Skip to main content

Ulcerative colitis: functional analysis of the in-depth proteome

Abstract

Background

Ulcerative colitis (UC) is one major form of inflammatory bowel disease. The cause and the pathophysiology of the disease are not fully understood and we therefor aim in this study to identify important pathophysiological features in UC from proteomics data.

Methods

Colon mucosa biopsies from inflamed tissue of untreated UC patients at diagnosis and from healthy controls were obtained during colonoscopy. Quantitative protein data was acquired by bottom-up proteomics and furthermore processed with MaxQuant. The quantitative proteome data was analyzed with Perseus and enrichment data was analyzed by ClueGO for Cytoscape.

Results

The generated proteome dataset is to-date the deepest from colon mucosa biopsies with 8562 identified proteins whereof 6818 were quantified in > 70% of the samples. We report abundance differences between UC and healthy controls and the respective p values for all quantified proteins in the supporting information. From this data set enrichment analysis revealed decreased protein abundances in UC for metallothioneins, PPAR-inducible proteins, fibrillar collagens and proteins involved in bile acid transport as well as metabolic functions of nutrients, energy, steroids, xenobiotics and carbonate. On the other hand increased abundances were enriched in immune response and protein processing in the endoplasmic reticulum, e.g. unfolded protein response and signal peptidase complex proteins.

Conclusions

This explorative study describes the most affected functions in UC tissue. Our results complemented previous findings substantially. Decreased abundances of signal peptidase complex proteins in UC are a new discovery.

Background

Ulcerative colitis (UC) is a form of inflammatory bowel disease (IBD). The chronic inflammation of the colon in UC starts at the rectum and can progress continuously to proximal colon parts. UC affects 0.51% of the European and 0.25% of the North American population, with increasing prevalence [1]. The disease has a complex pathophysiology and the exact disease causes remain unclear. A genetic component, environmental factors, defects of the epithelial barrier, and dysregulated immune responses are involved [2].

Pathophysiological changes likely lie or are reflected in the abundance and state of proteins, the major functional units in every tissue. The proteomic analysis of colon biopsies affected by UC gives data on these changes at the main site of the disease. Advances in sample preparation, instrumentation, and analysis software allow for increasing proteome coverage and improved quantification. Higher proteome coverage, growing and new pathway databases, and improved software packages benefit comprehensive enrichment analyses of tissues.

Proteomic studies with different designs regarding samples and analytical method were previously conducted on colon mucosa affected by UC. Inflamed mucosa from untreated patients at UC debut [3] as well as macroscopically normal mucosa from treated UC patients [4] have been compared to biopsies from healthy controls in previous studies. A comparison of inflamed mucosa from untreated UC patients with mucosa from healthy controls as well as from Crohn’s disease patients was conducted on pediatric patients [5]. Another study compared inflamed and non-inflamed tissue from the same patients [6]. Despite the differences in the study designs, the studies present remarkably similar proteome changes [3]. They revealed novel insights into the UC pathophysiology, e.g. the role of neutrophil extracellular traps [4] and S100 proteins [3]. They also identified biomarkers for differentiating between UC and Crohn’s disease [5] and between pancolitis and partial colitis [7]. Furthermore, multiple studies investigated specific proteins of interest and their functions in UC, such as inducible nitric oxide synthase (NOS2) [8] and histaminase (AOC1) [9].

However, despite the challenges in extracting biological information from omics data and the shortcomings of focusing on proteins with the highest fold-changes [10], enrichment analyses were not the main scope of previous proteomics studies on UC. Omics studies typically produce large amounts of data. Especially in experiments with strong phenotypes these datasets can contain so many significant findings that a sensible interpretation without additional statistical analyses is barely possible. Interpretations based on only the highest fold-changes in such a dataset disregard large amounts of data and fail to recognize functional changes as a result of multiple moderately different abundances. Moreover, the biological interpretation is prone to be arbitrary, and it has further disadvantages. Enrichment analyses can overcome these difficulties [10] and the enrichment factors give an additional indication for the importance of the respective functions for the disease pathophysiology.

Beside proteomics, transcriptomic approaches have been applied to investigate UC [11]. These studies show differential mRNA expressions in UC tissue. The pathophysiological implications of these finding however are less clear because the correlation of mRNA and protein abundances is poor. The cause for that low correlation is that both the translation and the degradation of proteins are subject to several regulation mechanisms that are independent of mRNA levels [12].

We conducted a comprehensive enrichment analysis on a proteomics dataset of 8562 identified proteins from colon mucosa biopsies.

Materials and methods

Patients included and biopsies collection

Mucosal biopsies were collected from newly diagnosed treatment-naïve UC patients in Norway. The UC diagnosis was established upon clinical, endoscopic and histological criteria defined by the European Crohn and Colitis Organization (ECCO) guidelines [13]. Furthermore, the degree of inflammation was evaluated during colonoscopy using the scoring system of ulcerative colitis disease activity index (UCDAI) [14]. Moreover, TNF mRNA levels were measured by real-time PCR to assess the level of UC activity [15]. Subjects admitted for a cancer screening and with normal colonoscopy and histological findings served as healthy controls. None of the recruited subjects suffered from irritable bowel disease and they were not on nonsteroidal anti-inflammatory drug medication prior to the colonoscopy. The biopsies from the UC patients were obtained from the most inflamed area in rectum or sigmoid colon. Biopsies from the control group were obtained from the rectum. From each study participant, 3 adjacent biopsies were obtained from the inflamed mucosa. One biopsy was immediately immersed in RNAlater (Qiagen, Germany). The second biopsy was frozen immediately in a dry cryotube tube at − 70 °C until further analysis. The third biopsy was obtained for ordinary histological examinations (haematoxylin and eosin staining). 17 patients with debut of UC and 15 healthy controls were recruited as shown in Table 1.

Table 1 Baseline characteristics in patients included at debut of ulcerative colitis and in healthy controls (see “Patients included and biopsies collection” and “Baseline characteristics” sections for further details)

Baseline characteristics

The baseline characteristics are shown in Table 1 (see Additional file 1: Table 1). All subjects included showed typical histological findings according to UC and normal, non-inflamed mucosa, respectively.

