“Microbiota Dynamics in Patient Treated with Fecal Microbiota Transplantation for Recurrent Clostridium Difficile Infection” by Yang Song, Shashank Gard

    “Microbiota Dynamics in Patient Treated with Fecal Microbiota Transplantation for Recurrent Clostridium Difficile Infection” by Yang Song, Shashank Gard

    Order Description

    I need summative article evaluation which consists of Introduction, Body-Discussion, Conclusion and Summary.
    Introduction – identify topic, explain why it is important. State the problem as simple as possible taking into account the broader view of the discipline as a whole.
    Body/discussion – review of research material, summarizes the material to support the theory of the research. Critically analyzed unbiased result of the research, present statistical data to judge the findings, may reference literature order to show how research builds upon previous work in the field of study.
    Conclusion – explain in expert-like detail what has been learned, explained, decided, proven. Reveal the ways in which the paper’s thesis might have significance in society. A conclusion should strive to answer questions that readers logically raise-“Why are you telling me this?”
    Summary: Point out importance or implication of the research on an area of societal concern. Mention the lack of conclusion in the field to demonstrate understanding of the subject’s complexity, may choose to propose what may be the natural next step to take in light of what the argument is attempting to convince.

    Microbiota Dynamics in Patients Treated with Fecal
    Microbiota Transplantation for Recurrent Clostridium
    difficile Infection
    Yang Song1, Shashank Garg2, Mohit Girotra2, Cynthia Maddox1, Erik C. von Rosenvinge3, Anand Dutta2,
    Sudhir Dutta2,4, W. Florian Fricke1*
    1 Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America, 2 Division of Gastroenterology, Sinai Hospital
    of Baltimore, Baltimore, Maryland, United States of America, 3 Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore,
    Maryland, United States of America, 4 Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
    Abstract
    Clostridium difficile causes antibiotic-associated diarrhea and pseudomembraneous colitis and is responsible for a large and
    increasing fraction of hospital-acquired infections. Fecal microbiota transplantation (FMT) is an alternate treatment option
    for recurrent C. difficile infection (RCDI) refractory to antibiotic therapy. It has recently been discussed favorably in the
    clinical and scientific communities and is receiving increasing public attention. However, short- and long-term health
    consequences of FMT remain a concern, as the effects of the transplanted microbiota on the patient remain unknown. To
    shed light on microbial events associated with RCDI and treatment by FMT, we performed fecal microbiota analysis by 16S
    rRNA gene amplicon pyrosequencing of 14 pairs of healthy donors and RCDI patients treated successfully by FMT. Post-FMT
    patient and healthy donor samples collected up to one year after FMT were studied longitudinally, including one post-FMT
    patient with antibiotic-associated relapse three months after FMT. This analysis allowed us not only to confirm prior reports
    that RCDI is associated with reduced diversity and compositional changes in the fecal microbiota, but also to characterize
    previously undocumented post-FMT microbiota dynamics. Members of the Streptococcaceae, Enterococcaceae, or
    Enterobacteriaceae were significantly increased and putative butyrate producers, such as Lachnospiraceae and
    Ruminococcaceae were significantly reduced in samples from RCDI patients before FMT as compared to post-FMT patient
    and healthy donor samples. RCDI patient samples showed more case-specific variations than post-FMT patient and healthy
    donor samples. However, none of the bacterial groups were invariably associated with RCDI or successful treatment by FMT.
    Overall microbiota compositions in post-FMT patients, specifically abundances of the above-mentioned Firmicutes,
    continued to change for at least 16 weeks after FMT, suggesting that full microbiota recovery from RCDI may take much
    longer than expected based on the disappearance of diarrheal symptoms immediately after FMT.
    Citation: Song Y, Garg S, Girotra M, Maddox C, von Rosenvinge EC, et al. (2013) Microbiota Dynamics in Patients Treated with Fecal Microbiota Transplantation
    for Recurrent Clostridium difficile Infection. PLoS ONE 8(11): e81330. doi:10.1371/journal.pone.0081330
    Editor: Gabriele Berg, Graz University of Technology (TU Graz), Austria
    Received August 29, 2013; Accepted October 20, 2013; Published November 26, 2013
    Copyright: ! 2013 Song et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
    unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
    Funding: This study or parts thereof were funded by the Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD and
    Gastroenterology Research Funds from the Division of Gastroenterology, Department of Medicine, Sinai Hospital of Baltimore, Baltimore, MD. The funders had no
    role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
    Competing Interests: The authors have declared that no competing interests exist.
    * E-mail: [email protected]
    Introduction
    Clostridium difficile, the pathogen associated with the majority of
    infective antibiotic-associated diarrhea and causative agent of
    pseudomembraneous colitis [1], is responsible for a large fraction
    of nosocomial, or hospital-acquired, disease [2]. Today, in parts of
    the U.S., the incidence of infections with C. difficile is higher than
    that of methicillin-resistant Staphylococcus aureus [3]. C. difficile
    infection (CDI) is believed to result from gastrointestinal dysbiosis,
    i.e., the disruption of the resident microbiota, often caused by
    antibiotic treatment, which enables C. difficile to establish an
    infection. C. difficile can be acquired via fecal-oral transmission of
    spores that survive atmospheric oxygen and gastric acid exposure
    and germinate in the large intestine. However, carriage of C.
    difficile is not always associated with disease, as asymptomatic C.
    difficile colonization is well recognized [4], especially in newborns
    and infants of ,1 year age [5].
    Besides treatment with almost any antibiotic [6–14], other
    factors associated with increased risk for C. difficile infection include
    old age, recent hospitalization, tube feeding, use of gastric acidsuppressing
    drugs and underlying chronic disease, including
    inflammatory bowel disease [15–19]. Recent evidence suggests
    that excessive inflammatory responses in the human host enhance
    the severity of CDI [20].
    Standard treatment for C. difficile infection consists of metronidazole
    or vancomycin administration and, more recently, fidaxomicin.
    However, the rate of recurrent C. difficile infection (RCDI)
    after initial therapy is about 20% [21] and even higher after
    subsequent antibiotic courses and recurrences [8,22]. Consequently,
    despite current therapeutic options, RCDI treatment has
    become increasingly challenging and the incidence of RCDI has
    been rising during the past decade resulting in increased
    healthcare cost and significant morbidity [23].
    PLOS ONE | www.plosone.org 1 November 2013 | Volume 8 | Issue 11 | e81330
    Fecal microbiota transplantation (FMT), which aims to restore a
    normal, functional intestinal microbiota from a healthy donor in
    the RCDI patient, has recently received increasing attention in
    clinical and research communities [24–27] and has also become a
    popular subject of discussion in other media. First documented in
    the fourth century in China and in 1958 in the U.S., FMT was
    shown in a recent systematic review of 317 patients in 27 separate
    studies to have an overall success rate of 92% [28]. The exact
    mechanism of action responsible for the success of FMT to treat
    RCDI remains unknown and there is no clinically validated set of
    parameters to define a suitable donor or ideal donor microbiota,
    although attempts in this direction have been made [29]. Shortand
    long-term effects of FMT on the recipient microbiota remain
    a concern, especially in light of the growing body of literature that
    implicates the gastrointestinal microbiota in a large number of
    diseases [30]. For the same reason, there is significant clinical
    interest in therapeutic options to target the microbiota to treat
    microbiota-associated health problems besides RCDI. As a result,
    attempts to treat IBD [31–33], metabolic syndrome [34] and other
    diseases [35,36] by FMT have been made.
