заявка
№ US 20160158295
МПК A61K35/744

Compositions and Methods

Авторы:
Noubar B. Afeyan Mary-Jane Lombardo McKenzie Kevin Daniel Litcofsky
Все (18)
Номер заявки
14765812
Дата подачи заявки
04.02.2014
Опубликовано
09.06.2016
Страна
US
Дата приоритета
13.12.2025
Номер приоритета
Страна приоритета
Как управлять
интеллектуальной собственностью
Чертежи 
7
Реферат

Provided are defined bacterial compositions for the maintenance or restoration of a healthy microbiota in the gastrointestinal tract of a mammalian subject, and methods for populating the gastrointestinal tract of a subject. Provided also are bacterial formulations for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition.

Формула изобретения

1. A therapeutic composition comprising a bacterial population comprising at least three but fewer than nine bacterial strains selected from the group consisting of Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition.

2. The composition of claim 1, wherein at least one of the bacterial strains is not a colonic bacterium.

3. The composition of claim 1, wherein at least one of the bacterial strains is not obtained from a fecal culture.

4. The composition of claim 1, wherein at least one Bacteroides species is detectably present in the mammalian subject prior to administration of the composition.

5. The composition of claim 1, wherein at least one Bacteroides species is not detectably present in the mammalian subject prior to administration of the composition, but is detectably present in the mammalian subject at least one hour after administration of the composition.

6. The composition of claim 1, wherein the mammalian subject has not received at least two doses of vancomycin, metronidazole and/or or similar antibiotic compound within one week prior to administration of the therapeutic composition.

7. The composition of claim 1, wherein a single administration is substantially effective to reduce C. difficile and/or C. difficile toxin content in a mammalian subject to whom the composition is administered.

8. The composition of claim 1, wherein one strain of E. coli is present in amounts at least 2, 5, 10, 50, 100 or more than 100 times greater than any other strain of E. coli present in the composition.

9. The composition of claim 1, wherein at least three bacterial strains are not present in the composition in equal ratios.

10. The composition of claim 1, wherein at least three bacterial strains are present in a ratio equivalent to the ratio of the bacterial strains in a reference mammalian subject.

11. The composition of claim 1, wherein at least one of the bacterial strains is provided in a concentration of greater than 1×109viable bacteria per gram of composition

12. The composition of claim 1, wherein at least one of the bacterial strains is provided in a concentration of less than 1×108viable bacteria per gram of composition.

13. The composition of claim 1, wherein a plurality of the bacterial strains is provided in a concentration of less than 1×108viable bacteria per gram of composition.

14. The composition of claim 1, comprising no more than nine different bacterial strains, wherein at least two strains are from the group selected from: Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroided ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta

15. The composition of claim 1, formulated for oral administration as a solid, semi-solid, gel, or liquid form.

16. The composition of claim 1, formulated in the form of a pill, tablet, capsule, or lozenge.

17. The composition of claim 1, wherein the bacterial strains are substantially encapsulated in an enteric coating.

18. The composition of claim 1, wherein the bacterial strains are substantially inactive prior to localization in the gastrointestinal tract of a mammalian subject to whom the composition is administered.

19. The composition of claim 1, wherein no more than two of the bacterial strains are strains whose growth is substantially inhibited by an equivalent amount of C. difficile bacteria.

20. A method of treating a mammalian subject suffering from or at risk of developing a gastrointestinal disease, disorder or condition, comprising the step of administering to the mammalian subject the therapeutic composition of claim 1.

21.-43. (canceled)

Описание

RELATED APPLICATIONS

[0001]

This application is related to U.S. Provisional Application No. 61/760,584, filed on Feb. 4, 2013, and to U.S. Provisional Application No. 61/760,585, filed on Feb. 4, 2013, and to U.S. Provisional Application No. 61/760,574, filed on Feb. 4, 2013, and to U.S. Provisional Application No. 61/760,606, filed on Feb. 4, 2013, and to U.S. Provisional Application No. 61/926,928, filed on Jan. 13, 2014, which are each incorporated by reference in its entirety for all purposes.

REFERENCE TO A SEQUENCE LISTING

[0002]

This application includes a Sequence Listing submitted electronically as a text file named 25968PCT_CRF_sequencelisting.txt, created on Feb. 4, 2014, with a size of 911,051 bytes. The sequence listing is incorporated by reference.

BACKGROUND

[0003]

Mammals are colonized by microbes in the gastrointestinal (GI) tract, on the skin, and in other epithelial and tissue niches such as the oral cavity, eye surface and vagina. The gastrointestinal tract harbors an abundant and diverse microbial community. It is a complex system, providing an environment or niche for a community of many different species or organisms, including diverse strains of bacteria. Hundreds of different species may form a commensal community in the GI tract in a healthy person, and this complement of organisms evolves from the time of birth to ultimately form a functionally mature microbial population by about 3 years of age. Interactions between microbial strains in these populations and between microbes and the host, e.g. the host immune system, shape the community structure, with availability of and competition for resources affecting the distribution of microbes. Such resources may be food, location and the availability of space to grow or a physical structure to which the microbe may attach. For example, host diet is involved in shaping the GI tract flora.

[0004]

A healthy microbiota provides the host with multiple benefits, including colonization resistance to a broad spectrum of pathogens, essential nutrient biosynthesis and absorption, and immune stimulation that maintains a healthy gut epithelium and an appropriately controlled systemic immunity. In settings of ‘dysbiosis’ or disrupted symbiosis, microbiota functions can be lost or deranged, resulting in increased susceptibility to pathogens, altered metabolic profiles, or induction of proinflammatory signals that can result in local or systemic inflammation or autoimmunity. Thus, the intestinal microbiota plays a significant role in the pathogenesis of many diseases and disorders, including a variety of pathogenic infections of the gut. For instance, subjects become more susceptible to pathogenic infections when the normal intestinal microbiota has been disturbed due to use of broad-spectrum antibiotics. Many of these diseases and disorders are chronic conditions that significantly decrease a subject's quality of life and can be ultimately fatal.

[0005]

Manufacturers of probiotics have asserted that their preparations of bacteria promote mammalian health by preserving the natural microflora in the GI tract and reinforcing the normal controls on aberrant immune responses. See, e.g., U.S. Pat. No. 8,034,601. Probiotics, however, have been limited to a very narrow group of genera and a correspondingly limited number of species; as such, they do not adequately replace the missing natural microflora of the GI tract in many situations.

[0006]

Thus, there is a need for a method of populating a subject's gastrointestinal tract with a diverse and useful selection of microbiota in order to alter a dysbiosis. In response to the need for durable, efficient, and effective compositions and methods for treatment of GI diseases by way of restoring or enhancing microbiota functions, Applicants address these and other shortcomings of the art by providing compositions and methods for treating subjects.

SUMMARY OF THE INVENTION

[0007]

Disclosed herein are therapeutic compositions comprising a bacterial population comprising at least three but fewer than nine bacterial strains selected from the group consisting of Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition. In some embodiments, the bacterial strains are not a colonic bacterium and/or are not obtained from a fecal culture. In some embodiments, at least one Bacteroides species is detectably present in the mammalian subject prior to administration of the composition or, in other embodiments at least one Bacteroides species is not detectably present in the mammalian subject prior to administration of the composition, but is detectably present in the mammalian subject at least one hour after administration of the composition. In some embodiments, the mammalian subject has not received at least two doses of vancomycin, metronidazole and/or or similar antibiotic compound within one week prior to administration of the therapeutic composition. In some embodiments, a single administration is substantially effective to reduce C. difficile and/or C. difficile toxin content in a mammalian subject to whom the composition is administered. In some embodiments, one strain of E. coli is present in amounts at least 2, 5, 10, 50, 100 or more than 100 times greater than any other strain of E. coli present in the composition. In some embodiments, at least three bacterial strains are not present in the composition in equal ratios or are present in a ratio equivalent to the ratio of the bacterial strains in a reference mammalian subject. In some embodiments, at least one of the bacterial strains is provided in a concentration of greater than 1×109 viable bacteria per gram of composition or is provided in a concentration of less than 1×108 viable bacteria per gram of composition. In some embodiments, a plurality of the bacterial strains is provided in a concentration of less than 1×108 viable bacteria per gram of composition. In some embodiments, no more than nine different bacterial strains are used, wherein at least two strains are from the group selected from: Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroided ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia product. In some embodiments, the composition is formulated for oral administration as a solid, semi-solid, gel, or liquid form; in the form of a pill, tablet, capsule, or lozenge; the bacterial strains are substantially encapsulated in an enteric coating; and/or the bacterial strains are substantially inactive prior to localization in the gastrointestinal tract of a mammalian subject to whom the composition is administered. In some embodiments, no more than two of the bacterial strains are strains whose growth is substantially inhibited by an equivalent amount of C. difficile bacteria.

[0008]

Also disclosed is a method of treating a mammalian subject suffering from or at risk of developing a gastrointestinal disease, disorder or condition, comprising the step of administering to the mammalian subject a therapeutic composition comprising a bacterial population as described herein, wherein the composition is formulated for oral or gastric administration. In some embodiments, the gastrointestinal disease, disorder or condition is selected from the group consisting of relapsing diarrhea caused by C. difficile, ulcerative colitis, colitis, Crohn's disease, and irritable bowel disease. In some embodiments, the mammalian subject is an agricultural mammal. In some embodiments, the bacterial population comprises at least six bacterial strains or comprises at least six but fewer than ten bacterial strains. In some embodiments, the therapeutic composition is administered only once prior to improvement of the disease, disorder or condition; wherein the therapeutic composition is administered at intervals greater than two days; the mammalian subject has not received at least two doses of vancomycin, metronidazole and/or or similar antibiotic compound within one week prior to administration of the therapeutic composition; the mammalian subject is not suffering from relapsing diarrhea caused by C. difficile; and/or the mammalian subject has not previously received an antibiotic compound in the one month prior to administration of the therapeutic composition.

[0009]

Also disclosed is a method of treating a mammalian subject suffering from or at risk of developing a gastrointestinal disease, disorder or condition, comprising the step of administering to the mammalian subject any therapeutic composition disclosed herein.

[0010]

Also disclosed is a method of treating a mammalian subject suffering from or at risk of developing a gastrointestinal disease, disorder or condition, or a metabolic disease, disorder or condition selected from the group consisting of diabetes and autism, comprising the step of administering to the mammalian subject a therapeutic composition comprising a bacterial population comprising at least three bacterial strains, wherein no greater than nine of the bacterial strains are members of a bacterial species selected from the group consisting of Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroided ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta, wherein the composition is formulated for oral or gastric administration. In some embodiments, the method further comprises the step of detecting one or more of the administered bacterial strains in the mammalian subject subsequent to administration, for example, detecting a 16S nucleic acid sequence in at least one administered bacterial strain.

[0011]

Also disclosed is a therapeutic composition comprising a bacterial population comprising at least three but fewer than nine bacterial strains, wherein the bacterial strains comprise 16S nucleic acid sequences at least 97% identical to reference 16S nucleic acid sequences in bacterial strains selected from the group consisting of Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition.

[0012]

Also disclosed is a therapeutic composition comprising a bacterial population comprising at least three but fewer than nine bacterial strains, wherein the bacterial strains comprise Clostridium innocuum, Clostridium ramosum, and Clostridium bifermentans, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition.

[0013]

Also disclosed is a therapeutic composition comprising a bacterial population comprising at least three but fewer than nine bacterial strains, wherein the bacterial strains do not comprise detectable amounts of Bacteroides ovatus, Bacteroides vulgatus, or Bacteroides thetaiotaomicron, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for prevention or treatment of a gastrointestinal disease, disorder or condition.

[0014]

Also disclosed is a method of increasing diversity of a gastrointestinal microbiota in a mammalian subject in need thereof, comprising administering to the mammalian subject a therapeutic composition comprising a bacterial population comprising at least three but fewer than nine bacterial strains, wherein the bacterial strains comprise 16S nucleic acid sequences at least 97% identical to reference 16S nucleic acid sequences in bacterial strains selected from the group consisting of Escherichia coli, Enterococcus faecalis, Clostridium innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta, wherein the composition is formulated for oral or gastric administration to a mammalian subject in an effective amount for increasing the diversity of the gastrointestinal microbiota of the mammalian subject subsequent to administration. In some embodiments, the diversity is increased for at least 1 day after administration or for at least 4 days after administration or for at least 7 days after administration or for at least 14 days after administration or for at least 21 days after administration. In some embodiments, wherein the mammalian subject is suffering from or at risk of developing a disease, disorder or condition other than a gastrointestinal disease, disorder or condition.

BRIEF DESCRIPTION OF THE DRAWINGS

[0015]

FIG. 1A provides a schematic of 16S rRNA gene and denotes the coordinates of hypervariable regions 1-9 (V1-V9). Coordinates of V1-V9 are 69-99, 137-242, 433-497, 576-682, 822-879, 986-1043, 1117-1173, 1243-1294, and 1435-1465 respectively, based on numbering using E. coli system of nomenclature defined by Brosius et al., Complete nucleotide sequence of a 16S ribosomal RNA gene (16S rRNA) from Escherichia coli, PNAS 75(10):4801-4805 (1978).

[0016]

FIG. 1B highlights in bold the nucleotide sequences for each hypervariable region in the exemplary reference E. coli 16S sequence described by Brosius et al.

[0017]

FIG. 2 shows a photograph of a CsCl gradient demonstrating the spore separation from other residual habitat material.

[0018]

FIG. 3 shows three phase contrast image demonstrating the progressive enrichment of spores from a fecal suspension; ethanol treated, CsCl purified spore preparation; and an ethanol treated, CsCl purified, sucrose purified spore preparation.

[0019]

FIG. 4 shows a set of survival curves demonstrating efficacy of the spore population in a hamster prophylaxis model of C. difficile.

[0020]

FIG. 5 provides a set of survival curves demonstrating efficacy of the spore population in a hamster relapse prevention model of C. difficile.

[0021]

FIG. 6 shows the strong correlation and linear correspondence between the measurement of DPA concentration by a coupled fluorescence assay and the viable spore colony forming units.

[0022]

The figures depict various embodiments of the present invention for purposes of illustration only. One skilled in the art will readily recognize from the following discussion that alternative embodiments of the structures and methods illustrated herein may be employed without departing from the principles of the invention described herein.

DETAILED DESCRIPTION

Definitions

[0023]

“Microbiota” refers to the community of microorganisms that occur (sustainably or transiently) in and on an animal subject, typically a mammal such as a human, including eukaryotes, archaea, bacteria, and viruses (including bacterial viruses i.e., phage).

[0024]

“Microbiome” refers to the genetic content of the communities of microbes that live in and on the human body, both sustainably and transiently, including eukaryotes, archaea, bacteria, and viruses (including bacterial viruses (i.e., phage)), wherein “genetic content” includes genomic DNA, RNA such as ribosomal RNA, the epigenome, plasmids, and all other types of genetic information.

[0025]

“Microbial Carriage” or simply “Carriage” refers to the population of microbes inhabiting a niche within or on humans. Carriage is often defined in terms of relative abundance. For example, OTU1 comprises 60% of the total microbial carriage, meaning that OTU1 has a relative abundance of 60% compared to the other OTUs in the sample from which the measurement was made. Carriage is most often based on genomic sequencing data where the relative abundance or carriage of a single OTU or group of OTUs is defined by the number of sequencing reads that are assigned to that OTU/s relative to the total number of sequencing reads for the sample.

[0026]

“Microbial Augmentation” or simply “augmentation” refers to the establishment or significant increase of a population of microbes that are (i) absent or undetectable (as determined by the use of standard genomic and microbiological techniques) from the administered therapeutic microbial composition, (ii) absent, undetectable, or present at low frequencies in the host niche (as example: gastrointestinal tract, skin, anterior-nares, or vagina) before the delivery of the microbial composition, and (iii) are found after the administration of the microbial composition or significantly increase, for instance 2-fold, 5-fold, 1×102, 1×103, 1×104, 1×105, 1×106, 1×107, or greater than 1×108, in cases where they were present at low frequencies. The microbes that comprise an augmented ecology can be derived from exogenous sources such as food and the environment, or grow out from micro-niches within the host where they reside at low frequency.

[0027]

The administration of the therapeutic microbial composition induces an environmental shift in the target niche that promotes favorable conditions for the growth of these commensal microbes. In the absence of treatment with a therapeutic microbial composition, the host can be constantly exposed to these microbes; however, sustained growth and the positive health effects associated with the stable population of increased levels of the microbes comprising the augmented ecology are not observed.

[0028]

“Microbial Engraftment” or simply “engraftment” refers to the establishment of OTUs comprising a therapeutic microbial composition in a target niche that are absent in the treated host prior to treatment. The microbes that comprise the engrafted ecology are found in the therapeutic microbial composition and establish as constituents of the host microbial ecology upon treatment. Engrafted OTUs can establish for a transient period of time, or demonstrate long-term stability in the microbial ecology that populates the host post treatment with a therapeutic microbial composition. The engrafted ecology can induce an environmental shift in the target niche that promotes favorable conditions for the growth of commensal microbes capable of catalyzing a shift from a dysbiotic ecology to one representative of a health state.

[0029]

“Ecological Niche” or simply “Niche” refers to the ecological space in which a an organism or group of organisms occupies. Niche describes how an organism or population or organisms responds to the distribution of resources, physical parameters (e.g., host tissue space) and competitors (e.g., by growing when resources are abundant, and when predators, parasites and pathogens are scarce) and how it in turn alters those same factors (e.g., limiting access to resources by other organisms, acting as a food source for predators and a consumer of prey).

[0030]

“Dysbiosis” refers to a state of the microbiota of the gut or other body area in a subject, including mucosal or skin surfaces in which the normal diversity and/or function of the ecological network is disrupted. This unhealthy state can be due to a decrease in diversity, the overgrowth of one or more pathogens or pathobionts, symbiotic organisms able to cause disease only when certain genetic and/or environmental conditions are present in a subject, or the shift to an ecological microbial network that no longer provides an essential function to the host subject, and therefore no longer promotes health.

[0031]

“Pathobionts” or “Opportunistic Pathogens” refers to symbiotic organisms able to cause disease only when certain genetic and/or environmental conditions are present in a subject.

[0032]

“Phylogenetic tree” refers to a graphical representation of the evolutionary relationships of one genetic sequence to another that is generated using a defined set of phylogenetic reconstruction algorithms (e.g. parsimony, maximum likelihood, or Bayesian). Nodes in the tree represent distinct ancestral sequences and the confidence of any node is provided by a bootstrap or Bayesian posterior probability, which measures branch uncertainty.

[0033]

“Operational taxonomic units,” “OTU” (or plural, “OTUs”) refer to a terminal leaf in a phylogenetic tree and is defined by a nucleic acid sequence, e.g., the entire genome, or a specific genetic sequence, and all sequences that share sequence identity to this nucleic acid sequence at the level of species. In some embodiments the specific genetic sequence may be the 16S sequence or a portion of the 16S sequence. In other embodiments, the entire genomes of two entities are sequenced and compared. In another embodiment, select regions such as multilocus sequence tags (MLST), specific genes, or sets of genes may be genetically compared. In 16S embodiments, OTUs that share ≧97% average nucleotide identity across the entire 16S or some variable region of the 16S are considered the same OTU (see e.g. Claesson M J, Wang Q, O'Sullivan O, Greene-Diniz R, Cole J R, Ross R P, and O'Toole P W. 2010. Comparison of two next-generation sequencing technologies for resolving highly complex microbiota composition using tandem variable 16S rRNA gene regions. Nucleic Acids Res 38: e200. Konstantinidis K T, Ramette A, and Tiedje J M. 2006. The bacterial species definition in the genomic era. Philos Trans R Soc Lond B Biol Sci 361: 1929-1940.). In embodiments involving the complete genome, MLSTs, specific genes, or sets of genes OTUs that share ≧95% average nucleotide identity are considered the same OTU (see e.g. Achtman M, and Wagner M. 2008. Microbial diversity and the genetic nature of microbial species. Nat. Rev. Microbiol. 6: 431-440. Konstantinidis K T, Ramette A, and Tiedje J M. 2006. The bacterial species definition in the genomic era. Philos Trans R Soc Lond B Biol Sci 361: 1929-1940.). OTUs are frequently defined by comparing sequences between organisms. Generally, sequences with less than 95% sequence identity are not considered to form part of the same OTU. OTUs may also be characterized by any combination of nucleotide markers or genes, in particular highly conserved genes (e.g., “house-keeping” genes), or a combination thereof. Such characterization employs, e.g., WGS data or a whole genome sequence.

[0034]

Table 1 below shows a List of Operational Taxonomic Units (OTU) with taxonomic assignments made to Genus, Species, and Phylogenetic Clade. Clade membership of bacterial OTUs is based on 16S sequence data. Clades are defined based on the topology of a phylogenetic tree that is constructed from full-length 16S sequences using maximum likelihood methods familiar to individuals with ordinary skill in the art of phylogenetics. Clades are constructed to ensure that all OTUs in a given clade are: (i) within a specified number of bootstrap supported nodes from one another, and (ii) within 5% genetic similarity. OTUs that are within the same clade can be distinguished as genetically and phylogenetically distinct from OTUs in a different clade based on 16S-V4 sequence data, while OTUs falling within the same clade are closely related. OTUs falling within the same clade are evolutionarily closely related and may or may not be distinguishable from one another using 16S-V4 sequence data. Members of the same clade, due to their evolutionary relatedness, play similar functional roles in a microbial ecology such as that found in the human gut. Compositions substituting one species with another from the same clade are likely to have conserved ecological function and therefore are useful in the present invention. All OTUs are denoted as to their putative capacity to form spores and whether they are a Pathogen or Pathobiont (see Definitions for description of “Pathobiont”). NIAID Priority Pathogens are denoted as ‘Category-A’, ‘Category-B’, or ‘Category-C’, and Opportunistic Pathogens are denoted as ‘OP’. OTUs that are not pathogenic or for which their ability to exist as a pathogen is unknown are denoted as ‘N’. The ‘SEQ ID Number’ denotes the identifier of the OTU in the Sequence Listing File and ‘Public DB Accession’ denotes the identifier of the OTU in a public sequence repository.

[0035]

“Residual habitat products” refers to material derived from the habitat for microbiota within or on a human or animal. For example, microbiota live in feces in the gastrointestinal tract, on the skin itself, in saliva, mucus of the respiratory tract, or secretions of the genitourinary tract (i.e., biological matter associated with the microbial community). Substantially free of residual habitat products means that the bacterial composition no longer contains the biological matter associated with the microbial environment on or in the human or animal subject and is 100% free, 99% free, 98% free, 97% free, 96% free, or 95% free of any contaminating biological matter associated with the microbial community. Residual habitat products can include abiotic materials (including undigested food) or it can include unwanted microorganisms. Substantially free of residual habitat products may also mean that the bacterial composition contains no detectable cells from a human or animal and that only microbial cells are detectable. In one embodiment, substantially free of residual habitat products may also mean that the bacterial composition contains no detectable viral (including bacterial viruses (i.e., phage)), fungal, mycoplasmal contaminants. In another embodiment, it means that fewer than 1×10−2%, 1×10−3%, 1×10−4%, 1×10−5%, 1×10−6%, 1×10−7%, 1×10−8of the viable cells in the bacterial composition are human or animal, as compared to microbial cells. There are multiple ways to accomplish this degree of purity, none of which are limiting. Thus, contamination may be reduced by isolating desired constituents through multiple steps of streaking to single colonies on solid media until replicate (such as, but not limited to, two) streaks from serial single colonies have shown only a single colony morphology. Alternatively, reduction of contamination can be accomplished by multiple rounds of serial dilutions to single desired cells (e.g., a dilution of 10−8or 10−9), such as through multiple 10-fold serial dilutions. This can further be confirmed by showing that multiple isolated colonies have similar cell shapes and Gram staining behavior. Other methods for confirming adequate purity include genetic analysis (e.g. PCR, DNA sequencing), serology and antigen analysis, enzymatic and metabolic analysis, and methods using instrumentation such as flow cytometry with reagents that distinguish desired constituents from contaminants.

[0036]

“Clade” refers to the OTUs or members of a phylogenetic tree that are downstream of a statistically valid node in a phylogenetic tree. The clade comprises a set of terminal leaves in the phylogenetic tree that is a distinct monophyletic evolutionary unit and that share some extent of sequence similarity.

[0037]

16s Sequencing, 16s, 16s-rRNA, 16s-NGS: In microbiology, “16S sequencing” or “16S-rRNA” or “16S” refers to sequence derived by characterizing the nucleotides that comprise the 16S ribosomal RNA gene(s). The bacterial 16S rDNA is approximately 1500 nucleotides in length and is used in reconstructing the evolutionary relationships and sequence similarity of one bacterial isolate to another using phylogenetic approaches. 16S sequences are used for phylogenetic reconstruction as they are in general highly conserved, but contain specific hypervariable regions that harbor sufficient nucleotide diversity to differentiate genera and species of most bacteria.

[0038]

The “V1-V9 regions” of the 16S rRNA refers to the first through ninth hypervariable regions of the 16S rRNA gene that are used for genetic typing of bacterial samples. These regions in bacteria are defined by nucleotides 69-99, 137-242, 433-497, 576-682, 822-879, 986-1043, 1117-1173, 1243-1294 and 1435-1465 respectively using numbering based on the E. coli system of nomenclature. Brosius et al., Complete nucleotide sequence of a 16S ribosomal RNA gene from Escherichia coli, PNAS 75(10):4801-4805 (1978). In some embodiments, at least one of the V1, V2, V3, V4, V5, V6, V7, V8, and V9 regions are used to characterize an OTU. In one embodiment, the V1, V2, and V3 regions are used to characterize an OTU. In another embodiment, the V3, V4, and V5 regions are used to characterize an OTU. In another embodiment, the V4 region is used to characterize an OTU. A person of ordinary skill in the art can identify the specific hypervariable regions of a candidate 16S rRNA by comparing the candidate sequence in question to a reference sequence and identifying the hypervariable regions based on similarity to the reference hypervariable regions, or alternatively, one can employ Whole Genome Shotgun (WGS) sequence characterization of microbes or a microbial community.

[0039]

The term “subject” refers to any animal subject including humans, laboratory animals (e.g., primates, rats, mice), livestock (e.g., cows, sheep, goats, pigs, turkeys, and chickens), and household pets (e.g., dogs, cats, and rodents). The subject may be suffering from a dysbiosis, including, but not limited to, an infection due to a gastrointestinal pathogen or may be at risk of developing or transmitting to others an infection due to a gastrointestinal pathogen.

[0040]

The term “phenotype” refers to a set of observable characteristics of an individual entity. As example an individual subject may have a phenotype of “health” or “disease”. Phenotypes describe the state of an entity and all entities within a phenotype share the same set of characteristics that describe the phenotype. The phenotype of an individual results in part, or in whole, from the interaction of the entities genome and/or microbiome with the environment.

[0041]

The term “Network Ecology” refers to a consortium of OTUs that co-occur in some number of subjects. As used herein, a “network” is defined mathematically by a graph delineating how specific nodes (i.e. OTUs) and edges (connections between specific OTUs) relate to one another to define the structural ecology of a consortium of OTUs. Any given Network Ecology will possess inherent phylogenetic diversity and functional properties. A Network Ecology can also be defined in terms of function where for example the nodes would be comprised of elements such as, but not limited to, enzymes, clusters of orthologous groups (COGS; http://www.ncbi.nlm.nih.gov/books/NBK21090/), or KEGG pathways (www.genome.jp/kegg/).

[0042]

Network Class, Core Network, Core Network Ecology: The terms “Network Class”, “Core Network” and “Core Network Ecology” refer to a group of network ecologies that in general are computationally determined to comprise ecologies with similar phylogenetic and/or functional characteristics. A Core Network therefore contains important biological features, defined either phylogenetically or functionally, of a group (i.e., a cluster) of related network ecologies. One representation of a Core Network Ecology is a designed consortium of microbes, typically non-pathogenic bacteria, that represents core features of a set of phylogenetically or functionally related network ecologies seen in many different subjects. In many occurrences, a Core Network, while designed as described herein, exists as a Network Ecology observed in one or more subjects. Core Network ecologies are useful for reversing or reducing a dysbiosis in subjects where the underlying, related Network Ecology has been disrupted.

[0043]

The term “Keystone OTU” refers to one or more OTUs that are common to many network ecologies and are members of networks ecologies that occur in many subjects (i.e. are pervasive). Due to the ubiquitous nature of Keystone OTUs, they are central to the function of network ecologies in healthy subjects and are often missing or at reduced levels in subjects with disease. Keystone OTUs may exist in low, moderate, or high abundance in subjects.

[0044]

The term “non-Keystone OTU” refers to an OTU that is observed in a Network Ecology and is not a keystone OTU.

[0045]

The term “Phylogenetic Diversity” refers to the biodiversity present in a given Network Ecology or Core Network Ecology based on the OTUs that comprise the network. Phylogenetic diversity is a relative term, meaning that a Network Ecology or Core Network that is comparatively more phylogenetically diverse than another network contains a greater number of unique species, genera, and taxonomic families. Uniqueness of a species, genera, or taxonomic family is generally defined using a phylogenetic tree that represents the genetic diversity all species, genera, or taxonomic families relative to one another. In another embodiment phylogenetic diversity may be measured using the total branch length or average branch length of a phylogenetic tree.

[0046]

“Spore” or “endospore” refers to an entity, particularly a bacterial entity, which is in a dormant, non-vegetative and non-reproductive stage. Spores are generally resistant to environmental stress such as radiation, desiccation, enzymatic treatment, temperature variation, nutrient deprivation, and chemical disinfectants.

[0047]

A “spore population” refers to a plurality of spores present in a composition. Synonymous terms used herein include spore composition, spore preparation, ethanol treated spore fraction and spore ecology. A spore population may be purified from a fecal donation, e.g. via ethanol or heat treatment, or a density gradient separation or any combination of methods described herein to increase the purity, potency and/or concentration of spores in a sample. Alternatively, a spore population may be derived through culture methods starting from isolated spore former species or spore former OTUs or from a mixture of such species, either in vegetative or spore form.

[0048]

In one embodiment, the spore preparation comprises spore forming species wherein residual non-spore forming species have been inactivated by chemical or physical treatments including ethanol, detergent, heat, sonication, and the like; or wherein the non-spore forming species have been removed from the spore preparation by various separations steps including density gradients, centrifugation, filtration and/or chromatography; or wherein inactivation and separation methods are combined to make the spore preparation. In yet another embodiment, the spore preparation comprises spore forming species that are enriched over viable non-spore formers or vegetative forms of spore formers. In this embodiment, spores are enriched by 2-fold, 5-fold, 10-fold, 50-fold, 100-fold, 1000-fold, 10,000-fold or greater than 10,000-fold ompared to all vegetative forms of bacteria. In yet another embodiment, the spores in the spore preparation undergo partial germination during processing and formulation such that the final composition comprises spores and vegetative bacteria derived from spore forming species.

[0049]

A “germinant” is a material or composition or physical-chemical process capable of inducing vegetative growth of a bacterium that is in a dormant spore form, or group of bacteria in the spore form, either directly or indirectly in a host organism and/or in vitro.

[0050]

A “sporulation induction agent” is a material or physical-chemical process that is capable of inducing sporulation in a bacterium, either directly or indirectly, in a host organism and/or in vitro.

[0051]

To “increase production of bacterial spores” includes an activity or a sporulation induction agent. “Production” includes conversion of vegetative bacterial cells into spores and augmentation of the rate of such conversion, as well as decreasing the germination of bacteria in spore form, decreasing the rate of spore decay in vivo, or ex vivo, or to increasing the total output of spores (e.g. via an increase in volumetric output of fecal material).

[0052]

The “colonization” of a host organism includes the non-transitory residence of a bacterium or other microscopic organism. As used herein, “reducing colonization” of a host subject's gastrointestinal tract (or any other microbiotal niche) by a pathogenic bacterium includes a reduction in the residence time of the pathogen in the gastrointestinal tract as well as a reduction in the number (or concentration) of the pathogen in the gastrointestinal tract or adhered to the luminal surface of the gastrointestinal tract. Measuring reductions of adherent pathogens may be demonstrated, e.g., by a biopsy sample, or reductions may be measured indirectly, e.g., by measuring the pathogenic burden in the stool of a mammalian host.

[0053]

A “combination” of two or more bacteria includes the physical co-existence of the two bacteria, either in the same material or product or in physically connected products, as well as the temporal co-administration or co-localization of the two bacteria.

[0054]

A “cytotoxic” activity or bacterium includes the ability to kill a bacterial cell, such as a pathogenic bacterial cell. A “cytostatic” activity or bacterium includes the ability to inhibit, partially or fully, growth, metabolism, and/or proliferation of a bacterial cell, such as a pathogenic bacterial cell.

[0055]

To be free of “non-comestible products” means that a bacterial composition or other material provided herein does not have a substantial amount of a non-comestible product, e.g., a product or material that is inedible, harmful or otherwise undesired in a product suitable for administration, e.g., oral administration, to a human subject. Non-comestible products are often found in preparations of bacteria from the prior art.

[0056]

As used herein the term “vitamin” is understood to include any of various fat-soluble or water-soluble organic substances (non-limiting examples include vitamin A, Vitamin B1 (thiamine), Vitamin B2 (riboflavin), Vitamin B3 (niacin or niacinamide), Vitamin B5 (pantothenic acid), Vitamin B6 (pyridoxine, pyridoxal, or pyridoxamine, or pyridoxine hydrochloride), Vitamin B7 (biotin), Vitamin B9 (folic acid), and Vitamin B12 (various cobalamins; commonly cyanocobalamin in vitamin supplements), vitamin C, vitamin D, vitamin E, vitamin K, K1 and K2 (i.e. MK-4, MK-7), folic acid and biotin) essential in minute amounts for normal growth and activity of the body and obtained naturally from plant and animal foods or synthetically made, pro-vitamins, derivatives, analogs.

[0057]

As used herein, the term “minerals” is understood to include boron, calcium, chromium, copper, iodine, iron, magnesium, manganese, molybdenum, nickel, phosphorus, potassium, selenium, silicon, tin, vanadium, zinc, or combinations thereof.

[0058]

As used herein, the term “antioxidant” is understood to include any one or more of various substances such as beta-carotene (a vitamin A precursor), vitamin C, vitamin E, and selenium) that inhibit oxidation or reactions promoted by Reactive Oxygen Species (“ROS”) and other radical and non-radical species. Additionally, antioxidants are molecules capable of slowing or preventing the oxidation of other molecules. Non-limiting examples of antioxidants include astaxanthin, carotenoids, coenzyme Q10 (“CoQ10”), flavonoids, glutathione, Goji (wolfberry), hesperidin, lactowolfberry, lignan, lutein, lycopene, polyphenols, selenium, vitamin A, vitamin C, vitamin E, zeaxanthin, or combinations thereof.

Compositions of the Invention

[0059]

Bacterial Compositions

[0060]

We have identified combinations of commensal bacteria of the human gut microbiota with the capacity to meaningfully provide functions of a healthy microbiota when administered to mammalian hosts. Without being limited to a specific mechanism, it is thought that such compositions inhibit the growth of a pathogen such as C. difficile, Salmonella spp., enteropathogenic E. coli, and vancomycin-resistant Enterococcus spp. and other pathobionts, so that a healthy, diverse and protective microbiota can be maintained or, in the case of pathogenic bacterial infections such as recurrent C. difficile infection, repopulate the intestinal lumen to reestablish ecological control over potential pathogens. Furthermore, the bacterial compositions have the ability to shift the state of the gut microbiota toward a healthy state allowing for augmentation of the microbiota with commensal bacteria found in healthy microbiomes. The bacterial compositions may also engraft in the host themselves and remain present in the gut for 1 day, 1 week, 1 month, 1 year, or longer than 1 year.

[0061]

Preferred bacterial species include Escherichia coli, Streptococcus faecalis, Clostridium Innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta. As used herein, a “type” or more than one “types” of bacteria may be differentiated at the genus level, the species, level, the sub-species level, the strain level or by any other taxonomic method, as described herein and otherwise known in the art. Preferred bacterial species also include species that share 97% homology in the 16s rDNA region to the species of Escherichia coli, Streptococcus faecalis, Clostridium Innocuum, Clostridium ramosum, Clostridium bifermentans, Bacteroides ovatus, Bacteroides vulgatus, Bacteroides thetaiotaomicron, and Blautia producta as defined by the provided 16s sequences (See Table 1).

[0062]

Bacterial compositions may consist essentially of no greater than a number of types of these preferred bacteria. For instance, a bacterial composition may comprise no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20 types of bacteria, as defined by above species or operational taxonomic unit (OTU) encompassing such species.

[0063]

Bacterial compositions may consist essentially of a range of numbers of species of these preferred bacteria, but the precise number of species in a given composition is not known. For instance, a bacterial composition may consist essentially of between 2 and 10, 3 and 10, 4 and 10, 5 and 10, 6 and 10, 7 and 10, 8 and 10, or 9 and 10; or 2 and 9, 3 and 9, 4 and 9, 5 and 9, 6 and 9, 7 and 8 or 8 and 9; or 2 and 8, 3 and 8, 4 and 8, 5 and 8, 6 and 8 or 7 and 8; or 2 and 7, 3 and 7, 4 and 7, 5 and 7, or 6 and 7; or 2 and 6, 3 and 6, 4 and 6 or 5 and 6; or 2 and 5, 3 and 5 or 4 and 5; or 2 and 4 or 3 and 4; or 2 and 3, as defined by above species or operational taxonomic unit (OTU) encompassing such species.

[0064]

Bacterial compositions containing a plurality of species may be provided such that the relative concentration of a given species in the composition to any other species in the composition is known or unknown. Such relative concentrations of any two species, or OTUs, may be expressed as a ratio, where the ratio of a first species or OTU to a second species or OTU is 1:1 or any ratio other than 1:1, such as 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:15, 1:20, 1:25; 1:50; 1:75, 1:100, 1:200, 1:500; 1:1000, 1:10,000, 1:100,000 or greater than 1:100,000. The ratio of bacterial strains present in a bacterial composition may be determined by the ratio of the bacterial strains in a reference mammalian subject, e.g., a healthy human not suffering from or at known risk of developing a dysbiosis.

[0065]

Bacterial compositions comprising a plurality of species may be provided such that the concentration of a given strain, or the aggregate of all strains, is between 1×104and 1×1015viable bacteria per gram of composition or per administered dose. For example the concentration of a given strain, or the aggregate of all strains, is e.g., 1×104, 1×105, 1×106, 1×107, 1×108, 1×109, 1×1010, 1×1011, 1×1012, 1×1013, 1×1014, 1×1015, or greater than 1×1015viable bacteria per gram of composition or per administered dose. Alternatively, the concentration of a given strain, or the aggregate of all strains, is below a given concentration e.g., below 1×104, 1×105, 1×106, 1×107, 1×108, 1×109, 1×1010, 1×1011, 1×1012, 1×1013, 1×1014, 1×1015, or below 1×1015viable bacteria per gram of composition or per administered dose.

[0066]

In certain embodiments, the bacterial compositions comprise a plurality of strains of the same species of bacteria. In certain embodiments, the strain of bacteria is present in amounts at least 2, 5, 10, 50, 100 or more than 100 times greater than any other strain of that species present in the composition. For example, provided are compositions containing one strain of E. coli, where this strain is present in amounts at least 2, 5, 10, 50, 100 or more than 100 times greater than any other strain of E. coli present in the composition.

[0067]

Because pathogenic bacteria such as C. difficile bacteria may inhibit growth of the bacterial strains present in the composition, it is generally beneficial such that no more than two of the bacterial strains in the bacterial composition are strains whose growth is substantially inhibited, e.g., in vitro, by an equivalent amount of C. difficile bacteria.

[0068]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Barnesiella intestinihominis; Lactobacillus reuteri; a species characterized as one of Enterococcus hirae, Enterococus faecium, or Enterococcus durans; a species characterized as one of Anaerostipes caccae or Clostridium indolis; a species characterized as one of Staphylococcus warneri or Staphylococcus pasteuri; and Adlercreutzia equolifaciens. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0069]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Clostridium absonum, Clostridium argentinense, Clostridium baratii, Clostridium bifermentans, Clostridium botulinum, Clostridium butyricum, Clostridium cadaveris, Clostridium camis, Clostridium celaturn, Clostridium chauvoei, Clostridium clostridioforme, Clostridium cochlearium, Clostridium difficile, Clostridium fallax, Clostridium felsineum, Clostridium ghonii, Clostridium glycolicum, Clostridium haemolyticum, Clostridium hastiforme, Clostridium histolyticum, Clostridium indolis, Clostridium innocuum, Clostridium irregulare, Clostridium limosum, Clostridium malenominaturn, Clostridium novyi, Clostridium oroticum, Clostridium paraputrificum, Clostridium perfringens, Clostridium piliforme, Clostridium putrefaciens, Clostridium putrificum, Clostridium ramosum, Clostridium sardiniense, Clostridium sartagoforme, Clostridium scindens, Clostridium septicum, Clostridium sordeffii, Clostridium sphenoides, Clostridium spiroforme, Clostridium sporogenes, Clostridium subterminale, Clostridium symbiosum, Clostridium tedium, Clostridium tetani, Clostridium welchii, and Clostridium villosum. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0070]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Clostridium innocuum, Clostridum bifermentans, Clostridium butyricum, Bacteroides fragilis, Bacteroides thetaiotaomicron, Bacteroides uniformis, three strains of Escherichia coli, and Lactobacillus sp. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0071]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Clostridium bifermentans, Clostridium innocuum, Clostridium butyricum, three strains of Escherichia coli, three strains of Bacteroides, and Blautia producta. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0072]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Bacteroides sp., Escherichia coli, and non pathogenic Clostridia, including Clostridium innocuum, Clostridium bifermentans and Clostridium ramosum. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0073]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Bacteroides species, Escherichia coli and non-pathogenic Clostridia, such as Clostridium butyricum, Clostridium bifermentans and Clostridium innocuum. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0074]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Bacteroides caccae, Bacteroides capillosus, Bacteroides coagulans, Bacteroides distasonis, Bacteroides eggerthii, Bacteroides forsythus, Bacteroides fragilis, Bacteroides fragilis-ryhm, Bacteroides gracilis, Bacteroides levii, Bacteroides macacae, Bacteroides merdae, Bacteroides ovatus, Bacteroides pneumosintes, Bacteroides putredinis, Bacteroides pyogenes, Bacteroides splanchnicus, Bacteroides stercoris, Bacteroides tectum, Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides ureolyticus, and Bacteroides vulgatus. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0075]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Bacteroides, Eubacteria, Fusobacteria, Propionibacteria, Lactobacilli, anaerobic cocci, Ruminococcus, Escherichia coli, Gemmiger, Desulfomonas, and Peptostreptococcus. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0076]

In one embodiment, the bacterial composition comprises at least one and preferably more than one of the following species or a species that is 97% identical based on the 16s rDNA: Bacteroides fragilis ss. Vulgatus, Eubacterium aerofaciens, Bacteroides fragilis ss. Thetaiotaomicron, Blautia producta (previously known as Peptostreptococcus productus II), Bacteroides fragilis ss. Distasonis, Fusobacterium prausnitzii, Coprococcus eutactus, Eubacterium aerofaciens III, Blautia producta (previously known as Peptostreptococcus productus I), Ruminococcus bronii, Bifidobacterium adolescentis, Gemmiger formicilis, Bifidobacterium longum, Eubacterium siraeum, Ruminococcus torques, Eubacterium rectale III-H, Eubacterium rectale IV, Eubacterium eligens, Bacteroides eggerthii, Clostridium leptum, Bacteroides fragilis ss. A, Eubacterium biforme, Bifidobacterium infantis, Eubacterium rectale III-F, Coprococcus comes, Bacteroides capillosus, Ruminococcus albus, Eubacterium formicigenerans, Eubacterium haffii, Eubacterium ventriosum I, Fusobacterium russii, Ruminococcus obeum, Eubacterium rectale II, Clostridium ramosum I, Lactobacillus leichmanii, Ruminococcus cailidus, Butyrivibrio crossotus, Acidaminococcus fermentans, Eubacterium ventriosum, Bacteroides fragilis ss. fragilis, Bacteroides AR, Coprococcus catus, Eubacterium hadrum, Eubacterium cylindroides, Eubacterium ruminantium, Eubacterium CH-1, Staphylococcus epidermidis, Peptostreptococcus BL, Eubacterium limosum, Bacteroides praeacutus, Bacteroides L, Fusobacterium mortiferum I, Fusobacterium naviforme, Clostridium innocuum, Clostridium ramosum, Propionibacterium acnes, Ruminococcus flavefaciens, Ruminococcus AT, Peptococcus AU-1, Eubacterium AG, -AK, -AL, -AL-1, -AN; Bacteroides fragilis ss. ovatus, -ss. d, -ss. f, Bacteroides L-1, L-5; Fusobacterium nucleatum, Fusobacterium mortiferum, Escherichia coli, Streptococcus morbiliorum, Peptococcus magnus, Peptococcus G, AU-2; Streptococcus intermedius, Ruminococcus lactaris, Ruminococcus CO Gemmiger X, Coprococcus BH, -CC; Eubacterium tenue, Eubacterium ramulus, Eubacterium AE, -AG-H, -AG-M, -AJ, -BN-1; Bacteroides clostridiiformis ss. clostridliformis, Bacteroides coagulans, Bacteroides orails, Bacteroides ruminicola ss. brevis, -ss. ruminicola, Bacteroides splanchnicus, Desuifomonas pigra, Bacteroides L-4, -N-i; Fusobacterium H, Lactobacillus G, and Succinivibrio A. In an alternative embodiment, at least one of the preceding species is not substantially present in the bacterial composition.

[0077]

Bacterial Compositions Described by Operational Taxonomic Unit (OTUs)

[0078]

Bacterial compositions may be prepared comprising at least two types of isolated bacteria, chosen from the species in Table 1.

[0079]

A bacterial composition may be prepared comprising at least two types of isolated bacteria, wherein a first type is a first OTU comprising a species of E. coli, S. faecalis, Cl. Innocuum, Cl. ramosum, Cl. bifermentans, Bact. ovatus, Bact. vulgatus, Bact. thetaiotaomicron, or Blautia producta and the type is a second OTU is characterized by, i.e., at least 95%, 96%, 97%, 98%, 99% or including 100% sequence identity to, the first OTU. In some embodiments, two types of bacteria are provided in a composition, and the first bacteria and the second bacteria are not the same OTU.

[0080]

OTUs can be defined either by full 16S sequencing of the rRNA gene (Table 1), by sequencing of a specific hypervariable region of this gene (i.e. V1, V2, V3, V4, V5, V6, V7, V8, or V9), or by sequencing of any combination of hypervariable regions from this gene (e.g. V1-3 or V3-5). The bacterial 16S rDNA is approximately 1500 nucleotides in length and is used in reconstructing the evolutionary relationships and sequence similarity of one bacterial isolate to another using phylogenetic approaches. 16S sequences are used for phylogenetic reconstruction as they are in general highly conserved, but contain specific hypervariable regions that harbor sufficient nucleotide diversity to differentiate genera and species of most microbes.

[0081]

Using well known techniques, in order to determine the full 16S sequence or the sequence of any hypervariable region of the 16S sequence, genomic DNA is extracted from a bacterial sample, the 16S rDNA (full region or specific hypervariable regions) amplified using polymerase chain reaction (PCR), the PCR products cleaned, and nucleotide sequences delineated to determine the genetic composition of 16S gene or subdomain of the gene. If full 16S sequencing is performed, the sequencing method used may be, but is not limited to, Sanger sequencing. If one or more hypervariable regions are used, such as the V4 region, the sequencing can be, but is not limited to being, performed using the Sanger method or using a next-generation sequencing method, such as an Illumina (sequencing by synthesis) method using barcoded primers allowing for multiplex reactions.

[0082]

OTUs can be defined by a combination of nucleotide markers or genes, in particular highly conserved genes (e.g., “house-keeping” genes), or a combination thereof, full-genome sequence, or partial genome sequence generated using amplified genetic products, or whole genome sequence (WGS). Using well defined methods DNA extracted from a bacterial sample will have specific genomic regions amplified using PCR and sequenced to determine the nucleotide sequence of the amplified products. In the whole genome shotgun (WGS) method, extracted DNA will be directly sequenced without amplification. Sequence data can be generated using any sequencing technology including, but not limited to Sanger, Illumina, 454 Life Sciences, Ion Torrent, ABI, Pacific Biosciences, and/or Oxford Nanopore.

[0083]

In one embodiment, the OTUs can be characterized by one or more of the variable regions of the 16S sequence (V1-V9). These regions in bacteria are defined by nucleotides 69-99, 137-242, 433-497, 576-682, 822-879, 986-1043, 1117-1173, 1243-1294 and 1435-1465 respectively using numbering based on the E. coli system of nomenclature. (See, e.g., Brosius et al., Complete nucleotide sequence of a 16S ribosomal RNA gene from Escherichia coli, PNAS 75(10):4801-4805 (1978)). In some embodiments, at least one of the V1, V2, V3, V4, V5, V6, V7, V8, and V9 regions are used to characterize an OTU. In one embodiment, the V1, V2, and V3 regions are used to characterize an OTU. In another embodiment, the V3, V4, and V5 regions are used to characterize an OTU. In another embodiment, the V4 region is used to characterize an OTU.

[0084]

Inhibition of Bacterial Pathogens

[0085]

In some embodiments, the bacterial composition provides a protective or therapeutic effect against infection by one or more GI pathogens of interest. A list of exemplary bacterial pathogens and pathobionts is provided in Table 1.

[0086]

In some embodiments, the pathogenic bacterium is selected from the group consisting of Yersinia, Vibrio, Treponema, Streptococcus, Staphylococcus, Shigella, Salmonella, Rickettsia, Orientia, Pseudomonas, Neisseria, Mycoplasma, Mycobacterium, Listeria, Leptospira, Legionella, Klebsiella, Helicobacter, Haemophilus, Francisella, Escherichia, Ehrlichia, Enterococcus, Coxiella, Corynebacterium, Clostridium, Chlamydia, Chlamydophila, Campylobacter, Burkholderia, Brucella, Borrelia, Bordetella, Bifidobacterium, Bacillus, multi-drug resistant bacteria, extended spectrum beta-lactam resistant Enterococci (ESBL), Carbapenem-resistent Enterobacteriaceae (CRE), and vancomycin-resistant Enterococci (VRE).

[0087]

In some embodiments, these pathogens include, but are not limited to, Aeromonas hydrophila, Campylobacter fetus, Plesiomonas shigelloides, Bacillus cereus, Campylobacter jejuni, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, enteroaggregative Escherichia coli, enterohemorrhagic Escherichia coli, enteroinvasive Escherichia coli, enterotoxigenic Escherichia coli (such as, but not limited to, LT and/or ST), Escherichia coli 0157:H7, Helicobacter pylori, Klebsiellia pneumonia, Lysteria monocytogenes, Plesiomonas shigelloides, Salmonella spp., Salmonella typhi, Salmonella paratyphi, Shigella spp., Staphylococcus spp., Staphylococcus aureus, vancomycin-resistant enterococcus spp., Vibrio spp., Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, and Yersinia enterocolitica.

