Methods for the diagnosis and treatment of inflammatory bowel disease

Abstract
There is provided methods and compositions to diagnose, classify and treat inflammatory bowel disease including ulcerative colitis and Crohn's disease by measuring the levels of certain bacterial taxa and proteins collected from the gut.
Description

This application is a nonprovisional of U.S. provisional patent application 61/781,564 filed Mar. 14, 2013, the specification of which is hereby incorporated by reference.


TECHNICAL FIELD

This invention relates generally to methods and compounds for the diagnosis and treatment of inflammatory bowel disease (IBD).


BACKGROUND

An intricate and essential partnership is established early in life between the host and the intestinal microbiome, assuring the maintenance of microbiota homeostasis. Disturbance of this partnership is often associated with various pathological conditions including inflammatory bowel diseases (IBD) (Cho, I. & Blaser, M. J. The human microbiome: at the interface of health and disease. Nature reviews. Genetics 13, 260-270, doi:10.1038/nrg3182 (2012)). The microbiota of IBD patients are characterized by a decreased prevalence of protective microorganisms (i.e. Clostridium IXa and IV groups) and an expansion of detrimental bacteria (i.e. Enterobacteriaceae/Escherichia coli) (Manichanh, C., Borruel, N., Casellas, F. & Guarner, F. The gut microbiota in IBD. Nature reviews. Gastroenterology & hepatology 9, 599-608, doi:10.1038/nrgastro.2012.152 (2012).


Inflammatory Bowel Disease encompasses two principal conditions: ulcerative colitis (UC) and Crohn's disease (CD). Some patients have features of both subtypes and are classified as IBD-undefined (IBD-U) (Gastroenterology, 2007. 133(5): p. 1670-89). UC is defined by continuous mucosal inflammation starting in the rectum and restricted to the colon while CD inflammation can occur anywhere in the gastrointestinal tract, involves full thickness of the bowel wall and often with skip lesions (Gastroenterol Clin North Am, 2009. 38(4): p. 611-28; Gastroenterology, 2007. 133(5): p. 1670-89). Recent attempts to find new markers for IBD subtypes, such as conventional antibodies, have fared very poorly at differentiating colonic CD versus UC. As treatments and responses to medical therapies differ between CD and UC (J Pediatr Gastroenterol Nutr, 2010, S1-S13. The American journal of gastroenterology, 2011. 106 Suppl 1: p. S2-25; quiz S26. Gastroenterol Clin North Am, 2009. 38(4): p. 611-28) there is an urgent need for biomarkers to differentiate between CD and UC.


The primary tool used for both diagnosis and IBD management is endoscopy (World J Gastrointest Endosc, 2012. 4(6): p. 201-11). Endoscopy enables both visualization of the mucosa and access for mucosal biopsies to diagnose disease, to define disease extent and activity, and to monitor disease progression. The diagnostic accuracy from colonoscopy ranges from 60 to 74% (J Clin Pathol, 2002. 55: p. 955-60). Accurate and early diagnosis is essential for proper disease management. The goal of IBD treatment is to bring active disease into remission and to prevent follow-up relapse (flare-ups). The choice of treatment depends on disease type (CD versus UC), disease location, severity of disease, disease complications and individual host factors (e.g. nutritional and growth status, pubertal status, child's age and size, medication allergies) (J Pediatr Gastroenterol Nutr, 2010, S1-S13. The American journal of gastroenterology, 2011. 106 Suppl 1: p. S2-25; quiz S26. Gastroenterol Clin North Am, 2009. 38(4): p. 611-28). Current drug therapies consist of aminosalycylates, immune-modulators, corticosteroids, antibiotics and biological therapies (i.e. anti-TNFα monoclonal antibodies). The optimum therapeutic regimen for maintaining a disease free state still remains to be determined and the effectiveness of these drugs significantly differs between CD and UC (J Pediatr Gastroenterol Nutr, 2010, S1-S13. The American journal of gastroenterology, 2011. 106 Suppl 1: p. S2-25; quiz S26. Gastroenterol Clin North Am, 2009. 38(4): p. 611-28). For example, 5-aminosalicylic acid (5-ASA) drugs are moderately effective at inducing remission and preventing relapse in mild-to-moderate-active UC, while they are not recommended in the management of active CD (The American journal of gastroenterology, 2011. 106 Suppl 1: p. S2-25; quiz S26). Methotrexate is good evidence for use as maintenance therapy to prevent relapse in CD however, there is no evidence for its use in UC (The American journal of gastroenterology, 2011. 106 Suppl 1: p. S2-25; quiz S26). Greater doses of anti-TNFα therapies at more frequent intervals are being just now recognized to be required for successful treatment of severe UC as compared to standard treatment protocols in use for CD. One third of the cost associated with IBD is due to medical therapies (CCFC. 2008, report. p. 1-101) stressing the economic importance of an effective treatment and thereby an accurate diagnosis.


While the etiology of IBD is unknown, the gut microbiota is emerging as a key player in disease development and/or chronicity. Genome wide association studies in both adults and pediatric patients have identified novel IBD-associated genes but only define 25% of the genetic risk for developing IBD and excepting for very young infants (i.e. <2 years of age), no unique genes have been discovered that define pediatric IBD from adult-onset IBD. IBD is a complex polygenic disease involving multiple risk gene loci (Nature genetics, 2008. 40(8): p. 955-62. Nature genetics, 2009. 41(12): p. 1335-40. Nature genetics, 2010. 42(4): p. 332-7). These loci encode genes involved in innate and adaptive immunity, autophagy, and maintenance of epithelial barrier integrity for those genes that have known function. While these studies have shown us that multiple pathways are involved in the pathogenesis of IBD, we remain surprisingly ignorant on the root cause(s) and pathogenesis of IBD. A prevailing hypothesis is that IBD development is a consequence of functional abnormalities in the interplay between the intestinal microbiota and the host (World journal of gastroenterology: WJG, 2011. 17(5): p. 557-66). Some of the best evidence that the gut microbiota plays a key role in IBD comes from animal model studies (World journal of gastroenterology: WJG, 2011. 17(5): p. 557-66. Cell, 2007. 131(1): p. 33-45. Inflamm Bowel Dis, 2007. 13(12): p. 1457-66). Although the experimental animal models of IBD do not exactly mimic human IBD, these studies have shown that the development of the disease is dependent on the presence of resident bacteria (Cell, 2007. 131(1): p. 33-45. Inflamm Bowel Dis, 2007. 13(12): p. 1457-66). The loss of the transcriptional factor T-bet in mice, which regulates the differentiation and function of immune system cells, was shown to promote the microbiota to become colitogenic. Moreover, the induced colitis could be transmitted to other genetically intact hosts by vertical transfer of the colitogenic microbiota (Cell, 2007. 131(1): p. 33-45). Numerous studies have revealed alterations in the composition of the gut microbiota of patients with IBD (Proc Natl Acad Sci USA, 2007. 104(34): p. 13780-5. (9) Nature, 2010. 464(7285): p. 59-65. (10) Cell, 2012. 148(6): p. 1258-70; World journal of gastroenterology: WJG, 2011. 17(5): p. 557-66). However, we do not know what triggers IBD and the resulting gut microbiota dysbiosis and we have only a rudimentary understanding of the interplay between the gut microbiota and the host. Clearly, studies that longitudinally follow gut microbiota dysbiosis in humans during flare-ups and remissions could contribute important insights into the clinical significance of the gut microbiota composition.


IBD symptoms may include bloody diarrhea, abdominal pain, cramping, fatigue, various nutritional deficiencies including iron deficiency anemia, bone health problems and weight loss (Archives of disease in childhood, 2006). In children poor linear growth is also common. The onset of symptoms is slow, indolent and non-specific and so the disease may be present in certain regions of the bowel for very long periods of time prior to diagnosis. Following diagnosis, this chronic, life-long disease is characterized by episodes of flare-up and remission (quiescent, symptom-free state) (Gastroenterol Clin North Am, 2009. 38(4): p. 611-28; Archives of disease in childhood, 2006). The current therapeutic treatments aim to stop mucosal inflammation so as to maintain the quiescent period and to reduce flare-ups to reduce permanent bowel damage and alleviate the complications of disease. Corticosteroids (prednisone) remain a mainstay of treatment for IBD despite the well-known side effects of this medication (Journal of Crohn's & colitis, 2012. 6(4): p. 492-502). Alternatively, enteral nutrition (EN) is more commonly being used as a primary therapy in lieu of prednisone to induce CD remission (Current opinion in clinical nutrition and metabolic care, 2011. 14(5): p. 491-6). However, it is more difficult for most patients to adhere to these protocols that involve enteral formulas alone without eating foods for many weeks at a time. It is apparent that the microbiota composition correlates with disease and that an “abnormal” microbiota contributes to (if not triggers) mucosa alterations and immune system malfunctions (World journal of gastroenterology: WJG, 2011. 17(5): p. 557-66). It follows that interventions aimed at restoring microbiota equilibrium could promote health and/or prevent flare-up. Moreover, given that each patient is have a unique gut microbiota composition it follows that any interventions aimed at manipulating the gut microbiota should preferably be disease and patient-specific.


In view of the above there is a need for better diagnostic assays and treatments for the management of IBD.


SUMMARY

There is provided assays and methods to diagnose and treat IBD as well as to classify gut samples into IBD, UC or CD samples. There is also provided a device for classifying gut samples into IBD, UC or CD samples.


In an embodiment there is provided an assay comprising the steps of measuring a level of proteobacteria or H2S producing bacteria or both in a gut microbioata sample from a human subject to identify the likelihood of the human subject having inflammatory bowel disease (IBD), and comparing the level of proteobacteria or H2S producing bacteria or both to a reference level of proteobacteria or H2S producing bacteria or both from gut microbiota samples of healthy human subjects, wherein a level of proteobacteria or H2S producing bacteria or both higher than the reference level is indicative of disease.


In another embodiment there is provided an assay comprising the steps of measuring a level of A. parvulum in a gut microbiota sample from a human subject to identify the likelihood of the human subject having IBD, and comparing the level of A. parvulum to a reference level of A. parvulum from gut microbiota samples of healthy human subjects, wherein a level of A. parvulum higher than the reference level is indicative of disease.


In a further embodiment there is provided an assay comprising the steps of measuring a level of butyrate producing bacteria in a gut microbiota sample from a human subject to identify the likelihood of the human subject having IBD, and comparing the level of butyrate producing bacteria to a reference level of butyrate producing bacteria from gut microbiota samples of healthy human subjects, wherein a level of butyrate producing bacteria lower than the reference level is indicative of disease.


Advantageously, the invention provides a method for distinguishing between patients with UC or CD.


In yet a further embodiment there is provided an assay for determining a severity of CD disease comprising measuring a level of one or more bacterial taxa selected from Carnobacteriaceae, Granulicatella, Mogibacterium, Proprionibacterium, Bacillaceae and Atopobium in a gut microbioata sample from the human subject wherein a level higher than a predetermined level is indicative of moderate or severe inflammation.


There is further provided an assay comprising the steps of measuring a level of sulfur dioxygenase (ETHE1), thiosulfate sulfur transferase (TST), cytochrome c oxidase subunit IV, sulfide dehydrogenase (SQR) and complexes III and IV of mithochondrial respiratory chain in a gut mucus sample from a human subject to identify the likelihood of the human subject having IBD, and wherein a lower level relative to a reference level from a healthy subject is indicative of disease.


In another aspect there is provided a method of treating IBD in a patient the method comprising: performing an assay to determine the presence of disease (IBD or UC or CD) and administering to the patient a pharmaceutically effective amount of a compound selected from aminosalycylates, immunomodulators, anti-integrins, anti-cytokines, enteral feed programs, steroids, corticosteroids, antibiotics, anti-TNFα, bismuth or a combination thereof.


These and other embodiments of the invention are further described below with reference to the Drawings and the Detailed Description.





BRIEF DESCRIPTION OF THE DRAWINGS

The invention is better understood by way of the following detailed description of embodiments of the invention with reference to the appended drawings, in which:



FIG. 1 A is a PLS-DA of CD patients with severe inflammation (n=23) against CD patients with mild inflammation (n=9), to confirm the validation of the PLS-DA models, permutation tests (n=1000) were performed and the corresponding p value for prediction accuracy calculated.



FIG. 1 B is a biplot analysis of the first two components of the PLS-DA model in panel A showing the significant taxa relative to disease activity (arrows).



FIG. 1 C is a graph showing the Abundance of A. parvulum relative to total bacteria as determined by quantitative PCR as a function of CD severity (n=13 for controls and severe CD; n=6 for mild and moderate CD; statistical comparison by Kruskal-Wallis test; diamond indicates minimum or maximum; cross indicates mean; horizontal bar indicates median).



FIG. 2 A is a photomicrograph showing the cecum and colon from gnotobiotic 1110−/− mice that were either associated or not with A. parvulum.



FIG. 2 B is a photomicrograph showing inflammation monitored macroscopically with a murine endoscope.



FIG. 2 C is a photomicrograph showing representative histological sections of the distal colon and cecum.



FIG. 2 D is a graph showing a blinded histological score of inflammation (n=6 to 7 per group; horizontal lines indicate mean and crosses indicate median; comparison by Mann-Whitney two tailed test).



FIG. 3 A is a functional annotation (“cellular component’) analysis of the differentially expressed proteins; the 10 most significantly enriched functional groups (GO terms) are shown (P<10−13); asterisks denote classifications that were significantly enriched compared to the whole proteomic dataset, *P<0.05 and ***P<0.001 (Fisher's exact test).



FIG. 3 B is a PLS-DA analysis of CD patients as a function of disease activity and controls (10 fold external validation of the model was performed on separate holdout validation sets of 5 randomly selected samples showing prediction accuracy of 75%).



FIG. 3 C qRT-PCR analysis of TST normalized to control, n=5 for UC, 13 to 15 for CD and 10 to 15 for controls Ns, not significant statistical significance was assessed using a two-tailed Mann-Whitney test.



FIG. 3 D qRT-PCR analysis cytochrome c oxidase subunit IV (hCOX41) normalized to control, n=5 for UC, 13 to 15 for CD and 10 to 15 for controls Ns, not significant statistical significance was assessed using a two-tailed Mann-Whitney test.



FIG. 3 E qRT-PCR analysis sulfide dehydrogenase (SQR) normalized to control, n=5 for UC, 13 to 15 for CD and 10 to 15 for controls Ns, not significant statistical significance was assessed using a two-tailed Mann-Whitney test.



FIG. 4 A are representative murine endoscopies of II10−/− mice associated or not with A. parvulum, treated or not with bismuth and kept under SPF conditions for 6 weeks.



FIG. 4 B are blinded inflammation scores (n=7 to 8 per group) for II10−/− mice under SPF conditions, horizontal lines indicate means and crosses indicate median statistical significance was assessed using a Kruskal-Wallis test with a Dunn's post hoc test using the Conover-Iman procedure.



FIG. 4 C is a graph of number of GALT foci of II10−/− mice associated or not with A. parvulum and treated or not with bismuth, and kept under gnotobiotic or SPF conditions (n=6 to 11 per group) horizontal lines indicate means and crosses indicate median. Statistical significance was assessed using a Kruskal-Wallis test with a Dunn's post hoc test.



FIG. 4 D are representative histological Swiss-rolled sections of the colon showing GALT foci in gnotobiotic mice arrows indicate GALT foci.



FIG. 4 E is PCA analysis of microbiota from II10−/− mice kept under SPF conditions (light blue), bismuth-treated (blue), associated with A. parvulum (red), and associated with A. parvulum and treated with bismuth (green).



FIG. 5 A is a plot of the size of the core microbiota of control subjects, and CD and UC patients; (OTU: operating taxonomic unit).



FIG. 5 B is phylogenetic tree of the microbial taxa detected in at least 75% of the samples within each group wherein a total of 241 core OTUs were detected, which represent 90.2%±8.3% of the microbial population, the figure was generated using the iTOL (Interactive Tree of Life) web package in which taxa marked with inner circle were identified as members of the core microbiota of the control subjects, taxa marked with middle and outer circles were identified as members of the core microbiota of the UC or CD patients respectively, CD and UC microbial communities are characterized by a smaller core microbiota as compared to control with 179, 172 and 214 core OTUs for CD, UC and control subjects respectively.



FIG. 6 A represents the average relative abundance of bacterial phyla identified in patients with Crohn's disease (CD; n=9) and Ulcerative Colitis (UC; n=8) and control subjects (n=9); similar profiles were obtained with reads generated using Hiseq2500 sequencing.



FIG. 6 B represents the change in relative abundance of Proteobacteria (mean±SEM) in controls, CD and UC patients; Mann-Whitney two-tailed test was applied for statistical pairwise comparison.



FIG. 7 A represents the relative abundances of the bacterial taxa obtained from the analysis of the 454 pyrosequencing reads were analysed by linear discrimination analysis (LDA) followed by a Wilcoxon Mann-Whitney test to assess the effect size using LEfSe; histogram of the LDA effect size score for CD-specific differentially abundant taxa (n=9 for CD and controls).



FIG. 7 B is a histogram of the LDA effect size score for UC-specific differentially abundant taxa (n=9 for control and n=8 for UC), as shown in panel A, Atopobium was identified as a biomarker of CD; 454-pyrosequencing reads assigned as Atopobium by QIIME analysis were retrieved and found to match to A. parvulum following alignment of the reads against the RDB and NCBI databases (the aligned region covered the entire 454 sequence length with >99% sequence identity to A. parvulum and did not align to any other known bacterial species).



FIG. 8 A is a Functional annotation analysis of the differentially expressed proteins for BP: biological processes in which the 10 most significantly enriched functional groups (GO terms) are shown (p<10−13); all classifications were significantly enriched compared to the whole proteomic dataset with P<0.05 (Fisher's exact test).



FIG. 8 B is a Functional annotation analysis of the differentially expressed proteins for MF: molecular functions in which the 10 most significantly enriched functional groups (GO terms) are shown (p<10−13); all classifications were significantly enriched compared to the whole proteomic dataset with P<0.05 (Fisher's exact test).



FIG. 8 C is a Functional annotation analysis of the differentially expressed proteins for KEGG pathways in which the 10 most significantly enriched functional groups (GO terms) are shown (p<10−13); all classifications were significantly enriched compared to the whole proteomic dataset with P<0.05 (Fisher's exact test).



FIG. 9 A is a PLS-DA analysis of the mitochondrial protein profiles classified as CD patients (black) and control subjects (gray), the model was calculated based on the 95 differentially expressed mitochondrial proteins as determined by an ANOVA test and by selecting the proteins with the corresponding GO term (by using the DAVID functional GO annotation program), an acceptable PLS-DA model was obtained with 2 components (predictive ability parameter [Q2 cum]=0.77, goodness-of-fit parameter [R2Y cum]=0.92).



FIG. 9 B is a PLS-DA analysis of the 96 differentially expressed mitochondrial proteins from CD patients classified as a function of disease activity (mild, moderate and severe), arobust model with good predictive power was generated with four components (Predictive ability parameter [Q2 cum]=0.44, goodness-of-fit parameter [R2Y cum]=0.94).



FIG. 10 is a model of mitochondrial H2S catabolism where the membrane bound sulfide dehydrogenase (SQR) oxidizes sulfide (H2S) to persulfide (formed at one of the SQR's cysteines; SQR-SSH), the electrons are transferred to the mitochondrial respiratory chain (cytochrome c oxidase complex III and IV) via the quinone pool (Qox/Qred) the sulfur dioxygenase, ETHE1, oxidizes persulfides to sulfites (H2SO3) in the mitochondrial matrix, rhodanese (sulfur trans.) catalyzes the final reaction, which produces thiosulfite (H2S2O3) by transferring a second persulfide from the SQR to sulfite, the cytochrome c oxidase subunit IV (COX-IV) is required for the assembly of the cytochrome c oxidase, rhodanese comprises two iso-enzymes: thiosulfate sulfurtransferase (TST) and mercaptopyruvate sulfurtransferase (MST).



FIG. 11 A is a graph of Cxcl1 cytokine expression in conventionalized II10−/− mice (129/SvEv II10−/− mice), measured by qRT-PCR, which were associated or not with A. parvulum and kept under SPF conditions (n=7 to 8 per group), total RNA was extracted from colonic intestinal tissues 6 weeks post-association and A Mann-Whitney U test was performed to assess statistical significance, the horizontal lines indicate the mean and error bars the SD, n.s., non-significant.



FIG. 11 B is a graph of 11-17 cytokine expression in conventionalized II10−/− mice (129/SvEv II10−/− mice), measured by qRT-PCR, which were associated or not with A. parvulum and kept under SPF conditions (n=7 to 8 per group), total RNA was extracted from colonic intestinal tissues 6 weeks post-association and A Mann-Whitney U test was performed to assess statistical significance, the horizontal lines indicate the mean and error bars the SD, n.s., non-significant.



FIG. 11 C is a graph of 11-12 cytokine expression in conventionalized II10−/− mice (129/SvEv II10−/− mice), measured by qRT-PCR, which were associated or not with A. parvulum and kept under SPF conditions (n=7 to 8 per group), total RNA was extracted from colonic intestinal tissues 6 weeks post-association and A Mann-Whitney U test was performed to assess statistical significance, the horizontal lines indicate the mean and error bars the SD, n.s., non-significant.



FIG. 11 D is a graph of 1110 cytokine expression in conventionalized II10−/− mice (129/SvEv II10−/− mice), measured by qRT-PCR, which were associated or not with A. parvulum and kept under SPF conditions (n=7 to 8 per group), total RNA was extracted from colonic intestinal tissues 6 weeks post-association and A Mann-Whitney U test was performed to assess statistical significance, the horizontal lines indicate the mean and error bars the SD, n.s., non-significant.



FIG. 12 A is a hyperplasia score of 129/SvEv II10−/− mice mono-associated or not with A. parvulum and treated or not with bismuth kept under gnotobiotic conditions (n=6 to 11 per group), error bars indicate SD.



FIG. 12 B represents levels of chromosomal DNA was extracted from stool pellets obtained 6 week after mono-association or not of 129/SvEv II10−/− mice with A. parvulum, colonization level was estimated using real-time qPCR and reported as the number of 16S rDNA gene copies per mg of stool, error bars indicate SEM, for panels A and B, horizontal lines indicate means. Statistical significance was assessed using a Mann-Whitney U-test.



FIG. 13 shows the relative quantification of butyryl-CoA:CoA transferase (BCoAT) gene using qPCR. BCoAT was quantified from control and IBD samples. CD and UC samples were subclassified normal and inflamed based on colon appearance during sample collection. Result is expressed as number of BCoAT genes per 16S rRNA gene. Error bars represent the standard error of the mean.



FIG. 14 A shows the diversity of butyrate-producing bacteria. Number of Observed Operational Taxonomic Units (OTUs) from BCoAT sequencing at 95% sequence similarity.



FIG. 14 B shows Alpha diversity represented by Chao1 estimated OTUs (left panel) and Shannon diversity index (right panel). Number of reads was equalized between samples at 4,600 reads. Control (blue bar), CD (red bar), and UC (orange bar).



FIG. 14 C shows Beta diversity presented by two-dimensional principal coordinates analysis (PCoA) plot of weighted UniFrac distance. Left plot represents control group and CD patients beta diversity, and right plot is showing control group and UC patients beta diversity. Percentage of variance explained by each component is presented under each axis. Control samples (green squares), CD samples (red triangle), and UC samples (blue circles).



FIG. 15 A-D shows the identified butyrate-producing bacteria at the species level using BCoAT sequencing. (A-C) Pie charts of relative abundance of butyrate producers in each group combined. (A) Control group, (B) CD patients, and (C) UC patients. (D) Relative abundance of butyrate producers in individual samples. Each stacked bar represent one subject.



FIG. 16 A-F shows the butyrate-producers species with differential abundance in IBD. (A-F) The relative abundance of Eubacterium rectale, Faecalibacterium prausnitzii, Roseburia inulinivorans, total unclassified OTUs, unclassified OUT_34, and unclassified OUT_43; respectively. Each bar represents the average relative abundance in one group. Error bars represent the standard error of the mean. *, P<0.05; **, P<0.01; ***, P<0.001.



FIG. 17 A-D shows the butyrate producers' genera identified by V6 hypervariable region of 16S rRNA sequencing. (A-C) Pie charts of relative abundance of butyrate producers in each group combined. (A) Control group, (B) CD patients, and (C) UC patients. (D) Relative abundance of butyrate producers in individual samples. Each stacked bar represent one subject.



FIG. 18 A-F shows the quantitative PCR analysis of key butyrate producers. Eubacterium rectale and Faecalibacterium prausnitzii were quantified using BCoAT and 16S rRNA primers. Roseburia was quantified using 16S rRNA primers. (A-E) represent the ΔCt of targeted butyrate producers relative to total bacteria 16S rRNA. (F) Ct for total bacteria 16S rRNA is similar between groups. Error bars represent the standard error of the mean. *, P<0.05; **, P<0.01; ***, P<0.001; ****, P<0.0001.



FIG. 19 A shows the quantitative PCR analysis of Faecalibacterium prausnitzii from stool samples. Faecalibacterium prausnitzii was quantified using 16S rRNA primers abd represents the ΔCt of F. prausnitzii relative to total bacteria 16S rRNA.



FIG. 19 B shows Ct for total bacteria 16S rRNA is similar between groups. Error bars represent the standard error of the mean.





DETAILED DESCRIPTION

In the present description by microbiota it is meant an ensemble of microorganisms residing in an environment and in particular by gut microbiota it is meant microorganisms found in any part of the alimentary canal from lips to the anus.


By patients having Inflammatory bowel disease (IBD) it is meant patients with ulcerative colitis (UC) or patients with Crohn's disease (CD) or IBD-undefined (IBD-U).


By level or abundance of bacteria or bacterial taxa it is meant a level or abundance obtained by a means to quantify bacteria such as culture based methods, flow cytometry, microscopy, quantitative DNA analysis and any other means that would be obvious to a person skilled in the art.


By severity of the disease it is meant a level of symptoms as described in disease activity index such Crohn's disease activity index (CDAI), Pediatric Crohn's disease activity index (PCDAI) Harvey-Bradshaw index, Ulcerative colitis activity index (UCAI), Pediatric Ulcerative colitis activity index (PUCAI), Paris classification of pediatric Crohn's disease and the like. For example severe CD corresponds to a score of 450 in the CDAI index.


By “core” it is meant the bacterial taxa that are conserved between individuals (that are present in two or more individuals).


In an aspect of the invention there is provided a method in which IBD can be detected by measuring the levels (or relative abundance) of certain bacterial taxa in samples from the gut of patients. Microbiota samples from the gut may be obtained from stools, intestinal mucosal biopsies, gut lavage or combination thereof. In an embodiment of the invention, microbiota samples are collected such as to comprise the microbiota from the mucosa-luminal interface of the gut.


In one embodiment of the invention, the collection can be performed during endoscopy by flushing a physiological solution, such as sterile saline solution or sterile water, onto the mucosa to remove the strongly adherent mucus layer overlying the intestinal mucosal epithelial cells and the microbial community embedded within the mucus layer. Aspirates are then collected directly through a colonoscope at a specific location in the gut as for example from the terminal ileum right colon and left colon and the samples are preferably immediately put on ice right in the endoscopy suite. For example the following steps can be performed: 1) a regular protocol of bowel clean out in preparation for colonoscopy is first applied to the patient, 2) then the colonoscope (“scope”) is advanced to the ascending colon or a region of the colon distal to that of interest, 3) suction out fluid and particulate matter, using either the scope's wash system or with a syringe through biopsy port, 4) flush sterile water onto mucosa until shards of mucus are dislodged, 5) aspirate mucus containing fluid into sterile trap through scope aspiration system, 6) remove the trap from scope suction and cap it and immediately place on ice, 7) advance the scope to more proximal region of interest and repeat steps 3-6, 8) transport traps with mucus to lab within 15 minutes of collection. The sample can then be analyzed at the point of care or transferred to a laboratory. The samples can also be further processed and then stored at −80° C.


Collection of the gut microbiota can also be performed on stools. Collection of bacteria from stools is known in the art. In the case of fecal microbiota collection/analysis, fresh stools may be collected and immediately processed and stored at −80° C. for DNA extraction and sequence/quantification as part of a bacterial analysis as further described below.


Samples containing gut microbiota collected as described above can be assayed for determining their microbial composition. Identification of the bacteria present in samples can be performed using DNA sequencing techniques as described in the examples below. In one embodiment, total DNA can be extracted from intestinal aspirates or stool samples. The protocol may comprise the extraction of total DNA using an extraction step with mechanical disruption. The extracted DNA can then be subjected to sequencing to identify bacteria by comparing the sequences to sequences contained in databases. In a preferred embodiment metagenomic DNA can be subjected to multiplexed massively parallel sequencing on the hypervariable V6 region of the 16S rRNA gene. It is appreciated that the sequencing of regions other than the hypervariable V6 region of the 16S rRNA gene can be used provided that such regions provide discriminating power (taxonomic resolution) for at least some bacterial taxa or operational taxonomic units (OTU's) and in particular for bacterial taxa that are preferentially associated with IBD as is further described below.


It will also be appreciated that other methods can be used to identify bacteria from the gut samples including but not limited to microscopy, metabolites identification, Gram staining, flow cytometry, immunological techniques (antibodies), culture-based methods such a colony forming unit counting and the like as would be known to a person skilled in the art.


In an aspect of the invention the relative abundance of certain bacterial taxa namely phylum, class, order, family, genus or species or combination thereof in the gut (gut microbiota profile) of patients is used to assess the presence or absence of IBD disease. It has been found that the IBD microbiota is characterized by a smaller core as compared to controls (FIG. 5A-B and Table 1), indicating a loss of microbiota homeostasis. Also, the IBD microbiota is characterized by a depletion of butyrate producing microbes together with an increased abundance of H2S-generating bacteria. For example, increase in the levels of H2S producers such as Fusobacterium nucleatum, Veillonella parvula, and Atopobium parvulum is indicative of disease.


Assessment of the presence of CD and UC disease in a human subject can be achieved by measuring the relative abundance of taxa as exemplified in table 1. In this particular example, microbial operational taxonomic units (OTUs) that were detected in all the samples within each group and that vary significantly in abundance between CD, UC and/or controls are listed. The number of 16S rDNA reads in each sample was normalized by random subsampling to 500,000. Minimum and maximum correspond to the minimum and maximum number of reads obtained; mean corresponds to the mean of the number of reads obtained. P values were generated using a Kruskal-Wallis test with a Dunn's post hoc test. “p|Control” indicates the P values obtained by comparison to the controls; “p|UC” and “p|CD” indicate the P values obtained by comparison to the UC and CD patients respectively. Values in bold indicate significance (P<0.05). From the table it can be seen that certain taxa are more or less abundant in patients with disease than in healthy controls. Furthermore there it is also possible to distinguish between CD and UC based on the relative abundance.









TABLE 1







Core OTUs that varies significantly in abundance in at least one of the three pairwise


comparisons performed (controls vs. CD; controls vs. UC; and CD vs. UC).















OTU |




Std.
p |
p |
p |


Variable
Taxonomy | Variable
Min
Max
Mean
deviation
Control
UC
CD


















177005 |
k_Bacteria;
0.000
12876.000
1590.857
3303.847
1
0.304
0.022


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Blautia;



s_ | Control


177005 |
k_Bacteria;
1.000
8538.000
829.071
2266.987
0.304
1
0.388


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Blautia;



s_ | UC


177005 |
k_Bacteria;
0.000
24758.000
870.135
4086.298
0.022
0.388
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Blautia;



s_ | CD


541301 |
k_Bacteria;
0.000
34876.000
2923.571
7856.412
1
0.265
0.332


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Porphyromonadaceae;



g_Parabacteroides;



s_ | Control


541301 |
k_Bacteria;
2.000
4594.000
400.357
1221.873
0.265
1
0.038


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Porphyromonadaceae;



g_Parabacteroides;



s_ | UC


541301 |
k_Bacteria;
1.000
10433.000
1166.622
2439.874
0.332
0.038
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Porphyromonadaceae;



g_Parabacteroides;



s_ | CD


258691 |
k_Bacteria;
0.000
40719.000
2275.238
8846.849
1
0.812
0.038


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesovatus |



Control


258691 |
k_Bacteria;
1.000
86361.000
6325.500
23037.262
0.812
1
0.122


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesovatus |



UC


258691 |
k_Bacteria;
1.000
37102.000
3148.459
7654.185
0.038
0.122
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesovatus |



CD


182122 |
k_Bacteria;
3.000
13653.000
1649.619
3463.599
1
0.034
0.077


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | Control


182122 |
k_Bacteria;
0.000
5379.000
518.643
1417.505
0.034
1
0.427


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | UC


182122 |
k_Bacteria;
1.000
7805.000
691.216
1475.854
0.077
0.427
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | CD


261912 |
k_Bacteria;
12.000
23969.000
7682.238
8341.342
1
0.043
0.540


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Dorea;



s_Doreaformicigenerans |



Control


261912 |
k_Bacteria;
24.000
7473.000
1846.143
2506.511
0.043
1
0.091


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Dorea;



s_Doreaformicigenerans |



UC


261912 |
k_Bacteria;
4.000
92806.000
10038.811
17773.654
0.540
0.091
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Dorea;



s_Doreaformicigenerans |



CD


585419 |
k_Bacteria;
2.000
860.000
111.000
201.434
1
0.003
0.114


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | Control


585419 |
k_Bacteria;
17.000
29801.000
4900.714
9206.483
0.003
1
0.059


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | UC


585419 |
k_Bacteria;
5.000
14260.000
1293.811
3473.360
0.114
0.059
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | CD


566952 |
k_Bacteria;
1.000
559.000
152.381
171.608
1
0.011
0.137


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Clostridium;



s_ | Control


566952 |
k_Bacteria;
0.000
378.000
41.429
101.442
0.011
1
0.137


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Clostridium;



s_ | UC


566952 |
k_Bacteria;
0.000
720.000
106.108
165.187
0.137
0.137
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Clostridium;



s_ | CD


303772 |
k_Bacteria;
0.000
149466.000
8653.333
32546.409
1
0.163
0.177


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | Control


303772 |
k_Bacteria;
1.000
16984.000
1460.286
4486.377
0.163
1
0.007


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | UC


303772 |
k_Bacteria;
0.000
2852.000
193.811
613.649
0.177
0.007
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | CD


145149 |
k_Bacteria;
1.000
350.000
75.524
107.340
1
0.012
0.170


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | Control


145149 |
k_Bacteria;
2.000
15569.000
4072.786
6042.303
0.012
1
0.119


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | UC


145149 |
k_Bacteria;
1.000
9591.000
811.892
1841.236
0.170
0.119
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Veillonellaceae;



g_Veillonella;



s_ | CD


362168 |
k_Bacteria;
1.000
10035.000
709.286
2194.846
1
0.540
0.039


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | Control


362168 |
k_Bacteria;
2.000
17437.000
1507.500
4604.990
0.540
1
0.261


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | UC


362168 |
k_Bacteria;
2.000
35846.000
1950.054
6025.223
0.039
0.261
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | CD


470973 |
k_Bacteria;
3.000
73975.000
5461.905
16369.874
1
0.106
0.637


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Ruminococcus;



s_Ruminococcustorques |



Control


470973 |
k_Bacteria;
3.000
55265.000
4591.643
14678.969
0.106
1
0.029


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Ruminococcus;



s_Ruminococcustorques |



UC


470973 |
k_Bacteria;
0.000
109867.000
8736.757
20949.142
0.637
0.029
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_Ruminococcus;



s_Ruminococcustorques |



CD


138006 |
k_Bacteria;
0.000
2913.000
464.476
900.379
1
0.117
0.229


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Rikenellaceae;



g_Alistipes;



s_ | Control


138006 |
k_Bacteria;
0.000
1393.000
193.286
411.329
0.117
1
0.006


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Rikenellaceae;



g_Alistipes;



s_ | UC


138006 |
k_Bacteria;
1.000
7020.000
795.243
1586.474
0.229
0.006
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Rikenellaceae;



g_Alistipes;



s_ | CD


188900 |
k_Bacteria;
11.000
9087.000
1275.952
2844.824
1
0.266
0.042


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Ruminococcaceae;



g_Faecalibacterium;



s_ | Control


188900 |
k_Bacteria;
3.000
87401.000
7661.214
23050.226
0.266
1
0.585


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Ruminococcaceae;



g_Faecalibacterium;



s_ | UC


188900 |
k_Bacteria;
2.000
98077.000
8299.324
20200.719
0.042
0.585
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Ruminococcaceae;



g_Faecalibacterium;



s_ | CD


64396 |
k_Bacteria;
0.000
674.000
86.952
185.286
1
0.050
0.013


Control
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | Control


64396 |
k_Bacteria;
0.000
41529.000
3892.429
11049.181
0.050
1
0.993


UC
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | UC


64396 |
k_Bacteria;
1.000
164748.000
6092.135
27224.424
0.013
0.993
1


CD
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | CD


196731 |
k_Bacteria;
1.000
787.000
174.333
238.985
1
<0.0001
0.346


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | Control


196731 |
k_Bacteria;
0.000
0.000
0.000
0.000
<0.0001
1
<0.0001


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | UC


196731 |
k_Bacteria;
0.000
8748.000
666.892
1933.413
0.346
<0.0001
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | CD


469709 |
k_Bacteria;
4.000
113796.000
12561.190
32939.260
1
0.274
0.035


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesdorei |



Control


469709 |
k_Bacteria;
11.000
123061.000
14096.286
34346.257
0.274
1
0.529


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesdorei |



UC


469709 |
k_Bacteria;
2.000
221413.000
22583.189
42793.771
0.035
0.529
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_Bacteroidesdorei |



CD


183879 |
k_Bacteria;
0.000
25451.000
2080.381
5620.095
1
0.097
0.005


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | Control


183879 |
k_Bacteria;
1.000
26097.000
2375.786
6987.836
0.097
1
0.533


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | UC


183879 |
k_Bacteria;
0.000
2104.000
220.324
486.771
0.005
0.533
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Lachnospiraceae;



g_; s_ | CD


514611 |
k_Bacteria;
3.000
44090.000
2806.333
9573.507
1
0.154
0.019


Control
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Clostridiaceae;



g_Clostridium;



s_ | Control


514611 |
k_Bacteria;
0.000
3656.000
599.643
1154.057
0.154
1
0.633


UC
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Clostridiaceae;



g_Clostridium;



s_ | UC


514611 |
k_Bacteria;
0.000
173925.000
6113.162
29294.960
0.019
0.633
1


CD
p_Firmicutes;



c_Clostridia;



o_Clostridiales;



f_Clostridiaceae;



g_Clostridium;



s_ | CD


288565 |
k_Bacteria;
1.000
204469.000
18673.762
57773.500
1
0.343
<0.0001


Control
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | Control


288565 |
k_Bacteria;
0.000
21535.000
2582.714
6677.428
0.343
1
<0.0001


UC
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | UC


288565 |
k_Bacteria;
0.000
0.000
0.000
0.000
<0.0001
<0.0001
1


CD
p_Fusobacteria;



c_Fusobacteria;



o_Fusobacteriales;



f_Fusobacteriaceae;



g_Fusobacterium;



s_ | CD


171559 |
k_Bacteria;
1.000
697.000
109.762
201.308
1
<0.0001
<0.0001


Control
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | Control


171559 |
k_Bacteria;
0.000
0.000
0.000
0.000
<0.0001
1
1.000


UC
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | UC


171559 |
k_Bacteria;
0.000
0.000
0.000
0.000
<0.0001
1.000
1


CD
p_Bacteroidetes;



c_Bacteroidia;



o_Bacteroidales;



f_Bacteroidaceae;



g_Bacteroides;



s_ | CD









In table 2 results for relative abundance of taxa are presented. Taxa that varie significantly in abundance in at least one of the three pairwise comparisons performed (controls vs. CD; controls vs. UC; and CD vs. UC) are shown. In table 2, microbial OTUs that were detected in at least 75% of the samples within each group and that vary significantly in abundance between CD, UC and/or controls are listed. The number of 16S rDNA reads in each sample was normalized by random subsampling to 500,000. Minimum and maximum correspond to the minimum and maximum number of reads obtained; mean corresponds to the mean of the number of reads obtained. P values were generated using a Kruskal-Wallis test with a Dunn's post hoc test. “p|Control” indicates the P values obtained by comparison to the controls; “p|UC” and “p|CD” indicate the P values obtained by comparison to the UC and CD patients respectively. Values in bold indicate significance (P<0.05).
