Sample preparation

Samples were homogenized with 250 µl cooled lysis buffer [8 M urea, 5% sodium deoxycholate (SDC), 100 mM triethylammonium bicarbonate buffer pH 8.5 (TEAB)] in MagNA Lyser Green Bead tubes (Roche Diagnostics AG, Rotkreuz, Switzerland) with a MagNA Lyser Instrument (Roche Diagnostics AG, Rotkreuz, Switzerland) for 35 s at 6500 rpm. Lysates were frozen at − 70 °C until further sample preparation. All samples and standards were diluted/produced to contain 5 mM TEAB for the BCA assay (Pierce™ BCA Protein Assay Kit, Thermo Fisher Scientific). The assay was performed according to the manufacturer’s protocol. Lysate aliquots of 60 µg protein were transferred to Protein LoBind tubes (Eppendorf AG, Hamburg, Germany). Disulfide bridges were reduced with 1,4-dithiothreitol (DTT) at a concentration of 5 mM by incubation at 54 °C for 30 min. Cysteins were alkylated with 15 mM iodoacetamide (IAA) and incubation for 30 min at room temperature in the dark. DTT solution corresponding to a final concentration of 5 mM was added to remove excess IAA. Lys-C predigestion was performed under gentle agitation for 8 h at 37 °C with 0.6 µg Lys-C (enzyme-to-protein ratio 1:100, w/w, Wako Chemicals GmbH, Neuss, Germany) in a buffer containing 1 mM calcium chloride, 6 M urea, and 100 mM TEAB. Calcium chloride solution, 3 µg trypsin (enzyme-to-protein ratio 1:20, w/w, Sequencing Grade Modified Trypsin, Promega Corporation, Madison, USA), water, and 1 M TEAB were added to a final concentration of 1 mM calcium chloride, 1 M urea, and 100 mM TEAB. Tryptic digestion was performed under gentle agitation for 16 h at 37 °C.

Equal peptide amounts from each sample were combined to produce a standard mixture, which was subsequently labelled with TMT126. Aliquots corresponding to 25 µg peptides were labelled with the remaining TMTsixplex isotopes according to the manufacturer’s protocol. 0.4 mg TMT reagent were used for 25 µg peptides from samples with starting protein concentrations ≥ 2200 µg/ml. Higher amounts of TMT reagent were used for lower starting concentrations (see Additional file 1: Table 2).

The differently labelled peptides were mixed in equal amounts to in total 100 µg peptides. The digests were acidified with 50% formic acid (FA) to a final concentration of 2.5% FA and pH ≤ 2. The samples were centrifuged at 16,000 g for 15 min and the supernatants were carefully transferred to fresh tubes. Acetonitrile was removed by evaporation in a vacuum concentrator.

Trifluoroacetic acid (TFA) was added to a concentration of 0.1%. The samples were fractionated with Pierce™ High pH Reversed-Phase Peptide Fractionation Kit (Thermo Fisher, Rockford, USA) according to the manufacturer’s protocol. Samples exceeding 300 µl were stepwise loaded with 2 min centrifugation after each loading. The fractions were dried in a vacuum concentrator and redissolved in 10 µl 0.1% TFA for subsequent LC–MS/MS analyses.

LC–MS/MS

The nano-LC–MS/MS analysis was performed on a Q Exactive mass spectrometer coupled to an EASY-nLC 1000 system (Thermo Fisher Scientific, Bremen, Germany).

Shortly, 2.0 µg peptides, as measured with a Nanodrop 2000 (Thermo Fisher Scientific, Bremen, Germany) at a wavelength of 205 nm and extinction coefficient 31 mg/ml [16], per sample were injected. They were concentrated on a reversed-phase trap column and subsequently separated on a reversed-phase main column. A binary solvent gradient of 0.1% FA (solvent A) and 0.1% FA in acetonitrile (solvent B) was used. The ACN proportion was increased from 0 to 5% over 19 min, further to 30% at 180 min and to 100% at 200 min. A detailed description of the LC–MS/MS conditions can be found in the supporting information (see Additional file 1).

Data evaluation

The raw data was analyzed with MaxQuant against a UniProt fasta file of all human proteins (see Additional file 1) [17, 18]. Further analyses were performed with Perseus [19] and Cytoscape/ClueGO [20, 21].

The data processing and visualization in Perseus is partially based on a recently published protocol by the Perseus developers [22]. The intensities were log2-transformed in Perseus, and the entries were filtered for the labels “potential contaminant”, “reverse” and “only identified by site”. Standard intensities were subtracted from the corresponding sample intensities and the resulting intensities Z-score transformed (matrix access: columns), which returns the normalized values (see Additional file 1).

Statistics and generation of figures and lists

Statistical analyses were performed in Perseus. The samples were assigned to the groups UC patients and healthy controls, respectively, and the groups containing the separate values for each sample were compared to each other with a two-sample test. Proteins were annotated with gene ontology terms from the databases GOBP (biological process), GOMF (molecular function), GOCC (cellular component) [23], KEGG [24], GSEA [25], UniProt keywords [18], InterPro [26], Reactome [27], and PROSITE [28]. The lists and the article figures were prepared in Perseus and the Cytoscape app ClueGO (see Additional file 1). The WikiPathways database was used for the ClueGO analysis [29]. Pathways were mapped with HumanCyc (see Additional file 1) [30].

Results and discussion

Exploratory analysis

The proteomes of UC and healthy tissue differed strongly, as shown in Fig. 1. We present the 20 proteins with the highest abundance increases and decreases, respectively, in Table 2. The Pearson correlations of normalized intensities (see Additional file 1) were higher when samples of the same group were compared (i.e. UC to UC, or healthy control to healthy control) than when UC samples were compared to healthy controls (Fig. 1a). Also the hierarchical clustering of the sample correlations resulted in separate grouping of UC and healthy samples (Fig. 1a). In addition averaged profiles showed high Pearson correlations (> 0.7) for comparisons within the groups UC and healthy, respectively, and low correlations (< 0.1) for comparisons of UC to healthy (Fig. 1b). Several multi scatter plots were investigated with random samples from UC patients or healthy controls in each group, and all showed the same trend with high correlations internally in the UC and healthy sample groups. Upon unsupervised investigation by principal component analysis (PCA) (Fig. 1c) a strong separation of UC and healthy in component 1 which explains 39.9% of the variance was observed. To investigate possible biases we controlled for grouping by gender, and such grouping did not occur.