    Clinical concerns and the increasing number of FMT procedures
    performed by U.S. physicians recently led the U.S. Food
    and Drug Administration (FDA) to release new guidelines that
    define FMT as a biologic therapy that requires physicians to
    obtain an investigational new drug (IND) application [37]. Shortly
    after this guideline was a released, however, the FDA announced a
    decision to exercise enforcement discretion in order to allow
    physicians to perform FMT in patients with RCDI not responsive
    to standard therapy. The urgency for further research into the
    short- and long-term effects of FMT is highlighted by the fact that
    the public awareness of FMT as a treatment option for RCDI has
    increased to a degree where do-it-yourself protocols have become
    available over the Internet and the procedure is being performed
    without medical surveillance.
    In this study, we applied 16S rRNA amplicon pyrosequencing
    to analyze fecal samples from RCDI patients and their
    corresponding donors before and after FMT. For the first time,
    we included longitudinal simultaneous sampling of both post-FMT
    patients and healthy donors for up to one year after FMT. This
    unique sample set allowed us to describe previously undocumented
    microbiota dynamics in post-FMT patients after resolution of
    CDI. In addition, inclusion of a patient, who was initially treated
    successfully by FMT but experienced relapse after new antibiotic
    treatment, provided us with the unique opportunity to distinguish
    microbiota changes seen in a previously asymptomatic patients
    after relapse of CDI from those apparent in RCDI patients with
    long-term disease and multiple courses of anti-C. difficile antibiotic
    treatment.
    Materials and Methods
    Study cohort and sample collection
    The Institutional Review Board of Sinai Hospital Baltimore
    approved the study under protocol number #1826 and all subjects
    provided their written informed consent to participate in the study.
    FMT was performed at Sinai Hospital of Baltimore, Baltimore,
    MD by infusion of a fecal solution prepared by a predefined
    protocol (Dutta et al., submitted) based on Aas et al. [38]. Potential
    donors were thoroughly clinically evaluated based on history,
    physical examination and serological screening for HIV, syphilis,
    hepatitis A, B and C and Helicobacter pylori infection. Fecal
    specimens of patients and donors were tested 3–5 days before
    FMT for the presence of pathogenic bacteria (salmonella, shigella,
    yersinia), parasites (entamoeba, giardia, worms), and C. difficile.
    Patients were admitted to the hospital the day before and bowel
    prep administered the night before FMT. Patients were also
    administered a proton pump inhibitor (omeprazole, 20 mg) on the
    evening and morning before the procedure. Donor fecal samples
    (25–30 g) were mixed with 250 ml of sterile saline buffer, mixed
    into slurry and filtered once with surgical gauze for large particles
    and twice with a coffee filter. The volume of the filtrate was
    increased to 450 ml with sterile saline buffer and divided into 5
    aliquots of 90 ml. For FMT, two aliquots (180 ml) were
    endoscopically delivered by spray catheter into the jejunum. The
    remaining three aliquots were instilled by colonoscopy into the
    right colon (180 ml) and transverse and upper descending colon
    (90 ml).
    The clinical aspects of this study, including a comprehensive
    description and discussion of the FMT-treated patient population
    and individual case metadata, are provided in a separate
    publication (Dutta et al., submitted). Fecal samples were collected
    from 14 patient-donor pairs and used for this study (Fig. 1; Table
    1). All patients had at least three recurrences of C. difficile infection
    and were treated with at least three courses of antibiotics. Fecal
    samples were collected before and after FMT from patients and, at
    corresponding time points, from their respective donors, which
    included family members (spouses and children) and friends (Fig.
    1).
    Sample collection and nucleic acid isolation
    All fecal samples were self-collected by patients and donors
    without bowel preps, stored in the freezer and within 24 hours
    brought to Sinai Hospital, after which they were stored at –80uC.
    Patients stopped antibiotic use 5 days before the FMT procedure;
    RCDI patient samples were taken 1–2 days prior to FMT. For
    processing, samples were thawed at 4uC and in aliquots of 0.15 g
    per tube re-suspended in 1 ml of 1 6phosphate-buffered saline.
    Cell lysis was initiated with two enzymatic incubations, first using
    5 ml of lysozyme (10 mg ml21; Amresco, Solon, OH, USA), 13 ml
    of mutanolysin (11.7 U ml21; Sigma-Aldrich) and 3 ml of lysostaphin
    (4.5 U ml21; Sigma-Aldrich) for an incubation of 30 min at
    37uC and, second, using 10 ml Proteinase K (20 mg ml21;
    Research Products International, Mt Prospect, IL, USA), 50 ml
    10% SDS and 2 ml RNase (10 mg ml21) for an incubation of 45
    min at 56uC. After the enzyme treatments, cells were disrupted by
    Figure 1. Overview of analyzed patient and donor samples.
    RCDI patient samples are marked in red, post-FMT patient samples in
    blue and donor samples in green. *Patient #6a experienced antibioticinduced
    relapse of C. difficile infection and was treated successfully with
    a second round of FMT as patient #6b. In the NCBI short read archive,
    samples referred to as #6b are designated as #7 samples.
    doi:10.1371/journal.pone.0081330.g001
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 2 November 2013 | Volume 8 | Issue 11 | e81330
    bead beating in tubes with Lysing Matrix B (0.1 mm silica spheres,
    MP Biomedicals, Solon, OH, USA), at 6 m s21 for 40 s at room
    temperature in a FastPrep-24 (MP Biomedicals). The resulting
    crude lysate was processed using the ZR Fecal DNA mini-prep kit
    (Zymo, Irvine, CA, USA) according to the manufacturer’s
    recommendation. The samples were eluted with 100 ml of ultra
    pure water into separate tubes. DNA concentrations in the
    samples were measured using the Quant-iT PicoGreen dsDNA
    assay kit (Molecular Probes, Invitrogen, Carlsbad, CA, USA).
    Amplification and sequencing
    In brief, hypervariable regions V1–V3 of the bacterial 16S
    rRNA gene were amplified with primers 27F and 534R as
    described previously [39]. DNA amplification of 16S rRNA genes
    was performed using AccuPrime Taq DNA polymerase High
    Fidelity (Invitrogen) and 50 ng of template DNA in a total reaction
    volume of 25 ml, following the AccuPrime product protocol.
    Reactions were run in a PTC-100 thermal controller (MJ
    Research, Waltham, MA, USA) using the following protocol: 3
    min of denaturation at 94uC, followed by 30 cycles of 30 s at 94uC
    (denaturation), 30 s at 52uC (annealing) and 45 ss at 68uC
    (elongation), with a final extension at 68uC for 5 min.
    Equimolar amounts (50 ng) of the PCR amplicons were mixed
    in a single tube. Amplification primers and reaction buffer were
    removed using the AMPure Kit (Beckman Coulter, Brea, CA,
    USA) and purified amplicon mixtures sequenced at the Institute
    for Genome Sciences, University of Maryland, using 454 primer A
    and protocols recommended by the manufacturer (Roche,
    Branford, CT, USA). Raw sequences were deposited in the Short
    Read Archive Database (http://www.ncbi.nlm.nih.gov/sra; project
    number SRP016902). In the NCBI short read archive, samples
    referred to as #6a are designated as #6 samples and samples
    referred to as #6b as #7 samples.
    Sequence processing and analysis
    16S rRNA sequence reads were processed with QIIME [40]
    and CloVR [41], using the automated CloVR-16S pipeline as
    described in the corresponding standard operating procedure [42].
