METHODS FOR DIAGNOSING AND TREATING INFLAMMATORY BOWEL DISEASE

Abstract
Methods and materials are disclosed for testing biomarkers in a subject suffering from inflammatory bowel disease (IBD) are described herein. Such detection can be useful for diagnosing and treating ulcerative colitis (UC) and Crohn's disease (CD), two forms of IBD that are otherwise difficult to distinguish. The method includes measuring the level of one or more of several biomarkers, including HD5 or MMP-7, which are expressed differentially in patents with UC and CD. A treatment may be based on the determination of whether the subject has ulcerative colitis or Crohn's disease.
Description
BACKGROUND OF THE DISCLOSURE

Inflammatory bowel disease (“IBD”) is the chronic relapsing inflammation of all, or part of, the digestive tract. There are two types of IBD, ulcerative colitis (“UC”) and Crohn's disease (“CD”). Crohn's disease involving only colon is termed as Crohn's colitis (“CC”). When nondefinitive evaluations have been established for criteria for either UC or CC are labeled as “indeterminate colitis (IC)”. UC results in inflammation and ulceration of the mucosal and, to a lesser degree, the submucosal linings of the colon and rectum. CC differs from UC in that it may result in inflammation deeper within all the four colonic layers (transmural inflammation and skip lesions). Furthermore, CC may also affect other organs through fistulation.


UC and CD affect an estimated 2 million people in the US alone with associated annual health care costs of over $6.8 billion. While UC and CD are both types of IBDs, differences between patients having UC or CD has major implications. Currently, clinicians use inexact combined classification for patients having IBD, which include clinical, endoscopy, radiological, and histopathology in an effort to diagnose CD and UC. Nonetheless, differentiating patients having UC or CD among patients suffering from IBD remains challenging, so much so that cases of patients having IBD that are difficult classify as UC or CD are classified as having indeterminate colitis (“IC”). A significant subgroup of IBD patients are misdiagnosed or have a correct diagnosis delayed despite use of a state-of-the-art classification system applying clinical, endoscopic, radiologic and histologic tools. Indeed, it is estimated that 30% of patients suffering from IBD cannot currently be accurately diagnosed as CD or UC.


In addition, 15% of colonic IDB cases that undergo ileal pouch anal anastomosis surgery, as they are diagnosed with UC, will subsequently have their original diagnosis changed to CD based on their postoperative follow-up visits, clinical and histopathology changes, and development of de novo CD in the ileal pouch. Ileal pouch anal anastomosis, a treatment normally suitable for UC but not CD, restores gastrointestinal continuity after surgical removal of the colon and rectum, and involves the creation of a pouch of small intestine to recreate the removed rectum.


Implications of distinguishing cases of UC and CD include choice of medical treatment, timing of surgery, prognosis, whether to offer the patient an ileal pouch anal anastomosis, and lifestyle expectations. For these reasons, there is a need for improving the diagnosis, and subsequent treatment, of subjects having IBD.


SUMMARY

It has been discovered that Paneth cells secreted DEFA5 also known as HD5 serve as biomarkers for determining whether a patient suffering from IBD has UC or CD.


In a first aspect, a method of measuring DEFA5 (HD5) in a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; and measuring at least one of the expression of DEFA5 (HD5) and the concentration of DEFA5 (HD5) in the sample.


In a second aspect, a method of treating a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; measuring at least one of the expression of DEFA5 (HD5) and the concentration of DEFA5 (HD5) in the sample; and performing an intervention on the patient to treat one of Crohn's disease and ulcerative colitis.


In a third aspect, a method of measuring MMP-7 in a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; and measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample.


In a fourth aspect, a method of treating a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample; and performing an intervention on the patient to treat one of Crohn's disease and ulcerative colitis.


In a fifth aspect, a method of measuring biomarkers in a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; measuring at least one of the expression of DEFA5 (HD5) and the concentration of DEFA5 (HD5) in the sample; and measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample.


In a sixth aspect, a method of treating a patient suffering from or at risk of IBD is disclosed, said method comprising: obtaining a sample from the patient; measuring at least one of the expression of DEFA5 (HD5) and the concentration of DEFA5 (HD5) in the sample; measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample; and performing an intervention on the patient to treat one of Crohn's disease and ulcerative colitis.


In a seventh aspect, a kit for measuring DEFA5 (HD5) and MMP-7 in a sample is disclosed, the kit comprising: a first assay for measuring at least one of the expression of human DEFA5 (HD5) and the concentration of human DEFA5 (HD5) in a sample; and a second assay for measuring at least one of the expression of human MMP-7 and the concentration of human MMP-7 in a sample.


In an eighth aspect, a method of measuring a biomarker in a patient suffering from or at risk of inflammatory bowel disease (IBD) is disclosed, said method comprising: obtaining a sample from the patient; and measuring a level of the biomarker in the same, the level of the biomarker selected from the group consisting of: the expression of the biomarker, the activity of the biomarker, and the concentration of the biomarker; wherein said biomarker is selected from Table 1.


The above methods may include diagnosing the patient as suffering from CD if the level of DEFA5 (HD5) concentration or DEFA5 (HD5) expression is greater than a given threshold level, diagnosing the patient as suffering from UC if the level of DEFA5 (HD5) concentration or DEFA5 (HD5) expression is below a threshold level, or both.


The above methods may include diagnosing the patient as suffering from CD if the level of MMP-7 concentration or MMP-7 expression is less than a given threshold level, diagnosing the patient as suffering from UC if the level of MMP-7 concentration or MMP-7 expression is above a threshold level, or both.


The above presents a simplified summary in order to provide a basic understanding of some aspects of the claimed subject matter. This summary is not an extensive overview. It is not intended to identify key or critical elements or to delineate the scope of the claimed subject matter. Its sole purpose is to present concepts in a simplified form as a prelude to the more detailed description that is presented later.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A and 1B illustrate a comparison of new diagnoses according to an embodiment of an assay method of the present invention with previous attending physical diagnoses.



FIGS. 2A-H show differential expression and concentration of DEFA5 (HD5) in CC and UC subjects. 2A is a bar graph showing transcription levels of DEFA5 (HD5) in subjects suffering from moderate UC as compared to moderate CC. 2B is a western blot analysis of DEFA5 (HD5) levels in diverticulitis, UC, and CC subjects. 2C is a scatter plot graph quantifying the western blot DEFA5 (HD5) levels. 2D-G is histological staining of DEFA5 (HD5) tissue samples from control, diverticulitis, UC, and CC subjects, respectively. 2H is a bar graph showing quantified DEFA5 (HD5) staining counts of UC and CC subjects as compared to the control.



FIG. 3 is a bar graph showing differentially expressed MMP-7 in UC subjects as compared to CC subjects.



FIG. 4 is a western blot analysis of MMP-7 levels in diverticulitis, UC, and CC subjects.



FIG. 5 is a plot graph quantifying the western blot MMP-7 levels of FIG. 4.



FIGS. 6A-G show a series of plot graphs quantifying and comparing western blot DEFA5 (HD5) and MMP-7 levels in control, diverticulitis, UC, and CC subjects. 6A=diverticulitis, 6B=mild UC, 6C=moderate UC, 6D=severe UC, 6E=mild CC, 6F=moderate CC, and 6G=severe CC.



FIG. 7 shows the sequence details of canonical human HD5 protein.



FIGS. 8A-D illustrate that it is possible to use DEFA5 (HD5) to determine patient candidacy for IPAA. 8A=Representative results from a RPC-operated patient that did not change the diagnosis after surgery and was molecularly tested using DEFA5 (HD5) IHC. 8B=Representative results from a UC RPC and IPAA operated patients that did change the diagnosis from UC to de novo Crohn's was molecularly tested using DEFA5 (HD5) IHC. C. NL-Ileum, control. 8D=Quantification of NEARAS DEFA5 (HD5) IHC staining spot counts for UC RPC and IPAA-operated patients who did not have their original diagnosis changed versus those who did change from UC to de novo Crohn's. (Ctrl 1 Destaining control, UC=Ulcerative Colitis, CC=Crohn's Colitis, DV=Diverticulitis, DVL=Diverticulosis).



FIGS. 9A-I illustrate H&E staining on parallel sections the typical morphological appearance of Paneth cell (PCs) including the presence of dense apical eosinophilic granules. Upper panel: 9A, Diverticulitis (DV, no PCs), 9B, Diverticulosis (DVL, no PCs), 9C, Normal (NL-Colon, Control, no PCs). Middle panel: 9D, UC (found prodromal PC in one patient, arrow). 9E, CC, demonstrate abundance of PCs allover colonic basal crypts (arrows). 9F, Normal (NL-Ileum, Control), with abundance of PCs. Lower panel: IHC detection of Paneth cell markers α-defensin 5 (DEFA5) and lysozyme (LYZ) in the colon. 9G, NL-Colon, 9H, CC, and 9I, NL-Ileum, Control.



FIGS. 10A-J illustrate a double stain of PCs, lysosomes and DEFA5 (HD5). Double staining analyses from de novo Crohn's (10A and 10D), and normal ileum/control (10G) are presented. Image deconvolutions are displayed vertically to evaluate lysozyme-specific permanent red (10B, 10E and 10H) and HD5α-specific DAB (FIGS. 10C, 10F and 10I). The normal colon image (FIG. 10J), which lacks PCs, was not further processed.



FIGS. 11A-D illustrate an assessment of DEFA5 (HD5) and Paneth cells in inflamed and normal, adjacent tissue. DEFA5 (HD)5 staining of CC inflamed and normal, adjacent tissue shows expression of DEFA5 (HD5) in all patient samples examined (FIG. 11A), compared to inflamed and adjacent, normal tissue of UC patients (FIG. 11B).H&E stains for Paneth Cells (FIGS. 11C and 11D), were negative for PCs in all tissues.



FIGS. 12A-D show histological staining of colon tissue in various subjects. 12A=CC, 12B=UC, 12C=diverticulitis, 12D=normal colon.



FIG. 13 illustrates the results of an antibody specificity assay. Dot blots were performed on recombinant HD1-6 with various commercial antibodies to determine specificity to HD5. Ponceau S Stain is used as a loading control. We found that the antibody from Santa Cruz was the most specific for DEFA5 (HD5) of those tested.



FIG. 14 is a condensed list of the samples included in all experiments and the colon locations from which the samples were taken.



FIG. 15 shows an assessment of levels of HD5 in surgical pathology colectomy samples via IHC in patients described in FIG. 1A.





DETAILED DESCRIPTION
Definitions

Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art of this disclosure. It will be further understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the specification and should not be interpreted in an idealized or overly formal sense, unless expressly so defined herein. Well-known functions or constructions may not be described in detail for brevity or clarity.


The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. As used herein, the singular forms “a”, “an”, and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise.


The term “consisting essentially of” means that, in addition to the recited elements, what is claimed may also contain other elements (steps, structures, ingredients, components, etc.) that do not adversely affect the operability of what is claimed for its intended purpose as stated in this disclosure. This term excludes such other elements that adversely affect the operability of what is claimed for its intended purpose as stated in this disclosure, even if such other elements might enhance the operability of what is claimed for some other purpose.


The terms “about” and “approximately” shall generally mean an acceptable degree of error or variation for the quantity measured given the nature or precision of the measurements. Typical, exemplary degrees of error or variation are within 20%, preferably within 10%, and more preferably within 5% of a given value or range of values. For biological systems, the term “about” refers to an acceptable standard deviation of error, preferably not more than 2-fold of a given value. Numerical quantities in this detailed description are approximate unless stated otherwise, meaning that the term “about” or “approximately” can be inferred when not expressly stated.


The terms “individual”, “subject”, or “patient” as used herein refer to any animal, including mammals, such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. The terms may specify male or female or both, or exclude male or female.


The terms “treatment”, “treat”, and “treating”, as used herein, refer to a course of action (such as administering a compound or pharmaceutical composition) initiated after the onset of a clinical manifestation of a disease state or condition so as to eliminate or reduce such clinical manifestation of the disease state or condition. Such treating need not be absolute to be useful.


The terms “first”, “second”, and the like are used herein to describe various features or elements, but these features or elements should not be limited by these terms. These terms are only used to distinguish one feature or element from another feature or element. Thus, a first feature or element discussed below could be termed a second feature or element, and similarly, a second feature or element discussed below could be termed a first feature or element without departing from the teachings of the present disclosure.


Methods of Treatment and Diagnosis

An assay method of diagnosing UC and CD in a subject, such as a human, suffering from IBD is described. The method measures DEFA5 (HD5) in tissue taken from a subject having IBD. DEFA5 (HD5) is a small, microbicidal innate immune system protein belonging to the alpha defensing family of mammalian defensing peptides. DEFA5 (HD5) is expressed in various tissues and particularly on mucosal surfaces. DEFA5 (HD5) is encoded by the gene DEFA5. DEFA5 (HD)5 is involved in host defense mechanisms, and is highly expressed in secretory granules of Paneth cells of the small intestine (ileum). Like most secreted proteins, HD5 is synthesized as prepro-HD5 (1-94) that undergoes proteolytic processing first, to the inactive pro-HD5s (20-94), HD5 (23-94) and HD5 (29-94). HD5 (23-94) and HD5 (29-94) are found within tissues, while HD5 (20-94) is the predominant intracellular form. The pro-HD5s are then processed to two active or mature forms. HD5 (56-94) and HD5 (63-94) with HD5 (63-94) being the most abundant form. These mature forms of HD5 are cysteine-rich host defense peptides which exert a broad-spectrum antimicrobial activity and contribute to innate immunity in the human gut. As used herein, HD5 may refer to exclusively mature forms or inactive forms of HD5.


Matrix metalloproteinase-7 (MMP-7, encoded by the MMP7 gene) is responsible for cleaving and activating HD5. It is believed that there may be a dysfunction in the activation pathway of HD5 in patients suffering from moderate and severe CD, and thus, an excess amount of inactive form HD5 is a potential mechanism for inflammation in patients suffering from CD. This excessive amount of inactive form HD5 may cause increased damage to the epithelial lining and potentially even a dysregulation in the levels and make-up of gut flora. The canonical structure of human MMP-7 is a 54 residue polypeptide (see Uniprot accession number A5GZ72).


The sample can be taken from any suitable source for measuring HD5 concentration, HD5 expression levels, MMP-7 expression, or MMP-7 concentration, such as the tissue samples from the large intestine or rectum. In this disclosure the term “expression of HD5” should be interpreted to mean the expression of the DEFA5 gene; “levels of HD5” should be interpreted to mean the concentration of HD5; “expression of MMP-7” should be interpreted to mean the expression of the MMP7 gene; “levels of MMP-7” should be interpreted to mean the concentration of matrix metalloproteinase-7.


The sample may be taken from a subject who is suffering from or at risk of IBD. The subject may display one or more symptoms characteristic of IBD, such as severe diarrhea, abdominal pain, fatigue, and weight loss. In some embodiments of the method, the subject displays more than one of said symptoms. In further embodiments the subject displays 2, 3, or 4 of said symptoms.


It has been discovered that the genes encoding HD5 and MMP-7 are differentially expressed in subjects having UC and CD; and further, that the concentration of HD5 is significantly higher in tissues of CD patients than in UC patients. Used in this way, HD5 and MMP-7 concentrations, and HD5 expression and MMP-7 expression, can be utilized and measured as biomarkers for distinguishing UC and CD in patients having IBD. This can in turn be used to more effectively treat the subject. For example, as ileal pouch anal anastomosis is clinically much more successfully in patients having UC than in patients suffering from CD, patients identified as having levels of HD5 or MMP-7 levels indicative of UC, or not having CD, may be treated with ileal pouch anal anastomosis. Indeed, as HD5 is produced by Paneth cells only, one would not typically expect to find Paneth cells that secret HD5 in the colon. The inventors have discovered Paneth cells (secreting HD5) are abundantly found in subjects having UC. On the other hand, patients identified as having levels of HD5 or MMP-7 and HD5 or MMP-7 expression indicative of CD may be treated with any suitable treatment for CD. In an embodiment, a diagnosing step, such as diagnosing a subject with UC or CD, is optional.


The methods may include a step of comparing the level of the biomarker in question to a benchmark value. The benchmark value may be a measure of central tendency based on levels observed in one or more populations of subjects that are established to be unafflicted by either of UC or CD. For example, the benchmark value may be a mean level of the gene expression or protein concentration observed in samples from a population of subjects who are unafflicted by UC, unafflicted by CD, or both. The population may be defined by one or more of the patient's geography, age, ethnicity, sex, and medical history. The benchmark value may take into account a measure of variation combined with a measure of central tendency. For example, the benchmark value may be a mean level of the gene expression or protein concentration observed in a given tumor population, plus or minus a margin of error. The benchmark may be based on raw measurements (such as fragments of mRNA or cDNA per kb gene length per million reads) or normalized measurements (such as % of normal expression, or expression compared to a constitutively expressed or widely expressed gene with generally consistent expression, such as β-actin).


The benchmark may also be established by analysis of a control sample that is measured alongside the sample from the subject. Examples of suitable control samples are: a sample from a subject unafflicted with UC, a sample from a subject unafflicted with CD, a sample from a subject afflicted with UC (although unafflicted with CD), a sample from a subject afflicted with CD (although unafflicted with UC), a sample from a subject afflicted with diverticulitis (although unafflicted with either of UC or CD), and a sample from a subject unafflicted from IBD.


In an embodiment, an assay method of differentially diagnosing UC and CD in a patient suffering from IBD includes measuring the level of HD5 or MMP-7 or HD5 or MMP-7 expression present in a sample obtained from the patient. The level of HD5 or MMP-7 concentration or expression in the tissue may be measured by any suitable peptide analysis. For example, the measuring step may include one or more of enzyme-linked immunosorbent assay (ELISA), cation-ion exchange, NMR analysis, genome-wide transcriptome analysis, and mass spectrometry. The method may include comparing the concentration or expression of the biomarker in the sample to the benchmark, and making a diagnosis if the concentration or expression of the biomarker in the sample is significantly less than or significantly greater than the benchmark value. For the example, the method may comprise comparing the concentration or expression of HD5 in the sample to the benchmark, and making a diagnosis of CD if the concentration or expression of HD5 in the sample is significantly greater than the benchmark value. As another example, the method may comprise comparing the concentration or expression of HD5 in the sample to the benchmark, and making a diagnosis of UC if the concentration or expression of HD5 in the sample is not significantly greater than the benchmark value. As another example, the method may comprise comparing the concentration or expression of MMP-7 in the sample to the benchmark, and making a diagnosis of UC if the concentration or expression of MMP-7 in the sample is significantly greater than the benchmark value. As another example, the method may comprise comparing the concentration or expression of MMP-7 in the sample to the benchmark, and making a diagnosis of CD if the concentration or expression of MMP-7 in the sample is not significantly greater than the benchmark value. In a further example, the method comprises measuring the concentration or expression of both MMP-7 and HD5, and making a diagnosis of either: CD if the concentration or expression of MMP-7 in the sample is not significantly greater than the benchmark value and the concentration or expression of HD5 is significantly greater than the benchmark value; or UC if the concentration or expression of MMP-7 in the sample is significantly greater than the benchmark value and the concentration or expression of HD5 is not significantly greater than the benchmark value.


The difference in expression or concentration may be considered significant based on any of a variety of known statistical tests for significance. These are generally based on a collection of measurements made from a sampled population, and are affected by both the population size and the sampling size. Such statistical tests are well known in the art and are not further elaborated upon in this disclosure; outside references can be relied upon to enable those skilled in the art to determine statistical significance, such as Rosener's Fundamentals of Biostatistics, 8th ed. (2015), Cengage Learning, Boston, Mass.


The method may include diagnosing the patient as having UC if HD5 or HD5 expression is at any level that is indicative of a patient not having CD, such as less than 5× normal levels of HD5 (i.e., levels of HD5 typical of a subject unafflicted with CD), less than about 5×-30× normal levels of HD5 or HD5 expression, less than about 31× normal levels, or less than about 118× normal levels. In a further embodiment, the patient is diagnosed as having UC if HD5 expression is at a level of less than 106, 107, 1.9×107, 6×105, or 3×106 HD5 mRNA transcript per 10 ng RNA. In some embodiments of the method the patient may be diagnosed as having CD if the level of HD5 expression is at any level indicative of a patient having CD, such as at least 3×106, 107, 1.9×107, 7×107, 108, 1.2×108, or from about to 3×106 to 1.2×108 HD5 mRNA Transcript per 10 ng RNA. The diagnosing may diagnose the patient as having CD if the patient has a MMP-7 concentration or MMP-7 expression level indicative of a patient having CD, such as up to a threshold limit that is 10× a benchmark value of MMP-7 concentration or MMP-7 expression. In further embodiments, the diagnosis may be CD using a threshold limit of up to 5×, and up to 1× a benchmark value. As used herein, a “normal level” of HD5 or HD5 expression means a level of HD5 or HD5 expression in the digestive tract tissue from a subject not having CD or UC, or a subject suffering from IBD and specifically UC. Normal HD5 expression may refer to from 1×105 to 9×105 HD5 mRNA Transcript per 10 ng RNA, or about 6×105 HD5 mRNA Transcript per 10 ng RNA. As used herein, a “normal level” of MMP-7 or MMP-7 expression means a level of MMP-7 or MMP-7 expression in the digestive tract tissue from a subject not having CD or UC, or a subject suffering from IBD, specifically CD.


In another embodiment, an assay method for differentially diagnosing UC and CD in a patient suffering from, or at risk of, IBD includes measuring the level of MMP-7 or MMP-7 expression present in a sample obtained from the patient. The level of MMP-7 or MMP-7 expression in the tissue may be measured by any suitable peptide analysis. In an embodiment, the method of diagnosing may be performed ex vivo.


In one embodiment, the assay methods involve determining the status of a subject with respect to the activity and/or expression of HD-5 or MMP-7 or the activity and/or expression of a polypeptide regulated by HD-5 or MMP-7. In one embodiment, such methods comprise determining the level of expression or activity of HD-5 or MMP-7 or a polypeptide regulated by HD-5 or MMP-7 in a sample from the subject. The method may further comprise collecting the sample from the subject. As used herein, a biological sample which is subjected to testing is a sample derived from a subject and includes, but is not limited to, any biological material, such as a bodily fluid. Examples of bodily fluids include, but are not limited to, whole blood, serum, saliva, tissue infiltrate, pleural effusions, lung lavage fluid, bronchoalveolar lavage fluid, and the like. The biological fluid may be a cell culture medium or supernatant of cultured cells. For example, the sample can be a blood sample or a serum sample. As another example the sample may be tissue or fluids from the subject's digestive tract. Some embodiments of the method involve a sample of intestinal tissue. In specific embodiments, the biological sample is collected from the colon of a subject (such as colonic tissue) or the ileum of a subject (such as ileal tissue).


Some embodiments of the method comprise measuring the concentration of the biomarker protein by selectively staining or dying the sample form the subject and measuring the signal from the stain. The stain or dye may comprise an antibody or an antibody fragment to recognize the protein. The stain or dye may also comprise a reporter, such a colorimetric group, a radionuclide, a stable isotope, a fluorophore, a chromophore, an enzyme, a magnetic particle, and a quantum dot. The concentration of the protein can then be measured by observing the signal from the reporter, such as by microscopy, colorimetry, radiometry, fluoroscopy, magnetotaxis, or any combination of the foregoing. In a specific embodiment of the method, the concentration of HD5 or MMP-7 is measured by immunostaining the sample with an immunostain that recognizes the biomarker and counting the number of stained cells by microscopy. This approach has the advantage of relative simplicity, and only requires the types of equipment that are already present in typical clinical laboratories. In specific examples in which the biomarker is HD5, a diagnosis can be made based on a threshold number of cells that stain positive, such as 10%, 20%, and 30%. If the number of HD5 stained cells is significantly above the threshold value, than a diagnosis of CD can be made; whereas if the number of HD5 stained cells is significantly below the threshold value, than a diagnosis of UC can be made.


Those subjects in which HD-5 or MMP-7 activity and/or expression differs (increased or decreased) from a control or benchmark value or the activity of a polypeptide regulated by HD-5 or MMP-7 differs as compared to a control or benchmark value are determined to be suffering from or at risk for a disease states and conditions associated with or characterized by increased or decreased HD-5 or MMP-7 activity.