Fig. 1
figure 1

Exploratory analysis. a Hierarchical clustering of the correlation coefficients of 6829 protein intensities (proteins quantified in at least 70% of the samples) after normalization shows a clear separation between ulcerative colitis (UC) and healthy controls. The two replicates (labeled with repl) show the highest correlations. b Multi-scatter plot of the averaged profiles for UC2, 3, 4, 6, 7, 11, 13, 14, and 16 (UC a), UC1, 5, 8, 9, 10, 12, 15, and 17 (UC b), H3, 4, 5, 6, 7, 8, 10, and 11 (H a), and H1, 2, 9, 12, 13, 14, and 15 (H b). The sample assignments to the groups a and b, respectively, are random. Blue numbers indicate Pearson correlation values. The averaged profiles show high correlations within the groups, and low correlations between the groups. c Principal component analysis (PCA) separates UC strongly from healthy controls in component 1 (39.9%)

Table 2 20 proteins with highest abundance increase and decrease, respectively, in UC patients as compared to healthy controls

Comparison with previous proteomics studies on UC

The in total 8562 identified proteins provided the so far deepest view into the proteome of human colon mucosa and of UC. We report the abundance changes including p values for all quantified proteins (see Additional file 2: Excel file). 6818 of these proteins were quantified in at least 70% of the samples and were subject to enrichment analysis. Our data confirmed and extends previous findings, but also includes novel findings, such as the increased abundance of signal peptidase complex proteins. We searched PubMed for the terms “ulcerative colitis proteomics” and reviewed the titles and abstracts to select the relevant studies. Additionally we included studies previously known to the authors, totaling to 7 studies for the comparison [3,4,5,6,7,8,9]. The present study was largely in agreement with those previous studies about the direction of abundance changes of differently abundant proteins. However, the magnitudes of the changes differed.

Functional analysis

Hierarchical clustering of the 596 most differently abundant proteins (8.7% of all proteins quantified in > 70% of samples; s0 = 2, FDR = 0.01) revealed three major clusters (Fig. 2). The proteins which were higher abundant in UC divided into two clusters, a small cluster of 20 proteins with a more pronounced abundance change in a subgroup of samples, and a large cluster of 247 proteins. The lower abundant proteins formed one cluster of 313 proteins. In total 16 proteins were outside these three clusters.

Fig. 2
figure 2

Clustering of the 596 most differently abundant proteins between ulcerative colitis (UC) and healthy controls. a Hierarchical clustering reveals three main protein clusters. Low abundances are indicated in blue, high abundances in red. b Protein abundance profiles of the three main clusters. 1: minor cluster of 20 proteins (including calprotectin) with increased abundance in UC, with a subgroup of UC samples showing a more pronounced increase. 2: major cluster of 247 proteins with increased abundance in UC. 3: Cluster of 313 proteins with a decreased abundance in UC

The extraction of biological information from omics datasets is a major challenge. Enrichment analyses can overcome some of the difficulties [10]. Enrichment analyses annotate categorical terms (e.g. functions, localizations, pathways) to proteins and subsequently determine which terms are overrepresented among the differently abundant proteins compared to the entire dataset of quantified proteins. Knowledge about the involved pathways and functions can help to understand the disease, understand complications, and develop new treatment options.

Abundance changes found in tissue proteomics could result from differential regulation of the protein abundances in cells, but also from changes of the cell population (e.g. from the migration of immune cells into inflamed tissue in UC [11]) or the extracellular matrix. It seems plausible that each of these factors affects all measured abundances in such an experiment to varying degrees.

Lists of the enriched gene ontology terms among proteins which were found to be differently abundant in UC as compared to healthy tissue, and a list of the proteins which contributed to the functions discussed in the following are provided as Additional file 2: Excel file.

This article can neither cover all enriched terms we found, nor can it discuss the impact of each differently abundant protein on a function or the magnitude of its abundance change in detail. We therefore strongly encourage the reader to consult the above mentioned tables for further information on functions and proteins of interest.

Lower abundant in UC

321 proteins showed decreased abundances in UC as compared to healthy control samples. 313 of these grouped into one cluster (Fig. 2, cluster 3). Functional analysis revealed that metabolic pathways were overrepresented among the proteins with decreased abundance in UC. The enrichment of proteins with decreased abundances in our dataset affects short-chain and long-chain fatty acid, ketone body and amino acid (particularly tryptophan) metabolism as well as the tricarboxylic acid (TCA) cycle (see Additional file 1: SuppPathway1–4) and the electron transport chain (ETC; Fig. 3a).

Fig. 3
figure 3

Functional networks of differently abundant proteins in colon mucosa biopsies affected by ulcerative colitis compared to healthy controls. a Functional network of the 321 proteins with the strongest abundance decrease in ulcerative colitis compared to healthy tissue. Metabolic functions dominate this network, especially those located in the mitochondria. b Functional network of the 275 proteins with highest abundance increase in ulcerative colitis. Functions of the immune system characterize this network. Generated with ClueGO, Pathways: WikiPathways (updated: 10.03.2018), pV ≤ 0.01

The observed abundance decreases of butyrate utilizing proteins are in line with previous studies which show a decreased butyrate metabolism by UC tissue. Butyrate is a short-chain fatty acid (SCFA) and the main energy source of healthy colon mucosa [31]. It was previously hypothesized that ulcerative colitis is an energy-deficiency disease resulting from the failure to utilize butyrate [31]. Colonocytes convert a part of the butyrate to ketone bodies [32]. Our data showed that proteins related to this specific utilization of butyrate were lower abundant in UC. Several studies have investigated whether butyrate has a positive effect on inflamed colon tissue, however the results of these studies are conflicting [33].