    Briefly, using the QIIME split_libraries.py tool sequences were
    binned based on sample-specific barcodes, trimmed by removal of
    barcode and primer sequences and filtered for quality, using the
    default parameters, except for “—barcode-type “variable_length”.
    Chimeric sequences were removed with UCHIME [43] using
    MicrobiomeUtilities (http://microbiomeutil.sourceforge.net/) and
    the rRNA16S.gold.fasta reference database. Reads were clustered
    into operational taxonomic units (OTUs) using a similarity
    threshold of 95%. On average, OTUs were classified using the
    RDP Naive Bayesian Classifier [44] with a score filtering threshold
    of 0.5. Rarefaction curves were calculated based on OTU counts
    using the rarefaction.single routine of the Mothur package [45].
    Hierarchical clustering, boxplots, and statistical calculations
    (Wilcoxon rank sum tests, Jensen-Shannon divergence etc.) were
    performed in R. Differentially abundant OTUs were determined
    with Metastats [46]. Phylogenetic trees were created with
    FastTree2 [47] using trimmed alignments generated with NAST.
    Dot plots to evaluate phylogenetic distances and Jensen-Shannon
    divergence between sample pairs and changes in relative
    abundance of specific taxonomic families over time were
    generated with Prism5 (version 6 for Mac, GraphPad Software,
    San Diego CA, USA).
    Results and Discussion
    Patient population, sample set and sequence data
    For this longitudinal study, fecal samples were collected from 14
    pairs of RCDI patients, treated successfully by FMT, and their
    respective donors (Fig. 1). In addition to the 14 donor samples used
    for FMT, 11 samples from pre-FMT RCDI patients and 17
    samples from eight post-FMT patient samples, as well as 14
    samples from eight healthy donors collected after FMT were
    Table 1. RCDI patient study population.
    Case [#] Sex Age
    RCDI duration
    [months] Donor
    Time to resolution of
    symptoms [days] Follow up [months] Inciting antibiotic
    1 F 65 18 Husband 2 26 Beta-lactam1 + lincosamide2
    2 F 65 6 Husband 3 21 multiple
    3 F 61 5 Friend 2 22 Lincosamide2
    4 F 56 12 Friend 3 19 Fluoroquinolones
    5 F 76 72 Friend 2 7 Fluoroquinolones
    6a* F 57 8 Son 3 18 Fluoroquinolones
    6b* 2 Brother 4 Fluoroquinolones3
    8 F 72 5 Daughter 3 17 Unknown
    9 F 63 6 Husband 2 17 Lincosamide2 +
    fluoroquinolone4
    10 F 61 11 Husband 3 17 Clindamycin
    11 M 68 6 Wife 3 16 Unknown
    12 F 41 12 Husband 2 16 Lincosamide2
    13 F 79 12 Husband 3 12 Unknown
    14 M 57 4.5 Wife 2 12 Unknown
    *#6a had a relapse of RCDI one month after successful FMT and received a second FMT three months after the first (#6b). In the NCBI short read archive, samples
    referred to as #6b are designated as #7 samples.
    1Penicillin; 2 clindamycin; 3 ciprofloxacin; 4 levofloxacin.
    doi:10.1371/journal.pone.0081330.t001
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 3 November 2013 | Volume 8 | Issue 11 | e81330
    analyzed, collected between one week and one year after the
    procedure, (total number of samples: 56). This allowed us to
    perform the first characterization of long-term microbiota changes
    in patients after FMT. All treated RCDI patients experienced
    resolution of diarrheal symptoms within 2–3 days after FMT
    (Table 1), in accordance with previous reports [27]. Of the post-
    FMT samples collected from asymptomatic patients, 14 were
    paired with donor samples collected at the same time points to
    serve as a control for intra-individual, longitudinal variations not
    associated with RCDI. RCDI patient #6a was successfully treated
    by FMT but experienced recurrence of C. difficile infection one
    month later, after being treated for a urinary tract infection with
    ciprofloxacin. Subsequent oral vancomycin and intravenous
    immunoglobulin therapy did not resolve the problem. The patient
    #6a was treated successfully for a second time by FMT, three
    months after the first FMT (designated as case #6b). Selected
    characteristics of all cases for which samples were analyzed are
    summarized in Table 1. Additional clinical aspects of this study
    have been described in a separate publication [48] FMT donors
    for this study were chosen by the RCDI patients and included
    genetically unrelated individuals living in the same household (8x
    spouses), as well as genetically related (2x children) or unrelated (3x
    friends) individuals living in households separate from those of the
    RCDI patients (Table 1). On average, 3,315 sequence reads were
    obtained per sample using the Roche/454 GS FLX Titanium
    platform (average sequence length: 527 bp). A list of read numbers
    and identified operational taxonomic units (OTUs) for each of the
    samples is part of the supplement (Table S1).
    Reduced microbiota diversity in RCDI patients increases
    after FMT
    Reduced microbiota diversity associated with C. difficile infection
    is reported in humans [49-51] and mice [52,53]. This finding was
    confirmed in our study with multiple post-FMT samples collected
    up to one year after the procedure. Compared to healthy donors
    the fecal microbiota diversity of RCDI patients was reduced, as
    shown by rarefaction analysis of OTU counts (Fig. 2). Microbiota
    diversity increased significantly in post-FMT patient samples, as
    demonstrated by Shannon diversity index calculations (p,0.01,
    Wilcoxon rank sum test) between RCDI (mean 1.686 0.75) and
    post-FMT (mean 3.376 0.46) patient samples (Fig. 3). Microbial
    richness was also increased in post-FMT compared to RCDI
    patient samples, based on the comparison of mean ACE indices
    (46%; p , 0.001). Interestingly, no significant difference in
    microbial diversity or richness was noted between post-FMT
    patient and donor samples as determined by Shannon and ACE
    indices. Shannon diversity increased in all 17 post-FMT patients as
    soon as one week after FMT and remained stable and comparable
    among different patients for up to one year afterwards (Fig. S1).
    Compared to the RCDI sample collected before the first FMT
    treatment (#6a_P0), microbial diversity in the RCDI sample from
    the same patient collected three months later after RCDI relapse
    (#6b_P0) showed a 2-fold increase based on the Shannon index
    but was still low compared to healthy donor samples (Fig. 3).
    These results suggest that FMT restores the reduced microbiota
    diversity associated with RCDI. Furthermore, diversity increases
    immediately after FMT and remains stable over time.
    FMT shifts fecal microbiota towards healthy donor
    composition
    To gain further insights into the effects of FMT on the patient
    microbiota, shared OTUs between RCDI patients, post-FMT
    patients and healthy donor samples were determined (Fig. S2).
    Using a threshold of at least five supporting reads across all 38
    samples for OTUs to be considered in the comparison, a total of
    1,321 OTUs were identified of which 876 (65%) were only
    identified in post-FMT patient and healthy donor samples but
    never in RCDI patient samples. This finding could be interpreted
    to indicate that post-FMT patients acquired donor OTUs as a
    consequence of FMT. However, the applied analysis has a
    detection limit of approximately 0.03% and does not allow for
    the distinction of different bacterial strains from the same OTU. It
    is therefore impossible to distinguish between OTUs that might
    have been present in RCDI patients below the detection limit and
    those that were acquired from the donors.
