Skip to main content
Erschienen in: Molecular Cancer 1/2015

Open Access 01.12.2015 | Research

PinX1 inhibits the invasion and metastasis of human breast cancer via suppressing NF-κB/MMP-9 signaling pathway

verfasst von: Meilin Shi, Menghan Cao, Jun Song, Qinghua Liu, Hailong Li, Fei Meng, Zhenqiang Pan, Jin Bai, Junnian Zheng

Erschienen in: Molecular Cancer | Ausgabe 1/2015

Abstract

Background

PinX1 (PIN2/TRF1-interacting telomerase inhibitor 1) was suggested to be correlated with tumor progression. This study was designed to evaluate the role of PinX1 in human breast cancer.

Methods

To evaluate the function of PinX1 in breast cancer, we used a tissue microarray (TMA) of 405 human breast cancer patients and immunohistochemistry to analyze the correlation between PinX1 expression and clinicopathologic variables and patient survival. We also detected the abilities of cell migration and invasion in breast cancer by performing cell migration and invasion assay, gelatin zymography and western blot analysis. Lastly, we set up the nude mice model by Tail vein assay to exam the functional role of PinX1 in breast cancer metastasis.

Results

We found that low PinX1 expression was associated with lymph node metastasis (P = 0.002) and histology grade (P = 0.001) in patients, as well as with poorer overall and disease-specific survival (P = 0.010 and P = 0.003, respectively). Moreover, we identified that PinX1 inhibited the migration and invasion of breast cancer by suppressing MMP-9 expression and activity via NF-κB-dependent transcription in vitro. Finally, our mice model confirmed that PinX1 suppressed breast cancer metastasis in vivo.

Conclusions

Our data revealed that low PinX1 expression was an independent negative prognostic factor for breast cancer patients. These findings suggested that PinX1 might be function as a tumor metastasis suppressor in the development and progression of breast cancer by regulating the NF-κB/MMP-9 signaling pathway, and might be a prognostic marker as well as a therapeutic target for breast cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12943-015-0332-2) contains supplementary material, which is available to authorized users.
Meilin Shi, Menghan Cao and Jun Song contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

WS provided and managed the clinical samples. MS and MC did all the cell-based experiments. MS and HL set up the nude mice model of metastasis and analyzed the data. HL and FM performed the histological sectioning and IHC staining. QL provided professional pathological assessment of the staining. JB designed research and performed the statistical analysis. ZP and JZ provided their clinical input and advises. The manuscript is drafted by MS. All authors read and approved the final manuscript.
Abkürzungen
PinX1
PIN2/TRF1-interacting telomerase inhibitor 1
hTERT
Human telomerase reverse transcriptase
TRF1
Telomeric repeat binding factor 1
LOH
Loss of heterozygosity
MMP
Matrix metalloproteinase
TIMPs
The specific, endogenous tissue inhibitors of metalloproteinases
TID
Telomerase inhibitory domain
NRF
The NF-κB-repression factor
ECM
The extracellular matrix
TMA
The tissue microarray
IRS
The immunoreactive score

Introduction

Breast cancer is the most common malignancy of the female, and its survival rate falls from 90% for localized to 20% for metastatic disease [1]. Each year there are approximately 400 000 deaths because of breast cancer [2]. The high mortality is related to complications of tumor dissemination and distant metastasis. Metastasis is a multistep process requiring cell proliferation, migration, invasion, adhesion, vessel formation and colonization to a secondary site [3]. Therefore, interrupting the metastatic process is a key to decreasing breast cancer mortality. Alterations in chromatin play a critical role in breast cancer progression and metastasis, but the exact molecular mechanisms are still limited [4].
Human telomerase is a ribonucleoprotein, mainly consisting of catalytic subunit hTERT and RNA template hTR, which involves in malignant tumor formation [5-7]. Human telomerase reverse transcriptase (hTERT), containing two conserved N-terminal and four C-terminal domains essential for telomerase catalytic activity, is referred as the rate-limiting step of telomerase activation [8]. Telomerase expression is suppressed in most normal cells, whereas reactivated in more than 85% of human cancer cells [9,10]. In addition, the telomerase activity is regulated by telomeric repeat binding factor 1 (TRF1) and its associated proteins, including PinX1 [11]. PinX1 is a potent telomerase inhibitor and a putative tumor suppressor, firstly found as a Pin2/TRF1-binding protein [12]. However, unlike other TRF1-binding proteins, PinX1 is unique in that it can also directly bind to hTERT and inhibit telomerase activity [12].
PinX1 is a versatile gene at human chromosome 8p23, a region frequently associated with loss of heterozygosity (LOH) in a variety of human malignancies [13-15]. The full-length form of PinX1 is composed of 737 bp and encodes a 45-KDa nucleolar protein containing 328 amino acids [16]. It has been identified that Pinx1 deficiency could induce telomerase activation, telomere elongation and chromosome instability [17], whereas overexpression of PinX1 leads to a decrease in both telomerase activity and cancer cell tumorigenicity [18,19]. LOH of PinX1 resulted in gastric carcinoma development, which suggested PinX1 might have a potential inhibitory role in cancer metastasis [20]. Then increasing evidences demonstrate that PinX1 plays a key role as a putative tumor suppressor in human cancer progression [21-26]. However, the PinX1 expression status and its correlation with the clinicopathological features in breast cancer have never been investigated. In addition, the potential molecular mechanisms underlying the role of PinX1 in breast cancer are still unclear.
To evaluate the function of PinX1 in breast cancer, we used a tissue microarray (TMA) of human breast cancer patients and immunohistochemistry to analyze the correlation between PinX1 expression and clinicopathologic variables and patient survival. Furthermore, we demonstrated that PinX1 suppressed breast cancer migration and invasion by inhibiting the expression and activity of MMP-9 via NF-κB-dependent transcription in vitro and in vivo. These data suggested that PinX1 might be function as a tumor metastasis suppressor in the development and progression of breast cancer by regulating the NF-κB/MMP-9 signaling pathway, and might be a prognostic marker as well as a therapeutic target for breast cancer.

