Skip to main content
OMICS : a Journal of Integrative Biology logoLink to OMICS : a Journal of Integrative Biology
. 2017 May 1;21(5):257–265. doi: 10.1089/omi.2017.0016

HLJ1 (DNAJB4) Gene Is a Novel Biomarker Candidate in Breast Cancer

Tolga Acun 1,, Natalie Doberstein 2, Jens K Habermann 2, Timo Gemoll 2, Christoph Thorns 3, Emin Oztas 4, Thomas Ried 5
PMCID: PMC5586162  PMID: 28481734

Abstract

Breast cancer is the most common cancer type and cause of cancer-related mortality among women worldwide. New biomarker discovery is crucial for diagnostic innovation and personalized medicine in breast cancer. Heat shock proteins (HSPs) have been increasingly reported as biomarkers and potential drug targets for cancers. HLJ1 (DNAJB4) belongs to the DNAJ (HSP40) family of HSPs and is regarded as a tumor suppressor gene in lung, colon, and gastric cancers. However, the role of the HLJ1 gene in breast cancer is currently unknown. We evaluated the role of the HLJ1 gene in breast cancer progression by analyzing its in vitro and in vivo expression and its genetic/epigenetic alterations. HLJ1 expression was found to be reduced or lost in breast cancer cell lines (SK-BR-3, MDA-MB-231, ZR-75-1) compared with the nontumorigenic mammary epithelial cell line (MCF 10A). In a clinical context for breast cancer progression, the HLJ1 expression was significantly less frequent in invasive breast carcinoma samples (n = 230) compared with normal breast tissue (n = 100), benign neoplasia (n = 53), and ductal carcinoma in situ (n = 21). In methylation analyses by the combined bisulfite restriction analysis assay, the CpG island located in the 5′-flanking region of the HLJ1 gene was found to be methylated in breast cancer cell lines. HLJ1 expression was restored in the ZR-75-1 cell line by DNA demethylating agent 5-Aza-2′-deoxycytidine (5-AzadC) and histone deacetylase inhibitor trichostatin A. These new observations support the idea that HLJ1 is a tumor suppressor candidate and potential biomarker for breast cancer. Epigenomic mechanisms such as CpG methylation and histone deacetylation might contribute to downregulation of HLJ1 expression. We call for future functional, epigenomic, and clinical studies to ascertain the contribution of HLJ1 to breast cancer pathogenesis and, importantly, evaluate its potential for biomarker development in support of personalized medicine diagnostic innovation in clinical oncology.

Keywords: : breast cancer biomarkers, DNAJB4, HLJ1, epigenomics, precision medicine

Introduction

Despite advances in prevention, diagnosis, and treatment of breast cancer, it remains as the most common cancer type and cause of cancer-related mortality among women worldwide (Ferlay et al., 2015). New biomarker discovery is much needed for diagnostic innovation and personalized therapeutics in breast cancer, as well as in clinical oncology broadly (Dzobo et al., 2016; Thomford et al., 2016; Toss and Cristofanilli, 2015).

Heat shock proteins (HSPs) have been reported as biomarkers and potential drug targets for cancers (Khalil et al., 2011; Sterrenberg et al., 2011). In this context, HLJ1 (DNAJB4) belongs to the DNAJ (HSP40) family of HSPs and is regarded as a tumor suppressor gene in lung, colon, and gastric cancers (Liu et al., 2014; Simões-correia et al., 2014; Tsai et al., 2006; Wang et al., 2005). HLJ1 has also been suggested as a potential biomarker and therapeutic target in lung cancer (Chen et al., 2008; Lai et al., 2013; Tsai et al., 2006; Wang et al., 2007). Moreover, in vitro studies showed that induction of HLJ1 inhibits cancer cell invasion in hepatocellular carcinoma cells (Wang et al., 2007). Importantly, the role of HLJ1 remains unclear in breast cancer despite the growing importance of this molecular target in abovementioned cancer types.

There are several lines of evidence that collectively suggest HLJ1 as an antioncogenic factor: HLJ1 inhibits cell cycle progression through controlling the STAT1/P21WAF1 pathway (Tsai et al., 2006), suppresses invasion by inducing E-Cadherin (Wang et al., 2005), and enhances apoptosis by activating JNK and CASPASE-3 (Lin et al., 2010). HLJ1 also suppresses malignancy by inhibiting the catalytic activity of SRC proto-oncogene (Chen et al., 2016).

DNAJ (HSP40) HSPs comprise the largest and most diverse subgroup of the HSP family. The DNAJ subfamily consists of 49 members and is divided further into three subclasses: DNAJA (4 members), DNAJB (13 members), and DNAJC (32 members). These cochaperones regulate major chaperones (HSP70, HSP90), which have tumor suppressor or oncoprotein functions. These regulatory actions make the DNAJ members more specific and selective drug targets compared with other major chaperones (Sterrenberg et al., 2011). Besides, it has been suggested that DNAJ proteins may also possess chaperone-like activities independently of HSP70 chaperones (Hageman et al., 2010).

