Abstract
Background & Aims
Chronic inflammatory illnesses are debilitating and recurrent conditions associated with significant comorbidities, including an increased risk of developing cancer. Extensive tissue remodeling is a hallmark of such illnesses, and is both a consequence and a mediator of disease progression. Despite previous characterization of epithelial and stromal remodeling during inflammatory bowel disease, a complete understanding of its impact on disease progression still is lacking.
Methods
A comprehensive proteomic pipeline using data-independent acquisition was applied to decellularized colon samples from the Muc2 knockout (Muc2KO) mouse model of colitis for an in-depth characterization of extracellular matrix remodeling. Unique proteomic profiles of the matrisomal landscape were extracted from prepathologic and overt colitis. Integration of proteomics and transcriptomics data sets extracted from the same murine model produced network maps describing the orchestrating role of matrisomal proteins in tissue remodeling during the progression of colitis.
Results
The in-depth proteomic workflow used here allowed the addition of 34 proteins to the known colon matrisomal signature. Protein signatures of prepathologic and pathologic colitic states were extracted, differentiating the 2 states by expression of small leucine-rich proteoglycans. We outlined the role of this class and other matrisomal proteins in tissue remodeling during colitis, as well as the potential for coordinated regulation of cell types by matrisomal ligands.
Conclusions
Our work highlights a central role for matrisomal proteins in tissue remodeling during colitis and defines orchestrating nodes that can be exploited in the selection of therapeutic targets.
Keywords: Inflammatory Bowel Disease, DIA Proteomics, Cell-Matrix Interactions, Proteoglycans
Graphical abstract
Summary.
A novel proteomics methodology characterized extracellular matrix remodeling in colitis. Known changes in cell populations accompanying colitic disease were orchestrated by a subset of matrisomal proteins, defining interaction networks that can be exploited in disease prevention strategies.
Chronic inflammation generates compositional and adaptive changes at the cellular and molecular levels. Resulting tissue remodeling ultimately impairs epithelial function, leading to diverse disease states such as chronic liver disease, gastritis, or colitis, which are debilitating and life-altering conditions associated with an increased risk of developing cancer.1 Characterization of these chronic illnesses largely has been cell-centric, describing epithelial and stromal cell adaptations while viewing the extracellular matrix (ECM) and its associated proteins—the matrisome—as a supporting scaffold.
The matrisome comprises core ECM constituents (collagens, glycoproteins, proteoglycans, and polysaccharides), as well as associated regulators and secreted factors.2 It is produced by resident cell types that are found in unique combinations in different tissues.3 Evidence for an instructive capacity of the ECM originally came from studies in which separation of embryonic epithelium and mesenchyme before heterotypic recombination revealed that the mesenchyme was responsible for tissue differentiation.4 Subsequently, many studies have established that the matrisome is dynamically maintained and associated with varied cellular responses, including cell growth and differentiation through direct ligand-receptor interactions.5 Furthermore, it dictates biomechanical properties and is able to sequester and regulate the availability of cytokines and growth factors.6 More recently, embryonic precursor cells seeded in complete ECM scaffolds derived from decellularized tissues were found to differentiate into epithelia of the same tissue type as that from which the scaffold was derived.7, 8, 9
The relationship whereby the matrisome dynamically regulates the biology of the cells from which it arises has been termed dynamic reciprocity.10 During chronic inflammation, excessive degradation and inappropriate deposition of the ECM by proteases prevents restoration of normal matrix composition. This process, termed fibrosis, is linked to increased cancer risk..11,12 The ECM has been shown to influence all the hallmarks that typify development of cancer, and its remodeling is considered central to cancer initiation as a result of chronic inflammation.13,14, 15
Ulcerative colitis (UC) is a chronic inflammatory disorder prone to remission and relapse, with a complex and not yet entirely understood etiology involving both genetic and environmental factors.16 UC is defined by colonic mucosal inflammation associated with reduced quality and quantity of the mucins that make up the intestinal mucus layer, impaired epithelial barrier function, and engagement with both innate and adaptive immune systems.17,18 UC patients are at a greater risk of developing colitis-associated colorectal cancer, which increases with the duration and severity of disease, reaching 18% by 30 years.19,20
The complexity of tissue response and disease escalation in UC, as well as technological barriers in quantifying protein changes in fractions with low solubility/low abundance, have meant that the full repertoire of matrisomal changes as well as how and when these orchestrate cellular adaptation and recruitment in UC have not been appreciated. Recently, label-free mass spectrometry–based proteomic strategies based on comprehensive data-independent acquisition (DIA)21, 22, 23 have been used to identify proteins in ECM-enriched cancer tissue samples and provided deep, accurate, and reproducible quantification.24 Here, we apply this unbiased approach to a murine model of colitis to characterize alterations in matrisomal protein expression. Network analyses define how ECM changes converge on key signaling nodes in both prepathologic and pathologic states to identify vulnerabilities for therapeutic and preventive interventions.
Results
Experimental Approach to Investigate ECM Remodeling in Colitis
Germline genetic knockout of the Muc2 gene in mice has been described previously as causing colitis that arises from impaired barrier function.25,26 This model was assessed phenotypically and mice with homozygous deletion of Mucin 2 (Muc2hom) were confirmed to show stochastic onset of colitis (Figure 1A and B). Staining with both Alcian blue and periodic acid–Schiff showed a reduction in expression of both acid and neutral mucins, and the remaining staining showed that goblet cells were retained (Figure 1A). This mouse model faithfully recapitulated key features of human UC including crypt enlargement and hyperplasia, immune infiltration of the mucosa, and the presence of abscesses (Figure 1B), as well as preneoplastic features such as dysplasia and early invasive epithelial foci (Figure 1C). In mature Muc2hom mice (age, 5–8 mo), these phenotypes were present regionally in both the proximal and distal colon, whereas the middle colon remained histologically normal (Figure 1D and E).
Figure 1.
Characterization of the Muc2KOmouse model of colitis reveals spatial differences in pathology. (A) Immunohistochemistry images for Muc2 (top) and Alcian blue (AB)/periodic acid–Schiff (PAS) (bottom), showing Muc2 deletion and hindered mucin production in Muc2hom compared with Muc2wt and Muc2het mouse colon. (B) Bar plots showing average crypt length (left), as well as the number of immune infiltrates (middle) and abscesses (right) per millimeter in the Muc2hom compared with Muc2het colon (N = 5 Muc2hom mice; median survival, 5.3 mo; N = 3 Muc2het mice; median survival, 4.5 mo). Statistical significance was determined using the Mann–Whitney test, ∗P < .04. (C) Bar plots showing presence of preneoplastic hallmarks in the Muc2hom colon. Dysplasia (left) and invasive length (right) are quantified as the proportion of respective pathology over the colon length, with E-cadherin+ glands crossing the muscularis mucosae considered as invasive. (D) Representative H&E images showing the spatial aspect of pathology along the mature Muc2hom mouse colon. Proximal and distal regions are commonly inflamed, whereas the middle colon remains histologically normal. (E) Mature Muc2hom colon drawing and histograms representing the spatial heterogeneity seen in crypt length, number of immune infiltrates, and abscesses along the length of the colon (N = 5 Muc2hom mice used, normalization to N = 3 Muc2het mice). Quantification was performed over 12 colonic regions as shown on the y-axis; error bars = 95% CI).