    Microbiota compositions were analyzed based on phylogenetic
    distance calculations between samples using the unweighted, i.e.,
    comparing OTU presences/absences, and weighted, i.e., including
    quantitative information about detected OTUs, UniFrac metric
    (Fig. 4). Principal coordinate analyses (PCoA) of the unweighted
    UniFrac comparison showed that most of the compositional
    variation among samples is accounted for by post-FMT patient
    and healthy donor samples (Fig. 4A). In contrast, when OTU
    abundance is also taken into consideration (weighted UniFrac
    analysis) most of the variation within the entire sample set is
    observed among RCDI patient samples (Fig. 4B), suggesting that
    relative abundances of major microbiota members can vary
    substantially not only between RCDI patient and healthy donor
    samples but also among different RCDI patient samples.
    In most cases, FMT resulted in the adoption of a fecal
    microbiota composition in post-FMT samples that was similar to
    that of healthy donors. This is apparent in the clustering of post-
    FMT patient and healthy donor samples in unweighted UniFrac
    analysis (Fig. 4A). However, several patients appeared to at least
    temporarily return to pre-FMT fecal microbiota composition
    states (e.g., Patient #8 at 5 months and Patient #14 at 3 weeks
    after FMT), although all treated patients were reported to be
    symptom-free within 2–3 days after FMT. The adoption of a fecal
    microbiota composition in post-FMT patient samples similar to
    Figure 2. Microbiota rarefaction curves showing fecal microbiota
    diversity in RCDI (red) and post-FMT (blue) patient and
    donor (green) samples. Each curve shows the average number of
    OTUs found in a given number of sampled sequences. OTUs can be
    treated as equivalent to taxonomic species in the sequence space. RCDI
    samples are marked from patient #6a (*), who experienced antibioticinduced
    relapse and was treated by FMT again as patient #6b (**).
    doi:10.1371/journal.pone.0081330.g002
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 4 November 2013 | Volume 8 | Issue 11 | e81330
    that of healthy donors was also supported by comparing mean
    phylogenetic UniFrac distances. These were significantly larger
    between RCDI and post-FMT patient samples than between post-
    FMT and donor samples both in unweighted (p,0.05) and
    weighted (p,0.01) UniFrac analysis.
    Interestingly, the RCDI sample from the patient (#6a/b), who
    relapsed after unrelated antibiotic treatment, showed a microbiota
    composition that was similar to that of other post-FMT and
    healthy donor samples, especially in the weighted UniFrac analysis
    (Fig. 4). This second RCDI episode lasted only two months and
    included treatment with a single antibiotic (vancomycin) compared
    to 4.5–72 months duration and at least three different antibiotic
    treatments in other RCDI patients, It is therefore possible that
    several of the phenotypes observed in other RCDI samples are
    reflective of long-term disease and multiple antibiotic treatment
    courses. The data presented here suggest that RCDI is associated
    with the presence or absence of specific fecal microbiota members
    (i.e., co-clustering of all RCDI samples in unweighted UniFrac
    analysis, including #6b_P0), rather than significant changes in the
    relative abundance of major microbiome components (i.e.,
    separate clustering of different RCDI samples and of #6b_P0
    with healthy donor samples in weighted UniFrac analysis), which
    could represent a consequence of long-term disease.
    FMT affects predominantly Firmicutes and
    Proteobacteria
    The identification of specific microbiota members associated
    with RCDI and successful FMT treatment bears the potential to
    identify new diagnostic markers to predict susceptibility to C.
    difficile infection or infection relapse in at-risk populations. In
    addition, this knowledge may provide the insights required to
    assemble culture-based “probiotic” bacterial mixtures as substitutes
    for transplantation of fecal samples, as has recently been
    demonstrated in humans [54] and the mouse model [55]. Towards
    this goal, the relative abundances of all identified microbial taxa
    were compared between RCDI and post-FMT patient and healthy
    donor sample groups using Metastats [46]. Among these three
    groups, bacteria from only three taxonomic orders, belonging to
    two phyla, showed significant changes, i.e., Clostridiales and
    Figure 3. Microbiota diversity (Shannon) and richness (ACE) of
    RCDI and post-FMT patient and donor samples. (A) Shannon
    index; (B) ACE index. Significant differences are shown (*, p,0.01; **,
    p,0.001) as measured by Wilcoxon rank sum test. RCDI samples from
    patient #6a (+), who experienced antibiotic-induced relapse and was
    treated by FMT again as patient #6b (++) are marked.
    doi:10.1371/journal.pone.0081330.g003
    Figure 4. Unscaled principal coordinate analysis (PCoA) plots showing unweighted (A) and weighted (B) UniFrac analysis of RCDI
    (red) and post-FMT (blue) patient and healthy donor (green) samples. RCDI patient samples are circled in red. RCDI samples from patient
    #6a (*), who experienced antibiotic-induced relapse and was treated by FMT again as patient #6b (**) are marked in dark red. Sample names
    indicate case numbers, patient or donor source and time point of collection (“0” time point refers to pre-FMT sampling time points; other time points
    are abbreviated as weeks [w], months [m] and year [y]).
    doi:10.1371/journal.pone.0081330.g004
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 5 November 2013 | Volume 8 | Issue 11 | e81330
    Lactobacillales (both from phylum Firmicutes) and Enterobacteriales
    (phylum Proteobacteria) (Fig. 5). Clostridiales, which include
    the species C. difficile, were present at only 12.8% in RCDI patient
    samples and significantly increased in post-FMT samples (55%)
    but still remained lower compared to healthy donor samples (70%)
    (p,0.001, unpaired t-test with unequal variance). Lactobacillales,
    which were present at high abundance in RCDI patient samples
    (mean: 58%), were significantly decreased in post-FMT patient
    (22%) and healthy donor (5%) samples. However, abundance of
    Lactobacillales remained higher in post-FMT patient compared to
    donor samples (p,0.01). Enterobacteriales, present at 6.5% in
    RCDI patient samples, were less than 1% in post-FMT patient
    and donor samples (p,0.001).
    Three taxonomic families within the order Clostridiales
    (phylum: Firmicutes) significantly increased in relative abundance
    between RCDI and post-FMT patient samples (p,0.01), Lachnospiraceae,
    Peptostreptococcaceae, and Ruminococcaceae (Fig. 6). Most
    prominently, an uncharacterized genus within the Lachnospiraceae
    family (Lachnospiraceae Incertae Sedis) increased from on average
    3% in RCDI patient samples to 30% in post-FMT patient samples
    and was 39% in healthy donor samples (p,0.01). The dominant
    OTU within this genus (99% identical to GenBank Acc.-No.:
    EF399262) was identified in all 28 donor samples (27 samples with
    .4 reads), 15 out of 17 post-FMT patient samples (14 samples
    with .4 reads), and 8 out of 11 RCDI patient samples (#6b was
    the only sample with .4 reads). C. difficile is a member of the
    Peptostreptococcaceae [56], which increased in patients after FMT.
    Moreover, an unknown genus within this family accounts for .2%
    of the fecal microbiota in healthy donor samples (Fig. 6),
    demonstrating that taxonomically close relatives of C. difficile exert
    non-pathogenic or even beneficial functions in the healthy
    intestinal microbiota.
