Skip to main content
Erschienen in: Molecular Cancer 1/2017

Open Access 01.12.2017 | Research

SOX2 regulates multiple malignant processes of breast cancer development through the SOX2/miR-181a-5p, miR-30e-5p/TUSC3 axis

verfasst von: Kuancan Liu, Fuan Xie, Anding Gao, Rui Zhang, Long Zhang, Zhangwu Xiao, Qiong Hu, Weifeng Huang, Qiaojia Huang, Baoshun Lin, Jian Zhu, Haikun Wang, Jianwen Que, Xiaopeng Lan

Erschienen in: Molecular Cancer | Ausgabe 1/2017

Abstract

Background

High levels of SOX2 protein are correlated with increased dissemination of breast cancer. However, the underlying molecular mechanisms are not fully understood.

Methods

In this study we investigate the role of SOX2 in breast cancer metastasis using multiple in vitro and in vivo assays including cell culture, shRNA-mediated knockdown, wound healing, colony formation, transwell chamber, xenograft and tail vein injection. Moreover, western blot, immunostaining, microarray and real-time PCR were used to determine the change of protein and miRNA levels. Luciferase assays were also used to evaluate activity which TUSC3 is a target of miR-181a-5p and miR-30e-5p, and the clinical survival relevance was analyzed by Kaplan-Meier analysis.

Results

We identified a novel pathway involving SOX2 regulation of microRNAs to control the proliferation and migration of breast cancer cells. shRNA-mediated knockdown of SOX2 inhibits breast cancer cell expansion and migration. More importantly, we found that these changes are accompanied by significant reduction in the levels of two microRNAs, miR-181a-5p and miR-30e-5p. Overexpression of these two microRNAs leads to reduced protein levels of Tumor Suppressor Candidate 3 (TUSC3) in breast cancer cells; mutations of the potential binding sites in the 3’-UTR of TUSC3 abrogate the inhibitory effects of the microRNAs. We further found that upregulation of TUSC3 expression leads to reduced proliferation and migration of breast cancer cells. In human breast cancer samples the levels of TUSC3 protein are inversely correlated with those of SOX2 protein.

Conclusions

Taken together, our work reveals a novel SOX2-mediated regulatory axis that plays critical roles in the proliferation, migration and invasiveness of breast cancer cells. Targeting this axis may provide beneficial effect in the treatment of breast cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12943-017-0632-9) contains supplementary material, which is available to authorized users.
Abkürzungen
CCK-8
Cell Counting Kit 8
ER
Endoplasmic reticulum
ESCC
Esophageal squamous cell carcinomas
GO
Gene Ontology
HE
Haematoxylin and eosin
miRNAs
MicroRNAs
OST
Oligosaccharyltransferase
SAM
Significance Analysis of Microarrays
SD
Standard deviation
SOX
Sex determining region Y (SRY)-like box
SOX2-Kd
SOX2-knockdwon
TUSC3
Tumor Suppressor Candidate 3
UPR
Unfolded protein response

Background

Breast cancer is a leading cause of cancer-related death in women worldwide. Despite the significant progress that has been made in cancer treatment and early detection, approximately 30 to 70% of breast cancer patients die from breast cancer recurrence or metastasis [1]. Therefore it is important to investigate the mechanisms underlying cancer cell proliferation and metastasis-related cellular activities such as cell migration and invasion. Genetic mutations or epigenetic modifications that provide growth advantage are critical for tumour initiation and progression. In breast cancer, activation mutations of genes such as EGFR, HER2 have been found to transmit signals into the nucleus to up-regulate the transcription of downstream genes, promoting cancer cell growth [24]. However, little is known about the mediators in the nucleus that are involved in gene regulation. Interestingly, the transcription factor SOX2 has recently been associated with breast cancer formation and metastasis [5]. SOX2 belongs to the sex determining region Y (SRY)-like box (SOX) gene family, members of which are pivotal for normal development and maintenance of stemness [611], cell proliferation and differentiation [12]. Emerging studies demonstrated that the SOX2 protein is also involved in tumourigenesis [1315]. High levels of SOX2 closely correlate with multiple processes during tumour development, including initiation [16], maintenance [17, 18], invasion and metastasis [1921]. Accordingly, aberrant SOX2 expression was detected in multiple types of cancers at different stages. We and others have shown high levels of SOX2 in lung cancer, esophageal squamous cell carcinomas (ESCC) and ovarian cancer [13, 2226]. Interestingly, high levels of SOX2 proteins have also been found in 19% of breast cancer patients [5]. Moreover, SOX2 expression has been linked to tamoxifen resistance and relapse in breast cancer treatment [27]. Nevertheless, the mechanisms by which high levels of SOX2 regulate the progression and metastasis of breast cancer remain largely unexplored.
MicroRNAs (miRNAs) are small, noncoding RNAs that are important for the regulation of gene expressions at the post-transcriptional level. miRNAs are involved in almost all key cellular activities such as proliferation, differentiation and migration [28]. Dysregulation of miRNAs is also involved in the initiation and progression of human cancers including breast cancer [2931]. For example, miR-124 is down-regulated in breast cancer cell lines. Ectopic expression of miR-124 reduces the protein levels of Myc and phospho-Rb while up-regulating p27 in the breast cancer cell line MDA-MB-231 [32], suggesting that miRNAs function as a tumour suppressor. In this study we examined the molecular mechanisms used by SOX2 in the proliferation and migration of breast cancer cell lines and identified that SOX2 regulates two miRNAs (miR-181a-5p and miR30e-5p) which in turn influence the expression of a common downstream target TUSC3. Importantly, inversed expression of SOX2 and TUSC3 is associated with poor prognosis of a subpopulation of breast cancer patients. Therefore we demonstrate here a clinically relevant axis which involves SOX2/miRNAs/TUSC3 in breast cancer development.

Methods

Cell lines and mice

ZR7530 (Cat NO: CRL-1504™) and MDA-MB-231 (Cat NO: HTB-26™) breast cancer cell lines were purchased from ATCC (Manassas, VA, USA), Cell line authentication was conducted by short tandem repeat analysis, and mycoplasma contamination in cell lines was tested using multiplex PCR. They were infected with lentivirus carrying pLK0.1SOX2 (SOX2 knockdown plasmid), pCDH-SOX2-IRES-GFP (SOX2 overexpression plasmid), pLK0.1Scramble (control plasmid) [33], and pCDH-TUSC3-IRES-GFP (TUSC3 overexpression plasmid, primers used for construction were shown in Additional file 1: Table S1). Puromycin (Santa Cruz Biotechnology, Cat No: sc-108071, Dallas, TX, USA) selection was used to establish cell lines stably expressing these constructs. Cell lines were cultured in Dulbecco’s Modified Eagle Medium (DMEM, HyClone, Cat No: SH30022.01B, Beijing, China) with 10% foetal bovine serum (FBS, Gibco, Cat No:10270-106, Grand Island, NY, USA). Nude mice at 6-weeks old were purchased from SLRC Laboratory Company (Shanghai, China).

Western blot analysis

Western blot was performed as previously described [34]. PVDF membrane was used for protein transfer and probed with antibodies against human SOX2 (polyclonal rabbit anti-SOX2, SEVEN HILLS, Cat No: WRAB-1236, Cincinnati, USA), human β-actin (mouse anti-β-actin, Beyotime, Cat No: AA128, Nantong, China), CDK4 (BBI, Cat No: AB20396b, Shanghai, China), CDK6 (BBI, Cat No: AB20398a, Shanghai, China), CCND1 (BBI, Cat No: AB60236b, Shanghai, China), TUSC3 (Novus Biologicals, Cat No: NBP1-55630, Littleton, USA). HRP-conjugated goat anti-rabbit IgG (Abcam, Cat No: ab136817, Cambridge, MA, USA) and HRP-conjugated goat anti-mouse IgG (ZSGB-Bio, Cat No: ZB-2305, Beijing, China) were used as secondary antibodies to detect the proteins. The density of protein band was quantified with Image-Pro Plus 6.0 software, and the ratio of target protein to housekeeping protein, which reflects the change of expression level, was calculated.

Cell proliferation and colony formation assays

ZR7530 or MDA-MB-231 cells with genetic manipulation (knockdown or overexpression) were seeded in 96-well plates. The OD value of cells was measured under 450 nm and 630 nm at 24, 48, 72 and 96 h using the Cell Counting Kit 8 (CCK-8) (Beyotime) assay, and the absorbance difference between 450 nm and 630 nm represents cell proliferation rates. For soft agar colony formation assays, ZR7530 or MDA-MB-231 cells were seeded in 6-well plates and maintained for 2 weeks and colonies were fixed with 4% paraformaldehyde (DingGuo, Cat No: AR-0211, Beijing, China) and stained with 0.5% crystal violet. The number of total colonies and colonies with a diameter greater than 0.5 mm were calculated with Image J (Fiji-win32 software, University of Wisconsin-Madison), and the data were analysed as previously described [35, 36].