Assay techniques that can be used to determine levels of expression or activity in a sample are known. Such assay methods include, but are not limited to, radioimmunoassays, reverse transcriptase PCR (RT-PCR) assays, immunohistochemistry assays, in situ hybridization assays, competitive-binding assays, Western Blot analyses, ELISA assays and proteomic approaches, two-dimensional gel electrophoresis (2D electrophoresis) and non-gel based approaches such as mass spectrometry or protein interaction profiling. Assays also include, but are not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, enzyme immunoassays (EIA), enzyme linked immunosorbent assay (ELISA), sandwich immunoassays, precipitin reactions, gel diffusion reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and immunoelectrophoresis assays. For examples of immunoassay methods, see U.S. Pat. Nos. 4,845,026 and 5,006,459.


In an ELISA assay, an antibody is prepared, if not readily available from a commercial source, specific to an antigen, such as, for example, HD-5 or MMP-7 or a polypeptide regulated by HD-5 or MMP-7. In addition, a reporter antibody generally is prepared. The reporter antibody is attached to a detectable reagent such as a radioactive, fluorescent, or enzymatic reagent, for example horseradish peroxidase enzyme or alkaline phosphatase. In one embodiment of the ELISA, to carry out the ELISA, antibody specific to the antigen is incubated on a solid support that binds the antibody. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein. Next, the sample to be analyzed is incubated with the solid support, during which time the antigen binds to the specific antibody. Unbound sample is washed out with a buffer. A reporter antibody specifically directed to the antigen and linked to a detectable reagent is introduced resulting in binding of the reporter antibody to any antibody bound to the antigen. Unattached reporter antibody is then washed out. Reagents for detecting the presence of the reporter antibody are then added. The detectable reagent is then determined in order to determine the amount of antigen present. In an alternate embodiment, the antigen is incubated with the solid support, followed by incubation with one or more antibodies, wherein at least one of the antibodies comprises a detectable reagent. Quantitative results may be obtained by reference to a standard curve.


Optionally, a genetic sample from the biological sample can be obtained. The genetic sample comprises a nucleic acid, preferably RNA and/or DNA. For example, in determining the expression of genes mRNA can be obtained from the biological sample, and the mRNA may be reverse transcribed into cDNA for further analysis. Alternatively, the mRNA itself is used in determining the expression of genes. A genetic sample may be obtained from the biological sample using any techniques known in the art (Ausubel et al. Current Protocols in Molecular Biology (John Wiley & Sons, Inc., New York, 1999); Molecular Cloning: A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch, and Maniatis (Cold Spring Harbor Laboratory Press: 1989); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984) each of the foregoing being incorporated herein by reference). The nucleic acid may be purified from whole cells using DNA or RNA purification techniques. The genetic sample may also be amplified using PCR or in vivo techniques requiring subcloning. The genetic sample can be obtained by isolating mRNA from the cells of the biological sample and reverse transcribing the RNA into DNA in order to create cDNA (Khan et al. Biochem. Biophys. Acta 1423:17 28, 1999).


Once a genetic sample has been obtained, it can be analyzed. The analysis may be performed using any techniques known in the art including, but not limited to, sequencing, PCR, RT-PCR, quantitative PCR, restriction fragment length polymorphism, hybridization techniques, Northern blot, microarray technology, and similar techniques. In determining the expression level of a gene or genes in a genetic sample, the level of expression may be normalized by comparison to the expression of another gene such as a well-known, well characterized gene or a housekeeping gene (for example, actin). For example, reverse-transcriptase PCR (RT-PCR) can be used to detect the presence of a specific mRNA population in a complex mixture of thousands of other mRNA species. Hybridization to clones or oligonucleotides arrayed on a solid support (e.g., gridding) can be used to both detect the expression of and quantitate the level of expression of that gene. In this approach, a cDNA encoding an antigen is fixed to a substrate. The substrate may be of any suitable type including but not limited to glass, nitrocellulose, nylon, or plastic. At least a portion of the DNA encoding the antigen is attached to the substrate and then incubated with the analyte, which may be RNA or a complementary DNA (cDNA) copy of the RNA, isolated from the sample of interest. Hybridization between the substrate bound DNA and the analyte can be detected and quantitated by several means including but not limited to radioactive labeling or fluorescence labeling of the analyte or a secondary molecule designed to detect the hybrid. Quantitation of the level of gene expression can be done by comparison of the intensity of the signal from the analyte compared with that determined from known standards. The standards can be obtained by in vitro transcription of the target gene, quantifying the yield, and then using that material to generate a standard curve.


The method may include diagnosing the patient as having UC if MMP-7 concentration or MMP-7 expression is at any level indicative of a patient having UC, such as from 2×-100×, 10×, 2×-50×, 5×-15×, or about 10× normal MMP-7 concentration or MMP-7 expression levels. In some embodiments of the method, the patient may be diagnosed as having CD if the MMP-7 concentration or MMP-7 expression is at any level indicative of a patient having CD, such as less than 1×-10× or 2×-5× normal levels of MMP-7 or MMP-7 expression.


A method of treating IBD in a patient suffering from IBD may include: (a) measuring the level of HD5 or HD5 expression present in a sample obtained from the patient, said measuring step optionally comprising one of cation-ion exchange, NMR analysis, genome-wide transcriptome analysis, and mass spectrometry, whereby a level of HD5 or HD5 expression is obtained; (b) if the level of HD5 or HD5 expression is at a level indicative of a patient not having CD, treating the IBD in the patient with a suitable medical treatment for UC; if the level of HD5 or HD5 expression is at a level indicative of a patient having CD, treating the IBD in the patient with a suitable medical treatment for CD. In another embodiment, levels of MMP-7 or MMP-7 expression are measured rather than levels of HD5 to determine whether to treat UC or CD.


Suitable medical treatments for UC include ileal pouch anal anastomosis or the administration of pharmaceutical agents or salts thereof. Suitable pharmaceutical agents may be one or more of: an iron supplement; an oral 5-aminosalicylate, such as mesalamine, balsalazide and olsalazine; an anti-inflammatory; a corticosteroid; an immunosuppressant such as azathioprine, mercaptopurine, methotrexate, and cyclosporine; an anti-TNF-alpha antibody such as infliximab, adalimumab, and golimumab; an anti-α4-integrin antibody such as vedolizumab; and an antibacterial antibiotic, such as ciprofloxacin and metronidazole. Surgeries that are sometimes used to treat UC include a proctocolectomy, and an ileal pouch anal anastomosis. Note that ileal pouch anal anastomosis are recognized as relatively ineffective when used to treat CD, in contrast to UC. It should also be noted that cyclosporine and golimumab, while currently approved for the treatment of UC in the United States, are not currently approved for the treatment of CD. Some embodiments of the method involve performing an intervention that is effective to treat UC, but either ineffective to treat CD or not yet approved by regulatory authorities for the treatment of CD.


Suitable medical treatments for CD include the administration of pharmaceutical agents or salts thereof. Suitable pharmaceutical agents include: an oral 5-aminosalicylate, such as mesalamine; a vitamin supplement, such as a vitamin B-12 supplement and a vitamin D supplement; a mineral supplement, such as a calcium supplement; an anti-inflammatory; a corticosteroid such as prednisone and budesonide; an immunosuppressant such as azathioprine, tacrolimus, methotrexate, and mercaptopurine; an anti-TNF-α antibody, such as infliximab, adalimumab, and certolizumab pegol; an anti-α-4-integrin antibody, such as natalizumab and vedolizumab; an anti-interleukin antibody, such as ustekinumab; and an antibacterial antibiotic, such as metronidazole, and ciprofloxacin. Although certolizumab pegol, methotrexate, and natalizumab are approved in the US for the treatment of CD, they are not currently approved for the treatment of UC. Surgical approaches are sometimes used to treat severe cases of CD. Such surgeries include ostomy, colostomy, ileostomy, bowel resection, colectomy, proctocolectomy, and strictureplasty. In some embodiments of the method, the subject is treated using a diet that is advantageous for the management of CD, but not necessarily advantageous in the management of UC. One such diet is a low fat diet. Some embodiments of the method involve performing an intervention that is effective to treat CD, but either ineffective to treat UC or not yet approved by regulatory authorities for the treatment of UC.


In an embodiment, the level of HD5 or HD5 expression may be elevated above normal levels in patients who are likely to be diagnosed UC but, at the time the HD5 or HD5 expression level is measured, diagnosed as having IC. These patients may be treated with any suitable medical treatments for UC.


A kit is provided for measuring HD5 in a subject. The kit may include a detectable antibody that is capable of binding with HD5. The antibody may be capable of binding with HD5 yet not bind with other defensins, such as HD4 and HD5. The antibody may be a purified HD5-specific monoclonal or polyclonal antibody, such as the HDAC5 Antibody from GenWay Biotech, Inc. FIG. 7 shows the amino acid sequence of HD5, including a schematic showing HD5 antibody epitopes to distinguish pro-HD5 from mature HD5. FIG. 7 also shows the alignment of the primary sequence of HD5 with that of HD1 showing the differences between the two polypeptides. The methods herein can involve detecting any protein having the consensus sequence of HD5 such as to account for natural variation of HD5 in humans having different HD5 sequences.


A kit is provided for measuring HD5 and MMP-7 in a subject. The kit may find use in several of the methods provided above, as well as others. The kit may be, for example, used for the diagnosis of inflammatory bowel disease. The kit comprises an assay for measuring at least one of HD5 concentration and HD5 expression; and an assay for measuring at least one of MMP-7 concentration and MMP-7 expression.



FIG. 12 illustrates a histological staining of colon tissue in a subject having CC. FIG. 12B illustrates a histological staining of colon tissue in a subject having UC. FIG. 12C illustrates a histological staining of colon tissue in a subject having Diverticulitis. FIG. 12D illustrates a histological staining of colon tissue in a subject having a normal colon.


Working Example 1

Abstract


Inability to distinguish Crohn's colitis from ulcerative colitis leads to the diagnosis of indeterminate colitis. This greatly effects medical and surgical care of the patient because treatments for the two diseases vary. Approximately 30 percent of inflammatory bowel disease patients cannot be accurately diagnosed, increasing their risk of inappropriate treatment. We sought to determine whether transcriptomic patterns could be used to develop diagnostic biomarker(s) to delineate inflammatory bowel disease more accurately. Four patients groups were assessed via whole-transcriptome microarray, qPCR, Western blot, and immunohistochemistry for differential expression of Human α-Defensin-5. In addition, immunohistochemistry for Paneth cells and Lysozyme, a Paneth cell marker, was also performed. Aberrant expression of Human α-Defensin-5 levels using transcript, Western blot, and immunohistochemistry staining levels was significantly upregulated in Crohn's colitis, p<0.0001. Among patients with indeterminate colitis, Human α-Defensin-5 is a reliable differentiator with a positive predictive value of 96 percent. We also observed abundant ectopic crypt Paneth cells in all colectomy tissue samples of Crohn's colitis patients. In a retrospective study, we show that Human α-Defensin-5 could be used in indeterminate colitis patients to determine if they have either ulcerative colitis (low levels of Human α-Defensin-5) or Crohn's colitis (high levels of Human α-Defensin-5). Twenty of 67 patients (30 percent) who underwent restorative proctocolectomy for definitive ulcerative colitis were clinically changed to de novo Crohn's disease. These patients were profiled by Human α-Defensin-5 immunohistochemistry. All patients tested strongly positive. In addition, we observed by both hematoxylin and eosin and Lysozyme staining, a large number of ectopic Paneth cells in the colonic crypt of Crohn's colitis patient samples. Our experiments are the first to show that Human α-Defensin-5 is a potential candidate biomarker to molecularly differentiate Crohn's colitis from ulcerative colitis, to our knowledge. These data give us both a potential diagnostic marker in Human α-Defensin-5 and insight to develop future mechanistic studies to better understand crypt biology in Crohn's colitis.


Methods


Clinical Samples and Ethical Consideration


In order to carryout tissue profiling of differentially expressed proteins/genes in IBD, we first sought ethical approval from the Meharry Medical College (IRB file #: 100916AM206) and Vanderbilt University Medical Center (IRB file #s: 080898 and 100581) Institutional Review Boards [20]. Informed consent was provided, and patient participation in the study was voluntary. Patient samples comprised of surgical pathology colectomy tissues from adults with definitive UC and CC phenotypes as well as those diagnosed with IC at Vanderbilt University Medical Center (VUMC) between 2000 and 2007. The full thickness surgical samples of colectomy tissue were analyzed by pathology teams at MMC and VUMC, Schools of Medicine following established protocol criteria for IBD subtypes. For each selected sample, medical records data on patient demographics, preoperative variables prior to and after time of ileal pouch-anal anastomosis surgery, surveillance endoscopic and clinical findings, and medical and surgical treatment history were reviewed retrospectively. Samples included in all experiments were taken from various parts of the colon; all inflamed tissue unless otherwise indicated. A condensed list of samples and colon locations are included in Table 3, as shown in FIG. 13.


Diagnostic Criteria for Inflammatory Bowel Disease


Pathology teams at MMC and VUMC Schools of Medicine used the following protocol criteria for the final surgical pathology reporting.


For ulcerative colitis. Characteristic pattern of involvement of colon, worse distally in untreated patients; lack of perianal or fistulizing disease; no granulomas, except in association with ruptured/injured crypts; no transmural lymphoid aggregates or other transmural inflammation; no involvement of terminal ileum, except mild “backwash ileitis” in cases with severe cecal involvement and no pyloric metaplasia in terminal ileum.


For Crohn's disease. Involvement of other sites in the gastrointestinal tract (skip lesions, segmental disease); perianal or fistulizing disease; granulomas, not in association with ruptured/injured crypts and terminal ileum involvement.


For indeterminate colitis. Distribution favors UC, but focal transmural inflammation, or inflammation in ileum more than expected in backwash ileitis and no fistulizing disease.


Vanderbilt Patient Medical Records Database


The availability of a detailed IBD patient database registry at Vanderbilt University Medical Center (VUMC) made chart review and follow-up surveillance possible. Medical records data on patient demographics, preoperative variables prior to and after IPAA surgery, surveillance of endoscopic and clinical findings, and medical and surgical treatment history were retrieved retrospectively.


Indeterminate Colitis Clinical Retrospective Study


A retrospective investigation was conducted to identify a cohort of patients diagnosed with IC and registered in the IBD Center at VUMC. Twenty-one patients, initially classified as IC at the time of diagnosis between years 2000-2007, were identified and reevaluated for disease course in 2014, after a mean surveillance follow-up of 8.7±3.7 (range, 4-14) years, in order to identify the rates of diagnosis resolution to UC or CC. Diagnosis for each patient was determined based on standard clinical and pathologic features as previously described [21,22].


Three gastrointestinal pathologists blinded to clinical diagnosis reconciled and confirmed colitis diagnosis for each patient and represented a consensus among treating physicians. Patients who clinically did not changed and maintained the IC diagnosis were tested via IHC and Nikon Element Advanced Research Analysis Software (NEARAS) for HD5 levels to determine if HD5 could be used to identify CC from UC.


Restorative Proctocolectomy Operated Patients' Retrospective Study


One hundred twenty patients with definitive UC underwent RPC surgery between Apr. 18, 2001 and Jun. 18, 2008. Of the 120 patients, 67 had their diagnosis re-evaluated after a mean follow up of 9.4 (range, 6-13) years of functionally acceptable pouches. Compiled medical records allowed us to re-evaluate a progressive course of UC patients following RPC. Clinical information needed for each of these patients was available in the IBD medical records registry database at VUMC. The aim was to reevaluate patients who underwent RPC operation for definitive UC and had a change in diagnosis to de novo Crohn's ileitis. Patients who had a change in diagnosis should reconcile the molecular biometric test that delineates IC into CC; again using NEARAS for HD5 levels.


cDNA Microarray


We performed a whole-transcriptome microarray with RNA extracted and pooled from human full thickness colon samples from UC and CC patients (n=5/group) (Affymetrix, Santa Clara, Calif.).


NanoString nCounter Human Inflammation Kit Gene Expression


RNA from UC and CCI tissue (and diverticulitis tissue used as a control) was processed by NanoString (NanoString Technologies Inc., Seattle, Wash.) to determine gene expression level according to the manufacturer protocol [23].


Real-Time RT-PCR


Real-Time RT-PCR was used to measure transcript levels of HD5. RNA was extracted from three human colon biopsy samples each from moderate UC and CC, and diverticulitis (DV) as a non-IBD control (RNeasy Miniprep Kit, Qiagen, CA). cDNA was generated using iScript cDNA synthesis kit (Bio-Rad, Hercules, Calif.). Pre-designed TaqMan probes (Thermo Fisher Scientific, Waltham, Mass.) were purchased for HD5 and GAPHD control, and all samples were run in triplicate using a CFX96 qPCR thermocycler (Bio-Rad). Data were analyzed per the ΔΔCt method of analysis.


Western Blot and Immunohistochemistry


Western blot was used to assess any differences in HD5 protein levels. Protein was extracted from a minimum of 10 colon biopsy samples each from mild, moderate, and severe UC; mild, moderate, and severe CC; and non-IBD DV control. Whole cell lysates were extracted from full-thickness colon samples using T-PER (Thermo Fisher Scientific) per manufacturer's protocol. Bradford Assays (Bio-Rad) were run to determine protein concentration, and protein was loaded onto a 4-20% SDS-PAGE tris/glycine gel (Bio-Rad). Proteins were transferred to PVDF (Bio-Rad), and Western blots for HD5 and β-actin loading control were performed with primary and secondary antibodies (Santa Cruz, Dallas, Tex.) per manufacturer's protocol. Blots were visualized with Opti-4CN colorimetric detection kit (Bio-Rad) and imaged with ChemiDoc XRS+imaging system (Bio-Rad). Band intensities were measured and data analysis performed with Image Lab Software (Bio-Rad).


Five colon tissue protein extracts and staining of HD5 per disease by immunohistochemistry (IHC) was done as previously described.24 Quantification of HD5 staining was analyzed manually by microscopy and automatically quantified using Nikon's Eclipse Ti microscope with built-in NEARAS [24,25].


NEARAS Technology for Quantification of Immunohistochemistry Staining


NEARAS (Melville, N.Y.) was used to calculate the number of cells with HD5 staining in IHC tissue. A mean intensity threshold of 20 to 255 intensity units was established to eliminate a false-positive signal from background staining. A circularity parameter of 0.5 to 1 and equivalent diameter of 5-15 micrometer was used to select for cells. All threshold parameters were used in each image to count the number of HD5-positive cells in tissue samples.


Statistical Analysis


The Vanderbilt University Microarray Core Laboratory performed statistical analyses for the microarray. Transcriptome level fold changes and the significance of those changes were calculated using one way ANOVA with Bonferroni's correction for multiple comparisons. Significantly changed transcripts were defined as having >2.0 fold expression change from controls and a Benjamini-Hochberg (BH) false discovery rate corrected ANOVA p-value <0.05. All other statistical analyses were performed using GraphPad Prism v6 software [26]. qRT-PCR and IHC HD5 counts were examined by applying an unpaired two-tailed Student's t-test with the Welch correction, respectively. Western blots were analyzed by ANOVA followed by Fisher's test for multiple comparisons. Chi square tests were utilized for determining relatedness of HD5 levels to CC. For all statistical analyses, p<0.05 indicated a statistical significance.


Dual Staining of Human α-Defensin-5 and Lysozyme


DoubleStain IHC was performed on a Lab Vision autostainer 360 (Thermo fisher) using Abcam's M&R on human tissue (DAB & AP/Red) staining kit (ab210059, Abcam Biotechnology, Cambridge, UK). The manufacture's recommended conditions were used with the following modifications. The mouse anti-α-defensin 5 (sc-53997, Santa Cruz Biotechnology, Inc, Dallas, Tex.) and rabbit anti-lysozyme (ab-2408) were used at a 1:50 dilution in OP Quanto antibody Diluent (Thermo Fisher, Waltham, Wash.). Prior to addition of antibody for 45 minutes, tissues were incubated for 10 min with Utravision hydrogen peroxide block (Thermo Fisher) followed by a 5 min incubation with Ultravisoion Quanto protein block. A single incubation with Permanent Red was used for ileum tissue, whereas two consecutive 10 min permanent Red incubations were performed for colonic tissue. Following hematoxylin counter staining, tissue was exposed to Richard-Allen Scientific Blueing Reagent (Thermo Fisher).


Antigen retrieval was performed in 1 mM EDTA pH 8.4, 0.05% Tween 20 for 20 minutes at 98° C. (60° C. preheat/70° C. cool down) using the Labvision PT Module (Thermo Scioentific). Image color deconvolution was performed with Fiji ImageJ 1.51f (http://imagej.nih.gov/ij) using the Fast Red, Fast Blue and DAB built in stain vector plugin.


Results


Nearly 30% of Indeterminate Colitis Patients Cannot be Delineated into UC or CC


A retrospective investigation was conducted to identify a cohort of patients diagnosed with IC to determine if they could be properly delineated into UC or CC over time. We followed 21 patients who were diagnosed with IC between the years 2000-2007 and reevaluated in 2014. A mean surveillance follow-up period was 8.7±3.7 (range, 4-14) years. Fifteen of the 21 (71.4%) had their original diagnosis changed; 9 to UC (43%) and 6 to CC (28.5%). Six (28.5%) patients remained clinically inconclusive and retained their diagnosis of IC (FIG. 1A). These data were collected in the absence of any type of biomarker.


Thirty Percent of Restorative Proctocolectomy Operated Crohn's Colitis Patients were Misdiagnosed as Ulcerative Colitis


A retrospective investigation was conducted to identify a cohort of patients that underwent RPC and IPAA surgery for a definitive UC diagnosis to determine if they had been misdiagnosed. We identified 67 such patients. A mean surveillance follow-up period was 9.4 (range, 6-13) years. A change in diagnosis to de novo Crohn's disease of the ileal pouch was clinically observed in 20 (30%) patients (FIG. 1B). In the other 47 (70%) cases, the initial diagnosis of UC remained clinically unchanged. These data were collected in the absence of any type of biomarker. Because of these results, we sought to determine if there are potential genes that may be used to better differentiate between UC and CC at first clinical biopsy and prior to any surgical intervention.


There is Differential Expression of Human α-Defensin-5 in Inflammatory Bowel Disease


We initially performed whole-transcriptome microarray with RNA extracted and pooled from human full thickness colon samples from UC and CC patients (n=5) using the Affymetrix gene expression array according to the manufacturer's instructions (Affymetrix, Santa Clara, Calif.) Tissues from diverticulitis (DV) were used as control. This analysis showed a total of 484 genes that were upor down-regulated (˜2-fold) between the two diseases. Among the upregulated genes were α-defensin-5, other antimicrobial peptides, and mucins (Table 1). HD5 was increased the most: 31-fold in CC vs. UC (in a previous study HD5 increase by 118-fold in CC versus UC—data not shown). A full list of the microarray results can be found in Table 1.


To replicate these data in a different platform, an independent analysis by PCR array (NanoString Technologies Inc. Seattle, Wash.) was carried out on 5 different human full thickness colon samples from UC and CC patients. Although the NanoString array only specifically targeted inflammatory genes, the only gene to show up in both the microarray and the PCR array was HD5. The NanoString array determined that HD5 was increased 118-fold in CC vs. UC in these human samples, compared to 31-fold in the previous samples analyzed by microarray (Table 2).