TABLE 2





Variable
Minimum
Maximum
Mean
Std. dev.
p|Control
p|UC
p|CD






















PHYLUM









Firmicutes|Control
177570.000
471151.000
300049.381
95087.202
1

0.011

0.246


Firmicutes|UC
39474.000
359555.000
204394.000
82968.346

0.011

1
0.075


Firmicutes|CD
66149.000
447067.000
259197.486
102279.924
0.246
0.075
1


CLASS


Negativicutes|
23.000
16688.000
1555.333
3588.392
1
0.020

0.008



Control


Negativicutes|UC
95.000
39966.000
10613.357
14349.888
0.020
1
0.799


Negativicutes|CD
27.000
74088.000
8759.081
15370.608

0.008

0.799
1


Clostridia|Control
172358.000
466967.000
290267.667
92133.532
1

0.006

0.054


Clostridia|UC
37917.000
356417.000
186560.357
84887.686

0.006

1
0.173


Clostridia|CD
9293.000
413730.000
227236.162
109051.179
0.054
0.173
1


Verrucomicrobiae|
0.000
1098.000
85.381
251.309
1

0.014

0.296


Control


Verrucomicrobiae|
0.000
13.000
1.071
3.452

0.014

1
0.075


UC


Verrucomicrobiae|
0.000
2103.000
95.811
357.421
0.296
0.075
1


CD


Betaproteobacteria|
14.000
44407.000
4215.810
10576.956
1
0.120

0.002



Control


Betaproteobacteria|
27.000
74113.000
12641.071
23053.369
0.120
1
0.306


UC


Betaproteobacteria|
26.000
129123.000
14407.270
26352.478

0.002

0.306
1


CD


ORDER


Pasteurellales|
0.000
446.000
24.238
96.821
1

0.003


0.002



Control


Pasteurellales|UC
0.000
6141.000
661.857
1682.557

0.003

1
0.544


Pasteurellales|CD
0.000
998.000
75.135
190.092

0.002

0.544
1


Chromatiales|
0.000
1.000
0.048
0.218
1

0.003


0.009



Control


Chromatiales|UC
0.000
12.000
2.071
3.407

0.003

1
0.331


Chromatiales|CD
0.000
30.000
2.054
5.637

0.009

0.331
1


Burkholderiales|
9.000
44407.000
4150.000
10590.948
1
0.764

0.015



Control


Burkholderiales|UC
5.000
52910.000
6830.000
14990.496
0.764
1
0.073


Burkholderiales|CD
9.000
128561.000
13994.270
26364.706

0.015

0.073
1


Selenomonadales|
23.000
16688.000
1555.333
3588.392
1
0.020

0.008



Control


Selenomonadales|
95.000
39966.000
10613.357
14349.888
0.020
1
0.799


UC


Selenomonadales|
27.000
74088.000
8759.081
15370.608

0.008

0.799
1


CD


Clostridiales|Control
172357.000
466967.000
290267.619
92133.596
1

0.006

0.054


Clostridiales|UC
37917.000
356417.000
186558.857
84887.572

0.006

1
0.173


Clostridiales|CD
9292.000
413730.000
227233.919
109052.061
0.054
0.173
1


Hydrogenophilales|
0.000
0.000
0.000
0.000
1

0.008

0.660


Control


Hydrogenophilales|
0.000
2.000
0.286
0.611

0.008

1

0.011



UC


Hydrogenophilales|
0.000
2.000
0.054
0.329
0.660

0.011

1


CD


Oceanospirillales|
0.000
4777.000
381.000
1090.456
1

0.007

0.127


Control


Oceanospirillales|
1.000
8453.000
1310.500
2596.743

0.007

1
0.100


UC


Oceanospirillales|
0.000
2349.000
180.459
413.881
0.127
0.100
1


CD


Rhizobiales|Control
0.000
2.000
0.238
0.539
1

0.001

0.537


Rhizobiales|UC
0.000
170.000
23.786
55.134

0.001

1

0.002



Rhizobiales|CD
0.000
67.000
4.243
14.245
0.537

0.002

1


Verrucomicrobiales|
0.000
1098.000
85.381
251.309
1

0.014

0.296


Control


Verrucomicrobiales|
0.000
13.000
1.071
3.452

0.014

1
0.075


UC


Verrucomicrobiales|
0.000
2103.000
95.811
357.421
0.296
0.075
1


CD


FAMILY


Verrucomicrobiaceae|
0.000
1098.000
85.381
251.309
1

0.014

0.296


Control


Verrucomicrobiaceae|
0.000
13.000
1.071
3.452

0.014

1
0.075


UC


Verrucomicrobiaceae|
0.000
2103.000
95.811
357.421
0.296
0.075
1


CD


Staphylococcaceae|
0.000
4079.000
355.333
910.119
1
0.345

0.009



Control


Staphylococcaceae|
0.000
229.000
59.000
80.285
0.345
1
0.216


UC


Staphylococcaceae|
0.000
5529.000
222.270
934.404

0.009

0.216
1


CD


Lachnospiraceae|
1485.000
244085.000
73731.762
57312.252
1

0.006

0.058


Control


Lachnospiraceae|
1094.000
83206.000
26960.929
25720.429

0.006

1
0.178


UC


Lachnospiraceae|
388.000
160635.000
47128.243
45439.902
0.058
0.178
1


CD


Halomonadaceae|
0.000
55.000
2.667
11.993
1

0.014

0.263


Control


Halomonadaceae|
0.000
711.000
52.857
189.465

0.014

1
0.083


UC


Halomonadaceae|
0.000
12.000
0.811
2.246
0.263
0.083
1


CD


Pasteurellaceae|
0.000
446.000
24.238
96.821
1

0.003


0.002



Control


Pasteurellaceae|UC
0.000
6141.000
661.857
1682.557

0.003

1
0.544


Pasteurellaceae|CD
0.000
998.000
75.135
190.092

0.002

0.544
1


Paenibacillaceae|
0.000
181.000
32.000
44.159
1

0.014


0.009



Control


Paenibacillaceae|
0.000
53.000
8.286
14.435

0.014

1
0.658


UC


Paenibacillaceae|
0.000
617.000
23.730
100.869

0.009

0.658
1


CD


Listeriaceae|Control
0.000
11.000
1.381
3.008
1
0.310

0.011



Listeriaceae|UC
0.000
3.000
0.286
0.825
0.310
1
0.272


Listeriaceae|CD
0.000
2.000
0.054
0.329

0.011

0.272
1


Bradyrhizobiaceae|
0.000
2.000
0.190
0.512
1

0.006

0.604


Control


Bradyrhizobiaceae|
0.000
32.000
3.714
8.914

0.006

1

0.010



UC


Bradyrhizobiaceae|
0.000
57.000
3.216
11.814
0.604

0.010

1


CD


Methylococcaceae|
0.000
2.000
0.190
0.512
1

0.004

0.243


Control


Methylococcaceae|
0.000
351.000
30.929
94.613

0.004

1
0.032


UC


Methylococcaceae|
0.000
4.000
0.243
0.830
0.243
0.032
1


CD


Hydrogenophilaceae|
0.000
0.000
0.000
0.000
1

0.008

0.660


Control


Hydrogenophilaceae|
0.000
2.000
0.286
0.611

0.008

1

0.011



UC


Hydrogenophilaceae|
0.000
2.000
0.054
0.329
0.660

0.011

1


CD


GENUS



Porphyromonas|

0.000
1.000
0.143
0.359
1

0.002

0.443


Control



Porphyromonas|UC

0.000
42.000
6.286
12.073

0.002

1

0.007




Porphyromonas|CD

0.000
16.000
1.000
2.963
0.443

0.007

1



Lautropia|Control

0.000
1.000
0.048
0.218
1

0.001

0.555



Lautropia|UC

0.000
57.000
5.286
15.097

0.001

1

<0.0001




Lautropia|CD

0.000
0.000
0.000
0.000
0.555

<0.0001

1



Methylobacterium|

0.000
1.000
0.048
0.218
1

0.004

0.243


Control



Methylobacterium|

0.000
103.000
12.786
31.740

0.004

1
0.032


UC



Methylobacterium|

0.000
11.000
1.000
2.759
0.243
0.032
1


CD



Akkermansia|

0.000
1098.000
85.381
251.309
1

0.014

0.296


Control



Akkermansia|UC

0.000
13.000
1.071
3.452

0.014

1
0.075



Akkermansia|CD

0.000
2103.000
95.811
357.421
0.296
0.075
1



Tannerella|Control

0.000
60.000
2.857
13.093
1
0.042
0.431



Tannerella|UC

0.000
1.000
0.214
0.426
0.042
1

0.004




Tannerella|CD

0.000
0.000
0.000
0.000
0.431

0.004

1



Haemophilus|

0.000
11.000
1.381
2.941
1

0.007


0.010



Control



Haemophilus|UC

0.000
722.000
79.714
190.874

0.007

1
0.478



Haemophilus|CD

0.000
600.000
30.568
101.515

0.010

0.478
1



Finegoldia|Control

0.000
1.000
0.048
0.218
1

0.014

0.941



Finegoldia|UC

0.000
303.000
30.643
84.514

0.014

1

0.009




Finegoldia|CD

0.000
1.000
0.054
0.229
0.941

0.009

1



Turicibacter|Control

0.000
866.000
70.190
198.733
1

0.007


0.006




Turicibacter|UC

0.000
2.000
0.286
0.611

0.007

1
0.569



Turicibacter|CD

0.000
1456.000
39.919
239.274

0.006

0.569
1



Nitrincola|Control

0.000
88.000
6.762
20.630
1

0.008

0.021



Nitrincola|UC

0.000
4354.000
397.071
1158.944

0.008

1
0.360



Nitrincola|CD

0.000
1068.000
38.676
175.578
0.021
0.360
1



Hydrogenophilus|

0.000
0.000
0.000
0.000
1

0.008

0.660


Control



Hydrogenophilus|

0.000
2.000
0.286
0.611

0.008

1

0.011



UC



Hydrogenophilus|

0.000
2.000
0.054
0.329
0.660

0.011

1


CD



Listeria|Control

0.000
11.000
1.381
3.008
1
0.310

0.011




Listeria|UC

0.000
3.000
0.286
0.825
0.310
1
0.272



Listeria|CD

0.000
2.000
0.054
0.329

0.011

0.272
1



Actinobacillus|

0.000
6.000
0.571
1.535
1

0.005

0.018


Control



Actinobacillus|UC

0.000
1806.000
138.786
480.704

0.005

1
0.307



Actinobacillus|CD

0.000
243.000
15.649
45.935
0.018
0.307
1



Anaerococcus|

0.000
20.000
1.095
4.358
1

0.001

0.054


Control



Anaerococcus|UC

0.000
23670.000
1740.571
6312.547

0.001

1
0.065



Anaerococcus|CD

0.000
1602.000
46.324
263.052
0.054
0.065
1



Catonella|Control

0.000
8.000
0.524
1.750
1
0.133
0.231



Catonella|UC

0.000
13.000
2.071
3.990
0.133
1

0.007




Catonella|CD

0.000
19.000
0.595
3.149
0.231

0.007

1



Mobiluncus|Control

0.000
0.000
0.000
0.000
1

0.007

0.677



Mobiluncus|UC

0.000
3.000
0.500
1.092

0.007

1

0.009




Mobiluncus|CD

0.000
1.000
0.027
0.164
0.677

0.009

1



Pantoea|Control

0.000
0.000
0.000
0.000
1
0.058

0.002




Pantoea|UC

0.000
8.000
1.071
2.401
0.058
1
0.523



Pantoea|CD

0.000
99.000
3.432
16.194

0.002

0.523
1



Enterobacter|

0.000
17.000
1.810
3.829
1

0.003

0.109


Control



Enterobacter|UC

0.000
10592.000
778.786
2824.745

0.003

1
0.057



Enterobacter|CD

0.000
3590.000
105.892
588.904
0.109
0.057
1



Paenibacillus|

0.000
181.000
31.952
44.147
1

0.013


0.007



Control



Paenibacillus|UC

0.000
53.000
8.214
14.412

0.013

1
0.699



Paenibacillus|CD

0.000
617.000
23.595
100.894

0.007

0.699
1



Staphylococcus|

0.000
4079.000
353.762
910.667
1
0.224

0.010



Control



Staphylococcus|UC

0.000
229.000
58.357
80.759
0.224
1
0.370



Staphylococcus|CD

0.000
5529.000
214.649
925.461

0.010

0.370
1



Vitreoscilla|Control

0.000
37.000
2.286
8.057
1

0.009

0.319



Vitreoscilla|UC

0.000
1667.000
128.357
443.285

0.009

1
0.044



Vitreoscilla|CD

0.000
15.000
1.622
3.192
0.319
0.044
1



Alcanivorax|Control

0.000
0.000
0.000
0.000
1

0.012

1.000



Alcanivorax|UC

0.000
5.000
0.643
1.646

0.012

1

0.006




Alcanivorax|CD

0.000
0.000
0.000
0.000
1.000

0.006

1



Veillonella|Control

6.000
1213.000
187.571
292.401
1

0.003

0.106



Veillonella|UC

23.000
38298.000
8985.571
13795.955

0.003

1
0.063



Veillonella|CD

7.000
17002.000
2107.243
4464.809
0.106
0.063
1



Tatumella|Control

0.000
61.000
3.381
13.347
1

0.014

0.090



Tatumella|UC

0.000
1836.000
175.643
501.883

0.014

1
0.224



Tatumella|CD

0.000
71.000
5.054
13.894
0.090
0.224
1



Afipia|Control

0.000
1.000
0.095
0.301
1
0.027
0.913



Afipia|UC

0.000
30.000
2.429
7.949
0.027
1

0.012




Afipia|CD

0.000
3.000
0.135
0.536
0.913

0.012

1









Certain bacterial taxa exhibit higher levels (abundance) in UC or CD or IBD patients and some taxa exhibit lower levels in UC or CD or IBD patients. Therefore, an assay on a gut sample from a patient can be performed to measure an abundance (or level) of a bacterial taxa and by comparing this abundance to that of a predetermined abundance or an average abundance (as in tables 1 or 2) of the taxa derived from sample of patients with UC or CD or IBD. The result allows one to determine whether a patient has UC or CD or IBD.


The abundance or level of bacterial taxa can be determined for example by quantitative DNA analysis such as quantitative polymerase chain reaction. As described above the data can be normalized (example subsampling normalization) as would be known in the art. Therefore the results discussed in the present application can represent relative abundance. It will be appreciated that a person skilled in the art would know to interpret these values to determine the relative levels of bacteria.


A method is also provided in which a diagnosis of UC or CD or IBD is achieved by collecting a gut sample from a patient and from which bacterial taxa levels will be determined using an assay as described above. The gut sample may be from the flushing of the colon wall as described above and still further described below or from stools.


Certain taxa exhibit a statistically significant difference in their abundance between UC patients and CD patients. Therefore by comparing the relative abundance of one or more of these taxa between UC and CD patients it is possible to determine whether the patient has CD or UC disease. For example, Hydrogenophilus is more abundant in both CD and UC patients relative to healthy individuals and furthermore it is more abundant in UC patients than CD patients.


In another aspect of the invention the severity of the disease can also be assessed from the bacterial profile of the gut microbiota. Thus, the severity of CD can be established by measuring the relative abundance of certain bacterial taxa in a gut microbiota sample. In this respect, the relative abundance of one or more microbial taxa from the gut can be compared/correlated with a standard disease activity index. The resulting classification allows the use of relative abundance of bacterial taxa as an indicator of disease severity (Table 3). It will be appreciated that abundance measurements from one or more bacterial taxa can be used for that purpose.


Supplementary Table 6: Taxa that varies significantly in abundance in CD patients in at least one of the three pairwise comparisons performed (mild vs. moderate; mild vs. sever; and moderate vs. severe). In table 3 the number of 16S rDNA reads in each sample was normalized by random subsampling to 500,000. Minimum and maximum correspond to the minimum and maximum number of reads obtained; mean corresponds to the mean of the number of reads obtained. P values were generated using a Kruskal-Wallis test with a Dunn's post hoc test and a Bonferroni correction for multiple hypotheses. “p|mild” indicates the P values obtained by comparison to the CD patients with a mild inflammation; “p|moderate” and “p|severe” indicate the P values obtained by comparison to CD patients with a moderate and severe inflammation respectively. Values in bold indicate significance (P<0.05).
















TABLE 3









Std.
p|
p|
p|


Variable
Minimum
Maximum
Mean
deviation
mild
severe
moderate






















PHYLUM









None


CLASS


Clostridia|Mild
174469.000
413730.000
312892.000
87462.076
1

0.012

0.105


Clostridia|Severe
9293.000
407027.000
198223.435
105908.496

0.012

1
0.860


Clostridia|Moderate
59849.000
289226.000
206514.200
90299.691
0.105
0.860
1


Betaproteobacteria|Mild
26.000
29550.000
4640.333
9490.562
1
0.170

0.013



Betaproteobacteria|Severe
60.000
54643.000
10878.696
15121.993
0.170
1
0.084


Betaproteobacteria|
2685.000
129123.000
48219.200
55650.162

0.013

0.084
1


Moderate


ORDER


Clostridiales|Mild
174469.000
413730.000
312892.000
87462.076
1

0.012

0.105


Clostridiales|Severe
9292.000
407027.000
198219.870
105908.987

0.012

1
0.860


Clostridiales|Moderate
59849.000
289225.000
206514.000
90299.462
0.105
0.860
1


FAMILY


Staphylococcaceae|Mild
4.000
1507.000
255.222
507.068
1
0.025

0.002



Staphylococcaceae|Severe
0.000
5529.000
257.522
1149.930
0.025
1
0.089


Staphylococcaceae|
0.000
2.000
0.800
0.837

0.002

0.089
1


Moderate


Propionibacteriaceae|Mild
0.000
1.000
0.556
0.527
1

0.016

0.484


Propionibacteriaceae|Severe
0.000
363.000
33.652
88.327

0.016

1

0.007



Propionibacteriaceae|
0.000
1.000
0.200
0.447
0.484

0.007

1


Moderate


Acidaminococcaceae|Mild
0.000
4.000
1.111
1.691
1

0.003

0.030


Acidaminococcaceae|
0.000
36271.000
2476.261
7722.681

0.003

1
0.965


Severe


Acidaminococcaceae|
0.000
8395.000
1712.000
3736.459
0.030
0.965
1


Moderate


Bacillaceae|Mild
0.000
136.000
16.000
45.008
1
0.137
0.281


Bacillaceae|Severe
0.000
6167.000
427.174
1339.746
0.137
1

0.016



Bacillaceae|Moderate
0.000
3.000
0.600
1.342
0.281

0.016

1


Carnobacteriaceae|Mild
13.000
643.000
155.222
229.710
1
0.206
0.148


Carnobacteriaceae|Severe
5.000
76920.000
3723.870
15973.648
0.206
1

0.008



Carnobacteriaceae|
4.000
78.000
26.200
30.136
0.148

0.008

1


Moderate


Sutterellaceae|Mild
2.000
1157.000
136.889
382.665
1

0.004


0.004



Sutterellaceae|Severe
1.000
54546.000
7005.435
15182.702

0.004

1
0.342


Sutterellaceae|Moderate
9.000
128471.000
42805.000
59466.785

0.004

0.342
1


GENUS



Atopobium|Mild

1.000
257.000
60.778
81.208
1
0.704
0.056



Atopobium|Severe

0.000
1273.000
118.652
263.676
0.704
1

0.014




Atopobium|Moderate

0.000
7.000
4.800
2.775
0.056

0.014

1



Propionibacterium|Mild

0.000
1.000
0.556
0.527
1

0.016

0.484



Propionibacterium|Severe

0.000
363.000
33.652
88.327

0.016

1

0.007




Propionibacterium|Moderate

0.000
1.000
0.200
0.447
0.484

0.007

1



Trichococcus|Mild

0.000
3.000
0.556
1.014
1

0.002

0.031



Trichococcus|Severe

0.000
0.000
0.000
0.000

0.002

1
1.000



Trichococcus|Moderate

0.000
0.000
0.000
0.000
0.031
1.000
1



Pectobacterium|Mild

0.000
0.000
0.000
0.000
1
1.000
0.029



Pectobacterium|Severe

0.000
0.000
0.000
0.000
1.000
1

0.014




Pectobacterium|Moderate

0.000
80.000
16.000
35.777
0.029

0.014

1



Granulicatella|Mild

13.000
581.000
145.444
215.009
1
0.229
0.167



Granulicatella|Severe

5.000
74250.000
3494.130
15434.318
0.229
1

0.012




Granulicatella|Moderate

3.000
78.000
25.000
30.406
0.167

0.012

1



Jonquetella|Mild

0.000
0.000
0.000
0.000
1
1.000
0.029



Jonquetella|Severe

0.000
0.000
0.000
0.000
1.000
1

0.014




Jonquetella|Moderate

0.000
83.000
16.600
37.119
0.029

0.014

1



Riemerella|Mild

0.000
0.000
0.000
0.000
1
1.000

0.002




Riemerella|Severe

0.000
0.000
0.000
0.000
1.000
1

0.000




Riemerella|Moderate

0.000
3.000
0.800
1.304

0.002


0.000

1



Mogibacterium|Mild

0.000
45.000
7.556
14.934
1
0.187
0.207



Mogibacterium|Severe

0.000
376.000
34.478
82.168
0.187
1

0.013




Mogibacterium|Moderate

0.000
1.000
0.200
0.447
0.207

0.013

1



Staphylococcus|Mild

4.000
1247.000
226.333
428.093
1
0.024

0.002




Staphylococcus|Severe

0.000
5529.000
256.565
1150.035
0.024
1
0.090



Staphylococcus|Moderate

0.000
2.000
0.800
0.837

0.002

0.090
1



Sutterella|Mild

2.000
1157.000
136.889
382.665
1

0.004


0.004




Sutterella|Severe

1.000
54546.000
7005.435
15182.702

0.004

1
0.342



Sutterella|Moderate

9.000
128471.000
42805.000
59466.785

0.004

0.342
1



Phascolarctobacterium|Mild

0.000
4.000
1.111
1.691
1

0.003

0.030



Phascolarctobacterium|

0.000
36271.000
2476.261
7722.681

0.003

1
0.965


Severe



Phascolarctobacterium|

0.000
8395.000
1712.000
3736.459
0.030
0.965
1


Moderate



Comamonas|Mild

0.000
3.000
0.667
1.118
1

0.016

0.054



Comamonas|Severe

0.000
1.000
0.043
0.209

0.016

1
0.792



Comamonas|Moderate

0.000
0.000
0.000
0.000
0.054
0.792
1



Hylemonella|Mild

0.000
0.000
0.000
0.000
1
1.000
0.029



Hylemonella|Severe

0.000
0.000
0.000
0.000
1.000
1

0.014




Hylemonella|Moderate

0.000
1.000
0.200
0.447
0.029

0.014

1



Xenorhabdus|Mild

0.000
0.000
0.000
0.000
1
1.000
0.029



Xenorhabdus|Severe

0.000
0.000
0.000
0.000
1.000
1

0.014




Xenorhabdus|Moderate

0.000
1.000
0.200
0.447
0.029

0.014

1



Averyella|Mild

0.000
0.000
0.000
0.000
1
1.000
0.029



Averyella|Severe

0.000
0.000
0.000
0.000
1.000
1

0.014




Averyella|Moderate

0.000
11.000
2.200
4.919
0.029

0.014

1









It will be appreciated that it is possible to refine the assessment of the stage or severity of the disease by combining the measurement(s) of the abundance of bacterial taxa with the observation of a choice of symptoms underlying the classic disease indexes to arrive at the establishment of a diagnosis. For example it may be desirable or sometimes only possible to measure only a limited set of standard symptoms associated with disease indexes. This limited set of symptoms may not be sufficient to pose a diagnostic. In such cases it may be possible to combine an assay involving the measurement of bacterial taxa to provide additional information on the nature or stage of the disease.


In an aspect of the invention A. parvulum, an H2S producer, is a good marker of CD exhibiting a higher relative abundance in patient with CD than in controls. Furthermore, the relative abundance of A. parvulum compared to core bacterial taxa abundance is also a measure of the presence and severity of the disease. For example an abundance of A. parvulum relative to the core greater than 0.005% is indicative of moderate or severe stage of the disease (FIG. 1C). Furthermore severity of CD can also be characterized by a significant increased abundance of Proteobacteria microbes. Severe CD and UC can also be characterized by an increased in the relative abundance of H2S producers compared to controls. It will be appreciated that specific taxa can be used to assess the severity of disease as described in Table 3.


In yet another aspect of the invention a decrease in the relative abundance of butyrate producers such as Firmicutes, Clostridia, Clostridiales and Lachnopiraceae Eubacterium and Faecalibacterium is indicative of the presence of disease (CD or UC).


The measurements of the abundance of bacterial taxa using DNA quatification can generally be done by methods that are known in the art. However in one aspect of the invention there is provided a method for determining the abundance of A. parvulum by absolute quantitative DNA measurement by performing PCR on the extracted metagenomic DNA. The following primers for the quantitative measurements of A. parvulum were developed: Aparv-711F 5′-GGGGAGTATTTCTTCCGTGCCG-3′ (SEQ ID NO. 1) and Aparv-881R 5′-CTTCACCTAAATGTCAA GCCCTGG-3′ (SEQ ID NO. 2). The development of these primers enables the use of an assay for measuring the abundance of A. parvulum that is highly specific, rapid and reliable. Thus in an another aspect of the invention there is also provided kits that would comprise these primers and other reagents as would be known in the art to detect A. parvulum or other taxa useful for the diagnosis, assessment or staging of UC, CD or IBD as described herein.


In further embodiment of the invention, the presence of UC and CD disease can be assessed by the presence, absence and/or relative abundance of certain host proteins. Proteins can be identified and measured by techniques known in the art such as shotgun mass-spectrometry in conjunction with protein fractionation. Other method for detecting specific proteins such as, immunology based methods (antibodies), western blots, spectrophotometry, enzyme assays, ELISA and any other method as would be known to one skilled in the art may also be used.


Table 4 provides a list of all differentially expressed proteins and their variable importance in projection scores (VIP) derived from the calculated PLS-DA. (Control v. CD with increasing inflammation severity)












TABLE 4






Comp 1
Comp 2
Comp 3


Variable
VIP
VIP
VIP


















General transcription factor IIA subunit 1; TFIIA p19 subunit; TFIIA p35
2.513
1.975
1.814


subunit; TFIIAL; Transcription initiation factor IIA alpha chain; Transcription initiation factor





IIA beta chain; Transcription initiation factor IIA subunit 1; Transcription initiation factor





TFIIA 42 kDa subunit





Angiotensin-binding protein; Microsomal endopeptidase; Mitochondrial oligopeptidase
2.296
1.901
1.745


M; Neurolysin, mitochondrial; Neurotensin endopeptidase





Defensin, alpha 5; Defensin-5
2.013
1.575
1.399


Mineral dust-induced gene protein; MYC-induced nuclear antigen; Nucleolar protein 52
1.942
1.793
1.585


Glutaminase kidney isoform, mitochondrial; K-glutaminase; L-glutamine amidohydrolase
1.880
1.538
1.358


Ethanolaminephosphotransferase 1; Selenoprotein I; Putative uncharacterized protein
1.853
1.918
1.702


ENSP00000385426; Putative uncharacterized protein ENSP00000391804





18S rRNA dimethylase; DIM1 dimethyladenosine transferase 1-like; Probable
1.790
1.524
1.435


dimethyladenosine transferase; S-adenosylmethionine-6-N,N-adenosyl(rRNA)





dimethyltransferase





6PF-2-K/Fru-2,6-P2ase heart-type isozyme; 6-phosphofructo-2-kinase; 6-phosphofructo-2-
1.717
1.442
1.275


kinase/fructose-2,6-biphosphatase 2; Fructose-2,6-bisphosphatase





Armadillo repeat-containing protein 8; cDNA FLJ56387, highly similar to Mus musculus
1.692
1.548
1.376


armadillo repeat containing 8 (Armc8), mRNA; Putative uncharacterized protein





ARMC8; Armadillo repeat containing 8, isoform CRA_g; cDNA FLJ53383, highly similar to






Homo sapiens armadillo repeat containing 8 (ARMC8), transcript variant 2, mRNA






Aconitase 2, mitochondrial; Aconitate hydratase, mitochondrial; Citrate hydro-lyase; cDNA
1.645
1.338
1.191


FLJ60429, highly similar to Aconitate hydratase, mitochondrial (EC 4.2.1.3); cDNA





FLJ50886, highly similar to Aconitate hydratase, mitochondrial(EC 4.2.1.3)





2C4D; Class II mMOB1; Mob1 homolog 3; Mps one binder kinase activator-like
1.634
1.359
1.203


3; Preimplantation protein 3; cDNA FLJ52887, highly similar to Preimplantation protein 3





Iron-sulfur subunit of complex II; Succinate dehydrogenase [ubiquinone] iron-sulfur
1.612
1.266
1.216


subunit, mitochondrial





Reticulocalbin-1; cDNA FLJ55835, highly similar to Reticulocalbin-1
1.593
1.238
1.093


DRB sensitivity-inducing factor 14 kDa subunit; DRB sensitivity-inducing factor small
1.592
1.247
1.102


subunit; Transcription elongation factor SPT4





Rhodanese; Thiosulfate sulfurtransferase
1.590
1.257
1.112


22 kDa protein; CP-22; Sorcin; V19; Putative uncharacterized protein SRI; cDNA FLJ60640,
1.589
1.282
1.144


highly similar to Sorcin; cDNA FLJ54267, moderately similar to Sorcin





Flavoprotein subunit of complex II; Succinate dehydrogenase [ubiquinone] flavoprotein
1.576
1.225
1.169


subunit, mitochondrial





Acetylneuraminyl hydrolase; G9 sialidase; Lysosomal sialidase; N-acetyl-alpha-
1.562
1.955
1.773


neuraminidase 1; Sialidase-1





Beta-IV spectrin; Spectrin beta chain, brain 3; Spectrin, non-erythroid beta chain 3; Putative
1.557
1.291
1.194


uncharacterized protein SPTBN4





Translocation protein SEC63 homolog
1.542
1.555
1.373


Epidermal-type fatty acid-binding protein; Fatty acid-binding protein 5; Fatty acid-binding
1.540
1.206
1.071


protein, epidermal; Psoriasis-associated fatty acid-binding protein homolog





Complex I-51 kD; NADH dehydrogenase [ubiquinone] flavoprotein 1, mitochondrial; NADH
1.530
1.205
1.205


dehydrogenase flavoprotein 1; NADH-ubiquinone oxidoreductase 51 kDa subunit; cDNA





FLJ57949, highly similar to NADH-ubiquinone oxidoreductase 51 kDa subunit,





mitochondrial (EC 1.6.5.3); cDNA, FLJ79021, highly similar to NADH-ubiquinone





oxidoreductase 51 kDa subunit, mitochondrial (EC 1.6.5.3)





Calregulin; Calreticulin; CRP55; Endoplasmic reticulum resident protein
1.528
1.220
1.077


60; grp60; HACBP; cDNA FLJ58668, highly similar to Calreticulin





UDP-glucose 6-dehydrogenase; cDNA FLJ60093, highly similar to UDP-glucose 6-
1.522
1.183
1.052


dehydrogenase (EC 1.1.1.22)





4-alpha-glucanotransferase; Amylo-alpha-1,6-glucosidase; Dextrin 6-alpha-D-
1.516
1.200
1.072


glucosidase; Glycogen debrancher; Glycogen debranching enzyme; Oligo-1,4-1,4-





glucantransferase





Malic enzyme 2; NAD-dependent malic enzyme, mitochondrial
1.514
1.225
1.081


Delta(3),delta(2)-enoyl-CoA isomerase; Diazepam-binding inhibitor-related protein
1.513
1.178
1.046


1; Dodecenoyl-CoA isomerase; DRS-1; Hepatocellular carcinoma-associated antigen





88; Peroxisomal 3,2-trans-enoyl-CoA isomerase; Renal carcinoma antigen NY-REN-





1; Putative uncharacterized protein PECI





Complex I-75 kD; NADH-ubiquinone oxidoreductase 75 kDa subunit, mitochondrial; cDNA
1.510
1.324
1.181


FLJ60586, highly similar to NADH-ubiquinone oxidoreductase 75 kDa subunit,





mitochondrial (EC 1.6.5.3)





cDNA FLJ53665, highly similar to Four and a half LIM domains protein 1; Four and a half
1.500
1.168
1.031


LIM domains 1; Four and a half LIM domains protein 1; Skeletal muscle LIM-protein 1





Putative adenosylhomocysteinase 3; S-adenosylhomocysteine hydrolase-like protein 2;
1.499
1.165
1.031


S-adenosyl-L-homocysteine hydrolase 3





28S ribosomal protein S9, mitochondrial
1.489
1.198
1.121


150 kDa oxygen-regulated protein; 170 kDa glucose-regulated protein; Hypoxia
1.480
1.179
1.041


up-regulated protein 1; cDNA FLJ54708, highly similar to 150 kDa oxygen-regulated protein





(Orp150)





Complex I-39 kD; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9,
1.466
1.152
1.083


mitochondrial; NADH-ubiquinone oxidoreductase 39 kDa subunit





DDAHI; Dimethylargininase-1; N(G),N(G)-dimethylarginine dimethylaminohydrolase
1.465
1.177
1.072


1; cDNA FLJ54083, highly similar to NG,NG-dimethylarginine dimethylaminohydrolase 1





(EC 3.5.3.18); cDNA FLJ54119, highly similar to NG,NG-dimethylarginine





dimethylaminohydrolase 1 (EC 3.5.3.18)





Phenylalanine--tRNA ligase beta chain; Phenylalanyl-tRNA synthetase beta chain
1.462
1.154
1.080


ABP-278; ABP-280 homolog; Actin-binding-like protein; Beta-filamin; Filamin homolog
1.462
1.166
1.032


1; Filamin-3; Filamin-B; Thyroid autoantigen; Truncated actin-binding protein





GTP-specific succinyl-CoA synthetase subunit beta; Succinyl-CoA ligase [GDP-forming]
1.459
1.151
1.018


subunit beta, mitochondrial; Succinyl-CoA synthetase beta-G chain





TPPP/p20; Tubulin polymerization-promoting protein family member 3
1.455
1.376
1.275


F8W031; F8VXJ7; F8VP03
1.450
1.187
1.058


Protein NipSnap homolog 1
1.438
1.119
0.993


78 kDa gastrin-binding protein; Long chain 3-hydroxyacyl-CoA dehydrogenase; Long-chain
1.434
1.118
1.032


enoyl-CoA hydratase; TP-alpha; Trifunctional enzyme subunit alpha, mitochondrial





Antioxidant enzyme AOE372; Peroxiredoxin IV; Peroxiredoxin-4; Thioredoxin peroxidase
1.421
1.144
1.011


AO372; Thioredoxin-dependent peroxide reductase A0372





Calumenin; Crocalbin; IEF SSP 9302
1.418
1.286
1.136


GTPase IMAP family member 4; Immunity-associated nucleotide 1 protein; Immunity-
1.418
1.105
0.975


associated protein 4; cDNA FLJ51351, highly similar to GTPase, IMAP family member 4





Plakophilin-2; Truncated plakophilin-2
1.417
1.103
1.000


Adaptor protein complex AP-1 mu-2 subunit; Adaptor-related protein complex 1 mu-2
1.417
1.262
1.117


subunit; AP-1 complex subunit mu-2; AP-mu chain family member mu1B; Clathrin assembly





protein complex 1 medium chain 2; Golgi adaptor HA1/AP1 adaptin mu-2 subunit; Mu1B-





adaptin; Mu-adaptin 2





Complex III subunit 1; Core protein I; Cytochrome b-c1 complex subunit 1,
1.413
1.209
1.071


mitochondrial; Ubiquinol-cytochrome-c reductase complex core protein 1





90 kDa ribosomal protein S6 kinase 3; Insulin-stimulated protein kinase 1; MAP kinase-
1.410
1.200
1.080


activated protein kinase 1b; pp90RSK2; Ribosomal protein S6 kinase alpha-3; Ribosomal





S6 kinase 2; cDNA, FLJ79381, highly similar to Ribosomal protein S6 kinase alpha-3 (EC





2.7.11.1); cDNA FLJ56618, highly similar to Ribosomal protein S6 kinase alpha-3 (EC





2.7.11.1)





3-5 RNA exonuclease OLD35; PNPase old-35; Polynucleotide phosphorylase
1.407
1.255
1.155


1; Polynucleotide phosphorylase-like protein; Polyribonucleotide nucleotidyltransferase 1,





mitochondrial





Complex I-B15; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 4; NADH-
1.397
1.113
1.010


ubiquinone oxidoreductase B15 subunit; Putative uncharacterized protein NDUFB4





Carnitine O-palmitoyltransferase 2, mitochondrial; Carnitine palmitoyltransferase II
1.396
1.089
0.976


Aldehyde dehydrogenase 5; Aldehyde dehydrogenase family 1 member B1; Aldehyde
1.393
1.120
0.988


dehydrogenase X, mitochondrial; cDNA FLJ51238, highly similar to Aldehyde





dehydrogenase X, mitochondrial (EC 1.2.1.3)





Coatomer subunit epsilon; Epsilon-coat protein; Coatomer protein complex, subunit
1.388
1.080
0.959


epsilon, isoform CRA_e; Putative uncharacterized protein COPE





Ethylmalonic encephalopathy protein 1; Hepatoma subtracted clone one protein; Protein
1.383
1.090
0.974


ETHE1, mitochondrial





SRA stem-loop-interacting RNA-binding protein, mitochondrial
1.382
1.075
0.960


15-hydroxyprostaglandin dehydrogenase [NADP+]; Carbonyl reductase [NADPH]
1.379
1.085
1.043


1; NADPH-dependent carbonyl reductase 1; Prostaglandin 9-ketoreductase; Prostaglandin-





E(2) 9-reductase; Putative uncharacterized protein CBR1; Carbonyl reductase 1, isoform





CRA_c; cDNA FLJ60474, highly similar to Carbonyl reductase





ER-Golgi intermediate compartment 53 kDa protein; Gp58; Intracellular mannose-specific
1.374
1.129
1.046


lectin MR60; Lectin mannose-binding 1; Protein ERGIC-53





Intestinal trefoil factor; Polypeptide P1.B; Trefoil factor 3
1.369
1.093
0.971


78 kDa glucose-regulated protein; Endoplasmic reticulum lumenal Ca(2+)-binding protein
1.366
1.104
1.046


grp78; Heat shock 70 kDa protein 5; Immunoglobulin heavy chain-binding protein





Complex I-13 kD-A; NADH dehydrogenase [ubiquinone] iron-sulfur protein 6,
1.365
1.090
0.968


mitochondrial; NADH-ubiquinone oxidoreductase 13 kDa-A subunit





3-ketoacyl-CoA thiolase, mitochondrial; Acetyl-CoA acyltransferase; Beta-
1.365
1.097
1.028


ketothiolase; Mitochondrial 3-oxoacyl-CoA thiolase; T1





Endoplasmic reticulum resident protein 46; Thioredoxin domain-containing protein
1.363
1.103
0.994


5; Thioredoxin-like protein p46; TXNDC5 protein; cDNA, FLJ96678, Homo sapiens





thioredoxin domain containing 5 (TXNDC5), mRNA; HCG1811539, isoform CRA_b





Elongation factor Tu, mitochondrial; P43
1.361
1.065
0.985


Outer mitochondrial membrane protein porin 2; Voltage-dependent anion-selective
1.361
1.060
0.938


channel protein 2; Voltage-dependent anion channel 2; cDNA FLJ60120, highly similar to





Voltage-dependent anion-selective channel protein 2; cDNA, FLJ78818, highly similar to





Voltage-dependent anion-selective channel protein 2





63 kDa membrane protein; Cytoskeleton-associated protein 4
1.361
1.102
0.975


Cytovillin; Ezrin; p81; Villin-2
1.360
1.057
0.937


Myosin I beta; Myosin-Ic
1.359
1.071
0.948


250/210 kDa paraneoplastic pemphigus antigen; Desmoplakin
1.356
1.126
1.002


Very long-chain specific acyl-CoA dehydrogenase, mitochondrial
1.355
1.089
0.983


15-oxoprostaglandin 13-reductase; Prostaglandin reductase 2; Zinc-binding alcohol
1.353
1.160
1.057


dehydrogenase domain-containing protein 1





Complex III subunit 2; Core protein II; Cytochrome b-c1 complex subunit 2,
1.345
1.200
1.182


mitochondrial; Ubiquinol-cytochrome-c reductase complex core protein 2





Aspartate aminotransferase, mitochondrial; Fatty acid-binding protein; Glutamate
1.339
1.041
0.919


oxaloacetate transaminase 2; Plasma membrane-associated fatty acid-binding





protein; Transaminase A





CML33; Phenylalanine-tRNA ligase alpha chain; Phenylalanyl-tRNA synthetase alpha
1.337
1.179
1.051


chain; cDNA FLJ50378, highly similar to Phenylalanyl-tRNA synthetase alpha chain (EC





6.1.1.20)





Sodium pump subunit alpha-1; Sodium/potassium-transporting ATPase subunit alpha-
1.335
1.042
0.927


1; ATPase, Na+/K+ transporting, alpha 1 polypeptide, isoform CRA_a; cDNA FLJ52430,





highly similar to Sodium/potassium-transporting ATPase alpha-1 chain (EC 3.6.3.9)





Putative uncharacterized protein MLLT4; Afadin; ALL1-fused gene from chromosome 6
1.334
1.277
1.207


protein; Myeloid/lymphoid or mixed-lineage leukemia (Trithorax homolog, Drosophila);





translocated to, 4





Cytochrome c oxidase polypeptide Vb; Cytochrome c oxidase subunit 5B, mitochondrial
1.332
1.229
1.092


35 kDa lectin; Carbohydrate-binding protein 35; Galactose-specific lectin 3; Galactoside-
1.328
1.057
0.972


binding protein; Galectin-3; IgE-binding protein; L-31; Laminin-binding protein; Lectin L-





29; Mac-2 antigen





Complex I-B22; LYR motif-containing protein 3; NADH dehydrogenase [ubiquinone] 1 beta
1.327
1.179
1.045


subcomplex subunit 9; NADH-ubiquinone oxidoreductase B22 subunit





3-ketoacyl-CoA thiolase; Acetyl-CoA acyltransferase; Beta-ketothiolase; TP-
1.325
1.030
0.972


beta; Trifunctional enzyme subunit beta, mitochondrial; cDNA FLJ56214, highly similar to





Trifunctional enzyme subunit beta, mitochondrial; Putative uncharacterized protein





HADHB





Endoplasmic reticulum resident protein 28; Endoplasmic reticulum resident protein
1.322
1.061
0.938


29; Endoplasmic reticulum resident protein 31





Alu corepressor 1; Antioxidant enzyme B166; Liver tissue 2D-page spot 71B; Peroxiredoxin
1.316
1.071
0.947


V; Peroxiredoxin-5, mitochondrial; Peroxisomal antioxidant enzyme; PLP; Thioredoxin





peroxidase PMP20; Thioredoxin reductase; TPx type VI; Putative uncharacterized protein





PRDX5





ER-Golgi SNARE of 24 kDa; SEC22 vesicle-trafficking protein homolog B; SEC22 vesicle-
1.315
1.023
0.937


trafficking protein-like 1; Vesicle-trafficking protein SEC22b





Calcium-binding mitochondrial carrier protein Aralar2; Citrin; Mitochondrial aspartate
1.314
1.021
0.944


glutamate carrier 2; Solute carrier family 25 member 13





RRP12-like protein
1.311
1.083
0.961


Endoplasmic reticulum resident protein 70; Endoplasmic reticulum resident protein
1.311
1.110
0.980


72; Protein disulfide-isomerase A4





Myosin-Id
1.308
1.070
0.944


Actin-depolymerizing factor; Destrin
1.306
1.027
0.918


Complex I-B14.5a; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit
1.305
1.115
1.169


7; NADH-ubiquinone oxidoreductase subunit B14.5a





Beta-G1; Beta-glucuronidase
1.297
1.019
0.921


Chymotrypsin-like elastase family member 3A; Elastase IIIA; Elastase-3A; Protease E
1.297
1.020
0.910


