Skip to main content

BORIS: a key regulator of cancer stemness

Abstract

BORIS (CTCFL) is a DNA binding protein which is involved in tumorigenesis. Although, there are different opinions on the level of gene expression and function of BORIS in normal and cancer tissues, the results of many studies have classified BORIS as a protein belonging to cancer/testis (CT) genes, which are identified as a group of genes that are expressed normally in testis, and abnormally in various types of cancers. In testis, BORIS induces the expression of some male germ cell/testis specific genes, and plays crucial roles during spermatogenesis and production of sperm. In tumorigenesis, the role of BORIS in the expression induction of some CT genes and oncogenes, as well as increasing proliferation/viability of cancer cells has been demonstrated in many researches. In addition to cancer cells, some believe that BORIS is also expressed in normal conditions and plays a universal function in cell division and regulation of genes. The following is a comprehensive review on contradictory views on the expression pattern and biological function of BORIS in normal, as well as cancer cells/tissues, and presents some evidence that support the expression of BORIS in cancer stem cells (CSCs) and advanced stage/poorer differentiation grade of cancers. Boris is involved in the regulation of CSC cellular and molecular features such as self-renewal, chemo-resistance, tumorigenicity, sphere-forming ability, and migration capacity. Finally, the role of BORIS in regulating two important signaling pathways including Wnt/β-catenin and Notch in CSCs, and its ability in recruiting transcription factors or chromatin-remodeling proteins to induce tumorigenesis is discussed.

Background

Brother Of the Regulator of Imprinted Sites (BORIS) or CTCFL (CCCTC-binding factor like) protein is recognized as a paralog of CTCF (CCCTC-binding factor). CTCF is a DNA binding protein that is involved in chromatin insulation, genomic imprinting, intra/interchromosomal interactions, and global three-dimensional genome organization [1,2,3,4,5,6]. BORIS and CTCF have identical 11 Zinc finger DNA-binding domains, and both seem to bind to similar DNA target sequences [7]. However, a study by Pugacheva et al. showed that only a subset of CTCF binding regions in cancer is occupied by BORIS [7, 8]. In spite of the very similar DNA binding domain in these two proteins, their amino and carboxyl domains have very little sequence homology, leading them to interact with different partners. Therefore, it may be the protein partners of these two proteins that determine their different chromatin regulating abilities and functional outcomes [1, 9,10,11,12]. The human BORIS gene is located at 20q13 and is comprised of 11 exons, 10 of which are coding [1]. Pugacheva el al. characterized 23 transcript variants of BORIS resulting in 17 protein isoforms. Different isoforms contain different zinc-fingers in their DNA-binding domain, have different amino and carboxyl termini, and have distinct expression profiles in various normal and cancer cells [13].

Many studies have attempted to explain the roles of BORIS in different cell types. Problems in understanding the biological roles of BORIS can be attributed to the lack of knowledge about the expression patterns of its isoforms in diverse cell types, the unknown identity of its potential interacting partners, and the experimental, analytical, and biological variability of the experiments performed [14]. According to many reports, BORIS is generally classified as a member of cancer testis (CT) genes, a group of genes which are normally expressed in germ cells, notably in testis, and also in a wide range of cancer types [15,16,17,18]. High expression of BORIS in testis suggests its involvement in the regulation of specific testis genes and meiosis of sperm [7,8,9, 19,20,21,22]. Abnormal expression of BORIS in a variety of cancer cells/tissues has been the main reason to categorize it as an oncogene with pathogenic roles in cell proliferation and tumorigenesis [7, 11, 13, 15,16,17,18, 21, 23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38]. Specific expression of BORIS in cancer stem cell (CSC) population and its role in the induction and maintenance of some important CSC properties suggest an association with severe malignancy and advanced stages of cancer [14, 32, 34, 39,40,41,42,43,44,45,46,47,48,49,50]. Several researchers reinforce the view that the expression of BORIS might not be limited to cancer cells/tissues and it might also be expressed in normal tissues and cells, and have a universal function [16, 17, 25, 27, 30, 51,52,53].

In this review, we explain in detail the reports that are related to the expression and general function of BORIS in normal tissues/cells such as testis/male germ cells. Subsequently, the expression of BORIS in various cancer/cancer stem cells, and its role in cell proliferation, tumorigenesis, and maintenance of CSC properties will be discussed. Finally, a mechanism for BORIS-mediated function in cancer and CSCs to regulate the expression of target genes and to induce tumorigenesis will be discussed.

Expression pattern and role of BORIS in normal cells/tissues

The first reports demonstrated that in contrast to the ubiquitous expression of CTCF in all somatic cell types, BORIS expression is restricted to testis. They also showed that during male germ cell development, BORIS and CTCF are expressed in a mutually exclusive manner. While CTCF expression was detected in post-meiotic round spermatids and spermatozoa, the expression of BORIS was only detected in primary spermatocytes, a cell type without CTCF expression. This finding indicated that the activation of BORIS expression is linked with the final round of mitosis of male germ-line cells [1, 18]. However, in subsequent studies, it was shown that BORIS is also expressed in pre-meiotic spermatogonia and pre-leptotene spermatocytes, where the expression of CTCF was also detected [21].