[0088]

In one embodiment, the pathogen of interest is at least one pathogen chosen from Clostridium difficile, Salmonella spp., pathogenic Escherichia coli, vancomycin-resistant Enterococcus spp., and extended spectrum beta-lactam resistant Enterococci (ESBL).

[0089]

Generation and Formulation of Bacterial Compositions

[0090]

The bacterial compositions are generally formulated for oral or gastric administration, typically to a mammalian subject. Preferably, such formulations contain or are coated by an enteric coating to protect the bacteria through the stomach and small intestine. In particular embodiments, the composition is formulated for oral administration as a solid, semi-solid, powder, gel, or liquid form, such as in the form of a pill, tablet, capsule, or lozenge.

[0091]

The bacterial strains may, individually or together, be substantially inactive prior to localization in the gastrointestinal tract of the mammalian subject to whom the composition is administered. The bacterial strains may be lyophilized or otherwise acted upon to increase long-term storage viability.

[0092]

Preferential species include E. coli, S. faecalis, Cl. Innocuum, Cl. ramosum, Cl. bifermentans, Bact. ovatus, Bact. vulgatus, Bact. thetaiotaomicron, and Blautia producta, or species 97% identical to the 16s rDNA sequences of these species and are generally provided in bacterial compositions containing at least three strains. In certain embodiments, at least one of the bacterial strains is not a colonic bacterium. In another embodiment, at least one of the bacterial strains is not obtained from a fecal culture, e.g., the bacterial strain is obtained from culturing a non-fecal biological material from a subject, from an environmental source, or from repeatedly streaking and purifying strains from any source. Alternatively, at least one bacterial strain is genetically engineered. In another embodiment, at least one species is present in the composition in spore form. In an alternative embodiment, all of the species are present as vegetative forms, substantially free of spores.

[0093]

The bacterial compositions may be formulated to be effective in a given mammalian subject in a single administration or over multiple administrations. For example, a single administration is substantially effective to reduce C. difficile and/or C. difficile toxin content in a mammalian subject to whom the composition is administered. Alternatively, a single administration is substantially effective to reduce vancomycin resistant Enterococci, carbapenem resistant bacteria, or another pathobiont in a mammalian subject to whom the composition is administered (See Table 1). Substantially effective means that C. difficile and/or C. difficile toxin and/or pathobiont content in the subject is reduced by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% or greater than 99% following administration of the composition.

[0094]

In some embodiments, the bacterial compositions comprise purified spore populations. Purified spore populations comprise one or more commensal bacteria of the human gut microbiota present in the form of a spore. Without being limited to a specific mechanism, it is thought that such compositions inhibit the growth of a pathogen such as C. difficile, Salmonella spp., enteropathogenic E. coli, and vancomycin-resistant Enterococcus spp., so that a healthy, diverse and protective microbiota can be maintained or, in the case of pathogenic bacterial infections such as C. difficile infection, repopulate the intestinal lumen to reestablish ecological control over potential pathogens.

[0095]

In some embodiments, spore-forming bacteria are identified by the presence of nucleic acid sequences that modulate sporulation. In particular, signature sporulation genes are highly conserved across members of distantly related genera including Clostridium and Bacillus. Traditional approaches of forward genetics have identified many, if not all, genes that are essential for sporulation (spo). The developmental program of sporulation is governed in part by the successive action of four compartment-specific sigma factors (appearing in the order σF, σE, σG and σK), whose activities are confined to the forespore (σF and σG) or the mother cell (σE and σK).

[0096]

Provided are bacterial compositions comprising more than one type of spore forming bacterium. As used herein, a “type” or more than one “types” of bacteria may be differentiated at the genus level, the species, level, the sub-species level, the strain level or by any other taxonomic method, as described herein and otherwise known in the art.

[0097]

Pharmaceutical Compositions and Formulations of the Invention

[0098]

Provided are formulations for administration to humans and other subjects in need thereof. Generally the bacterial compositions are combined with additional active and/or inactive materials in order to produce a final product, which may be in single dosage unit or in a multi-dose format.

[0099]

In some embodiments, the composition comprises at least one carbohydrate. A “carbohydrate” refers to a sugar or polymer of sugars. The terms “saccharide,” “polysaccharide,” “carbohydrate,” and “oligosaccharide” may be used interchangeably. Most carbohydrates are aldehydes or ketones with many hydroxyl groups, usually one on each carbon atom of the molecule. Carbohydrates generally have the molecular formula CnH2nOn. A carbohydrate can be a monosaccharide, a disaccharide, trisaccharide, oligosaccharide, or polysaccharide. The most basic carbohydrate is a monosaccharide, such as glucose, sucrose, galactose, mannose, ribose, arabinose, xylose, and fructose. Disaccharides are two joined monosaccharides. Exemplary disaccharides include sucrose, maltose, cellobiose, and lactose. Typically, an oligosaccharide includes between three and six monosaccharide units (e.g., raffinose, stachyose), and polysaccharides include six or more monosaccharide units. Exemplary polysaccharides include starch, glycogen, and cellulose. Carbohydrates can contain modified saccharide units, such as 2′-deoxyribose wherein a hydroxyl group is removed, 2′-fluororibose wherein a hydroxyl group is replace with a fluorine, or N-acetylglucosamine, a nitrogen-containing form of glucose (e.g., 2′-fluororibose, deoxyribose, and hexose). Carbohydrates can exist in many different forms, for example, conformers, cyclic forms, acyclic forms, stereoisomers, tautomers, anomers, and isomers.

[0100]

In some embodiments, the composition comprises at least one lipid. As used herein, a “lipid” includes fats, oils, triglycerides, cholesterol, phospholipids, fatty acids in any form including free fatty acids. Fats, oils and fatty acids can be saturated, unsaturated (cis or trans) or partially unsaturated (cis or trans). In some embodiments, the lipid comprises at least one fatty acid selected from lauric acid (12:0), myristic acid (14:0), palmitic acid (16:0), palmitoleic acid (16:1), margaric acid (17:0), heptadecenoic acid (17:1), stearic acid (18:0), oleic acid (18:1), linoleic acid (18:2), linolenic acid (18:3), octadecatetraenoic acid (18:4), arachidic acid (20:0), eicosenoic acid (20:1), eicosadienoic acid (20:2), eicosatetraenoic acid (20:4), eicosapentaenoic acid (20:5) (EPA), docosanoic acid (22:0), docosenoic acid (22:1), docosapentaenoic acid (22:5), docosahexaenoic acid (22:6) (DHA), and tetracosanoic acid (24:0). In other embodiments, the composition comprises at least one modified lipid, for example, a lipid that has been modified by cooking.

[0101]

In some embodiments, the composition comprises at least one supplemental mineral or mineral source. Examples of minerals include, without limitation: chloride, sodium, calcium, iron, chromium, copper, iodine, zinc, magnesium, manganese, molybdenum, phosphorus, potassium, and selenium. Suitable forms of any of the foregoing minerals include soluble mineral salts, slightly soluble mineral salts, insoluble mineral salts, chelated minerals, mineral complexes, non-reactive minerals such as carbonyl minerals, and reduced minerals, and combinations thereof.

[0102]

In certain embodiments, the composition comprises at least one supplemental vitamin. In one embodiment, at least one vitamin can be fat-soluble or water soluble vitamins. Suitable vitamins include but are not limited to vitamin C, vitamin A, vitamin E, vitamin B12, vitamin K, riboflavin, niacin, vitamin D, vitamin B6, folic acid, pyridoxine, thiamine, pantothenic acid, and biotin. Suitable forms of any of the foregoing are salts of the vitamin, derivatives of the vitamin, compounds having the same or similar activity of the vitamin, and metabolites of the vitamin.

[0103]

In other embodiments, the composition comprises an excipient. Non-limiting examples of suitable excipients include a buffering agent, a preservative, a stabilizer, a binder, a compaction agent, a lubricant, a dispersion enhancer, a disintegration agent, a flavoring agent, a sweetener, and a coloring agent.

[0104]

In another embodiment, the excipient is a buffering agent. Non-limiting examples of suitable buffering agents include sodium citrate, magnesium carbonate, magnesium bicarbonate, calcium carbonate, and calcium bicarbonate.

[0105]

In some embodiments, the excipient comprises a preservative. Non-limiting examples of suitable preservatives include antioxidants, such as alpha-tocopherol and ascorbate, and antimicrobials, such as parabens, chlorobutanol, and phenol.

[0106]

In other embodiments, the composition comprises a binder as an excipient. Non-limiting examples of suitable binders include starches, pregelatinized starches, gelatin, polyvinylpyrolidone, cellulose, methylcellulose, sodium carboxymethylcellulose, ethylcellulose, polyacrylamides, polyvinyloxoazolidone, polyvinylalcohols, C12-C18fatty acid alcohol, polyethylene glycol, polyols, saccharides, oligosaccharides, and combinations thereof.

[0107]

In another embodiment, the composition comprises a lubricant as an excipient. Non-limiting examples of suitable lubricants include magnesium stearate, calcium stearate, zinc stearate, hydrogenated vegetable oils, sterotex, polyoxyethylene monostearate, talc, polyethyleneglycol, sodium benzoate, sodium lauryl sulfate, magnesium lauryl sulfate, and light mineral oil.

[0108]

In other embodiments, the composition comprises a dispersion enhancer as an excipient. Non-limiting examples of suitable dispersants include starch, alginic acid, polyvinylpyrrolidones, guar gum, kaolin, bentonite, purified wood cellulose, sodium starch glycolate, isoamorphous silicate, and microcrystalline cellulose as high HLB emulsifier surfactants.

[0109]

In some embodiments, the composition comprises a disintegrant as an excipient. In other embodiments, the disintegrant is a non-effervescent disintegrant. Non-limiting examples of suitable non-effervescent disintegrants include starches such as corn starch, potato starch, pregelatinized and modified starches thereof, sweeteners, clays, such as bentonite, micro-crystalline cellulose, alginates, sodium starch glycolate, gums such as agar, guar, locust bean, karaya, pecitin, and tragacanth. In another embodiment, the disintegrant is an effervescent disintegrant. Non-limiting examples of suitable effervescent disintegrants include sodium bicarbonate in combination with citric acid, and sodium bicarbonate in combination with tartaric acid.

[0110]

In another embodiment, the excipient comprises a flavoring agent. Flavoring agents can be chosen from synthetic flavor oils and flavoring aromatics; natural oils; extracts from plants, leaves, flowers, and fruits; and combinations thereof. In some embodiments the flavoring agent is selected from cinnamon oils; oil of wintergreen; peppermint oils; clover oil; hay oil; anise oil; eucalyptus; vanilla; citrus oil such as lemon oil, orange oil, grape and grapefruit oil; and fruit essences including apple, peach, pear, strawberry, raspberry, cherry, plum, pineapple, and apricot.

[0111]

In other embodiments, the excipient comprises a sweetener. Non-limiting examples of suitable sweeteners include glucose (corn syrup), dextrose, invert sugar, fructose, and mixtures thereof (when not used as a carrier); saccharin and its various salts such as the sodium salt; dipeptide sweeteners such as aspartame; dihydrochalcone compounds, glycyrrhizin; Stevia Rebaudiana (Stevioside); chloro derivatives of sucrose such as sucralose; and sugar alcohols such as sorbitol, mannitol, sylitol, and the like. Also contemplated are hydrogenated starch hydrolysates and the synthetic sweetener 3,6-dihydro-6-methyl-1,2,3-oxathiazin-4-one-2,2-dioxide, particularly the potassium salt (acesulfame-K), and sodium and calcium salts thereof.

[0112]

In yet other embodiments, the composition comprises a coloring agent. Non-limiting examples of suitable color agents include food, drug and cosmetic colors (FD&C), drug and cosmetic colors (D&C), and external drug and cosmetic colors (Ext. D&C). The coloring agents can be used as dyes or their corresponding lakes.

[0113]

The weight fraction of the excipient or combination of excipients in the formulation is usually about 99% or less, such as about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 75% or less, about 70% or less, about 65% or less, about 60% or less, about 55% or less, 50% or less, about 45% or less, about 40% or less, about 35% or less, about 30% or less, about 25% or less, about 20% or less, about 15% or less, about 10% or less, about 5% or less, about 2% or less, or about 1% or less of the total weight of the composition.

[0114]

The bacterial compositions disclosed herein can be formulated into a variety of forms and administered by a number of different means. The compositions can be administered orally, rectally, or parenterally, in formulations containing conventionally acceptable carriers, adjuvants, and vehicles as desired. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, or intrasternal injection and infusion techniques. In an exemplary embodiment, the bacterial composition is administered orally.

[0115]

Solid dosage forms for oral administration include capsules, tablets, caplets, pills, troches, lozenges, powders, and granules. A capsule typically comprises a core material comprising a bacterial composition and a shell wall that encapsulates the core material. In some embodiments, the core material comprises at least one of a solid, a liquid, and an emulsion. In other embodiments, the shell wall material comprises at least one of a soft gelatin, a hard gelatin, and a polymer. Suitable polymers include, but are not limited to: cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methyl cellulose (HPMC), methyl cellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropylmethyl cellulose phthalate, hydroxypropylmethyl cellulose succinate and carboxymethylcellulose sodium; acrylic acid polymers and copolymers, such as those formed from acrylic acid, methacrylic acid, methyl acrylate, ammonio methylacrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate (e.g., those copolymers sold under the trade name “Eudragit”); vinyl polymers and copolymers such as polyvinyl pyrrolidone, polyvinyl acetate, polyvinylacetate phthalate, vinylacetate crotonic acid copolymer, and ethylene-vinyl acetate copolymers; and shellac (purified lac). In yet other embodiments, at least one polymer functions as taste-masking agents.

[0116]

Tablets, pills, and the like can be compressed, multiply compressed, multiply layered, and/or coated. The coating can be single or multiple. In one embodiment, the coating material comprises at least one of a saccharide, a polysaccharide, and glycoproteins extracted from at least one of a plant, a fungus, and a microbe. Non-limiting examples include corn starch, wheat starch, potato starch, tapioca starch, cellulose, hemicellulose, dextrans, maltodextrin, cyclodextrins, inulins, pectin, mannans, gum arabic, locust bean gum, mesquite gum, guar gum, gum karaya, gum ghatti, tragacanth gum, funori, carrageenans, agar, alginates, chitosans, or gellan gum. In some embodiments the coating material comprises a protein. In another embodiment, the coating material comprises at least one of a fat and an oil. In other embodiments, the at least one of a fat and an oil is high temperature melting. In yet another embodiment, the at least one of a fat and an oil is hydrogenated or partially hydrogenated. In one embodiment, the at least one of a fat and an oil is derived from a plant. In other embodiments, the at least one of a fat and an oil comprises at least one of glycerides, free fatty acids, and fatty acid esters. In some embodiments, the coating material comprises at least one edible wax. The edible wax can be derived from animals, insects, or plants. Non-limiting examples include beeswax, lanolin, bayberry wax, carnauba wax, and rice bran wax. Tablets and pills can additionally be prepared with enteric coatings.

[0117]

Alternatively, powders or granules embodying the bacterial compositions disclosed herein can be incorporated into a food product. In some embodiments, the food product is a drink for oral administration. Non-limiting examples of a suitable drink include fruit juice, a fruit drink, an artificially flavored drink, an artificially sweetened drink, a carbonated beverage, a sports drink, a liquid diary product, a shake, an alcoholic beverage, a caffeinated beverage, infant formula and so forth. Other suitable means for oral administration include aqueous and nonaqueous solutions, emulsions, suspensions and solutions and/or suspensions reconstituted from non-effervescent granules, containing at least one of suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, coloring agents, and flavoring agents.

[0118]

In some embodiments, the food product can be a solid foodstuff. Suitable examples of a solid foodstuff include without limitation a food bar, a snack bar, a cookie, a brownie, a muffin, a cracker, an ice cream bar, a frozen yogurt bar, and the like.

[0119]

In other embodiments, the compositions disclosed herein are incorporated into a therapeutic food. In some embodiments, the therapeutic food is a ready-to-use food that optionally contains some or all essential macronutrients and micronutrients. In another embodiment, the compositions disclosed herein are incorporated into a supplementary food that is designed to be blended into an existing meal. In one embodiment, the supplemental food contains some or all essential macronutrients and micronutrients. In another embodiment, the bacterial compositions disclosed herein are blended with or added to an existing food to fortify the food's protein nutrition. Examples include food staples (grain, salt, sugar, cooking oil, margarine), beverages (coffee, tea, soda, beer, liquor, sports drinks), snacks, sweets and other foods.

[0120]

In one embodiment, the formulations are filled into gelatin capsules for oral administration. An example of an appropriate capsule is a 250 mg gelatin capsule containing from 10 (up to 100 mg) of lyophilized powder (108to 1011bacteria), 160 mg microcrystalline cellulose, 77.5 mg gelatin, and 2.5 mg magnesium stearate. In an alternative embodiment, from 105to 1012bacteria may be used, 105to 107, 106to 107, or 108to 1010, with attendant adjustments of the excipients if necessary. In an alternative embodiment, an enteric-coated capsule or tablet or with a buffering or protective composition can be used.

Methods of the Invention

[0121]

Administration of Bacterial Compositions

[0122]

The bacterial compositions of the invention are suitable for administration to mammals and non-mammalian animals in need thereof. The bacterial compositions can be administered to animals, including humans, laboratory animals (e.g., primates, rats, mice), livestock (e.g., cows, sheep, goats, pigs, turkeys, chickens), and household pets (e.g., dogs, cats, rodents). In certain embodiments, the mammalian subject is a human subject who has one or more symptoms of a dysbiosis.

[0123]

When the mammalian subject is suffering from a disease, disorder or condition characterized by an aberrant microbiota, the bacterial compositions described herein are suitable for treatment thereof. In some embodiments, the mammalian subject has not received antibiotics in advance of treatment with the bacterial compositions. For example, the mammalian subject has not been administered at least two doses of vancomycin, metronidazole and/or or similar antibiotic compound within one week prior to administration of the therapeutic composition. In other embodiments, the mammalian subject has not previously received an antibiotic compound in the one month prior to administration of the therapeutic composition.

[0124]

The bacterial compositions are useful in methods of treating a mammalian subject suffering from or at risk of developing a gastrointestinal disease, disorder or condition. Therefore, a therapeutic benefit is provided by orally administering to the mammalian subject a therapeutic composition containing a bacterial population comprising at least three bacterial strains, wherein each bacterial strain is a member of a bacterial species selected from the group consisting of E. coli, S. faecalis, C. ramosum, C. bifermentans, Bact. ovatus, Bact. vulgatus, Bact. thetaiotaomicron, and Blautia producta, wherein the therapeutic composition is formulated for oral administration. In some embodiments, the gastrointestinal disease, disorder or condition is relapsing diarrhea caused by C. difficile, ulcerative colitis, colitis, Crohn's disease, or irritable bowel disease. Beneficially, the therapeutic composition is administered only once prior to improvement of the disease, disorder or condition. In some embodiments the therapeutic composition is administered at intervals greater than two days, such as once every three, four, five or six days, or every week or less frequently than every week.

[0125]

Also provided are methods of treating or preventing a mammalian subject suffering from or at risk of developing a metabolic disease, disorder or condition selected from the group consisting of diabetes, metabolic syndrome, obesity, and autism using the therapeutic compositions provided herein. The bacterial compositions can be administered as a complementary treatment to antibiotics when a patient is suffering from an acute infection, to reduce the risk of recurrence after an acute infection has subsided, or when a patient will be in close proximity to others with or at risk of serious gastrointestinal infections (physicians, nurses, hospital workers, family members of those who are ill or hospitalized).

[0126]

An In Vivo Assay Establishing that a Bacterial Composition Populates a Subject's Gastrointestinal Tract

[0127]

In order to determine that the bacterial composition populates the gastrointestinal tract of a subject, an animal model, such as a mouse model, may be used. The model may begin by evaluating the microbiota of the mice. Qualitative assessments may be accomplished using 16S profiling of the microbial community in the feces of normal mice. It may also be accomplished by full genome sequencing, whole genome shotgun sequencing (WGS), or traditional microbiological techniques. Quantitative assessments may be conducted using quantitative PCR (qPCR), described in section below, or by using traditional microbiological techniques and counting colony formation.

[0128]

Optionally, the mice may receive an antibiotic treatment to mimic the condition of dysbiosis. Antibiotic treatment can decrease the taxonomic richness, diversity, and evenness of the community, including a reduction of abundance of a significant number of bacterial taxa. Dethlefsen et al., The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing, PLoS Biology 6(11):3280 (2008). At least one antibiotic may be used and antibiotics are well known. Antibiotics may include aminoglycoside antibiotics (amikacin, arbekacin, gentamicin, kanamycin, neomycin, netilmicin, paromomycin, rhodostreptomycin, streptomycin, tobramycin, and apramycin), amoxicillin, ampicillin, Augmentin (an amoxicillin/clavulanate potassium combination), cephalosporin (cefaclor, defadroxil, cefazolin, cefixime, fefoxitin, cefprozil, ceftazimdime, cefuroxime, cephalexin), clavulanate potassium, clindamycin, colistin, gentamycin, kanamycin, metronidazole, or vancomycin. As an individual, nonlimiting specific example, the mice may be provided with drinking water containing a mixture of the antibiotics kanamycin, colistin, gentamycin, metronidazole and vancomycin at 40 mg/kg, 4.2 mg/kg, 3.5 mg/kg, 21.5 mg/kg, and 4.5 mg/kg (mg per average mouse body weight), respectively, for 7 days. Alternatively, mice may be administered ciprofloxacin at a dose of 15-20 mg/kg (mg per average mouse body weight), for 7 days.

[0129]

If the mice are provided with an antibiotic, a wash out period of from one day to three days may be provided with no antibiotic treatment and no bacterial composition treatment.

[0130]

Subsequently, the test bacterial composition is administered to the mice by oral gavage. The test bacterial composition may be administered in a volume of 0.2 ml containing 104CFUs of each type of bacteria in the bacterial composition. Dose-response may be assessed by using a range of doses, including, but not limited to 102, 103, 104, 105, 106, 107, 108, 109, and/or 1010.

[0131]

The mice may be evaluated using 16S sequencing, full genome sequencing, whole genome shotgun sequencing (WGS), or traditional microbiological techniques to determine whether the test bacterial composition has populated the gastrointestinal tract of the mice. For example only, one day, three days, one week, two weeks, and one month after administration of the bacterial composition to the mice, 16S profiling is conducted to determine whether the test bacterial composition has populated the gastrointestinal tract of the mice. Quantitative assessments, including qPCR and traditional microbiological techniques such as colony counting, may additionally or alternatively be performed, at the same time intervals.

[0132]

Furthermore, the number of sequence counts that correspond exactly to those in the bacterial composition over time may be assessed to determine specifically which components of the bacterial composition reside in the gastrointestinal tract over a particular period of time. In one embodiment, the strains of the bacterial composition persist for a desired period of time; in another embodiment, the components of the bacterial composition persist for a desired period of time while also increasing the ability of other microbes (such as those present in the environment, food, etc.) to populate the gastrointestinal tract, further increasing overall diversity, as discussed below.

[0133]

Ability of Bacterial cCompositions to Populate Different Regions of the Gastrointestinal Tract

[0134]

The present bacterial compositions may also be assessed for their ability to populate different regions on the gastrointestinal tract. In one embodiment, a bacterial composition may be chosen for its ability to populate one or more than one region of the gastrointestinal tract, including, but not limited to the stomach, the small intestine (duodenum, jejunum, and ileum), the large intestine (the cecum, the colon (the ascending, transverse, descending, and sigmoid colon), and the rectum).

[0135]

An in vivo study may be conducted to determine which regions of the gastrointestinal tract a given bacterial composition will populate. A mouse model similar to the one described above in section II.A may be conducted, except instead of assessing the feces produced by the mice, particular regions of the gastrointestinal tract may be removed and studied individually. For example, at least one particular region of the gastrointestinal tract may be removed and a qualitative or quantitative determination may be performed on the contents of that region of the gastrointestinal tract. In another embodiment, the contents may optionally be removed and the qualitative or quantitative determination may be conducted on the tissue removed from the mouse.

[0136]

Methods for Testing Sensitivity of Bacterial Composition

[0137]

In certain embodiments, methods for testing the sensitivity of bacterial compositions in order to select for particular desirable characteristics may be employed. For example, the constituents in the bacterial composition may be tested for pH resistance, bile acid resistance, and/or antibiotic sensitivity, either individually on a constituent-by-constituent basis or collectively as a bacterial composition comprised of multiple bacterial constituents (collectively referred to in this section as bacterial composition).

[0138]

1. pH Sensitivity Testing

[0139]

For oral administration of the bacterial compositions, optionally testing for pH resistance enhances the selection of bacterial compositions that will survive at the highest yield possible through the varying pH environments of the distinct regions of the GI tract. Understanding how the bacterial compositions react to the pH of the GI tract also assists in formulation, so that the number of bacteria in a dosage form can be increased if beneficial and/or so that the composition may be administered in an enteric-coated capsule or tablet or with a buffering composition. As the pH of the stomach can drop to a pH of 1 to 2 after a high-protein meal for a short time before physiological mechanisms adjust it to a pH of 3 to 4 and often resides at a resting pH of 4 to 5, and as the pH of the small intestine can range from a pH of 6 to 7.4, bacterial compositions can be prepared that survive these varying pH ranges (specifically wherein at least 1%, 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or as much as 100% of the bacteria can survive gut transit times through various pH ranges).

[0140]

This may be tested by exposing the bacterial composition to varying pH ranges for the expected gut transit times through those pH ranges. Therefore, as a nonlimiting example only, 18-hour cultures of bacterial compositions may be grown in standard media, such as gut microbiota medium (“GMM”, see Goodman et al., Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice, PNAS 108(15):6252-6257 (2011)) or another animal-products-free medium, with the addition of pH adjusting agents for a pH of 1 to 2 for 30 minutes, a pH of 3 to 4 for 1 hour, a pH of 4 to 5 for 1 to 2 hours, and a pH of 6 to 7.4 for 2.5 to 3 hours.

[0141]

Alternatively, bacterial cells may be grown in a standard media, such as the GMM media described above, and adjusted to pH 2.5 with 1M HCl. The cells may be incubated anaerobically at 37° C. and their survival measured at intervals of 0, 30, 60, 120, 240, and/or 360 minutes.

[0142]

A further alternative method for testing stability to acid is described in U.S. Pat. No. 4,839,281.

[0143]

Survival of bacteria may be determined by culturing the bacteria and counting colonies on appropriate selective or non-selective media.

[0144]

2. Bile Acid Sensitivity Testing

[0145]

Additionally, in some embodiments, testing for bile-acid resistance enhances the selection of bacterial compositions that will survive exposures to bile acid during transit through the GI tract. Bile acids are secreted into the small intestine and can, like pH, affect the survival of bacterial compositions.

[0146]

This may be tested by exposing the bacterial compositions to bile acids for the expected gut exposure time to bile acids. For example, bile acid solutions may be prepared at desired concentrations using 0.05 mM Tris at pH 9 as the solvent. After the bile acid is dissolved, the pH of the solution may be adjusted to 7.2 with 10% HCl. Bacterial compositions may be cultured in 2.2 ml of a bile acid composition mimicking the concentration and type of bile acids in the subject, 1.0 ml of 10% sterile-filtered feces media and 0.1 ml of an 18-hour culture of the given strain of bacteria. Incubations may be conducted for from 2.5 to 3 hours or longer. Survival of bacteria may be determined by culturing the bacteria and counting colonies on appropriate selective or non-selective media.

[0147]

As another alternative, bacterial strains may be streaked onto agar plates supplemented with porcine bile (Sigma) at 0.5%, 1%, and 5% (w/v). Plates may be incubated at 37° C. under anaerobic conditions and the growth recorded after 48 hours. Growth may be compared with control plates by an experienced observer and the growth of colonies scored as: 0=no growth, 1=hazy translucent growth (<33% control plates with 0% bile), 2=definite growth but not as good as controls (>33% but <66%), 3=growth equivalent to controls (>66%).

[0148]

A further alternative method for testing stability to bile acid is described in U.S. Pat. No. 4,839,281. Survival of bacteria may be determined by culturing the bacteria and counting colonies on appropriate selective or non-selective media.

[0149]

3. Antibiotic Sensitivity Testing

[0150]

As a further optional sensitivity test, bacterial compositions may be tested for sensitivity to antibiotics. In one embodiment, bacterial compositions may be chosen so that the bacterial constituents are sensitive to antibiotics such that if necessary they can be eliminated or substantially reduced from the subject's gastrointestinal tract by at least one antibiotic targeting the bacterial composition.

[0151]

4. Adherence to Gastrointestinal Cells

[0152]

The bacterial compositions may optionally be tested for the ability to adhere to gastrointestinal cells. Without being bound by theory, in some instances, adherence can enhance the ability to repopulate a subject's gastrointestinal tract and thus may be used as a criterion in some embodiments. As a first embodiment, this may be conducted in a tissue culture model, where gastrointestinal epithelial cells, such as but not limited to CACO-2 cells (ATCC HTB-37), are grown in tissue culture flasks to differentiation in an antibiotic containing media, grown for at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, or at least 7 days in an antibiotic-free medium, incubated with bacterial cells for 30 minutes, 60 minutes, 90 minutes, or 120 minutes, and washed three times. After washing, the number of adherent bacteria may be assessed by lysing the epithelial cells and enumerating the bacteria using the plate count method or qPCR, as described herein.

[0153]

As another mode, bacterial adherence may be evaluated using an engineered tissue model of the lining of the gastrointestinal tract. Viney et al., “Co-culture of Intestinal Epithelial and Stromal Cells in 3D Collagen-based Environments,” Regen Med 4(3):397-406 (2009). After preparation of the engineered tissue model, the bacterial cells may be incubated, washed, and adherence enumerated, as described above.

[0154]

Furthermore, bacterial adherence may be evaluated using a tissue explant from the gastrointestinal tract of a subject. Gastrointestinal tract tissue may be surgically removed from a healthy donor. Alternatively, gastrointestinal tract tissue may be surgically removed from a donor who has a gastrointestinal disease, such as from an unused portion of a biopsy. After surgical excision, the bacterial cells may be incubated, washed, and adherence enumerated, as described above.

[0155]

As an alternative, one technique involves the collection of the effluent from a subject with a well functioning ileostomy by saline lavage, as described in U.S. Pat. No. 4,839,281.

[0156]

Assessment of Microbiota Diversity

[0157]

Microbiota diversity, as assessed by deep 16S rRNA sequencing or metagenomic sequencing, may be evaluated at a variety of time points to assess the effect of microbial compositions on restoring microbiota diversity. Microbial diversity may optionally be assessed before administration of an antibiotic or removal of microbiota by a colon-cleansing preparation. Microbial diversity may also be assessed before administration of a microbial composition and after administration of a microbial composition at any frequency (e.g., more frequently than 1 day, 1 day, 3 days, 1 week, 2 weeks, one month, or more than one month). One may provide for any temporal resolution that has more or less frequent sampling intervals, and/or samples taken for a longer time period (e.g., at least 1 week, at least 1 month, at least three months, at least six months, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or over more than 10 years.

[0158]

Diversity may be measured according to known sequencing methods, including, but not limited to Dethlefsen et al., The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing, PLoS Biology 6(11):3280 (2008). Sequence data used for diversity analysis may comprise 16S rRNA, whole genome sequence, any subset of a genome as well as whole genome shotgun metagenomic sequence (WGS). Sequence data may be generated using any sequencing technology including, but not limited to Sanger, Illumina, 454 Life Sciences, Ion Torrent, ABI, Pacific Bioscienses, and/or Oxford Nanopore. Subjects may be assessed for a variety of diversity metrics, including, but not limited to, observed taxon richness, Shannon diversity index, Shannon equitability index, alpha diversity, beta diversity, Chao1 index, Simpson diversity index, normalized abundance over time, and/or phylogenetic breadth.

[0159]

In another embodiment, subjects may be assessed for a variety of diversity metrics, including, but not limited to, Simpson diversity or by plotting rarefaction curve comparisons of reference OTUs for subjects before and after treatment with microbial compositions. Phylogenetic diversity may be plotted against the number of sequence reads (full 16S sequence or one or any combination of more hypervariable regions of the 16S sequence, whole genome sequence, or specific marker loci). Shahinas, et al., Toward an understanding of changes in diversity associated with fecal microbiome transplantation based on 16S rRNA Gene Deep Sequencing, mBio 3(5):e00338-12 (2012).

[0160]

In another embodiment, diversity may be shown by graphing a bar plot of abundance at the family level and showing an increase in either the number of families or the balance of abundance between families.

[0161]

In another embodiment, diversity may be shown by plotting heatmaps of the abundance of an OTU, species, genus, and/or higher taxonomic assignment showing sets of samples that are enriched or depleted for specific constituents.

[0162]

In another embodiment, diversity may be shown by generation of a phylogenetic tree of OTUs, species, genera, and/or higher or lower clade assignments. In this embodiment genetic distances may be computed between all OTUs, and summary statistics including, but not limited to average genetic distance and number of unique clades may be computed.

[0163]

In another embodiment, beta-diversity may be computed using diversity metrics familiar to those with ordinary skill in the art that include but are not limited to Bray-Curtis Dissimilarity Indices or Jaccard Distances and plotted using Principal Coordinates Analysis.

[0164]

Beneficial bacterial compositions may result in an increase in diversity after their administration that is 10%, 20%, 30%, 40%, or 50% higher than diversity before their administration. In another embodiment bacterial compositions may result in restoration of diversity to at least 70%, 80%, 90%, 95%, or up to 100% of the diversity either measured before administration of an antibiotic or removal of microbiota by a colon-cleansing preparation, or alternatively, as compared to the diversity of a reference mammalian subject.

[0165]

Methods for Preparing a Bacterial Composition for Administration to a Subject

[0166]

Methods for producing bacterial compositions may include three main processing steps, combined with one or more mixing steps. The steps are: organism banking, organism production, and preservation.

[0167]

For banking, the strains included in the bacterial composition may be (1) isolated directly from a specimen or taken from a banked stock, (2) optionally cultured on a nutrient agar or broth that supports growth to generate viable biomass, and (3) the biomass optionally preserved in multiple aliquots in long-term storage.

[0168]

In embodiments using a culturing step, the agar or broth may contain nutrients that provide essential elements and specific factors that enable growth. An example would be a medium composed of 20 g/L glucose, 10 g/L yeast extract, 10 g/L soy peptone, 2 g/L citric acid, 1.5 g/L sodium phosphate monobasic, 100 mg/L ferric ammonium citrate, 80 mg/L magnesium sulfate, 10 mg/L hemin chloride, 2 mg/L calcium chloride, 1 mg/L menadione. Medium can be added to the culture at the start, may be added during the culture, or may be intermittently/continuously flowed through the culture. The strains in the bacterial composition may be cultivated alone, as a subset of the bacterial composition, or as an entire collection comprising the bacterial composition. As an example, a first strain may be cultivated together with a second strain in a mixed continuous culture, at a dilution rate lower than the maximum growth rate of either cell to prevent the culture from washing out of the cultivation.

[0169]

The inoculated culture is incubated under favorable conditions for a time sufficient to build biomass. For bacterial compositions for human use this is often at 37° C. temperature, pH, and other parameter with values similar to the normal human niche. The environment may be actively controlled, passively controlled (e.g., via buffers), or allowed to drift. For example, for anaerobic bacterial compositions (e.g., gut applications), an anoxic/reducing environment may be employed. This can be accomplished by addition of reducing agents such as cysteine to the broth, and/or stripping it of oxygen. As an example, a culture of a bacterial composition may be grown at 37° C., pH 7, in the medium above, pre-reduced with 1 g/L cysteineŸHCl.

[0170]

When the culture has generated sufficient biomass, it may be preserved for banking. The organisms may be placed into a chemical milieu that protects from freezing (adding ‘cryoprotectants’), drying (‘lyoprotectants’), and/or osmotic shock (‘osmoprotectants’), dispensing into multiple (optionally identical) containers to create a uniform bank, and then treating the culture for preservation. Containers are generally impermeable and have closures that assure isolation from the environment. Cryopreservation treatment is accomplished by freezing a liquid at ultra-low temperatures (e.g., at or below −80° C.). Dried preservation removes water from the culture by evaporation (in the case of spray drying or ‘cool drying’) or by sublimation (e.g., for freeze drying, spray freeze drying). Removal of water improves long-term bacterial composition storage stability at temperatures elevated above cryogenic. Bacterial composition banking may be done by culturing and preserving the strains individually, or by mixing the strains together to create a combined bank. As an example of cryopreservation, a bacterial composition culture may be harvested by centrifugation to pellet the cells from the culture medium, the supernatant decanted and replaced with fresh culture broth containing 15% glycerol. The culture can then be aliquoted into 1 mL cryotubes, sealed, and placed at −80° C. for long-term viability retention. This procedure achieves acceptable viability upon recovery from frozen storage.

[0171]

Organism production may be conducted using similar culture steps to banking, including medium composition and culture conditions. It may be conducted at larger scales of operation, especially for clinical development or commercial production. At larger scales, there may be several subcultivations of the bacterial composition prior to the final cultivation. At the end of cultivation, the culture is harvested to enable further formulation into a dosage form for administration. This can involve concentration, removal of undesirable medium components, and/or introduction into a chemical milieu that preserves the bacterial composition and renders it acceptable for administration via the chosen route. For example, a bacterial composition may be cultivated to a concentration of 1010CFU/mL, then concentrated 20-fold by tangential flow microfiltration; the spent medium may be exchanged by diafiltering with a preservative medium consisting of 2% gelatin, 100 mM trehalose, and 10 mM sodium phosphate buffer. The suspension can then be freeze-dried to a powder and titrated. After drying, the powder may be blended to an appropriate potency, and mixed with other cultures and/or a ‘filler’ such as microcrystalline cellulose for consistency and ease of handling, and the bacterial composition filled into gelatin capsules for oral administration.

[0172]

An example of an appropriate capsule is a 250 mg gelatin capsule containing from 10 (up to 100 mg) of lyophilized powder (108to 1011bacteria), 160 mg microcrystalline cellulose, 77.5 mg gelatin, and 2.5 mg magnesium stearate. In an alternative embodiment, from 105to 1012bacteria may be used, 105to 107, 106to 107, or 108to 1010, with attendant adjustments of the excipients if necessary.

[0173]

In one embodiment, the number of bacteria of each type may be present in the same amount or in different amounts. For example, in a bacterial composition with two types of bacteria, the bacteria may be present in from a 1:10,000 ratio to a 1:1 ratio, from a 1:10,000 ratio to a 1:1,000 ratio, from a 1:1,000 ratio to a 1:100 ratio, from a 1:100 ratio to a 1:50 ratio, from a 1:50 ratio to a 1:20 ratio, from a 1:20 ratio to a 1:10 ratio, from a 1:10 ratio to a 1:1 ratio. For bacterial compositions comprising at least three types of bacteria, the ratio of type of bacteria may be chosen pairwise from ratios for bacterial compositions with two types of bacteria. For example, in a bacterial composition comprising bacteria A, B, and C, at least one of the ratio between bacteria A and B, the ratio between bacteria B and C, and the ratio between bacteria A and C may be chosen, independently, from the pairwise combinations above.

[0174]

Methods of Treating a Subject

[0175]

A. Overview of Method

[0176]

The bacterial compositions may be used to populate the gastrointestinal tract of a subject, resulting in one or more of a changed constitution of a subject's microbiota and improvement or correction of a dysbiosis. Without being bound by theory, bacterial compositions can promote mammalian health by restoring the natural microflora in the GI tract and reinforcing the normal controls on aberrant immune responses.

[0177]

Bacterial compositions can also improve fiber and protein digestion. Improving fiber and protein digestion is desirable as it promotes the growth of microbiota. A probiotic composition with multiple species has been shown to reduce production of toxic metabolites from protein fermentation. Rehman, Effects of Probiotics and antibiotics on intestinal homeostasis in a computer controlled model of the large intestine, BMC Microbiology 12:47 (2012). Carbohydrate fermentation is, for the most part, believed to be a beneficial process in the large gut, because the growth of saccharolytic bacteria stimulates their requirements for toxic products associated with putrefaction, for incorporation into cellular proteins, thereby protecting the host. However, as digestive materials move along the gut, carbohydrates become depleted, which may be linked to the increased prevalence of colonic disease in the distal bowel. Macfarlane, et al., Bacteria, colonic fermentation, and gastrointestinal health, J AOAC Int. 95(1):50-60 (2012). Thus, restoring microbiota, including, but not limited to, restoring microbiota in the distal bowel can provide health benefits.

[0178]

Fiber digestion may be determined using the method described in Vickers et al., Comparison of fermentation of selected fructooligosaccharides and other fiber substrates by canine colonic microflora, Am. J. Vet. Res. 61 (4), 609-615 (2001), with the exception that instead of inoculating using diluted fecal samples each experiment may use the bacterial compositions of interest.

[0179]

In one embodiment, the pathogen may be Clostridium difficile, Salmonella spp., pathogenic Escherichia coli, or vancomycin-resistant Enterococcus spp. In yet another embodiment, the pathogen may be Clostridium difficile, or other pathogen or pathobiont listed in Table 1.

[0180]

The present bacterial compositions may be useful in a variety of clinical situations. For example, the bacterial compositions may be administered as a complementary treatment to antibiotics when a subject is suffering from an acute infection, to reduce the risk of recurrence after an acute infection has subsided, or when a subject will be in close proximity to others with or at risk of serious gastrointestinal infections (physicians, nurses, hospital workers, family members of those who are ill or hospitalized).

[0181]

The present bacterial compositions may be administered to animals, including humans, laboratory animals (e.g., primates, rats, mice), livestock (e.g., cows, sheep, goats, pigs, turkeys, chickens), and household pets (e.g., dogs, cats, rodents).

[0182]

In the present method, the bacterial composition is administered enterically, in other words by a route of access to the gastrointestinal tract. This includes oral administration, rectal administration (including enema, suppository, or colonoscopy), by an oral or nasal tube (nasogastric, nasojejunal, oral gastric, or oral jejunal), as detailed more fully in the section below.

[0183]

B. Pretreatment Protocols

[0184]

Prior to administration of the bacterial composition, the subject may optionally have a pretreatment protocol to prepare the gastrointestinal tract to receive the bacterial composition. In certain embodiments, the pretreatment protocol is advisable, such as when a subject has an acute infection with a highly resilient pathogen or when the microbiota resident in the subject's gastrointestinal tract is likely to resist colonization by the bacterial composition. In other embodiments, the pretreatment protocol is entirely optional, such as when the dysbiosis is not associated with a pathogenic infection; when, if an infection is present, the pathogen causing the infection is not resilient, or when the subject has had an acute infection that has been successfully treated but where the physician is concerned that the infection may recur. In these instances, the pretreatment protocol may enhance the ability of the bacterial composition to affect the subject's microbiome.

[0185]

As one way of preparing the subject for administration of the microbial ecosystem, a standard colon-cleansing preparation may be administered to the subject to substantially empty the contents of the colon, such as used to prepare a subject for a colonscopy. By “substantially emptying the contents of the colon,” this application means removing at least 75%, at least 80%, at least 90%, at least 95%, or about 100% of the contents of the ordinary volume of colon contents.

[0186]

If a subject has received an antibiotic for treatment of an infection, or if a subject has received an antibiotic as part of a specific pretreatment protocol, in one embodiment the antibiotic should be stopped in sufficient time to allow the antibiotic to be substantially reduced in concentration in the gut before the bacterial composition is administered. In one embodiment, the antibiotic may be discontinued 1, 2, or 3 days before the administration of the bacterial composition. In one embodiment, the antibiotic may be discontinued 3, 4, 5, 6, or 7 antibiotic half-lives before administration of the bacterial composition. In another embodiment, the antibiotic may be chosen so the constituents in the bacterial composition have an MIC50 that is higher than the concentration of the antibiotic in the gut.

[0187]

MIC50 of a bacterial composition or the elements in the composition may be determined by methods well known in the art. Reller et al., Antimicrobial Susceptibility Testing: A Review of General Principles and Contemporary Practices, Clinical Infectious Diseases 49(11):1749-1755 (2009). In such an embodiment, the additional time between antibiotic administration and administration of the bacterial composition is not necessary. If the pretreatment protocol is part of treatment of an acute infection, the antibiotic may be chosen so that the infection is sensitive to the antibiotic, but the constituents in the bacterial composition are not sensitive to the antibiotic.

[0188]

C. Routes of Administration

[0189]

In the present method, the bacterial composition is administered enterically, in other words by a route of access to the gastrointestinal tract. This preferentially includes oral administration, or by an oral or nasal tube (including nasogastric, nasojejunal, oral gastric, or oral jejunal). In other embodiments, administration includes rectal administration (including enema, suppository, or colonoscopy). The bacterial composition may be administered to at least one region of the gastrointestinal tract, including the mouth, esophagus, stomach, small intestine, large intestine, and rectum. In some embodiments it is administered to all regions of the gastrointestinal tract. The bacterial compositions may be administered orally in the form of medicaments such as powders, capsules, tablets, gels or liquids. The bacterial compositions may also be administered in gel or liquid form by the oral route or through a nasogastric tube, or by the rectal route in a gel or liquid form, by enema or instillation through a colonoscope or by a suppository.

[0190]

If the composition is administered colonoscopically and, optionally, if the bacterial composition is administered by other rectal routes (such as an enema or suppository) or even if the subject has an oral administration, the subject may have a colonic-cleansing preparation. The colon-cleansing preparation can facilitate proper use of the colonoscope or other administration devices, but even when it does not serve a mechanical purpose it can also maximize the proportion of the bacterial composition relative to the other organisms previously residing in the gastrointestinal tract of the subject. Any ordinarily acceptable colonic-cleansing preparation may be used such as those typically provided when a subject undergoes a colonoscopy.

[0191]

D. Dosages and Schedule for Administration

[0192]

In one embodiment, from 108and 1011microorganisms total may be administered to the subject in a given dosage form. In one mode, an effective amount may be provided in from 1 to 500 ml or from 1 to 500 grams of the bacterial composition having from 108to 1011bacteria per ml or per gram, or a capsule, tablet or suppository having from 1 mg to 1000 mg lyophilized powder having from 108to 1015bacteria. Those receiving acute treatment may receive higher doses than those who are receiving chronic administration (such as hospital workers or those admitted into long-term care facilities).

[0193]

Any of the preparations described herein may be administered once on a single occasion or on multiple occasions, such as once a day for several days or more than once a day on the day of administration (including twice daily, three times daily, or up to five times daily). Or the preparation may be administered intermittently according to a set schedule, e.g., once a day, once weekly, or once monthly, or when the subject relapses from the primary illness. In another embodiment, the preparation may be administered on a long-term basis to individuals who are at risk for infection with or who may be carriers of these pathogens, including individuals who will have an invasive medical procedure (such as surgery), who will be hospitalized, who live in a long-term care or rehabilitation facility, who are exposed to pathogens by virtue of their profession (livestock and animal processing workers), or who could be carriers of pathogens (including hospital workers such as physicians, nurses, and other health care professionals).

[0194]

E. Subject Selection

[0195]

Particular bacterial compositions may be selected for individual subjects or for subjects with particular profiles. For example, 16S sequencing may be performed for a given subject to identify the bacteria present in his or her microbiota. The sequencing may either profile the subject's entire microbiome using 16S sequencing (to the family, genera, or species level), a portion of the subject's microbiome using 16S sequencing, or it may be used to detect the presence or absence of specific candidate bacteria that are biomarkers for health or a particular disease state. Based on the biomarker data, a particular composition may be selected for administration to a subject to supplement or complement a subject's microbiota in order to restore health or treat or prevent disease.

[0196]

The specification is most thoroughly understood in light of the teachings of the references cited within the specification. The embodiments within the specification provide an illustration of embodiments and should not be construed to limit the scope. The skilled artisan readily recognizes that many other embodiments are encompassed. All publications and patents cited in this disclosure are incorporated by reference in their entirety. To the extent the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art.

[0197]

Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification, including claims, are to be understood as being modified in all instances by the term “about.” Accordingly, unless otherwise indicated to the contrary, the numerical parameters are approximations and may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.

[0198]

Unless otherwise indicated, the term “at least” preceding a series of elements is to be understood to refer to every element in the series.

Examples

[0199]

Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.

[0200]

The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., T. E. Creighton, Proteins: Structures and Molecular Properties (W.H. Freeman and Company, 1993); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition); Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.); Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pa.: Mack Publishing Company, 1990); Carey and Sundberg Advanced Organic Chemistry 3rdEd. (Plenum Press) Vols A and B(1992).

Example 1

Administration of Bacterial Compositions to Mammalian Subjects Having C. difficile Infection

[0201]

An in vivo mouse model was employed to demonstrate the protective effect of the bacterial compositions against C. difficile. In this model (based on Chen, et al., A mouse model of Clostridium difficile associated disease, Gastroenterology 135(6):1984-1992 (2008)), mice were made susceptible to C. difficile by a 7 or 9 day treatment (days −14 or −12 until −5 of experiment) with 5 to 7 antibiotics (kanamycin, colistin, gentamycin, metronidazole and vancomycin, and optionally including ampicillin and ciprofloxacin) delivered via their drinking water, followed by a single dose with clindamycin on day −3. The mice were then challenged three days later (on day 0) with 104spores of C. difficile via oral gavage (i.e., oro-gastric lavage). The outcomes assessed each day from day 0 to day 6 were weight, clinical signs, mortality and shedding of C. difficile in the feces. Weight, clinical signs and mortality were also evaluated on day −1. Weight loss, clinical signs of disease, and C. difficile shedding were typically observed without treatment. Vancomycin treatment protects against these outcomes and serves as a positive control when provided by oral gavage on days −1 to 4. Clinical signs were subjective, and scored each day by the same experienced observer. Mortality includes euthanasia of animals that lose greater than or equal to 25% of their body weight or that display severe morbidity as assessed by clinical signs. Feces were gathered from the mouse cages (5 mice per cage) each day, and suspended in 5 mL of PBS by vortexing. The shedding of C. difficile spores is detected in the feces using a selective plating assay as described for the in vitro assay above, after a 1 hour treatment with 50% ethanol at room temperature to kill C. difficile vegetative cells. C. difficile may also be detected in the mouse fecal suspension via qPCR for the toxin gene as described herein. The effects of various administrations, including 10% suspension of human feces in PBS (as a positive control), microbial compositions, or PBS (as a negative vehicle control), were determined by introducing the bacterial composition in a 0.2 mL volume into the mice via oral gavage on day −1, one day prior to C. difficile challenge. Vancomycin, as discussed above, is given on days −1 to −4 as another positive control. Alternative dosing schedules can be employed, including multiple doses of bacterial composition, and 103to 1010of a given organism or composition may be delivered.

[0202]

Exemplary bacterial compositions as described herein were administered as follows.