17-beta-hydroxysteroid dehydrogenase 11; 17-beta-hydroxysteroid dehydrogenase
1.294
1.013
0.895


XI; Cutaneous T-cell lymphoma-associated antigen HD-CL-03; Dehydrogenase/reductase





SDR family member 8; Estradiol 17-beta-dehydrogenase 11; Retinal short-chain





dehydrogenase/reductase 2





Interleukin-25; Stromal cell-derived growth factor SF20; UPF0556 protein C19orf10
1.292
1.121
0.990


Complex III subunit 3; Complex III subunit III; Cytochrome b; Cytochrome b-c1 complex
1.287
1.237
1.209


subunit 3; Ubiquinol-cytochrome-c reductase complex cytochrome b subunit





Cellular thyroid hormone-binding protein; p55; Prolyl 4-hydroxylase subunit beta; Protein
1.285
1.018
0.995


disulfide-isomerase; cDNA FLJ59430, highly similar to Protein disulfide-isomerase (EC





5.3.4.1)





Importin-4; Importin-4b; Ran-binding protein 4
1.280
1.268
1.137


94 kDa glucose-regulated protein; Endoplasmin; gp96 homolog; Heat shock protein 90 kDa
1.277
1.040
1.005


beta member 1; Tumor rejection antigen 1





Amine oxidase [flavin-containing] A; Monoamine oxidase type A; cDNA FLJ61220, highly
1.276
1.023
0.903


similar to Amine oxidase (flavin-containing) A (EC 1.4.3.4)





Ubiquitin-fold modifier 1
1.269
1.042
1.089


Antigen NY-CO-4; Elongation factor 1-delta
1.267
1.160
1.025


3-phosphoadenosine-5-phosphosulfate synthase; Adenosine-5-phosphosulfate 3-
1.263
0.990
0.874


phosphotransferase; Adenylylsulfate 3-phosphotransferase; Adenylyl-sulfate kinase; APS





kinase; ATP-sulfurylase; Bifunctional 3-phosphoadenosine 5-phosphosulfate synthase





2; Sulfate adenylate transferase; Sulfate adenylyltransferase; Sulfurylase kinase 2





Alpha-adducin; Erythrocyte adducin subunit alpha; Adducin 1 (Alpha); Adducin 1 (Alpha),
1.259
1.110
0.981


isoform CRA_e; ADD1 protein





Microsomal signal peptidase 25 kDa subunit; Signal peptidase complex subunit 2
1.257
0.978
0.863


Quiescin Q6; Sulfhydryl oxidase 1
1.257
1.111
1.053


Acetoacetyl-CoA thiolase; Acetyl-CoA acetyltransferase, mitochondrial; T2
1.255
0.994
0.887


2-oxoglutarate dehydrogenase complex component E1; 2-oxoglutarate dehydrogenase,
1.254
0.996
0.933


mitochondrial; Alpha-ketoglutarate dehydrogenase





Complex III subunit 7; Complex III subunit VII; Cytochrome b-c1 complex subunit 7; QP-
1.251
1.078
0.966


C; Ubiquinol-cytochrome c reductase complex 14 kDa protein; cDNA FLJ52271,





moderately similar to Ubiquinol-cytochrome c reductase complex 14 kDa protein (EC





1.10.2.2)





Calcium-activated chloride channel family member 1; Calcium-activated chloride channel
1.250
0.983
0.905


protein 1; Calcium-activated chloride channel regulator 1





Complement component 4A (Rodgers blood group); Putative uncharacterized protein
1.249
0.982
0.981


C4A; Complement component C4B (Childo blood group); Complement component C4B





(Childo blood group) 2; C4B1; Complement component 4B (Childo blood group)





Actin-interacting protein 1; NORI-1; WD repeat-containing protein 1; cDNA FLJ58303,
1.249
1.050
1.050


highly similar to WD repeat protein 1





Catalase
1.245
0.986
0.951


Proteasome subunit alpha type-7; Proteasome subunit RC6-1; Proteasome subunit
1.244
0.988
1.122


XAPC7; Proteasome subunit alpha type





Heat shock-related 70 kDa protein 2; cDNA FLJ40505 fis, clone TESTI2045562, highly
1.244
1.032
0.922


similar to HEAT SHOCK-RELATED 70 kDa PROTEIN 2





Endopeptidase SP18; Microsomal signal peptidase 18 kDa subunit; SEC11 homolog
1.240
1.009
1.057


A; SEC11-like protein 1; Signal peptidase complex catalytic subunit SEC11A; SPC18; cDNA





FLJ51313, highly similar to Microsomal signal peptidase 18 kDa subunit(EC 3.4.—.—);





SEC11-like 1 (S. cerevisiae), isoform CRA_d





Inorganic pyrophosphatase; Pyrophosphate phospho-hydrolase; Pyrophosphatase
1.238
0.962
0.998


(Inorganic) 1





Myosin heavy chain 11; Myosin heavy chain, smooth muscle isoform; Myosin-
1.234
0.963
0.862


11; SMMHC; Myosin heavy chain 11 smooth muscle isoform





Clathrin heavy chain 1; Clathrin heavy chain on chromosome 17
1.232
0.982
0.869


Villin-1; cDNA FLJ57609, highly similar to Villin-1
1.228
0.979
0.871


Centromere protein V; Nuclear protein p30; Proline-rich protein 6
1.227
1.172
1.067


Cathepsin C; Cathepsin J; Dipeptidyl peptidase 1; Dipeptidyl peptidase 1 exclusion domain
1.224
0.962
0.855


chain; Dipeptidyl peptidase 1 heavy chain; Dipeptidyl peptidase 1 light chain; Dipeptidyl





peptidase I; Dipeptidyl peptidase I exclusion domain chain; Dipeptidyl peptidase I heavy





chain; Dipeptidyl peptidase I light chain; Dipeptidyl transferase





Hydroxyacyl-coenzyme A dehydrogenase, mitochondrial; Medium and short-chain L-3-
1.224
1.009
0.903


hydroxyacyl-coenzyme A dehydrogenase; Short-chain 3-hydroxyacyl-CoA dehydrogenase





Aldo-keto reductase family 1 member B10; Aldose reductase-like; Aldose reductase-
1.223
0.954
0.851


related protein; ARL-1; Small intestine reductase





Carbonate dehydratase II; Carbonic anhydrase 2; Carbonic anhydrase C; Carbonic
1.223
1.031
0.910


anhydrase II





Putative uncharacterized protein PRRC1; Protein PRRC1
1.223
0.951
0.938


Cytosolic malate dehydrogenase; Malate dehydrogenase, cytoplasmic; Malate
1.222
0.988
0.875


dehydrogenase; Putative uncharacterized protein MDH1





Cadherin-associated Src substrate; Catenin delta-1; p120 catenin; p120(cas); Putative
1.222
1.013
0.894


uncharacterized protein CTNND1





Metavinculin; Vinculin
1.221
0.977
0.955


Amphiregulin-associated protein; Midgestation and kidney protein; Midkine; Neurite
1.220
1.206
1.070


outgrowth-promoting factor 2; Neurite outgrowth-promoting protein





Putative uncharacterized protein APEH; Acylamino-acid-releasing enzyme; Acylaminoacyl-
1.219
0.973
0.871


peptidase; Acyl-peptide hydrolase; Oxidized protein hydrolase





Kallikrein inhibitor; Kallistatin; Peptidase inhibitor 4; Serpin A4
1.218
1.258
1.166


Dihydrolipoamide dehydrogenase; Dihydrolipoyl dehydrogenase, mitochondrial; Glycine
1.213
0.980
0.911


cleavage system L protein; cDNA FLJ50515, highly similar to Dihydrolipoyl





dehydrogenase, mitochondrial (EC 1.8.1.4); Dihydrolipoyl dehydrogenase





5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase; AICAR
1.212
0.943
0.864


transformylase; ATIC; Bifunctional purine biosynthesis protein PURH; IMP





cyclohydrolase; IMP synthase; Inosinicase; Phosphoribosylaminoimidazolecarboxamide





formyltransferase





Glioma pathogenesis-related protein 2; Golgi-associated plant pathogenesis-related
1.210
1.027
0.947


protein 1; GLI pathogenesis-related 2





Signal transducer and activator of transcription 1-alpha/beta; Transcription factor ISGF-3
1.209
1.167
1.046


components p91/p84





PDHE1-A type I; Pyruvate dehydrogenase E1 component subunit alpha, somatic form,
1.205
0.948
0.932


mitochondrial





Glucose phosphomutase 1; Phosphoglucomutase-1; cDNA FLJ50606, highly similar to
1.202
0.935
0.998


Phosphoglucomutase-1 (EC 5.4.2.2)





18 kDa Alu RNA-binding protein; Signal recognition particle 14 kDa protein
1.200
1.015
0.912


Isovaleryl-CoA dehydrogenase, mitochondrial; cDNA FLJ16602 fis, clone TESTI4007816,
1.198
1.177
1.045


highly similar to Isovaleryl-CoA dehydrogenase, mitochondrial (EC 1.3.99.10); Isovaleryl





Coenzyme A dehydrogenase, isoform CRA_b





Cullin-associated and neddylation-dissociated protein 1; Cullin-associated NEDD8-
1.197
1.039
0.994


dissociated protein 1; p120 CAND1; TBP-interacting protein of 120 kDa A





Cytochrome c oxidase polypeptide VIc; Cytochrome c oxidase subunit 6C
1.196
1.056
0.935


Beta-hexosaminidase subunit alpha; Beta-N-acetylhexosaminidase subunit alpha; N-
1.194
1.105
1.110


acetyl-beta-glucosaminidase subunit alpha





HCNPpp; Hippocampal cholinergic neurostimulating peptide; Neuropolypeptide
1.193
0.958
0.996


h3; Phosphatidylethanolamine-binding protein 1; Prostatic-binding protein; Raf kinase





inhibitor protein; cDNA FLJ51535, highly similar to Phosphatidylethanolamine-binding





protein 1





59 kDa serine/threonine-protein kinase; ILK-1; ILK-2; Integrin-linked protein
1.192
1.090
0.965


kinase; p59ILK; cDNA FLJ50979, moderately similar to Integrin-linked protein kinase (EC





2.7.11.1); cDNA FLJ53825, highly similar to Integrin-linked protein kinase 1 (EC 2.7.11.1)





Collagen alpha-1 (XV) chain; Endostatin; Endostatin-XV; Related to endostatin; Restin
1.188
0.994
0.894


Desmoyokin; Neuroblast differentiation-associated protein AHNAK
1.187
0.968
0.893


HBeAg-binding protein 2 binding protein A; Mannose-P-dolichol utilization defect 1
1.183
0.933
0.904


protein; Suppressor of Lec15 and Lec35 glycosylation mutation homolog; My008





protein; cDNA FLJ57793, moderately similar to Mannose-P-dolichol utilization defect 1





protein; cDNA FLJ14836 fis, clone OVARC1001702





Outer mitochondrial membrane protein porin 1; Plasmalemmal porin; Porin 31HL; Porin
1.182
0.920
0.858


31HM; Voltage-dependent anion-selective channel protein 1





Alpha E-catenin; Cadherin-associated protein; Catenin alpha-1; Renal carcinoma antigen
1.180
0.933
0.847


NY-REN-13; cDNA FLJ54047, highly similar to Alpha-1 catenin (Cadherin-associated





protein); Catenin (Cadherin-associated protein), alpha 1, 102 kDa, isoform





CRA_c; CTNNA1 protein





Antigen KI-67
1.180
1.623
1.483


Glutamate dehydrogenase 1, mitochondrial; cDNA FLJ55203, highly similar to Glutamate
1.179
0.981
0.896


dehydrogenase 1, mitochondrial (EC 1.4.1.3); cDNA FLJ16138 fis, clone BRALZ2017531,





highly similar to Glutamate dehydrogenase 1, mitochondrial (EC 1.4.1.3); Glutamate





dehydrogenase 1, isoform CRA_a; Glutamate dehydrogenase 2, mitochondrial





Complex I-PDSW; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit
1.176
1.263
1.126


10; NADH-ubiquinone oxidoreductase PDSW subunit; NADH dehydrogenase (Ubiquinone)





1 beta subcomplex, 10, 22 kDa, isoform CRA_a; NDUFB10 protein





Hydroxysteroid dehydrogenase-like protein 2; cDNA FLJ61200, highly similar to Homo sapiens
1.175
0.941
0.868


hydroxysteroid dehydrogenase like 2 (HSDL2), mRNA





Elongation factor Ts, mitochondrial; Elongation factor Ts
1.169
1.294
1.165


5H9 antigen; CD9 antigen; Cell growth-inhibiting gene 2 protein; Leukocyte antigen
1.169
0.916
0.826


MIC3; Motility-related protein; p24; Tetraspanin-29; Putative uncharacterized protein





CD9; cDNA FLJ51032, highly similar to CD9 antigen





180 kDa ribosome receptor homolog; ES/130-related protein; Ribosome receptor
1.168
0.928
1.046


protein; Ribosome-binding protein 1





Hematopoietic cell-specific LYN substrate 1; Hematopoietic lineage cell-specific
1.167
1.083
1.017


protein; LckBP1; p75





DDAHII; Dimethylargininase-2; N(G),N(G)-dimethylarginine dimethylaminohydrolase
1.166
0.968
0.901


2; Protein G6a; S-phase protein; Dimethylarginine dimethylaminohydrolase 2





Glutathione S-transferase omega-1; Glutathione S-transferase omega 1, isoform
1.162
0.906
0.890


CRA_a; Glutathione S-transferase omega 1





Apoptotic chromatin condensation inducer in the nucleus
1.160
1.040
0.920


Aldehyde dehydrogenase family 6 member A1; Methylmalonate-semialdehyde
1.158
0.938
0.845


dehydrogenase [acylating], mitochondrial





Protein disulfide isomerase P5; Protein disulfide-isomerase A6; Thioredoxin domain-
1.158
0.924
0.953


containing protein 7; cDNA FLJ58502, highly similar to Protein disulfide-isomerase A6 (EC





5.3.4.1)





Nuclear transport factor 2; Placental protein 15
1.157
1.016
0.914


Complex I-23 kD; NADH dehydrogenase [ubiquinone] iron-sulfur protein 8,
1.151
1.148
1.025


mitochondrial; NADH-ubiquinone oxidoreductase 23 kDa subunit; TYKY subunit





Complex III subunit 5; Complex III subunit IX; Cytochrome b-c1 complex subunit
1.150
1.059
0.936


11; Cytochrome b-c1 complex subunit 5; Cytochrome b-c1 complex subunit Rieske,





mitochondrial; Rieske iron-sulfur protein; Ubiquinol-cytochrome c reductase 8 kDa





protein; Ubiquinol-cytochrome c reductase iron-sulfur subunit; Putative cytochrome b-c1





complex subunit Rieske-like protein 1





Nicotinamide phosphoribosyltransferase; Pre-B-cell colony-enhancing factor
1.148
0.893
0.823


1; Visfatin; Novel protein similar to Pre-B cell enhancing factor (PBEF)





Basic leucine zipper and W2 domain-containing protein 2; Putative uncharacterized
1.147
0.987
0.974


protein BZW2; Basic leucine zipper and W2 domains 2, isoform CRA_b





ATP-specific succinyl-CoA synthetase subunit beta; Renal carcinoma antigen NY-REN-
1.147
0.928
0.922


39; Succinyl-CoA ligase [ADP-forming] subunit beta, mitochondrial; Succinyl-CoA





synthetase beta-A chain; Succinate-CoA ligase, ADP-forming, beta subunit





3-hydroxyisobutyryl-CoA hydrolase, mitochondrial; 3-hydroxyisobutyryl-coenzyme A
1.145
1.152
1.058


hydrolase





E3 UFM1-protein ligase 1
1.143
1.066
1.053


UPF0197 transmembrane protein C11orf10
1.140
0.886
0.782


Interferon-induced protein 53; T1-TrpRS; T2-TrpRS; Tryptophan--tRNA
1.139
1.201
1.084


ligase; Tryptophanyl-tRNA synthetase, cytoplasmic





Tumor protein D52-like 2; Tumor protein D52-like 2, isoform CRA_e; Tumor protein D54
1.136
1.002
0.885


Glycogen phosphorylase, brain form
1.131
0.884
0.787


Cytosolic thyroid hormone-binding protein; Opa-interacting protein 3; p58; Pyruvate kinase
1.131
1.043
0.931


2/3; Pyruvate kinase isozymes M1/M2; Pyruvate kinase muscle isozyme; Thyroid hormone-





binding protein 1; Tumor M2-PK





58 kDa glucose-regulated protein; 58 kDa microsomal protein; Disulfide isomerase ER-
1.129
0.910
1.022


60; Endoplasmic reticulum resident protein 57; Endoplasmic reticulum resident protein





60; Protein disulfide-isomerase A3; cDNA PSEC0175 fis, clone OVARC1000169, highly





similar to Protein disulfide-isomerase A3 (EC 5.3.4.1)





Glutathione S-transferase kappa 1; Glutathione S-transferase subunit 13; GST 13-13; GST
1.128
0.978
0.995


class-kappa; GSTK1-1





Enoyl-CoA hydratase 1; Enoyl-CoA hydratase, mitochondrial; Short-chain enoyl-CoA
1.125
0.890
0.787


hydratase





Active breakpoint cluster region-related protein; cDNA FLJ54747, highly similar to Active
1.124
0.918
0.913


breakpoint cluster region-related protein





HLA-DR-associated protein II; Inhibitor of granzyme A-activated
1.118
1.145
1.099


DNase; PHAPII; Phosphatase 2A inhibitor I2PP2A; Protein SET; Template-activating factor





I; SET nuclear oncogene; Putative uncharacterized protein SET





Tubulin beta-2 chain; Tubulin beta-2C chain
1.117
0.900
0.805


Beta-II spectrin; Fodrin beta chain; Spectrin beta chain, brain 1; Spectrin, non-erythroid beta
1.117
0.905
0.800


chain 1





Cyclophilin B; CYP-S1; Peptidyl-prolyl cis-trans isomerase B; Rotamase B; S-cyclophilin
1.115
0.920
0.903


ATP synthase subunit a; F-ATPase protein 6
1.112
0.888
0.826


Putative uncharacterized protein ARPC4; Actin-related protein 2/3 complex subunit
1.106
0.931
0.919


4; Arp2/3 complex 20 kDa subunit





Carboxymethylenebutenolidase homolog
1.106
0.927
0.820


Vasodilator-stimulated phosphoprotein
1.106
1.087
0.995


47 kDa mannose 6-phosphate receptor-binding protein; Cargo selection protein
1.102
0.978
0.917


TIP47; Mannose-6-phosphate receptor-binding protein 1; Perilipin-3; Placental protein 17





All-trans-13,14-dihydroretinol saturase; All-trans-retinol 13,14-reductase
1.099
1.249
1.121


Beta-coat protein; Coatomer subunit beta; p102; cDNA FLJ56271, highly similar to
1.097
0.880
0.905


Coatomer subunit beta; Coatomer protein complex, subunit beta 2 (Beta prime), isoform





CRA_b





BPG-dependent PGAM 1; Phosphoglycerate mutase 1; Phosphoglycerate mutase isozyme B
1.096
1.042
0.940


Cadherin family member 5; Desmoglein-2; HDGC
1.096
1.083
1.010


Superoxide dismutase [Mn], mitochondrial; Superoxide dismutase
1.095
0.871
0.872


Interferon-induced 15 kDa protein; Interferon-induced 17 kDa protein; Ubiquitin cross-
1.093
0.918
0.810


reactive protein





Transmembrane and coiled-coil domain-containing protein 1; Transmembrane and coiled-
1.092
0.849
0.786


coil domains protein 4; Xenogeneic cross-immune protein PCIA3; Putative uncharacterized





protein TMCO1





Actin-binding protein 280; Alpha-filamin; Endothelial actin-binding protein; Filamin-
1.091
0.939
0.851


1; Filamin-A; Non-muscle filamin; Filamin A, alpha (Actin binding protein 280)





ABP-280-like protein; ABP-L; Actin-binding-like protein; Filamin-2; Filamin-C; Gamma-filamin
1.089
0.855
0.761


1-acylglycerophosphocholine O-acyltransferase; 1-acylglycerophosphoserine O-
1.088
1.242
1.118


acyltransferase; Lysophosphatidylcholine acyltransferase; Lysophosphatidylcholine





acyltransferase 3; Lysophosphatidylserine acyltransferase; Lysophospholipid





acyltransferase 5; Membrane-bound O-acyltransferase domain-containing protein 5; cDNA





FLJ55747, highly similar to Membrane bound O-acyltransferase domain-containing





protein 5 (EC 2.3.—.—)





Alcohol dehydrogenase 1C; Alcohol dehydrogenase subunit gamma
1.086
0.976
0.875


Beta-hexosaminidase subunit beta; Beta-hexosaminidase subunit beta chain A; Beta-
1.081
0.841
0.811


hexosaminidase subunit beta chain B; Beta-N-acetylhexosaminidase subunit beta; Cervical





cancer proto-oncogene 7 protein; N-acetyl-beta-glucosaminidase subunit beta; ENC-1AS





E3 ubiquitin/ISG15 ligase TRIM25; Estrogen-responsive finger protein; RING finger protein
1.079
0.982
0.925


147; Tripartite motif-containing protein 25; Ubiquitin/ISG15-conjugating enzyme





TRIM25; Zinc finger protein 147





p195; Ras GTPase-activating-like protein IQGAP1
1.078
0.877
0.859


Cytochrome c oxidase polypeptide IV; Cytochrome c oxidase subunit 4 isoform 1,
1.074
1.097
0.969


mitochondrial; Cytochrome c oxidase subunit IV isoform 1; COX4I1 protein





Intramembrane protease 1; Minor histocompatibility antigen H13; Presenilin-like protein
1.074
0.847
0.768


3; Signal peptide peptidase





Complex I-ASHI; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 8,
1.073
0.986
1.194


mitochondrial; NADH-ubiquinone oxidoreductase ASHI subunit; NADH dehydrogenase





(Ubiquinone) 1 beta subcomplex, 8, 19 kDa; NADH dehydrogenase (Ubiquinone) 1 beta





subcomplex, 8, 19 kDa, isoform CRA_a; cDNA FLJ52503, highly similar to NADH





dehydrogenase (ubiquinone) 1 beta subcomplex subunit 8, mitochondrial (EC 1.6.5.3)





(EC 1.6.99.3) (NADH-ubiquinone oxidoreductase ASHI subunit) (Complex I-ASHI) (CI-ASHI)





Cathepsin D; Cathepsin D heavy chain; Cathepsin D light chain
1.070
0.864
0.790


Golgi transport 1 homolog B; hGOT1a; Putative NF-kappa-B-activating protein 470; Vesicle
1.069
0.834
0.763


transport protein GOT1B





ADP-ribosylation factor 4; Putative uncharacterized protein ARF4
1.069
0.890
0.832


Calnexin; IP90; Major histocompatibility complex class I antigen-binding protein
1.068
0.857
0.983


p88; p90; cDNA FLJ54242, highly similar to Calnexin





Macropain subunit C8; Multicatalytic endopeptidase complex subunit C8; Proteasome
1.066
0.845
0.953


component C8; Proteasome subunit alpha type-3





Endoplasmic oxidoreductin-1-like protein; ERO1-like protein alpha; Oxidoreductin-1-L-
1.066
0.875
0.912


alpha





Elastin microfibril interface-located protein 1; EMILIN-1
1.064
0.846
0.788


Membrane protein p24A; Transmembrane emp24 domain-containing protein 2; cDNA
1.059
0.828
0.826


FLJ52153, highly similar to Transmembrane emp24 domain-containing protein 2





60 kDa SS-A/Ro ribonucleoprotein; Ro 60 kDa autoantigen; Sjoegren syndrome antigen
1.057
1.024
0.928


A2; Sjoegren syndrome type A antigen; TROVE domain family member 2; TROVE domain





family, member 2; TROVE domain family, member 2, isoform CRA_c; TROVE domain





family, member 2, isoform CRA_e; TROVE domain family, member 2, isoform CRA_d





Serine/threonine-protein phosphatase PP1-beta catalytic subunit
1.049
0.850
0.916


Complex I-49 kD; NADH dehydrogenase [ubiquinone] iron-sulfur protein 2,
1.048
0.883
0.931


mitochondrial; NADH-ubiquinone oxidoreductase 49 kDa subunit; cDNA, FLJ78876, highly





similar to NADH-ubiquinone oxidoreductase 49 kDa subunit, mitochondrial (EC 1.6.5.3)





Glycoprotein GP36b; Lectin mannose-binding 2; Vesicular integral-membrane protein
1.039
0.809
0.779


VIP36; cDNA FLJ52285, highly similar to Vesicular integral-membrane protein VIP36





Azoreductase; DT-diaphorase; Menadione reductase; NAD(P)H dehydrogenase [quinone]
1.039
0.880
0.848


1; NAD(P)H: quinone oxidoreductase 1; Phylloquinone reductase; Quinone reductase





1; cDNA FLJ50573, highly similar to Homo sapiens NAD(P)H dehydrogenase, quinone 1





(NQO1), transcript variant 3, mRNA





Fortilin; Histamine-releasing factor; p23; Translationally-controlled tumor protein; TPT1
1.037
0.854
0.770


protein; Tumor protein, translationally-controlled 1; Tumor protein, translationally-controlled





1, isoform CRA_a





Adapter protein CMS; Cas ligand with multiple SH3 domains; CD2-associated protein
1.033
1.063
0.962


Oxysterol-binding protein 1
1.032
1.125
1.001


B5; Dolichyl-diphosphooligosaccharide-protein glycosyltransferase subunit
1.029
0.966
0.888


STT3A; lntegral membrane protein 1; Transmembrane protein TMC





11S regulator complex subunit alpha; Activator of multicatalytic protease subunit
1.028
1.069
0.953


1; Interferon gamma up-regulated I-5111 protein; Proteasome activator 28 subunit





alpha; Proteasome activator complex subunit 1; Putative uncharacterized protein PSME1





CFR-1; Cysteine-rich fibroblast growth factor receptor; E-selectin ligand 1; Golgi apparatus
1.028
0.879
0.890


protein 1; Golgi sialoglycoprotein MG-160





Ubiquitin carrier protein D3; Ubiquitin-conjugating enzyme E2 D3; Ubiquitin-conjugating
1.028
0.940
0.831


enzyme E2(17)KB 3; Ubiquitin-conjugating enzyme E2-17 kDa 3; Ubiquitin-protein ligase





D3; Ubiquitin carrier protein D2; Ubiquitin-conjugating enzyme E2 D2; Ubiquitin-conjugating





enzyme E2(17)KB 2; Ubiquitin-conjugating enzyme E2-17 kDa 2; Ubiquitin-protein ligase





D2; Ubiquitin carrier protein





Activated RNA polymerase II transcriptional coactivator p15; p14; Positive cofactor 4; SUB1
1.024
0.913
0.818


homolog





HLA-B-associated transcript 3; Large proline-rich protein BAT3; Protein G3; HLA-B
1.024
0.942
0.836


associated transcript 3; HLA-B associated transcript 3, isoform CRA_a





Cytochrome c oxidase polypeptide II; Cytochrome c oxidase subunit 2
1.024
1.008
1.048


Histone H1; Histone H1(0); Histone H1.0
1.022
0.846
0.748


Echinoderm microtubule-associated protein-like 4; Restrictedly overexpressed
1.021
0.899
0.822


proliferation-associated protein; Ropp 120; Putative uncharacterized protein EML4





Fructose-bisphosphate aldolase A; Lung cancer antigen NY-LU-1; Muscle-type aldolase
1.019
0.828
0.750


High density lipoprotein-binding protein; Vigilin
1.018
0.791
0.705


32 kDa accessory protein; Vacuolar proton pump subunit d 1; V-ATPase 40 kDa accessory
1.015
0.810
0.900


protein; V-ATPase AC39 subunit; V-type proton ATPase subunit d 1





NADH dehydrogenase [ubiquinone] flavoprotein 2, mitochondrial; NADH-ubiquinone
1.015
0.833
0.956


oxidoreductase 24 kDa subunit





Maternal-embryonic 3; Vacuolar protein sorting-associated protein 35; Vesicle protein
1.013
0.795
0.951


sorting 35





Histone H3
1.013
0.825
0.729


17-beta-hydroxysteroid dehydrogenase type 2; 20 alpha-hydroxysteroid
1.013
1.457
1.303


dehydrogenase; E2DH; Estradiol 17-beta-dehydrogenase 2; Microsomal 17-beta-





hydroxysteroid dehydrogenase; Testosterone 17-beta-dehydrogenase





56 kDa selenium-binding protein; Selenium-binding protein 1; cDNA FLJ61035, highly
1.013
0.789
0.706


similar to Selenium-binding protein 1; Selenium binding protein 1





Cell proliferation-inducing gene 19 protein; LDH muscle subunit; L-lactate dehydrogenase
1.006
0.936
1.001


A chain; Renal carcinoma antigen NY-REN-59





Tight junction protein 1; Tight junction protein ZO-1; Zona occludens protein 1; Zonula
1.002
1.323
1.170


occludens protein 1





Ras-related protein Rab-18; RAB18, member RAS oncogene family
1.000
0.849
1.001


Epithelial protein lost in neoplasm; LIM domain and actin-binding protein 1; cDNA
1.000
0.980
0.875


FLJ55990, highly similar to LIM domain and actin-binding protein 1





Iron regulatory protein 2; Iron-responsive element-binding protein 2
0.999
0.786
0.761


G protein subunit beta-2; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-
0.995
0.799
0.785


2; Transducin beta chain 2; Putative uncharacterized protein GNB2





40S ribosomal protein S13
0.991
0.857
0.891


Heat shock 70 kDa protein 1/2; Heat shock 70 kDa protein 1A/1B; Heat shock 70 kDa
0.989
1.199
1.082


protein 1A





LIM and SH3 domain protein 1; Metastatic lymph node gene 50 protein; Putative
0.981
0.977
0.872


uncharacterized protein LASP1; cDNA FLJ51834, highly similar to LIM and SH3 domain





protein 1; cDNA FLJ52195, highly similar to LIM and SH3 domain protein 1





60S ribosomal protein L26
0.981
0.765
0.972


20 kDa myosin light chain; MLC-2C; Myosin regulatory light chain 2, smooth muscle
0.981
1.027
0.924


isoform; Myosin regulatory light chain 9; Myosin regulatory light chain MRLC1; Myosin





regulatory light polypeptide 9; Myosin RLC





Guanine nucleotide-binding protein G(y) subunit alpha; Guanine nucleotide-binding protein
0.978
0.764
0.780


subunit alpha-11





Meg-3; Niban-like protein 1; Protein FAM129B
0.972
0.829
0.789


ADP-ribosylation factor-like protein 6-interacting protein 5; Cytoskeleton-related vitamin A-
0.969
1.128
1.069


responsive protein; Dermal papilla-derived protein 11; Glutamate transporter EAAC1-





interacting protein; GTRAP3-18; JM5; PRA1 family protein 3; Prenylated Rab acceptor





protein 2; Protein JWa; Putative MAPK-activating protein PM27





60S ribosomal protein L27
0.968
0.799
0.741


Beta-globin; Hemoglobin beta chain; Hemoglobin subunit beta; LVV-hemorphin-7
0.967
0.996
1.014


GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase-4-reductase; GDP-L-fucose
0.964
0.927
0.848


synthase; Protein FX; Red cell NADP(H)-binding protein; Short-chain





dehydrogenase/reductase family 4E member 1





26S protease regulatory subunit 6A; 26S proteasome AAA-ATPase subunit
0.963
1.125
1.059


RPT5; Proteasome 26S subunit ATPase 3; Proteasome subunit P50; Tat-binding protein 1





Apolipoprotein A-l-binding protein; YjeF N-terminal domain-containing protein 1
0.958
1.024
0.970


APEX nuclease; Apurinic-apyrimidinic endonuclease 1; DNA-(apurinic or apyrimidinic site)
0.954
1.016
0.900


lyase; Protein REF-1





Signal recognition particle 54 kDa protein
0.953
0.743
0.667


5F7; Basigin; Collagenase stimulatory factor; Extracellular matrix metalloproteinase
0.952
0.794
0.849


inducer; Leukocyte activation antigen M6; OK blood group antigen; Tumor cell-derived





collagenase stimulatory factor





AIR carboxylase; Multifunctional protein ADE2; Phosphoribosylaminoimidazole
0.952
0.740
0.673


carboxylase; Phosphoribosylaminoimidazole-succinocarboxamide synthase; SAICAR





synthetase





70 kDa peroxisomal membrane protein; ATP-binding cassette sub-family D member 3
0.951
0.926
0.853


Actin, aortic smooth muscle; Alpha-actin-2; Cell growth-inhibiting gene 46 protein; Actin,
0.951
0.935
0.971


alpha 1, skeletal muscle





3-hydroxybutyrate dehydrogenase type 2; Dehydrogenase/reductase SDR family member
0.946
0.753
0.896


6; Oxidoreductase UCPA; R-beta-hydroxybutyrate dehydrogenase





Kinesin light chain 4; Kinesin-like protein 8; cDNA FLJ58264, highly similar to Kinesin light
0.945
1.267
1.218


chain 4





Deubiquitinating enzyme 15; Ubiquitin carboxyl-terminal hydrolase 15; Ubiquitin
0.944
1.145
1.049


thioesterase 15; Ubiquitin-specific-processing protease 15; Unph-2; Unph4





Putative uncharacterized protein KIAA0664; Protein KIAA0664
0.933
0.738
0.830


Protein SCO2 homolog, mitochondrial
0.928
0.744
0.657


35-alpha calcimedin; Annexin A3; Annexin III; Annexin-3; Inositol 1,2-cyclic phosphate 2-
0.926
1.006
0.966


phosphohydrolase; Lipocortin III; Placental anticoagulant protein III





Inosine phosphorylase; Purine nucleoside phosphorylase
0.919
0.803
0.816


Complex I-B8; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 2; NADH-
0.916
0.719
0.780


ubiquinone oxidoreductase B8 subunit





Lamin-B2
0.914
0.712
0.910


Signal transducing adapter molecule 1; Putative uncharacterized protein STAM
0.906
0.965
0.883


Plasma membrane calcium ATPase isoform 1; Plasma membrane calcium pump isoform
0.896
0.797
0.826


1; Plasma membrane calcium-transporting ATPase 1





Carbonyl reductase [NADPH] 3; NADPH-dependent carbonyl reductase 3
0.896
1.061
1.298


DNA-directed RNA polymerase II subunit H; DNA-directed RNA polymerases I, II, and III
0.892
1.439
1.271


17.1 kDa polypeptide; DNA-directed RNA polymerases I, II, and III subunit





RPABC3; RPB17; RPB8 homolog; Putative uncharacterized protein POLR2H





70 kDa lamin; Lamin-A/C; Renal carcinoma antigen NY-REN-32; Lamin
0.888
0.914
0.853


A/C; Progerin; Rhabdomyosarcoma antigen MU-RMS-40.12





22 kDa neuronal tissue-enriched acidic protein; Brain acid soluble protein 1; Neuronal
0.888
0.915
0.984


axonal membrane protein NAP-22





ADP-ribosylation factor-like protein 3
0.884
1.055
1.005


Acetylglucosamine phosphomutase; N-acetylglucosamine-phosphate
0.884
0.712
0.632


mutase; Phosphoacetylglucosamine mutase; Phosphoglucomutase-3





Cytochrome c oxidase polypeptide VIIc; Cytochrome c oxidase subunit 7C, mitochondrial
0.879
0.913
0.839


Dolichyl-diphosphooligosaccharide--protein glycosyltransferase 67 kDa subunit; Dolichyl-
0.878
0.715
0.974


diphosphooligosaccharide--protein glycosyltransferase subunit 1; Ribophorin I; Ribophorin-





1; cDNA FLJ50809, highly similar to Dolichyl-diphosphooligosaccharide--protein





glycosyltransferase 67 kDa subunit (EC 2.4.1.119); cDNA FLJ51908, highly similar to





Dolichyl-diphosphooligosaccharide--proteinglycosyltransferase 67 kDa subunit (EC





2.4.1.119)





Cytochrome c oxidase subunit 7A2, mitochondrial; Cytochrome c oxidase subunit VIIa-
0.875
0.705
0.933


liver/heart





21 kDa transmembrane-trafficking protein; p24delta; S31III125; Tmp-21-I; Transmembrane
0.875
0.690
0.754


emp24 domain-containing protein 10; Transmembrane protein Tmp21





Cell cycle control protein 50A; Transmembrane protein 30A; cDNA FLJ55687, highly
0.868
2.133
1.883


similar to Cell cycle control protein 50A





Oxidative stress-responsive 1 protein; Serine/threonine-protein kinase OSR1; Putative
0.864
1.203
1.066


uncharacterized protein OXSR1





Acyl-CoA-binding domain-containing protein 3; Golgi complex-associated protein 1; Golgi
0.861
1.201
1.111


phosphoprotein 1; Golgi resident protein GCP60; PBR- and PKA-associated protein





7; Peripheral benzodiazepine receptor-associated protein PAP7





Pyruvate carboxylase, mitochondrial; Pyruvic carboxylase; cDNA FLJ60715, highly similar
0.857
1.001
0.885


to Pyruvate carboxylase, mitochondrial (EC 6.4.1.1)





Double-stranded RNA-binding protein Staufen homolog 1; Staufen, RNA binding protein,
0.855
1.316
1.229


homolog 1 (Drosophila)





Q15149-6
0.851
0.704
1.087


40S ribosomal protein S19
0.848
0.732
0.944


Intestine-specific plastin; Plastin-1
0.845
1.185
1.053


Nidogen-2; Osteonidogen
0.836
1.244
1.104


Gastric cancer antigen Ga19; N-alpha-acetyltransferase 15, NatA auxiliary subunit; NMDA
0.833
0.751
0.704


receptor-regulated protein 1; N-terminal acetyltransferase; Protein tubedown-1; Tbdn100





40S ribosomal protein S14
0.832
0.654
0.615


Heterogeneous nuclear ribonucleoprotein L; cDNA FLJ75895, highly similar to Homo sapiens
0.829
0.747
0.663


heterogeneous nuclear ribonucleoprotein L (HNRPL), transcript variant 2,





mRNA; Putative uncharacterized protein HNRNPL





cDNA FLJ56102, highly similar to Homo sapiens calpastatin (CAST), transcript variant 8,
0.826
0.782
0.694


mRNA; Calpain inhibitor; Calpastatin; Sperm BS-17 component; cDNA FLJ56123, highly





similar to Calpastatin





Tropomyosin 3; Tropomyosin 3, isoform CRA_b; cDNA FLJ35393 fis, clone
0.826
0.829
0.732


SKNSH2000971, highly similar to TROPOMYOSIN, CYTOSKELETAL TYPE





Macropain chain Z; Multicatalytic endopeptidase complex chain Z; Proteasome subunit
0.825
0.745
0.685


beta type-7; Proteasome subunit Z; Proteasome (Prosome, macropain) subunit, beta type,





7; cDNA FLJ60039, highly similar to Proteasome subunit beta type 7 (EC





3.4.25.1); Proteasome (Prosome, macropain) subunit, beta type, 7, isoform CRA_b





DNA-directed RNA polymerase II 140 kDa polypeptide; DNA-directed RNA polymerase II
0.824
1.499
1.425


subunit B; DNA-directed RNA polymerase II subunit RPB2; RNA polymerase II subunit





2; RNA polymerase II subunit B2; Putative uncharacterized protein POLR2B





40S ribosomal protein S7; Putative uncharacterized protein RPS7
0.824
0.813
0.982


Thyroid hormone receptor-associated protein 3; Thyroid hormone receptor-associated
0.823
1.280
1.131


protein complex 150 kDa component





Large tumor suppressor homolog 1; Serine/threonine-protein kinase LATS1; WARTS
0.822
0.768
0.718


protein kinase; LATS1 protein





11S regulator complex subunit beta; Activator of multicatalytic protease subunit
0.819
1.303
1.157


2; Proteasome activator 28 subunit beta; Proteasome activator complex subunit 2





Leukocyte common antigen; Receptor-type tyrosine-protein phosphatase C; T200
0.819
0.976
0.975


Tight junction protein 2; Tight junction protein ZO-2; Zona occludens protein 2; Zonula
0.811
0.919
0.836


occludens protein 2





CDC42 GTPase-activating protein; GTPase-activating protein rhoOGAP; p50-
0.805
0.963
1.011


RhoGAP; Rho GTPase-activating protein 1; Rho-related small GTPase protein





activator; Rho-type GTPase-activating protein 1





Calcium-activated neutral proteinase 2; Calpain large polypeptide L2; Calpain M-
0.804
1.056
1.017


type; Calpain-2 catalytic subunit; Calpain-2 large subunit; Millimolar-calpain





ABP125; ABP130; Protein transport protein Sec31A; SEC31-like protein 1; SEC31-related
0.798
0.622
0.895


protein A; Web1-like protein; SEC31A protein





Electron transfer flavoprotein subunit beta
0.797
0.696
0.886


Collapsin response mediator protein 2; Dihydropyrimidinase-related protein 2; N2A3; Unc-
0.796
0.874
1.048


33-like phosphoprotein 2





DEAD box protein 27; Probable ATP-dependent RNA helicase DDX27
0.781
1.299
1.171


Copper amine oxidase; HPAO; Membrane primary amine oxidase; Semicarbazide-sensitive
0.778
0.845
0.888


amine oxidase; Vascular adhesion protein 1





Dolichyl-diphosphooligosaccharide--protein glycosyltransferase 63 kDa subunit; Dolichyl-
0.777
0.766
1.033


diphosphooligosaccharide-protein glycosyltransferase subunit 2; RIBIIR; Ribophorin





II; Ribophorin-2





Argininosuccinate synthase; Citrulline--aspartate ligase
0.776
0.987
0.880


Glycoprotein 25L2; Transmembrane emp24 domain-containing protein 9
0.772
1.404
1.242


NAP-1-related protein; Nucleosome assembly protein 1-like 1; cDNA FLJ30458 fis, clone
0.763
1.080
0.954


BRACE2009421, highly similar to NUCLEOSOME ASSEMBLY PROTEIN 1-LIKE 1; cDNA





FLJ58569, highly similar to Nucleosome assembly protein 1-like 1; cDNA FLJ16112 fis,





clone 3NB692001853, highly similar to NUCLEOSOME ASSEMBLY PROTEIN 1-LIKE





1; Nucleosome assembly protein 1-like 1, isoform CRA_c





TESS; Testin
0.761
1.013
0.896


14 kDa phosphohistidine phosphatase; Phosphohistidine phosphatase 1; Protein janus-A
0.761
0.888
0.888


homolog





Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Transducin beta chain
0.758
1.071
1.024