The decreased protein abundances also affected the transport and metabolism of the long-chain fatty acids (LCFA), i.e. the long carbon chain analogs of SCFAs. LCFAs have an ambiguous role in ulcerative colitis. Linoleic acid increases the chances of developing ulcerative colitis [34]. Omega-3 fatty acids, such as docosahexaenoic acid (DHA), may reduce the chance of developing the disease [35]. Studies on the efficacy of omega-3 fatty acids in the treatment of ulcerative colitis show controversial results. Systematic reviews however conclude that large high-quality studies fail to show positive effects, and that there is no sufficient evidence for the efficiacy of omega-3 fatty acids in the treatment of UC [36, 37].

Mitochondrial enzymes involved in the metabolism of SCFA and LCFA as well as the tricarboxylic acid (TCA) and the electron transport chain (ETC) were found in our dataset to be low abundant in UC. These findings reflect a mitochondrial dysfunction which possibly contributes to the proposed energy deficiency [38].

Proteins related to nuclear receptors were of decreased abundance in UC. Peroxisome proliferator-activated receptor (PPAR)-inducible proteins were lower abundant in UC (Fig. 3). The changes in the LCFA transport and the PPAR pathway are interdependent. Activation of the intracellular PPAR receptors by LCFA demands the import of the fatty acids into the cell and the transport within the cell. One protein involved in the transport into the cell is CD36 which is low abundant in UC tissue. Fatty acid binding proteins (FABP) facilitate the transport within the cell [39]. Four FABPs (FABP1, FABP2, FABP4, and FABP5) are among the proteins with the lowest relative abundance in UC tissue compared to healthy tissue. A decreased transport of LCFA might in turn be partially causative for the decreased abundances of PPAR-inducible proteins, such as ACOX1, HMGCS2, FABP1, and PCK1 [40]. In accordance with a previous study [41], we found the vitamin D3 receptor NR1i1 to be lower abundant in UC tissue. However, we observed no general abundance changes of proteins regulated by this receptor. Previous studies found decreased abundances of hepatocyte nuclear factor 4 alpha (HNF4A) in UC and suggested a role in the disease pathophysiology [42]. Our data supports these findings.

All detected metallothioneins—MT1H, MT2A, and MT1F—were found to be lower abundant in UC tissue compared to healthy controls. Metallothioneins are small, cysteine-rich proteins, which bind heavy metal cations. Our findings confirmed several previous studies, which concluded that the decreased metallothionein abundances may result in an inefficient antioxidant response in the mucosa and in turn contributes to the IBD pathophysiology [43]. However, other studies present conflicting results and conclusions [44].

Histamine is the major mediator of mast cells and contributes to the immuno-inflammatory reaction in IBD. Previous studies showed increased histamine secretion and levels in UC affected tissues [45] and decreased histaminase (AOC1) activity [9]. The present study revealed that both major histamine degrading enzymes—Histaminase (AOC1) and histamine N-methyltransferase (HNMT)—were lower abundant in UC tissue compared to healthy tissue (not based on enrichment analysis). This finding constitutes a plausible causative explanation for some of the previously reported findings on histamine in UC.

Chemical compounds undergo phase I and phase II biotransformation reactions which make them more hydrophilic [46]. This allows their excretion with urine because the polar compounds are not reabsorbed in the kidneys [47]. Phase I reactions introduce or change functional groups by oxidation, reduction, or hydrolysis. In phase II reactions, enzymes attach hydrophilic endogenous compounds [46]. Our data showed that the abundances of a large proportion of both phase I and phase II enzymes are decreased in UC. A decrease of xenobiotic metabolism gene products in UC was shown earlier on mRNA level [48]. The affected phase II enzymes are involved in sulfonation, glucuronidation, and glutathione conjugation. Proteins related to both phase 1 (monoamine oxidase A, MAOA) and phase 2 metabolism (sulfotransferases) of catecholamines were found in our dataset to be less abundant in UC. This could contribute to previously found increases of norepinephrine in rectal UC mucosa [49].

Synthetic steroids are an important treatment option for ulcerative colitis. Our data showed impairments in the steroid metabolism in UC. This suggests a role of endogenous steroids in the pathophysiology of the disease. The hydroxysteroid dehydrogenases HSD11B2, HSD17B11, HSD17B2, and HSD17B7 were among the most down-regulated proteins in UC (HSD17B8 is downregulated to a lesser degree). The abundances of HSD17B4, HSD17B10, and HSD17B12 were not different in UC from healthy tissue. Decreased abundances of HSD11B2 [50] and HSD17B7 [3] have been reported previously. Our data furthermore showed that proteins related to the transport of bile acids are enriched among the lower abundant proteins, which supports findings of a previous study [41]. Pathway mapping shows abundance decreases of proteins involved in the neutral pathway of bile acids biosynthesis (Additional file 1: SuppPathway5). However, the implications of this finding are not clear, because bile acids are primarily synthesized in hepatocytes, whereas there is no evidence of bile acid synthesis in enterocytes [51].

The carbonic anhydrases CA1, CA2, CA4, and CA12 were found to be lower abundant in UC. CA3 was the only quantified but not significantly lower abundant carbonic anhydrase. A decreased abundance in UC was reported earlier for CA1 [52].

Fibrillar collagens are a subgroup of collagens and provide three-dimensional frameworks for tissues and organs [53]. The fibrillar collagens were enriched among the proteins that were of decreased abundance in UC. Proteins that are involved in collagen degradation, e.g. metalloproteinases, on the other hand were enriched among the proteins with increased abundance in UC (see below). This finding is in accordance with a previous study on the role of collagen degradation in IBD [54].

Higher abundant proteins in UC

As expected, many proteins with increased abundance in UC are involved in inflammatory and immune processes (Fig. 3b). Proteins related to neutrophils (e.g. NADPH oxidase complex proteins which generate superoxide, metalloproteinases) and B cells (e.g. V and C regions of immunoglobulins) were highly enriched. This could result from a tissue infiltration by these immune cells.