Wound healing assay

1 × 106 ZR7530 or MDA-MB-231 cells (SOX2 knockdown, scramble and TUSC3 overexpression) were seeded in 6-well plates and grown to 90% confluence. Cell monolayers were scraped with a sterile pipette tip. Floating cells were removed and the cultures were maintained in DMEM supplemented with 5% FBS. The wound area was recorded 0 h and 48 h after the scrape. The healing index was calculated and analysed using the formula: (S0 − Sn)/S0 × 100%, which represents the ability of cellular migration. S0 represents the blank area 0 h after scraping, and Sn represents the blank area n h after scraping [37].

Matrigel invasion assay

Invasion assays were performed in chambers with a 6.5-mm insert in 24-well plates (Corning company, Cat No: 3422, Corning, NY, USA). An 8-μm polycarbonate membrane was coated with 12 μl of ice-cold Matrigel™ Basement Membrane Matrix (BD company, Cat No: 356234, Bedford, MA, USA). Cells (5x104 per well) were added to the upper chamber in 200 μl of the medium containing 5% FBS. The lower chamber was filled with 500 μl of medium containing 20% FBS. The culture was maintained for 24 h. Cells were then fixed in methanol and stained with 1% crystal violet. Cells at the lower side of the filter were counted under a light microscope and dissolved with 50% acetic acid.

Xenograft studies

1x105 ZR7530 cells were re-suspended in 200 μl of DMEM and subcutaneously injected into the flanks of 6-week old BALB/c nude mice, which were maintained in the SPF environment. Five female nude mice per cohort (SOX2 knockdown and control) were randomly divided to receive cancer cells. Tumour size was measured every 3 days using a Vernier calliper after injection, and tumour volumes were calculated with the formula 0.52 × width2 × length [38, 39]. Tumours were collected 7 weeks after cell inoculation.
For lung metastatic experiments, 2 × 106 tumour cells were re-suspended in 100 μl of PBS and injected into BALB/c nude mice via the tail vein. Five female nude mice per cohort, which were also maintained in the SPF environment, received the tumour cells through tail vein. These nude mice were sacrificed 8 weeks after tail vein injection. Lungs were fixed with 4% PFA and sectioned for haematoxylin and eosin (HE) staining to determine the metastatic capabilities of cancer cells. The metastatic capabilities were assessed as previously described [40, 41].

miRNA microarray assay and real-time PCR

Total RNAs of ZR7530 cells (SOX2 knockdown and control) were extracted and purified using Trizol (Invitrogen, Carlsbad, CA, USA) and miRNeasy kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. The human miRNA microarray miRCURY™ LNA Expression Array (v.18.0, Exiqon) was used to detect the level changes after SOX2 knockdown. The low intensity miRNAs were filtered, and original signal intensities were normalised using the Median Normalization Method. The miRNA data were analysed with Significance Analysis of Microarrays (SAM) version 2.1 to generate a list of miRNAs that up-regulate or down-regulate with a fold change ≥1.5 and a p-value ≤0.05. Quantitative real-time PCR analysis was used to validate the level changes of miRNA. Additional file 1: Table S2 and Table S3 describe the stem-loop RT primers and specific PCR primers used for quantitative PCR.

GO and pathway analyses

Differentially expressed miRNAs were clustered using the Cluster software, and we performed miRNA target gene predictions using the microRNA target prediction tools miRBase, TargetScan, and miRanda, and all common target genes which are regulated by two or multiple miRNAs were analyzed with cytoscape program. We also performed Gene Ontology (GO) analysis (http://​www.​geneontology.​org) of the target genes that were predicted using the three databases, and the KEGG database (http://​www.​genome.​jp/​kegg/​) was selected for pathway analysis based on the target genes of these differentially expressed miRNAs. The significance of the GO terms and Pathway was selected based on a p-value cut-off of 0.05.

Luciferase reporter assays

Luciferase reporter activities were determined using the Dual-Luciferase Reporter Assay System (Promega, Cat No. E1910, Madison, USA). The primers used for obtaining mutated 3’UTR of TUSC3 were described in Additional file 1: Table S4. pGL3 with wild-type or mutated 3’UTR of TUSC3, miRNA mimics and pRL-TK were co-transfected into cells for all luciferase assays, and Renilla luciferase activities were measured to assess the transfection efficiency, sequences of miRNA mimics, inhibitors and relevant controls were shown in Additional file 1: Table S5.

Statistical analysis

The data represent at least three independent experiments using cells or extracts from a minimum of three separate isolations. Differences between groups were compared using analysis of variance for repeated measures. All statistical analyses were performed using GraphPad PRISM. v5.0 software (San Diego, CA, USA). Data were presented as mean ± SD (standard deviation). Statistical significance between two groups was calculated by unpaired student’s t-test, and P value < 0.05 is considered significant.

Results

SOX2 knockdown leads to reduced proliferation and migration of breast cancer cells

The breast cancer cells ZR-7530 and MDA-MB-231 express high levels of SOX2 protein. To test whether SOX2 is required for the proliferation of these cell lines, we performed lentivirus-delivered SOX2 shRNA knockdown in ZR-7530 as previously described [33]. Knockdown leads to up to 85–92% reduction in the levels of SOX2 protein (p < 0.01 for ZR-7530 and p < 0.001 for MDA-MB-231) (Fig. 1a). More importantly, decreased protein levels are associated with decreased proliferation of cancer cells, especially 96 h after knockdown (p < 0.001 for ZR-7530 and p < 0.001 for MDA-MB-231) (Fig. 1b). The decrease in cell proliferation is correlated with reduced levels of cell cycle protein CCND1, CDK4 and CDK6 (Fig. 1d). Similarly, SOX2 knockdown inhibits the proliferation of MDA-MB-231 cells (Fig. 1b right). We also performed colony forming assay to test whether SOX2 knockdown affects colony formation from single cells. Reduced levels of SOX2 affect the number and size of colonies. The number of colonies with a diameter greater than 0.5 mm formed from ZR7530 knockdown cells was significantly reduced in the SOX2-knockdwon (SOX2-Kd) group compared to scramble controls (P < 0.05) (Fig. 1c). Similarly, SOX2 knockdown in MDA-MB-231 cells also significantly reduced the number of colonies with diameters greater than 0.5 mm (P < 0.05). Together these data confirm that SOX2 is required for the proliferation of breast cancer cells.
High levels of SOX2 have been found in metastasized breast cancer nodules [42]. We next asked whether SOX2 is involved in cellular activities (e.g. cell migration) relevant to metastasis. SOX2 knockdown results in a decrease in the migration capability of ZR7530 and MDA-MB-231 cells (Fig. 2a). Wound-healing assay showed that the healing index was reduced from 24.75% (controls) to 3.33% for ZR7530 cells (p < 0.001), and from 57.39 to 43.8% for MDA-MB-231 cells (p < 0.05) (Fig. 2b). In addition, SOX2 knockdown inhibits the invasion of breast cancer cells in transwell assay. The number of cancer cells passing through the filter of the transwell are significantly reduced after SOX2 knockdown (Fig. 2c, d, p < 0.01 for ZR7530 and p < 0.05 for MDA-MB-231). The changed tendency of the OD value of eluted crystal violet was consistent with the number of migrated cells in these groups (p < 0.01 for ZR7530 and p < 0.05 for MDA-MB-231) (Fig. 2e). Together, these in vitro results demonstrate that high levels of SOX2 protein are critical for migration and invasion of breast cancer cells.