To date, many DNAJ members have been implicated in cancer development and metastasis of different organs, such as DNAJA1 (glioblastoma, prostate), DNAJA3 (osteosarcoma, melanoma, kidney, hematopoietic, breast), DNAJB6 (breast), DNAJB11 (KSHV-related tumors, ovarian), DNAJC9 (cervical), DNAJC10 (neuroblastoma), and DNAJC15 (ovarian, brain, breast) (Khalil et al., 2011; Sterrenberg et al., 2011).

In this study, we evaluated the potential role of HLJ1 in breast cancer progression by analyzing its in vitro and in vivo expression and its genetic/epigenetic alterations. We showed that HLJ1 expression is reduced or lost in breast cancer cell lines and invasive breast carcinoma samples. Moreover, we suggest that epigenetic mechanisms in the form of CpG methylation and histone deacetylation could contribute to downregulation of HLJ1 expression.

Materials and Methods

Cell lines and clinical study samples

Breast cancer cell lines, BT-20 (ATCC® HTB-19™), SK-BR-3 (ATCC® HTB-30™), MDA-MB-231 (ATCC® HTB-26™), and ZR-75-1 (ATCC® CRL-1500™), and nontumorigenic mammary epithelial cell line MCF 10A (ATCC® CRL-10317™) were used and cultured according to American Type Culture Collection (ATCC) recommendations (ATCC, Manassas, VA, USA) (Table 1).

Table 1.

The Clinicopathological Features of the Breast Cell Lines Used in the Study

    Immunoprofilea    
Cell line Subtypea,b ER PR HER2 Diseasec Tumorigenicityc
BT-20 Basal A/basal Carcinoma Yes
SK-BR-3 Luminal/HER2+ + Adenocarcinoma Yes
MDA-MB-231 Basal B/claudin-low Adenocarcinoma Yes
ZR-75-1 Luminal/luminal + Ductal carcinoma Yes
MCF 10A Basal B/nonmalignant basal Fibrocystic disease No
a

Kao et al. (2009).

b

Heiser et al. (2012).

ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; PR, progesterone receptor.

For the clinical study, we evaluated 404 breast tissue samples (tissue microarray [TMA] sample set) from 245 patients (study cohort) (Tables 2 and 3). In a subset of 118 patients, supplementary samples were collected from the tissue adjacent to the sample of the primary histological diagnosis. These supplementary samples have been used for comparison between histological groups. The normal tissue samples (n = 100) were obtained from adjacent tissue of samples of the 245 patients with invasive breast carcinoma (n = 203), ductal carcinoma in situ (DCIS; n = 6), and benign neoplasia (n = 36). For association and survival analysis, only samples of the primary breast cancer diagnosis were evaluated.

Table 2.

Histopathological Features and Numbers of Patients (Study Cohort) and the Tissue Microarray Samples Used in the Study

Histopathology Patients TMA samples
Normal tissue 100
Benign neoplasia 36 53
DCIS 6 21
Invasive carcinoma 203 230
TOTAL 245 404

DCIS, ductal carcinoma in situ; TMA, tissue microarray.

Table 3.

Characteristics of the Clinical Study Cohort

  Total (n = 245) Benign neoplasia (n = 36) DCISa(n = 6) Invasive carcinoma (n = 203)
Age at diagnosis (years)
 Mean 59.71 45.96 69.60 61.63
 Median 58.00 47.00 71.00 60.00
 Range 18–93 18–77 59–75 30–93
 95% CI 57.46–61.96 39.55–52.38 61.87–77.33 59.32–63.95
Overall survival (years)
 Mean 11.77 16.58 12.55 10.97
 Median 12.84 20.45 14.75 9.32
 Range 0–22.45 0.56–21.45 2.32–20.45 0–22.45
 95% CI 10.69–12.86 14.05–19.12 3.26–21.83 9.79–12.16
Disease-free survival (years)
 Mean 11.37 16.58 12.55 10.50
 Median 11.05 20.45 14.75 8.07
 Range 0–22.45 0.56–21.45 2.32–20.45 0–22.45
 95% CI 10.25–12.49 14.05–19.12 3.26–21.83 9.28–11.71
a

Ductal carcinoma in situ.

CI, confidence interval.

Formalin-fixed and paraffin-embedded (FFPE) tissue samples were collected between 1989 and 1993 and archived in the Institute of Pathology at the University Hospital Schleswig Holstein in Luebeck, Germany. The study was performed in accordance with the approval of the local Ethics Committee at the same institute (#08–012).