[0203]

In a first demonstration, the results of which are provided in tabular form as Table 2, Treatment 1 is the vehicle control of phosphate buffered saline (PBS), Treatment 2 is a positive control of 10% fecal suspension in PBS which has total anaerobic cfu/ml of 4.5×109, Treatment 3 is a positive control of 10% fecal suspension in PBS which has total anaerobic cfu/ml of 6.2×108, Treatment 4 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 7.1×109. Treatment 5 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 7.1×108. Treatment 6 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family EnterococcaceaeATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 7.1×107. Treatment 7 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 3.7×109. Treatment 8 is a bacterial composition comprising Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483) with a total anaerobic cfu/ml of 3.5×109. Treatment 9 is a bacterial composition comprising Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 6.7×109. Treatment 10 is a bacterial composition comprising Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), and Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 1.2×109. Treatment 11 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 6.1×109. Treatment 12 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 1×1010.

[0204]

In a second demonstration, the results of which are provided in tabular form as Table 3A, Treatment 1 is the vehicle control of phosphate buffered saline (PBS), Treatment 2 is a positive control of 10% fecal suspension in PBS which has total anaerobic cfu/ml of 5×109, Treatment 3 is a positive control of 10% fecal suspension in PBS which has total anaerobic cfu/ml of 7×108.

[0205]

Treatment 4 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 2.2×109, Treatment 5 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 2.2×108. Treatment 6 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 2.2×107, Treatment 7 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, ATCC 35320), Enterococcus faecalis (family Enterococcaceae ATCC, 19433), Blautia producta (family Lachnospiraceae, DSM 14466), Bacteroides vulgatus (family Bacteroidaceae, ATCC 8482), Bacteroides thetaiotaomicron (family Bacteroidaceae, ATCC 29148), Bacteroides ovatus (family Bacteroidaceae, ATCC 8483), Clostridium innocuum (family Erysipelotrichaceae, DSM 22910) with a total anaerobic cfu/ml of 5×109, Treatment 8 is a bacterial composition comprising Escherichia coli (family, Enterobacteriaceae, SPC21221), Escherichia coli (family, Enterobacteriaceae, SPC21248), Enterococcus faecalis (family, Enterococcaceae, SPC21240), Enterococcus faecalis (family, Enterococcaceae, SPC21252), Blautia producta (family, Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21122), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides thetaiotaomicron, (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family, Bacteroidaceae, SPC21141), Bacteroides ovatus (family, Bacteroidaceae, SPC21181), Clostridium innocuum (family, Erysipelotrichaceae, SPC21112), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), with a total anaerobic cfu/ml of 1.7×1010, Treatment 9 is a bacterial composition comprising Escherichia coli (family, Enterobacteriaceae, SPC21221), Escherichia coli (family, Enterobacteriaceae, SPC21248), Enterococcus faecalis (family, Enterococcaceae, SPC21240), Enterococcus faecalis (family, Enterococcaceae, SPC21252), Blautia producta (family, Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21122), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides thetaiotaomicron, (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family, Bacteroidaceae, SPC21141), Bacteroides ovatus (family, Bacteroidaceae, SPC21181), Clostridium innocuum (family, Erysipelotrichaceae, SPC21112), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), with a total anaerobic cfu/ml of 1.7×109, Treatment 10 is a bacterial composition comprising Escherichia coli (family, Enterobacteriaceae, SPC21221), Escherichia coli (family, Enterobacteriaceae, SPC21248), Enterococcus faecalis (family, Enterococcaceae, SPC21240), Enterococcus faecalis (family, Enterococcaceae, SPC21252), Blautia producta (family, Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21122), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides thetaiotaomicron, (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family, Bacteroidaceae, SPC21141), Bacteroides ovatus (family, Bacteroidaceae, SPC21181), Clostridium innocuum (family, Erysipelotrichaceae, SPC21112), Clostridium bifermentans (family Peptostreptococcaceae, ATCC 638), and Clostridium ramosum (family Erysipelotrichaceae, ATCC 25582), with a total anaerobic cfu/ml of 1.7×108, Treatment 11 is a bacterial composition comprising Escherichia coli (family, Enterobacteriaceae, SPC21221), Escherichia coli (family, Enterobacteriaceae, SPC21248), Enterococcus faecalis (family, Enterococcaceae, SPC21240), Enterococcus faecalis (family, Enterococcaceae, SPC21252), Blautia producta (family, Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21122), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides thetaiotaomicron, (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family, Bacteroidaceae, SPC21141), Bacteroides ovatus (family, Bacteroidaceae, SPC21181), and Clostridium innocuum (family, Erysipelotrichaceae, SPC21112) with a total anaerobic cfu/ml of 1.2×1010, Treatment 12 is a bacterial composition comprising Escherichia coli (family, Enterobacteriaceae, SPC21221), Escherichia coli (family, Enterobacteriaceae, SPC21248), Enterococcus faecalis (family, Enterococcaceae, SPC21240), Enterococcus faecalis (family, Enterococcaceae, SPC21252), Blautia producta (family, Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21122), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides thetaiotaomicron, (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family, Bacteroidaceae, SPC21141), Bacteroides ovatus (family, Bacteroidaceae, SPC21181), Clostridium innocuum (family, Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 9×109, Treatment 13 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21122), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), and Clostridium innocuum (family Erysipelotrichaceae, SPC21112) with a total anaerobic cfu/ml of 6×109. Treatment 14 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21248), Enterococcus faecalis (family Enterococcaceae, SPC21252), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family, Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21133), Bacteroides ovatus (family Bacteroidaceae, SPC21181), and Clostridium innocuum (family Erysipelotrichaceae, SPC21112) with a total anaerobic cfu/ml of 5×109.

[0206]

In a third demonstration, the results of which are provided in tabular form as Table 3B, Treatment 1 is the vehicle control of phosphate buffered saline (PBS), Treatment 2 is a positive control of 10% fecal suspension in PBS which has total anaerobic cfu/ml of 2.3×1011, Treatment 3 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 2.5×109, Treatment 4 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 2.5×108, Treatment 5 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 2.5×107, Treatment 6 is a bacterial composition comprising Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 2.8×109, Treatment 7 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 6×109, Treatment 8 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278) with a total anaerobic cfu/ml of 1.8×109, Treatment 9 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), and Bacteroides ovatus (family Bacteroidaceae, SPC21141) with a total anaerobic cfu/ml of 2×109, Treatment 10 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), Clostridium celatum (family Clostridiaceae, SPC21278), Clostridium butyricum (family Clostridiaceae, SPC21367), and Clostridium glycolicum (family Peptostreptococcaceae, SPC21349) with a total anaerobic cfu/ml of 8×109, Treatment 11 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), Clostridium celatum (family Clostridiaceae, SPC21278), Clostridium butyricum (family Clostridiaceae, SPC21367), Clostridium glycolicum (family Peptostreptococcaceae, SPC21349), Dorea formicigerans (family Lachnospiraceae, SPC21308), Ruminococcus torques (family Ruminococcaceae, SPC21344), Eubacterium tenue (family Peptostreptococcaceae, SPC21391) and Eubacterium cylindroides (family Erysipelotrichacea, SPC21300) with a total anaerobic cfu/ml of 2.5×109. Treatment 12 is a bacterial composition comprising Blautia producta (family Lachnospiraceae, SPC2115104), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), and Clostridium celatum (family Clostridiaceae, SPC21278), with a total anaerobic cfu/ml of 2.1×107. Treatment 13 is a bacterial composition comprising Blautia producta (family Lachnospiraceae, SPC2115104), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), Clostridium celatum (family Clostridiaceae, SPC21278), Clostridium butyricum (family Clostridiaceae, SPC21367), and Clostridium glycolicum (family Peptostreptococcaceae, SPC21349) with a total anaerobic cfu/ml of 1.4×108. Treatment 14 is a bacterial composition comprising Blautia producta (family Lachnospiraceae, SPC2115104), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), Clostridium celatum (family Clostridiaceae, SPC21278), Clostridium butyricum (family Clostridiaceae, SPC21367), Clostridium glycolicum (family Peptostreptococcaceae, SPC21349), Dorea formicigerans (family Lachnospiraceae, SPC21308), Ruminococcus torques (family Ruminococcaceae, SPC21344), Eubacterium tenue (family Peptostreptococcaceae, SPC21391) and Eubacterium cylindroides (family Erysipelotrichacea, SPC21300) with a total anaerobic cfu/ml of 3.1×107. Treatment 15 is a bacterial composition comprising Escherichia coli (family Enterobacteriaceae, SPC21221), Enterococcus faecalis (family Enterococcaceae, SPC21240), Blautia producta (family Lachnospiraceae, SPC2115104), Bacteroides vulgatus (family Bacteroidaceae, SPC21192), Bacteroides thetaiotaomicron (family Bacteroidaceae, SPC21132), Bacteroides ovatus (family Bacteroidaceae, SPC21141), Clostridium innocuum (family Erysipelotrichaceae, SPC21112), Clostridium celatum (family Clostridiaceae, SPC21278), Clostridium butyricum (family Clostridiaceae, SPC21367), Clostridium glycolicum (family Peptostreptococcaceae, SPC21349), Dorea formicigerans (family Lachnospiraceae, SPC21308), Ruminococcus torques (family Ruminococcaceae, SPC21344), Eubacterium tenue (family Peptostreptococcaceae, SPC21391) and Eubacterium cylindroides (family Erysipelotrichacea, SPC21300) that was treated with 50% EtOH for 1 hour at 37 C, centrifuged to pellet the bacterial cells and resuspended in sterile PBS at the equivalent original volume and having a total anaerobic cfu/ml of 1.3×107.

[0207]

For each of the three demonstrations, scores for each treatment range from 0 to 9. Scores were calculated as follows. Groups of animals (N=10/group) were scored for % mortality. Mortality was normalized in a given demonstration by setting the observed mortality of the PBS vehicle control group=3. A treatment group was then scored based on the observed mortality for that group divided by the mortality of the PBS vehicle control times 3. Thus, if the mortality of the PBS vehicle group was 60% (score=3), then a treatment group with mortality of 30% would receive a score of 1.5. Similarly, mean weight loss on Day 3 was normalized to a score of 3 for the PBS vehicle control group. A score for each treatment was calculated by dividing the actual mean weight loss by the mean weight loss for the PBS vehicle control group and multiplying by 3. A similar procedure was used for the clinical scores evaluation. Finally, the individual values for mortality, mean weight loss on Day 3 and clinical scores were summed to give a final value of 0-9 for each treatment, with 0 being best (no death, no weight loss, no clinical symptoms) and 9 being equivalent to the vehicle control.

Example 2

Species Identification

[0208]

The identity of the bacterial species which grew up from a complex fraction can be determined in multiple ways. First, individual colonies can be picked into liquid media in a 96 well format, grown up and saved as 15% glycerol stocks at −80C. Aliquots of the cultures can be placed into cell lysis buffer and colony PCR methods can be used to amplify and sequence the 16S rDNA gene (described below in Example 3). Alternatively, colonies may be streaked to purity in several passages on solid media. Well separated colonies are streaked onto the fresh plates of the same kind and incubated for 48-72 hours at 37 C. The process is repeated multiple times in order to ensure purity. Pure cultures can be analyzed by phenotypic- or sequence-based methods, including 16S rDNA amplification and sequencing as described in Examples 3 and 4. Sequence characterization of pure isolates or mixed communities e.g. plate scrapes and spore fractions can also include whole genome shotgun sequencing. The latter is valuable to determine the presence of genes associated with sporulation, antibiotic resistance, pathogenicity, and virulence. Colonies can also be scraped from plates en masse and sequenced using a massively parallel sequencing method as described in Examples 3 & 4 such that individual 16S signatures can be identified in a complex mixture. Optionally, the sample can be sequenced prior to germination (if appropriate DNA isolation procedures are used to lsye and release the DNA from spores) in order to compare the diversity of germinable species with the total number of species in a spore sample. As an alternative or complementary approach to 16S analysis, MALDI-TOF-mass spec can also be used for species identification (as reviewed in Anaerobe 22:123).

Example 3

16s Sequencing to Determine Operational Taxonomic Unit (OTU)

[0209]

Method for Determining 16S Sequence

[0210]

OTUs may be defined either by full 16S sequencing of the rRNA gene, by sequencing of a specific hypervariable region of this gene (i.e. V1, V2, V3, V4, V5, V6, V7, V8, or V9), or by sequencing of any combination of hypervariable regions from this gene (e.g. V1-3 or V3-5). The bacterial 16S rDNA is approximately 1500 nucleotides in length and is used in reconstructing the evolutionary relationships and sequence similarity of one bacterial isolate to another using phylogenetic approaches. 16S sequences are used for phylogenetic reconstruction as they are in general highly conserved, but contain specific hypervariable regions that harbor sufficient nucleotide diversity to differentiate genera and species of most microbes.

[0211]

Using well known techniques, in order to determine the full 16S sequence or the sequence of any hypervariable region of the 16S sequence, genomic DNA is extracted from a bacterial sample, the 16S rDNA (full region or specific hypervariable regions) amplified using polymerase chain reaction (PCR), the PCR products cleaned, and nucleotide sequences delineated to determine the genetic composition of 16S gene or subdomain of the gene. If full 16S sequencing is performed, the sequencing method used may be, but is not limited to, Sanger sequencing. If one or more hypervariable regions are used, such as the V4 region, the sequencing may be, but is not limited to being, performed using the Sanger method or using a next-generation sequencing method, such as an Illumina (sequencing by synthesis) method using barcoded primers allowing for multiplex reactions.

[0212]

In addition to the 16S rRNA gene, one may define an OTU by sequencing a selected set of genes that are known to be marker genes for a given species or taxonomic group of OTUs. These genes may alternatively be assayed using a PCR-based screening strategy. As example, various strains of pathogenic Escherichia coli can be distinguished using DNAs from the genes that encode heat-labile (LTI, LTIIa, and LTIIb) and heat-stable (STI and STII) toxins, verotoxin types 1, 2, and 2e (VT1, VT2, and VT2e, respectively), cytotoxic necrotizing factors (CNF1 and CNF2), attaching and effacing mechanisms (eaeA), enteroaggregative mechanisms (Eagg), and enteroinvasive mechanisms (Einv). The optimal genes to utilize for taxonomic assignment of OTUs by use of marker genes will be familiar to one with ordinary skill of the art of sequence based taxonomic identification and may include, but will not be limited, to highly conserved “house-keeping” genes or a combination thereof, full-genome sequence, or partial genome sequence generated using amplified genetic products, or whole genome sequence (WGS). Using well defined methods DNA extracted from a bacterial sample will have specific genomic regions amplified using PCR and sequenced to determine the nucleotide sequence of the amplified products. In the whole genome shotgun (WGS) method, extracted DNA will be directly sequenced without amplification. Sequence data can be generated using any sequencing technology including, but not limited to Sanger, Illumina, 454 Life Sciences, Ion Torrent, ABI, Pacific Biosciences, and/or Oxford Nanopore.

[0213]

Genomic DNA Extraction

[0214]

Genomic DNA is extracted from pure microbial cultures using a hot alkaline lysis method. 1 μl of microbial culture is added to 9 μl of Lysis Buffer (25 mM NaOH, 0.2 mM EDTA) and the mixture is incubated at 95° C. for 30 minutes. Subsequently, the samples are cooled to 4° C. and neutralized by the addition of 10 μl of Neutralization Buffer (40 mM Tris-HCl) and then diluted 10-fold in Elution Buffer (10 mM Tris-HCl). Alternatively, genomic DNA is extracted from pure microbial cultures using commercially available kits such as the Mo Bio Ultraclean® Microbial DNA Isolation Kit (Mo Bio Laboratories, Carlsbad, Calif.) or by standard methods known to those skilled in the art.

[0215]

Amplification of 16S Sequences for Downstream Sanger Sequencing

[0216]

To amplify bacterial 16S rDNA (FIG. 1A), 2 μl of extracted gDNA is added to a 20 μl final volume PCR reaction. For full-length 16 sequencing the PCR reaction also contains 1× HotMasterMix (5PRIME, Gaithersburg, Md.), 250 nM of 27f (AGRGTTTGATCMTGGCTCAG, IDT, Coralville, Iowa), and 250 nM of 1492r (TACGGYTACCTTGTTAYGACTT, IDT, Coralville, Iowa), with PCR Water (Mo Bio Laboratories, Carlsbad, Calif.) for the balance of the volume. Alternatively, other universal bacterial primers or thermostable polymerases known to those skilled in the art are used. For example primers are available to those skilled in the art for the sequencing of the the “V1-V9 regions” of the 16S rRNA (FIG. 1A). These regions refer to the first through ninth hypervariable regions of the 16S rRNA gene that are used for genetic typing of bacterial samples. These regions in bacteria are defined by nucleotides 69-99, 137-242, 433-497, 576-682, 822-879, 986-1043, 1117-1173, 1243-1294 and 1435-1465 respectively using numbering based on the E. coli system of nomenclature. Brosius et al., Complete nucleotide sequence of a 16S ribosomal RNA gene from Escherichia coli, PNAS 75(10):4801-4805 (1978). In some embodiments, at least one of the V1, V2, V3, V4, V5, V6, V7, V8, and V9 regions are used to characterize an OTU. In one embodiment, the V1, V2, and V3 regions are used to characterize an OTU. In another embodiment, the V3, V4, and V5 regions are used to characterize an OTU. In another embodiment, the V4 region is used to characterize an OTU. A person of ordinary skill in the art can identify the specific hypervariable regions of a candidate 16S rRNA (in FIG. 1A) by comparing the candidate sequence in question to the reference sequence (FIG. 1B) and identifying the hypervariable regions based on similarity to the reference hypervariable regions.

[0217]

The PCR is performed on commercially available thermocyclers such as a BioRad MyCycler™ Thermal Cycler (BioRad, Hercules, Calif.). The reactions are run at 94° C. for 2 minutes followed by 30 cycles of 94° C. for 30 seconds, 51° C. for 30 seconds, and 68° C. for 1 minute 30 seconds, followed by a 7 minute extension at 72° C. and an indefinite hold at 4° C. Following PCR, gel electrophoresis of a portion of the reaction products is used to confirm successful amplification of a ˜1.5 kb product.

[0218]

To remove nucleotides and oligonucleotides from the PCR products, 2 μl of HT ExoSap-IT (Affymetrix, Santa Clara, Calif.) is added to 5 μl of PCR product followed by a 15 minute incubation at 37° C. and then a 15 minute inactivation at 80° C.

[0219]

Amplification of 16S Sequences for Downstream Characterization by Massively Parallel Sequencing Technologies

[0220]

Amplification performed for downstream sequencing by short read technologies such as Illumina require amplification using primers known to those skilled in the art that additionally include a sequence-based barcoded tag. As example, to amplify the 16s hypervariable region V4 region of bacterial 16S rDNA, 2 μl of extracted gDNA is added to a 20 μl final volume PCR reaction. The PCR reaction also contains 1× HotMasterMix (5PRIME, Gaithersburg, Md.), 200 nM of V4_515 f_adapt (AATGATACGGCGACCACCGAGATCTACACTATGGTAATTGTGTGCCAGCMGCCGC GGTAA, IDT, Coralville, Iowa), and 200 nM of barcoded 806rbc (CAAGCAGAAGACGGCATACGAGAT_12bpGolayBarcode_AGTCAGTCAGCCGGACT ACHVGGGTWTCTAAT, IDT, Coralville, Iowa), with PCR Water (Mo Bio Laboratories, Carlsbad, Calif.) for the balance of the volume. These primers incorporate barcoded adapters for Illumina sequencing by synthesis. Optionally, identical replicate, triplicate, or quadruplicate reactions may be performed. Alternatively other universal bacterial primers or thermostable polymerases known to those skilled in the art are used to obtain different amplification and sequencing error rates as well as results on alternative sequencing technologies.

[0221]

The PCR amplification is performed on commercially available thermocyclers such as a BioRad MyCycler™ Thermal Cycler (BioRad, Hercules, Calif.). The reactions are run at 94° C. for 3 minutes followed by 25 cycles of 94° C. for 45 seconds, 50° C. for 1 minute, and 72° C. for 1 minute 30 seconds, followed by a 10 minute extension at 72° C. and a indefinite hold at 4° C. Following PCR, gel electrophoresis of a portion of the reaction products is used to confirm successful amplification of a ˜1.5 kb product. PCR cleanup is performed as specified in the previous example.

[0222]

Sanger Sequencing of Target Amplicons from Pure Homogeneous Samples

[0223]

To detect nucleic acids for each sample, two sequencing reactions are performed to generate a forward and reverse sequencing read. For full-length 16s sequencing primers 27f and 1492r are used. 40 ng of ExoSap-IT-cleaned PCR products are mixed with 25 pmol of sequencing primer and Mo Bio Molecular Biology Grade Water (Mo Bio Laboratories, Carlsbad, Calif.) to 15 μl total volume. This reaction is submitted to a commercial sequencing organization such as Genewiz (South Plainfield, N.J.) for Sanger sequencing.

[0224]

Massively Parallel Sequencing of Target Amplicons from Heterogeneous Samples

[0225]

DNA Quantification & Library Construction.

[0226]

The cleaned PCR amplification products are quantified using the Quant-iT™ PicoGreen® dsDNA Assay Kit (Life Technologies, Grand Island, N.Y.) according to the manufacturer's instructions. Following quantification, the barcoded cleaned PCR products are combined such that each distinct PCR product is at an equimolar ratio to create a prepared Illumina library.

[0227]

Nucleic Acid Detection.

[0228]

The prepared library is sequenced on Illumina HiSeq or MiSeq sequencers (Illumina, San Diego, Calif.) with cluster generation, template hybridization, iso-thermal amplification, linearization, blocking and denaturization and hybridization of the sequencing primers performed according to the manufacturer's instructions. 16SV4SeqFw (TATGGTAATTGTGTGCCAGCMGCCGCGGTAA), 16SV4SeqRev (AGTCAGTCAGCCGGACTACHVGGGTWTCTAAT), and 16SV4Index (ATTAGAWACCCBDGTAGTCCGGCTGACTGACT) (IDT, Coralville, Iowa) are used for sequencing. Other sequencing technologies can be used such as but not limited to 454, Pacific Biosciences, Helicos, Ion Torrent, and Nanopore using protocols that are standard to someone skilled in the art of genomic sequencing.

Example 4

Sequence Read Annotation

[0229]

Primary Read Annotation

[0230]

Nucleic acid sequences are analyzed and annotations are to define taxonomic assignments using sequence similarity and phylogenetic placement methods or a combination of the two strategies. A similar approach can be used to annotate protein names, transcription factor names, and any other classification schema for nucleic acid sequences. Sequence similarity based methods include those familiar to individuals skilled in the art including, but not limited to BLAST, BLASTx, tBLASTn, tBLASTx, RDP-classifier, DNAclust, and various implementations of these algorithms such as Qiime or Mothur. These methods rely on mapping a sequence read to a reference database and selecting the match with the best score and e-value. Common databases include, but are not limited to the Human Microbiome Project, NCBI non-redundant database, Greengenes, RDP, and Silva. Phylogenetic methods can be used in combination with sequence similarity methods to improve the calling accuracy of an annotation or taxonomic assignment. Here tree topologies and nodal structure are used to refine the resolution of the analysis. In this approach we analyze nucleic acid sequences using one of numerous sequence similarity approaches and leverage phylogenetic methods that are well known to those skilled in the art, including but not limited to maximum likelihood phylogenetic reconstruction (see e.g. Liu K, Linder C R, and Warnow T. 2011. RAxML and FastTree: Comparing Two Methods for Large-Scale Maximum Likelihood Phylogeny Estimation. PLoS ONE 6: e27731. McGuire G, Denham M C, and Balding D J. 2001. Models of sequence evolution for DNA sequences containing gaps. Mol. Biol. Evol 18: 481-490. Wróbel B. 2008. Statistical measures of uncertainty for branches in phylogenetic trees inferred from molecular sequences by using model-based methods. J. Appl. Genet. 49: 49-67.) Sequence reads are placed into a reference phylogeny comprised of appropriate reference sequences. Annotations are made based on the placement of the read in the phylogenetic tree. The certainty or significance of the OTU annotation is defined based on the OTU's sequence similarity to a reference nucleic acid sequence and the proximity of the OTU sequence relative to one or more reference sequences in the phylogeny. As an example, the specificity of a taxonomic assignment is defined with confidence at the the level of Family, Genus, Species, or Strain with the confidence determined based on the position of bootstrap supported branches in the reference phylogenetic tree relative to the placement of the OTU sequence being interrogated.

[0231]

Clade Assignments

[0232]

The ability of 16S-V4 OTU identification to assign an OTU as a specific species depends in part on the resolving power of the 16S-V4 region of the 16S gene for a particular species or group of species. Both the density of available reference 16S sequences for different regions of the tree as well as the inherent variability in the 16S gene between different species will determine the definitiveness of a taxonomic annotation. Given the topological nature of a phylogenetic tree and the fact that tree represents hierarchical relationships of OTUs to one another based on their sequence similarity and an underlying evolutionary model, taxonomic annotations of a read can be rolled up to a higher level using a clade-based assignment procedure (Table 1). Using this approach, clades are defined based on the topology of a phylogenetic tree that is constructed from full-length 16S sequences using maximum likelihood or other phylogenetic models familiar to individuals with ordinary skill in the art of phylogenetics. Clades are constructed to ensure that all OTUs in a given clade are: (i) within a specified number of bootstrap supported nodes from one another (generally, 1-5 bootstraps), and (ii) within a 5% genetic similarity. OTUs that are within the same clade can be distinguished as genetically and phylogenetically distinct from OTUs in a different clade based on 16S-V4 sequence data. OTUs falling within the same clade are evolutionarily closely related and may or may not be distinguishable from one another using 16S-V4 sequence data. The power of clade based analysis is that members of the same clade, due to their evolutionary relatedness, are likely to play similar functional roles in a microbial ecology such as that found in the human gut. Compositions substituting one species with another from the same clade are likely to have conserved ecological function and therefore are useful in the present invention.

[0233]

Notably, 16S sequences of isolates of a given OTU are phylogenetically placed within their respective clades, sometimes in conflict with the microbiological-based assignment of species and genus that may have preceded 16S-based assignment. Discrepancies between taxonomic assignment based on microbiological characteristics versus genetic sequencing are known to exist from the literature.

Example 5

Germinating Spores

[0234]

Mixtures of bacteria can include species that are in spore form. Germinating a spore fraction increases the number of viable bacteria that will grow on various media types. To germinate a population of spores, the sample is moved to the anaerobic chamber, resuspended in prereduced PBS, mixed and incubated for 1 hour at 37 C to allow for germination. Germinants can include amino-acids (e.g., alanine, glycine), sugars (e.g., fructose), nucleosides (e.g., inosine), bile salts (e.g., cholate and taurocholate), metal cations (e.g., Mg2+, Ca2+), fatty acids, and long-chain alkyl amines (e.g., dodecylamine, Germination of bacterial spores with alkyl primary amines” J. Bacteriology, 1961.). Mixtures of these or more complex natural mixtures, such as rumen fluid or Oxgall, can be used to induce germination. Oxgall is dehydrated bovine bile composed of fatty acids, bile acids, inorganic salts, sulfates, bile pigments, cholesterol, mucin, lecithin, glycuronic acids, porphyrins, and urea. The germination can also be performed in a growth medium like prereduced BHIS/oxgall germination medium, in which BHIS (Brain heart infusion powder (37 g/L), yeast extract (5 g/L), L-cysteine HCl (1 g/L)) provides peptides, amino acids, inorganic ions and sugars in the complex BHI and yeast extract mixtures and Oxgall provides additional bile acid germinants.

[0235]

In addition, pressure may be used to germinate spores. The selection of germinants can vary with the microbe being sought. Different species require different germinants and different isolates of the same species can require different germinants for optimal germination. Finally, it is important to dilute the mixture prior to plating because some germinants are inhibitory to growth of the vegetative-state microorganisms. For instance, it has been shown that alkyl amines must be neutralized with anionic lipophiles in order to promote optimal growth. Bile acids can also inhibit growth of some organisms despite promoting their germination, and must be diluted away prior to plating for viable cells.

[0236]

For example, BHIS/oxgall solution is used as a germinant and contains 0.5×BHIS medium with 0.25% oxgall (dehydrated bovine bile) where 1×BHIS medium contains the following per L of solution: 6 g Brain Heart Infusion from solids, 7 g peptic digest of animal tissue, 14.5 g of pancreatic digest of casein, 5 g of yeast extract, 5 g sodium chloride, 2 g glucose, 2.5 g disodium phosphate, and 1 g cysteine. Additionally, Ca-DPA is a germinant and contains 40 mM CaCl2, and 40 mM dipicolinic acid (DPA). Rumen fluid (Bar Diamond, Inc.) is also a germinant. Simulated gastric fluid (Ricca Chemical) is a germinant and is 0.2% (w/v) Sodium Chloride in 0.7% (v/v) Hydrochloric Acid. Mucin medium is a germinant and prepared by adding the following items to 1 L of distilled sterile water: 0.4 g KH2PO4, 0.53 g Na2HPO4, 0.3 g NH4Cl, 0.3 g NaCl, 0.1 g MgCl2×6H2O, 0.11 g CaCl2, 1 ml alkaline trace element solution, 1 ml acid trace element solution, 1 ml vitamin solution, 0.5 mg resazurin, 4 g NaHCO3, 0.25 g Na2S×9H2O. The trace element and vitamin solutions prepared as described previously (Stams et al., 1993). All compounds were autoclaved, except the vitamins, which were filter-sterilized. The basal medium was supplemented with 0.7% (v/v) clarified, sterile rumen fluid and 0.25% (v/v) commercial hog gastric mucin (Type III; Sigma), purified by ethanol precipitation as described previously (Miller & Hoskins, 1981). This medium is referred herein as mucin medium.

[0237]

Fetal Bovine Serum (Gibco) can be used as a germinant and contains 5% FBS heat inactivated, in Phosphate Buffered Saline (PBS, Fisher Scientific) containing 0.137M Sodium Chloride, 0.0027M Potassium Chloride, 0.0119M Phosphate Buffer. Thioglycollate is a germinant as described previously (Kamiya et al Journal of Medical Microbiology 1989) and contains 0.25M (pH10) sodium thioglycollate. Dodecylamine solution containing 1 mM dodecylamine in PBS is a germinant. A sugar solution can be used as a germinant and contains 0.2% fructose, 0.2% glucose, and 0.2% mannitol. Amino acid solution can also be used as a germinant and contains 5 mM alanine, 1 mM arginine, 1 mM histidine, 1 mM lysine, 1 mM proline, 1 mM asparagine, 1 mM aspartic acid, 1 mM phenylalanine. A germinant mixture referred to herein as Germix 3 can be a germinant and contains 5 mM alanine, 1 mM arginine, 1 mM histidine, 1 mM lysine, 1 mM proline, 1 mM asparagine, 1 mM aspartic acid, 1 mM phenylalanine, 0.2% taurocholate, 0.2% fructose, 0.2% mannitol, 0.2% glucose, 1 mM inosine, 2.5 mM Ca-DPA, and 5 mM KCl. BHIS medium+ DPA is a germinant mixture and contains BHIS medium and 2 mM Ca-DPA. Escherichia coli spent medium supernatant referred to herein as EcSN is a germinant and is prepared by growing E. coli MG1655 in SweetB/Fos inulin medium anaerobically for 48 hr, spinning down cells at 20,000 rcf for 20 minutes, collecting the supernatant and heating to 60 C for 40 min. Finally, the solution is filter sterilized and used as a germinant solution.

Example 6

Selection of Media for Growth

[0238]

It is important to select appropriate media to support growth, including preferred carbon sources. For example, some organisms prefer complex sugars such as cellobiose over simple sugars. Examples of media used in the isolation of sporulating organisms include EYA, BHI, BHIS, and GAM (see below for complete names and references). Multiple dilutions are plated out to ensure that some plates will have well isolated colonies on them for analysis, or alternatively plates with dense colonies may scraped and suspended in PBS to generate a mixed diverse community.

[0239]

Plates are incubated anaerobically or aerobically at 37° C. for 48-72 or more hours, targeting anaerobic or aerobic spore formers, respectively.

[0240]

Solid plate media include:

    • Gifu Anaerobic Medium (GAM, Nissui) without dextrose supplemented with fructooligosaccharides/inulin (0.4%), mannitol (0.4%), inulin (0.4%), or fructose (0.4%), or a combination thereof.
    • Sweet GAM [Gifu Anaerobic Medium (GAM, Nissui)] modified, supplemented with glucose, cellobiose, maltose, L-arabinose, fructose, fructooligosaccharides/inulin, mannitol and sodium lactate)
    • Brucella Blood Agar (BBA, Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010)
    • PEA sheep blood (Anaerobe Systems; 5% Sheep Blood Agar with Phenylethyl Alcohol)
    • Egg Yolk Agar (EYA) (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010)
    • Sulfite polymyxin milk agar (Mevissen-Verhage et al., J. Clin. Microbiol. 25:285-289 (1987))
    • Mucin agar (Derrien et al., IJSEM 54: 1469-1476 (2004))
    • Polygalacturonate agar (Jensen & Canale-Parola, Appl. Environ. Microbiol. 52:880-997 (1986))
    • M2GSC (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010)
    • M2 agar (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) supplemented with starch (1%), mannitol (0.4%), lactate (1.5 g/L) or lactose (0.4%)
    • Sweet B-Brain Heart Infusion agar (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) supplemented with yeast extract (0.5%), hemin, cysteine (0.1%), maltose (0.1%), cellobiose (0.1%), soluble starch (sigma, 1%), MOPS (50 mM, pH 7).
    • PY-salicin (peptone-yeast extract agar supplemented with salicin) (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010).
    • Modified Brain Heart Infusion (M-BHI) [[sweet and sour]] contains the following per L: 37.5 g Brain Heart Infusion powder (Remel), 5 g yeast extract, 2.2 g meat extract, 1.2 g liver extract, 1 g cystein HCl, 0.3 g sodium thioglycolate, 10 mg hemin, 2 g soluble starch, 2 g FOS/Inulin, 1 g cellobiose, 1 g L-arabinose, 1 g mannitol, 1 Na-lactate, 1 mL Tween 80, 0.6 g MgSO4×7H2O, 0.6 g CaCl2, 6 g (NH4)2SO4, 3 g KH2PO4, 0.5 g K2HPO4, 33 mM Acetic acid, 9 mM propionic acid, 1 mM Isobutyric acid, 1 mM isovaleric acid, 15 g agar, and after autoclaving add 50 mL of 8% NaHCO3 solution and 50 mL 1M MOPS-KOH (pH 7).
    • Noack-Blaut Eubacterium agar (See Noack et al. J. Nutr. (1998) 128:1385-1391)
    • BHIS az1/ge2-BHIS az/ge agar (Reeves et. al. Infect. Immun. 80:3786-3794 (2012)) [Brain Heart Infusion agar (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) supplemented with yeast extract 0.5%, cysteine 0.1%, 0.1% cellobiose, 0.1% inulin, 0.1% maltose, aztreonam 1 mg/L, gentamycin 2 mg/L]
    • BHIS CInM az1/ge2-BHIS CInM [Brain Heart Infusion agar (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) supplemented with yeast extract 0.5%, cysteine 0.1%, 0.1% cellobiose, 0.1% inulin, 0.1% maltose, aztreonam 1 mg/L, gentamycin 2 mg/L]

Example 7

The Purification and Isolation of a Spore Forming Fraction from Feces

[0257]

To purify and selectively isolate efficacious spores from fecal material a donation is first blended with saline using a homogenization device (e.g., laboratory blender) to produce a 20% slurry (w/v). 100% ethanol is added for an inactivation treatment that lasts 10 seconds to 1 hour. The final alcohol concentration can range from 30-90%, preferably 50-70%. High speed centrifugation (3200 rcf for 10 min) is performed to remove solvent and the pellet is retained and washed. Subsequently, once the washed pellet is resuspended, a low speed centrifugation step (200 rcf for 4 min) is performed to remove large particulate vegetative matter and the supernatant containing the spores is retained. High speed centrifugation (3200 rcf for 10 min) is performed on the supernatant to concentrate the spore material. The pellet is then washed and resuspended to generate a 20% slurry. This is the ethanol treated spore preparation. The concentrated slurry is then separated with a density based gradient e.g. a CsCl gradient, sucrose gradient or combination of the two generating a ethanol treated, gradient-purified spore preparation. For example, a CsCl gradient is performed by loading a 20% volume of spore suspension on top a 80% volume of a stepwise CsCl gradient (w/v) containing the steps of 64%, 50%, 40% CsCl (w/v) and centrifuging for 20 min at 3200 rcf. The spore fraction is then run on a sucrose step gradient with steps of 67%, 50%, 40%, and 30% (w/v). When centrifuged in a swinging bucket rotor for 10 min at 3200 rcf. The spores run roughly in the 30% and 40% sucrose fractions. The lower spore fraction (FIG. 2) is then removed and washed to produce a concentrated ethanol treated, gradient-purified spore preparation. Taking advantage of the refractive properties of spores observed by phase contrast microscopy (spores are bright and refractive while germinated spores and vegetative cells are dark) one can see an enrichment of the spore fraction from a fecal bacterial cell suspension (FIG. 3, left) compared to an ethanol treated, CsCl gradient purified, spore preparation (FIG. 3, center), and to an ethanol treated, CsCl gradient purified, sucrose gradient purified, spore preparation (FIG. 3, right).

[0258]

Furthermore, growth of spores after treatment with a germinant can also be used to quantify a viable spore population. Briefly, samples were incubated with a germinant (Oxgall, 0.25% for up to 1 hour), diluted and plated anaerobically on BBA (Brucella Blood Agar) or similar media (e.g. see Examples 5 and 6). Individual colonies were picked and DNA isolated for full-length 16S sequencing to identify the species composition (e.g. see Examples 3 and 4). Analysis revealed that 22 species were observed in total (Table 4) with a vast majority present in both the material purified with the gradient and without the gradient, indicating no or inconsequential shift in the ecology as a result of gradient purification. Spore yield calculations demonstrate an efficient recovery of 38% of the spores from the initial fecal material as measured by germination and plating of spores on BBA or measuring DPA count in the sample.

Example 8

Bacterial Compositions Prevent C. difficile Infection in a Mouse Model

[0259]

To test the therapeutic potential of the bacterial composition such as but not limited to a spore population, a prophylactic mouse model of C. difficile infection (model based on Chen, et al., A mouse model of Clostridium difficile associated disease, Gastroenterology 135(6):1984-1992) was used. Two cages of five mice each were tested for each arm of the experiment. All mice received an antibiotic cocktail consisting of 10% glucose, kanamycin (0.5 mg/ml), gentamicin (0.044 mg/ml), colistin (1062.5 U/ml), metronidazole (0.269 mg/ml), ciprofloxacin (0.156 mg/ml), ampicillin (0.1 mg/ml) and Vancomycin (0.056 mg/ml) in their drinking water on days −14 through −5 and a dose of 10 mg/kg Clindamycin by oral gavage on day −3. On day −1, they received either the test article or vehicle control via oral gavage. On day 0 they were challenged by administration of approximately 4.5 log 10 cfu of C. difficile (ATCC 43255) via oral gavage. Optionally a positive control group received vancomycin from day −1 through day 3 in addition to the antibiotic protocol and C. difficile challenge specified above. Feces were collected from the cages for analysis of bacterial carriage, mortality was assessed every day from day 0 to day 6 and the weight and subsequent weight change of the animal was assessed with weight loss being associated with C. difficile infection. Mortality and reduced weight loss of the test article compared to the vehicle were used to assess the success of the test article. Additionally, a C. difficile symptom scoring was performed each day from day −1 through day 6. Clinical Score was based on a 0-4 scale by combining scores for Appearance (0-2 pts based on normal, hunched, piloerection, or lethargic), and Clinical Signs (0-2 points based on normal, wet tail, cold-to-the-touch, or isolation from other animals).

[0260]

In a naive control arm, animals were challenged with C. difficile. In the vancomycin positive control arm animals were dosed with C. difficile and treated with vancomycin from day −1 through day 3. The negative control was gavaged with PBS alone and no bacteria. The test arms of the experiment tested 1×, 0.1×, 0.01× dilutions derived from a single donor preparation of ethanol treated spores (e.g. see Example 7) or the heat treated feces prepared by treating a 20% slurry for 30 min at 80 C. Dosing for CFU counts was determined from the final ethanol treated spores and dilutions of total spores were administered at 1×, 0.1×, 0.01× of the spore mixture for the ethanol treated fraction and a 1× dose for the heat treated fraction.

[0261]

Weight loss and mortality were assessed on day 3. The negative control, treated with C. difficile only, exhibits 20% mortality and weight loss on Day 3, while the positive control of 10% human fecal suspension displays no mortality or weight loss on Day 3 (Table 5). EtOH-treated feces prevents mortality and weight loss at three dilutions, while the heat-treated fraction was protective at the only dose tested. These data indicate that the spore fraction is efficacious in preventing C. difficile infection in the mouse.

Example 9

The Prophylactic and Relapse Prevention Hamster Models

[0262]

Previous studies with hamsters using toxigenic and nontoxigenic strains of C. difficile demonstrated the utility of the hamster model in examining relapse post antibiotic treatment and the effects of prophylaxis treatments with cecal flora in C. difficile infection (Wilson et al. 1981, Wilson et al. 1983, Borriello et al. 1985) and more broadly gastrointestinal infectious disease. To demonstrate prophylactic use of a bacterial composition such as but not limited to a spore population, spore preparation, vegetative cell population, to ameliorate C. difficile infection, the following hamster model is used. In a prophylactic model, Clindamycin (10 mg/kg s.c.) is given on day −5, the bacterial composition or control is administered on day −3, and C. difficile challenge occurs on day 0. In the positive control arm, vancomycin is then administered on day 1-5 (and vehicle control is delivered on day −3). Feces are collected on day −5, −4, −1, 1, 3, 5, 7, 9 and fecal samples are assessed for pathogen carriage and reduction by microbiological methods, 16S sequencing approaches or other methods utilized by one skilled in the art. Mortality is assessed throughout the experiment through 21 days post C. difficile challenge. The percentage survival curves show that ethanol treated spores and ethanol treated, gradient-purified spores better protect the hamsters compared to the Vancomycin control, and vehicle control.

[0263]

FIG. 4 shows prophylaxis model with the ethanol treated spore preparation and the ethanol treated, gradient-purified spore preparation.

[0264]

In the relapse prevention model, hamsters are challenged with toxigenic C. difficile strains on day 0, and treated with clindamycin by oral gavage on day 1, and vancomycin dosing day 2-6. Test or control treatment was then administered on day 7, 8, and 9. The groups of hamsters for each arm consist of 8 hamsters per group. Fecal material is collected on day −1, 1, 3, 5, 7, 10 and 13 and hamster mortality is assessed throughout. Survival curves are used to assess the success of the test article e.g. ethanol treated or ethanol treated, gradient purified spores versus the control treatment in preventing hamster death. The survival curves demonstrate maximum efficacy for the ethanol treated, gradient-purified spores followed by the ethanol treated spores. Both treatments improved survival percentage over vancomycin treatment alone.

[0265]

FIG. 5 shows relapse prevention model with ethanol treated spores and ethanol treated, gradient purified spores

Example 10

Clinical Treatment of Recurrent C. difficile in Patients

[0266]

To assess the efficacy of test articles like bacterial compositions including but not limited to a ethanol treated spore preparations (e.g. see Example 7) to treat recurrent C. difficile in human patients, the following procedure was performed to take feces from a healthy donor, inactivate via the ethanol treated spore preparation protocol described below, and treat recurrent C. difficile in patients presenting with this indication. Non-related donors were screened for general health history for absence of chronic medical conditions (including inflammatory bowel disease; irritable bowel syndrome; Celiac disease; or any history of gastrointestinal malignancy or polyposis), absence of risk factors for transmissible infections, antibiotic non-use in the previous 6 months, and negative results in laboratory assays for blood-borne pathogens (HIV, HTLV, HCV, HBV, CMV, HAV and Treponema pallidum) and fecal bacterial pathogens (Salmonella, Shigella, Yersinia, Campylobacter, E. coli 0157), ova and parasites, and other infectious agents (Giardia, Cryptosporidium Cyclospora, Isospora) prior to stool donation.

[0267]

Donor stool was frozen shortly after donation and sampled for testing. At the time of use, approximately 75 g of donor stool was thawed and resuspended in 500 mL of non-bacteriostatic normal saline and mixed in a single use glass or plastic blender. The resulting slurry was sequentially passed through sterile, disposable mesh screens that remove particles of size 600, 300 and 200 microns. The slurry was then centrifuged briefly (200 rcf for 4 min) to separate fibrous and particulate materials, and the supernatant (containing bacterial cells and spores) was transferred to a fresh container. Ethanol was added to a final concentration of 50% and the resulting ˜1500 ml slurry was incubated at room temperature for 1 hr with continuous mixing to inactivate vegetative bacterial cells. Midway through inactivation the slurry was transferred to a new bottle to ensure complete contact with the ethanol. The solid matter was pelleted in a centrifuge and washed 3 times with normal saline to remove residual ethanol. The final pellet was resuspended in 100% sterile, USP glycerol at a minimum volume, and filled into approximately 30 size 0 delayed release capsules (hypromellose DRcaps, Capsugel, Inc.) at 0.65 mL suspension each. The capsules were immediately capped and placed onto an aluminum freezing block held at −80° C. via dry ice to freeze. The frozen capsules were in turn over-capsulated with size 00 DRcaps to enhance capsule stability, labeled, and placed into <−65° C. storage immediately. The final product was stored at <−65° C. until the day and time of use. Encapsulated product may be stored for indefinitely at <−65° C. On the day of dosing capsules were warmed on wet ice for 1 to 2 hours to improve tolerability, and were then dosed with water ad libitium.

[0268]

Patient 1 is a 45-year old woman with a history of C. difficile infection and diarrhea for at least 1 year prior to treatment. She has been previously treated with multiple courses of antibiotics followed each time by recurrence of C. difficile-associated diarrhea.

[0269]

Patient 2 is an 81-year old female who has experienced recurrent C. difficile infection for 6 months prior to treatment despite adequate antibiotic therapy following each recurrence.

[0270]

24 hours prior to starting oral treatment, CDAD antibiotic therapy was discontinued. Each patient received a colon preparation procedure intended to reduce the competing microbial burden in the gastrointestinal tract and to facilitate repopulation by the spore forming organisms in the investigational product.

[0271]

On the morning of the first treatment day, the patients received a dose of delayed release capsules containing the investigational product with water ad libitum. Patients were requested to avoid food for 1 hour thereafter. The next day, the patient returned to the clinic to receive an additional dose. Patients were asked to avoid food for 4 hours prior to receiving their second dose and for 1 hour following dosing.

[0272]

Both patients were followed closely for evidence of relapse or adverse symptoms following treatment. Patients were contacted by phone on Day 2, Day 4, and Weeks 1, 2 and 4 and each was queried about her general status and the condition of her CDAD and related symptoms. Stool samples were collected at baseline and Weeks 1, 2, 4 and 8 post-treatment to assess changes in the gut microbiota via 16S sequencing and spore count with methods explained previously (e.g. see Examples AAAB and AAAC). Through 4 weeks post treatment, each patient has gradually improved with no evidence of C. difficile recurrence.

[0273]

Six other patients with recurrent C. difficile-associated diarrhea were treated in a similar fashion, with no CU recurrence and no requirement for resumption of antibiotics (total of 8 patients). Additionally, there were no treatment-related serious adverse events.

[0274]

The above protocol could be modified to deliver other bacterial compositions e.g. vegetative cells, spore preparations, combinations thereof.

Example 11

Enrichment and Purification of Bacteria

[0275]

To purify individual bacterial strains, dilution plates were selected in which the density enables distinct separation of single colonies. Colonies were picked with a sterile implement (either a sterile loop or toothpick) and re-streaked to BBA or other solid media. Plates were incubated at 37° C. for 3-7 days. One or more well-isolated single colonies of the major morphology type were re-streaked. This process was repeated at least three times until a single, stable colony morphology is observed. The isolated microbe was then cultured anaerobically in liquid media for 24 hours or longer to obtain a pure culture of 106-1010cfu/ml. Liquid growth medium might include Brain Heart Infusion-based medium (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) supplemented with yeast extract, hemin, cysteine, and carbohydrates (for example, maltose, cellobiose, soluble starch) or other media described previously (e.g. see example 6). The culture was centrifuged at 10,000×g for 5 min to pellet the bacteria, the spent culture media was removed, and the bacteria were resuspended in sterile PBS. Sterile 75% glycerol was added to a final concentration of 20%. An aliquot of glycerol stock was titered by serial dilution and plating. The remainder of the stock was frozen on dry ice for 10-15 min and then placed at −80 C for long term storage.

Example 12

Cell Bank Preparation

[0276]

Cell banks (RCBs) of bacterial strains were prepared as follows. Bacterial strains were struck from −80° C. frozen glycerol stocks to Brucella blood agar with Hemin or Vitamin K (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010), M2GSC (Atlas, Handbook of Microbiological Media, 4th ed, ASM Press, 2010) or other solid growth media and incubated for 24 to 48 h at 37° C. in an anaerobic chamber with a gas mixture of H2:CO2:N2of 10:10:80. Single colonies were then picked and used to inoculate 250 ml to 1 L of Wilkins-Chalgren broth, Brain-Heart Infusion broth, M2GSC broth or other growth media, and grown to mid to late exponential phase or into the stationary phase of growth. Alternatively, the single colonies may be used to inoculate a pilot culture of 10 ml, which were then used to inoculate a large volume culture. The growth media and the growth phase at harvest were selected to enhance cell titer, sporulation (if desired) and phenotypes that might be associated desired in vitro or in vivo. Optionally, Cultures were grown static or shaking, depending which yielded maximal cell titer. The cultures were then concentrated 10 fold or more by centrifugation at 5000 rpm for 20 min, and resuspended in sterile phosphate buffered saline (PBS) plus 15% glycerol. 1 ml aliquots were transferred into 1.8 ml cryovials which were then frozen on dry ice and stored at −80 C. The identity of a given cell bank was confirmed by PCR amplification of the 16S rDNA gene, followed by Sanger direct cycle sequencing, and comparison to a curated rDNA database to determine a taxonomic ID. Each bank was confirmed to yield colonies of a single morphology upon streaking to Brucella blood agar or M2GSC agar. When more than one morphology was observed, colonies were confirmed to be the expected species by PCR and sequencing analysis of the 16S rDNA gene. Variant colony morphologies can be observed within pure cultures, and in a variety of bacteria the mechanisms of varying colony morphologies have been well described (van der Woude, Clinical Microbiology Reviews, 17:518, 2004), including in Clostridium species (Wadsworth-KTL Anaerobic Bacteriology Manual, 6th Ed, Jousimie-Somer, et al 2002). For obligate anaerobes, RCBs were confirmed to lack aerobic colony forming units at a limit of detection of 10 cfu/ml.