Results

Correlation of PinX1 staining with clinicopathologic parameters in breast cancer patients

To investigate PinX1 expression in breast cancer, immunohistochemistry was carried out in TMA slides (Figure 1A). Samples with IRS 0–3 and IRS 4–12 were classified as low and high expression of PinX1. Of the 405 breast cancer analyzed, low and high expression of PinX1 staining were 52.3% (212/405) and 47.7% (193/405), respectively (Table 1). We then analyzed the correlations between PinX1 expression and characteristics of the breast carcinomas, and found that PinX1 staining was dramatically decreased in histology grade II and III compared with histology grade I (P = 0.001, χ2 test, Table 1). We also found that PinX1 expression is significantly correlated with lymph node metastasis (P = 0.002, χ2 test, Table 1). However, we did not find any significant correlations between PinX1 expression and other clinicopathologic variables, including patient age, tumor size, ER status, PR status, HER2 status and p53 status.
Table 1
PinX1 staining and clinicopathological characteristics of 405 breast cancer patients
Variables
PinX1 staining
Low (%)
High (%)
Total
P *
Age
    
≤50 years
99 (52.7)
89 (47.3)
188
0.906
>50 years
113 (52.1)
104 (47.9)
217
 
Tumor size
    
T1 (<2 cm)
47 (61.8)
29 (38.2)
76
0.090
T2 (2-5 cm)
148 (51.2)
141 (48.8)
289
 
T3 (>5 cm)
11 (39.3)
17 (60.7)
28
 
Lymph node metastasis
    
Negative
83 (43.5)
108 (56.5)
191
0.002
Positive
110 (59.5)
75 (40.5)
185
 
Histology grade
    
I
11 (30.6)
25 (69.4)
36
0.001
II
103 (50.5)
101 (49.5)
204
 
III
50 (68.5)
23 (31.5)
73
 
ER status
    
Negative
40 (54.8)
33 (45.2)
73
0.359
Positive
63 (48.1)
68 (51.9)
131
 
PR status
    
Negative
48 (54.5)
40 (45.5)
88
0.313
Positive
55 (47.4)
61 (52.6)
116
 
HER2 status
    
Negative
14 (66.7)
7 (33.3)
21
0.206
Positive
79 (52.0)
73 (48.0)
152
 
p53 status
    
Negative
41 (50.0)
41 (50.0)
82
0.347
Positive
52 (57.1)
39 (42.9)
91
 
* P values are from χ2 test.
Some cases were not available for the information.

PinX1 functions as an independent prognostic factor for human breast cancer

To further study whether PinX1 expression correlates with the survival of patients, Kaplan-Meier survival curves were constructed using 5-year overall and disease-specific cumulative survival to compare the patients with high PinX1 staining to those with low PinX1 staining (n = 236, follow-up time, 60 months). Our data revealed that low PinX1 staining correlated with both poorer overall and disease-specific patient survival (P = 0.010 and P = 0.003, respectively, log-rank test; Figure 1B & C). The 5-year overall cumulative survival rate dropped from 65.3% in patients with high PinX1 expression to 48.0% in those with low PinX1 expression, and the 5-year disease-specific cumulative survival rate dropped from 78.4% in patients with high PinX1 expression to 57.5% in those with low PinX1 expression.
Moreover, we examined whether PinX1 expression was an independent prognostic factor for breast cancer. The univariate Cox regression analyses revealed that PinX1 expression was an independent prognostic marker for breast cancer patients overall survival (hazard ratio, 0.573; 95% CI, 0.371-0.884; P = 0.012; Table  2 ) and disease-specific survival (hazard ratio, 0.417; 95% CI, 0.230-0.755; P = 0.004; Table  2 ). In multivariate Cox regression analysis, we found that PinX1 expression was also an independent prognostic marker for both 5-year overall survival (hazard ratio, 0.527; 95% CI, 0.404-0.656; P = 0.027; Table 3) and disease-specific survival (hazard ratio, 0.429; 95% CI, 0.222-0.630; P = 0.012; Table 3). Our results definitely confirmed that low PinX1 expression is associated with poor prognosis, suggesting that PinX1 may function as a prognostic marker for breast cancer.
Table 2
Univariate Cox proportional regression analysis on 5-year overall and disease-specific survival of 405 breast cancer patients
Variable *
Overall survival
Disease-specific survival
Hazard ratio
95% CI
P *
Hazard ratio
95% CI
P *
PinX1
      
Low
1.000
 
0.012
1.000
 
0.004
High
0.573
0.371-0.884
 
0.417
0.230-0.755
 
Age
      
≤50 years
1.000
 
0.443
1.000
 
0.630
>50 years
0.864
0.594-1.225
 
0.855
0.632-1.030
 
Tumor size
      
≤5 cm
1.000
 
0.002
1.000
 
0.000
>5 cm
2.289
1.363-3.843
 
3.463
2.730-3.935
 
Lymph node metastasis
      
Negative
1.000
 
0.000
1.000
 
0.000
positive
4.994
3.032-8.227
 
3.564
2.594-4.505
 
Histology Grade
      
I
1.000
 
0.002
1.000
 
0.005
II/III
2.309
1.740-3.456
 
2.554
1.618-3.551
 
* P values are from Log-rank test.
CI: confidence interval.
Table 3
Multivariate Cox regression analysis on 5-year overall and disease-specific survival of 405 breast cancer patients
Variable *
Overall survival
Disease-specific survival
Hazard ratio
95% CI
P
Hazard ratio
95% CI
P
PinX1
0.527
0.404 to 0.656
0.027
0.429
0.222 to 0.630
0.012
Age
0.993
0.740 to 1.345
0.836
0.856
0.652 to 1.222
0.884
Tumor size
2.730
1.947 to 3.785
0.003
2.913
1.882 to 4.146
0.001
Lymph node metastasis
3.183
1.911 to 4.505
0.001
3.962
2.975 to 4.756
0.000
Histology Grade
1.840
1.184 to 2.843
0.023
1.958
1.514 to 2.511
0.032
*Coding of variables: PinX1 was coded as 1 (low), and 2 (high). Age was coded as 1 (≤50 years), and 2 (>50 years). Tumor size was coded as 1 (≤5 cm), and 2 (>5 cm). Lymph node metastasis was coded as 1 (negative), and 2 (positive). Histology grade was coded as 1 (I), and 2 (II and III).
CI: confidence interval.

PinX1 inhibits migration and invasion of human breast cancer cells in vitro

Because low PinX1 expression is associated with poor prognosis, supporting PinX1 may play important roles in one or more steps of tumor metastasis. We investigated the involvement of PinX1 in breast cancer cells migration and invasion. We transiently transfected MDA-MB-231 and BT-549 cells with control siRNA and PinX1 siRNA or pEGFP-C3 and pEGFP-C3-PinX1 plasmids, respectively. Forty-eight or twenty-four hours after transfection, PinX1 protein was significantly knockdown or overexpressed in cancer cells, respectively (Figure 2A & B). In cell migration assay, we found that the ability of cell migration was drastically increased after PinX1 knockdown in both MDA-MB-231 and BT-549 cell line (Figure 2C). In contrast, overexpression of PinX1 inhibited cell migration (Figure 2D). Meanwhile, the results of the cell invasion assay corresponded with the cell migration assay (Figure 2E & F). However, overexpression or knockdown of PinX1 had no detectable effect on the proliferation of breast cancer cells under normal culture conditions (data not shown).