Real-time quantitative reverse transcription PCR (qRT-PCR)

Total RNA was extracted from the breast cell lines by using RNeasy Mini Kit (Qiagen, Germany). Isolated RNAs were then used to synthesize cDNA by using the Verso cDNA Synthesis Kit (Thermo Fisher Scientific, Inc., MA, USA). Relative quantification of HLJ1 transcripts was performed with Power SYBR Green Master Mix (Thermo Fisher Scientific, Inc.) on ABI Prism 7000 Sequence Detection System (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. HLJ1 and TBP (TATA box-binding protein) -specific primer pairs were used with cDNAs from breast cell lines. The TBP gene was used as an internal control (Gur-Dedeoglu et al., 2009). Relative levels of HLJ1 transcripts were measured by the comparative CT method (2−ΔΔCT method) (Livak and Schmittgen, 2001). Statistical analyses were performed using Student's t-test.

Western blot

Total cell lysates from breast cell lines were prepared in RIPA buffer (150 mM sodium chloride, 1.0% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM Tris [pH 8.0]) containing protease inhibitor cocktail (Roche cOmplete Mini; Sigma-Aldrich Co. LLC., St. Louis, MO, USA). Insoluble material was removed by centrifugation at 10,000 g for 20 min at 4°C and protein contents were measured with bicinchoninic acid (BCA) assay (Pierce™ BCA Protein Assay Kit; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Equalized protein samples were resolved by 4–12% SDS-PAGE (NuPAGE® Novex®Bis-Tris Gels; Thermo Fisher Scientific, Inc.) and then transferred onto polyvinylidene fluoride membrane.

DNAJB4 rabbit polyclonal antibody (Cat No: 13064-1-AP; Proteintech Group, Inc., IL, USA) (dilution, 1:1000) was used as the primary antibody, and HRP-linked anti-rabbit IgG (Cat. No: 7074; Cell Signaling Technology, Inc., MA, USA) was used as the secondary antibody (dilution, 1:2000). Membrane was developed using SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific, Inc.). α-Tubulin (11H10) Rabbit mAb (Cat. No: 2125; Cell Signaling Technology, Inc.) was used as the loading control (dilution, 1:2000).

Immunohistochemistry

Clinical specimens were formalin-fixed, paraffin-embedded, and implemented into a TMA. The TMAs were constructed using a semiautomated arrayer (TMArrayer; Pathology Devices, MD, USA) as described previously (Oberländer et al., 2014). Tissue cores with a diameter of 1.5 mm were taken from selected regions of FFPE donor tissue blocks and transferred into an empty paraffin recipient block. Detailed information of immunohistochemistry (IHC) method was supplied in Supplementary Data.

Combined bisulfite restriction analysis

DNeasy tissue kit (Qiagen, Germany) was used to extract genomic DNA from breast cell lines. Genomic DNAs were treated with bisulfite by using Methylamp™ DNA Modification Kit (EpiGentek, NY, USA) according to the manufacturer's instructions. Seminested primer pairs targeting HLJ1 CpG islands were used to amplify bisulfite-treated DNAs. To analyze the methylation status, amplicons were restriction digested with TaqI and Hpy188I (New England BioLabs, Ipswich, MA), as previously described (Xiong and Laird, 1997).

5-Aza-2′-deoxycytidine and trichostatin A treatments

ZR-75-1 cells were plated in 75-cm2 plastic tissue culture flasks (T-75) at a density of 8 × 105 cells/flask and grown for 24 h before treatment. ZR-75-1 cells were treated with 10 μM 5-Aza-2′-deoxycytidine (5-AzadC, Cat No: A3656; Sigma-Aldrich Co. LLC.) or 300 nM trichostatin A (TSA, Cat No: T1952; Sigma-Aldrich Co. LLC.]). TSA treatment was performed in the last 24 h. All media were replenished every 12 h over a period of 72 h. Control cultures were left untreated or received a mock treatment with dimethyl sulfoxide (DMSO) (final concentration, 0.1%) since both drugs were dissolved in DMSO (Cat No: D2650; Sigma-Aldrich Co. LLC.). Treatments were performed in triplicates.

Statistical analysis

The semiquantitatively scored HLJ1 expression and nonbinary clinical characteristics were dichotomized according to survival outcomes, group sizes, and median cutoff (e.g., age). For dichotomization, only samples with primary diagnosis were included. Other categorical nonbinary variables were also stratified by either survival outcomes or group sizes.

Associations between HLJ1 expression and clinical characteristics were examined using the two-sided χ2-test or Fisher's exact test. Kaplan–Meier plots and log-rank tests were used to assess the differences between groups in overall survival (OS). Associations with OS were determined using univariate Cox regression, providing a hazard ratio and 95% confidence interval for each variable. Multivariate Cox regression analysis was used to determine whether HLJ1 expression was independent of other prognostic factors. A p ≤ 0.05 was considered as statistically significant. Statistical analysis was performed using SPSS Statistics, version 22 (IBM Corporation, Somers, NY, USA).