SOX2 level changes are correlated with tumour development and metastasis in murine xenograft models

We next used a xenograft model to test whether SOX2 knockdown reduces tumour growth in vivo. SOX2 knockdown ZR7530 and control cells were transplanted into immunocompromised nude mice. Significantly, SOX2 knockdown leads to 4.4 and 4.7 fold decreases in the volume and weight of tumour, respectively (Fig. 3ac) (n = 5). In addition, SOX2 overexpression enables higher tumourigenesis capability of breast cancer cells. We transplanted SOX2-overexpressing ZR7530 cells into nude mice, and SOX2 overexpression results in a 2.3 fold increase in tumour volume and a 3.6 fold increase in tumour weight (Fig. 3ac) (n = 5). These results suggest that the levels of SOX2 are correlated with increased growth capability of breast cancer cells in vivo.
Our in vitro results show that SOX2 knockdown reduces the migration of breast cancer cell lines. To test whether decreased levels of SOX2 also block metastasis capability in vivo, we performed tail vein injection of ZR7530 and studied tumour seeding and development in the lung. Upon SOX2 knockdown, the number of tumours seeded in the lung are significantly reduced (33 ± 2 Vs 7 ± 3) (p < 0.001) (n = 5) (Fig. 3d, e). In addition, SOX2 knockdown also reduces the ratio of tumour/total lung area (10.4% Vs 1.8%) (p < 0.001) (Fig. 3f). By contrast, ectopic SOX2 expression enables high metastastic capability of ZR7530 cells, leading to an increased number of seeding tumours (33 ± 2 Vs 51 ± 4) (p < 0.01) and enlarged metastatic foci in the lung (10.4% vs 27.5%) (p < 0.001) (Fig. 3df). Together, these in vivo findings support the important roles of SOX2 in promoting metastasis of breast cancer cells.

SOX2 knockdown leads to decreased levels of miR-181a-5p and miR-30e-5p

miRNAs have been implicated in the regulation of multiple steps in breast cancer progression [4345]. To test whether SOX2 regulates proliferation and metastasis through miRNAs, we performed microRNA microarray analysis (miRCURY™ LNA Expression Array v.18.0, Exiqon). Total RNAs were isolated from scramble control and SOX2 knockdown ZR7530 cells. SOX2 knockdown significantly changed the levels of multiple miRNAs, among which 11 were up-regulated and 10 were significantly down-regulated (Fig. 4a and Additional file 1: Table S6, p < 0.05). We validated the expression levels with real-time PCR (Fig. 4b). Although the levels of miR-338-3p are significantly down-regulated in the array analysis, the levels of miR-30e-5p are most dramatically down-regulated upon SOX2 knockdown, which was validated by real-time PCR (Fig. 4b). Notably, the levels of miR-181a-5p which has previously been associated with breast cancer metastasis are also down-regulated (p < 0.001) (Fig. 4a, b) [46]. Down-regulation of miR-30e-5p and miR-181a-5p was also observed in MDA-MB-231 cells after SOX2 knockdown (data not shown).

miR-181a-5p and miR-30e-5p co-regulate TUSC3 in breast cancer cells

Several miRNAs down-regulated upon SOX2 knockdown, have been implicated in the regulation of genes involved in metabolism, signalling pathway activation (Fig. 4c, d). Intriguingly, miR-181a-5p and miR-30e-5p share a common downstream target, TUSC3 as identified by the cytoscape program. TUSC3 is a putative tumor suppressor gene, and hypermethylation or loss of this gene has been found in cancers such as ovarian and breast cancer [4749]. Intriguingly, two potential binding sites of miR-181a-5p and miR-30e-5p were identified at the 3’-UTR of TUSC3 (Fig. 5a). Consistently, transfection of miR-181a-5p or miR-30e-5p mimics reduces the protein levels of TUSC3 in ZR7530 cells (Fig. 5b). Conversely, inhibition of these two miRNAs with specific inhibitors leads to increased protein levels of TUSC3 (Fig. 5c). We then cloned the 3’-UTR of TUSC3 containing the potential miRNA binding sites into the luciferase reporter vectors and tested whether these two miRNAs regulate TUSC3 through the potential binding sites (Fig. 5d, Additional file 2: Figure S1). Interestingly, co-transfection of miR-181a-5p or miR-30e-5p mimics reduces the luciferase activities to 47 and 55.4% of control, respectively (Fig. 5e). More importantly, mutation of these potential binding sites leads to attenuated luciferase activities, suggesting that miR-181a-5p or miR-30e-5p controls TUSC3 expression through the binding sites. As expected, SOX2 knockdown leads to increased protein levels of TUSC3 protein (Fig. 5f). Moreover, in the xenografts of SOX2 knockdown group the transcript levels of SOX2 remain low and the level of miR-181a-5p or miR-30e-5p are also significantly lower than the control group (p < 0.05 for hsa-miR-30e-5p and p < 0.01 for hsa-miR-181a-5p), suggesting that SOX2 is required for maintaining the expression of two microRNAs in vivo. In addition, we did not see significant change of TUSC3 transcript levels, which is consistent with the finding that the two microRNA regulate translation but not transcription of TUSC3 (Fig. 5g). Taken together, these data indicate a regulatory axis where SOX2 modulates the levels of the tumor suppressor protein TUSC3 through miR-181a-5p and miR-30e-5p.

TUSC3 regulates the proliferation, migration and invasion of breast cancer cells

SOX2 knockdown leads to decreased levels of miR-181a-5p and miR-30e-5p and increased levels of TUSC3. To test whether TUSC3 acts as a negative regulator of proliferation and migration of breast cancer cells, we performed TUSC3 gain-of-function study (Fig. 6a). TUSC3 overexpression leads to decreased proliferation of ZR7530 cells (p < 0.01) (Fig. 6b). In addition, TUSC3 overexpression significantly inhibits the colony formation efficiency (274 ± 6 Vs 119 ± 4 colonies/well) in 6-well plates (p < 0.01) and reduces the size of individual colonies (p < 0.05) (Fig. 6c), suggesting that TUSC3 acts as a negative regulator of proliferation of breast cancer cells.
We next asked whether increased levels of TUSC3 are also able to reduce the migration of breast cancer cells. We first used a wound-healing assay to address the issue and observed that TUSC3 overexpression significantly reduced the healing index of ZR7530 cells as compared to control (24.3% Vs 12.4%) (p < 0.01) (Fig. 6d). We then used the transwell assay to further test the effect of TUSC3 overexpression on cell invasion. Similar to SOX2 knockdown, TUSC3 overexpression dramatically decreased the invasion capability of ZR7530 (p < 0.01) (Fig. 6e). Collectively, these results demonstrated that high levels of TUSC3 protein reduce the invasive potential of breast cancer cells.

Increased SOX2 protein levels correlate with poor survival in breast cancer patients

Previous studies revealed that approximately 16% of breast cancer patients exhibit SOX2-positive expression in clinical samples, and SOX2 is expressed in the early stage of breast tumours [17]. However, other studies revealed that SOX2 expression was associated with subtypes and tumour grades of breast cancer [5, 50]. To analyse whether Sox2 expression level (by probe 214178_s_at) correlates with distant metastasis free survival in breast cancer patients, we used the Kaplan-Meier analysis from IBM SPSS software to analyse the relationship between SOX2 protein levels and 5-year survival rates in 471 patients, and the patients data used for analysis derives from previous study [51]. As shown in Fig. 7a, SOX2 expression level significantly converts poor prognosis (Chisquare p-value = 0.014, Logrank p-value = 0.02268), and the distant metastasis-free survival of breast cancer patients expressing high, middle and low levels of SOX2 protein were 50.4, 70.5 and 78.6%, suggesting that high levels of SOX2 are correlated with poor prognosis in breast cancer patients. Further analysis revealed that patients with high and middle levels of SOX2 have a significant worse survival rate than those with low levels (Fig. 7a, b) (p < 0.05). However, the survival difference between the middle and low level groups is not significant (p > 0.05). Moreover, we found that SOX2 levels are inversely correlated with those of TUSC3. We examined 12 pairs of normal and breast cancer biopsies, and found that 9 breast cancer biopsies have increased expression of SOX2 proteins concomitant with low levels of TUSC3 (Fig. 7c). In keeping with this line, high levels of TUSC3 protein were detected in adjacent normal tissue samples but were very low in cancer biopsies (Fig. 7d).