1; Guanine nucleotide binding protein (G protein), beta polypeptide 1





130 kDa leucine-rich protein; GP130; Leucine-rich PPR motif-containing protein,
0.756
0.741
0.954


mitochondrial





Cellugyrin; Synaptogyrin-2
0.754
0.685
0.839


c-Ki-ras; c-K-ras; GTPase KRas; GTPase KRas, N-terminally processed; Ki-Ras; K-Ras 2
0.750
1.185
1.075


80K-H protein; Glucosidase 2 subunit beta; Glucosidase II subunit beta; Protein kinase C
0.749
0.874
1.036


substrate 60.1 kDa protein heavy chain





Haptoglobin; Haptoglobin alpha chain; Haptoglobin beta chain; HP protein
0.746
0.838
1.044


49 kDa TATA box-binding protein-interacting protein; 54 kDa erythrocyte cytosolic
0.742
0.585
0.524


protein; INO80 complex subunit H; Nuclear matrix protein 238; Pontin 52; RuvB-like





1; TIP49a; TIP60-associated protein 54-alpha





Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1; Peptidyl-prolyl cis-trans isomerase
0.741
0.769
0.689


Pin1; Rotamase Pin1





Acid ceramidase; Acid ceramidase subunit alpha; Acid ceramidase subunit
0.740
1.345
1.559


beta; Acylsphingosine deacylase; N-acylsphingosine amidohydrolase; Putative 32 kDa





heart protein





Annexin A2; Annexin II; Annexin-2; Calpactin I heavy chain; Calpactin-1 heavy
0.740
0.626
0.871


chain; Chromobindin-8; Lipocortin II; p36; Placental anticoagulant protein IV; Protein





I; Annexin A2 pseudogene 2; Lipocortin II pseudogene; Putative annexin A2-like





protein; cDNA FLJ34687 fis, clone MESAN2000620, highly similar to Annexin A2





Translocation protein 1; Translocation protein SEC62
0.735
1.515
1.341


Smu-1 suppressor of mec-8 and unc-52 protein homolog; WD40 repeat-containing protein
0.723
1.363
1.272


SMU1; cDNA FLJ54259, highly similar to Smu-1 suppressor of mec-8 and unc-52 protein





homolog





UPF0568 protein C14orf166
0.722
0.730
0.998


NADPH-cytochrome P450 reductase
0.721
1.142
1.009


60S ribosomal protein L3; HIV-1 TAR RNA-binding protein B; Putative uncharacterized
0.715
0.561
0.512


protein RPL3





Actin-related protein 2/3 complex subunit 5; Arp2/3 complex 16 kDa subunit
0.709
0.599
0.724


Citrate synthase, mitochondrial; Citrate synthase
0.706
0.689
0.932


ATP-dependent 61 kDa nucleolar RNA helicase; DEAD box protein 21; DEAD box protein
0.705
0.549
0.971


56; Probable ATP-dependent RNA helicase DDX56; Putative uncharacterized protein





DDX56





Ribosome maturation protein SBDS; Shwachman-Bodian-Diamond syndrome protein
0.705
0.681
0.704


Selenide, water dikinase 1; Selenium donor protein 1; Selenophosphate synthase 1; cDNA
0.699
1.296
1.294


FLJ60186, highly similar to Selenide, water dikinase 1 (EC 2.7.9.3); Selenophosphate





synthetase 1; Selenophosphate synthetase 1, isoform CRA_a





eIF-2B GDP-GTP exchange factor subunit epsilon; Translation initiation factor eIF-2B
0.691
1.419
1.313


subunit epsilon





C-terminal LIM domain protein 1; Elfin; LIM domain protein CLP-36; PDZ and LIM domain
0.673
0.568
0.950


protein 1





Calcyclin; Growth factor-inducible protein 2A9; MLN 4; Prolactin receptor-associated
0.672
0.522
0.519


protein; Protein S100-A6; S100 calcium-binding protein A6





Dolichyl-diphosphooligosaccharide--protein glycosyltransferase 48 kDa subunit
0.670
0.522
0.898


ICD-M; IDP; Isocitrate dehydrogenase [NADP], mitochondrial; NADP(+)-specific
0.669
1.139
1.036


ICDH; Oxalosuccinate decarboxylase





100 kDa coactivator; EBNA2 coactivator p100; p100 co-activator; Staphylococcal nuclease
0.665
0.600
0.914


domain-containing protein 1; Tudor domain-containing protein 11





Glutaredoxin-1; Thioltransferase-1
0.656
1.176
1.038


Microsomal triglyceride transfer protein large subunit
0.639
1.044
1.044


Beta-coat protein; Coatomer subunit beta
0.638
0.631
0.946


60S ribosomal protein L23a; Putative uncharacterized protein RPL23A; Ribosomal protein
0.634
0.711
0.945


L23a, isoform CRA_a





14-3-3 protein eta; Protein AS1; Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase
0.626
1.253
1.129


activation protein, eta polypeptide





Putative uncharacterized protein SUMO1; GAP-modifying protein 1; Sentrin; Small
0.623
0.485
0.694


ubiquitin-related modifier 1; SMT3 homolog 3; Ubiquitin-homology domain protein





PIC1; Ubiquitin-like protein SMT3C; Ubiquitin-like protein UBL1; SMT3 suppressor of mif





two 3 homolog 1 (Yeast), isoform CRA_c; SMT3 suppressor of mif two 3 homolog 1





(Yeast), isoform CRA_b





Complex I-15 kDa; NADH dehydrogenase [ubiquinone] iron-sulfur protein 5; NADH-
0.622
0.602
0.912


ubiquinone oxidoreductase 15 kDa subunit





Importin-7; Ran-binding protein 7
0.621
0.754
0.722


Dnm1p/Vps1p-like protein; Dynamin family member proline-rich carboxyl-terminal domain
0.614
1.328
1.172


less; Dynamin-1-like protein; Dynamin-like protein; Dynamin-like protein 4; Dynamin-like





protein IV; Dynamin-related protein 1; cDNA FLJ59504, highly similar to Dynamin-1-like





protein (EC 3.6.5.5)





Activated-platelet protein 1; Inducible poly(A)-binding protein; Polyadenylate-binding
0.611
0.710
0.994


protein 4; Poly(A) binding protein, cytoplasmic 4 (Inducible form); Poly(A) binding protein,





cytoplasmic 4 (Inducible form), isoform CRA_e





Succinyl-CoA ligase [GDP-forming] subunit alpha, mitochondrial; Succinyl-CoA synthetase
0.604
0.498
0.906


subunit alpha





27 kDa prosomal protein; Macropain iota chain; Multicatalytic endopeptidase complex iota
0.603
1.148
1.138


chain; Proteasome iota chain; Proteasome subunit alpha type-6; cDNA FLJ51729, highly





similar to Proteasome subunit alpha type 6 (EC 3.4.25.1); Proteasome (Prosome,





macropain) subunit, alpha type, 6, isoform CRA_a; cDNA FLJ52022, highly similar to





Proteasome subunit alpha type 6 (EC 3.4.25.1); cDNA, FLJ79122, highly similar to





Proteasome subunit alpha type 6 (EC 3.4.25.1)





Cytosol aminopeptidase; Leucine aminopeptidase 3; Leucyl aminopeptidase; Peptidase
0.599
0.963
0.857


S; Proline aminopeptidase; Prolyl aminopeptidase





60S ribosomal protein L14; CAG-ISL 7; cDNA FLJ51325, highly similar to 60S ribosomal
0.589
0.522
0.744


protein L14





DNA topoisomerase 1; DNA topoisomerase I
0.575
0.715
0.997


40S ribosomal protein S26; Putative 40S ribosomal protein S26-like 1
0.571
0.777
0.753


Rho GDP-dissociation inhibitor 1; Rho-GDI alpha; cDNA FLJ50748, highly similar to Rho
0.562
0.438
1.074


GDP-dissociation inhibitor 1; Putative uncharacterized protein ARHGDIA





Hsc70/Hsp90-organizing protein; Renal carcinoma antigen NY-REN-11; Stress-induced-
0.550
0.715
1.000


phosphoprotein 1; Transformation-sensitive protein IEF SSP 3521





Signal sequence receptor subunit delta; Translocon-associated protein subunit
0.548
0.602
0.556


delta; Putative uncharacterized protein SSR4





Putative uncharacterized protein MUTED
0.547
0.734
0.906


ATPase family AAA domain-containing protein 3A
0.544
0.612
0.978


Nuclear distribution protein C homolog; Nuclear migration protein nudC
0.540
0.429
0.779


Protein LSM12 homolog
0.530
0.932
1.102


Protein transport protein Sec61 subunit beta
0.528
0.579
0.791


Macropain subunit C5; Multicatalytic endopeptidase complex subunit C5; Proteasome
0.509
1.172
1.039


component C5; Proteasome gamma chain; Proteasome subunit beta type-1





Alcohol dehydrogenase 5; Alcohol dehydrogenase class chi chain; Alcohol dehydrogenase
0.508
0.456
0.936


class-3; Alcohol dehydrogenase class-III; Glutathione-dependent formaldehyde





dehydrogenase; S-(hydroxymethyl)glutathione dehydrogenase





DEAD box protein 47; Probable ATP-dependent RNA helicase DDX47
0.502
0.638
0.755


Macropain delta chain; Multicatalytic endopeptidase complex delta chain; Proteasome
0.499
1.013
1.008


delta chain; Proteasome subunit beta type-6; Proteasome subunit Y





Proteasome chain 13; Proteasome component C10-II; Proteasome subunit beta type-
0.499
1.147
1.422


3; Proteasome theta chain





Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-5
0.493
0.673
1.014


60S ribosomal protein L35
0.480
0.523
0.899


Putative uncharacterized protein DBN1; Developmentally-regulated brain protein; Drebrin
0.468
0.770
0.774


tRNA pseudouridine synthase A; tRNA pseudouridylate synthase I; tRNA-uridine
0.467
0.877
0.963


isomerase I





Protein NDRG2; Protein Syld709613; cDNA FLJ55190, highly similar to Protein NDRG2
0.467
1.647
1.485


AKAP 120-like protein; A-kinase anchor protein 350 kDa; A-kinase anchor protein 450
0.465
0.785
0.703


kDa; A-kinase anchor protein 9; Centrosome- and Golgi-localized PKN-associated





protein; Protein hyperion; Protein kinase A-anchoring protein 9; Protein yotiao





Anandamide amidohydrolase 1; Fatty-acid amide hydrolase 1; Oleamide hydrolase 1
0.462
0.612
1.046


Acyl-coenzyme A thioesterase 13; Thioesterase superfamily member 2
0.461
1.134
1.032


40S ribosomal protein S21; RPS21 protein; Ribosomal protein S21; Ribosomal protein S21,
0.456
0.594
0.980


isoform CRA_e





NSFL1 cofactor p47; p97 cofactor p47; UBX domain-containing protein 2C
0.448
0.600
1.023


Glutaredoxin-3; PKC-interacting cousin of thioredoxin; PKC-theta-interacting
0.445
0.808
0.971


protein; Thioredoxin-like protein 2





Charcot-Leyden crystal protein; CLC; Eosinophil lysophospholipase; Galectin-
0.445
1.579
1.477


10; Lysolecithin acylhydrolase





LanC-like protein 2; Testis-specific adriamycin sensitivity protein
0.441
1.599
1.423


Dimethylallyltranstransferase; Farnesyl diphosphate synthase; Farnesyl pyrophosphate
0.441
0.469
0.821


synthase; Geranyltranstransferase





26S protease regulatory subunit 7; 26S proteasome AAA-ATPase subunit
0.436
0.409
0.774


RPT1; Proteasome 26S subunit ATPase 2; Protein MSS1; cDNA FLJ52353, highly similar





to 26S protease regulatory subunit 7





CCT-epsilon; T-complex protein 1 subunit epsilon
0.432
0.768
0.859


Glyceraldehyde-3-phosphate dehydrogenase
0.428
0.605
0.994


Ras-related protein Rab-6A
0.427
0.874
1.046


Nucleosome assembly protein 1-like 4b; Putative uncharacterized protein
0.426
0.331
0.996


NAP1L4; Nucleosome assembly protein 1-like 4; Nucleosome assembly protein 2





Myosin-VI; Unconventional myosin-6
0.422
0.789
0.865


26S proteasome non-ATPase regulatory subunit 3; 26S proteasome regulatory subunit
0.420
0.562
0.992


RPN3; 26S proteasome regulatory subunit S3; Proteasome subunit p58; cDNA FLJ54148,





highly similar to 26S proteasome non-ATPase regulatory subunit 3





Placental ribonuclease inhibitor; Ribonuclease inhibitor; Ribonuclease/angiogenin
0.419
0.470
0.883


inhibitor 1





30 kDa prosomal protein; Macropain subunit C2; Multicatalytic endopeptidase complex
0.402
0.563
0.994


subunit C2; Proteasome component C2; Proteasome nu chain; Proteasome subunit alpha





type-1; Proteasome subunit alpha type





IMPDH-II; Inosine-5-monophosphate dehydrogenase 2
0.401
0.356
0.652


Bax antagonist selected in saccharomyces 1; Negative regulatory element-binding
0.401
0.560
0.906


protein; Protein DBP-5; Protein SON; SON3





AGX-1; AGX-2; Antigen X; Sperm-associated antigen 2; UDP-N-acetylgalactosamine
0.400
1.176
1.039


pyrophosphorylase; UDP-N-acetylglucosamine pyrophosphorylase; UDP-N-





acetylhexosamine pyrophosphorylase; UDP-N-acteylglucosamine pyrophosphorylase 1





Heat shock 70 kDa protein 4; Heat shock 70-related protein APG-2; HSP70RY
0.390
1.029
1.069


Autocrine motility factor; Glucose-6-phosphate isomerase; Neuroleukin; Phosphoglucose
0.383
0.317
0.849


isomerase; Phosphohexose isomerase; Sperm antigen 36





ATP-dependent helicase SMARCA2; BRG1-associated factor 190B; Probable global
0.379
0.669
1.095


transcription activator SNF2L2; Protein brahma homolog; SNF2-alpha; SWI/SNF-related





matrix-associated actin-dependent regulator of chromatin subfamily A member 2





PEP11 homolog; Vacuolar protein sorting-associated protein 29; Vesicle protein sorting 29
0.366
1.199
1.100


Deubiquitinating enzyme 7; Herpesvirus-associated ubiquitin-specific protease; Ubiquitin
0.365
0.730
0.991


carboxyl-terminal hydrolase 7; Ubiquitin thioesterase 7; Ubiquitin-specific-processing





protease 7; Ubiquitin carboxyl-terminal hydrolase





Butyrate-induced protein 1; Protein tyrosine phosphatase-like protein PTPLAD1; Protein-
0.365
0.301
0.736


tyrosine phosphatase-like A domain-containing protein 1; cDNA FLJ54138, highly similar





to Homo sapiens butyrate-induced transcript 1 (HSPC121), mRNA





Amine oxidase [flavin-containing] B; Monoamine oxidase type B; cDNA FLJ51821, highly
0.364
1.217
1.126


similar to Amine oxidase (flavin-containing) B (EC 1.4.3.4); cDNA FLJ52418, highly similar





to Amine oxidase (flavin-containing) B (EC 1.4.3.4)





Cell proliferation-inducing gene 21 protein; Guanine nucleotide-binding protein subunit
0.361
0.644
0.979


beta-2-like 1; Guanine nucleotide-binding protein subunit beta-like protein 12.3; Human





lung cancer oncogene 7 protein; Receptor for activated C kinase; Receptor of activated





protein kinase C 1





CAAX farnesyltransferase subunit alpha; FTase-alpha; Protein
0.356
1.417
1.499


farnesyltransferase/geranylgeranyltransferase type-1 subunit alpha; Ras proteins





prenyltransferase subunit alpha; Type I protein geranyl-geranyltransferase subunit alpha





1F5 antigen; 20 kDa homologous restriction factor; CD59 glycoprotein; MAC-inhibitory
0.354
1.131
1.368


protein; MEM43 antigen; Membrane attack complex inhibition factor; Membrane inhibitor of





reactive lysis; Protectin





Aldehyde dehydrogenase family 1 member A1; Aldehyde dehydrogenase, cytosolic; ALDH-
0.348
0.678
1.002


E1; ALHDII; Retinal dehydrogenase 1





Glycine hydroxymethyltransferase; Serine hydroxymethyltransferase, mitochondrial; Serine
0.345
0.677
0.978


methylase; cDNA FLJ58585, highly similar to Serine hydroxymethyltransferase,





mitochondrial (EC 2.1.2.1); Serine hydroxymethyltransferase 2 (Mitochondrial), isoform





CRA_h





Macropain zeta chain; Multicatalytic endopeptidase complex zeta chain; Proteasome
0.339
0.954
0.900


subunit alpha type-5; Proteasome zeta chain; cDNA FLJ52182, highly similar to





Proteasome subunit alpha type 5 (EC 3.4.25.1); Proteasome (Prosome, macropain)





subunit, alpha type, 5, isoform CRA_c





Methionine--tRNA ligase; Methionyl-tRNA synthetase, cytoplasmic; Putative
0.325
0.537
0.977


uncharacterized protein MARS; cDNA FLJ16674 fis, clone THYMU3008136, highly similar





to Methionyl-tRNA synthetase (EC 6.1.1.10)





Importin-9; Ran-binding protein 9
0.320
0.530
0.801


Pre-mRNA-splicing factor SRP75; Splicing factor, arginine/serine-rich 4; SRP001LB
0.310
1.096
1.336


Anterior gradient protein 2 homolog; HPC8; Secreted cement gland protein XAG-2
0.310
1.524
1.365


homolog; Putative uncharacterized protein AGR2





Coagulation factor XIII A chain; Protein-glutamine gamma-glutamyltransferase A
0.306
1.379
1.397


chain; Transglutaminase A chain





DNA replication licensing factor MCM2; Minichromosome maintenance protein 2
0.303
0.555
0.981


homolog; Nuclear protein BM28





Phosphate carrier protein, mitochondrial; Phosphate transport protein; Solute carrier family
0.299
0.569
0.911


25 member 3





CCT-beta; T-complex protein 1 subunit beta
0.291
0.540
0.980


Protein ftsJ homolog 3; Putative rRNA methyltransferase 3; rRNA (uridine-2-O-)-
0.286
0.995
1.042


methyltransferase 3





High mobility group-like nuclear protein 2 homolog 1; NHP2-like protein 1; OTK27; SNU13
0.280
0.676
0.992


homolog; U4/U6.U5 tri-snRNP 15.5 kDa protein; NHP2 non-histone chromosome protein 2-





like 1 (S. cerevisiae)





CCT-gamma; hTRiC5; T-complex protein 1 subunit gamma
0.278
0.521
0.867


Nuclear matrix protein 200; Pre-mRNA-processing factor 19; PRP19/PSO4
0.276
0.291
0.352


homolog; Senescence evasion factor





Protein mago nashi homolog; cDNA FLJ55283, moderately similar to Protein mago nashi
0.272
1.125
1.047


homolog; Mago-nashi homolog, proliferation-associated (Drosophila); Mago-nashi





homolog, proliferation-associated (Drosophila), isoform CRA_a





Mannose-6-phosphate isomerase; Phosphohexomutase; Phosphomannose
0.261
0.847
0.931


isomerase; Mannose phosphate isomerase isoform





Carnitine/acylcarnitine translocase; Mitochondrial carnitine/acylcarnitine carrier
0.254
1.064
1.024


protein; Solute carrier family 25 member 20; cDNA FLJ53016, highly similar to





Mitochondrial carnitine/acylcarnitine carrier protein





Caspase-1; Caspase-1 subunit p10; Caspase-1 subunit p20; Interleukin-1 beta
0.252
1.158
1.068


convertase; Interleukin-1 beta-converting enzyme; p45; cDNA FLJ59442, highly similar to





Caspase-1 (EC 3.4.22.36)





40S ribosomal protein S4, X isoform; SCR10; Single copy abundant mRNA protein
0.252
0.522
0.958


Dipeptidyl aminopeptidase II; Dipeptidyl peptidase 2; Dipeptidyl peptidase 7; Dipeptidyl
0.248
1.042
0.922


peptidase II; Quiescent cell proline dipeptidase





Protein mago nashi homolog 2; Putative uncharacterized protein MAGOHB
0.240
1.337
1.259


Brain-type aldolase; Fructose-bisphosphate aldolase C; Fructose-bisphosphate
0.236
0.954
0.892


aldolase; Putative uncharacterized protein ALDOC





Ras-related protein Rab-1A; YPT1-related protein; cDNA FLJ57768, highly similar to Ras-
0.230
0.572
0.900


related protein Rab-1A





Brush border myosin I; Myosin I heavy chain; Myosin-Ia
0.222
1.238
1.241


CDC21 homolog; DNA replication licensing factor MCM4; P1-CDC21
0.220
0.172
0.646


28 kDa heat shock protein; Estrogen-regulated 24 kDa protein; Heat shock 27 kDa
0.208
0.621
0.941


protein; Heat shock protein beta-1; Stress-responsive protein 27; cDNA FLJ52243, highly





similar to Heat-shock protein beta-1





ATP-dependent RNA helicase DDX19A; DDX19-like protein; DEAD box protein 19A; ATP-
0.207
0.443
0.447


dependent RNA helicase DDX19B; DEAD box protein 19B; DEAD box RNA helicase





DEAD5; cDNA FLJ52463, highly similar to ATP-dependent RNA helicase DDX19A (EC 3.6.1.—)





Double-stranded RNA-binding protein 76; Interleukin enhancer-binding factor 3; M-phase
0.202
0.696
0.934


phosphoprotein 4; Nuclear factor associated with dsRNA; Nuclear factor of activated T-





cells 90 kDa; Translational control protein 80; Putative uncharacterized protein ILF3; cDNA





FLJ58801, highly similar to Interleukin enhancer-binding factor 3





PHD finger protein 6; PHD-like zinc finger protein; cDNA FLJ60207, highly similar to PHD
0.201
1.197
1.485


finger protein 6; PHD finger protein 6, isoform CRA_d





Glycosyltransferase 25 family member 1; Hydroxylysine galactosyltransferase
0.197
0.183
1.063


1; Procollagen galactosyltransferase 1





D-fructose-6-phosphate amidotransferase 2; Glucosamine--fructose-6-phosphate
0.197
1.720
1.634


aminotransferase [isomerizing] 2; Glutamine: fructose 6 phosphate amidotransferase





2; Hexosephosphate aminotransferase 2





CCT-delta; Stimulator of TAR RNA-binding; T-complex protein 1 subunit delta
0.189
0.562
0.972


5-3 exoribonuclease 2; DHM1-like protein; cDNA FLJ55645, highly similar to 5-3
0.187
0.495
0.838


exoribonuclease 2 (EC 3.1.11.—)





High mobility group protein 2a; High mobility group protein 4; High mobility group protein
0.182
0.609
1.217


B3





CCT-theta; Renal carcinoma antigen NY-REN-15; T-complex protein 1 subunit theta; cDNA
0.181
0.581
0.962


FLJ53379, highly similar to T-complex protein 1 subunit theta; cDNA FLJ59382, highly





similar to T-complex protein 1 subunit theta





CD63 antigen; Granulophysin; Lysosomal-associated membrane protein 3; Melanoma-
0.180
0.953
0.884


associated antigen ME491; Ocular melanoma-associated antigen; OMA81H; Tetraspanin-





30; Putative uncharacterized protein CD63; Lysosome-associated membrane protein-3





variant





Sideroflexin-1; Tricarboxylate carrier protein
0.178
0.600
0.965


Phosphopantothenate--cysteine ligase; Phosphopantothenoylcysteine synthetase
0.178
0.853
1.016


CCT-alpha; T-complex protein 1 subunit alpha
0.175
0.864
0.990


Aldehyde dehydrogenase family 18 member A1; Delta-1-pyrroline-5-carboxylate
0.174
0.757
0.868


synthase; Gamma-glutamyl kinase; Gamma-glutamyl phosphate reductase; Glutamate 5-





kinase; Glutamate-5-semialdehyde dehydrogenase; Glutamyl-gamma-semialdehyde





dehydrogenase





Alpha-II spectrin; Fodrin alpha chain; Spectrin alpha chain, brain; Spectrin, non-erythroid
0.173
0.562
0.960


alpha chain; Putative uncharacterized protein SPTAN1; cDNA FLJ59116, highly similar to





Spectrin alpha chain, brain





Delta(3,5)-Delta(2,4)-dienoyl-CoA isomerase, mitochondrial
0.167
0.512
0.972


Farnesyl-diphosphate farnesyltransferase; FPP: FPP farnesyltransferase; Squalene
0.167
0.280
0.600


synthase; cDNA FLJ50548, highly similar to Squalene synthetase (EC 2.5.1.21); cDNA





FLJ50447, highly similar to Squalene synthetase (EC 2.5.1.21); cDNA, FLJ78892, highly





similar to Squalene synthetase (EC 2.5.1.21); cDNA, FLJ79250, highly similar to Squalene





synthetase (EC 2.5.1.21); cDNA, FLJ79430, highly similar to Squalene synthetase (EC





2.5.1.21); cDNA FLJ50660, highly similar to Squalene synthetase (EC 2.5.1.21); cDNA,





FLJ79433, highly similar to Squalene synthetase (EC 2.5.1.21); cDNA FLJ33164 fis, clone





UTERU2000542, highly similar to Squalene synthetase (EC 2.5.1.21)





DEAH box protein 30; Putative ATP-dependent RNA helicase DHX30
0.162
0.794
1.002


Bifunctional coenzyme A synthase; Dephospho-CoA kinase; Dephospho-CoA
0.162
1.139
1.115


pyrophosphorylase; Dephosphocoenzyme A kinase; NBP; Pantetheine-phosphate





adenylyltransferase; Phosphopantetheine adenylyltransferase; POV-2





DNA-dependent protein kinase catalytic subunit; DNPK1; p460
0.161
0.579
0.967


CDC46 homolog; DNA replication licensing factor MCM5; P1-CDC46; MCM5
0.159
0.349
0.925


minichromosome maintenance deficient 5, cell division cycle 46 (S. cerevisiae), isoform





CRA_c; Minichromosome maintenance complex component 5





Protein A1S9; Ubiquitin-activating enzyme E1; Ubiquitin-like modifier-activating enzyme
0.156
0.683
0.940


1; cDNA FLJ54582, highly similar to Ubiquitin-activating enzyme E1





CNDP dipeptidase 2; Cytosolic non-specific dipeptidase; Glutamate carboxypeptidase-like
0.155
1.122
0.999


protein 1; Peptidase A





Ras-related protein Rab-5B; cDNA FLJ60627, highly similar to Ras-related protein Rab-
0.152
0.305
0.369


5B; RAB5B, member RAS oncogene family, isoform CRA_b





358 kDa nucleoporin; E3 SUMO-protein ligase RanBP2; Nuclear pore complex protein
0.152
0.806
0.988


Nup358; Nucleoporin Nup358; p270; Ran-binding protein 2





Paraspeckle protein 2; RNA-binding motif protein 14; RNA-binding protein 14; RRM-
0.142
0.762
0.779


containing coactivator activator/modulator; Synaptotagmin-interacting protein





11-zinc finger protein; CCCTC-binding factor; CTCFL paralog; Transcriptional repressor
0.131
0.524
1.286


CTCF; Putative uncharacterized protein CTCF





6-phosphofructokinase, muscle type; Phosphofructo-1-kinase isozyme
0.124
0.812
1.019


A; Phosphofructokinase 1; Phosphohexokinase





Cyclin-A/CDK2-associated protein p19; Organ of Corti protein 2; Organ of Corti protein
0.123
0.644
0.779


II; p19A; p19skp1; RNA polymerase II elongation factor-like protein; SIII; S-phase kinase-





associated protein 1; Transcription elongation factor B





DRG family-regulatory protein 1; Likely ortholog of mouse immediate early response
0.121
0.869
0.890


erythropoietin 4; Zinc finger CCCH domain-containing protein 15





39S ribosomal protein L53, mitochondrial
0.119
0.960
0.919


Met-induced mitochondrial protein; Mitochondrial carrier homolog 2
0.116
0.476
0.951


Protein H105e3; Sterol-4-alpha-carboxylate 3-dehydrogenase, decarboxylating; Putative
0.105
1.222
1.087


uncharacterized protein NSDHL





Bw-45; HLA class I histocompatibility antigen, B-45 alpha chain; MHC class I antigen B*45
0.095
0.796
0.733


Hsc70-interacting protein; Progesterone receptor-associated p48 protein; Protein
0.095
0.714
0.961


FAM10A1; Putative tumor suppressor ST13; Renal carcinoma antigen NY-REN-





33; Suppression of tumorigenicity 13 protein; ST13 protein; Putative protein





FAM10A5; Putative protein FAM10A4





BRCA1-A complex subunit MERIT40; Mediator of RAP80 interactions and targeting
0.091
0.275
0.504


subunit of 40 kDa; New component of the BRCA1-A complex





Ezrin-radixin-moesin-binding phosphoprotein 50; Na(+)/H(+) exchange regulatory cofactor
0.088
0.557
0.968


NHE-RF1; Regulatory cofactor of Na(+)/H(+) exchanger; Sodium-hydrogen exchanger





regulatory factor 1; Solute carrier family 9 isoform A3 regulatory factor 1





Valyl-tRNA synthetase; Protein G7a; Valine--tRNA ligase
0.084
0.710
0.910


HCV F-transactivated protein 2; Up-regulated during skeletal muscle growth protein 5
0.083
0.468
0.994


Adenylate cyclase-stimulating G alpha protein; Extra large alphas protein; Guanine
0.073
1.209
1.091


nucleotide-binding protein G(s) subunit alpha isoforms XLas; Putative uncharacterized





protein GNAS; Guanine nucleotide-binding protein G(s) subunit alpha isoforms short





CCT-eta; HIV-1 Nef-interacting protein; T-complex protein 1 subunit eta; Putative
0.071
0.574
0.934


uncharacterized protein CCT7; cDNA FLJ59454, highly similar to T-complex protein 1





subunit eta; Chaperonin containing TCP1, subunit 7 (Eta), isoform CRA_a





Collagen alpha-1(VI) chain; cDNA FLJ61362, highly similar to Collagen alpha-1(VI) chain
0.068
1.608
1.496


ARF-binding protein 1; E3 ubiquitin-protein ligase HUWE1; HECT, UBA and WWE domain-
0.066
0.661
0.963


containing protein 1; Homologous to E6AP carboxyl terminus homologous protein 9; Large





structure of UREB1; Mcl-1 ubiquitin ligase E3; Upstream regulatory element-binding protein 1





Alpha-1-acid glycoprotein 2; Orosomucoid-2
0.060
1.836
1.804


Protein NipSnap homolog 3A; Protein NipSnap homolog 4; Target for Salmonella secreted
0.060
0.931
1.090


protein C





Antioxidant protein 1; HBC189; Peroxiredoxin III; Peroxiredoxin-3; Protein MER5
0.058
0.578
0.972


homolog; Thioredoxin-dependent peroxide reductase, mitochondrial





Aspartate carbamoyltransferase; CAD protein; Dihydroorotase; Glutamine-dependent
0.049
0.820
0.923


carbamoyl-phosphate synthase





Nuclear mitotic apparatus protein 1; SP-H antigen
0.045
0.486
0.961


50 kDa nucleoporin; Nuclear pore complex protein Nup50; Nuclear pore-associated protein
0.043
0.036
0.745


60 kDa-like; Nucleoporin Nup50





[Acyl-carrier-protein] S-acetyltransferase; [Acyl-carrier-protein] S-malonyltransferase; 3-
0.035
0.582
0.581


hydroxypalmitoyl-[acyl-carrier-protein] dehydratase; 3-oxoacyl-[acyl-carrier-protein]





reductase; 3-oxoacyl-[acyl-carrier-protein] synthase; Enoyl-[acyl-carrier-protein]





reductase; Fatty acid synthase; Oleoyl-[acyl-carrier-protein] hydrolase





DNA mismatch repair protein Msh6; G/T mismatch-binding protein; MutS-alpha 160 kDa
0.029
0.674
0.714


subunit; cDNA FLJ55677, highly similar to DNA mismatch repair protein MSH6





Heterogeneous nuclear ribonucleoprotein H; Heterogeneous nuclear ribonucleoprotein H,
0.025
0.583
0.972


N-terminally processed





Protein-tyrosine phosphatase 1D; Protein-tyrosine phosphatase 2C; SH-PTP2; SH-
0.018
0.430
0.913


PTP3; Tyrosine-protein phosphatase non-receptor type 11





ATP-dependent RNA helicase A; DEAH box protein 9; Nuclear DNA helicase II
0.006
0.562
0.967


Cysteine dioxygenase type 1; Cysteine dioxygenase type I
0.005
1.364
1.209









It has been observed that certain mitochondrial proteins are differentially expressed and their levels can be associated with the presence or absence of UC or CD disease. For example, sulfur dioxygenase (ETHE1), thiosulfate sulfur transferase (TST), cytochrome c oxidase subunit IV, sulfide dehydrogenase genes (SQR) and complexes III and IV of mithochondrial respiratory chain obtained from a gut mucus sample of a human subject can be indicative of the presence of UC or CD or IBD in the subject. However it will be appreciated that any other protein(s) listed in table 4 or 5 alone or in combination that is or are differentially expressed can also be used to assess the presence/absence/severity of UC or CD disease.


Expression of certain cytokines above normal levels can also be used to detect the presence of A. parvulum. For example the presence of A. parvulum is correlated with expression (or overexpression) of Cxcl1, II17a, II12 and II1β. Therefore there is provided an assay for identifying the likelihood of an individual of having UC or CD or IBD by measuring a relative abundance of A. parvulum by measuring the expression of Cxcl1, II17a, II12 or II1β. This correlation can also be used to provide a method of diagnostic that comprises collecting samples to measure one or more cytokines, determining the presence of A. parvulum based on the cytokine(s) measurement and establishing a diagnosis.


Table 5 List of all differentially expressed mitochondrial proteins and their variable importance in projection scores (VIP) derived from the calculated PLS-DA model.












TABLE 5






Comp. 1
Comp. 2
Comp. 3


Variable
VIP
VIP
VIP


















Complex I-PDSW; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit
1.834
1.448
1.178


10; NADH-ubiquinone oxidoreductase PDSW subunit; NADH dehydrogenase (Ubiquinone)





1 beta subcomplex, 10, 22 kDa, isoform CRA_a; NDUFB10 protein





Complex I-75 kD; NADH-ubiquinone oxidoreductase 75 kDa subunit, mitochondrial; cDNA
1.788
1.323
1.100


FLJ60586, highly similar to NADH-ubiquinone oxidoreductase 75 kDa subunit,





mitochondrial (EC 1.6.5.3)





Isovaleryl-CoA dehydrogenase, mitochondrial; cDNA FLJ16602 fis, clone TESTI4007816,
1.748
1.559
1.324


highly similar to Isovaleryl-CoA dehydrogenase, mitochondrial (EC 1.3.99.10); Isovaleryl





Coenzyme A dehydrogenase, isoform CRA_b





Complex I-39 kD; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9,
1.706
1.331
1.100


mitochondrial; NADH-ubiquinone oxidoreductase 39 kDa subunit





Cytochrome c oxidase polypeptide Vb; Cytochrome c oxidase subunit 5B, mitochondrial
1.666
1.237
1.006


Complex III subunit 5; Complex III subunit IX; Cytochrome b-c1 complex subunit
1.658
1.249
1.038


11; Cytochrome b-c1 complex subunit 5; Cytochrome b-c1 complex subunit Rieske,





mitochondrial; Rieske iron-sulfur protein; Ubiquinol-cytochrome c reductase 8 kDa





protein; Ubiquinol-cytochrome c reductase iron-sulfur subunit; Putative cytochrome b-c1





complex subunit Rieske-like protein 1





Complex III subunit 7; Complex III subunit VII; Cytochrome b-c1 complex subunit 7; QP-
1.625
1.220
1.007


C; Ubiquinol-cytochrome c reductase complex 14 kDa protein; cDNA FLJ52271, moderately





similar to Ubiquinol-cytochrome c reductase complex 14 kDa protein (EC 1.10.2.2)





Complex III subunit 2; Core protein II; Cytochrome b-c1 complex subunit 2,
1.551
1.167
1.207


mitochondrial; Ubiquinol-cytochrome-c reductase complex core protein 2





Angiotensin-binding protein; Microsomal endopeptidase; Mitochondrial oligopeptidase
1.547
1.997
1.627


M; Neurolysin, mitochondrial; Neurotensin endopeptidase





Rhodanese; Thiosulfate sulfurtransferase
1.504
1.169
0.954


Complex I-ASHI; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 8,
1.489
1.117
1.207


mitochondrial; NADH-ubiquinone oxidoreductase ASHI subunit; NADH dehydrogenase





(Ubiquinone) 1 beta subcomplex, 8, 19 kDa; NADH dehydrogenase (Ubiquinone) 1 beta





subcomplex, 8, 19 kDa, isoform CRA_a; cDNA FLJ52503, highly similar to NADH





dehydrogenase (ubiquinone) 1 beta subcomplex subunit 8, mitochondrial (EC 1.6.5.3) (EC





1.6.99.3) (NADH-ubiquinone oxidoreductase ASHI subunit) (Complex I-ASHI) (CI-ASHI)





Iron-sulfur subunit of complex II; Succinate dehydrogenase [ubiquinone] iron-sulfur subunit,
1.486
1.167
1.040


mitochondrial





Complex III subunit 1; Core protein I; Cytochrome b-c1 complex subunit 1,
1.474
1.124
0.922


mitochondrial; Ubiquinol-cytochrome-c reductase complex core protein 1





Complex I-B14.5a; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit
1.470
1.088
1.194


7; NADH-ubiquinone oxidoreductase subunit B14.5a





Complex I-23 kD; NADH dehydrogenase [ubiquinone] iron-sulfur protein 8,
1.443
1.178
0.982


mitochondrial; NADH-ubiquinone oxidoreductase 23 kDa subunit; TYKY subunit





Cytochrome c oxidase polypeptide IV; Cytochrome c oxidase subunit 4 isoform 1,
1.432
1.063
0.934


mitochondrial; Cytochrome c oxidase subunit IV isoform 1; COX4I1 protein





Cytochrome c oxidase polypeptide VIc; Cytochrome c oxidase subunit 6C
1.416
1.074
0.929


Glutathione S-transferase kappa 1; Glutathione S-transferase subunit 13; GST 13-13; GST
1.404
1.172
0.975


class-kappa; GSTK1-1





Cytochrome c oxidase polypeptide II; Cytochrome c oxidase subunit 2
1.375
1.120
0.943


GTP-specific succinyl-CoA synthetase subunit beta; Succinyl-CoA ligase [GDP-forming]
1.363
1.127
0.961


subunit beta, mitochondrial; Succinyl-CoA synthetase beta-G chain





250/210 kDa paraneoplastic pemphigus antigen; Desmoplakin
1.354
1.040
0.867


Complex I-51 kD; NADH dehydrogenase [ubiquinone] flavoprotein 1, mitochondrial; NADH
1.353
1.034
1.066


dehydrogenase flavoprotein 1; NADH-ubiquinone oxidoreductase 51 kDa subunit; cDNA





FLJ57949, highly similar to NADH-ubiquinone oxidoreductase 51 kDa subunit,





mitochondrial (EC 1.6.5.3); cDNA, FLJ79021, highly similar to NADH-ubiquinone





oxidoreductase 51 kDa subunit, mitochondrial (EC 1.6.5.3)





Alu corepressor 1; Antioxidant enzyme B166; Liver tissue 2D-page spot 71B; Peroxiredoxin
1.336
0.994
0.821


V; Peroxiredoxin-5, mitochondrial; Peroxisomal antioxidant enzyme; PLP; Thioredoxin





peroxidase PMP20; Thioredoxin reductase; TPx type VI; Putative uncharacterized protein





PRDX5





ICD-M; IDP; Isocitrate dehydrogenase [NADP], mitochondrial; NADP(+)-specific
1.320
1.210
1.078


ICDH; Oxalosuccinate decarboxylase





Flavoprotein subunit of complex II; Succinate dehydrogenase [ubiquinone] flavoprotein
1.292
1.057
1.032


subunit, mitochondrial





Brain-type aldolase; Fructose-bisphosphate aldolase C; Fructose-bisphosphate
1.289
1.115
0.906


aldolase; Putative uncharacterized protein ALDOC





Complex I-13 kD-A; NADH dehydrogenase [ubiquinone] iron-sulfur protein 6,
1.261
0.935
0.781


mitochondrial; NADH-ubiquinone oxidoreductase 13 kDa-A subunit





Ethylmalonic encephalopathy protein 1; Hepatoma subtracted clone one protein; Protein
1.252
0.951
0.851


ETHE1, mitochondrial





Complex I-B15; NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 4; NADH-
1.241
0.922
0.749


ubiquinone oxidoreductase B15 subunit; Putative uncharacterized protein NDUFB4





DEAH box protein 30; Putative ATP-dependent RNA helicase DHX30
1.213
1.450
1.220


Cytochrome c oxidase polypeptide VIIc; Cytochrome c oxidase subunit 7C, mitochondrial
1.211
0.897
0.751


Amine oxidase [flavin-containing] A; Monoamine oxidase type A; cDNA FLJ61220, highly
1.204
0.970
0.866


similar to Amine oxidase (flavin-containing) A (EC 1.4.3.4)





Carnitine O-palmitoyltransferase 2, mitochondrial; Carnitine palmitoyltransferase II
1.141
0.869
0.871


Amine oxidase [flavin-containing] B; Monoamine oxidase type B; cDNA FLJ51821, highly
1.066
1.289
1.107


similar to Amine oxidase (flavin-containing) B (EC 1.4.3.4); cDNA FLJ52418, highly similar





to Amine oxidase (flavin-containing) B (EC 1.4.3.4)





Glutaminase kidney isoform, mitochondrial; K-glutaminase; L-glutamine amidohydrolase
1.033
1.317
1.087


Acyl-coenzyme A thioesterase 13; Thioesterase superfamily member 2
1.006
1.393
1.190