Thus far, some functions have been attributed to BORIS in testis. In fact, an extensive overlap has been recorded between the genome-wide erasure of methylation, re-setting of paternal DNA methylation patterns, and BORIS expression/silencing of CTCF [18], indicating that in testis, BORIS may play a role in the reprogramming of the paternal DNA [4, 18]. BORIS has also been implicated to be involved in the resetting of imprinting at the Igf2/H19 imprinting control region (ICR) in male germ cells [10]. In contrast, in somatic cells, CTCF is recognized as reader and protector of Igf2/H19 imprinting marks [11,12,13, 18, 21]. In addition, during spermatogenesis, BORIS has been detected as an inducer of multiple testis-specific genes which are suppressed by CTCF in somatic cells [7,8,9]. For example, important roles of BORIS in the induction of expression of some male germ cell/testis specific genes including ALF, SPANX-N, Gal3st1, and Prss50 which play crucial roles in meiosis and spermatogenesis have been reported [19,20,21,22]. This is consistent with the findings in BORIS knockout male mice which show subfertility and multiple defects in spermatogenesis, including a reduction in testis size, defective sperm production and a significant delay in the production of sperm [21, 22]. Overall, these studies show that in testis tissue, BORIS regulates gene expression, and exerts an important role in meiosis and production of the haploid sperm.

Although according to some reports, repressive effects of CTCF, p53, and promoter DNA methylation has restricted the expression of BORIS to testicular germ cells [1, 7, 11, 13, 15, 24, 25, 28], a few other studies have shown that in addition to male germ cells, BORIS transcripts are also expressed in other normal tissues such as human oocyte and ovary, and in various fetal tissues [13], indicating a role in meiosis during oogenesis [13, 44, 54], and early stages of preimplantation development [44] (Table 1). Significant levels of BORIS were also found in normal human skin and freshly isolated whole dermis, epidermis, or disaggregated primary keratinocytes [52] (Table 1).

Table 1 Cells/tissues that normally express BORIS

Several general regulatory functions have been proposed for BORIS in normal cells. Rosa-Garrido et al. exhibited that BORIS is involved in RNA transcription, cell cycle progression, and genome instability [52]. Experiments using the ectopic expression or inactivation of BORIS demonstrated that optimal levels of BORIS is needed to support normal cell division. In addition, BORIS knock-down caused a reduction in the synthesis of rRNA and global RNA, suggesting a role for BORIS in the licensing of RNA transcription [52]. BORIS has also been recognized as a RNA binding protein which is associated with actively translating ribosomes. These properties display its role in the regulation of genes at both the transcriptional and post-transcriptional levels [55]. Moreover, localization of BORIS within the nucleolus of cancer and normal cells suggested a role for this protein in nucleolar function [51].

Expression pattern of BORIS in cancer cells/tissues

In many tumors and cancer cell lines, hypomethylation of BORIS promoter leads to overexpression of BORIS [11, 13, 15,16,17, 23,24,25,26,27,28,29,30,31,32,33]. For instance, Vatolin et al. and Hong et al. demonstrated that the suppressed expression of BORIS (observed in normal somatic tissues and cell lines), is abrogated in various breast, neuroblastoma, prostate, melanoma, colon, and lung cancers [11]. In other studies, the comparative expression analysis of several cancer/testis genes revealed a high incidence of BORIS expression in uterine/endometrial, ovarian and cervical cancers in comparison with their normal tissues [17, 24, 32, 56]. In similar reports, analysis of BORIS in esophageal squamous cancers, pancreatic and hepatocellular carcinoma indicated that the expression of BORIS was significantly higher in these cancers than that in the adjacent non-cancerous tissues and normal cells [46, 47, 49, 57]. Some reports on prostate cancer, glioblastoma, and laryngeal squamous cell carcinoma indicate that BORIS protein is absent or present at low levels in non-tumorigenic cells and tissues, but it is present at variable higher levels in all cancer cell lines and tumors, indicating that BORIS might be used as a cancer biomarker [48, 50, 58]. D’Arcy and colleagues also showed that BORIS is expressed in all types of breast cancer cell lines, whereas primary normal breast cells and normal breast tissues do not express this protein [15]. The other evidence in support of BORIS as a tumor marker was obtained by detecting significantly higher level of BORIS in the leukocyte fraction in patients with different types of breast tumors compared to the control group [23]. Although, there is no report about BORIS expression in leukemic patients, some isoforms of BORIS are detected at high levels in leukemic cell lines [13]. Overall, the expression of BORIS in testis and many cancers (Table 2) led to its classification as a CT gene [15,16,17,18].

Table 2 Expression of BORIS in different cancer cells/tissues

In contrast to numerous reports indicating the expression of BORIS in cancers, some researchers report different findings. For instance, although BORIS is activated in a substantial fraction of melanoma samples, it does not appear to be present in all tumors of this kind [16]. Another research indicated that immortalized human ovarian surface epithelial cells (IOSE121) and four ovarian cancer cell lines (OVCAR3, SKOV3, A2780, and OVCAR429) do not express BORIS or other CT genes at significantly higher levels [17]. In a research by Hines et al., it was revealed that neither mature BORIS transcripts nor spliced variants are commonly expressed at detectable levels in human breast cancer cell lines and high grade breast carcinomas. There are also reports that show the absence of a significant difference in BORIS transcript levels in cancer and non-cancer cells. For example, expression of BORIS mRNA showed no significant difference between normal and cancerous prostate and bladder tissues [30], and also between some mouse cancer and non-cancer cell lines [59]. Similarly, according to the findings of Sheer and colleagues, the expression of BORIS was not restricted to the germ/cancer cells and its expression was also detected within the nucleolus of normal and cancer cells [51, 53].