Discussion

High levels of SOX2 proteins have been linked to breast cancer metastasis. However, the underlying mechanisms remain largely unknown. Here, we use in vitro and in vivo models to demonstrate that SOX2 is required for multiple steps in metastatic processes. We showed that SOX2 regulates the proliferation, migration and seeding of breast cancer cells. More importantly, we have identified a miRNA-mediated regulatory axis downstream of SOX2 to control these processes. Specifically, we found that SOX2 regulates the levels of miR-181a-5p and miR-30e-5p which in turn modulate the levels of the tumor suppressor TUSC3. Consistently, overexpression of TUSC3 reduces the proliferation and migration of SOX2-high breast cancer cells.
SOX2 is a key transcription factor important for the self-renewal of stem cells in multiple tissues including the trachea and brain [6]. We and others have shown that SOX2 overexpression promotes the proliferation of stem cells in the esophagus and lung, leading to malignancy [15, 33], suggesting that SOX2 acts as an oncogene. In this study, we found that SOX2 is also required for the proliferation of breast cancer cells. SOX2 knockdown leads to decreased levels of cell cycle proteins including CCND1, CDK4 and CDK6. Interestingly, SOX2 knockdown also leads to reduced levels of multiple microRNAs. We found that the levels of miR-30e-5p (previously named as miR-30e) are most dramatically down-regulated upon SOX2 knockdown, which was validated by real-time PCR. Interestingly, we also found that miR-181a-5p (previously named as miR-181a) which has previously been linked to breast cancer development is also dramatically downregulated. More importantly, further analysis using the cytoscape program we found that miR-30e-5p together with miR-181a-5p co-regulate a common downstream target, TUSC3. Both miR-30e-5p and miR-181a-5p have been implicated in the regulation of cancer cell growth and migration, invasion and metastasis [5254]. miR-181a-5p is a member of the miR-181 s family which includes three other highly conserved miRNAs, miR-181b, c, and d [55]. Previous studies have shown increased levels of miR-181a-5p in multiple types of cancer including ovarian cancer and gastric cancer [56, 57]. The levels of miR-181a-5p in breast cancer were recently assessed. While Li et al. demonstrated that miR-181a-5p is down-regulated in aggressive human breast and colon cancers [58], a recent study showed that the levels of miR-181a-5p are high in breast cancer samples [59]. By contrast, we have just begun to elicit the role of miR-30e-5p in tumour development. Tissue microarray analysis suggests that high levels of miR-30e-5p in primary tumours are associated with a favourable prognosis in breast cancer [60]. In our study, we observed increased levels of miR-181a-5p and miR-30e-5p in breast cancer biopsies as compared to normal mammary gland tissue. We found that SOX2 knockdown leads to decreased levels of miR-181a-5p and miR-30e-5p accompanied by decreased cell proliferation, suggesting that these two microRNAs are required for the proliferation of breast cancer cells.
Consistently, we identified that miR-181a-5p and miR-30e-5p target the tumor suppressor TUSC3. Overexpression of these two miRNAs reduces the protein levels of TUSC3. Moreover, the luciferase assay showed that miR-181a-5p and miR-30e-5p regulate the 3’-UTR of TUSC3 transcripts. TUSC3 is a putative tumour suppressor often lost in epithelial cancers e.g. ovarian cancer and head and neck squamous cell carcinomas [47, 61]. Promoter methylation leading to silencing of TUSC3 gene was considered as a poor prognostic factor in ovarian cancer [48]. Here, we show decreased but not complete loss of TUSC3 protein in breast cancers, suggesting that promoter methylation is unlikely a significant contributor to disease progression. We found that the protein levels of TUSC3 are decreased in breast tumour tissues as compared to surrounding normal tissues. In addition, overexpression of TUSC3 decreases the proliferation and migration capabilities of breast cancer cells. These findings are consistent with the tumor suppressor function of TUSC3 [62]. TUSC3 is an important component of the oligosaccharyltransferase (OST) complex that catalyzes N-linked glycosylation of proteins in the endoplasmic reticulum (ER) [63, 64]. Loss of TUSC3 in mammalian cells leads to accumulation of unfolded or misfolded proteins in the ER, resulting in ER stress. In ovarian cancer cells loss of TUSC3 induces ER stress and morphological alterations in the ER. Interestingly, ovarian cancer cells lacking TUSC3 seem able to alleviate the massive ER stress response through the PERK-mediated adaptive pathway [65], which is consistent with the finding that accumulated unfolded proteins in cancer cells induce the unfolded protein response (UPR), facilitating cancer cells to adapt to ER stress [66]. miR-181a-5p overexpression has been shown to promote ER stress and leads to myogenic differentiation in C2C12 cells [67]. Along these lines our study indicates that high levels of TUSC3 expression may induce a cellular response of breast cancer cells, resulting in reduced cell proliferation and migration.
Previous studies revealed that SOX2 protein levels are correlated with the overall survival of patients with multiple types of cancer including esophageal [33] and lung cancer [24]. Moreover, SOX2 protein has been assessed as a diagnostic marker and therapeutic target for multiple cancer treatments [6870]. Our results suggest that high levels of SOX2 protein are associated with poor prognosis of breast cancer.

Conclusions

In summary, we showed in this study that SOX2 protein is critical for the proliferation, migration and metastasis of breast cancer. We identified that SOX2 exerts these functions through miR-181a-5p and miR-30e-5p which share a common downstream target, the putative tumour suppressor TUSC3 (Fig. 8). Knockdown SOX2 leads to decreased levels of miR-181a-5p and miR-30e-5p accompanied by increased levels of TUSC3. Consistently, overexpression of TUSC3 suppresses the proliferation and migration of breast cancer cells. Together these findings support a regulatory axis orchestrated by high levels of SOX2, and this axis is critical for breast cancer development. In future studies it will be interesting to explore potential avenues to target this SOX2/microRNA/TUSC3 axis for therapeutic gains.

Acknowledgments

The authors are grateful for inputs from colleagues in Liu and Que laboratory.

Funding

This work was supported by the National Natural Science Foundation of China (No.81302068 to K.L), the National High Technology Research and Development Program of China (863 Program, No. 2014AA020541 to K. L), and the Program for the Top Young Innovative Talents of Fujian Province (to K. L), the National Institutes of Health (R01-DK100342 to J. Q), the New York State Stem Cell Research Program (C029555 to J. Q), and the National Key Research and Development Program (2016YFA0502500 to L.Z).

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article, and the materials will be available once request.

