MMP dysregulation in TNBC is associated with relapse-free and overall survival.
Given the relevance of MMPs in the first step of the metastatic process, we explored the differences in mRNA expression levels between non-metastatic TNBC and normal tissue samples in The Cancer Genome Atlas (TCGA) cohort. Seven MMPs were downregulated and ten MMPs were upregulated in TNBC tumors (n = 90) compared to normal tissues (n = 99, Figure S1a). Interestingly, six consecutive MMP genes with increased expression in TNBC tumors are encoded on the same genomic locus (Fig. 1a), in a region that harbors nine different MMP genes, located at Chr11q22.2 (Figure S1b). Furthermore, coexpression patterns were observed in TNBC tumors between genes located on each side of the MMP region, revealing two different expression clusters (Fig. 1b), which were defined as 5’MMP region (containing MMP7, MMP8, MMP20, and MMP27; in addition to neighbor non-MMP genes TMEM123 and BIRC3) and 3’MMP region (containing MMP1, MMP3, MMP10, MMP12, and MMP13). The ratio of the expression levels between MMP genes located at Chr11q22.2 (3’MMP/5’MMP) was significantly higher in TNBC (Figure S1c). Thus, we hypothesized that the alternative expression of these two clusters might be associated with survival in TNBC patients. Using the Kaplan-Meier analysis in patients with TNBC (n = 417), we observed that tumors with higher expression of MMPs located at the 3’MMP region have a significantly shorter relapse-free survival (RFS; Fig. 1c, [log rank P = 0.023; hazard ratio (HR) = 1.57, 95% CI = 1.06 − 2.33]) and overall survival (Fig. 1c, OS; [log rank P = 0.0048; HR = 2.65, 95% CI = 1.31 − 5.37]). We examined if the expression of these MMP genes was also linked to differential prognosis in non-TNBC cohorts. No differences were observed in RFS neither in hormone receptor-positive nor HER2-positive breast cancer subtypes. Regarding OS, the MMP signature was associated with a shorter survival in hormone receptor-positive, but a better prognosis was found in HER2-positive breast cancer. Interestingly, when considering non-breast solid tumors, this signature was associated with a worse prognosis in liver cancer, lung adenocarcinoma, and sarcoma, whereas it correlated with better survival in gastric cancer (Figure S2). Thus, the upregulation of 3’MMP genes appears to be associated with worse survival outcomes in patients beyond TNBC, so we will focus on this breast cancer subtype as a model for the study of this alteration.
An insulator element nearby the MMP8 gene promoter region is involved in regional MMP gene regulation.
Considering the expression pattern exhibited by the MMP genes at Chr11q22.2, we explored the distribution of gene regulatory elements in this region ( Figure S3a). We found 29 potential insulator elements (IEs) and 13 potential enhancer elements (EEs) at Chr11q22.2 (Fig. 2a). The IEs play a pivotal role in the topologically associating domain (TAD) formation, being DNA elements recognized by the CCCTC-binding factor (CTCF) and contributing to chromatin loop formation [32]. We explored potential TADs at Chr11q22.2 using data from CTCF ChIA-PET (Figure S3b). This data suggested that the IE located between the MMP8 promoter region and the MMP10 gene body (chr11:102,732,800 − 102,733,900; hg38), hereinafter IE8, demarcates a boundary for a TAD contributing to the expression signature observed by MMP genes at Chr11q22.2. This is supported by data from eQTLs. We observed that SNPs tend to modulate the expression of genes located on the same side TADs at Chr11q22.2 (Figure S3c). It is worth noting that the CTCF binding site of the IE8 was depleted in mutations according to data from the Pan-Cancer Analysis of Whole Genomes (PCAWAG) database (N = 2,658) and whole genome sequencing of TNBC samples (N = 237) (Figure S3d), indicating that its normal function is required in cancer.
To explore the role of IE8 activation on MMP expression, we stably disrupted the CTCF binding motif on IE8 using CRISPR/Cas9 technology in TNBC cell lines. We initially explored the expression levels of MMP genes in a panel of well-characterized TNBC cell lines (n = 17; Fig. 2b). We selected MDA-MB-231 and MDA-MB-436, two cell lines that showed an increased expression of the 3’MMP genes when compared to the 5’MMP genes, which recapitulates the expression pattern observed in TNBC-patient samples (Fig. 1a and S1c). We characterized chromatin interactions in these two cell lines using Hi-C. We observed that IE8 colocalizes with TAD boundaries in both models, in concordance with data from ChIA-PET (Fig. 2c).