Samples for proteomic analysis were collected from 5 mature Muc2hom mice aged until they presented with diarrhea, a symptom of active colitis, and culled when this was accompanied by other symptoms such as signs of pain (hunch, piloerection) and/or development of an anal prolapse. Tissue from 5 age-matched heterozygous (Muc2het) mice were used concurrently as controls (Figure 2A). The colon tissue was taken from proximal, actively inflamed regions and from middle uninvolved regions (Figure 2B). Samples were processed for enrichment of ECM proteins by sequential fractionation that was validated at each stage by Western blot (Figure 3A). The final protein fraction was solubilized, digested, and subjected to mass spectrometry (MS) analysis as described previously24 (Figure 3B). A spectral library was built directly from DIA MS/MS spectra and used to identify and quantify all proteins across all conditions. In total, 2328 protein groups were identified and quantified, with at least 2 unique peptides and a 1% false discovery rate. A clustering analysis based on all quantified protein groups led to removal of 2 outlier samples that clustered apart from their own sample group (Figure 3C). For the remaining samples, correlation coefficients for protein expression within the sample group was greater than 85% (Figure 3D). Principal component analysis revealed genotype and location along the colon as the main factors contributing to variance between sample groups (Figure 3E). Of the 2328 proteins identified, 953 (∼41% of the total protein fraction) were annotated as extracellular using the Gene Ontology term Cellular Component (Figure 1A). Within this subset, 125 proteins were identified as components or associated proteins of the colon matrisome (MatrisomeDB2.0),27 accounting for 70% of all annotated proteins in this data set. In addition, 37 identified proteins were part of the wider, pan-organ matrisome database, which now can be added to the known colon matrisomal signature (Supplementary Table 4).
Figure 2.
Timeline and sample location for ECM proteomics. (A) Timeline showing age of mice used for histopathologic analysis and ECM proteomics. (B) Scheme representing the location of samples used for ECM proteomics and associated level of pathology.
Figure 3.
Experimental approach to investigate ECM remodeling in colitis. (A) Experimental outline for label-free MS on decellularized fractions of Muc2KO mouse colon. (B) Western blot showing sequential purification of the extracellular tissue fraction. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used to probe for removal of cytoplasmic proteins, Hmga1 of nuclear proteins, β1-integrin of membrane proteins, and actin of cytoskeletal proteins. Enrichment for ECM is seen with appearance of a band for collagen 1. (C) Heatmap displaying identified and quantified protein groups for all samples, showing 2 out layers outlined in black that were removed from the analysis. (D) Box plots representing the distribution of Pearson correlation coefficients for samples in each condition. (E) Principal component analysis using the abundance of all quantified proteins between sample groups, where PC1 (x-axis) separates samples based on genotype and PC2 (y-axis) based on location along the colon. (F) Stacked bar plots showing the number of genes present in each extended cell type signatures, coming from each of 3 colon single-cell RNA sequencing published data sets (see details in Methods).
Proteomic Analysis Reflects Disease State
Proteins with expression altered significantly in colitis (defined as q-value < 0.05), identified from the comparison of proximal colonic regions of Muc2hom inflamed and Muc2het control mice, were assessed for alterations in biological processes using the molecular interaction database ConsensusPathDB.28 The analysis identified positive enrichment for ECM organization as well as different immune-related functions (Figure 1B and C), confirming ,the resulting data set being informative of ECM remodeling during colitis.
The analysis focused on proteins with low solubility, which, as reported previously, include cellular remnants such as keratins and desmosomal proteins that relate to overall tissue and cellular identity.29 To evaluate how protein alterations reflect cell population level changes accompanying inflammatory disease, differentially expressed proteins (defined as q-value < 0.05) from the comparison of Muc2hom and Muc2het proximal colons were assigned to specific cell types, using previously described signatures30, 31, 32 (see Methods and Figure 3F). This analysis identified enrichment for immune cell types and underrepresentation of epithelial secretory cells, notably goblet cells, reflecting well-known features of colitis (Figure 1D and E). These findings indicated that proteomic analysis of the extracellular protein fraction captured the complexity of tissue changes associated with disease state at the level of both biological processes and cell type identity.
Deriving a Colitis Signature
To derive a robust colitis signature, we selected the top 200 proteins with the highest significance for differential expression in the proximal colon of Muc2hom vs location-matched Muc2het samples (Supplementary Table 3). Many up-regulated proteins were associated with specific immune cell types (e.g., eosinophils peroxidase, myeloperoxydase or mast cell protease 1) or immune function, such as Immunoglobulin heavy constant alpha (Figure 1F). Included in this signature was also the down-regulation of small leucine-rich proteoglycans (SLRPs) asporin and osteoglycin, both present in the subset of proteins presented in Figure 1F, as well as decorin (Dcn), present in the extended inflammation signature Supplementary Table 5. SLRPs are known to bind and alter the properties of collagens.33 At the level of protein groups, proteins classified as proteoglycans showed the most dramatic change in expression, with the majority being down-regulated with inflammation (Figure 1G).
Midcolon in Mature Muc2hom Mice Displays a Prepathologic State
Comparison of the midcolonic regions from Muc2hom and Muc2het mice identified 1712 differentially expressed proteins (Figure 5A). Of these, 721 were also differentially expressed in the proximal colonic region of Muc2hom mice that showed signs of active colitis (Figure 5A). The similarity between proximal inflamed and middle histologically normal colon samples was apparent in the principal component analysis, where those samples clustered together (Figure 3E).
Figure 5.
Midcolon regions in mature Muc2KOmice are in a prepathologic state. (A) Venn diagram showing overlap in proteins up-downregulated and down-regulated (q-value < 0.05) in proximal and middle mature Muc2hom mouse colon (inflamed and histologically normal, respectively, N = 4 samples per condition) compared with location-matched Muc2het control samples (N = 5 for each location). Proteins sharing the same direction of change in both comparisons are circled in grey (see Supplementary Table 5), proteins showing the opposite direction of change are circled in black (see Supplementary Table 5). Fill color from blue to red: lower to higher number of proteins in each subset of the Venn diagram. (B) Bar plot showing a subset of proteins extracted from the shared signature (q-value < 0.01, log2 fold change >1 or <-1). (C and D) Highlighted proteins are shown. (C) Bar plots showing normalized protein abundance for sulfotransferase family 1A member 1 (Sult1a1), down-regulated in both proximal and middle mature Muc2hom compared with location-matched Muc2het control samples. (D) Bar plots representing normalized protein abundance for gremlin (Grem1), showing a similar pattern to Sult1a1, with a stronger sequential up-regulation from mid- to proximal Muc2hom mouse colon. (E) Representative immunofluorescence images of Grem1 (in red) in Muc2het, midcolon Muc2hom, and proximal Muc2hom tissue. 4′,6-diamidino-2-phenylindole (DAPI) marks nuclei, and E-cadherin marks colonic epithelial cells. Bottom: Magnification of the white boxes in the top panel. (F) Bar plot showing a subset of proteins showing opposing signatures in middle and proximal Muc2homcolon samples(q-value < 0.05, log2 fold change >1 or <-1). (G and H) Highlighted proteins are shown. (G) Bar plots showing normalized protein abundance for chemokine (C-C motif) ligand 6 (Ccl6), down-regulated in Muc2hom midcolon and up-regulated in Muc2hom proximal colon compared with Muc2het control samples. (H) Normalized protein abundance for proline and arginine-rich end leucine-rich repeat protein (Prelp), up-regulated in Muc2hom midcolon and down-regulated in Muc2hom proximal colon compared with Muc2het control samples.
Comparing differentially expressed proteins in proximal and middle colon samples from Muc2hom (compared with location-matched Muc2het samples) revealed a linear relationship with a positive correlation, showing that shared alterations also follow the same direction of change (Figure 6A). We hypothesized that the substantial overlap in the number of proteins sharing the same direction of change might be explained by 2 opposing interpretations. First, it may reflect stable adaptive changes accompanying loss of Muc2 expression. Alternatively, it could indicate an early prepathologic stage in the development of inflammatory disease. To discriminate between these possibilities, a cohort of Muc2hom mice were maintained with colitis symptoms (median survival, 12 mo; range, 11.7–12.6 mo) and analyzed for inflammation along the length of the colon (Figure 2). These older animals showed an overall increase in pathology-related metrics, as well as a trend toward pancolitis (Figure 6B and C). This observation supported that the middle region of the colon in mature Muc2hom mice constituted a prepathologic state.
Figure 6.