Results

HLJ1 expression in breast cancer cell lines and clinical breast samples

We analyzed HLJ1 mRNA and protein expression levels in breast cell lines by real-time qRT-PCR and western blotting, respectively. HLJ1 mRNA expression was found to be reduced or very low in MDA-MB-231, SK-BR-3, and ZR-75-1 cell lines compared with MCF 10A cell line, which is a nontumorigenic mammary epithelial cell line (Fig. 1A). Moreover, HLJ1 protein expression was found to be lost in ZR-75-1 cell line (Fig. 1B). We could not observe a good correlation between mRNA and protein levels of SK-BR-3 and MDA-MB-231 cell lines (Fig. 1A, B).

FIG. 1.

FIG. 1.

HLJ1 expression in breast cell lines. (A) Real-time qRT-PCR results showing HLJ1 mRNA expression in breast cancer cell lines (BT-20, SK-BR-3, MDA-MB-231, ZR-75-1) and nontumorigenic mammary epithelial cell line (MCF 10A). TBP was used as internal control. Standard deviations of three biological replicates are shown. (B) HLJ1 expression was also assessed with western blot analysis by using total cell lysates from breast cell lines. α-Tubulin was used as loading control. Twenty micrograms of total protein was loaded per sample.

According to the IHC results, HLJ1 protein expression (score 1 + 2 + 3) was also significantly less frequent in invasive breast carcinoma samples (47.3%) compared with normal breast samples (78.1%, p = 0.00001), benign neoplasia samples (74.5%, p = 0.0007), and DCIS samples (75%, p = 0.039) (Fig. 2, Supplementary Fig. S1).

FIG. 2.

FIG. 2.

HLJ1 protein expression in breast samples. Comparison between histological groups regarding HLJ1 expression in breast samples (n = 404). DCIS, ductal carcinoma in situ.

In addition, we found significant associations between HLJ1 expression and clinical characteristics of breast carcinomas: HLJ1 expression was more frequent in ER+ and PR+ samples compared with ER− and PR− samples (p = 0.0048 and p = 0.016, respectively) (Table 4). Furthermore, a significant association with the molecular subtype could be determined: while HLJ1 was more frequently expressed in the samples of luminal A subtype, it was less frequent in triple negative samples and HER2-overexpressing samples (p = 0.031, Table 4). However, a significant association between HLJ1 expression and OS of breast cancer patients could not be observed (Fig. 3).

Table 4.

Associations Between HLJ1 Expression and Clinical Characteristics in Invasive Breast Carcinomas (n = 203)

  HLJ1 expression
Variable Score 0, n (%) Score 1 + 2 + 3, n (%)
Age at diagnosisa,(years)
 <58 47 (53) 42 (47)
 >58 56 (53) 49 (47)
P 1.000
Recurrence
 No 87 (51) 84 (49)
 Yes 16 (70) 7 (30)
P 0.120
Tumor stage
 pT1 22 (46) 26 (54)
 pT2–pT4 66 (56) 52 (44)
P 0.304
Lymph node stage
 pN0 39 (53) 34 (47)
 pN1–pN3 40 (50) 40 (50)
P 0.747
Metastasis stage
 pM0 90 (52) 84 (48)
 pM1 13 (65) 7 (35)
P 0.345
UICC stage
 Early 50 (48) 54 (52)
 Late 38 (61) 24 (39)
P 0.110
Grade
 G1–G2 70 (51) 68 (49)
 G3 21 (62) 13 (38)
P 0.258
Ki67 status
 Low (<20%) 89 (51) 81 (49)
 High (>20%) 13 (57) 10 (43)
P 0.825
ER
 Negative 50 (66) 26 (34)
 Positive 51 (44) 64 (56)
P 0.0048
PR
 Negative 46 (64) 26 (36)
 Positive 52 (45) 63 (55)
P 0.016
Her2
 Negative 88 (51) 83 (49)
 Positive 15 (68) 7 (32)
P 0.175
Molecular class
 Luminal A 32 (41) 47 (59)
 Luminal B Her2− 12 (55) 10 (45)
 Luminal B Her2+ 5 (50) 5 (50)
 HER2 overexpr. 8 (89) 1 (11)
 Triple negative 27 (61) 17 (39)
P 0.031
a

Median age at diagnosis of the full study cohort.

b

Statistically significant values are highlighted in bold.

FIG. 3.

FIG. 3.

Kaplan–Meier overall survival plot of HLJ1 protein expression in invasive breast carcinoma (n = 203). Log-rank test and univariate Cox regression analysis were used to assess the difference between groups of patients stratified by dichotomized HLJ1 protein expression (Score 0 and Score 1 + 2 + 3). The survival rates for patients with HLJ1-negative breast carcinomas were 62% (95% CI, 52–72%) and for patients with HLJ1-positive carcinomas 73% (95% CI, 63–84%) at the time point of 5 years. No significant differences were found in the present sample (p > 0.05). CI, confidence interval.