Cancer cells were transiently transfected with a Cas9-containing plasmid and a sgRNA against the IE8. A single clone for each condition was selected to perform further experiments (Figure S4). To evaluate the disruption, CTCF binding ability to IE8 was tested in both cell lines through Cleavage Under Targets and Release Using Nuclease (CUT&RUN) followed by qPCR [33]. Basal levels of CTCF occupancy were higher in MDA-MB-231 than in MDA-MB-436. After IE8 disruption, a significant decrease in CTCF binding was observed in both MDA-MB-231 and MDA-MB-436 when compared to wild-type counterparts (Fig. 2d).
Hi-C was also performed on TNBC cell lines after IE8 disruption. We found that disruption of IE8 did not change TAD distribution at Chr11q22.2 (Figure S5a). However, we observed that IE8 tends to interact with 3’ distal regions in both models and conditions. Interestingly, some of these potential interactions contain other IEs disposed of in a convergent orientation to IE8. It has been reported that 90% of IE interactions are convergently oriented. Thus, we identified some potential partners of IE8 located at the 3’MMP region of Chr11q22.2 (Figure S5b).
Disruption of IE8 leads to local chromatin accessibility changes.
To address the implications of the IE8 disruption on chromatin accessibility we performed ATAC-seq on our TNBC cell line models. 34,047 common peaks between all replicates were identified in MDA-MB-231, whereas 35,347 common peaks were found in MDA-MB-436. Accessibility analysis was performed by assessing differential accessibility peaks, which were defined by either their presence in only one condition (WT or IE8 dis) or by the significant change in intensity of shared peaks between conditions (Figure S6a). Thus, we identified 3,083 and 2,232 regions that gained and lost accessibility upon IE8 disruption in MDA-MB-231, respectively. Regarding MDA-MB-436, 8,673 and 370 regions were more and less accessible after IE8 disruption, respectively (Fig. 3a). Importantly, a significant overlap between the differentially accessible regions (n = 1,033) of both cell lines was observed.
Despite changes in chromatin accessibility being detected across the genome, we examined whether they were also specifically enriched around IE8. We explored the differentially accessible regions located on chromosome 11- where our region of interest is located – as well as in different width windows around IE8 (± 10MB to ± 0.5MB). Significant enrichment in the number of differentially accessible regions was observed in the IE8 genome vicinity whereas no differences were observed across chromosome 11 (Fig S6b).
Moreover, we aimed to identify potential gene regulatory elements associated with differential chromatin accessibility reprogramming after IE8 disruption. The relative abundance of promoter, enhancer, and insulator elements that exhibited differential accessibility after IE8 disruption was similar between MDA-MB-231 and MDA-MB-436 (Figure S6c). We focused on changes in chromatin accessibility around (± 2 kb) the gene transcription start sites (TSS). As expected, the resulting heatmap displayed a similar accessibility profile on all conditions with increased peak density on the TSS (Fig. 3b). Similar profiles were also observed between all conditions when peaks were centered in insulators and enhancers (Figure S6d). However, we found interesting differences when we focused on the TSS of the genes located at the Chr11q22.2. Promoters in the 5’MMP region were more accessible upon IE8 disruption, but no changes were observed in promoter regions located toward the 3’MMP region (Fig. 3b and Fig. 3c). In this regard, we also found changes in the accessibility of enhancer elements in both 5’ and 3’ MMP regions (Figure S6e). We additionally identified a decrease in chromatin accessibility at the IE8 CTCF binding site disrupted by CRISPR/Cas9 (Fig. 3d). In agreement with a higher CTCF occupancy detected by CUT&RUN (Fig. 2d), the decrease in chromatin accessibility of this site was more evident in the MDA-MB-231 cells than in the MDA-MB-436 cells.
IE8 disruption modulates regional MMP expression patterns
We determined whether the observed alterations in chromatin accessibility resulted in differential expression of the MMP genes nearby IE8. We assessed the mRNA expression levels of MMP genes in the wild-type and IE8-disrupted cells through RNA-seq. We observed 237 mRNAs with a significant differential expression upon IE8 impairment (Fig. 4a). We performed a gene ontology (GO) analysis to classify the 166 significantly upregulated and 71 downregulated genes by biological process. Significant pathways, such as extracellular matrix organization or Ca2+-dependent cell-cell adhesion were found significantly modulated (Figure S7a). Since the CRISPR/Cas9-mediated IE8 disruption occurred in chromosome 11, we assessed whether significant changes were enriched on this chromosome. We did not observe significant variation in gene expression when considering all the genes, but the genes located 1Mb around IE8 were significantly modulated after IE8 disruption in MDA-MB-231 (Fig. 4b).