Cathepsin D; Cathepsin D heavy chain; Cathepsin D light chain
1.004
0.784
0.652


78 kDa gastrin-binding protein; Long chain 3-hydroxyacyl-CoA dehydrogenase; Long-chain
0.999
0.742
0.836


enoyl-CoA hydratase; TP-alpha; Trifunctional enzyme subunit alpha, mitochondrial





Elongation factor Tu, mitochondrial; P43
0.991
0.754
0.929


Enoyl-CoA hydratase 1; Enoyl-CoA hydratase, mitochondrial; Short-chain enoyl-CoA
0.968
0.842
0.714


hydratase





Dihydrolipoamide dehydrogenase; Dihydrolipoyl dehydrogenase, mitochondrial; Glycine
0.956
0.716
0.728


cleavage system L protein; cDNA FLJ50515, highly similar to Dihydrolipoyl dehydrogenase,





mitochondrial (EC 1.8.1.4); Dihydrolipoyl dehydrogenase





Delta(3),delta(2)-enoyl-CoA isomerase; Diazepam-binding inhibitor-related protein
0.945
0.706
0.578


1; Dodecenoyl-CoA isomerase; DRS-1; Hepatocellular carcinoma-associated antigen





88; Peroxisomal 3,2-trans-enoyl-CoA isomerase; Renal carcinoma antigen NY-REN-





1; Putative uncharacterized protein PECI





Catalase
0.924
0.881
0.801


3-ketoacyl-CoA thiolase; Acetyl-CoA acyltransferase; Beta-ketothiolase; TP-
0.892
0.696
0.981


beta; Trifunctional enzyme subunit beta, mitochondrial; cDNA FLJ56214, highly similar to





Trifunctional enzyme subunit beta, mitochondrial; Putative uncharacterized protein HADHB





Aldehyde dehydrogenase family 6 member A1; Methylmalonate-semialdehyde
0.882
0.756
0.616


dehydrogenase [acylating], mitochondrial





Malic enzyme 2; NAD-dependent malic enzyme, mitochondrial
0.872
1.419
1.325


Outer mitochondrial membrane protein porin 2; Voltage-dependent anion-selective channel
0.871
0.649
0.538


protein 2; Voltage-dependent anion channel 2; cDNA FLJ60120, highly similar to Voltage-





dependent anion-selective channel protein 2; cDNA, FLJ78818, highly similar to Voltage-





dependent anion-selective channel protein 2





Aspartate aminotransferase, mitochondrial; Fatty acid-binding protein; Glutamate
0.869
0.668
0.605


oxaloacetate transaminase 2; Plasma membrane-associated fatty acid-binding





protein; Transaminase A





Aldehyde dehydrogenase 5; Aldehyde dehydrogenase family 1 member B1; Aldehyde
0.861
0.893
0.729


dehydrogenase X, mitochondrial; cDNA FLJ51238, highly similar to Aldehyde





dehydrogenase X, mitochondrial (EC 1.2.1.3)





Protein NipSnap homolog 1
0.849
0.635
0.541


Calcium-binding mitochondrial carrier protein Aralar2; Citrin; Mitochondrial aspartate
0.840
0.650
0.691


glutamate carrier 2; Solute carrier family 25 member 13





Elongation factor Ts, mitochondrial; Elongation factor Ts
0.822
0.893
0.726


Cytosol aminopeptidase; Leucine aminopeptidase 3; Leucyl aminopeptidase; Peptidase
0.797
0.590
0.848


S; Proline aminopeptidase; Prolyl aminopeptidase





Outer mitochondrial membrane protein porin 1; Plasmalemmal porin; Porin 31HL; Porin
0.766
0.602
0.533


31HM; Voltage-dependent anion-selective channel protein 1





Superoxide dismutase [Mn], mitochondrial; Superoxide dismutase
0.723
0.581
0.916


Sideroflexin-1; Tricarboxylate carrier protein
0.721
0.705
1.238


Aldehyde dehydrogenase family 18 member A1; Delta-1-pyrroline-5-carboxylate
0.716
0.559
1.164


synthase; Gamma-glutamyl kinase; Gamma-glutamyl phosphate reductase; Glutamate 5-





kinase; Glutamate-5-semialdehyde dehydrogenase; Glutamyl-gamma-semialdehyde





dehydrogenase





39S ribosomal protein L53, mitochondrial
0.705
0.812
0.703


Glycine hydroxymethyltransferase; Serine hydroxymethyltransferase, mitochondrial; Serine
0.687
0.690
1.139


methylase; cDNA FLJ58585, highly similar to Serine hydroxymethyltransferase,





mitochondrial (EC 2.1.2.1); Serine hydroxymethyltransferase 2 (Mitochondrial), isoform





CRA_h





Antioxidant enzyme AOE372; Peroxiredoxin IV; Peroxiredoxin-4; Thioredoxin peroxidase
0.651
1.017
1.015


AO372; Thioredoxin-dependent peroxide reductase A0372





Hematopoietic cell-specific LYN substrate 1; Hematopoietic lineage cell-specific
0.635
0.872
0.708


protein; LckBP1; p75





Acetoacetyl-CoA thiolase; Acetyl-CoA acetyltransferase, mitochondrial; T2
0.592
0.646
0.573


Complex I-B8; NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 2; NADH-
0.570
1.106
0.937


ubiquinone oxidoreductase B8 subunit





Heat shock-related 70 kDa protein 2; cDNA FLJ40505 fis, clone TESTI2045562, highly
0.551
1.027
0.850


similar to HEAT SHOCK-RELATED 70 kDa PROTEIN 2





Pyruvate carboxylase, mitochondrial; Pyruvic carboxylase; cDNA FLJ60715, highly similar to
0.542
0.477
0.394


Pyruvate carboxylase, mitochondrial (EC 6.4.1.1)





Hydroxysteroid dehydrogenase-like protein 2; cDNA FLJ61200, highly similar to Homo sapiens
0.509
0.403
0.419


hydroxysteroid dehydrogenase like 2 (HSDL2), mRNA





PDHE1-A type I; Pyruvate dehydrogenase E1 component subunit alpha, somatic form,
0.498
0.372
0.801


mitochondrial





Antioxidant protein 1; HBC189; Peroxiredoxin III; Peroxiredoxin-3; Protein MER5
0.490
0.707
1.120


homolog; Thioredoxin-dependent peroxide reductase, mitochondrial





3-hydroxybutyrate dehydrogenase type 2; Dehydrogenase/reductase SDR family member
0.476
0.807
0.883


6; Oxidoreductase UCPA; R-beta-hydroxybutyrate dehydrogenase





3-5 RNA exonuclease OLD35; PNPase old-35; Polynucleotide phosphorylase
0.470
1.236
1.035


1; Polynucleotide phosphorylase-like protein; Polyribonucleotide nucleotidyltransferase 1,





mitochondrial





3-ketoacyl-CoA thiolase, mitochondrial; Acetyl-CoA acyltransferase; Beta-
0.461
0.677
1.061


ketothiolase; Mitochondrial 3-oxoacyl-CoA thiolase; T1





Delta(3,5)-Delta(2,4)-dienoyl-CoA isomerase, mitochondrial
0.458
0.638
1.143


Carnitine/acylcarnitine translocase; Mitochondrial carnitine/acylcarnitine carrier
0.453
2.048
1.697


protein; Solute carrier family 25 member 20; cDNA FLJ53016, highly similar to Mitochondrial





carnitine/acylcarnitine carrier protein





Protein SCO2 homolog, mitochondrial
0.439
1.323
1.107


HCNPpp; Hippocampal cholinergic neurostimulating peptide; Neuropolypeptide
0.411
0.517
0.981


h3; Phosphatidylethanolamine-binding protein 1; Prostatic-binding protein; Raf kinase





inhibitor protein; cDNA FLJ51535, highly similar to Phosphatidylethanolamine-binding





protein 1





Collapsin response mediator protein 2; Dihydropyrimidinase-related protein 2; N2A3;
0.403
0.309
1.150


Unc-33-like phosphoprotein 2





28S ribosomal protein S9, mitochondrial
0.356
2.072
1.692


Met-induced mitochondrial protein; Mitochondrial carrier homolog 2
0.353
0.461
1.075


Phosphate carrier protein, mitochondrial; Phosphate transport protein; Solute carrier family
0.353
0.514
1.140


25 member 3





Acyl-CoA-binding domain-containing protein 3; Golgi complex-associated protein 1; Golgi
0.351
1.694
1.801


phosphoprotein 1; Golgi resident protein GCP60; PBR- and PKA-associated protein





7; Peripheral benzodiazepine receptor-associated protein PAP7





HCV F-transactivated protein 2; Up-regulated during skeletal muscle growth protein 5
0.344
0.707
1.117


Protein-tyrosine phosphatase 1D; Protein-tyrosine phosphatase 2C; SH-PTP2; SH-
0.303
0.699
1.024


PTP3; Tyrosine-protein phosphatase non-receptor type 11





ATP synthase subunit a; F-ATPase protein 6
0.260
0.193
0.279


Complex III subunit 3; Complex III subunit III; Cytochrome b; Cytochrome b-c1 complex
0.235
0.489
0.692


subunit 3; Ubiquinol-cytochrome-c reductase complex cytochrome b subunit





Clathrin heavy chain 1; Clathrin heavy chain on chromosome 17
0.226
0.477
0.730


Glutamate dehydrogenase 1, mitochondrial; cDNA FLJ55203, highly similar to Glutamate
0.216
0.315
0.598


dehydrogenase 1, mitochondrial (EC 1.4.1.3); cDNA FLJ16138 fis, clone BRALZ2017531,





highly similar to Glutamate dehydrogenase 1, mitochondrial (EC 1.4.1.3); Glutamate





dehydrogenase 1, isoform CRA_a; Glutamate dehydrogenase 2, mitochondrial





Succinyl-CoA ligase [GDP-forming] subunit alpha, mitochondrial; Succinyl-CoA synthetase
0.189
0.452
1.125


subunit alpha





Complex I-15 kDa; NADH dehydrogenase [ubiquinone] iron-sulfur protein 5; NADH-
0.163
1.344
1.142


ubiquinone oxidoreductase 15 kDa subunit





Complex I-49 kD; NADH dehydrogenase [ubiquinone] iron-sulfur protein 2,
0.137
0.301
1.161


mitochondrial; NADH-ubiquinone oxidoreductase 49 kDa subunit; cDNA, FLJ78876, highly





similar to NADH-ubiquinone oxidoreductase 49 kDa subunit, mitochondrial (EC 1.6.5.3)





3-hydroxyisobutyryl-CoA hydrolase, mitochondrial; 3-hydroxyisobutyryl-coenzyme A
0.106
1.378
1.513


hydrolase





Alcohol dehydrogenase 5; Alcohol dehydrogenase class chi chain; Alcohol dehydrogenase
0.093
0.471
1.107


class-3; Alcohol dehydrogenase class-III; Glutathione-dependent formaldehyde





dehydrogenase; S-(hydroxymethyl)glutathione dehydrogenase





Electron transfer flavoprotein subunit beta
0.091
0.479
1.047


130 kDa leucine-rich protein; GP130; Leucine-rich PPR motif-containing protein,
0.057
0.480
1.135


mitochondrial





ATP-specific succinyl-CoA synthetase subunit beta; Renal carcinoma antigen NY-REN-
0.024
0.865
0.725


39; Succinyl-CoA ligase [ADP-forming] subunit beta, mitochondrial; Succinyl-CoA





synthetase beta-A chain; Succinate-CoA ligase, ADP-forming, beta subunit





Citrate synthase, mitochondrial; Citrate synthase
0.014
0.627
1.020









In yet another embodiment of the invention there is provided an assay that allows the measurement of the gut microbiota composition and the meta-proteome from a same sample. More specifically the assay comprises the collection of mucus at the luminal interface of the gut during endoscopy by flushing a physiological solution, such as sterile saline, onto the mucosa to remove the strongly adherent mucus layer overlaying the intestinal mucosal epithelial cells thereby sampling the microbial community and host and bacterial proteins embedded within the mucus layer. Aspirates are then collected directly through the colonoscope and the samples are preferably immediately put on ice right in the endoscopy suite. The sample can then be analyzed at the point of care or transferred to a laboratory. Bacteria and proteins can then be identified and/or measured as described above. This method advantageously permits the establishment of a protein and bacterial profile in the same patient at a pre-determined time point.


The establishment of the presence of disease using bacterial taxa can be used to determine a course of treatment in a patient. Treatment is normally based on accepted diseases indexes. The methods and assays provided by the invention can complement or replace such disease indexes to provide more accurate diagnosis and thereby permit more efficacious treatments.


It will be appreciated that the above described assays for identifying and measuring gut proteins and bacteria can be performed as a function of time thereby allowing an assessment of the progression of the disease as well as of the efficacy of a treatment. Staging of IBD (and CD and UC) is particularly useful for choosing the appropriate treatment to be delivered. For example, treatment regimen may advantageously be adjusted taking in consideration the levels of H2S producing bacteria, which as described above, are more elevated as the severity of the disease increases. Thus regimens that are more aggressive towards mitigating the effects of the H2S producing bacteria can be timely administered to optimize the therapeutic dose. Treatment optimization using the information on the presence/stage of IBD, UC and CD provided by the measuring of bacteria and protein as described above, can be applied to known therapeutic agents such as but not limited to aminosalycylates, immunomodulators, anti-integrins, anti-cytokines, enteral feed programs, steroids, corticosteroids, antibiotics, anti-TNFα, bismuth and the like. In particular, as further described below, bismuth can be used effectively as treatment when A. parvulum is detected in a patient and or assessed to be above certain critical abundance levels.


In table 6 taxa that vary significantly in abundance in II10−/− mice in response to A. parvulum colonization and/or bismuth administration are listed. As can be seen from the table bismuth treatment may be indicated or beneficial when the relative abundance of taxa other than A. parvulum are within levels indicative of disease.

















TABLE 6









Std.






Variable
Minimum
Maximum
Mean
dev.
p|Atopo
p|AtopoBis
p|Bis
p|SPF























PHYLUM










Basidiomycota|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

0.000



Basidiomycota|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
1.000

0.000



Basidiomycota|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

0.000



Basidiomycota|SPF
0.000
80.000
25.429
32.103

0.000


0.000


0.000

1


Firmicutes|Atopo
57699.000
101958.000
79769.875
13542.546
1
0.869

0.002

0.194


Firmicutes|AtopoBis
49410.000
184098.000
104984.625
60115.166
0.869
1

0.003

0.255


Firmicutes|Bis
105913.000
199488.000
175443.625
40788.918

0.002


0.003

1
0.084


Firmicutes|SPF
64133.000
198934.000
121645.143
57520.397
0.194
0.255
0.084
1


Cyanobacteria|Atopo
1.000
136.000
56.125
53.731
1
0.023

0.007


0.006



Cyanobacteria|AtopoBis
63.000
553.000
216.125
157.070
0.023
1
0.680
0.563


Cyanobacteria|Bis
52.000
669.000
261.000
194.269

0.007

0.680
1
0.857


Cyanobacteria|SPF
30.000
872.000
370.000
326.109

0.006

0.563
0.857
1


Fusobacteria|Atopo
0.000
20.000
3.500
6.845
1

0.008

0.089

0.007



Fusobacteria|AtopoBis
1.000
34434.000
6670.125
12103.868

0.008

1
0.341
0.889


Fusobacteria|Bis
0.000
3175.000
445.125
1109.012
0.089
0.341
1
0.289


Fusobacteria|SPF
5.000
96.000
38.571
31.921

0.007

0.889
0.289
1


Bacteroidetes|Atopo
86976.000
133781.000
108741.375
14150.040
1
0.216

<0.0001

0.011


Bacteroidetes|AtopoBis
232.000
117882.000
64729.375
53917.801
0.216
1

0.004

0.175


Bacteroidetes|Bis
98.000
519.000
209.500
134.340

<0.0001


0.004

1
0.151


Bacteroidetes|SPF
71.000
107705.000
28633.857
48179.254
0.011
0.175
0.151
1


CLASS


Fusobacteria (class)|Atopo
0.000
20.000
3.500
6.845
1

0.008

0.089

0.007



Fusobacteria (class)|
1.000
34434.000
6670.125
12103.868

0.008

1
0.341
0.889


AtopoBis


Fusobacteria (class)|Bis
0.000
3175.000
445.125
1109.012
0.089
0.341
1
0.289


Fusobacteria (class)|SPF
5.000
96.000
38.571
31.921

0.007

0.889
0.289
1


Erysipelotrichi|Atopo
998.000
7663.000
4385.500
2215.367
1
0.409
0.011

0.004



Erysipelotrichi|AtopoBis
928.000
8147.000
3320.375
2780.457
0.409
1
0.088
0.037


Erysipelotrichi|Bis
2.000
10136.000
1779.000
3443.700
0.011
0.088
1
0.660


Erysipelotrichi|SPF
6.000
3618.000
861.857
1483.609

0.004

0.037
0.660
1


Negativicutes|Atopo
1.000
2446.000
783.625
939.756
1
0.741
0.078
0.198


Negativicutes|AtopoBis
5.000
32.000
16.375
8.568
0.741
1
0.036
0.333


Negativicutes|Bis
0.000
10.000
3.625
3.335
0.078
0.036
1

0.003



Negativicutes|SPF
1.000
7076.000
2222.286
2915.263
0.198
0.333

0.003

1


Clostridia|Atopo
49447.000
95800.000
72002.750
15417.245
1
0.826

0.002

0.111


Clostridia|AtopoBis
41041.000
155491.000
88416.125
47521.587
0.826
1

0.005

0.167


Clostridia|Bis
95673.000
193932.000
161745.625
37810.173

0.002


0.005

1
0.184


Clostridia|SPF
54427.000
194395.000
116338.286
57644.557
0.111
0.167
0.184
1


Agaricomycetes|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

0.000



Agaricomycetes|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
1.000

0.000



Agaricomycetes|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

0.000



Agaricomycetes|SPF
0.000
80.000
25.429
32.103

0.000


0.000


0.000

1


Bacteroidia|Atopo
86976.000
133781.000
108741.375
14150.040
1
0.216

<0.0001

0.011


Bacteroidia|AtopoBis
232.000
117882.000
64729.375
53917.801
0.216
1

0.004

0.175


Bacteroidia|Bis
96.000
519.000
209.250
134.578

<0.0001


0.004

1
0.151


Bacteroidia|SPF
71.000
107705.000
28633.857
48179.254
0.011
0.175
0.151
1


Betaproteobacteria|Atopo
0.000
295.000
44.375
101.677
1
0.067
0.837

0.001



Betaproteobacteria|
11.000
61.000
30.375
15.638
0.067
1
0.105
0.102


AtopoBis


Betaproteobacteria|Bis
5.000
27.000
13.625
8.297
0.837
0.105
1

0.001



Betaproteobacteria|SPF
38.000
133.000
62.143
33.810

0.001

0.102

0.001

1


ORDER


Clostridiales|Atopo
49447.000
95800.000
72002.750
15417.245
1
0.826

0.002

0.111


Clostridiales|AtopoBis
41041.000
155488.000
88397.875
47505.759
0.826
1

0.005

0.167


Clostridiales|Bis
95673.000
193932.000
161745.500
37810.379

0.002


0.005

1
0.84


Clostridiales|SPF
54427.000
194394.000
116338.000
57644.323
0.111
0.167
0.184
1


Alteromonadales|Atopo
0.000
1.000
0.125
0.354
1
0.724
0.282

0.000



Alteromonadales|AtopoBis
0.000
1.000
0.250
0.463
0.724
1
0.470

0.001



Alteromonadales|Bis
0.000
126.000
24.875
47.975
0.282
0.470
1

0.007



Alteromonadales|SPF
1.000
915.000
264.143
345.489

0.000


0.001


0.007

1


Bacteroidales|Atopo
86976.000
133781.000
108741.375
14150.040
1
0.216

<0.0001

0.011


Bacteroidales|AtopoBis
232.000
117882.000
64729.375
53917.801
0.216
1

0.004

0.175


Bacteroidales|Bis
96.000
519.000
209.250
134.578

<0.0001


0.004

1
0.151


Bacteroidales|SPF
71.000
107705.000
28633.857
48179.254
0.011
0.175
0.151
1


Oceanospirillales|Atopo
0.000
5.000
2.000
1.852
1
0.393
0.576

0.004



Oceanospirillales|AtopoBis
0.000
74.000
23.125
27.910
0.393
1
0.158
0.037


Oceanospirillales|Bis
0.000
56.000
11.250
21.645
0.576
0.158
1

0.001



Oceanospirillales|SPF
3.000
4829.000
1391.571
1770.378

0.004

0.037

0.001

1


Agaricales|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

0.000



Agaricales|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
1.000

0.000



Agaricales|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

0.000



Agaricales|SPF
0.000
80.000
25.429
32.103

0.000


0.000


0.000

1


Actinomycetales|Atopo
0.000
1.000
0.500
0.535
1
0.446
0.796

0.002



Actinomycetales|AtopoBis
0.000
4.000
1.250
1.488
0.446
1
0.615
0.020


Actinomycetales|Bis
0.000
11.000
1.875
3.834
0.796
0.615
1

0.005



Actinomycetales|SPF
1.000
61.000
12.286
21.593

0.002

0.020

0.005

1


Erysipelotrichales|Atopo
998.000
7663.000
4385.500
2215.367
1
0.409
0.011

0.004



Erysipelotrichales|AtopoBis
928.000
8147.000
3320.375
2780.457
0.409
1
0.088
0.037