CpG methylation in the 5′-flanking region of HLJ1

Our in silico analysis (Li and Dahiya, 2002) showed that there are CpG islands in the 5′-flanking region and in a region driving basal transcriptional activity of the HLJ1 gene (Wang et al., 2005). Those regions were designated as region-1 and region-2, respectively (Fig. 4). Genomic DNAs from the cell lines treated with bisulfite were amplified using the seminested primer pairs targeting those two regions. PCR amplicons were restriction digested by TaqI and Hpy188I restriction enzymes to analyze methylation status of region-1 and region-2, respectively (Fig. 5A, B). We did not detect CpG methylation in a region driving basal transcriptional activity. However, region-1, which contains some sequences similar to heat shock transcription factor 1 (HSF1) binding motifs (Jaeger et al., 2014; Trinklein et al., 2004), was found to be methylated in all cell lines (Fig. 5A), suggesting that methylation of this region could have some regulatory effects.

FIG. 4.

FIG. 4.

CpG islands in 5’-region of HLJ1. In silico analysis of HLJ1 locus by MethPrimer program (Li and Dahiya, 2002) showed CpG islands. Location of the primers (F, forward; R, reverse) used in COBRA assay is shown by arrows. Vertical red lines denote CpG sites, numbered boxes denote exons. CpG islands are shown as light blue areas. COBRA, combined bisulfite restriction analysis.

FIG. 5.

FIG. 5.

COBRA. TaqI (A) and Hpy188I (B) restriction enzymes were used for region-1 and region-2, respectively. (−), No restriction digestion; M, marker. An amplicon having an Hpy188I cutting site was used with (+) and without (Uncut) a restriction enzyme.

Restoration of HLJ1 expression in ZR-75-1 cell line

To explore the role of DNA methylation and/or histone deacetylation as mechanisms that might operate on HLJ1 downregulation in breast cancers, we treated ZR-75-1 cells with DNA methyl transferase inhibitor (5-AzadC) or histone deacetylase inhibitor (TSA). Both drugs successfully restored HLJ1 expression in ZR-75-1 cells, which implies that epigenetic mechanisms could play a role in the downregulation of HLJ1 (Fig. 6A, B).

FIG. 6.

FIG. 6.

Restoration of HLJ1 expression in ZR-75-1 cell line. Real-time qRT-PCR (A) and western blotting (B) results showing restoration of HLJ1 expression in ZR-75-1 cells treated with 5-AzadC and TSA. TBP and α-tubulin were used as internal controls in real-time qRT-PCR and western blotting, respectively. DMSO was used as mock treatment. Thirty micrograms of total protein was loaded per sample. 5-AzadC, 5-Aza-2′-deoxycytidine; DMSO, dimethyl sulfoxide; TSA, trichostatin A.

Discussion

The tumor suppressor role of HLJ1 has been well documented in lung cancer, colorectal cancer, and gastric cancer (Liu et al., 2014; Simões-correia et al., 2014; Tsai et al., 2006; Wang et al., 2005). Moreover, high expression of HLJ1 was noted as a significant prognostic factor for reduced cancer recurrence and longer OS in NSCLC patients (Tsai et al., 2006). HLJ1 shows its antioncogenic properties through the STAT1/P21WAF1 pathway (Tsai et al., 2006), E-Cadherin (Wang et al., 2005), JNK and CASPASE-3 (Lin et al., 2010), and SRC (Chen et al., 2016).

HLJ1 was also demonstrated to inhibit invasion and metastasis by reducing nucleophosmin (NPM1) oligomerization through direct binding, which results in increased AP-2α function and decreased matrix metalloproteinase-2 (MMP2) and signal transducer and activator of transcription 3 (STAT3) activities (Chang et al., 2010). Most of these pathways or genes are important for breast carcinogenesis, which highlighted the tumor suppressive role of HLJ1 for breast cancer (Banerjee and Resat, 2016; Berx et al., 2001; Cellurale et al., 2012; Duffy et al., 2000; Finn, 2008; Pellikainen et al., 2002).

Two herbal chemicals, curcumin (Chen et al., 2008) and andrographolide (Lai et al., 2013), were shown to induce HLJ1 expression and suppress tumorigenesis in lung adenocarcinoma and nonsmall cell lung cancer, respectively. High concentration of DMSO (≥1%) was also shown to induce HLJ1 and inhibit malignant properties of lung adenocarcinoma cells (Wang et al., 2012). These results and the above studies strongly suggest that targeted induction of HLJ1 may represent a promising approach for cancer therapy.