Therefore, the above findings demonstrate the widespread expression of BORIS in normal and cancer cells. In reality, one reason for detection of BORIS in a variety of cell lines can be related to loss of the q arm of chromosome 16 (the locus of CTCF as a suppressor of BORIS) and gain of chromosome 20q13 (the locus of BORIS) during prolonged growth of normal and cancer cell lines in culture, a phenomenon that occur throughout adaptation of hES cells to growth in culture for a long time [60, 61]. Moreover, the use of different techniques to measure the expression of BORIS, isolated detection of its various isoforms, and various invalid commercial antibodies against BORIS or its specific isoforms have resulted in incomplete/contradictory findings on the expression pattern of BORIS in cancer cells/tissues. On the other hand, tumors are composed of heterogeneous combination of cells that exhibit distinct phenotypic characteristics and proliferative potentials, with having only a fraction of cells expressing BORIS. Accordingly, the level of BORIS transcript/protein might also depend on the grade of malignancy/benignity of tissues, leading to the detection of various expression levels for BORIS in different samples of the same type of tumor.

Function of BORIS in cancer cells/tissues

Expression of BORIS in cancer cells likely leads to its interference with CTCF function by competition for binding to CTCF DNA binding target sites. Due to their distinct amino- and carboxy-termini and different interacting proteins, the two proteins of BORIS and CTCF have opposite effects on gene expression [1]. While CTCF represses gene expression and blocks cell proliferation by arresting cells in a senescence-like state throughout the cell cycle [18, 62], BORIS associates with relatively open chromatin of active genes, and appears to activate a unique class of genes like oncogenes and CT genes (Fig. 1). Furthermore, the enforced expression of BORIS in fibroblasts leads to a significant decrease in apoptosis induction, increased anchorage-independent cell growth, and extended lifespan [7, 11, 18, 21, 25, 26, 34]. In contrast, the down-regulation of BORIS with specific siRNAs results in decreased cell proliferation/viability and induced cell death/apoptosis [49, 63] (Fig. 1).

Fig. 1
figure 1

Role of BORIS in tumorigenesis. Increased expression of BORIS, shifts the competition between CTCF and BORIS for binding to CTCF DNA-binding site in favor of BORIS. This leads to the replacement of CTCF by BORIS at promoters of some cancer-testis (CT) genes including MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-B1, MAGE-B4, GAGE-3-8, RAGE-2, NY-ESO-1 (CTAG1B), LAGE-1 (CTAG2), FerT and TSP50, and some non-CT genes such as BRCA1, Oct-3/4 (POU5F1), MYC, Rb2/p130, SBSN, and hTERT, and androgen, progesterone and estrogen receptors. Expression of target genes leads to cancer progression via activation of the network of CT genes, inhibition of apoptosis, induced cell growth, and increased proliferation and invasiveness of cancer cells

The results of some studies seem to support this theory that the ectopic expression of BORIS in normal human fibroblasts or low expressing cell lines would induce the replacement of CTCF by BORIS at promoters of several CT genes including MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-B1, MAGE-B4, GAGE-3-8, RAGE-2, NY-ESO-1 (CTAG1B), LAGE-1 (CTAG2), FerT and TSP50, resulting in the de-repression of the target CT genes [11, 18, 25, 26, 29, 64,65,66,67]. In addition to CT genes, BORIS participates in regulation of some non-CT genes such as BRCA1, Oct-3/4 (POU5F1), MYC, Rb2/p130, SBSN and hTERT (human telomerase reverse transcriptase) which are known to be involved in cancer progression [30, 34, 68, 69] (Figs. 1, 2).

Fig. 2
figure 2

BORIS function in sustaining cancer stem cell (CSC) properties. BORIS induces the expression of some important CSC markers such as ALDH1, ABCG2, hTERT, NANOG, OCT4 and SOX2 in cancer cells. BORIS is also recognized as an inducer of Wnt and Notch signaling pathways that play important roles in the maintenance of CSC properties such as self-renewal, tumor-sphere formation, chemoresistance, anchorage independent growth, and migration/invasion capacity

Maintenance of telomeres is necessary to inhibit replicative senescence. Telomerase activity which is required to stabilize telomere length has not been detected in differentiated somatic cells, but is detected in proliferative immortal cells, such as germ cells, stem cells, and cancer cells [70,71,72]. In the majority of telomerase-positive cells such as cancer cell lines and tumors, hypermethylation of hTERT exon 1 region prevents the binding and prevents the repressive effects of CTCF [73,74,75,76]. Although methylation of exon 1 region is the most prevalent mechanism to regulate hTERT in tumor cells and tissues, it is found that in some cancer cells, the expression of BORIS prevents the repressive effects of CTCF on hTERT gene, and permits its transcription [34]. Therefore, the expression of BORIS could be an alternative mechanism for the induction of hTERT in cancer cells. This indicates that BORIS might have important regulatory roles in tumor immortalization during tumorigenesis. In breast tumors, estrogen and progesterone have been demonstrated to promote tumorigenesis [35, 36]. It is accepted that BORIS activates the promoters of genes for progesterone and estrogen receptors, suggesting a role for BORIS in the progression of breast tumors [15]. In a similar role, in prostate cancer, androgen receptor (AR) mediates various functions of androgens essential for cell viability, development and invasion in both androgen dependent and independent prostate cancers [37, 38]. BORIS is capable to activate the expression of endogenous AR gene in prostate cell lines. This indicates that BORIS might be involved in the growth and proliferation of prostate tumors [48]. Taken together, these results show the involvement of BORIS in tumorigenesis, cell proliferation and invasiveness of cancer cells and could point to an oncogenic role for BORIS in cancer (Fig. 1).