PinX1 inhibits human breast cancer cells’ migration and invasion abilities by suppressing MMP-9 expression and activity

To investigate the mechanisms of PinX1 regulating migration and invasion in breast cancer cells, we performed western blot to detect the MMPs protein levels and gelatin zymography to observe the MMPs activity. Our result showed that the MMP-9 expression and activity were negatively regulated by PinX1 in MDA-MB-231 and BT-549 cells,but not MMP-2 (Figure 3A & B & C). So we supposed PinX1 suppress migration and invasion of breast cancer cells by regulating MMP-9 expression and activity. To further validate our assumption, we added MMP-9 inhibitor I (sc-311437, Santa Cruz) at the same time of PinX1 siRNA transfecting into cells. As expected, the upregulation of MMP-9 was blocked by MMP-9 inhibitor I (Figure 3D & E). We also validated this hypothesis by migration and invasion analysis, the migration and invasion ability can be enhanced by knocking down PinX1 in MDA-MB-231 and BT-549 cells, nevertheless, these regulations were blocked by MMP-9 inhibitor I (Figure 3F & G).
As we know TIMP-1 and TIMP-2 is the tissue inhibitor of MMP-9 and MMP-2, so we detected the expression of TIMP protein. Our data showed that MMP-9 expression was up-regulated or down-regulated corresponded with PinX1 knockdown or overexpression, however, the expression of TIMP-1 had not changed correspondingly. Moreover, neither overexpressing nor silencing PinX1 had any effect on the protein levels of MMP-2 and TIMP-2 (Figure 3A & B).

PinX1 suppress MMP-9 expression via NF-κB-dependent transcription

Furthermore, increasing evidence demonstrate that several MMPs (including MMP-9) expression and activation were regulated by NF-κB activation in many human cancers [27,28]. G-patch domain of PinX1 is also an important nucleic acids binding domain which could combine with the C-terminus of the NF-κB-repression factor (NRF) [29]. Thus, PinX1 may also inhibit the transcriptional activity of NF-κB proteins by direct protein-protein interaction with its G-patch domain. Western blot results showed that the level of NF-κB-p65 protein was dramatically increased in PinX1KD-MDA-MB-231 cells and PinX1KD-BT-549 cells. In contrast, NF-κB-p65 expression was down-regulated sharply in PinX1OE-MDA-MB-231 cells and PinX1OE-BT-549 cells (Figure 4A & B). To further confirm whether PinX1 regulated MMP-9 expression via the NF-κB signaling pathway in human breast cancer cells, we transfected NF-κB-p65 siRNA (Santa Cruz) into PinX1KD-MDA-MB-231 cells and PinX1KD-BT-549 cells. Our result indicated that the MMP-9 expression was up-regulated in PinX1KD-MDA-MB-231 cells and PinX1KD-BT-549 cells, but these effects were further blocked by silencing NF-κB-p65 with the specific siRNA (Figure 4A & B). These data provide definite evidence that PinX1 may modulate MMP-9 expression by NF-κB transcription factor.
We also validated the mechanism by migration and invasion analysis. The migration and invasion ability can be enhanced in PinX1KD-MDA-MB-231 cells and PinX1KD-BT-549 cells, however, these effects were dramatically reversed by treatment with NF-κB-p65 siRNA (Figure 4C & D). These data suggested that PinX1 may regulate the migration and invasion via the NF-κB/MMP-9 signaling pathway.

PinX1 inhibits breast cancer cells metastasis in vivo

Lastly, we examined whether PinX1 suppressed breast cancer metastasis in vivo. PinX1OE-MDA-MB-231, PinX1KD-MDA-MB-231 and Ctrl-MDA-MB-231 cell lines were established previously. After 3 weeks selection following with lentivirus infection, the PinX1 protein levels of these cell lines were confirmed by western blot. Then continuing to incubate them without adding puromycin for 2 months, we determined that the PinX1 protein expression levels of three stable cell lines had not been changed (Figure 5A).
The BALB/c nude mice were randomly divided into three groups: PinX1OE, PinX1KD and Control group, each group consisted of 10 mice. Three groups of nude mice were injected through tail vein with PinX1OE-MDA-MB-231, PinX1KD-MDA-MB-231 and Ctrl-MDA-MB-231 cells respectively. After 2 months, three groups of mice were sacrificed and their lungs were resected and fixed in 10% buffered formalin for metastatic nodules counting and further histopathological analysis. Randomly selected metastatic nodules had been validated by H&E staining (Figure 5B). Extensive tumor formation was found in PinX1KD group. In contrast, the lungs in PinX1OE group had fewer and smaller detectable tumor nodules (Figure 5C). Compared with PinX1OE group, the dramatic increase in the number of the tumor nodules was observed in PinX1KD group, meanwhile, PinX1OE group and PinX1KD group had significant differences compared with Control group respectively (Figure 5D).