To further validate these data, we assessed the expression of HD5 by semi-quantitative RT-PCR using RNA extracted from moderate CC and moderate UC tissues (n=3). This analysis also showed a significant increase in transcript levels of HD5 in CC compared to UC (FIG. 2A, SEM, p<0.05). Several commercially available HD5 antibodies have been developed. Due to the sequence homology of the alpha defensin class of proteins, we tested a set of antibodies to assess specificity to HD5. We performed dot blots using commercially available antibodies against recombinant HD1-6. We determined that the monoclonal antibody from Santa Cruz Biotechnology, Inc. (Santa Cruz, Calif.) showed the highest level of specificity for HD5, and was therefore used in subsequent assays (FIG. 13). Next, we assessed the expression of HD5 by Western blotting (n=10 for each disease state). Samples were run individually on western blots, with a combination of disease states on each blot, and we show an example of an individual sample per disease state in a representative blot (FIG. 2B). When we take each individual sample into consideration across all western blots, protein densitometry analysis also shows significantly higher levels of HD5 in moderate and severe CC compared to all other disease states (FIG. 2C, p<0.0001). Finally, we examined the expression of HD5 in moderate disease activity of IBD and control tissues by IHC using FFPE sections. This analysis revealed that HD5 levels are indeed increased in CC (FIG. 2G) when compared to DV, and UC and normal (NL) control tissue (FIGS. 2D, 2E and 2F). Quantification of the HD5 IHC staining spot counts by NEARAS revealed a 5.6-fold increase of HD5 in CC vs. UC (FIG. 2H, p<0.0001). We believe the IHC data explains the weak western blot banding patterns. Because the western blots were run with full-thickness samples, there is a low overall abundance of HD5 in the tissue; the IHC shows that it is much localized in the base of individual colonic crypts. Because of this, further analysis is done using IHC instead of western blots.


Table 1 shows a list of targets from an affymetrix cDNA microarray. A total of 484 genes were highlighted in the microarray as potential markers for distinguishing UC from CC. The gene showing the largest fold change between the two diseases was Human Defensin 5 (HD5).


Table 2 shows a full list of targets from NanoString Human Inflammation PCR array. 16 inflammatory genes were changed in this subset of samples. HD5 was the only gene to appear in both the microarray and the NanoString PCR array.













TABLE 2







NanoString
NanoString
Microarray



Gene
p-value
Fold Change
Fold Change



Symbol
(CC vs UC)
(CC vs UC)
(CC vs UC)




















DEFA5
0.00182525
118.145
31.0374



RBP2
0.282548
6.8909




CD53
0.417119
−1.32516




SAA2
0.575901
−1.36908




SNORD13P2
0.0839705
−1.42879




SMAD4
0.00233383
−1.49572




SNORD28
0.00995582
−1.58122




ALOX5AP
0.153036
−1.61452




SCARNA8
0.132287
−1.63997




SNORD13
0.00409278
−1.87394




UNQ2550
0.0386757
−1.97314




CLEC4D
0.168864
−2.03025




STAP1
0.211401
−2.03524




CYP4F3LP
0.0584598
−2.37697




SAA1
0.0988763
−2.42023




IL6
0.167391
−4.90534










Human α-Defensin-5 Levels are Aberrant in Indeterminate Colitis and Restorative Proctocolectomy Operated Patients


In order to determine if HD5 could be used to assess whether IC patients could be delineated into a diagnosis of either UC or CC, we assessed levels of HD5 in surgical pathology colectomy samples via IHC in patients described in FIG. 1A. In each instance of a final diagnosis of CC, HD5 high NEARAS counts were in agreement with that diagnosis. We also found that when the 6 patients with unchanged IC diagnoses were analyzed via HD5 IHC NEARAS profile tests, 3 showed high HD5 count and agreed with the final diagnosis of CC, and 3 showed low HD5 count and were in agreement with the final diagnosis of UC (Table 4, as shown in FIG. 13). Table 4 shows that IHC staining for HD5 agrees with final diagnostic outcome in a sample of IC patients even when there was no agreement with the attending physician.


Further, RPC and IPAA-operated patients described in FIG. 1B who had a clinical change in diagnosis to de novo Crohn's (n=20) and those whose diagnoses did not change (n=47) were also analyzed molecularly for HD5 levels via NEARAS IHC counts. The patients whose diagnosis remained unchanged showed only trace levels of HD5 (FIG. 8A). Patients whose diagnoses clinically changed from UC to de novo Crohn's showed prominent HD5 staining (FIG. 8B).


These images can be compared to normal ileum control (FIG. 8C). Differential quantification of HD5 levels by NEARAS counts for UC RPC and IPAA-operated patients who did not have their original diagnoses changed vs. those with de novo Crohn's (FIG. 8A vs. 3B) were statistically significant (p<0.0001) (FIG. 8D). In addition, statistical analysis to determine positive predictive values (PPVs) of HD5 in patient tissue are 95.8% for CC and only 76.9% for UC. Chi squared analysis shows significant relatedness between high levels of HD5 and a diagnosis of CC (p<0.0001). These data indicate that HD5 could be developed into a diagnostic tool to better distinguish CC from UC.


Aberrantly Regulated Human α-Defensin-5 in Crohn's Colitis Patients May be Caused by Ectopic Colonic Crypt Paneth Cells HD5 is a Paneth cell product; therefore, we wanted to determine if Paneth cells were present in the colon crypt of Crohn's colitis patients. All 20 UC RPC operated patients with de novo Crohn's showed pools of ectopic crypt PCs in the colectomy samples, as demonstrated by H&E representative photomicrography (FIG. 9E). This was validated by IHC labeling of PCs using lysozyme by microscopy, which confirmed the abundant presence of PCs in CC colonic crypts (FIG. 9H, arrow). To validate whether the pool of HD5 expressed in CC and in de novo Crohn's colectomy samples was indeed coming from colonic epithelial crypt PCs, we used immunohistochemically detection of PC markers α-Defensin 5 (DEFA5) and lysozyme (LYZ) and double staining IHC to colocalize PCs and HD5 on colectomy samples. Lysozyme alone detects PCs. We demonstrate the presence of abundant crypt PCs in CC colectomy patients (FIG. 9H) compared to all other colonic conditions analyzed (UC, DV, DVL and NL). Further, double staining analyses from de novo Crohn's (FIGS. 10A and 5D), normal colon (FIG. 10J) and normal-ileum/control (FIG. 10G) are presented. Image deconvolutions are displayed vertically to evaluate lysozyme-specific permanent red (FIGS. 10B, 5E and 5H) and HD5-specific DAB (FIGS. 10C, 5F and 5I). The normal colon image (FIG. 10J), which lacks PCs, was not further processed. The results reconcile and represent a consensus among treating physicians.


Human α-Defensin-5 (DEFA5) is a Better Candidate Biomarker than Paneth Cells for Crohn's Colitis.


Finally, we sought to determine if HD5 and Paneth cells were both upregulated in the normal, adjacent tissue of CC patients compared to UC patients (FIG. 11). Immunohistochemistry for HD5 shows positive staining in the base of the crypts in both inflamed and normal, adjacent tissue of CC patient samples (FIG. 11A). We were even able to see some positive HD5 staining when the crypt structure is abolished due to excessive inflammation and tissue damage (FIG. 11A, patient WD-12919, arrows). Unsurprisingly, in UC tissue, we saw either very low levels of HD5 or no expression at all (FIG. 11B). We were very surprised, however, to find that we could not see any Paneth cells in either the inflamed or normal adjacent tissues of any of the CC or UC patients surveyed (CC, n=3; UC, n=2) (FIGS. 11C and 6D). These results are surprising, considering our earlier experiments surveying Paneth cells in a larger number of patients (FIG. 9). Because we can detect HD5 in the normal, adjacent tissue more readily than visualize Paneth cells, we believe that HD5 will serve as a better candidate biomarker than Paneth cells for CC.


Studies of MMP-7


Semi-quantitative real-time PCR (qPCR) was used to measure transcript levels of MMP-7. To do this, RNA was extracted from three human colon biopsy samples per condition; three each from moderate UC and CC, and from DV biopsy samples as a non-IBD control using the Qiagen RNeasy Miniprep Kit, (Valencia, Calif.). cDNA was generated using the iScript cDNA synthesis kit (Bio-Rad, Hercules, Calif.), then used in the qPCR reactions using pre-designed TaqMan probes for MMP-7, and GAPHD control, and universal PCR master mix (Thermo Fisher Scientific, Waltham, Mass.). The reactions were run in triplicate using a CFX96 real-time PCR thermocycler (Bio-Rad). Data were analyzed according to the ΔΔCt method.


To assess any differences in the expression of HD5 and MMP-7 at the protein level, colon surgical resections (n=10) were used from mild, moderate, and severe UC; mild, moderate, and severe CC; and non-IBD DV control. Whole tissue protein extracts were prepared from full-thickness colon samples (n=10) using T-PER protein extraction kit according to manufacturer's protocol (Thermo Fisher Scientific). Bradford Assays (Bio-Rad) were used to determine protein concentration, and equal amounts of protein were separated in 4-20% SDS-PAGE tris/glycine gel (sodium dodecyl sulphate-polyacrylamide gel electrophoresis) (Bio-Rad), then transferred to PVDF (polyvinylidene difluoride) membranes (Bio-Rad). The membranes were probed with antibodies against HD5, MMP-7, and β-actin loading control according to manufacturer's protocol. Blots were visualized with Opti-4CN colorimetric detection kit (Bio-Rad) and imaged with ChemiDoc XRS+imaging system (Bio-Rad). Band intensities were measured and data analysis performed with Image Lab Software (Bio-Rad).



FIG. 3 is a bar graph of a qRT-PCR result showing a decrease in MMP-7 levels in moderate CC compared to moderate UC, the inverse of HD5 levels (p<0.001). Western blot data for all disease states (n 10) shows a decrease in MMP-7 levels in CC compared to UC, significant when moderate and severe CC is compared to mild UC. FIG. 4 is a representative western blot of MMP-7 in subjects having IBD. Expression of HD5 in IBD tissues was examined by IHC using FFPE thin sections. MMP-7 levels (top) appear to decrease progressing from left to right. β-actin loading control is shown on bottom. FIG. 5 is a graphical representation of MMP-7 levels shown in FIG. 3. Band intensities were measured and normalized to β-actin loading control. Moderate and severe CC are statistically significant when compared to mild UC (p<0.05 and p<0.005, respectively). FIGS. 8A-G are graphical representations showing MMP-7 and HD5 levels are inversely expressed in IBD. Patient samples were matched and levels of HD5 and MMP-7 were compared. In all disease states, MMP-7 and HD5 levels are inversely expressed, and all differences are statistically significant. As MMP-7 levels decrease, HD5 levels increase.


Discussion


Colectomy surgical pathology samples of patients with unambiguous CC and UC undergoing colectomy in connection with RPC and IPAA were analyzed [6,7]. We those protein profiles which had the necessary (i) specificity; (ii) sensitivity; (iii) discriminatory; and (iv) predictive capacity to determine the heterogeneity of IBD [6,7] were identified and compared. It was possible to molecularly delineate UC and CC with molecular signatures of HD5 using IHC and quantified by NEARAS. Alpha-Defensins HD5 and HD6 are PC products and their altered expression has been linked to IBD pathogenesis.


It was not expected that one could not visualize PCs in these tissues even though one could detect HD5 in the same tissue (FIG. 2H), especially compared to earlier experiments showing high levels of PCs in all CC patients surveyed (FIG. 2C). Whether the PCs are essential for stem cell maintenance in vivo remains debatable [27].


To date, there is no diagnostic gold standard tool for IBD. Differentiating UC and CC among patients with IC has remained painstaking and is a major challenge in endoscopic medicine and colorectal surgery [1,12,28,29]. Clinicians use an inexact classification system of clinical, endoscopy, radiologic, and histopathology findings in order to diagnose CC and UC [21,30,31]. Even with a combination of these diagnostic modalities, up to 15% of IBD patients are labeled as IC when no definitive evaluations can be made [13,30,32]. In addition, CC is mistakenly diagnosed and RPC and IPAA-operated as definitive UC in another 15% of IBD patients because of overlap in the clinical, endoscopic, radiological and histologic findings [12,33-36]. Further, most IC patients who undergo RPC and IPAA surgery for presumed UC are subsequently found to develop a recurrent de novo Crohn's disease in the ileal pouch [1,12,33]. This is a serious consequence that may hinder the restoration of intestinal continuity and its intractable nature leads to pouch failure, often requiring pouch diversion or excision with a permanent terminal-ileostomy, resulting in negative psycho-sociological implications and poorer quality of life [1,29,31,36-41]. Curative treatment for UC is often surgical [42].


Success of RPC and IPAA surgery is largely dependent on careful patient selection combined with meticulous surgical technique and diagnostic accuracy [9-11,13]. Available clinical presentations and experience suggest that it is difficult to identify patients with CC who are likely to have a successful outcome after RPC and IPAA surgery [10,13,34,43]. However, in highly selected patients with CC, RPC and IPAA has been indicated [44-47]. Thus, RPC operation may be considered and should remain a careful option for certain subgroup of patients with CC, but an acceptable care option for patients with UC and for those IC patients predicted to develop UC [9,42].


These studies of HD5 as a candidate biomarker for CC suggest it could be a diagnostic signature to efficiently distinguish CC from UC. Newly published data shows that patients with small bowel Crohn's disease (Crohn's ileitis) are characterized with a deficiency of HD5, as shown by a reduced expression and secretion of the Paneth cell HD5, a fundamental feature of Crohn's ileitis [48-51]. Based on this study, in CC, the reverse is true. It was found that Paneth cell HD5 is a predominantly expressed antimicrobial peptide. This indicates that definitive CC and Crohn's ileitis may have distinct etiologies and mechanisms. In these studies, all IC patient samples have been reconciled into UC and CC using molecular biomarker, HD5, and verified the reconciliation by patient outcomes (FIGS. 1 and 13 (Table 4)).


Accurately distinguishing CC from UC is of utmost importance when determining the candidacy of a patient for RPC [1,42]. Early diagnostic accuracy of IBD will lead to timely appropriate medical options. This study confirms that HD5 can differentiate CC and UC and reclassify IC into CC. In addition to distinguishing the colitides, HD5 could objectively be used to evaluate biophysiological processes and therapeutic outcomes and potentially play a pivotal role in IBD clinics as an attractive, non-invasive avenue [52,53].


Thus, this working example shows that tissue samples taken from patients suffering from CD have levels of HD5 significantly higher than the HD5 levels in samples from patients suffering from UC. Additionally, this working example shows that samples taken from patients suffering from CD have levels of MMP-7 significantly lower than the MMP-7 levels in samples of patients suffering from UC.


REFERENCES

The following references were cited in the above working example. Such citation is not to be construed as an admission that any reference meets the legal definition of “prior art” in any country, nor as an admission that any reference is relevant to the patentability of anything claimed. Any such reference shall be incorporated herein by reference only to the extent it is necessary for a person of ordinary skill in the art to make and use anything claimed.

  • 1. Jackson K L, Stocchi L, Duraes L, Rencuzogullari A, Bennett A E, Remzi F H. Long-Term Outcomes in Indeterminate Colitis Patients Undergoing Ileal Pouch-Anal Anastomosis: Function, Quality of Life, and Complications. J Gastrointest Surg 2017; 21:56-61. https://doi.org/10.1007/s11605-016-3306-9 PMID: 27832426
  • 2. Carvalho R S, Abadom V, Dilworth H P, Thompson R, Oliva-Hemker M, Cuffari C. Indeterminate colitis: a significant subgroup of pediatric IBD. Inflamm Bowel Dis 2006; 12:258-62. https://doi.org/10.1097/01. MIB.0000215093.62245.b9 PMID: 16633047
  • 3. Cantoro L, Di Sabatino A, Papi C, Margagnoni G, Ardizzone S, Giuffrida P, et al. The time course of diagnostic delay in inflammatory bowel disease over the last sixty years: an Italian multicentre study. J Crohn's Colitis 2017 March 18 [Epub ahead of print].
  • 4. Clark C, Turner J. Diagnostic Modalities for Inflammatory Bowel Disease: Serologic Markers and Endoscopy. Surg Clin North Am 2015; 95:1123-41. https://doi.org/10.1016/j.suc.2015.07.008 PMID: 26596918
  • 5. Odze R D. A contemporary and critical appraisal of ‘indeterminate colitis’. Mod Pathol 2015; 28 Suppl 1: S30-46.
  • 6. M'Koma A E, Seeley E H, Washington M K, Schwartz D A, Muldoon R L, Herline A J, et al. Proteomic profil-ing of mucosal and submucosal colonic tissues yields protein signatures that differentiate the inflamma-tory colitides. Inflamm Bowel Dis 2011; 17:875-83. https://doi.org/10.1002/ibd.21442 PMID: 20806340
  • 7. Seeley E H, Washington M K, Caprioli R M, M'Koma A E. Proteomic patterns of colonic mucosal tissues delineate Crohn's colitis and ulcerative colitis. Proteomics Clin Appl 2013; 7:541-9. https://doi.org/10.1002/prca.201200107 PMID: 23382084
  • 8. M'Koma A E, Blum D L, Norris J L, Koyama T, Billheimer D, Motley S, et al. Detection of pre-neoplastic and neoplastic prostate disease by MALDI profiling of urine. Biochem Biophys Res Commun 2007; 353:829-34. https://doi.org/10.1016/j.bbrc.2006.12.111 PMID: 17194448
  • 9. M'Koma A E, Wise P E, Muldoon R L, Schwartz D A, Washington M K, Herline A J. Evolution of the restor-ative proctocolectomy and its effects on gastrointestinal hormones. Int J Colorectal Dis 2007; 22:1143-63. https://doi.org/10.1007/s00384-007-0331-x PMID: 17576578
  • 10. Shen B, Remzi F H, Brzezinski A, Lopez R, Bennett A E, Lavery I C, et al. Risk factors for pouch failure in patients with different phenotypes of Crohn's disease of the pouch. Inflamm Bowel Dis 2008; 14:942-8. https://doi.org/10.1002/ibd.20409 PMID: 18300279
  • 11. Shen B, Remzi F H, Lavery I C, Lashner B A, Fazio V W. A proposed classification of ileal pouch disorders and associated complications after restorative proctocolectomy. Clin Gastroenterol Hepatol 2008; 6:145-58. https://doi.org/10.1016/j.cgh.2007.11.006 PMID: 18237865
  • 12. Zaghiyan K, Kaminski J P, Barmparas G, Fleshner P. De novo Crohn's Disease after Ileal Pouch-Anal Anastomosis for Ulcerative Colitis and Inflammatory Bowel Disease Unclassified: Long-Term Follow-Up of a Prospective Inflammatory Bowel Disease Registry. Am Surg 2016; 82:977-81. PMID: 27779987
  • 13. Shen B. Crohn's disease of the ileal pouch: reality, diagnosis, and management. Inflamm Bowel Dis 2009; 15:284-94. https://doi.org/10.1002/ibd.20661 PMID: 18816633
  • 14. Lee H S, Choe J, Lee H J, Hwang S W, Park S H, Yang D H, et al. Change in the diagnosis of inflammatory bowel disease: a hospital-based cohort study from Korea. Intest Res 2016; 14:258-63. https://doi.org/10.5217/ir.2016.14.3.258 PMID: 27433148
  • 15. Nakamura K, Sakuragi N, Takakuwa A, Ayabe T. Paneth cell alpha-defensins and enteric microbiota in health and disease. Biosci Microbiota Food Health 2016; 35:57-67. https://doi.org/10.12938/bmfh. 2015-019 PMID: 27200259
  • 16. Durand A, Donahue B, Peignon G, Letourneur F, Cagnard N, Slomianny C, et al. Functional intestinal stem cells after Paneth cell ablation induced by the loss of transcription factor Math1 (Atoh1). Proc Natl Acad Sci USA 2012; 109:8965-70. https://doi.org/10.1073/pnas.1201652109 PMID: 22586121
  • 17. Li X, LeBlanc J, Elashoff D, McHardy I, Tong M, Roth B, et al. Microgeographic Proteomic Networks of the Human Colonic Mucosa and Their Association With Inflammatory Bowel Disease. Cell Mol Gastro-enterol Hepatol 2016; 2:567-83. https://doi.org/10.1016/j.jcmgh.2016.05.003 PMID: 28174738
  • 18. VanDussen K L, Liu T C, Li D, Towfic F, Modiano N, Winter R, et al. Genetic variants synthesize to pro-duce paneth cell phenotypes that define subtypes of Crohn's disease. Gastroenterology 2014; 146:200-9. https://doi.org/10.1053/j.gastro.2013.09.048 PMID: 24076061
  • 19. Lawrance I C, Fiocchi C, Chakravarti S. Ulcerative colitis and Crohn's disease: distinctive gene expres-sion profiles and novel susceptibility candidate genes. Hum Mol Genet 2001; 10:445-56. PMID: 11181568
  • 20. Puri K S, Suresh K R, Gogtay N J, Thatte U M. Declaration of Helsinki, 2008: implications for stakeholders in research. J Postgrad Med 2009; 55:131-4. https://doi.org/10.4103/0022-3859.52846 PMID: 19550060
  • 21. Targan S R, Karp L C. Inflammatory bowel disease diagnosis, evaluation and classification: state-of-the art approach. Curr Opin Gastroenterol 2007; 23:390-4. https://doi.org/10.1097/MOG. 0b013e3281722271 PMID: 17545774
  • 22. James S D, Wise P E, Zuluaga-Toro T, Schwartz D A, Washington M K, Shi C. Identification of pathologic features associated with “ulcerative colitis-like” Crohn's disease. World J Gastroenterol 2014; 20:13139-45. https://doi.org/10.3748/wjg.v20.i36.13139 PMID: 25278708
  • 23. Jong V L, Novianti P W, Roes K C, Eijkemans M J. Selecting a classification function for class prediction with gene expression data. Bioinformatics 2016; 32:1814-22. https://doi.org/10.1093/bioinformatics/btw034 PMID: 26873933
  • 24. Myers J N, Schaffer M W, Korolkova O Y, Williams A D, Gangula P R, M'Koma A E. Implications of the Colonic Deposition of Free Hemoglobin-alpha Chain: A Previously Unknown Tissue By-product in Inflammatory Bowel Disease. Inflamm Bowel Dis 2014; 20:1530-47. https://doi.org/10.1097/MIB. 0000000000000144 PMID: 25078150
  • 25. Gmyr V, Bonner C, Lukowiak B, Pawlowski V, Dellaleau N, Belaich S, et al. Automated digital image analysis of islet cell mass using Nikon's inverted eclipse Ti microscope and software to improve engraftment may help to advance the therapeutic efficacy and accessibility of islet transplantation across centers. Cell Transplant 2015; 24:1-9. https://doi.org/10.3727/096368913X667493 PMID: 23683575
  • 26. Tulay P, Naja R P, Cascales-Roman O, Doshi A, Serhal P, SenGupta S B. Investigation of microRNA expression and DNA repair gene transcripts in human oocytes and blastocysts. J Assist Reprod Genet 2015; 32:1757-64. https://doi.org/10.1007/s10815-015-0585-0 PMID: 26438643
  • 27. Farin H F, Karthaus W R, Kujala P, Rakhshandehroo M, Schwank G, Vries R G, et al. Paneth cell extru-sion and release of antimicrobial products is directly controlled by immune cell-derived IFN-gamma. J Exp Med 2014; 211:1393-405. https://doi.org/10.1084/jem.20130753 PMID: 24980747
  • 28. Ballard B R, M'Koma A E. Gastrointestinal endoscopy biopsy derived proteomic patterns predict indeterminate colitis into ulcerative colitis and Crohn's colitis. World J Gastrointest Endos 2015; 7:670-4.
  • 29. Zaharie R, Tantau A, Zaharie F, Tantau M, Gheorghe L, Gheorghe C, et al. Diagnostic Delay in Roma-nian Patients with Inflammatory Bowel Disease: Risk Factors and Impact on the Disease Course and Need for Surgery. J Crohn's Colitis 2016; 10:306-14.
  • 30. M'Koma A E. Diagnosis of inflammatory bowel disease: Potential role of molecular biometrics. World J Gastrointest Surg 2014; 6:208-19. https://doi.org/10.4240/wjgs.v6.i11.208 PMID: 25429322
  • 31. Matsui T. [Diagnosis of inflammatory bowel diseases. 1. Diagnosis, disease type and severity classifica-tion]. Nihon Naika Gakkai Zasshi 2009; 98:31-6. PMID: 19334353
  • 32. Feakins R M. Ulcerative colitis or Crohn's disease? Pitfalls and problems. Histopathology 2014; 64:317-35. https://doi.org/10.1111/his.12263 PMID: 24266813
  • 33. Gu J, Stocchi L, Kiran R P, Shen B, Remzi F H. Do clinical characteristics of de novo pouch Crohn's dis-ease after restorative proctocolectomy affect ileal pouch retention? Dis Colon Rectum 2014; 57:76-82. https://doi.org/10.1097/01.dcr.0000437691.52109.f2 PMID: 24316949
  • 34. Wagner-Bartak N A, Levine M S, Rubesin S E, Laufer I, Rombeau J L, Lichtenstein G R. Crohn's disease in the ileal pouch after total colectomy for ulcerative colitis: findings on pouch enemas in six patients. AJR Am J Roentgenol 2005; 184:1843-7. https://doi.org/10.2214/ajr.184.6.01841843 PMID: 15908540
  • 35. Price A B. Overlap in the spectrum of non-specific inflammatory bowel disease—‘colitis indeterminate’. J Clin Pathol 1978; 31:567-77. PMID: 670413
  • 36. Langner C, Aust D, Ensari A, Villanacci V, Becheanu G, Miehlke S, et al. Histology of microscopic coli-tis-review with a practical approach for pathologists. Histopathology 2015; 66:613-26. https://doi.org/10.1111/his.12592 PMID: 25381724
  • 37. Langner C, Magro F, Driessen A, Ensari A, Mantzaris G J, Villanacci V, et al. The histopathological approach to inflammatory bowel disease: a practice guide. Virchows Arch 2014; 464:511-27. https://doi.org/10.1007/s00428-014-1543-4 PMID: 24487791
  • 38. Abdelaal K, Jaffray B. Variables associated with loss of ileoanal pouches constructed in childhood. J Pediatr Surg 2017; 52:281-5. https://doi.org/10.1016/j.jpedsurg.2016.11.017 PMID: 27890312
  • 39. Robbins L, Zaghiyan K, Melmed G, et al. Outcomes with Anti-Tumour Necrosis Factor-Alpha Therapy and Serology in Patients with Denovo Crohn's Disease After Ileal Pouch Anal Anastomosis. J Crohn's Colitis 2017; 11:77-83.
  • 40. Das P, Smith J J, Tekkis P P, Heriot A G, Antropoli M, John Nicholls R. Quality of life after indefinite diver-sion/pouch excision in ileal pouch failure patients. Colorectal Dis 2007; 9:718-24. https://doi.org/10.1111/j.1463-1318.2007.01216.x PMID: 17764535
  • 41. Lepisto A, Luukkonen P, Jarvinen H J. Cumulative failure rate of ileal pouch-anal anastomosis and qual-ity of life after failure. Dis Colon Rectum 2002; 45:1289-94. https://doi.org/10.1097/01.DCR. 0000027032.95753.11 PMID: 12394424
  • 42. Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A, Barreiro-de Acosta M, et al. Third European Evidence-Based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions, diagnosis, extra-intestinal manifestations, pregnancy, cancer surveillance, surgery, and ileo-anal pouch disorders. J Crohn's Colitis 2017; 11:649-670
  • 43. Keighley M R. The final diagnosis in pouch patients for presumed ulcerative colitis may change to Crohn's disease: patients should be warned of the consequences. Acta Chir Iugosl 2000; 47:27-31. PMID: 11432239
  • 44. Turina M, Remzi F H. The J-pouch for patients with Crohn's disease and indeterminate colitis: (when) is it an option? J Gastrointest Surg 2014; 18:1343-4. https://doi.org/10.1007/s11605-014-2498-0 PMID: 24777433
  • 45. Le Q, Melmed G, Dubinsky M, McGovern D, Vasiliauskas E A, Murrell Z, et al. Surgical outcome of ileal pouch-anal anastomosis when used intentionally for well-defined Crohn's disease. Inflamm Bowel Dis 2013; 19:30-6. https://doi.org/10.1002/ibd.22955 PMID: 22467562
  • 46. Li Y, Wu B, Shen B. Diagnosis and differential diagnosis of Crohn's disease of the ileal pouch. Curr Gastroenterol Rep 2012; 14:406-13. https://doi.org/10.1007/s11894-012-0282-4 PMID: 22855236
  • 47. Shen B, Patel S, Lian L. Natural history of Crohn's disease in patients who underwent intentional restor-ative proctocolectomy with ileal pouch-anal anastomosis. Aliment Pharmacol Ther 2010; 31:745-53. https://doi.org/10.1111/j.1365-2036.2009.04227.x PMID: 20047579
  • 48. Courth L F, Ostaff M J, Mailander-Sanchez D, Malek N P, Stange E F, Wehkamp J. Crohn's disease-derived monocytes fail to induce Paneth cell defensins. Proc Natl Acad Sci USA 2015; 112:14000-5. https://doi.org/10.1073/pnas.1510084112 PMID: 26512113
  • 49. Tan G, Zeng B, Zhi F C. Regulation of human enteric alpha-defensins by NOD2 in the Paneth cell line-age. Eur J Cell Biol 2015; 94:60-6. https://doi.org/10.1016/j.ejcb.2014.10.007 PMID: 25433720
  • 50. Wang C, Shen M, Gohain N, Tolbert W D, Chen F, Zhang N, et al. Design of a potent antibiotic peptide based on the active region of human defensin 5. J Med Chem 2015; 58:3083-93. https://doi.org/10.1021/jm501824a PMID: 25782105
  • 51. Hayashi R, Tsuchiya K, Fukushima K, Horita N, Hibiya S, Kitagaki K, et al. Reduced Human alpha-defensin 6 in Noninflamed Jejunal Tissue of Patients with Crohn's Disease. Inflamm Bowel Dis 2016; 22:1119-28. https://doi.org/10.1097/MIB.0000000000000707 PMID: 26891258
  • 52. Wagner J A, Williams S A, Webster C J. Biomarkers and surrogate end points for fit-for-purpose develop-ment and regulatory evaluation of new drugs. Clin Pharmacol Ther 2007; 81:104-7. https://doi.org/10.1038/sj.clpt.6100017 PMID: 17186007
  • 53. Vermeire S, Van Assche G, Rutgeerts P. Laboratory markers in IBD: useful, magic, or unnecessary toys? Gut 2006; 55:426-31. https://doi.org/10.1136/gut.2005.069476 PMID: 16474109.