On the other hand, there are also opposite viewpoints on the role of BORIS in cancer. Several findings discuss that BORIS is not a leading CT gene and its presence is not necessary for the expression of CT genes [11, 16, 25, 66, 77,78,79]. For example; it has been shown that the expression of some MAGE-A family and of other CT genes in melanoma, glioma stem cells and head and neck squamous cell carcinoma (HNSCC) is observed in the absence of BORIS, suggesting that BORIS might not be an obligatory factor for the activation of CT genes. Accordingly, BORIS positive tumors do not necessarily express high levels of other CT genes and the exogenous expression of BORIS does not always lead to the hypomethylation of promoter in CT genes [16, 27, 64, 78, 80]. Furthermore, in contrast to the previous findings that show opposing roles for BORIS and CTCF, in one study it has been detected that BORIS similar to CTCF caused a significant reduction in cell proliferation and clonogenic capacity. Thus, against previous hypothesis that considers an oncogenic property for BORIS, these data indicate that BORIS and CTCF might act as a tumor suppressor. Accordingly, expression of BORIS in many cancers implies that genetic and epigenetic dysregulation in cancer might result in BORIS induction. Therefore, BORIS can be an effect rather than a cause of transformation [6, 7].

Opposite results in the overexpression of BORIS in different cell types might be a cell context-dependent phenomenon. For example, it is possible that along with BORIS, other transcription or epigenomic regulatory factors be effective to induce CT gene expression and these factors may be expressed in specific cell types. It is also likely that a particular isoform of BORIS is necessary for the regulation of some CT genes. Furthermore, some of the differential effects of BORIS may be attributable to a dose-dependent effect of BORIS on activation of downstream targets and the number of CTCF sites occupied by BORIS. For instance, Gaykalova et al. showed that only lower BORIS concentrations stimulate the highest expression of suprabasin gene as a non-CT target of BORIS, while higher concentrations of BORIS has less inducer effects [79].

In conclusion, according to some reports, BORIS is recognized as a main participator in the induction of some important CT and non-CT genes in cancer, and thus has a role in growth, proliferation, invasion and tumorigenesis of cancer cells. However, the epigenetic state of the cell, the level of expression of genes in CT gene network, and the level of expression of BORIS itself may affect proliferation and tumorigenesis of cancer cells (Fig. 1).

BORIS in cancer stem cells

In addition to cancer cells, BORIS expression has been detected in some pluripotent cells including human embryonic stem (ES) [13, 44] and embryonal carcinoma (EC) cells (TERA-1, TERA-2, NT2 and NCCIT) [30, 43]. Pluripotent cells and undifferentiated tumor cells share several hallmark traits including self-renewal and differentiation ability, which provide the basis of unlimited proliferative capacity, immortality, and capacity to produce progenitors that differentiate into other cell types [81,82,83,84]. Furthermore, expression of some important pluripotent markers including OCT4, SOX2, KIF4 and c-MYC genes which are essential for the maintenance of pluripotency, have been found in many cancer cells and tumors [85,86,87,88,89,90,91,92,93,94,95,96], indicating that transformation to a cancerous state requires some characteristics found in stem cells [97]. As a result, BORIS might be involved in the establishment of a state of pluripotency, which is also present in a subpopulation of cancer cells called cancer stem cells (CSCs). This hypothesis has been proved to be true by detection of BORIS expression in cancer stem cells (CSCs) and identification of its role in the induction of some important CSC markers and maintenance of CSC properties (Fig. 2).

Cancer stem cells are a group of pluripotent cells that have been detected in most types of solid and hematologic cancers. Similar to normal stem cells, CSCs have uncontrolled proliferation ability, enhanced potential to self-renew, and differentiation capacity into non-CSC progeny [83, 98, 99]. Moreover CSCs, have a higher intrinsic resistance to conventional therapies, such as chemotherapy and radiotherapy through a variety of mechanisms such as increased expression of detoxifying ALDH enzymes, enhanced DNA repair activity, reduced drug activation via quiescence, and increased drug efflux by up regulation of ATP-binding cassette (ABC) transporters [100,101,102,103,104]. Indeed, enhanced activation of one or more signal transduction pathways including the Notch, Hedgehog (HH), and Wnt pathways has been observed in CSCs of many different cancer types. These pathways play an important role in the maintenance of self-renewal potential and ability to avoid being affected by chemo and radio therapy in CSCs [83, 105,106,107,108,109,110,111,112] (Fig. 2). Accordingly, this sub-population of tumor cells plays an important role in tumor growth, recurrence, metastasis, and resistance to treatment. There are various methods for the identification and isolation of CSCs including cell sorting by fluorescence-activated cell sorting (FACS) or magnetic activated cell sorting (MACS) based on the expression of specific surface biomarkers [113,114,115], sphere-forming assay which is an in vitro method to evaluate the ability of CSCs to form spheres in serum-free medium by anchorage independent growth in suspension [116,117,118,119], and finally, functional cell sorting based on biological characteristics of the cells (side population (SP) cell sorting). SP phenotype is a CSC property that defines the cells that express ABC drug transporters, such as MDR1 (P-glycoprotein) and ABCG2. SP cells are characterized by the efflux of fluorescence dyes such as Hoechst 33342 and Rhodamin 123 [120,121,122].

There is some evidence that support a potential relationship between BORIS expression and CSCs. Expression of BORIS has been found in some EC  [30, 43], and CSCs from neuroblastoma, cervical, colon and breast cancers [39,40,41,42]. In addition, expression of BORIS shows a positive correlation with specific stemness and CSC markers [40, 43, 44]. Thus, BORIS is considered to be a positive regulator of cancer/stem cell markers, and to have a role in the maintenance of CSC population in tumors [34, 39, 40, 44].