The higher abundant proteins divided into two main clusters under hierarchical clustering (Fig. 2, clusters 1 and 2). The UC patients with high cluster 1 protein abundances (UC1, UC2, UC5, UC8, UC15, UC17) grouped together (Fig. 2a, b). Cluster 1 contained proteins occurring in neutrophils, among these Protein S100A8 and Protein S100A9 which together form the calprotectin complex (see Additional file 1: Table 3). Several proteins (calprotectin, S100A12, lactotransferrin) of the minor cluster have been used to differentiate between IBD and non-IBD [55]. Interestingly, the average Geboes index in the patient cluster with high cluster 1 protein abundances is 11.0 (SD = 3.1), while it is 6.5 (SD = 3.2) for the other patients.

Our data showed that inducible nitric oxide synthase (NOS2) is higher abundant in UC tissue (not based on enrichment analysis). NOS2 produces nitric oxide (NO) which has various physiological roles as a messenger molecule. It functions most prominently as a vasodilator [56], but it is also involved in the immune system [57]. NO is directly toxic to pathogens, induces or suppresses apoptosis, and regulates the immune reaction [58]. Excess NO is cytotoxic and induces cell death. NOS2 is inducible by inflammation, infection and other stimuli. Increased NOS2 abundances in UC were shown earlier by immunostaining [8] and our results confirm these findings.

Proteins with functions in protein processing in the endoplasmic reticulum (ER) are overrepresented among the proteins which were more abundant in UC. Also unfolded protein response (UPR) and signal peptidase complex (SPC) proteins are related to protein processing, located in the ER, and enriched among proteins with increased abundances in UC. UPR has previously been identified as one factor in the pathophysiology of ulcerative colitis [59]. The enrichment of SPC proteins among proteins with increased abundances in UC is a novel finding from our data.

However, ER proteins were not generally higher abundant. For instance, many metabolic enzymes are located in the ER and enriched among the proteins with decreased abundances (e.g. UDP-glucuronosyltransferases, see “Lower abundant in UC” section).

The SPC is not inherently inflammatory. Nonetheless its proteins were among those with the highest abundance increases in UC. The SPC is located in the membrane of the endoplasmic reticulum and cleaves the signal peptide cotranslationally from nascent proteins [60]. The SPC subunits SPCS1, SPCS2, and SPCS3 and the SPC catalytic subunit SEC11C were more abundant in UC. They furthermore showed highly similar abundance profiles (Additional file 1: Figure 3). The SPC catalytic subunit SEC11 however does not follow this pattern and was similarly abundant in UC and healthy tissue.

Accumulation of unfolded and misfolded proteins causes stress to the endoplasmic reticulum. The UPR aims to remove these proteins. To achieve this, the cell stops the translation of further proteins, degrades misfolded proteins, and produces chaperones, which correct the protein folding. Our data showed strong enrichment of proteins related to UPR, response to ER stress, and the ER chaperone complex among the higher abundant proteins in UC. Specifically, several chaperones that can be induced by ATF6 alpha (HSP90B1, calreticulin [61], and HSPA5 [62]), which plays a central role in UPR, were enriched. Our findings support previous assumptions that the UPR may play a role in the pathophysiology of UC.

Foreseeable applications

The functional changes we observed in the present study allow for hypothesis generation for treatment approaches.

Alterations of metabolic pathways in ulcerative colitis imply that nutritional interventions directed at the respective metabolites could be effective in the treatment of the disease. For instance, decreased metallothionein abundances indicate that the detoxification of heavy metals and the utilization of polyvalent metal ions could be impaired. Considering the decreased detoxification capacities, reduced intake or chelation of toxic heavy metals might have beneficial effects. On the other hand, the supplementation of essential polyvalent metal ions could be advantageous. Furthermore, the decreased metabolic and transport capabilities related to butyrate contribute probably to the energy deprivation of colonocytes. Previous studies investigated the treatment of UC with butyrate and other SCFA. But the pathophysiological changes we observed related to the utilization of those compounds imply that supplementation with a later metabolite such as beta-hydroxybutyrate might be preferable. Beta-hydroxybutyrate is a ketone body that becomes systemically available after oral administration and has been used in the treatment of other pathophysiological conditions [63, 64].

The enrichment of proteins related to signaling pathways such as PPAR designates the respective pathways as potential drug targets.

Abundance changes of single proteins may have similar implications, especially if they are known to be drugable. Proteins with high abundance differences between UC patients and healthy controls and high corresponding − Log p values are of potential interest as biomarker candidates. Proteins with these characteristics are for instance SLC26A2, HMGCS2, and CD38 (Additional file 1: Figure 4; Additional file 2).

However, further testing, clinical studies, and validation will be necessary before any clinical application.

Conclusions

Our study presents new evidence and complements previous findings about changes of biological functions in UC. The major novel findings of this study were the increased abundances of SPC proteins and the presence of two distinguished clusters of higher abundant proteins, with the calprotectin complex proteins S100A8 and S100A9 in the minor cluster 1. This cluster of 20 proteins diverged from the abundance changes of the majority of proteins with increased abundances in UC that we observed in cluster 2.

In conclusion, abundances in UC tissue were increased compared to healthy controls for proteins related to the immune system and to protein processing in the ER, e.g. UPR and SPC proteins. Immune cell infiltration into the inflamed tissue contributes probably to the increased immune system protein abundances. The high abundance of UPR proteins indicates an ER stress response. Moreover, NOS2 which produces NO was higher abundant in UC tissue.

Lower abundant in UC were predominantly metabolic proteins. These metabolic proteins comprise mitochondrial enzymes for the metabolization of SCFA and LCFA as well as proteins of the TCA cycle and the ETC. These changes are probably contributing to the previously reported mitochondrial dysfunction and energy deficiency of colonocytes in UC. The abundances of LCFA transport proteins were decreased which may be partially causative for the decreased abundances of PPAR-inducible proteins. Decreased abundances of histamine degrading enzymes probably contribute to the increased histamine levels in UC tissue which were found in previous studies. Furthermore, abundances of proteins involved in phase I and phase II biotransformation were decreased, as well as the abundances of several metallothioneins, hydroxysteroid dehydrogenases, and carbonic anhydrases.