Example 13

Titer Determination

[0277]

The number of viable cells per ml was determined on the freshly harvested, washed and concentrated culture by plating serial dilutions of the RCB to Brucella blood agar or other solid media, and varied from 106 to 1010 cfu/ml. The impact of freezing on viability was determined by titering the banks after one or two freeze-thaw cycles on dry ice or at −80° C., followed by thawing in an anaerobic chamber at room temperature. Some strains displayed a 1-3 log drop in viable cfu/ml after the 1st and/or 2nd freeze thaw, while the viability of others were unaffected.

Example 14

Preparation of Bacterial Compositions

[0278]

Individual strains were typically thawed on ice and combined in an anaerobic chamber to create mixtures, followed by a second freeze at −80° C. to preserve the mixed samples. When making combinations of strains for in vitro or in vivo assays, the cfu in the final mixture was estimated based on the second freeze-thaw titer of the individual strains. For experiments in rodents, strains may be combined at equal counts in order to deliver between 1e4 and 1e10 per strain. Additionally, some bacteria may not grow to sufficient titer to yield cell banks that allowed the production of compositions where all bacteria were present at 1e10.

Example 15

Provision of Out Microbiome Sample Material

[0279]

For sourcing of microbial cultures and for use as a positive control in in vivo studies, fresh gut microbiome samples, e.g. fecal samples, were obtained from healthy human donors who have been screened for general good health and for the absence of infectious diseases, and meet inclusion and exclusion criteria, inclusion criteria include being in good general health, without significant medical history, physical examination findings, or clinical laboratory abnormalities, regular bowel movements with stool appearance typically Type 2, 3, 4, 5 or 6 on the Bristol Stool Scale, and having a BMI≧18 kg/m2and ≦25 kg/m2. Exclusion criteria generally included significant chronic or acute medical conditions including renal, hepatic, pulmonary, gastrointestinal, cardiovascular, genitourinary, endocrine, immunologic, metabolic, neurologic or hematological disease, a family history of, inflammatory bowel disease including Crohn's disease and ulcerative colitis, Irritable bowel syndrome, colon, stomach or other gastrointestinal malignancies, or gastrointestinal polyposis syndromes, or recent use of yogurt or commercial probiotic materials in which an organism(s) is a primary component. Samples were collected directly using a commode specimen collection system, which contains a plastic support placed on the toilet seat and a collection container that rests on the support. Gut microbiome samples e.g. feces were deposited into the container, and the lid was then placed on the container and sealed tightly. The sample was then delivered on ice within 1-4 hours for processing. Samples were mixed with a sterile disposable tool, and 2-4 g aliquots were weighed and placed into tubes and flash frozen in a dry ice/ethanol bath. Aliquots are frozen at −80 degrees Celsius until use.

[0280]

Optionally, the microbiome sample was suspended in a solution, and/or fibrous and/or particulate materials were removed. A frozen aliquot containing a known weight of sample was removed from storage at −80 degrees Celsius and allowed to thaw at room temperature. Sterile 1×PBS was added to create a 10% w/v suspension, and vigorous vortexing was performed to suspend the sample until the material appeared homogeneous. The sample was then left to sit for 10 minutes at room temperature to sediment fibrous and particulate matter. The suspension above the sediment was then carefully removed into a new tube and contains a purified spore population. Optionally, the suspension was then centrifuged at a low speed, e.g., 1000×g, for 5 minutes to pellet particulate matter including fibers. The pellet was discarded and the supernatant, which contained vegetative organisms and spores, was removed into a new tube. The supernatant was then centrifuged at 6000×g for 10 minutes to pellet the vegetative organisms and spores. The pellet was then resuspended in 1× PBS with vigorous vortexing until the sample material appears homogenous.

Example 16

Quantification of Spore Concentrations Using DPA Assay

[0281]

Methods to assess spore concentration in complex mixtures typically require the separation and selection of spores and subsequent growth of individual species to determine the colony forming units. The art does not teach how to quantitatively germinate all the spores in a complex mixture as there are many species for which appropriate germinants have not been identified. Furthermore, sporulation is thought to be a stochastic process as a result of evolutionary selection, meaning that not all spores from a single species germinate with same response to germinant concentration, time and other environmental conditions. Alternatively, a key metabolite of bacterial spores, dipicolinic acid (DPA) has been developed to quantify spores particles in a sample and avoid interference from fecal contaminants. The assay utilizes the fact that DPA chelates Terbium 3+ to form a luminescent complex (Fichtel et al, FEMS Microbiology Ecology, 2007; Kort et al, Applied and Environmental Microbiology, 2005; Shafaat and Ponce, Applied and Environmental Microbiology, 2006; Yang and Ponce, International Journal of Food Microbiology, 2009; Hindle and Hall, Analyst, 1999). A time-resolved fluorescence assay detects terbium luminescence in the presence of DPA giving a quantitative measurement of DPA concentration in a solution.

[0282]

To perform the assay 1 mL of the spore standard to be measured was transferred to a 2 mL microcentrifuge tube. The samples were centrifuged at 13000 RCF for 10 min and the sample is washed in 1 mL sterile deionized H2O. Wash an additional time by repeating the centrifugation. Transfer the 1 mL solution to hungate tubes and autoclave samples on a steam cycle for 30 min at 250 C. Add 100 uL of 30 uM TbCl3solution (400 mM sodium acetate, pH 5.0, 30 μM TbCl3) to the sample. Make serial dilutions of of the autoclaved material and measure the fluorescence of each sample by exciting with 275 nm light and measuring the emission wavelength of 543 nm for an integration time of 1.25 ms and a 0.1 ms delay.

[0283]

Purified spores are produced as described previously (e.g. see http://www.epa.gov/pesticides/methods/MB-28-00.pdf). Serial dilutions of purified spores from C. bifermentans, C. sporogenes, and C. butyricum cultures were prepared and measured by plating on BBA media and incubating overnight at 37 C to determine CFU/ml. FIG. 6 shows the linear correspondence across different spore producing bacteria across several logs demonstrating the DPA assay as means to assess spore content (linear range of DPA assay compared to CFU counts/ml).

[0284]

The discrepancy for complex spore populations between spore counts measured by germinable spore CFU and by DPA has important implications for determining the potency of an ethanol treated spore preparation for clinical use. Table 6 shows spore content data from 3 different ethanol treated spore preparations used to successfully treat 3 patients suffering from recurrent C. difficile infection. The spore content of each spore preparation is characterized using the two described methods.

[0285]

What is immediately apparent is that spore content varies greatly per 30 capsules. As measured by germinable SCFU, spore content varies by greater than 10,000-fold. As measured by DPA, spore content varies by greater than 100-fold. In the absence of the DPA assay, it would be difficult to set a minimum dose for administration to a patient. For instance, without data from the DPA assay, one would conclude that a minimum effective dose of spores is 4×105 or less using the SCFU assay (e.g. Preparation 1, Table 7). If that SCFU dose was used to normalize dosing in a clinical setting, however, then the actual spore doses given to patients would be much lower for other ethanol treated spore preparations as measured as by the DPA assay (Table 7).

[0286]

It becomes immediately obvious from the variability of SCFU and DPA counts across various donations that using SCFU as the measure of potency would lead to significant underdosing in certain cases. For instance, setting a dose specification of 4×105SCFU (the apparent effective dose from donor Preparation 1) for product Preparation 3 would lead to a potential underdosing of more than 100-fold. This can be rectified only by setting potency specifications based on the DPA assay which better reflects total spore counts in an ethanol treated spore preparation. The unexpected finding of this work is that the DPA assay is uniquely suited to set potency and determine dosing for an ethanol treated spore preparation.

Example 17

Identification of Keystone OTUs and Functions

[0287]

The human body is an ecosystem in which the microbiota, and the microbiome, play a significant role in the basic healthy function of human systems (e.g. metabolic, immunological, and neurological). The microbiota and resulting microbiome comprise an ecology of microorganisms that co-exist within single subjects interacting with one another and their host (i.e., the mammalian subject) to form a dynamic unit with inherent biodiversity and functional characteristics. Within these networks of interacting microbes (i.e. ecologies), particular members can contribute more significantly than others; as such these members are also found in many different ecologies, and the loss of these microbes from the ecology can have a significant impact on the functional capabilities of the specific ecology. Robert Paine coined the concept “Keystone Species” in 1969 (see Paine R T. 1969. A note on trophic complexity and community stability. The American Naturalist 103: 91-93.) to describe the existence of such lynchpin species that are integral to a given ecosystem regardless of their abundance in the ecological community. Paine originally describe the role of the starfish Pisaster ochraceus in marine systems and since the concept has been experimentally validated in numerous ecosystems.

[0288]

Keystone OTUs and/or Functions are computationally-derived by analysis of network ecologies elucidated from a defined set of samples that share a specific phenotype. Keystone OTUs and/or Functions are defined as all Nodes within a defined set of networks that meet two or more of the following criteria. Using Criterion 1, the node is frequently observed in networks, and the networks in which the node is observed are found in a large number of individual subjects; the frequency of occurrence of these Nodes in networks and the pervasiveness of the networks in individuals indicates these Nodes perform an important biological function in many individuals. Using Criterion 2, the node is frequently observed in networks, and each the networks in which the node is observed contain a large number of Nodes—these Nodes are thus “super-connectors”, meaning that they form a nucleus of a majority of networks and as such have high biological significance with respect to their functional contributions to a given ecology. Using Criterion 3, the node is found in networks containing a large number of Nodes (i.e. they are large networks), and the networks in which the node is found occur in a large number of subjects; these networks are potentially of high interest as it is unlikely that large networks occurring in many individuals would occur by chance alone strongly suggesting biological relevance. Optionally, the required thresholds for the frequency at which a node is observed in network ecologies, the frequency at which a given network is observed across subject samples, and the size of a given network to be considered a Keystone node are defined by the 50th, 70th, 80th, or 90th percentiles of the distribution of these variables. Optionally, the required thresholds are defined by the value for a given variable that is significantly different from the mean or median value for a given variable using standard parametric or non-parametric measures of statistical significance. In another embodiment a Keystone node is defined as one that occurs in a sample phenotype of interest such as but not limited to “health” and simultaneously does not occur in a sample phenotype that is not of interest such as but not limited to “disease.” Optionally, a Keystone Node is defined as one that is shown to be significantly different from what is observed using permuted test datasets to measure significance.

Example 18

Prophylactic Use and Treatment in a Mouse Model of Vancomycin Resistant Enterococcus (VRE) Colonization

[0289]

The emergence and spread of highly antibiotic-resistant bacteria represent a major clinical challenge (Snitkin et al Science Translational Medicine, 2012). In recent years, the numbers of infections caused by organisms such as methicillin-resistant Staphylococcus aureus, carbapenem-resistant Enterobacteriaceae, vancomycin-resistant Enterococcus (VRE), and Clostridium difficile have increased markedly, and many of these strains are acquiring resistance to the few remaining active antibiotics. Most infections produced by highly antibiotic-resistant bacteria are acquired during hospitalizations, and preventing patient-to-patient transmission of these pathogens is one of the major challenges confronting hospitals and clinics. Most highly antibiotic-resistant bacterial strains belong to genera that colonize mucosal surfaces, usually at low densities. The highly complex microbiota that normally colonizes mucosal surfaces inhibits expansion of and domination by bacteria such as Enterobacteriaceae and Enterococcaceae. Destruction of the normal flora by antibiotic administration, however, disinhibition antibiotic-resistant members of these bacterial families, leading to their expansion to very high densities (Ubeda et al Journal of Clinical Investigation 2010). High-density colonization by these organisms can be calamitous for the susceptible patient, resulting in bacteremia and sepsis (Taur et al, Clinical Infectious Disease, 2012).

[0290]

To test prophylactic use and treatment of a bacterial composition test article, a VRE infection mouse model is used as previously described (Ubeda et al, Infectious Immunity 2013, Ubeda et al, Journal of clinical investigation, 2010). Briefly, experiments are done with 7-week-old C57BL/6 J female mice purchased from Jackson Laboratory, housed with irradiated food, and provided with acidified water. Mice are individually housed to avoid contamination between mice due to coprophagia. For experimental infections with VRE, mice are treated with ampicillin (0.5 g/liter) in their drinking water, which is changed every 3 days.

[0291]

In the treatment model, on day 1, mice are infected by means of oral gavage with 108 CFU of the vancomycin-resistant Enterococcus faecium strain purchased from ATCC (ATCC 700221). One day after infection (day 1), antibiotic treatment is stopped and VRE levels are determined at different time points by plating serial dilutions of fecal pellets on Enterococcosel agar plates (Difco) with vancomycin (8 ug/ml; Sigma). VRE colonies are identified by appearance and confirmed by Gram staining or other methods previously described (e.g. see examples 2, 3 and 4). In addition, as previously described (Ubeda et al, Journal of Clinical Investigation 2010), PCR of the vanA gene, which confers resistance to vancomycin, confirms the presence of VRE in infected mice. The bacterial composition test article such as but not limited to an ethanol treated, gradient purified spore preparation (as described herein), fecal suspension, or antibiotic treatment is delivered in PBS on days 1-3 while the negative control contains only PBS and is also delivered on days 1-3 by oral gavage. Fresh fecal stool pellets are obtained daily for the duration of the experiment from days −7 to day 10. The samples are immediately frozen and stored at −80° C. DNA was extracted using standard techniques and analyzed with 16S or comparable methods (e.g. see example 3 and 4).

[0292]

In the colonization model, ampicillin is administered as described above for day −7 to day 1, treatment with the test article or vehicle control is administered on day 0-2 and the VRE resistant bacteria at 108 CFU are administered on day 14. Fecal samples are taken throughout the experiment daily from −7 to day 21 and submitted for 16S sequencing as previously described (e.g. see examples 3 and 4).

[0293]

In both models titers of VRE in feces are used to evaluate the success of the test article versus the negative control. Furthermore, microbiota composition is assessed for the ability of the bacterial composition test article to induce a healthy microbiome.

Example 19

Prophylactic Use and Treatment of a Mouse Model of Carbapenem Resistant Klebsiella (CRKB) Colonization

[0294]

The emergence of Klebsiella pneumoniae strains with decreased susceptibility to carbapenems is a significant threat to hospitalized patients. Resistance to carbapenems in these organisms is most frequently mediated by K. pneumoniae carbapenemase (KPC), a class A beta-lactamase that also confers resistance to broad-spectrum cephalosporins and commercially available beta-lactam/beta-lactamase inhibitor combinations (Queenan et al, Clinical Microbiology Review, 2007). KPC-producing K. pneumoniae (KPC-Kp) strains often harbor resistance determinants against several other classes of antimicrobials, including aminoglycosides and fluoroquinolones, resulting in truly multidrug-resistant (MDR) organisms (Hirsch et al, Journal of Antimicrobial Chemotherapy, 2009). Considering the limited antimicrobial options, infections caused by KPC-Kp pose a tremendous therapeutic challenge and are associated with poor clinical outcomes

[0295]

A treatment protocol in a mouse model as previously described (e.g. Perez et al, Antimicrobial Agents Chemotherapy, 2011) is used to evaluate the bacterial composition (test article) for treating carbapenem resistant Klebsiella and reducing carriage in the GI tract. Female CF1 mice (Harlan Sprague-Dawley, Indianapolis, Ind.) are used and are individually housed and weighed between 25 and 30 g.

[0296]

The thoroughly characterized strain of K. pneumoniae, VA-367 (8, 9, 25) is used in this study. This clinical isolate is genetically related to the KPC-Kp strain circulating in the Eastern United States. Characterization of the resistance mechanisms in K. pneumoniae VA-367 with PCR and DNA sequence analysis revealed the presence of blaKPC-3, blaTEM-1, blaSHV-11, and blaSHV-12as well as qnrB19 and aac(6′)-lb. Additionally, PCR and DNA sequencing revealed disruptions in the coding sequences of the following outer membrane protein genes: ompK35, ompK36, and ompK37. Antibiotic susceptibility testing (AST) was performed with the agar dilution method and interpreted according to current recommendations from the Clinical and Laboratory Standards Institute (CLSI). A modified Hodge test were performed, according to a method described previously (e.g. see Anderson et al, Journal of Clinical Microbiology, 2007) with ertapenem, meropenem, and imipenem. Tigecycline and polymyxin E were evaluated by Etest susceptibility assays (AB bioM'erieux, Solna, Sweden). Results for tigecycline were interpreted as suggested by the U.S. Food and Drug Administration (FDA) and according to CLSI recommendations (criteria for Pseudomonas) for polymyxin E.

[0297]

Mice (10 per group) are assigned to either a bacterial composition (test article), ethanol treated, spore preparation (e.g. see example 7), antibiotic clindamycin, piperacillin-tazobactam, tigecycline, ertapenem, cefepime, ciprofloxacin, or combination thereof or control group receiving only the vehicle. They are administered the test article daily from day −10 to day 0, On day 0, 103CFU of KPC-Kp VA-367 diluted in 0.5 ml phosphate-buffered saline (PBS) was administered by oral gavage using a stainless-steel feeding tube (Perfektum; Popper & Sons, New Hyde Park, N.Y.). Stool samples were collected 1, 4, 6, and 11 days after the administration of KPC-Kp in order to measure the concentration of carbapenem-resistant K. pneumoniae. Stool samples (100 mg diluted in 800 ml of PBS) are plated onto MacConkey agar with and without 0.5 ug/ml of imipenem, and the number of CFU per gram of stool was determined. Alternatively other methods may be used to measure the levels of carbapenem-resistant K. pneumoniae e.g. per, antigen testing, as one who's skilled in the art could perform.

[0298]

Stool samples were collected after 5 days of treatment to assess the effects of the antibiotics on the stool microflora and to measure antibiotic levels in stool. To assess the effects on the microflora, fresh stool samples as previously described (e.g. see examples AAAB and AAAC). Additional experiments are performed to examine whether the administration the bacterial composition (test article) resulted in the elimination or persistence of colonization with KPC-Kp VA-367.

[0299]

Mice are treated with subcutaneous clindamycin to reduce the normal intestinal flora 1 day before receiving 104 CFU of KPC-Kp VA-367 by oral gavage, and the mice continued to receive subcutaneous clindamycin every other day for 7 days. Concurrently, for 7 days after oral gavage with KPC-Kp, mice received oral gavage of normal saline (control group), or the bacterial composition as specified. An additional dose of subcutaneous clindamycin was administered 20 days after the administration of KPC-Kp VA-367 to assess whether low levels of carbapenem-resistant K. pneumoniae were present that could be augmented by the elimination of the anaerobic microflora. Stool samples were collected at baseline and at 3, 6, 8, 11, 16, and 21 days after KPC-Kp VA-367 was given by gavage. The bacterial composition will be examined by the reduction of CRKB in feces.

Example 20

Methods of Construction and Quantification

[0300]

Construction of Binary Pairs in a High-Throughput 96-Well Format.

[0301]

To allow high-throughput screening of binary pairs, vials of −80° C. glycerol stock banks were thawed and diluted to 1e8 CFU/mL. Each strain was then diluted 10× (to a final concentration of 1e7 CFU/mL of each strain) into 200 uL of PBS+15% glycerol in the wells of a 96-well plate. Plates were then frozen at −80° C. When needed, plates were removed from −80° C. and thawed at room temperature under anaerobic conditions when testing in an In vitro inhibition assay with Clostridium difficile.

[0302]

Construction of Ternary Combinations in a High-Throughput 96-Well Format

[0303]

To allow high-throughput screening of ternary combinations, vials of −80° C. glycerol stock banks were thawed and diluted to 1e8 CFU/mL. Each strain was then diluted 10× (to a final concentration of 1e7 CFU/mL of each strain) into 200 uL of PBS+15% glycerol in the wells of a 96-well plate. Plates were then frozen at −80° C. When needed for the assay, plates were removed from −80° C. and thawed at room temperature under anaerobic conditions when testing in an In vitro inhibition assay with Clostridium difficile.

[0304]

Construction of an In Vitro Inhibition Assay to Screen for Ecobiotic™ Compositions Inhibitory to the Growth of Clostridium difficile

[0305]

An overnight culture of Clostridium difficile was grown under anaerobic conditions in SweetB-FosIn or other suitable media for the growth of C. difficile. SweetB-FosIn is a complex media composed of brain heart infusion, yeast extract, cysteine, cellobiose, maltose, soluble starch, and fructooligosaccharides/inulin, and hemin, and is buffered with MOPs. After 24 hr of growth the culture was diluted 100,000 fold into a complex media such as SweetB-FosIn which is suitable for the growth of a wide variety of anaerobic bacterial species. The diluted C. difficile mixture was then aliquoted to wells of a 96-well plate (180 uL to each well). 20 uL of a unique binary pair of potential inhibitory species was then added to each well at a final concentration of 1e6 CFU/mL of each species. Alternatively the assay can be tested with binary pairs at different initial concentrations (1e9 CFU/mL, 1e8 CFU/mL, 1e7 CFU/mL, 1e5 CFU/mL, 1e4 CFU/mL, 1e3 CFU/mL, 1e2 CFU/mL). Control wells only inoculated with C. difficile were included for a comparison to the growth of C. difficile without inhibition. Additional wells were used for controls that either inhibit or do not inhibit the growth of C. difficile. One example of a positive control that inhibits growth was a combination of Blautia producta, Clostridium bifermentans and Escherichia coli. One example of a control that shows reduced inhibition of C. difficile growth as a combination of Bacteroides thetaiotaomicron, Bacteroides ovatus and Bacteroides vulgatus. Plates were wrapped with parafilm and incubated for 24 hr at 37° C. under anaerobic conditions. After 24 hr the wells containing C. difficile alone were serially diluted and plated to determine titer. The 96-well plate was then frozen at −80 C before quantifying C. difficile by qPCR assay.

[0306]

Construction of an In Vitro Inhibition Assay to Screen for Bacterial Compositions that Produce Diffusible Products Inhibitory to the Growth of Clostridium difficile Using a Filter Insert.

[0307]

The In vitro inhibition assay described above was modified by using a 0.22 uM filter insert (Millipore™ MultiScreen™ 96-Well Assay Plates—Item MAGVS2210) in 96-well format to physically separate C. difficile from the bacterial compositions. The C. difficile was aliquoted into the 96-well plate while the bacterial compositions were aliquoted into media on the filter overlay. The nutrient media as in contact on both sides of the 0.22 uM filter, allowing exchange of nutrients, small molecules and many macromolecules (e.g., bacteriocins, cell-surface proteins, or polysaccharides) by diffusion. In this embodiment, after 24 hr incubation, the filter insert containing the bacterial compositions was removed. The plate containing C. difficile was then transferred to a 96-well plate reader suitable for measuring optical density (OD) at 600 nm. The growth of C. difficile in the presence of different bacterial compositions was compared based on the OD measurement.

[0308]

Construction of an In Vitro Inhibition Assay to Screen for Bacterial Compositions Inhibitory to the Growth of Clostridium difficile Using Clostridium difficile Selective Media for Quantification

[0309]

The In vitro inhibition assay described above can be modified to determine final C. difficile titer by serially diluting and plating to C. difficile selective media (Bloedt et al 2009) such as CCFA (cycloserine cefoxitin fructose agar, Anaerobe Systems), CDSA (Clostridium difficile selective agar, which is cycloserine cefoxitin mannitol agar, Becton Dickinson).

[0310]

Quantification of C. difficile Using Quantitative PCR (qPCR) Standard Curve Preparation

[0311]

The standard curve was generated from a well on each assay plate containing only pathogenic C. difficile grown in SweetB+FosIn media as provided herein and quantified by selective spot plating. Serial dilutions of the culture were performed in sterile phosphate-buffered saline. Genomic DNA was extracted from the standard curve samples along with the other wells.

[0312]

Genomic DNA Extraction

[0313]

Genomic DNA was extracted from 5 μl of each sample using a dilution, freeze/thaw, and heat lysis protocol. 5 μL of thawed samples were added to 45 μL of UltraPure water (Life Technologies, Carlsbad, Calif.) and mixed by pipetting. The plates with diluted samples were frozen at −20° C. until use for qPCR which includes a heated lysis step prior to amplification. Alternatively the genomic DNA could be isolated using the Mo Bio Powersoil®-htp 96 Well Soil DNA Isolation Kit (Mo Bio Laboratories, Carlsbad, Calif.), Mo Bio Powersoil® DNA Isolation Kit (Mo Bio Laboratories, Carlsbad, Calif.), or the QIAamp DNA Stool Mini Kit (QIAGEN, Valencia, Calif.) according to the manufacturer's instructions.

[0314]

qPCR Composition and Conditions

[0315]

The qPCR reaction mixture contained 1× SsoAdvanced Universal Probes Supermix, 900 nM of Wr-tcdB-F primer (AGCAGTTGAATATAGTGGTTTAGTTAGAGTTG, IDT, Coralville, Iowa), 900 nM of Wr-tcdB-R primer (CATGCTTTTTTAGTTTCTGGATTGAA, IDT, Coralville, Iowa), 250 nM of Wr-tcdB-P probe (6FAM-CATCCAGTCTCAATTGTATATGTTTCTCCA-MGB, Life Technologies, Grand Island, N.Y.), and Molecular Biology Grade Water (Mo Bio Laboratories, Carlsbad, Calif.) to 18 μl (Primers adapted from: Wroblewski, D. et al., Rapid Molecular Characterization of Clostridium difficile and Assessment of Populations of C. difficile in Stool Specimens, Journal of Clinical Microbiology 47:2142-2148 (2009)). This reaction mixture was aliquoted to wells of a Hard-shell Low-Profile Thin Wall 96-well Skirted PCR Plate (BioRad, Hercules, Calif.). To this reaction mixture, 2 μl of diluted, frozen, and thawed samples were added and the plate sealed with a Microseal ‘B’ Adhesive Seal (BioRad, Hercules, Calif.). The qPCR was performed on a BioRad C1000™ Thermal Cycler equipped with a CFX96™ Real-Time System (BioRad, Hercules, Calif.). The thermocycling conditions were 95° C. for 15 minutes followed by 45 cycles of 95° C. for 5 seconds, 60° C. for 30 seconds, and fluorescent readings of the FAM channel. Alternatively, the qPCR could be performed with other standard methods known to those skilled in the art.

[0316]

Data Analysis

[0317]

The Cq value for each well on the FAM channel was determined by the CFX Manager™ 3.0 software. The log10(cfu/mL) of C. difficile each experimental sample was calculated by inputting a given sample's Cq value into a linear regression model generated from the standard curve comparing the Cq values of the standard curve wells to the known log10(cfu/mL) of those samples. The log inhibition was calculated for each sample by subtracting the log10(cfu/mL) of C. difficile in the sample from the log10(cfu/mL) of C. difficile in the sample on each assay plate used for the generation of the standard curve that has no additional bacteria added. The mean log inhibition was calculated for all replicates for each composition.

[0318]

A histogram of the range and standard deviation of each composition was plotted. Ranges or standard deviations of the log inhibitions that were distinct from the overall distribution were examined as possible outliers. If the removal of a single log inhibition datum from one of the binary pairs that were identified in the histograms would bring the range or standard deviation in line with those from the majority of the samples, that datum was removed as an outlier, and the mean log inhibition was recalculated.

[0319]

The pooled variance of all samples evaluated in the assay was estimated as the average of the sample variances weighted by the sample's degrees of freedom. The pooled standard error was then calculated as the square root of the pooled variance divided by the square root of the number of samples. Confidence intervals for the null hypothesis were determined by multiplying the pooled standard error to the z score corresponding to a given percentage threshold. Mean log inhibitions outside the confidence interval were considered to be inhibitory if positive or stimulatory if negative with the percent confidence corresponding to the interval used. Samples with mean log inhibition greater than the 99% confidence interval (C.I) of the null hypothesis are reported as ++++, those with a 95%<C.I. <99% as +++, those with a 90%<C.I. <95% as ++, those with a 80%<C.I. <90% as + while samples with mean log inhibition less than than the 99% confidence interval (C.I) of the null hypothesis are reported as −−−−, those with a 95%<C.I. <99% as −−−, those with a 90%<C.I. <95% as −−, those with a 80%<C.I. <90% as −.

[0320]

Many binary pairs inhibit C. difficile Table 8. 622 of 989 combinations show inhibition with a confidence interval >80%; 545 of 989 with a C.I. >90%; 507 of 989 with a C.I. >95%; 430 of 989 with a C.I. of >99%. Non-limiting but exemplary binary pairs include those with mean log reduction greater than 0.366, e.g. Allistipes shahii paired with Blautia producta, Clostridium hathaweyi, or Colinsella aerofaciens, or Clostidium mayombei paired with C. innocuum, C. tertium, Colinsella aerofaciens, or any of the other 424 combinations shown in Table 8. Equally important, the In vitro inhibition assay describes binary pairs that do not effectively inhibit C. difficile. 188 of 989 combinations promote growth with >80% confidence; 52 of 989 show a lack of inhibition with >90% confidence; 22 of 989 show a lack of inhibition with >95% confidence; 3 of 989, including B. producta combined with Coprococcus catus, Alistipes shahii combined with Dorea formicigenerans, and Eubacterium rectale combined with Roseburia intestinalis, show a lack of inhibition with >99% confidence. 249 of 989 combinations are neutral in the assay, meaning they neither promote nor inhibit C. difficile growth to the limit of measurement.

[0321]

Ternary combinations with mean log inhibition greater than 0.312 are reported as ++++99% confidence interval (C.I.) of the null hypothesis), those with mean log inhibition between 0.221 and 0.312 as +++(95%<C.I. <99%), those with mean log inhibition between 0.171 and 0.221 as ++(90%<C.I. <95%), those with mean log inhibition between 0.113 and 0.171 as +(80%<C.I. <90%), those with mean log inhibition between −0.113 and −0.171 as −(80%<C.I. <90%), those with mean log inhibition between −0.171 and −0.221 as −−(90%<C.I. <95%), those with mean log inhibition between −0.221 and −0.312 as −−−(95%<C.I. <99%), and those with mean log inhibition less than −0.312 as −−−−(99%<C.I.).

[0322]

The In vitro inhibition assay shows that many ternary combinations inhibit C. difficile. 39 of 56 combinations show inhibition with a confidence interval >80%; 36 of 56 with a C.I. >90%; 36 of 56 with a C.I. >95%; 29 of 56 with a C.I. of >99%. Non-limiting but exemplary ternary combinations include those with mean log reduction greater than 0.171, e.g. any combination shown in Table 9 with a score of ++++, such as Colinsella aerofaciens, Coprococcus comes, and Blautia producta. Equally important, the In vitro inhibition assay describes ternary combinations that do not effectively inhibit C. difficile. 5 of 56 combinations promote growth with >80% confidence; 2 of 56 promote growth with >90% confidence; 1 of 56, Coprococcus comes, Clostridium symbiosum and Eubacterium rectale, promote growth with >95% confidence. 12 of 56 combinations are neutral in the assay, meaning they neither promote nor inhibit C. difficile growth to the limit of measurement.

[0323]

Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification, including claims, are to be understood as being modified in all instances by the term “about.” Accordingly, unless otherwise indicated to the contrary, the numerical parameters are approximations and may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.

[0324]

Unless otherwise indicated, the term “at least” preceding a series of elements is to be understood to refer to every element in the series.

[0325]

While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.

[0326]

All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes.

TABLES

[0327]

[0000]