Discussion

PinX1 is a functional gene at human chromosome 8p23, a region frequently associated with loss of heterozygosity (LOH) in a variety of human malignancies [13]. Increasing evidence demonstrate that PinX1 plays a key role as a putative tumor suppressor in human cancer progression [18,19]. However, the expression and function of PinX1 in breast cancer and its correlation with the clinicopathological features of breast cancer patients have never been investigated.
In the present study, we used a breast cancer TMA containing 405 tumor samples with specific clinical data to investigate the role of PinX1 in human breast cancer. Our data showed that low PinX1 expression was associated with lymph node metastasis and histology grade in patients, as well as with poorer overall and disease-specific survival (Figure 1B & C; Table 1). Cox regression analysis revealed that low PinX1 expression was an independent negative prognostic indicator for breast cancer patients (Table  2 ; Table 3). These findings suggested a potential role of PinX1 in regulating breast cancer metastasis and functioning as a breast cancer candidate clinical prognostic marker. Our clinical data urged us to carry out a series of in vitro and in vivo experiments to explore the potential mechanisms.
The matrix metalloproteinase (MMP) family can degrade the extracellular matrix (ECM) in the major early stages of a number of malignant tumors, which plays an important role in cancer invasion and metastasis [30]. High expression of MMP-9 and MMP-2 were associated with lymph node metastasis as well as with poorer survival in breast cancer [31]. Our data demonstrated that PinX1 inhibited breast cancer cells’ migration and invasion abilities by down-regulating MMP-9 expression and activity in vitro (Figure 2; Figure 3). MMPs activity is controlled by specific, endogenous tissue inhibitors of metalloproteinases (TIMPs), and the imbalance between MMPs and TIMPs is responsible for cancer metastasis [32]. TIMP-1 is the tissue inhibitor of MMP-9, which negatively regulating MMP-9 enzyme activity were involved in several tumor metastasis processes, including breast cancer [33]. But in our study, TIMP-1 expression was not seemed to be the regulator of MMP-9 activation in breast cancer cells (Figure 3A & B). These result suggested us to investigate the potential mechanism of how PinX1 regulates MMP-9 expression and activity.
NF-κB is a critical transcription factor activated in various types of human cancers and plays a crucial role in tumor development and progression [34,35]. The NF-κB signaling modulates several key biological processes during the development and progression of cancer by inducing transcription of a variety of target genes that regulate cell proliferation, survival, invasion and angiogenesis [36-38]. NF-κB is constitutively present in cells as a heterodimer, consisting of a p50 DNA-binding subunit and a p65 transactivating subunit [39]. Furthermore, increasing evidence demonstrate that MMP-9 expression and activation were regulated by p65 up-regulation and nuclear translocation which induced NF-κB activation in many human cancers [27,28]. Our data indicated that the NF-κB specific siRNA dramatically prevented the up-regulation of MMP-9 expression induced by PinX1 knockdown (Figure 4A & B). Consistently, the increased ability of migration and invasion induced by PinX1 knockdown was also suppressed by inhibition of NF-κB-p65 expression in breast cancer cells (Figure 4C & D). Thus, these results suggested that PinX1 may regulate the migration and invasion via the NF-κB/MMP-9 signaling pathway.
We regret to investigate the potential regulation mechanism between PinX1 and NF-κB pathway, however, we have found some conceivable relations between them. Human PinX1 protein contains an N-terminal Gly-rich patch (G-patch) and a C-terminal TID (telomerase inhibitory domain) [12]. G-patch domain existed in a number of putative RNA-binding proteins involved in tumor suppression and DNA-damage repair [40], but the function of them had been researched rarely. Moreover, G-patch domain of PinX1 is also an important nucleic acids binding domain which could combine with the C-terminus of the NF-κB-repression factor (NRF) [29]. NRF is a nuclear inhibitor of NF-κB, which can inhibit the transcriptional activity of NF-κB proteins by direct protein-protein interaction [41]. Thus, we conclude that PinX1 also can inhibit the transcriptional activity of NF-κB proteins by direct protein-protein interaction with its G-patch domain (Figure 6). These hypothesizes have never been definitely validated. However, the association between PinX1 and NF-κB in breast cancer cells exists indeed. So we will investigate the further molecular mechanisms between PinX1 and NF-κB signaling pathway continually.
To further observe the functional role of PinX1 in breast cancer metastasis in vivo, three groups of nude mice experimental model were constructed. We investigated that PinX1 overexpression in breast cancer cells significantly inhibited the formation of metastasis nodules in lung of nude mice. In contrast, PinX1 knockdown dramatically enhanced the metastasis process (Figure 5).
In conclusion, loss of PinX1 expression was significantly correlated with breast cancer progression and was an independent negative prognostic factor in breast cancer patients. PinX1 suppressed breast cancer migration and invasion by inhibiting the expression and activity of MMP-9 via NF-κB-dependent transcription. Therefore PinX1 may be an attractive therapeutic target for the treatment of breast cancer.

Materials and methods

Patient specimens and tissue microarray construction

The collection of patient specimens and construction of the tissue microarray (TMA) have been previously described [42]. The study material consists of a series of 405 consecutive cases of primary breast carcinoma, from The First Affiliated Hospital of Nanjing Medical University, between 1996 and 2005. All these patients were treated with surgery only or with postoperative adjuvant therapy. The patients’ clinicopathologic information including age at diagnosis, tumor size, histology grade, lymph node metastasis, ER status, PR status, HER2 status and p53 status was obtained from the archive of the pathology department and confirmed by the medical record of the hospital. The histologic grade was assessed using Bloom-Richardson classification. Five-year clinical follow-up results were available for 236 patients. The use of these tissue specimens was approved by the Ethics Committee of the Hospital.

Immunohistochemistry of TMA

Immunohistochemistry was carried out as described before [43]. According to the streptavidin-peroxidase (Sp) method using a standard Sp Kit (Zhongshan biotech, Beijing, China). The TMA slides were dewaxed at 60°C for 20 min followed by three 5-min washes with xylene. The tissues were then rehydrated by washing the slides for 5-min each with 100%, 95%, 80% ethanol and finally with distilled water. The slides were then heated to 95°C for 30 min in 10 mmol/L sodium citrate (pH 6.0) for antigen retrieval and then treated with 3% hydrogen peroxide for 1 hour to block the endogenous peroxidase activity. Then subsequently the TMA slides were incubated with a polyclonal rabbit anti-PinX1 antibody (1:50 dilution; Novus Biologicals, Littleton, CO, USA) at 4°C overnight, and 3, 3′-diaminobenzidine (DAB; Zhongshan Biotech, Beijing, China) was used to produce a brown precipitate. Negative controls were performed by substituting primary antibodies with non-immune serum.

Evaluation of immunostaining

The evaluation of PinX1 staining was blindly and independently examined by two pathologists. Positive PinX1 immunostaining is defined mainly in the nucleus area and also can be observed in the cytoplasm. We grade it according to both the intensity and percentage of cells with positive staining. The PinX1 staining intensity was scored 0 to 3 (0 = negative; 1 = weak; 2 = moderate; 3 = strong). The percentage of PinX1-positive stained cells was also scored into four categories: 1 (0%-25%), 2 (26%-50%), 3 (51%-75%) and 4 (76%-100%). The level of PinX1 staining was evaluated by the immunoreactive score (IRS), which is calculated by multiplying the scores of staining intensity and the percentage of positive cells. Based on the IRS, the PinX1 staining pattern was categorized as negative (IRS: 0), weak (IRS: 1–3), moderate (IRS: 4–6) and strong (IRS: 8–12). The concordance for IRS staining score of PinX1 between the two pathologists was 363 in 405 samples (90%), and the few discrepancies were resolved by consensus using a multihead microscope.
The optimum cutoff value of IRS was obtained by receiver-operator characteristic analysis, and the areas under the curves at different cutoff values of the PinX1 IRS for 1, 3 and 5 years of overall survival time were also calculated. The optimum value of cutoff point of the PinX1 IRS was shown to be 3 since it had the best predictive value for survival (Additional file 1: Figure S1). Under these conditions, samples with IRS 0–3 and 4–12 were classified as having low or high expression of PinX1, respectively.

Animals and cell lines

Female BALB/c nude mice, 6 weeks old, were purchased from the Shanghai Laboratory Animal Center (Shanghai, China) for studies approved by the Animal Care Committee of Xuzhou Medical College. Two human breast cancer cell lines MDA-MB-231 and BT-549 were purchased from the Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, China). MDA-MB-231 cells were cultured in Leibovitz’ s L-15 Medium (Gibco, USA) supplemented with 10% fetal calf serum (Gibco, USA), BT-549 cells were cultured in RPMI 1640 Medium (Invitrogen, Shanghai, China) supplemented with 10% fetal calf serum. These two cells were both incubated in a 37°C humidified incubator with 5% CO2.

siRNA and DNA transfections, and stable cell line generation

Cells were grown to 50% confluence before small interfering RNA (siRNA) transfection. PinX1 siRNA (GenePharma, Shanghai, China), NF-κB-p65 siRNA (Santa Cruz, CA, USA) or Nonspecific control siRNA (GenePharma, Shanghai, China) was transfected by siLentFect Lipid Reagent (Bio-Rad, CA, USA) according to the manufacturer’s protocol. The pEGFP-C3 and pEGFP-C3-PinX1 expression plasmids were obtained from Dr Xiao-Fen Lai (Southern Medical University, Guangzhou, China). Transfection of the pEGFP-C3-PinX1 plasmid and the pEGFP-C3 vector into the MDA-MB-231 and BT-549 cells were carried out using Lipofectamine 2000 transfection reagent (Invitrogen) following the manufacturer’s instructions.
PinX1 overexpression MDA-MB-231 cell lines (PinX1OE-MDA-MB-231), PinX1 knockdown MDA-MB-231 cell lines (PinX1KD-MDA-MB-231) and control MDA-MB-231 cell lines (Ctrl-MDA-MB-231) were established by infecting with lentivirus packing PinX1 expression vector, PinX1 shRNA expression vector and control vector respectively (GenePharma, Shanghai, China). It is the same as the construction of PinX1 overexpression BT-549 cell lines (PinX1OE-BT-549), PinX1 knockdown BT-549 cell lines (PinX1KD-BT-549) and control BT-549 cell lines (Ctrl-BT-549). Target cells were infected with lentivirus for 48 hours then selected with puromycin (Santa Cruz) for three weeks.

Western blot analysis

Western blot analysis was performed as described previousely [44]. The following antibodies were used for western blot: rabbit anti-PinX1 (Novus Biologicals); rabbit anti-MMP-2, anti-MMP-9, anti-TIMP-1, anti-TIMP-2 (all from Cell Signaling Technology); rabbit anti-NF-κB-p65 (Santa Cruz Biotechnology); mouse anti-β-actin (Cell Signaling Technology); Infrared IRDye-labeled secondary antibody (LI-COR, NE, USA) was applied to the blot for 2 hour at room temperature, the signals were detected with Odyssey Infrared Imaging system (LI-COR).

Cell migration and invasion assay

Cell migration and invasion assay were performed using modified two-chamber plates with a pore size of 8 μm. The transwell filter coating with or without Matrigel (BD Biosciences, Mississauga, Canada) were used respectively for invasion and migration assay. The detailed conditions were described previously [45].

Gelatin zymography

Gelatin zymography was performed as described before [46]. Thirty-six hours after transfection, cells were incubated in serum-free medium for 24 h. The proteins in the conditioned medium were concentrated with Amicon Ultra-4-30 k centrifugal filters (Millipore, MA, USA) at 7500 g for 20 min at 4°C. Fifty micrograms of the proteins were loaded on a 10% polyacrylamide gel containing 0.1% gelatin (Sigma, MO, USA). Gelatinolytic activity was shown as clear areas in the gel. Gels were photographed and then quantitatively measured by scanning densitometry.

Tail vein assay of metastasis

The BALB/c nude mice were randomly divided into three groups: PinX1OE, PinX1KD and Control group, each group consisted of 10 mice. The mice were injected intravenously with 2.5 × 106 MDA-MB-231 cells in 200 μl of PBS through tail vein. After 2 months, the three groups of mice were sacrificed, their lungs were resected and fixed in 10% buffered formalin for metastatic nodules counting and further histopathological analysis. The number of metastatic nodules presented on the surface of each group of lungs was counted by visual inspection using a stereoscopic dissecting microscope.

Statistical analysis

For the TMA, statistical analysis was performed with SPSS 20 software (SPSS, Inc, Chicago, IL). The association between PinX1 staining and the clinicopathologic parameters of the breast cancer patients was evaluated by χ2 test. The Kaplan-Meier method and log-rank test were used to evaluate the correlation between PinX1 expression and patient survival. Cox regression model was used for multivariate analysis. Data are expressed as the means ± SD. Two-factor analysis of variance procedures and the Dunnett’s t-test were used to assess differences within treatment groups. Differences were considered significant when P < 0.05.

Acknowledgements

This project is supported by grants from the National Natural Science Foundation of China (No.81472663, 81201636), the Science and Technology Department of Jiangsu Province (No.BK2012139), and China Postdoctoral Science Foundation (No. 2014 T70549, 2012 M511323).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

WS provided and managed the clinical samples. MS and MC did all the cell-based experiments. MS and HL set up the nude mice model of metastasis and analyzed the data. HL and FM performed the histological sectioning and IHC staining. QL provided professional pathological assessment of the staining. JB designed research and performed the statistical analysis. ZP and JZ provided their clinical input and advises. The manuscript is drafted by MS. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Wang X, Lu H, Urvalek AM, Li T, Yu L, Lamar J, et al. KLF8 promotes human breast cancer cell invasion and metastasis by transcriptional activation of MMP9. Oncogene. 2011;30:1901–11.CrossRefPubMedCentralPubMed Wang X, Lu H, Urvalek AM, Li T, Yu L, Lamar J, et al. KLF8 promotes human breast cancer cell invasion and metastasis by transcriptional activation of MMP9. Oncogene. 2011;30:1901–11.CrossRefPubMedCentralPubMed
2.
Zurück zum Zitat DeSantis C, Siegel R, Bandi P, Jemal A. Breast cancer statistics. CA Cancer J Clin. 2011;61:409–18.CrossRefPubMed DeSantis C, Siegel R, Bandi P, Jemal A. Breast cancer statistics. CA Cancer J Clin. 2011;61:409–18.CrossRefPubMed
3.
Zurück zum Zitat Chen J, Gallo KA. MLK3 regulates paxillin phosphorylation in chemokine-mediated breast cancer cell migration and invasion to drive metastasis. Cancer Res. 2012;72:4130–40.CrossRefPubMed Chen J, Gallo KA. MLK3 regulates paxillin phosphorylation in chemokine-mediated breast cancer cell migration and invasion to drive metastasis. Cancer Res. 2012;72:4130–40.CrossRefPubMed
4.
Zurück zum Zitat Cowper-Sal Lari R, Zhang X, Wright JB, Bailey SD, Cole MD, Eeckhoute J, et al. Breast cancer risk-associated SNPs modulate the affinity of chromatin for FOXA1 and alter gene expression. Nat Genet. 2012;44:1191–8.CrossRefPubMedCentralPubMed Cowper-Sal Lari R, Zhang X, Wright JB, Bailey SD, Cole MD, Eeckhoute J, et al. Breast cancer risk-associated SNPs modulate the affinity of chromatin for FOXA1 and alter gene expression. Nat Genet. 2012;44:1191–8.CrossRefPubMedCentralPubMed
5.
Zurück zum Zitat Shay JW, Wright WE. Telomerase: a target for cancer therapeutics. Cancer Cell. 2002;2:257–65.CrossRefPubMed Shay JW, Wright WE. Telomerase: a target for cancer therapeutics. Cancer Cell. 2002;2:257–65.CrossRefPubMed
6.
Zurück zum Zitat Dong CK, Masutomi K, Hahn WC. Telomerase: regulation, function and transformation. Crit Rev Oncol Hematol. 2005;54:85–93.CrossRefPubMed Dong CK, Masutomi K, Hahn WC. Telomerase: regulation, function and transformation. Crit Rev Oncol Hematol. 2005;54:85–93.CrossRefPubMed
7.
Zurück zum Zitat Smogorzewska A, de Lange T. Regulation of telomerase by telomeric proteins. Annu Rev Biochem. 2004;73:177–208.CrossRefPubMed Smogorzewska A, de Lange T. Regulation of telomerase by telomeric proteins. Annu Rev Biochem. 2004;73:177–208.CrossRefPubMed
8.
Zurück zum Zitat Lin SY, Elledge SJ. Multiple tumor suppressor pathways negatively regulate telomerase. Cell. 2003;113:881–9.CrossRefPubMed Lin SY, Elledge SJ. Multiple tumor suppressor pathways negatively regulate telomerase. Cell. 2003;113:881–9.CrossRefPubMed
9.
Zurück zum Zitat Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266:2011–5.CrossRefPubMed Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266:2011–5.CrossRefPubMed
10.
11.
Zurück zum Zitat Lee TH, Tun-Kyi A, Shi R, Lim J, Soohoo C, Finn G, et al. Essential role of Pin1 in the regulation of TRF1 stability and telomere maintenance. Nat Cell Biol. 2009;11:97–105.CrossRefPubMedCentralPubMed Lee TH, Tun-Kyi A, Shi R, Lim J, Soohoo C, Finn G, et al. Essential role of Pin1 in the regulation of TRF1 stability and telomere maintenance. Nat Cell Biol. 2009;11:97–105.CrossRefPubMedCentralPubMed
12.
Zurück zum Zitat Zhou XZ, Lu KP. The Pin2/TRF1-interacting protein PinX1 is a potent telomerase inhibitor. Cell. 2001;107:347–59.CrossRefPubMed Zhou XZ, Lu KP. The Pin2/TRF1-interacting protein PinX1 is a potent telomerase inhibitor. Cell. 2001;107:347–59.CrossRefPubMed
13.
Zurück zum Zitat Baffa R, Santoro R, Bullrich F, Mandes B, Ishii H, Croce CM. Definition and refinement of chromosome 8p regions of loss of heterozygosity in gastric cancer. Clin Cancer Res. 2000;6:1372–7.PubMed Baffa R, Santoro R, Bullrich F, Mandes B, Ishii H, Croce CM. Definition and refinement of chromosome 8p regions of loss of heterozygosity in gastric cancer. Clin Cancer Res. 2000;6:1372–7.PubMed
14.
Zurück zum Zitat Bova GS, MacGrogan D, Levy A, Pin SS, Bookstein R, Isaacs WB. Physical mapping of chromosome 8p22 markers and their homozygous deletion in a metastatic prostate cancer. Genomics. 1996;35:46–54.CrossRefPubMed Bova GS, MacGrogan D, Levy A, Pin SS, Bookstein R, Isaacs WB. Physical mapping of chromosome 8p22 markers and their homozygous deletion in a metastatic prostate cancer. Genomics. 1996;35:46–54.CrossRefPubMed
15.
Zurück zum Zitat Kishimoto Y, Shiota G, Wada K, Kitano M, Nakamoto K, Kamisaki Y, et al. Frequent loss in chromosome 8p loci in liver cirrhosis accompanying hepatocellular carcinoma. J Cancer Res Clin Oncol. 1996;122:585–9.CrossRefPubMed Kishimoto Y, Shiota G, Wada K, Kitano M, Nakamoto K, Kamisaki Y, et al. Frequent loss in chromosome 8p loci in liver cirrhosis accompanying hepatocellular carcinoma. J Cancer Res Clin Oncol. 1996;122:585–9.CrossRefPubMed
16.
Zurück zum Zitat MacGrogan D, Levy A, Bova GS, Isaacs WB, Bookstein R. Structure and methylation-associated silencing of a gene within a homozygously deleted region of human chromosome band 8p22. Genomics. 1996;35:55–65.CrossRefPubMed MacGrogan D, Levy A, Bova GS, Isaacs WB, Bookstein R. Structure and methylation-associated silencing of a gene within a homozygously deleted region of human chromosome band 8p22. Genomics. 1996;35:55–65.CrossRefPubMed
17.
Zurück zum Zitat Zhang B, Bai YX, Ma HH, Feng F, Jin R, Wang ZL. Silencing PinX1 compromises telomere length maintenance as well as tumorigenicity in telomerase-positive human cancer cells. Cancer Res. 2009;69:75–83.CrossRefPubMed Zhang B, Bai YX, Ma HH, Feng F, Jin R, Wang ZL. Silencing PinX1 compromises telomere length maintenance as well as tumorigenicity in telomerase-positive human cancer cells. Cancer Res. 2009;69:75–83.CrossRefPubMed
18.
Zurück zum Zitat Zhou XZ, Huang PY, Shi R, Lee TH, Lu GN, Zhang ZH, et al. The telomerase inhibitor PinX1 is a major haploinsufficient tumor suppressor essential for chromosome stability in mice. J Clin Invest. 2011;121:1266–82.CrossRefPubMedCentralPubMed Zhou XZ, Huang PY, Shi R, Lee TH, Lu GN, Zhang ZH, et al. The telomerase inhibitor PinX1 is a major haploinsufficient tumor suppressor essential for chromosome stability in mice. J Clin Invest. 2011;121:1266–82.CrossRefPubMedCentralPubMed
19.
20.
Zurück zum Zitat Ma YY, Wu L, Liu C, Xu L, Li D, Li JC. The correlation of genetic instability of PINX1 gene to clinico-pathological features of gastric cancer in the Chinese population. J Cancer Res Clin Oncol. 2009;135:431–7.CrossRefPubMed Ma YY, Wu L, Liu C, Xu L, Li D, Li JC. The correlation of genetic instability of PINX1 gene to clinico-pathological features of gastric cancer in the Chinese population. J Cancer Res Clin Oncol. 2009;135:431–7.CrossRefPubMed
21.
Zurück zum Zitat Shi R, Zhao Z, Zhou H, Wei M, Ma WL, Zhou JY, et al. Reduced expression of PinX1 correlates to progressive features in patients with prostate cancer. Cancer Cell Int. 2014;14:46.CrossRefPubMedCentralPubMed Shi R, Zhao Z, Zhou H, Wei M, Ma WL, Zhou JY, et al. Reduced expression of PinX1 correlates to progressive features in patients with prostate cancer. Cancer Cell Int. 2014;14:46.CrossRefPubMedCentralPubMed
22.
Zurück zum Zitat Zuo J, Wang DH, Zhang YJ, Liu L, Liu FL, Liu W. Expression and mechanism of PinX1 and telomerase activity in the carcinogenesis of esophageal epithelial cells. Oncol Rep. 2013;30:1823–31.PubMed Zuo J, Wang DH, Zhang YJ, Liu L, Liu FL, Liu W. Expression and mechanism of PinX1 and telomerase activity in the carcinogenesis of esophageal epithelial cells. Oncol Rep. 2013;30:1823–31.PubMed
23.
Zurück zum Zitat Liu JY, Qian D, He LR, Li YH, Liao YJ, Mai SJ, et al. PinX1 suppresses bladder urothelial carcinoma cell proliferation via the inhibition of telomerase activity and p16/cyclin D1 pathway. Mol Cancer. 2013;12:148.CrossRefPubMedCentralPubMed Liu JY, Qian D, He LR, Li YH, Liao YJ, Mai SJ, et al. PinX1 suppresses bladder urothelial carcinoma cell proliferation via the inhibition of telomerase activity and p16/cyclin D1 pathway. Mol Cancer. 2013;12:148.CrossRefPubMedCentralPubMed
24.
Zurück zum Zitat Lai XF, Shen CX, Wen Z, Qian YH, Yu CS, Wang JQ, et al. PinX1 regulation of telomerase activity and apoptosis in nasopharyngeal carcinoma cells. J Exp Clin Cancer Res. 2012;31:12.CrossRefPubMedCentralPubMed Lai XF, Shen CX, Wen Z, Qian YH, Yu CS, Wang JQ, et al. PinX1 regulation of telomerase activity and apoptosis in nasopharyngeal carcinoma cells. J Exp Clin Cancer Res. 2012;31:12.CrossRefPubMedCentralPubMed
25.
Zurück zum Zitat Zhang L, Jiang Y, Zheng Y, Zeng Y, Yang Z, Huang G, et al. Selective killing of Burkitt’s lymphoma cells by mBAFF-targeted delivery of PinX1. Leukemia. 2011;25:331–40.CrossRefPubMedCentralPubMed Zhang L, Jiang Y, Zheng Y, Zeng Y, Yang Z, Huang G, et al. Selective killing of Burkitt’s lymphoma cells by mBAFF-targeted delivery of PinX1. Leukemia. 2011;25:331–40.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Cai MY, Zhang B, He WP, Yang GF, Rao HL, Rao ZY, et al. Decreased expression of PinX1 protein is correlated with tumor development and is a new independent poor prognostic factor in ovarian carcinoma. Cancer Sci. 2010;101:1543–9.CrossRefPubMed Cai MY, Zhang B, He WP, Yang GF, Rao HL, Rao ZY, et al. Decreased expression of PinX1 protein is correlated with tumor development and is a new independent poor prognostic factor in ovarian carcinoma. Cancer Sci. 2010;101:1543–9.CrossRefPubMed
27.
Zurück zum Zitat Shih YW, Chien ST, Chen PS, Lee JH, Wu SH, Yin LT. Alpha-mangostin suppresses phorbol 12-myristate 13-acetate-induced MMP-2/MMP-9 expressions via alphavbeta3 integrin/FAK/ERK and NF-kappaB signaling pathway in human lung adenocarcinoma A549 cells. Cell Biochem Biophys. 2010;58:31–44.CrossRefPubMed Shih YW, Chien ST, Chen PS, Lee JH, Wu SH, Yin LT. Alpha-mangostin suppresses phorbol 12-myristate 13-acetate-induced MMP-2/MMP-9 expressions via alphavbeta3 integrin/FAK/ERK and NF-kappaB signaling pathway in human lung adenocarcinoma A549 cells. Cell Biochem Biophys. 2010;58:31–44.CrossRefPubMed
28.
Zurück zum Zitat Fukuyama R, Ng KP, Cicek M, Kelleher C, Niculaita R, Casey G, et al. Role of IKK and oscillatory NFkappaB kinetics in MMP-9 gene expression and chemoresistance to 5-fluorouracil in RKO colorectal cancer cells. Mol Carcinog. 2007;46:402–13.CrossRefPubMed Fukuyama R, Ng KP, Cicek M, Kelleher C, Niculaita R, Casey G, et al. Role of IKK and oscillatory NFkappaB kinetics in MMP-9 gene expression and chemoresistance to 5-fluorouracil in RKO colorectal cancer cells. Mol Carcinog. 2007;46:402–13.CrossRefPubMed
29.
Zurück zum Zitat Jianfeng D, Feng J, Chaoneng J, Zhongzhou Z, Shaohua G, Qihan W, et al. Cloning of the correct full length cDNA of NF-kappaB-repressing factor. Mol Cells. 2003;16:397–401.PubMed Jianfeng D, Feng J, Chaoneng J, Zhongzhou Z, Shaohua G, Qihan W, et al. Cloning of the correct full length cDNA of NF-kappaB-repressing factor. Mol Cells. 2003;16:397–401.PubMed
30.
Zurück zum Zitat Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25:9–34.CrossRefPubMed Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25:9–34.CrossRefPubMed
31.
Zurück zum Zitat Li HC, Cao DC, Liu Y. Prognostic value of matrix metalloproteinases (MMP2 and MMP9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 2004;88:75–85.CrossRefPubMed Li HC, Cao DC, Liu Y. Prognostic value of matrix metalloproteinases (MMP2 and MMP9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 2004;88:75–85.CrossRefPubMed