EXEMPLARY EMBODIMENTS

In addition to anything described above or currently claimed, it is specifically contemplated that any of the following embodiments may be claimed:


Emb. 1: A method of measuring HD5 in a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: obtaining a sample from the patient; and measuring at least one of the expression of HD5 and the concentration of HD5 in the sample.


Emb. 2: A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: performing the method of measuring HD5 in the patient according to embodiment 1; and performing an intervention on the patient to treat Crohn's disease.


Emb. 3: Any one of the methods of embodiments 1-2, comprising: comparing the expression of HD5 or the concentration of HD5 in the sample to a benchmark value that is typical of a subject not suffering from Crohn's disease;


and diagnosing Crohn's disease if the expression of HD5 or the concentration of HD5 in the sample significantly exceeds the benchmark value.


Emb. 4: Any one of the methods of embodiments 1-3, wherein the expression of HD5 or the concentration of HD5 in the sample exceeds a benchmark value that is typical of a subject not suffering from Crohn's disease.


Emb. 5: Any one of the methods of embodiments 1-4, wherein the expression of HD5 is measured to be significantly greater in the sample than in a control sample from a subject not suffering from Crohn's disease.


Emb. 6: Any one of the methods of embodiments 1-5, wherein the expression of HD5 is measured to be at least about 31 times greater in the sample than in a control sample from a subject not suffering from Crohn's disease.


Emb. 7: Any one of the methods of embodiments 1-6, wherein the expression of HD5 is measured to be at least about 118 times greater in the sample than in a control sample from a subject not suffering from Crohn's disease.


Emb. 8: Any one of the methods of embodiments 1-7, wherein the expression of HD5 is measured to be greater than about 106 HD5 mRNA transcripts per 10 ng RNA.


Emb. 9: Any one of the methods of embodiments 1-8, wherein the expression of HD5 is measured to be greater than 107 HD5 mRNA transcripts per 10 ng RNA.


Emb. 10: Any one of the methods of embodiments 1-9, wherein the expression of HD5 is measured to be greater than 1.9×107 HD5 mRNA transcripts per 10 ng RNA.


Emb. 11: Any one of the methods of embodiments 1-10, wherein the expression of HD5 is measured to be greater than 7×107 HD5 mRNA transcripts per 10 ng RNA.


Emb. 12: Any one of the methods of embodiments 1-11, wherein the expression of HD5 is measured by qRT-PCR, wherein the method comprises measuring the expression of HD5 mRNA in a control sample from a subject not suffering from Crohn's disease, and wherein the expression of HD5 mRNA in the sample is significantly greater than the expression of HD5 mRNA in the control sample.


Emb. 13: Any one of the methods of embodiments 1-12, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is at least 10%.


Emb. 14: Any one of the methods of embodiments 1-13, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is at least 20%.


Emb. 15: Any one of the methods of embodiments 1-14, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is at least about 30%.


Emb. 16: Any one of the methods of embodiments 1-15, wherein the intervention is not effective to treat ulcerative colitis.


Emb. 17: Any one of the methods of embodiments 1-16, wherein the intervention is a surgery.


Emb. 18: Any one of the methods of embodiments 1-17, wherein the intervention is a surgery selected from the group consisting of: ostomy, colostomy, ileostomy, bowel resection, colectomy, proctocolectomy, and strictureplasty.


Emb. 19: Any one of the methods of embodiments 1-18, wherein the intervention is administration of a drug.


Emb. 20: Any one of the methods of embodiments 1-19, wherein the intervention is administration of a drug, to the exclusion of a surgery.


Emb. 21: Any one of the methods of embodiments 1-20, wherein the intervention is administration of a drug selected from the group consisting of: a vitamin supplement, vitamin B12, vitamin D, a mineral supplement, calcium, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, an anti-TNF-alpha antibody, infliximab, adalimumab, certolizumab pegol, methotrexate, an anti-α4-integrin antibody, natalizumab, vedolizumab, an anti-interleukin antibody, ustekinumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.


Emb. 22: Any one of the methods of embodiments 1-21, wherein the intervention is administration of a drug selected from the group consisting of: certolizumab pegol, methotrexate, and natalizumab.


Emb. 23: Any one of the methods of embodiments 1-22, wherein the intervention is placement of the subject on a low fat diet.


Emb. 24: A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising:


performing the method of measuring HD5 in the patient according to embodiment 1; and; and


performing an intervention on the patient to treat ulcerative colitis.


Emb. 25: The method of embodiment 24, comprising: comparing the expression of HD5 or the concentration of HD5 in the sample to a benchmark value that is typical of a subject not suffering from Crohn's disease; and diagnosing ulcerative colitis if the expression of HD5 or the concentration of HD5 in the sample does not significantly exceed the benchmark value.


Emb. 26: The method of any one of embodiments 24-25, wherein the expression of HD5 or the concentration of HD5 in the sample is below a benchmark value that is typical of a subject suffering from Crohn's disease.


Emb. 27: The method of any one of embodiments 24-25, wherein the expression or concentration of HD5 in the sample is measured to be significantly less than in a control sample from a subject suffering from Crohn's disease.


Emb. 28: The method of any one of embodiments 24-27, wherein the expression of HD5 is measured in the sample is no more than about 1/31 of expression of HD5 measured in a control sample from a subject not suffering from Crohn's disease.


Emb. 29: The method of any one of embodiments 24-28, wherein the expression of HD5 is measured in the sample is no more than about 1/118 of expression of HD5 measured in a control sample from a subject not suffering from Crohn's disease.


Emb. 30: The method of any one of embodiments 24-29, wherein the expression of HD5 is measured to be less than 106 HD5 mRNA transcripts per 10 ng RNA.


Emb. 31: The method of any one of embodiments 24-30, wherein the expression of HD5 is measured to be less than 107 HD5 mRNA transcripts per 10 ng RNA.


Emb. 32: The method of any one of embodiments 24-31, wherein the expression of HD5 is measured to be less than 1.9×107 HD5 mRNA transcripts per 10 ng RNA.


Emb. 33: The method of any one of embodiments 24-32, wherein the expression of HD5 is measured to be less than 6×105 HD5 mRNA transcripts per 10 ng RNA.


Emb. 34: The method of any one of embodiments 24-33, wherein the expression of HD5 is measured by qRT-PCR, wherein the method comprises measuring the expression of HD5 in a control sample from a subject suffering from Crohn's disease, and wherein the expression of HD5 mRNA in the sample is significantly less than the expression of HD5 in the control sample.


Emb. 35: The method of any one of embodiments 24-34, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is less than 10%.


Emb. 36: The method of any one of embodiments 24-35, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is less than 20%.


Emb. 37: The method of any one of embodiments 24-36, wherein the sample is intestinal tissue, and comprising measuring the concentration of HD5 by: immunostaining the sample with an anti-HD5 immunostaining agent; and measuring the percentage of cells in the sample that stain positive; wherein the percentage of cells in the sample that stain positive is less than about 30%.


Emb. 38: The method of any one of embodiments 24-37, wherein the intervention is not effective to treat Crohn's disease.


Emb. 39: The method of any one of embodiments 24-38, wherein the intervention is a surgery.


Emb. 40: The method of any one of embodiments 24-39, wherein the intervention is a surgery combined with the administration of a drug.


Emb. 41: The method of any one of embodiments 24-40, wherein the intervention is a surgery selected from the group consisting of: a proctocolectomy, and an ileal pouch anal anastomosis.


Emb. 42: The method of any one of embodiments 24-41, wherein the intervention is administration of a drug selected from the group consisting of: an iron supplement, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, cyclosporine, an anti-TNF-alpha antibody, infliximab, adalimumab, golimumab, methotrexate, an anti-α4-integrin antibody, vedolizumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.


Emb. 43: The method of any one of embodiments 24-42, wherein the intervention is administration of a drug selected from the group consisting of: cyclosporine, and golimumab.


Emb. 44: The method of any one of embodiments 1-43, wherein the expression of HD5 is measured.


Emb. 45: The method of any one of embodiments 1-44, wherein the concentration of HD5 is measured.


Emb. 46: The method of any one of embodiments 1-45, wherein the sample is intestinal tissue.


Emb. 47: The method of any one of embodiments 1-46, wherein the sample is from the subject's large intestine.


Emb. 48: The method of any one of embodiments 1-47, wherein the sample is colonic tissue.


Emb. 49: The method of any one of embodiments 1-48, wherein the sample is ileal tissue.


Emb. 50: The method of any one of embodiments 1-49, wherein the expression of HD5 or the concentration of HD5 is measured in the sample ex vivo.


Emb. 51: The method of any one of embodiments 1-50, wherein HD5 expression is measured by a technique selected from the group consisting of: whole transcriptome analysis, whole-transcriptome microarray, Northern blot, DNA microarray, PCR, sequencing PCR, RT-PCR, quantitative PCR, restriction fragment length polymorphism, in situ hybridization assay, and a competitive-binding assay.


Emb. 52: The method of any one of embodiments 1-51, wherein HD5 concentration is measured by a technique selected from the group consisting of: Western blot, ELISA, two-dimensional gel electrophoresis, mass spectrometry, protein interaction profiling, a competitive binding assay, a non-competitive binding assay, a radioimmunoassay, an enzyme immunoassays, an enzyme linked immunosorbent assay (ELISA), a sandwich immunoassay, a precipitation reaction, a gel diffusion reaction, an immunodiffusion assay, an agglutination assay, a complement-fixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, NMR analysis, and an immunoelectrophoresis assay.


Emb. 53: The method of any one of embodiments 1-52, wherein the patient is suffering from IBD.


Emb. 54: The method of any one of embodiments 1-53, wherein the patient displays a symptom selected from the group consisting of: severe diarrhea, abdominal pain, fatigue, and weight loss.


Emb. 55: The method of any one of embodiments 1-54, wherein the patient displays severe diarrhea, abdominal pain, fatigue, and weight loss.


Emb. 56: The method of any one of embodiments 1-55, wherein the expression of HD5 is measured.


Emb. 57: The method of any one of embodiments 1-56, wherein the concentration of HD5 is measured.


Emb. 58: The method of any one of embodiments 1-57, further comprising measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample.


Emb. 59: A method of measuring MMP-7 in a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: obtaining a sample from the patient; and measuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample.


Emb. 60: A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising:


performing the method of measuring MMP-7 in the patient according to any one of embodiments 58-59; and


performing an intervention on the patient to treat Crohn's disease.


Emb. 61: The method of any one of embodiments 58-60, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis; and diagnosing Crohn's disease if the expression of MMP-7 or the concentration of MMP-7 in the sample does not significantly exceed the benchmark value.


Emb. 62: The method of any one of embodiments 58-61, wherein the expression of MMP-7 or the concentration of MMP-7 in the sample is below a benchmark value that is typical of a subject suffering from ulcerative colitis.


Emb. 63: The method of any one of embodiments 58-61, wherein the expression of MMP-7 is measured to be significantly less in the sample than in a control sample from a subject suffering from ulcerative colitis.


Emb. 64: The method of any one of embodiments 58-63, wherein the expression of MMP-7 is measured in the sample to be at most about 1/10 of the expression of MMP-7 measured in a control sample from a subject not suffering from ulcerative colitis.


Emb. 65: The method of any one of embodiments 58-64, wherein the expression of MMP-7 is measured by qRT-PCR, wherein the method comprises measuring the expression of MMP-7 in a control sample from a subject suffering from ulcerative colitis, and wherein the expression of MMP-7 in the sample is significantly less than the expression of MMP-7 in the control sample.


Emb. 66: The method of any one of embodiments 58-65, wherein the sample is intestinal tissue, and comprising measuring the concentration of MMP-7 by: immunostaining the sample with an antiMMP-7 immunostaining agent; and measuring the percentage of cells in the sample that stain positive.


Emb. 67: The method of any one of embodiments 58-66, wherein the intervention is not effective to treat ulcerative colitis.


Emb. 68: The method of any one of embodiments 58-67, wherein the intervention is a surgery.


Emb. 69: The method of any one of embodiments 58-68, wherein the intervention is a surgery selected from the group consisting of: ostomy, colostomy, ileostomy, bowel resection, colectomy, proctocolectomy, and strictureplasty.


Emb. 70: The method of any one of embodiments 58-69, wherein the intervention is administration of a drug.


Emb. 71: The method of any one of embodiments 58-70, wherein the intervention is administration of a drug, to the exclusion of a surgery.


Emb. 72: The method of any one of embodiments 58-71, wherein the intervention is administration of a drug selected from the group consisting of: a vitamin supplement, vitamin B12, vitamin D, a mineral supplement, calcium, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, an anti-TNF-alpha antibody, infliximab, adalimumab, certolizumab pegol, methotrexate, an anti-α4-integrin antibody, natalizumab, vedolizumab, an anti-interleukin antibody, ustekinumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.


Emb. 73: The method of any one of embodiments 58-72, wherein the intervention is administration of a drug selected from the group consisting of: certolizumab pegol, methotrexate, and natalizumab.


Emb. 74: The method of any one of embodiments 58-73, wherein the intervention is placement of the subject on a low fat diet.


Emb. 75: A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising:


performing the method of measuring MMP-7 in the patient according to embodiment 58; and


performing an intervention on the patient to treat ulcerative colitis.


Emb. 76: The method of any one of embodiments 58, 59 and 75, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis; and diagnosing ulcerative colitis if the expression of MMP-7 or the concentration of MMP-7 in the sample significantly exceeds the benchmark value.


Emb. 77: The method of any one of embodiments 58, 59 and 75-76, wherein the expression of MMP-7 or the concentration of MMP-7 in the sample is above a benchmark value that is typical of a subject not suffering from ulcerative colitis.


Emb. 78: The method of any one of embodiments 58, 59 and 75-76, wherein the expression or concentration of MMP-7 in the sample is measured to be significantly greater than in a control sample from a subject not suffering from ulcerative colitis.


Emb. 79: The method of any one of embodiments 58, 59 and 75-78, wherein the expression of MMP-7 measured in the sample is at least about 5 times the expression of MMP-7 measured in a control sample from a subject not suffering from ulcerative colitis.


Emb. 80: The method of any one of embodiments 58, 59 and 75-79, wherein the expression of MMP-7 measured in the sample is at least about 10 times the expression of MMP-7 measured in a control sample from a subject not suffering from ulcerative colitis.


Emb. 81: The method of any one of embodiments 58, 59 and 75-80, wherein the expression of MMP-7 is measured by qRT-PCR, wherein the method comprises measuring the expression of MMP-7 in a control sample from a subject not suffering from ulcerative colitis, and wherein the expression of MMP-7 in the sample is significantly greater than the expression of MMP-7 in the control sample.


Emb. 82: The method of any one of embodiments 58, 59 and 75-81, wherein the sample is intestinal tissue, and comprising measuring the concentration of MMP-7 by: immunostaining the sample with an antiMMP-7 immunostaining agent; and measuring the percentage of cells in the sample that stain positive.


Emb. 83: The method of any one of embodiments 58, 59 and 75-82, wherein the intervention is not effective to treat Crohn's disease.


Emb. 84: The method of any one of embodiments 58, 59 and 75-83, wherein the intervention is a surgery.


Emb. 85: The method of any one of embodiments 58, 59 and 75-84, wherein the intervention is a surgery combined with the administration of a drug.


Emb. 86: The method of any one of embodiments 58, 59 and 75-85, wherein the intervention is a surgery selected from the group consisting of: a proctocolectomy, and an ileal pouch anal anastomosis.


Emb. 87: The method of any one of embodiments 58, 59 and 75-86, wherein the intervention is administration of a drug selected from the group consisting of: an iron supplement, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, cyclosporine, an anti-TNF-alpha antibody, infliximab, adalimumab, golimumab, methotrexate, an anti-α4-integrin antibody, vedolizumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.


Emb. 88: The method of any one of embodiments 58, 59 and 75-87, wherein the intervention is administration of a drug selected from the group consisting of: cyclosporine, and golimumab.


Emb. 89: The method of any one of embodiments 58-88, wherein the expression of MMP-7 or the concentration of MMP-7 is measured in the sample ex vivo.


Emb. 90: The method of any one of embodiments 58-89, wherein MMP-7 expression is measured by a technique selected from the group consisting of: whole transcriptome analysis, whole-transcriptome microarray, Northern blot, DNA microarray, PCR, sequencing PCR, RT-PCR, quantitative PCR, restriction fragment length polymorphism, in situ hybridization assay, and a competitive-binding assay.


Emb. 91: The method of any one of embodiments 58-90, wherein MMP-7 concentration is measured by a technique selected from the group consisting of: Western blot, ELISA, two-dimensional gel electrophoresis, mass spectrometry, protein interaction profiling, a competitive binding assay, a non-competitive binding assay, a radioimmunoassay, an enzyme immunoassays, an enzyme linked immunosorbent assay (ELISA), a sandwich immunoassay, a precipitation reaction, a gel diffusion reaction, an immunodiffusion assay, an agglutination assay, a complement-fixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, NMR analysis, and an immunoelectrophoresis assay.


Emb. 92: The method of any one of embodiments 58-91, wherein the patient is suffering from IBD.


Emb. 93: The method of any one of embodiments 58-92, wherein the patient displays a symptom selected from the group consisting of: severe diarrhea, abdominal pain, fatigue, and weight loss.


Emb. 94: The method of any one of embodiments 58-93, wherein the patient displays severe diarrhea, abdominal pain, fatigue, and weight loss.


Emb. 95: The method of any one of embodiments 58-94, wherein the sample is intestinal tissue.


Emb. 96: The method of any one of embodiments 58-95, wherein the sample is from the subject's large intestine.


Emb. 97: The method of any one of embodiments 58-96, wherein the sample is colonic tissue.


Emb. 98: The method of any one of embodiments 58-97, wherein the sample is ileal tissue.


Emb. 99: A kit for measuring HD5 and MMP-7 in a sample, the kit comprising: a first assay for measuring at least one of the expression of human HD5 and the concentration of human HD5 in a sample; and a second assay for measuring at least one of the expression of human MMP-7 and the concentration of human MMP-7 in a sample.


Emb. 100: The kit of embodiment 99, wherein said kit is for the diagnosis of inflammatory bowel disease.