List of Operational Taxonomic Units (OTU) with taxonomic
assignments made to Genus, Species, and Phylogenetic Clade.
SEQ IDPublic DBSporePathogen
OTUNumberAccessionCladeFormerStatus
Eubacterium saburreum858AB525414clade_178YN
Eubacterium sp. oral clone IR009866AY349376clade_178YN
Lachnospiraceae bacterium ICM621061HQ616401clade_178YN
Lachnospiraceae bacterium1062HQ616384clade_178YN
MSX33
Lachnospiraceae bacterium oral1063ADDS01000069clade_178YN
taxon 107
Alicyclobacillus acidocaldarius122NR_074721clade_179YN
Clostridium baratii555NR_029229clade_223YN
Clostridium colicanis576FJ957863clade_223YN
Clostridium paraputrificum611AB536771clade_223YN
Clostridium sardiniense621NR_041006clade_223YN
Eubacterium budayi837NR_024682clade_223YN
Eubacterium moniliforme851HF558373clade_223YN
Eubacterium multiforme852NR_024683clade_223YN
Eubacterium nitritogenes853NR_024684clade_223YN
Anoxybacillus flavithermus173NR_074667clade_238YN
Bacillus aerophilus196NR_042339clade_238YN
Bacillus aestuarii197GQ980243clade_238YN
Bacillus amyloliquefaciens199NR_075005clade_238YN
Bacillus anthracis200AAEN01000020clade_238YCategory-A
Bacillus atrophaeus201NR_075016clade_238YOP
Bacillus badius202NR_036893clade_238YOP
Bacillus cereus203ABDJ01000015clade_238YOP
Bacillus circulans204AB271747clade_238YOP
Bacillus firmus207NR_025842clade_238YOP
Bacillus flexus208NR_024691clade_238YOP
Bacillus fordii209NR_025786clade_238YOP
Bacillus halmapalus211NR_026144clade_238YOP
Bacillus herbersteinensis213NR_042286clade_238YOP
Bacillus idriensis215NR_043268clade_238YOP
Bacillus lentus216NR_040792clade_238YOP
Bacillus licheniformis217NC_006270clade_238YOP
Bacillus megaterium218GU252124clade_238YOP
Bacillus nealsonii219NR_044546clade_238YOP
Bacillus niabensis220NR_043334clade_238YOP
Bacillus niacini221NR_024695clade_238YOP
Bacillus pocheonensis222NR_041377clade_238YOP
Bacillus pumilus223NR_074977clade_238YOP
Bacillus safensis224JQ624766clade_238YOP
Bacillus simplex225NR_042136clade_238YOP
Bacillus sonorensis226NR_025130clade_238YOP
Bacillus sp. 10403023227CAET01000089clade_238YOP
MM10403188
Bacillus sp. 2_A_57_CT2230ACWD01000095clade_238YOP
Bacillus sp. 2008724126228GU252108clade_238YOP
Bacillus sp. 2008724139229GU252111clade_238YOP
Bacillus sp. 7_16AIA231FN397518clade_238YOP
Bacillus sp. AP8233JX101689clade_238YOP
Bacillus sp. B27(2008)234EU362173clade_238YOP
Bacillus sp. BT1B_CT2235ACWC01000034clade_238YOP
Bacillus sp. GB1.1236FJ897765clade_238YOP
Bacillus sp. GB9237FJ897766clade_238YOP
Bacillus sp. HU19.1238FJ897769clade_238YOP
Bacillus sp. HU29239FJ897771clade_238YOP
Bacillus sp. HU33.1240FJ897772clade_238YOP
Bacillus sp. JC6241JF824800clade_238YOP
Bacillus sp. oral taxon F79248HM099654clade_238YOP
Bacillus sp. SRC_DSF1243GU797283clade_238YOP
Bacillus sp. SRC_DSF10242GU797292clade_238YOP
Bacillus sp. SRC_DSF2244GU797284clade_238YOP
Bacillus sp. SRC_DSF6245GU797288clade_238YOP
Bacillus sp. tc09249HQ844242clade_238YOP
Bacillus sp. zh168250FJ851424clade_238YOP
Bacillus sphaericus251DQ286318clade_238YOP
Bacillus sporothermodurans252NR_026010clade_238YOP
Bacillus subtilis253EU627588clade_238YOP
Bacillus thermoamylovorans254NR_029151clade_238YOP
Bacillus thuringiensis255NC_008600clade_238YOP
Bacillus weihenstephanensis256NR_074926clade_238YOP
Geobacillus kaustophilus933NR_074989clade_238YN
Geobacillus stearothermophilus936NR_040794clade_238YN
Geobacillus thermodenitrificans938NR_074976clade_238YN
Geobacillus thermoglucosidasius939NR_043022clade_238YN
Lysinibacillus sphaericus1193NR_074883clade_238YN
Clostridiales sp. SS3_4543AY305316clade_246YN
Clostridium beijerinckii557NR_074434clade_252YN
Clostridium botulinum560NC_010723clade_252YCategory-A
Clostridium butyricum561ABDT01000017clade_252YN
Clostridium chauvoei568EU106372clade_252YN
Clostridium favososporum582X76749clade_252YN
Clostridium histolyticum592HF558362clade_252YN
Clostridium isatidis597NR_026347clade_252YN
Clostridium limosum602FR870444clade_252YN
Clostridium sartagoforme622NR_026490clade_252YN
Clostridium septicum624NR_026020clade_252YN
Clostridium sp. 7_2_43FAA626ACDK01000101clade_252YN
Clostridium sporogenes645ABKW02000003clade_252YN
Clostridium tertium653Y18174clade_252YN
Clostridium carnis564NR_044716clade_253YN
Clostridium celatum565X77844clade_253YN
Clostridium disporicum579NR_026491clade_253YN
Clostridium gasigenes585NR_024945clade_253YN
Clostridium quinii616NR_026149clade_253YN
Clostridium hylemonae593AB023973clade_260YN
Clostridium scindens623AF262238clade_260YN
Lachnospiraceae bacterium1054ACTR01000020clade_260YN
5_1_57FAA
Clostridium glycyrrhizinilyticum588AB233029clade_262YN
Clostridium nexile607X73443clade_262YN
Coprococcus comes674ABVR01000038clade_262YN
Lachnospiraceae bacterium1048ACTM01000065clade_262YN
1_1_57FAA
Lachnospiraceae bacterium1049ACTN01000028clade_262YN
1_4_56FAA
Lachnospiraceae bacterium1057ACWQ01000079clade_262YN
8_1_57FAA
Ruminococcus lactaris1663ABOU02000049clade_262YN
Ruminococcus torques1670AAVP02000002clade_262YN
Paenibacillus lautus1397NR_040882clade_270YN
Paenibacillus polymyxa1399NR_037006clade_270YN
Paenibacillus sp. HGF51402AEXS01000095clade_270YN
Paenibacillus sp. HGF71403AFDH01000147clade_270YN
Eubacterium sp. oral clone JI012868AY349379clade_298YN
Alicyclobacillus contaminans124NR_041475clade_301YN
Alicyclobacillus herbarius126NR_024753clade_301YN
Alicyclobacillus pomorum127NR_024801clade_301YN
Blautia coccoides373AB571656clade_309YN
Blautia glucerasea374AB588023clade_309YN
Blautia glucerasei375AB439724clade_309YN
Blautia hansenii376ABYU02000037clade_309YN
Blautia luti378AB691576clade_309YN
Blautia producta379AB600998clade_309YN
Blautia schinkii380NR_026312clade_309YN
Blautia sp. M25381HM626178clade_309YN
Blautia stercoris382HM626177clade_309YN
Blautia wexlerae383EF036467clade_309YN
Bryantella formatexigens439ACCL02000018clade_309YN
Clostridium coccoides573EF025906clade_309YN
Eubacterium cellulosolvens839AY178842clade_309YN
Lachnospiraceae bacterium1056ACTV01000014clade_309YN
6_1_63FAA
Ruminococcus hansenii1662M59114clade_309YN
Ruminococcus obeum1664AY169419clade_309YN
Ruminococcus sp. 5_1_39BFAA1666ACII01000172clade_309YN
Ruminococcus sp. K_11669AB222208clade_309YN
Syntrophococcus sucromutans1911NR_036869clade_309YN
Bacillus alcalophilus198X76436clade_327YN
Bacillus clausii205FN397477clade_327YOP
Bacillus gelatini210NR_025595clade_327YOP
Bacillus halodurans212AY144582clade_327YOP
Bacillus sp. oral taxon F26246HM099642clade_327YOP
Clostridium innocuum595M23732clade_351YN
Clostridium sp. HGF2628AENW01000022clade_351YN
Clostridium perfringens612ABDW01000023clade_353YCategory-B
Sarcina ventriculi1687NR_026146clade_353YN
Clostridium bartlettii556ABEZ02000012clade_354YN
Clostridium bifermentans558X73437clade_354YN
Clostridium ghonii586AB542933clade_354YN
Clostridium glycolicum587FJ384385clade_354YN
Clostridium mayombei605FR733682clade_354YN
Clostridium sordellii625AB448946clade_354YN
Clostridium sp. MT4 E635FJ159523clade_354YN
Eubacterium tenue872M59118clade_354YN
Clostridium argentinense553NR_029232clade_355YN
Clostridium sp. JC122630CAEV01000127clade_355YN
Clostridium sp. NMBHI_1636JN093130clade_355YN
Clostridium subterminale650NR_041795clade_355YN
Clostridium sulfidigenes651NR_044161clade_355YN
Dorea formicigenerans773AAXA02000006clade_360YN
Dorea longicatena774AJ132842clade_360YN
Lachnospiraceae bacterium1050ADLB01000035clade_360YN
2_1_46FAA
Lachnospiraceae bacterium1051ACTO01000052clade_360YN
2_1_58FAA
Lachnospiraceae bacterium1053ADCR01000030clade_360YN
4_1_37FAA
Lachnospiraceae bacterium1058ACTX01000023clade_360YN
9_1_43BFAA
Ruminococcus gnavus1661X94967clade_360YN
Ruminococcus sp. ID81668AY960564clade_360YN
Blautia hydrogenotrophica377ACBZ01000217clade_368YN
Lactonifactor longoviformis1147DQ100449clade_368YN
Robinsoniella peoriensis1633AF445258clade_368YN
Eubacterium infirmum849U13039clade_384YN
Eubacterium sp. WAL 14571864FJ687606clade_384YN
Erysipelotrichaceae bacterium823ACZW01000054clade_385YN
5_2_54FAA
Eubacterium biforme835ABYT01000002clade_385YN
Eubacterium cylindroides842FP929041clade_385YN
Eubacterium dolichum844L34682clade_385YN
Eubacterium sp. 3_1_31861ACTL01000045clade_385YN
Eubacterium tortuosum873NR_044648clade_385YN
Bulleidia extructa441ADFR01000011clade_388YN
Solobacterium moorei1739AECQ01000039clade_388YN
Coprococcus catus673EU266552clade_393YN
Lachnospiraceae bacterium oral1064HM099641clade_393YN
taxon F15
Clostridium cochlearium574NR_044717clade_395YN
Clostridium malenominatum604FR749893clade_395YN
Clostridium tetani654NC_004557clade_395YN
Acetivibrio ethanolgignens6FR749897clade_396YN
Anaerosporobacter mobilis161NR_042953clade_396YN
Bacteroides pectinophilus288ABVQ01000036clade_396YN
Clostridium aminovalericum551NR_029245clade_396YN
Clostridium phytofermentans613NR_074652clade_396YN
Eubacterium hallii848L34621clade_396YN
Eubacterium xylanophilum875L34628clade_396YN
Ruminococcus callidus1658NR_029160clade_406YN
Ruminococcus champanellensis1659FP929052clade_406YN
Ruminococcus sp. 18P131665AJ515913clade_406YN
Ruminococcus sp. 9SE511667FM954974clade_406YN
Anaerostipes caccae162ABAX03000023clade_408YN
Anaerostipes sp. 3_2_56FAA163ACWB01000002clade_408YN
Clostridiales bacterium541ABQR01000074clade_408YN
1_7_47FAA
Clostridiales sp. SM4_1542FP929060clade_408YN
Clostridiales sp. SSC_2544FP929061clade_408YN
Clostridium aerotolerans546X76163clade_408YN
Clostridium aldenense547NR_043680clade_408YN
Clostridium algidixylanolyticum550NR_028726clade_408YN
Clostridium amygdalinum552AY353957clade_408YN
Clostridium asparagiforme554ACCJ01000522clade_408YN
Clostridium bolteae559ABCC02000039clade_408YN
Clostridium celerecrescens566JQ246092clade_408YN
Clostridium citroniae569ADLJ01000059clade_408YN
Clostridium clostridiiformes571M59089clade_408YN
Clostridium clostridioforme572NR_044715clade_408YN
Clostridium hathewayi590AY552788clade_408YN
Clostridium indolis594AF028351clade_408YN
Clostridium lavalense600EF564277clade_408YN
Clostridium saccharolyticum620CP002109clade_408YN
Clostridium sp. M62_1633ACFX02000046clade_408YN
Clostridium sp. SS2_1638ABGC03000041clade_408YN
Clostridium sphenoides643X73449clade_408YN
Clostridium symbiosum652ADLQ01000114clade_408YN
Clostridium xylanolyticum658NR_037068clade_408YN
Eubacterium hadrum847FR749933clade_408YN
Lachnospiraceae bacterium1052ACTP01000124clade_408YN
3_1_57FAA_CT1
Lachnospiraceae bacterium1055ACTS01000081clade_408YN
5_1_63FAA
Lachnospiraceae bacterium A41059DQ789118clade_408YN
Lachnospiraceae bacterium DJF1060EU728771clade_408YN
VP30
Lachnospiraceae genomosp. C11065AY278618clade_408YN
Clostridium difficile578NC_013315clade_409YOP
Eubacterium sp. AS15b862HQ616364clade_428YN
Eubacterium sp. OBRC9863HQ616354clade_428YN
Eubacterium sp. oral clone OH3A871AY947497clade_428YN
Eubacterium yurii876AEES01000073clade_428YN
Clostridium acetobutylicum545NR_074511clade_430YN
Clostridium algidicarnis549NR_041746clade_430YN
Clostridium cadaveris562AB542932clade_430YN
Clostridium carboxidivorans563FR733710clade_430YN
Clostridium estertheticum580NR_042153clade_430YN
Clostridium fallax581NR_044714clade_430YN
Clostridium felsineum583AF270502clade_430YN
Clostridium frigidicarnis584NR_024919clade_430YN
Clostridium kluyveri598NR_074165clade_430YN
Clostridium magnum603X77835clade_430YN
Clostridium putrefaciens615NR_024995clade_430YN
Clostridium sp. HPB_46629AY862516clade_430YN
Clostridium tyrobutyricum656NR_044718clade_430YN
Sutterella parvirubra1899AB300989clade_432YN
Acetanaerobacterium elongatum4NR_042930clade_439YN
Clostridium cellulosi567NR_044624clade_439YN
Ethanoligenens harbinense832AY675965clade_439YN
Eubacterium rectale856FP929042clade_444YN
Eubacterium sp. oral clone GI038865AY349374clade_444YN
Lachnobacterium bovis1045GU324407clade_444YN
Roseburia cecicola1634GU233441clade_444YN
Roseburia faecalis1635AY804149clade_444YN
Roseburia faecis1636AY305310clade_444YN
Roseburia hominis1637AJ270482clade_444YN
Roseburia intestinalis1638FP929050clade_444YN
Roseburia inulinivorans1639AJ270473clade_444YN
Brevibacillus brevis410NR_041524clade_448YN
Brevibacillus laterosporus414NR_037005clade_448YN
Bacillus coagulans206DQ297928clade_451YOP
Sporolactobacillus inulinus1752NR_040962clade_451YN
Kocuria palustris1041EU333884clade_453YN
Nocardia farcinica1353NC_006361clade_455YN
Bacillus sp. oral taxon F28247HM099650clade_456YOP
Catenibacterium mitsuokai495AB030224clade_469YN
Clostridium sp. TM_40640AB249652clade_469YN
Coprobacillus cateniformis670AB030218clade_469YN
Coprobacillus sp. 29_1671ADKX01000057clade_469YN
Clostridium rectum618NR_029271clade_470YN
Eubacterium nodatum854U13041clade_476YN
Eubacterium saphenum859NR_026031clade_476YN
Eubacterium sp. oral clone JH012867AY349373clade_476YN
Eubacterium sp. oral clone JS001870AY349378clade_476YN
Faecalibacterium prausnitzii880ACOP02000011clade_478YN
Gemmiger formicilis932GU562446clade_478YN
Subdoligranulum variabile1896AJ518869clade_478YN
Clostridiaceae bacterium JC13532JF824807clade_479YN
Clostridium sp. MLG055634AF304435clade_479YN
Erysipelotrichaceae bacterium822ACTJ01000113clade_479YN
3_1_53
Clostridium cocleatum575NR_026495clade_481YN
Clostridium ramosum617M23731clade_481YN
Clostridium saccharogumia619DQ100445clade_481YN
Clostridium spiroforme644X73441clade_481YN
Coprobacillus sp. D7672ACDT01000199clade_481YN
Clostridiales bacterium SY8519535AB477431clade_482YN
Clostridium sp. SY8519639AP012212clade_482YN
Eubacterium ramulus855AJ011522clade_482YN
Erysipelothrix inopinata819NR_025594clade_485YN
Erysipelothrix rhusiopathiae820ACLK01000021clade_485YN
Erysipelothrix tonsillarum821NR_040871clade_485YN
Holdemania filiformis1004Y11466clade_485YN
Mollicutes bacterium pACH931258AY297808clade_485YN
Coxiella burnetii736CP000890clade_486YCategory-B
Clostridium hiranonis591AB023970clade_487YN
Clostridium irregulare596NR_029249clade_487YN
Clostridium orbiscindens609Y18187clade_494YN
Clostridium sp. NML 04A032637EU815224clade_494YN
Flavonifractor plautii886AY724678clade_494YN
Pseudoflavonifractor capillosus1591AY136666clade_494YN
Ruminococcaceae bacterium D161655ADDX01000083clade_494YN
Acetivibrio cellulolyticus5NR_025917clade_495YN
Clostridium aldrichii548NR_026099clade_495YN
Clostridium clariflavum570NR_041235clade_495YN
Clostridium stercorarium647NR_025100clade_495YN
Clostridium straminisolvens649NR_024829clade_495YN
Clostridium thermocellum655NR_074629clade_495YN
Fusobacterium nucleatum901ADVK01000034clade_497YN
Eubacterium barkeri834NR_044661clade_512YN
Eubacterium callanderi838NR_026330clade_512YN
Eubacterium limosum850CP002273clade_512YN
Anaerotruncus colihominis164ABGD02000021clade_516YN
Clostridium methylpentosum606ACEC01000059clade_516YN
Clostridium sp. YIT 12070642AB491208clade_516YN
Hydrogenoanaerobacterium1005NR_044425clade_516YN
saccharovorans
Ruminococcus albus1656AY445600clade_516YN
Ruminococcus flavefaciens1660NR_025931clade_516YN
Clostridium haemolyticum589NR_024749clade_517YN
Clostridium novyi608NR_074343clade_517YN
Clostridium sp. LMG 16094632X95274clade_517YN
Eubacterium ventriosum874L34421clade_519YN
Bacteroides galacturonicus280DQ497994clade_522YN
Eubacterium eligens845CP001104clade_522YN
Lachnospira multipara1046FR733699clade_522YN
Lachnospira pectinoschiza1047L14675clade_522YN
Lactobacillus rogosae1114GU269544clade_522YN
Bacillus horti214NR_036860clade_527YOP
Bacillus sp. 9_3AIA232FN397519clade_527YOP
Eubacterium brachy836U13038clade_533YN
Filifactor alocis881CP002390clade_533YN
Filifactor villosus882NR_041928clade_533YN
Clostridium leptum601AJ305238clade_537YN
Clostridium sp. YIT 12069641AB491207clade_537YN
Clostridium sporosphaeroides646NR_044835clade_537YN
Eubacterium coprostanoligenes841HM037995clade_537YN
Ruminococcus bromii1657EU266549clade_537YN
Eubacterium siraeum860ABCA03000054clade_538YN
Clostridium viride657NR_026204clade_540YN
Oscillibacter sp. G21386HM626173clade_540YN
Oscillibacter valericigenes1387NR_074793clade_540YN
Oscillospira guilliermondii1388AB040495clade_540YN
Butyrivibrio crossotus455ABWN01000012clade_543YN
Clostridium sp. L2_50631AAYW02000018clade_543YN
Coprococcus eutactus675EF031543clade_543YN
Coprococcus sp. ART55_1676AY350746clade_543YN
Eubacterium ruminantium857NR_024661clade_543YN
Collinsella aerofaciens659AAVN02000007clade_553YN
Alkaliphilus metalliredigenes137AY137848clade_554YN
Alkaliphilus oremlandii138NR_043674clade_554YN
Clostridium sticklandii648L04167clade_554YN
Turicibacter sanguinis1965AF349724clade_555YN
Fulvimonas sp. NML 060897892EF589680clade_557YN
Desulfitobacterium frappieri753AJ276701clade_560YN
Desulfitobacterium hafniense754NR_074996clade_560YN
Desulfotomaculum nigrificans756NR_044832clade_560YN
Lutispora thermophila1191NR_041236clade_564YN
Brachyspira pilosicoli405NR_075069clade_565YN
Eggerthella lenta778AF292375clade_566YN
Streptomyces albus1888AJ697941clade_566YN
Chlamydiales bacterium NS11505JN606074clade_567YN
Anaerofustis stercorihominis159ABIL02000005clade_570YN
Butyricicoccus pullicaecorum453HH793440clade_572YN
Eubacterium desmolans843NR_044644clade_572YN
Papillibacter cinnamivorans1415NR_025025clade_572YN
Sporobacter termitidis1751NR_044972clade_572YN
Deferribacteres sp. oral clone744AY349371clade_575YN
JV006
Clostridium colinum577NR_026151clade_576YN
Clostridium lactatifermentans599NR_025651clade_576YN
Clostridium piliforme614D14639clade_576YN
Saccharomonospora viridis1671X54286clade_579YN
Thermobifida fusca1921NC_007333clade_579YN
Leptospira licerasiae1164EF612284clade_585YOP
Moorella thermoacetica1259NR_075001clade_590YN
Thermoanaerobacter1920CP000924clade_590YN
pseudethanolicus
Flexistipes sinusarabici888NR_074881clade_591YN
Gloeobacter violaceus942NR_074282clade_596YN
Eubacterium sp. oral clone JN088869AY349377clade_90YN
Clostridium oroticum610FR749922clade_96YN
Clostridium sp. D5627ADBG01000142clade_96YN
Eubacterium contortum840FR749946clade_96YN
Eubacterium fissicatena846FR749935clade_96YN
Corynebacterium coyleae692X96497clade_100NN
Corynebacterium mucifaciens711NR_026396clade_100NN
Corynebacterium ureicelerivorans733AM397636clade_100NN
Corynebacterium appendicis684NR_028951clade_102NN
Corynebacterium genitalium698ACLJ01000031clade_102NN
Corynebacterium glaucum699NR_028971clade_102NN
Corynebacterium imitans703AF537597clade_102NN
Corynebacterium riegelii719EU848548clade_102NN
Corynebacterium sp. L_2012475723HE575405clade_102NN
Corynebacterium sp. NML724GU238409clade_102NN
93_0481
Corynebacterium sundsvallense728Y09655clade_102NN
Corynebacterium tuscaniae730AY677186clade_102NN
Prevotella maculosa1504AGEK01000035clade_104NN
Prevotella oris1513ADDV01000091clade_104NN
Prevotella salivae1517AB108826clade_104NN
Prevotella sp. ICM551521HQ616399clade_104NN
Prevotella sp. oral clone AA0201528AY005057clade_104NN
Prevotella sp. oral clone GI0321538AY349396clade_104NN
Prevotella sp. oral taxon G701558GU432179clade_104NN
Prevotella corporis1491L16465clade_105NN
Bacteroides sp. 4_1_36312ACTC01000133clade_110NN
Bacteroides sp. AR20315AF139524clade_110NN
Bacteroides sp. D20319ACPT01000052clade_110NN
Bacteroides sp. F_4322AB470322clade_110NN
Bacteroides uniformis329AB050110clade_110NN
Prevotella nanceiensis1510JN867228clade_127NN
Prevotella sp. oral taxon 2991548ACWZ01000026clade_127NN
Prevotella bergensis1485ACKS01000100clade_128NN
Prevotella buccalis1489JN867261clade_129NN
Prevotella timonensis1564ADEF01000012clade_129NN
Prevotella oralis1512AEPE01000021clade_130NN
Prevotella sp. SEQ0721525JN867238clade_130NN
Leuconostoc carnosum1177NR_040811clade_135NN
Leuconostoc gasicomitatum1179FN822744clade_135NN
Leuconostoc inhae1180NR_025204clade_135NN
Leuconostoc kimchii1181NR_075014clade_135NN
Edwardsiella tarda777CP002154clade_139NN
Photorhabdus asymbiotica1466Z76752clade_139NN
Psychrobacter arcticus1607CP000082clade_141NN
Psychrobacter cibarius1608HQ698586clade_141NN
Psychrobacter cryohalolentis1609CP000323clade_141NN
Psychrobacter faecalis1610HQ698566clade_141NN
Psychrobacter nivimaris1611HQ698587clade_141NN
Psychrobacter pulmonis1612HQ698582clade_141NN
Pseudomonas aeruginosa1592AABQ07000001clade_154NN
Pseudomonas sp. 2_1_261600ACWU01000257clade_154NN
Corynebacterium confusum691Y15886clade_158NN
Corynebacterium propinquum712NR_037038clade_158NN
Corynebacterium713X84258clade_158NN
pseudodiphtheriticum
Bartonella bacilliformis338NC_008783clade_159NN
Bartonella grahamii339CP001562clade_159NN
Bartonella henselae340NC_005956clade_159NN
Bartonella quintana341BX897700clade_159NN
Bartonella tamiae342EF672728clade_159NN
Bartonella washoensis343FJ719017clade_159NN
Brucella abortus430ACBJ01000075clade_159NCategory-B
Brucella canis431NR_044652clade_159NCategory-B
Brucella ceti432ACJD01000006clade_159NCategory-B
Brucella melitensis433AE009462clade_159NCategory-B
Brucella microti434NR_042549clade_159NCategory-B
Brucella ovis435NC_009504clade_159NCategory-B
Brucella sp. 83_13436ACBQ01000040clade_159NCategory-B
Brucella sp. BO1437EU053207clade_159NCategory-B
Brucella suis438ACBK01000034clade_159NCategory-B
Ochrobactrum anthropi1360NC_009667clade_159NN
Ochrobactrum intermedium1361ACQA01000001clade_159NN
Ochrobactrum pseudintermedium1362DQ365921clade_159NN
Prevotella genomosp. C21496AY278625clade_164NN
Prevotella multisaccharivorax1509AFJE01000016clade_164NN
Prevotella sp. oral clone1543AY550997clade_164NN
IDR_CEC_0055
Prevotella sp. oral taxon 2921547GQ422735clade_164NN
Prevotella sp. oral taxon 3001549GU409549clade_164NN
Prevotella marshii1505AEEI01000070clade_166NN
Prevotella sp. oral clone IK0531544AY349401clade_166NN
Prevotella sp. oral taxon 7811554GQ422744clade_166NN
Prevotella stercorea1562AB244774clade_166NN
Prevotella brevis1487NR_041954clade_167NN
Prevotella ruminicola1516CP002006clade_167NN
Prevotella sp. sp241560AB003384clade_167NN
Prevotella sp. sp341561AB003385clade_167NN
Prevotella albensis1483NR_025300clade_168NN
Prevotella copri1490ACBX02000014clade_168NN
Prevotella oulorum1514L16472clade_168NN
Prevotella sp. BI_421518AJ581354clade_168NN
Prevotella sp. oral clone P4PB_831546AY207050clade_168NN
P2
Prevotella sp. oral taxon G601557GU432133clade_168NN
Prevotella amnii1484AB547670clade_169NN
Bacteroides caccae268EU136686clade_170NN
Bacteroides finegoldii277AB222699clade_170NN
Bacteroides intestinalis283ABJL02000006clade_171NN
Bacteroides sp. XB44A326AM230649clade_171NN
Bifidobacteriaceae genomosp. C1345AY278612clade_172NN
Bifidobacterium adolescentis346AAXD02000018clade_172NN
Bifidobacterium angulatum347ABYS02000004clade_172NN
Bifidobacterium animalis348CP001606clade_172NN
Bifidobacterium breve350CP002743clade_172NN
Bifidobacterium catenulatum351ABXY01000019clade_172NN
Bifidobacterium dentium352CP001750clade_172NOP
Bifidobacterium gallicum353ABXB03000004clade_172NN
Bifidobacterium infantis354AY151398clade_172NN
Bifidobacterium kashiwanohense355AB491757clade_172NN
Bifidobacterium longum356ABQQ01000041clade_172NN
Bifidobacterium357ABXX02000002clade_172NN
pseudocatenulatum
Bifidobacterium pseudolongum358NR_043442clade_172NN
Bifidobacterium scardovii359AJ307005clade_172NN
Bifidobacterium sp. HM2360AB425276clade_172NN
Bifidobacterium sp. HMLN12361JF519685clade_172NN
Bifidobacterium sp. M45362HM626176clade_172NN
Bifidobacterium sp. MSX5B363HQ616382clade_172NN
Bifidobacterium sp. TM_7364AB218972clade_172NN
Bifidobacterium thermophilum365DQ340557clade_172NN
Leuconostoc citreum1178AM157444clade_175NN
Leuconostoc lactis1182NR_040823clade_175NN
Alicyclobacillus acidoterrestris123NR_040844clade_179NN
Alicyclobacillus cycloheptanicus125NR_024754clade_179NN
Acinetobacter baumannii27ACYQ01000014clade_181NN
Acinetobacter calcoaceticus28AM157426clade_181NN
Acinetobacter genomosp. C129AY278636clade_181NN
Acinetobacter haemolyticus30ADMT01000017clade_181NN
Acinetobacter johnsonii31ACPL01000162clade_181NN
Acinetobacter junii32ACPM01000135clade_181NN
Acinetobacter lwoffii33ACPN01000204clade_181NN
Acinetobacter parvus34AIEB01000124clade_181NN
Acinetobacter schindleri36NR_025412clade_181NN
Acinetobacter sp. 56A137GQ178049clade_181NN
Acinetobacter sp. CIP 10193438JQ638573clade_181NN
Acinetobacter sp. CIP 10214339JQ638578clade_181NN
Acinetobacter sp. M16_2241HM366447clade_181NN
Acinetobacter sp. RUH262442ACQF01000094clade_181NN
Acinetobacter sp. SH02443ADCH01000068clade_181NN
Lactobacillus jensenii1092ACQD01000066clade_182NN
Alcaligenes faecalis119AB680368clade_183NN
Alcaligenes sp. CO14120DQ643040clade_183NN
Alcaligenes sp. S3121HQ262549clade_183NN
Oligella ureolytica1366NR_041998clade_183NN
Oligella urethralis1367NR_041753clade_183NN
Eikenella corrodens784ACEA01000028clade_185NN
Kingella denitrificans1019AEWV01000047clade_185NN
Kingella genomosp. P1 oral cone1020DQ003616clade_185NN
MB2_C20
Kingella kingae1021AFHS01000073clade_185NN
Kingella oralis1022ACJW02000005clade_185NN
Kingella sp. oral clone ID0591023AY349381clade_185NN
Neisseria elongata1330ADBF01000003clade_185NN
Neisseria genomosp. P2 oral clone1332DQ003630clade_185NN
MB5_P15
Neisseria sp. oral clone JC0121345AY349388clade_185NN
Neisseria sp. SMC_A91991342FJ763637clade_185NN
Simonsiella muelleri1731ADCY01000105clade_185NN
Corynebacterium glucuronolyticum700ABYP01000081clade_193NN
Corynebacterium716FJ185225clade_193NN
pyruviciproducens
Rothia aeria1649DQ673320clade_194NN
Rothia dentocariosa1650ADDW01000024clade_194NN
Rothia sp. oral taxon 1881653GU470892clade_194NN
Corynebacterium accolens681ACGD01000048clade_195NN
Corynebacterium macginleyi707AB359393clade_195NN
Corynebacterium714ABYQ01000237clade_195NN
pseudogenitalium
Corynebacterium729ACVP01000009clade_195NN
tuberculostearicum
Lactobacillus casei1074CP000423clade_198NN
Lactobacillus paracasei1106ABQV01000067clade_198NN
Lactobacillus zeae1143NR_037122clade_198NN
Prevotella dentalis1492AB547678clade_205NN
Prevotella sp. oral clone ASCG101529AY923148clade_206NN
Prevotella sp. oral clone HF0501541AY349399clade_206NN
Prevotella sp. oral clone ID0191542AY349400clade_206NN
Prevotella sp. oral clone IK0621545AY349402clade_206NN
Prevotella genomosp. P9 oral1499DQ003633clade_207NN
clone MB7_G16
Prevotella sp. oral clone AU0691531AY005062clade_207NN
Prevotella sp. oral clone CY0061532AY005063clade_207NN
Prevotella sp. oral clone FL0191534AY349392clade_207NN
Actinomyces genomosp. C156AY278610clade_212NN
Actinomyces genomosp. C257AY278611clade_212NN
Actinomyces genomosp. P1 oral58DQ003632clade_212NN
clone MB6_C03
Actinomyces georgiae59GU561319clade_212NN
Actinomyces israelii60AF479270clade_212NN
Actinomyces massiliensis61AB545934clade_212NN
Actinomyces meyeri62GU561321clade_212NN
Actinomyces odontolyticus66ACYT01000123clade_212NN
Actinomyces orihominis68AJ575186clade_212NN
Actinomyces sp. CCUG 3729071AJ234058clade_212NN
Actinomyces sp. ICM3475HQ616391clade_212NN
Actinomyces sp. ICM4176HQ616392clade_212NN
Actinomyces sp. ICM4777HQ616395clade_212NN
Actinomyces sp. ICM5478HQ616398clade_212NN
Actinomyces sp. oral clone IP08187AY349366clade_212NN
Actinomyces sp. oral taxon 17891AEUH01000060clade_212NN
Actinomyces sp. oral taxon 18092AEPP01000041clade_212NN
Actinomyces sp. TeJ580GU561315clade_212NN
Haematobacter sp. BC14248968GU396991clade_213NN
Paracoccus denitrificans1424CP000490clade_213NN
Paracoccus marcusii1425NR_044922clade_213NN
Grimontia hollisae967ADAQ01000013clade_216NN
Shewanella putrefaciens1723CP002457clade_216NN
Afipia genomosp. 4111EU117385clade_217NN
Rhodopseudomonas palustris1626CP000301clade_217NN
Methylobacterium extorquens1223NC_010172clade_218NN
Methylobacterium podarium1224AY468363clade_218NN
Methylobacterium radiotolerans1225GU294320clade_218NN
Methylobacterium sp. 1sub1226AY468371clade_218NN
Methylobacterium sp. MM41227AY468370clade_218NN
Achromobacter denitrificans18NR_042021clade_224NN
Achromobacter piechaudii19ADMS01000149clade_224NN
Achromobacter xylosoxidans20ACRC01000072clade_224NN
Bordetella bronchiseptica384NR_025949clade_224NOP
Bordetella holmesii385AB683187clade_224NOP
Bordetella parapertussis386NR_025950clade_224NOP
Bordetella pertussis387BX640418clade_224NOP
Microbacterium chocolatum1230NR_037045clade_225NN
Microbacterium flavescens1231EU714363clade_225NN
Microbacterium lacticum1233EU714351clade_225NN
Microbacterium oleivorans1234EU714381clade_225NN
Microbacterium oxydans1235EU714348clade_225NN
Microbacterium paraoxydans1236AJ491806clade_225NN
Microbacterium phyllosphaerae1237EU714359clade_225NN
Microbacterium schleiferi1238NR_044936clade_225NN
Microbacterium sp. 7681239EU714378clade_225NN
Microbacterium sp. oral strain1240AF287752clade_225NN
C24KA
Microbacterium testaceum1241EU714365clade_225NN
Corynebacterium atypicum686NR_025540clade_229NN
Corynebacterium mastitidis708AB359395clade_229NN
Corynebacterium sp. NML725GU238411clade_229NN
97_0186
Mycobacterium elephantis1275AF385898clade_237NOP
Mycobacterium paraterrae1288EU919229clade_237NOP
Mycobacterium phlei1289GU142920clade_237NOP
Mycobacterium sp. 17761293EU703152clade_237NN
Mycobacterium sp. 17811294EU703147clade_237NN
Mycobacterium sp. AQ1GA41297HM210417clade_237NN
Mycobacterium sp. GN_105461299FJ497243clade_237NN
Mycobacterium sp. GN_108271300FJ497247clade_237NN
Mycobacterium sp. GN_111241301FJ652846clade_237NN
Mycobacterium sp. GN_91881302FJ497240clade_237NN
Mycobacterium sp. GR_2007_2101303FJ555538clade_237NN
Anoxybacillus contaminans172NR_029006clade_238NN
Bacillus aeolius195NR_025557clade_238NN
Brevibacterium frigoritolerans422NR_042639clade_238NN
Geobacillus sp. E263934DQ647387clade_238NN
Geobacillus sp. WCH70935CP001638clade_238NN
Geobacillus thermocatenulatus937NR_043020clade_238NN
Geobacillus thermoleovorans940NR_074931clade_238NN
Lysinibacillus fusiformis1192FN397522clade_238NN
Planomicrobium koreense1468NR_025011clade_238NN
Sporosarcina newyorkensis1754AFPZ01000142clade_238NN
Sporosarcina sp. 26811755GU994081clade_238NN
Ureibacillus composti1968NR_043746clade_238NN
Ureibacillus suwonensis1969NR_043232clade_238NN
Ureibacillus terrenus1970NR_025394clade_238NN
Ureibacillus thermophilus1971NR_043747clade_238NN
Ureibacillus thermosphaericus1972NR_040961clade_238NN
Prevotella micans1507AGWK01000061clade_239NN
Prevotella sp. oral clone DA0581533AY005065clade_239NN
Prevotella sp. SEQ0531523JN867222clade_239NN
Treponema socranskii1937NR_024868clade_240NOP
Treponema sp. 6:H:D15A_41938AY005083clade_240NN
Treponema sp. oral taxon 2651953GU408850clade_240NN
Treponema sp. oral taxon G851958GU432215clade_240NN
Porphyromonas endodontalis1472ACNN01000021clade_241NN
Porphyromonas sp. oral clone1478AY005068clade_241NN
BB134
Porphyromonas sp. oral clone1479AY005069clade_241NN
F016
Porphyromonas sp. oral clone1480AY207054clade_241NN
P2PB_52_P1
Porphyromonas sp. oral clone1481AY207057clade_241NN
P4GB_100 P2
Acidovorax sp. 98_6383326AY258065clade_245NN
Comamonadaceae bacterium663JN585335clade_245NN
NML000135
Comamonadaceae bacterium664JN585331clade_245NN
NML790751
Comamonadaceae bacterium665JN585332clade_245NN
NML910035
Comamonadaceae bacterium666JN585333clade_245NN
NML910036
Comamonas sp. NSP5668AB076850clade_245NN
Delftia acidovorans748CP000884clade_245NN
Xenophilus aerolatus2018JN585329clade_245NN
Oribacterium sp. oral taxon 0781380ACIQ02000009clade_246NN
Oribacterium sp. oral taxon 1021381GQ422713clade_246NN
Weissella cibaria2007NR_036924clade_247NN
Weissella confusa2008NR_040816clade_247NN
Weissella hellenica2009AB680902clade_247NN
Weissella kandleri2010NR_044659clade_247NN
Weissella koreensis2011NR_075058clade_247NN
Weissella paramesenteroides2012ACKU01000017clade_247NN
Weissella sp. KLDS 7.07012013EU600924clade_247NN
Mobiluncus curtisii1251AEPZ01000013clade_249NN
Enhydrobacter aerosaccus785ACYI01000081clade_256NN
Moraxella osloensis1262JN175341clade_256NN
Moraxella sp. GM21264JF837191clade_256NN
Brevibacterium casei420JF951998clade_257NN
Brevibacterium epidermidis421NR_029262clade_257NN
Brevibacterium sanguinis426NR_028016clade_257NN
Brevibacterium sp. H15427AB177640clade_257NN
Acinetobacter radioresistens35ACVR01000010clade_261NN
Lactobacillus alimentarius1068NR_044701clade_263NN
Lactobacillus farciminis1082NR_044707clade_263NN
Lactobacillus kimchii1097NR_025045clade_263NN
Lactobacillus nodensis1101NR_041629clade_263NN
Lactobacillus tucceti1138NR_042194clade_263NN
Pseudomonas mendocina1595AAUL01000021clade_265NN
Pseudomonas pseudoalcaligenes1598NR_037000clade_265NN
Pseudomonas sp. NP522b1602EU723211clade_265NN
Pseudomonas stutzeri1603AM905854clade_265NN
Paenibacillus barcinonensis1390NR_042272clade_270NN
Paenibacillus barengoltzii1391NR_042756clade_270NN
Paenibacillus chibensis1392NR_040885clade_270NN
Paenibacillus cookii1393NR_025372clade_270NN
Paenibacillus durus1394NR_037017clade_270NN
Paenibacillus glucanolyticus1395D78470clade_270NN
Paenibacillus lactis1396NR_025739clade_270NN
Paenibacillus pabuli1398NR_040853clade_270NN
Paenibacillus popilliae1400NR_040888clade_270NN
Paenibacillus sp. CIP 1010621401HM212646clade_270NN
Paenibacillus sp. JC661404JF824808clade_270NN
Paenibacillus sp. R_274131405HE586333clade_270NN
Paenibacillus sp. R_274221406HE586338clade_270NN
Paenibacillus timonensis1408NR_042844clade_270NN
Rothia mucilaginosa1651ACVO01000020clade_271NN
Rothia nasimurium1652NR_025310clade_271NN
Prevotella sp. oral taxon 3021550ACZK01000043clade_280NN
Prevotella sp. oral taxon F681556HM099652clade_280NN
Prevotella tannerae1563ACIJ02000018clade_280NN
Prevotellaceae bacterium P4P_621566AY207061clade_280NN
P1
Porphyromonas asaccharolytica1471AENO01000048clade_281NN
Porphyromonas gingivalis1473AE015924clade_281NN
Porphyromonas macacae1475NR_025908clade_281NN
Porphyromonas sp. UQD 3011477EU012301clade_281NN
Porphyromonas uenonis1482ACLR01000152clade_281NN
Leptotrichia buccalis1165CP001685clade_282NN
Leptotrichia hofstadii1168ACVB02000032clade_282NN
Leptotrichia sp. oral clone HE0121173AY349386clade_282NN
Leptotrichia sp. oral taxon 2231176GU408547clade_282NN
Bacteroides fluxus278AFBN01000029clade_285NN
Bacteroides helcogenes281CP002352clade_285NN
Parabacteroides johnsonii1419ABYH01000014clade_286NN
Parabacteroides merdae1420EU136685clade_286NN
Treponema denticola1926ADEC01000002clade_288NOP
Treponema genomosp. P5 oral1929DQ003624clade_288NN
clone MB3_P23
Treponema putidum1935AJ543428clade_288NOP
Treponema sp. oral clone1942AY207055clade_288NN
P2PB_53 P3
Treponema sp. oral taxon 2471949GU408748clade_288NN
Treponema sp. oral taxon 2501950GU408776clade_288NN
Treponema sp. oral taxon 2511951GU408781clade_288NN
Anaerococcus hydrogenalis144ABXA01000039clade_289NN
Anaerococcus sp. 8404299148HM587318clade_289NN
Anaerococcus sp. gpac215156AM176540clade_289NN
Anaerococcus vaginalis158ACXU01000016clade_289NN
Propionibacterium acidipropionici1569NC_019395clade_290NN
Propionibacterium avidum1571AJ003055clade_290NN
Propionibacterium granulosum1573FJ785716clade_290NN
Propionibacterium jensenii1574NR_042269clade_290NN
Propionibacterium propionicum1575NR_025277clade_290NN
Propionibacterium sp. H4561577AB177643clade_290NN
Propionibacterium thoenii1581NR_042270clade_290NN
Bifidobacterium bifidum349ABQP01000027clade_293NN
Leuconostoc mesenteroides1183ACKV01000113clade_295NN
Leuconostoc1184NR_040814clade_295NN
pseudomesenteroides
Johnsonella ignava1016X87152clade_298NN
Propionibacterium acnes1570ADJM01000010clade_299NN
Propionibacterium sp. 434_HC21576AFIL01000035clade_299NN
Propionibacterium sp. LG1578AY354921clade_299NN
Propionibacterium sp. S555a1579AB264622clade_299NN
Alicyclobacillus sp. CCUG 53762128HE613268clade_301NN
Actinomyces cardiffensis53GU470888clade_303NN
Actinomyces funkei55HQ906497clade_303NN
Actinomyces sp. HKU3174HQ335393clade_303NN
Actinomyces sp. oral taxon C5594HM099646clade_303NN
Kerstersia gyiorum1018NR_025669clade_307NN
Pigmentiphaga daeguensis1467JN585327clade_307NN
Aeromonas allosaccharophila104S39232clade_308NN
Aeromonas enteropelogenes105X71121clade_308NN
Aeromonas hydrophila106NC_008570clade_308NN
Aeromonas jandaei107X60413clade_308NN
Aeromonas salmonicida108NC_009348clade_308NN
Aeromonas trota109X60415clade_308NN
Aeromonas veronii110NR_044845clade_308NN
Marvinbryantia formatexigens1196AJ505973clade_309NN
Rhodobacter sp. oral taxon C301620HM099648clade_310NN
Rhodobacter sphaeroides1621CP000144clade_310NN
Lactobacillus antri1071ACLL01000037clade_313NN
Lactobacillus coleohominis1076ACOH01000030clade_313NN
Lactobacillus fermentum1083CP002033clade_313NN
Lactobacillus gastricus1085AICN01000060clade_313NN
Lactobacillus mucosae1099FR693800clade_313NN
Lactobacillus oris1103AEKL01000077clade_313NN
Lactobacillus pontis1111HM218420clade_313NN
Lactobacillus reuteri1112ACGW02000012clade_313NN
Lactobacillus sp. KLDS 1.07071127EU600911clade_313NN
Lactobacillus sp. KLDS 1.07091128EU600913clade_313NN
Lactobacillus sp. KLDS 1.07111129EU600915clade_313NN
Lactobacillus sp. KLDS 1.07131131EU600917clade_313NN
Lactobacillus sp. KLDS 1.07161132EU600921clade_313NN
Lactobacillus sp. KLDS 1.07181133EU600922clade_313NN
Lactobacillus sp. oral taxon 0521137GQ422710clade_313NN
Lactobacillus vaginalis1140ACGV01000168clade_313NN
Brevibacterium aurantiacum419NR_044854clade_314NN
Brevibacterium linens423AJ315491clade_314NN
Lactobacillus pentosus1108JN813103clade_315NN
Lactobacillus plantarum1110ACGZ02000033clade_315NN
Lactobacillus sp. KLDS 1.07021123EU600906clade_315NN
Lactobacillus sp. KLDS 1.07031124EU600907clade_315NN
Lactobacillus sp. KLDS 1.07041125EU600908clade_315NN
Lactobacillus sp. KLDS 1.07051126EU600909clade_315NN
Agrobacterium radiobacter115CP000628clade_316NN
Agrobacterium tumefaciens116AJ389893clade_316NN
Corynebacterium argentoratense685EF463055clade_317NN
Corynebacterium diphtheriae693NC_002935clade_317NOP
Corynebacterium715NR_037070clade_317NN
pseudotuberculosis
Corynebacterium renale717NR_037069clade_317NN
Corynebacterium ulcerans731NR_074467clade_317NN
Aurantimonas coralicida191AY065627clade_318NN
Aureimonas altamirensis192FN658986clade_318NN
Lactobacillus acidipiscis1066NR_024718clade_320NN
Lactobacillus salivarius1117AEBA01000145clade_320NN
Lactobacillus sp. KLDS 1.07191134EU600923clade_320NN
Lactobacillus buchneri1073ACGH01000101clade_321NN
Lactobacillus genomosp. C11086AY278619clade_321NN
Lactobacillus genomosp. C21087AY278620clade_321NN
Lactobacillus hilgardii1089ACGP01000200clade_321NN
Lactobacillus kefiri1096NR_042230clade_321NN
Lactobacillus parabuchneri1105NR_041294clade_321NN
Lactobacillus parakefiri1107NR_029039clade_321NN
Lactobacillus curvatus1079NR_042437clade_322NN
Lactobacillus sakei1116DQ989236clade_322NN
Aneurinibacillus aneurinilyticus167AB101592clade_323NN
Aneurinibacillus danicus168NR_028657clade_323NN
Aneurinibacillus migulanus169NR_036799clade_323NN
Aneurinibacillus terranovensis170NR_042271clade_323NN
Staphylococcus aureus1757CP002643clade_325NCategory-B
Staphylococcus auricularis1758JQ624774clade_325NN
Staphylococcus capitis1759ACFR01000029clade_325NN
Staphylococcus caprae1760ACRH01000033clade_325NN
Staphylococcus carnosus1761NR_075003clade_325NN
Staphylococcus cohnii1762JN175375clade_325NN
Staphylococcus condimenti1763NR_029345clade_325NN
Staphylococcus epidermidis1764ACHE01000056clade_325NN
Staphylococcus equorum1765NR_027520clade_325NN
Staphylococcus haemolyticus1767NC_007168clade_325NN
Staphylococcus hominis1768AM157418clade_325NN
Staphylococcus lugdunensis1769AEQA01000024clade_325NN
Staphylococcus pasteuri1770FJ189773clade_325NN
Staphylococcus pseudintermedius1771CP002439clade_325NN
Staphylococcus saccharolyticus1772NR_029158clade_325NN
Staphylococcus saprophyticus1773NC_007350clade_325NN
Staphylococcus sp. clone bottae71777AF467424clade_325NN
Staphylococcus sp. H2921775AB177642clade_325NN
Staphylococcus sp. H7801776AB177644clade_325NN
Staphylococcus succinus1778NR_028667clade_325NN
Staphylococcus warneri1780ACPZ01000009clade_325NN
Staphylococcus xylosus1781AY395016clade_325NN
Cardiobacterium hominis490ACKY01000036clade_326NN
Cardiobacterium valvarum491NR_028847clade_326NN
Pseudomonas fluorescens1593AY622220clade_326NN
Pseudomonas gessardii1594FJ943496clade_326NN
Pseudomonas monteilii1596NR_024910clade_326NN
Pseudomonas poae1597GU188951clade_326NN
Pseudomonas putida1599AF094741clade_326NN
Pseudomonas sp. G12291601DQ910482clade_326NN
Pseudomonas tolaasii1604AF320988clade_326NN
Pseudomonas viridiflava1605NR_042764clade_326NN
Listeria grayi1185ACCR02000003clade_328NOP
Listeria innocua1186JF967625clade_328NN
Listeria ivanovii1187X56151clade_328NN
Listeria monocytogenes1188CP002003clade_328NCategory-B
Listeria welshimeri1189AM263198clade_328NOP
Capnocytophaga sp. oral clone484AY923149clade_333NN
ASCH05
Capnocytophaga sputigena489ABZV01000054clade_333NN
Leptotrichia genomosp. C11166AY278621clade_334NN
Leptotrichia shahii1169AY029806clade_334NN
Leptotrichia sp. neutropenic Patient1170AF189244clade_334NN
Leptotrichia sp. oral clone GT0181171AY349384clade_334NN
Leptotrichia sp. oral clone GT0201172AY349385clade_334NN
Bacteroides sp. 20_3296ACRQ01000064clade_335NN
Bacteroides sp. 3_1_19307ADCJ01000062clade_335NN
Bacteroides sp. 3_2_5311ACIB01000079clade_335NN
Parabacteroides distasonis1416CP000140clade_335NN
Parabacteroides goldsteinii1417AY974070clade_335NN
Parabacteroides gordonii1418AB470344clade_335NN
Parabacteroides sp. D131421ACPW01000017clade_335NN
Capnocytophaga genomosp. C1477AY278613clade_336NN
Capnocytophaga ochracea480AEOH01000054clade_336NN
Capnocytophaga sp. GEJ8481GU561335clade_336NN
Capnocytophaga sp. oral strain486AY005077clade_336NN
A47ROY
Capnocytophaga sp. S1b482U42009clade_336NN
Paraprevotella clara1426AFFY01000068clade_336NN
Bacteroides heparinolyticus282JN867284clade_338NN
Prevotella heparinolytica1500GQ422742clade_338NN
Treponema genomosp. P4 oral1928DQ003618clade_339NN
clone MB2_G19
Treponema genomosp. P6 oral1930DQ003625clade_339NN
clone MB4_G11
Treponema sp. oral taxon 2541952GU408803clade_339NN
Treponema sp. oral taxon 5081956GU413616clade_339NN
Treponema sp. oral taxon 5181957GU413640clade_339NN
Chlamydia muridarum502AE002160clade_341NOP
Chlamydia trachomatis504U68443clade_341NOP
Chlamydia psittaci503NR_036864clade_342NCategory-B
Chlamydophila pneumoniae509NC_002179clade_342NOP
Chlamydophila psittaci510D85712clade_342NOP
Anaerococcus octavius146NR_026360clade_343NN
Anaerococcus sp. 8405254149HM587319clade_343NN
Anaerococcus sp. 9401487150HM587322clade_343NN
Anaerococcus sp. 9403502151HM587325clade_343NN
Gardnerella vaginalis923CP001849clade_344NN
Campylobacter lari466CP000932clade_346NOP
Anaerobiospirillum142NR_026075clade_347NN
succiniciproducens
Anaerobiospirillum thomasii143AJ420985clade_347NN
Ruminobacter amylophilus1654NR_026450clade_347NN
Succinatimonas hippei1897AEVO01000027clade_347NN
Actinomyces europaeus54NR_026363clade_348NN
Actinomyces sp. oral clone GU00982AY349361clade_348NN
Moraxella catarrhalis1260CP002005clade_349NN
Moraxella lincolnii1261FR822735clade_349NN
Moraxella sp. 162851263JF682466clade_349NN
Psychrobacter sp. 139831613HM212668clade_349NN
Actinobaculum massiliae49AF487679clade_350NN
Actinobaculum schaalii50AY957507clade_350NN
Actinobaculum sp. BM#10134251AY282578clade_350NN
Actinobaculum sp. P2P_19 P152AY207066clade_350NN
Actinomyces sp. oral clone IO07684AY349363clade_350NN
Actinomyces sp. oral taxon 84893ACUY01000072clade_350NN
Actinomyces neuii65X71862clade_352NN
Mobiluncus mulieris1252ACKW01000035clade_352NN
Blastomonas natatoria372NR_040824clade_356NN
Novosphingobium aromaticivorans1357AAAV03000008clade_356NN
Sphingomonas sp. oral clone1745AY349411clade_356NN
FI012
Sphingopyxis alaskensis1749CP000356clade_356NN
Oxalobacter formigenes1389ACDQ01000020clade_357NN
Veillonella atypica1974AEDS01000059clade_358NN
Veillonella dispar1975ACIK02000021clade_358NN
Veillonella genomosp. P1 oral1976DQ003631clade_358NN
clone MB5_P17
Veillonella parvula1978ADFU01000009clade_358NN
Veillonella sp. 3_1_441979ADCV01000019clade_358NN
Veillonella sp. 6_1_271980ADCW01000016clade_358NN
Veillonella sp. ACP11981HQ616359clade_358NN
Veillonella sp. AS161982HQ616365clade_358NN
Veillonella sp. BS32b1983HQ616368clade_358NN
Veillonella sp. ICM51a1984HQ616396clade_358NN
Veillonella sp. MSA121985HQ616381clade_358NN
Veillonella sp. NVG 100cf1986EF108443clade_358NN
Veillonella sp. OK111987JN695650clade_358NN
Veillonella sp. oral clone ASCG011990AY923144clade_358NN
Veillonella sp. oral clone ASCG021991AY953257clade_358NN
Veillonella sp. oral clone OH1A1992AY947495clade_358NN
Veillonella sp. oral taxon 1581993AENU01000007clade_358NN
Kocuria marina1040GQ260086clade_365NN
Kocuria rhizophila1042AY030315clade_365NN
Kocuria rosea1043X87756clade_365NN
Kocuria varians1044AF542074clade_365NN
Clostridiaceae bacterium END_2531EF451053clade_368NN
Micrococcus antarcticus1242NR_025285clade_371NN
Micrococcus luteus1243NR_075062clade_371NN
Micrococcus lylae1244NR_026200clade_371NN
Micrococcus sp. 1851245EU714334clade_371NN
Lactobacillus brevis1072EU194349clade_372NN
Lactobacillus parabrevis1104NR_042456clade_372NN
Pediococcus acidilactici1436ACXB01000026clade_372NN
Pediococcus pentosaceus1437NR_075052clade_372NN
Lactobacillus dextrinicus1081NR_036861clade_373NN
Lactobacillus perolens1109NR_029360clade_373NN
Lactobacillus rhamnosus1113ABWJ01000068clade_373NN
Lactobacillus saniviri1118AB602569clade_373NN
Lactobacillus sp. BT61121HQ616370clade_373NN
Mycobacterium mageritense1282FR798914clade_374NOP
Mycobacterium neoaurum1286AF268445clade_374NOP
Mycobacterium smegmatis1291CP000480clade_374NOP
Mycobacterium sp. HE51304AJ012738clade_374NN
Dysgonomonas gadei775ADLV01000001clade_377NN
Dysgonomonas mossii776ADLW01000023clade_377NN
Porphyromonas levii1474NR_025907clade_377NN
Porphyromonas somerae1476AB547667clade_377NN
Bacteroides barnesiae267NR_041446clade_378NN
Bacteroides coprocola272ABIY02000050clade_378NN
Bacteroides coprophilus273ACBW01000012clade_378NN
Bacteroides dorei274ABWZ01000093clade_378NN
Bacteroides massiliensis284AB200226clade_378NN
Bacteroides plebeius289AB200218clade_378NN
Bacteroides sp. 3_1_33FAA309ACPS01000085clade_378NN
Bacteroides sp. 3_1_40A310ACRT01000136clade_378NN
Bacteroides sp. 4_3_47FAA313ACDR02000029clade_378NN
Bacteroides sp. 9_1_42FAA314ACAA01000096clade_378NN
Bacteroides sp. NB_8323AB117565clade_378NN
Bacteroides vulgatus331CP000139clade_378NN
Bacteroides ovatus287ACWH01000036clade_38NN
Bacteroides sp. 1_1_30294ADCL01000128clade_38NN
Bacteroides sp. 2_1_22297ACPQ01000117clade_38NN
Bacteroides sp. 2_2_4299ABZZ01000168clade_38NN
Bacteroides sp. 3_1_23308ACRS01000081clade_38NN
Bacteroides sp. D1318ACAB02000030clade_38NN
Bacteroides sp. D2321ACGA01000077clade_38NN
Bacteroides sp. D22320ADCK01000151clade_38NN
Bacteroides xylanisolvens332ADKP01000087clade_38NN
Treponema lecithinolyticum1931NR_026247clade_380NOP
Treponema parvum1933AF302937clade_380NOP
Treponema sp. oral clone JU0251940AY349417clade_380NN
Treponema sp. oral taxon 2701954GQ422733clade_380NN
Parascardovia denticolens1428ADEB01000020clade_381NN
Scardovia inopinata1688AB029087clade_381NN
Scardovia wiggsiae1689AY278626clade_381NN
Clostridiales bacterium 9400853533HM587320clade_384NN
Mogibacterium diversum1254NR_027191clade_384NN
Mogibacterium neglectum1255NR_027203clade_384NN
Mogibacterium pumilum1256NR_028608clade_384NN
Mogibacterium timidum1257Z36296clade_384NN
Borrelia burgdorferi389ABGI01000001clade_386NOP
Borrelia garinii392ABJV01000001clade_386NOP
Borrelia sp. NE49397AJ224142clade_386NOP
Caldimonas manganoxidans457NR_040787clade_387NN
Comamonadaceae bacterium oral667HM099651clade_387NN
taxon F47
Lautropia mirabilis1149AEQP01000026clade_387NN
Lautropia sp. oral clone AP0091150AY005030clade_387NN
Peptoniphilus asaccharolyticus1441D14145clade_389NN
Peptoniphilus duerdenii1442EU526290clade_389NN
Peptoniphilus harei1443NR_026358clade_389NN
Peptoniphilus indolicus1444AY153431clade_389NN
Peptoniphilus lacrimalis1446ADDO01000050clade_389NN
Peptoniphilus sp. gpac0771450AM176527clade_389NN
Peptoniphilus sp. JC1401447JF824803clade_389NN
Peptoniphilus sp. oral taxon 3861452ADCS01000031clade_389NN
Peptoniphilus sp. oral taxon 8361453AEAA01000090clade_389NN
Peptostreptococcaceae bacterium1454JN837495clade_389NN
ph1
Dialister pneumosintes765HM596297clade_390NN
Dialister sp. oral taxon 502767GQ422739clade_390NN
Cupriavidus metallidurans741GU230889clade_391NN
Herbaspirillum seropedicae1001CP002039clade_391NN
Herbaspirillum sp. JC2061002JN657219clade_391NN
Janthinobacterium sp. SY121015EF455530clade_391NN
Massilia sp. CCUG 43427A1197FR773700clade_391NN
Ralstonia pickettii1615NC_010682clade_391NN
Ralstonia sp. 5_7_47FAA1616ACUF01000076clade_391NN
Francisella novicida889ABSS01000002clade_392NN
Francisella philomiragia890AY928394clade_392NN
Francisella tularensis891ABAZ01000082clade_392NCategory-A
Ignatzschineria indica1009HQ823562clade_392NN
Ignatzschineria sp. NML 95_02601010HQ823559clade_392NN
Streptococcus mutans1814AP010655clade_394NN
Lactobacillus gasseri1084ACOZ01000018clade_398NN
Lactobacillus hominis1090FR681902clade_398NN
Lactobacillus iners1091AEKJ01000002clade_398NN
Lactobacillus johnsonii1093AE017198clade_398NN
Lactobacillus senioris1119AB602570clade_398NN
Lactobacillus sp. oral clone HT0021135AY349382clade_398NN
Weissella beninensis2006EU439435clade_398NN
Sphingomonas echinoides1744NR_024700clade_399NN
Sphingomonas sp. oral taxon A091747HM099639clade_399NN
Sphingomonas sp. oral taxon F711748HM099645clade_399NN
Zymomonas mobilis2032NR_074274clade_399NN
Arcanobacterium haemolyticum174NR_025347clade_400NN
Arcanobacterium pyogenes175GU585578clade_400NN
Trueperella pyogenes1962NR_044858clade_400NN
Lactococcus garvieae1144AF061005clade_401NN
Lactococcus lactis1145CP002365clade_401NN
Brevibacterium mcbrellneri424ADNU01000076clade_402NN
Brevibacterium paucivorans425EU086796clade_402NN
Brevibacterium sp. JC43428JF824806clade_402NN
Selenomonas artemidis1692HM596274clade_403NN
Selenomonas sp. FOBRC91704HQ616378clade_403NN
Selenomonas sp. oral taxon 1371715AENV01000007clade_403NN
Desmospora activa751AM940019clade_404NN
Desmospora sp. 8437752AFHT01000143clade_404NN
Paenibacillus sp. oral taxon F451407HM099647clade_404NN
Corynebacterium ammoniagenes682ADNS01000011clade_405NN
Corynebacterium aurimucosum687ACLH01000041clade_405NN
Corynebacterium bovis688AF537590clade_405NN
Corynebacterium canis689GQ871934clade_405NN
Corynebacterium casei690NR_025101clade_405NN
Corynebacterium durum694Z97069clade_405NN
Corynebacterium efficiens695ACLI01000121clade_405NN
Corynebacterium falsenii696Y13024clade_405NN
Corynebacterium flavescens697NR_037040clade_405NN
Corynebacterium glutamicum701BA000036clade_405NN
Corynebacterium jeikeium704ACYW01000001clade_405NOP
Corynebacterium kroppenstedtii705NR_026380clade_405NN
Corynebacterium lipophiloflavum706ACHJ01000075clade_405NN
Corynebacterium matruchotii709ACSH02000003clade_405NN
Corynebacterium minutissimum710X82064clade_405NN