    Within the orders Lactobacillales (phylum: Firmicutes) and
    Enterobacteriales (phylum: Proteobacteria), the genera Enterococcus
    and Klebsiella, which were present on average at 18% and 4% in
    RCDI patient samples, respectively, were significantly reduced to
    less than 0.1% in post-FMT patient samples (p,0.01). Members of
    the Streptococcaceae (phylum: Firmicutes), the dominant taxonomic
    family in RCDI patient samples (mean: 30.1%), were reduced on
    average by more than 10% after FMT, although this change was
    not statistically significant due to large variations between RCDI
    patients. With the exception of the genus Streptococcus, none of these
    families or genera showed significant differences in relative
    abundance between post-FMT patient and healthy donor samples
    (p,0.05). Streptococcus was the only genus with a significant
    difference in relative abundance between both RCDI patient
    and donor samples and between post-FMT patient and donor
    samples. As post-FMT patients appear to show increased
    susceptibility to C. difficile infection compared to healthy donors,
    if additional antibiotic medication to treat unrelated infections
    becomes necessary [27], the increased abundance of the Streptococcus
    genus in this population could play a role for this
    susceptibility. However, not all RCDI samples contained high
    counts of Streptococcus sequences (range: 0.1% to 82.4%). In
    general, different RCDI samples showed more variation in the
    abundance of microbiota members that were increased relative to
    healthy donors (e.g., Enterococcaceae and Streptococcaceae) than of
    microbiota members that were reduced (see error bars in Fig. 6).
    This may suggest that the second group provides a better target for
    the identification of diagnostic markers for RCDI (e.g., among the
    Lachnospiraceae, Peptostreptococcaceae, and Ruminococcaceae).
    In contrast to all other cases, the fecal RCDI microbiota from
    patient #6b, who experienced antibiotic-induced relapse of C.
    difficile infection, contained large fractions of Lachnospiraceae (11%
    compared to no detection before the first FMT and on average 1%
    in other RCDI samples) and Akkermansia (60% compared to on
    average 0.1% in other RCDI samples and 1.8% in healthy donor
    samples) (Fig. S3). This atypical composition could be responsible
    for the clustering of this sample with healthy donor and post-FMT
    patient samples in the weighted UniFrac analysis (Fig. 4B). It is
    therefore possible that the reductions in Lachnospiraceae characteristic
    of the other RCDI samples, rather than being a cause of
    disease susceptibility, represent an effect of disease duration and
    number of antibiotic treatment regimens exceeding those that
    patient #6b experienced after recurrence. Interestingly, Akkermansia
    spp. have recently received special attention in human
    microbiome research because of their ability to colonize the
    intestinal mucosa and to utilize mucus as a sole carbon and
    nitrogen source [57,58]. While A. municiphila has been proposed as
    a marker of a healthy intestine, due to its production of short chain
    fatty acids and its negative correlation with inflammatory bowel
    diseases, appendicitis and obesity (reviewed here:[58]), its high
    abundance in the fecal sample of patient #6b might also be an
    indicator of high concentrations of mucus in the stool, which could
    be the result of acute diarrhea.
    The fecal microbiota continues to change in
    asymptomatic post-FMT patients
    Asymptomatic post-FMT patients appear to be at higher risk for
    recurrence of C. difficile infection compared to patients without a
    history of RCDI, if additional antibiotic medication to treat
    unrelated infections becomes necessary [27]. Whether specific
    microbiota features, such as the increased abundance of Streptococcus
    in post-FMT patient compared to healthy donor samples,
    are responsible for this susceptibility is unknown, but the
    susceptibility of post-FMT patients to RCDI may decrease over
    time and little is known about the long-term dynamics of FMTinduced
    microbiota changes. In order to characterize microbiota
    changes after FMT over time, fecal samples from post-FMT
    patients, all of which were asymptomatic with respect to RCDI,
    were compared longitudinally. Microbiota diversity in post-FMT
    patient samples did not change significantly over time, as
    measured by comparing the Shannon diversity index (Fig. S1).
    Figure 5. Microbiota changes between RCDI and post-FMT
    patient and healthy donor sample groups at the taxonomic
    order level. Significant differences between sample groups as
    calculated with the Metastats tool are marked with asterisks (p,0.01).
    doi:10.1371/journal.pone.0081330.g005
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 6 November 2013 | Volume 8 | Issue 11 | e81330
    To study changes in microbiota composition over time, weighted
    and unweighted UniFrac distances and the Jensen-Shannon
    divergence were calculated between (i) RCDI and post-FMT
    patient sample pairs, (ii) donor and post-FMT patient samples
    pairs and, as a control for temporal variations in healthy
    individuals, between (iii) sample pairs collected from the same
    donor before and after FMT (Fig. 7). For the comparison of post-
    FMT and RCDI patient samples, both unweighted UniFrac and
    Jensen-Shannon distance metrics displayed a significant linear
    change over time when plotted on a logarithmic scale. However,
    comparison of post-FMT patient and donor samples or of donor
    samples collected before and after FMT did not. That this
    correlation is only apparent if temporal changes are plotted on a
    logarithmic scale shows that the most significant changes happen
    immediately after FMT and that the microbiota continues to
    evolve over time albeit at a decreasing rate.
    Individual taxonomic families showed similar trends in post-
    FMT patients over time, if compared case-by-case, i.e. increases in
    Lachnospiraceae and Ruminococcaceae and decreases in Streptococcaceae
    (Fig. 8). However, in contrast to changes in relative abundance
    between the pre- and post-FMT patient microbiota (Fig. 6),
    changes in post-FMT patients over time were not significant for
    the three studied Firmicutes families. This suggests that, while
    changes in the abundance of Lachnospiraceae and/or Streptococcaceae
    might play important roles for RCDI or successful recovery after
    FMT in some patients, general post-FMT microbiota dynamics
    across the entire patient population are better described using
    metrics that take account of the microbiota as a whole, i.e.,
    UniFrac distances and Jensen-Shannon divergence.
    ‘Keystone’ species are not identified in RCDI or FMT
    The concept of keystone species has been used to describe the
    disproportionate importance of a single or a few organisms for the
    structure or function of an entire environment [59,60], e.g. in the
    oral cavity where colonization with the commensal bacterium
    Porphyromonas gingivalis even at low abundance can play a major
    role for microbiota changes associated with periodontitis [61]. In
    the context of RCDI and FMT, keystone bacteria could be crucial
    for the identification of diagnostic markers to predict susceptibility
    to C. difficile infection and as substitutes for fecal samples of largely
    unknown composition to be used in transplantation. That RCDI
    can principally be treated by transplantation of in vitro-assembled
    microbial communities instead of fecal material was shown
    recently in humans [54] and mice [52], although little justification
    was provided for the selection of specific bacterial species or
    strains. While, based on our findings and previous data, members
    of the Lachnospiraceae family, for example, might present themselves
    as keystone candidates [50,62,63], at least one case was found in
    our cohort where RCDI was associated with relatively high counts
    of Lachnospiraceae (i.e., #6b). In another case (#9), Lachnospiraceae
    did only increase temporarily six weeks after FMT but dropped to
    pre-FMT levels 12 weeks after FMT. Khoruts et al. found a
    Figure 6. Microbiota changes between RCDI and post-FMT patient and healthy donor sample groups at the taxonomic family and
    genus levels. Significant differences between sample groups as calculated with the Metastats tool are marked with asterisks (p,0.01). Note that
    standard deviations are smaller for genera that increased in post-FMT relative to RCDI patient samples (e.g., Lachnospiraceae Incertae Sedis)
    compared to those that decreased (e.g. Streptococcus), which reflects differences in the relative abundances of major microbiota members among
    RCDI patient samples.
    doi:10.1371/journal.pone.0081330.g006
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 7 November 2013 | Volume 8 | Issue 11 | e81330
    relatively large proportion (.5%) of Lachnospiraceae Inc. Sed. in an
    RCDI sample before FMT treatment [25]. Interestingly, the
    dominant representative of the genus Lachnospiraceae Inc. Sed.
    associated with successful FMT treatment, which was identified in
    the Canadian study by Shahinas et al. [50], is different from the
    one identified here (Shahinas: 97% identical to GenBank Acc.-No.