Emb. 101: The kit of any one of embodiments 99-100, wherein the first assay comprises an antibody that recognizes human HD5; and wherein the second assay comprises antibody that recognizes human MMP-7.


Emb. 102: The kit of any one of embodiments 99-101, wherein the first assay comprises an oligonucleotide probe that binds to human HD5 cDNA; and wherein the second assay comprises an oligonucleotide probe that binds to human MMP-7 cDNA.


Emb. 103: The kit of any one of embodiments 99-102, wherein the first assay comprises a pair of primers complementary to a region of human HD5 cDNA; and wherein the second assay comprises a pair of primers complementary to a region of human MMP-7 cDNA.


Emb. 104: The kit of any one of embodiments 99-103, wherein: the first assay comprises a means for detecting HD5 protein; and the second assay comprises a means for detecting MMP-7 protein.


Emb. 105: The kit of embodiment 104, wherein: the means for detecting the HD5 protein is a first probe comprising a first ligand group that specifically binds to HD5 protein; and the means for detecting MMP-7 protein is a second probe comprising a second ligand group that specifically binds to MMP-7 protein.


Emb. 106: The kit of embodiment 105, wherein the first ligand group is an immunoglobulin.


Emb. 107: The kit of any one of embodiments 105-106, wherein the second ligand group is an immunoglobulin.


Emb. 108: The kit of any one of embodiments 105-107, wherein the first probe and the second probe are immobilized to a surface.


Emb. 109: The kit of any one of embodiments 99-108, wherein: the assay for measuring the expression of HD5 detects a first target sequence of at least 15 bp that is present in a first cDNA or mRNA of HD5; and the assay for measuring the expression of MMP-7 detects a second target sequence of at least 15 bp that is present in a second cDNA or mRNA of MMP-7.


Emb. 110: The kit of embodiment 109, wherein: the assay for detecting the first target sequence comprises a first probe comprising a first polynucleotide of at least 15 bp that hybridizes under highly stringent conditions with the first target sequence of at least 15 bp that is present in the first cDNA or mRNA of HD5; and the assay for detecting the second target sequence comprises a second probe comprising a second polynucleotide of at least 15 bp that hybridizes under highly stringent conditions with the second target sequence of at least 15 bp that is present in the second cDNA or mRNA of MMP-7.


Emb. 111: The kit of any one of embodiments 109-110, comprising a container of a reverse transcriptase.


Emb. 112: The kit of any one of embodiments 102-111, wherein the first probe comprises a first reporter, and the second probe comprises a second reporter.


Emb. 113: The kit of embodiment 112, wherein the first reporter is selected from the group consisting of: a radionuclide, a stable isotope, a fluorophore, a chromophore, an enzyme, a magnetic particle, and a quantum dot; and the second reporter selected from the group consisting of: a radionuclide, a fluorophore, a chromophore, an enzyme, a magnetic particle, and a quantum dot.


Emb. 114: The kit of any one of embodiments 110-113, wherein the first polynucleotide is single stranded DNA; and wherein the second polynucleotide is single stranded DNA.


Emb. 115: The kit of any one of embodiments 102-114, wherein the first probe and the second probe are components of a DNA array.


Emb. 116: The kit of any one of embodiments 102-115, wherein the first probe and the second probe are components of a DNA microarray.


Emb. 117: The kit of any one of embodiments 110-116, wherein the first polynucleotide is at least 20 bp and the second polynucleotide is at least 20 bp.


Emb. 118: The kit of any one of embodiments 110-117, wherein the first polynucleotide is at least 25 bp and the second polynucleotide is at least 25 bp.


Emb. 119: A method of diagnosing and treating Crohn's disease in a subject suffering from inflammatory bowel disease, the method comprising:


obtaining a sample from the patient;


measuring at least one of the expression of HD5 and the concentration of HD5 in the sample;


comparing the expression of HD5 or the concentration of HD5 in the sample to a benchmark value that is typical of a subject not suffering from Crohn's disease;


diagnosing Crohn's disease if the expression of HD5 or the concentration of HD5 in the sample significantly exceeds the benchmark value; and treating the subject for Crohn's disease by way of a non-surgical intervention.


Emb. 120: A method of diagnosing Crohn's disease in a subject suffering from inflammatory bowel disease, comprising: measuring the level of HD5 or HD5 expression in a sample from the subject, wherein the measuring is selected from the group consisting of radioimmunoassays, reverse transcriptase PCR (RT-PCR) assays, immunohistochemistry assays, in situ hybridization assays, competitive-binding assays, Western Blot analyses, ELISA assays and proteomic approaches, cation-ion exchange, NMR analysis, genome-wide transcriptome analysis, mass spectrometry, and combinations thereof; and diagnosing the subject as suffering from Crohn's disease if the level of HD5 is indicative of a subject having Crohn's disease.


Emb. 121: The method of embodiment 120, wherein the subject is diagnosed as suffering from Crohn's disease if the level of HD5 or HD5 expression is from about 1.9×107 HD5 mRNA Transcript per 10 ng RNA to about 7×107 HD5 mRNA Transcript per 10 ng RNA.


Emb. 122: A method of diagnosing ulcerative colitis in a subject suffering from inflammatory bowel disease, comprising: measuring the level of HD5 or HD5 expression in a sample from the subject, wherein the measuring is selected from the group consisting of radioimmunoassays, reverse transcriptase PCR (RT-PCR) assays, immunohistochemistry assays, in situ hybridization assays, competitive-binding assays, Western Blot analyses, ELISA assays and proteomic approaches, cation-ion exchange, NMR analysis, genome-wide transcriptome analysis, mass spectrometry, and combinations thereof; and diagnosing the subject as suffering from ulcerative colitis if the level of HD5 or HD5 expression is indicative of a subject having ulcerative colitis.


Emb. 123: The method of embodiment 122, wherein the subject is diagnosed as suffering from ulcerative colitis if the level of HD5 or HD5 expression is from about 6×105 HD5 mRNA Transcript per 10 ng RNA to about 1.8×107 HD5 mRNA Transcript per 10 ng RNA.


Emb. 124: A method of treating inflammatory bowel disease in a subject, comprising: measuring the level of HD5 or HD5 expression present in a sample obtained from the subject, said measuring step wherein the measuring is selected from the group consisting of radioimmunoassays, reverse transcriptase PCR (RT-PCR) assays, immunohistochemistry assays, in situ hybridization assays, competitive-binding assays, Western Blot analyses, ELISA assays and proteomic approaches, cation-ion exchange, NMR analysis, genome-wide transcriptome analysis, mass spectrometry, and combinations thereof, whereby a level of HD5 or HD5 expression is obtained; and if the level of HD5 or HD5 expression is at a level indicative of a subject not having Crohn's disease, treating the inflammatory bowel disease in the subject with a suitable medical treatment for ulcerative colitis; if the level of HD5 or HD5 expression is at a level indicative of a subject having Crohn's disease, treating the inflammatory bowel disease in the subject with a suitable medical treatment for Crohn's disease.


Emb. 125: The method according to embodiment 122, wherein the suitable medical treatment for ulcerative colitis comprises performing ileal pouch anal anastomosis in the subject.


Emb. 126: The method according to embodiment 122, wherein the suitable medical treatment for Crohn's disease comprises the administration of one or more of 5aminosalicylate, a corticosteroid, and an immunosuppressant to the subject.


Emb. 127: The method according to embodiment 122, wherein the sample is collected from the large intestine, and the subject is human.


Emb. 128: An assay for detecting elevated levels of HD5, comprising an HD5 antibody capable of binding with HD5.


Emb. 129: The assay according to embodiment 128, wherein the assay is provided in a kit.


Emb. 130: The novel and non-obvious embodiments and features disclosed herein.


Emb. 131: A method of measuring a biomarker in a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: obtaining a sample from the patient; and measuring a level of the biomarker in the same, the level of the biomarker selected from the group consisting of: the expression of the biomarker, the activity of the biomarker, and the concentration of the biomarker; wherein said biomarker is selected from Table 1.


Emb. 132: A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: performing the method of measuring the level of the biomarker in the patient according to embodiment 131; and performing an intervention on the patient to treat Crohn's disease.


Emb. 133: Any one of the methods of embodiments 131-132, comprising: comparing the level of the biomarker in the sample to a benchmark value that is typical of a subject not suffering from Crohn's disease; and diagnosing Crohn's disease if the expression of the biomarker in the sample significantly differs from the benchmark value.


CONCLUSIONS

It is to be understood that any given elements of the disclosed embodiments of the invention may be embodied in a single structure, a single step, a single substance, or the like. Similarly, a given element of the disclosed embodiment may be embodied in multiple structures, steps, substances, or the like.


The foregoing description illustrates and describes the processes, machines, manufactures, compositions of matter, and other teachings of the present disclosure. Additionally, the disclosure shows and describes only certain embodiments of the processes, machines, manufactures, compositions of matter, and other teachings disclosed, but as mentioned above, it is to be understood that the teachings of the present disclosure are capable of use in various other combinations, modifications and environments and are capable of changes or modifications within the scope of the teachings as expressed herein, commensurate with the skill and/or knowledge of a person having ordinary skill in the relevant art. The embodiments described hereinabove are further intended to explain certain best modes known of practicing the processes, machines, manufactures, compositions of matter, and other teachings of the present disclosure and to enable others skilled in the art to utilize the teachings of the present disclosure in such, or other, embodiments and with the various modifications required by the particular applications or uses. Accordingly, the processes, machines, manufactures, compositions of matter, and other teachings of the present disclosure are not intended to limit the exact embodiments and examples disclosed herein. Any section headings herein are provided only for consistency with the suggestions of 37 C.F.R. § 1.77, or otherwise to provide organizational queues. These headings shall not limit or characterize the invention(s) set forth herein.













TABLE 1





Gene
Gene

p-value
Fold


Information
Symbol
RefSeq
(CC vs UC)
Increase



















NM_021010 // DEFA5 // defensin, alpha 5,
DEFA5
NM_021010
7.23E−05
31.0374


Paneth cell-specific // 8p23.1 // 1670






NM_002909 // REG1A // regenerating islet-
REG1A
NM_002909
0.00321456
21.9439


derived 1 alpha // 2p12 // 5967 /// ENS






NM_138938 // REG3A // regenerating islet-
REG3A
NM_138938
0.000310891
17.3268


derived 3 alpha // 2p12 // 5068 /// NM_






NM_001926 // DEFA6 // defensin, alpha 6,
DEFA6
NM_001926
0.0024893
16.139


Paneth cell-specific // 8p23.1 // 1671






NM_058186 // FAM3B // family with
FAM3B
NM_058186
0.00116588
14.6887


sequence similarity 3, member B // 21q22.3 //






NM_006507 // REG1B // regenerating islet-
REG1B
NM_006507
0.0120953
13.9675


derived 1 beta // 2p12 // 5968 /// ENST






NM_001074 // UGT2B7 // UDP
UGT2B7
NM_001074
0.0154146
9.92532


glucuronosyltransferase 2 family,






polypeptide B7 // 4






NM_001285 // CLCA1 // chloride channel
CLCA1
NM_001285
0.00297816
9.07579


accessory 1 // 1p22.3 // 1179 /// ENST000






NM_003122 // SPINK1 // serine peptidase
SPINK1
NM_003122
0.007176
7.60063


inhibitor, Kazal type 1 // 5q32 // 6690






NM_001076 // UGT2B15 // UDP
UGT2B15
NM_001076
0.0169187
7.12294


glucuronosyltransferase 2 family,






polypeptide B15 //






NM_001076 // UGT2B15 // UDP
UGT2B15
NM_001076
0.0169187
7.12294


glucuronosyltransferase 2 family,






polypeptide B15 //






NM_000343 // SLC5A1 // solute carrier
SLC5A1
NM_000343
0.00447091
7.0494


family 5 (sodium/glucose cotransporter), m






NM_000134 // FABP2 // fatty acid binding
FABP2
NM_000134
0.0300574
6.63756


protein 2, intestinal // 4q28-q31 // 21






NM_000035 // ALDOB // aldolase B,
ALDOB
NM_000035
0.0444145
6.30502


fructose-bisphosphate // 9q21.3-q22.2 // 229/






NM_002770 // PRSS2 // protease, serine, 2
PRSS2
NM_002770
0.0052665
6.27999


(trypsin 2) // 7q34 // 5645 /// ENST00






NM_005379 // MYO1A // myosin IA //
MYO1A
NM_005379
0.00588172
5.72861


12q13-q14 // 4640 /// ENST00000300119 //






MYO1






NM_007329 // DMBT1 // deleted in
DMBT1
NM_007329
0.0365636
5.56609


malignant brain tumors 1 // 10q26.13 // 1755 //






NM_031457 // MS4A8B // membrane-spanning
MS4A8B
NM_031457
0.00577952
5.34254


4-domains, subfamily A, member 8B // 11






NM_001041 // SI // sucrase-isomaltase
SI
NM_001041
0.0417578
5.23854


(alpha-glucosidase) // 3q25.2-q26.2 // 647






NM_000482 // APOA4 // apolipoprotein A-IV //
APOA4
NM_000482
0.0468523
5.15957


11q23 // 337 /// ENST00000357780 //






NM_006418 // OLFM4 // olfactomedin 4 //
OLFM4
NM_006418
0.038931
5.05883


13q14.3 // 10562 /// ENST00000219022 //






NM_000482 // APOA4 // apolipoprotein A-IV //
APOA4
NM_000482
0.0472178
4.92519


11q23 // 337 /// ENST00000357780 //






NM_004133 // HNF4G // hepatocyte nuclear
HNF4G
NM_004133
0.0113549
4.8964


factor 4, gamma // 8q21.11 // 3174 ///






NM_017675 // CDHR2 // cadherin-related
CDHR2
NM_017675
0.00253568
4.82206


family member 2 // 5q35.2 // 54825 /// NM






NM_005588 // MEP1A // meprin A, alpha
MEP1A
NM_005588
0.0198087
4.78504


(PABA peptide hydrolase) // 6p12-p11 // 42






NM_002354 // EPCAM // epithelial cell
EPCAM
NM_002354
0.0242383
4.77321


adhesion molecule // 2p21 // 4072 /// ENST






NM_001172312 // PLS1 // plastin 1 // 3q23 //
PLS1
NM_001172312
0.0155248
4.73894


5357 /// NM_001145319 // PLS1 // pl






NM_002354 // EPCAM // epithelial cell
EPCAM
NM_002354
0.0297878
4.72533


adhesion molecule // 2p21 // 4072 /// ENST






NM_001150 // ANPEP // alanyl (membrane)
ANPEP
NM_001150
0.0203087
4.58929


aminopeptidase // 15q25-q26 // 290 /// E






NM_001077 // UGT2B17 // UDP
UGT2B17
NM_001077
0.0267812
4.51157


glucuronosyltransferase 2 family,






polypeptide B17 //






NM_002591 // PCK1 //
PCK1
NM_002591
0.0333639
4.50793


phosphoenolpyruvate carboxykinase 1






(soluble) // 20q13.31 /






NM_021804 // ACE2 // angiotensin I
ACE2
NM_021804
0.0271919
4.49025


converting enzyme (peptidyl-dipeptidase A) 2






NM_024308 // DHRS11 // dehydrogenase/
DHRS11
NM_024308
0.0176773
4.41914


reductase (SDR family) member 11 // 17q12/






NM_019010 // KRT20 // keratin 20 //
KRT20
NM_019010
0.026162
4.35459


17q21.2 // 54474 /// ENST00000167588 // KRT2






ENST00000319509 // MUC3A // mucin 3A,
MUC3A
ENST00000319509
0.00353785
4.28484


cell surface associated // 7q22 // 4584 //






NM_000379 // XDH // xanthine
XDH
NM_000379
0.00289109
4.17476


dehydrogenase // 2p23.1 // 7498 ///






ENST00000379416






NM_007127 // VIL1 // villin 1 // 2q35 // 7429 ///
VIL1
NM_007127
0.00825691
4.16925


ENST00000248444 // VIL1 // vil






NM_025130 // HKDC1 // hexokinase domain
HKDC1
NM_025130
0.00344261
4.13874


containing 1 // 10q22.1 // 80201 /// ENS






NR_029578 // MIR192 // microRNA 192 //
MIR192
NR_029578
0.00199884
4.12467


11q13.1 // 406967






NM_004063 // CDH17 // cadherin 17, LI
CDH17
NM_004063
0.0331015
4.12001


cadherin (liver-intestine) // 8q22.1 // 10






NM_024922 // CES3 // carboxylesterase 3 //
CES3
NM_024922
0.0022354
4.11886


16q22.1 // 23491 ///NM_001185177 //






NM_033049 // MUC13 // mucin 13, cell
MUC13
NM_033049
0.0271079
4.11287


surface associated // 3q21.2 // 56667 /// E






NM_000888 // ITGB6 // integrin, beta 6 //
ITGB6
NM_000888
0.000602949
4.09738


2q24.2 // 3694 /// ENST00000283249 //






NM_004963 // GUCY2C // guanylate
GUCY2C
NM_004963
0.00645462
4.0793


cyclase 2C (heat stable enterotoxin receptor)/






NM_004293 // GDA // guanine deaminase //
GDA
NM_004293
0.0208862
4.0739


9q21.13 // 9615 /// ENST00000358399 //






NM_001307 // CLDN7 // claudin 7 // 17p13 //
CLDN7
NM_001307
0.0213404
4.06183


1366 /// NM_001185022 // CLDN7 // cl






NR_033807 // CYP3A5 // cytochrome P450,
CYP3A5
NR_033807
0.0046334
4.04376


family 3, subfamily A, polypeptide 5 //






NM_021924 // CDHR5 // cadherin-related
CDHR5
NM_021924
0.00480695
3.97925


family member 5 // 11p15.5 // 53841 /// N






NM_001010922 // BCL2L15 // BCL2-like 15 //
BCL2L15
NM_001010922
0.027053
3.96946


1p13.2 // 440603 /// ENST00000393316






NM_020770 // CGN // cingulin // 1q21 //
CGN
NM_020770
0.00129584
3.94184


57530 /// ENST00000271636 // CGN // cing






NM_032787 // GPR128 // G protein-coupled
GPR128
NM_032787
0.00779494
3.93937


receptor 128 // 3q12.2 // 84873 /// ENS






NM_138933 // A1CF // APOBEC1
A1CF
NM_138933
0.00976589
3.79699


complementation factor // 10q11.23 // 29974 /// NM_






NM_152311 // CLRN3 // clarin 3 // 10q26.2 //
CLRN3
NM_152311
0.0132404
3.74982


119467 /// ENST00000368671 // CLRN3






NM_007072 // HHLA2 // HERV-H LTR-
HHLA2
NM_007072
0.0139075
3.74668


associating 2 // 3q13.13 // 11148 /// ENST00000






NM_003399 // XPNPEP2 // X-prolyl
XPNPEP2
NM_003399
0.0359348
3.73179


aminopeptidase (aminopeptidase P) 2,






membrane-b






NM_021258 // IL22RA1 // interleukin 22
IL22RA1
NM_021258
0.00520995
3.72759


receptor, alpha 1 // 1p36.11 // 58985 ///






NM_000149 // FUT3 // fucosyltransferase 3
FUT3
NM_000149
0.0106419
3.70158


(galactoside 3(4)-L-fucosyltransferase






NM_002644 // PIGR // polymeric
PIGR
NM_002644
0.0363588
3.68869


immunoglobulin receptor // 1q31-q41 // 5284 /// E






NM_001136503 // C19orf77 // chromosome
C19orf77
NM_001136503
0.0114867
3.6586


19 open reading frame 77 // 19p13.3 // 28






NR_024626 // C17orf73 // chromosome 17
C17orf73
NR_024626
0.00240775
3.64138


open reading frame 73 // 17q21.33 // 5501






NM_020973 // GBA3 // glucosidase, beta,
GBA3
NM_020973
0.0362758
3.63402


acid 3 (cytosolic) // 4p15.2 // 57733 //






NM_023944 // CYP4F12 // cytochrome
CYP4F12
NM_023944
0.00468827
3.62246


P450, family 4, subfamily F, polypeptide 12/






NM_024320 // PRR15L // proline rich 15-like //
PRR15L
NM_024320
0.0331566
3.60367


17q21.32 // 79170 /// ENST0000030






NM_005495 // SLC17A4 // solute carrier
SLC17A4
NM_005495
0.0299201
3.59753


family 17 (sodium phosphate), member 4 //






NM_001135099 // TMPRSS2 // transmembrane
TMPRSS2
NM_001135099
0.0351257
3.57585


protease, serine 2 // 21q22.3 // 7113/






NM_001193434 // C10orf81 // chromosome
C10orf81
NM_001193434
0.00228381
3.5687


10 open reading frame 81 // 10q25.3 // 79






NM_001935 // DPP4 // dipeptidyl-peptidase
DPP4
NM_001935
0.0302652
3.49144


4 // 2q24.3 // 1803 /// ENST0000036053






NM_001644 // APOBEC1 // apolipoprotein B
APOBEC1
NM_001644
0.0138008
3.48792


mRNA editing enzyme, catalytic polypept






NM_004360 // CDH1 // cadherin 1, type 1,
CDH1
NM_004360
0.010781
3.48059


E-cadherin (epithelial) // 16q22.1 // 9






NM_024921 // POF1B // premature ovarian
POF1B
NM_024921
0.0313161
3.44457


failure, 1B // Xq21.2 // 79983 /// ENST0






NM_002416 // CXCL9 // chemokine (C-X-C
CXCL9
NM_002416
0.00248734
3.44146


motif) ligand 9 // 4q21 // 4283 /// ENST0






NM_014479 // ADAMDEC1 // ADAM-like,
ADAMDEC1
NM_014479
0.00203661
3.42469


decysin 1 // 8p21.2 // 27299 ///






NM_00114527






NM_001112706 // SCIN // scinderin //
SCIN
NM_001112706
0.00493508
3.3952


7p21.3 // 85477 /// NM_033128 // SCIN // sc






NR_024345 // NCRNA00262 // non-protein
NCRNA00262
NR_024345
0.037473
3.39502


coding RNA 262 // 12q24.31 // 283460






NM_002273 // KRT8 // keratin 8 // 12q13 //
KRT8
NM_002273
0.0146545
3.39222


3856 /// ENST00000293308 // KRT8 // k






NM_001038603 // MARVELD2 // MARVEL
MARVELD2
NM_001038603
0.0179974
3.37682


domain containing 2 // 5q13.2 // 153562 /// E






NM_001038603 // MARVELD2 // MARVEL
MARVELD2
NM_001038603
0.0179974
3.37682


domain containing 2 // 5q13.2 // 153562 /// E






NM_144575 // CAPN13 // calpain 13 //
CAPN13
NM_144575
0.013239
3.36885


2p22-p21 // 92291 /// ENST00000295055 // CA






NM_022129 // PBLD // phenazine
PBLD
NM_022129
0.00497915
3.3666


biosynthesis-like protein domain containing // 10






NM_000775 // CYP2J2 // cytochrome P450,
CYP2J2
NM_000775
0.0196093
3.36302


family 2, subfamily J, polypeptide 2 //






NM_001135195 // SLC39A5 // solute carrier
SLC39A5
NM_001135195
0.00623473
3.34227