Despite the significance of HLJ1 in cancer progression, HLJ1 genetic/epigenetic alterations and functional importance in breast cancer are currently unknown. Breast cancer is the most common cancer type and the most common cause of cancer-related mortality among women worldwide (Ferlay et al., 2015). Therefore, identification of new diagnostic/prognostic biomarkers and drug targets is of high interest for this malignancy, highlighting the significance of evaluating HLJ1 in this context.

We first evaluated HLJ1 expression level among breast cell lines (Fig. 1A, B). The malignant cell lines, MDA-MB-231, SK-BR-3, and ZR-75-1, showed reduced or very low levels of HLJ1 mRNA compared with the nonmalignant breast cell line, MCF 10A. Moreover, HLJ1 protein was found to be lost in the ZR-75-1 cell line. Immunohistochemical analysis of clinical breast samples confirmed our in vitro observations and showed that HLJ1 was significantly less frequently expressed in invasive breast carcinoma samples than in normal breast samples (Fig. 2, Supplementary Fig. S1).

The HLJ1 expression was significantly correlated with estrogen receptor (ER) and progesterone receptor (PR) positivity, as well as with the luminal A subtype, all of which are associated with a better prognosis. Triple negative and HER2-overexpressing samples less frequently expressed HLJ1, which are related to worse prognosis (Table 4) (Dai et al., 2015). Although we could not found a significant association between HLJ1 expression and OS of breast cancer patients (Fig. 3), a study with a larger sample size might be necessary to ascertain the impact of HLJ1 expression on OS.

According to the COSMIC database, genetic alterations of HLJ1 in the form of mutations or copy number variations (CNVs) are very rare in cancers. In the case of breast cancer, HLJ1 mutation rate is only 0.19%. CNVs are seen at the HLJ1 locus (1p31.1) with a rate of 0.06% and 0.46%, either as loss or gain, respectively (Forbes et al., 2015).

It is known that DNA methylation and alterations of chromatin structure are predominant epigenetic mechanisms that downregulate tumor suppressor genes in cancers (Esteller, 2007). So, epigenetic mechanisms could also be responsible for HLJ1 downregulation in breast cancers. We found CpG islands in the 5′-flanking region and in a region having basal transcriptional activity (Wang et al., 2005) (Fig. 4). According to our combined bisulfite restriction analysis (COBRA) results, the 5′-flanking region having sequences similar to HSF1 binding motifs (heat shock elements [HSEs]) was found to be methylated in all breast cell lines (Jaeger et al., 2014; Trinklein et al., 2004) (Fig. 5A). It is important to note that findings regarding these HSEs are mainly based on our in silico observations. HSEs may vary in their consensus sequence, length, and/or orientation (Crinelli et al., 2015; Jaeger et al., 2014).

Moreover, we successfully restored HLJ1 expression in ZR-75-1 cells with 5-AzadC and TSA (Fig. 6A, B). It is therefore plausible to suggest that epigenetic mechanisms in the form of CpG methylation and histone deacetylation may contribute to the downregulation of HLJ1 expression. HLJ1 could also be regulated by miRNAs in breast cancer. A study employing the CellMiner web-based application suggested that some miRNAs could be involved in post-transcriptional regulation of HLJ1 in some breast cell lines (Ozgur et al., 2014; Reinhold et al., 2012).

Conclusions

HLJ1 was found to be downregulated in breast cancers and epigenomic events such as CpG methylation and histone deacetylation could be responsible for this downregulation. Consistent with previous studies showing tumor suppressor properties of HLJ1, our findings suggest that HLJ1 is a tumor suppressor candidate and potential biomarker in breast cancer. HLJ1 was previously regarded as a promising therapeutic target in lung cancers. In this regard, we call for future functional, epigenomic, and clinical studies to ascertain the contribution of HLJ1 to breast cancer pathogenesis and, importantly, evaluate its potential for biomarker development in support of personalized medicine diagnostic innovation in clinical oncology.

Supplementary Material

Supplemental data
Supp_Data.pdf (212.8KB, pdf)

Abbreviations Used

5-AzadC

5-Aza-2′-deoxycytidine

ATCC

American Type Culture Collection

CNV

copy number variations

COBRA

combined bisulfite restriction analysis

DCIS

ductal carcinoma in situ

DMSO

dimethyl sulfoxide

FFPE

formalin-fixed and paraffin-embedded

HSF1

heat shock transcription factor 1

HSPs

heat shock proteins

IHC

immunohistochemistry

OS

overall survival

TMA

tissue microarray

TSA

trichostatin A

Acknowledgments

This project was supported by Bülent Ecevit University (Zonguldak, Turkey) (Grant No. 2013-50737594-01) and the Fulbright Visiting Scholar Program to Dr. Acun. The study was conducted in cooperation with the US National Cancer Institute (MD). The authors thank Dr. Darawalee Wangsa Zong, Danny Wangsa, Dr. Nicole E. McNeil, Buddy Chen, and Mai T. Tran for scientific and technical support to the project.