Authors’ contributions

KCL did the most experiments and contributed the writing of manuscript. FAX, ADG, RZ, QH, BSL conducted cell culture, western blot and qPCR experiments. KCL, JWQ and XPL analysed the data and designed the study, prepared the figures and performed the statistical analysis. LZ, ZWX, WFH, QJH, JZ and HKW planned the project and participated coordination. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
All authors read and approved the final manuscript.
We confirmed that all the protocols on human tissue examination and animal experiments were approved by the Medical Ethics Committee of Fuzhou General Hospital.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis 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.
Literatur
1.
Zurück zum Zitat Cardoso F, Harbeck N, Fallowfield L, Kyriakides S, Senkus E. Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23 Suppl 7:vii11–9.CrossRefPubMed Cardoso F, Harbeck N, Fallowfield L, Kyriakides S, Senkus E. Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23 Suppl 7:vii11–9.CrossRefPubMed
2.
Zurück zum Zitat Teng YH, Tan WJ, Thike AA, Cheok PY, Tse GM, Wong NS, Yip GW, Bay BH, Tan PH. Mutations in the epidermal growth factor receptor (EGFR) gene in triple negative breast cancer: possible implications for targeted therapy. Breast Cancer Res. 2011;13:R35.CrossRefPubMedPubMedCentral Teng YH, Tan WJ, Thike AA, Cheok PY, Tse GM, Wong NS, Yip GW, Bay BH, Tan PH. Mutations in the epidermal growth factor receptor (EGFR) gene in triple negative breast cancer: possible implications for targeted therapy. Breast Cancer Res. 2011;13:R35.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, Goel N, Aronson AB, Li S, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–37.CrossRefPubMed Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, Goel N, Aronson AB, Li S, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–37.CrossRefPubMed
4.
Zurück zum Zitat Zabransky DJ, Yankaskas CL, Cochran RL, Wong HY, Croessmann S, Chu D, Kavuri SM, Red Brewer M, Rosen DM, Dalton WB, et al. HER2 missense mutations have distinct effects on oncogenic signaling and migration. Proc Natl Acad Sci U S A. 2015;112:E6205–14.CrossRefPubMedPubMedCentral Zabransky DJ, Yankaskas CL, Cochran RL, Wong HY, Croessmann S, Chu D, Kavuri SM, Red Brewer M, Rosen DM, Dalton WB, et al. HER2 missense mutations have distinct effects on oncogenic signaling and migration. Proc Natl Acad Sci U S A. 2015;112:E6205–14.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Li X, Xu Y, Chen Y, Chen S, Jia X, Sun T, Liu Y, Xiang R, Li N. SOX2 promotes tumor metastasis by stimulating epithelial-to-mesenchymal transition via regulation of WNT/beta-catenin signal network. Cancer Lett. 2013;336:379–89.CrossRefPubMed Li X, Xu Y, Chen Y, Chen S, Jia X, Sun T, Liu Y, Xiang R, Li N. SOX2 promotes tumor metastasis by stimulating epithelial-to-mesenchymal transition via regulation of WNT/beta-catenin signal network. Cancer Lett. 2013;336:379–89.CrossRefPubMed
6.
7.
Zurück zum Zitat Li XL, Eishi Y, Bai YQ, Sakai H, Akiyama Y, Tani M, Takizawa T, Koike M, Yuasa Y. Expression of the SRY-related HMG box protein SOX2 in human gastric carcinoma. Int J Oncol. 2004;24:257–63.PubMed Li XL, Eishi Y, Bai YQ, Sakai H, Akiyama Y, Tani M, Takizawa T, Koike M, Yuasa Y. Expression of the SRY-related HMG box protein SOX2 in human gastric carcinoma. Int J Oncol. 2004;24:257–63.PubMed
8.
Zurück zum Zitat Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.CrossRefPubMed Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.CrossRefPubMed
9.
Zurück zum Zitat Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.CrossRefPubMed Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.CrossRefPubMed
10.
Zurück zum Zitat Fong H, Hohenstein KA, Donovan PJ. Regulation of self-renewal and pluripotency by Sox2 in human embryonic stem cells. Stem Cells. 2008;26:1931–8.CrossRefPubMed Fong H, Hohenstein KA, Donovan PJ. Regulation of self-renewal and pluripotency by Sox2 in human embryonic stem cells. Stem Cells. 2008;26:1931–8.CrossRefPubMed
11.
Zurück zum Zitat Wakamatsu Y, Endo Y, Osumi N, Weston JA. Multiple roles of Sox2, an HMG-box transcription factor in avian neural crest development. Dev Dyn. 2004;229:74–86.CrossRefPubMed Wakamatsu Y, Endo Y, Osumi N, Weston JA. Multiple roles of Sox2, an HMG-box transcription factor in avian neural crest development. Dev Dyn. 2004;229:74–86.CrossRefPubMed
12.
Zurück zum Zitat Schepers GE, Teasdale RD, Koopman P. Twenty pairs of sox: extent, homology, and nomenclature of the mouse and human sox transcription factor gene families. Dev Cell. 2002;3:167–70.CrossRefPubMed Schepers GE, Teasdale RD, Koopman P. Twenty pairs of sox: extent, homology, and nomenclature of the mouse and human sox transcription factor gene families. Dev Cell. 2002;3:167–70.CrossRefPubMed
13.
Zurück zum Zitat Bass AJ, Watanabe H, Mermel CH, Yu S, Perner S, Verhaak RG, Kim SY, Wardwell L, Tamayo P, Gat-Viks I, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 2009;41:1238–42.CrossRefPubMedPubMedCentral Bass AJ, Watanabe H, Mermel CH, Yu S, Perner S, Verhaak RG, Kim SY, Wardwell L, Tamayo P, Gat-Viks I, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 2009;41:1238–42.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Gen Y, Yasui K, Zen Y, Zen K, Dohi O, Endo M, Tsuji K, Wakabayashi N, Itoh Y, Naito Y, et al. SOX2 identified as a target gene for the amplification at 3q26 that is frequently detected in esophageal squamous cell carcinoma. Cancer Genet Cytogenet. 2010;202:82–93.CrossRefPubMed Gen Y, Yasui K, Zen Y, Zen K, Dohi O, Endo M, Tsuji K, Wakabayashi N, Itoh Y, Naito Y, et al. SOX2 identified as a target gene for the amplification at 3q26 that is frequently detected in esophageal squamous cell carcinoma. Cancer Genet Cytogenet. 2010;202:82–93.CrossRefPubMed
15.
Zurück zum Zitat Lu Y, Futtner C, Rock JR, Xu X, Whitworth W, Hogan BL, Onaitis MW. Evidence that SOX2 overexpression is oncogenic in the lung. Plos One. 2010;5:e11022.CrossRefPubMedPubMedCentral Lu Y, Futtner C, Rock JR, Xu X, Whitworth W, Hogan BL, Onaitis MW. Evidence that SOX2 overexpression is oncogenic in the lung. Plos One. 2010;5:e11022.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Santini R, Pietrobono S, Pandolfi S, Montagnani V, D’Amico M, Penachioni JY, Vinci MC, Borgognoni L, Stecca B. SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells. Oncogene. 2014;33:4697–708.CrossRefPubMedPubMedCentral Santini R, Pietrobono S, Pandolfi S, Montagnani V, D’Amico M, Penachioni JY, Vinci MC, Borgognoni L, Stecca B. SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells. Oncogene. 2014;33:4697–708.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Leis O, Eguiara A, Lopez-Arribillaga E, Alberdi MJ, Hernandez-Garcia S, Elorriaga K, Pandiella A, Rezola R, Martin AG. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene. 2012;31:1354–65.CrossRefPubMed Leis O, Eguiara A, Lopez-Arribillaga E, Alberdi MJ, Hernandez-Garcia S, Elorriaga K, Pandiella A, Rezola R, Martin AG. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene. 2012;31:1354–65.CrossRefPubMed
18.
Zurück zum Zitat Basu-Roy U, Seo E, Ramanathapuram L, Rapp TB, Perry JA, Orkin SH, Mansukhani A, Basilico C. Sox2 maintains self renewal of tumor-initiating cells in osteosarcomas. Oncogene. 2012;31:2270–82.CrossRefPubMed Basu-Roy U, Seo E, Ramanathapuram L, Rapp TB, Perry JA, Orkin SH, Mansukhani A, Basilico C. Sox2 maintains self renewal of tumor-initiating cells in osteosarcomas. Oncogene. 2012;31:2270–82.CrossRefPubMed
19.