Erysipelotrichales|Bis
2.000
10136.000
1779.000
3443.700
0.011
0.088
1
0.660


Erysipelotrichales|SPF
6.000
3618.000
861.857
1483.609

0.004

0.037
0.660
1


Neisseriales|Atopo
0.000
0.000
0.000
0.000
1

0.004

0.585
0.441


Neisseriales|AtopoBis
0.000
2.000
0.750
0.707

0.004

1
0.022
0.048


Neisseriales|Bis
0.000
1.000
0.125
0.354
0.585
0.022
1
0.809


Neisseriales|SPF
0.000
8.000
1.143
3.024
0.441
0.048
0.809
1


Pasteurellales|Atopo
0.000
1.000
0.125
0.354
1
0.594
0.530

0.000



Pasteurellales|AtopoBis
0.000
4.000
0.750
1.488
0.594
1
0.925

0.001



Pasteurellales|Bis
0.000
14.000
2.000
4.899
0.530
0.925
1

0.001



Pasteurellales|SPF
2.000
56.000
20.143
21.836

0.000


0.001


0.001

1


Chromatiales|Atopo
0.000
0.000
0.000
0.000
1
1.000
0.560

0.000



Chromatiales|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
0.560

0.000



Chromatiales|Bis
0.000
2.000
0.250
0.707
0.560
0.560
1

0.001



Chromatiales|SPF
0.000
14.000
4.571
5.255

0.000


0.000


0.001

1


Vibrionales|Atopo
0.000
13.000
2.375
4.534
1
0.191
0.374

<0.0001



Vibrionales|AtopoBis
1.000
14.000
5.000
5.155
0.191
1
0.677

0.006



Vibrionales|Bis
0.000
23.000
6.125
8.097
0.374
0.677
1

0.002



Vibrionales|SPF
20.000
48532.000
15351.143
18959.865

<0.0001


0.006


0.002

1


Burkholderiales|Atopo
0.000
294.000
43.875
101.490
1
0.052
0.978

0.001



Burkholderiales|AtopoBis
10.000
58.000
27.750
15.351
0.052
1
0.056
0.137


Burkholderiales|Bis
5.000
19.000
11.125
5.357
0.978
0.056
1

0.001



Burkholderiales|SPF
32.000
133.000
58.571
35.156

0.001

0.137

0.001

1


Fusobacteriales|Atopo
0.000
20.000
3.500
6.845
1

0.008

0.089

0.007



Fusobacteriales|AtopoBis
1.000
34434.000
6670.125
12103.868

0.008

1
0.341
0.889


Fusobacteriales|Bis
0.000
3175.000
445.125
1109.012
0.089
0.341
1
0.289


Fusobacteriales|SPF
5.000
96.000
38.571
31.921

0.007

0.889
0.289
1


Bacillales|Atopo
73.000
348.000
155.000
97.999
1

0.000


0.007

0.115


Bacillales|AtopoBis
1326.000
30228.000
8866.125
9642.601

0.000

1
0.336
0.050


Bacillales|Bis
46.000
13607.000
5683.500
4962.745

0.007

0.336
1
0.304


Bacillales|SPF
163.000
7884.000
1604.857
2795.624
0.115
0.050
0.304
1


Selenomonadales|Atopo
1.000
2446.000
783.625
939.756
1
0.741
0.078
0.198


Selenomonadales|AtopoBis
5.000
32.000
16.375
8.568
0.741
1
0.036
0.333


Selenomonadales|Bis
0.000
10.000
3.625
3.335
0.078
0.036
1

0.003



Selenomonadales|SPF
1.000
7076.000
2222.286
2915.263
0.198
0.333

0.003

1


Lactobacillales|Atopo
295.000
7198.000
2442.750
2396.791
1
0.783
0.296
0.032


Lactobacillales|AtopoBis
187.000
18776.000
4365.625
6472.746
0.783
1
0.187
0.060


Lactobacillales|Bis
848.000
22750.000
6231.875
7499.344
0.296
0.187
1

0.002



Lactobacillales|SPF
66.000
1769.000
444.000
610.249
0.032
0.060

0.002

1


FAMILY


Staphylococcaceae|Atopo
0.000
9.000
1.375
3.114
1

0.002


0.004

0.010


Staphylococcaceae|
6.000
82.000
32.625
28.213

0.002

1
0.793
0.637


AtopoBis


Staphylococcaceae|Bis
0.000
132.000
40.000
45.854

0.004

0.793
1
0.827


Staphylococcaceae|SPF
2.000
838.000
191.000
333.003
0.010
0.637
0.827
1


Enterococcaceae|Atopo
11.000
38.000
20.750
9.588
1

0.000

0.014
0.040


Enterococcaceae|AtopoBis
122.000
1580.000
725.125
549.030

0.000

1
0.178
0.106


Enterococcaceae|Bis
0.000
1440.000
567.125
531.694
0.014
0.178
1
0.753


Enterococcaceae|SPF
39.000
1383.000
261.286
495.744
0.040
0.106
0.753
1


Eubacteriaceae|Atopo
3.000
43.000
8.875
13.861
1
0.061
0.518

0.001



Eubacteriaceae|AtopoBis
10.000
32.000
18.500
8.783
0.061
1
0.220
0.111


Eubacteriaceae|Bis
1.000
2387.000
313.625
838.348
0.518
0.220
1

0.005



Eubacteriaceae|SPF
13.000
1107.000
485.143
399.257

0.001

0.111

0.005

1


Ferrimonadaceae|Atopo
0.000
0.000
0.000
0.000
1
0.625
1.000

<0.0001



Ferrimonadaceae|AtopoBis
0.000
1.000
0.125
0.354
0.625
1
0.625

0.000



Ferrimonadaceae|Bis
0.000
0.000
0.000
0.000
1.000
0.625
1

<0.0001



Ferrimonadaceae|SPF
0.000
623.000
140.714
227.571

<0.0001


0.000


<0.0001

1


Alcanivoracaceae|Atopo
0.000
4.000
1.500
1.773
1
0.477
0.314
0.064


Alcanivoracaceae|AtopoBis
0.000
73.000
22.125
28.119
0.477
1
0.086
0.245


Alcanivoracaceae|Bis
0.000
4.000
0.500
1.414
0.314
0.086
1

0.005



Alcanivoracaceae|SPF
0.000
554.000
143.286
208.723
0.064
0.245

0.005

1


Bacteroidaceae|Atopo
86967.000
133771.000
108707.250
14136.218
1
0.226

<0.0001


0.007



Bacteroidaceae|AtopoBis
211.000
117745.000
64625.875
53942.157
0.226
1

0.005

0.125


Bacteroidaceae|Bis
74.000
515.000
171.750
143.702

<0.0001


0.005

1
0.239


Bacteroidaceae|SPF
57.000
107600.000
28388.714
48274.613

0.007

0.125
0.239
1


Oceanospirillaceae|Atopo
0.000
1.000
0.250
0.463
1
0.424
0.722

0.000



Oceanospirillaceae|
0.000
4.000
1.000
1.414
0.424
1
0.657

0.003



AtopoBis


Oceanospirillaceae|Bis
0.000
16.000
3.250
6.228
0.722
0.657
1

0.001



Oceanospirillaceae|SPF
3.000
3579.000
1056.714
1313.005

0.000


0.003


0.001

1


Halomonadaceae|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

0.000



Halomonadaceae|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
1.000

0.000



Halomonadaceae|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

0.000



Halomonadaceae|SPF
0.000
18.000
5.857
7.010

0.000


0.000


0.000

1


Lactobacillaceae|Atopo
263.000
7137.000
2406.750
2385.720
1

0.004


0.000


0.001



Lactobacillaceae|AtopoBis
3.000
27.000
12.375
9.226

0.004

1
0.440
0.586


Lactobacillaceae|Bis
0.000
1026.000
133.000
360.889

0.000

0.440
1
0.840


Lactobacillaceae|SPF
0.000
81.000
19.571
30.127

0.001

0.586
0.840
1


Neisseriaceae|Atopo
0.000
0.000
0.000
0.000
1

0.004

0.585
0.441


Neisseriaceae|AtopoBis
0.000
2.000
0.750
0.707

0.004

1
0.022
0.048


Neisseriaceae|Bis
0.000
1.000
0.125
0.354
0.585
0.022
1
0.809


Neisseriaceae|SPF
0.000
8.000
1.143
3.024
0.441
0.048
0.809
1


Halothiobacillaceae|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

<0.0001



Halothiobacillaceae|
0.000
0.000
0.000
0.000
1.000
1
1.000

<0.0001



AtopoBis


Halothiobacillaceae|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

<0.0001



Halothiobacillaceae|SPF
0.000
14.000
4.571
5.255

<0.0001


<0.0001


<0.0001

1


Pasteurellaceae|Atopo
0.000
1.000
0.125
0.354
1
0.594
0.530

0.000



Pasteurellaceae|AtopoBis
0.000
4.000
0.750
1.488
0.594
1
0.925

0.001



Pasteurellaceae|Bis
0.000
14.000
2.000
4.899
0.530
0.925
1

0.001



Pasteurellaceae|SPF
2.000
56.000
20.143
21.836

0.000


0.001


0.001

1


Erysipelotrichaceae|Atopo
998.000
7663.000
4385.500
2215.367
1
0.409
0.011

0.004



Erysipelotrichaceae|
928.000
8147.000
3320.375
2780.457
0.409
1
0.088
0.037


AtopoBis


Erysipelotrichaceae|Bis
2.000
10136.000
1779.000
3443.700
0.011
0.088
1
0.660


Erysipelotrichaceae|SPF
6.000
3618.000
861.857
1483.609

0.004

0.037
0.660
1


Fusobacteriaceae|Atopo
0.000
20.000
3.500
6.845
1

0.008

0.089

0.007



Fusobacteriaceae|AtopoBis
1.000
34434.000
6670.125
12103.868

0.008

1
0.341
0.889


Fusobacteriaceae|Bis
0.000
3175.000
445.125
1109.012
0.089
0.341
1
0.289


Fusobacteriaceae|SPF
5.000
96.000
38.571
31.921

0.007

0.889
0.289
1


Bacillaceae|Atopo
71.000
336.000
147.875
94.253
1

0.001

0.024
0.311


Bacillaceae|AtopoBis
1182.000
28393.000
8495.875
9032.956
0.001
1
0.284
0.028


Bacillaceae|Bis
6.000
13296.000
5596.125
4907.619
0.024
0.284
1
0.244


Bacillaceae|SPF
65.000
7836.000
1401.571
2847.097
0.311
0.028
0.244
1


Listeriaceae|Atopo
0.000
1.000
0.375
0.518
1

0.006

0.029
0.477


Listeriaceae|AtopoBis
0.000
17.000
7.250
6.497

0.006

1
0.590
0.055


Listeriaceae|Bis
0.000
14.000
5.000
4.751
0.029
0.590
1
0.161


Listeriaceae|SPF
0.000
29.000
4.571
10.799
0.477
0.055
0.161
1


Streptococcaceae|Atopo
0.000
21.000
7.500
7.521
1
0.182

0.001

0.056


Streptococcaceae|AtopoBis
0.000
17141.000
3433.000
6026.709
0.182
1
0.046
0.532


Streptococcaceae|Bis
9.000
22660.000
5396.500
7790.156

0.001

0.046
1
0.193


Streptococcaceae|SPF
8.000
237.000
84.714
100.521
0.056
0.532
0.193
1


Vibrionaceae|Atopo
0.000
13.000
2.375
4.534
1
0.191
0.374

<0.0001



Vibrionaceae|AtopoBis
1.000
14.000
5.000
5.155
0.191
1
0.677

0.006



Vibrionaceae|Bis
0.000
23.000
6.125
8.097
0.374
0.677
1

0.002



Vibrionaceae|SPF
20.000
48532.000
15351.143
18959.865

<0.0001


0.006


0.002

1


Methylococcaceae|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000
0.027


Methylococcaceae|
0.000
0.000
0.000
0.000
1.000
1
1.000
0.027


AtopoBis


Methylococcaceae|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1
0.027


Methylococcaceae|SPF
0.000
2.000
0.429
0.787
0.027
0.027
0.027
1


Sutterellaceae|Atopo
0.000
14.000
3.125
5.592
1
0.020
0.708

0.007



Sutterellaceae|AtopoBis
1.000
37.000
12.125
12.253
0.020
1
0.050
0.645


Sutterellaceae|Bis
0.000
10.000
2.500
3.381
0.708
0.050
1
0.019


Sutterellaceae|SPF
1.000
125.000
33.571
44.071

0.007

0.645
0.019
1


Veillonellaceae|Atopo
1.000
2444.000
780.750
938.048
1
0.815
0.085
0.197


Veillonellaceae|AtopoBis
5.000
26.000
12.625
7.689
0.815
1
0.050
0.287


Veillonellaceae|Bis
0.000
10.000
3.375
3.378
0.085
0.050
1

0.003



Veillonellaceae|SPF
1.000
7076.000
2221.857
2915.640
0.197
0.287

0.003

1


Catabacteriaceae|Atopo
0.000
2.000
0.250
0.707
1
0.643
0.220

0.004



Catabacteriaceae|AtopoBis
0.000
0.000
0.000
0.000
0.643
1
0.091

0.001



Catabacteriaceae|Bis
0.000
1472.000
314.000
592.517
0.220
0.091
1
0.097


Catabacteriaceae|SPF
0.000
525.000
113.857
201.222

0.004


0.001

0.097
1


Shewanellaceae|Atopo
0.000
0.000
0.000
0.000
1
1.000
0.352

0.000



Shewanellaceae|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
0.352

0.000



Shewanellaceae|Bis
0.000
1.000
0.250
0.463
0.352
0.352
1

0.003



Shewanellaceae|SPF
0.000
118.000
24.429
42.637

0.000


0.000


0.003

1


Hahellaceae|Atopo
0.000
1.000
0.250
0.463
1
0.427
0.947

0.002



Hahellaceae|AtopoBis
0.000
0.000
0.000
0.000
0.427
1
0.389

0.000



Hahellaceae|Bis
0.000
4.000
0.625
1.408
0.947
0.389
1

0.003



Hahellaceae|SPF
0.000
200.000
56.143
70.099

0.002


0.000


0.003

1


Actinomycetaceae|Atopo
0.000
1.000
0.125
0.354
1
0.190
0.325

0.000



Actinomycetaceae|
0.000
3.000
0.875
1.126
0.190
1
0.743
0.017


AtopoBis


Actinomycetaceae|Bis
0.000
9.000
1.375
3.114
0.325
0.743
1

0.007



Actinomycetaceae|SPF
1.000
60.000
11.571
21.454

0.000

0.017

0.007

1


Alteromonadaceae|Atopo
0.000
1.000
0.125
0.354
1
1.000
0.274

0.001



Alteromonadaceae|
0.000
1.000
0.125
0.354
1.000
1
0.274

0.001



AtopoBis


Alteromonadaceae|Bis
0.000
125.000
24.625
47.533
0.274
0.274
1
0.027


Alteromonadaceae|SPF
0.000
280.000
99.000
107.201

0.001


0.001

0.027
1


Lycoperdaceae|Atopo
0.000
0.000
0.000
0.000
1
1.000
1.000

0.000



Lycoperdaceae|AtopoBis
0.000
0.000
0.000
0.000
1.000
1
1.000

0.000



Lycoperdaceae|Bis
0.000
0.000
0.000
0.000
1.000
1.000
1

0.000



Lycoperdaceae|SPF
0.000
80.000
25.429
32.103

0.000


0.000


0.000

1


Peptostreptococcaceae|
0.000
0.000
0.000
0.000
1
0.308
0.352

0.006



Atopo


Peptostreptococcaceae|
0.000
3.000
0.625
1.188
0.308
1
0.929
0.078


AtopoBis


Peptostreptococcaceae|Bis
0.000
2.000
0.500
0.926
0.352
0.929
1
0.065


Peptostreptococcaceae|
0.000
11.000
3.714
4.348

0.006

0.078
0.065
1


SPF


GENUS



Morganella|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

0.000




Morganella|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

0.000




Morganella|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

0.000




Morganella|SPF

0.000
80.000
25.429
32.103

0.000


0.000


0.000

1



Erwinia|Atopo

0.000
24.000
12.750
8.714
1
0.826
0.710

0.003




Erwinia|AtopoBis

0.000
145.000
49.625
60.985
0.826
1
0.880

0.006




Erwinia|Bis

0.000
50896.000
10198.375
19632.394
0.710
0.880
1
0.010



Erwinia|SPF

146.000
2984.000
919.857
992.120

0.003


0.006

0.010
1



Peptostreptococcus|Atopo

0.000
0.000
0.000
0.000
1
0.308
0.352

0.006




Peptostreptococcus|

0.000
3.000
0.625
1.188
0.308
1
0.929
0.078


AtopoBis



Peptostreptococcus|Bis

0.000
2.000
0.500
0.926
0.352
0.929
1
0.065



Peptostreptococcus|SPF

0.000
11.000
3.714
4.348

0.006

0.078
0.065
1



Dorea|Atopo

0.000
9.000
2.625
2.973
1
0.757
0.527
0.041



Dorea|AtopoBis

0.000
20.000
5.750
7.888
0.757
1
0.346
0.081



Dorea|Bis

0.000
10.000
2.375
3.926
0.527
0.346
1

0.008




Dorea|SPF

0.000
30.000
13.429
10.114
0.041
0.081

0.008

1



Ruminococcus|Atopo

13.000
281.000
62.375
89.427
1
0.030
0.021

0.002




Ruminococcus|AtopoBis

29.000
1586.000
441.500
610.453
0.030
1
0.891
0.341



Ruminococcus|Bis

33.000
1006.000
398.250
401.712
0.021
0.891
1
0.412



Ruminococcus|SPF

69.000
20346.000
4684.143
8151.122

0.002

0.341
0.412
1



Kangiella|Atopo

0.000
0.000
0.000
0.000
1
1.000
0.548

0.003




Kangiella|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
0.548

0.003




Kangiella|Bis

0.000
1.000
0.125
0.354
0.548
0.548
1
0.015



Kangiella|SPF

0.000
23.000
4.429
8.344

0.003


0.003

0.015
1



Enterovibrio|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

<0.0001




Enterovibrio|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

<0.0001




Enterovibrio|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

<0.0001




Enterovibrio|SPF

0.000
239.000
88.000
92.454

<0.0001


<0.0001


<0.0001

1



Coprobacillus|Atopo

331.000
2302.000
1152.125
678.666
1

0.003


0.008


0.004




Coprobacillus|AtopoBis

0.000
1735.000
257.375
605.539

0.003

1
0.772
0.970



Coprobacillus|Bis

0.000
1309.000
200.625
452.797

0.008

0.772
1
0.750



Coprobacillus|SPF

0.000
924.000
177.571
348.761

0.004

0.970
0.750
1



Actinomyces|Atopo

0.000
1.000
0.125
0.354
1
0.190
0.325

0.000




Actinomyces|AtopoBis

0.000
3.000
0.875
1.126
0.190
1
0.743
0.017



Actinomyces|Bis

0.000
9.000
1.375
3.114
0.325
0.743
1

0.007




Actinomyces|SPF

1.000
60.000
11.571
21.454

0.000

0.017

0.007

1



Marinomonas|Atopo

0.000
1.000
0.250
0.463
1
0.694
0.694

0.002




Marinomonas|AtopoBis

0.000
1.000
0.375
0.518
0.694
1
0.431

0.007




Marinomonas|Bis

0.000
1.000
0.125
0.354
0.694
0.431
1

0.001




Marinomonas|SPF

0.000
494.000
144.857
185.634

0.002


0.007


0.001

1



Vibrio|Atopo

0.000
13.000
2.375
4.534
1
0.216
0.381

<0.0001




Vibrio|AtopoBis

1.000
12.000
4.750
4.683
0.216
1
0.718

0.006




Vibrio|Bis

0.000
23.000
6.125
8.097
0.381
0.718
1

0.002




Vibrio|SPF

19.000
48256.000
15217.857
18829.126

<0.0001


0.006


0.002

1



Dialister|Atopo

0.000
9.000
1.375
3.114
1
0.020
0.534
0.129



Dialister|AtopoBis

0.000
18.000
6.625
6.675
0.020
1

0.003

0.465



Dialister|Bis

0.000
1.000
0.250
0.463
0.534

0.003

1
0.034



Dialister|SPF

0.000
7.000
3.571
3.101
0.129
0.465
0.034
1



Halothiobacillus|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

<0.0001




Halothiobacillus|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

<0.0001




Halothiobacillus|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

<0.0001




Halothiobacillus|SPF

0.000
14.000
4.571
5.255

<0.0001


<0.0001


<0.0001

1



Trichococcus|Atopo

0.000
6.000
1.125
2.031
1

0.007

0.041
0.009



Trichococcus|AtopoBis

0.000
0.000
0.000
0.000

0.007

1
0.519
1.000



Trichococcus|Bis

0.000
1.000
0.125
0.354
0.041
0.519
1
0.533



Trichococcus|SPF

0.000
0.000
0.000
0.000
0.009
1.000
0.533
1



Nitrincola|Atopo

0.000
0.000
0.000
0.000
1
0.577
1.000

0.001




Nitrincola|AtopoBis

0.000
3.000
0.375
1.061
0.577
1
0.577

0.003




Nitrincola|Bis

0.000
0.000
0.000
0.000
1.000
0.577
1

0.001




Nitrincola|SPF

0.000
226.000
55.143
83.762

0.001


0.003


0.001

1



Serratia|Atopo

0.000
0.000
0.000
0.000
1
0.198
1.000

0.000




Serratia|AtopoBis

0.000
3.000
0.750
1.165
0.198
1
0.198
0.009



Serratia|Bis

0.000
0.000
0.000
0.000
1.000
0.198
1

0.000




Serratia|SPF

0.000
621.000
192.286
237.182

0.000

0.009

0.000

1



Ferrimonas|Atopo

0.000
0.000
0.000
0.000
1
0.625
1.000

<0.0001




Ferrimonas|AtopoBis

0.000
1.000
0.125
0.354
0.625
1
0.625

0.000




Ferrimonas|Bis

0.000
0.000
0.000
0.000
1.000
0.625
1

<0.0001




Ferrimonas|SPF

0.000
623.000
140.714
227.571

<0.0001


0.000


<0.0001

1



Butyrivibrio|Atopo

0.000
1.000
0.125
0.354
1
1.000
0.534
0.032



Butyrivibrio|AtopoBis

0.000
1.000
0.125
0.354
1.000
1
0.534
0.032



Butyrivibrio|Bis

0.000
0.000
0.000
0.000
0.534
0.534
1

0.006




Butyrivibrio|SPF

0.000
1.000
0.571
0.535
0.032
0.032

0.006

1



Oscillospira|Atopo

0.000
3.000
0.750
1.165
1
0.055
0.144

0.000




Oscillospira|AtopoBis

1.000
7.000
3.125
2.167
0.055
1
0.646
0.083



Oscillospira|Bis

0.000
3538.000
715.625
1371.803
0.144
0.646
1
0.030



Oscillospira|SPF

2.000
3855.000
713.000
1439.678

0.000

0.083
0.030
1



Epulopiscium|Atopo

0.000
9.000
1.625
3.068
1
0.434
0.099
0.014



Epulopiscium|AtopoBis

0.000
2.000
0.375
0.744
0.434
1
0.015

0.001




Epulopiscium|Bis

0.000
8385.000
1199.875
2932.011
0.099
0.015
1
0.393



Epulopiscium|SPF

2.000
60.000
17.429
24.110
0.014

0.001

0.393
1



Escherichia|Atopo

0.000
7.000
2.000
2.507
1
0.029
0.557
0.351



Escherichia|AtopoBis

0.000
0.000
0.000
0.000
0.029
1
0.110

0.002




Escherichia|Bis

0.000
52.000
10.375
19.799
0.557
0.110
1
0.133



Escherichia|SPF

0.000
5.000
2.714
1.799
0.351

0.002

0.133
1



Alkalimonas|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

0.006




Alkalimonas|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

0.006




Alkalimonas|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

0.006




Alkalimonas|SPF

0.000
30.000
7.714
11.339

0.006


0.006


0.006

1



Listeria|Atopo

0.000
1.000
0.375
0.518
1

0.006

0.029
0.477



Listeria|AtopoBis

0.000
17.000
7.250
6.497

0.006

1
0.590
0.055



Listeria|Bis

0.000
14.000
5.000
4.751
0.029
0.590
1
0.161



Listeria|SPF

0.000
29.000
4.571
10.799
0.477
0.055
0.161
1



Streptococcus|Atopo

0.000
21.000
7.500
7.521
1
0.182

0.001

0.056



Streptococcus|AtopoBis

0.000
17141.000
3433.000
6026.709
0.182
1
0.046
0.532



Streptococcus|Bis

9.000
22660.000
5396.500
7790.156

0.001

0.046
1
0.193



Streptococcus|SPF

8.000
237.000
84.714
100.521
0.056
0.532
0.193
1



Actinobacillus|Atopo

0.000
0.000
0.000
0.000
1
0.681
0.328

0.000




Actinobacillus|AtopoBis

0.000
1.000
0.125
0.354
0.681
1
0.571

0.001




Actinobacillus|Bis

0.000
8.000
1.125
2.800
0.328
0.571
1

0.005




Actinobacillus|SPF

0.000
39.000
12.000
15.011

0.000


0.001


0.005

1



Roseburia|Atopo

10.000
46.000
23.125
12.722
1
0.773
0.040
0.321



Roseburia|AtopoBis

1.000
56.000
22.875
17.836
0.773
1
0.078
0.203



Roseburia|Bis

1.000
22.000
7.875
7.453
0.040
0.078
1

0.003




Roseburia|SPF

8.000
192.000
65.429
64.714
0.321
0.203

0.003

1



Bacillus|Atopo

70.000
331.000
145.500
92.705
1

0.001

0.023
0.298



Bacillus|AtopoBis

1162.000
24417.000
7572.750
7652.213

0.001

1
0.284
0.029



Bacillus|Bis

5.000
11338.000
4861.250
4100.801
0.023
0.284
1
0.250



Bacillus|SPF

57.000
7717.000
1376.429
2805.259
0.298
0.029
0.250
1



Parabacteroides|Atopo

0.000
40.000
8.875
13.559
1
0.195
0.562
0.036



Parabacteroides|AtopoBis

1.000
35.000
12.750
10.820
0.195
1
0.061
0.397



Parabacteroides|Bis

0.000
9.000
3.750
3.284
0.562
0.061
1

0.008




Parabacteroides|SPF

0.000
43.000
23.571
16.662
0.036
0.397

0.008

1



Sarcina|Atopo

0.000
1.000
0.125
0.354
1

0.005

0.009
0.317



Sarcina|AtopoBis

0.000
1929.000
424.250
698.617

0.005

1
0.815
0.082



Sarcina|Bis

0.000
2282.000
700.125
966.646
0.009
0.815
1
0.131



Sarcina|SPF

0.000
8.000
2.286
3.147
0.317
0.082
0.131
1



Enterococcus|Atopo

11.000
38.000
20.750
9.588
1

0.000

0.016
0.040



Enterococcus|AtopoBis

121.000
1543.000
718.125
539.764

0.000

1
0.161
0.100



Enterococcus|Bis

0.000
1425.000
558.625
527.899
0.016
0.161
1
0.773



Enterococcus|SPF

39.000
1382.000
260.429
495.621
0.040
0.100
0.773
1



Carnobacterium|Atopo

0.000
1.000
0.125
0.354
1

0.006

0.045
0.543



Carnobacterium|AtopoBis

0.000
42.000
17.500
18.769

0.006

1
0.451
0.040



Carnobacterium|Bis

0.000
29.000
6.500
10.100
0.045
0.451
1
0.185



Carnobacterium|SPF

0.000
4.000
1.000
1.732
0.543
0.040
0.185
1



Coprococcus|Atopo

244.000
35300.000
7793.625
11731.118
1

0.001

0.012
0.220



Coprococcus|AtopoBis

10.000
79.000
40.625
22.671

0.001

1
0.433
0.050



Coprococcus|Bis

6.000
5234.000
893.750
1803.125
0.012
0.433
1
0.228



Coprococcus|SPF

20.000
31009.000
6867.571
12275.631
0.220
0.050
0.228
1



Enterobacter|Atopo

0.000
3.000
0.500
1.069
1
0.849
0.333

0.000




Enterobacter|AtopoBis

0.000
1.000
0.375
0.518
0.849
1
0.437

0.001




Enterobacter|Bis

0.000
12.000
2.875
4.794
0.333
0.437
1

0.007




Enterobacter|SPF

3.000
2140.000
682.571
877.138

0.000


0.001


0.007

1



Neisseria|Atopo

0.000
0.000
0.000
0.000
1

0.003

0.538
0.387



Neisseria|AtopoBis

0.000
2.000
0.750
0.707

0.003

1
0.014
0.032



Neisseria|Bis

0.000
1.000
0.125
0.354
0.538
0.014
1
0.785



Neisseria|SPF

0.000
5.000
0.714
1.890
0.387
0.032
0.785
1



Photobacterium|Atopo

0.000
0.000
0.000
0.000
1
0.222
1.000

<0.0001




Photobacterium|AtopoBis

0.000
2.000
0.250
0.707
0.222
1
0.222

<0.0001




Photobacterium|Bis

0.000
0.000
0.000
0.000
1.000
0.222
1

<0.0001




Photobacterium|SPF

1.000
121.000
42.286
49.671

<0.0001


<0.0001


<0.0001

1



Brenneria|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

0.000




Brenneria|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

0.000




Brenneria|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

0.000




Brenneria|SPF

0.000
595.000
156.429
226.566

0.000


0.000


0.000

1



Oceanobacillus|Atopo

0.000
0.000
0.000
0.000
1
0.465

0.005

1.000



Oceanobacillus|AtopoBis

0.000
1.000
0.125
0.354
0.465
1
0.026
0.480



Oceanobacillus|Bis

0.000
2.000
0.625
0.744

0.005

0.026
1

0.006




Oceanobacillus|SPF

0.000
0.000
0.000
0.000
1.000
0.480

0.006

1



Lactobacillus|Atopo

263.000
7137.000
2406.750
2385.720
1

0.000

<0.0001

<0.0001




Lactobacillus|AtopoBis

3.000
27.000
12.375
9.226

0.000

1
0.277
0.441



Lactobacillus|Bis

0.000
1026.000
133.000
360.889

<0.0001

0.277
1
0.774



Lactobacillus|SPF

0.000
81.000
19.571
30.127

<0.0001

0.441
0.774
1



Xanthomonas|Atopo

0.000
1.000
0.125
0.354
1
0.620
0.138

0.007




Xanthomonas|AtopoBis

0.000
0.000
0.000
0.000
0.620
1
0.052

0.002




Xanthomonas|Bis

0.000
1005.000
207.500
395.202
0.138
0.052
1
0.163



Xanthomonas|SPF

0.000
29.000
6.000
10.360

0.007


0.002

0.163
1



Sutterella|Atopo

0.000
14.000
3.125
5.592
1
0.009
0.657

0.003




Sutterella|AtopoBis

1.000
37.000
12.125
12.253
0.009
1
0.026
0.586



Sutterella|Bis

0.000
10.000
2.500
3.381
0.657
0.026
1
0.009



Sutterella|SPF

1.000
125.000
33.571
44.071

0.003

0.586
0.009
1



Staphylococcus|Atopo

0.000
8.000
1.250
2.765
1

0.000


0.001


0.003




Staphylococcus|AtopoBis

6.000
82.000
32.625
28.213

0.000

1
0.744
0.558



Staphylococcus|Bis

0.000
132.000
40.000
45.854

0.001

0.744
1
0.785



Staphylococcus|SPF

2.000
838.000
191.000
333.003

0.003

0.558
0.785
1



Lachnobacterium|Atopo

0.000
22.000
4.125
7.605
1
0.072
0.275
0.068



Lachnobacterium|AtopoBis

0.000
0.000
0.000
0.000
0.072
1

0.006


0.001




Lachnobacterium|Bis

0.000
11871.000
2128.000
4324.494
0.275

0.006

1
0.418



Lachnobacterium|SPF

0.000
809.000
230.857
372.166
0.068

0.001

0.418
1



Vagococcus|Atopo

0.000
0.000
0.000
0.000
1

0.001


0.004

0.129



Vagococcus|AtopoBis

0.000
37.000
7.000
12.387

0.001

1
0.570
0.049



Vagococcus|Bis

0.000
34.000
8.500
12.166

0.004

0.570
1
0.144



Vagococcus|SPF

0.000
3.000
0.857
1.215
0.129
0.049
0.144
1



Leclercia|Atopo

0.000
2.000
0.500
0.756
1
0.298
0.501

<0.0001




Leclercia|AtopoBis

0.000
4.000
0.500
1.414
0.298
1
0.707

<0.0001




Leclercia|Bis

0.000
1.000
0.250
0.463
0.501
0.707
1

<0.0001




Leclercia|SPF

12.000
4650.000
1419.143
1875.855

<0.0001


<0.0001


<0.0001

1



Fusobacterium|Atopo

0.000
20.000
3.500
6.845
1

0.006

0.065

0.005




Fusobacterium|AtopoBis

1.000
34434.000
6670.000
12103.946

0.006

1
0.307
0.865



Fusobacterium|Bis

0.000
3175.000
445.125
1109.012
0.065
0.307
1
0.249



Fusobacterium|SPF

4.000
94.000
38.000
31.559

0.005

0.865
0.249
1



Citrobacter|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

0.000




Citrobacter|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

0.000




Citrobacter|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

0.000




Citrobacter|SPF

0.000
41.000
11.857
16.477

0.000


0.000


0.000

1



Hahella|Atopo

0.000
1.000
0.250
0.463
1
0.265
0.925

0.000




Hahella|AtopoBis

0.000
0.000
0.000
0.000
0.265
1
0.229

<0.0001




Hahella|Bis

0.000
4.000
0.625
1.408
0.925
0.229
1

0.000




Hahella|SPF

0.000
200.000
56.143
70.099

0.000


<0.0001


0.000

1



Alcanivorax|Atopo

0.000
4.000
1.500
1.773
1
0.481
0.085
0.045



Alcanivorax|AtopoBis

0.000
73.000
22.125
28.119
0.481
1
0.019
0.170



Alcanivorax|Bis

0.000
0.000
0.000
0.000
0.085
0.019
1

0.001




Alcanivorax|SPF

0.000
531.000
138.857
200.897
0.045
0.170

0.001

1



Facklamia|Atopo

0.000
0.000
0.000
0.000
1
1.000
1.000

0.003




Facklamia|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
1.000

0.003




Facklamia|Bis

0.000
0.000
0.000
0.000
1.000
1.000
1

0.003




Facklamia|SPF

0.000
56.000
11.571
21.509

0.003


0.003


0.003

1



Faecalibacterium|Atopo

0.000
252.000
39.625
86.169
1
0.010
0.687

0.007




Faecalibacterium|AtopoBis

8.000
120.000
57.000
37.413
0.010
1

0.004

0.811



Faecalibacterium|Bis

1.000
31.000
13.375
11.476
0.687

0.004

1

0.003




Faecalibacterium|SPF

8.000
212.000
88.571
76.868

0.007

0.811

0.003

1



Eubacterium|Atopo

3.000
43.000
8.875
13.861
1
0.024
0.417

0.000




Eubacterium|AtopoBis

10.000
32.000
18.500
8.783
0.024
1
0.130
0.052



Eubacterium|Bis

1.000
2387.000
313.625
838.348
0.417
0.130
1

0.001




Eubacterium|SPF

13.000
1107.000
485.143
399.257

0.000

0.052

0.001

1



Shewanella|Atopo

0.000
0.000
0.000
0.000
1
1.000
0.144

<0.0001




Shewanella|AtopoBis

0.000
0.000
0.000
0.000
1.000
1
0.144

<0.0001




Shewanella|Bis

0.000
1.000
0.250
0.463
0.144
0.144
1

<0.0001




Shewanella|SPF

0.000
118.000
24.429
42.637

<0.0001


<0.0001


<0.0001

1



Tatumella|Atopo

0.000
0.000
0.000
0.000
1
0.454
1.000

<0.0001




Tatumella|AtopoBis

0.000
1.000
0.125
0.354
0.454
1
0.454

0.000




Tatumella|Bis

0.000
0.000
0.000
0.000
1.000
0.454
1

<0.0001




Tatumella|SPF

0.000
76.000
26.714
33.674

<0.0001


0.000


<0.0001

1



Bacteroides|Atopo

86967.000
133771.000
108707.250
14136.218
1
0.075

<0.0001


0.000




Bacteroides|AtopoBis

211.000
117745.000
64625.875
53942.157
0.075
1

0.000

0.027



Bacteroides|Bis

74.000
515.000
171.750
143.702

<0.0001


0.000

1
0.083



Bacteroides|SPF

57.000
107600.000
28388.714
48274.613

0.000

0.027
0.083
1









It has also been found that A. parvulum is correlated with the presence/abundance of GALT foci. Therefore there is provided an assay for identifying the likelihood of an individual of having UC or CD or IBD by measuring a relative abundance of A. parvulum by measuring the abundance of GALT foci. This correlation can also be used to provide a method of diagnostic that comprises collecting samples to measure the abundance of GAT foci, determining the presence of A. parvulum based on the cytokine(s) measurement and establishing a diagnosis.


In other aspect of the invention it has been shown that certain OTU's and/or taxa are indicative of improve therapeutic response. Table 7 exemplifies OTU's and/or taxa that exhibit a significant difference between the levels of bacteria between patients that responded to treatment. The patients in the two groups (responded to treatment/failed to respond to treatment) received a systemic corticosteroid medication (prednisone) as their acute anti-inflammatory therapy. Two patients (in the group that responded) received the mucosally active corticosteroid medication Entocort instead. Azathioprine (n=11) or methotrexate (n=4) immunomodulator medication was initiated in the patients for maintenance therapy. The clinical failure of response was determined by Physician Global Assessment and Pediatric Crohn's Disease Activity Index scoring determinations.


Clearly from the data of table 7 there is link between the level of bacteria and the efficacy of treatment. Thus when a patient exhibits bacterial levels in one or more taxa or OTU's from table 7 that are more elevated than a predetermined level or average corresponding to responders level the patient is likely not to respond to treatment. Alternatively patients exhibiting levels of bacteria lower that a predetermined level or average corresponding to non responders will profit the most from the treatment. The patient that did not respond have a different physiological or pathological status as assessed by standard diagnostic tests. For example patients with levels of Erwinia greater than about 3431 or preferably greater than about 13482 (one std dev) are likely not to respond and patient with lower levels than these likely to benefit most.

















TABLE 7











Std.
p|
p|



Observations
Minimum
Maximum
Mean
deviation
responded
Failed























Analysis at OTU level Variable










Eubacterium (OTU589746)|responded

9
0.000
4.000
1.333
1.732
1

0.011




Eubacterium (OTU589746)|Failed

6
2.000
20.000
9.667
7.174

0.011

1



Oribacteriumsinus (OTU470747)|

9
0.000
11.000
2.778
3.598
1

0.005



responded



Oribacteriumsinus (OTU470747)|Failed

6
2.000
77.000
28.000
25.954

0.005

1



Veillonellaceae (OTU535825)|

9
0.000
40.000
9.000
14.186
1

0.024



responded



Veillonellaceae (OTU535825)|Failed

6
2.000
93.000
49.500
38.667

0.024

1



Enterobacteriaceae (OTU323418)|

9
0.000
180.000
23.889
59.675
1

0.035



responded



Enterobacteriaceae (OTU323418)|

6
1.000
140.000
34.000
55.714

0.035

1


Failed



Lachnospiraceae (OTU71387)|

9
0.000
137.000
16.000
45.418
1

0.015



responded



Lachnospiraceae (OTU71387)|Failed

6
2.000
259.000
73.500
95.663

0.015

1



Atopobium (OTU529659)|responded

9
0.000
101.000
17.889
32.259
1

0.018




Atopobium (OTU529659)|Failed

6
19.000
187.000
78.667
62.516

0.018

1



Mogibacterium (OTU46159)|responded

9
0.000
163.000
21.556
53.346
1

0.043




Mogibacterium (OTU46159)|Failed

6
3.000
376.000
79.167
146.389

0.043

1



Propionibacteriumacnes (OTU368907)|

9
0.000
192.000
32.667
68.431
1

0.044



responded



Propionibacteriumacnes (OTU368907)|

6
1.000
363.000
63.500
146.761

0.044

1


Failed



Alteromonadaceae; BD2-13

9
0.000
44.000
8.444
15.001
1

0.022



(OTU110075)|responded



Alteromonadaceae; BD2-13

6
1.000
715.000
142.000
281.637

0.022

1


(OTU110075)|Failed



Coprococcus (OTU182512)|responded

9
0.000
133.000
23.778
43.249
1

0.045




Coprococcus (OTU182512)|Failed

6
3.000
282.000
129.167
128.205

0.045

1



Lachnospiraceae (OTU303772)|

9
1.000
78.000
14.778
25.806
1

0.043



responded



Lachnospiraceae (OTU303772)|Failed

6
2.000
509.000
167.667
205.745

0.043

1



Clostridiales (OTU204932)|responded

9
0.000
2054.000
237.222
681.670
1

0.024




Clostridiales (OTU204932)|Failed

6
11.000
5639.000
1037.833
2257.516

0.024

1



Bacteroidales (OTU183618)|responded

9
0.000
2059.000
246.667
681.482
1

0.023




Bacteroidales (OTU183618)|Failed

6
9.000
6706.000
1295.167
2666.325

0.023

1



Ruminococcaceae (OTU195252)|

9
0.000
120.000
21.889
40.946
1

0.044



responded



Ruminococcaceae (OTU195252)|

6
2.000
9494.000
1716.333
3814.455

0.044

1


Failed



Sutterella (OTU295422)|responded

9
2.000
1132.000
145.556
371.004
1

0.018




Sutterella (OTU295422)|Failed

6
17.000
20759.000
6729.833
8755.580

0.018

1



Enterobacteriaceae (OTU307080)|

9
6.000
13849.000
1846.333
4559.821
1

0.025



responded



Enterobacteriaceae (OTU307080)|

6
165.000
15872.000
4616.833
5840.027

0.025

1


Failed



Ruminococcus (OTU174136)|

9
0.000
24.000
6.667
9.000
1

0.033



responded



Ruminococcus (OTU174136)|Failed

6
1.000
48877.000
8170.833
19941.884

0.033

1



Clostridiumramosum (OTU470139)|

9
5.000
10411.000
1186.444
3459.360
1

0.045



responded



Clostridiumramosum (OTU470139)|

6
10.000
139273.000
36964.333
53876.950

0.045

1


Failed



Erwinia (OTU289103)|responded

9
3.000
30231.000
3425.667
10052.419
1

0.010




Erwinia (OTU289103)|Failed

6
78.000
164661.000
42988.167
62253.786

0.010

1


Analysis at genus level


Variable



Erwinia|responded

9
3.000
30231.000
3430.778
10050.512
1

0.013




Erwinia|Failed

6
78.000
164661.000
42989.333
62253.169

0.013

1



Atopobium|responded

9
0.000
307.000
51.556
101.187
1

0.045




Atopobium|Failed

6
23.000
231.000
90.167
76.434

0.045

1



Propionibacterium|

9
0.000
196.000
40.444
79.783
1

0.042



responded



Propionibacterium|Failed

6
2.000
363.000
63.833
146.593

0.042

1









It will be appreciated that more than one taxa and/or OTU can be combined to identify patients that are more likely to respond to treatment. For example one could combine the measurement of OTU295422 and of taxa Erwinia in a patient and if the levels are below about 145 and about 3430 respectively then the patient is considered likely to respond. It should be noted that OTU's are most of the time closely related to a taxa therefore the above described approach would also be applicable using taxa associated with an OTU.


Thus in one aspect the present invention provides a method to test or assay or measure the levels of gut bacteria obtained directly from the gut or from stools and in which the actual measurement of bacterial levels is done in vitro. The measured levels can be used to assess the nature, severity or stage of IBD, CD or UC disease and determine treatment course such as the administration of certain drugs.


Furthermore there is also provided a method in which a test to measure the level(s) of bacteria as described above is requested to provide the results of an analysis to determine whether a patient has IBD, CD or UC or to determine the severity or stage of such disease by assessing bacterial levels as described above and administering a treatment if the patient exhibit the type and levels of bacteria associated with disease or the severity or stage of the disease.


Thus the present invention provides to the identification of pathological states or characteristics of patients by identifying bacteria associated with disease and of physiological states by providing levels of bacteria present in disease or at different stages or severity of disease.


The bacterial taxa and proteins described above can be referred to as diagnostic markers. These diagnostic markers can be used in a method for classifying a sample as being associated with IBD, UC or CD. The method comprises the steps of determining a presence or level of one or more of the diagnostic markers and comparing the presence or level to samples from IBD, UC or CD patients and/or normal patients. A combination of diagnostic markers may be combined together and may also further be combined with a standard diagnostic results derived from a disease activity index.


The algorithm can be a statistical algorithm which may comprise a learning statistical classifier system (or combination of such systems) such as neural network, random forest, interactive tree and the like, as would be known to a person skilled in the art. The predictive value of the classifying system maybe predetermined and may for example be at least 60%, 70%, 80%, 90% or 95%. The classification result may be provided to a clinician such as a gastroenterologist or general practitioner.


In yet a further aspect of the invention there is provided a method of classifying a gut sample to determine an association with IBD, UC or CD that comprises determining a diagnostic marker profile by detecting a presence or level of at least one gut diagnostic marker and classifying the sample as IBD, UC or CD by comparing the diagnostic marker profile to samples from IBD, UC or CD patients or normal subjects or combination thereof. The profile can be combined with a diagnostic based on a disease activity index specific for IBD, UC or CD.


The diagnostic marker can be selected from H2S producing bacteria, Proteobacteria, butyrate producing bacteria, Fusobacterium nucleatum, Veillonella parvula, Atopobium parvulum, Firmicutes, Clostridia, Clostridiales, Lachnopiraceae, Eubacterium, Roseburia, Coprococcus, Clostridium, Eubacterium rectale, Clostridium coccoides, Roseburia inulivorans, Verrucomicrobiae, Clostridiales, Verrucomicrobiales, Verrucomicrobiacae, Lachnospiraceae, Paenibacillaceae, Akkermansia, Turicibacter, Paenibacillus, Pasteurellales, Chromatialles, Hydrogenophilales, Oceanospirillales, Rhizobiales, Halomonadaceae, Pasteurellaceae, Bradyrhizobiaceae, Methylococcaceae, Hydrogenophilaceae, Porphyromonas, Lautropia, Methylobacterium, Haemophilus, Finegoldia, Nitrincola, Hydrogenophilu, Actinobacillus, Anaerococcus, Mobiluncus, Enterobacter, Vitreoscilla, Alcanivorax, Veillonella, Tatumella, Staphylococcaceae, Paenibacillaceae, Listeriaceae, Listeria, Paenibacillus, Staphylococcus, Negativicutes, Betaproteobacteria, Pasteurellales, Chromatialles, Burkholderiales, Selenomonadales, Pasteurellaceae, Haemophilus, Pantoea, Carnobacteriaceae, Granulicatella, Mogibacterium, Proprionibacterium, Bacillaceae, Atopobium, Hydrogenophilales, Rhizobiales, Bradyrhyzobiaceae, Hydrogenophylaceae, Porphyromonas, Lautropia, Tannarella, Finegoldia, Hydrogenophilus, Catonella, Mobilumcus, Alcanivorax, Afipia, sulfur dioxygenase (ETHE1), thiosulfate sulfur transferase (TST), cytochrome c oxidase subunit IV, sulfide dehydrogenase (SQR), complexes III and IV of mithochondrial respiratory chain, Cxcl1, IL17a, II12, II1β and combination thereof.


The profile may consist of level(s) of a marker or the combination of levels from different markers or the relative levels (ratios) of markers combined or not with a diagnosis based on a disease activity index. The profile may also comprise levels of markers over time or stages of the disease(s) or severity of the disease(s). The profile may also be weighted with respect to the markers or the diagnosis.


There is also provided an apparatus comprising a diagnostic marker detector capable of detecting one or more of the markers described above for example by methods described in this application, a processor configured to classify the sample as an IBD, UC or CD sample by comparing the diagnostic marker profile to samples from IBD, CD, UC or normal subjects or combination thereof and a result display unit to display to a user a classification obtained from the processor. The processor may also receive from an input a diagnostic result based on a disease activity index specific for IBD, UC or CD and combine this diagnostic result with the diagnostic marker profile to generate the classification. Thus the processor may use training data or a training cohort to identify the characterisitics of the diagnostic marker that provides a reliable classification.The data provided here (levels of bacteria and proteins for example) with their correlation to presence of disease or disease severity or progression can be used as a training cohort. However it will be appreciated that additional data could be generated to improve the training data based on the guidance of the results presented in this application.


It will be understood that the processor may use algorithms as described above.


EXAMPLES
Example 1

An inception cohort of 157 patients (84 Crohn's disease (CD), 20 ulcerative colitis (UC) and 53 controls; Table 8) was recruited.

















TABLE 8









Site
Visual
Paris
PCDAI/



#SampleID
Description
M/F
age
Sampled
Appearance
Classification
PUCAI
Experiment























HMC002RC
UC
F
16
RC
normal
E1, S0
20 (Mild)
Illumina


HMC003RC
Control
F
9
RC
normal
n/a
n/a
Illumina


HMC004RC
Control
M
12
RC
normal
n/a
n/a
454Pyro


HMC005RC
Control
F
10
RC
normal
n/a
n/a
Illumina


HMC006RC
Control
M
15
RC
normal
n/a
n/a
Illumina


HMC012RC
CD
M
13
RC
normal
A1b, L1, B1, G0
12.5 (Mild)
Illumina


HMC013RC
UC
M
12
RC
inflammed
E4, S1
65
Illumina









(Severe)


HMC014RC
CD
F
14
RC
normal
A1b, L3, L4a, B1,
37.5
454pyro








G1
(Moderate


HMC015RC
CD
F
14
RC
normal
A1b, L1, B2, G0
10 (Mild)
Illumina


HMC016RC
CD
M
13
RC
normal
A1b, L3, B1, G0
20 (Mild)
Illumina


HMC017RC
CD
M
13
RC
inflammed
A1b, L2, L4a, B1,
57.5
Illumina








G1
(Severe)


HMC018RC
Control
M
13
RC
normal
n/a
n/a
Illumina


HMC019RC
UC
M
14
RC
inflammed
E4, S1
80
Illumina/454pyro









(Severe)


HMC020RC
Control
F
12
RC
normal
n/a
n/a
454Pyro


HMC022RC
CD
M
14
RC
normal
A1b, L1, B1, G1
37.5
Illumina









(Moderate


HMC023RC
UC
M
14
RC
normal
E4, S1
50
Illumina/454pyro









(Moderate


HMC024RC
UC
M
16
RC
normal
E3, S1
40
454pyro









(Moderate


HMC025RC
CD
F
15
RC
normal
A1b, L4b, B1, G0
20 (Mild)
Illumina


HMC026RC
Control
F
16
RC
normal
n/a
n/a
454Pyro


HMC027RC
Control
F
16
RC
normal
n/a
n/a
Illumina


HMC028RC
Control
M
13
RC
normal
n/a
n/a
Illumina/454pyro


HMC029RC
CD
M
14
RC
inflammed
A1b, L3, L4a, B1,
32.5
Illumina








G0
(Moderate


HMC030RC
CD
F
17
RC
inflammed
A1b, L3, L4a, B1,
45
Illumina








G0
(Severe)


HMC038RC
CD
F
16
RC
inflammed
A1b, L2, B1p, G0
20 (Mild)
Illumina


HMC039RC
CD
F
13
RC
normal
A1b, L1, L4a, B1,
60
Illumina








G1
(Severe)


HMC041RC
CD
F
15
RC
normal
A1b, L3, B1, G0
57.5
Illumina









(Severe)


HMC042RC
Control
M
17
RC
normal
n/a
n/a
Illumina


HMC043RC
Control
F
16
RC
normal
n/a
n/a
Illumina


HMC044RC
CD
F
15
RC
normal
A1b, L1, B3, G1
52.5
Illumina









(Severe)


HMC045RC
UC
M
17
RC
normal
E3
45
Illumina









(Moderate


HMC046RC
UC
F
4
RC
inflammed
E4, S1
65
Illumina/454pyro









(Severe)


HMC047RC
CD
M
16
RC
inflammed
A1b, L3, L4a, B1,
65
Illumina








G1
(Severe)


HMC049RC
CD
M
12
RC
normal
A1b, L1, B1, G1
40
Illumina/454pyro









(Severe)


HMC050RC
CD
F
16
RC
normal
A1b, L1, B1, G0
12.5 (Mild)
Illumina


HMC051RC
CD
F
16
RC
inflammed
A1b, L3, L4a, B1,
65
Illumina








G0
(Severe)


HMC052RC
Control
F
8
RC
normal
n/a
n/a
454Pyro


HMC055RC
Control
M
6
RC
normal
n/a
n/a
Illumina/454pyro


HMC056RC
Control
F
8
RC
normal
n/a
n/a
Illumina


HMC059RC
Control
M
14
RC
normal
n/a
n/a
Illumina


HMC061RC
CD
M
9
RC
inflammed
A1a, L2, B1, G0
32.5
Illumina/454pyro









(Moderate


HMC062RC
CD
F
15
RC
inflammed
A1b, L3, L4a, B3,
57.5
Illumina








G0
(Severe)


HMC063RC
CD
M
13
RC
normal
A1b, L1, L4a, B1,
65
Illumina/454pyro








G1
(Severe)


HMC064RC
UC
M
18
RC
inflammed
E4, S1
0
Illumina









(Inactive)


HMC065RC
CD
M
16
RC
normal
A1b, L1, B2, B3, G0
50
Illumina









(Severe)


HMC066RC
UC
F
18
RC
inflammed
E4, S0
35
Illumina/454pyro









(Moderate


HMC067RC
Control
F
17
RC
normal
n/a
n/a
Illumina


HMC068RC
CD
M
17
RC
inflammed
A1b, L2, B1, G0
32.5
454Pyro









(Moderate


HMC069RC
Control
F
17
RC
normal
n/a
n/a
Illumina


HMC070RC
Control
F
8
RC
normal
n/a
n/a
454Pyro


HMC071RC
Control
M
11
RC
normal
n/a
n/a
Illumina


HMC072RC
CD
M
12
RC
inflammed
A1b, L3, L4a, B1,
55
Illumina








G1
(Severe)


HMC073RC
Control
F
16
RC
normal
n/a
n/a
Illumina


HMC074RC
Control
F
16
RC
normal
n/a
n/a
454Pyro


HMC075RC
CD
M
14
RC
inflammed
A1b, L2, B1, G1
50
Illumina









(Severe)


HMC076RC
UC
F
17
RC
inflammed
E4, S0
55
Illumina/454pyro









(Moderate


HMC077RC
UC
M
17
RC
normal
E3, S0
50
Illumina/454pyro









(Moderate


HMC078RC
CD
F
15
RC
normal
A1b, L1, B1, G0
45
Illumina









(Severe)


HMC079RC
CD
M
16
RC
inflammed
A1b, L3, B1, G0
45
Illumina









(Severe)


HMC081RC
CD
M
11
RC
normal
A1b, L1, B1, G1
67.5
Illumina









(Severe)


HMC082RC
CD
F
15
RC
inflammed
A1a, L3, B1, G0
27.5 (Mild)
Illumina


HMC085RC
CD
M
16
RC
normal
A1b, L2, B1, G1
62.5
Illumina/454pyro









(Severe)


HMC086RC
CD
M
8
RC
normal
A1a, L1, L4a, L4b,
22.5 (Mild)
Illumina








B1, G0


HMC087RC
Control
F
18
RC
normal
n/a
n/a
Illumina/454pyro


HMC088RC
UC
M
16
RC
inflammed
E1, S0
20 (Mild)
Illumina


HMC090RC
CD
M
16
RC
inflammed
A1b, L3, L4, B1, G0
52.5
Illumina/454pyro









(Severe)


HMC091RC
Control
M
12
RC
normal
n/a
n/a
Illumina


HMC092RC
UC
M
12
RC
normal
E3, S1
80
Illumina/454pyro









(Severe)


HMC093RC
CD
M
12
RC
normal
A1b, L1, B1p, G0
27.5 (Mild)
Illumina/454pyro


HMC094RC
CD
M
11
RC
normal
A1b, L1, B1p, G0
45
Illumina









(Severe)


HMC095RC
CD
M
11
RC
normal
A1b, L3, B1p, G1
65
Illumina









(Severe)


HMC097RC
CD
M
12
RC
inflammed
A1b, L3, L4a,
40
454Pyro








B1, G0
(Severe)


HMC098RC
Control
F
16
RC
normal
n/a
n/a
Illumina


HMC100RC
Control
M
15
RC
normal
n/a
n/a
Illumina


HMC102RC
Control
F
17
RC
normal
n/a
n/a
Illumina


HMC103RC
UC
F
17
RC
inflammed
E4, S0
50
Illumina









(Moderate


HMC106DC
Control
F
16
DC
normal
n/a
n/a
qPCR/qRTPCR


HMC109DC
Control
F
3
DC
normal
n/a
n/a
qPCR/qRTPCR


HMC112DC
Control
M
9
DC
normal
n/a
n/a
qPCR/qRTPCR


HMC113DC
UC
M
15
DC
inflammed
E2, S1
0
qRTPCR









((Inactive))


HMC117DC
Control
F
15
DC
normal
n/a
n/a
qRTPCR


HMC201RC
CD
F
11
RC
inflammed
A1b, L2, B1, G1
37.5
Illumina/Massspec/









(Moderate
qPCR/qRTP


HMC202RC
CD
M
17
RC
inflammed
A1b, L3, B1, G0
37.5
Illumina/Massspec/









(Moderate
qPCR/qRTP


HMC203RC
CD
M
11
RC
inflammed
A1b, L3, B1, G0
45
Illumina/Massspec/









(Severe)
qPCR/qRTP


HMC204RC
CD
M
13
RC
normal
A1b, L1, L4b, B1,
45
Illumina/qRTPCR








G1
(Severe)


HMC205RC
CD
M
13
RC
normal
A1b, L1, L4a, B1,
45
Illumina/qPCR/qRTPCR








P, G1
(Severe)


HMC206RC
CD
M
14
RC
inflammed
A1b, L3, B1, G1
45
qRTPCR









(Severe)


HMC207RC
UC
F
13
RC
inflammed
E4, S1
70
Illumina









(Severe)


HMC208RC
Control
F
16
RC
normal
n/a
n/a
Illumina/qPCR/qRTPCR


HMC210RC
CD
F
17
RC
normal
A1b, L1, B1, G0
37.5
qRTPCR









(Moderate


HMC211RC
control
M
13
RC
normal
n/a
n/a
qRTPCR


HMC212RC
control
F
15
RC
normal
n/a
n/a
qRTPCR


HMC213RC
CD
M
10
RC
inflammed
A1b, L3, B1, G1
60
qPCR/MassSpec









(Severe)


HMC214RC
UC
M
17
RC
inflammed
E3, S0
35
qRTPCR









(Moderate


HMC215RC
UC
F
12
RC
inflammed
E4, S1
65
qRTPCR









(Severe)


HMC217RC
CD
F
14
RC
normal
A1b, L1, L4a,
55
qPCR/qRTPCR








B1, G0
(Severe)


HMC219RC
CD
M
14
RC
inflammed
A1b, L3, B1, G1
37.5
MassSpec/qRTPCR









(Moderate


HMC220RC
CD
M
13
RC
inflammed
A1b, L3, B1, G0
60
MassSpec/qRTPCR









(Severe)


HMC221RC
control
F
9
RC
normal
n/a
n/a
qPCR/qRTPCR


HMC222RC
control
M
10
RC
normal
n/a
n/a
qPCR/qRTPCR


HMC223RC
CD
F
9
RC
inflammed
A1a, L2, B1, G1
45
MassSpec/qRTPCR









(Severe)


HMC224RC
control
M
15
RC
normal
n/a
n/a
qPCR/qRTPCR/










MassSpec


HMC225RC
control
F
15
RC
normal
n/a
n/a
qRTPCR


HMC227RC
CD
F
13
RC
inflammed
A1b, L3, B1, G0
32.5
qPCR/qRTPCR/









(Moderate
MassSpec


HMC228RC
CD
M
13
RC
inflammed
A1b, L3, B1, P,
62.5
MassSpec/qRTPCR








G0
(Severe)


HMC229RC
CD
M
10
RC
inflammed
A1b, L2, L4a,
57.5
qPCR/qRTPCR








B1, G0
(Severe)


HMC230RC
CD
M
14
RC
inflammed
A1b, L3, L4a,
52.5
qPCR/qRTPCR








B1, P, G0
(Severe)


HMC231RC
CD
F
15
RC
normal
A1b, L1, B1, G0
37.5
qPCR/qRTPCR









(Moderate


HMC232RC
control
F
15
RC
normal
n/a
n/a
qPCR/qRTPCR/










MassSpec


HMC234RC
CD
F
13
RC
inflammed
A1b, L3, B1, G0
52.5
qRTPCR









(Severe)


HMC235RC
control
M
11
RC
normal
n/a
n/a
qRTPCR


HMC237RC
control
M
14
RC
normal
n/a
n/a
MassSpec/qRTPCR


HMC238RC
Control
M
14
RC
normal
n/a
n/a
qPCR/qRTPCR


HMC239RC
CD
M
3
RC
infamed
A1a, L3, B1p,
22.5 (Mild)
MassSpec/qRTPCR








GO


HMC240RC
CD
M
10
RC
normal
A1b, L1, B1, G0
55
qRTPCR









(Severe)


HMC241RC
Control
F
12
RC
normal
n/a
n/a
qRTPCR


HMC243RC
UC
M
8
RC
inflammed
E1, S0
45
qRTPCR









(Moderate


HMC245RC
Control
M
16
RC
normal
n/a
n/a
qRTPCR


HMC246RC
Control
M
13
RC
normal
n/a
n/a
qRTPCR


HMC247RC
UC
F
13
RC
inflammed
E4, S1
80
qRTPCR









(Severe)


HMC249RC
UC
M
14
RC
inflammed
E4, S0
45
qRTPCR









(Moderate


HMC252RC
CD
M
9
RC
inflammed
A1a, L3, B1, G0
37.5
MassSpec









(Moderate


HMC253RC
CD
F
11
RC
inflammed
A1b, L2, L4a, B1
40
qPCR









(Severe)


HMC254RC
CD
M
12
RC
normal
A1a, L1, L4a,
27.5 (Mild)
MassSpec








B2B3


HMC256RC
CD
F
10
RC
inflammed
A1b, L2, B1
30
MassSpec









(Moderate


HMC258RC
Control
M
15
RC
normal
n/a
n/a
MassSpec


HMC260RC
CD
M
13
RC
inflammed
A1b, L2, B1p
47.5
qPCR









(Severe)


HMC261RC
CD
F
13
RC
normal
A1b, L3, L4, B1p
32.5
qPCR









(Moderate


HMC264RC
CD
F
15
RC
normal
A1bL3L4aB1G0
30
qPCR









(Moderate


HMC266RC
CD
F
10
RC
inflammed
A1a, L2, B1
27.5 (Mild)
qPCR


HMC269RC
CD
M
8
RC
normal
A1a, L3, L4a, B1
2.5
qPCR









(Inactive)


HMC270RC
Control
F
17
RC
normal
n/a
n/a
qPCR


HMC271RC
CD
M
15
RC
inflammed
A1a, L3, B1p,
62.5
qPCR/MassSpec








GO
(Severe)


HMC272RC
CD
M
12
RC
inflammed
A1a, L3, B1p,
50
MassSpec








G1
(Severe)


HMC280RC
CD
M
15
RC
inflammed
A1b, L3, B1
35
MassSpec









(Moderate


HMC281RC
Control
F
15
RC
normal
n/a
n/a
MassSpec


HMC285RC
CD
F
9
RC
inflammed
A1a, L3, B1, G0
17.5 (Mild)
qPCR


HMC286RC
CD
M
15
RC
inflammed
A1b, L3, L4a, B1
5
qPCR









(Inactive)


HMC288RC
CD
M
14
RC
inflammed
A1b, L3, B1p
62.5
qPCR/MassSpec









(Severe)


HMC289RC
CD
M
12
RC
inflammed
A1b, L3, B1p
67.5
qPCR









(Severe)


HMC293RC
CD
F
12
RC
inflammed
A1b, L2, B1, G1
22.5 (Mild)
qPCR


HMC295RC
CD
F
14
RC
inflammed
L2, B1, G0
20 (Mild)
qPCR


HMC297RC
Control
F
17
RC
normal
n/a
n/a
qPCR


HMC298RC
CD
M
12
RC
normal
A1b, L1, B1
52.5
qPCR









(Severe)


HMC300RC
CD
M
10
RC
inflammed
A1a, L3, L4a,
27.5 (Mild)
MassSpec








B1, G1


HMC301RC
CD
F
14
RC
inflammed
A1b, L3, L4a,
60
MassSpec








B1, G0
(Severe)


HMC305RC
CD
M
4
RC
normal
A1a, L3, L4a,
12.5 (Mild)
qPCR








B2p, G0


HMC307RC
Control
F
16
RC
normal
n/a
n/a
qPCR/MassSpec


HMC309RC
Control
M
10
RC
normal
n/a
n/a
qPCR/MassSpec


HMC313RC
Control
M
16
RC
normal
n/a
n/a
qPCR


HMC315RC
Control
M
16
RC
normal
n/a
n/a
MassSpec


HMC316RC
CD
M
14
RC
inflammed
A1b, L3, L4a, B1
17.5 (Mild)
qPCR/MassSpec


HMC317RC
CD
M
15
RC
normal
A1b, L1, L4a,
0
qPCR








B1p
(Inactive)


HMC319RC
CD
M
12
RC
normal
L1, B1, G1
40
qPCR









(Severe)


HMC321RC
Control
F
16
RC
normal
n/a
n/a
qPCR


HMC322RC
CD
M
12
RC
inflammed
A1bL3B1P
27.5 (Mild)
qPCR


HMC323RC
CD
F
16
RC
inflammed
A1bL3B1G0
35
qPCR









(Moderate


HMC327RC
CD
F
10
RC
inflammed
A1aL2B1
25 (Mild)
MassSpec









The microbiota at the intestinal mucosal interface embedded within the mucus layer and in direct contact with the site of disease was collected, and the microbial composition was characterized. The IBD microbiota was characterized by a smaller core as compared to controls (FIG. 5A, 5B and Table 1), indicating a loss of microbiota homeostasis. In addition, two taxa from the core microbiota that are potent H2S producers, Fusobacterium nucleatum and Veillonella parvula were found to be more abundant in CD and UC patients, respectively, as compared to controls (P<0.013; Table 1). Notably, F. nucleatum has been previously associated with adult IBD and shown to promote tumorigenesis in Apcmin/+ mice4-6. To identify microbes associated with IBD, the microbial taxa composition of CD, UC and control communities were compared using a nonparametric Kruskal-Wallis test, which indicated significant discriminating factors in 48 taxa (P<0.015; Supplementary Table 5). The relative abundances of Firmicutes, Clostridia, Clostridiales, and Lachnospiraceae, which are the major producers of butyrate, were decreased in CD and UC as compared to control microbiota while the relative abundances of Negativicutes, Selenomonadales, Veillonella and Betaproteobacteria were increased (Table 2). Taxa associated with disease activity were identified to identify the microbes modulating IBD severity. A Kruskal-Wallis test identified 11 unique taxa exhibiting differential abundance between CD patients with mild, moderate or severe inflammation (Table 3). Partial least square discriminant analysis (PLS-DA) clustered CD patients with severe inflammation separately from those with mild inflammation (FIG. 1a; p=0.001). Taxa biplot analysis indicated that certain taxa namely, Carnobacteriaceae, Granulicatella, Mogibacterium, Proprionibacterium, Bacillaceae and Atopobium were more abundant in patients with severe as compared to mild inflammation (FIG. 1b). In contrast, Clostridia, which are major butyrate-producers, were drivers of mild inflammation and exhibited a significant decline in relative abundance with increased disease severity. Atopobium was further classified as Atopobium parvulum (OTU#529659), a potent H2S producer implicated in halitosis7,8. H2S is now recognized as an important mediator of many physiological and pathological processes and has been associated with IBD and colorectal cancer9,10. Sulfide inhibits butyrate metabolism, the major energy source for colonocytes, and cytochrome c oxidase activity, the site of ATP production9. Therefore, a higher concentration of H2S might severely impair cellular bioenergetics. This would induce colonocyte starvation and death, disrupt the epithelial barrier and potentially lead to inflammation.