Authors' Contributions

All authors met the ICMJE criteria for authorship. T.A. conceived the study hypotheses, designed and implemented all experiments except IHC, interpreted the data, and drafted the first version of the manuscript. N.D., J.K.H, T.G., and C.T. performed the IHC study and interpreted the results. T.G., J.K.H., E.O., and T.R. contributed to data interpretation and participated in drafting and finalizing the manuscript.

Author Disclosure Statement

The authors declare that no conflicting financial interests exist.

References

  1. Banerjee K, and Resat H. (2016). Constitutive activation of STAT3 in breast cancer cells: A review. Int J Cancer 138, 2570–2578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Berx G, Roy FV, Thoreson M, et al. (2001). The E-cadherin/catenin complex: An important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast Cancer Res 3, 289–293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Cellurale C, Girnius N, Jiang F, et al. (2012). Role of JNK in mammary gland development and breast cancer. Cancer Res 72, 472–481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Chang TP, Yu SL, Lin SY, et al. (2010). Tumor suppressor HLJ1 binds and functionally alters nucleophosmin via activating enhancer binding protein 2α complex formation. Cancer Res 70, 1656–1667 [DOI] [PubMed] [Google Scholar]
  5. Chen CH, Chang WH, Su KY, et al. (2016). HLJ1 is an endogenous Src inhibitor suppressing cancer progression through dual mechanisms. Oncogene 35, 5674–5685 [DOI] [PubMed] [Google Scholar]
  6. Chen HW, Lee JY, Huang JY, et al. (2008). Curcumin inhibits lung cancer cell invasion and metastasis through the tumor suppressor HLJ1. Cancer Res 68, 7428–7438 [DOI] [PubMed] [Google Scholar]
  7. Crinelli R, Bianchi M, Radici L, Carloni E, Giacomini E, and Magnani M. (2015). Molecular dissection of the human ubiquitin C promoter reveals heat shock element architectures with activating and repressive functions. PLoS One 10, e0136882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Dai X, Li T, Bai Z, et al. (2015). Breast cancer intrinsic subtype classification, clinical use and future trends. Am J Cancer Res 5, 2929–2943 [PMC free article] [PubMed] [Google Scholar]
  9. Duffy MJ, Maguire TM, Hill A, McDermott E, and O'Higgins N. (2000). Metalloproteinases: Role in breast carcinogenesis, invasion and metastasis. Breast Cancer Res 2, 252–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Dzobo K, Senthebane DA, Rowe A, et al. (2016) Cancer stem cell hypothesis for therapeutic innovation in clinical oncology? Taking the root out, not chopping the leaf. OMICS 20, 681–691 [DOI] [PubMed] [Google Scholar]
  11. Esteller M. (2007). Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8, 286–298 [DOI] [PubMed] [Google Scholar]
  12. Ferlay J, Soerjomataram I, Dikshit R, et al. (2015). Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136, E359–E386 [DOI] [PubMed] [Google Scholar]
  13. Finn RS. (2008). Targeting Src in breast cancer. Ann Oncol 19, 1379–1386 [DOI] [PubMed] [Google Scholar]
  14. Gur-Dedeoglu B, Konu O, Bozkurt B, Ergul G, Seckin S, and Yulug IG. (2009). Identification of endogenous reference genes for qRT-PCR analysis in normal matched breast tumor tissues. Oncol Res 17, 353–365 [DOI] [PubMed] [Google Scholar]
  15. Forbes SA, Beare D, Gunasekaran P, et al. (2015). COSMIC: Exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res 43, D805–D811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Hageman J, Rujano MA, van Waarde MA, et al. (2010). A DNAJB chaperone subfamily with HDAC-dependent activities suppresses toxic protein aggregation. Mol Cell 37, 355–369 [DOI] [PubMed] [Google Scholar]
  17. Heiser LM, Sadanandam A, Kuo WL, et al. (2012). Subtype and pathway specific responses to anticancer compounds in breast cancer. Proc Natl Acad Sci U S A 109, 2724–2729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Jaeger AM, Makley LN, Gestwicki JE, and Thiele DJ. (2014). Genomic heat shock element sequences drive cooperative human heat shock factor 1 DNA binding and selectivity. J Biol Chem 289, 30459–30469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Kao J, Salari K, Bocanegra M, et al. (2009). Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS One 4, e6146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Khalil AA, Kabapy NF, Deraz SF, and Smith C. (2011). Heat shock proteins in oncology: Diagnostic biomarkers or therapeutic targets? Biochim Biophys Acta 1816, 89–104 [DOI] [PubMed] [Google Scholar]