Muc2KOmice evolve from discrete to pancolitis with age. (A) Dotplot showing log2 fold changes for each protein expressed differentially in Muc2hom midcolon (y-axis) and proximal (x-axis) samples compared with location-matched Muc2het colon sample (q-value < 0.05). (B) Survival analysis showing heterogeneity in Muc2hom mice survival, from 100 days up to 1 year old (study end point). (C) Bar plot showing crypt height for young (graybars; median age, 1.8 mo) and old (pink bars; median age, 12.4 mo) Muc2hom mice, normalized to age-matched Muc2het controls (NFig. 4 = 3 mice for each condition). (D) Representative H&E pictures of the whole swiss rolled colon (top) and specific colonic regions in Muc2het, (left) young Muc2hom(middle) and old Muc2hom mice (right), showing development of pathology in the proximal colon in young Muc2hom mice, which extends to the whole colon length in old Muc2hom mice. Boxes indicate magnified regions.
A shared representative signature was derived from proteins expressed differentially in the comparison of inflamed and prepathologic Muc2hom vs location-matched Muc2het samples and that shared the same direction of change (Figure 2A and B and Supplementary Table 2). Sulfotransferase family 1A member 1 (Sult1a1), a protein involved in small-molecule metabolism and previously found to be down-regulated in DSS colitis,30 was down-regulated in both prepathologic and inflamed states (Figure 2B and C). In contrast, Gremlin 1 (Grem1) expression increased incrementally in the respective states (Figure 2B and D). Notably, immunohistochemistry analysis showed that although Grem1 expression was restricted to the basement membrane in the prepathologic state, it extended to the mucosa in inflamed tissues (Figure 2E).
Because the midcolonic mucosa remained histologically normal in 5- to 8-month-old Muc2hom mice on initial presentation, we speculated that the observed molecular changes may illustrate an adaptation of the epithelium to inflammatory signals. Proteins were identified that were differentially expressed and changed in the opposite direction when comparing mid- and proximal inflamed regions of Muc2hom mice to their respective control regions in Muc2het samples (Figure 2A and F and Supplementary Table 2). Reciprocating patterns of expression were identified for ECM proteins including collagens (Col15a1, Col6a5, Col6a6) and SLRP proteoglycans such as proline and arginine-rich end leucine-rich repeat protein (Prelp) and Decorin (Dcn), as well as proteins implicated in mitochondrial function (Voltage-dependent anion-selective channel 1 and 3 or NADH-ubiquinone oxidoreductase 75 kDa subunit,) that all were up-regulated in midcolon and down-regulated in proximal inflamed colon (Figure 2F and H). Intriguingly, also showing this pattern was Vascular Adhesion Protein 1 (Vap-1), also known as Aoc3, a protein known to exist in tissue-bound and soluble forms and detected primarily in endothelial cells, adipocytes or serum, respectively. The tissue-bound form has been associated with tissue differentiation and ECM deposition whereas the soluble form has been shown to be proinflammatory and implicated in vascular diseases through involvement in leukocyte recruitment.31 Showing the opposing pattern (ie, down-regulated in midcolon and up-regulated in inflamed proximal) was chemokine ligand 6 (Figure 2F and G). Increased expression of this chemoattractant chemokine is consistent with the increased immune cell recruitment observed in inflammation.
Differential Expression of Proteoglycans Distinguishes Pathologic States
Expression of 6 SLRPs in mid- and proximal inflamed regions of mature Muc2hom mice was assessed relative to their respective control regions in Muc2het samples. These canonical SLRPs33 play diverse roles in collagen deposition, bind to receptor tyrosine kinases, innate immune receptors, and modulate the transforming growth factor-β (TGF-β) signaling pathway.34
All identified SLRPs, in particular Prelp and Dcn, were down-regulated with inflammation whilst up-regulated in midcolon, illustrating a clear distinction in differential proteoglycan expression associated with early (prepathologic) and advanced pathologic states (Figure 7A and B). The Bader Laboratory CellCellInteraction database35 was used to explore the consequences of altered SLRP expression by identifying differentially expressed interactors in both states. Dcn and biglycan (Bgn) presented with the highest number of known interactors (Figure 7C). Pathway analysis based on differentially expressed SLRP interactors (see Methods) showed a strong enrichment for inflammation-related pathways such as Inflammatory Response and Allograft Rejection in the inflamed proximal but not the midcolon of Muc2hom mice (Figure 7D). Interestingly, an enrichment for the p53 pathway was observed in the midcolon of Muc2hom mice, contrasting with an up-regulation of the Kras pathway in inflamed proximal samples. Heightened p53 activity in midcolon was accompanied by positive enrichment for apoptosis, suggesting a possible role in control of cellular integrity. No interactors of SLRPs could be mapped to the p53 or apoptosis pathway in proximal inflamed samples, suggesting that this pathway might be down-regulated to allow for cell proliferation necessary for tissue repair. Taken together, these findings highlight the central role that down-regulation of proteoglycans plays in the progression of chronic inflammation.
Figure 7.
Differential expression of proteoglycans distinguishes tissues in prepathologic and pathologic states. (A) Bar plot showing distribution of log2 fold changes for Small Leucine Rich Proteoglycans (SLRPs) found significantly differentially expressed in the comparison of middle Muc2hom vs Muc2het and/or proximal Muc2hom vs Muc2het samples (q-value < 0.05). (B) Box plots representing differential protein expression as in panel A, showing down-regulation from the comparison of middle Muc2hom vs Muc2het to proximal Muc2hom vs Muc2het for SLRPs identified in both comparisons. Dcn and proline and Prelp are highlighted as candidates up-regulated in the middle Muc2hom vs Muc2het and down-regulated in proximal Muc2hom vs Muc2het. (C) Bar plots showing the number of known interactors to SLRPs identified (left, in black) and those differentially expressed in the comparison of proximal Muc2hom vs Muc2het or middle Muc2hom vs Muc2het samples (right, in grey). (D) Circos plots displaying SLRPs on the bottom, differentially expressed in proximal Muc2hom vs Muc2het samples (left) or middle Muc2hom vs Muc2het samples (right). SLRPs are mapped to hallmark pathways (top) involving their known interactors, enriched (see Methods), either in proximal Muc2hom vs Muc2het samples (left) or middle Muc2hom vs Muc2het samples (right).
Matrisomal Orchestration of Tissue Remodeling in Colitis
Next, to infer the orchestrating role of matrisomal proteins in tissue remodeling, the 162 matrisomal proteins identified (Supplementary Table 1, Figure 4A) were interrogated for differential expression in mature Muc2hom mice, selecting only proteins annotated as ligands in the CellCellInteraction database.35 The same database was then was used to infer potential interactions using receptors found to be expressed differentially in Muc2hom mice at the transcriptomic level. In inflamed proximal Muc2hom tissue, 47 matrisomal proteins were identified as ligands to 27 differentially expressed receptors. In contrast, in midcolon, only 10 receptors and 37 matrisomal proteins were differentially expressed (Figure 8A, Supplementary Table 1), showing that the matrisomal regulome in prepathology maps to significantly fewer interactors than in inflamed tissue. For example, fibronectin (Fn1) interacted with 25 receptors in inflamed tissue, but with only 7 in prepathology (Figure 9B). Furthermore, the repertoire of differentially expressed integrins was reduced in prepathology. The only integrins engaging with matrisomal ligands were Integrin alpha and beta 6, compared with the inflamed state in which Integrin av, ae, al, az and b2 2 additionally were engaged. Taken together, this shows a greater complexity of regulation in overt inflammation compared to prepathology.
Figure 4.