Related to the presence of BORIS in CSC-enriched population of cancer cells, BORIS mRNA was detected at significantly higher levels in SP cells and tumor spheres compared to non-SP and parental cells [39]. Furthermore, Garikapati et al. showed that CD44/CD133 positive cells that are recognized as CSCs in neuroblastoma have higher levels of BORIS in comparison with CD44/CD133 negative cells [40]. In other studies, BORIS was found to be expressed in cervical and colon CSCs [41, 42]. These findings are in accordance with the findings of Yamada and colleagues indicating that considerable numbers of CT genes including BORIS are expressed in cancer stem like cells. They classified these CT genes in a sub-category called cancer/testis/stem (CTS) genes which define a class of genes expressed in the testis and CSCs [42]. In another research by Pugacheva el al. it was observed that BORIS expression in hES cells disappears upon differentiation, indicating an association with pluripotency [13].

In addition to detecting BORIS in pluripotent and CSCs population, a mutual relationship was found between the expression of BORIS and some fundamental CSC markers and traits. As a noteworthy example, BORIS positive cells express ABCG2 and do not take up Hoechst, so are defined as SP cells [39]. More investigations showed higher level of stem cell (NANOG, OCT4, SOX2) and cancer stem cell (CD44 and ALDH1) markers in BORIS-positive cells in comparition to BORIS-negative cancer cells [40, 43]. In hepatocellular carcinoma tissues, correlation of BORIS expression with liver CSC marker CD90 is another reason for its correlation with CSC markers [46]. Association of BORIS and CSC markers were reinforced when it was observed that some CSC markers such as ALDH1, NANOG, OCT4, SOX2 and ABCG2 were generally down-regulated in all tumor cells after BORIS silencing. In addition, overexpression of BORIS also significantly increased the expression of previously mentioned CSC markers [39,40,41,42, 123]. As it was previously implied, one of the cancer/stem cell markers that is inducible by BORIS is hTERT telomerase gene [34]. It is shown that the expression of telomerase is essential for self-renewal of CSCs [124,125,126]. Therefore, BORIS might be an important factor in self-renewal and immortal capacity of CSCs by induction of hTERT. BORIS has also been recognized as an essential factor for maintaining CSC properties. For example, correlation of BORIS with sphere formation, tumor-initiating ability and maintenance of CSCs in cervical, colon, and breast cancer has been shown in separate reports [39, 41]. Moreover, Garikapati et al. Showed that the expression of BORIS effectively controlled tumurosphere formation and anchorage independent growth in neuroblastoma CSCs [40]. A recent study has reported that BORIS affects the CSC-like traits of human liver cancer cells such as self-renewal, tumor sphere-forming ability, tumorigenicity, chemo resistance and migration/invasion capacity through regulating of OCT4 gene expression [123]. The POU domain transcription factor OCT4 by regulating target genes such as NANOG and SOX2 has been recognized as the most important pluripotency factor and master regulator in the maintenance of CSC-like phenotypes such as self-renewal, chemo-resistance migration and invasion [127,128,129,130]. Consequently, in accordance with these findings, BORIS may serve as a biomarker of CSCs and has a probable role in sustaining the stemness properties of CSCs (Fig. 2).

In a research by Soltanian et al. retinoic acid induced differentiation of P19 (as a pluripotent embryonal carcinoma cell line), was concomitant by significant depression of some important pluripotency markers such as OCT4, NANOG and SSEA1, and was not accompanied with significant variations of BORIS expression. In fact, P19 cell line is a heterogeneous population of cells comprising a small population of BORIS-expressing cancer stem cells. Therefore, in order to investigate the changes in the expression level of BORIS during retinoic acid induced differentiation of CSCs, stem-like cells must be first isolated according to their markers and properties [39, 40].

Association of BORIS with advanced stages of cancers

According to CSC hypothesis, CSCs typically represent a small proportion of total cells of a given tumor that involve in tumor growth, recurrence, metastasis, and treatment resistance. Therefore, it has been shown that CSCs are more frequent in highly aggressive and refractory tumors [118, 131]. Applying this hypothesis to many studies that highlighted the correlation of BORIS expression with poor overall survival of different cancer patients/poorer differentiation grade and recurrence of cancer emphasize that BORIS has a decisive role in maintaining CSCs.

Furthermore, function of BORIS as an inducer of CSC markers and CSC-like traits is consistent with a lot of reports that show expression of BORIS to be associated with poor overall survival/more severe malignancy and advanced stages in different cancers [14]. For instance, in epithelial ovarian cancer and cervical cancer, high level of BORIS is associated with poorer prognosis/less median survival times of patients and advanced cancer stages [41, 45]. BORIS expression was also correlated with tumor size, differentiation grade and tumor recurrences in hepatocellular carcinoma. In this kind of cancer, patients with high BORIS expression had reduced overall survival rate which suggests that BORIS could be used as a diagnostic index of liver cancer [46, 47]. In prostate cancer a positive correlation has also been detected between higher levels of BORIS and higher Gleason score (which measures prostate tumor differentiation and predicts the aggressiveness of the disease), higher T-stage (which reflects the progression of the cancer, e.g., tumor size, metastatic potential, invasiveness), and increased androgen receptor protein levels [48]. Moreover, in prostate cancer, greater BORIS/CTCF ratio was detected in cancer and metastases compared to benign tissue, and an increase in this ratio correlated with higher Gleason’s grade, positive surgical margins, and increased tumor volume [132]. In another report, BORIS expression was significantly associated with lymph node metastasis in esophageal squamous cell cancer (ESCC), and patients with BORIS-positive tumors had a poor overall survival in this cancer, suggesting that BORIS is associated with metastatic activity of ESCC cells in the early stage and BORIS can be considered as a potential biomarker for esophageal cancer patients with a poor prognosis [49]. A research on endometrial cancer showed that increased BORIS mRNA expression level associates with cancer progression and poor survival, so that all the clinically established markers for aggressive endometrial carcinoma including high age, non-endometroid histology, high grade, and hormone receptor loss were significantly associated with high BORIS mRNA levels [32]. In addition, Schwarzenbach el al. indicated that serum levels of cell-free BORIS mRNA were significantly higher in patients with invasive carcinomas than in patients with benign breast diseases or healthy women [133]. Another study on laryngeal squamous cell carcinomas revealed that patients having BORIS 7+ (BORIS transcript variants containing exon 7)/BORIS 7− (BORIS transcript variants lacking exon 7) ratio ≥ 1 had a higher rate of disease relapse than patients with BORIS 7+/BORIS 7− ratio < 1 [50].

