation and…

Figure 4

Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of…

Figure 4
Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of CDI (A) Schematic of a mouse model of CDI. Dysbiosis was induced in conventional Swiss Webster mice with an antibiotic cocktail in drinking water (kanamycin 0.4 mg/mL, gentamicin 0.035 mg/mL, colistin 850 U/mL, metronidazole [0.215 mg/mL], and vancomycin [0.045 mg/mL]) for 3 days, beginning 6 days before inoculation (gray box, pre-treatment). Mice were switched to regular water for 2 days and then treated with 1 mg of clindamycin via oral gavage on day −1. On day 0, mice were randomly divided into an uninfected control group (n = 5) or four treatment groups (n = 7 for each) treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS), ebselen (100 mg/kg), vancomycin (100 mg/kg), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively). Mice were orally challenged with approximately 108 CFUs of C. difficile strain 630 (treatment groups) or PBS control. Treatments were dosed once daily via oral gavage for five treatments (acute infection and treatment phase) starting on day 0 after sampling. Fecal samples were collected for 28 days post-infection. (B) PCA plot of weighted UniFrac distances of GI microbiota at day 28. Fecal samples from each animal were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between vehicle/vancomycin (n = 42), vehicle/ebselen and vancomycin (n = 42), vancomycin/ebslen and vancomycin (n = 42), and intra groups (n = 21 each) at day 28. Colored spheres indicate groups from which distances were measured. Analysis of comparison between weighted UniFrac distances were made via one-way ANOVA, ∗∗p 
Similar articles
Cited by
References
    1. Lessa F.C., Mu Y., Bamberg W.M., Beldavs Z.G., Dumyati G.K., Dunn J.R., Farley M.M., Holzbauer S.M., Meek J.I., Phipps E.C. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 2015;372:825–834. - PubMed
    1. Desai M.S., Seekatz A.M., Koropatkin N.M., Kamada N., Hickey C.A., Wolter M., Pudlo N.A., Kitamoto S., Terrapon N., Muller A. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell. 2016;167:1339–1353.e21. - PMC - PubMed
    1. Gerding D.N., Kelly C.P., Rahav G., Lee C., Dubberke E.R., Kumar P.N., Yachyshyn B., Kao D., Eves K., Ellison M.C. Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis. 2018;67:649–656. - PMC - PubMed
    1. Hryckowian A.J., Pruss K.M., Sonnenburg J.L. The emerging metabolic view of Clostridium difficile pathogenesis. Curr. Opin. Microbiol. 2017;35:42–47. - PMC - PubMed
    1. Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–215. - PMC - PubMed
Show all 45 references
Publication types
MeSH terms
Associated data
LinkOut - more resources
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Ebselen Attenuates Host-Derived Inflammation and Promotes Microbiome Recovery in a Mouse Model of CDI (A) Schematic of a mouse model of CDI. Dysbiosis was induced in conventional Swiss Webster mice with an antibiotic cocktail in drinking water (kanamycin 0.4 mg/mL, gentamicin 0.035 mg/mL, colistin 850 U/mL, metronidazole [0.215 mg/mL], and vancomycin [0.045 mg/mL]) for 3 days, beginning 6 days before inoculation (gray box, pre-treatment). Mice were switched to regular water for 2 days and then treated with 1 mg of clindamycin via oral gavage on day −1. On day 0, mice were randomly divided into an uninfected control group (n = 5) or four treatment groups (n = 7 for each) treated with one of four treatments: vehicle (6.7% DMSO, 1% Tween80 in PBS), ebselen (100 mg/kg), vancomycin (100 mg/kg), or vancomycin and ebselen (100 mg/kg and 100 mg/kg, respectively). Mice were orally challenged with approximately 108 CFUs of C. difficile strain 630 (treatment groups) or PBS control. Treatments were dosed once daily via oral gavage for five treatments (acute infection and treatment phase) starting on day 0 after sampling. Fecal samples were collected for 28 days post-infection. (B) PCA plot of weighted UniFrac distances of GI microbiota at day 28. Fecal samples from each animal were analyzed for GI microbial diversity via 16S rRNA amplicon analysis. (C) Weighted UniFrac distances between vehicle/vancomycin (n = 42), vehicle/ebselen and vancomycin (n = 42), vancomycin/ebslen and vancomycin (n = 42), and intra groups (n = 21 each) at day 28. Colored spheres indicate groups from which distances were measured. Analysis of comparison between weighted UniFrac distances were made via one-way ANOVA, ∗∗p 