family 39 (metal ion transporter), mem






NM_138788 // TMEM45B // transmembrane
TMEM45B
NM_138788
0.0306305
3.33725


protein 45B // 11q24.3 // 120224 /// ENST0






NM_176813 // AGR3 // anterior gradient
AGR3
NM_176813
0.0400823
3.32266


homolog 3 (Xenopus laevis) // 7p21.1 // 1






NM_022901 // LRRC19 // leucine rich repeat
LRRC19
NM_022901
0.0294679
3.31296


containing 19 // 9p21.2 // 64922 ///






NM_139053 // EPS8L3 // EPS8-like 3 //
EPS8L3
NM_139053
0.00371579
3.29224


1p13.3 // 79574 /// NM_133181 // EPS8L3 //






NM_017697 // ESRP1 // epithelial splicing
ESRP1
NM_017697
0.0234665
3.27492


regulatory protein 1 // 8q22.1 // 5484






NM_002457 // MUC2 // mucin 2, oligomeric
MUC2
NM_002457
0.0182535
3.26416


mucus/gel-forming // 11p15.5 // 4583 //






NR_001296 // TRY6 // trypsinogen C // 7q34 //
TRY6
NR_001296
0.0203767
3.24356


154754 /// NM_002770 // PRSS2 // p






NM_002773 // PRSS8 // protease, serine, 8 //
PRSS8
NM_002773
0.0131026
3.2405


16p11.2 // 5652 /// ENST00000317508






NM_025214 // CCDC68 // coiled-coil domain
CCDC68
NM_025214
0.00627753
3.2264


containing 68 // 18q21 // 80323 /// NM






NM_001943 // DSG2 // desmoglein 2 //
DSG2
NM_001943
0.0357587
3.22627


18q12.1 // 1829 /// ENST00000261590 // DSG2






NM_000772 // CYP2C18 // cytochrome
CYP2C18
NM_000772
0.0100284
3.20876


P450, family 2, subfamily C, polypeptide 18/






NM_000767 // CYP2B6 // cytochrome P450,
CYP2B6
NM_000767
0.00589423
3.19484


family 2, subfamily B, polypeptide 6 //






NM_016234 // ACSL5 // acyl-CoA synthetase
ACSL5
NM_016234
0.00353915
3.19242


long-chain family member 5 // 10q25.1-






NM_145865 // ANKS4B // ankyrin repeat
ANKS4B
NM_145865
0.027168
3.16823


and sterile alpha motif domain containing






NM_032579 // RETNLB // resistin like beta //
RETNLB
NM_032579
0.0226491
3.14305


3q13.1 // 84666 /// ENST00000295755






NM_021978 // ST14 // suppression of
ST14
NM_021978
0.0143682
3.14171


tumorigenicity 14 (colon carcinoma) // 11q24






NM_000492 // CFTR // cystic fibrosis
CFTR
NM_000492
0.0330127
3.13524


transmembrane conductance regulator






(ATP-bi






NM_018842 // BAIAP2L1 // BAI1-associated
BAIAP2L1
NM_018842
0.00626097
3.13099


protein 2-like 1 // 7q22.1 // 55971 ///






NM_001165958 // GSDMB // gasdermin B //
GSDMB
NM_001165958
0.0013942
3.1309


17q12 // 55876 /// NM_001042471 // GSDMB






NM_024422 // DSC2 // desmocollin 2 //
DSC2
NM_024422
0.0115939
3.11862


18q12.1 // 1824 /// NM_004949 // DSC2 // d






NM_006017 // PROM1 // prominin 1 //
PROM1
NM_006017
0.0116042
3.10273


4p15.32 // 8842 /// NM_001145847 //






PROM1 //






NM_017878 // HRASLS2 // HRAS-like
HRASLS2
NM_017878
0.0267887
3.09847


suppressor 2 // 11q12.3 // 54979 ///






ENST00000






NM_002203 // ITGA2 // integrin, alpha 2
ITGA2
NM_002203
0.00793505
3.07141


(CD49B, alpha 2 subunit of VLA-2 recepto






NM_005123 // NR1H4 // nuclear receptor
NR1H4
NM_005123
0.0456782
3.06865


subfamily 1, group H, member 4 // 12q23.1






NM_001145862 // MTMR11 // myotubularin
MTMR11
NM_001145862
0.00116554
3.03455


related protein 11 // 1q12-q21 // 10903/






NM_018414 // ST6GALNAC1 // ST6 (alpha-
ST6GALNAC1
NM_018414
0.0240185
3.0202


N-acetyl-neuraminyl-2,3-beta-galactosyl-1,






NM_001080527 // MYO7B // myosin VIIB //
MYO7B
NM_001080527
0.00130692
2.99927


2q21.1 // 4648 /// ENST00000428314 // MY






NM_002153 // HSD17B2 // hydroxysteroid
HSD17B2
NM_002153
0.0213389
2.99803


(17-beta) dehydrogenase 2 // 16q24.1-q24.






AK095678 // LOC151009 // hypothetical
LOC151009
AK095678
0.000466288
2.99502


LOC151009 // 2q13 // 151009 /// AK056084/






NM_000769 // CYP2C19 // cytochrome
CYP2C19
NM_000769
0.0193957
2.99186


P450, family 2, subfamily C, polypeptide 19/






NM_000790 // DDC // dopa decarboxylase
DDC
NM_000790
0.0257511
2.98778


(aromatic L-amino acid decarboxylase) //






NM_001143948 // C6orf105 // chromosome
C6orf105
NM_001143948
0.0220945
2.95786


6 open reading frame 105 // 6p24.1 // 848






NM_001015001 // CKMT1A // creatine
CKMT1A
NM_001015001
0.042629
2.95709


kinase, mitochondrial 1A // 15q15 // 548596/






NM_001015001 // CKMT1A // creatine
CKMT1A
NM_001015001
0.042629
2.95709


kinase, mitochondrial 1A // 15q15 // 548596/






NM_019893 // ASAH2 // N-acylsphingosine
ASAH2
NM_019893
0.0167497
2.95643


amidohydrolase (non-lysosomal ceramidase






NM_001002236 // SERPINA1 // serpin
SERPINA1
NM_001002236
0.0170929
2.94245


peptidase inhibitor, clade A (alpha-1 antipro






NM_002031 // FRK // fyn-related kinase //
FRK
NM_002031
0.0177896
2.93608


6q21-q22.3 // 2444 /// ENST00000368626






NM_001190482 // PCSK5 // proprotein
PCSK5
NM_001190482
0.00160967
2.92603


convertase subtilisin/kexin type 5 // 9q21.3






NM_004415 // DSP // desmoplakin // 6p24 //
DSP
NM_004415
0.0116502
2.91732


1832 /// NM_001008844 // DSP // desmo






NM_004591 // CCL20 // chemokine (C-C
CCL20
NM_004591
0.0229351
2.91511


motif) ligand 20 // 2q33-q37 // 6364 /// NM






NM_000561 // GSTM1 // glutathione S-
GSTM1
NM_000561
0.032505
2.91233


transferase mu 1 // 1p13.3 // 2944 /// NM_14






NM_000927 // ABCB1 // ATP-binding
ABCB1
NM_000927
0.03279
2.89709


cassette, sub-family B (MDR/TAP), member 1 //






NM_000187 // HGD // homogentisate 1,2-
HGD
NM_000187
0.0180393
2.8961


dioxygenase // 3q13.33 // 3081 /// ENST000






NM_000187 // HGD // homogentisate 1,2-
HGD
NM_000187
0.0180393
2.8961


dioxygenase // 3q13.33 // 3081 /// ENST000






NM_153676 // USH1C // Usher syndrome
USH1C
NM_153676
0.00547469
2.88241


1C (autosomal recessive, severe) // 11p14.3






NM_005624 // CCL25 // chemokine (C-C
CCL25
NM_005624
0.0492359
2.86049


motif) ligand 25 // 19p13.2 // 6370 /// ENS






NM_004174 // SLC9A3 // solute carrier
SLC9A3
NM_004174
0.0173616
2.8567


family 9 (sodium/hydrogen exchanger), memb






NM_001306 // CLDN3 // claudin 3 // 7q11.23 //
CLDN3
NM_001306
0.0490185
2.84657


1365 /// ENST00000395145 // CLDN3






NM_001114309 // ELF3 // E74-like factor 3
ELF3
NM_001114309
0.00265363
2.84098


(ets domain transcription factor, epit






NM_000507 // FBP1 // fructose-1,6-
FBP1
NM_000507
0.022351
2.83767


bisphosphatase 1 // 9q22.3 // 2203 ///






NM_0011






NM_025257 // SLC44A4 // solute carrier
SLC44A4
NM_025257
0.0415598
2.83697


family 44, member 4 // 6p21.3 // 80736 //






NM_025257 // SLC44A4 // solute carrier
SLC44A4
NM_025257
0.0415598
2.83697


family 44, member 4 // 6p21.3 // 80736 //






NM_025257 // SLC44A4 // solute carrier
SLC44A4
NM_025257
0.0415598
2.83697


family 44, member 4 // 6p21.3 // 80736 //






NM_001017970 // TMEM30B // transmembrane
TMEM30B
NM_001017970
0.00717685
2.83259


protein 30B // 14q23.1 // 161291 /// EN






NM_003963 // TM4SF5 // transmembrane 4
TM4SF5
NM_003963
0.0295851
2.82875


L six family member 5 // 17p13.3 // 9032






NM_002242 // KCNJ13 // potassium
KCNJ13
NM_002242
0.0400838
2.82471


inwardly-rectifying channel, subfamily J,






membe






NM_017655 // GIPC2 // GIPC PDZ domain
GIPC2
NM_017655
0.0155498
2.81938


containing family, member 2 // 1p31.1 // 5






NM_001127605 // LIPA // lipase A,
LIPA
NM_001127605
0.000449938
2.81611


lysosomal acid, cholesterol esterase // 10q23.






NM_001249 // ENTPD5 // ectonucleoside
ENTPD5
NM_001249
0.0118697
2.81265


triphosphate diphosphohydrolase 5 // 14q24






NM_005358 // LMO7 // LIM domain 7 //
LMO7
NM_005358
0.00460576
2.80795


13q22.2 // 4008 /// NM_015842 // LMO7 // LI






NM_018667 // SMPD3 // sphingomyelin
SMPD3
NM_018667
0.00228114
2.80665


phosphodiesterase 3, neutral membrane






(neutr






NM_004563 // PCK2 // phosphoenolpyruvate
PCK2
NM_004563
0.00983672
2.79262


carboxykinase 2 (mitochondrial) // 14q1






NM_003657 // BCAS1 // breast carcinoma
BCAS1
NM_003657
0.0213345
2.78368


amplified sequence 1 // 20q13.2 // 8537/






NM_024850 // BTNL8 // butyrophilin-like 8 //
BTNL8
NM_024850
0.0446038
2.7769


5q35.3 // 79908 /// NM_001040462 //






NM_020672 // S100A14 // S100 calcium
S100A14
NM_020672
0.0202797
2.77156


binding protein A14 // 1q21.3 // 57402 ///






NM_033229 // TRIM15 // tripartite motif-
TRIM15
NM_033229
0.0097609
2.77095


containing 15 // 6p21.3 // 89870 /// ENS






NM_033229 // TRIM15 // tripartite motif-
TRIM15
NM_033229
0.0097609
2.77095


containing 15 // 6p21.3 // 89870 /// ENS






NM_033229 // TRIM15 // tripartite motif-
TRIM15
NM_033229
0.0097609
2.77095


containing 15 // 6p21.3 // 89870 /// ENS






NM_001144060 // NHSL1 // NHS-like 1 //
NHSL1
NM_001144060
0.0124428
2.7705


6q23.3 // 57224 /// NM_020464 // NHSL1 //






NM_003869 // CES2 // carboxylesterase 2 //
CES2
NM_003869
0.0197746
2.76326


16q22.1 // 8824 /// NR_036684 // CES2






NM_199187 // KRT18 // keratin 18 // 12q13 //
KRT18
NM_199187
0.0272938
2.7567


3875 /// NM_000224 // KRT18 // kera






NM_002842 // PTPRH // protein tyrosine
PTPRH
NM_002842
0.00126103
2.75623


phosphatase, receptor type, H // 19q13.4






NM_001105248 // TMC5 // transmembrane
TMC5
NM_001105248
0.015439
2.74553


channel-like 5 // 16p12.3 // 79838 /// NM_






NM_001145809 // MYH14 // myosin, heavy
MYH14
NM_001145809
0.00203315
2.74198


chain 14, non-muscle // 19q13.33 // 79784






NM_001054 // SULT1A2 // sulfotransferase
SULT1A2
NM_001054
0.0273843
2.73


family, cytosolic, 1A, phenol-preferrin






NM_024850 // BTNL8 // butyrophilin-like 8 //
BTNL8
NM_024850
0.0433332
2.7165


5q35.3 // 79908 /// NM_001159708 //






NM_006147 // IRF6 // interferon regulatory
IRF6
NM_006147
0.00663477
2.71435


factor 6 // 1q32.3-q41 // 3664 /// EN






NM_000457 // HNF4A // hepatocyte nuclear
HNF4A
NM_000457
0.00414138
2.70616


factor 4, alpha // 20q13.12 // 3172 ///






NM_138809 // CMBL //
CMBL
NM_138809
0.0336993
2.69623


carboxymethylenebutenolidase homolog






(Pseudomonas) // 5p15.






NM_001080467 // MYO5B // myosin VB //
MYO5B
NM_001080467
0.00639465
2.69568


18q21 // 4645 /// ENST00000285039 // MYO5B






NM_153274 // BEST4 // bestrophin 4 //
BEST4
NM_153274
0.0313639
2.68747


1p33-p32.3 // 266675 /// ENST00000372207/






NM_020775 // KIAA1324 // KIAA1324 //
KIAA1324
NM_020775
0.0214297
2.68133


1p13.3 // 57535 /// ENST00000234923 // KIAA






NM_001004320 // TMEM195 // transmembrane
TMEM195
NM_001004320
0.0149666
2.67293


protein 195 // 7p21.2 // 392636 /// ENS






NM_001091 // ABP1 // amiloride binding
ABP1
NM_001091
0.0487109
2.66772


protein 1 (amine oxidase (copper-containi






NM_016245 // HSD17B11 // hydroxysteroid
HSD17B11
NM_016245
0.0216559
2.66473


(17-beta) dehydrogenase 11 // 4q22.1 //






NM_006144 // GZMA // granzyme A
GZMA
NM_006144
0.00618242
2.66284


(granzyme 1, cytotoxic T-lymphocyte-






associated s






NM_001039372 // HEPACAM2 // HEPACAM family
HEPACAM2
NM_001039372
0.0201907
2.6524


member 2 // 7q21.3 // 253012 /// NM_1






NM_001197097 // PRSS3 // protease,
PRSS3
NM_001197097
0.0173103
2.63924


serine, 3 // 9p11.2 // 5646 /// NM_007343 //






NM_012214 // MGAT4A // mannosyl (alpha-
MGAT4A
NM_012214
0.00113208
2.62742


1,3-)-glycoprotein beta-1,4-N-acetylgluco






NM_019894 // TMPRSS4 // transmembrane
TMPRSS4
NM_019894
0.0362683
2.60764


protease, serine 4 // 11q23.3 // 56649 ///






NM_003810 // TNFSF10 // tumor necrosis
TNFSF10
NM_003810
0.0129809
2.60509


factor (ligand) superfamily, member 10 //






NM_022842 // CDCP1 // CUB domain
CDCP1
NM_022842
0.0167874
2.60268


containing protein 1 // 3p21.31 // 64866 /// NM






NM_001136493 // MFSD2A // major
MFSD2A
NM_001136493
0.00343618
2.59815


facilitator superfamily domain containing 2A //






NM_018265 // C1orf 106 // chromosome 1
C1orf106
NM_018265
0.00613223
2.59677


open reading frame 106 // 1q32.1 // 55765






NM_000063 // C2 // complement component
C2
NM_000063
0.0117239
2.59406


2 // 6p21.3 // 717 /// NM_001145903 // C






NM_000063 // C2 // complement component
C2
NM_000063
0.0117239
2.59406


2 // 6p21.3 // 717 /// NM_001145903 // C






NM_000625 // NOS2 // nitric oxide synthase
NOS2
NM_000625
0.0089305
2.59304


2, inducible // 17q11.2-q12 // 4843/






NM_001677 // ATP1B1 // ATPase, Na+/K+
ATP1B1
NM_001677
0.0131783
2.58871


transporting, beta 1 polypeptide // 1q24/






NM_004751 // GCNT3 // glucosaminyl (N-
GCNT3
NM_004751
0.0432197
2.58761


acetyl) transferase 3, mucin type // 15q21






NM_002021 // FMO1 // flavin containing
FMO1
NM_002021
0.0408097
2.57646


monooxygenase 1 // 1q24.3 // 2326 /// ENS






NM_033292 // CASP1 // caspase 1,
CASP1
NM_033292
0.00634065
2.57013


apoptosis-related cysteine peptidase






(interleuk






NM_147161 // ACOT11 // acyl-CoA
ACOT11
NM_147161
0.0462671
2.53682


thioesterase 11 // 1p32.3 // 26027 ///






ENST00000






NM_001039112 // FER1L6 // fer-1-like 6
FER1L6
NM_001039112
0.0413201
2.53444


(C. elegans) // 8q24.1 // 654463 /// ENST






NM_212543 // B4GALT4 // UDP-
B4GALT4
NM_212543
0.00083206
2.53146


Gal:betaGIcNAc beta 1,4-






galactosyltransferase, poly






NM_182762 // MACC1 // metastasis
MACC1
NM_182762
0.0113734
2.52994


associated in colon cancer 1 // 7p21.1 //






34638






NM_001461 // FMO5 // flavin containing
FMO5
NM_001461
0.0227505
2.52925


monooxygenase 5 // 1q21.1 // 2330 /// NM_






NM_031219 // HDHD3 // haloacid
HDHD3
NM_031219
0.00048055
2.52696


dehalogenase-like hydrolase domain






containing 3/






NM_001010872 // FAM83B // family with
FAM83B
NM_001010872
0.00806204
2.52496


sequence similarity 83, member B // 6p12.1






NM_024533 // CHST5 // carbohydrate (N-
CHST5
NM_024533
0.026327
2.51739


acetylglucosamine 6-O) sulfotransferase 5






NM_000063 // C2 // complement component
C2
NM_000063
0.0114041
2.51419


2 // 6p21.3 // 717 /// NM_001145903 // C






NM_004624 // VIPR1 // vasoactive intestinal
VIPR1
NM_004624
0.00331244
2.50863


peptide receptor 1 // 3p22 // 7433 /






NM_004572 // PKP2 // plakophilin 2 // 12p11 //
PKP2
NM_004572
0.042448
2.49612


5318 /// NM_001005242 // PKP2 //






NM_032521 // PARD6B // par-6 partitioning
PARD6B
NM_032521
0.00395798
2.49598


defective 6 homolog beta (C. elegans)






NM_024915 // GRHL2 // grainyhead-like 2
GRHL2
NM_024915
0.00624177
2.49455


(Drosophila) // 8q22.3 // 79977 /// ENST






NM_003982 // SLC7A7 // solute carrier
SLC7A7
NM_003982
0.00813405
2.49274


family 7 (cationic amino acid transporter,






NM_198584 // CA13 // carbonic anhydrase
CA13
NM_198584
0.00510852
2.48988


XIII // 8q21.2 // 377677 /// ENST0000032






ENST00000319509 // MUC3A // mucin 3A,
MUC3A
ENST00000319509
0.0135883
2.4817


cell surface associated // 7q22 // 4584 //






NM_021102 // SPINT2 // serine peptidase
SPINT2
NM_021102
0.0219176
2.48131


inhibitor, Kunitz type, 2 // 19q13.1 //






NM_080489 // SDCBP2 // syndecan binding
SDCBP2
NM_080489
0.000789754
2.47862


protein (syntenin) 2 // 20p13 // 27111/






NM_001144967 // NEDD4L // neural
NEDD4L
NM_001144967
0.0227827
2.47791


precursor cell expressed, developmentally






down-






NM_001982 // ERBB3 // v-erb-b2
ERBB3
NM_001982
0.0175723
2.47531


erythroblastic leukemia viral oncogene






homolog 3






NM_000240 // MAOA // monoamine oxidase
MAOA
NM_000240
0.0446884
2.47082


A // Xp11.3 // 4128 /// ENST00000338702/






NM_182960 // PRELID2 // PRELI domain
PRELID2
NM_182960
0.00837834
2.47032


containing 2 // 5q32 // 153768 /// NM_13849






NM_017720 // STAP2 // signal transducing
STAP2
NM_017720
0.016285
2.46781


adaptor family member 2 // 19p13.3 // 5






NM_138700 // TRIM40 // tripartite motif-
TRIM40
NM_138700
0.0336507
2.45989


containing 40 // 6p22.1 // 135644 /// EN






NM_000050 // ASS1 // argininosuccinate
ASS1
NM_000050
0.0132614
2.43678


synthase 1 // 9q34.1 // 445 /// NM_054012






NM_005021 // ENPP3 // ectonucleotide
ENPP3
NM_005021
0.0149678
2.43651


pyrophosphatase/phosphodiesterase 3 //






6q22






NM_001130080 // IFI27 // interferon, alpha-
IFI27
NM_001130080
0.0140236
2.43613


inducible protein 27 // 14q32 // 3429






NM_001979 // EPHX2 // epoxide hydrolase
EPHX2
NM_001979
0.00690804
2.43531


2, cytoplasmic // 8p21 // 2053 /// BC011






NM_017700 // ARHGEF38 // Rho guanine
ARHGEF38
NM_017700
0.00476968
2.42966


nucleotide exchange factor (GEF) 38 // 4q24






NM_019080 // NDFIP2 // Nedd4 family
NDFIP2
NM_019080
0.00576011
2.42832


interacting protein 2 // 13q31.1 // 54602 //






NM_001135181 // SLC5A9 // solute carrier
SLC5A9
NM_001135181
0.0296431
2.42215


family 5 (sodium/glucose cotransporter)