BORIS and its mechanistic connections to tumorigenesis

The mechanism of BORIS function in regulating cancer stemness as well as tumorigenesis has been shown by its involvement in modulating two important signaling pathways in CSCs including Wnt/β-catenin and Notch (Fig. 2). It has been confirmed that abnormal Wnt/β-catenin signaling pathway plays an important role in the maintenance of CSC properties and epithelial–mesenchymal transition (EMT) in various cancers [134,135,136,137,138,139]. EMT is a process by which epithelial cells gain migratory and invasive properties to acquire features similar to mesenchymal stem cells. This process plays a critical role in cancer metastasis [140]. It is proved that EMT produces CSC like cells with self-renewal and migratory capability [96, 141, 142]. In this regard, it has been shown that BORIS can modulate the levels of Wnt5a, β-catenin, TCF, and pLRP as key players of Wnt/β-catenin signaling pathway. Hence, by regulation of metastasis/EMT through Wnt/β-catenin pathway, BORIS is responsible for cancer stemness [40]. A relationship between BORIS and EMT phenotype has also been confirmed in BCM1 cells as micrometastatic breast cancer cells gathered from bone marrow of breast cancer patients. Interestingly, these cells which express high level of BORIS have some cancer stem cell characteristics and EMT like invasive phenotype [133, 143, 144]. Additionally, BORIS has also been recognized as an inducer of Notch pathway and its expression has been detected in cell lines derived from several solid tumors overexpressing NOTCH3 [110, 145,146,147,148,149]. All together, these results indicate that BORIS plays a principle role in the maintenance of cancer stemness by interacting with WNT/ß-catenin and Notch signaling pathway.

It has been reported that BORIS is an epigenetic modifier, and its binding to promoters of target genes leads to the recruitment of additional transcription factors or chromatin-remodeling proteins that alter the epigenetic status, chromatin conformation, and transcription of these genes (Fig. 3). For example, it has been demonstrated that BORIS/CTCF expression ratio is associated with DNA hypomethylation [45]. Moreover, the overexpression of BORIS is correlated with aberrant expression of multiple proto-oncogenes and CT genes such as NY-ESO-1, MAGE-A1 and MAGE-A3 via induction of promoter demethylation [11, 25, 26, 69, 79, 150]. Furthermore, in vitro studies show recruitment of PRMT7 and SET1A to chromatin induced by BORIS [12]. SET1A and PRMT7 have been recognized as a H3K4 methyltransferase and arginine methyltransferase, respectively [68, 151]. Therefore, BORIS induces the expression of MAGEA1-A4, BAG1, BRCA1, SBSN, NY-ESO-1 and MYC genes via recruitment of histone modifiers onto the promoters of target genes which results in permissive/active histone modifications such as trimethylation of lysine 4 of histone H3 tail (H3K4me3) and acetylation of lysine 14 of histone H3 tail (H3K14Ac) [25, 66, 68, 79, 152, 153]. The role of BORIS as a chromatin regulator protein was confirmed by its ability to bind in NOTCH3 promoter and increasing the H3K4me3/H3K27me3 ratio leading to abnormal upregulation of NOTCH3 in cancer cells [145]. A recent study by Liu el al. showed that BORIS promotes CSC-like traits of human liver cancer cells by epigenetic up-regulating of OCT4. In fact, BORIS regulates OCT4 gene expression via histone methylation modification as reflected by increasing the H3K4me2/H3K27me3 ratio and creates a permissive/active chromatin conformation [123] (Fig. 3). Another mechanism for BORIS-mediated activation of genes has been reported for NY-ESO-1. Kang et al. provided evidence that recruitment of Sp1 to NY-ESO-1 promoter is a mechanism by which BORIS induces NY-ESO-1 in lung cancer [29]. Sp1 is a transcription factor that activates promoters via recruitment of additional regulatory proteins, leading to the formation of a functional transcriptional machinery [154, 155]. Altogether, the results of these studies suggest that BORIS acts as a type of genomic recruiter/chromatin scaffold that recruits many interacting partners and induces open chromatin conformation in the promoter of target genes. This conclusion was further supported by the finding that BORIS is co localized with H3K4me3 and Pol II at transcriptionally active promoters [21].

Fig. 3
figure 3

Role of BORIS as an epigenetic modifier regulating the expression of multiple proto-oncogenes and CT genes. BORIS recruits histone modifier complexes to the promoter of target genes to induce tri-methylation of lysine 4 of histone H3 tail, and methylation of arginine and acetylation of lysine 14 of histone H3 tail. BORIS also is involved in promoter methylation/demethylation of target genes. Therefore, BORIS creates a permissive/active chromatin conformation via epigenetic modifications that lead to the upregulation of target genes such as CT genes, Oct4 and Notch3

Conclusion

In this review we describe the expression pattern and functions of BORIS or CTCF-like protein which has been identified as a paralog of CTCF, an old protein with known functions and pattern of expression. Although there are contradictory reports on the expression pattern and function of BORIS, but it has been recognized as a CT gene that is normally expressed in male germ line cells in testis, and is frequently deregulated in many cancers. In cancer cells, BORIS appears to regulate the activation of other CT genes and oncogenes, affecting cell proliferation and invasive ability of cancer cells. Recent reports show a correlation between BORIS and CSCs. According to these finding, BORIS has also been recognized as an inducer of some important CSC markers and as a probable player in the maintenance of CSCs in advanced cancers. However, further studies are needed to clarify the role of BORIS in sustaining CSC properties, and in advanced stage/poorer differentiation grade cancers.