References

    1. Lessa F.C., Mu Y., Bamberg W.M., Beldavs Z.G., Dumyati G.K., Dunn J.R., Farley M.M., Holzbauer S.M., Meek J.I., Phipps E.C. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 2015;372:825–834.
    1. Desai M.S., Seekatz A.M., Koropatkin N.M., Kamada N., Hickey C.A., Wolter M., Pudlo N.A., Kitamoto S., Terrapon N., Muller A. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell. 2016;167:1339–1353.e21.
    1. Gerding D.N., Kelly C.P., Rahav G., Lee C., Dubberke E.R., Kumar P.N., Yachyshyn B., Kao D., Eves K., Ellison M.C. Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis. 2018;67:649–656.
    1. Hryckowian A.J., Pruss K.M., Sonnenburg J.L. The emerging metabolic view of Clostridium difficile pathogenesis. Curr. Opin. Microbiol. 2017;35:42–47.
    1. Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–215.
    1. Hryckowian A.J., Van Treuren W., Smits S.A., Davis N.M., Gardner J.O., Bouley D.M., Sonnenburg J.L. Microbiota-accessible carbohydrates suppress Clostridium difficile infection in a murine model. Nat. Microbiol. 2018;3:662–669.
    1. Trifan A., Stanciu C., Girleanu I., Stoica O.C., Singeap A.M., Maxim R., Chiriac S.A., Ciobica A., Boiculese L. Proton pump inhibitors therapy and risk of Clostridium difficile infection: Systematic review and meta-analysis. World J. Gastroenterol. 2017;23:6500–6515.
    1. Ng K.M., Ferreyra J.A., Higginbottom S.K., Lynch J.B., Kashyap P.C., Gopinath S., Naidu N., Choudhury B., Weimer B.C., Monack D.M., Sonnenburg J.L. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature. 2013;502:96–99.
    1. Schubert A.M., Rogers M.A., Ring C., Mogle J., Petrosino J.P., Young V.B., Aronoff D.M., Schloss P.D. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. MBio. 2014;5:e01021. e14.
    1. Schubert A.M., Sinani H., Schloss P.D. Antibiotic-Induced Alterations of the Murine Gut Microbiota and Subsequent Effects on Colonization Resistance against Clostridium difficile. MBio. 2015;6:e00974.
    1. Zaura E., Brandt B.W., Teixeira de Mattos M.J., Buijs M.J., Caspers M.P., Rashid M.U., Weintraub A., Nord C.E., Savell A., Hu Y. Same Exposure but Two Radically Different Responses to Antibiotics: Resilience of the Salivary Microbiome versus Long-Term Microbial Shifts in Feces. MBio. 2015;6:e01693-e15.
    1. Cohen S.H., Gerding D.N., Johnson S., Kelly C.P., Loo V.G., McDonald L.C., Pepin J., Wilcox M.H., Society for Healthcare Epidemiology of America. Infectious Diseases Society of America Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA) Infect. Control Hosp. Epidemiol. 2010;31:431–455.
    1. McDonald L.C., Gerding D.N., Johnson S., Bakken J.S., Carroll K.C., Coffin S.E., Dubberke E.R., Garey K.W., Gould C.V., Kelly C. Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: 2017 Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA) Clin. Infect. Dis. 2018;66:987–994.
    1. Johnson S., Louie T.J., Gerding D.N., Cornely O.A., Chasan-Taber S., Fitts D., Gelone S.P., Broom C., Davidson D.M., Polymer Alternative for CDI Treatment (PACT) investigators Vancomycin, metronidazole, or tolevamer for Clostridium difficile infection: results from two multinational, randomized, controlled trials. Clin. Infect. Dis. 2014;59:345–354.