NM_032717 // AGPAT9 // 1-acylglycerol-3-
AGPAT9
NM_032717
0.0147877
2.41843


phosphate O-acyltransferase 9 // 4q21.23






NM_001145303 // TMC4 // transmembrane
TMC4
NM_001145303
0.00110774
2.41442


channel-like 4 // 19q13.42 // 147798 /// N






NM_138700 // TRIM40 // tripartite motif-
TRIM40
NM_138700
0.0250665
2.41358


containing 40 // 6p22.1 // 135644 /// EN






NM_138700 // TRIM40 // tripartite motif-
TRIM40
NM_138700
0.0250665
2.41358


containing 40 // 6p22.1 // 135644 /// EN






NM_203463 // LASS6 // LAG1 homolog,
LASS6
NM_203463
0.00156196
2.41203


ceramide synthase 6 // 2q24.3 // 253782 ///






NM_001730 // KLF5 // Kruppel-like factor 5
KLF5
NM_001730
0.0129015
2.40278


(intestinal) // 13q22.1 // 688 /// EN






NM_001265 // CDX2 // caudal type
CDX2
NM_001265
0.0471437
2.402


homeobox 2 // 13q12.3 // 1045 ///






ENST000003810






NM_000239 // LYZ // lysozyme // 12q15 //
LYZ
NM_000239
0.0118582
2.39899


4069 /// ENST00000261267 // LYZ // lyso






NM_022772 // EPS8L2 // EPS8-like 2 //
EPS8L2
NM_022772
0.00191717
2.39231


11p15.5 // 64787 /// ENST00000318562 // EP






NM_025153 // ATP10B // ATPase, class V,
ATP10B
NM_025153
0.0273664
2.38677


type 10B // 5q34 // 23120 /// ENST000003






NM_178445 // CCRL1 // chemokine (C-C
CCRL1
NM_178445
0.0328488
2.38032


motif) receptor-like 1 // 3q22 // 51554 ///






NM_001031803 // LLGL2 // lethal giant
LLGL2
NM_001031803
0.00351395
2.36948


larvae homolog 2 (Drosophila) // 17q25.1/






NM_175058 // PLEKHA7 // pleckstrin
PLEKHA7
NM_175058
0.00170237
2.36502


homology domain containing, family A






member 7






NM_006714 // SMPDL3A // sphingomyelin
SMPDL3A
NM_006714
0.0236138
2.36218


phosphodiesterase, acid-like 3A // 6q22.31






NR_024158 // LOC25845 // hypothetical
LOC25845
NR_024158
0.0297858
2.35341


LOC25845 // 5p15.33 // 25845 ///






ENST00000






NM_016339 // RAPGEFL1 // Rap guanine
RAPGEFL1
NM_016339
0.026897
2.3526


nucleotide exchange factor (GEF)-like 1 //






NM_015888 // HOOK1 // hook homolog 1
HOOK1
NM_015888
0.0336071
2.34842


(Drosophila) // 1p32.1 // 51361 /// ENST000






NM_138737 // HEPH // hephaestin // Xq11-
HEPH
NM_138737
0.0118198
2.34595


q12 // 9843 /// NM_001130860 // HEPH //






NM_012079 // DGAT1 // diacylglycerol O-
DGAT1
NM_012079
0.023252
2.34522


acyltransferase 1 // 8q24.3 // 8694 /// E






NM_012079 // DGAT1 // diacylglycerol O-
DGAT1
NM_012079
0.023252
2.34522


acyltransferase 1 // 8q24.3 // 8694 /// E






NM_001017535 // VDR // vitamin D (1,25-
VDR
NM_001017535
0.0115491
2.34153


dihydroxyvitamin D3) receptor // 12q13.1






NM_001029874 // REP15 // RAB15 effector
REP15
NM_001029874
0.0477963
2.33656


protein // 12p11.22 // 387849 /// ENST00






NM_198495 // CTAGE4 // CTAGE family,
CTAGE4
NM_198495
0.00065154
2.33596


member 4 // 7q35 // 100128553 ///






NM_001145






NM_006548 // IGF2BP2 // insulin-like growth
IGF2BP2
NM_006548
8.80E−05
2.33476


factor 2 mRNA binding protein 2 // 3






NM_002985 // CCL5 // chemokine (C-C
CCL5
NM_002985
0.0247261
2.33002


motif) ligand 5 // 17q11.2-q12 // 6352 /// E






NM_001005328 // OR2A7 // olfactory
OR2A7
NM_001005328
0.00337105
2.32021


receptor, family 2, subfamily A, member 7 //






NM_018284 // GBP3 // guanylate binding
GBP3
NM_018284
0.013933
2.31798


protein 3 // 1p22.2 // 2635 /// ENST00000






NM_002829 // PTPN3 // protein tyrosine
PTPN3
NM_002829
0.0212048
2.31511


phosphatase, non-receptor type 3 // 9q31






NM_021073 // BMP5 // bone morphogenetic
BMP5
NM_021073
0.0201876
2.31001


protein 5 // 6p12.1 // 653 /// ENST00000






NM_178176 // MOGAT3 // monoacylglycerol
MOGAT3
NM_178176
0.00641018
2.30988


O-acyltransferase 3 // 7q22.1 // 346606






NM_000666 // ACY1 // aminoacylase 1 //
ACY1
NM_000666
0.0261486
2.30581


3p21.1 // 95 /// L07548 // ACY1 // aminoa






NM_001098634 // RBM47 // RNA binding
RBM47
NM_001098634
0.00857247
2.30203


motif protein 47 // 4p14 // 54502 /// NM_01






NM_080658 // ACY3 // aspartoacylase
ACY3
NM_080658
0.0498753
2.301


(aminocyclase) 3 // 11q13.2 // 91703 /// ENS






NR_003587 // MYO15B // myosin XVB
MYO15B
NR_003587
0.00759021
2.29754


pseudogene // 17q25.1 // 80022 ///






BC027875 //






NM_005435 // ARHGEF5 // Rho guanine
ARHGEF5
NM_005435
0.00766916
2.29684


nucleotide exchange factor (GEF) 5 // 7q33-q






NM_005435 // ARHGEF5 // Rho guanine
ARHGEF5
NM_005435
0.00846455
2.29311


nucleotide exchange factor (GEF) 5 // 7q33-q






NM_001017967 // MARVELD3 // MARVEL
MARVELD3
NM_001017967
0.0124186
2.2921


domain containing 3 // 16q22.2 // 91862 /// N






NM_003389 // CORO2A // coronin, actin
CORO2A
NM_003389
0.0203606
2.28709


binding protein, 2A // 9q22.3 // 7464 ///






NM_031469 // SH3BGRL2 // SH3 domain
SH3BGRL2
NM_031469
0.0214373
2.27245


binding glutamic acid-rich protein like 2 //






NM_030766 // BCL2L14 // BCL2-like 14
BCL2L14
NM_030766
0.0037691
2.26634


(apoptosis facilitator) // 12p13-p12 // 793






NR_002713 // NPY6R // neuropeptide Y
NPY6R
NR_002713
0.0429642
2.26407


receptor Y6 (pseudogene) // 5q31 // 4888 //






NM_001114086 // CLIC5 // chloride
CLIC5
NM_001114086
0.0269601
2.25433


intracellular channel 5 // 6p12.3 // 53405 ///






NM_003645 // SLC27A2 // solute carrier
SLC27A2
NM_003645
0.040906
2.2539


family 27 (fatty acid transporter), membe






NM_001136050 // DHRS1 //
DHRS1
NM_001136050
0.000608529
2.23931


dehydrogenase/reductase (SDR family)






member 1 // 14q12






NM_002164 // IDO1 // indoleamine 2,3-
IDO1
NM_002164
0.00532092
2.2314


dioxygenase 1 // 8p12-p11 // 3620 /// ENST0






NM_001171192 // GDPD2 //
GDPD2
NM_001171192
0.0455387
2.23073


glycerophosphodiester phosphodiesterase






domain containi






NM_016445 // PLEK2 // pleckstrin 2 //
PLEK2
NM_016445
0.0184048
2.22972


14q23.3 // 26499 /// ENST00000216446 // PL






NR_033122 // PDZD3 // PDZ domain
PDZD3
NR_033122
0.0104609
2.2269


containing 3 // 11q23.3 // 79849 ///






NM_0011684






NM_000932 // PLCB3 // phospholipase C,
PLCB3
NM_000932
0.01393
2.22018


beta 3 (phosphatidylinositol-specific) //






NM_018235 // CNDP2 // CNDP dipeptidase
CNDP2
NM_018235
0.000958173
2.20566


2 (metallopeptidase M20 family) // 18q22.






NM_032562 // PLA2G12B // phospholipase
PLA2G12B
NM_032562
0.0420214
2.20423


A2, group XIIB // 10q22.1 // 84647 /// EN






NM_021080 // DAB1 // disabled homolog 1
DAB1
NM_021080
0.04076
2.20106


(Drosophila) // 1p32-p31 // 1600 /// ENS






NM_001710 // CFB // complement factor B //
CFB
NM_001710
0.00181667
2.19954


6p21.3 // 629 /// ENST00000425368 //






NM_183240 // TM EM37 // transmembrane
TMEM37
NM_183240
0.0487149
2.19842


protein 37 // 2q14.2 // 140738 /// ENST0000






AK127847 // FLJ45950 // FLJ45950 protein //
FLJ45950
AK127847
0.00195329
2.198


11q24.3 // 399975






NM_001710 // CFB // complement factor B //
CFB
NM_001710
0.00220919
2.19758


6p21.3 // 629 /// ENST00000417261 //






NM_144590 // ANKRD22 // ankyrin repeat
ANKRD22
NM_144590
0.0445105
2.19752


domain 22 // 10q23.31 // 118932 /// ENST0






NM_002067 // GNA11 // guanine nucleotide
GNA11
NM_002067
0.014093
2.19185


binding protein (G protein), alpha 11 (






NM_006579 // EBP // emopamil binding
EBP
NM_006579
0.0115147
2.18786


protein (sterol isomerase) // Xp11.23-p11.2






NM_014873 // LPGAT1 //
LPGAT1
NM_014873
0.000550666
2.18469


lysophosphatidylglycerol acyltransferase 1 //






1q32 // 992






NM_030943 // AMN // amnionless homolog
AMN
NM_030943
0.00168811
2.18289


(mouse) // 14q32.3 // 81693 /// ENST00000






NM_016548 // GOLM1 // golgi membrane
GOLM1
NM_016548
0.0424472
2.18243


protein 1 // 9q21.33 // 51280 /// NM_177937






NM_032148 // SLC41A2 // solute carrier
SLC41A2
NM_032148
0.0301277
2.17752


family 41, member 2 // 12q23.3 // 84102/






NM_000949 // PRLR // prolactin receptor //
PRLR
NM_000949
0.0313649
2.17608


5p13.2 // 5618 /// ENST00000382002 //






NM_181642 // SPINT1 // serine peptidase
SPINT1
NM_181642
0.0361797
2.17498


inhibitor, Kunitz type 1 // 15q15.1 // 6






NM_001113567 // C17orf76 // chromosome
C17orf76
NM_001113567
0.0248369
2.17219


17 open reading frame 76 // 17p11.2 // 38






NM_000355 // TCN2 // transcobalamin II //
TCN2
NM_000355
0.0233279
2.17134


22q12.2 // 6948 /// NM_001184726 // TC






NM_015198 // COBL // cordon-bleu
COBL
NM_015198
0.0208672
2.1656


homolog (mouse) // 7p12.1 // 23242 ///






ENST0000






NM_024616 // C3orf52 // chromosome 3
C3orf52
NM_024616
0.00881101
2.16302


open reading frame 52 // 3q13.2 // 79669 //






NM_020469 // ABO // ABO blood group
ABO
NM_020469
0.00222828
2.16292


(transferase A, alpha 1-3-N-acetylgalactosam






NM_030908 // OR2A4 // olfactory receptor,
OR2A4
NM_030908
0.00568966
2.15894


family 2, subfamily A, member 4 // 6q2






NM_003980 // MAP7 // microtubule-
MAP7
NM_003980
0.0037529
2.15742


associated protein 7 // 6q23.3 // 9053 /// NM_0






NM_017417 // GALNT8 // UDP-N-acetyl-
GALNT8
NM_017417
0.013696
2.15417


alpha-D-galactosamine: polypeptide N-acetylga






NM_005410 // SEPP1 // selenoprotein P,
SEPP1
NM_005410
0.0133071
2.15347


plasma, 1 // 5q31 // 6414 /// NM_00108548






NM_152573 // RASEF // RAS and EF-hand
RASEF
NM_152573
0.0366785
2.15133


domain containing // 9q21.32 // 158158 ///






NM_006633 // IQGAP2 // IQ motif containing
IQGAP2
NM_006633
0.00969849
2.1509


GTPase activating protein 2 // 5q13.3






NM_152550 // SH3RF2 // SH3 domain
SH3RF2
NM_152550
0.00614396
2.15072


containing ring finger 2 // 5q32 // 153769 ///






NM_018686 // CMAS // cytidine
CMAS
NM_018686
0.0124234
2.14998


monophosphate N-acetylneuraminic acid






synthetase/






NM_025045 // BAIAP2L2 // BA11-associated
BAIAP2L2
NM_025045
0.0129162
2.14195


protein 2-like 2 // 22q13.1 // 80115 //






NM_001859 // SLC31A1 // solute carrier
SLC31A1
NM_001859
0.00838827
2.13821


family 31 (copper transporters), member 1






NM_016614 // TDP2 // tyrosyl-DNA
TDP2
NM_016614
0.0246156
2.13573


phosphodiesterase 2 // 6p22.3-p22.1 //






51567 //






NM_003848 // SUCLG2 // succinate-CoA
SUCLG2
NM_003848
0.00569037
2.13077


ligase, GDP-forming, beta subunit // 3p14.1






NM_017904 // TTC22 // tetratricopeptide
TTC22
NM_017904
0.0153126
2.12827


repeat domain 22 // 1p32.3 // 55001 ///






NM_003060 // SLC22A5 // solute carrier
SLC22A5
NM_003060
0.02024
2.12394


family 22 (organic cation/carnitine trans






NM_002662 // PLD1 // phospholipase D1,
PLD1
NM_002662
0.0135876
2.12113


phosphatidylcholine-specific // 3q26 // 5






NM_018964 // SLC37A1 // solute carrier
SLC37A1
NM_018964
0.0229039
2.12062


family 37 (glycerol-3-phosphate transport






NM_001251 // CD68 // CD68molecule //
CD68
NM_001251
0.00105743
2.11575


17p13 // 968 /// NM_001040059 // CD68 // C






NM_174941 // CD163L1 // CD163 molecule-
CD163L1
NM_174941
0.00407203
2.11396


like 1 // 12p13.3 // 283316 /// ENST00000






NM_016029 // DHRS7 //
DHRS7
NM_016029
0.0124063
2.11159


dehydrogenase/reductase (SDR family)






member 7 // 14q23.1/






NM_024101 // MLPH // melanophilin //
MLPH
NM_024101
0.00197625
2.10533


2q37.3 // 79083 /// NM_001042467 // MLPH //






NM_004670 // PAPSS2 // 3′-
PAPSS2
NM_004670
0.0403309
2.10272


phosphoadenosine 5′-phosphosulfate






synthase 2 // 10q24






AK172782 // GPAM // glycerol-3-phosphate
GPAM
AK172782
0.0314353
2.09633


acyltransferase, mitochondrial // 10q25






NM_001142685 // ARHGAP32 // Rho
ARHGAP32
NM_001142685
0.00415504
2.09203


GTPase activating protein 32 // 11q24.3 // 9743






NM_198495 // CTAGE4 // CTAGE family,
CTAGE4
NM_198495
0.00141321
2.0906


member 4 // 7q35 // 100128553 /// NM_001145






ENST00000439698 // P4HA2 // prolyl 4-
P4HA2
ENST00000439698
0.0142839
2.08741


hydroxylase, alpha polypeptide II // 5q31/






NM_015020 // PHLPP2 // PH domain and
PHLPP2
NM_015020
0.013905
2.08634


leucine rich repeat protein phosphatase 2/






NM_004252 // SLC9A3R1 // solute carrier
SLC9A3R1
NM_004252
0.00776993
2.0857


family 9 (sodium/hydrogen exchanger), me






NM_012243 // SLC35A3 // solute carrier
SLC35A3
NM_012243
0.0307101
2.07986


family 35 (UDP-N-acetylglucosamine (UDP-G






NM_020184 // CNNM4 // cyclin M4 // 2q11 //
CNNM4
NM_020184
0.02685
2.07897


26504 /// ENST00000377075 // CNNM4 //






NM_001490 // GCNT1 // glucosaminyl (N-
GCNT1
NM_001490
0.00172819
2.07671


acetyl) transferase 1, core 2 // 9q13 // 2






NM_003667 // LGR5 // leucine-rich repeat-
LGR5
NM_003667
0.0237574
2.07254


containing G protein-coupled receptor 5






NM_001966 // EHHADH // enoyl-CoA,
EHHADH
NM_001966
0.0130422
2.07114


hydratase/3-hydroxyacyl CoA






dehydrogenase // 3






NM_017726 // PPP1R14D // protein
PPP1R14D
NM_017726
0.0497008
2.07017


phosphatase 1, regulatory (inhibitor) subunit 1






NM_006994 // BTN3A3 // butyrophilin,
BTN3A3
NM_006994
0.00121808
2.06925


subfamily 3, member A3 // 6p21.3 // 10384/






NM_001039724 // NOSTRIN // nitric oxide
NOSTRIN
NM_001039724
0.00986343
2.06731


synthase trafficker // 2q31.1 // 115677






NR_026912 // ABHD11 // abhydrolase
ABHD11
NR_026912
0.000593971
2.05896


domain containing 11 // 7q11.23 // 83451 ///






NM_001145206 // KIAA1671 // KIAA1671 //
KIAA1671
NM_001145206
0.00446756
2.05612


22q11.23 // 85379 /// ENST00000358431 //






NM_153345 // TM EM 139 // transmembrane
TMEM139
NM_153345
0.00505302
2.05293


protein 139 // 7q34 // 135932 /// ENST0000






NM_001164694 // IYD // iodotyrosine
IYD
NM_001164694
0.022189
2.05208


deiodinase // 6q25.1 // 389434 /// NM_203395






NM_016472 // C14orf 129 // chromosome 14
C14orf129
NM_016472
0.048055
2.04519


open reading frame 129 // 14q32.2 // 515






NM_001017402 // LAMB3 // laminin, beta 3 //
LAMB3
NM_001017402
0.0267716
2.04174


1q32 // 3914 /// NM_001127641 // LAM






NM_004999 // MYO6 // myosin VI // 6q13 //
MYO6
NM_004999
0.00369349
2.04095


4646 /// ENST00000369977 // MYO6 // my






NR_027244 // LOC151009 // hypothetical
LOC151009
NR_027244
0.0115721
2.04078


LOC151009 // 2q13 // 151009 /// NR_027244






AB065085 // TOM1L1 // target of myb1
TOM1L1
AB065085
0.04656
2.03713


(chicken)-like 1 // 17q23.2 // 10040






NM_017750 // RETSAT // retinol saturase
RETSAT
NM_017750
0.0184264
2.03345


(all-trans-retinol 13,14-reductase) // 2






NM_004721 // MAP3K13 // mitogen-
MAP3K13
NM_004721
0.00937615
2.03148


activated protein kinase kinase kinase 13 // 3q2






NM_018677 // ACSS2 // acyl-CoA
ACSS2
NM_018677
0.0306269
2.02661


synthetase short-chain family member 2 // 20q11.2






NM_014317 // PDSS1 // prenyl (decaprenyl)
PDSS1
NM_014317
0.0365076
2.02171


diphosphate synthase, subunit 1 // 10p






NM_014498 // GOLIM4 // golgi integral
GOLIM4
NM_014498
0.00240934
2.02056


membrane protein 4 // 3q26.2 // 27333 ///






NM_033429 // CALML4 // calmodulin-like 4 //
CALML4
NM_033429
0.0419784
2.01981


15q23 // 91860 /// NM_001031733 // C






NR_036751 // HSP90AA6P // heat shock
HSP90AA6P
NR_036751
0.0220954
2.01604


protein 90 kDa alpha (cytosolic), class A me






NM_012120 // CD2AP // CD2-associated
CD2AP
NM_012120
0.00502091
2.0122


protein // 6p12 // 23607 ///






ENST0000035931






NM_005536 // IMPA1 // inositol(myo)-1 (or
IMPA1
NM_005536
0.0194688
2.01203


4)-monophosphatase 1 // 8q21.13-q21.3/






NM_001153 // ANXA4 // annexinA4 // 2p13 //
ANXA4
NM_001153
0.0255723
2.01151


307 /// ENST00000394295 // ANXA4 //






NM_000147 // FUCA1 // fucosidase, alpha-
FUCA1
NM_000147
0.00469253
2.0105


L-1, tissue // 1p34 // 2517 /// ENST000






NM_003774 // GALNT4 // UDP-N-acetyl-
GALNT4
NM_003774
0.00622316
2.00871


alpha-D-galactosamine: polypeptide N-






acetylga






NM_001122890 // GGT6 // gamma-
GGT6
NM_001122890
0.0328357
2.00627


glutamyltransferase 6 // 17p13.2 // 124975 /// NM_






NM_001164277 // SLC37A4 // solute carrier
SLC37A4
NM_001164277
0.0068184
2.00477


family 37 (glucose-6-phosphate transpo






NM_001565 // CXCL10 // chemokine (C-X-C
CXCL10
NM_001565
0.0468134
2.00368


motif) ligand 10 // 4q21 // 3627 /// ENS






NM_005030 // PLK1 // polo-like kinase 1 //
PLK1
NM_005030
0.0109795
2.00251


16p12.2 // 5347 /// ENST00000300093/






NM_001012631 // IL32 // interleukin 32 //
IL32
NM_001012631
0.0214868
2.00238


16p13.3 // 9235 /// NM_004221 // IL32






NM_005309 // GPT // glutamic-pyruvate
GPT
NM_005309
0.0098254
2.00201


transaminase (alanine aminotransferase) //






NM_005159 // ACTC1 // actin, alpha,
ACTC1
NM_005159
0.00451989
−2.00712


cardiac muscle 1 // 15q11 -q14 // 70 /// ENST






NM_130385 // MRVI1 // murine retrovirus
MRVI1
NM_130385
0.0186352
−2.00908


integration site 1 homolog // 11p15 // 1






NR_003329 // SNORD116-14 // small
SNORD116-14
NR_003329
0.00710694
−2.01066


nucleolar RNA, C/D box 116-14 // 15q11.2 // 10






NM_030751 // ZEB1 // zinc finger E-box
ZEB1
NM_030751
0.0190641
−2.01665


binding homeobox 1 // 10p11.2 // 6935 ///






NM_001321 // CSRP2 // cysteine and
CSRP2
NM_001321
0.0130189
−2.01975


glycine-rich protein 2 // 12q21.1 // 1466 ///






NM_199460 // CACNA1C // calcium
CACNA1C
NM_199460
0.0164629
−2.03364


channel, voltage-dependent, L type, alpha 1C sub






NM_007078 // LDB3 // LIM domain binding 3 //
LDB3
NM_007078
0.013344
−2.03636


10q22.3-q23.2 // 11155 /// NM_00117






ENST00000436525 // C15orf51 // dynamin 1
C15orf51
ENST00000436525
0.0479813
−2.04311


pseudogene // 15q26.3 // 196968






ENST00000436525 // C15orf51 // dynamin 1
C15orf51
ENST00000436525
0.0479813
−2.04311


pseudogene // 15q26.3 // 196968






NM_001042454 // TGFB111 // transforming
TGFB1I1
NM_001042454
0.0141045
−2.0503


growth factor beta 1 induced transcript






NM_201266 // NRP2 // neuropilin 2 // 2q33.3 //
NRP2
NM_201266
0.0231808
−2.05329


8828 /// NM_003872 // NRP2 // neu






NM_014286 // NCS1 // neuronal calcium
NCS1
NM_014286
0.0400809
−2.05571


sensor 1 // 9q34 // 23413 /// NM_001128826






NR_002960 // SNORA20 // small nucleolar
SNORA20
NR_002960
0.0102255
−2.05618


RNA, H/ACA box 20 // 6q25.3 // 677806






NR_023343 // RNU4ATAC // RNA, U4atac
RNU4ATAC
NR_023343
0.0114016
−2.05953


small nuclear (U12-dependent splicing) // 2






NM_003829 // MPDZ // multiple PDZ
MPDZ
NM_003829
0.0230169
−2.06542


domain protein // 9p23 // 8777 /// ENST0000038






NM_182734 // PLCB1 // phospholipase C,
PLCB1
NM_182734
0.0285626
−2.0675


beta 1 (phosphoinositide-specific) // 20p






NM_212482 // FN1 // fibronectin 1 // 2q34 //
FN1
NM_212482
0.0289963
−2.06817


2335 /// NM_002026 // FN1 // fibron






NM_001166292 // PTCH2 // patched 2 //
PTCH2
NM_001166292
0.0155977
−2.06949


1p34.1 // 8643 /// ENST00000438067 // PTCH






NM_001128310 // SPARCL1 // SPARC-like
SPARCL1
NM_001128310
0.0275433
−2.0695


1 (hevin) // 4q22.1 // 8404 /// NM_004684






NR_003332 // SNORD116-17 // small
SNORD116-17
NR_003332
0.00123218
−2.07085


nucleolar RNA, C/D box 116-17 // 15q11.2 // 10






NR_003332 // SNORD116-17 // small
SNORD116-17
NR_003332
0.00123218
−2.07085


nucleolar RNA, C/D box 116-17 // 15q11.2 // 10






NM_001390 // DTNA // dystrobrevin, alpha //
DTNA
NM_001390
0.0140008
−2.07227


18q12 // 1837 /// NM_032975 // DTNA






NM_172316 // MEIS2 // Meis homeobox 2 //
MEIS2
NM_172316
0.012629
−2.07482


15q14 // 4212 /// NM_170677 // MEIS2 //






NM_032801 // JAM3 // junctional adhesion
JAM3
NM_032801
0.00375191
−2.08055


molecule 3 // 11q25 // 83700 /// ENST00






NM_001496 // GFRA3 // GDNF family
GFRA3
NM_001496
0.0143176
−2.08436


receptor alpha 3 // 5q31.1-q31.3 // 2676 /// E






NM_003116 // SPAG4 // sperm associated
SPAG4
NM_003116
0.0370178
−2.09743


antigen 4 // 20q11.21 // 6676 /// ENST000






NR_002754 // RNU5E // RNA, U5E small
RNU5E
NR_002754
0.0153145
−2.10499


nuclear // 1p36.22 // 26829 /// M77839 // R






NM_000109 // DM D // dystrophin // Xp21.2 //
DMD
NM_000109
0.0305823
−2.10535


1756 /// NM_004010 // DMD // dystrop






NM_005725 // TSPAN2 // tetraspanin 2 //
TSPAN2
NM_005725
0.00484522
−2.10726


1p13.2 // 10100 /// ENST00000369516 // T






ENST00000436525 // C15orf51 // dynamin 1
C15orf51
ENST00000436525
0.0401346
−2.11861