Abbreviations

CT:

cancer/testis

CSCs:

cancer stem cells

BORIS:

Brother Of the Regulator of Imprinted Sites

CTCFL:

CCCTC-binding factor like

ICR:

imprinting control region

hTERT:

human telomerase reverse transcriptase

HNSCC:

head and neck squamous cell carcinoma

EC:

embryonal carcinoma

ABC:

ATP-binding cassette

FACS:

fluorescence-activated cell sorting

MACS:

magnetic activated cell sorting

SP:

side population

CTS:

cancer/testis/stem

ESCC:

esophageal squamous cell cancer

EMT:

epithelial–mesenchymal transition

References

  1. Loukinov DI, Pugacheva E, Vatolin S, Pack SD, Moon H, Chernukhin I, et al. BORIS, a novel male germ-line-specific protein associated with epigenetic reprogramming events, shares the same 11-zinc-finger domain with CTCF, the insulator protein involved in reading imprinting marks in the soma. Proc Natl Acad Sci USA. 2002;99(10):6806–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wallace JA, Felsenfeld G. We gather together: insulators and genome organization. Curr Opin Genet Dev. 2007;17(5):400–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lewis A, Murrell A. Genomic imprinting: CTCF protects the boundaries. Curr Biol. 2004;14(7):R284–6.

    Article  CAS  PubMed  Google Scholar 

  4. Hore TA, Deakin JE, Marshall Graves JA. The evolution of epigenetic regulators CTCF and BORIS/CTCFL in amniotes. PLoS Genet. 2008;4(8):e1000169.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Phillips JE, Corces VG. CTCF: master weaver of the genome. Cell. 2009;137(7):1194–211.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Marshall AD, Bailey CG, Rasko JE. CTCF and BORIS in genome regulation and cancer. Curr Opin Genet Dev. 2014;24:8–15.

    Article  CAS  PubMed  Google Scholar 

  7. Tiffen JC, Bailey CG, Marshall AD, Metierre C, Feng Y, Wang Q, et al. The cancer-testis antigen BORIS phenocopies the tumor suppressor CTCF in normal and neoplastic cells. Int J Cancer. 2013;133(7):1603–13.

    Article  CAS  PubMed  Google Scholar 

  8. Pugacheva EM, Rivero-Hinojosa S, Espinoza CA, Méndez-Catalá CF, Kang S, Suzuki T, et al. Comparative analyses of CTCF and BORIS occupancies uncover two distinct classes of CTCF binding genomic regions. Genome Biol. 2015;16(1):161.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Campbell AE, Martinez SR, Miranda JJ. Molecular architecture of CTCFL. Biochem Biophys Res Commun. 2010;396(3):648–50.

    Article  CAS  PubMed  Google Scholar 

  10. Ohlsson R, Lobanenkov V, Klenova E. Does CTCF mediate between nuclear organization and gene expression? BioEssays. 2010;32(1):37–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vatolin S, Abdullaev Z, Pack SD, Flanagan PT, Custer M, Loukinov DI, et al. Conditional expression of the CTCF-paralogous transcriptional factor BORIS in normal cells results in demethylation and derepression of MAGE-A1 and reactivation of other cancer-testis genes. Cancer Res. 2005;65(17):7751–62.

    Article  CAS  PubMed  Google Scholar 

  12. Jelinic P, Stehle JC, Shaw P. The testis-specific factor CTCFL cooperates with the protein methyltransferase PRMT7 in H19 imprinting control region methylation. PLoS Biol. 2006;4(11):e355.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Pugacheva EM, Suzuki T, Pack SD, Kosaka-Suzuki N, Yoon J, Vostrov AA, et al. The structural complexity of the human BORIS gene in gametogenesis and cancer. PLoS ONE. 2010;5(11):e13872.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Martin-Kleiner I. BORIS in human cancers—a review. Eur J Cancer. 2012;48(6):929–35.

    Article  CAS  PubMed  Google Scholar 

  15. D’Arcy V, Pore N, Docquier F, Abdullaev ZK, Chernukhin I, Kita GX, et al. BORIS, a paralogue of the transcription factor, CTCF, is aberrantly expressed in breast tumours. Br J Cancer. 2008;98(3):571–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kholmanskikh O, Loriot A, Brasseur F, De Plaen E, De Smet C. Expression of BORIS in melanoma: lack of association with MAGE-A1 activation. Int J Cancer. 2008;122(4):777–84.

    Article  CAS  PubMed  Google Scholar 

  17. Woloszynska-Read A, James SR, Link PA, Yu J, Odunsi K, Karpf AR. DNA methylation-dependent regulation of BORIS/CTCFL expression in ovarian cancer. Cancer Immun. 2007;7:21.

    PubMed  PubMed Central  Google Scholar 

  18. Klenova EM, Morse HC 3rd, Ohlsson R, Lobanenkov VV. The novel BORIS + CTCF gene family is uniquely involved in the epigenetics of normal biology and cancer. Semin Cancer Biol. 2002;12(5):399–414.