We provide a list of all 8562 identified proteins including the fold changes UC/H and p values as supporting information (see Additional file 2: Excel file).

Abbreviations

BCA:

bicinchoninic acid

DHA:

docosahexaenoic acid

ER:

endoplasmic reticulum

ETC:

electron transport chain

FA:

formic acid

FABP:

fatty acid binding protein

IBD:

inflammatory bowel disease

LCFA:

long-chain fatty acid

Lys-C:

endoproteinase Lys-C

NO:

nitric oxide

PCA:

principal component analysis

PPAR:

peroxisome proliferator-activated receptor

SCFA:

short-chain fatty acid

SDC:

sodium deoxycholate

SPC:

signal peptidase complex

TCA cycle:

tricarboxylic acid cycle

TEAB:

triethylammonium bicarbonate buffer

TMT:

tandem mass tag

UC:

ulcerative colitis

UPR:

unfolded protein response

References

  1. Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time: based on systematic review. Gastroenterology. 2012;142(1):46.e42–54.e42.

    Article  Google Scholar 

  2. Ungaro R, Mehandru S, Allen PB, Peyrin-Biroulet L, Colombel JF. Ulcerative colitis. Lancet. 2017;389(10080):1756–70.

    Article  Google Scholar 

  3. Schniers A, Anderssen E, Fenton Christopher G, Goll R, Pasing Y, Paulssen Ruth H, Florholmen J, Hansen T. The proteome of ulcerative colitis in colon biopsies from adults—optimized sample preparation and comparison with healthy controls. Proteomics Clin Appl. 2017;11(11–12):1700053.

    Article  Google Scholar 

  4. Bennike TB, Carlsen TG, Ellingsen T, Bonderup OK, Glerup H, Bøgsted M, Christiansen G, Birkelund S, Stensballe A, Andersen V. Neutrophil extracellular traps in ulcerative colitis: a proteome analysis of intestinal biopsies. Inflamm Bowel Dis. 2015;21(9):2052–67.

    Article  Google Scholar 

  5. Starr AE, Deeke SA, Ning Z, Chiang C-K, Zhang X, Mottawea W, Singleton R, Benchimol EI, Wen M, Mack DR, et al. Proteomic analysis of ascending colon biopsies from a paediatric inflammatory bowel disease inception cohort identifies protein biomarkers that differentiate Crohn’s disease from UC. Gut. 2016;66:1573–83.

    Article  Google Scholar 

  6. Poulsen NA, Andersen V, Møller JC, Møller HS, Jessen F, Purup S, Larsen LB. Comparative analysis of inflamed and non-inflamed colon biopsies reveals strong proteomic inflammation profile in patients with ulcerative colitis. BMC Gastroenterol. 2012;12(1):76.

    Article  CAS  Google Scholar 

  7. Deeke SA, Starr AE, Ning Z, Ahmadi S, Zhang X, Mayne J, Chiang C-K, Singleton R, Benchimol EI, Mack DR, et al. Mucosal–luminal interface proteomics reveals biomarkers of pediatric inflammatory bowel disease-associated colitis. Am J Gastroenterol. 2018;113(5):713–24.

    Article  CAS  Google Scholar 

  8. Leonard N, Bishop AE, Polak JM, Talbot IC. Expression of nitric oxide synthase in inflammatory bowel disease is not affected by corticosteroid treatment. J Clin Pathol. 1998;51(10):750–3.

    Article  CAS  Google Scholar 

  9. Mennigen R, Kusche J, Streffer C, Krakamp B. Diamine oxidase activities in the large bowel mucosa of ulcerative colitis patients. Agents Actions. 1990;30(1–2):264–6.

    Article  CAS  Google Scholar 

  10. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102(43):15545–50.

    Article  CAS  Google Scholar 

  11. Taman H, Fenton CG, Hensel IV, Anderssen E, Florholmen J, Paulssen RH. Transcriptomic landscape of treatment—Naïve ulcerative colitis. J Crohn’s Colitis. 2018;12(3):327–36.

    Article  Google Scholar 

  12. Maier T, Güell M, Serrano L. Correlation of mRNA and protein in complex biological samples. FEBS Lett. 2009;583(24):3966–73.

    Article  CAS  Google Scholar 

  13. Stange EF, Travis SP, Vermeire S, Reinisch W, Geboes K, Barakauskiene A, Feakins R, Flejou JF, Herfarth H, Hommes DW, et al. European evidence-based Consensus on the diagnosis and management of ulcerative colitis: definitions and diagnosis. J Crohns Colitis. 2008;2(1):1–23.

    Article  CAS  Google Scholar 

  14. Sutherland LR, Martin F, Greer S, Robinson M, Greenberger N, Saibil F, Martin T, Sparr J, Prokipchuk E, Borgen L. 5-Aminosalicylic acid enema in the treatment of distal ulcerative colitis, proctosigmoiditis, and proctitis. Gastroenterology. 1987;92(6):1894–8.

    Article  CAS  Google Scholar 

  15. Olsen T, Goll R, Cui G, Husebekk A, Vonen B, Birketvedt G, Florholmen J. Tissue levels of tumor necrosis factor-alpha correlates with grade of inflammation in untreated ulcerative colitis. Scand J Gastroenterol. 2007;42(11):1312–20.

    Article  CAS  Google Scholar 

  16. Scopes RK. Measurement of protein by spectrophotometry at 205 nm. Anal Biochem. 1974;59(1):277–82.

    Article  CAS  Google Scholar 

  17. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26(12):1367–72.

    Article  CAS  Google Scholar 

  18. The UniProt Consortium. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 2017;45(D1):D158–69.

    Article  Google Scholar 

  19. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, Mann M, Cox J. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13(9):731–40.