Corynebacterium resistens718ADGN01000058clade_405NN
Corynebacterium simulans720AF537604clade_405NN
Corynebacterium singulare721NR_026394clade_405NN
Corynebacterium sp. 1 ex sheep722Y13427clade_405NN
Corynebacterium sp. NML726GU238413clade_405NN
99_0018
Corynebacterium striatum727ACGE01000001clade_405NOP
Corynebacterium urealyticum732X81913clade_405NOP
Corynebacterium variabile734NR_025314clade_405NN
Aerococcus sanguinicola98AY837833clade_407NN
Aerococcus urinae99CP002512clade_407NN
Aerococcus urinaeequi100NR_043443clade_407NN
Aerococcus viridans101ADNT01000041clade_407NN
Fusobacterium naviforme898HQ223106clade_408NN
Moryella indoligenes1268AF527773clade_408NN
Selenomonas genomosp. P51697AY341820clade_410NN
Selenomonas sp. oral clone IQ0481710AY349408clade_410NN
Selenomonas sputigena1717ACKP02000033clade_410NN
Hyphomicrobium sulfonivorans1007AY468372clade_411NN
Methylocella silvestris1228NR_074237clade_411NN
Legionella pneumophila1153NC_002942clade_412NOP
Lactobacillus coryniformis1077NR_044705clade_413NN
Arthrobacter agilis178NR_026198clade_414NN
Arthrobacter arilaitensis179NR_074608clade_414NN
Arthrobacter bergerei180NR_025612clade_414NN
Arthrobacter globiformis181NR_026187clade_414NN
Arthrobacter nicotianae182NR_026190clade_414NN
Mycobacterium abscessus1269AGQU01000002clade_418NOP
Mycobacterium chelonae1273AB548610clade_418NOP
Bacteroides salanitronis291CP002530clade_419NN
Paraprevotella xylaniphila1427AFBR01000011clade_419NN
Barnesiella intestinihominis336AB370251clade_420NN
Barnesiella viscericola337NR_041508clade_420NN
Parabacteroides sp. NS31_31422JN029805clade_420NN
Porphyromonadaceae bacterium1470EF184292clade_420NN
NML 060648
Tannerella forsythia1913CP003191clade_420NN
Tannerella sp. 6_1_58FAA_CT11914ACWX01000068clade_420NN
Mycoplasma amphoriforme1311AY531656clade_421NN
Mycoplasma genitalium1317L43967clade_421NN
Mycoplasma pneumoniae1322NC_000912clade_421NN
Mycoplasma penetrans1321NC_004432clade_422NN
Ureaplasma parvum1966AE002127clade_422NN
Ureaplasma urealyticum1967AAYN01000002clade_422NN
Treponema genomosp. P11927AY341822clade_425NN
Treponema sp. oral taxon 2281943GU408580clade_425NN
Treponema sp. oral taxon 2301944GU408603clade_425NN
Treponema sp. oral taxon 2311945GU408631clade_425NN
Treponema sp. oral taxon 2321946GU408646clade_425NN
Treponema sp. oral taxon 2351947GU408673clade_425NN
Treponema sp. ovine footrot1959AJ010951clade_425NN
Treponema vincentii1960ACYH01000036clade_425NOP
Burkholderiales bacterium 1_1_47452ADCQ01000066clade_432NOP
Parasutterella excrementihominis1429AFBP01000029clade_432NN
Parasutterella secunda1430AB491209clade_432NN
Sutterella morbirenis1898AJ832129clade_432NN
Sutterella sanguinus1900AJ748647clade_432NN
Sutterella sp. YIT 120721901AB491210clade_432NN
Sutterella stercoricanis1902NR_025600clade_432NN
Sutterella wadsworthensis1903ADMF01000048clade_432NN
Propionibacterium freudenreichii1572NR_036972clade_433NN
Propionibacterium sp. oral taxon1580GQ422728clade_433NN
192
Tessaracoccus sp. oral taxon F041917HM099640clade_433NN
Peptoniphilus ivorii1445Y07840clade_434NN
Peptoniphilus sp. gpac0071448AM176517clade_434NN
Peptoniphilus sp. gpac018A1449AM176519clade_434NN
Peptoniphilus sp. gpac1481451AM176535clade_434NN
Flexispira rappini887AY126479clade_436NN
Helicobacter bilis993ACDN01000023clade_436NN
Helicobacter cinaedi995ABQT01000054clade_436NN
Helicobacter sp. None998U44756clade_436NN
Brevundimonas subvibrioides429CP002102clade_438NN
Hyphomonas neptunium1008NR_074092clade_438NN
Phenylobacterium zucineum1465AY628697clade_438NN
Streptococcus downei1793AEKN01000002clade_441NN
Streptococcus sp. SHV5151848Y07601clade_441NN
Acinetobacter sp. CIP 53.8240JQ638584clade_443NN
Halomonas elongata990NR_074782clade_443NN
Halomonas johnsoniae991FR775979clade_443NN
Butyrivibrio fibrisolvens456U41172clade_444NN
Roseburia sp. 11SE371640FM954975clade_444NN
Roseburia sp. 11SE381641FM954976clade_444NN
Shuttleworthia satelles1728ACIP02000004clade_444NN
Shuttleworthia sp. MSX8B1729HQ616383clade_444NN
Shuttleworthia sp. oral taxon G691730GU432167clade_444NN
Bdellovibrio sp. MPA344AY294215clade_445NN
Desulfobulbus sp. oral clone755AY005036clade_445NN
CH031
Desulfovibrio desulfuricans757DQ092636clade_445NN
Desulfovibrio fairfieldensis758U42221clade_445NN
Desulfovibrio piger759AF192152clade_445NN
Desulfovibrio sp. 3_1_syn3760ADDR01000239clade_445NN
Geobacter bemidjiensis941CP001124clade_445NN
Brachybacterium alimentarium401NR_026269clade_446NN
Brachybacterium conglomeratum402AB537169clade_446NN
Brachybacterium tyrofermentans403NR_026272clade_446NN
Dermabacter hominis749FJ263375clade_446NN
Aneurinibacillus thermoaerophilus171NR_029303clade_448NN
Brevibacillus agri409NR_040983clade_448NN
Brevibacillus centrosporus411NR_043414clade_448NN
Brevibacillus choshinensis412NR_040980clade_448NN
Brevibacillus invocatus413NR_041836clade_448NN
Brevibacillus parabrevis415NR_040981clade_448NN
Brevibacillus reuszeri416NR_040982clade_448NN
Brevibacillus sp. phR417JN837488clade_448NN
Brevibacillus thermoruber418NR_026514clade_448NN
Lactobacillus murinus1100NR_042231clade_449NN
Lactobacillus oeni1102NR_043095clade_449NN
Lactobacillus ruminis1115ACGS02000043clade_449NN
Lactobacillus vini1141NR_042196clade_449NN
Gemella haemolysans924ACDZ02000012clade_450NN
Gemella morbillorum925NR_025904clade_450NN
Gemella morbillorum926ACRX01000010clade_450NN
Gemella sanguinis927ACRY01000057clade_450NN
Gemella sp. oral clone ASCE02929AY923133clade_450NN
Gemella sp. oral clone ASCF04930AY923139clade_450NN
Gemella sp. oral clone ASCF12931AY923143clade_450NN
Gemella sp. WAL 1945J928EU427463clade_450NN
Sporolactobacillus nakayamae1753NR_042247clade_451NN
Gluconacetobacter entanii945NR_028909clade_452NN
Gluconacetobacter europaeus946NR_026513clade_452NN
Gluconacetobacter hansenii947NR_026133clade_452NN
Gluconacetobacter oboediens949NR_041295clade_452NN
Gluconacetobacter xylinus950NR_074338clade_452NN
Auritibacter ignavus193FN554542clade_453NN
Dermacoccus sp. Ellin185750AEIQ01000090clade_453NN
Janibacter limosus1013NR_026362clade_453NN
Janibacter melonis1014EF063716clade_453NN
Acetobacter aceti7NR_026121clade_454NN
Acetobacter fabarum8NR_042678clade_454NN
Acetobacter lovaniensis9NR_040832clade_454NN
Acetobacter malorum10NR_025513clade_454NN
Acetobacter orientalis11NR_028625clade_454NN
Acetobacter pasteurianus12NR_026107clade_454NN
Acetobacter pomorum13NR_042112clade_454NN
Acetobacter syzygii14NR_040868clade_454NN
Acetobacter tropicalis15NR_036881clade_454NN
Gluconacetobacter azotocaptans943NR_028767clade_454NN
Gluconacetobacter diazotrophicus944NR_074292clade_454NN
Gluconacetobacter johannae948NR_024959clade_454NN
Nocardia brasiliensis1351AIHV01000038clade_455NN
Nocardia cyriacigeorgica1352HQ009486clade_455NN
Nocardia puris1354NR_028994clade_455NN
Nocardia sp. 01_Je_0251355GU574059clade_455NN
Rhodococcus equi1623ADNW01000058clade_455NN
Oceanobacillus caeni1358NR_041533clade_456NN
Oceanobacillus sp. Ndiop1359CAER01000083clade_456NN
Ornithinibacillus bavariensis1384NR_044923clade_456NN
Ornithinibacillus sp. 7_10AIA1385FN397526clade_456NN
Virgibacillus proomii2005NR_025308clade_456NN
Corynebacterium amycolatum683ABZU01000033clade_457NOP
Corynebacterium hansenii702AM946639clade_457NN
Corynebacterium xerosis735FN179330clade_457NOP
Staphylococcaceae bacterium1756AY841362clade_458NN
NML 92_0017
Staphylococcus fleurettii1766NR_041326clade_458NN
Staphylococcus sciuri1774NR_025520clade_458NN
Staphylococcus vitulinus1779NR_024670clade_458NN
Stenotrophomonas maltophilia1782AAVZ01000005clade_459NN
Stenotrophomonas sp. FG_61783EF017810clade_459NN
Mycobacterium africanum1270AF480605clade_46NOP
Mycobacterium alsiensis1271AJ938169clade_46NOP
Mycobacterium avium1272CP000479clade_46NOP
Mycobacterium colombiense1274AM062764clade_46NOP
Mycobacterium gordonae1276GU142930clade_46NOP
Mycobacterium intracellulare1277GQ153276clade_46NOP
Mycobacterium kansasii1278AF480601clade_46NOP
Mycobacterium lacus1279NR_025175clade_46NOP
Mycobacterium leprae1280FM211192clade_46NOP
Mycobacterium lepromatosis1281EU203590clade_46NOP
Mycobacterium mantenii1283FJ042897clade_46NOP
Mycobacterium marinum1284NC_010612clade_46NOP
Mycobacterium microti1285NR_025234clade_46NOP
Mycobacterium parascrofulaceum1287ADNV01000350clade_46NOP
Mycobacterium seoulense1290DQ536403clade_46NOP
Mycobacterium sp. 17611292EU703150clade_46NN
Mycobacterium sp. 17911295EU703148clade_46NN
Mycobacterium sp. 17971296EU703149clade_46NN
Mycobacterium sp.1298HQ174245clade_46NN
B10_07.09.0206
Mycobacterium sp. NLA0010007361305HM627011clade_46NN
Mycobacterium sp. W1306DQ437715clade_46NN
Mycobacterium tuberculosis1307CP001658clade_46NCategory-C
Mycobacterium ulcerans1308AB548725clade_46NOP
Mycobacterium vulneris1309EU834055clade_46NOP
Xanthomonas campestris2016EF101975clade_461NN
Xanthomonas sp. kmd_4892017EU723184clade_461NN
Dietzia natronolimnaea769GQ870426clade_462NN
Dietzia sp. BBDP51770DQ337512clade_462NN
Dietzia sp. CA149771GQ870422clade_462NN
Dietzia timorensis772GQ870424clade_462NN
Gordonia bronchialis951NR_027594clade_463NN
Gordonia polyisoprenivorans952DQ385609clade_463NN
Gordonia sp. KTR9953DQ068383clade_463NN
Gordonia sputi954FJ536304clade_463NN
Gordonia terrae955GQ848239clade_463NN
Leptotrichia goodfellowii1167ADAD01000110clade_465NN
Leptotrichia sp. oral clone IK0401174AY349387clade_465NN
Leptotrichia sp. oral clone1175AY207053clade_465NN
P2PB_51 P1
Bacteroidales genomosp. P7 oral264DQ003623clade_466NN
clone MB3_P19
Butyricimonas virosa454AB443949clade_466NN
Odoribacter laneus1363AB490805clade_466NN
Odoribacter splanchnicus1364CP002544clade_466NN
Capnocytophaga gingivalis478ACLQ01000011clade_467NN
Capnocytophaga granulosa479X97248clade_467NN
Capnocytophaga sp. oral clone483AY005074clade_467NN
AH015
Capnocytophaga sp. oral strain S3487AY005073clade_467NN
Capnocytophaga sp. oral taxon488AEXX01000050clade_467NN
338
Capnocytophaga canimorsus476CP002113clade_468NN
Capnocytophaga sp. oral clone485AY349368clade_468NN
ID062
Lactobacillus catenaformis1075M23729clade_469NN
Lactobacillus vitulinus1142NR_041305clade_469NN
Cetobacterium somerae501AJ438155clade_470NN
Fusobacterium gonidiaformans896ACET01000043clade_470NN
Fusobacterium mortiferum897ACDB02000034clade_470NN
Fusobacterium necrogenes899X55408clade_470NN
Fusobacterium necrophorum900AM905356clade_470NN
Fusobacterium sp. 12_1B905AGWJ01000070clade_470NN
Fusobacterium sp. 3_1_5R911ACDD01000078clade_470NN
Fusobacterium sp. D12918ACDG02000036clade_470NN
Fusobacterium ulcerans921ACDH01000090clade_470NN
Fusobacterium varium922ACIE01000009clade_470NN
Mycoplasma arthritidis1312NC_011025clade_473NN
Mycoplasma faucium1314NR_024983clade_473NN
Mycoplasma hominis1318AF443616clade_473NN
Mycoplasma orale1319AY796060clade_473NN
Mycoplasma salivarium1324M24661clade_473NN
Mitsuokella jalaludinii1247NR_028840clade_474NN
Mitsuokella multacida1248ABWK02000005clade_474NN
Mitsuokella sp. oral taxon 5211249GU413658clade_474NN
Mitsuokella sp. oral taxon G681250GU432166clade_474NN
Selenomonas genomosp. C11695AY278627clade_474NN
Selenomonas genomosp. P8 oral1700DQ003628clade_474NN
clone MB5_P06
Selenomonas ruminantium1703NR_075026clade_474NN
Veillonellaceae bacterium oral1994GU402916clade_474NN
taxon 131
Alloscardovia omnicolens139NR_042583clade_475NN
Alloscardovia sp. OB7196140AB425070clade_475NN
Bifidobacterium urinalis366AJ278695clade_475NN
Prevotella loescheii1503JN867231clade_48NN
Prevotella sp. oral clone ASCG121530DQ272511clade_48NN
Prevotella sp. oral clone GU0271540AY349398clade_48NN
Prevotella sp. oral taxon 4721553ACZS01000106clade_48NN
Selenomonas dianae1693GQ422719clade_480NN
Selenomonas flueggei1694AF287803clade_480NN
Selenomonas genomosp. C21696AY278628clade_480NN
Selenomonas genomosp. P6 oral1698DQ003636clade_480NN
clone MB3_C41
Selenomonas genomosp. P7 oral1699DQ003627clade_480NN
clone MB5_C08
Selenomonas infelix1701AF287802clade_480NN
Selenomonas noxia1702GU470909clade_480NN
Selenomonas sp. oral clone FT0501705AY349403clade_480NN
Selenomonas sp. oral clone GI0641706AY349404clade_480NN
Selenomonas sp. oral clone1707AY349405clade_480NN
GT010
Selenomonas sp. oral clone1708AY349406clade_480NN
HU051
Selenomonas sp. oral clone IK0041709AY349407clade_480NN
Selenomonas sp. oral clone JI0211711AY349409clade_480NN
Selenomonas sp. oral clone JS0311712AY349410clade_480NN
Selenomonas sp. oral clone OH4A1713AY947498clade_480NN
Selenomonas sp. oral clone1714AY207052clade_480NN
P2PA_80 P4
Selenomonas sp. oral taxon 1491716AEEJ01000007clade_480NN
Veillonellaceae bacterium oral1995GU470897clade_480NN
taxon 155
Agrococcus jenensis117NR_026275clade_484NN
Microbacterium gubbeenense1232NR_025098clade_484NN
Pseudoclavibacter sp. Timone1590FJ375951clade_484NN
Tropheryma whipplei1961BX251412clade_484NN
Zimmermannella bifida2031AB012592clade_484NN
Legionella hackeliae1151M36028clade_486NOP
Legionella longbeachae1152M36029clade_486NOP
Legionella sp. D39231154JN380999clade_486NOP
Legionella sp. D40881155JN381012clade_486NOP
Legionella sp. H631156JF831047clade_486NOP
Legionella sp. NML 93L0541157GU062706clade_486NOP
Legionella steelei1158HQ398202clade_486NOP
Tatlockia micdadei1915M36032clade_486NN
Helicobacter pullorum996ABQU01000097clade_489NN
Acetobacteraceae bacterium16AGEZ01000040clade_490NN
AT_5844
Roseomonas cervicalis1643ADVL01000363clade_490NN
Roseomonas mucosa1644NR_028857clade_490NN
Roseomonas sp. NML94_01931645AF533357clade_490NN
Roseomonas sp. NML97_01211646AF533359clade_490NN
Roseomonas sp. NML98_00091647AF533358clade_490NN
Roseomonas sp. NML98_01571648AF533360clade_490NN
Rickettsia akari1627CP000847clade_492NOP
Rickettsia conorii1628AE008647clade_492NOP
Rickettsia prowazekii1629M21789clade_492NCategory-B
Rickettsia rickettsii1630NC_010263clade_492NOP
Rickettsia slovaca1631L36224clade_492NOP
Rickettsia typhi1632AE017197clade_492NOP
Anaeroglobus geminatus160AGCJ01000054clade_493NN
Megasphaera genomosp. C11201AY278622clade_493NN
Megasphaera micronuciformis1203AECS01000020clade_493NN
Clostridiales genomosp. BVAB3540CP001850clade_495NN
Tsukamurella paurometabola1963X80628clade_496NN
Tsukamurella tyrosinosolvens1964AB478958clade_496NN
Abiotrophia para_adiacens2AB022027clade_497NN
Carnobacterium divergens492NR_044706clade_497NN
Carnobacterium maltaromaticum493NC_019425clade_497NN
Enterococcus avium800AF133535clade_497NN
Enterococcus caccae801AY943820clade_497NN
Enterococcus casseliflavus802AEWT01000047clade_497NN
Enterococcus durans803AJ276354clade_497NN
Enterococcus faecalis804AE016830clade_497NN
Enterococcus faecium805AM157434clade_497NN
Enterococcus gallinarum806AB269767clade_497NN
Enterococcus gilvus807AY033814clade_497NN
Enterococcus hawaiiensis808AY321377clade_497NN
Enterococcus hirae809AF061011clade_497NN
Enterococcus italicus810AEPV01000109clade_497NN
Enterococcus mundtii811NR_024906clade_497NN
Enterococcus raffinosus812FN600541clade_497NN
Enterococcus sp. BV2CASA2813JN809766clade_497NN
Enterococcus sp. CCRI_16620814GU457263clade_497NN
Enterococcus sp. F95815FJ463817clade_497NN
Enterococcus sp. RfL6816AJ133478clade_497NN
Enterococcus thailandicus817AY321376clade_497NN
Fusobacterium canifelinum893AY162222clade_497NN
Fusobacterium genomosp. C1894AY278616clade_497NN
Fusobacterium genomosp. C2895AY278617clade_497NN
Fusobacterium periodonticum902ACJY01000002clade_497NN
Fusobacterium sp. 1_1_41FAA906ADGG01000053clade_497NN
Fusobacterium sp. 11_3_2904ACUO01000052clade_497NN
Fusobacterium sp. 2_1_31907ACDC02000018clade_497NN
Fusobacterium sp. 3_1_27908ADGF01000045clade_497NN
Fusobacterium sp. 3_1_33909ACQE01000178clade_497NN
Fusobacterium sp. 3_1_36A2910ACPU01000044clade_497NN
Fusobacterium sp. AC18912HQ616357clade_497NN
Fusobacterium sp. ACB2913HQ616358clade_497NN
Fusobacterium sp. AS2914HQ616361clade_497NN
Fusobacterium sp. CM1915HQ616371clade_497NN
Fusobacterium sp. CM21916HQ616375clade_497NN
Fusobacterium sp. CM22917HQ616376clade_497NN
Fusobacterium sp. oral clone919AY923141clade_497NN
ASCF06
Fusobacterium sp. oral clone920AY953256clade_497NN
ASCF11
Granulicatella adiacens959ACKZ01000002clade_497NN
Granulicatella elegans960AB252689clade_497NN
Granulicatella paradiacens961AY879298clade_497NN
Granulicatella sp. oral clone963AY923126clade_497NN
ASC02
Granulicatella sp. oral clone964DQ341469clade_497NN
ASCA05
Granulicatella sp. oral clone965AY953251clade_497NN
ASCB09
Granulicatella sp. oral clone966AY923146clade_497NN
ASCG05
Tetragenococcus halophilus1918NR_075020clade_497NN
Tetragenococcus koreensis1919NR_043113clade_497NN
Vagococcus fluvialis1973NR_026489clade_497NN
Chryseobacterium anthropi514AM982793clade_498NN
Chryseobacterium gleum515ACKQ02000003clade_498NN
Chryseobacterium hominis516NR_042517clade_498NN
Treponema refringens1936AF426101clade_499NOP
Treponema sp. oral clone JU0311941AY349416clade_499NN
Treponema sp. oral taxon 2391948GU408738clade_499NN
Treponema sp. oral taxon 2711955GU408871clade_499NN
Alistipes finegoldii129NR_043064clade_500NN
Alistipes onderdonkii131NR_043318clade_500NN
Alistipes putredinis132ABFK02000017clade_500NN
Alistipes shahii133FP929032clade_500NN
Alistipes sp. HGB5134AENZ01000082clade_500NN
Alistipes sp. JC50135JF824804clade_500NN
Alistipes sp. RMA 9912136GQ140629clade_500NN
Mycoplasma agalactiae1310AF010477clade_501NN
Mycoplasma bovoculi1313NR_025987clade_501NN
Mycoplasma fermentans1315CP002458clade_501NN
Mycoplasma flocculare1316X62699clade_501NN
Mycoplasma ovipneumoniae1320NR_025989clade_501NN
Arcobacter butzleri176AEPT01000071clade_502NN
Arcobacter cryaerophilus177NR_025905clade_502NN
Campylobacter curvus461NC_009715clade_502NOP
Campylobacter rectus467ACFU01000050clade_502NOP
Campylobacter showae468ACVQ01000030clade_502NOP
Campylobacter sp. FOBRC14469HQ616379clade_502NOP
Campylobacter sp. FOBRC15470HQ616380clade_502NOP
Campylobacter sp. oral clone471AY005038clade_502NOP
BB120
Campylobacter sputorum472NR_044839clade_502NOP
Bacteroides ureolyticus330GQ167666clade_504NN
Campylobacter gracilis463ACYG01000026clade_504NOP
Campylobacter hominis464NC_009714clade_504NOP
Dialister invisus762ACIM02000001clade_506NN
Dialister micraerophilus763AFBB01000028clade_506NN
Dialister microaerophilus764AENT01000008clade_506NN
Dialister propionicifaciens766NR_043231clade_506NN
Dialister succinatiphilus768AB370249clade_506NN
Megasphaera elsdenii1200AY038996clade_506NN
Megasphaera genomosp. type_11202ADGP01000010clade_506NN
Megasphaera sp. BLPYG_071204HM990964clade_506NN
Megasphaera sp. UPII 199_61205AFIJ01000040clade_506NN
Chromobacterium violaceum513NC_005085clade_507NN
Laribacter hongkongensis1148CP001154clade_507NN
Methylophilus sp. ECd51229AY436794clade_507NN
Finegoldia magna883ACHM02000001clade_509NN
Parvimonas micra1431AB729072clade_509NN
Parvimonas sp. oral taxon 1101432AFII01000002clade_509NN
Peptostreptococcus micros1456AM176538clade_509NN
Peptostreptococcus sp. oral clone1460AY349390clade_509NN
FJ023
Peptostreptococcus sp. P4P_311458AY207059clade_509NN
P3
Helicobacter pylori997CP000012clade_510NOP
Anaplasma marginale165ABOR01000019clade_511NN
Anaplasma phagocytophilum166NC_007797clade_511NN
Ehrlichia chaffeensis783AAIF01000035clade_511NOP
Neorickettsia risticii1349CP001431clade_511NN
Neorickettsia sennetsu1350NC_007798clade_511NN
Pseudoramibacter alactolyticus1606AB036759clade_512NN
Veillonella montpellierensis1977AF473836clade_513NN
Veillonella sp. oral clone ASCA081988AY923118clade_513NN
Veillonella sp. oral clone ASCB031989AY923122clade_513NN
Inquilinus limosus1012NR_029046clade_514NN
Sphingomonas sp. oral clone1746AY349412clade_514NN
FZ016
Anaerococcus lactolyticus145ABYO01000217clade_515NN
Anaerococcus prevotii147CP001708clade_515NN
Anaerococcus sp. gpac104152AM176528clade_515NN
Anaerococcus sp. gpac126153AM176530clade_515NN
Anaerococcus sp. gpac155154AM176536clade_515NN
Anaerococcus sp. gpac199155AM176539clade_515NN
Anaerococcus tetradius157ACGC01000107clade_515NN
Bacteroides coagulans271AB547639clade_515NN
Clostridiales bacterium 9403326534HM587324clade_515NN
Clostridiales bacterium ph2539JN837487clade_515NN
Peptostreptococcus sp. 9succ11457X90471clade_515NN
Peptostreptococcus sp. oral clone1459AB175072clade_515NN
AP24
Tissierella praeacuta1924NR_044860clade_515NN
Helicobacter canadensis994ABQS01000108clade_518NN
Peptostreptococcus anaerobius1455AY326462clade_520NN
Peptostreptococcus stomatis1461ADGQ01000048clade_520NN
Bilophila wadsworthia367ADCP01000166clade_521NN
Desulfovibrio vulgaris761NR_074897clade_521NN
Actinomyces nasicola64AJ508455clade_523NN
Cellulosimicrobium funkei500AY501364clade_523NN
Lactococcus raffinolactis1146NR_044359clade_524NN
Bacteroidales genomosp. P1258AY341819clade_529NN
Bacteroidales genomosp. P2 oral259DQ003613clade_529NN
clone MB1_G13
Bacteroidales genomosp. P3 oral260DQ003615clade_529NN
clone MB1_G34
Bacteroidales genomosp. P4 oral261DQ003617clade_529NN
clone MB2_G17
Bacteroidales genomosp. P5 oral262DQ003619clade_529NN
clone MB2_P04
Bacteroidales genomosp. P6 oral263DQ003634clade_529NN
clone MB3_C19
Bacteroidales genomosp. P8 oral265DQ003626clade_529NN
clone MB4_G15
Bacteroidetes bacterium oral taxon333HM099638clade_530NN
D27
Bacteroidetes bacterium oral taxon334HM099643clade_530NN
F31
Bacteroidetes bacterium oral taxon335HM099649clade_530NN
F44
Flavobacterium sp. NF2_1885FJ195988clade_530NN
Myroides odoratimimus1326NR_042354clade_530NN
Myroides sp. MY151327GU253339clade_530NN
Chlamydiales bacterium NS16507JN606076clade_531NN
Chlamydophila pecorum508D88317clade_531NOP
Parachlamydia sp. UWE251423BX908798clade_531NN
Fusobacterium russii903NR_044687clade_532NN
Streptobacillus moniliformis1784NR_027615clade_532NN
Eubacteriaceae bacterium P4P_50833AY207060clade_533NN
P4
Abiotrophia defectiva1ACIN02000016clade_534NN
Abiotrophia sp. oral clone3AY207063clade_534NN
P4PA_155 P1
Catonella genomosp. P1 oral clone496DQ003629clade_534NN
MB5_P12
Catonella morbi497ACIL02000016clade_534NN
Catonella sp. oral clone FL037498AY349369clade_534NN
Eremococcus coleocola818AENN01000008clade_534NN
Facklamia hominis879Y10772clade_534NN
Granulicatella sp. M658_99_3962AJ271861clade_534NN
Campylobacter coli459AAFL01000004clade_535NOP
Campylobacter concisus460CP000792clade_535NOP
Campylobacter fetus462ACLG01001177clade_535NOP
Campylobacter jejuni465AL139074clade_535NCategory-B
Campylobacter upsaliensis473AEPU01000040clade_535NOP
Atopobium minutum183HM007583clade_539NN
Atopobium parvulum184CP001721clade_539NN
Atopobium rimae185ACFE01000007clade_539NN
Atopobium sp. BS2186HQ616367clade_539NN
Atopobium sp. F0209187EU592966clade_539NN
Atopobium sp. ICM42b10188HQ616393clade_539NN
Atopobium sp. ICM57189HQ616400clade_539NN
Atopobium vaginae190AEDQ01000024clade_539NN
Coriobacteriaceae bacterium677JN809768clade_539NN
BV3Ac1
Actinomyces naeslundii63X81062clade_54NN
Actinomyces oricola67NR_025559clade_54NN
Actinomyces oris69BABV01000070clade_54NN
Actinomyces sp. 740094270EU484334clade_54NN
Actinomyces sp. ChDC B19772AF543275clade_54NN
Actinomyces sp. GEJ1573GU561313clade_54NN
Actinomyces sp. M2231_94_179AJ234063clade_54NN
Actinomyces sp. oral clone GU06783AY349362clade_54NN
Actinomyces sp. oral clone IO07785AY349364clade_54NN
Actinomyces sp. oral clone IP07386AY349365clade_54NN
Actinomyces sp. oral clone JA06388AY349367clade_54NN
Actinomyces sp. oral taxon 17089AFBL01000010clade_54NN
Actinomyces sp. oral taxon 17190AECW01000034clade_54NN
Actinomyces urogenitalis95ACFH01000038clade_54NN
Actinomyces viscosus96ACRE01000096clade_54NN
Orientia tsutsugamushi1383AP008981clade_541NOP
Megamonas funiformis1198AB300988clade_542NN
Megamonas hypermegale1199AJ420107clade_542NN
Aeromicrobium marinum102NR_025681clade_544NN
Aeromicrobium sp. JC14103JF824798clade_544NN
Luteococcus sanguinis1190NR_025507clade_544NN
Propionibacteriaceae bacterium1568EF599122clade_544NN
NML 02_0265
Rhodococcus corynebacterioides1622X80615clade_546NN
Rhodococcus erythropolis1624ACNO01000030clade_546NN
Rhodococcus fascians1625NR_037021clade_546NN
Segniliparus rotundus1690CP001958clade_546NN
Segniliparus rugosus1691ACZI01000025clade_546NN
Exiguobacterium acetylicum878FJ970034clade_547NN
Macrococcus caseolyticus1194NR_074941clade_547NN
Streptomyces sp. 1 AIP_20091890FJ176782clade_548NN
Streptomyces sp. SD 5241892EU544234clade_548NN
Streptomyces sp. SD 5281893EU544233clade_548NN
Streptomyces thermoviolaceus1895NR_027616clade_548NN
Borrelia afzelii388ABCU01000001clade_549NOP
Borrelia crocidurae390DQ057990clade_549NOP
Borrelia duttonii391NC_011229clade_549NOP
Borrelia hermsii393AY597657clade_549NOP
Borrelia hispanica394DQ057988clade_549NOP
Borrelia persica395HM161645clade_549NOP
Borrelia recurrentis396AF107367clade_549NOP
Borrelia spielmanii398ABKB01000002clade_549NOP
Borrelia turicatae399NC_008710clade_549NOP
Borrelia valaisiana400ABCY01000002clade_549NOP
Providencia alcalifaciens1586ABXW01000071clade_55NN
Providencia rettgeri1587AM040492clade_55NN
Providencia rustigianii1588AM040489clade_55NN
Providencia stuartii1589AF008581clade_55NN
Treponema pallidum1932CP001752clade_550NOP
Treponema phagedenis1934AEFH01000172clade_550NN
Treponema sp. clone DDKL_41939Y08894clade_550NN
Acholeplasma laidlawii17NR_074448clade_551NN
Mycoplasma putrefaciens1323U26055clade_551NN
Mycoplasmataceae genomosp. P11325DQ003614clade_551NN
oral clone MB1_G23
Spiroplasma insolitum1750NR_025705clade_551NN
Collinsella intestinalis660ABXH02000037clade_553NN
Collinsella stercoris661ABXJ01000150clade_553NN
Collinsella tanakaei662AB490807clade_553NN
Caminicella sporogenes458NR_025485clade_554NN
Acidaminococcus fermentans21CP001859clade_556NN
Acidaminococcus intestini22CP003058clade_556NN
Acidaminococcus sp. D2123ACGB01000071clade_556NN
Phascolarctobacterium faecium1462NR_026111clade_556NN
Phascolarctobacterium sp. YIT1463AB490812clade_556NN
12068
Phascolarctobacterium1464AB490811clade_556NN
succinatutens
Acidithiobacillus ferrivorans25NR_074660clade_557NN
Xanthomonadaceae bacterium2015EU313791clade_557NN
NML 03_0222
Catabacter hongkongensis494AB671763clade_558NN
Christensenella minuta512AB490809clade_558NN
Clostridiales bacterium oral clone536AY207065clade_558NN
P4PA_66 P1
Clostridiales bacterium oral taxon537GQ422712clade_558NN
093
Heliobacterium modesticaldum1000NR_074517clade_560NN
Alistipes indistinctus130AB490804clade_561NN
Bacteroidales bacterium ph8257JN837494clade_561NN
Candidatus Sulcia muelleri475CP002163clade_561NN
Cytophaga xylanolytica742FR733683clade_561NN
Flavobacteriaceae genomosp. C1884AY278614clade_561NN
Gramella forsetii958NR_074707clade_561NN
Sphingobacterium faecium1740NR_025537clade_562NN
Sphingobacterium mizutaii1741JF708889clade_562NN
Sphingobacterium multivorum1742NR_040953clade_562NN
Sphingobacterium spiritivorum1743ACHA02000013clade_562NN
Jonquetella anthropi1017ACOO02000004clade_563NN
Pyramidobacter piscolens1614AY207056clade_563NN
Synergistes genomosp. C11904AY278615clade_563NN
Synergistes sp. RMA 145511905DQ412722clade_563NN
Synergistetes bacterium ADV8971906GQ258968clade_563NN
Candidatus Arthromitus sp.474NR_074460clade_564NN
SFB_mouse_Yit
Gracilibacter thermotolerans957NR_043559clade_564NN
Brachyspira aalborgi404FM178386clade_565NN
Brachyspira sp. HIS3406FM178387clade_565NN
Brachyspira sp. HIS4407FM178388clade_565NN
Brachyspira sp. HIS5408FM178389clade_565NN
Adlercreutzia equolifaciens97AB306661clade_566NN
Coriobacteriaceae bacterium678CAEM01000062clade_566NN
JC110
Coriobacteriaceae bacterium phl679JN837493clade_566NN
Cryptobacterium curtum740GQ422741clade_566NN
Eggerthella sinensis779AY321958clade_566NN
Eggerthella sp. 1_3_56FAA780ACWN01000099clade_566NN
Eggerthella sp. HGA1781AEXR01000021clade_566NN
Eggerthella sp. YY7918782AP012211clade_566NN
Gordonibacter pamelaeae680AM886059clade_566NN
Gordonibacter pamelaeae956FP929047clade_566NN
Slackia equolifaciens1732EU377663clade_566NN
Slackia exigua1733ACUX01000029clade_566NN
Slackia faecicanis1734NR_042220clade_566NN
Slackia heliotrinireducens1735NR_074439clade_566NN
Slackia isoflavoniconvertens1736AB566418clade_566NN
Slackia piriformis1737AB490806clade_566NN
Slackia sp. NATTS1738AB505075clade_566NN
Chlamydiales bacterium NS13506JN606075clade_567NN
Victivallaceae bacterium NML2003FJ394915clade_567NN
080035
Victivallis vadensis2004ABDE02000010clade_567NN
Streptomyces griseus1889NR_074787clade_573NN
Streptomyces sp. SD 5111891EU544231clade_573NN
Streptomyces sp. SD 5341894EU544232clade_573NN
Cloacibacillus evryensis530GQ258966clade_575NN
Deferribacteres sp. oral clone743AY349370clade_575NN
JV001
Deferribacteres sp. oral clone745AY349372clade_575NN
JV023
Synergistetes bacterium1907GQ258969clade_575NN
LBVCM1157
Synergistetes bacterium oral taxon1909GU410752clade_575NN
362
Synergistetes bacterium oral taxon1910GU430992clade_575NN
D48
Peptococcus sp. oral clone JM0481439AY349389clade_576NN
Helicobacter winghamensis999ACDO01000013clade_577NN
Wolinella succinogenes2014BX571657clade_577NN
Olsenella genomosp. C11368AY278623clade_578NN
Olsenella profusa1369FN178466clade_578NN
Olsenella sp. F00041370EU592964clade_578NN
Olsenella sp. oral taxon 8091371ACVE01000002clade_578NN
Olsenella uli1372CP002106clade_578NN
Nocardiopsis dassonvillei1356CP002041clade_579NN
Peptococcus niger1438NR_029221clade_580NN
Peptococcus sp. oral taxon 1671440GQ422727clade_580NN
Akkermansia muciniphila118CP001071clade_583NN
Opitutus terrae1373NR_074978clade_583NN
Clostridiales bacterium oral taxon538HM099644clade_584NN
F32
Leptospira borgpetersenii1161NC_008508clade_585NOP
Leptospira broomii1162NR_043200clade_585NOP
Leptospira interrogans1163NC_005823clade_585NOP
Methanobrevibacter gottschalkii1213NR_044789clade_587NN
Methanobrevibacter millerae1214NR_042785clade_587NN
Methanobrevibacter oralis1216HE654003clade_587NN
Methanobrevibacter thaueri1219NR_044787clade_587NN
Methanobrevibacter smithii1218ABYV02000002clade_588NN
Deinococcus radiodurans746AE000513clade_589NN
Deinococcus sp. R_43890747FR682752clade_589NN
Thermus aquaticus1923NR_025900clade_589NN
Actinomyces sp. c10981AB167239clade_590NN
Syntrophomonadaceae genomosp.1912AY341821clade_590NN
P1
Anaerobaculum hydrogeniformans141ACJX02000009clade_591NN
Microcystis aeruginosa1246NC_010296clade_592NN
Prochlorococcus marinus1567CP000551clade_592NN
Methanobrevibacter acididurans1208NR_028779clade_593NN
Methanobrevibacter arboriphilus1209NR_042783clade_593NN
Methanobrevibacter curvatus1210NR_044796clade_593NN
Methanobrevibacter cuticularis1211NR_044776clade_593NN
Methanobrevibacter filiformis1212NR_044801clade_593NN
Methanobrevibacter woesei1220NR_044788clade_593NN
Roseiflexus castenholzii1642CP000804clade_594NN
Methanobrevibacter olleyae1215NR_043024clade_595NN
Methanobrevibacter ruminantium1217NR_042784clade_595NN
Methanobrevibacter wolinii1221NR_044790clade_595NN
Methanosphaera stadtmanae1222AY196684clade_595NN
Chloroflexi genomosp. P1511AY331414clade_596NN
Halorubrum lipolyticum992AB477978clade_597NN
Methanobacterium formicicum1207NR_025028clade_597NN
Acidilobus saccharovorans24AY350586clade_598NN
Hyperthermus butylicus1006CP000493clade_598NN
Ignicoccus islandicus1011X99562clade_598NN
Metallosphaera sedula1206D26491clade_598NN
Thermofilum pendens1922X14835clade_598NN
Prevotella melaninogenica1506CP002122clade_6NN
Prevotella sp. ICM11520HQ616385clade_6NN
Prevotella sp. oral clone FU0481535AY349393clade_6NN
Prevotella sp. oral clone GI0301537AY349395clade_6NN
Prevotella sp. SEQ1161526JN867246clade_6NN
Streptococcus anginosus1787AECT01000011clade_60NN
Streptococcus milleri1812X81023clade_60NN
Streptococcus sp. 163621829JN590019clade_60NN
Streptococcus sp. 691301832X78825clade_60NN
Streptococcus sp. AC151833HQ616356clade_60NN
Streptococcus sp. CM71839HQ616373clade_60NN
Streptococcus sp. OBRC61847HQ616352clade_60NN
Burkholderia ambifaria442AAUZ01000009clade_61NOP
Burkholderia cenocepacia443AAHI01000060clade_61NOP
Burkholderia cepacia444NR_041719clade_61NOP
Burkholderia mallei445CP000547clade_61NCategory-B
Burkholderia multivorans446NC_010086clade_61NOP
Burkholderia oklahomensis447DQ108388clade_61NOP
Burkholderia pseudomallei448CP001408clade_61NCategory-B
Burkholderia rhizoxinica449HQ005410clade_61NOP
Burkholderia sp. 383450CP000151clade_61NOP
Burkholderia xenovorans451U86373clade_61NOP
Prevotella buccae1488ACRB01000001clade_62NN
Prevotella genomosp. P8 oral1498DQ003622clade_62NN
clone MB3_P13
Prevotella sp. oral clone FW0351536AY349394clade_62NN
Prevotella bivia1486ADFO01000096clade_63NN
Prevotella disiens1494AEDO01000026clade_64NN
Bacteroides faecis276GQ496624clade_65NN
Bacteroides fragilis279AP006841clade_65NN
Bacteroides nordii285NR_043017clade_65NN
Bacteroides salyersiae292EU136690clade_65NN
Bacteroides sp. 1_1_14293ACRP01000155clade_65NN
Bacteroides sp. 1_1_6295ACIC01000215clade_65NN
Bacteroides sp. 2_1_56FAA298ACWI01000065clade_65NN
Bacteroides sp. AR29316AF139525clade_65NN
Bacteroides sp. B2317EU722733clade_65NN
Bacteroides thetaiotaomicron328NR_074277clade_65NN
Actinobacillus minor45ACFT01000025clade_69NN
Haemophilus parasuis978GU226366clade_69NN
Vibrio cholerae1996AAUR01000095clade_71NCategory-B
Vibrio fluvialis1997X76335clade_71NCategory-B
Vibrio furnissii1998CP002377clade_71NCategory-B
Vibrio mimicus1999ADAF01000001clade_71NCategory-B
Vibrio parahaemolyticus2000AAWQ01000116clade_71NCategory-B
Vibrio sp. RC3412001ACZT01000024clade_71NCategory-B
Vibrio vulnificus2002AE016796clade_71NCategory-B
Lactobacillus acidophilus1067CP000033clade_72NN
Lactobacillus amylolyticus1069ADNY01000006clade_72NN
Lactobacillus amylovorus1070CP002338clade_72NN
Lactobacillus crispatus1078ACOG01000151clade_72NN
Lactobacillus delbrueckii1080CP002341clade_72NN
Lactobacillus helveticus1088ACLM01000202clade_72NN
Lactobacillus kalixensis1094NR_029083clade_72NN
Lactobacillus kefiranofaciens1095NR_042440clade_72NN
Lactobacillus leichmannii1098JX986966clade_72NN
Lactobacillus sp. 66c1120FR681900clade_72NN
Lactobacillus sp. KLDS 1.07011122EU600905clade_72NN
Lactobacillus sp. KLDS 1.07121130EU600916clade_72NN
Lactobacillus sp. oral clone HT0701136AY349383clade_72NN
Lactobacillus ultunensis1139ACGU01000081clade_72NN
Prevotella intermedia1502AF414829clade_81NN
Prevotella nigrescens1511AFPX01000069clade_81NN
Prevotella pallens1515AFPY01000135clade_81NN
Prevotella sp. oral taxon 3101551GQ422737clade_81NN
Prevotella genomosp. C11495AY278624clade_82NN
Prevotella sp. CM381519HQ610181clade_82NN
Prevotella sp. oral taxon 3171552ACQH01000158clade_82NN
Prevotella sp. SG121527GU561343clade_82NN
Prevotella denticola1493CP002589clade_83NN
Prevotella genomosp. P7 oral1497DQ003620clade_83NN
clone MB2_P31
Prevotella histicola1501JN867315clade_83NN
Prevotella multiformis1508AEWX01000054clade_83NN
Prevotella sp. JCM 63301522AB547699clade_83NN
Prevotella sp. oral clone GI0591539AY349397clade_83NN
Prevotella sp. oral taxon 7821555GQ422745clade_83NN
Prevotella sp. oral taxon G711559GU432180clade_83NN
Prevotella sp. SEQ0651524JN867234clade_83NN
Prevotella veroralis1565ACVA01000027clade_83NN
Bacteroides acidifaciens266NR_028607clade_85NN
Bacteroides cellulosilyticus269ACCH01000108clade_85NN
Bacteroides clarus270AFBM01000011clade_85NN
Bacteroides eggerthii275ACWG01000065clade_85NN
Bacteroides oleiciplenus286AB547644clade_85NN
Bacteroides pyogenes290NR_041280clade_85NN
Bacteroides sp. 315_5300FJ848547clade_85NN
Bacteroides sp. 31SF15301AJ583248clade_85NN
Bacteroides sp. 31SF18302AJ583249clade_85NN
Bacteroides sp. 35AE31303AJ583244clade_85NN
Bacteroides sp. 35AE37304AJ583245clade_85NN
Bacteroides sp. 35BE34305AJ583246clade_85NN
Bacteroides sp. 35BE35306AJ583247clade_85NN
Bacteroides sp. WH2324AY895180clade_85NN
Bacteroides sp. XB12B325AM230648clade_85NN
Bacteroides stercoris327ABFZ02000022clade_85NN
Actinobacillus pleuropneumoniae46NR_074857clade_88NN
Actinobacillus ureae48AEVG01000167clade_88NN
Haemophilus aegyptius969AFBC01000053clade_88NN
Haemophilus ducreyi970AE017143clade_88NOP
Haemophilus haemolyticus973JN175335clade_88NN
Haemophilus influenzae974AADP01000001clade_88NOP
Haemophilus parahaemolyticus975GU561425clade_88NN
Haemophilus parainfluenzae976AEWU01000024clade_88NN
Haemophilus977M75076clade_88NN
paraphrophaemolyticus
Haemophilus somnus979NC_008309clade_88NN
Haemophilus sp. 70334980HQ680854clade_88NN
Haemophilus sp. HK445981FJ685624clade_88NN
Haemophilus sp. oral clone982AY923117clade_88NN
ASCA07
Haemophilus sp. oral clone983AY923147clade_88NN
ASCG06
Haemophilus sp. oral clone BJ021984AY005034clade_88NN
Haemophilus sp. oral clone BJ095985AY005033clade_88NN
Haemophilus sp. oral taxon 851987AGRK01000004clade_88NN
Haemophilus sputorum988AFNK01000005clade_88NN
Histophilus somni1003AF549387clade_88NN
Mannheimia haemolytica1195ACZX01000102clade_88NN
Pasteurella bettyae1433L06088clade_88NN
Moellerella wisconsensis1253JN175344clade_89NN
Morganella morganii1265AJ301681clade_89NN
Morganella sp. JB_T161266AJ781005clade_89NN
Proteus mirabilis1582ACLE01000013clade_89NN
Proteus penneri1583ABVP01000020clade_89NN
Proteus sp. HS75141584DQ512963clade_89NN
Proteus vulgaris1585AJ233425clade_89NN
Oribacterium sinus1374ACKX01000142clade_90NN
Oribacterium sp. ACB11375HM120210clade_90NN
Oribacterium sp. ACB71376HM120211clade_90NN
Oribacterium sp. CM121377HQ616374clade_90NN
Oribacterium sp. ICM511378HQ616397clade_90NN
Oribacterium sp. OBRC121379HQ616355clade_90NN
Oribacterium sp. oral taxon 1081382AFIH01000001clade_90NN
Actinobacillus44AY362885clade_92NN
actinomycetemcomitans
Actinobacillus succinogenes47CP000746clade_92NN
Aggregatibacter112CP001733clade_92NN
actinomycetemcomitans
Aggregatibacter aphrophilus113CP001607clade_92NN
Aggregatibacter segnis114AEPS01000017clade_92NN
Averyella dalhousiensis194DQ481464clade_92NN
Bisgaard Taxon368AY683487clade_92NN
Bisgaard Taxon369AY683489clade_92NN
Bisgaard Taxon370AY683491clade_92NN
Bisgaard Taxon371AY683492clade_92NN
Buchnera aphidicola440NR_074609clade_92NN
Cedecea davisae499AF493976clade_92NN
Citrobacter amalonaticus517FR870441clade_92NN
Citrobacter braakii518NR_028687clade_92NN
Citrobacter farmeri519AF025371clade_92NN
Citrobacter freundii520NR_028894clade_92NN
Citrobacter gillenii521AF025367clade_92NN
Citrobacter koseri522NC_009792clade_92NN
Citrobacter murliniae523AF025369clade_92NN
Citrobacter rodentium524NR_074903clade_92NN
Citrobacter sedlakii525AF025364clade_92NN
Citrobacter sp. 30_2526ACDJ01000053clade_92NN
Citrobacter sp. KMSI_3527GQ468398clade_92NN
Citrobacter werkmanii528AF025373clade_92NN
Citrobacter youngae529ABWL02000011clade_92NN
Cronobacter malonaticus737GU122174clade_92NN
Cronobacter sakazakii738NC_009778clade_92NN
Cronobacter turicensis739FN543093clade_92NN
Enterobacter aerogenes786AJ251468clade_92NN
Enterobacter asburiae787NR_024640clade_92NN
Enterobacter cancerogenus788Z96078clade_92NN
Enterobacter cloacae789FP929040clade_92NN
Enterobacter cowanii790NR_025566clade_92NN
Enterobacter hormaechei791AFHR01000079clade_92NN
Enterobacter sp. 247BMC792HQ122932clade_92NN
Enterobacter sp. 638793NR_074777clade_92NN
Enterobacter sp. JC163794JN657217clade_92NN
Enterobacter sp. SCSS795HM007811clade_92NN
Enterobacter sp. TSE38796HM156134clade_92NN
Enterobacteriaceae bacterium797ADCU01000033clade_92NN
9_2_54FAA
Enterobacteriaceae bacterium798AJ489826clade_92NN
CF01Ent_1
Enterobacteriaceae bacterium799AY538694clade_92NN
Smarlab 3302238
Escherichia albertii824ABKX01000012clade_92NN
Escherichia coli825NC_008563clade_92NCategory-B
Escherichia fergusonii826CU928158clade_92NN
Escherichia hermannii827HQ407266clade_92NN
Escherichia sp. 1_1_43828ACID01000033clade_92NN
Escherichia sp. 4_1_40B829ACDM02000056clade_92NN
Escherichia sp. B4830EU722735clade_92NN
Escherichia vulneris831NR_041927clade_92NN
Ewingella americana877JN175329clade_92NN
Haemophilus genomosp. P2 oral971DQ003621clade_92NN
clone MB3_C24
Haemophilus genomosp. P3 oral972DQ003635clade_92NN
clone MB3_C38
Haemophilus sp. oral clone JM053986AY349380clade_92NN
Hafnia alvei989DQ412565clade_92NN
Klebsiella oxytoca1024AY292871clade_92NOP
Klebsiella pneumoniae1025CP000647clade_92NOP
Klebsiella sp. AS101026HQ616362clade_92NN
Klebsiella sp. Co99351027DQ068764clade_92NN
Klebsiella sp. enrichment culture1036HM195210clade_92NN
clone SRC_DSD25
Klebsiella sp. OBRC71028HQ616353clade_92NN
Klebsiella sp. SP_BA1029FJ999767clade_92NN
Klebsiella sp. SRC_DSD11033GU797254clade_92NN
Klebsiella sp. SRC_DSD111030GU797263clade_92NN
Klebsiella sp. SRC_DSD121031GU797264clade_92NN
Klebsiella sp. SRC_DSD151032GU797267clade_92NN
Klebsiella sp. SRC_DSD21034GU797253clade_92NN
Klebsiella sp. SRC_DSD61035GU797258clade_92NN
Klebsiella variicola1037CP001891clade_92NN
Kluyvera ascorbata1038NR_028677clade_92NN
Kluyvera cryocrescens1039NR_028803clade_92NN
Leminorella grimontii1159AJ233421clade_92NN
Leminorella richardii1160HF558368clade_92NN
Pantoea agglomerans1409AY335552clade_92NN
Pantoea ananatis1410CP001875clade_92NN
Pantoea brenneri1411EU216735clade_92NN
Pantoea citrea1412EF688008clade_92NN
Pantoea conspicua1413EU216737clade_92NN
Pantoea septica1414EU216734clade_92NN
Pasteurella dagmatis1434ACZR01000003clade_92NN
Pasteurella multocida1435NC_002663clade_92NN
Plesiomonas shigelloides1469X60418clade_92NN
Raoultella ornithinolytica1617AB364958clade_92NN
Raoultella planticola1618AF129443clade_92NN
Raoultella terrigena1619NR_037085clade_92NN
Salmonella bongori1683NR_041699clade_92NCategory-B
Salmonella enterica1672NC_011149clade_92NCategory-B
Salmonella enterica1673NC_011205clade_92NCategory-B
Salmonella enterica1674DQ344532clade_92NCategory-B
Salmonella enterica1675ABEH02000004clade_92NCategory-B
Salmonella enterica1676ABAK02000001clade_92NCategory-B
Salmonella enterica1677NC_011080clade_92NCategory-B
Salmonella enterica1678EU118094clade_92NCategory-B
Salmonella enterica1679NC_011094clade_92NCategory-B
Salmonella enterica1680AE014613clade_92NCategory-B
Salmonella enterica1682ABFH02000001clade_92NCategory-B
Salmonella enterica1684ABEM01000001clade_92NCategory-B
Salmonella enterica1685ABAM02000001clade_92NCategory-B
Salmonella typhimurium1681DQ344533clade_92NCategory-B
Salmonella typhimurium1686AF170176clade_92NCategory-B
Serratia fonticola1718NR_025339clade_92NN
Serratia liquefaciens1719NR_042062clade_92NN
Serratia marcescens1720GU826157clade_92NN
Serratia odorifera1721ADBY01000001clade_92NN
Serratia proteamaculans1722AAUN01000015clade_92NN
Shigella boydii1724AAKA01000007clade_92NCategory-B
Shigella dysenteriae1725NC_007606clade_92NCategory-B
Shigella flexneri1726AE005674clade_92NCategory-B
Shigella sonnei1727NC_007384clade_92NCategory-B
Tatumella ptyseos1916NR_025342clade_92NN
Trabulsiella guamensis1925AY373830clade_92NN
Yersinia aldovae2019AJ871363clade_92NOP
Yersinia aleksiciae2020AJ627597clade_92NOP
Yersinia bercovieri2021AF366377clade_92NOP
Yersinia enterocolitica2022FR729477clade_92NCategory-B
Yersinia frederiksenii2023AF366379clade_92NOP
Yersinia intermedia2024AF366380clade_92NOP
Yersinia kristensenii2025ACCA01000078clade_92NOP
Yersinia mollaretii2026NR_027546clade_92NOP
Yersinia pestis2027AE013632clade_92NCategory-A
Yersinia pseudotuberculosis2028NC_009708clade_92NOP
Yersinia rohdei2029ACCD01000071clade_92NOP
Yokenella regensburgei2030AB273739clade_92NN
Conchiformibius kuhniae669NR_041821clade_94NN
Morococcus cerebrosus1267JN175352clade_94NN
Neisseria bacilliformis1328AFAY01000058clade_94NN
Neisseria cinerea1329ACDY01000037clade_94NN
Neisseria flavescens1331ACQV01000025clade_94NN
Neisseria gonorrhoeae1333CP002440clade_94NOP
Neisseria lactamica1334ACEQ01000095clade_94NN
Neisseria macacae1335AFQE01000146clade_94NN
Neisseria meningitidis1336NC_003112clade_94NOP
Neisseria mucosa1337ACDX01000110clade_94NN
Neisseria pharyngis1338AJ239281clade_94NN
Neisseria polysaccharea1339ADBE01000137clade_94NN
Neisseria sicca1340ACKO02000016clade_94NN
Neisseria sp. KEM2321341GQ203291clade_94NN
Neisseria sp. oral clone AP1321344AY005027clade_94NN
Neisseria sp. oral strain B33KA1346AY005028clade_94NN
Neisseria sp. oral taxon 0141347ADEA01000039clade_94NN
Neisseria sp. TM10_11343DQ279352clade_94NN
Neisseria subflava1348ACEO01000067clade_94NN
Okadaella gastrococcus1365HQ699465clade_98NN
Streptococcus agalactiae1785AAJO01000130clade_98NN
Streptococcus alactolyticus1786NR_041781clade_98NN
Streptococcus australis1788AEQR01000024clade_98NN
Streptococcus bovis1789AEEL01000030clade_98NN
Streptococcus canis1790AJ413203clade_98NN
Streptococcus constellatus1791AY277942clade_98NN
Streptococcus cristatus1792AEVC01000028clade_98NN
Streptococcus dysgalactiae1794AP010935clade_98NN
Streptococcus equi1795CP001129clade_98NN
Streptococcus equinus1796AEVB01000043clade_98NN
Streptococcus gallolyticus1797FR824043clade_98NN
Streptococcus genomosp. C11798AY278629clade_98NN
Streptococcus genomosp. C21799AY278630clade_98NN
Streptococcus genomosp. C31800AY278631clade_98NN
Streptococcus genomosp. C41801AY278632clade_98NN
Streptococcus genomosp. C51802AY278633clade_98NN
Streptococcus genomosp. C61803AY278634clade_98NN
Streptococcus genomosp. C71804AY278635clade_98NN
Streptococcus genomosp. C81805AY278609clade_98NN
Streptococcus gordonii1806NC_009785clade_98NN
Streptococcus infantarius1807ABJK02000017clade_98NN
Streptococcus infantis1808AFNN01000024clade_98NN
Streptococcus intermedius1809NR_028736clade_98NN
Streptococcus lutetiensis1810NR_037096clade_98NN
Streptococcus massiliensis1811AY769997clade_98NN
Streptococcus mitis1813AM157420clade_98NN
Streptococcus oligofermentans1815AY099095clade_98NN
Streptococcus oralis1816ADMV01000001clade_98NN
Streptococcus parasanguinis1817AEKM01000012clade_98NN
Streptococcus pasteurianus1818AP012054clade_98NN
Streptococcus peroris1819AEVF01000016clade_98NN
Streptococcus pneumoniae1820AE008537clade_98NN
Streptococcus porcinus1821EF121439clade_98NN
Streptococcus pseudopneumoniae1822FJ827123clade_98NN
Streptococcus pseudoporcinus1823AENS01000003clade_98NN
Streptococcus pyogenes1824AE006496clade_98NOP
Streptococcus ratti1825X58304clade_98NN
Streptococcus sanguinis1827NR_074974clade_98NN
Streptococcus sinensis1828AF432857clade_98NN
Streptococcus sp. 2_1_36FAA1831ACOI01000028clade_98NN
Streptococcus sp. 2285_971830AJ131965clade_98NN
Streptococcus sp. ACS21834HQ616360clade_98NN
Streptococcus sp. AS201835HQ616366clade_98NN
Streptococcus sp. BS35a1836HQ616369clade_98NN
Streptococcus sp. C1501837ACRI01000045clade_98NN
Streptococcus sp. CM61838HQ616372clade_98NN
Streptococcus sp. ICM101840HQ616389clade_98NN
Streptococcus sp. ICM121841HQ616390clade_98NN
Streptococcus sp. ICM21842HQ616386clade_98NN
Streptococcus sp. ICM41844HQ616387clade_98NN
Streptococcus sp. ICM451843HQ616394clade_98NN
Streptococcus sp. M1431845ACRK01000025clade_98NN
Streptococcus sp. M3341846ACRL01000052clade_98NN
Streptococcus sp. oral clone1849AY923121clade_98NN
ASB02
Streptococcus sp. oral clone1850DQ272504clade_98NN
ASCA03
Streptococcus sp. oral clone1851AY923116clade_98NN
ASCA04
Streptococcus sp. oral clone1852AY923119clade_98NN
ASCA09
Streptococcus sp. oral clone1853AY923123clade_98NN
ASCB04
Streptococcus sp. oral clone1854AY923124clade_98NN
ASCB06
Streptococcus sp. oral clone1855AY923127clade_98NN
ASCC04
Streptococcus sp. oral clone1856AY923128clade_98NN
ASCC05
Streptococcus sp. oral clone1857DQ272507clade_98NN
ASCC12
Streptococcus sp. oral clone1858AY923129clade_98NN
ASCD01
Streptococcus sp. oral clone1859AY923130clade_98NN
ASCD09
Streptococcus sp. oral clone1860DQ272509clade_98NN
ASCD10
Streptococcus sp. oral clone1861AY923134clade_98NN
ASCE03
Streptococcus sp. oral clone1862AY953253clade_98NN
ASCE04
Streptococcus sp. oral clone1863DQ272510clade_98NN
ASCE05
Streptococcus sp. oral clone1864AY923135clade_98NN
ASCE06
Streptococcus sp. oral clone1865AY923136clade_98NN
ASCE09
Streptococcus sp. oral clone1866AY923137clade_98NN
ASCE10
Streptococcus sp. oral clone1867AY923138clade_98NN
ASCE12
Streptococcus sp. oral clone1868AY923140clade_98NN
ASCF05
Streptococcus sp. oral clone1869AY953255clade_98NN
ASCF07
Streptococcus sp. oral clone1870AY923142clade_98NN
ASCF09
Streptococcus sp. oral clone1871AY923145clade_98NN
ASCG04
Streptococcus sp. oral clone1872AY005042clade_98NN
BW009
Streptococcus sp. oral clone1873AY005044clade_98NN
CH016
Streptococcus sp. oral clone1874AY349413clade_98NN
GK051
Streptococcus sp. oral clone1875AY349414clade_98NN
GM006
Streptococcus sp. oral clone1876AY207051clade_98NN
P2PA_41 P2
Streptococcus sp. oral clone1877AY207064clade_98NN
P4PA_30 P4
Streptococcus sp. oral taxon 0711878AEEP01000019clade_98NN
Streptococcus sp. oral taxon G591879GU432132clade_98NN
Streptococcus sp. oral taxon G621880GU432146clade_98NN
Streptococcus sp. oral taxon G631881GU432150clade_98NN
Streptococcus suis1882FM252032clade_98NN
Streptococcus thermophilus1883CP000419clade_98NN
Streptococcus salivarius1826AGBV01000001clade_98NN
Streptococcus uberis1884HQ391900clade_98NN
Streptococcus urinalis1885DQ303194clade_98NN
Streptococcus vestibularis1886AEKO01000008clade_98NN
Streptococcus viridans1887AF076036clade_98NN
Synergistetes bacterium oral clone1908GU227192clade_98NN
03 5 D05
Clade membership of bacterial OTUs is based on 16S sequence data. Clades are defined based on the topology of a phylogenetic tree that is constructed from full-length 16S sequences using maximum likelihood methods familiar to individuals with ordinary skill in the art of phylogenetics. Clades are constructed to ensure that all OTUs in a given clade are: (i) within a specified number of bootstrap supported nodes from one another, and (ii) within 5% genetic similarity. OTUs that are within the same clade can be distinguished as genetically and phylogenetically distinct from OTUs in a different clade based on 16S-V4 sequence data, while OTUs falling within the same clade are closely related. OTUs falling within the same clade are evolutionarily closely related and may or may not be distinguishable from one another using 16S-V4 sequence data. Members of the same clade, due to their evolutionary relatedness, play similar functional roles in a microbial ecology such as that found in the human gut. Compositions substituting one species with another from the same clade are likely to have conserved ecological function and therefore are useful in the present invention. All OTUs are denoted as to their putative capacity to form spores and whether they are a Pathogen or Pathobiont (see Definitions for description of “Pathobiont”). NIAID Priority Pathogens are denoted as ‘Category-A’, ‘Category-B’, or ‘Category-C’, and Opportunistic Pathogens are denoted as ‘OP’. OTUs that are not pathogenic or for which their ability to exist as a pathogen is unknown are denoted as ‘N’. The ‘SEQ ID Number’ denotes the identifier of the OTU in the Sequence Listing File and ‘Public DB Accession’ denotes the identifier of the OTU in a public sequence repository.