The relative abundance of A. parvulum was validated by quantitative polymerase chain reaction (qPCR) and found to be positively correlated with disease severity (FIG. 1c). The correlation between A. parvulum and CD was also confirmed by 454 pyrosequencing of a subset of samples followed by linear discriminant analysis effect size11 (LEfSe; Supplementary FIG. 2-3). Importantly, the increased abundance of A. parvulum was not observed in UC and therefore is not simply a consequence of inflammation, suggesting a causal effect in CD.


To evaluate the colitogenic potential of A. parvulum, we utilized colitic-susceptible II10−/− mice12,13. Germ-free II10−/− mice were transferred to specific pathogen free (SPF) housing and gavaged with A. parvulum (108 CFU/mouse/week) for 6 weeks. Compared to control uninfected II10−/− mice, A. parvulum-colonized II10−/− mice showed macroscopic evidence of cecal atrophy and colon length reduction (FIG. 2a). Colonoscopy imaging revealed mucosal erythema, friability and mucosal ulceration in A. parvulum-colonized II10−/− mice compared to the healthy mucosa observed in controlled mice (FIG. 2b). Histological assessment of the intestinal tract showed evidence of inflammation with crypt hyperplasia, ulcers, goblet cell depletion and immune cell infiltration observed in the cecum and the distal part of the colon of A. parvulum-associated II10−/− mice compared to uninfected control II10−/− mice (FIG. 2c). Accordingly, histologic inflammation scores were significantly higher in A. parvulum associated II10−/− mice compared to uninfected mice (P<0.05; FIG. 2d). At the molecular level, the colon of A. parvulum-infected II10−/− mice showed increased Cxcl1 and II17 mRNA accumulation compared to uninfected II10−/− mice (fold increases of 8 and 5 respectively; P<0.01). These results indicate that the H2S-producing bacterium A. parvulum induces pancolitis in a genetically susceptible mouse model of IBD.


To gain mechanistic insights into the role of H2S-producing microbes in IBD severity, an unbiased, quantitative proteomic analysis of mucosal biopsies of IBD subjects of various disease severity (n=21) and controls (n=8) was conducted. Measurements for 3880 proteins were obtained of which 490 were identified as differentially expressed by comparing the 3 major groups, severe vs. moderate vs. control (one-way ANOVA with P<0.05). Mitochondrial proteins were identified as a major discriminant feature representing 21.7% of all differentially expressed proteins (FIG. 3A and FIGS. 8A, 8B and 8C). Proteins driving disease activity were identified by PLS-DA and the analysis of their variable importance projection (VIP) scores (Tables 4 and 5 and FIGS. 7B and 9A and 9B). Notably, components of the mitochondrial hydrogen sulfide detoxification complex (9 and FIG. 10) were found to be the main proteins driving the separation based on disease severity (Table 5). These proteins, namely the sulfur dioxygenase (ETHE1), the thiosulfate sulfur transferase (TST), and the components of complexes III and IV of the mitochondrial respiratory chain, were down-regulated in CD patients compared to controls (P<0.05). Secondary validation by qRT-PCR confirmed the repression of the TST transcript (5 fold decrease, P=0.002) in CD and UC patients (FIG. 3C). Moreover, the expression levels of the cytochrome c oxidase subunit IV and the sulfide dehydrogenase genes (SQR), which also contribute to the detoxification of H2S, were significantly down-regulated in CD and/or UC patients, as measured by qRT-PCR (FIG. 3D-E). These findings indicate that transcriptional regulation contributes to the observed change in protein abundance and that the decreased abundance of these H2S-detoxification proteins is a hallmark of CD disease activity and possibly UC. Importantly, these results would explain the previously observed increase of fecal sulfide levels in IBD patients14.


The findings demonstrate an alteration of the balance between bacterial-derived H2S production and host-mediated detoxification of H2S at the mucosal-luminal interface. To test the causative role of H2S-producing microbes in colitis, we assessed whether an H2S scavenger (bismuth) could alleviate Atopobium-induced colitis in II10−/− mice. Consistent with the first cohort, Atopobium-associated SPF mice developed severe colitis (FIG. 4A-B) and exhibited significant increases of pro-inflammatory cytokine expression (FIG. 11A-D). Treatment with bismuth prevented colitis as evidenced by colonoscopy visualization (FIG. 4A) and by decrease inflammatory score (P=0.007; FIG. 4B). Atopobium-associated mice also exhibited an increased number of GALT (gut associated lymphoid tissue) foci as compared to non-associated mice (FIG. 4C; P=0.012). However bismuth treatment did not prevent GALT formation indicating that A. parvulum induces GALT neogenesis in II10−/− mice. It should be noted that the intestines of IBD patients also display a similar increased number of lymphoid follicles15. Interestingly, elimination of GALT with LTβR-Ig treatment protects mice from developing colitis suggesting a role for GALT formation in the development of chronic intestinal inflammation16. Increased GALT foci in Atopobium-associated mice could lead to an aberrant expression of lymphoid adhesion molecules and unwanted T cell activation towards commensal microbes. To assess the role of these commensal microbes in colitis development, germ-free mice were mono-associated with A. parvulum and kept under gnotobiotic conditions. While these mice showed crypt hyperplasia and increased GALT foci (FIG. 12A and FIG. 4C), they had no signs of ulcerations, goblet cell depletion or immune cell infiltration (FIG. 4D). This result indicates that the gut microbiota is required for the development of Atopobium-induced colitis. While bismuth treatment prevented GALT neogenesis in mice mono-associated with A. parvulum, the observed effect might not necessarily be due to H2S scavenging but instead due to a potential antimicrobial activity of bismuth on A. parvulum, as evidenced by a reduced colonization level (P=0.0001; FIG. 12B). Because both A. parvulum and the gut microbiota are required for colitis-development and because bismuth exhibits antimicrobial properties17, we assessed the effect of bismuth on the gut microbiota composition of our SPF and Atopobium-associated mice. PCA analysis of the gut microbiota composition revealed a significant alteration in the microbial profile of the Atopobium-associated mice as compared to the SPF mice (FIG. 4E). Concomitantly to colitis prevention, bismuth administration altered the microbiota composition of these 2 groups of mice (Table 6). Altogether, these results indicate that (1) A. parvulum colonization altered the composition of the gut microbiota (with a significant decrease in abundance of the major butyrate-producers including Eubacterium and Faecalibacterium (P<0.02)) analogously to the microbiota composition of pediatric IBD patients; (2) the aberrant composition of the gut microbiota in Atopobium-associated mice is a major inducer of colitis; and (3) bismuth restores the microbiota of these mice toward a healthier community (with an increase abundance of butyrate-producers).


Collectively the findings shed light on the pathogenic mechanisms of early IBD onset. The emerging picture is that the pediatric IBD microbiota is characterized by a depletion in butyrate producing microbes together with an increased abundance of H2S-generating bacteria, namely A. parvulum, Fusobacterium and Veillonella, which produce H2S by protein fermentation18. Because IBD patients exhibit increased levels of fecal H2S14, sulfate-reducing bacteria (SRB) have long been proposed to be involved in the etiology of IBD, although studies have failed to demonstrate a link between SRB and IBD10. Instead, our study demonstrates a key role for microbes producing H2S through protein fermentation in CD pathogenesis. Butyrate is known to activate the expression of the genes encoding the host mitochondrial H2S detoxification components19 and our proteomic analyses indicate a diminished capacity for H2S detoxification by IBD patients. Therefore, we postulate that the depletion of butyrate-producing microbes from the gut microbiota would disable the host H2S defense systems. This “disarmed” host would be highly susceptible to further damage caused by enhanced H2S production, resulting in metabolic stress and subsequently increased mucosal inflammation. Interestingly, variants in mitochondrial DNA, which result in increased metabolic activities, protect mice from colitis20. This is in agreement with the important role of the mitochondria in modulating the mucosal barrier. More recently, excess H2S has been shown to act as an autocrine T-cell activator, potentially contributing to unwanted T-cell responses against commensal bacteria21, consistent with our observation that the gut microbiota is required for A. parvulum-induced experimental colitis. Given the essential role of butyrate in regulating regulatory T cells (Treg) homeostasis and the critical role of Treg in limiting intestinal inflammation22, H2S production may also interfere with this process by impairing butyrate oxidation and thus might lead to increased colitis severity. This result emphasises the importance of the microbial community and its interaction with the host in the pathogenesis of IBD. Altogether, our findings provide new avenues for diagnostics as well as therapies to treat IBD.


Methods
Example 2

Colonic mucosal lavages and/or mucosal biopsies were collected from 157 pediatric subjects (84 Crohn's Disease, 20 Ulcerative Colitis, and 53 controls). All IBD cases were newly diagnosed with IBD and met the standard diagnostic criteria for either CD or UC. Metagenomic DNA from the intestinal lavages was extracted using the FastDNA SPIN Kit. Microbial communities were surveyed by deep sequencing the 16S rRNA V6 hypervariable region using Illumina HiSeq2500 and 454-Pyrosequencing. Reads were quality filtered and QIIME23 was used to assign reads into operational taxonomic units (OTUs) against the Greengenes reference set. Several statistical approaches (Kruskal-Wallis tests, LEFSe, PCA, PLS-DA) were used to determine differentially abundant OTUs. The correlation between A. parvulum relative abundance and CD severity was confirmed by qPCR. Proteomic analysis of mucosal biopsies was conducted using super-SILAC-based HPLC-ESI-MS/MS. The generated raw data was processed and analyzed by MaxQuant against the decoy Uniport-human database with the protein-group file imported into Persus for statistical analysis. Pathway analysis was done using the DAVID Bioinformatics Resources. The transcript levels of TST, SQRDL and COX4-1 were quantified by RT-qPCR. Gnotobiotic and specific pathogen free II10−/− mice were gavaged once weekly with A. parvulum (108 cfu) for 6 weeks. Bismuth (III) subsalicylate (7 g/kg) was added to the diet of the assigned groups one week before the gavage. Tissue samples from the colon were collected for RNA and histology as described previously24 Mouse colonoscopies were performed and histological inflammation was blindly scored as previously described25. Mice mucosal cytokines (Cxcl1, II12p40, II1β and II17a) were quantified by RT-qPCR.


Example 3

Patient Cohort and Study Design:


This study involved the enrollment, detailed assessment, and biological sampling of 157 pediatric subjects (84 CD, 20 UC, and 53 controls; Table 7). All patients under 18 years of age scheduled to undergo their first diagnostic colonoscopy at the Children's Hospital of Eastern Ontario (CHEO) were potentially eligible for recruitment to this study, with the following exclusions which are known to affect the gut microbiota composition: (1) body mass index (BMI) greater than 95th percentile for age; (2) diagnosis with diabetes mellitus; (3) diagnosis with infectious gastroenteritis within the preceding 2 months; and (4) use of any antibiotics or probiotics within the last 4 weeks. All cases were newly diagnosed with IBD (inception cohort prior to the initiation of treatment) and met the standard diagnostic criteria for either Ulcerative Colitis or Crohn's Disease following thorough clinical, microbiologic, endoscopic, histologic and radiologic evaluation26; most had active inflammatory luminal disease involving the terminal ileum and/or the colon+/−perianal disease. Phenotyping of disease was based on endoscopy and clinical disease activity scores. The Simplified-Endoscopy Score-Crohn's disease was used to record macroscopic activity in each segment of the intestinal tract in Crohn's disease27, the site of involvement in CD was recorded utilizing the Paris IBD Classification28 and clinical disease activity of CD was determined using the Pediatric Crohn's Disease Activity Index (PCDAI)29. For UC, the site of disease was recorded using the Paris Classification system28, endoscopic activity was recorded using the Mayo Score Flexible Proctosigmoidoscopy Assessment in ulcerative colitis and clinical activity of UC was determined using and Pediatric Ulcerative Colitis Activity Index (PUCAI)30. The clinical activity scores are both validated for use in Pediatric IBD. All controls had a macroscopically and microscopically normal colon, and did not carry a diagnosis for any known inflammatory intestinal disorder and did not have a well-defined infectious etiology for the bowel inflammation. Data collected on all participants included: demographics (age, gender, BMI, country of birth, age of diagnosis), environmental exposures (cigarette smoke, diet, previous antibiotic exposure), and all clinical features. This study was performed in compliance with the protocol approved by the Research Ethic Board of the Children's Hospital of Eastern Ontario.


Example 4

Biopsies and Mucosa-Luminal Sample Collection:


Mucosal-luminal interface samples were collected from the right colon at the time of endoscopy. Colonoscopy preparation was done the day before the procedure as per standard protocol31. During endoscopy, once the correct position is reached, loose fluid and debris was aspirated. Thereafter sterile water was flushed onto the mucosa and the collection of water, mucus and intestinal cells of the colonic mucosa was aspirated into sterile container through the colonoscope. These samples were immediately place on ice in the endoscopy suite, promptly transferred to the lab to minimize delay for processing and then storing at −80° C. Up to 2 biopsies were collected from macroscopically involved area of the right colon. Biopsies were flash frozen on dry-ice in the endoscopy suite and immediately stored at −80° C. until further processing.


Example 5

Microbiota DNA Extraction and Sequencing of 16S rDNA Amplicons:


Metagenomic DNA was extracted from the mucosa-luminal samples using the Fast DNA SPIN Kit (MP Biomedicals) and the FastPrep machine (MP Biomedicals) with two mechanical lysis cycles at speed 6.0 for 40 seconds. Extracted DNA was then used for the construction of the sequencing libraries.


Two sequencing-by-synthesis platforms were used in this study: (1) pyrosequencing (Roche 454 GS-FLX) and (2) Illumina Hiseq 2500. Samples for both sequencing techniques were PCR amplified to target the same V6 hypervariable region. Samples for sequencing by Roche 454 were independently sequenced using the FLX chemistry on 12 lanes of a 16-lane sub-divided 454 FLX PicoTiter plate (70×75 mm) and using a total of 3 plates. The 454 amplicons libraries were constructed using the conserved V6 primers pair 16S-V6_907-F (5′-AAACTCAAAKGAATTGACGG-3′) (SEQ ID NO. 16) and 16S-V6_1073-R (5′-ACGAGCTGACGACARCCATG-3′)32 (SEQ ID NO. 17). The hypervariable V6 region of 16S rDNA gene was amplified using two successive PCR reactions to reduce PCR bias as previously described33. The first PCR used 16S-V6 specific primers and the 2nd PCR involved 454 fusion-tailed primers. In the first PCR, ten amplicons were generated from each extracted DNA sample. Each PCR reaction contained 2 μL DNA template, 17.5 μL molecular biology grade water, 2.5 μL 10× reaction buffer (200 mM Tris-HCl, 500 mM KCl, pH 8.4), 0.5 μL dNTPs (10 mM), 1 μl 50 mM MgCl2, 1 μL of both forward and reverse primers (10 mM each) and 0.5 μL Invitrogen's Platinum Taq polymerase (5 U/μL) in total volume of 25 μL. The PCR conditions were initiated with heated lid at 95° C. for 5 min, followed by a total of 15 cycles of 94° C. for 40 sec, 48° C. for 1 min, and 72° C. for 30 sec, and a final extension at 72° C. for 5 min, and hold at 4° C. Amplicons generated from each sample were pooled and purified to remove the excess unused primers using Qiagen's MiniElute PCR purification columns and eluted in 30 μL molecular biology grade water. The purified amplicons from the first PCR were used as templates in a second PCR with the same amplification conditions used in the first PCR with the exception of using 454 fusion-tailed primers in a 30-cycle amplification regime. An Eppendorf Mastercycler ep gradient S thermalcycler was used in all PCRs. A negative control reaction (no DNA template) was included in all experiments. PCR success was checked by agarose gel electrophoresis. The 16S-V6 amplicon of each sample was quantified by fluorometer and purified with AMPure magnetic beads. The amplicon libraries were sequenced on a 454 Genome Sequencer FLX System (Roche Diagnostics GmbH) following the amplicon sequencing protocol. Amplicons of each sample was bi-directionally sequenced in 1/16th of full sequencing run (70×75 picotiter plate).


For samples to be sequenced by Illumina Hiseq 2500, the V6 hypervariable region of the 16S rDNA gene was amplified using two successive PCR reactions as described previously34. The universal 16S rDNA-V6 primers for the first PCR step were modified from Sundquist et al32 to include the Illumina paired-end sequencing adapters, and a 4-6 nucleotide barcode sequence (Supplementary Table 10). Each PCR reaction was performed in a total volume of 50 μL using 50 ng of the extracted DNA, 1× Phusion HF PCR buffer, 0.5 μM of each primer, 0.2 mM dNTPs, and 1 U Phusion High-Fidelity DNA polymerase (Thermo Scientific). The PCR conditions included initial denaturation at 94° C. for 30 s, 10 cycles of 94° C. for 10 s, 61° C. for 10 s with a 1° C. drop each cycle and 72° C. for 15 s followed by an additional 15 cycles using an annealing temperature of 51° C. for 45 s, and a final extension at 72° C. for 2 min. The second PCR was carried out using 10 μL of the first PCR products in a final volume of 50 μL using the primers PCRFWD1/PCRRVS1 (Supplementary Table 10). The second PCR conditions were 30 s at 94° C., 15 cycles of 10 s at 94° C., 10 s at 65° C., and 15 s at 72° C. followed by a final extension step at 72° C. for 2 min. The amplicons of each sample were visualized on a 1.5% agarose gel and purified using the Montage PCR96 Cleanup Kit (Millipore). Next, the DNA concentration in each reaction was quantified using the Qubit® dsDNA BR Assay Kit (Invitrogen) following the manufacturer instructions and 100 ng of amplicons from each sample were pooled. Finally, the library consisting of the pooled amplicons was gel purified using the QIAquick Gel Extraction Kit (Qiagen), quantified and subjected to Illumina HiSeq 2500 sequencing at The Center for Applied Genomics (TCAG, Toronto) generating paired-end reads of 2×100 bases.


Example 6

Microbiota Analysis:


454 Pyrosequencing Data Analysis:


A total of 346,160 reads were generated from 454 pyrosequencing of 16S rDNA-V6 region from 26 right colon samples. The generated reads were submitted to NCBI Sequence Read Archive under accession number SRP034632. The raw sequences were processed to remove low quality and short reads using Quantitative Insights Into Microbial Ecology pipeline release 1.4.0 (QIIME 1.4.0)23 according to the following parameters: (1) Minimum read length of 100 bp, (2) Exact matching to the sequencing primers, (3) No ambiguous nucleotides, and (4) The minimum average quality score of 20. This resulted in a total of 266,006 high quality reads with an average of ˜10,224 sequences per sample and a mean length of 169.58 bases including the primers. Next, sequences were clustered into operational taxonomic units (OTUs) using UCLUST based on average percentage of identity of 97%. The most abundant read from each OTU was picked as a representative sequence for that cluster, while singletons were discarded. PyNAST was used to align the representative sequences with a minimum alignment length of 100 and a minimum percentage identity of 75%, followed by identification of chimeric OTUs with the Blast Fragments Algorithm implemented in QIIME. Only 6 representative sequences were identified as chimeras and therefore were removed from the aligned representative set. Taxonomy assignments were made with BLAST by searching the representative sequences against the Greengenes database (release 4 Feb. 2011) with an e value of 1e-8 and a confidence score of ≥0.5. The resulting OTU table was used to determine the alpha and beta diversity within and between the samples using the default criteria of QIIME. Taxa significantly associated with disease status (CD, UC and control) or disease severity (mild, moderate and severe) were identified using the linear discriminant effect size (LEFSe) algorithm (http://huttenhower.org/galaxy/)11. To assign taxonomy at the species level, representative reads from OTUs of interest were retrieved from QIIME and aligned against the NCBI and RDB databases35,36.


Illumina Sequencing Data Analysis:


Paired-end sequences obtained by Illumina HiSeq 2500 (2×101 nucleotides) were merged into longer reads (with an average length per sequence of 165 nucleotides) using Fast Length Adjustment of Short reads (FLASh) software avoiding any mismatch in the overlap region that ranges from 20 to 80 nucleotides37. More than 95% of the reads was merged successfully, while the sequences that failed to merge were discarded. The merged reads were then quality filtered with a minimum quality score of 20 using the fastq_quality_filter command from the Fastx toolkit (http://hannonlab.cshl.edu/). High quality reads were sorted according to the forward and the reverse barcode sequences with barcodes trimming using the NovoBarCode software (Novocraft.com). Sequences with mismatched primers were excluded. The sorted reads were submitted to NCBI Sequence Read Archive under accession number SRP034595. Next, the reads were fed to QIIME 1.5.023 pipeline and clustered into OTUs using a closed-reference OTU picking workflow with UCLUST against the Greengenes reference set (release 4 Feb. 2011) based on average percentage of identity of 97%. The OTUs were assigned the taxonomy associated with the corresponding Greengenes reference sequence. Singletons and doubletons were removed and a table of OTU counts per sample was generated. Next, the OTU table was randomly subsampled to a total number of reads per sample of 500,000. The resulting rarefied OTU table was used to analyze the microbiota structure and diversity using the microbial ecology tools available in the QIIME package and for all other downstream analyses. For the identification of the core microbiota, OTUs detected in at least 75% of the samples within a clinical group (CD patients, UC patients or control subjects) were considered as members of the core microbiota for that particular group.


Multivariate Statistical Analysis:


Several statistical approaches were employed to identify taxa significantly associated with disease status and severity. A Kruskal-Wallis test with post hoc Dunn's test was performed to compare the relative abundance of taxa as a function of disease status (CD vs. UC vs. control) and disease severity (mild vs. moderate vs severe). A Bonferroni correction was employed to account for multiple hypotheses with a P<0.05 considered significant. The relative abundances of the taxa identified were also analyzed by principal component analysis (PCA) and partial least square discriminant analysis (PLS-DA). For PLS-DA calculation the data were log-transformed and scaled to unit variance as described in Durbin et al.38. The PLS-DA models were validated by cross-validation and permutation tests. The variable importance in projection (VIP) value was used to identify features which contribute the most to the clustering (taxa with VIP>1.0 were considered influential and with VIP>1.5 highly influential). All statistical analyses were performed using XLSTAT and/or R software package.


Example 7


Atopobium parvulum qPCR Quantification:


The relative abundance of A. parvulum was determined by conducting absolute quantitative PCR on the extracted metagenomic DNA using the Applied Biosystems 7300 DNA analyzer and A. parvulum-specific 16S rRNA primers developed for the current study; Aparv-711F 5′-GGGGAGTATTTCTTCCGTGCCG-3′ (SEQ ID NO. 1) and Aparv-881R 5′-CTTCACCTAAATGTCAA GCCCTGG-3′ (SEQ ID NO. 2). Each sample was tested in duplicate in a total volume of 25 μL per reaction. 100 ng of template DNA was added to a reaction mixture containing 1 μM of each primer, and 1× QuantiFast SYBR Green PCR master mix (Qiagen). The amplification conditions were 5 min at 95° C. followed by 40 cycles of 95° C. for 10 sec and 66° C. for 1 min with data collection at the second step of each cycle. To normalize between samples, the total 16S rRNA in each sample was simultaneously quantified using the universal primers; 331F 5′-TCCTACGGGAGGCAGCAGT-3′ (SEQ ID NO. 18) and 797R 5′-GGACTACCAGGGTATCTAATCCTGTT-3′ 39 (SEQ ID NO. 19). The positive standards for A. parvulum and the total 16S rRNA quantification were prepared by conducting PCR on the DNA extracted from A. parvulum ATCC 33793 strain and one mucosal aspirate sample from a healthy subject, respectively. The amplicons were purified using PureLink™ PCR Purification Kit (Invitrogen) and quantified by Qubit® dsDNA BR Assay Kit (Invitrogen). Afterward, 106, 107, 108 and 109 copies from each gene fragment were prepared, assuming the average molecular weight of the base pair is 660, and the Ct values were determined for each concentration by qPCR following the same conditions described above. The standard curves of both A. parvulum and the total 16S rRNA gene copy numbers against Ct values were established and the relative abundance of A. parvulum in each sample was calculated as A. parvulum 16S-rRNA divided by the total 16S-rRNA copy number. To validate the specificity of Apar-711F and Aparv-881R, fresh PCR amplicons from the total DNA extracted from two different mucosal aspirates was cloned using TOPO TA cloning kit (Invitrogen) according to the manufacturer's instructions, and then, the plasmid containing the 16S rRNA gene fragment was extracted from 6 different clones by QIAprep Spin Miniprep kit (Qiagen) using its standard protocol followed by Sanger sequencing using M13F and M13R primers.


Example 8

Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC):


Human hepatic HuH7 cells (HuH-7), human embryonic kidney 293 cells (HEK-293) and human colorectal cancer 116 cells (HCT-116) were individually grown at 37° C. in a 5% CO2 humidified incubator. SILAC medium was prepared as follows: DMEM lacking lysine, arginine and methionine was custom prepared by AthenaES (Baltimore, Md., USA) and supplemented with 30 mg/L methionine (Sigma Aldrich; Oakville, ON, CAN), 10% (v/v) dialyzed FBS (GIBCO-Invitrogen; Burlington, ON, CAN), 1 mM sodium pyruvate (Gibco-Invitrogen), 28 μg/mL gentamicin (Gibco-Invitrogen), and [13C6, 15N2]-L-lysine, [13C6, 15N4]-L-arginine (heavy form of amino acids; Heavy Media) from Sigma Aldrich (Oakville, ON, CAN) at final concentrations of 42 mg/L and 146 mg/L for arginine and lysine respectively. For HCT-116, the concentration of arginine was increased to 84 mg/L. Cells were grown for at least 10 doublings in SILAC media to allow for complete incorporation of the isotopically labeled amino acids into the cells.


Example 9

Determination of the Rate of SILAC Amino Acids Incorporation into HuH-7, HEK-293 and HCT-116 Cells:


Cells were grown to 80% confluency in SILAC medium (5×106 cells were plated in 10-cm dish). Next, the cells were washed twice with ice-cold phosphate-buffered saline and lyzed by addition of 1 mL of 1×RIPA buffer (50 mM Tris (pH 7.6), 150 mM NaCl, 1% (v/v) NP-40, 0.5% (w/v) deoxycholate, 0.1% (w/v) SDS with protease inhibitor cocktail (Complete Mini Roche; Mississauga, ON,CAN) and phosphatase inhibitor (PhosStop Roche tablet). The lysates were then transferred to 15 mL conical tubes and the proteins were precipitated by addition of 5 mL ice-cold acetone followed by incubation at −20° C. overnight. Proteins were collected by centrifugation (3000×g, 10 min, 4° C.), washed with ice-cold acetone two times, and the protein pellets were resolubilized in 300 μL of a 50 mM NH4HCO3 solution containing 8 M urea. Protein concentrations were determined by the Bradford dye-binding method using Bio-Rad's Protein Assay Kit (Mississauga, ON, CAN). For the general in-solution digestion, 200 pg of protein lysates were reconstituted in 50 mM NH4HCO3 (200 μL) and proteins were reduced by mixing with 5 μL of 400 mM DTT at 56° C. for 15 min. The proteins were then subjected to alkylation by mixing with 20 μL of 400 mM iodoacetamide in darkness (15 min at room temperature) followed by addition of 800 μL of 50 mM NH4HCO3 to reduce the urea concentration to ˜0.8 M. Next, the proteins were digested with TPCK-trypsin solution (final ratio of 1:20 (w/w, trypsin: protein) at 37° C. for 18 h. Finally, the digested peptides were desalted using C18 Sep-Pack cartridges (Waters), dried down in a speed-vac, and reconstituted in 0.5% formic acid prior to mass spectrometric analysis (as described below) and the determination of labeling efficiency. The incorporation efficiency was calculated according to the following equation: (1−1/Ratio(H/L)); where H and L represents the intensity of heavy and light peptides detected by mass-spectrometry, respectively. Labeling was considered complete when values reached at least 95% for each cell type.


Example 10

Proteomic Analysis of Biopsies Using Super-SILAC-Based Quantitative Mass Spectrometry:


Biopsies were lysed in 4% SDS (sodium dodecyl sulfate), 50 mM Tris-HCl (pH 8.0) supplemented with proteinase inhibitor cocktail (Roche) and homogenized with a Pellet pestle. The lysates were sonicated 3 times with 10 s pulses each with at least 30 s on ice between each pulse. Protein concentrations were determined using the Bio-Rad DC Protein Assay. The proteins were processed using the Filter Aided Sample Preparation Method (FASP) as previously described with some modifications40. Colon tissue lysates (45 μg of proteins) and heavy SILAC-labeled cell lysates (15 μg from each HuH-7, HEK-293 and HCT-116 cells) were mixed at a 1:1 weight ratio and transferred into the filter. The samples were centrifuged (16,000×g, 10 min), followed by two washes of 200 μL 8 M urea, 50 mM Tris-HCl pH 8.0. Samples were then reduced by incubation in 200 μL of 8 M urea, 50 mM Tris-HCl (pH 8.0) supplemented with 20 mM dithiothreitol. After centrifugation, samples were subjected to alkylation by adding 200 μL of 8 M urea, 50 mM Tris-HCl pH 8.0, containing 20 mM iodoacetamide (30 min at room temperature protected from light). Samples were washed using 200 μL 8 M urea, 50 mM Tris-HCl pH 8.0 (twice) to remove excess SDS. To further dilute urea, two washes of 200 μL 50 mM Tris-HCl pH 8.0 were performed. For the trypsin digest, samples were incubated in 200 μL of 50 mM Tris-HCl pH 8.0, containing 5 μg of Trypsin (TPCK Treated, Worthington) on a shaker (250 rpm) at 37° C. overnight. Finally, 200 μL of 50 mM Tris-HCl pH 8.0 was added to elute the peptides by centrifugation (twice). Peptides were fractionated, using an in-house constructed SCX column with five pH fractions (pH 4.0, 6.0, 8.0, 10.0, 12.0). The buffer composition was 20 mM boric acid, 20 mM phosphoric acid, and 20 mM acetic acid, with the pH adjusted by using 1 M NaOH). Finally, the fractionated samples were desalted using in-house C18 desalting cartridges and dried in a speed-vac prior to LC-MS analysis.


Mass-Spectrometry Analyses:


All resulting peptide mixtures were analyzed by high-performance liquid chromatography/electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS). The HPLC-ESI-MS/MS consisted of an automated Ekspert™ nanoLC 400 system (Eksigent, Dublin, Calif., USA) coupled with an LTQ Velos Pro Orbitrap Elite mass spectrometer (ThermoFisher Scientific, San Jose, Calif.) equipped with a nano-electrospray interface operated in positive ion mode. Briefly, each peptide mixture was reconstituted in 20 μL of 0.5% (v/v) formic acid and 12 μL was loaded on a 200 μm×50 mm fritted fused silica pre-column packed in-house with reverse phase Magic C18AQ resins (5 μm; 200 Å pore size; Dr. Maisch GmbH, Ammerbuch, Germany). The separation of peptides was performed on an analytical column (75 μm×10 cm) packed with reverse phase beads (3 μm; 120 Å pore size; Dr. Maisch GmbH, Ammerbuch, Germany) using a 120 min gradient of 5-30% acetonitrile (v/v) containing 0.1% formic acid (v/v) (JT Baker, Phillipsburg N.J., USA) at an eluent flow rate of 300 nL/min. The spray voltage was set to 2.2 kV and the temperature of heated capillary was 300° C. The instrument method consisted of one full MS scan from 400 to 2000 m/z followed by data-dependent MS/MS scan of the 20 most intense ions, a dynamic exclusion repeat count of 2, and a repeat duration of 90 s. The full mass was scanned in an Orbitrap analyzer with R=60,000 (defined at m/z 400), and the subsequent MS/MS analyses were performed in LTQ analyzer. To improve the mass accuracy, all the measurements in the Orbitrap mass analyzer were performed with on-the-fly internal recalibration (“Lock Mass”). The charge state rejection function was enabled with charge states “unassigned” and “single” states rejected. All data were recorded with Xcalibur software (ThermoFisher Scientific, San Jose, Calif.).


Database Search and Bioinformatic Analysis:


All raw files were processed and analyzed by MaxQuant, Version 1.2.2.5 against the decoy Uniport-human database (86,749 entries), including commonly observed contaminants. The following parameters were used: cysteine carbamidomethylation was selected as a fixed modification, with methionine oxidation, protein N-terminal acetylation and heavy proline set as variable modifications. Enzyme specificity was set to trypsin. Up to two missing cleavages of trypsin were allowed. SILAC double labeling (light: K0R0; heavy: K8R10) was set as the search parameter in order to assess the conversion efficiency. The precursor ion mass tolerances were 7 ppm and the fragment ion mass tolerance was 0.5 Da for MS/MS spectra. The false discovery rate (FDR) for peptides and proteins was set at 1% and a minimum length of six amino acids was used for peptide identification. The peptides file was imported into Persus (version 1.2.0.17) to extract the lysine- and arginine-containing peptides, respectively.


The protein-group file was imported into Persus (version 1.3.0.4) for data statistical analysis and an ANOVA—test was chosen for the protein profile with p values of less than 0.05 considered significant. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was achieved using the DAVID Bioinformatics Resources (http://david.abcc.ncifcrf.gov/). DAVID statistical analyses were performed against the whole genome. Proteomics has a tendency to oversample proteins from the cytosol and nucleus while under-sampling membrane-associated proteins. Therefore, the results from DAVID were retested against the set of proteins that were not changing in our dataset in order to eliminate any pathway/GO enrichment biases.


Example 11

Total RNA Extraction and qRT-PCR Quantification of Mitochondrial Genes Expression:


RNA integrity was preserved by adding the mucosal aspirates to an equal volume of RNAlater (Ambion) before freezing at −80° C. The frozen aliquot (2 mL) was thawed on ice and the total RNA was extracted following a hot phenol protocol as described previously41. Briefly, 4 mL of each sample in RNAlater was pelleted by centrifugation at 13,000×g for 5 min at 4° C. The pellets were washed once in 50% RNAlater/PBS buffer and resuspended with lysis in 2 mL of denaturing buffer (4 M guanidium thiocyanate, 25 mM sodium citrate, 0.5% N-laurylsarcosine, 1% N-acetyl cysteine, 0.1 M 2-mercaptoethanol). The lysate was divided into 500 μL aliquots, to which 4 μL of 1M sodium acetate (pH 5.2) was added. Each aliquot was then incubated with 500 μL of buffer saturated phenol (pH 4.3) at 64° C. for 10 minutes, with intermittent mixing. One ml of chloroform was added to the solution and incubated for 15 minutes on ice, followed by centrifugation at 18,000×g for 30 min at 4° C. Afterward, RNA was precipitated from the aqueous layer by adding 1/10 volume 3M sodium acetate, 500 mM DEPC treated EDTA and 2 volumes of cold ethanol followed by overnight incubation at −80° C. The RNA was then pelleted by centrifugation at 4° C., washed with 80% cold ethanol and resuspended in 100 μL of RNAse free ddH2O. The extracted RNA was treated twice with DNase I (Epicentre) followed by PCR amplification using the 16S rRNA universal primers; Bact-8F and 1391-R; to confirm the absence of genomic DNA. The quality and the quantity of the extracted RNA was determined by NanoDrop 2000 spectrophotometer (Thermoscientific) and confirmed by BioRad's Experion StdSens RNA system according to the manufacturer's description and stored at −80° C. until use.


The quantification of the expression level of TST (Thiosulfate Sulfurtransferase), SQRDL (Sulfide Quinone Reductase Like) and COX4-1 (Cytochrome C oxidase subunit IV isoform 1) relative to GAPDH (Glyceraldehyde-3-Phosphate Dehydrogenase) genes was determined using the Applied Biosystems 7300 DNA analyzer and Quantitect SYBR Green RT-PCR kit (Qiagen). The primers used were either designed by NCBI Primer-BLAST tool42 or extracted from a literature source as detailed in Supplementary Table 11. Each reaction contained 100 ng RNA template, 0.5 μM of each primer, 1× Quantitect SYBR Green RT-PCR master mix and 0.25 μL Quantitect RT-mix in a final volume of 25 μL. The one step RT-PCR conditions were 50° C. for 30 min, 95° C. for 15 min followed by 40 cycles of 15 sec at 94° C., 30 sec at 60° C. and 30 sec at 72° C. with data collection at the third step of each cycle. The amplification specificity was checked by the melting profile of the amplicon and 2% agarose gel electrophoresis. Ct values were then extracted using the Applied Biosystems 7300 sequence detection software versions 1.3.1. Ct values of TST, SQRDL, or COX4-1 were normalized to the Ct values of GAPDH generating ΔCt. Next, ΔΔCt was calculated by subtracting the average ΔCt of the control group from the ΔCt of each sample. The relative quantification were then calculated by 2−ΔΔCt as mentioned previously34.


Example 12

II10−/− Mice Experiments and Tissue Processing:


Germ-free SvEv129/C57BL6 II10−/−; NF-κBEGFP mice (8-12 weeks old, n=12) were transferred to specific pathogen free (SPF) conditions and mice from one cohort (n=6) were gavaged once weekly with A. parvulum (1×108 CFUs) for 6 weeks. Atopobium parvulum ATCC 33793 was grown in fastidious anaerobic broth (FAB) (Lab M, Canada).