Proteomic analysis of an extracellular matrix–enriched tissue fraction reflects disease state. (A) Venn diagram showing the proportion of proteins identified through DIA-MS classed as extracellular based on Gene Ontology terms (953; 40.9% of the total protein fraction) and the proportion of matrisomal proteins identified (162; amounting to 5% of the total protein fraction). (B) Volcano plot showing differentially expressed proteins in the proximal colon of Muc2hom (N = 4) vs Muc2het control (N = 5) mice. Red indicates significant up-regulation and blue indicates significant down-regulation (log2 fold change >1 or <-1; q-value < 0.05). (C) Dot plot showing ConsensusPathDB output,assessing pathway enrichment for Muc2hom vs Muc2het proximal colon samples. The top significantly enriched pathways are represented (P < .01). (D) Immunofluorescence pictures showing enrichment for CD4+ T cells (left) and B220/CD45R+ B cells (right) in Muc2hom compared with Muc2het proximal colon (4′,6-diamidino-2-phenylindole [DAPI] marking nuclei, and E-cadherin marking colonic epithelial cells). (E) Violin plots showing the distribution of protein log2 fold changes in Muc2hom compared with Muc2hetproximal samples (q-value < 0.05), where proteins are mapped to different cell type signatures (details in Methods). Positive enrichment is indicated by the median (black dot) above the red line, and negative enrichment when under this line. (F) Protein differential expression signature was extracted from the comparison of Muc2hom compared with Muc2het proximal samples (see Supplementary Table 2). A subset is shown here where log2 FC >2 or <-2, and q-value < 5 × 10e-7. (G) Box plots showing differential expression of proteins in Muc2hom compared with Muc2het proximal samples (q-value < 0.05), where ECM proteins are classed into categories. ∗P < .05, pairwise t test.
Figure 8.
Building networks representing matrisomal orchestration of cell type remodeling. (A) Circos plot showing differentially expressed matrisomal ligands (bottom, q-value < 0.05) and their known interactors expressed differentially at the transcriptomic level (top, adjusted P value < .05), in proximal Muc2hom (left) and middle Muc2hom (right) compared with location-matched Muc2het control samples. (B) Histograms showing the number of receptors expressed differentially at the transcriptomic level (x axis, adjusted P value < .05), for each matrisomal ligand differentially expressed in the comparison of proximal Muc2hom vs Muc2het (left) or middle Muc2hom vs Muc2het (right). (C) Bar plot representing the number of genes present in each combined cell type signatures, combining discrete cell types (see Methods). (D) Histogram showing the number (x-axis) and name of matrisomal proteins (as described in panel B) controlling each cell type present on the y-axis (combined cell type signature). (E) Histogram showing the number of cell types (combined cell type signature) controlled by each matrisomal protein as described in panel B.
Figure 9.
Matrisomal orchestration of tissue remodeling in colitis. (A) Circos plot showing matrisomal ligands differentially expressed (q-value < 0.05) in proximal Muc2hom vs Muc2het (left) or middle Muc2hom vs Muc2het (right), organized by groups (bottom) linked to cell types (top) containing receptors with which they are known to interact and are expressed differentially at the transcriptomic level (adjusted P value < .05, see Methods for details). The heat on the top panel represends mean log2 fold change for interactors to matrisomal ligands mapping to each cell type. (B) Heatmap showing matrisomal proteins part of the inflamed matrisome (y axis) and the cell types within which they have potential interactors (x-axis). The heat represents the number of interactors of each matrisomal ligand within each cell type, and is normalized per row, giving more power to unique interactions. (C) Immunofluorescence images showing the location of CD31+ endothelial cells (left panel, in red), CD68+ macrophages (middle panel, in yellow), and Serpinh1+ fibroblasts (right panel, in green) in proximal and middle Muc2hom colon. (D) Heatmaps showing the percentage of positive cells for each of the 3 cell types in 3 regions of the same area along the crypt axis (bottom, middle, and top) in proximal and middle Muc2hom colon (N = 3). (E) Heatmaps showing the expression of 4 ECM ligands in proximal and middle Muc2hom colon, expressed as intensity per area (N = 3). (D) Representative immunofluorescence image showing the distribution of Lamc1 along the crypt axis in proximal and middle Muc2hom colon. DAPI, 4′,6-diamidino-2-phenylindole.
Receptors interacting with matrisomal ligands were then mapped to cell types to reveal the extent of the matrisomal contribution to regulation of tissue function in the context of colitis (Figure 9C). This analysis indicated that 10 cell types were potentially regulated by differentially expressed matrisomal proteins in inflamed tissue, compared with 8 in prepathology (Figure 9D). In both states, endothelial cells had the most connections to matrisomal proteins. Matrisomal regulation of macrophages and B cells appeared limited to inflamed tissue (Figure 5A). Proteins such as fibronectin (Fn1) were implicated in engagement of 8 different cell types in inflammation. In prepathology, engagement of 4 cell types was the maximum for any matrisomal protein (Figure 9E).
Finally, an integrated network was created to link differentially expressed matrisomal proteins to the cell types they regulate (Figure 9A). This network revealed greater involvement of ECM regulators and secreted factors in inflammation compared with prepathology, and more widespread tissue landscape alterations extending to include immune cell types such as B cells, T cells, and macrophages.
Hierarchical clustering was used to recognize similarities in the regulation of different cell types by matrisomal proteins altered in inflammation (Figure 9B). This analysis revealed a possible coordinated regulation of fibroblasts and endothelial cells by a subset of matrisomal proteins, including lysyl oxidase homolog 2 (Loxl2) and neutrophil elastase (Elane). Of note, the presence of matrisomal proteins such as S100 calcium-binding protein A9 (S100A9), plasminogen (Plg), and Loxl2 in this signature suggests a shared function in increasing collagen cross-linking (Figure 9B).
To gain direct evidence for this coordinated regulation by matrisomal ligands, the distribution of macrophages, fibroblasts, and endothelial cells along the colonic crypt axis was first compared using immunofluorescence in Muc2hom prepathologic midcolon and inflamed proximal tissue (Figure 9C and D). In the former, CD31+ endothelial cells, CD68+ macrophages, and Serpin H family member 1 (Serpinh1)+ fibroblasts are located primarily in a region adjacent to the lower crypt epithelium and submucosa. With inflammation, fibroblasts and endothelial cells show a marked displacement toward the luminal surface at the top of the crypts, whereas macrophages largely remain in a more basal location. Next, the expression profile of 4 matrisomal ligands (Laminin subunit gamma 1 (Lamc1), Annexin 1, S100A9, and Elane) that are associated predominantly with fibroblast and endothelial regulation along the crypt axis was assessed similarly (Figure 4B and E). This revealed a trend for increased expression of these ligands in the same luminal region with inflammation. Of note, Lamc1 expression also was displaced toward the basal side of crypts (Figure 9F). These findings suggest that spatial changes in the frequency of fibroblasts and endothelial cells can be tracked to the ligands associated with their regulation. Together, these results define the orchestrating role of the matrisome in the development of colonic inflammatory disease.
Discussion
Despite widespread acceptance of the fundamental role of the matrisome in regulating tissue homeostasis, as well as remodeling in chronic disease states such as colitis, relatively few studies have focused on understanding and integrating the full repertoire of matrisomal orchestration.36 Here, the development of label-free mass spectrometry (DIA-MS) approaches to quantify proteins in ECM-enriched samples provided the opportunity to build an integrated picture of matrisomal regulation in a mouse model that captures much of the pathology and dysfunction seen in UC patients.25,26,37 Importantly, the choice of model also enabled identification of ECM remodeling events and candidate pathways involved in early tissue adaptation to chronic inflammation before any histologic manifestation.
Combining protein fractionation with comprehensive and sensitive data-independent acquisition–mass spectrometry revealed the extensive tissue remodeling accompanying colitis and related it to mediators of the inflammatory phenotype. For example, within the colitis signature, latent TGF-β proteins are enriched and are responsible for directing latent TGF-β to extracellular matrix microfibrils, where it becomes bioavailable upon tissue remodeling to mediate both inflammatory and fibrotic responses seen both in the Muc2KO model and in UC patients.38,39 In addition, the depth of this unbiased proteomic analysis allowed deconvolution of cell populations known to be associated with colonic inflammation in both murine models of colitis as well as in human disease, using approaches previously applied at the transcriptomic level using single-cell technologies.40, 41, 42
Many differentially expressed proteins were detected in the apparently histologically normal middle colon, which led to the finding that this tissue already had acquired a prepathologic state. This finding is reminiscent of recent observations in a delayed-onset model of induced colitis that identified protein changes before the onset of inflammatory disease.36 Analysis of colitis in older animals confirmed the development of a pancolitis, allowing interrogation of the proteomic data for adaptive responses that may restrain histologic manifestations of inflammation at earlier disease stages.