    JX230866, compared to this study: 99% identical to EF399262).
    This difference could either result from variations in the applied
    pyrosequencing protocols (e.g., Shahinas et al. used primers
    specific for hypervariable regions V5–V6 instead of primers
    specific for V1–V3 used here) or indicate that different species or
    strains of the genus Lachnospiraceae Inc. Sed. circulate in U.S. and
    Canadian human populations. In any case, it seems as if neither
    RCDI nor FMT are associated with the presence or absence of a
    single specific microbiota fraction.
    Figure 7. Post-FMT microbiota changes. Unweighted (A) and
    weighted (B) UniFrac distances and Jensen-Shannon divergence (C)
    metrics were calculated between post-FMT and RCDI patient sample
    pairs (red), post-FMT patient and donor sample pairs (green) and
    between donor sample pairs collected over time (blue) and plotted on
    logarithmic scales. R2 values and p-values to establish whether the
    slope of the curve was significantly different from zero are shown with
    asterisks indicating significance (p,0.05, F-test). The 20-week data
    point of patient #8 was classified as outlier and not included in the
    analyses, based on the Bonferroni-adjusted outlier test, and is shown
    with parentheses. One-year time points (patient and donor #1) were
    also classified as outliers and omitted from the analysis and plot. A plot
    showing all data points including those omitted is part of the
    supplement (Fig. S4).
    doi:10.1371/journal.pone.0081330.g007
    Figure 8. Post-FMT changes in selected microbiota members
    by case (genus level). (A) Lachnospira Incertae Sedis; (B) Ruminococcus;
    (C) Streptococcus. Genus-specific changes in relative abundance
    over time were not significant (p.0.05)when samples were grouped by
    time periods (1 week, 2–4 weeks, 6–8 weeks, 12–20 weeks) and groups
    compared with a non-parametric statistical test (Wilcoxon rank sum
    test).
    doi:10.1371/journal.pone.0081330.g008
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 8 November 2013 | Volume 8 | Issue 11 | e81330
    Instead of bacterial keystone taxa, specific microbial microbiota
    genes or transcripts could be associated with health and disease
    and, thus, serve as “keystone functions” with potential as
    diagnostic markers. A redundancy and similarity of functional
    microbiota compositions between individuals despite significant
    taxonomic variation has previously been demonstrated for the
    healthy human microbiota [64]. These functions could be
    predominantly but not exclusively associated with certain members
    of the fecal microbiota, which would then still show statistical
    correlations with health and disease states. Short-chain fatty acid
    (SCFA) production plays an important role in the regulation of
    intestinal inflammatory processes [65] and intestinal barrier
    maintenance [66–68] and has been discussed in the context of
    RCDI, as C. difficile infection in the mouse model was shown to
    alter SCFA profiles [52]. Consequently, the reduction of
    Lachnospiraceae and Ruminococcaceae has been interpreted as a
    depletion in butyrate-producing bacteria [51]. Shotgun sequencing
    of total metagenomic DNA and/or metatranscriptomic RNA
    isolates will be needed to confirm the lack of butyrate production
    in the fecal RCDI microbiota or to associated other “keystone
    functions” with RCDI and FMT.
    Concomitant effects of antibiotics and diarrhea
    Previous RCDI microbiota studies have had difficulty determining
    the chain of events leading to disease as well as the
    relationship between observed microbiota phenotypes and disease.
    C. difficile infection is typically initiated by antibiotic treatment and
    phenotypically characterized by severe diarrhea. Both events by
    themselves have a massive impact on the fecal microbiota
    independent of the disease caused by the C. difficile infection
    [69,70]. It is therefore difficult to distinguish between microbiota
    changes that play a causative role in RCDI and those that simply
    co-occur. The data presented here also include an RCDI patient
    with successful FMT and subsequent relapse of CDI after
    antibiotic treatment, whose fecal microbiota showed characteristics
    described for healthy individuals as opposed to RCDI patients
    (e.g. relatively high Lachnospiraceae abundance). This single patient
    may therefore suggest that multiple rounds of antibiotic treatment
    and/or long-term duration of the disease are needed to induce
    some of the microbiota changes previously reported to be
    associated with CDI. In order to determine the exact time line
    of events, prospective studies are needed starting before antibiotic
    treatment and following patients during the onset and course of
    CDI.
    Conclusion
    In accordance with previous reports, we found a reduction in
    microbiota diversity and richness in fecal samples from RCDI
    patients compared to healthy donors, which was restored after
    FMT. Similarly, our results confirm previous findings that FMT
    changes the RCDI fecal microbiota to become more similar to
    that of healthy donors. We extend current knowledge by
    demonstrating that there are different varieties of dysbiosis in
    RCDI patient samples, that FMT predominantly affects Firmicutes
    and Proteobacteria, and that the fecal microbiota continues
    to change in post-FMT patients. We did not identify a ‘keystone’
    species in RCDI or FMT, but our findings suggest that butyrate
    producing bacteria may be important. We believe that additional
    longitudinal studies, ideally beginning before initial infection and
    including metagenomic and metatranscriptomic analyses, will lead
    to improved outcomes in C. difficile infection.
    Supporting Information
    Figure S1 Fecal microbiota diversity in patient and donor
    samples depending on collection time points. The Shannon index
    of all samples is plotted over time, split into donor (A, blue) and
    patient (B, red) samples.
    (PDF)
    Figure S2 Venn diagram showing shared OTUs between RCDI
    and post-FMT patient and donor samples. Only OTUs represented
    by at least 5 reads across all 56 samples are shown.
    (PDF)
    Figure S3 Microbiota changes between RCDI samples collected
    from the same patient before the first FMT (#6a) and, after
    antibiotic-induced relapse, before the second FMT (#6b). Relative
    abundances of all taxonomic genera (.1%) are shown.
    (PDF)
    Figure S4 Post-FMT microbiota changes. Unweighted (A) and
    weighted (B) UniFrac distances and Jensen-Shannon divergence
    (C) metrics were calculated between post-FMT and RCDI patient
    sample pairs (red), post-FMT patient and donor sample pairs
    (green) and between donor sample pairs collected over time (blue).
    This figures shows that both patient and donor samples from case
    #1 collected one year aft FMT show an unusual small divergence
    (Unweighted/weighted UniFrac distances and Jensen-Shannon
    divergence) from the donor sample collected before FMT.
    (PDF)
    Table S1 Numbers of reads and identified operational taxonomic
    units (OTUs) by sample.
    (XLSX)
    Acknowledgments
    We are grateful for generous support from the Weinberg Foundation, the
    Friedman and Friedman Group and Eric Cowan.
    Author Contributions
    Conceived and designed the experiments: YS SD WFF. Performed the
    experiments: YS SG MG CM AD SD. Analyzed the data: YS WFF.
    Contributed reagents/materials/analysis tools: SD WFF. Wrote the paper:
    YS ECvR SD WFF.
    References
    1. Bartlett JG (2002) Clinical practice. Antibiotic-associated diarrhea. N Engl J
    Med 346: 334–339.
    2. Jobe BA, Grasley A, Deveney KE, Deveney CW, Sheppard BC (1995)
    Clostridium difficile colitis: an increasing hospital-acquired illness. Am J Surg
    169: 480–483.
    3. Miller BA, Chen LF, Sexton DJ, Anderson DJ (2011) 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
    32: 387–390.