    Article  CAS  Google Scholar 

  20. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  CAS  Google Scholar 

  21. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pages F, Trajanoski Z, Galon J. ClueGO: a Cytoscape plug-into decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics (Oxford, England). 2009;25(8):1091–3.

    Article  CAS  Google Scholar 

  22. Tyanova S, Cox J. Perseus: a bioinformatics platform for integrative analysis of proteomics data in cancer research. Methods Mol Biol (Clifton, NJ). 2018;1711:133–48.

    Article  CAS  Google Scholar 

  23. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000;25(1):25–9.

    Article  CAS  Google Scholar 

  24. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.

    Article  CAS  Google Scholar 

  25. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci. 2005;102(43):15545.

    Article  CAS  Google Scholar 

  26. Finn RD, Attwood TK, Babbitt PC, Bateman A, Bork P, Bridge AJ, Chang H-Y, Dosztányi Z, El-Gebali S, Fraser M, et al. InterPro in 2017-beyond protein family and domain annotations. Nucleic Acids Res. 2017;45(D1):D190–9.

    Article  CAS  Google Scholar 

  27. Fabregat A, Jupe S, Matthews L, Sidiropoulos K, Gillespie M, Garapati P, Haw R, Jassal B, Korninger F, May B, et al. The Reactome pathway knowledgebase. Nucleic Acids Res. 2018;46(D1):D649–55.

    Article  CAS  Google Scholar 

  28. Sigrist CJA, de Castro E, Cerutti L, Cuche BA, Hulo N, Bridge A, Bougueleret L, Xenarios I. New and continuing developments at PROSITE. Nucleic Acids Res. 2013;41(D1):D344–7.

    Article  CAS  Google Scholar 

  29. Slenter DN, Kutmon M, Hanspers K, Riutta A, Windsor J, Nunes N, Mélius J, Cirillo E, Coort SL, Digles D, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018;46(D1):D661–7.

    Article  CAS  Google Scholar 

  30. Romero P, Wagg J, Green ML, Kaiser D, Krummenacker M, Karp PDJGB. Computational prediction of human metabolic pathways from the complete human genome. Genome Biol. 2004;6(1):R2.

    Article  Google Scholar 

  31. Roediger WEW. The colonic epithelium in ulcerative colitis: an energy-deficiency disease? Lancet. 1980;316(8197):712–5.

    Article  Google Scholar 

  32. Roediger WE. Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut. 1980;21(9):793–8.

    Article  CAS  Google Scholar 

  33. Vinolo MAR, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011;3(10):858–76.

    Article  CAS  Google Scholar 

  34. Tjonneland A, Overvad K, Bergmann MM, Nagel G, Linseisen J, Hallmans G, Palmqvist R, Sjodin H, Hagglund G, Berglund G, et al. Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case-control study within a European prospective cohort study. Gut. 2009;58(12):1606–11.

    Article  CAS  Google Scholar 

  35. John S, Luben R, Shrestha SS, Welch A, Khaw K-T, Hart AR. Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study. Eur J Gastroenterol Hepatol. 2010;22(5):602–6.

    Article  CAS  Google Scholar 

  36. Ungaro F, Rubbino F, Danese S, D’Alessio S. Actors and factors in the resolution of intestinal inflammation: lipid mediators as a new approach to therapy in inflammatory bowel diseases. Front Immunol. 2017;8:1331.

    Article  Google Scholar 

  37. Turner D, Shah PS, Steinhart AH, Zlotkin S, Griffiths AM. Maintenance of remission in inflammatory bowel disease using omega-3 fatty acids (fish oil): a systematic review and meta-analyses. Inflamm Bowel Dis. 2011;17(1):336–45.

    Article  Google Scholar 

  38. Novak EA, Mollen KP. Mitochondrial dysfunction in inflammatory bowel disease. Front Cell Dev Biol. 2015;3:62.

    Article  Google Scholar 

  39. Georgiadi A, Kersten S. Mechanisms of gene regulation by fatty acids. Adv Nutr. 2012;3(2):127–34.

    Article  CAS  Google Scholar 

  40. Rakhshandehroo M, Hooiveld G, Müller M, Kersten S. Comparative analysis of gene regulation by the transcription factor PPARα between mouse and human. PLoS ONE. 2009;4(8):e6796.

    Article  Google Scholar 

  41. Jahnel J, Fickert P, Hauer AC, Hogenauer C, Avian A, Trauner M. Inflammatory bowel disease alters intestinal bile acid transporter expression. Drug Metab Dispos Biol Fate Chem. 2014;42(9):1423–31.

    Article  Google Scholar 

  42. Ahn SH, Shah YM, Inoue J, Morimura K, Kim I, Yim S, Lambert G, Kurotani R, Nagashima K, Gonzalez FJ, et al. Hepatocyte nuclear factor 4alpha in the intestinal epithelial cells protects against inflammatory bowel disease. Inflamm Bowel Dis. 2008;14(7):908–20.

    Article  Google Scholar 

  43. Kruidenier L, Kuiper I, van Duijn W, Mieremet-Ooms MAC, van Hogezand RA, Lamers CBHW, Verspaget HW. Imbalanced secondary mucosal antioxidant response in inflammatory bowel disease. J Pathol. 2003;201:17–27.

    Article  CAS  Google Scholar 

  44. Rahman MT, Karim MM. Metallothionein: a potential link in the regulation of zinc in nutritional immunity. Biol Trace Elem Res. 2018;182(1):1–13.

    Article  CAS  Google Scholar 

  45. Raithel M, Matek M, Baenkler HW, Jorde W, Hahn EG. Mucosal histamine content and histamine secretion in Crohn’s disease, ulcerative colitis and allergic enteropathy. Int Arch Allergy Immunol. 1995;108(2):127–33.

    Article  CAS  Google Scholar 

  46. Omiecinski CJ, Vanden Heuvel JP, Perdew GH, Peters JM. Xenobiotic metabolism, disposition, and regulation by receptors: from biochemical phenomenon to predictors of major toxicities. Toxicol Sci. 2011;120(1):S49–75.