One class of proteins that may be involved in restricting inflammatory disease are SLRPs, which are increased in prepathology before their reduction in overt inflammation. Down-regulation of small leucine-rich proteoglycans is associated with poor outcomes in invasive breast cancer,43 and in chronic inflammation–associated cancers, including lung squamous cell carcinoma.24 Interrogating interactors of SLRPs differentially expressed in prepathology, we found evidence for increased p53 activity that was not seen in the inflamed state. This is in accord with previous studies showing that the proliferative phase of tissue regeneration requires the coordinated up-regulation of trophic pathways (as seen with Kras here) while down-regulating that of p53.44 Hence, the transient increased expression of SLRPs in the prepathologic state may coincide with the activation of a p53 checkpoint control that subsequently is lost as inflammatory disease develops, resulting in a corresponding increase in cancer risk.
Notably, the prepathologic state was associated with increased expression of many mitochondrial proteins. In inflammatory bowel disease patients, genetic risk factors affecting mitochondrial function have been identified and their altered metabolism proposed as causative in inflammatory disease.45, 46, 47 Recently, altered mitochondrial function has been shown to predict disease recurrence in Crohn’s disease, supporting a role for mitochondrial dysfunction developing before the onset of active inflammation.48
To uncover the contribution of the matrisome in orchestrating alterations in tissue ecology during colitis, an integrative approach was used to map ECM ligands to receptors with altered expression at the transcriptomic level. This ligand-receptor interaction analysis showed positive interactions between matrisomal proteins and T-cell, B-cell, and macrophage populations consistent with their expansion in inflamed tissues. Moreover, common matrisomal protein subsets could be associated with specific cell types such as fibroblasts and endothelial cells. For example, in the inflamed state, of the 38 proteins interacting with fibroblasts and 39 interacting with endothelial cells, only EGF-like domain protein 7 (Egfl7) is specific to endothelial cells. The other 38 proteins interact with both endothelial cells and fibroblasts. Spatial analysis showed a displacement of specific matrisomal ligands toward the top of crypts in inflamed regions, which was associated with a displacement of both cell types, illustrating a coordinated regulation driven by ECM remodeling accompanying colitis. Matrisomal ligands associated with remodeling of these cell types were produced by immune cells or inflammatory fibroblasts, representing an example of dynamic reciprocity in the context of colitis.
In contrast, in the prepathologic state, fewer matrisomal ligands and interactors are engaged and they are predicted to have an impact on a smaller number of cell types, primarily epithelial and endothelial cells. Therefore, assessing the prepathologic tissue landscape to determine how altered ECM composition engages with and regulates different cell types might provide new insights into how early tissue adaptations delay the subsequent onset of colitis. These observations will benefit from performing complementary studies of uninvolved mucosa in UC patients to establish if episodic recurrence of colitis in defined regions relates to local loss of protective mechanisms rather than an acquisition of susceptibility.
Methods
Mice
Animal care and procedures were performed at the Cancer Research UK Cambridge Institute Biological Resource Unit according to UK Home Office guidelines. Mice were of a C57BL/6 background. The Muc2KO line used was described by Velcich et al.25 Genotyping of Muc2KO mice was outsourced to Transnetyx (Cordova, TN).
Treatment of animals
The mice were housed under controlled conditions (temperature, 21°C ± 2°C; humidity, 55% ± 10%; 12-hour light/dark cycle) in a specific-pathogen free facility (tested according to the recommendations for health monitoring by the Federation of European Laboratory Animal Science Associations). Animals had unrestricted access to food and water.
Histology
Immunohistochemistry/immunofluorescence
Mouse colons were opened longitudinally, fixed overnight in 4% paraformaldehyde, and processed for histology by conventional means. Sections were dewaxed and rehydrated, followed by heat-induced epitope-retrieval using 10 mmol/L Tri-sodium citrate buffer pH 6.0. Immunochemistry or immunofluorescence was performed as described previously.49 Antibodies used in immunohistochemistry were Muc2 (cat. sc-15334; Santa Cruz) and Ki67 (ab15580; Abcam). Antibodies used in immunofluorescence were E-cadherin (cat. 610182; BD Bioscience), B220/CD45R (cat. MAB1217; R&D Systems), Grem1 (cat. AF956; R&D Systems), CD68a (cat. 98941; Cell Signaling), Serpinh1 (cat. ab109117; Abcam), CD31 (cat. ab28364; Abcam), and Lamc1 (cat. ab233389; Abcam).
Spatial phenotyping
Fixed colons were Swiss-rolled luminal side in, starting from the proximal end. On-edge sections were cut at 3 different levels and stained with H&E. Immune hallmarks were scored and normalized along the colon length.
Image analysis
The Indica Laboratories HALO image analysis platform was used. The Indica Laboratories Area Quantification FL v.1.0 program was used for quantification of fluorescent staining (percentage of tissue stained and intensity of staining). The Indica Laboratories Highplex FL v.2.2.3 program was used for cell deconvolution based on 4′,6-diamidino-2-phenylindole nuclear expression and quantification of the percentage of cells expressing specific fluorescent markers. For spatial analysis along the crypt axis, layers of the same area were drawn on the bottom, middle, and top of the crypt, within which the analysis was performed.
Extracellular Matrix Enrichment
Decellularization
Colon tissue was dissected from 6-month-old Muc2hom mice and Muc2het mice (n = 5 mice per genotype). After flushing with phosphate-buffered saline, 40–60 mg colon tissue pellets were flash-frozen and stored at -80°C. On the day of decellularization, pellets were homogenized using a Qiagen Lyser, before processing using a Compartmental Extraction Kit (2145; Millipore) as per the manufacturer’s protocol. This allowed extraction of extracellular matrix proteins through stepwise washes with salt solutions and detergents.
Western blot
Purification of each cellular compartment was tested by Western blot with antibodies for glyceraldehyde-3-phosphate dehydrogenase (cat. G8795; Sigma), High Mobility Group AT-Hook 1 (Hmga1) (cat. ab129153; Abcam), β1 integrin (cat. 610467; BD Bioscience), β-actin (cat. ab6276; Abcam), and collagen 1 (cat. PA5-95137; Invitrogen). One fifth of the extracellular protein pellet was dissolved in 30 uL of 4× sodium dodecyl sulfate (SDS) buffer supplemented with 100 mmol/L dithiothreitol (DTT). For other compartments, 10 uL sample was mixed with 10× lithium dodecyl sulfate (LDS) and 4× SDS.
Label-Free Mass Spectrometry / Data Independent Acquisition
Chemicals
Liquid chromatography–MS grade acetonitrile (ACN), methanol, and water were obtained from Burdick & Jackson (Muskegon, MI). Reagents for protein chemistry, including SDS, ammonium bicarbonate, triethylammonium bicarbonate (TEAB), iodoacetamide, DTT, sequencing-grade endoproteinase Lys-C, and formic acid (FA) were purchased from Sigma-Aldrich (St. Louis, MO). Sequencing-grade trypsin was purchased from Promega (Madison, WI). Glycerol-free Peptide:N-glycosidase F was purchased from New England BioLabs (Ipswich, MA).
Solubilization of ECM proteins
The extracted ECM pellets were solubilized by agitation for 10 minutes in a solution containing 1% SDS and 50 mmol/L DTT, followed by sonication for 10 minutes, and, finally, heating at 85°C for 1 hour with agitation.