  21. Lai YH, Yu SL, Chen HY, Wang CC, Chen HW, and Chen JJW. (2013). The HLJ1-targeting drug screening identified Chinese herb andrographolide that can: Suppress tumour growth and invasion in non-small-cell lung cancer. Carcinogenesis 34, 1069–1080 [DOI] [PubMed] [Google Scholar]
  22. Li LC, and Dahiya R. (2002). MethPrimer: Designing primers for methylation PCRs. Bioinformatics 18, 1427–1431 [DOI] [PubMed] [Google Scholar]
  23. Lin SY, Hsueh CM, Yu SL, et al. (2010). HLJ1 is a novel caspase-3 substrate and its expression enhances UV-induced apoptosis in non-small cell lung carcinoma. Nucleic Acids Res 38, 6148–6158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Liu Y, Zhou J, Zhang C, et al. (2014). HLJ1 is a novel biomarker for colorectal carcinoma progression and overall patient survival. Int J Clin Exp Pathol 7, 969–977 [PMC free article] [PubMed] [Google Scholar]
  25. Livak KJ, and Schmittgen TD. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods 25, 402–408 [DOI] [PubMed] [Google Scholar]
  26. Oberländer M, Alkemade H, Bünger S, et al. (2014). A “waterfall” transfer-based workflow for improved quality of tissue microarray construction and processing in breast cancer research. Pathol Oncol Res 20, 719–726 [DOI] [PubMed] [Google Scholar]
  27. Ozgur A, Tutar L, and Tutar Y. (2014). Regulation of Heat Shock Proteins by miRNAs in human breast cancer. MicroRNA 3, 118–135 [DOI] [PubMed] [Google Scholar]
  28. Pellikainen J, Kataja V, Ropponen K, et al. (2002). Reduced nuclear expression of transcription factor AP-2 associates with aggressive breast cancer. Clin Cancer Res 8, 3487–3495 [PubMed] [Google Scholar]
  29. Reinhold WC, Sunshine M, Liu H, et al. (2012). CellMiner: A web-based suite of genomic and pharmacologic tools to explore transcript and drug patterns in the NCI-60 cell line set. Cancer Res 72, 3499–3511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Simões-correia J, Silva DI, Melo S, et al. (2014). DNAJB4 molecular chaperone distinguishes WT from mutant E-cadherin, determining their fate in vitro and in vivo. Hum Mol Genet 23, 2094–2105 [DOI] [PubMed] [Google Scholar]
  31. Sterrenberg JN, Blatch GL, and Edkins AL. (2011). Human DNAJ in cancer and stem cells. Cancer Lett 312, 129–142 [DOI] [PubMed] [Google Scholar]
  32. Thomford NE, Mkhize B, Dzobo K, et al. (2016) African lettuce (Launaea taraxacifolia) displays possible anticancer effects and herb-drug interaction potential by CYP1A2, CYP2C9, and CYP2C19 inhibition. OMICS 20, 528–537 [DOI] [PubMed] [Google Scholar]
  33. Toss A, and Cristofanilli M. (2015). Molecular characterization and targeted therapeutic approaches in breast cancer. Breast Cancer Res 17, 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Trinklein ND, Murray JI, Hartman SJ, Botstein D, and Myers RM. (2004). The role of heat shock transcription factor 1 in the genome-wide regulation of the mammalian heat shock response. Mol Biol Cell 15, 1254–1261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Tsai MF, Wang CC, Chang GC, et al. (2006). A new tumor suppressor DnaJ-like heat shock protein, HLJ1, and survival of patients with non-small-cell lung carcinoma. J Natl Cancer Inst 98, 825–838 [DOI] [PubMed] [Google Scholar]
  36. Wang CC, Lin SY, Lai YH, Liu YJ, Hsu YL, and Chen JJW. (2012). Dimethyl Sulfoxide Promotes the Multiple Functions of the Tumor Suppressor HLJ1 through Activator Protein-1 Activation in NSCLC Cells. PLoS One7, e33772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Wang CC, Tsai MF, Dai TH, et al. (2007). Synergistic activation of the tumor suppressor, HLJ1, by the transcription factors YY1 and activator protein 1. Cancer Res 67, 4816–4826 [DOI] [PubMed] [Google Scholar]
  38. Wang CC, Tsai MF, Hong TM, et al. (2005). The transcriptional factor YY1 upregulates the novel invasion suppressor HLJ1 expression and inhibits cancer cell invasion. Oncogene 24, 4081–4093 [DOI] [PubMed] [Google Scholar]
  39. Xiong Z, and Laird PW. (1997). COBRA: A sensitive and quantitative DNA methylation assay. Nucleic Acids Res 25, 2532–2534 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental data
Supp_Data.pdf (212.8KB, pdf)

Articles from OMICS : a Journal of Integrative Biology are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES