Skip to main content
Erschienen in: Molecular Cancer 1/2019

Open Access 01.12.2019 | Research

RETRACTED ARTICLE: Activation of LncRNA TINCR by H3K27 acetylation promotes Trastuzumab resistance and epithelial-mesenchymal transition by targeting MicroRNA-125b in breast Cancer

verfasst von: Huaying Dong, Jianguo Hu, Kejian Zou, Mulin Ye, Yuanwen Chen, Chengyi Wu, Xin Chen, Mingli Han

Erschienen in: Molecular Cancer | Ausgabe 1/2019

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Trastuzumab resistance followed by metastasis is a major obstacle for improving the clinical outcome of patients with advanced human epidermal growth factor receptor 2-positive (HER-2+) breast cancer. While long non-coding RNAs (lncRNAs) can modulate cell behavior, the contribution of these RNAs in trastuzumab resistance and metastasis of HER-2+ breast cancer is not well known. In this study, we sought to identify the regulatory role of lncRNA in trastuzumab resistance and accompanied Epithelial-mesenchymal Transition (EMT) process in advanced HER-2+ breast cancer.

Methods

Trastuzumab-resistant SKBR-3-TR and BT474-TR cell lines were established by grafting SKBR-3 and BT474 cells into mouse models and subjected to trastuzumab treatment. LncRNA microarray followed by quantitative reverse transcription PCR (qRT-PCR) was carried out to verify the differentially expressed lncRNAs. Western blotting, bioinformatics analysis, immunofluorescence assay and immunoprecipitation assays (ChIP and RIP) were performed to identify the involvement and functional interactions between H3K27 acetylation and terminal differentiation-induced non-coding RNA (TINCR) or between TINCR and its downstream genes including miR-125b, HER-2 and Snail-1. In addition, a series of in vitro and in vivo assays were performed to assess the functions of TINCR.

Results

An increase in both, IC50 value of trastuzumab and EMT was observed in the established trastuzumab-resistant cell lines. The expression level of TINCR was significantly increased in trastuzumab-resistant cells when compared with sensitive cells. Knockdown of TINCR reversed the trastuzumab resistance and the acquired EMT in these cells. TINCR was detected in the cytoplasm of breast cancer cells and could sponge miR-125b, thereby releasing HER-2 and inducing trastuzumab resistance. In addition, Snail-1 was found to be the target gene of miR-125b and overexpression of Snail-1 could reverse the suppressed migration, invasion, and EMT caused by TINCR silencing. The upregulation of TINCR in breast cancer was attributed to the CREB-binding protein (CBP)-mediated H3K27 acetylation at the promoter region of TINCR. Clinically, HER-2+ breast cancer patients with high TINCR expression levels were associated with poor response to trastuzumab therapy and shorter survival time.

Conclusion

TINCR could promote trastuzumab resistance and the accompanied EMT process in breast cancer. Therefore, TINCR might be a potential indicator for prognosis and a therapeutic target to enhance the clinical efficacy of trastuzumab treatment.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12943-018-0931-9) contains supplementary material, which is available to authorized users.
A Correction to this article is available online at https://​doi.​org/​10.​1186/​s12943-021-01385-9.
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s12943-022-01609-6.
This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12943-022-01609-6"
Abkürzungen
AUC
Area under the curve
CBP
CREB-binding protein
ChIP
Chromatin immunoprecipitation
DAPI
4′,6-diamidino-2-phenylindole
DMEM
Dulbecco’s modified Eagle medium
EMT
Epithelial-mesenchymal Transition
FBS
Fetal bovine serum
FISH
Fluorescence in situ hybridization
GAPDH
Glyceraldehyde 3-phosphate dehydrogenase
HAT
Histone acetyltransferase
HER-2
Human epidermal growth factor receptor 2
IHC
Immunohistochemistry
lncRNA
Long non-coding RNA
MyD88
Myeloid differentiation factor 88
NC
Negative control
PCR
Polymerase chain reaction
RIP
RNA immunoprecipitation
ROC
Receiver operating characteristic
SD
Standard deviation

Background

Cancer therapy is becoming increasingly personalized and molecularly targeted by using biomarkers to identify patients most likely to respond to therapy [1]. Breast cancer patients expressing the human epidermal growth factor receptor-2 (HER-2) protein were traditionally associated with poor prognosis [2]. Several advances have been made in HER-2-targeted treatment among these patients, such as trastuzumab, which is an antibody-drug conjugate that has been approved for the treatment of HER-2+ metastatic breast cancer [3]. However, issues of poor response to therapy and subsequent metastasis have become prevalent in recent years. In fact, only less than 35% of patients with HER-2+ breast cancer initially respond to trastuzumab [4, 5]. Therefore, in addition to new treatment strategies, there is an immediate need for reliable predictive biomarkers to treat patients who will benefit from such treatments.
Long non-coding RNAs (lncRNAs) are a class of poor conserved endogenous RNAs longer than 200 nucleotides that do not encode proteins but regulate gene expression [6]. On a functional level, lncRNAs are involved in complex biological processes through diverse mechanisms. These comprise, among others, gene regulation by titration of transcription factors, alternative splicing, sponging of microRNAs, and recruitment of chromatin modifying enzymes [710]. In addition, lncRNAs can influence cancer progression and chemoresistance in cancer patients, in whom they are dysregulated [11]. Recently, a group of lncRNAs such as UCA1, GAS5 and lncRNA-ATB were identified as critical regulators of trastuzumab resistance [1214]. However, the specific role of lncRNAs in trastuzumab resistance and subsequent metastasis is still not well known.
TINCR, (terminal differentiation-induced non-coding RNA) is a spliced, long non-coding RNA that produces a 3.7 kb transcript. It is isolated from human somatic tissues that are well-differentiated and is required for normal epidermal differentiation [15]. There are many evidences to suggest that aberrant expression of TINCR is associated with a variety of human cancers [1619]. Although Liu et al. revealed an oncogenic role for TINCR in breast cancer [20], whether TINCR plays a role in trastuzumab resistance and resistance-induced metastasis is not defined.
In our previous study, we identified some specific lncRNAs that might participate in trastuzumab resistance [2123]. In this study, we established trastuzumab-resistant cell lines by planting SKBR-3 and BT474 cells into nude mice and performed courses of trastuzumab treatment in vivo. We compared the lncRNA expression in trastuzumab-resistant cells and parental cells using microarray analysis. Among the significantly dysregulated lncRNAs, we selected TINCR lncRNA because it was associated with HER-2 expression [24]. We verified that TINCR was upregulated in chemoresistant cells in contrast to the un-treated parental cells. Functionally, knockdown of TINCR partially reversed resistance to trastuzumab and the accompanied epithelial-mesenchymal transition (EMT) by the regulation of miR-125b targeting HER-2 and Snail-1, respectively. Moreover, upregulation of TINCR was attributed to transcriptional activation by H3K27 acetylation (H3K27ac) enrichment. Clinically, TINCR was correlated with poor prognosis of breast cancer patients who received trastuzumab therapy.

Methods

Ethics statement and tissue samples

The study included 60 patients with HER-2+ breast cancer (female/male: 60/0, range of age (median, years): 27–63 (45)) who underwent surgical resection followed by trastuzumab treatment at Hainan General Hospital, The Fifth People’s Hospital of Chongqing, The Frist Affiliated Hospital of Chongqing Medical University and The First Affiliated Hospital of Zhengzhou University between Jan 2010 and Jun 2013. The diagnosis of recruited patients was pathologically confirmed, and primary cancer tissues were collected before performing trastuzumab treatment. The obtained upon resection tissue samples were immediately snap-frozen in liquid nitrogen and then stored at − 80 °C until further use. This study was approved by Research Scientific Ethics Committee of Hainan General Hospital, The Fifth People’s Hospital of Chongqing, The Frist Affiliated Hospital of Chongqing Medical University and The First Affiliated Hospital of Zhengzhou University. All participants signed informed consent prior to using the tissues for scientific research.

Cell culture and reagents

The human HER-2+ breast cancer cell lines SKBR-3, BT474 and human normal breast epithelial cell line MCF-10A were purchased from American Tissue Culture Collection (ATCC, Manassas, VA, USA). SKBR-3 and BT474 cells were cultured in Dulbecco’s modified Eagle (DMEM, Gibco, Carlsbad, CA) medium with 10% fetal bovine serum (FBS) (Gibco BRL, Grand Island, NY, USA). Normal breast epithelial MCF-10A cells were grown in DMEM/F-12 medium (HyClone) containing 10% FBS, 100 ng/ml cholera toxin (Sigma-Aldrich, St Louis, MO, USA), 5 μg/ml hydrocortisone (Sigma-Aldrich) and 10 μg/ml insulin (Sigma-Aldrich). The cultures were incubated at 37 °C in 5% CO2. Trastuzumab (Herceptin) was purchased from Roche (Shanghai, China) and dissolved in enclosed sterile water.

Establishment of trastuzumab-resistant cell lines

The trastuzumab-resistant cell lines were established according to the method as previously reported [25]. Briefly, 5 × 106 SKBR-3 or BT474 cells were injected subcutaneously into the flanks of nude mice. When the volume of xenograft reached 200 mm3, mice were intraperitoneally injected either with trastuzumab (3 mg/kg) or PBS once every two days for two weeks followed by another two weeks without the drug treatment (one course). Altogether, the mice received four courses of trastuzumab treatment and breast cancer cells were isolated from xenografts after completion of four courses of treatment and confirmed for resistance to trastuzumab.

Expression profile analysis of lncRNAs

Total RNA was extracted from trastuzumab-resistant cells and parental cells by using the RNeasy plus mini kit (Qiagen, Waltham, MA) according to the manufacturer’s protocol. LncRNAs were sequenced and microarray was performed using Agilent human lncRNA microarray V.2.0 platform (GPL18109). The data were analyzed by GeneSpring 12.6 software (Agilent) and the raw signals were log transformed and normalized using the Percentile shift normalization method, the value was set at 75th percentile. cDNA was fragmented (Bioruptor, Diagenode) to an average size of 250 bp to build the cDNA library. Data processing and statistical analysis for RNA-sequencing data were performed and heat maps were generated.

Vector construction and cell transduction

The synthetic oligonucleotides used for silencing TINCR (sh-TINCR) and oligonucleotides for overexpression of TINCR (Lv-TINCR) were synthesized by Sangon Biotech Co. Ltd. (Shanghai, China). Negative control sh-NC or Lv-NC were also obtained from Sangon Biotech Co. Ltd. The above synthetic oligonucleotides were cloned into lentiviral vector to guarantee stable infection. Ribo™ Biotech (Guangzhou, China) synthesized the overexpression plasmid containing Snail-1 coding sequences (p-Snail-1) and mimics of miR-125b and anti-miR-125b. Cells were transduced with the above vectors by using TransFast transfection reagent (Promega; Madison, WI, USA) according to the manufacturer’s protocol. A total of 5 × 105 cells were seeded into each well of a 6-well plate and transfected with respective oligoribonucleotides (final concentration 100 nM) upon reaching 70 to 80% confluence. The altered expression of target genes was measured after 24 h of transfection. The cells were then subjected to RNA/protein extraction and further functional assays.

Reverse transcription (RT) and quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted from breast cancer tissues or cells by using the RNeasy plus mini kit (Qiagen) according to the manufacturer’s protocol. RT and qPCR kits were used to evaluate the expression of target RNAs. RT (20 μl) reactions were performed using the PrimeScript® RT reagent kit (Takara, Dalian, China) and incubated for 30 min at 37 °C followed by 5 s at 85 °C. For qPCR, 2 μl of diluted RT product was mixed with 23 μl reaction buffer provided by Takara (Takara Inc., Dalian, China) to a final volume of 25 μl. All reactions were carried out using an Eppendorf Mastercycler EP Gradient S (Eppendorf, Germany) under the following conditions: 95 °C for 30 s followed by 45 cycles of 95 °C for 5 s and 60 °C for 30 s. The internal expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used for the normalization of detected RNAs using the comparative 2-ΔΔCq method. The primer sequences for qPCR are presented in Additional file 1: Table S1.

Immunofluorescence

Cells were permeabilized with 0.3% Triton X-100 (Beyotime, Shanghai, China) for 15 min after being fixed with 4% paraformaldehyde. The cells were blocked by using goat serum followed by incubation with anti-Ki67 antibody (1:100, ab15580, Abcam, Cambridge, MA) overnight at 4 °C. Subsequently, the slides were incubated with anti-rabbit Alexa Fluor 488 (Jackson Immunoresearch, West Grove, PA, USA) for 1 h at room temperature. DAPI was used for nuclear counterstaining. The slides were observed under a fluorescence microscope (DMI4000B, Leica).

Cell viability assay

The altered cell viability after treatment with trastuzumab or (and) sh-TINCR was assayed using the MTT Kit (Dojindo, Rockville, MD, USA). Cells were seeded onto 96-well plates at a density of 3000 cells/well and cultured in 200 μL cell culture medium. Ten microlitre MTT (5 mg/mL, pH = 7.4, pre-pared with PBS) was added to culture the cells for 2 h. Afterthe medium was turned away, the precipitate was made soluble in 100 μL DMSO. An enzyme-linked immunosorbent plate reader was utilized to determine the absorbance of each well.

Cell migration and invasion assay

Cell migration ability was evaluated by performing wound-healing assay. Cells were seeded onto six-well plates at a density of 500,000 cells/well. Twelve hours after treatment with trastuzumab or transduction with respective vectors, the layer of cells was scratched to form wounds by using a sterile 20-μl pipette tip; the non-adherent cells were washed away with culture medium and then the cells were further incubated for 48 h and photographed to identify the gap area. Cell invasive ability was evaluated using the transwell invasion assay with Boyden chambers (BD Biosciences) that had 8 μm pore size membranes with Matrigel. Cells in serum-free media were placed in the upper chamber of an insert. Medium containing 10% FBS was added to the lower chamber. After 12 h of incubation, the cells that had invaded through the membrane were stained with methanol and 0.1% crystal violet and imaged using an inverted microscope (Leica, DM IRBE).

Immunohistochemical (IHC) staining and scoring analyses

Immunohistochemical staining was performed on 4 μm-thick TMA slides as previously described [23]. Anti-HER-2 antibody (1:100, cat. no. ab16901, Abcam, Cambridge, MA) and anti-Snail-1 antibody (1:100, cat. no. ab53519, Abcam) were used to detect their protein level in xenografts tissues. Images were visualized using a Nikon ECLIPSE Ti (Tokyo, Japan) microscope system and processed using Nikon software.

Nucleocytoplasmic separation

The PARIS™ kit (Ambion, Austin, TX) was used for the nucleo-cytoplasmic separation experiment. Briefly, 5 × 106 cells were re-suspended in 0.6 ml resuspension buffer and incubated for 15 min followed by homogenization. After centrifugation at 400×g for 15 min, the cytoplasmic fraction was obtained in the supernatant. The pellet was then resuspended in 0.3 ml PBS, 0.3 ml nuclear isolation buffer, and 0.3 ml RNase-free H2O, followed by 20 min incubation on ice. The pellet was the nuclear fraction after centrifugation. TINCR expression was determined by qPCR with GAPDH as cytoplasmic control and U1 as nuclear control. The primers used are shown in Additional file 1: Table S1.

Fluorescence in situ hybridization analysis (FISH)

Sangon Biotech synthesized the specific TINCR probe. Briefly, the cells were fixed in 1 ml of 4% formaldehyde for 10 min at room temperature, washed twice with 1× PBS and permeabilized with 70% EtOH in two-chamber dishes. The probes (0.3–0.6 μM final concentration) were hybridized in 10% dextran sulfate (Sigma, cat. no. D8906), 10% formamide and 2× SSC at 37 °C overnight followed by thorough washing. Imaging was performed immediately using a fluorescence microscope (DMI4000B, Leica).

RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP)

For RIP assay, cells were rinsed with cold PBS and fixed in 1% formaldehyde for 10 min. After centrifugation (1500×g for 15 min at 4 °C), cell pellets were collected and re-suspended in NP-40 lysis buffer. The RIP assay was performed using the Magna RIP™ RNA-Binding Protein Immunoprecipitation Kit (Millipore, Billerica, MA, USA) according to the manufacturer’s instructions. Briefly, cells were harvested and lysed in RIP lysis buffer. RNA was immunoprecipitated with antibody against Ago2 (Abcam, cat. no. ab32381), HER-2 (Abcam, cat. no. ab16901) or negative control IgG (EMD Millipore, cat. no. 12–371, Burlington, MA, USA).
An EZ-Magna ChIP kit (Millipore) was used for the ChIP assay according to the manufacturer’s protocol. Briefly, cells were treated with formaldehyde and incubated for 10 min to generate DNA–protein cross-links. Cell lysates were then sonicated to generate chromatin fragments of 200–300 bp and immunoprecipitated with H3K27 antibody (Abcam, cat. no. ab4729), CBP antibody (Abcam, cat. no. ab2832) or the negative control IgG antibody (EMD Millipore, cat. no. 12–371). RNA was recovered and analyzed by qPCR.

Western blots and antibodies

RIPA buffer (Sigma Aldrich, Cambridge, MA) was used to lyse the cells to obtain total protein lysates. Protein concentration was measured using the BCA method (Sigma Aldrich). The quantified protein (25 μg) was transferred onto polyvinylidene fluoride (PVDF) membranes following SDS-PAGE gel electrophoresis. Then, the membrane was blocked with 5% nonfat dry milk in tri-buffered saline plus Tween (TBS-T) buffer for 2 h at room temperature and incubated with respective primary antibodies (1:1000 dilution) at 4 °C overnight, followed by Horseradish peroxidase-conjugated (HRP) secondary antibody (1:5000, Abcam, cat. no. ab7090) at room temperature for 1 h. The following primary antibodies were used: anti-HER-2 antibody (Abcam, cat. no. ab227383), anti-E-cadherin antibody (Abcam, cat. no. ab186533), anti-Snail-1 antibody (Abcam, cat. no. ab8614), anti-N-cadherin antibody (Abcam, cat. no. ab182651), anti-vimentin antibody (Abcam, cat. no. ab8805), anti-β-catenin antibody (Abcam, cat. no. ab8932), anti-GAPDH antibody (Invitrogen, cat. no. PA1–987).

In vivo animal experiment

Ten male BALB/c nude mice (19–22 g, 6 weeks old) were obtained from the Animal Center of Chinese Academy of Science (Shanghai, China). They were randomly divided into two groups of five each and housed three per cage in pathogen-free conditions at 28 °C, 50% humidity and were housed in a specific sterile environment suitable and regularly observed. The experimental protocol was approved by the Committee on the Ethics of Animal Experiments of Hainan General Hospital. SKBR-3-TR cells (1 × 107) that were stably transduced with sh-TINCR or sh-NC were subcutaneously injected into the flanks. The mice were housed for 25 days, then the formed tumors were stripped and the tumor mass was measured.
Experimental lung metastases were induced by injections of single-cell suspension (2 × 106 cells in 100 μl) into the mouse lateral tail vein. Cells were stably transduced with sh-TINCR or sh-NC, and all cell injections were administered in a total volume of 500 μL PBS containing 0.1% BSA over a 60 s duration [26]. Five weeks later, prior to in vivo imaging, the mice were anaesthetized with phenobarbital sodium and the established lung metastases images were observed by LB983 NIGHTOWL II system (Berthold Technologies GmbH, Calmbacher, Germany).

Statistical analysis

Kolmogorov-Smirnov test was applied for data analysis with the distribution of each group samples. Data were presented as median (interquartile range). Mann-Whitney U test was carried out to compare the datasets of the two groups. The Kruskal-Wallis test followed by post-hoc test with Bonferroni correction was used for evaluating the difference among multiple groups. Receiver operation characteristic (ROC) analysis was performed to evaluate the diagnostic performance of TINCR. The correlation between TINCR and miR-125b expression and TINCR and Snail-1 expression was analyzed using Spearman’s correlation test. Kaplan-Meier analysis was performed to determine the prognostic performance of TINCR. A two-sided P < 0.05 was considered as statistically significant. Statistical analysis was performed using Prism 5 (GraphPad Software Inc., San Diego, CA, USA).

Results

Identification of trastuzumab resistance and resistance-induced EMT in breast cancer cells

Trastuzumab-resistant breast cancer cell lines were established by grafting SKBR-3 and BT474 cells into nude mice followed by cycles of trastuzumab treatment (3 mg/kg, intraperitoneal injection) as described in Methods (Fig. 1a). Breast cancer xenografts that received the fourth course of trastuzumab treatment displayed poor response to trastuzumab therapy. Cells were isolated from these resistant xenografts and named SKBR-3-TR and BT474-TR, respectively. We then validated the trastuzumab-resistant feature of these two sub-lines. As shown in Fig. 1b, the cell viability of both trastuzumab-resistant cell lines was much higher than their respective parental cell lines after treatment with trastuzumab at 3 μg/ml for 48 h. Furthermore, the IC50 values of trastuzumab were much higher in trastuzumab-resistant cell sub-lines when compared to the respective parental ones (Fig. 1c). A different morphological feature was identified between the trastuzumab-resistant cells and parental cells (Fig. 1d). The resistant cells showed increased intercellular separation and formation of pseudopodia, suggesting that these cells may undergo EMT. Moreover, we used TUNEL assay to assess apoptosis after treatment with trastuzumab (3 μg/ml for 48 h). We observed a dramatic decrease in TUNEL intensity in trastuzumab-resistant cells when compared to the respective parental cells (Fig. 1e). It is well-known that chemoresistant cells treated by chemotherapeutics (e.g. oxaliplatin, 5-fluorouracil, etc.) undergo EMT; however, whether this phenomenon applies to cells treated with monoclonal antibody (e.g. trastuzumab) is still unknown. We investigated the migratory and invasive abilities of these cells and demonstrated that both trastuzumab-resistant cell lines showed enhanced migratory and invasive capacities (Fig. 1f, g). In addition, the epithelial proteins (E-cadherin and β-catenin) were downregulated whereas mesenchymal proteins (vimentin, N-cadherin) were upregulated in trastuzumab-resistant cells when compared to non-resistant cells (Fig. 1h). This suggests that trastuzumab resistance induces EMT of breast cancer cells.

LncRNA TINCR is upregulated in trastuzumab-resistant cells

To identify the potential lncRNA that may influence trastuzumab resistance in breast cancer cells, we performed microarray analysis by using trastuzumab-resistant cells and respective parental cells. According to the microarray data, we identified 187 transcripts that were upregulated with more than 2-fold change in SKBR-3-TR cells in contrast to SKBR-3 cells while 66 transcripts were downregulated by more than 2-fold. Moreover, we identified 224 transcripts that were upregulated with more than 2-fold change and 78 transcripts that were downregulated by 2-fold in BT474-TR cells in contrast to BT474-TR cells (Fig. 2a). Among these upregulated lncRNAs, we focused on TINCR, which has been previously reported to be linked to HER-2 expression and breast cancer tumorigenesis [24]. We validated the expression of TINCR in trastuzumab-resistant cells by using qPCR. As shown in Fig. 2b, TINCR is significantly upregulated in SKBR-3-TR and BT474-TR cells than in SKBR-3 and BT474 cells, respectively. To further validate the upregulation of TINCR, we included 30 tissue samples from HER-2+ breast cancer patients exhibiting poor response to trastuzumab therapy and another 30 tissue samples from HER-2+ breast cancer patients responding to trastuzumab therapy. As expected, TINCR expression was increased in trastuzumab-resistant patients compared to trastuzumab-responsive patients (Fig. 2c).

Silencing TINCR reverses trastuzumab resistance of breast cancer cells

We then investigated the biological function of TINCR by constructing two kinds of shRNAs and cloned them in lentiviral vectors. We observed that both shRNAs induced significant downregulation of TINCR in SKBR-3-TR and BT474-TR cells (Fig. 3a). The MTT assay demonstrated that TINCR silencing significantly promotes trastuzumab-induced inhibition of cell viability (Fig. 3b). As trastuzumab mainly affects HER-2-regulated cell proliferation, we detected the cell proliferation protein Ki67 in these cells. Immunofluorescence assay revealed that TINCR knockdown decreased Ki67 expression level in both trastuzumab-resistant cell lines (Fig. 3c). In addition, TINCR knockdown damaged the acquired cell migration and invasion properties of trastuzumab-resistant cells as evidenced by wound healing and the matrigel-transwell assay (Fig. 3d, e). Western blot analysis of EMT proteins revealed that TINCR knockdown suppressed EMT in trastuzumab-resistant cells (Fig. 3f).
Furthermore, we overexpressed TINCR in parental SKBR-3 and BT474 cells by transduction of lentiviral vector containing TINCR sequence (Lv-TINCR) (Fig. 3g). We treated the cells with trastuzumab for 48 h at a concentration of 3 μg/ml and found that the cells transduced with Lv-TINCR decreased the trastuzumab-induced cell cytotoxicity compared to that of Lv-NC-transduced cells (Fig. 3h). Interestingly, we found that enhanced TINCR expression showed minimal effect on cell migration, invasion, and EMT of SKBR-3 and BT474 cells (data not shown), indicating that TINCR is a critical regulator of trastuzumab resistance and resistance-induced EMT process.

TINCR promotes HER-2 expression, thereby induces trastuzumab resistance

As TINCR is associated with trastuzumab resistance, we hypothesized that TINCR may regulate HER-2 expression. To confirm this hypothesis, we measured the expression level of HER-2 in cells by performing immunofluorescence assay. As shown in Fig. 4a, HER-2 expression was increased in both the trastuzumab-resistant cell lines. To further validate the upregulated HER-2 expression that was induced by TINCR, we detected the expression level of HER-2 in SKBR-3-TR and BT474-TR cells upon TINCR knockdown. HER-2 expression was significantly inhibited at both transcript and protein levels in these cells (Fig. 4b). Similarly, overexpression of TINCR in SKBR-3 and BT474 cells increased HER-2 expression (Fig. 4c). Our data suggests that TINCR induces trastuzumab resistance by increasing HER-2 expression.

TINCR regulates HER-2 expression by sponging miR-125b

LncRNA in cytoplasm could compete for MREs with the driver genes closely related to cancer occurrence and development as competing endogenous RNAs (ceRNAs), which would weaken the inhibition of miRNA upon target genes, indirectly regulate the expression level of target genes. To elucidate the regulatory mechanism of TINCR, we identified the subcellular location of TINCR in breast cancer cells. qPCR analysis of nuclear and cytoplasmic lncRNA showed that TINCR was mainly detected in the cytoplasm of SKBR-3-TR and BT474-TR cells (Fig. 5a). By performing fluorescence in situ hybridization of TINCR, we found that TINCR in breast cancer cells was mainly located in the cytoplasm (Fig. 5b). MiRNAs are present in the form of miRNA ribonucleoprotein complexes (miRNPs) which contains AGO2, a core component of the RNA-induced silencing complex [27]. Hence, we performed RIP assay with Ago2 to verify whether TINCR exerts its function through sponging with miRNAs. Clearly, both TINCR and HER-2 were enriched with Ago2 (Fig. 5c). In addition, TINCR knockdown led to decreased enrichment of Ago2 and TINCR but showed an increased recruitment of Ago2 to HER-2 transcript (Fig. 5d), suggesting that there is a competition between TINCR and HER-2 genes for the silencing effects induced by Ago2-based miRNAs.
Subsequently, we sought to identify the miRNA that is responsible for sponging TINCR and HER-2. Interestingly, the microRNA miR-125b was predicted to target both HER-2 and TINCR according to miRcode (http://mircode.org/) (Fig. 5e). TINCR knockdown increased miR-125b expression (Fig. 5f). Moreover, co-transfection of anti-miR-125b abrogated the downregulated the expression of HER-2 that was induced by TINCR knockdown (Fig. 5g). Luciferase reporter assay showed that enhanced expression of miR-125b significantly suppressed the luciferase activity in both TINCR and HER-2 wild type reporters but was relatively unaffected in presence of mutant HER-2 reporter (Fig. 5h-j). RIP assay revealed that TINCR silencing promoted the enrichment of miR-125b bound to HER-2 (Fig. 5k). Collectively, our data indicates TINCR as a molecular sponge for miR-125b to modulate HER-2 expression.

TINCR promotes trastuzumab resistance-induced EMT by directly targeting Snail-1

We further investigated the underlying mechanism by which TINCR mediates the EMT of trastuzumab-resistant cells. As we identified miR-125b as a sponge target of TINCR, we were interested to see whether other downstream mRNA targets of miR-125b are involved in the EMT process. Among the potential targets, we focused on Snail-1, which is well-known as a crucial regulator of EMT [28]. Bioinformatics analysis using miRcode indicated a potential binding site of miR-125b at the 3′UTR of Snail-1 (NM_005985.3, position: 1245–1251) (Fig. 6a). Overexpression of miR-125b significantly suppressed Snail-1 expression in SKBR-3-TR and BT474-TR cells (Fig. 6b). In contrast, anti-miR-125b reversed the TINCR knockdown-induced inhibition of Snail-1 expression (Fig. 6c). Furthermore, co-expression of p-Snail-1 dramatically rescued Snail-1 expression that was -downregulated by sh-TINCR (Fig. 6d), as well as the migratory and invasive abilities of SKBR-3-TR and BT474-TR cells (Fig. 6e-f). Western blot analysis showed that enhanced Snail-1 expression dramatically abrogated the sh-TINCR-induced suppression of EMT in both trastuzumab-resistant cell lines (Fig. 6g). To conclude, our data indicates that Snail-1 is a functional target of TINCR/miR-125b in the regulation of trastuzumab resistance-induced EMT.

TINCR is transcriptionally activated by H3K27 acetylation in breast cancer

Recent studies have shown that aberrant expression of lncRNAs is attributed to acetylation mediated-transcriptional activation [29]. To further understand the reason for increased expression of TINCR in trastuzumab-resistant cells, we explored the probable mechanisms using genome bioinformatics analysis (http://genome.ucsc.edu/), and found that the promoter region of TINCR had high enrichment of H3K27ac (Fig. 7a). To verify the existence of histone acetylation, we used ChIP experiment. As shown in Fig. 7b, there is H3K27ac enrichment at the promoter region of TINCR gene in both trastuzumab-resistant cells and parental cells. Moreover, the enriched intensity of H3K27ac was dramatically higher in trastuzumab-resistant cells compared to the respective parental cells. We also performed ChIP assay using five tissues from trastuzumab-resistant patients and five tissues from trastuzumab-responsive patients, and observed TINCR expression to be aberrantly upregulated in resistant tissues compared to responsive tissues. As expected, the resistant tissues showed an increased enrichment level when compared to that in responsive tissues (Fig. 7c). Moreover, treatment with histone acetyltransferase (HAT) inhibitor C646 significantly decreased the expression level of TINCR (Fig. 7d).
It is well known that CREB-binding protein (CBP) is essential for chromatin acetylation, we then performed ChIP assay and confirmed the enrichment of CBP at the TINCR promoter (Fig. 7e). In addition, knockdown of CBP using specific siRNA (Fig. 7f) dramatically decreased the enrichment of H3K27ac at the TINCR promoter region (Fig. 7g), thereby downregulating TINCR (Fig. 7h). Together, our results strongly indicate that the upregulated levels of TINCR are due to histone acetylation at the TINCR promoter region mediated by CBP enzyme.

Silencing of TINCR impairs trastuzumab resistance and metastasis in vivo

To validate the functional role of TINCR in vitro, we established in vivo xenografts in BALB/cnude mice by grafting SKBR-3-TR cells that are stably transduced with sh-TINCR or sh-NC. When tumors were palpable, mice were intraperitoneally injected with 3 mg/kg trastuzumab once every two days for three weeks. The mouse models and tumor xenografts stripped from nude mice are presented in Fig. 8a and b, respectively. The quantitative data showed that TINCR-silenced mice exhibited significantly less tumor growth than did mice transduced with sh-NC (Fig. 8c). In addition, both the luciferase flux count of lung metastases and visible number of metastases on the lung surface were significantly less in sh-TINCR group compared to sh-NC group (Fig. 8d, e). Moreover, qPCR analysis showed that the expression of TINCR, HER-2, and Snail-1 in implanted breast cancer tissues was dramatically decreased, while miR-125b level was significantly increased in the sh-TINCR group in contrast to that in the sh-NC group (Fig. 8f). Immunohistochemical analysis further revealed a significant downregulation of HER-2 and Snail-1 in the tumor tissues of the sh-TINCR group compared with that in the sh-NC group (Fig. 8g). To conclude, our results confirmed the functional role of TINCR in trastuzumab resistance in in vivo xenografts.

High TINCR expression is associated with poor prognosis of patients receiving trastuzumab therapy

Finally, we performed a preliminary study to find the clinical role of TINCR in 60 primary breast cancer tissues from patients receiving trastuzumab treatment (30 responsive and 30 non-responsive cases according to the Immune-related Response Evaluation Criteria In Solid Tumors (irRECIST) [30]). The clinicopathological analysis showed that expression of TINCR correlated with advanced TNM stage, lymph node invasion, and distant metastasis (Additional file 2: Table S2). As TINCR expression was upregulated in non-responsive patients compared to responsive patients (previously shown in Fig. 2c), we performed ROC curve analysis to investigate the predictive value of TINCR in differentiating trastuzumab responsive patients from non-responsive patients. As shown in Fig. 9a, area under the curve (AUC) was 0.833 with the diagnostic sensitivity and specificity reaching 76.7 and 70.0% with the cut-off value of 0.138, respectively. Under this criterion (0.138), we divide the patients into low- and high-TINCR expression groups and found that the number of patients with low TINCR expression was significantly higher in the responsive group than in the non-responsive group (Fig. 9b). Kaplan-Meier analysis showed that patients exhibiting a high level of TINCR were correlated with shorter overall survival time and progression-free survival time (Fig. 9c). In addition, Cox proportional-hazards model based univariate and multivariate analysis for progression-free survival showed that TINCR is an independent prognostic factor (Additional file 3: Table S3). By analyzing TCGA breast cancer dataset with the online database Starbase (http://​starbase.​sysu.​edu.​cn/​panCancer.​php), we identified positive correlations between the RNA expression of TINCR and HER-2, TINCR and Snail-1 in breast cancer, while a negative correlation was found between TINCR and miR-125b expression (Fig. 9d). More importantly, the correlation coefficients were much higher in our 30 non-responding samples (Fig. 9e), which strongly supports the regulatory role of TINCR in trastuzumab resistance.
Collectively, our study demonstrates that the H3K27 acetylation-induced upregulation of TINCR sponged miR-125b, thereby inducing trastuzumab resistance by upregulating HER-2 expression and promoting trastuzumab resistance-induced EMT process by increasing Snail-1 expression (Fig. 9f). Therefore, TINCR might serve as a potential treatment target for breast cancer patients to enhance the benefit of trastuzumab treatment.

Discussion

Numerous studies in recent years have helped to gain a better understanding of the molecular mechanisms during cancer progression and chemoresistance. However, the specific regulatory model is still largely unknown in cancer, one such being breast cancer. Therefore, it is of much importance to discover new molecular signatures which may be useful for improving the therapeutic efficacy. To this end, we screened potential lncRNAs that may be crucial for trastuzumab resistance in breast cancer. We verified that lncRNA TINCR was significantly upregulated in trastuzumab-resistant cells compared to sensitive parental cells. Moreover, TINCR could promote trastuzumab resistance by directly targeting HER-2 and inducing EMT process via targeting Snail-1 in a miR-125b-dependent manner. Clinically, increased TINCR expression was associated with shorter survival time in breast cancer patients receiving trastuzumab therapy.
Plenty studies have demonstrated that breast cancer patients with mutated HER-2 are correlated with poor survival [31]. Slamon et al. initially demonstrated the association between HER-2 amplification and poor prognosis and related studies following this found that breast cancer patients in Asia-Pacific regions were associated with worse clinical outcomes due to high occurrence of HER-2 amplification [3234]. Trastuzumab is the first Food and Drug Administration (FDA)-approved targeted and personalized drug for HER-2+ metastatic breast cancer. Addition of trastuzumab to adjuvant chemotherapy has dramatically reduced the risk of recurrence and has become a standard treatment for HER-2+ patients [35]. However, the presence of acquired and de novo resistance is a serious concern. In addition, clinical metastasis is always associated with the occurrence of chemoresistance and the underlying mechanism for this resistance, needs comprehensive investigation. Recent findings showed that chemoresistant cells undergo EMT or mesenchymal-like transition, an important process by which cancer cells may potentially acquire chemoresistance [36]. Resistant cells may switch their “molecular machinery” from a proliferative, epithelial phenotype to a more invasive and migratory mode. Because proliferation is required for most drug-induced cytotoxicity, the decrease in proliferation along with an increase in invasion may be one way, whereby resistant cells can escape the effects of therapy [37]. In this study, the two trastuzumab-resistant cell lines established in nude mice were verified to have an enhanced invasive ability and EMT, which is consistent with previous reports. By using these two resistant cell lines, we hope to identify the molecular pathway that might be critical for trastuzumab resistance and EMT.
The functional role of lncRNAs in cancer progression and resistance has been widely investigated. In our previous study, we have identified lncRNA SNHG14 and AGAP2-AS1 as an important regulator of trastuzumab resistance via regulating several downstream pathways [21, 23]. However, not all pathways controlled by lncRNAs established a direct functional link to HER-2, the target gene of trastuzumab therapy, and the translational relevance was thereby weakened. Therefore, we sought to find lncRNAs that may have direct connections with HER-2 expression. By probing for abnormally expressed lncRNAs in trastuzumab-resistant cells compared to non-resistant cells, we identified TINCR to be significantly upregulated. We found that TINCR is essential for trastuzumab resistance and resistance-accompanied EMT process, whereas minimal effect was observed in the migration and invasion of parental cells. More importantly, gain- or loss-function assays showed that silencing TINCR dramatically suppressed HER-2 expression at both transcript and protein levels, which strongly indicated that TINCR may be a critical regulator of trastuzumab resistance and a potential therapeutic target for trastuzumab therapy.
To further uncover the regulatory mechanism by which TINCR modulates HER-2 expression and EMT, we examined the subcellular location of TINCR and identified it in the cytoplasm. As TINCR also regulates the expression of HER-2 transcripts, we hypothesized that TINCR may promote HER-2 expression by serving as a miRNA sponge. Previous reports showed that TINCR could serve as a ceRNA and sponge miR-375 in gastric cancer [38]. In lung cancer, Liu et al. demonstrated that TINCR suppresses proliferation and invasion by regulating miR-544a/FBXW7 axis [39]. As expected, the RIP and luciferase reporter assays validated that TINCR regulated HER-2 expression by sponging miR-125b, which is widely studied and verified as a potential tumor suppressor gene in glioma [40], bladder cancer [41], breast cancer [42], osteosarcoma [43], hepatocellular carcinoma [44], and melanoma [45]. More importantly, Ferracin et al. revealed that miR-125b suppressed cancer progression through direct regulation of ERBB2/HER-2 expression [46], suggesting a direct interaction between miR-125b and HER-2, which is consistent with our results.
Cells resistant to the treatment of chemo-drugs undergo EMT. We attempted to unravel the molecular switch of TINCR controlling this malignant phenotype and elucidate the underlying mechanisms of metastatic invasion in breast cancer. Snail-1 belongs to the Snail superfamily of zinc-finger transcription factors, which also includes Snail-2 (Slug) and Snail-3 (Smuc). Snail-1 has a pivotal role in the regulation of EMT, chemo and immune resistance in cancer. Our results confirmed the direct interaction between miR-125b and Snail-1 as evidenced by RIP and gain- or loss-function assays. In addition, dysregulated Snail-1 could reverse the trastuzumab resistance-related metastasis and EMT process. The functional role of TINCR was also validated by using an in vivo mouse xenograft model. Therefore, we demonstrate that TINCR induces trastuzumab resistance and the accompanied EMT process by promoting HER-2 and Snail-1, respectively.
We also explored the reason for the upregulated expression of TINCR in trastuzumab-resistant cells. Recent studies revealed that alteration of chromatin structure via the various modifications is the major factor that controls gene expression in a temporal and spatial manner, resulting in the establishment and maintenance of epigenetic cellular memory [47]. We analyzed the promoter region of TINCR by genome bioinformatics analysis (http://genome.ucsc.edu/) and identified that H3K27ac was highly enriched in this region. Histone proteins have long flexible N-terminal tails that are subject to several covalent modifications, including acetylation. Many Lys residues of histones are involved in interacting with DNA, and this acetylation neutralizes the positive charge of Lys, leading to the weakening of the DNA-histone interaction and subsequent activation of transcription [48]. Luckily, the ChIP results confirmed the enrichment of H3K27ac at the TINCR promoter region, and the enriched concentration was dramatically increased in trastuzumab-resistant cells when compared to non-resistant cells. Notably, the enrichment of H3K27ac was mediated by CBP, which strongly supports a CBP-mediated histone acetylation regulation at TINCR promoter region [49, 50].

Conclusions

We verified the prognostic influence of TINCR and showed that high TINCR expression was associated with poor survival in patients receiving trastuzumab therapy. In summary, the present study demonstrates that H3K27ac-activated TINCR regulates miR-125b-HER-2/Snail-1 pathway, which contributes to trastuzumab resistance and accompanied EMT. Our findings suggest the TINCR might be a potential target for the prediction and treatment of HER-2+ breast cancer.

Acknowledgements

Not applicable.

Funding

This study is supported by National Science Foundation of China(81702557, 81860101); Hainan provincial health fund(02A2150014P1).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
This study was approved by Research Scientific Ethics Committee of Hainan General Hospital, The Fifth People’s Hospital of Chongqing, The Frist Affiliated Hospital of Chongqing Medical University and The First Affiliated Hospital of Zhengzhou University. All participants signed informed consent prior to using the tissues for scientific research.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

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.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
Zurück zum Zitat Veneziani BM, Criniti V, Cavaliere C, Corvigno S, Nardone A, Picarelli S, Tortora G, Ciardiello F, Limite G, De Placido S. In vitro expansion of human breast cancer epithelial and mesenchymal stromal cells: optimization of a coculture model for personalized therapy approaches. Mol Cancer Ther. 2007;6(12 Pt 1):3091–100.PubMedCrossRef Veneziani BM, Criniti V, Cavaliere C, Corvigno S, Nardone A, Picarelli S, Tortora G, Ciardiello F, Limite G, De Placido S. In vitro expansion of human breast cancer epithelial and mesenchymal stromal cells: optimization of a coculture model for personalized therapy approaches. Mol Cancer Ther. 2007;6(12 Pt 1):3091–100.PubMedCrossRef
2.
Zurück zum Zitat Hayashi N, Niikura N, Yamauchi H, Nakamura S, Ueno NT. Adding hormonal therapy to chemotherapy and trastuzumab improves prognosis in patients with hormone receptor-positive and human epidermal growth factor receptor 2-positive primary breast cancer. Breast Cancer Res Treat. 2013;137(2):523–31.PubMedCrossRef Hayashi N, Niikura N, Yamauchi H, Nakamura S, Ueno NT. Adding hormonal therapy to chemotherapy and trastuzumab improves prognosis in patients with hormone receptor-positive and human epidermal growth factor receptor 2-positive primary breast cancer. Breast Cancer Res Treat. 2013;137(2):523–31.PubMedCrossRef
3.
Zurück zum Zitat Hicks DG, Kulkarni S. Trastuzumab as adjuvant therapy for early breast cancer: the importance of accurate human epidermal growth factor receptor 2 testing. Arch Pathol Lab Med. 2008;132(6):1008–15.PubMedCrossRef Hicks DG, Kulkarni S. Trastuzumab as adjuvant therapy for early breast cancer: the importance of accurate human epidermal growth factor receptor 2 testing. Arch Pathol Lab Med. 2008;132(6):1008–15.PubMedCrossRef
4.
Zurück zum Zitat Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol. 2007;25(1):118–45.PubMedCrossRef Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol. 2007;25(1):118–45.PubMedCrossRef
5.
Zurück zum Zitat Narayan M, Wilken JA, Harris LN, Baron AT, Kimbler KD, Maihle NJ. Trastuzumab-induced HER reprogramming in "resistant" breast carcinoma cells. Cancer Res. 2009;69(6):2191–4.PubMedCrossRef Narayan M, Wilken JA, Harris LN, Baron AT, Kimbler KD, Maihle NJ. Trastuzumab-induced HER reprogramming in "resistant" breast carcinoma cells. Cancer Res. 2009;69(6):2191–4.PubMedCrossRef
7.
Zurück zum Zitat Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21.PubMedCrossRef Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21.PubMedCrossRef
8.
Zurück zum Zitat Nakagawa S, Kageyama Y. Nuclear lncRNAs as epigenetic regulators-beyond skepticism. Biochim Biophys Acta. 2014;1839(3):215–22.PubMedCrossRef Nakagawa S, Kageyama Y. Nuclear lncRNAs as epigenetic regulators-beyond skepticism. Biochim Biophys Acta. 2014;1839(3):215–22.PubMedCrossRef
10.
Zurück zum Zitat Ni W, Zhang Y, Zhan Z, Ye F, Liang Y, Huang J, Chen K, Chen L, Ding Y. A novel lncRNA uc.134 represses hepatocellular carcinoma progression by inhibiting CUL4A-mediated ubiquitination of LATS1. J Hematol Oncol. 2017;10(1):91.PubMedPubMedCentralCrossRef Ni W, Zhang Y, Zhan Z, Ye F, Liang Y, Huang J, Chen K, Chen L, Ding Y. A novel lncRNA uc.134 represses hepatocellular carcinoma progression by inhibiting CUL4A-mediated ubiquitination of LATS1. J Hematol Oncol. 2017;10(1):91.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Han P, Li JW, Zhang BM, Lv JC, Li YM, Gu XY, Yu ZW, Jia YH, Bai XF, Li L, et al. The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/beta-catenin signaling. Mol Cancer. 2017;16(1):9.PubMedPubMedCentralCrossRef Han P, Li JW, Zhang BM, Lv JC, Li YM, Gu XY, Yu ZW, Jia YH, Bai XF, Li L, et al. The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/beta-catenin signaling. Mol Cancer. 2017;16(1):9.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Li W, Zhai L, Wang H, Liu C, Zhang J, Chen W, Wei Q. Downregulation of LncRNA GAS5 causes trastuzumab resistance in breast cancer. Oncotarget. 2016;7(19):27778–86.PubMedPubMedCentralCrossRef Li W, Zhai L, Wang H, Liu C, Zhang J, Chen W, Wei Q. Downregulation of LncRNA GAS5 causes trastuzumab resistance in breast cancer. Oncotarget. 2016;7(19):27778–86.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Shi SJ, Wang LJ, Yu B, Li YH, Jin Y, Bai XZ. LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget. 2015;6(13):11652–63.PubMedPubMedCentralCrossRef Shi SJ, Wang LJ, Yu B, Li YH, Jin Y, Bai XZ. LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget. 2015;6(13):11652–63.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Zhu HY, Bai WD, Ye XM, Yang AG, Jia LT. Long non-coding RNA UCA1 desensitizes breast cancer cells to trastuzumab by impeding miR-18a repression of yes-associated protein 1. Biochem Biophys Res Commun. 2018;496(4):1308–13.PubMedCrossRef Zhu HY, Bai WD, Ye XM, Yang AG, Jia LT. Long non-coding RNA UCA1 desensitizes breast cancer cells to trastuzumab by impeding miR-18a repression of yes-associated protein 1. Biochem Biophys Res Commun. 2018;496(4):1308–13.PubMedCrossRef
16.
Zurück zum Zitat Xu TP, Wang YF, Xiong WL, Ma P, Wang WY, Chen WM, Huang MD, Xia R, Wang R, Zhang EB, et al. E2F1 induces TINCR transcriptional activity and accelerates gastric cancer progression via activation of TINCR/STAU1/CDKN2B signaling axis. Cell Death Dis. 2017;8(6):e2837.PubMedPubMedCentralCrossRef Xu TP, Wang YF, Xiong WL, Ma P, Wang WY, Chen WM, Huang MD, Xia R, Wang R, Zhang EB, et al. E2F1 induces TINCR transcriptional activity and accelerates gastric cancer progression via activation of TINCR/STAU1/CDKN2B signaling axis. Cell Death Dis. 2017;8(6):e2837.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Zhu ZJ, He JK. TINCR facilitates non-small cell lung cancer progression through BRAF-activated MAPK pathway. Biochem Biophys Res Commun. 2018;497(4):971–7.PubMedCrossRef Zhu ZJ, He JK. TINCR facilitates non-small cell lung cancer progression through BRAF-activated MAPK pathway. Biochem Biophys Res Commun. 2018;497(4):971–7.PubMedCrossRef
18.
Zurück zum Zitat Zheng Y, Yang C, Tong S, Ding Y, Deng W, Song D, Xiao K. Genetic variation of long non-coding RNA TINCR contribute to the susceptibility and progression of colorectal cancer. Oncotarget. 2017;8(20):33536–43.PubMedPubMedCentralCrossRef Zheng Y, Yang C, Tong S, Ding Y, Deng W, Song D, Xiao K. Genetic variation of long non-coding RNA TINCR contribute to the susceptibility and progression of colorectal cancer. Oncotarget. 2017;8(20):33536–43.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Chen Z, Liu Y, He A, Li J, Chen M, Zhan Y, Lin J, Zhuang C, Liu L, Zhao G, et al. Theophylline controllable RNAi-based genetic switches regulate expression of lncRNA TINCR and malignant phenotypes in bladder cancer cells. Sci Rep. 2016;6:30798.PubMedPubMedCentralCrossRef Chen Z, Liu Y, He A, Li J, Chen M, Zhan Y, Lin J, Zhuang C, Liu L, Zhao G, et al. Theophylline controllable RNAi-based genetic switches regulate expression of lncRNA TINCR and malignant phenotypes in bladder cancer cells. Sci Rep. 2016;6:30798.PubMedPubMedCentralCrossRef
20.
21.
Zurück zum Zitat Dong H, Wang W, Mo S, Liu Q, Chen X, Chen R, Zhang Y, Zou K, Ye M, He X, et al. Long non-coding RNA SNHG14 induces trastuzumab resistance of breast cancer via regulating PABPC1 expression through H3K27 acetylation. J Cell Mol Med. 2018;22(10):4935–47. Dong H, Wang W, Mo S, Liu Q, Chen X, Chen R, Zhang Y, Zou K, Ye M, He X, et al. Long non-coding RNA SNHG14 induces trastuzumab resistance of breast cancer via regulating PABPC1 expression through H3K27 acetylation. J Cell Mol Med. 2018;22(10):4935–47.
22.
Zurück zum Zitat Dong H, Wang W, Chen R, Zhang Y, Zou K, Ye M, He X, Zhang F, Han J. Exosome-mediated transfer of lncRNASNHG14 promotes trastuzumab chemoresistance in breast cancer. Int J Oncol. 2018;53(3):1013–26.PubMedPubMedCentral Dong H, Wang W, Chen R, Zhang Y, Zou K, Ye M, He X, Zhang F, Han J. Exosome-mediated transfer of lncRNASNHG14 promotes trastuzumab chemoresistance in breast cancer. Int J Oncol. 2018;53(3):1013–26.PubMedPubMedCentral
23.
Zurück zum Zitat Dong H, Wang W, Mo S, Chen R, Zou K, Han J, Zhang F, Hu J. SP1-induced lncRNA AGAP2-AS1 expression promotes chemoresistance of breast cancer by epigenetic regulation of MyD88. J Exp Clin Cancer Res. 2018;37(1):202.PubMedPubMedCentralCrossRef Dong H, Wang W, Mo S, Chen R, Zou K, Han J, Zhang F, Hu J. SP1-induced lncRNA AGAP2-AS1 expression promotes chemoresistance of breast cancer by epigenetic regulation of MyD88. J Exp Clin Cancer Res. 2018;37(1):202.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al. Exosome-transmitted lncARSR promotes Sunitinib resistance in renal Cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29(5):653–68.PubMedCrossRef Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al. Exosome-transmitted lncARSR promotes Sunitinib resistance in renal Cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29(5):653–68.PubMedCrossRef
26.
Zurück zum Zitat Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436(7050):518–24.PubMedPubMedCentralCrossRef Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436(7050):518–24.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Karginov FV, Hannon GJ. Remodeling of Ago2-mRNA interactions upon cellular stress reflects miRNA complementarity and correlates with altered translation rates. Genes Dev. 2013;27(14):1624–32.PubMedPubMedCentralCrossRef Karginov FV, Hannon GJ. Remodeling of Ago2-mRNA interactions upon cellular stress reflects miRNA complementarity and correlates with altered translation rates. Genes Dev. 2013;27(14):1624–32.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Kaufhold S, Bonavida B. Central role of Snail1 in the regulation of EMT and resistance in cancer: a target for therapeutic intervention. J Exp Clin Cancer Res. 2014;33:62.PubMedPubMedCentralCrossRef Kaufhold S, Bonavida B. Central role of Snail1 in the regulation of EMT and resistance in cancer: a target for therapeutic intervention. J Exp Clin Cancer Res. 2014;33:62.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, Aronow B, Lin C, Li W, Yang L, et al. LncPRESS1 is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated De-acetylation of histone H3K56. Mol Cell. 2016;64(5):967–81.PubMedPubMedCentralCrossRef Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, Aronow B, Lin C, Li W, Yang L, et al. LncPRESS1 is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated De-acetylation of histone H3K56. Mol Cell. 2016;64(5):967–81.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.PubMedCrossRef Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.PubMedCrossRef
31.
Zurück zum Zitat Yu D, Hung MC. Overexpression of ErbB2 in cancer and ErbB2-targeting strategies. Oncogene. 2000;19(53):6115–21.PubMedCrossRef Yu D, Hung MC. Overexpression of ErbB2 in cancer and ErbB2-targeting strategies. Oncogene. 2000;19(53):6115–21.PubMedCrossRef
32.
Zurück zum Zitat Parise C, Caggiano V. Disparities in the risk of the ER/PR/HER2 breast cancer subtypes among Asian Americans in California. Cancer Epidemiol. 2014;38(5):556–62.PubMedCrossRef Parise C, Caggiano V. Disparities in the risk of the ER/PR/HER2 breast cancer subtypes among Asian Americans in California. Cancer Epidemiol. 2014;38(5):556–62.PubMedCrossRef
33.
Zurück zum Zitat Telli ML, Chang ET, Kurian AW, Keegan TH, McClure LA, Lichtensztajn D, Ford JM, Gomez SL. Asian ethnicity and breast cancer subtypes: a study from the California Cancer registry. Breast Cancer Res Treat. 2011;127(2):471–8.PubMedCrossRef Telli ML, Chang ET, Kurian AW, Keegan TH, McClure LA, Lichtensztajn D, Ford JM, Gomez SL. Asian ethnicity and breast cancer subtypes: a study from the California Cancer registry. Breast Cancer Res Treat. 2011;127(2):471–8.PubMedCrossRef
34.
Zurück zum Zitat Kurian AW, Fish K, Shema SJ, Clarke CA. Lifetime risks of specific breast cancer subtypes among women in four racial/ethnic groups. Breast Cancer Res. 2010;12(6):R99.PubMedPubMedCentralCrossRef Kurian AW, Fish K, Shema SJ, Clarke CA. Lifetime risks of specific breast cancer subtypes among women in four racial/ethnic groups. Breast Cancer Res. 2010;12(6):R99.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Hamberg P, Bos MM, Braun HJ, Stouthard JM, van Deijk GA, Erdkamp FL, Van der Stelt-Frissen IN, Bontenbal M, Creemers GJ, Portielje JE, et al. Randomized phase II study comparing efficacy and safety of combination-therapy trastuzumab and docetaxel vs. sequential therapy of trastuzumab followed by docetaxel alone at progression as first-line chemotherapy in patients with HER2+ metastatic breast cancer: HERTAX trial. Clin Breast Cancer. 2011;11(2):103–13.PubMedCrossRef Hamberg P, Bos MM, Braun HJ, Stouthard JM, van Deijk GA, Erdkamp FL, Van der Stelt-Frissen IN, Bontenbal M, Creemers GJ, Portielje JE, et al. Randomized phase II study comparing efficacy and safety of combination-therapy trastuzumab and docetaxel vs. sequential therapy of trastuzumab followed by docetaxel alone at progression as first-line chemotherapy in patients with HER2+ metastatic breast cancer: HERTAX trial. Clin Breast Cancer. 2011;11(2):103–13.PubMedCrossRef