Protein digestion and desalting
Samples were solubilized with 4% SDS and 50 mmol/L TEAB at pH 8. Proteins were reduced with 20 mmol/L DTT (10 min at 50°C followed by 10 min at room temperature) and then alkylated with 40 mmol/L iodoacetamide (30 min at room temperature in the dark). Samples were acidified with a final concentration of 1.2% phosphoric acid, and diluted with 7 volumes S-trap buffer (90% methanol in 100 mmol/L TEAB, pH 8). Samples then were loaded onto the S-trap microspin columns (Protifi, Farmingdale, NY) and spun at 4000 × g for 10 seconds. The S-trap columns were washed with S-trap buffer twice at 4000 × g for 10 seconds each, before incubating the proteins with 250 ng sequencing-grade endoproteinase Lys-C in 50 mmol/L TEAB (pH 8) at 37°C for 2 hours. Then, 2.4 μg sequencing grade trypsin in 50 mmol/L TEAB (pH 8) for 1 hour at 47°C were added to the sample. After a 1-hour digestion at 47°C, the same amount of trypsin was added again and proteins were digested overnight at 37°C. Peptides were eluted sequentially with 50 mmol/L TEAB (pH 8), 0.5% FA in water, 50% acetronitrile, and 0.5% FA in water. After vacuum drying, samples were resuspended in 300 μL of 25 mmol/L ammonium bicarbonate in water, and spot-checked to ensure a pH of 7–8. Subsequently, 9 μL (4500 U) glycerol-free PNGase F were added, and samples were incubated for 3 hours at 37⁰C with agitation. This reaction was quenched with 10% FA in water for a final concentration of 1%, and spot-checked again to ensure a pH of 2–3. The quenched peptide samples were vacuum dried and resuspended in 20 μL of 0.2% FA, before desalting them using ZipTip (Merck, C5737) with 0.6 μL C18 resin (Sigma-Aldrich). Finally, samples were concentrated in a vacuum concentrator and resuspended in aqueous 0.2% FA containing indexed retention time (iRT) peptide standards (Biognosys, Schlieren, Switzerland).50
Mass spectrometric analysis
Liquid chromatography–tandem MS analyses were performed on a Dionex UltiMate 3000 system coupled to an Orbitrap Eclipse Tribrid mass spectrometer (both from Thermo Fisher Scientific, San Jose, CA). The solvent system consisted of 2% ACN, 0.1% FA in water (solvent A) and 98% ACN, 0.1% FA in water (solvent B). Proteolytic peptides were loaded onto an Acclaim PepMap 100 C18 trap column (0.1 × 20 mm, 5 μm particle size; Thermo Fisher Scientific) over 5 minutes at 5 μL/min with 100% solvent A. Peptides were eluted on an Acclaim PepMap 100 C18 analytical column (75 μm × 50 cm, 3 μm particle size; Thermo Fisher Scientific) at 0.3 μL/min using the following gradient of solvent B: 2% for 5 minutes, linear from 2% to 20% in 125 minutes, linear from 20% to 32% in 40 minutes, up to 80% in 1 minute, 80% for 9 minutes, and down to 2% in 1 minute. The column was equilibrated at 2% for 29 minutes (total gradient length = 210 min).
Every sample was acquired in data-independent acquisition (DIA) mode [20-22] using the following settings: full MS spectra were collected at 120,000 resolution (Automatic Gain Control (AGC) target: 3e6 ions, maximum injection time: 60 ms, 350–1650 m/z), and MS/MS spectra at 30,000 resolution (AGC target: 3e6 ions, maximum injection time: auto, Normalised Collision Energy: 27, fixed first mass 200 m/z). The isolation scheme consisted in 26 variable windows covering the 350–1650 m/z range with an overlap of 1 m/z22 (Supplementary Table 2).
DIA-MS data processing with Spectronaut
All DIA data was processed in Spectronaut version 14.10.201222.47784 (Biognosys) using direct DIA. Data were searched against the Mus musculus proteome with 58,430 protein entries (UniProtKB-TrEMBL), accessed on January 31, 2018. Trypsin/P was set as digestion enzyme and two missed cleavages were allowed. Cysteine carbamidomethylation was set as fixed modification, and methionine oxidation and protein N-terminus acetylation as variable modifications. Data extraction parameters were selected as dynamic, and nonlinear iRT calibration with precision iRT was selected. Identification was performed using a 1% precursor and protein q-value, and iRT profiling was selected. Quantification was based on the tandem MS peak area of the 3–6 best fragment ions per precursor ion, peptide abundances were obtained by summing precursor abundances and protein abundances by summing peptide abundances. Interference correction was selected, and local normalization was applied. Differential protein abundance analysis was performed using the paired t test, and P values were corrected for multiple testing, specifically applying groupwise testing corrections using the Storey51 method.52 For differential analysis, protein groups with at least 2 unique peptides and a q-value ≤ 0.05 were considered to be altered significantly (Supplementary Table 5).
Bulk RNA Sequencing
Colon samples from three 10-month-old Muc2hom and Muc2het mice were used for bulk RNA sequencing. RNA extraction was performed following instructions from the Qiagen RNA extraction kit. Library preparation was performed by the genomics core at the Cancer Research UK Cambridge Institute using the Illumina Stranded messenger RNA Prep kit (20040532; Illumina) according to the manufacturer’s instructions. Samples were submitted for sequencing in the Illumina Novaseq platform with 50-bp paired-end reads. Differential expression analysis was performed using DESeq253. An interaction model was used to identify differentially expressed genes in proximal or mid-Muc2hom colon compared with location-matched Muc2het samples.
Bioinformatic Analysis
Plots were generated using either the ggplot2 package54 in R or GraphPad Prism.
Pearson correlation
The Pearson coefficients of correlation were determined between the different replicates using the stats package in R (version 4.0.2, version 1.3.1093; RStudio) and the abundances of all quantifiable protein groups as input.
Pathway analysis
An overrepresentation analysis was performed using ConsensusPathDB (release 35, 05.06.2021)28 to determine which Gene Ontology terms were enriched significantly. Gene Ontology terms identified from the overrepresentation or underrepresentation analysis were subjected to the following filters: q-value < 0.01, number of background genes ≥ 5, number of candidate genes ≥ 2, and log2 fold enrichment >1 or <-1.
Cell type deconvolution
Cell type signatures were created by combining single-cell RNA sequencing data sets provided in Kinchen et al,40 Smillie et al,41 and Mitsialis et al42 [DATASETS]. Two different data sets were created, with more- or less-specific cell types (Figures 7F and 9B). Significantly differentially expressed proteins (q-value < 0.05) or genes (from transcriptomic data, adjusted P value < 0.05) were mapped to cell type signatures and the spread of log2 fold changes for each cell type was used to extrapolate information on enrichment.
Protein interaction analysis
The ligand-receptor interaction set from the CellCellInteractions database (version 1.0, built April 25, 2017, containing iRefIndex version 14, Pathway Commons version 8, and BioGRID version 3.4.147)35 was used to find potential interactors to matrisomal proteins.
SLRP pathway enrichment
Pathway enrichment scores were calculated as follows: , where Enrichement Scoreinteractors is calculated as the average (log2 fold change) ∗ the average (q-value) for all interactors found in the pathway; Ndifferentially expressed interactors represents the number of interactors to SLRPs mapped to the pathway and expressed differentially in the relevant comparison; and Ninteractors represents the total number of potential interactors to all SLRPs with interactors in the pathway.
Integrative network analysis
Protein parts of the colon matrisome55 and expressed differentially in the comparison of either proximal inflamed vs true normal or middle matched normal vs true normal were used for this analysis. Those data sets were processed to keep proteins annotated as ligands (Bader Laboratory data set of protein types). Bulk RNA sequencing performed on Muc2hom vs Muc2het colon samples was used to identify differentially expressed genes in the same comparisons. Those data sets were processed to keep genes annotated as receptors, and mapped to matrisomal ligands with which they have a described interaction.
Circos plots
The circlize package was used to produce circos plots,56 in which matrisomal proteins were aligned on the bottom and linked to potential interactors at the top, which were either mapped to pathways or cell types.
Acknowledgments
The authors thank the Histopathology core, Genomics core, and the Biological Resources Unit at the Cancer Research UK Cambridge Institute for technical support; and the members of Winton Laboratories for their help. The authors acknowledge the generous support from SCIEX for the ZenoTOF 7600 system and a Waters M-class high-performance liquid chromatography system at the Buck Institute.