    4. Rivera EV, Woods S (2003) Prevalence of asymptomatic Clostridium difficile
    colonization in a nursing home population: a cross-sectional study. J Gend Specif
    Med 6: 27–30.
    5. Jangi S, Lamont JT (2010) Asymptomatic colonization by Clostridium difficile in
    infants: implications for disease in later life. J Pediatr Gastroenterol Nutr 51: 2–7.
    6. McFarland LV, Elmer GW, Surawicz CM (2002) Breaking the cycle: treatment
    strategies for 163 cases of recurrent Clostridium difficile disease. Am J
    Gastroenterol 97: 1769–1775.
    7. Pepin J, Routhier S, Gagnon S, Brazeau I (2006) Management and outcomes of
    a first recurrence of Clostridium difficile-associated disease in Quebec, Canada.
    Clin Infect Dis 42: 758–764.
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 9 November 2013 | Volume 8 | Issue 11 | e81330
    8. Surawicz CM, McFarland LV, Greenberg RN, Rubin M, Fekety R, et al. (2000)
    The search for a better treatment for recurrent Clostridium difficile disease: use
    of high-dose vancomycin combined with Saccharomyces boulardii. Clin Infect
    Dis 31: 1012–1017.
    9. Musher DM, Logan N, Mehendiratta V, Melgarejo NA, Garud S, et al. (2007)
    Clostridium difficile colitis that fails conventional metronidazole therapy:
    response to nitazoxanide. J Antimicrob Chemother 59: 705–710.
    10. Buggy BP, Fekety R, Silva J Jr (1987) Therapy of relapsing Clostridium difficileassociated
    diarrhea and colitis with the combination of vancomycin and
    rifampin. J Clin Gastroenterol 9: 155–159.
    11. Berman AL (2007) Efficacy of rifaximin and vancomycin combination therapy in
    a patient with refractory Clostridium difficile-associated diarrhea. J Clin
    Gastroenterol 41: 932–933.
    12. Johnson S, Schriever C, Galang M, Kelly CP, Gerding DN (2007) Interruption
    of recurrent Clostridium difficile-associated diarrhea episodes by serial therapy
    with vancomycin and rifaximin. Clin Infect Dis 44: 846–848.
    13. Garey KW, Jiang ZD, Bellard A, Dupont HL (2009) Rifaximin in treatment of
    recurrent Clostridium difficile-associated diarrhea: an uncontrolled pilot study. J
    Clin Gastroenterol 43: 91–93.
    14. Johnson S, Schriever C, Patel U, Patel T, Hecht DW, et al. (2009) Rifaximin
    Redux: treatment of recurrent Clostridium difficile infections with rifaximin
    immediately post-vancomycin treatment. Anaerobe 15: 290–291.
    15. Navaneethan U, Venkatesh PG, Shen B (2010) Clostridium difficile infection
    and inflammatory bowel disease: understanding the evolving relationship. World
    J Gastroenterol 16: 4892–4904.
    16. Forster AJ, Taljaard M, Oake N, Wilson K, Roth V, et al. (2012) The effect of
    hospital-acquired infection with Clostridium difficile on length of stay in hospital.
    CMAJ 184: 37–42.
    17. Daneman N, Stukel TA, Ma X, Vermeulen M, Guttmann A (2012) Reduction
    in Clostridium difficile infection rates after mandatory hospital public reporting:
    findings from a longitudinal cohort study in Canada. PLoS Med 9: e1001268.
    18. O’Keefe SJ (2010) Tube feeding, the microbiota, and Clostridium difficile
    infection. World J Gastroenterol 16: 139–142.
    19. Pohl JF (2012) Clostridium difficile infection and proton pump inhibitors. Curr
    Opin Pediatr 24: 627–631.
    20. Sartor RB (2008) Microbial influences in inflammatory bowel diseases.
    Gastroenterology 134: 577–594.
    21. Kelly CP, LaMont JT (2008) Clostridium difficile—more difficult than ever. N
    Engl J Med 359: 1932–1940.
    22. Maroo S, Lamont JT (2006) Recurrent clostridium difficile. Gastroenterology
    130: 1311–1316.
    23. McDonald LC, Killgore GE, Thompson A, Owens RC Jr, Kazakova SV, et al.
    (2005) An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J
    Med 353: 2433–2441.
    24. Lund-Tonnesen S, Berstad A, Schreiner A, Midtvedt T (1998) [Clostridium
    difficile-associated diarrhea treated with homologous feces]. Tidsskr Nor
    Laegeforen 118: 1027–1030.
    25. Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ (2010) Changes in the
    composition of the human fecal microbiome after bacteriotherapy for recurrent
    Clostridium difficile-associated diarrhea. J Clin Gastroenterol 44: 354–360.
    26. Wettstein A, Borody TJ, Leis S, Chongan J, Torres M, et al. (2007) Faecal
    bacteriotherapy – an effective treatment for relapsing symptomatic Clostridium
    difficile infection. 15th Gut United European Gastroenterology Week (France)
    39(Suppl 1): A303 Vienna: United European Gastroenterology Federation.
    27. Brandt LJ, Aroniadis OC, Mellow M, Kanatzar A, Kelly C, et al. (2012) Longterm
    follow-up of colonoscopic fecal microbiota transplant for recurrent
    Clostridium difficile infection. Am J Gastroenterol 107: 1079–1087.
    28. Gough E, Shaikh H, Manges AR (2011) Systematic review of intestinal
    microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium
    difficile infection. Clin Infect Dis 53: 994–1002.
    29. Bakken JS, Borody T, Brandt LJ, Brill JV, Demarco DC, et al. (2011) Treating
    Clostridium difficile infection with fecal microbiota transplantation. Clin
    Gastroenterol Hepatol 9: 1044–1049.
    30. Vrieze A, de Groot PF, Kootte RS, Knaapen M, van Nood E, et al. (2013) Fecal
    transplant: a safe and sustainable clinical therapy for restoring intestinal
    microbial balance in human disease? Best Pract Res Clin Gastroenterol 27: 127–
    137.
    31. Anderson JL, Edney RJ, Whelan K (2012) Systematic review: faecal microbiota
    transplantation in the management of inflammatory bowel disease. Aliment
    Pharmacol Ther 36: 503–516.
    32. Musa S, Thomson S, Cowan M, Rahman T (2010) Clostridium difficile
    infection and inflammatory bowel disease. Scand J Gastroenterol 45: 261–272.
    33. Kump PK, Grochenig HP, Lackner S, Trajanoski S, Reicht G, et al. (2013)
    Alteration of Intestinal Dysbiosis by Fecal Microbiota Transplantation Does not
    Induce Remission in Patients with Chronic Active Ulcerative Colitis. Inflamm
    Bowel Dis 19: 2155–2165.
    34. Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS, et al. (2012)
    Transfer of intestinal microbiota from lean donors increases insulin sensitivity in
    individuals with metabolic syndrome. Gastroenterology 143: 913–916 e917.
    35. Borody TJ, Paramsothy S, Agrawal G (2013) Fecal microbiota transplantation:
    indications, methods, evidence, and future directions. Curr Gastroenterol Rep
    15: 337.
    36. Borody TJ, Khoruts A (2012) Fecal microbiota transplantation and emerging
    applications. Nat Rev Gastroenterol Hepatol 9: 88–96.
    37. U.S. Department of Health and Human Services FaDA, Center for Biologics
    Evaluation and Research (2013) Guidance for Industry: Enforcement Policy
    Regarding Investigational New Drug Requirements for Use of Fecal Microbiota
    for Transplantation to Treat Clostridium difficile Infection Not Responsive to
    Standard Therapies. 1–4.