Zurück zum Zitat Neumann J, Bahr F, Horst D, Kriegl L, Engel J, Luque RM, Gerhard M, Kirchner T, Jung A. SOX2 expression correlates with lymph-node metastases and distant spread in right-sided colon cancer. BMC Cancer. 2011;11:518.CrossRefPubMedPubMedCentral Neumann J, Bahr F, Horst D, Kriegl L, Engel J, Luque RM, Gerhard M, Kirchner T, Jung A. SOX2 expression correlates with lymph-node metastases and distant spread in right-sided colon cancer. BMC Cancer. 2011;11:518.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Xiang R, Liao D, Cheng T, Zhou H, Shi Q, Chuang TS, Markowitz D, Reisfeld RA, Luo Y. Downregulation of transcription factor SOX2 in cancer stem cells suppresses growth and metastasis of lung cancer. Br J Cancer. 2011;104:1410–7.CrossRefPubMedPubMedCentral Xiang R, Liao D, Cheng T, Zhou H, Shi Q, Chuang TS, Markowitz D, Reisfeld RA, Luo Y. Downregulation of transcription factor SOX2 in cancer stem cells suppresses growth and metastasis of lung cancer. Br J Cancer. 2011;104:1410–7.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Sun C, Sun L, Li Y, Kang X, Zhang S, Liu Y. Sox2 expression predicts poor survival of hepatocellular carcinoma patients and it promotes liver cancer cell invasion by activating Slug. Med Oncol. 2013;30:503.CrossRefPubMed Sun C, Sun L, Li Y, Kang X, Zhang S, Liu Y. Sox2 expression predicts poor survival of hepatocellular carcinoma patients and it promotes liver cancer cell invasion by activating Slug. Med Oncol. 2013;30:503.CrossRefPubMed
22.
Zurück zum Zitat Hussenet T, Dali S, Exinger J, Monga B, Jost B, Dembele D, Martinet N, Thibault C, Huelsken J, Brambilla E, du Manoir S. SOX2 is an oncogene activated by recurrent 3q26.3 amplifications in human lung squamous cell carcinomas. Plos One. 2010;5:e8960.CrossRefPubMedPubMedCentral Hussenet T, Dali S, Exinger J, Monga B, Jost B, Dembele D, Martinet N, Thibault C, Huelsken J, Brambilla E, du Manoir S. SOX2 is an oncogene activated by recurrent 3q26.3 amplifications in human lung squamous cell carcinomas. Plos One. 2010;5:e8960.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Mccaughan F, Pole JC, Bankier AT, Konfortov BA, Carroll B, Falzon M, Rabbitts TH, George PJ, Dear PH, Rabbitts PH. Progressive 3q amplification consistently targets SOX2 in preinvasive squamous lung cancer. Am J Respir Crit Care Med. 2010;182:83–91.CrossRefPubMedPubMedCentral Mccaughan F, Pole JC, Bankier AT, Konfortov BA, Carroll B, Falzon M, Rabbitts TH, George PJ, Dear PH, Rabbitts PH. Progressive 3q amplification consistently targets SOX2 in preinvasive squamous lung cancer. Am J Respir Crit Care Med. 2010;182:83–91.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Wilbertz T, Wagner P, Petersen K, Stiedl AC, Scheble VJ, Maier S, Reischl M, Mikut R, Altorki NK, Moch H, et al. SOX2 gene amplification and protein overexpression are associated with better outcome in squamous cell lung cancer. Mod Pathol. 2011;24:944–53.CrossRefPubMed Wilbertz T, Wagner P, Petersen K, Stiedl AC, Scheble VJ, Maier S, Reischl M, Mikut R, Altorki NK, Moch H, et al. SOX2 gene amplification and protein overexpression are associated with better outcome in squamous cell lung cancer. Mod Pathol. 2011;24:944–53.CrossRefPubMed
25.
Zurück zum Zitat Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, Bergbower EA, Guan Y, Shin J, Guillory J, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet. 2012;44:1111–6.CrossRefPubMedPubMedCentral Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, Bergbower EA, Guan Y, Shin J, Guillory J, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet. 2012;44:1111–6.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Belotte J, Fletcher NM, Alexis M, Morris RT, Munkarah AR, Diamond MP, Saed GM. Sox2 gene amplification significantly impacts overall survival in serous epithelial ovarian cancer. Reprod Sci. 2015;22:38–46.CrossRefPubMedPubMedCentral Belotte J, Fletcher NM, Alexis M, Morris RT, Munkarah AR, Diamond MP, Saed GM. Sox2 gene amplification significantly impacts overall survival in serous epithelial ovarian cancer. Reprod Sci. 2015;22:38–46.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Piva M, Domenici G, Iriondo O, Rabano M, Simoes BM, Comaills V, Barredo I, Lopez-Ruiz JA, Zabalza I, Kypta R, Vivanco M. Sox2 promotes tamoxifen resistance in breast cancer cells. EMBO Mol Med. 2014;6:66–79.CrossRefPubMed Piva M, Domenici G, Iriondo O, Rabano M, Simoes BM, Comaills V, Barredo I, Lopez-Ruiz JA, Zabalza I, Kypta R, Vivanco M. Sox2 promotes tamoxifen resistance in breast cancer cells. EMBO Mol Med. 2014;6:66–79.CrossRefPubMed
28.
29.
Zurück zum Zitat Mulrane L, Mcgee SF, Gallagher WM, O’Connor DP. miRNA dysregulation in breast cancer. Cancer Res. 2013;73:6554–62.CrossRefPubMed Mulrane L, Mcgee SF, Gallagher WM, O’Connor DP. miRNA dysregulation in breast cancer. Cancer Res. 2013;73:6554–62.CrossRefPubMed
30.
Zurück zum Zitat Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, Li C, Chong M, Ibrahim T, Mercatali L, et al. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15:284–94.CrossRefPubMedPubMedCentral Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, Li C, Chong M, Ibrahim T, Mercatali L, et al. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15:284–94.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Hu J, Sun T, Wang H, Chen Z, Wang S, Yuan L, Liu T, Li HR, Wang P, Feng Y, et al. MiR-215 Is Induced Post-transcriptionally via HIF-Drosha Complex and Mediates Glioma-Initiating Cell Adaptation to Hypoxia by Targeting KDM1B. Cancer Cell. 2016;29:49–60.CrossRefPubMedPubMedCentral Hu J, Sun T, Wang H, Chen Z, Wang S, Yuan L, Liu T, Li HR, Wang P, Feng Y, et al. MiR-215 Is Induced Post-transcriptionally via HIF-Drosha Complex and Mediates Glioma-Initiating Cell Adaptation to Hypoxia by Targeting KDM1B. Cancer Cell. 2016;29:49–60.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Seviour EG, Sehgal V, Lu Y, Luo Z, Moss T, Zhang F, Hill SM, Liu W, Maiti SN, Cooper L, et al. Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene. 2016;35:691–701.CrossRefPubMed Seviour EG, Sehgal V, Lu Y, Luo Z, Moss T, Zhang F, Hill SM, Liu W, Maiti SN, Cooper L, et al. Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene. 2016;35:691–701.CrossRefPubMed
33.
Zurück zum Zitat Liu K, Jiang M, Lu Y, Chen H, Sun J, Wu S, Ku WY, Nakagawa H, Kita Y, Natsugoe S, et al. Sox2 cooperates with inflammation-mediated Stat3 activation in the malignant transformation of foregut basal progenitor cells. Cell Stem Cell. 2013;12:304–15.CrossRefPubMedPubMedCentral Liu K, Jiang M, Lu Y, Chen H, Sun J, Wu S, Ku WY, Nakagawa H, Kita Y, Natsugoe S, et al. Sox2 cooperates with inflammation-mediated Stat3 activation in the malignant transformation of foregut basal progenitor cells. Cell Stem Cell. 2013;12:304–15.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Jiang M, Ku WY, Zhou Z, Dellon ES, Falk GW, Nakagawa H, Wang ML, Liu K, Wang J, Katzka DA, et al. BMP-driven NRF2 activation in esophageal basal cell differentiation and eosinophilic esophagitis. J Clin Invest. 2015;125:1557–68.CrossRefPubMedPubMedCentral Jiang M, Ku WY, Zhou Z, Dellon ES, Falk GW, Nakagawa H, Wang ML, Liu K, Wang J, Katzka DA, et al. BMP-driven NRF2 activation in esophageal basal cell differentiation and eosinophilic esophagitis. J Clin Invest. 2015;125:1557–68.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Ikezoe T, Gery S, Yin D, O’Kelly J, Binderup L, Lemp N, Taguchi H, Koeffler HP. CCAAT/enhancer-binding protein delta: a molecular target of 1,25-dihydroxyvitamin D3 in androgen-responsive prostate cancer LNCaP cells. Cancer Res. 2005;65:4762–8.CrossRefPubMed Ikezoe T, Gery S, Yin D, O’Kelly J, Binderup L, Lemp N, Taguchi H, Koeffler HP. CCAAT/enhancer-binding protein delta: a molecular target of 1,25-dihydroxyvitamin D3 in androgen-responsive prostate cancer LNCaP cells. Cancer Res. 2005;65:4762–8.CrossRefPubMed
36.