[0000]

Mouse studies with bacterial compositions
MortalityMorbiditySummary
ScoreD3 Weight Score>3 ScoreScore
Treatment 133.03.09
Treatment 200.60.01
Treatment 30−0.50.0−1
Treatment 40.52.40.63
Treatment 500.50.00
Treatment 612.00.64
Treatment 70.50.70.42
Treatment 800.60.01
Treatment 90.51.30.62
Treatment 100.51.50.63
Treatment 110.51.00.62
Treatment 1201.30.01
Treatment 130.51.30.42
Treatment 140.51.20.62

[0000]

Table 3A. Mouse studies with bacterial compositions
MortalityMorbiditySummary
ScoreD3 Weight Score>3 ScoreScore
Treatment 133.03.09
Treatment 200.00.00
Treatment 332.12.78
Treatment 41.51.11.14
Treatment 500.70.21
Treatment 601.20.01
Treatment 732.42.18
Treatment 800.80.61
Treatment 900.90.31
Treatment 101.52.52.77
Treatment 1101.30.82
Treatment 1200.10.00
Treatment 1331.60.35
Treatment 1400.42.33
Treatment 151.53.42.67

[0000]

Table 3B. Mouse studies with bacterial compositions
MortalityMorbiditySummary
ScoreD3 Weight Score>3 ScoreScore
Treatment 133.03.09
Treatment 21.80.72.15
Treatment 300.00.00
Treatment 400.50.00
Treatment 500.70.01
Treatment 60.62.30.74
Treatment 70.61.70.63
Treatment 82.43.02.07
Treatment 90.61.40.63
Treatment 1001.10.01
Treatment 1101.70.62
Treatment 1233.13.29

[0000]

Table 4: Species isolated from ethanol treated spore preparation
preparation before (left) and after (right) CsCl gradient step
ethanol treated,
ethanol treatedgradient purified
Isolatesspore preparationspore preparation
Bacillus coagulans72
Blautia luti11
Blautia sp1413
Blautia wexlerae31
Ruminococcus obeum42
Clostridiales sp12
Clostridium aerotolerans12
Clostridium disporicum01
Clostridium sp11
Clostridium symbiosum01
Dorea longicatena86
Eubacterium cellulosolvens10
Eubacterium ventriosum22
Gemmiger formicilis01
Robinsoniella peoriensis01
Roseburia hominis36
Roseburia intestinalis97
Ruminococcus sp52
Syntrophococcus11
sucromutans
Turicibacter sanguinis34
Clostridiales sp79
Clostridium bartlettii811
Clostridium irregulare01
Clostridium sordellii46
Lachnospiraceae sp10

[0000]

Mortality and weight change in mice challenged with C. difficile
with or without ethanol treated, spore preparation treatment.
% weight
mortalitychange on
Test article(n = 10)Day 3
vehicle20%−10.5% 
(negative
control)
Donor feces0−0.1% 
(positive control)
EtOH-treated02.3%
spore
preparation 1x
EtOH-treated02.4%
spore
preparation 0.1x
EtOH-treated0 −3%
spore
preparation
0.01x
heat-treated00.1%
spore
preparation

[0000]

Preparation 14.0 × 1056.8 × 1075.9 × 10−3
Preparation 22.1 × 1079.2 × 1080.023
Preparation 36.9 × 1099.6 × 1090.72

[0328]

Table 6 shows spore quantitation for ethanol treated spore preparations using spore CFU (SCFU) assay and DPA assay.

[0000]

DPA doses in Table 7 when normalized to 4 × 105SCFU per dose
Fraction of
SCFU/30DPA SEq/30Preparation 1
PreparationcapsulescapsulesDose
Preparation 14.0 × 1056.8 × 1071.0
Preparation 24.0 × 1051.8 × 1070.26
Preparation 34.0 × 1055.6 × 1050.0082

[0000]

SPC10325SPC10415SPC10567Clostridium_bolteaeBlautia productaEubacterium_rectale++++
SPC10325SPC10355SPC10415Clostridium_bolteaeClostridium_symbiosumBlautia producta++++
SPC10325SPC10355SPC10567Clostridium_bolteaeClostridium_symbiosumEubacterium_rectale
SPC10325SPC10355SPC10386Clostridium_bolteaeClostridium_symbiosumFaecalibacterium_prausnitzii
SPC10325SPC10355SPC10390Clostridium_bolteaeClostridium_symbiosumLachnospiraceae_bacterium
5_1_57FAA
SPC10325SPC10386SPC10415Clostridium_bolteaeFaecalibacterium_prausnitziiBlautia producta++++
SPC10325SPC10386SPC10567Clostridium_bolteaeFaecalibacterium_prausnitziiEubacterium_rectale
SPC10325SPC10386SPC10390Clostridium_bolteaeFaecalibacterium_prausnitziiLachnospiraceae_bacterium++++
5_1_57FAA
SPC10325SPC10390SPC10415Clostridium_bolteaeLachnospiraceae_bacteriumBlautia producta++++
5_1_57FAA
SPC10325SPC10390SPC10567Clostridium_bolteaeLachnospiraceae_bacteriumEubacterium_rectale+
5_1_57FAA
SPC10355SPC10415SPC10567Clostridium_symbiosumBlautia productaEubacterium_rectale++++
SPC10355SPC10386SPC10415Clostridium_symbiosumFaecalibacterium_prausnitziiBlautia producta++++
SPC10355SPC10386SPC10567Clostridium_symbiosumFaecalibacterium_prausnitziiEubacterium_rectale
SPC10355SPC10386SPC10390Clostridium_symbiosumFaecalibacterium_prausnitziiLachnospiraceae_bacterium+
5_1_57FAA
SPC10355SPC10390SPC10415Clostridium_symbiosumLachnospiraceae_bacteriumBlautia producta++++
5_1_57FAA
SPC10355SPC10390SPC10567Clostridium_symbiosumLachnospiraceae_bacteriumEubacterium_rectale
5_1_57FAA
SPC10097SPC10415SPC10567Collinsella_aerofaciensBlautia productaEubacterium_rectale++++
SPC10097SPC10325SPC10415Collinsella_aerofaciensClostridium_bolteaeBlautia producta++++
SPC10097SPC10325SPC10355Collinsella_aerofaciensClostridium_bolteaeClostridium_symbiosum++++
SPC10097SPC10325SPC10567Collinsella_aerofaciensClostridium_bolteaeEubacterium_rectale++++
SPC10097SPC10325SPC10386Collinsella_aerofaciensClostridium_bolteaeFaecalibacterium_prausnitzii++++
SPC10097SPC10325SPC10390Collinsella_aerofaciensClostridium_bolteaeLachnospiraceae_bacterium++++
5_1_57FAA
SPC10097SPC10355SPC10415Collinsella_aerofaciensClostridium_symbiosumBlautia producta++++
SPC10097SPC10355SPC10567Collinsella_aerofaciensClostridium_symbiosumEubacterium_rectale
SPC10097SPC10355SPC10386Collinsella_aerofaciensClostridium_symbiosumFaecalibacterium_prausnitzii
SPC10097SPC10355SPC10390Collinsella_aerofaciensClostridium_symbiosumLachnospiraceae_bacterium+
5_1_57FAA
SPC10097SPC10304SPC10415Collinsella_aerofaciensCoprococcus_comesBlautia producta++++
SPC10097SPC10304SPC10325Collinsella_aerofaciensCoprococcus_comesClostridium_bolteae++++
SPC10097SPC10304SPC10355Collinsella_aerofaciensCoprococcus_comesClostridium_symbiosum+++
SPC10097SPC10304SPC10567Collinsella_aerofaciensCoprococcus_comesEubacterium_rectale+++
SPC10097SPC10304SPC10386Collinsella_aerofaciensCoprococcus_comesFaecalibacterium_prausnitzii++++
SPC10097SPC10304SPC10390Collinsella_aerofaciensCoprococcus_comesLachnospiraceae_bacterium+++
5_1_57FAA
SPC10097SPC10386SPC10415Collinsella_aerofaciensFaecalibacterium_prausnitziiBlautia producta++++
SPC10097SPC10386SPC10567Collinsella_aerofaciensFaecalibacterium_prausnitziiEubacterium_rectale+++
SPC10097SPC10386SPC10390Collinsella_aerofaciensFaecalibacterium_prausnitziiLachnospiraceae_bacterium+++
5_1_57FAA
SPC10097SPC10390SPC10415Collinsella_aerofaciensLachnospiraceae_bacteriumBlautia producta++++
5_1_57FAA
SPC10097SPC10390SPC10567Collinsella_aerofaciensLachnospiraceae_bacteriumEubacterium_rectale++++
5_1_57FAA
SPC10304SPC10415SPC10567Coprococcus_comesBlautia productaEubacterium_rectale++++
SPC10304SPC10325SPC10415Coprococcus_comesClostridium_bolteaeBlautia producta++++
SPC10304SPC10325SPC10355Coprococcus_comesClostridium_bolteaeClostridium_symbiosum
SPC10304SPC10325SPC10567Coprococcus_comesClostridium_bolteaeEubacterium_rectale−−
SPC10304SPC10325SPC10386Coprococcus_comesClostridium_bolteaeFaecalibacterium_prausnitzii+++
SPC10304SPC10325SPC10390Coprococcus_comesClostridium_bolteaeLachnospiraceae_bacterium+++
5_1_57FAA
SPC10304SPC10355SPC10415Coprococcus_comesClostridium_symbiosumBlautia producta++++
SPC10304SPC10355SPC10567Coprococcus_comesClostridium_symbiosumEubacterium_rectale−−−
SPC10304SPC10355SPC10386Coprococcus_comesClostridium_symbiosumFaecalibacterium_prausnitzii
SPC10304SPC10355SPC10390Coprococcus_comesClostridium_symbiosumLachnospiraceae_bacterium
5_1_57FAA
SPC10304SPC10386SPC10415Coprococcus_comesFaecalibacterium_prausnitziiBlautia producta++++
SPC10304SPC10386SPC10567Coprococcus_comesFaecalibacterium_prausnitziiEubacterium_rectale
SPC10304SPC10386SPC10390Coprococcus_comesFaecalibacterium_prausnitziiLachnospiraceae_bacterium
5_1_57FAA
SPC10304SPC10390SPC10415Coprococcus_comesLachnospiraceae_bacteriumBlautia producta++++
5_1_57FAA
SPC10304SPC10390SPC10567Coprococcus_comesLachnospiraceae_bacteriumEubacterium_rectale
5_1_57FAA
SPC10386SPC10415SPC10567Faecalibacterium_prausnitziiBlautia productaEubacterium_rectale++++
SPC10386SPC10390SPC10415Faecalibacterium_prausnitziiLachnospiraceae_bacteriumBlautia producta++++
5_1_57FAA
SPC10386SPC10390SPC10567Faecalibacterium_prausnitziiLachnospiraceae_bacteriumEubacterium_rectale
5_1_57FAA
SPC10390SPC10415SPC10567Lachnospiraceae_bacteriumBlautia productaEubacterium_rectale++++
5_1_57FAA

[0000]