To investigate involvement of complex biota and H2S in the development of colitis, we performed two subsequent experiments using 129/SvEv II-10−/− mice. In the first experimental setting, gnotobiotic II10−/− mice (n=37) were randomized into 4 groups; 1: GF only (n=6), 2: GF+bismuth (III) subsalicylate (n=10); 3: A. parvulum (1×108 CFUs) (n=10) and 4: A. parvulum+bismuth (III) subsalicylate (n=11). Mice were euthanized after 6 weeks of mono-association. Bismuth (III) subsalicylate (Sigma-Aldrich, Saint Louis, Mo.) was incorporated to the chow (Teklan Global 18% Protein Rodent Diet) at a concentration of 7 g/kg (Harlan Laboratories, Madison, Wis.) and then irradiated for gnotobiotic experiments. Mice were fed with this diet starting 1 week before the colonization with A. parvulum. In the second experimental setting, gnotobiotic II10−/− mice (n=31) were transferred to SPF conditions and randomized into 4 groups; 1: SPF only (n=7), 2: SPF+bismuth (III) subsalicylate (n=8); 3: SPF plus A. parvulum (1×108 CFUs) (n=8) and 4: A. parvulum+bismuth (III) subsalicylate (n=8). Mice were euthanized after 6 weeks of weekly infection with A. parvulum. Bismuth (III) subsalicylate (Sigma-Aldrich, Saint Louis, Mo.) was incorporated to the chow (Teklan Global 18% Protein Rodent Diet) at a concentration of 7 g/kg (Harlan Laboratories, Madison, Wis.). Mice were fed with this diet starting 1 week before the colonization with A. parvulum.


All animal protocols were approved by the Institutional Animal Care and Use Committee of the University of North Carolina at Chapel Hill. Tissue samples from the colon were collected for RNA and histology as described previously24. Histological images were acquired using a DP71 camera and DP Controller 3.1.1.276 (Olympus), and intestinal inflammation was scored as previously described12. The tissue was divided into 4 quarters, a score was given to each quarter separately and then added to generate a final inflammation score on a scale of 0-16.


Example 13

Mouse Endoscopy:


Colonoscopy was performed using a “Coloview System” (Karl Storz Veterinary Endoscopy) as described previously25. Mice were anesthetized using 1.5% to 2% isoflurane and ˜4 cm of the colon from the anal verge from the splenic flexure was visualized. The procedures were digitally recorded on an AIDA Compaq PC.


Example 14

Real Time RT-PCR on Mouse Intestinal Samples:


Total RNA from intestinal tissues was extracted using TRIzol (Invitrogen) following the manufacturers protocol. cDNA was reverse-transcribed using M-MLV (Invitrogen) and mRNA expression levels were measured using SYBR Green PCR Master mix (Applied Biosystems) on an ABI 7900HT Fast Real-Time PCR System and normalized to β-actin. The primers used were as follows: β-actin (5′-TGGAATCCTGTGGCATCCATGAAAC-3′ (SEQ ID NO. 20) and 5′-TAAAACGCAGCTCAGTAACAGTCCG-3′ (SEQ ID NO. 21)), cxcl1 (5′-GCTGGGATTCACCTCAAGAA-3′ (SEQ ID NO. 22) and 5′-TCTCCGTTACTTGGGGACAC-3′ (SEQ ID NO. 23)), tnf (5′-ATGAGCACAGAAAGCATGATC-3′ (SEQ ID NO. 24) and 5′-TACAGGCTTGTCACTCGAATT-3′ (SEQ ID NO. 25)), il12p40 (5′-GGAAGCACGGCAGCAGCAGAATA-3′ (SEQ ID NO. 26) and 5′-AACTTGAGGGAGAAGTAGGAATGG-3′ (SEQ ID NO. 27)), il1β (5′-GCCCATCCTCTGTGACTCAT-3′ (SEQ ID NO. 28) and 5′-AGGCCACAGGTATTTTGTCG-3′ (SEQ ID NO. 29)), il-17a (5′-TCCAGAAGGCCCTCAGACTA-3′ (SEQ ID NO. 30) and 5′-ACACCCACCAGCATCTTCTC-3′ (SEQ ID NO. 31)). The PCR reactions were performed for 40 cycles according to the manufacturer's recommendation, and RNA fold changes were calculated using the ΔΔCt method.


Statistical Analyses Of II10−/− Mice Results:


Unless specifically noted, statistical analyses were performed using GraphPad Prism version 5.0a (GraphPad, La Jolla, Calif.). Comparisons of mouse studies were made with a nonparametric analysis of variance, and then a Mann-Whitney U test. All graphs depict mean±SEM. Experiments were considered statistically significant if p<0.05.


Example 15

Sample Collection.


Mucosal aspirates (washings) were collected from the right colon of 40 control children, 41 crohn's disease (CD) and 20 ulcerative colitis (UC) patients aged 3-18 years old at initial diagnosis, at Children's Hospital of Eastern Ontario (CHEO), Ottawa, Canada, using a standardized protocol.28 The right colon was selected in particular because it is thought to be the most active site for butyrate synthesis.29,30 In addition, fresh stool samples were collected from a subset of patients (5 control and 10 CD) at least 24 h prior to the endoscopy procedure (Table S1 for participating patient information). Immediately following collection, samples were transported on ice to the lab where they were either processed for DNA extraction or stored at −80° C. until further use.


Extraction of Metagenomic DNA.


5 ml aliquots of the mucosal washes were spun at 20,000×g for 10 min at 4° C. Then, DNA was extracted from the sediments (or stool samples) using the FastDNA® Spin Kit (MP Biomedicals) utilizing two mechanical lysis cycles in a FastPrep® Instrument (MP Biomedicals) set at speed of 6.0 for 40 seconds. Extracted DNA was then stored at −20° C. until further use.


Characterize the Diversity of Butyrate-Producing Bacteria in Healthy and IBD Children


Relative Quantification of BCoAT Gene from Mucosal-Washes Metagenomic DNA Using qPCR.


The overall abundance of butyrate-producing bacteria was determined by quantifying the amount of BCoAT gene utilizing the primers BCoATscrF/BCoATscrR as described elsewhere.27 50 ng of metagenomic DNA from each sample was used in a 25 μl qPCR reaction containing 1× QuantiTect SYBR Green PCR Master Mix (QIAGEN) and 0.5 μM of BCoATscrF/BCoATscrR primers. The amplification conditions were as follows: 1 cycle of 95° C. for 15 min; 40 cycles of 94° C. for 15 sec, 53° C., and 72° C. each for 30 sec with data acquisition at 72° C. For the melting curve analysis, a stepwise temperature increase from 55° C. to 95° C. was performed. Quantification standards were prepared by purifying and quantifying the BCoAT gene fragment from healthy subjects using PureLink™ PCR Purification Kit (Invitrogen) and Qubit® dsDNA BR Assay Kit (Invitrogen), respectively. Then, 106, 107, and 108 gene copies were prepared assuming an average molecular weight of 660 per nucleotide. Simultaneously, the number of 16S rRNA gene copies was quantified in parallel to the BCoAT gene as described previously31, and results were expressed as copy number of BCoAT genes per 16S rRNA gene.


Preparation of BCoAT Gene and 16S rRNA Libraries from Mucosal-Washes Metagenomic DNA for Deep Sequencing.


BCoAT library construction was carried out using a two-step PCR strategy. In the 1st step, 50 ng of metagenomic DNA was used in a 50 μl PCR reaction containing 1.5 mM MgCl2, 0.5 μM of BCoATscrF/BCoATscrR primers, 0.2 mM dNTPs, and 2.5 U HotStarTaq DNA polymerase (QIAGEN). Amplification started with an initial enzyme activation step at 95° C. for 15 min. Then, amplification was carried out using 25 cycles at 94° C., 53° C., and 72° C. (each for 30 sec), and a 10 min final extension at 72° C. For the second PCR, 13 fusion primers were designed (12 forward and one reverse, SEQ ID 3-15) following Roche's Amplicon Fusion Primer Design Guidelines for GS FLX Titanium Series Lib-L Chemistry. Briefly, the forward primers contain (from 5′-3′) GS FLX Titanium Primer A, a four-base library key, 12 different Multiplex Identifiers (MIDs), and a BCoATscrF primer sequence. The reverse primer contains (from 5′-3′) GS FLX Titanium Primer B, a four-base library key, and a BCoATscrR primer sequence (Table 9). 10 μl of product from the 1st PCR was utilized in 50 μl for the second PCR reaction using a unique MID fusion primer for every 12 samples and the same concentration of PCR component as the 1st PCR. A total of 15 amplification cycles were performed utilizing the same amplification conditions as the first PCR. For each sample, a total of 5 reaction tubes were prepared. Following amplification, PCR products from the same sample were pooled together, inspected on 1.5% agarose gel, and purified using Montage PCR96 Cleanup Kit (Millipore). Finally, an equimolar amount of samples with unique MIDs (a total of 4 libraries) were pooled together and sequenced on a Roche 454 platform using a full plate of GS FLX Titanium chemistry (each library in ¼ plate) at The McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada. A 16S rRNA library was constructed. The 16S rRNA library was sequenced at The Centre for Applied Genomics (TCAG) at the Hospital for Sick Children in Toronto (Canada) using a HiSeq 2500 platform to generate paired-end reads of 2×100 bases.











TABLE 9






Primer




Name
Primer Sequence







1st
BCoATs
5′-GCIGAICATTTCACITGGAAYWSITGGCAYATG-3′ (SEQ ID NO. 32)


PCR
crF







BCoATs
5′-CCTGCCTTTGCAATRTCIACRAANGC-3′ (SEQ ID NO. 33)



crR






2nd
BCoAT-
5′-


PCR
F-1
CCATCTCATCCCTGCGTGTCTCCGACTCAGACGAGTGCGTGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 3)






BCoAT-
5′-



F-2
CCATCTCATCCCTGCGTGTCTCCGACTCAGACGCTCGACAGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 4)






BCoAT-
5′-



F-3
CCATCTCATCCCTGCGTGTCTCCGACTCAGAGACGCACTCGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 5)






BCoAT-
5′-



F-4
CCATCTCATCCCTGCGTGTCTCCGACTCAGAGCACTGTAGGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 6)






BCoAT-
5′-



F-5
CCATCTCATCCCTGCGTGTCTCCGACTCAGATCAGACACGGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 7)






BCoAT-
5′-



F-6
CCATCTCATCCCTGCGTGTCTCCGACTCAGATATCGCGAGGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 8)






BCoAT-
5′-



F-7
CCATCTCATCCCTGCGTGTCTCCGACTCAGCGTGTCTCTAGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 9)






BCoAT-
5′-



F-8
CCATCTCATCCCTGCGTGTCTCCGACTCAGCTCGCGTGTCGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 10)






BCoAT-
5′-



F-10
CCATCTCATCCCTGCGTGTCTCCGACTCAGTCTCTATGCGGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 11)






BCoAT-
5′-



F-11
CCATCTCATCCCTGCGTGTCTCCGACTCAGTGATACGTCTGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 12)






BCoAT-
5′-



F-13
CCATCTCATCCCTGCGTGTCTCCGACTCAGCATAGTAGTGGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 13)






BCoAT-
5′-



F-14
CCATCTCATCCCTGCGTGTCTCCGACTCAGCGAGAGATACGCIGAICATTTCACITGGAAYWSITGG




CAYATG-3′ (SEQ ID NO. 14)






BCoAT-
5′-CCTATCCCCTGTGTGCCTTGGCAGTCTCAGCCTGCCTTTGCAATRTCIACRAANGC-3′



R
(SEQ ID NO. 15)









Data Analysis.


For BCoAT sequencing, demultiplexed reads from each sample were filtered using RDP's Pyrosequencing Pipeline32 based on a minimum quality score of 20 and 200 nucleotides read length cutoff. Operational Taxonomic Units (OTUs) clustering at 95% sequence similarity was achieved using a de novo UCLUST algorithm integrated in Quantitative Insights Into Microbial Ecology (QIIME) software package V 1.7.0,33 after which, singleton OTUs were removed. QIIME was also used to compute alpha and beta diversity between samples using a fixed number of reads/sample of 4,600. The longest sequence from each OTU was then selected and used for taxonomy assignment as described previously.23 Sequences with <75% identity to functional gene reference database were considered unclassified OTUs. Finally, the relative abundance (RA) of assigned species was calculated and differences in butyrate producing bacteria RA were analyzed.


16S rRNA paired-end sequences were merged using Fast Length Adjustment of SHort reads (FLASH) software.34 During this step, most reads overlapped perfectly by about 10-80 nucleotides, and less than 5% of the reads failed to combine. Uncombined reads were discarded from further analysis. Subsequently, Novobarcode command from Novocraft Technologies was used to demultiplex merged reads according to the 5′ and 3′ barcode sequences and trim the barcode sequence from the corresponding read. Reads with minimum quality score of 20 were selected for further analysis using fastq_quality_filter command line from the Fastx-toolkit V 0.0.13. Taxonomy assignment to the genus level was done using QIIME V 1.5.0 aligning against Greengenes database (release 4 Feb. 2011) using UCLUST Reference-based OTU picking method at 97% sequence identity. Butyrate producers were selected from the overall micobiota using the list of bacteria that produce butyrate through BCoAT pathway found in reference23. Since each bacteria has a different copy number of the 16S rRNA gene and only one copy of the BCoAT gene, the copy number of 16S rRNA was normalized to 1 by dividing the number of reads of a given genus by its average 16S rRNA copy number obtained from rrnDB.35 The RA of identified butyrate producer genera was then calculated. Finally, correlation between BCoAT and 16S rRNA datasets was analyzed.


For phylogenetic analysis, full nucleotide sequences of BCoAT genes for the assigned butyrate producer species were obtained from the National Center for Biotechnology Information (NCBI's) Reference Sequence Database and MUSCLE aligned with the unclassified OTUs sequences. Then, a phylogenetic tree of aligned sequences was constructed using a maximum-likelihood algorithm with FastTree tool integrated into QIIME. Visual display of the rooted tree was achieved using Interactive Tree Of Life (iTOL) tool.36 Using the same strategy, another phylogenetic tree was constructed from MUSCLE aligned unclassified OTU sequences and but nucleotide database sequences (a dataset of all predicted BCoAT gene sequences (4,041 sequences) obtained from the Functional Gene Pipeline/Repository.26


Confirmation of BCoAT Sequencing Results Using qPCR.


35 control, 37 CD and 19 UC mucosal aspirate samples were used to validate the sequencing result by qPCR. Primers specific to BCoAT gene of E. rectale and F. prausnitzii were used in the qPCR. In addition, primers targeting the 16S rRNA gene of Eubacterium rectale/Clostridium coccoides group (XIVa) (20-21), F. prausnitzii 22-23, and Roseburia 24-25 were used. For stool samples, 5 control and 10 CD subjects were used to determine the relative amount of F. prausnitzii using 16S rRNA specific primers only (table 10). The complete 16S rRNA gene was amplified using the universal primer UniF/UniR (18-19) adapted from reference37. Fifty ng of metagenomic DNA was used in a 25 μl PCR reaction using QuantiTect SYBR Green PCR Master Mix (QIAGEN) as described in the previous section using 55° C. instead of 53° C. annealing temperature. The assay was done in duplicate for each sample. Delta-Ct (ΔCt) for each target was calculated by subtracting the Ct of the total 16S rRNA from the target Ct. Then, the ΔCt values were compared between groups.












TABLE 10






Primer




Target
Name
Primer Sequence
Reference







16S rRNA gene from 
UniF
5′-GTGSTGCAYGGYYGTCGTCA-3′
ISME J 2011; 5:


all bacteria

(SEQ ID NO. 34)
220-230



UniR
5′-ACGTCRTCCMCNCCTTCCTC-3′





(SEQ ID NO. 35)






Eubacterium rectale/
UniF338
5′-ACTCCTACGGGAGGCAGC-3′
Infect. Immun. 



Clostridium coccoides


(SEQ ID NO. 36)
2008


group 16S rRNA gene
C.cocR491
5′-GCTTCTTAGTCAGGTACCGTCAT-3′





(SEQ ID NO. 37)







Faecalibacterium 

Fprau 07
5′-CCATGAATTGCCTTCAAAACTGTT-3′
FEMS Microbiol. 



prausnitzii


(SEQ ID NO. 38)
Ecol. 2012; 79:


16S rRNA gene
Fprau 02
5′-GAGCCTCAGCGTCAGTTGGT-3′
685-696




(SEQ ID NO. 39)







Roseburia spp. 16S 

Ros-F1
5′-GCGGTRCGGCAAGTCTGA-3′
FEMS Microbiol. 


rRNA gene

(SEQ ID NO. 40)
Ecol. 2012; 79:



Ros-R1
5′-CCTCCGACACTCTAGTMCGAC-3′
685-6964




(SEQ ID NO. 41)






BCoAT gene from all
BCoATscrF
5′-GCIGAICATTTCACITGGAAYWSITGGCAYATG-
Appl. Environ.


bacteria

3′ (SEQ ID NO. 42)
Microbiol.



BCoATscrR
5′-CCTGCCTTTGCAATRTCIACRAANGC-3′
2007; 73:




(SEQ ID NO. 43)
2009-2012.






Eubacterium rectale 

RosEub_F
5′-TCAAATCMGGIGACTGGGTWGA-3′
Microbiome 


BCoAT gene

(SEQ ID NO. 44)
2013, 1:8



Eub_R
5′-TCATAACCGCCCATATGCCATGAG-3′





(SEQ ID NO. 45)







Faecalibacterium 

Fprsn_F
5′-GACAAGGGCCGTCAGGTCTA-3′
Microbiome 



prausnitzii


(SEQ ID NO. 46)
2013, 1:8.


BCoAT gene
Fprsn_R
5′-GGACAGGCAGATRAAGCTCTTGC-3′





(SEQ ID NO. 47)









Statistical Analysis.


Unless otherwise specified, result from the qPCR and sequencing was analyzed using two-tailed Mann-Whitney test comparing IBD subtypes to the control group. A P value less than 0.05 was considered significant. Correlation between BCoAT and 16S rRNA sequencing was done by calculating Spearman's rank correlation coefficient (r) of paired RA of bacterial taxa identified by the two approaches.


Butyrate Producers are Reduced in UC Patients with Colonic Inflammation.


In order to determine the relative amount of butyrate producers, the copy number of BCoAT genes to 16S rRNA was assayed. The difference in the relative number of BCoAT genes between control subjects (2.15×10−4±2.46×10−4) and IBD subgroups (CD, 2.17×10−4±1.97×10−4; UC, 1.74×10−4±2.78×10−4) was not statistically significant (P>0.2) (FIG. 13). However, analyzing each IBD group based on macroscopic appearance during colonoscopy, revealed that UC patients with an inflamed colon had a lower number of butyrate producers (3.13×10−5±3.10×10−5) compared to control subjects (P=0.029) (FIG. 13).


Diversity of Butyrate Producers is Different in IBD Patients Compared to Healthy Subjects.


A total of 670,287 high quality reads were generated from 43 samples (13 control, 20 CD, and 10 UC) with an average of 15,570 reads per sample (range, 44,158-2,938) and an average read length of 465 nucleotides (summarized in Table S4). Clustering reads at a 0.05 distance resulted in a total of 965 OTUs from all samples with a total OTU number of 714 for controls, 804 for CD, and 744 for UC. The majority of observed OTUs were shared between the three groups (486 OTUs), and only a few were unique to each individual group (FIG. 14A). Comparing samples alpha diversity, using Chao1 estimated OTUs and the Shannon diversity index with equalized data sets to 4,600 reads, revealed that CD samples had significantly lower Chao1 estimated OTUs compared to controls (P=0.04) (FIG. 14B). In contrast, no difference was observed in the Shannon index (FIG. 14B). This signifies that IBD subjects have a similar evenness to controls, but CD patients show a lower richness.


Furthermore, multidimensional scaling analysis of UniFrac metrics, presented by principal coordinates analysis (PCoA) plot, indicates that the IBD group is different than controls. Although no separation was observed with unweighted UniFrac (data not shown), PCoA showed good separation of CD and UC samples from control with weighted UniFrac. When clustering CD samples with controls, most control and CD subjects were grouped into two distinct clusters with 52.6% of the variance accounted for by coordinate 1 (PCoA1) and an additional 12.7% of variance attributable to PCoA2 (FIG. 14C). Similar separation was observed when comparing UC against control, with 62.1% of variance explained by PCoA1 and 10.2% by PCoA2 (FIG. 14C).



Eubacterium Rectale is Depleted and Faecalibacterium prausnitzii Thrives in IBD Patients.


In order to take a more in depth look at butyrate producer diversity, we looked at the assigned bacterial taxa. Overall, OTUs from all samples were assigned to 12 classified bacterial species that belong to the Firmicutes phylum in addition to 67 unclassified OTUs (FIG. 15). In the control group, the majority of butyrate-producing bacteria belonged to Clostridium cluster XIVa. These included Eubacterium rectale (58.7%±29.7), Eubacterium hallii (8.5%±8.9), Roseburia inulinivorans (2.7%±7.2), Roseburia hominis (1%±2.9), Coprococcus catus (0.18%±0.65), Roseburia intestinalis (0.06%±0.17), and Clostridium symbiosum (8.4×10−5%±2.8×10−4). Faecalibacterium prausnitzii, a member of the Clostridium cluster IV, was also found to be a major contributor to healthy butyrate producers' consortium with an abundance of 14.3%±17.4. Other members of the healthy core of butyrate producers are Clostridium sp. SS2/1 (11.2%±22.1), butyrate-producing bacterium SS3/4 (0.03%±0.08), Clostridium sp. M62/1 (0.019%±0.068), Ruminococcus bacterium D16 (2.7×10−6%±9.8×10−6), and 49 different unclassified OTUs (3.1%±4.4).


a. Comparing the identified bacterial species RA of the control group to IBD patients (both CD or UC) revealed that the control group is characterized by a higher RA of E. rectale (P<0.05) (FIG. 16A). Subclassifying IBD patients based on endoscopic appearance showed that E. rectale was reduced in CD patients with either inflamed or non-inflamed colon (P<0.02). Conversely, only UC patients with an inflamed colon had reduced E. rectale RA (P<0.05) (FIG. 16A). Another butyrate producer with reduced RA in CD is R. inulinivorans. R. inulinivorans reduction was solely restricted to CD patients with an inflamed colon (P=0.04) (FIG. 16C). Unexpectedly, F. prausnitzii, which is one of the most abundant butyrate producers in the healthy human gut, was increased in CD, particularly CD patients with an inflamed colon (P=0.04) (FIG. 16B). Although F. prausnitzii RA was also high in UC patients compared to control, the difference did not reach statistical significance (P=0.07). In conclusion, this highlights the presence of a unique signature of butyrate producers that distinguishes IBD from controls. Comparison of bacterial RA was also carried-out for unclassified OTUs. In total, 61 unclassified OTUs were found in CD samples that represent 7.2%±20.2 of all reads and 39 for UC that represent 0.7%±1 of total reads. The overall number of unclassified OTUs was significantly lower in UC (P<0.05; FIG. 16D). Unclassified OTU_34, which shares 59% identity with E. rectale, was reduced in CD patients with either normal or inflamed colons (CD, P=0.006; CD_normal, P=0.01; CD_inflamed, P=0.05; FIG. 16E). Another unclassified OTU, OTU_43 that share 61% identity with E. rectale, was reduced in all IBD subsets (P<0.01). OTU_43 was reduced in both CD subtypes as well as UC patients with an inflamed colon (FIG. 16F).


b. In order to test the possibility that the unclassified OTUs could represent novel butyrate producers, the complete sequences of the BCoAT gene for the assigned bacterial species were MUSCLE aligned with the unclassified OTUs sequences. The aligned sequences were then used to construct a phylogeny tree using a maximum-likelihood algorithm. Twenty-five of the 67 unclassified OTUs were clustered with known butyrate producers. Among these, the unclassified OTUs 34 and 43, which were found to be deficient in IBD, clustered with E. rectale. Interestingly, 42 of the unclassified OTUs did not cluster with any of the assigned butyrate producers. In a second step, the sequences of unclassified OTUs were MUSCLE aligned with the but nucleotide database downloaded from the Functional Gene Pipeline/Repository. The but database contains all nucleotide sequences of probable BCoAT genes identified by Hidden Markov Model searches of the NCBI bacterial protein database. Subsequently the aligned sequences were subjected to phylogenetic tree construction. This time, only 4 of the 67 unclassified OTUs were clustered with classified bacteria. The remaining OTUs clustered only with partial BCoAT coding sequences isolated from human samples that belong to unclassified uncultured bacterium. Hence, this suggests that the 63 unclassified OTUs might belong to novel butyrate producers.


Diversity of Butyrate-Producing Bacteria Revealed by 16S rRNA Sequencing:


Analyzing the diversity of butyrate producing bacteria at the genus level using 16S rRNA sequencing reveals similar results to BCoAT sequencing with minor differences. In total, 10 genera of butyrate producers were identified with the 16S rRNA approach compared to 6 genera using a functional gene approach. The majority of reads were assigned to 4 genera: Eubacterium, Faecalibacterium, Roseburia, and Coprococcus (FIG. 17). In accordance with our functional gene findings, Eubacterium was higher in the control (21.66%±22.95) group compared to IBD (CD, 6.89%±9.47; UC, 9.48±18.72). Furthermore, Faecalibacterium dominated both CD (55.6%±35.56) and UC (60.12%±29.29) compared to controls (18.09%±22.13). In contrast to BCoAT data, Roseburia was the most abundant butyrate-producing bacteria in the control group (34.52%±28.55) and had a higher abundance compared to CD (23.82%±24.76) and UC (16.99%±19.3). This further supports a previous hypothesis that butyrate production is restricted to certain members of the same genus.26 Similarly, Coprococcus abundance was higher in 16S rRNA data compared to the BCoAT approach (FIG. 17). However, it is worth noting that Coprococcus can produce butyrate not only via the BCoAT pathway but also via a butyrate kinase pathway.26 This could explain the reduced abundance of Coprococcus in the BCoAT data. The 16S rRNA approach identified 6 low abundant butyrate producers (total abundance of the 6 genera<1%) that were missed by BCoAT sequencing. These include: Peptoniphilus, Anaerofustis, Anaerostipes, Butyrivibrio, Megasphaera, and Treponema. This could be attributed to the higher sequencing depth of the HiSeq2500 Illumina platform used for 16S rRNA that can generate up to 50 times more reads than 454 pyrosequencing, which was used for the BCoAT sequencing. Lack of species level resolution by the 16S rRNA data made it impossible to identify some important butyrate producers that were identified by the functional gene approach. These include: Clostridium sp. M62/1, Clostridium sp. SS2/1, and Clostridium symbiosum. Calculating paired Spearman's rank correlation coefficient (r) for the relative abundance of butyrate producers identified by 16S rRNA and BCoAT sequencing revealed a strong correlation between the two datasets (r=0.73).


Relative Quantification of Key Butyrate Producers Revealed by qPCR:


BCoAT sequencing results were further validated using qPCR utilizing BCoAT and 16S rRNA specific primers. Eubacterium rectale/Clostridium coccoides group (XIVa), which is dominated by E. rectale, was reduced in both CD subtypes compared to controls. On the other hand, the UC group had similar levels of group XIVa compared to the controls (FIG. 18B). This finding in the UC group is not unexpected since Clostridium cluster XIVa harbors other major butyrate producers and non-butyrate producers, and an increase in other members of group XIVa could obscure the reduction of E. rectale in UC patients. Absence of species-specific, or even genus specific, primers to E. rectale 16S rRNA made it impossible to target this bacteria only using the 16S rRNA qPCR. When targeting the BCoAT gene of E. rectale, a clear reduction in all IBD subtypes compared to controls was observed (P<0.02; FIG. 18A). Nonetheless, BCoAT sequencing showed a decrease in E. rectale RA in UC patients with an inflamed colon only, and the reduction of E. rectale RA in UC patients with normal colon did not reach statistical significance (FIG. 16A). Although F. prausnitzii was high in CD with inflamed colons by sequencing, qPCR (using both BCoAT and 16S rRNA primers specific to F. prausnitzii) revealed that it is increased not only in CD but also in UC patients with an inflamed colon (FIGS. 18C and 18D). However, the slight difference in qPCR statistical significance compared to sequencing is due to higher sample numbers used in qPCR compared to sequencing. Finally, the reduction in R. inulinivorans, demonstrated by sequencing, was only validated at the genus level using 16S rRNA specific primers due to the lack of species-specific primers. qPCR shows that Roseburia is reduced in CD (both with normal and inflamed colon) (FIG. 8E), however, the statistical significance was higher in CD with inflamed colons (CD-normal, P<0.017; CD-inflamed, P<0.009). Taken together, the qPCR findings are in keeping with those of the BCoAT sequencing.


In order to investigate if sample type (stool versus mucosal aspirate) could affect the level of detected bacteria, stool collected from 5 control and 10 CD (6 with normal and 4 with inflamed colons, table S1) patients were subjected to qPCR using 16S rRNA specific primers to F. prausnitzii. Contrary to the mucosal aspirate finding, F. prausnitzii showed similar levels to the controls in both CD subtypes (FIG. 19).


REFERENCES



  • 1 Cho, I. & Blaser, M. J. Nature reviews. Genetics 13, 260-270, doi:10.1038/nrg3182 (2012).

  • 2 Manichanh, C., Borruel, N., Casellas, F. & Guarner, F. Nature reviews. Gastroenterology & hepatology 9, 599-608, doi:10.1038/nrgastro.2012.152 (2012).

  • 3 Benchimol, E. I. et al. Gut 58, 1490-1497, doi:10.1136/gut.2009.188383 (2009).

  • 4 Kostic, A. D. et al. Cell host & microbe 14, 207-215, doi:10.1016/j.chom.2013.07.007 (2013).

  • 5 Rubinstein, M. R. et al. Cell host & microbe 14, 195-206, doi:10.1016/j.chom.2013.07.012 (2013).

  • 6 Strauss, J. et al. Inflammatory bowel diseases 17, 1971-1978, doi:10.1002/ibd.21606 (2011).

  • 7 Copeland, A. et al. Standards in genomic sciences 1, 166-173, doi:10.4056/sigs.29547 (2009).

  • 8 Kazor, C. E. et al. Journal of clinical microbiology 41, 558-563 (2003).

  • 9 Blachier, F. et al. Amino acids 39, 335-347, doi:10.1007/s00726-009-0445-2 (2010).

  • 10 Linden, D. R. Antioxidants & redox signaling, doi:10.1089/ars.2013.5312 (2013).

  • 11 Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome biology 12, R60, doi:10.1186/gb-2011-12-6-r60 (2011).

  • 12 Karrasch, T., Kim, J. S., Muhlbauer, M., Magness, S. T. & Jobin, C. Journal of immunology 178, 6522-6532 (2007).

  • 13 Kim, S. C., Tonkonogy, S. L., Karrasch, T., Jobin, C. & Sartor, R. B. Inflammatory bowel diseases 13, 1457-1466, doi: 10. 1002/ibd.20246 (2007).

  • 14 Levine, J., Ellis, C. J., Fume, J. K., Springfield, J. & Levitt, M. D. The American journal of gastroenterology 93, 83-87, doi:10.1111/j.1572-0241.1998.083_c.x (1998).

  • 15 Kaiserling, E. Newly-formed lymph nodes in the submucosa in chronic inflammatory bowel disease. Lymphology 34, 22-29 (2001).

  • 16 Mackay, F. et al. Both the lymphotoxin and tumor necrosis factor pathways are involved in experimental murine models of colitis. Gastroenterology 115, 1464-1475 (1998).

  • 17 Salvador, J. A., Figueiredo, S. A., Pinto, R. M. & Silvestre, S. M. Future medicinal chemistry 4, 1495-1523, doi:10.4155/fmc.12.95 (2012).

  • 18 Rajilic-Stojanovic, M. Clinical gastroenterology 27, 5-16, doi:10.1016/j.bpg.2013.03.006 (2013).

  • 19 Ramasamy, S., Singh, S., Taniere, P., Langman, M. J. & Eggo, M. C. American journal of physiology. Gastrointestinal and liver physiology 291, G288-296, doi:10.1152/ajpgi.00324.2005 (2006).

  • 20 Bar, F. et al. Gastroenterology 145, 1055-1063 e1053, doi:10.1053/j.gastro.2013.07.015 (2013).

  • 21 Miller, T. W. et al. The Journal of biological chemistry 287, 4211-4221, doi:10.1074/jbc.M 111.307819 (2012).

  • 22 Smith, P. M. et al. Science 341, 569-573, doi:10.1126/science.1241165 (2013).

  • 23 Caporaso, J. G. et al. Nature methods 7, 335-336, doi:10.1038/nmeth.f.303 (2010).

  • 24 Sun, X., Threadgill, D. & Jobin, C. Gastroenterology 142, 86-95 e85, doi:10. 1053/j.gastro.2011.09.042 (2012).

  • 25 Lippert, E. et al. PloS one 4, e7413, doi:10.1371/journal.pone.0007413 (2009).

  • 26 Vital M, Penton C R, Wang Q, et al. 2013; 1:1.

  • 27 Louis P, Flint HJ. Appl. Environ. Microbiol. 2007; 73:2009-2012. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1828812&tool=pmcentrez&rend ertype=abstract [Accessed Nov. 11, 2013].

  • 28. Jimenez-Rivera C, Haas D, Boland M, et al. Gastroenterol Res Pr. 2009; 2009:1-4. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2796226&tool=pmcentrez&rend ertype=abstract [Accessed Nov. 11, 2013].

  • 29. Louis P, Flint H J. FEMS Microbiol. Lett. 2009; 294:1-8.

  • 30. Duncan S H, Louis P, Thomson J M, et al. Environ. Microbiol. 2009; 11:2112-2122.

  • 31. Bourne D G, Muirhead A, Sato Y. ISME J 2011; 5:559-564. Available at: http://www.pubmedcentral. nih. gov/articlerender.fcgi?artid=3105726&tool=pmcentrez&rend ertype=abstract [Accessed Nov. 11, 2013].

  • 32. Cole J R, Wang Q, Cardenas E, et al. Nucleic Acids Res 2009; 37:D141-D145.

  • 33. Caporaso J G, Kuczynski J, Stombaugh J, et al. Nat Methods 2010; 7:335-336.

  • 34. Magoc T, Salzberg S L, Mago6 T. Bioinformatics 2011; 27:2957-2963. Available at: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3198573&tool=pmcentrez&rend ertype=abstract [Accessed Nov. 11, 2013].

  • 35. Lee ZM-P, Bussema C, SchmidtT M rrnDB: Nucleic Acids Res 2009; 37:D489-93.

  • 36. Letunic I, Bork P. Interactive Tree Of Life (iTOL): Bioinformatics 2007; 23:127-128. Available at: http://www.ncbi.nlm.nih.gov/pubmed/17050570 [Accessed Dec. 13, 2013].


Claims
  • 1. A method for determining a severity of inflammatory bowel disease, Crohn's disease, or ulcerative colitis in a human subject, wherein the determining of the severity includes determining if there is an absence of inflammatory bowel disease, Crohn's disease, or ulcerative colitis, comprising: measuring a level of Atopobium parvulum in a gut microbiota sample from the human subject using quantitative polymerase reaction, wherein the quantitative polymerase chain reaction uses a forward and reverse primer for targeting A. parvulum and wherein the forward primer is SEQ ID 1 and the reverse primer is SEQ ID 2, andcomparing the measured level of A. parvulum to a predetermined level of A. parvulum, wherein the measured level of A. parvulum relative to the predetermined level is indicative of the severity of the disease.
  • 2. A method for determining a severity of Crohn's disease, wherein a degree of inflammation is indicative of the severity of Crohn's disease, comprising: measuring a level of Atopobium parvulum in a gut microbiota sample from a human subject using quantitative polymerase chain reaction, wherein a level higher than a predetermined level of A. parvulum is indicative of moderate or severe inflammation, and wherein the quantitative polymerase chain reaction uses a forward and reverse primer for targeting A. parvulum and wherein the forward primer is SEQ ID 1 and the reverse primer is SEQ ID 2.
  • 3. The method of claim 2 wherein the predetermined level is a level corresponding to mild inflammation.
  • 4. The method of claim 2, wherein the predetermined level is an abundance of A. parvulum greater than about 0.005 relative abundance of total bacteria from the gut microbiota sample.
  • 5. A method of treating inflammatory bowel disease in a patient comprising: (1) performing an assay to determine the presence and severity of the inflammatory bowel disease of the patient, the assay comprising (i) measuring the level of Atopobium parvulum in a gut microbiota sample from the patient, wherein the measuring comprises using quantitative polymerase chain reaction, wherein the quantitative polymerase chain reaction uses a forward and reverse primer for targeting A. parvulum, and wherein the forward primer is SEQ ID 1 and the reverse primer is SEQ ID 2, and(ii) comparing the measured level of A. parvulum to a predetermined level of A. parvulum from gut microbiota samples of healthy human subjects, wherein the measured level of A. parvulum relative to the predetermined level is indicative of the presence and severity of the disease, and(2) administering to the patient a pharmaceutically effective amount of a compound selected from the group consisting of aminosalycylates, immunomodulators, anti-integrins, anti-cytokines, enteral feed programs, steroids, corticosteroids, antibiotics, anti-TNFa, and bismuth, or a combination thereof.
  • 6. The method of claim 5, wherein bismuth is administered.
PCT Information
Filing Document Filing Date Country Kind
PCT/CA2014/050245 3/14/2014 WO 00
Publishing Document Publishing Date Country Kind
WO2014/138999 9/18/2014 WO A
US Referenced Citations (2)
Number Name Date Kind
5225329 Marks Jul 1993 A
20070269813 Dewhirst et al. Nov 2007 A1
Foreign Referenced Citations (2)
Number Date Country
WO 2007056680 May 2007 WO
WO 2013133298 Sep 2013 WO
Non-Patent Literature Citations (28)
Entry
N. Katarzyna et al., Visfatin/PBEF/Nampt and Other Adipocytokines in Inflammatory Bowel Disease, Advances in Clinical and Experimental Medicine 19(3):399-404 ⋅ Apr. 2010.
PCT/CA2015/050992 international search report with related claims.
PCT/CA2015/050992 written opinion.
Alessandro Armuzzi et al., Results of the 2nd scientific workshop of the ECCO (IV): Therapeutic strategies to enhance intestinal healing in inflammatory bowel disease, Journal of Crohn's and Colitis (2012) 6, 492-502.
Bhupinder K. Sandhu et al., Guidelines for the Management of Inflammatory Bowel Disease in Children in the United Kingdom, JPGN 2010;50: S1-S13.
Cary G. Sauer et al., Pediatric Inflammatory Bowel Disease: Highlighting Pediatric Differences in IBD, Gastroenterol Clin N Am 38 (2009) 611-628.
Daniel N. Frank et al., Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases, 13780-13785 □ PNAS □ Aug. 21, 2007 □ vol. 104 □ No. 34.
Dermot P.B. McGovern et al., Genome-wide association identifies multiple ulcerative colitis susceptibility loci, Nat Genet. Apr. 2010; 42(4): 332-337.
E Bentley et al., How could pathologists improve the initial diagnosis of colitis? Evidence from an international workshop, J Clin Pathol 2002;55:955-960.
Francesca Fava, Silvio Danese, Intestinal microbiota in inflammatory bowel disease: Friend of foe?, World J Gastroenterol Feb. 7, 2011; 17(5): 557-566.
Ilseung Cho et al., The human microbiome: at the interface of health and disease, Nature review 13, 260-270 2012.
Jeffrey C. Barrett et al., Genome-wide association defines more than thirty distinct susceptibility loci for Crohn's disease, Nat Genet. Aug. 2008; 40(8): 955-962.
Jimmy et al., Fecal Hydrogen sulfide production in ulcerative colitis, American journal of gastraoenterology, vol. 93 No. 1 1998, 83-87.
Jose C. Clemente et al., The Impact of the Gut Microbiota on Human Health: An Integrative View, Cell 148, Mar. 16, 2012, 1258-1269.
Junjie Qin et al., A human gut microbial gene catalog established by metagenomic sequencing, Nature. Mar. 4, 2010; 464(7285): 59-65.
Kaminska B et al., Colonic microflora in inflammatory bowel disease, Pediatria Wspolozesna, vol. 7 issue 3 2005 167-170.
Kim SC et al., Dual-association of gnotobiotic IL-10-/- mice with 2 nonpathogenic commensal bacteria induces aggressive pancolitis, Inflammatory Bowel Diseases [2007, 13(12):1457-1466].
Manichanh C et al., The gut microbiota in IBD, Nat Rev Gastroenterol Hepatol. Oct. 2012;9(10):599-608, abstract.
Marcin Imielinski et al., Common variants at five new loci associated with early-onset inflammatory bowel disease, Nat Genet. Dec. 2009; 41(12): 1335-1340.
Mekki Medani et al., Emerging Role of Hydrogen Sulfide in Colonic Physiology and Pathophysiology, Inflamm Bowel Dis ⋅ vol. 17. No. 7. Jul. 2011.
Nicholas J. Talley et al., An Evidence-Based Systematic Review on Medical Therapies for Infl ammatory Bowel Disease, Am J Gastroenterol 2011; 106:S2-S25; doi: 10.1038/ajg.2011.58.
PCT/CA2014/050245 international preliminary report.
PCT/CA2014/050245 international search report.
PCT/CA2014/050245 search strategy.
Rajaratnam Rameshshanker et al., Endoscopy in inflammatory bowel disease when and why ,World J Gastrointest Endosc Jun. 16, 2012; 4(6): 201-211.
Susanna Nikolaus et al., Diagnostics of Inflammatory Bowel Disease, Gastroenterology 2007;133:1670-1689.
Tighe MP et al., Nutrition and inflammatory bowel disease: primary or adjuvant therapy, Current Opinion in Clinical Nutrition and Metabolic Care [2011, 14(5):491-496] Abstract.
Wendy S. Garrett et al., Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system, Cell. Oct. 5, 2007; 131(1): 33-45.
Related Publications (1)
Number Date Country
20160032363 A1 Feb 2016 US
Provisional Applications (1)
Number Date Country
61781564 Mar 2013 US