36.
Zurück zum Zitat Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–51.PubMedPubMedCentralCrossRef Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–51.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Liu J, Pan C, Guo L, Wu M, Guo J, Peng S, Wu Q, Zuo Q. A new mechanism of trastuzumab resistance in gastric cancer: MACC1 promotes the Warburg effect via activation of the PI3K/AKT signaling pathway. J Hematol Oncol. 2016;9(1):76.PubMedPubMedCentralCrossRef Liu J, Pan C, Guo L, Wu M, Guo J, Peng S, Wu Q, Zuo Q. A new mechanism of trastuzumab resistance in gastric cancer: MACC1 promotes the Warburg effect via activation of the PI3K/AKT signaling pathway. J Hematol Oncol. 2016;9(1):76.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Chen Z, Liu H, Yang H, Gao Y, Zhang G, Hu J. The long noncoding RNA, TINCR, functions as a competing endogenous RNA to regulate PDK1 expression by sponging miR-375 in gastric cancer. Onco Targets Ther. 2017;10:3353–62.PubMedPubMedCentralCrossRef Chen Z, Liu H, Yang H, Gao Y, Zhang G, Hu J. The long noncoding RNA, TINCR, functions as a competing endogenous RNA to regulate PDK1 expression by sponging miR-375 in gastric cancer. Onco Targets Ther. 2017;10:3353–62.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Liu X, Ma J, Xu F, Li L. TINCR suppresses proliferation and invasion through regulating miR-544a/FBXW7 axis in lung cancer. Biomed Pharmacother. 2018;99:9–17.PubMedCrossRef Liu X, Ma J, Xu F, Li L. TINCR suppresses proliferation and invasion through regulating miR-544a/FBXW7 axis in lung cancer. Biomed Pharmacother. 2018;99:9–17.PubMedCrossRef
40.
Zurück zum Zitat Xia HF, He TZ, Liu CM, Cui Y, Song PP, Jin XH, Ma X. MiR-125b expression affects the proliferation and apoptosis of human glioma cells by targeting Bmf. Cell Physiol Biochem. 2009;23(4–6):347–58.PubMedCrossRef Xia HF, He TZ, Liu CM, Cui Y, Song PP, Jin XH, Ma X. MiR-125b expression affects the proliferation and apoptosis of human glioma cells by targeting Bmf. Cell Physiol Biochem. 2009;23(4–6):347–58.PubMedCrossRef
41.
Zurück zum Zitat Avellini C, Licini C, Lazzarini R, Gesuita R, Guerra E, Tossetta G, Castellucci C, Giannubilo SR, Procopio A, Alberti S, et al. The trophoblast cell surface antigen 2 and miR-125b axis in urothelial bladder cancer. Oncotarget. 2017;8(35):58642–53.PubMedPubMedCentralCrossRef Avellini C, Licini C, Lazzarini R, Gesuita R, Guerra E, Tossetta G, Castellucci C, Giannubilo SR, Procopio A, Alberti S, et al. The trophoblast cell surface antigen 2 and miR-125b axis in urothelial bladder cancer. Oncotarget. 2017;8(35):58642–53.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Zhang Y, Yan LX, Wu QN, Du ZM, Chen J, Liao DZ, Huang MY, Hou JH, Wu QL, Zeng MS, et al. miR-125b is methylated and functions as a tumor suppressor by regulating the ETS1 proto-oncogene in human invasive breast cancer. Cancer Res. 2011;71(10):3552–62.PubMedCrossRef Zhang Y, Yan LX, Wu QN, Du ZM, Chen J, Liao DZ, Huang MY, Hou JH, Wu QL, Zeng MS, et al. miR-125b is methylated and functions as a tumor suppressor by regulating the ETS1 proto-oncogene in human invasive breast cancer. Cancer Res. 2011;71(10):3552–62.PubMedCrossRef
43.
Zurück zum Zitat Wang F, Yu D, Liu Z, Wang R, Xu Y, Cui H, Zhao T. MiR-125b functions as a tumor suppressor and enhances Chemosensitivity to cisplatin in osteosarcoma. Technol Cancer Res Treat. 2016;15(6):NP105–12.PubMedCrossRef Wang F, Yu D, Liu Z, Wang R, Xu Y, Cui H, Zhao T. MiR-125b functions as a tumor suppressor and enhances Chemosensitivity to cisplatin in osteosarcoma. Technol Cancer Res Treat. 2016;15(6):NP105–12.PubMedCrossRef
44.
Zurück zum Zitat Kim JK, Noh JH, Jung KH, Eun JW, Bae HJ, Kim MG, Chang YG, Shen Q, Park WS, Lee JY, et al. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology. 2013;57(3):1055–67.PubMedCrossRef Kim JK, Noh JH, Jung KH, Eun JW, Bae HJ, Kim MG, Chang YG, Shen Q, Park WS, Lee JY, et al. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology. 2013;57(3):1055–67.PubMedCrossRef
45.
Zurück zum Zitat Glud M, Manfe V, Biskup E, Holst L, Dirksen AM, Hastrup N, Nielsen FC, Drzewiecki KT, Gniadecki R. MicroRNA miR-125b induces senescence in human melanoma cells. Melanoma Res. 2011;21(3):253–6.PubMedCrossRef Glud M, Manfe V, Biskup E, Holst L, Dirksen AM, Hastrup N, Nielsen FC, Drzewiecki KT, Gniadecki R. MicroRNA miR-125b induces senescence in human melanoma cells. Melanoma Res. 2011;21(3):253–6.PubMedCrossRef
46.
Zurück zum Zitat Ferracin M, Bassi C, Pedriali M, Pagotto S, D'Abundo L, Zagatti B, Corra F, Musa G, Callegari E, Lupini L, et al. miR-125b targets erythropoietin and its receptor and their expression correlates with metastatic potential and ERBB2/HER2 expression. Mol Cancer. 2013;12(1):130.PubMedPubMedCentralCrossRef Ferracin M, Bassi C, Pedriali M, Pagotto S, D'Abundo L, Zagatti B, Corra F, Musa G, Callegari E, Lupini L, et al. miR-125b targets erythropoietin and its receptor and their expression correlates with metastatic potential and ERBB2/HER2 expression. Mol Cancer. 2013;12(1):130.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Piunti A, Pasini D. Epigenetic factors in cancer development: polycomb group proteins. Future Oncol. 2011;7(1):57–75.PubMedCrossRef Piunti A, Pasini D. Epigenetic factors in cancer development: polycomb group proteins. Future Oncol. 2011;7(1):57–75.PubMedCrossRef
48.
49.
Zurück zum Zitat Chen G, Zou X, Watanabe H, van Deursen JM, Shen J. CREB binding protein is required for both short-term and long-term memory formation. J Neurosci. 2010;30(39):13066–77.PubMedPubMedCentralCrossRef Chen G, Zou X, Watanabe H, van Deursen JM, Shen J. CREB binding protein is required for both short-term and long-term memory formation. J Neurosci. 2010;30(39):13066–77.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Valor LM, Pulopulos MM, Jimenez-Minchan M, Olivares R, Lutz B, Barco A. Ablation of CBP in forebrain principal neurons causes modest memory and transcriptional defects and a dramatic reduction of histone acetylation but does not affect cell viability. J Neurosci. 2011;31(5):1652–63.PubMedPubMedCentralCrossRef Valor LM, Pulopulos MM, Jimenez-Minchan M, Olivares R, Lutz B, Barco A. Ablation of CBP in forebrain principal neurons causes modest memory and transcriptional defects and a dramatic reduction of histone acetylation but does not affect cell viability. J Neurosci. 2011;31(5):1652–63.PubMedPubMedCentralCrossRef
Metadaten
Titel
RETRACTED ARTICLE: Activation of LncRNA TINCR by H3K27 acetylation promotes Trastuzumab resistance and epithelial-mesenchymal transition by targeting MicroRNA-125b in breast Cancer
verfasst von
Huaying Dong
Jianguo Hu
Kejian Zou
Mulin Ye
Yuanwen Chen
Chengyi Wu
Xin Chen
Mingli Han
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2019
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0931-9

Weitere Artikel der Ausgabe 1/2019

Molecular Cancer 1/2019 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.