CRediT Authorship Contributions
Elisa Brune Moutin, PhD (Conceptualization: Equal; Data curation: Lead; Formal analysis: Lead; Investigation: Lead; Methodology: Equal; Software: Equal; Validation: Lead; Visualization: Lead; Writing – original draft: Lead; Writing – review & editing: Lead)
Joanna Bons, PhD (Data curation: Equal; Formal analysis: Equal; Methodology: Equal; Software: Lead; Validation: Supporting; Writing – review & editing: Supporting)
Giada Giavara, MSc (Investigation: Supporting; Methodology: Supporting; Project administration: Supporting; Validation: Supporting)
Filipe Lourenco, PhD (Conceptualization: Equal; Funding acquisition: Supporting; Investigation: Supporting; Methodology: Supporting; Writing – review & editing: Equal)
Deng Pan, PhD (Investigation: Supporting; Methodology: Supporting; Writing – review & editing: Equal)
Jordan Bruce Burton, PhD (Investigation: Supporting; Methodology: Supporting)
Samah Shah, BSc (Investigation: Supporting; Methodology: Supporting)
Mathilde Colombe, MSc (Investigation: Supporting; Methodology: Supporting)
Philippe Gascard, PhD (Conceptualization: Supporting; Data curation: Supporting; Funding acquisition: Equal; Project administration: Equal; Writing – review & editing: Equal)
Thea Tlsty, PhD (Conceptualization: Equal; Data curation: Supporting; Funding acquisition: Lead; Project administration: Equal; Writing – review & editing: Equal)
Birgit Schilling, PhD (Conceptualization: Supporting; Data curation: Supporting; Methodology: Equal; Writing – review & editing: Equal)
Doug James Winton, BSc, PhD (Conceptualization: Equal; Funding acquisition: Lead; Investigation: Equal; Project administration: Lead; Resources: Lead; Supervision: Lead; Writing – original draft: Lead; Writing – review & editing: Lead)
Footnotes
Conflicts of interest The authors disclose no conflicts.
Funding This work was supported by Cancer Research UK grant awards A29074 (D.W.) and A27145 (T.D.T.) as part of STORMing Cancer (Cancer Grand Challenge); National Cancer Institute Awards 5R35CA197694 (T.D.T.) and 5R50CA211543 (P.G.); CRUK Cambridge Institute core funding A24456 (F.L., R.K., and D.J.W.); and Wellcome Trust grant 103805 (M.C.).
Data Availability All data relevant to this study are available from the corresponding authors upon reasonable request. Raw data and complete mass spectrometry data sets have been uploaded to the Mass-Spectrometry Interactive Virtual Environment (MassIVE), repository of the Center for Computational Mass Spectrometry at University of California San Diego, and can be downloaded using the following link: ftp://MSV000092056@massive.ucsd.edu (MassIVE ID number: MSV000092056; ProteomeXchange ID: PXD042551).
Note: To access the supplementary material accompanying this article, go to the full text version at http://doi.org/10.1016/j.jcmgh.2024.01.003.
Supplementary Material
References
- 1.Coussens L.M., Werb Z. Inflammation and cancer. Environmental Health and Preventive Medicine. 2018;23:(860–867) doi: 10.1186/s12199-018-0740-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pompili S., Latella G., Gaudio E., Sferra R., Vetuschi A. The charming world of the extracellular matrix: a dynamic and protective network of the intestinal wall. Front Med. 2021;8:1–19. doi: 10.3389/fmed.2021.610189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.McCabe M.C., Saviola A.J., Hansen K.C. Mass spectrometry-based atlas of extracellular matrix proteins across 25 mouse organs. J Proteome Res. 2023;22:790–801. doi: 10.1021/acs.jproteome.2c00526. [DOI] [PubMed] [Google Scholar]
- 4.Sakakura T., Nishizuka Y., Dawe C.J. Mesenchyme-dependent morphogenesis and epithelium-specific cytodifferentiation in mouse mammary gland. Science. 1976;194:1439–1441. doi: 10.1126/science.827022. [DOI] [PubMed] [Google Scholar]
- 5.Mott J.D., Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol. 2004;16:558–564. doi: 10.1016/j.ceb.2004.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Muncie J.M., Weaver V.M. The physical and biochemical properties of the extracellular matrix regulate cell fate. Curr Top Dev Biol. 2019;130:1–30. doi: 10.1016/bs.ctdb.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Nakayama K.H., Batchelder C.A., Lee C.I., Tarantal A.F. Decellularized rhesus monkey kidney as a three-dimensional scaffold for renal tissue engineering. Tissue Eng. 2010;16:2207–2216. doi: 10.1089/ten.tea.2009.0602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rana D., Zreiqat H., Benkirane-Jessel N., et al. Development of decellularized scaffolds for stem cell-driven tissue engineering. J Tissue Eng Regen Med. 2017;11:942–965. doi: 10.1002/term.2061. [DOI] [PubMed] [Google Scholar]
- 9.Ullah I., Busch J.F., Rabien A., et al. Adult tissue extracellular matrix determines tissue specification of human iPSC-derived embryonic stage mesodermal precursor cells. Adv Sci. 2020;7:1–10. doi: 10.1002/advs.201901198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bissell M.J., Hall H.G., Parry G. How does the extracellular matrix direct gene expression? J Theor Biol. 1982;99:31–68. doi: 10.1016/0022-5193(82)90388-5. [DOI] [PubMed] [Google Scholar]
- 11.Kirov S., Sasson A., Zhang C., et al. Degradation of the extracellular matrix is part of the pathology of ulcerative colitis. Mol Omics. 2019;15:67–76. doi: 10.1039/c8mo00239h. [DOI] [PubMed] [Google Scholar]
- 12.Baugh M.D., Perry M.J., Hollander A.P., et al. Matrix metalloproteinase levels are elevated in inflammatory bowel disease. Gastroenterology. 1999;117:814–822. doi: 10.1016/s0016-5085(99)70339-2. [DOI] [PubMed] [Google Scholar]
- 13.Ingber D.E. Cancer as a disease of epithelial-mesenchymal interactions and extracellular matrix regulation. Differentiation. 2002;70:547–560. doi: 10.1046/j.1432-0436.2002.700908.x. [DOI] [PubMed] [Google Scholar]
- 14.Pickup M.W., Mouw J.K., Weaver V.M. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014;15:1243–1253. doi: 10.15252/embr.201439246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Marangio A., Biccari A., D’Angelo E., et al. The study of the extracellular matrix in chronic inflammation: a way to prevent cancer initiation? Cancers. 2022;14:5903. doi: 10.3390/cancers14235903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ordás I., Eckmann L., Talamini M., et al. Ulcerative colitis. Lancet. 2012;380:1606–1619. doi: 10.1016/S0140-6736(12)60150-0. [DOI] [PubMed] [Google Scholar]
- 17.Mortensen J.H., Lindholm M., Langholm L.L., et al. The intestinal tissue homeostasis–the role of extracellular matrix remodeling in inflammatory bowel disease. Gastroenterol Hepatol. 2019;10:977–993. doi: 10.1080/17474124.2019.1673729. [DOI] [PubMed] [Google Scholar]
- 18.Koelink P.J., Overbeek S.A., Braber A., et al. Collagen degradation and neutrophilic infiltration: a vicious circle in inflammatory bowel disease. Gut. 2014;63:578–587. doi: 10.1136/gutjnl-2012-303252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gordon I.O., Agrawal N., Willis E., et al. Fibrosis in ulcerative colitis is directly linked to severity and chronicity of mucosal inflammation. Aliment Pharmacol Ther. 2018;47:922–939. doi: 10.1111/apt.14526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ullman T.A., Itzkowitz S.H. Intestinal inflammation and cancer. Gastroenterology. 2011;140:1807–1816. doi: 10.1053/j.gastro.2011.01.057. [DOI] [PubMed] [Google Scholar]