Regarding the Chr11q22.2 encoded genes, the modification of regional gene expression patterns supports the promoter accessibility changes (Fig. 3c, Figure S6d). RNA-seq revealed two interesting changes at the mRNA level, an increase in MMP8 and a decrease in MMP1 (Fig. 4c) after IE8 disruption. These alterations were confirmed by qPCR (Fig. 4d). MDA-MB-231 showed a shift between MMP1 and MMP8 after the IE8 disruption, exhibiting a decrease in the pro-invasive enzyme MMP1 and an increase in MMP8, associated with antitumor activity. Although variations were not statistically significant, RNA expression levels displayed a similar tendency in MDA-MB-436 (Fig. 4d). Thus, the ratio between MMP1 and MMP8 is decreased after IE8 disruption, resembling the profile observed in healthy breast samples as opposed to tumor samples (Figure S7b).
We explored whether dysregulation of the MMPs located in the genomic vicinity of IE8 triggered any compensatory mechanism either modulating the expression of tissue inhibitor of metalloproteinases (TIMP) genes, a four-member family that balances the MMP activity or altering the RNA levels of two other relevant metalloproteinases, MMP2 and MMP9. TIMP1, TIMP2, TIMP3, and TIMP4 did not show significant changes at the mRNA level upon IE8 disruption (Figure S7c). IE8 depletion was not associated with changes in chromatin accessibility at MMP2 or MMP9 promoter regions. However, an important increase in accessibility was reported on enhancer elements located 8kb and 15kb upstream of TSS of the MMP2 and MMP9 genes, respectively (Fig. 4e). These changes were translated into a cell line-dependent increase in MMP mRNA levels. MMP2 was upregulated only in MDA-MB-231 whereas MMP9 was increased in MDA-MB-436 after IE8-disruption (Fig. 4f).
We considered whether changes in chromatin accessibility and the concomitant modulation of gene expression may also be observed in TNBC patient samples. We explored the six TCGA TNBC samples with ATAC-seq and mRNA-expression data. We found different levels of chromatin accessibility at IE8 (Figure S8a). Interestingly, we could associate these changes to variations in 3’MMP/5’MMP expression. We observed that those patient-derived samples with higher levels of accessibility at IE8 showed a higher ratio of 3’MMP vs 5’ MMP genes (Figure S8b)
IE8 disruption interferes with MMP1 release and decreases invasive potential in breast cancer.
We observed that IE8 disruption triggered a significant decrease in MMP1 protein levels both in MDA-MB-231 and MDA-MB-436 (Fig. 5a). We checked the functional consequences of IE8 disruption on relevant features of cancer cells. We did not observe differences either in cell proliferation (Fig. 5b) or in clonogenic ability (Figure S9a). Migration capacity assessed by wound healing assay showed no changes between wild-type and IE8 disruption conditions (Fig. 5c, Figure S9b). However, the invasion competence interrogated through 3D spheroid invasion assay was significantly reduced in IE8-disrupted MDA-MB-231 cells, but no differences were observed in the IE8-disrupted MDA-MB-436 cells (Fig. 5d). However, when we employed collagen I – which is a major component of ECM and the breast basement membrane [34]- we observed a significant decrease in the number of invasive cells after IE8 disruption in both the MDA-MB-231 and MDA-MB-436 cells (Fig. 5e).
Therefore, we explored whether the ratios between pro-invasive and antitumorigenic MMPs are linked to the progression of ductal carcinomas in situ (DCIS) to invasive ductal carcinoma (IDC) regardless of the breast cancer subtype. We compared clinical specimens of normal breast tissue, pure DCIS, and IDC from different TNM stages. The MMP (3’MMP/5’MMP) ratio was significantly higher in DCIS and IDC when compared to normal breast tissue (PDCIS<0.001, PIDC<0.001), (Figure S9c). Importantly, DCIS that eventually progressed to invasive disease displayed a significantly higher MMP ratio than those that did not progress (Figure S9d). Remarkably when considering the ratio between the pro-invasive MMP1 and the antimetastatic MMP8, the differences in DCIS that progressed to invasive disease were even more pronounced (P = 0.002, Fig. 5e). There were no changes between IDC stratified by tumor size. High MMP ratios were also associated with shorter relapse-free survival in patients with DCIS (p-value < 0.001, Fig. 5f and Figure S9e). Importantly, MMP ratios significantly predicted which DCIS patients will progress to invasive disease (AUC = 0.67–0.77, Fig. 5g, Figure S9f). Importantly, in an independent cohort of DCIS patients [31] we found that MMP1 is significantly upregulated in DCIS that progressed ipsilateral breast event (Figure S9g). Altogether, our results suggest that IE8 disruption diminishes invasiveness in the presence of collagen I and that MMP gene expression reprogramming significantly predicts the progression of DCIS to invasive disease.