Zurück zum Zitat Chang PH, Hwang-Verslues WW, Chang YC, Chen CC, Hsiao M, Jeng YM, Chang KJ, Lee EY, Shew JY, Lee WH. Activation of Robo1 signaling of breast cancer cells by Slit2 from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/beta-catenin pathway. Cancer Res. 2012;72:4652–61.CrossRefPubMedPubMedCentral Chang PH, Hwang-Verslues WW, Chang YC, Chen CC, Hsiao M, Jeng YM, Chang KJ, Lee EY, Shew JY, Lee WH. Activation of Robo1 signaling of breast cancer cells by Slit2 from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/beta-catenin pathway. Cancer Res. 2012;72:4652–61.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Kliment CR, Englert JM, Gochuico BR, Yu G, Kaminski N, Rosas I, Oury TD. Oxidative stress alters syndecan-1 distribution in lungs with pulmonary fibrosis. J Biol Chem. 2009;284:3537–45.CrossRefPubMedPubMedCentral Kliment CR, Englert JM, Gochuico BR, Yu G, Kaminski N, Rosas I, Oury TD. Oxidative stress alters syndecan-1 distribution in lungs with pulmonary fibrosis. J Biol Chem. 2009;284:3537–45.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Basile KJ, Abel EV, Dadpey N, Hartsough EJ, Fortina P, Aplin AE. In vivo MAPK reporting reveals the heterogeneity in tumoral selection of resistance to RAF inhibitors. Cancer Res. 2013;73:7101–10.CrossRefPubMed Basile KJ, Abel EV, Dadpey N, Hartsough EJ, Fortina P, Aplin AE. In vivo MAPK reporting reveals the heterogeneity in tumoral selection of resistance to RAF inhibitors. Cancer Res. 2013;73:7101–10.CrossRefPubMed
39.
Zurück zum Zitat Veitonmaki N, Hansson M, Zhan F, Sundberg A, Lofstedt T, Ljungars A, Li ZC, Martinsson-Niskanen T, Zeng M, Yang Y, et al. A human ICAM-1 antibody isolated by a function-first approach has potent macrophage-dependent antimyeloma activity in vivo. Cancer Cell. 2013;23:502–15.CrossRefPubMed Veitonmaki N, Hansson M, Zhan F, Sundberg A, Lofstedt T, Ljungars A, Li ZC, Martinsson-Niskanen T, Zeng M, Yang Y, et al. A human ICAM-1 antibody isolated by a function-first approach has potent macrophage-dependent antimyeloma activity in vivo. Cancer Cell. 2013;23:502–15.CrossRefPubMed
40.
Zurück zum Zitat Cooke VG, Lebleu VS, Keskin D, Khan Z, O’Connell JT, Teng Y, Duncan MB, Xie L, Maeda G, Vong S, et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell. 2012;21:66–81.CrossRefPubMedPubMedCentral Cooke VG, Lebleu VS, Keskin D, Khan Z, O’Connell JT, Teng Y, Duncan MB, Xie L, Maeda G, Vong S, et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell. 2012;21:66–81.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Lebleu VS, O’Connell JT, Gonzalez Herrera KN, Wikman H, Pantel K, Haigis MC, de Carvalho FM, Damascena A, Domingos Chinen LT, Rocha RM, et al. PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol. 2014;16:992–1003. 1001–1015.CrossRefPubMedPubMedCentral Lebleu VS, O’Connell JT, Gonzalez Herrera KN, Wikman H, Pantel K, Haigis MC, de Carvalho FM, Damascena A, Domingos Chinen LT, Rocha RM, et al. PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol. 2014;16:992–1003. 1001–1015.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Huang YH, Luo MH, Ni YB, Tsang JY, Chan SK, Lui PC, Yu AM, Tan PH, Tse GM. Increased SOX2 expression in less differentiated breast carcinomas and their lymph node metastases. Histopathology. 2014;64:494–503.CrossRefPubMed Huang YH, Luo MH, Ni YB, Tsang JY, Chan SK, Lui PC, Yu AM, Tan PH, Tse GM. Increased SOX2 expression in less differentiated breast carcinomas and their lymph node metastases. Histopathology. 2014;64:494–503.CrossRefPubMed
44.
Zurück zum Zitat Bockhorn J, Yee K, Chang YF, Prat A, Huo D, Nwachukwu C, Dalton R, Huang S, Swanson KE, Perou CM, et al. MicroRNA-30c targets cytoskeleton genes involved in breast cancer cell invasion. Breast Cancer Res Treat. 2013;137:373–82.CrossRefPubMed Bockhorn J, Yee K, Chang YF, Prat A, Huo D, Nwachukwu C, Dalton R, Huang S, Swanson KE, Perou CM, et al. MicroRNA-30c targets cytoskeleton genes involved in breast cancer cell invasion. Breast Cancer Res Treat. 2013;137:373–82.CrossRefPubMed
45.
Zurück zum Zitat Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, Chang YF, Huo D, Wen Y, Swanson KE, et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 2013;4:1393.CrossRefPubMedPubMedCentral Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, Chang YF, Huo D, Wen Y, Swanson KE, et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 2013;4:1393.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Neel JC, Lebrun JJ. Activin and TGFbeta regulate expression of the microRNA-181 family to promote cell migration and invasion in breast cancer cells. Cell Signal. 2013;25:1556–66.CrossRefPubMed Neel JC, Lebrun JJ. Activin and TGFbeta regulate expression of the microRNA-181 family to promote cell migration and invasion in breast cancer cells. Cell Signal. 2013;25:1556–66.CrossRefPubMed
47.
Zurück zum Zitat Vanhara P, Horak P, Pils D, Anees M, Petz M, Gregor W, Zeillinger R, Krainer M. Loss of the oligosaccharyl transferase subunit TUSC3 promotes proliferation and migration of ovarian cancer cells. Int J Oncol. 2013;42:1383–9.PubMed Vanhara P, Horak P, Pils D, Anees M, Petz M, Gregor W, Zeillinger R, Krainer M. Loss of the oligosaccharyl transferase subunit TUSC3 promotes proliferation and migration of ovarian cancer cells. Int J Oncol. 2013;42:1383–9.PubMed
48.
Zurück zum Zitat Pils D, Horak P, Vanhara P, Anees M, Petz M, Alfanz A, Gugerell A, Wittinger M, Gleiss A, Auner V, et al. Methylation status of TUSC3 is a prognostic factor in ovarian cancer. Cancer. 2013;119:946–54.CrossRefPubMed Pils D, Horak P, Vanhara P, Anees M, Petz M, Alfanz A, Gugerell A, Wittinger M, Gleiss A, Auner V, et al. Methylation status of TUSC3 is a prognostic factor in ovarian cancer. Cancer. 2013;119:946–54.CrossRefPubMed
49.
Zurück zum Zitat Conway K, Edmiston SN, Tse CK, Bryant C, Kuan PF, Hair BY, Parrish EA, May R, Swift-Scanlan T. Racial variation in breast tumor promoter methylation in the Carolina Breast Cancer Study. Cancer Epidemiol Biomarkers Prev. 2015;24:921–30.CrossRefPubMedPubMedCentral Conway K, Edmiston SN, Tse CK, Bryant C, Kuan PF, Hair BY, Parrish EA, May R, Swift-Scanlan T. Racial variation in breast tumor promoter methylation in the Carolina Breast Cancer Study. Cancer Epidemiol Biomarkers Prev. 2015;24:921–30.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Chen Y, Shi L, Zhang L, Li R, Liang J, Yu W, Sun L, Yang X, Wang Y, Zhang Y, Shang Y. The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer. J Biol Chem. 2008;283:17969–78.CrossRefPubMed Chen Y, Shi L, Zhang L, Li R, Liang J, Yu W, Sun L, Yang X, Wang Y, Zhang Y, Shang Y. The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer. J Biol Chem. 2008;283:17969–78.CrossRefPubMed
51.
Zurück zum Zitat Schmidt ME, Steindorf K, Mutschelknauss E, Slanger T, Kropp S, Obi N, Flesch-Janys D, Chang-Claude J. Physical activity and postmenopausal breast cancer: effect modification by breast cancer subtypes and effective periods in life. Cancer Epidemiol Biomarkers Prev. 2008;17:3402–10.CrossRefPubMed Schmidt ME, Steindorf K, Mutschelknauss E, Slanger T, Kropp S, Obi N, Flesch-Janys D, Chang-Claude J. Physical activity and postmenopausal breast cancer: effect modification by breast cancer subtypes and effective periods in life. Cancer Epidemiol Biomarkers Prev. 2008;17:3402–10.CrossRefPubMed
52.
Zurück zum Zitat Kwak SY, Kim BY, Ahn HJ, Yoo JO, Kim J, Bae IH, Han YH. Ionizing radiation-inducible miR-30e promotes glioma cell invasion through EGFR stabilization by directly targeting CBL-B. FEBS J. 2015;282:1512–25.CrossRefPubMed Kwak SY, Kim BY, Ahn HJ, Yoo JO, Kim J, Bae IH, Han YH. Ionizing radiation-inducible miR-30e promotes glioma cell invasion through EGFR stabilization by directly targeting CBL-B. FEBS J. 2015;282:1512–25.CrossRefPubMed
53.
Zurück zum Zitat Ji D, Chen Z, Li M, Zhan T, Yao Y, Zhang Z, Xi J, Yan L, Gu J. MicroRNA-181a promotes tumor growth and liver metastasis in colorectal cancer by targeting the tumor suppressor WIF-1. Mol Cancer. 2014;13:86.CrossRefPubMedPubMedCentral Ji D, Chen Z, Li M, Zhan T, Yao Y, Zhang Z, Xi J, Yan L, Gu J. MicroRNA-181a promotes tumor growth and liver metastasis in colorectal cancer by targeting the tumor suppressor WIF-1. Mol Cancer. 2014;13:86.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Taylor MA, Sossey-Alaoui K, Thompson CL, Danielpour D, Schiemann WP. TGF-beta upregulates miR-181a expression to promote breast cancer metastasis. J Clin Invest. 2013;123:150–63.CrossRefPubMed Taylor MA, Sossey-Alaoui K, Thompson CL, Danielpour D, Schiemann WP. TGF-beta upregulates miR-181a expression to promote breast cancer metastasis. J Clin Invest. 2013;123:150–63.CrossRefPubMed
55.