    Article  CAS  Google Scholar 

  47. Bardal SK, Waechter JE, Martin DS. Chapter 2—Pharmacokinetics. In: Bardal SK, Waechter JE, Martin DS, editors. Applied pharmacology. Philadelphia: Content Repository Only; 2011. p. 17–34.

    Google Scholar 

  48. Langmann T, Moehle C, Mauerer R, Scharl M, Liebisch G, Zahn A, Stremmel W, Schmitz G. Loss of detoxification in inflammatory bowel disease: dysregulation of pregnane X receptor target genes. Gastroenterology. 2004;127(1):26–40.

    Article  CAS  Google Scholar 

  49. Penttila O, Kyosola K, Klinge E, Ahonen A, Tallqvist G. Studies of rectal mucosal catecholamines in ulcerative colitis. Ann Clin Res. 1975;7(1):32–6.

    CAS  PubMed  Google Scholar 

  50. Takahashi K-I, Fukushima K, Sasano H, Sasaki I, Matsuno S, Krozowski ZS, Nagura H. Type II 11β-hydroxysteroid dehydrogenase expression in human colonic epithelial cells of inflammatory bowel disease. Dig Dis Sci. 1999;44(12):2516–22.

    Article  CAS  Google Scholar 

  51. Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res. 2015;56(6):1085–99.

    Article  CAS  Google Scholar 

  52. Giovanni RF, Renzo L, Giuseppe C, Adriana F, Sambuy MY. Carbonic anhydrase I reduction in colonic mucosa of patients with active ulcerative colitis. Dig Dis Sci. 1998;43(9):2086–92.

    Article  Google Scholar 

  53. Bella J, Hulmes DJS. Fibrillar collagens. In: Parry DAD, Squire JM, editors. Fibrous proteins: structures and mechanisms. Cham: Springer; 2017. p. 457–90.

    Chapter  Google Scholar 

  54. Koelink PJ, Overbeek SA, Braber S, Morgan ME, Henricks PAJ, Roda MA, Verspaget HW, Wolfkamp SC, te Velde AA, Jones CW, et al. Collagen degradation and neutrophilic infiltration: a vicious circle in inflammatory bowel disease. Gut. 2014;63(4):578–87.

    Article  CAS  Google Scholar 

  55. Iskandar HN, Ciorba MA. Biomarkers in inflammatory bowel disease: current practices and recent advances. Transl Res. 2012;159(4):313–25.

    Article  CAS  Google Scholar 

  56. Chen K, Pittman RN, Popel AS. Nitric oxide in the vasculature: where does it come from and where does it go? A quantitative perspective. Antioxiod Redox Signal. 2008;10(7):1185–98.

    Article  CAS  Google Scholar 

  57. Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001;2:907.

    Article  CAS  Google Scholar 

  58. Coleman JW. Nitric oxide in immunity and inflammation. Int Immunopharmacol. 2001;1(8):1397–406.

    Article  CAS  Google Scholar 

  59. Cao SS. Epithelial ER stress in Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis. 2016;22(4):984–93.

    Article  Google Scholar 

  60. Sakaguchi M. Eukaryotic protein secretion. Curr Opin Biotechnol. 1997;8(5):595–601.

    Article  CAS  Google Scholar 

  61. Fox RM, Andrew DJ. Transcriptional regulation of secretory capacity by bZip transcription factors. Front Biol. 2015;10(1):28–51.

    Article  CAS  Google Scholar 

  62. Thuerauf DJ, Morrison L, Glembotski CC. Opposing roles for ATF6α and ATF6β in endoplasmic reticulum stress response gene induction. J Biol Chem. 2004;279(20):21078–84.

    Article  CAS  Google Scholar 

  63. Tieu K, Perier C, Caspersen C, Teismann P, Wu D-C, Yan S-D, Naini A, Vila M, Jackson-Lewis V, Ramasamy R, et al. D-β-Hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson disease. J Clin Investig. 2003;112(6):892–901.

    Article  CAS  Google Scholar 

  64. Plecko B, Stoeckler-Ipsiroglu S, Schober E, Harrer G, Mlynarik V, Gruber S, Moser E, Moeslinger D, Silgoner H, Ipsiroglu O. Oral beta-hydroxybutyrate supplementation in two patients with hyperinsulinemic hypoglycemia: monitoring of beta-hydroxybutyrate levels in blood and cerebrospinal fluid, and in the brain by in vivo magnetic resonance spectroscopy. Pediatr Res. 2002;52(2):301–6.

    CAS  PubMed  Google Scholar 

  65. Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006;55(6):749–53.

    Article  CAS  Google Scholar 

Download references

Authors’ contributions

AS, YP and TH planned the experiments. AS and TH analyzed and interpreted the proteome data with input from YP. SWS performed the histological examination to determine the Geboes scores. JF and RG recruited patients, provided colon mucosa biopsies, and reported the baseline characteristics. All authors contributed with writing to their parts as well as approved the final version of the manuscript.

Acknowledgements

We thank Jack-Ansgar Bruun for fruitful discussions about mass spectrometric analyses. We thank Mary Ann Lauger and Hal Roach for proofreading of the manuscript.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Consent for publication

Not applicable.

Ethics approval and consent to participate

The Regional Committee of Medical Ethics of North Norway and the Norwegian Social Science Data Services approved the study and the storage of biological material under the number REK NORD 2012/1349. In addition, all enrolled subjects have signed an informed and written consent form.

Funding

This study was funded by Helse Nord RHF and UiT The Arctic University of Norway.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Terkel Hansen.

Additional files

Additional file 1.

Detailed method descriptions, additional figures and tables, mapping of metabolic pathways.

Additional file 2.

Complete list of identified proteins including UC/Healthy ratios and p-values, lists of gene ontology term enrichments, enriched functions discussed in the article and their proteins.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schniers, A., Goll, R., Pasing, Y. et al. Ulcerative colitis: functional analysis of the in-depth proteome. Clin Proteom 16, 4 (2019). https://doi.org/10.1186/s12014-019-9224-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12014-019-9224-6

Keywords