    38. Aas J, Gessert CE, Bakken JS (2003) Recurrent Clostridium difficile colitis: case
    series involving 18 patients treated with donor stool administered via a
    nasogastric tube. Clin Infect Dis 36: 580–585.
    39. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, et al. (2011) Vaginal
    microbiome of reproductive-age women. Proc Natl Acad Sci U S A 108 Suppl 1:
    4680–4687.
    40. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, et al. (2010)
    QIIME allows analysis of high-throughput community sequencing data. Nat
    Methods 7: 335–336.
    41. Angiuoli SV, Matalka M, Gussman A, Galens K, Vangala M, et al. (2011)
    CloVR: a virtual machine for automated and portable sequence analysis from
    the desktop using cloud computing. BMC Bioinformatics 12: 356.
    42. James White WFF, Cesar Arze, Malcolm Matalka, The CloVR Team, Owen
    White, et al. (2011) CloVR-16S: Phylogenetic microbial community composition
    analysis based on 16S ribosomal RNA amplicon sequencing – standard
    operating procedure, version1.0. Nature Precedings.
    43. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R (2011) UCHIME
    improves sensitivity and speed of chimera detection. Bioinformatics 27: 2194–
    2200.
    44. Wang Q, Garrity GM, Tiedje JM, Cole JR (2007) Naive Bayesian classifier for
    rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl
    Environ Microbiol 73: 5261–5267.
    45. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, et al. (2009)
    Introducing mothur: open-source, platform-independent, community-supported
    software for describing and comparing microbial communities. Appl Environ
    Microbiol 75: 7537–7541.
    46. White JR, Nagarajan N, Pop M (2009) Statistical methods for detecting
    differentially abundant features in clinical metagenomic samples. PLoS Comput
    Biol 5: e1000352.
    47. Price MN, Dehal PS, Arkin AP (2010) FastTree 2—approximately maximumlikelihood
    trees for large alignments. PLoS One 5: e9490.
    48. Dutta SK, Garg S, Dutta A, von Rosenvinge EC, Maddox C, et al. (2013)
    Jejunal and Colonic Fecal Microbiota Transplantation for Recurrent Clostridium
    difficile Infection with genomic analysis. Gastroenterology ID: AJG-13-
    1181submitted.
    49. Skraban J, Dzeroski S, Zenko B, Mongus D, Gangl S, et al. (2013) Gut
    microbiota patterns associated with colonization of different Clostridium difficile
    ribotypes. PLoS One 8: e58005.
    50. Shahinas D, Silverman M, Sittler T, Chiu C, Kim P, et al. (2012) Toward an
    understanding of changes in diversity associated with fecal microbiome
    transplantation based on 16S rRNA gene deep sequencing. MBio 3.
    51. Antharam VC, Li E, Ishmael A, Sharma A, Mai V, et al. (2013) Intestinal
    dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection
    and nosocomial diarrhea. J Clin Microbiol.
    52. Lawley TD, Clare S, Walker AW, Stares MD, Connor TR, et al. (2012)
    Targeted restoration of the intestinal microbiota with a simple, defined
    bacteriotherapy resolves relapsing Clostridium difficile disease in mice. PLoS
    Pathog 8: e1002995.
    53. Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA, et al. (2009)
    Antibiotic treatment of clostridium difficile carrier mice triggers a supershedder
    state, spore-mediated transmission, and severe disease in immunocompromised
    hosts. Infect Immun 77: 3661–3669.
    54. Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, et al. (2012) Stool
    substitute transplant therapy for the eradication of Clostridium difficile infection:
    ‘RePOOPulating’ the gut. Microbiome 1.
    55. Lawley B, Tannock GW (2012) Nucleic acid-based methods to assess the
    composition and function of the bowel microbiota. Gastroenterol Clin North
    Am 41: 855–868.
    56. Ludwig W, Schleifer K-H, Whitman WB (2009) Revised road map to the
    phylum Firmicutes. In: Bergey’s Manual of Systematic Bacteriology, ed., vol. 3
    (The Firmicutes) (P. De Vos, G. Garrity, D. Jones, N.R. Krieg, W. Ludwig, F.A.
    Rainey, K.-H. Schleifer, and W.B. Whitman, eds.). Springer-Verlag, New York:
    1–13.
    57. Derrien M, Vaughan EE, Plugge CM, de Vos WM (2004) Akkermansia
    muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium.
    Int J Syst Evol Microbiol 54: 1469–1476.
    58. Belzer C, de Vos WM (2012) Microbes inside– –from diversity to function: the
    case of Akkermansia. ISME J 6: 1449–1458.
    59. Power ME, Tilman D, Estes JA, Menge BA, Bond WJ, et al. (1996) Challenges
    in the quest for keystones. Bioscience 46: 609–620.
    60. Ebenman B, Jonsson T (2005) Using community viability analysis to identify
    fragile systems and keystone species. Trends Ecol Evol 20: 568–575.
    61. Hajishengallis G, Liang S, Payne MA, Hashim A, Jotwani R, et al. (2011) Lowabundance
    biofilm species orchestrates inflammatory periodontal disease
    through the commensal microbiota and complement. Cell Host Microbe 10:
    497–506.
    62. Hamilton MJ, Weingarden AR, Unno T, Khoruts A, Sadowsky MJ (2013) Highthroughput
    DNA sequence analysis reveals stable engraftment of gut microbiota
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 10 November 2013 | Volume 8 | Issue 11 | e81330
    following transplantation of previously frozen fecal bacteria. Gut Microbes 4:
    125–135.
    63. Reeves AE, Koenigsknecht MJ, Bergin IL, Young VB (2012) Suppression of
    Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated
    with a murine isolate from the family Lachnospiraceae. Infect Immun 80: 3786–
    3794.
    64. Human Microbiome Project C (2012) Structure, function and diversity of the
    healthy human microbiome. Nature 486: 207–214.
    65. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, et al. (2009) Regulation
    of inflammatory responses by gut microbiota and chemoattractant receptor
    GPR43. Nature 461: 1282–1286.
    66. Roediger WE (1982) Utilization of nutrients by isolated epithelial cells of the rat
    colon. Gastroenterology 83: 424–429.
    67. Koruda MJ, Rolandelli RH, Bliss DZ, Hastings J, Rombeau JL, et al. (1990)
    Parenteral nutrition supplemented with short-chain fatty acids: effect on the
    small-bowel mucosa in normal rats. Am J Clin Nutr 51: 685–689.
    68. Peng L, He Z, Chen W, Holzman IR, Lin J (2007) Effects of butyrate on
    intestinal barrier function in a Caco-2 cell monolayer model of intestinal barrier.
    Pediatr Res 61: 37–41.
    69. Dethlefsen L, Relman DA (2011) Incomplete recovery and individualized
    responses of the human distal gut microbiota to repeated antibiotic perturbation.
    Proc Natl Acad Sci U S A 108 Suppl 1: 4554–4561.
    70. Gorkiewicz G, Thallinger GG, Trajanoski S, Lackner S, Stocker G, et al. (2013)
    Alterations in the colonic microbiota in response to osmotic diarrhea. PLoS One
    8: e55817.
    Post-Fecal Transplant Microbiota Characterization
    PLOS ONE | www.plosone.org 11 November 2013 | Volume 8 | Issue 11 | e81330

                                                                                                                                      Order Now