Zurück zum Zitat Ji J, Yamashita T, Budhu A, Forgues M, Jia HL, Li C, Deng C, Wauthier E, Reid LM, Ye QH, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology. 2009;50:472–80.CrossRefPubMedPubMedCentral Ji J, Yamashita T, Budhu A, Forgues M, Jia HL, Li C, Deng C, Wauthier E, Reid LM, Ye QH, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology. 2009;50:472–80.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Parikh A, Lee C, Joseph P, Marchini S, Baccarini A, Kolev V, Romualdi C, Fruscio R, Shah H, Wang F, et al. microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition. Nat Commun. 2014;5:2977.CrossRefPubMedPubMedCentral Parikh A, Lee C, Joseph P, Marchini S, Baccarini A, Kolev V, Romualdi C, Fruscio R, Shah H, Wang F, et al. microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition. Nat Commun. 2014;5:2977.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Zhang X, Nie Y, Du Y, Cao J, Shen B, Li Y. MicroRNA-181a promotes gastric cancer by negatively regulating tumor suppressor KLF6. Tumour Biol. 2012;33:1589–97.CrossRefPubMed Zhang X, Nie Y, Du Y, Cao J, Shen B, Li Y. MicroRNA-181a promotes gastric cancer by negatively regulating tumor suppressor KLF6. Tumour Biol. 2012;33:1589–97.CrossRefPubMed
58.
Zurück zum Zitat Li Y, Kuscu C, Banach A, Zhang Q, Pulkoski-Gross A, Kim D, Liu J, Roth E, Li E, Shroyer KR, et al. miR-181a-5p Inhibits Cancer Cell Migration and Angiogenesis via Downregulation of Matrix Metalloproteinase-14. Cancer Res. 2015;75:2674–85.CrossRefPubMedPubMedCentral Li Y, Kuscu C, Banach A, Zhang Q, Pulkoski-Gross A, Kim D, Liu J, Roth E, Li E, Shroyer KR, et al. miR-181a-5p Inhibits Cancer Cell Migration and Angiogenesis via Downregulation of Matrix Metalloproteinase-14. Cancer Res. 2015;75:2674–85.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Ouyang M, Li Y, Ye S, Ma J, Lu L, Lv W, Chang G, Li X, Li Q, Wang S, Wang W. MicroRNA profiling implies new markers of chemoresistance of triple-negative breast cancer. Plos One. 2014;9:e96228.CrossRefPubMedPubMedCentral Ouyang M, Li Y, Ye S, Ma J, Lu L, Lv W, Chang G, Li X, Li Q, Wang S, Wang W. MicroRNA profiling implies new markers of chemoresistance of triple-negative breast cancer. Plos One. 2014;9:e96228.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat D’Aiuto F, Callari M, Dugo M, Merlino G, Musella V, Miodini P, Paolini B, Cappelletti V, Daidone MG. miR-30e* is an independent subtype-specific prognostic marker in breast cancer. Br J Cancer. 2015;113:290–8.CrossRefPubMedPubMedCentral D’Aiuto F, Callari M, Dugo M, Merlino G, Musella V, Miodini P, Paolini B, Cappelletti V, Daidone MG. miR-30e* is an independent subtype-specific prognostic marker in breast cancer. Br J Cancer. 2015;113:290–8.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Guervos MA, Marcos CA, Hermsen M, Nuno AS, Suarez C, Llorente JL. Deletions of N33, STK11 and TP53 are involved in the development of lymph node metastasis in larynx and pharynx carcinomas. Cell Oncol. 2007;29:327–34.PubMedPubMedCentral Guervos MA, Marcos CA, Hermsen M, Nuno AS, Suarez C, Llorente JL. Deletions of N33, STK11 and TP53 are involved in the development of lymph node metastasis in larynx and pharynx carcinomas. Cell Oncol. 2007;29:327–34.PubMedPubMedCentral
62.
Zurück zum Zitat Horak P, Tomasich E, Vanhara P, Kratochvilova K, Anees M, Marhold M, Lemberger CE, Gerschpacher M, Horvat R, Sibilia M, et al. TUSC3 loss alters the ER stress response and accelerates prostate cancer growth in vivo. Sci Rep. 2014;4:3739.CrossRefPubMedPubMedCentral Horak P, Tomasich E, Vanhara P, Kratochvilova K, Anees M, Marhold M, Lemberger CE, Gerschpacher M, Horvat R, Sibilia M, et al. TUSC3 loss alters the ER stress response and accelerates prostate cancer growth in vivo. Sci Rep. 2014;4:3739.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Mohorko E, Glockshuber R, Aebi M. Oligosaccharyltransferase: the central enzyme of N-linked protein glycosylation. J Inherit Metab Dis. 2011;34:869–78.CrossRefPubMed Mohorko E, Glockshuber R, Aebi M. Oligosaccharyltransferase: the central enzyme of N-linked protein glycosylation. J Inherit Metab Dis. 2011;34:869–78.CrossRefPubMed
64.
Zurück zum Zitat Mohorko E, Owen RL, Malojcic G, Brozzo MS, Aebi M, Glockshuber R. Structural basis of substrate specificity of human oligosaccharyl transferase subunit N33/Tusc3 and its role in regulating protein N-glycosylation. Structure. 2014;22:590–601.CrossRefPubMed Mohorko E, Owen RL, Malojcic G, Brozzo MS, Aebi M, Glockshuber R. Structural basis of substrate specificity of human oligosaccharyl transferase subunit N33/Tusc3 and its role in regulating protein N-glycosylation. Structure. 2014;22:590–601.CrossRefPubMed
65.
Zurück zum Zitat Kratochvilova K, Horak P, Esner M, Soucek K, Pils D, Anees M, Tomasich E, Drafi F, Jurtikova V, Hampl A, et al. Tumor suppressor candidate 3 (TUSC3) prevents the epithelial-to-mesenchymal transition and inhibits tumor growth by modulating the endoplasmic reticulum stress response in ovarian cancer cells. Int J Cancer. 2015;137:1330–40.CrossRefPubMed Kratochvilova K, Horak P, Esner M, Soucek K, Pils D, Anees M, Tomasich E, Drafi F, Jurtikova V, Hampl A, et al. Tumor suppressor candidate 3 (TUSC3) prevents the epithelial-to-mesenchymal transition and inhibits tumor growth by modulating the endoplasmic reticulum stress response in ovarian cancer cells. Int J Cancer. 2015;137:1330–40.CrossRefPubMed
66.
Zurück zum Zitat Hetz C, Martinon F, Rodriguez D, Glimcher LH. The unfolded protein response: integrating stress signals through the stress sensor IRE1alpha. Physiol Rev. 2011;91:1219–43.CrossRefPubMed Hetz C, Martinon F, Rodriguez D, Glimcher LH. The unfolded protein response: integrating stress signals through the stress sensor IRE1alpha. Physiol Rev. 2011;91:1219–43.CrossRefPubMed
67.
Zurück zum Zitat Wei Y, Tao X, Xu H, Chen Y, Zhu L, Tang G, Li M, Jiang A, Shuai S, Ma J, et al. Role of miR-181a-5p and endoplasmic reticulum stress in the regulation of myogenic differentiation. Gene. 2016;592:60–70.CrossRefPubMed Wei Y, Tao X, Xu H, Chen Y, Zhu L, Tang G, Li M, Jiang A, Shuai S, Ma J, et al. Role of miR-181a-5p and endoplasmic reticulum stress in the regulation of myogenic differentiation. Gene. 2016;592:60–70.CrossRefPubMed
68.
Zurück zum Zitat Li X, Chen S, Sun T, Xu Y, Chen Y, Liu Y, Xiang R, Li N. The transcriptional regulation of SOX2 on FOXA1 gene and its application in diagnosis of human breast and lung cancers. Clin Lab. 2014;60:909–18.PubMed Li X, Chen S, Sun T, Xu Y, Chen Y, Liu Y, Xiang R, Li N. The transcriptional regulation of SOX2 on FOXA1 gene and its application in diagnosis of human breast and lung cancers. Clin Lab. 2014;60:909–18.PubMed
69.
Zurück zum Zitat Polakova I, Duskova M, Smahel M. Antitumor DNA vaccination against the Sox2 transcription factor. Int J Oncol. 2014;45:139–46.PubMed Polakova I, Duskova M, Smahel M. Antitumor DNA vaccination against the Sox2 transcription factor. Int J Oncol. 2014;45:139–46.PubMed
70.
Zurück zum Zitat Finicelli M, Benedetti G, Squillaro T, Pistilli B, Marcellusi A, Mariani P, Santinelli A, Latini L, Galderisi U, Giordano A. Expression of stemness genes in primary breast cancer tissues: the role of SOX2 as a prognostic marker for detection of early recurrence. Oncotarget. 2014;5:9678–88.CrossRefPubMedPubMedCentral Finicelli M, Benedetti G, Squillaro T, Pistilli B, Marcellusi A, Mariani P, Santinelli A, Latini L, Galderisi U, Giordano A. Expression of stemness genes in primary breast cancer tissues: the role of SOX2 as a prognostic marker for detection of early recurrence. Oncotarget. 2014;5:9678–88.CrossRefPubMedPubMedCentral
Metadaten
Titel
SOX2 regulates multiple malignant processes of breast cancer development through the SOX2/miR-181a-5p, miR-30e-5p/TUSC3 axis
verfasst von
Kuancan Liu
Fuan Xie
Anding Gao
Rui Zhang
Long Zhang
Zhangwu Xiao
Qiong Hu
Weifeng Huang
Qiaojia Huang
Baoshun Lin
Jian Zhu
Haikun Wang
Jianwen Que
Xiaopeng Lan
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2017
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-017-0632-9

Weitere Artikel der Ausgabe 1/2017

Molecular Cancer 1/2017 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.