    1. Kelly C.P. Can we identify patients at high risk of recurrent Clostridium difficile infection? Clinical microbiology and infection: the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2012;18(Suppl 6):21–27.
    1. van Nood E., Vrieze A., Nieuwdorp M., Fuentes S., Zoetendal E.G., de Vos W.M., Visser C.E., Kuijper E.J., Bartelsman J.F., Tijssen J.G. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013;368:407–415.
    1. Seekatz A.M., Aas J., Gessert C.E., Rubin T.A., Saman D.M., Bakken J.S., Young V.B. Recovery of the gut microbiome following fecal microbiota transplantation. MBio. 2014;5:e00893-e14.
    1. Shen A. Clostridium difficile toxins: mediators of inflammation. J. Innate Immun. 2012;4:149–158.
    1. Symphony Health . 2019. Zinplava - US Estimates 2019.
    1. Tam J., Hamza T., Ma B., Chen K., Beilhartz G.L., Ravel J., Feng H., Melnyk R.A. Host-targeted niclosamide inhibits C. difficile virulence and prevents disease in mice without disrupting the gut microbiota. Nat. Commun. 2018;9:5233.
    1. Bender K.O., Garland M., Ferreyra J.A., Hryckowian A.J., Child M.A., Puri A.W., Solow-Cordero D.E., Higginbottom S.K., Segal E., Banaei N. A small-molecule antivirulence agent for treating Clostridium difficile infection. Sci. Transl. Med. 2015;7:306ra148.
    1. Beilhartz G.L., Tam J., Zhang Z., Melnyk R.A. Comment on “A small-molecule antivirulence agent for treating Clostridium difficile infection”. Sci Transl Med. 2016;8:370tc2.
    1. Sies H. Ebselen, a selenoorganic compound as glutathione peroxidase mimic. Free Radic. Biol. Med. 1993;14:313–323.
    1. Antony S., Bayse C.A. Modeling the mechanism of the glutathione peroxidase mimic ebselen. Inorg. Chem. 2011;50:12075–12084.
    1. Best E.L., Freeman J., Wilcox M.H. Models for the study of Clostridium difficile infection. Gut Microbes. 2012;3:145–167.
    1. Anton P.M., O’Brien M., Kokkotou E., Eisenstein B., Michaelis A., Rothstein D., Paraschos S., Kelly C.P., Pothoulakis C. Rifalazil treats and prevents relapse of clostridium difficile-associated diarrhea in hamsters. Antimicrob. Agents Chemother. 2004;48:3975–3979.
    1. Kokkotou E., Moss A.C., Michos A., Espinoza D., Cloud J.W., Mustafa N., O’Brien M., Pothoulakis C., Kelly C.P. Comparative efficacies of rifaximin and vancomycin for treatment of Clostridium difficile-associated diarrhea and prevention of disease recurrence in hamsters. Antimicrob. Agents Chemother. 2008;52:1121–1126.
    1. Weiss W., Pulse M., Vickers R. In vivo assessment of SMT19969 in a hamster model of clostridium difficile infection. Antimicrob. Agents Chemother. 2014;58:5714–5718.
    1. Ochsner U.A., Bell S.J., O’Leary A.L., Hoang T., Stone K.C., Young C.L., Critchley I.A., Janjic N. Inhibitory effect of REP3123 on toxin and spore formation in Clostridium difficile, and in vivo efficacy in a hamster gastrointestinal infection model. J. Antimicrob. Chemother. 2009;63:964–971.
    1. See I., Mu Y., Cohen J., Beldavs Z.G., Winston L.G., Dumyati G., Holzbauer S., Dunn J., Farley M.M., Lyons C. NAP1 strain type predicts outcomes from Clostridium difficile infection. Clin. Infect. Dis. 2014;58:1394–1400.