32.
Zurück zum Zitat Giannelli G, Erriquez R, Fransvea E, Daniele A, Trerotoli P, Schittulli F, et al. Proteolytic imbalance is reversed after therapeutic surgery in breast cancer patients. Int J Cancer. 2004;109:782–5.CrossRefPubMed Giannelli G, Erriquez R, Fransvea E, Daniele A, Trerotoli P, Schittulli F, et al. Proteolytic imbalance is reversed after therapeutic surgery in breast cancer patients. Int J Cancer. 2004;109:782–5.CrossRefPubMed
33.
Zurück zum Zitat Jinga DC, Blidaru A, Condrea I. MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: correlations with prognostic factors. J Cell Mol Med. 2006;10:499–510.CrossRefPubMed Jinga DC, Blidaru A, Condrea I. MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: correlations with prognostic factors. J Cell Mol Med. 2006;10:499–510.CrossRefPubMed
34.
Zurück zum Zitat Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431–6.CrossRefPubMed Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431–6.CrossRefPubMed
35.
Zurück zum Zitat Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–10.CrossRefPubMed Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–10.CrossRefPubMed
36.
Zurück zum Zitat Kim YR, Kim IJ, Kang TW, Choi C, Kim KK, Kim MS, et al. HOXB13 downregulates intracellular zinc and increases NF-κB signaling to promote prostate cancer metastasis. Oncogene. 2014;33:4558–67.CrossRefPubMed Kim YR, Kim IJ, Kang TW, Choi C, Kim KK, Kim MS, et al. HOXB13 downregulates intracellular zinc and increases NF-κB signaling to promote prostate cancer metastasis. Oncogene. 2014;33:4558–67.CrossRefPubMed
37.
Zurück zum Zitat Jiang L, Lin C, Song L, Wu J, Chen B, Ying Z, et al. MicroRNA-30e* promotes human glioma cell invasiveness in an orthotopic xenotransplantation model by disrupting the NF-kappaB/IkappaBalpha negative feedback loop. J Clin Invest. 2012;122:33–47.CrossRefPubMedCentralPubMed Jiang L, Lin C, Song L, Wu J, Chen B, Ying Z, et al. MicroRNA-30e* promotes human glioma cell invasiveness in an orthotopic xenotransplantation model by disrupting the NF-kappaB/IkappaBalpha negative feedback loop. J Clin Invest. 2012;122:33–47.CrossRefPubMedCentralPubMed
39.
Zurück zum Zitat Lee WR, Chung CL, Hsiao CJ, Chou YC, Hsueh PJ, Yang PC, et al. Suppression of matrix metalloproteinase-9 expression by andrographolide in human monocytic THP-1 cells via inhibition of NF-κB activation. Phytomedicine. 2012;19:270–7.CrossRefPubMed Lee WR, Chung CL, Hsiao CJ, Chou YC, Hsueh PJ, Yang PC, et al. Suppression of matrix metalloproteinase-9 expression by andrographolide in human monocytic THP-1 cells via inhibition of NF-κB activation. Phytomedicine. 2012;19:270–7.CrossRefPubMed
40.
Zurück zum Zitat Aravind L, Koonin EV. G-patch: a new conserved domain in eukaryotic RNA-processing proteins and type D retroviral polyproteins. Trends Biochem Sci. 1999;24:342–4.CrossRefPubMed Aravind L, Koonin EV. G-patch: a new conserved domain in eukaryotic RNA-processing proteins and type D retroviral polyproteins. Trends Biochem Sci. 1999;24:342–4.CrossRefPubMed
41.
Zurück zum Zitat Nourbakhsh M, Oumard A, Schwarzer M, Hauser H. NRF, a nuclear inhibitor of NF-kappaB proteins silencing interferon-beta promoter. Eur Cytokine Netw. 2000;11:500–1.PubMed Nourbakhsh M, Oumard A, Schwarzer M, Hauser H. NRF, a nuclear inhibitor of NF-kappaB proteins silencing interferon-beta promoter. Eur Cytokine Netw. 2000;11:500–1.PubMed
42.
Zurück zum Zitat Bai J, Yong HM, Chen FF, Mei PJ, Liu H, Li C, et al. Cullin1 is a novel marker of poor prognosis and a potential therapeutic target in human breast cancer. Ann Oncol. 2013;24:2016–22.CrossRefPubMed Bai J, Yong HM, Chen FF, Mei PJ, Liu H, Li C, et al. Cullin1 is a novel marker of poor prognosis and a potential therapeutic target in human breast cancer. Ann Oncol. 2013;24:2016–22.CrossRefPubMed
43.
Zurück zum Zitat Bai J, Mei PJ, Liu H, Li C, Li W, Wu YP, et al. BRG1 expression is increased in human glioma and controls glioma cell proliferation, migration and invasion in vitro. J Cancer Res Clin Oncol. 2012;138:991–8.CrossRefPubMed Bai J, Mei PJ, Liu H, Li C, Li W, Wu YP, et al. BRG1 expression is increased in human glioma and controls glioma cell proliferation, migration and invasion in vitro. J Cancer Res Clin Oncol. 2012;138:991–8.CrossRefPubMed
44.
Zurück zum Zitat Bai J, Zhou Y, Chen GD, Zeng JY, Ding JJ, Tan YF, et al. Overexpression of Cullin1 is associated with poor prognosis of patients with gastric cancer. Hum Pathol. 2011;42:375–83.CrossRefPubMed Bai J, Zhou Y, Chen GD, Zeng JY, Ding JJ, Tan YF, et al. Overexpression of Cullin1 is associated with poor prognosis of patients with gastric cancer. Hum Pathol. 2011;42:375–83.CrossRefPubMed
45.
Zurück zum Zitat Chen FF, Bai J, Li W, Mei PJ, Liu H, Li LL, et al. RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS One. 2013;8:e56241.CrossRefPubMedCentralPubMed Chen FF, Bai J, Li W, Mei PJ, Liu H, Li LL, et al. RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS One. 2013;8:e56241.CrossRefPubMedCentralPubMed
46.
Zurück zum Zitat Bai J, Zhang J, Wu J, Shen L, Zeng J, Ding J, et al. JWA regulates melanoma metastasis by integrin alphaVbeta3 signaling. Oncogene. 2010;29:1227–37.CrossRefPubMed Bai J, Zhang J, Wu J, Shen L, Zeng J, Ding J, et al. JWA regulates melanoma metastasis by integrin alphaVbeta3 signaling. Oncogene. 2010;29:1227–37.CrossRefPubMed
Metadaten
Titel
PinX1 inhibits the invasion and metastasis of human breast cancer via suppressing NF-κB/MMP-9 signaling pathway
verfasst von
Meilin Shi
Menghan Cao
Jun Song
Qinghua Liu
Hailong Li
Fei Meng
Zhenqiang Pan
Jin Bai
Junnian Zheng
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2015
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0332-2

Weitere Artikel der Ausgabe 1/2015

Molecular Cancer 1/2015 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

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.

Update Onkologie

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