pseudogene // 15q26.3 // 196968






NM_001190839 // MGP // matrix Gia protein //
MGP
NM_001190839
0.0229696
−2.13146


12p12.3 // 4256 /// NM_000900 // MG






NM_031442 // TMEM47 // transmembrane
TMEM47
NM_031442
0.0162367
−2.16059


protein 47 // Xp11.4 // 83604 /// ENST00000






NM_002776 // KLK10 // kallikrein-related
KLK10
NM_002776
0.0131782
−2.16442


peptidase 10 // 19q13 // 5655 /// NM_14






NM_134269 // SMTN // smoothelin //
SMTN
NM_134269
0.0278447
−2.16615


22q12.2 // 6525 /// NM_134270 // SMTN // smoo






NM_002742 // PRKD1 // protein kinase D1 //
PRKD1
NM_002742
0.0208525
−2.17797


14q11 // 5587 /// ENST00000331968 //






NM_001001396 // ATP2B4 // ATPase, Ca++
ATP2B4
NM_001001396
0.0372252
−2.18014


transporting, plasma membrane 4 // 1q32.1






NM_005451 // PDLIM7 // PDZ and LIM
PDLIM7
NM_005451
0.00654348
−2.18595


domain 7 (enigma) // 5q35.3 // 9260 /// NM_20






NR_002952 // SNORA9 // small nucleolar
SNORA9
NR_002952
0.0244704
−2.19918


RNA, H/ACA box 9 // 7p13 // 677798 /// AK






NM_003069 // SMARCA1 // SWI/SNF
SMARCA1
NM_003069
0.00571381
−2.2109


related, matrix associated, actin dependent






regu






NR_003330 // SNORD116-15 // small
SNORD116-15
NR_003330
6.72E−05
−2.21218


nucleolar RNA, C/D box 116-15 // 15q11.2 //






10






NM_002398 // MEIS1 // Meis homeobox 1 //
MEIS1
NM_002398
0.0208728
−2.21341


2p14 // 4211 /// ENST00000272369 // MEI






ENST00000436525 // C15orf51 // dynamin 1
C15orf51
ENST00000436525
0.0297132
−2.22015


pseudogene // 15q26.3 // 196968






ENST00000436525 // C15orf51 // dynamin 1
C15orf51
ENST00000436525
0.0297132
−2.22015


pseudogene // 15q26.3 // 196968






NM_003734 // AOC3 // amine oxidase,
AOC3
NM_003734
0.0151647
−2.22019


copper containing 3 (vascular adhesion prote






AF391113 // C21orf70 // chromosome 21
C21orf70
AF391113
0.00109586
−2.22308


open reading frame 70 // 21q22.3 // 85395






NM_001937 // DPT // dermatopontin // 1q12-
DPT
NM_001937
0.0379186
−2.22359


q23 // 1805 /// ENST00000367817 // DPT






NM_012232 // PTRF // polymerase I and
PTRF
NM_012232
0.0194925
−2.23107


transcript release factor // 17q21.2 // 28






NM_024605 // ARHGAP10 // Rho GTPase
ARHGAP10
NM_024605
0.00832518
−2.23204


activating protein 10 // 4q31.23 // 79658 //






NM_022117 // TSPYL2 // TSPY-like2 //
TSPYL2
NM_022117
0.0134024
−2.23502


Xp11.2 // 64061 /// ENST00000375442 // TSP






NM_005100 // AKAP12 // A kinase (PRKA)
AKAP12
NM_005100
0.0357306
−2.24089


anchor protein 12 // 6q24-q25 // 9590 ///






AY423733 // DDR2 // discoidin domain
DDR2
AY423733
0.0358613
−2.2447


receptor tyrosine kinase 2 // 1q23.3 // 492






NM_153703 // PODN // podocan // 1p32.3 //
PODN
NM_153703
0.0277365
−2.26923


127435 /// ENST00000312553 // PODN //






NM_004370 // COL12A1 // collagen, type
COL12A1
NM_004370
0.0499701
−2.27002


XII, alpha 1 // 6q12-q13 // 1303 /// NM_0






NM_004137 // KCNMB1 // potassium large
KCNMB1
NM_004137
0.0277682
−2.27584


conductance calcium-activated channel, su






NM_014575 // SCHIP1 // schwannomin
SCHIP1
NM_014575
0.00470657
−2.28272


interacting protein 1 // 3q25.32-q25.33 // 29






NM_001753 // CAV1 // caveolin 1, caveolae
CAV1
NM_001753
0.0368534
−2.29054


protein, 22 kDa // 7q31.1 // 857 /// NM






NM_002338 // LSAMP // limbic system-
LSAMP
NM_002338
0.0456749
−2.30408


associated membrane protein // 3q13.2-q21 //






NM_058229 // FBXO32 // F-box protein 32 //
FBXO32
NM_058229
0.0422526
−2.30763


8q24.13 // 114907 /// NM_148177 // FB






NM_006765 // TUSC3 // tumor suppressor
TUSC3
NM_006765
0.00173576
−2.32217


candidate 3 // 8p22 // 7991 /// NM_178234






NM_015687 // FILIP1 // filamin A interacting
FILIP1
NM_015687
0.0158717
−2.32321


protein 1 // 6q14.1 // 27145 /// EN






NM_006080 // SEMA3A // sema domain,
SEMA3A
NM_006080
0.0142131
−2.32699


immunoglobulin domain (Ig), short basic doma






NM_000922 // PDE3B // phosphodiesterase
PDE3B
NM_000922
0.00420057
−2.33135


3B, cGMP-inhibited // 11p15.1 // 5140 //






NM_000722 // CACNA2D1 // calcium
CACNA2D1
NM_000722
0.0107345
−2.33411


channel, voltage-dependent, alpha 2/delta subun






NM_001197294 // DPYSL3 // dihydropyrimidinase-
DPYSL3
NM_001197294
0.0231385
−2.33517


like 3 // 5q32 // 1809 /// NM_0013






NM_172311 // STON1-GTF2A1L // STON1-
TON1-GTF2A1
NM_172311
0.0264382
−2.33729


GTF2A1L readthrough // 2p16.3 // 286749 ///






NM_000857 // GUCY1B3 // guanylate
GUCY1B3
NM_000857
0.0141507
−2.34285


cyclase 1, soluble, beta 3 // 4q31.3-q33 // 29






NR_033662 // CSF3 // colony stimulating
CSF3
NR_033662
0.036854
−2.35397


factor 3 (granulocyte) // 17q11.2-q12 //






NM_001706 // BCL6 // B-cell CLL/lymphoma
BCL6
NM_001706
0.0395014
−2.37213


6 // 3q27 // 604 /// NM_001130845 // BC






NM_014112 // TRPS1 // trichorhinophalangeal
TRPS1
NM_014112
0.021813
−2.37338


syndrome I // 8q24.12 // 7227 /// EN






NM_003275 // TMOD1 // tropomodulin 1 //
TMOD1
NM_003275
0.00926909
−2.39163


9q22.3 // 7111 /// NM_001166116 // TMOD1






NM_004040 // RHOB // ras homolog gene
RHOB
NM_004040
0.00209611
−2.39166


family, member B // 2p24 // 388 /// ENST00






NM_007281 // SCRG1 // stimulator of
SCRG1
NM_007281
0.0449505
−2.42771


chondrogenesis 1 // 4q34.1 // 11341 /// ENST






NM_053025 // MYLK // myosin light chain
MYLK
NM_053025
0.0334323
−2.44896


kinase // 3q21 // 4638 /// NM_053026 //






NM_133646 // ZAK // sterile alpha motif and
ZAK
NM_133646
0.0101002
−2.45225


leucine zipper containing kinase AZK






NM_001123364 // C6orf 186 // chromosome
C6orf186
NM_001123364
0.0338175
−2.45305


6 open reading frame 186 // 6q21 // 72846






NM_005909 // MAPI B // microtubule-
MAP1B
NM_005909
0.00199713
−2.45363


associated protein 1B // 5q13 // 4131 /// ENST






NM_001136191 // KANK2 // KN motif and
KANK2
NM_001136191
0.00418
−2.45823


ankyrin repeat domains 2 // 19p13.2 // 259






NR_002836 // PGM5P2 // phosphoglucomutase
PGM5P2
NR_002836
0.0106051
−2.46207


5 pseudogene 2 // 9q12 // 595135 /// N






NM_006988 // ADAMTS1 // ADAM
ADAMTS1
NM_006988
0.0212926
−2.47602


metallopeptidase with thrombospondin type






1 motif,






NM_001897 // CSPG4 // chondroitin sulfate
CSPG4
NM_001897
0.000233664
−2.47738


proteoglycan 4 // 15q24.2 // 1464 ///






NM_012134 // LMOD1 // leiomodin 1
LMOD1
NM_012134
0.0254164
−2.48821


(smooth muscle) // 1q32 // 25802 ///






ENST00000






NM_000856 // GUCY1A3 // guanylate
GUCY1A3
NM_000856
0.0154068
−2.49669


cyclase 1, soluble, alpha 3 // 4q31.3-q33|4q31






NR_002196 // H19 // H19, imprinted
H19
NR_002196
0.0422207
−2.49895


maternally expressed transcript (non-protein






NM_002667 // PLN // phospholamban //
PLN
NM_002667
0.0458219
−2.50528


6q22.1 // 5350 /// ENST00000357525 // PLN/






NM_004078 // CSRP1 // cysteine and
CSRP1
NM_004078
0.0389579
−2.51599


glycine-rich protein 1 // 1q32 // 1465 /// NM






NM_001141945 // ACTA2 // actin, alpha2,
ACTA2
NM_001141945
0.00367966
−2.51621


smooth muscle, aorta // 10q23.3 // 59/






NM_002986 // CCL11 // chemokine(C-C
CCL11
NM_002986
0.0132628
−2.5178


motif) ligand 11 // 17q21.1-q21.2 // 6356/






NM_033138 // CALD1 // caldesmon 1 //
CALD1
NM_033138
0.0229067
−2.51869


7q33 // 800 /// NM_033157 // CALD1 // calde






NM_001164836 // FXYD6 // FXYD domain
FXYD6
NM_001164836
0.0202065
−2.53004


containing ion transport regulator 6 // 11q






NM_003725 // HSD17B6 // hydroxysteroid
HSD17B6
NM_003725
0.0196889
−2.54527


(17-beta) dehydrogenase 6 homolog (mouse)






NM_001146312 // MYOCD // myocardin //
MYOCD
NM_001146312
0.0298805
−2.59465


17p11.2 // 93649 /// NM_153604 // MYOCD //






NM_015225 // PRUNE2 // prune homolog 2
PRUNE2
NM_015225
0.0217217
−2.59492


(Drosophila) // 9q21.2 // 158471 /// AB53






NM_001168278 // WWTR1 // WW domain
WWTR1
NM_001168278
0.014475
−2.60243


containing transcription regulator 1 // 3q23-






NM_001008711 // RBPMS // RNA binding
RBPMS
NM_001008711
0.00600769
−2.60406


protein with multiple splicing // 8p12 // 1






NM_001014796 // DDR2 // discoidin domain
DDR2
NM_001014796
0.00523497
−2.61121


receptor tyrosine kinase 2 // 1q23.3 //






NM_018640 // LMO3 // UM domain only 3
LMO3
NM_018640
0.042971
−2.63105


(rhombotin-like 2) // 12p12.3 // 55885 //






NR_002836 // PGM5P2 // phosphoglucomutase
PGM5P2
NR_002836
0.00678244
−2.64929


5 pseudogene 2 // 9q12 // 595135 /// N






NM_021914 // CFL2 // cofilin 2 (muscle) //
CFL2
NM_021914
0.0261349
−2.65343


14q12 // 1073 /// NM_138638 // CFL2/






NM_016277 // RAB23 // RAB23, member
RAB23
NM_016277
0.035448
−2.66122


RAS oncogene family // 6p11 // 51715 /// NM_






NM_145234 // CHRDL1 // chordin-like 1 //
CHRDL1
NM_145234
0.00265317
−2.67563


Xq23 // 91851 /// NM_001143981 // CHRDL






NM_001134439 // PHLDB2 // pleckstrin
PHLDB2
NM_001134439
0.0258326
−2.67775


homology-like domain, family B, member 2 //






NM_006832 // FERMT2 // fermitin family
FERMT2
NM_006832
0.0205617
−2.7145


member 2 // 14q22.1 // 10979 /// NM_00113






NM_001128205 // SULF1 // sulfatase 1 //
SULF1
NM_001128205
0.0335496
−2.73234


8q13.1 // 23213 /// NM_015170 // SULF1/






NM_194272 // RBPMS2 // RNA binding
RBPMS2
NM_194272
0.012053
−2.74286


protein with multiple splicing 2 // 15q22.31






NM_014476 // PDLIM3 // PDZ and LIM
PDLIM3
NM_014476
0.0110612
−2.7574


domain 3 // 4q35 // 27295 /// NM_001114107 //






NM_015886 // P115 // peptidase inhibitor 15 //
PI15
NM_015886
0.0312943
−2.78937


8q21.11 // 51050 /// ENST00000260






NM_003289 // TPM2 // tropomyosin 2 (beta) //
TPM2
NM_003289
0.0272347
−2.80338


9p13 // 7169 /// NM_213674 // TPM2






NM_001458 // FLNC // filamin C, gamma //
FLNC
NM_001458
0.0113027
−2.80588


7q32-q35 // 2318 /// NM_001127487 // FL






NM_006097 // MYL9 // myosin, light chain 9,
MYL9
NM_006097
0.0412118
−2.81849


regulatory // 20q11.23 // 10398 ///






NM_199460 // CACNA1C // calcium
CACNA1C
NM_199460
0.00694625
−2.83404


channel, voltage-dependent, L type, alpha






1C sub






NM_001232 // CASQ2 // calsequestrin 2
CASQ2
NM_001232
0.0349505
−2.84886


(cardiac muscle) // 1p13.3-p11 // 845 ///






NM_001193460 // MSRB3 // methionine
MSRB3
NM_001193460
0.0108076
−2.84899


sulfoxide reductase B3 // 12q14.3 // 253827






NM_001456 // FLNA // filamin A, alpha //
FLNA
NM_001456
0.0164878
−2.86026


Xq28 // 2316 /// NM_001110556 // FLNA/






NM_006366 // CAP2 // CAP, adenylate
CAP2
NM_006366
0.00596997
−2.89059


cyclase-associated protein, 2 (yeast) // 6p2






NM_001031701 // NT5DC3 // 5′-
NT5DC3
NM_001031701
0.0464686
−2.90347


nucleotidase domain containing 3 // 12q22-q23.1 //






NM_003999 // OSMR // oncostatin M
OSMR
NM_003999
0.0324297
−2.92605


receptor // 5p13.1 // 9180 /// NM_001168355 //






NM_001885 // CRYAB // crystallin, alpha B //
CRYAB
NM_001885
0.0163674
−2.96044


11q22.3-q23.1 // 1410 /// ENST00000






NM_000517 // HBA2 // hemoglobin, alpha 2 //
HBA2
NM_000517
0.0195505
−3.10109


16p13.3 // 3040 /// BC101846 // HBA1






NM_000558 // HBA1 // hemoglobin, alpha 1 //
HBA1
NM_000558
0.0195505
−3.10109


16p13.3 // 3039 /// BC101846 // HBA1






NM_004282 // BAG2 // BCL2-associated
BAG2
NM_004282
0.0108668
−3.11097


athanogene 2 // 6p12.1-p11.2 // 9532 /// EN






NM_022135 // POPDC2 // popeye domain
POPDC2
NM_022135
0.0219995
−3.1427


containing 2 // 3q13.33 // 64091 /// ENST00






NM_001001522 // TAGLN // transgelin //
TAGLN
NM_001001522
0.0148609
−3.35842


11q23.2 // 6876 /// NM_003186 // TAGLN //






NM_212482 // FN1 // fibronectin 1 // 2q34 //
FN1
NM_212482
0.00987492
−3.43741


2335 /// NM_002026 // FN1 // fibron






NM_133477 // SYNPO2 // synaptopodin 2 //
SYNPO2
NM_133477
0.0241716
−3.56252


4q26 // 171024 /// NM_001128933 // SYNP






NM_000450 // SELE // selectin E // 1q22-q25 //
SELE
NM_000450
0.0460446
−3.56423


6401 /// ENST00000333360 // SELE






NR_029686 // MIR145 // microRNA 145 //
MIR145
NR_029686
0.0119026
−3.58867


5q32 // 406937 /// NR_027180 // LOC728264






NM_022648 // TNS1 // tensin 1 // 2q35-q36 //
TNS1
NM_022648
0.00555851
−3.61273


7145 /// ENST00000171887 // TNS1 //






NM_001615 // ACTG2 // actin, gamma 2,
ACTG2
NM_001615
0.0379131
−3.62826


smooth muscle, enteric // 2p13.1 // 72 ///






NM_022844 // MYH11 // myosin, heavy
MYH11
NM_022844
0.0240032
−3.66415


chain 11, smooth muscle // 16p13.11 // 4629






NM_002205 // ITGA5 // integrin, alpha 5
ITGA5
NM_002205
0.0207749
−3.82521


(fibronectin receptor, alpha polypeptide






NM_001299 // CNN1 // calponin 1, basic,
CNN1
NM_001299
0.0413103
−3.84711


smooth muscle // 19p13.2-p13.1 // 1264/






NM_001034954 // SORBS1 // sorbin and
SORBS1
NM_001034954
0.00399907
−3.89048


SH3 domain containing 1 // 10q23.33 // 1058






NM_001927 // DES // desmin // 2q35 // 1674 ///
DES
NM_001927
0.0268126
−3.90558


ENST00000373960 // DES // desmin






NM_144617 // HSPB6 // heat shock protein,
HSPB6
NM_144617
0.0145209
−3.90993


alpha-crystallin-related, B6 // 19q13.






NM_015424 // CHRDL2 // chordin-like 2 //
CHRDL2
NM_015424
0.0247555
−4.23746


11q14 // 25884 /// ENST00000263671 // C






NM_000518 // HBB // hemoglobin, beta //
HBB
NM_000518
0.0255665
−4.3277


11p15.5 // 3043 /// ENST00000335295 // H






NM_002160 // TNC // tenascin C // 9q33 //
TNC
NM_002160
0.0126641
−4.4403


3371 /// ENST00000350763 // TNC // ten






NM_006198 // PCP4 // Purkinje cell protein
PCP4
NM_006198
0.0340302
−4.51736


4 // 21q22.2 // 5121 /// ENST00000328








Claims
  • 1. A method of measuring HD5 in a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: obtaining a sample from the patient; andmeasuring at least one of an expression of HD5 and a concentration of HD5 in the sample.
  • 2. A method of measuring MMP-7 in a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: obtaining a sample from the patient; andmeasuring at least one of the expression of MMP-7 and the concentration of MMP-7 in the sample.
  • 3. A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: performing the method of measuring MMP-7 in the patient according to claim 2; andperforming an intervention on the patient to treat Crohn's disease.
  • 4. The method of claim 2, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis or severe ulcerative colitis; and diagnosing Crohn's disease if the expression of MMP-7 or the concentration of MMP-7 in the sample does not significantly exceed the benchmark value.
  • 5. The method of claim 3, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis or severe ulcerative colitis; and diagnosing Crohn's disease if the expression of MMP-7 or the concentration of MMP-7 in the sample does not significantly exceed the benchmark value.
  • 6. The method of claim 2, wherein the expression of MMP-7 or the concentration of MMP-7 in the sample is below a benchmark value that is typical of a subject suffering from ulcerative colitis or severe ulcerative colitis.
  • 7. The method of claim 2, wherein the expression of MMP-7 is measured to be significantly less in the sample than in a control sample from a subject suffering from ulcerative colitis or severe ulcerative colitis.
  • 8. The method of claim 2, wherein the expression of MMP-7 is measured in the sample to be at most about 1/10 of the expression of MMP-7 measured in a control sample from a subject not suffering from ulcerative colitis or severe ulcerative colitis.
  • 9. The method of claim 2, wherein the sample is intestinal tissue, and comprising measuring the concentration of MMP-7 by: immunostaining the sample with an antiMMP-7 immunostaining agent; and measuring the percentage of cells in the sample that stain positive.
  • 10. The method of claim 2, wherein the intervention is not effective to treat ulcerative colitis or severe ulcerative colitis.
  • 11. The method of claim 2, wherein the intervention is administration of a drug, to the exclusion of a surgery.
  • 12. The method of claim 2, wherein the intervention is administration of a drug selected from the group consisting of: a vitamin supplement, vitamin B12, vitamin D, a mineral supplement, calcium, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, an anti-TNF-alpha antibody, infliximab, adalimumab, certolizumab pegol, methotrexate, an anti-α4-integrin antibody, natalizumab, vedolizumab, an anti-interleukin antibody, ustekinumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.
  • 13. The method of claim 2, wherein the intervention is administration of a drug selected from the group consisting of: vitamin B12, vitamin D, calcium, certolizumab pegol, methotrexate, and natalizumab.
  • 14. The method of claim 2, wherein the intervention is placement of the subject on a low fat diet.
  • 15. A method of treating a patient suffering from or at risk of inflammatory bowel disease (IBD), said method comprising: performing the method of measuring MMP-7 in the patient according to claim 2; andperforming an intervention on the patient to treat ulcerative colitis or severe ulcerative colitis.
  • 16. The method of claim 2, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis or severe ulcerative colitis; and diagnosing ulcerative colitis or severe ulcerative colitis if the expression of MMP-7 or the concentration of MMP-7 in the sample significantly exceeds the benchmark value.
  • 17. The method of claim 15, comprising: comparing the expression of MMP-7 or the concentration of MMP-7 in the sample to a benchmark value that is typical of a subject not suffering from ulcerative colitis or severe ulcerative colitis; and diagnosing ulcerative colitis or severe ulcerative colitis if the expression of MMP-7 or the concentration of MMP-7 in the sample significantly exceeds the benchmark value.
  • 18. The method of claim 15, wherein the sample is intestinal tissue, and comprising measuring the concentration of MMP-7 by: immunostaining the sample with an anti-MMP-7 immunostaining agent; and measuring the percentage of cells in the sample that stain positive.
  • 19. The method of claim 15, wherein the intervention is not effective to treat Crohn's disease.
  • 20. The method of claim 15, wherein the intervention is a surgery selected from the group consisting of: a proctocolectomy, and an ileal pouch anal anastomosis.
  • 21. The method of claim 15, wherein the intervention is administration of a drug selected from the group consisting of: an iron supplement, an anti-inflammatory, a corticosteroid, a 5-aminosalicylate, an immunosuppressant, azathioprine, mercaptopurine, cyclosporine, an anti-TNF-alpha antibody, infliximab, adalimumab, golimumab, methotrexate, an anti-α4-integrin antibody, vedolizumab, an antibacterial antibiotic, ciprofloxacin, and metronidazole.
  • 22. The method of claim 15, wherein the intervention is administration of a drug selected from the group consisting of: cyclosporine, and golimumab.
  • 23. A kit for measuring HD5 and MMP-7 in a sample, the kit comprising: a first assay for measuring at least one of the expression of human HD5 and the concentration of human HD5 in a sample; and a second assay for measuring at least one of the expression of human MMP-7 and the concentration of human MMP-7 in a sample.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. patent application Ser. No. 16/571,034, filed Sep. 13, 2019, which is a continuation of PCT/US2018/024069 filed Mar. 23, 2018, which claims the benefit of U.S. Provisional Patent Application No. 62/475,506, filed Mar. 23, 2017, each of which is relied upon for priority and incorporated by reference herein in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

This invention was made with government support under grant numbers R21DK095186; U54CA091408-09S1; U54CA091408-0952; U54RR026140; U54MD007593; UL1RR024975; UL1TR000445; G12MD007586; U54CA163069; R24 DA036420; and S10RR0254970 awarded by the National Institute of Health. The government has certain rights in the invention. In this context “government” refers to the government of the United States of America.

Provisional Applications (1)
Number Date Country
62475506 Mar 2017 US
Continuations (2)
Number Date Country
Parent 16571034 Sep 2019 US
Child 17872674 US
Parent PCT/US2018/024069 Mar 2018 US
Child 16571034 US