    1. Bignardi G.E. Risk factors for Clostridium difficile infection. J. Hosp. Infect. 1998;40:1–15.
    1. Dethlefsen L., Relman D.A. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl 1):4554–4561.
    1. Theriot C.M., Koenigsknecht M.J., Carlson P.E., Jr., Hatton G.E., Nelson A.M., Li B., Huffnagle G.B., Z Li J., Young V.B. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun. 2014;5:3114.
    1. Chen X., Katchar K., Goldsmith J.D., Nanthakumar N., Cheknis A., Gerding D.N., Kelly C.P. A mouse model of Clostridium difficile-associated disease. Gastroenterology. 2008;135:1984–1992.
    1. Studer N., Desharnais L., Beutler M., Brugiroux S., Terrazos M.A., Menin L., Schürch C.M., McCoy K.D., Kuehne S.A., Minton N.P. Functional Intestinal Bile Acid 7α-Dehydroxylation by Clostridium scindens Associated with Protection from Clostridium difficile Infection in a Gnotobiotic Mouse Model. Front. Cell. Infect. Microbiol. 2016;6:191.
    1. Chassaing B., Srinivasan G., Delgado M.A., Young A.N., Gewirtz A.T., Vijay-Kumar M. Fecal lipocalin 2, a sensitive and broadly dynamic non-invasive biomarker for intestinal inflammation. PLoS ONE. 2012;7:e44328.
    1. Warren C.A., van Opstal E.J., Riggins M.S., Li Y., Moore J.H., Kolling G.L., Guerrant R.L., Hoffman P.S. Vancomycin treatment’s association with delayed intestinal tissue injury, clostridial overgrowth, and recurrence of Clostridium difficile infection in mice. Antimicrob. Agents Chemother. 2013;57:689–696.
    1. Khanna S., Pardi D.S., Kelly C.R., Kraft C.S., Dhere T., Henn M.R., Lombardo M.J., Vulic M., Ohsumi T., Winkler J. A Novel Microbiome Therapeutic Increases Gut Microbial Diversity and Prevents Recurrent Clostridium difficile Infection. J. Infect. Dis. 2016;214:173–181.
    1. U.S. Food & Drug Administration . 2019. Important Safety Alert Regarding Use of Fecal Microbiota for Transplantation and Risk of Serious Adverse Reactions Due to Transmission of Multi-Drug Resistant Organisms.
    1. Tran V., Phan J., Nulsen B., Huang L., Kaneshiro M., Weiss G., Ho W., Sack J., Ha C., Uslan D., Sauk J.S. Severe Ileocolonic Crohn’s Disease Flare Associated with Fecal Microbiota Transplantation Requiring Diverting Ileostomy. ACG Case Rep. J. 2018;5:e97.
    1. Shaw L.P., Bassam H., Barnes C.P., Walker A.S., Klein N., Balloux F. Modelling microbiome recovery after antibiotics using a stability landscape framework. ISME J. 2019;13:1845–1856.
    1. Lloyd-Price J., Abu-Ali G., Huttenhower C. The healthy human microbiome. Genome Med. 2016;8:51.
    1. Vázquez-Baeza Y., Pirrung M., Gonzalez A., Knight R. EMPeror: a tool for visualizing high-throughput microbial community data. Gigascience. 2013;2:16.
    1. Pawlowski S.W., Calabrese G., Kolling G.L., Platts-Mills J., Freire R., AlcantaraWarren C., Liu B., Sartor R.B., Guerrant R.L. Murine model of Clostridium difficile infection with aged gnotobiotic C57BL/6 mice and a BI/NAP1 strain. J. Infect. Dis. 2010;202:1708–1712.
    1. Callahan B.J., McMurdie P.J., Rosen M.J., Han A.W., Johnson A.J., Holmes S.P. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581–583.

Source: PubMed

3
S'abonner