SPC10414SPC10414Alistipes_shahiiAlistipes_shahii
SPC10211SPC10414Bacteroides_caccaeAlistipes_shahii
SPC10213SPC10414Bacteroides_eggerthiiAlistipes_shahii−−
SPC10030SPC10414Bacteroides_ovatusAlistipes_shahii
SPC00006SPC10414Bacteroides_sp_1_1_6Alistipes_shahii++++
SPC00007SPC10414Bacteroides_sp_3_1_23Alistipes_shahii+
SPC10019SPC10414Bacteroides_sp_D20Alistipes_shahii
SPC00005SPC10414Bacteroides_vulgatusAlistipes_shahii+++
SPC10081SPC10414Bacteroides_vulgatusAlistipes_shahii+
SPC10301SPC10414Bifidobacterium_adolescentisAlistipes_shahii++++
SPC10298SPC10414Bifidobacterium_pseudocatenulatumAlistipes_shahii
SPC00021SPC10414Blautia_productaAlistipes_shahii++++
SPC10403SPC10414Blautia_schinkiiAlistipes_shahii
SPC10243SPC10414Clostridium_hathewayiAlistipes_shahii++++
SPC00026SPC10414Clostridium_nexileAlistipes_shahii
SPC00027SPC10414Clostridium_sp_HGF2Alistipes_shahii
SPC10355SPC10414Clostridium_symbiosumAlistipes_shahii
SPC10097SPC10414Collinsella_aerofaciensAlistipes_shahii++++
SPC00009SPC10414Coprobacillus_sp_D7Alistipes_shahii++++
SPC00080SPC10414Coprococcus_catusAlistipes_shahii
SPC10304SPC10414Coprococcus_comesAlistipes_shahii
SPC00018SPC10414Dorea_formicigeneransAlistipes_shahii−−−−
SPC00057SPC10414Dorea_longicatenaAlistipes_shahii++++
SPC00008SPC10414Enterococcus_faecalisAlistipes_shahii++++
SPC10001SPC10414Erysipelotrichaceae_bacteriumAlistipes_shahii−−−
SPC00001SPC10414Escherichia_coliAlistipes_shahii++++
SPC10110SPC10414Escherichia_coliAlistipes_shahii++++
SPC00022SPC10414Eubacterium_eligensAlistipes_shahii−−
SPC10363SPC10414Eubacterium_rectaleAlistipes_shahii
SPC00054SPC10414Faecalibacterium_prausnitziiAlistipes_shahii
SPC10386SPC10414Faecalibacterium_prausnitziiAlistipes_shahii+
SPC10390SPC10414Lachnospiraceae_bacterium_5_1_57FAAAlistipes_shahii
SPC00056SPC10414Odoribacter_splanchnicusAlistipes_shahii
SPC10388SPC10414Odoribacter_splanchnicusAlistipes_shahii
SPC10048SPC10414Parabacteroides_merdaeAlistipes_shahii
SPC00061SPC10414Roseburia_intestinalisAlistipes_shahii
SPC10197SPC10414Ruminococcus_obeumAlistipes_shahii
SPC10233SPC10414Ruminococcus_torquesAlistipes_shahii
SPC00015SPC10414Streptococcus_thermophilusAlistipes_shahii
SPC10211SPC10211Bacteroides_caccaeBacteroides_caccae++++
SPC10030SPC10211Bacteroides_ovatusBacteroides_caccae
SPC00006SPC10211Bacteroides_sp_1_1_6Bacteroides_caccae++++
SPC00007SPC10211Bacteroides_sp_3_1_23Bacteroides_caccae+++
SPC10019SPC10211Bacteroides_sp_D20Bacteroides_caccae+++
SPC00005SPC10211Bacteroides_vulgatusBacteroides_caccae++++
SPC10081SPC10211Bacteroides_vulgatusBacteroides_caccae+
SPC00021SPC10211Blautia_productaBacteroides_caccae++++
SPC00026SPC10211Clostridium_nexileBacteroides_caccae
SPC00027SPC10211Clostridium_sp_HGF2Bacteroides_caccae
SPC10097SPC10211Collinsella_aerofaciensBacteroides_caccae++++
SPC00009SPC10211Coprobacillus_sp_D7Bacteroides_caccae+++
SPC00080SPC10211Coprococcus_catusBacteroides_caccae++++
SPC00018SPC10211Dorea_formicigeneransBacteroides_caccae+++
SPC00057SPC10211Dorea_longicatenaBacteroides_caccae
SPC00008SPC10211Enterococcus_faecalisBacteroides_caccae++++
SPC10001SPC10211Erysipelotrichaceae_bacteriumBacteroides_caccae++
SPC00001SPC10211Escherichia_coliBacteroides_caccae++++
SPC10110SPC10211Escherichia_coliBacteroides_caccae++++
SPC00022SPC10211Eubacterium_eligensBacteroides_caccae++
SPC00054SPC10211Faecalibacterium_prausnitziiBacteroides_caccae
SPC00056SPC10211Odoribacter_splanchnicusBacteroides_caccae
SPC10048SPC10211Parabacteroides_merdaeBacteroides_caccae+
SPC00061SPC10211Roseburia_intestinalisBacteroides_caccae+
SPC10197SPC10211Ruminococcus_obeumBacteroides_caccae++++
SPC00015SPC10211Streptococcus_thermophilusBacteroides_caccae++
SPC10211SPC10213Bacteroides_caccaeBacteroides_eggerthii++++
SPC10213SPC10213Bacteroides_eggerthiiBacteroides_eggerthii++++
SPC10030SPC10213Bacteroides_ovatusBacteroides_eggerthii
SPC00006SPC10213Bacteroides_sp_1_1_6Bacteroides_eggerthii+++
SPC00007SPC10213Bacteroides_sp_3_1_23Bacteroides_eggerthii++
SPC10019SPC10213Bacteroides_sp_D20Bacteroides_eggerthii
SPC00005SPC10213Bacteroides_vulgatusBacteroides_eggerthii++++
SPC10081SPC10213Bacteroides_vulgatusBacteroides_eggerthii+
SPC00021SPC10213Blautia_productaBacteroides_eggerthii++++
SPC00026SPC10213Clostridium_nexileBacteroides_eggerthii
SPC00027SPC10213Clostridium_sp_HGF2Bacteroides_eggerthii
SPC10097SPC10213Collinsella_aerofaciensBacteroides_eggerthii++++
SPC00009SPC10213Coprobacillus_sp_D7Bacteroides_eggerthii
SPC00080SPC10213Coprococcus_catusBacteroides_eggerthii+
SPC00018SPC10213Dorea_formicigeneransBacteroides_eggerthii
SPC00057SPC10213Dorea_longicatenaBacteroides_eggerthii
SPC00008SPC10213Enterococcus_faecalisBacteroides_eggerthii++++
SPC10001SPC10213Erysipelotrichaceae_bacteriumBacteroides_eggerthii
SPC00001SPC10213Escherichia_coliBacteroides_eggerthii++++
SPC10110SPC10213Escherichia_coliBacteroides_eggerthii++++
SPC00022SPC10213Eubacterium_eligensBacteroides_eggerthii
SPC00054SPC10213Faecalibacterium_prausnitziiBacteroides_eggerthii
SPC00056SPC10213Odoribacter_splanchnicusBacteroides_eggerthii
SPC10048SPC10213Parabacteroides_merdaeBacteroides_eggerthii
SPC00061SPC10213Roseburia_intestinalisBacteroides_eggerthii
SPC10197SPC10213Ruminococcus_obeumBacteroides_eggerthii++++
SPC00015SPC10213Streptococcus_thermophilusBacteroides_eggerthii
SPC10030SPC10030Bacteroides_ovatusBacteroides_ovatus+++
SPC00006SPC10030Bacteroides_sp_1_1_6Bacteroides_ovatus++++
SPC00007SPC10030Bacteroides_sp_3_1_23Bacteroides_ovatus
SPC10019SPC10030Bacteroides_sp_D20Bacteroides_ovatus
SPC00005SPC10030Bacteroides_vulgatusBacteroides_ovatus+
SPC00021SPC10030Blautia_productaBacteroides_ovatus++++
SPC00026SPC10030Clostridium_nexileBacteroides_ovatus
SPC00027SPC10030Clostridium_sp_HGF2Bacteroides_ovatus
SPC00009SPC10030Coprobacillus_sp_D7Bacteroides_ovatus
SPC00080SPC10030Coprococcus_catusBacteroides_ovatus
SPC00018SPC10030Dorea_formicigeneransBacteroides_ovatus
SPC00057SPC10030Dorea_longicatenaBacteroides_ovatus
SPC00008SPC10030Enterococcus_faecalisBacteroides_ovatus++++
SPC10001SPC10030Erysipelotrichaceae_bacteriumBacteroides_ovatus
SPC00001SPC10030Escherichia_coliBacteroides_ovatus++++
SPC00022SPC10030Eubacterium_eligensBacteroides_ovatus
SPC00054SPC10030Faecalibacterium_prausnitziiBacteroides_ovatus
SPC00056SPC10030Odoribacter_splanchnicusBacteroides_ovatus
SPC00061SPC10030Roseburia_intestinalisBacteroides_ovatus
SPC00015SPC10030Streptococcus_thermophilusBacteroides_ovatus++
SPC00006SPC00006Bacteroides_sp_1_1_6Bacteroides_sp_1_1_6++++
SPC00005SPC00006Bacteroides_vulgatusBacteroides_sp_1_1_6++++
SPC00001SPC00006Escherichia_coliBacteroides_sp_1_1_6++++
SPC00006SPC00007Bacteroides_sp_1_1_6Bacteroides_sp_3_1_23++++
SPC00007SPC00007Bacteroides_sp_3_1_23Bacteroides_sp_3_1_23
SPC00005SPC00007Bacteroides_vulgatusBacteroides_sp_3_1_23+++
SPC00001SPC00007Escherichia_coliBacteroides_sp_3_1_23++++
SPC00006SPC10019Bacteroides_sp_1_1_6Bacteroides_sp_D20++++
SPC00007SPC10019Bacteroides_sp_3_1_23Bacteroides_sp_D20++++
SPC10019SPC10019Bacteroides_sp_D20Bacteroides_sp_D20
SPC00005SPC10019Bacteroides_vulgatusBacteroides_sp_D20+
SPC00021SPC10019Blautia_productaBacteroides_sp_D20++++
SPC00026SPC10019Clostridium_nexileBacteroides_sp_D20
SPC00027SPC10019Clostridium_sp_HGF2Bacteroides_sp_D20
SPC00009SPC10019Coprobacillus_sp_D7Bacteroides_sp_D20
SPC00080SPC10019Coprococcus_catusBacteroides_sp_D20
SPC00018SPC10019Dorea_formicigeneransBacteroides_sp_D20
SPC00057SPC10019Dorea_longicatenaBacteroides_sp_D20
SPC00008SPC10019Enterococcus_faecalisBacteroides_sp_D20++++
SPC10001SPC10019Erysipelotrichaceae_bacteriumBacteroides_sp_D20
SPC00001SPC10019Escherichia_coliBacteroides_sp_D20++++
SPC00022SPC10019Eubacterium_eligensBacteroides_sp_D20
SPC00054SPC10019Faecalibacterium_prausnitziiBacteroides_sp_D20
SPC00056SPC10019Odoribacter_splanchnicusBacteroides_sp_D20
SPC00061SPC10019Roseburia_intestinalisBacteroides_sp_D20
SPC00015SPC10019Streptococcus_thermophilusBacteroides_sp_D20+
SPC10030SPC10081Bacteroides_ovatusBacteroides_vulgatus
SPC00006SPC10081Bacteroides_sp_1_1_6Bacteroides_vulgatus
SPC00007SPC10081Bacteroides_sp_3_1_23Bacteroides_vulgatus
SPC10019SPC10081Bacteroides_sp_D20Bacteroides_vulgatus
SPC00005SPC00005Bacteroides_vulgatusBacteroides_vulgatus+
SPC00005SPC10081Bacteroides_vulgatusBacteroides_vulgatus++
SPC10081SPC10081Bacteroides_vulgatusBacteroides_vulgatus
SPC00021SPC10081Blautia_productaBacteroides_vulgatus++++
SPC00026SPC10081Clostridium_nexileBacteroides_vulgatus
SPC00027SPC10081Clostridium_sp_HGF2Bacteroides_vulgatus+++
SPC00009SPC10081Coprobacillus_sp_D7Bacteroides_vulgatus
SPC00080SPC10081Coprococcus_catusBacteroides_vulgatus++
SPC00018SPC10081Dorea_formicigeneransBacteroides_vulgatus
SPC00057SPC10081Dorea_longicatenaBacteroides_vulgatus
SPC00008SPC10081Enterococcus_faecalisBacteroides_vulgatus++++
SPC10001SPC10081Erysipelotrichaceae_bacteriumBacteroides_vulgatus
SPC00001SPC00005Escherichia_coliBacteroides_vulgatus++++
SPC00001SPC10081Escherichia_coliBacteroides_vulgatus++++
SPC00022SPC10081Eubacterium_eligensBacteroides_vulgatus
SPC00054SPC10081Faecalibacterium_prausnitziiBacteroides_vulgatus
SPC00056SPC10081Odoribacter_splanchnicusBacteroides_vulgatus
SPC10048SPC10081Parabacteroides_merdaeBacteroides_vulgatus+
SPC00061SPC10081Roseburia_intestinalisBacteroides_vulgatus
SPC00015SPC10081Streptococcus_thermophilusBacteroides_vulgatus−−
SPC10211SPC10301Bacteroides_caccaeBifidobacterium_adolescentis++++
SPC10213SPC10301Bacteroides_eggerthiiBifidobacterium_adolescentis++++
SPC10030SPC10301Bacteroides_ovatusBifidobacterium_adolescentis++++
SPC00006SPC10301Bacteroides_sp_1_1_6Bifidobacterium_adolescentis++++
SPC00007SPC10301Bacteroides_sp_3_1_23Bifidobacterium_adolescentis++++
SPC10019SPC10301Bacteroides_sp_D20Bifidobacterium_adolescentis++++
SPC00005SPC10301Bacteroides_vulgatusBifidobacterium_adolescentis++++
SPC10081SPC10301Bacteroides_vulgatusBifidobacterium_adolescentis++++
SPC10301SPC10301Bifidobacterium_adolescentisBifidobacterium_adolescentis++++
SPC10298SPC10301Bifidobacterium_pseudocatenulatumBifidobacterium_adolescentis++++
SPC00021SPC10301Blautia_productaBifidobacterium_adolescentis++++
SPC10243SPC10301Clostridium_hathewayiBifidobacterium_adolescentis++++
SPC00026SPC10301Clostridium_nexileBifidobacterium_adolescentis++++
SPC00027SPC10301Clostridium_sp_HGF2Bifidobacterium_adolescentis++++
SPC10097SPC10301Collinsella_aerofaciensBifidobacterium_adolescentis++++
SPC00009SPC10301Coprobacillus_sp_D7Bifidobacterium_adolescentis++++
SPC00080SPC10301Coprococcus_catusBifidobacterium_adolescentis
SPC00018SPC10301Dorea_formicigeneransBifidobacterium_adolescentis++++
SPC00057SPC10301Dorea_longicatenaBifidobacterium_adolescentis++++
SPC00008SPC10301Enterococcus_faecalisBifidobacterium_adolescentis++++
SPC10001SPC10301Erysipelotrichaceae_bacteriumBifidobacterium_adolescentis++++
SPC00001SPC10301Escherichia_coliBifidobacterium_adolescentis++++
SPC10110SPC10301Escherichia_coliBifidobacterium_adolescentis++++
SPC00022SPC10301Eubacterium_eligensBifidobacterium_adolescentis++++
SPC00054SPC10301Faecalibacterium_prausnitziiBifidobacterium_adolescentis+
SPC00056SPC10301Odoribacter_splanchnicusBifidobacterium_adolescentis+++
SPC10048SPC10301Parabacteroides_merdaeBifidobacterium_adolescentis++++
SPC00061SPC10301Roseburia_intestinalisBifidobacterium_adolescentis+++
SPC10197SPC10301Ruminococcus_obeumBifidobacterium_adolescentis++++
SPC10233SPC10301Ruminococcus_torquesBifidobacterium_adolescentis++++
SPC00015SPC10301Streptococcus_thermophilusBifidobacterium_adolescentis++++
SPC10211SPC10298Bacteroides_caccaeBifidobacterium_pseudocatenulatum++++
SPC10213SPC10298Bacteroides_eggerthiiBifidobacterium_pseudocatenulatum++++
SPC10030SPC10298Bacteroides_ovatusBifidobacterium_pseudocatenulatum++++
SPC00006SPC10298Bacteroides_sp_1_1_6Bifidobacterium_pseudocatenulatum++++
SPC00007SPC10298Bacteroides_sp_3_1_23Bifidobacterium_pseudocatenulatum++++
SPC10019SPC10298Bacteroides_sp_D20Bifidobacterium_pseudocatenulatum−−
SPC00005SPC10298Bacteroides_vulgatusBifidobacterium_pseudocatenulatum++++
SPC10081SPC10298Bacteroides_vulgatusBifidobacterium_pseudocatenulatum++++
SPC10298SPC10298Bifidobacterium_pseudocatenulatumBifidobacterium_pseudocatenulatum++++
SPC00021SPC10298Blautia_productaBifidobacterium_pseudocatenulatum+
SPC10243SPC10298Clostridium_hathewayiBifidobacterium_pseudocatenulatum++++
SPC00026SPC10298Clostridium_nexileBifidobacterium_pseudocatenulatum++++
SPC00027SPC10298Clostridium_sp_HGF2Bifidobacterium_pseudocatenulatum+++
SPC10097SPC10298Collinsella_aerofaciensBifidobacterium_pseudocatenulatum++++
SPC00009SPC10298Coprobacillus_sp_D7Bifidobacterium_pseudocatenulatum+++
SPC00080SPC10298Coprococcus_catusBifidobacterium_pseudocatenulatum
SPC00018SPC10298Dorea_formicigeneransBifidobacterium_pseudocatenulatum+++
SPC00057SPC10298Dorea_longicatenaBifidobacterium_pseudocatenulatum++++
SPC00008SPC10298Enterococcus_faecalisBifidobacterium_pseudocatenulatum++++
SPC10001SPC10298Erysipelotrichaceae_bacteriumBifidobacterium_pseudocatenulatum
SPC00001SPC10298Escherichia_coliBifidobacterium_pseudocatenulatum++++
SPC10110SPC10298Escherichia_coliBifidobacterium_pseudocatenulatum++++
SPC00022SPC10298Eubacterium_eligensBifidobacterium_pseudocatenulatum++++
SPC00054SPC10298Faecalibacterium_prausnitziiBifidobacterium_pseudocatenulatum++
SPC00056SPC10298Odoribacter_splanchnicusBifidobacterium_pseudocatenulatum+
SPC10048SPC10298Parabacteroides_merdaeBifidobacterium_pseudocatenulatum++++
SPC00061SPC10298Roseburia_intestinalisBifidobacterium_pseudocatenulatum+++
SPC10197SPC10298Ruminococcus_obeumBifidobacterium_pseudocatenulatum++++
SPC10233SPC10298Ruminococcus_torquesBifidobacterium_pseudocatenulatum++++
SPC00015SPC10298Streptococcus_thermophilusBifidobacterium_pseudocatenulatum++++
SPC10414SPC10415Alistipes_shahiiBlautia_producta
SPC10211SPC10415Bacteroides_caccaeBlautia_producta+
SPC10213SPC10415Bacteroides_eggerthiiBlautia_producta
SPC10030SPC10415Bacteroides_ovatusBlautia_producta
SPC00006SPC00021Bacteroides_sp_1_1_6Blautia_producta++++
SPC00006SPC10415Bacteroides_sp_1_1_6Blautia_producta++++
SPC00007SPC00021Bacteroides_sp_3_1_23Blautia_producta++++
SPC00007SPC10415Bacteroides_sp_3_1_23Blautia_producta++
SPC10019SPC10415Bacteroides_sp_D20Blautia_producta
SPC00005SPC00021Bacteroides_vulgatusBlautia_producta++++
SPC00005SPC10415Bacteroides_vulgatusBlautia_producta++++
SPC10081SPC10415Bacteroides_vulgatusBlautia_producta++++
SPC10301SPC10415Bifidobacterium_adolescentisBlautia_producta++++
SPC10298SPC10415Bifidobacterium_pseudocatenulatumBlautia_producta
SPC00021SPC00021Blautia_productaBlautia_producta++++
SPC00021SPC10415Blautia_productaBlautia_producta++++
SPC10415SPC10415Blautia_productaBlautia_producta+
SPC10415SPC10415Blautia_productaBlautia_producta++++
SPC10403SPC10415Blautia_schinkiiBlautia_producta
SPC10256SPC10415Clostridium butyricumBlautia_producta++++
SPC10358SPC10415Clostridium orbiscindensBlautia_producta++++
SPC10325SPC10415Clostridium_bolteaeBlautia_producta++++
SPC10167SPC10415Clostridium_disporicumBlautia_producta++++
SPC10243SPC10415Clostridium_hathewayiBlautia_producta+++
SPC10313SPC10415Clostridium_hylemonaeBlautia_producta++++
SPC10202SPC10415Clostridium_innocuumBlautia_producta++++
SPC10238SPC10415Clostridium_mayombeiBlautia_producta++++
SPC00026SPC10415Clostridium_nexileBlautia_producta
SPC00027SPC10415Clostridium_sp_HGF2Blautia_producta
SPC10355SPC10415Clostridium_symbiosumBlautia_producta
SPC10355SPC10415Clostridium_symbiosumBlautia_producta++++
SPC10155SPC10415Clostridium_tertiumBlautia_producta++++
SPC10097SPC10415Collinsella_aerofaciensBlautia_producta++++
SPC10097SPC10415Collinsella_aerofaciensBlautia_producta++++
SPC00009SPC00021Coprobacillus_sp_D7Blautia_producta++++
SPC00009SPC10415Coprobacillus_sp_D7Blautia_producta++++
SPC00080SPC10415Coprococcus_catusBlautia_producta−−−−
SPC10304SPC10415Coprococcus_comesBlautia_producta
SPC10304SPC10415Coprococcus_comesBlautia_producta++++
SPC00018SPC00021Dorea_formicigeneransBlautia_producta++++
SPC00018SPC10415Dorea_formicigeneransBlautia_producta−−
SPC00057SPC10415Dorea_longicatenaBlautia_producta+++
SPC00008SPC00021Enterococcus_faecalisBlautia_producta++++
SPC00008SPC10415Enterococcus_faecalisBlautia_producta++++
SPC10001SPC10415Erysipelotrichaceae_bacteriumBlautia_producta−−−
SPC00001SPC00021Escherichia_coliBlautia_producta++++
SPC00001SPC10415Escherichia_coliBlautia_producta++++
SPC10110SPC10415Escherichia_coliBlautia_producta++++
SPC00022SPC10415Eubacterium_eligensBlautia_producta−−−
SPC10363SPC10415Eubacterium_rectaleBlautia_producta+
SPC00054SPC10415Faecalibacterium_prausnitziiBlautia_producta
SPC10386SPC10415Faecalibacterium_prausnitziiBlautia_producta+
SPC10386SPC10415Faecalibacterium_prausnitziiBlautia_producta++++
SPC10390SPC10415Lachnospiraceae_bacterium_5_1_57FAABlautia_producta+
SPC10390SPC10415Lachnospiraceae_bacterium_5_1_57FAABlautia_producta++++
SPC00056SPC10415Odoribacter_splanchnicusBlautia_producta
SPC10388SPC10415Odoribacter_splanchnicusBlautia_producta+
SPC10048SPC10415Parabacteroides_merdaeBlautia_producta+++
SPC00061SPC10415Roseburia_intestinalisBlautia_producta−−
SPC10468SPC10415Ruminococcus_gnavusBlautia_producta++++
SPC10197SPC10415Ruminococcus_obeumBlautia_producta
SPC10233SPC10415Ruminococcus_torquesBlautia_producta
SPC00015SPC00021Streptococcus_thermophilusBlautia_producta++++
SPC00015SPC10415Streptococcus_thermophilusBlautia_producta
SPC10211SPC10403Bacteroides_caccaeBlautia_schinkii
SPC10213SPC10403Bacteroides_eggerthiiBlautia_schinkii−−
SPC10030SPC10403Bacteroides_ovatusBlautia_schinkii
SPC00006SPC10403Bacteroides_sp_1_1_6Blautia_schinkii+++
SPC00007SPC10403Bacteroides_sp_3_1_23Blautia_schinkii+
SPC10019SPC10403Bacteroides_sp_D20Blautia_schinkii−−
SPC00005SPC10403Bacteroides_vulgatusBlautia_schinkii++
SPC10081SPC10403Bacteroides_vulgatusBlautia_schinkii
SPC10301SPC10403Bifidobacterium_adolescentisBlautia_schinkii++
SPC10298SPC10403Bifidobacterium_pseudocatenulatumBlautia_schinkii
SPC00021SPC10403Blautia_productaBlautia_schinkii++++
SPC10403SPC10403Blautia_schinkiiBlautia_schinkii
SPC10243SPC10403Clostridium_hathewayiBlautia_schinkii++++
SPC00026SPC10403Clostridium_nexileBlautia_schinkii−−
SPC00027SPC10403Clostridium_sp_HGF2Blautia_schinkii
SPC10355SPC10403Clostridium_symbiosumBlautia_schinkii
SPC10097SPC10403Collinsella_aerofaciensBlautia_schinkii++++
SPC00009SPC10403Coprobacillus_sp_D7Blautia_schinkii++++
SPC00080SPC10403Coprococcus_catusBlautia_schinkii−−−
SPC10304SPC10403Coprococcus_comesBlautia_schinkii+
SPC00018SPC10403Dorea_formicigeneransBlautia_schinkii
SPC00057SPC10403Dorea_longicatenaBlautia_schinkii+++
SPC00008SPC10403Enterococcus_faecalisBlautia_schinkii++++
SPC10001SPC10403Erysipelotrichaceae_bacteriumBlautia_schinkii−−−
SPC00001SPC10403Escherichia_coliBlautia_schinkii++++
SPC10110SPC10403Escherichia_coliBlautia_schinkii++++
SPC00022SPC10403Eubacterium_eligensBlautia_schinkii
SPC10363SPC10403Eubacterium_rectaleBlautia_schinkii+
SPC00054SPC10403Faecalibacterium_prausnitziiBlautia_schinkii
SPC10386SPC10403Faecalibacterium_prausnitziiBlautia_schinkii
SPC10390SPC10403Lachnospiraceae_bacterium_5_1_57FAABlautia_schinkii
SPC00056SPC10403Odoribacter_splanchnicusBlautia_schinkii
SPC10388SPC10403Odoribacter_splanchnicusBlautia_schinkii
SPC10048SPC10403Parabacteroides_merdaeBlautia_schinkii
SPC00061SPC10403Roseburia_intestinalisBlautia_schinkii
SPC10197SPC10403Ruminococcus_obeumBlautia_schinkii
SPC10233SPC10403Ruminococcus_torquesBlautia_schinkii
SPC00015SPC10403Streptococcus_thermophilusBlautia_schinkii
SPC10256SPC10256Clostridium butyricumClostridium butyricum++++
SPC10167SPC10256Clostridium_disporicumClostridium butyricum++++
SPC10202SPC10256Clostridium_innocuumClostridium butyricum++++
SPC10238SPC10256Clostridium_mayombeiClostridium butyricum++++
SPC10155SPC10256Clostridium_tertiumClostridium butyricum++++
SPC10097SPC10256Collinsella_aerofaciensClostridium butyricum++++
SPC10304SPC10256Coprococcus_comesClostridium butyricum++++
SPC10256SPC10358Clostridium butyricumClostridium++++
orbiscindens
SPC10358SPC10358Clostridium orbiscindensClostridium+
orbiscindens
SPC10325SPC10358Clostridium_bolteaeClostridium++++
orbiscindens
SPC10167SPC10358Clostridium_disporicumClostridium++++
orbiscindens
SPC10313SPC10358Clostridium_hylemonaeClostridium
orbiscindens
SPC10202SPC10358Clostridium_innocuumClostridium++++
orbiscindens
SPC10238SPC10358Clostridium_mayombeiClostridium++++
orbiscindens
SPC10355SPC10358Clostridium_symbiosumClostridium++++
orbiscindens
SPC10155SPC10358Clostridium_tertiumClostridium++++
orbiscindens
SPC10097SPC10358Collinsella_aerofaciensClostridium++++
orbiscindens
SPC10304SPC10358Coprococcus_comesClostridium++++
orbiscindens
SPC10386SPC10358Faecalibacterium_prausnitziiClostridium
orbiscindens
SPC10256SPC10325Clostridium butyricumClostridium_bolteae++++
SPC10325SPC10325Clostridium_bolteaeClostridium_bolteae++++
SPC10167SPC10325Clostridium_disporicumClostridium_bolteae++++
SPC10313SPC10325Clostridium_hylemonaeClostridium_bolteae
SPC10202SPC10325Clostridium_innocuumClostridium_bolteae++++
SPC10238SPC10325Clostridium_mayombeiClostridium_bolteae++++
SPC10355SPC10325Clostridium_symbiosumClostridium_bolteae++++
SPC10155SPC10325Clostridium_tertiumClostridium_bolteae++++
SPC10097SPC10325Collinsella_aerofaciensClostridium_bolteae++++
SPC10304SPC10325Coprococcus_comesClostridium_bolteae++++
SPC10167SPC10167Clostridium_disporicumClostridium_disporicum+++
SPC10202SPC10167Clostridium_innocuumClostridium_disporicum+++
SPC10155SPC10167Clostridium_tertiumClostridium_disporicum++++
SPC10097SPC10167Collinsella_aerofaciensClostridium_disporicum
SPC10211SPC10243Bacteroides_caccaeClostridium_hathewayi++++
SPC10213SPC10243Bacteroides_eggerthiiClostridium_hathewayi++++
SPC10030SPC10243Bacteroides_ovatusClostridium_hathewayi++++
SPC00006SPC10243Bacteroides_sp_1_1_6Clostridium_hathewayi++++
SPC00007SPC10243Bacteroides_sp_3_1_23Clostridium_hathewayi++++
SPC10019SPC10243Bacteroides_sp_D20Clostridium_hathewayi++++
SPC00005SPC10243Bacteroides_vulgatusClostridium_hathewayi++++
SPC10081SPC10243Bacteroides_vulgatusClostridium_hathewayi++++
SPC00021SPC10243Blautia_productaClostridium_hathewayi++++
SPC10243SPC10243Clostridium_hathewayiClostridium_hathewayi++++
SPC00026SPC10243Clostridium_nexileClostridium_hathewayi
SPC00027SPC10243Clostridium_sp_HGF2Clostridium_hathewayi
SPC10097SPC10243Collinsella_aerofaciensClostridium_hathewayi++++
SPC00009SPC10243Coprobacillus_sp_D7Clostridium_hathewayi++++
SPC00080SPC10243Coprococcus_catusClostridium_hathewayi+++
SPC00018SPC10243Dorea_formicigeneransClostridium_hathewayi++++
SPC00057SPC10243Dorea_longicatenaClostridium_hathewayi+
SPC00008SPC10243Enterococcus_faecalisClostridium_hathewayi++++
SPC10001SPC10243Erysipelotrichaceae_bacteriumClostridium_hathewayi++++
SPC00001SPC10243Escherichia_coliClostridium_hathewayi++++
SPC10110SPC10243Escherichia_coliClostridium_hathewayi++++
SPC00022SPC10243Eubacterium_eligensClostridium_hathewayi+
SPC00054SPC10243Faecalibacterium_prausnitziiClostridium_hathewayi
SPC00056SPC10243Odoribacter_splanchnicusClostridium_hathewayi
SPC10048SPC10243Parabacteroides_merdaeClostridium_hathewayi+
SPC00061SPC10243Roseburia_intestinalisClostridium_hathewayi+++
SPC10197SPC10243Ruminococcus_obeumClostridium_hathewayi++++
SPC10233SPC10243Ruminococcus_torquesClostridium_hathewayi++++
SPC00015SPC10243Streptococcus_thermophilusClostridium_hathewayi++
SPC10256SPC10313Clostridium butyricumClostridium_hylemonae++++
SPC10325SPC10313Clostridium_bolteaeClostridium_hylemonae
SPC10167SPC10313Clostridium_disporicumClostridium_hylemonae
SPC10313SPC10313Clostridium_hylemonaeClostridium_hylemonae
SPC10202SPC10313Clostridium_innocuumClostridium_hylemonae++++
SPC10238SPC10313Clostridium_mayombeiClostridium_hylemonae++++
SPC10155SPC10313Clostridium_tertiumClostridium_hylemonae++++
SPC10097SPC10313Collinsella_aerofaciensClostridium_hylemonae+++
SPC10304SPC10313Coprococcus_comesClostridium_hylemonae+
SPC10167SPC10202Clostridium_disporicumClostridium_innocuum+++
SPC10202SPC10202Clostridium_innocuumClostridium_innocuum++++
SPC10238SPC10202Clostridium_mayombeiClostridium_innocuum++++
SPC10155SPC10202Clostridium_tertiumClostridium_innocuum++++
SPC10097SPC10202Collinsella_aerofaciensClostridium_innocuum+++
SPC10256SPC10238Clostridium butyricumClostridium_mayombei++++
SPC10167SPC10238Clostridium_disporicumClostridium_mayombei++++
SPC10202SPC10238Clostridium_innocuumClostridium_mayombei++++
SPC10238SPC10238Clostridium_mayombeiClostridium_mayombei++++
SPC10155SPC10238Clostridium_tertiumClostridium_mayombei++++
SPC10097SPC10238Collinsella_aerofaciensClostridium_mayombei++++
SPC00006SPC00026Bacteroides_sp_1_1_6Clostridium_nexile++++
SPC00007SPC00026Bacteroides_sp_3_1_23Clostridium_nexile++++
SPC00005SPC00026Bacteroides_vulgatusClostridium_nexile++++
SPC00021SPC00026Blautia_productaClostridium_nexile++++
SPC00026SPC00026Clostridium_nexileClostridium_nexile++
SPC00009SPC00026Coprobacillus_sp_D7Clostridium_nexile
SPC00018SPC00026Dorea_formicigeneransClostridium_nexile
SPC00008SPC00026Enterococcus_faecalisClostridium_nexile++++
SPC00001SPC00026Escherichia_coliClostridium_nexile++++
SPC00022SPC00026Eubacterium_eligensClostridium_nexile+
SPC00015SPC00026Streptococcus_thermophilusClostridium_nexile+
SPC00006SPC00027Bacteroides_sp_1_1_6Clostridium_sp_HGF2++++
SPC00007SPC00027Bacteroides_sp_3_1_23Clostridium_sp_HGF2++++
SPC00005SPC00027Bacteroides_vulgatusClostridium_sp_HGF2++
SPC00021SPC00027Blautia_productaClostridium_sp_HGF2++++
SPC00026SPC00027Clostridium_nexileClostridium_sp_HGF2++++
SPC00027SPC00027Clostridium_sp_HGF2Clostridium_sp_HGF2++++
SPC00009SPC00027Coprobacillus_sp_D7Clostridium_sp_HGF2
SPC00018SPC00027Dorea_formicigeneransClostridium_sp_HGF2
SPC00008SPC00027Enterococcus_faecalisClostridium_sp_HGF2++++
SPC00001SPC00027Escherichia_coliClostridium_sp_HGF2++++
SPC00022SPC00027Eubacterium_eligensClostridium_sp_HGF2
SPC00015SPC00027Streptococcus_thermophilusClostridium_sp_HGF2+
SPC10211SPC10355Bacteroides_caccaeClostridium_symbiosum+++
SPC10213SPC10355Bacteroides_eggerthiiClostridium_symbiosum++++
SPC10030SPC10355Bacteroides_ovatusClostridium_symbiosum
SPC00006SPC10355Bacteroides_sp_1_1_6Clostridium_symbiosum++++
SPC00007SPC10355Bacteroides_sp_3_1_23Clostridium_symbiosum++++
SPC10019SPC10355Bacteroides_sp_D20Clostridium_symbiosum
SPC00005SPC10355Bacteroides_vulgatusClostridium_symbiosum+++
SPC10081SPC10355Bacteroides_vulgatusClostridium_symbiosum
SPC10301SPC10355Bifidobacterium_adolescentisClostridium_symbiosum++++
SPC10298SPC10355Bifidobacterium_pseudocatenulatumClostridium_symbiosum+
SPC00021SPC10355Blautia_productaClostridium_symbiosum++++
SPC10256SPC10355Clostridium butyricumClostridium_symbiosum++++
SPC10358SPC10355Clostridium orbiscindensClostridium_symbiosum++++
SPC10325SPC10355Clostridium_bolteaeClostridium_symbiosum++++
SPC10167SPC10355Clostridium_disporicumClostridium_symbiosum++++
SPC10243SPC10355Clostridium_hathewayiClostridium_symbiosum++++
SPC10313SPC10355Clostridium_hylemonaeClostridium_symbiosum+++
SPC10202SPC10355Clostridium_innocuumClostridium_symbiosum++++
SPC10238SPC10355Clostridium_mayombeiClostridium_symbiosum++++
SPC00026SPC10355Clostridium_nexileClostridium_symbiosum+
SPC00027SPC10355Clostridium_sp_HGF2Clostridium_symbiosum
SPC10355SPC10355Clostridium_symbiosumClostridium_symbiosum+
SPC10355SPC10355Clostridium_symbiosumClostridium_symbiosum++++
SPC10155SPC10355Clostridium_tertiumClostridium_symbiosum+
SPC10097SPC10355Collinsella_aerofaciensClostridium_symbiosum+++
SPC10097SPC10355Collinsella_aerofaciensClostridium_symbiosum++++
SPC00009SPC10355Coprobacillus_sp_D7Clostridium_symbiosum
SPC00080SPC10355Coprococcus_catusClostridium_symbiosum
SPC10304SPC10355Coprococcus_comesClostridium_symbiosum
SPC10304SPC10355Coprococcus_comesClostridium_symbiosum++++
SPC00018SPC10355Dorea_formicigeneransClostridium_symbiosum
SPC00057SPC10355Dorea_longicatenaClostridium_symbiosum++++
SPC00008SPC10355Enterococcus_faecalisClostridium_symbiosum++++
SPC10001SPC10355Erysipelotrichaceae_bacteriumClostridium_symbiosum
SPC00001SPC10355Escherichia_coliClostridium_symbiosum++++
SPC10110SPC10355Escherichia_coliClostridium_symbiosum++++
SPC00022SPC10355Eubacterium_eligensClostridium_symbiosum+
SPC00054SPC10355Faecalibacterium_prausnitziiClostridium_symbiosum
SPC00056SPC10355Odoribacter_splanchnicusClostridium_symbiosum
SPC10048SPC10355Parabacteroides_merdaeClostridium_symbiosum
SPC00061SPC10355Roseburia_intestinalisClostridium_symbiosum−−
SPC10197SPC10355Ruminococcus_obeumClostridium_symbiosum++++
SPC10233SPC10355Ruminococcus_torquesClostridium_symbiosum++
SPC00015SPC10355Streptococcus_thermophilusClostridium_symbiosum
SPC10167SPC10155Clostridium_disporicumClostridium_tertium++++
SPC10155SPC10155Clostridium_tertiumClostridium_tertium++++
SPC10097SPC10155Collinsella_aerofaciensClostridium_tertium
SPC10030SPC10097Bacteroides_ovatusCollinsella_aerofaciens++++
SPC00006SPC10097Bacteroides_sp_1_1_6Collinsella_aerofaciens++++
SPC00007SPC10097Bacteroides_sp_3_1_23Collinsella_aerofaciens++++
SPC10019SPC10097Bacteroides_sp_D20Collinsella_aerofaciens++++
SPC00005SPC10097Bacteroides_vulgatusCollinsella_aerofaciens++++
SPC10081SPC10097Bacteroides_vulgatusCollinsella_aerofaciens++++
SPC00021SPC10097Blautia_productaCollinsella_aerofaciens++++
SPC00026SPC10097Clostridium_nexileCollinsella_aerofaciens+
SPC00027SPC10097Clostridium_sp_HGF2Collinsella_aerofaciens++++
SPC10155SPC10097Clostridium_tertiumCollinsella_aerofaciens
SPC10097SPC10097Collinsella_aerofaciensCollinsella_aerofaciens++++
SPC10097SPC10097Collinsella_aerofaciensCollinsella_aerofaciens
SPC00009SPC10097Coprobacillus_sp_D7Collinsella_aerofaciens+++
SPC00080SPC10097Coprococcus_catusCollinsella_aerofaciens++++
SPC00018SPC10097Dorea_formicigeneransCollinsella_aerofaciens++
SPC00057SPC10097Dorea_longicatenaCollinsella_aerofaciens++++
SPC00008SPC10097Enterococcus_faecalisCollinsella_aerofaciens++++
SPC10001SPC10097Erysipelotrichaceae_bacteriumCollinsella_aerofaciens++++
SPC00001SPC10097Escherichia_coliCollinsella_aerofaciens++++
SPC00022SPC10097Eubacterium_eligensCollinsella_aerofaciens+++
SPC00054SPC10097Faecalibacterium_prausnitziiCollinsella_aerofaciens+++
SPC00056SPC10097Odoribacter_splanchnicusCollinsella_aerofaciens+++
SPC10048SPC10097Parabacteroides_merdaeCollinsella_aerofaciens++++
SPC00061SPC10097Roseburia_intestinalisCollinsella_aerofaciens++
SPC00015SPC10097Streptococcus_thermophilusCollinsella_aerofaciens+
SPC00006SPC00009Bacteroides_sp_1_1_6Coprobacillus_sp_D7+++
SPC00007SPC00009Bacteroides_sp_3_1_23Coprobacillus_sp_D7
SPC00005SPC00009Bacteroides_vulgatusCoprobacillus_sp_D7+
SPC00009SPC00009Coprobacillus_sp_D7Coprobacillus_sp_D7
SPC00008SPC00009Enterococcus_faecalisCoprobacillus_sp_D7++++
SPC00001SPC00009Escherichia_coliCoprobacillus_sp_D7++
SPC00006SPC00080Bacteroides_sp_1_1_6Coprococcus_catus++++
SPC00007SPC00080Bacteroides_sp_3_1_23Coprococcus_catus
SPC00005SPC00080Bacteroides_vulgatusCoprococcus_catus+
SPC00021SPC00080Blautia_productaCoprococcus_catus++++
SPC00026SPC00080Clostridium_nexileCoprococcus_catus
SPC00027SPC00080Clostridium_sp_HGF2Coprococcus_catus−−−
SPC00009SPC00080Coprobacillus_sp_D7Coprococcus_catus−−−
SPC00080SPC00080Coprococcus_catusCoprococcus_catus
SPC00018SPC00080Dorea_formicigeneransCoprococcus_catus
SPC00057SPC00080Dorea_longicatenaCoprococcus_catus
SPC00008SPC00080Enterococcus_faecalisCoprococcus_catus++++
SPC00001SPC00080Escherichia_coliCoprococcus_catus++++
SPC00022SPC00080Eubacterium_eligensCoprococcus_catus
SPC00054SPC00080Faecalibacterium_prausnitziiCoprococcus_catus
SPC00056SPC00080Odoribacter_splanchnicusCoprococcus_catus
SPC00061SPC00080Roseburia_intestinalisCoprococcus_catus
SPC00015SPC00080Streptococcus_thermophilusCoprococcus_catus
SPC10211SPC10304Bacteroides_caccaeCoprococcus_comes+++
SPC10213SPC10304Bacteroides_eggerthiiCoprococcus_comes+++
SPC10030SPC10304Bacteroides_ovatusCoprococcus_comes
SPC00006SPC10304Bacteroides_sp_1_1_6Coprococcus_comes+++
SPC00007SPC10304Bacteroides_sp_3_1_23Coprococcus_comes++++
SPC10019SPC10304Bacteroides_sp_D20Coprococcus_comes
SPC00005SPC10304Bacteroides_vulgatusCoprococcus_comes++++
SPC10081SPC10304Bacteroides_vulgatusCoprococcus_comes
SPC10301SPC10304Bifidobacterium_adolescentisCoprococcus_comes++++
SPC10298SPC10304Bifidobacterium_pseudocatenulatumCoprococcus_comes++++
SPC00021SPC10304Blautia_productaCoprococcus_comes++++
SPC10256SPC10304Clostridium butyricumCoprococcus_comes++++
SPC10167SPC10304Clostridium_disporicumCoprococcus_comes++++
SPC10243SPC10304Clostridium_hathewayiCoprococcus_comes++++
SPC10313SPC10304Clostridium_hylemonaeCoprococcus_comes+
SPC10202SPC10304Clostridium_innocuumCoprococcus_comes++++
SPC10238SPC10304Clostridium_mayombeiCoprococcus_comes++++
SPC00026SPC10304Clostridium_nexileCoprococcus_comes
SPC00027SPC10304Clostridium_sp_HGF2Coprococcus_comes
SPC10155SPC10304Clostridium_tertiumCoprococcus_comes++++
SPC10097SPC10304Collinsella_aerofaciensCoprococcus_comes++++
SPC10097SPC10304Collinsella_aerofaciensCoprococcus_comes+++
SPC00009SPC10304Coprobacillus_sp_D7Coprococcus_comes+++
SPC00080SPC10304Coprococcus_catusCoprococcus_comes−−
SPC10304SPC10304Coprococcus_comesCoprococcus_comes
SPC10304SPC10304Coprococcus_comesCoprococcus_comes++
SPC00018SPC10304Dorea_formicigeneransCoprococcus_comes
SPC00057SPC10304Dorea_longicatenaCoprococcus_comes
SPC00008SPC10304Enterococcus_faecalisCoprococcus_comes++++
SPC10001SPC10304Erysipelotrichaceae_bacteriumCoprococcus_comes
SPC00001SPC10304Escherichia_coliCoprococcus_comes++++
SPC10110SPC10304Escherichia_coliCoprococcus_comes++++
SPC00022SPC10304Eubacterium_eligensCoprococcus_comes++
SPC00054SPC10304Faecalibacterium_prausnitziiCoprococcus_comes
SPC00056SPC10304Odoribacter_splanchnicusCoprococcus_comes
SPC10048SPC10304Parabacteroides_merdaeCoprococcus_comes
SPC00061SPC10304Roseburia_intestinalisCoprococcus_comes
SPC10197SPC10304Ruminococcus_obeumCoprococcus_comes++++
SPC10233SPC10304Ruminococcus_torquesCoprococcus_comes++++
SPC00015SPC10304Streptococcus_thermophilusCoprococcus_comes++
SPC00006SPC00018Bacteroides_sp_1_1_6Dorea_formicigenerans+++
SPC00007SPC00018Bacteroides_sp_3_1_23Dorea_formicigenerans
SPC00005SPC00018Bacteroides_vulgatusDorea_formicigenerans++
SPC00009SPC00018Coprobacillus_sp_D7Dorea_formicigenerans
SPC00018SPC00018Dorea_formicigeneransDorea_formicigenerans−−
SPC00008SPC00018Enterococcus_faecalisDorea_formicigenerans++++
SPC00001SPC00018Escherichia_coliDorea_formicigenerans++
SPC00015SPC00018Streptococcus_thermophilusDorea_formicigenerans
SPC00006SPC00057Bacteroides_sp_1_1_6Dorea_longicatena++++
SPC00007SPC00057Bacteroides_sp_3_1_23Dorea_longicatena+++
SPC00005SPC00057Bacteroides_vulgatusDorea_longicatena++++
SPC00021SPC00057Blautia_productaDorea_longicatena++++
SPC00026SPC00057Clostridium_nexileDorea_longicatena
SPC00027SPC00057Clostridium_sp_HGF2Dorea_longicatena−−
SPC00009SPC00057Coprobacillus_sp_D7Dorea_longicatena
SPC00018SPC00057Dorea_formicigeneransDorea_longicatena++
SPC00057SPC00057Dorea_longicatenaDorea_longicatena
SPC00008SPC00057Enterococcus_faecalisDorea_longicatena++++
SPC00001SPC00057Escherichia_coliDorea_longicatena++++
SPC00022SPC00057Eubacterium_eligensDorea_longicatena++
SPC00054SPC00057Faecalibacterium_prausnitziiDorea_longicatena
SPC00056SPC00057Odoribacter_splanchnicusDorea_longicatena
SPC00015SPC00057Streptococcus_thermophilusDorea_longicatena+
SPC00006SPC00008Bacteroides_sp_1_1_6Enterococcus_faecalis++++
SPC00007SPC00008Bacteroides_sp_3_1_23Enterococcus_faecalis++++
SPC00005SPC00008Bacteroides_vulgatusEnterococcus_faecalis++++
SPC00008SPC00008Enterococcus_faecalisEnterococcus_faecalis++++
SPC00001SPC00008Escherichia_coliEnterococcus_faecalis++++
SPC00006SPC10001Bacteroides_sp_1_1_6Erysipelotrichaceae_bacterium++++
SPC00007SPC10001Bacteroides_sp_3_1_23Erysipelotrichaceae_bacterium
SPC00005SPC10001Bacteroides_vulgatusErysipelotrichaceae_bacterium+
SPC00021SPC10001Blautia_productaErysipelotrichaceae_bacterium++++
SPC00026SPC10001Clostridium_nexileErysipelotrichaceae_bacterium
SPC00027SPC10001Clostridium_sp_HGF2Erysipelotrichaceae_bacterium−−
SPC00009SPC10001Coprobacillus_sp_D7Erysipelotrichaceae_bacterium
SPC00080SPC10001Coprococcus_catusErysipelotrichaceae_bacterium
SPC00018SPC10001Dorea_formicigeneransErysipelotrichaceae_bacterium−−
SPC00057SPC10001Dorea_longicatenaErysipelotrichaceae_bacterium
SPC00008SPC10001Enterococcus_faecalisErysipelotrichaceae_bacterium++++
SPC10001SPC10001Erysipelotrichaceae_bacteriumErysipelotrichaceae_bacterium
SPC00001SPC10001Escherichia_coliErysipelotrichaceae_bacterium++++
SPC00022SPC10001Eubacterium_eligensErysipelotrichaceae_bacterium
SPC00054SPC10001Faecalibacterium_prausnitziiErysipelotrichaceae_bacterium
SPC00056SPC10001Odoribacter_splanchnicusErysipelotrichaceae_bacterium
SPC00061SPC10001Roseburia_intestinalisErysipelotrichaceae_bacterium
SPC00015SPC10001Streptococcus_thermophilusErysipelotrichaceae_bacterium
SPC10030SPC10110Bacteroides_ovatusEscherichia_coli++++
SPC00006SPC10110Bacteroides_sp_1_1_6Escherichia_coli++++
SPC00007SPC10110Bacteroides_sp_3_1_23Escherichia_coli++++
SPC10019SPC10110Bacteroides_sp_D20Escherichia_coli++++
SPC00005SPC10110Bacteroides_vulgatusEscherichia_coli++++
SPC10081SPC10110Bacteroides_vulgatusEscherichia_coli++++
SPC00021SPC10110Blautia_productaEscherichia_coli++++
SPC00026SPC10110Clostridium_nexileEscherichia_coli++++
SPC00027SPC10110Clostridium_sp_HGF2Escherichia_coli++++
SPC10097SPC10110Collinsella_aerofaciensEscherichia_coli++++
SPC00009SPC10110Coprobacillus_sp_D7Escherichia_coli++
SPC00080SPC10110Coprococcus_catusEscherichia_coli++++
SPC00018SPC10110Dorea_formicigeneransEscherichia_coli++++
SPC00057SPC10110Dorea_longicatenaEscherichia_coli++++
SPC00008SPC10110Enterococcus_faecalisEscherichia_coli++++
SPC10001SPC10110Erysipelotrichaceae_bacteriumEscherichia_coli++++
SPC00001SPC00001Escherichia_coliEscherichia_coli++++
SPC00001SPC10110Escherichia_coliEscherichia_coli++++
SPC10110SPC10110Escherichia_coliEscherichia_coli++++
SPC00022SPC10110Eubacterium_eligensEscherichia_coli++++
SPC00054SPC10110Faecalibacterium_prausnitziiEscherichia_coli+++
SPC00056SPC10110Odoribacter_splanchnicusEscherichia_coli+++
SPC10048SPC10110Parabacteroides_merdaeEscherichia_coli++++
SPC00061SPC10110Roseburia_intestinalisEscherichia_coli+++
SPC00015SPC10110Streptococcus_thermophilusEscherichia_coli+++
SPC00006SPC00022Bacteroides_sp_1_1_6Eubacterium_eligens++++
SPC00007SPC00022Bacteroides_sp_3_1_23Eubacterium_eligens
SPC00005SPC00022Bacteroides_vulgatusEubacterium_eligens+++
SPC00021SPC00022Blautia_productaEubacterium_eligens++++
SPC00009SPC00022Coprobacillus_sp_D7Eubacterium_eligens
SPC00018SPC00022Dorea_formicigeneransEubacterium_eligens−−
SPC00008SPC00022Enterococcus_faecalisEubacterium_eligens++++
SPC00001SPC00022Escherichia_coliEubacterium_eligens++
SPC00022SPC00022Eubacterium_eligensEubacterium_eligens
SPC00015SPC00022Streptococcus_thermophilusEubacterium_eligens
SPC10211SPC10363Bacteroides_caccaeEubacterium_rectale
SPC10213SPC10363Bacteroides_eggerthiiEubacterium_rectale
SPC10030SPC10363Bacteroides_ovatusEubacterium_rectale
SPC00006SPC10363Bacteroides_sp_1_1_6Eubacterium_rectale++++
SPC00007SPC10363Bacteroides_sp_3_1_23Eubacterium_rectale+++
SPC10019SPC10363Bacteroides_sp_D20Eubacterium_rectale−−
SPC00005SPC10363Bacteroides_vulgatusEubacterium_rectale++++
SPC10081SPC10363Bacteroides_vulgatusEubacterium_rectale
SPC10301SPC10363Bifidobacterium_adolescentisEubacterium_rectale++++
SPC10298SPC10363Bifidobacterium_pseudocatenulatumEubacterium_rectale
SPC00021SPC10363Blautia_productaEubacterium_rectale++++
SPC10415SPC10567Blautia_productaEubacterium_rectale++++
SPC10256SPC10567Clostridium butyricumEubacterium_rectale++++
SPC10358SPC10567Clostridium orbiscindensEubacterium_rectale+
SPC10325SPC10567Clostridium_bolteaeEubacterium_rectale++
SPC10167SPC10567Clostridium_disporicumEubacterium_rectale++++
SPC10243SPC10363Clostridium_hathewayiEubacterium_rectale++++
SPC10313SPC10567Clostridium_hylemonaeEubacterium_rectale
SPC10202SPC10567Clostridium_innocuumEubacterium_rectale++++
SPC10238SPC10567Clostridium_mayombeiEubacterium_rectale++++
SPC00026SPC10363Clostridium_nexileEubacterium_rectale
SPC00027SPC10363Clostridium_sp_HGF2Eubacterium_rectale−−
SPC10355SPC10363Clostridium_symbiosumEubacterium_rectale++
SPC10355SPC10567Clostridium_symbiosumEubacterium_rectale+
SPC10155SPC10567Clostridium_tertiumEubacterium_rectale++++
SPC10097SPC10363Collinsella_aerofaciensEubacterium_rectale++++
SPC10097SPC10567Collinsella_aerofaciensEubacterium_rectale++++
SPC00009SPC10363Coprobacillus_sp_D7Eubacterium_rectale+++
SPC00080SPC10363Coprococcus_catusEubacterium_rectale−−−
SPC10304SPC10363Coprococcus_comesEubacterium_rectale+
SPC10304SPC10567Coprococcus_comesEubacterium_rectale++++
SPC00018SPC10363Dorea_formicigeneransEubacterium_rectale
SPC00057SPC10363Dorea_longicatenaEubacterium_rectale++++
SPC00008SPC10363Enterococcus_faecalisEubacterium_rectale++++
SPC10001SPC10363Erysipelotrichaceae_bacteriumEubacterium_rectale
SPC00001SPC10363Escherichia_coliEubacterium_rectale++++
SPC10110SPC10363Escherichia_coliEubacterium_rectale++++
SPC00022SPC10363Eubacterium_eligensEubacterium_rectale
SPC10363SPC10363Eubacterium_rectaleEubacterium_rectale+++
SPC10567SPC10567Eubacterium_rectaleEubacterium_rectale
SPC00054SPC10363Faecalibacterium_prausnitziiEubacterium_rectale−−
SPC10386SPC10567Faecalibacterium_prausnitziiEubacterium_rectale
SPC10390SPC10567Lachnospiraceae_bacterium_5_1_57FAAEubacterium_rectale+++
SPC00056SPC10363Odoribacter_splanchnicusEubacterium_rectale
SPC10048SPC10363Parabacteroides_merdaeEubacterium_rectale
SPC00061SPC10363Roseburia_intestinalisEubacterium_rectale−−−−
SPC10470SPC10567Ruminococcus_bromiiEubacterium_rectale+
SPC10468SPC10567Ruminococcus_gnavusEubacterium_rectale++++
SPC10197SPC10363Ruminococcus_obeumEubacterium_rectale++
SPC10233SPC10363Ruminococcus_torquesEubacterium_rectale+
SPC00015SPC10363Streptococcus_thermophilusEubacterium_rectale
SPC10211SPC10386Bacteroides_caccaeFaecalibacterium_prausnitzii
SPC10213SPC10386Bacteroides_eggerthiiFaecalibacterium_prausnitzii
SPC10030SPC10386Bacteroides_ovatusFaecalibacterium_prausnitzii
SPC00006SPC00054Bacteroides_sp_1_1_6Faecalibacterium_prausnitzii++++
SPC00006SPC10386Bacteroides_sp_1_1_6Faecalibacterium_prausnitzii+++
SPC00007SPC00054Bacteroides_sp_3_1_23Faecalibacterium_prausnitzii++
SPC00007SPC10386Bacteroides_sp_3_1_23Faecalibacterium_prausnitzii
SPC10019SPC10386Bacteroides_sp_D20Faecalibacterium_prausnitzii−−
SPC00005SPC00054Bacteroides_vulgatusFaecalibacterium_prausnitzii++++
SPC00005SPC10386Bacteroides_vulgatusFaecalibacterium_prausnitzii+++
SPC10081SPC10386Bacteroides_vulgatusFaecalibacterium_prausnitzii−−−
SPC10301SPC10386Bifidobacterium_adolescentisFaecalibacterium_prausnitzii+
SPC10298SPC10386Bifidobacterium_pseudocatenulatumFaecalibacterium_prausnitzii
SPC00021SPC00054Blautia_productaFaecalibacterium_prausnitzii++++
SPC00021SPC10386Blautia_productaFaecalibacterium_prausnitzii++++
SPC10256SPC10386Clostridium butyricumFaecalibacterium_prausnitzii++++
SPC10358SPC10386Clostridium orbiscindensFaecalibacterium_prausnitzii
SPC10325SPC10386Clostridium_bolteaeFaecalibacterium_prausnitzii++
SPC10167SPC10386Clostridium_disporicumFaecalibacterium_prausnitzii
SPC10243SPC10386Clostridium_hathewayiFaecalibacterium_prausnitzii+++
SPC10313SPC10386Clostridium_hylemonaeFaecalibacterium_prausnitzii
SPC10202SPC10386Clostridium_innocuumFaecalibacterium_prausnitzii++++
SPC10238SPC10386Clostridium_mayombeiFaecalibacterium_prausnitzii++++
SPC00026SPC00054Clostridium_nexileFaecalibacterium_prausnitzii
SPC00026SPC10386Clostridium_nexileFaecalibacterium_prausnitzii
SPC00027SPC00054Clostridium_sp_HGF2Faecalibacterium_prausnitzii++
SPC00027SPC10386Clostridium_sp_HGF2Faecalibacterium_prausnitzii−−
SPC10355SPC10386Clostridium_symbiosumFaecalibacterium_prausnitzii+++
SPC10355SPC10386Clostridium_symbiosumFaecalibacterium_prausnitzii++++
SPC10155SPC10386Clostridium_tertiumFaecalibacterium_prausnitzii++++
SPC10097SPC10386Collinsella_aerofaciensFaecalibacterium_prausnitzii++++
SPC10097SPC10386Collinsella_aerofaciensFaecalibacterium_prausnitzii++++
SPC00009SPC00054Coprobacillus_sp_D7Faecalibacterium_prausnitzii−−−
SPC00009SPC10386Coprobacillus_sp_D7Faecalibacterium_prausnitzii
SPC00080SPC10386Coprococcus_catusFaecalibacterium_prausnitzii−−−
SPC10304SPC10386Coprococcus_comesFaecalibacterium_prausnitzii
SPC10304SPC10386Coprococcus_comesFaecalibacterium_prausnitzii+++
SPC00018SPC00054Dorea_formicigeneransFaecalibacterium_prausnitzii
SPC00018SPC10386Dorea_formicigeneransFaecalibacterium_prausnitzii−−−
SPC00057SPC10386Dorea_longicatenaFaecalibacterium_prausnitzii+++
SPC00008SPC00054Enterococcus_faecalisFaecalibacterium_prausnitzii++++
SPC00008SPC10386Enterococcus_faecalisFaecalibacterium_prausnitzii++++
SPC10001SPC10386Erysipelotrichaceae_bacteriumFaecalibacterium_prausnitzii−−
SPC00001SPC00054Escherichia_coliFaecalibacterium_prausnitzii++++
SPC00001SPC10386Escherichia_coliFaecalibacterium_prausnitzii++++
SPC10110SPC10386Escherichia_coliFaecalibacterium_prausnitzii++
SPC00022SPC00054Eubacterium_eligensFaecalibacterium_prausnitzii
SPC00022SPC10386Eubacterium_eligensFaecalibacterium_prausnitzii
SPC10363SPC10386Eubacterium_rectaleFaecalibacterium_prausnitzii+
SPC00054SPC00054Faecalibacterium_prausnitziiFaecalibacterium_prausnitzii+
SPC00054SPC10386Faecalibacterium_prausnitziiFaecalibacterium_prausnitzii
SPC10386SPC10386Faecalibacterium_prausnitziiFaecalibacterium_prausnitzii+
SPC10386SPC10386Faecalibacterium_prausnitziiFaecalibacterium_prausnitzii
SPC10390SPC10386Lachnospiraceae_bacterium_5_1_57FAAFaecalibacterium_prausnitzii++++
SPC00056SPC10386Odoribacter_splanchnicusFaecalibacterium_prausnitzii−−
SPC10048SPC10386Parabacteroides_merdaeFaecalibacterium_prausnitzii
SPC00061SPC10386Roseburia_intestinalisFaecalibacterium_prausnitzii
SPC10197SPC10386Ruminococcus_obeumFaecalibacterium_prausnitzii
SPC10233SPC10386Ruminococcus_torquesFaecalibacterium_prausnitzii
SPC00015SPC00054Streptococcus_thermophilusFaecalibacterium_prausnitzii
SPC00015SPC10386Streptococcus_thermophilusFaecalibacterium_prausnitzii
SPC10211SPC10390Bacteroides_caccaeLachnospiraceae_bacterium_5_1_57FAA
SPC10213SPC10390Bacteroides_eggerthiiLachnospiraceae_bacterium_5_1_57FAA
SPC10030SPC10390Bacteroides_ovatusLachnospiraceae_bacterium_5_1_57FAA
SPC00006SPC10390Bacteroides_sp_1_1_6Lachnospiraceae_bacterium_5_1_57FAA+++
SPC00007SPC10390Bacteroides_sp_3_1_23Lachnospiraceae_bacterium_5_1_57FAA
SPC10019SPC10390Bacteroides_sp_D20Lachnospiraceae_bacterium_5_1_57FAA−−−
SPC00005SPC10390Bacteroides_vulgatusLachnospiraceae_bacterium_5_1_57FAA+++
SPC10081SPC10390Bacteroides_vulgatusLachnospiraceae_bacterium_5_1_57FAA−−
SPC10301SPC10390Bifidobacterium_adolescentisLachnospiraceae_bacterium_5_1_57FAA++++
SPC10298SPC10390Bifidobacterium_pseudocatenulatumLachnospiraceae_bacterium_5_1_57FAA
SPC00021SPC10390Blautia_productaLachnospiraceae_bacterium_5_1_57FAA++++
SPC10415SPC10390Blautia_productaLachnospiraceae_bacterium_5_1_57FAA++++
SPC10256SPC10390Clostridium butyricumLachnospiraceae_bacterium_5_1_57FAA++++
SPC10358SPC10390Clostridium orbiscindensLachnospiraceae_bacterium_5_1_57FAA++++
SPC10325SPC10390Clostridium_bolteaeLachnospiraceae_bacterium_5_1_57FAA++++
SPC10167SPC10390Clostridium_disporicumLachnospiraceae_bacterium_5_1_57FAA++++
SPC10243SPC10390Clostridium_hathewayiLachnospiraceae_bacterium_5_1_57FAA++++
SPC10313SPC10390Clostridium_hylemonaeLachnospiraceae_bacterium_5_1_57FAA++++
SPC10202SPC10390Clostridium_innocuumLachnospiraceae_bacterium_5_1_57FAA++++
SPC10238SPC10390Clostridium_mayombeiLachnospiraceae_bacterium_5_1_57FAA++++
SPC00026SPC10390Clostridium_nexileLachnospiraceae_bacterium_5_1_57FAA
SPC00027SPC10390Clostridium_sp_HGF2Lachnospiraceae_bacterium_5_1_57FAA
SPC10355SPC10390Clostridium_symbiosumLachnospiraceae_bacterium_5_1_57FAA+
SPC10355SPC10390Clostridium_symbiosumLachnospiraceae_bacterium_5_1_57FAA++++
SPC10155SPC10390Clostridium_tertiumLachnospiraceae_bacterium_5_1_57FAA++++
SPC10097SPC10390Collinsella_aerofaciensLachnospiraceae_bacterium_5_1_57FAA++++
SPC10097SPC10390Collinsella_aerofaciensLachnospiraceae_bacterium_5_1_57FAA++++
SPC00009SPC10390Coprobacillus_sp_D7Lachnospiraceae_bacterium_5_1_57FAA++++
SPC00080SPC10390Coprococcus_catusLachnospiraceae_bacterium_5_1_57FAA
SPC10304SPC10390Coprococcus_comesLachnospiraceae_bacterium_5_1_57FAA
SPC10304SPC10390Coprococcus_comesLachnospiraceae_bacterium_5_1_57FAA++++
SPC00018SPC10390Dorea_formicigeneransLachnospiraceae_bacterium_5_1_57FAA−−
SPC00057SPC10390Dorea_longicatenaLachnospiraceae_bacterium_5_1_57FAA++++
SPC00008SPC10390Enterococcus_faecalisLachnospiraceae_bacterium_5_1_57FAA++++
SPC10001SPC10390Erysipelotrichaceae_bacteriumLachnospiraceae_bacterium_5_1_57FAA−−−
SPC00001SPC10390Escherichia_coliLachnospiraceae_bacterium_5_1_57FAA++++
SPC10110SPC10390Escherichia_coliLachnospiraceae_bacterium_5_1_57FAA++++
SPC00022SPC10390Eubacterium_eligensLachnospiraceae_bacterium_5_1_57FAA
SPC10363SPC10390Eubacterium_rectaleLachnospiraceae_bacterium_5_1_57FAA
SPC00054SPC10390Faecalibacterium_prausnitziiLachnospiraceae_bacterium_5_1_57FAA
SPC10386SPC10390Faecalibacterium_prausnitziiLachnospiraceae_bacterium_5_1_57FAA
SPC10386SPC10390Faecalibacterium_prausnitziiLachnospiraceae_bacterium_5_1_57FAA++++
SPC10390SPC10390Lachnospiraceae_bacterium_5_1_57FAALachnospiraceae_bacterium_5_1_57FAA
SPC10390SPC10390Lachnospiraceae_bacterium_5_1_57FAALachnospiraceae_bacterium_5_1_57FAA++++
SPC00056SPC10390Odoribacter_splanchnicusLachnospiraceae_bacterium_5_1_57FAA−−
SPC10388SPC10390Odoribacter_splanchnicusLachnospiraceae_bacterium_5_1_57FAA
SPC10048SPC10390Parabacteroides_merdaeLachnospiraceae_bacterium_5_1_57FAA
SPC00061SPC10390Roseburia_intestinalisLachnospiraceae_bacterium_5_1_57FAA
SPC10197SPC10390Ruminococcus_obeumLachnospiraceae_bacterium_5_1_57FAA
SPC10233SPC10390Ruminococcus_torquesLachnospiraceae_bacterium_5_1_57FAA
SPC00015SPC10390Streptococcus_thermophilusLachnospiraceae_bacterium_5_1_57FAA
SPC10211SPC10388Bacteroides_caccaeOdoribacter_splanchnicus
SPC10213SPC10388Bacteroides_eggerthiiOdoribacter_splanchnicus
SPC10030SPC10388Bacteroides_ovatusOdoribacter_splanchnicus−−
SPC00006SPC00056Bacteroides_sp_1_1_6Odoribacter_splanchnicus++++
SPC00006SPC10388Bacteroides_sp_1_1_6Odoribacter_splanchnicus+
SPC00007SPC00056Bacteroides_sp_3_1_23Odoribacter_splanchnicus+
SPC00007SPC10388Bacteroides_sp_3_1_23Odoribacter_splanchnicus
SPC10019SPC10388Bacteroides_sp_D20Odoribacter_splanchnicus−−−
SPC00005SPC00056Bacteroides_vulgatusOdoribacter_splanchnicus+++
SPC00005SPC10388Bacteroides_vulgatusOdoribacter_splanchnicus+++
SPC10081SPC10388Bacteroides_vulgatusOdoribacter_splanchnicus
SPC10301SPC10388Bifidobacterium_adolescentisOdoribacter_splanchnicus++++
SPC10298SPC10388Bifidobacterium_pseudocatenulatumOdoribacter_splanchnicus+++
SPC00021SPC00056Blautia_productaOdoribacter_splanchnicus++++
SPC00021SPC10388Blautia_productaOdoribacter_splanchnicus++++
SPC10243SPC10388Clostridium_hathewayiOdoribacter_splanchnicus++++
SPC00026SPC00056Clostridium_nexileOdoribacter_splanchnicus
SPC00026SPC10388Clostridium_nexileOdoribacter_splanchnicus−−−
SPC00027SPC00056Clostridium_sp_HGF2Odoribacter_splanchnicus
SPC00027SPC10388Clostridium_sp_HGF2Odoribacter_splanchnicus−−−
SPC10355SPC10388Clostridium_symbiosumOdoribacter_splanchnicus++
SPC10097SPC10388Collinsella_aerofaciensOdoribacter_splanchnicus++++
SPC00009SPC00056Coprobacillus_sp_D7Odoribacter_splanchnicus
SPC00009SPC10388Coprobacillus_sp_D7Odoribacter_splanchnicus+++
SPC00080SPC10388Coprococcus_catusOdoribacter_splanchnicus−−
SPC10304SPC10388Coprococcus_comesOdoribacter_splanchnicus
SPC00018SPC00056Dorea_formicigeneransOdoribacter_splanchnicus
SPC00018SPC10388Dorea_formicigeneransOdoribacter_splanchnicus
SPC00057SPC10388Dorea_longicatenaOdoribacter_splanchnicus++++
SPC00008SPC00056Enterococcus_faecalisOdoribacter_splanchnicus++++
SPC00008SPC10388Enterococcus_faecalisOdoribacter_splanchnicus++++
SPC10001SPC10388Erysipelotrichaceae_bacteriumOdoribacter_splanchnicus−−
SPC00001SPC00056Escherichia_coliOdoribacter_splanchnicus++++
SPC00001SPC10388Escherichia_coliOdoribacter_splanchnicus++++
SPC10110SPC10388Escherichia_coliOdoribacter_splanchnicus++++
SPC00022SPC00056Eubacterium_eligensOdoribacter_splanchnicus
SPC00022SPC10388Eubacterium_eligensOdoribacter_splanchnicus
SPC10363SPC10388Eubacterium_rectaleOdoribacter_splanchnicus+
SPC00054SPC00056Faecalibacterium_prausnitziiOdoribacter_splanchnicus
SPC00054SPC10388Faecalibacterium_prausnitziiOdoribacter_splanchnicus
SPC10386SPC10388Faecalibacterium_prausnitziiOdoribacter_splanchnicus+
SPC00056SPC00056Odoribacter_splanchnicusOdoribacter_splanchnicus
SPC00056SPC10388Odoribacter_splanchnicusOdoribacter_splanchnicus−−−
SPC10388SPC10388Odoribacter_splanchnicusOdoribacter_splanchnicus+
SPC10048SPC10388Parabacteroides_merdaeOdoribacter_splanchnicus
SPC00061SPC10388Roseburia_intestinalisOdoribacter_splanchnicus
SPC10197SPC10388Ruminococcus_obeumOdoribacter_splanchnicus+
SPC10233SPC10388Ruminococcus_torquesOdoribacter_splanchnicus
SPC00015SPC00056Streptococcus_thermophilusOdoribacter_splanchnicus
SPC00015SPC10388Streptococcus_thermophilusOdoribacter_splanchnicus+
SPC10030SPC10048Bacteroides_ovatusParabacteroides_merdae
SPC00006SPC10048Bacteroides_sp_1_1_6Parabacteroides_merdae++++
SPC00007SPC10048Bacteroides_sp_3_1_23Parabacteroides_merdae+++
SPC10019SPC10048Bacteroides_sp_D20Parabacteroides_merdae
SPC00005SPC10048Bacteroides_vulgatusParabacteroides_merdae++++
SPC00021SPC10048Blautia_productaParabacteroides_merdae++++
SPC00026SPC10048Clostridium_nexileParabacteroides_merdae++
SPC00027SPC10048Clostridium_sp_HGF2Parabacteroides_merdae+++
SPC00009SPC10048Coprobacillus_sp_D7Parabacteroides_merdae
SPC00080SPC10048Coprococcus_catusParabacteroides_merdae+++
SPC00018SPC10048Dorea_formicigeneransParabacteroides_merdae
SPC00057SPC10048Dorea_longicatenaParabacteroides_merdae
SPC00008SPC10048Enterococcus_faecalisParabacteroides_merdae++++
SPC10001SPC10048Erysipelotrichaceae_bacteriumParabacteroides_merdae
SPC00001SPC10048Escherichia_coliParabacteroides_merdae++++
SPC00022SPC10048Eubacterium_eligensParabacteroides_merdae
SPC00054SPC10048Faecalibacterium_prausnitziiParabacteroides_merdae+
SPC00056SPC10048Odoribacter_splanchnicusParabacteroides_merdae
SPC10048SPC10048Parabacteroides_merdaeParabacteroides_merdae+++
SPC00061SPC10048Roseburia_intestinalisParabacteroides_merdae
SPC00015SPC10048Streptococcus_thermophilusParabacteroides_merdae
SPC00006SPC00061Bacteroides_sp_1_1_6Roseburia_intestinalis++++
SPC00007SPC00061Bacteroides_sp_3_1_23Roseburia_intestinalis+
SPC00005SPC00061Bacteroides_vulgatusRoseburia_intestinalis+
SPC00021SPC00061Blautia_productaRoseburia_intestinalis++++
SPC00026SPC00061Clostridium_nexileRoseburia_intestinalis
SPC00027SPC00061Clostridium_sp_HGF2Roseburia_intestinalis−−−
SPC00009SPC00061Coprobacillus_sp_D7Roseburia_intestinalis
SPC00018SPC00061Dorea_formicigeneransRoseburia_intestinalis
SPC00057SPC00061Dorea_longicatenaRoseburia_intestinalis
SPC00008SPC00061Enterococcus_faecalisRoseburia_intestinalis++++
SPC00001SPC00061Escherichia_coliRoseburia_intestinalis++++
SPC00022SPC00061Eubacterium_eligensRoseburia_intestinalis
SPC00054SPC00061Faecalibacterium_prausnitziiRoseburia_intestinalis
SPC00056SPC00061Odoribacter_splanchnicusRoseburia_intestinalis
SPC00061SPC00061Roseburia_intestinalisRoseburia_intestinalis
SPC00015SPC00061Streptococcus_thermophilusRoseburia_intestinalis
SPC10415SPC10470Blautia_productaRuminococcus_bromii++++
SPC10256SPC10470Clostridium butyricumRuminococcus_bromii++++
SPC10358SPC10470Clostridium orbiscindensRuminococcus_bromii
SPC10325SPC10470Clostridium_bolteaeRuminococcus_bromii+++
SPC10167SPC10470Clostridium_disporicumRuminococcus_bromii
SPC10313SPC10470Clostridium_hylemonaeRuminococcus_bromii
SPC10202SPC10470Clostridium_innocuumRuminococcus_bromii++++
SPC10238SPC10470Clostridium_mayombeiRuminococcus_bromii++++
SPC10355SPC10470Clostridium_symbiosumRuminococcus_bromii++++
SPC10155SPC10470Clostridium_tertiumRuminococcus_bromii++++
SPC10097SPC10470Collinsella_aerofaciensRuminococcus_bromii++++
SPC10304SPC10470Coprococcus_comesRuminococcus_bromii++++
SPC10567SPC10470Eubacterium_rectaleRuminococcus_bromii+
SPC10386SPC10470Faecalibacterium_prausnitziiRuminococcus_bromii
SPC10390SPC10470Lachnospiraceae_bacterium_5_1_57FAARuminococcus_bromii++++
SPC10470SPC10470Ruminococcus_bromiiRuminococcus_bromii
SPC10468SPC10470Ruminococcus_gnavusRuminococcus_bromii++++
SPC10415SPC10468Blautia_productaRuminococcus_gnavus++++
SPC10256SPC10468Clostridium butyricumRuminococcus_gnavus++++
SPC10358SPC10468Clostridium orbiscindensRuminococcus_gnavus++++
SPC10325SPC10468Clostridium_bolteaeRuminococcus_gnavus++++
SPC10167SPC10468Clostridium_disporicumRuminococcus_gnavus++++
SPC10313SPC10468Clostridium_hylemonaeRuminococcus_gnavus+++
SPC10202SPC10468Clostridium_innocuumRuminococcus_gnavus++++
SPC10238SPC10468Clostridium_mayombeiRuminococcus_gnavus++++
SPC10355SPC10468Clostridium_symbiosumRuminococcus_gnavus++++
SPC10155SPC10468Clostridium_tertiumRuminococcus_gnavus++++
SPC10097SPC10468Collinsella_aerofaciensRuminococcus_gnavus++++
SPC10304SPC10468Coprococcus_comesRuminococcus_gnavus++++
SPC10386SPC10468Faecalibacterium_prausnitziiRuminococcus_gnavus++++
SPC10390SPC10468Lachnospiraceae_bacterium_5_1_57FAARuminococcus_gnavus++++
SPC10470SPC10468Ruminococcus_bromiiRuminococcus_gnavus++++
SPC10468SPC10468Ruminococcus_gnavusRuminococcus_gnavus+++
SPC10030SPC10197Bacteroides_ovatusRuminococcus_obeum
SPC00006SPC10197Bacteroides_sp_1_1_6Ruminococcus_obeum+++
SPC00007SPC10197Bacteroides_sp_3_1_23Ruminococcus_obeum+++
SPC10019SPC10197Bacteroides_sp_D20Ruminococcus_obeum
SPC00005SPC10197Bacteroides_vulgatusRuminococcus_obeum++++
SPC10081SPC10197Bacteroides_vulgatusRuminococcus_obeum
SPC00021SPC10197Blautia_productaRuminococcus_obeum++++
SPC00026SPC10197Clostridium_nexileRuminococcus_obeum
SPC00027SPC10197Clostridium_sp_HGF2Ruminococcus_obeum−−
SPC10097SPC10197Collinsella_aerofaciensRuminococcus_obeum++++
SPC00009SPC10197Coprobacillus_sp_D7Ruminococcus_obeum+
SPC00080SPC10197Coprococcus_catusRuminococcus_obeum
SPC00018SPC10197Dorea_formicigeneransRuminococcus_obeum++++
SPC00057SPC10197Dorea_longicatenaRuminococcus_obeum
SPC00008SPC10197Enterococcus_faecalisRuminococcus_obeum++++
SPC10001SPC10197Erysipelotrichaceae_bacteriumRuminococcus_obeum
SPC00001SPC10197Escherichia_coliRuminococcus_obeum+++
SPC10110SPC10197Escherichia_coliRuminococcus_obeum++++
SPC00022SPC10197Eubacterium_eligensRuminococcus_obeum+
SPC00054SPC10197Faecalibacterium_prausnitziiRuminococcus_obeum
SPC00056SPC10197Odoribacter_splanchnicusRuminococcus_obeum
SPC10048SPC10197Parabacteroides_merdaeRuminococcus_obeum
SPC00061SPC10197Roseburia_intestinalisRuminococcus_obeum
SPC10197SPC10197Ruminococcus_obeumRuminococcus_obeum++++
SPC00015SPC10197Streptococcus_thermophilusRuminococcus_obeum+++
SPC10211SPC10233Bacteroides_caccaeRuminococcus_torques++++
SPC10213SPC10233Bacteroides_eggerthiiRuminococcus_torques++++
SPC10030SPC10233Bacteroides_ovatusRuminococcus_torques++++
SPC00006SPC10233Bacteroides_sp_1_1_6Ruminococcus_torques++++
SPC00007SPC10233Bacteroides_sp_3_1_23Ruminococcus_torques++++
SPC10019SPC10233Bacteroides_sp_D20Ruminococcus_torques++
SPC00005SPC10233Bacteroides_vulgatusRuminococcus_torques++++
SPC10081SPC10233Bacteroides_vulgatusRuminococcus_torques++++
SPC00021SPC10233Blautia_productaRuminococcus_torques++++
SPC00026SPC10233Clostridium_nexileRuminococcus_torques+
SPC00027SPC10233Clostridium_sp_HGF2Ruminococcus_torques
SPC10097SPC10233Collinsella_aerofaciensRuminococcus_torques++++
SPC00009SPC10233Coprobacillus_sp_D7Ruminococcus_torques++++
SPC00080SPC10233Coprococcus_catusRuminococcus_torques+
SPC00018SPC10233Dorea_formicigeneransRuminococcus_torques++++
SPC00057SPC10233Dorea_longicatenaRuminococcus_torques
SPC00008SPC10233Enterococcus_faecalisRuminococcus_torques++++
SPC10001SPC10233Erysipelotrichaceae_bacteriumRuminococcus_torques+
SPC00001SPC10233Escherichia_coliRuminococcus_torques++++
SPC10110SPC10233Escherichia_coliRuminococcus_torques++++
SPC00022SPC10233Eubacterium_eligensRuminococcus_torques++
SPC00054SPC10233Faecalibacterium_prausnitziiRuminococcus_torques
SPC00056SPC10233Odoribacter_splanchnicusRuminococcus_torques
SPC10048SPC10233Parabacteroides_merdaeRuminococcus_torques+
SPC00061SPC10233Roseburia_intestinalisRuminococcus_torques+
SPC10197SPC10233Ruminococcus_obeumRuminococcus_torques++++
SPC10233SPC10233Ruminococcus_torquesRuminococcus_torques++++
SPC00015SPC10233Streptococcus_thermophilusRuminococcus_torques+
SPC00006SPC00015Bacteroides_sp_1_1_6Streptococcus_thermophilus+++
SPC00007SPC00015Bacteroides_sp_3_1_23Streptococcus_thermophilus+++
SPC00005SPC00015Bacteroides_vulgatusStreptococcus_thermophilus+
SPC00009SPC00015Coprobacillus_sp_D7Streptococcus_thermophilus+
SPC00008SPC00015Enterococcus_faecalisStreptococcus_thermophilus++++
SPC00001SPC00015Escherichia_coliStreptococcus_thermophilus+
SPC00015SPC00015Streptococcus_thermophilusStreptococcus_thermophilus

Как компенсировать расходы
на инновационную разработку
Похожие патенты