- 21.Gillet L.C., Navarro P., Tate S., et al. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: a new concept for consistent and accurate proteome analysis. Mol Cell Proteomics. 2012;11:1–17. doi: 10.1074/mcp.O111.016717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Collins B.C., Hunter C.L., Liu Y., et al. Multi-laboratory assessment of reproducibility, qualitative and quantitative performance of SWATH-mass spectrometry. Nat Commun. 2017;8:1–11. doi: 10.1038/s41467-017-00249-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bruderer R., Bernhardt O.M., Gandhi T., et al. Optimization of experimental parameters in data-independent mass spectrometry significantly increases depth and reproducibility of results. Mol Cell Proteomics. 2017;16:2296–2309. doi: 10.1074/mcp.RA117.000314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bons J., Pan D., Shah S., et al. Data-independent acquisition and quantification of extracellular matrix from human lung in chronic inflammation-associated carcinomas. Proteomics. 2023;23 doi: 10.1002/pmic.202200021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Velcich A., Yang W., Heyer J., et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 2002;295:1726–1729. doi: 10.1126/science.1069094. [DOI] [PubMed] [Google Scholar]
- 26.Van der Sluis M., De Koning B.A.E., De Bruijn A.C.J.M., et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006;131:117–129. doi: 10.1053/j.gastro.2006.04.020. [DOI] [PubMed] [Google Scholar]
- 27.Shao X., et al. MatrisomeDB 2.0: 2023 updates to the ECM-protein knowledge database. Nucleic Acids Res. 2023;51:D1519–D1530. doi: 10.1093/nar/gkac1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kamburov A., Weirling C., Lehrach H., Herwig R. ConsensusPathDB - a database for integrating human functional interaction networks. Nucleic Acids Res. 2009;37(Suppl 1):623–628. doi: 10.1093/nar/gkn698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Moll R., Divo M., Langbein L. The human keratins: biology and pathology. Histochem Cell Biol. 2008;129:705–733. doi: 10.1007/s00418-008-0435-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Czarnewski P., Parigi S., Sorini C., et al. Conserved transcriptomic profile between mouse and human colitis allows unsupervised patient stratification. Nat Commun. 2019;10:1–11. doi: 10.1038/s41467-019-10769-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Solé M., Unzeta M. Vascular cell lines expressing SSAO/VAP-1: a new experimental tool to study its involvement in vascular diseases. Biol Cell. 2011:103543–103557. doi: 10.1042/BC20110049. [DOI] [PubMed] [Google Scholar]
- 32.Schaefer L., Iozzo R. Biological functions of the small leucine-rich proteoglycans: from genetics to signal transduction. J Biol Chem. 2008;283:21305–21309. doi: 10.1074/jbc.R800020200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hocking A.M., Shinomura T., McQuillan D.J. Leucine-rich repeat glycoproteins of the extracellular matrix. Matrix Biol. 1998;17:1–19. doi: 10.1016/s0945-053x(98)90121-4. [DOI] [PubMed] [Google Scholar]
- 34.Iozzo R.V., Schaefer L. Proteoglycan form and function: a comprehensive nomenclature of proteoglycans. Matrix Biol. 2015;42:11–55. doi: 10.1016/j.matbio.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Isserlin R, Voisin V, Ailles L, et al. Cell-cell interaction database [DATASET]. Zenodo, 2020. 10.5281/zenodo.7589953 [DOI]
- 36.Shimshoni E., Adir I., Afi R., et al. Distinct extracellular–matrix remodeling events precede symptoms of inflammation. Matrix Biol. 2020;96:47–68. doi: 10.1016/j.matbio.2020.11.001. [DOI] [PubMed] [Google Scholar]
- 37.Johansson M.E.V., Gustafsson J.K., Holmen-Larsson J., et al. Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis. Gut. 2014;63:281–291. doi: 10.1136/gutjnl-2012-303207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rifkin D.B., Rifkin W.J., Zilberberg L. LTBPs in biology and medicine: LTBP diseases. Matrix Biol. 2018;71–72:90–99. doi: 10.1016/j.matbio.2017.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mak J.W.Y., Ng S.C. Epidemiology of fibrostenosing inflammatory bowel disease. J Dig Dis. 2020;21:332–335. doi: 10.1111/1751-2980.12853. [DOI] [PubMed] [Google Scholar]
- 40.Kinchen J., Chen H.H., Parikh K., et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell. 2018;175:372–386.e17. doi: 10.1016/j.cell.2018.08.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Smillie C.S., Biton M., Ordovas-Montanes J., et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019;178:714–730. doi: 10.1016/j.cell.2019.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mitsialis V., Wall S., Liu P., et al. Single-cell analyses of colon and blood reveal distinct immune cell signatures of ulcerative colitis and Crohn’s disease. Gastroenterology. 2020;159:591–608. doi: 10.1053/j.gastro.2020.04.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Troup S., Njue C., Kliewer E.V., et al. Reduced expression of the small leucine-rich proteoglycans, lumican, and decorin is associated with poor outcome in node-negative invasive breast cancer. Clin Cancer Res. 2003;9:207–214. [PubMed] [Google Scholar]
- 44.Charni M., Aloni-Grinstein R., Molchadsky A., et al. P53 on the crossroad between regeneration and cancer. Cell Death Differ. 2017;24:8–14. doi: 10.1038/cdd.2016.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Fukushima K., Fiocchi C. Paradoxical decrease of mitochondrial DNA deletions in epithelial cells of active ulcerative colitis patients. Am J Physiol Gastrointest Liver Physiol. 2004;286:804–813. doi: 10.1152/ajpgi.00398.2003. [DOI] [PubMed] [Google Scholar]
- 46.Beltran B., Nos P., Dasi F., et al. Mitochondrial dysfunction, persistent oxidative damage, and catalase inhibition in immune cells of naïve and treated Crohn’s disease. Inflamm Bowel Dis. 2010;16:76–86. doi: 10.1002/ibd.21027. [DOI] [PubMed] [Google Scholar]
- 47.Rath E., Haller D. Mitochondria at the interface between danger signaling and metabolism : role of unfolded protein responses in chronic inflammation. Basic Sci Rev. 2012;18:1364–1377. doi: 10.1002/ibd.21944. [DOI] [PubMed] [Google Scholar]
- 48.Khaloian S., Rath E., Hammoudi N., et al. Mitochondrial impairment drives intestinal stem cell transition into dysfunctional Paneth cells predicting Crohn’s disease recurrence. Gut. 2020;69:1939–1951. doi: 10.1136/gutjnl-2019-319514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tomic G., Morrissey E., Kozar S., et al. Phospho-regulation of ATOH1 is required for plasticity of secretory progenitors and tissue regeneration. Cell Stem Cell. 2018;23:436–443. doi: 10.1016/j.stem.2018.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Escher C., Reiter L., MacLean B., et al. Using iRT, a normalized retention time for more targeted measurement of peptides. Proteomics. 2012;12:1111–1121. doi: 10.1002/pmic.201100463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Storey J.D. A direct approach to false discovery rates. J R Stat Soc B Stat Methodol. 2002;64:479–498. [Google Scholar]
- 52.Burger T. Gentle introduction to the statistical foundations of false discovery rate in quantitative proteomics. J Proteome Res. 2018;17:12–22. doi: 10.1021/acs.jproteome.7b00170. [DOI] [PubMed] [Google Scholar]
- 53.Love M.I., Huber W., Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology. 2014:1–21. doi: 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Elegant Graphics for Data Analysis. Journal of the Royal Statistical Society Series A: Statistics in Society ggplot2. 2016;174(1):245–246. [Google Scholar]
- 55.Naba A., Clauser K.R., Hoersh S., et al. The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol Cell Proteomics. 2012;11 doi: 10.1074/mcp.M111.014647. M111.014647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gu Z., Gu L., Eils R., et al. Circlize implements and enhances circular visualization in R. Bioinformatics. 2014;30:2811–2812. doi: 10.1093/bioinformatics/btu393. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.