    Article  CAS  PubMed  Google Scholar 

  19. Kouprina N, Noskov VN, Pavlicek A, Collins NK, Schoppee Bortz PD, Ottolenghi C, et al. Evolutionary diversification of SPANX-N sperm protein gene structure and expression. PLoS ONE. 2007;2(4):e359.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Kim M, Li D, Cui Y, Mueller K, Chears WC, DeJong J. Regulatory factor interactions and somatic silencing of the germ cell-specific ALF gene. J Biol Chem. 2006;281(45):34288–98.

    Article  CAS  PubMed  Google Scholar 

  21. Sleutels F, Soochit W, Bartkuhn M, Heath H, Dienstbach S, Bergmaier P, et al. The male germ cell gene regulator CTCFL is functionally different from CTCF and binds CTCF-like consensus sites in a nucleosome composition-dependent manner. Epigenetics Chromatin. 2012;5(1):8.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Suzuki T, Kosaka-Suzuki N, Pack S, Shin DM, Yoon J, Abdullaev Z, et al. Expression of a testis-specific form of Gal3st1 (CST), a gene essential for spermatogenesis, is regulated by the CTCF paralogous gene BORIS. Mol Cell Biol. 2010;30(10):2473–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. D’Arcy V, Abdullaev ZK, Pore N, Docquier F, Torrano V, Chernukhin I, et al. The potential of BORIS detected in the leukocytes of breast cancer patients as an early marker of tumorigenesis. Clin Cancer Res. 2006;12(20 Pt 1):5978–86.

    Article  PubMed  CAS  Google Scholar 

  24. Risinger JI, Chandramouli GV, Maxwell GL, Custer M, Pack S, Loukinov D, et al. Global expression analysis of cancer/testis genes in uterine cancers reveals a high incidence of BORIS expression. Clin Cancer Res. 2007;13(6):1713–9.

    Article  CAS  PubMed  Google Scholar 

  25. Hong JA, Kang Y, Abdullaev Z, Flanagan PT, Pack SD, Fischette MR, et al. Reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter coincides with derepression of this cancer-testis gene in lung cancer cells. Cancer Res. 2005;65(17):7763–74.

    Article  CAS  PubMed  Google Scholar 

  26. Smith IM, Glazer CA, Mithani SK, Ochs MF, Sun W, Bhan S, et al. Coordinated activation of candidate proto-oncogenes and cancer testes antigens via promoter demethylation in head and neck cancer and lung cancer. PLoS ONE. 2009;4(3):e4961.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Cuffel C, Rivals JP, Zaugg Y, Salvi S, Seelentag W, Speiser DE, et al. Pattern and clinical significance of cancer-testis gene expression in head and neck squamous cell carcinoma. Int J Cancer. 2011;128(11):2625–34.

    Article  CAS  PubMed  Google Scholar 

  28. Renaud S, Pugacheva EM, Delgado MD, Braunschweig R, Abdullaev Z, Loukinov D, et al. Expression of the CTCF-paralogous cancer-testis gene, brother of the regulator of imprinted sites (BORIS), is regulated by three alternative promoters modulated by CpG methylation and by CTCF and p53 transcription factors. Nucleic Acids Res. 2007;35(21):7372–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kang Y, Hong JA, Chen GA, Nguyen DM, Schrump DS. Dynamic transcriptional regulatory complexes including BORIS, CTCF and Sp1 modulate NY-ESO-1 expression in lung cancer cells. Oncogene. 2007;26(30):4394–403.

    Article  CAS  PubMed  Google Scholar 

  30. Hoffmann MJ, Muller M, Engers R, Schulz WA. Epigenetic control of CTCFL/BORIS and OCT4 expression in urogenital malignancies. Biochem Pharmacol. 2006;72(11):1577–88.

    Article  CAS  PubMed  Google Scholar 

  31. Chen K, Huang W, Huang B, Wei Y, Li B, Ge Y, et al. BORIS, brother of the regulator of imprinted sites, is aberrantly expressed in hepatocellular carcinoma. Genet Test Mol Biomarkers. 2012;17(2):160–5.

    Article  CAS  PubMed  Google Scholar 

  32. Hoivik EA, Kusonmano K, Halle MK, Berg A, Wik E, Werner HM, et al. Hypomethylation of the CTCFL/BORIS promoter and aberrant expression during endometrial cancer progression suggests a role as an Epi-driver gene. Oncotarget. 2014;5(4):1052.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett. 2017;396:130–7.

    Article  PubMed  CAS  Google Scholar 

  34. Renaud S, Loukinov D, Alberti L, Vostrov A, Kwon YW, Bosman FT, et al. BORIS/CTCFL-mediated transcriptional regulation of the hTERT telomerase gene in testicular and ovarian tumor cells. Nucleic Acids Res. 2010;39(3):862–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Medina D. Mammary developmental fate and breast cancer risk. Endocr Relat Cancer. 2005;12(3):483–95.

    Article  CAS  PubMed  Google Scholar 

  36. Girard GM, Vanzulli SI, Cerliani JP, Bottino MC, Bolado J, Vela J, et al. Association of estrogen receptor-α and progesterone receptor A expression with hormonal mammary carcinogenesis: role of the host microenvironment. Breast Cancer Res. 2007;9(2):R22.

    Article  CAS  Google Scholar 

  37. Hara T, Miyazaki H, Lee A, Tran CP, Reiter RE. Androgen receptor and invasion in prostate cancer. Can Res. 2008;68(4):1128–35.

    Article  CAS  Google Scholar 

  38. Hååg P, Bektic J, Bartsch G, Klocker H, Eder IE. Androgen receptor down regulation by small interference RNA induces cell growth inhibition in androgen sensitive as well as in androgen independent prostate cancer cells. J Steroid Biochem Mol Biol. 2005;96(3):251–8.

    Article  PubMed  CAS  Google Scholar 

  39. Alberti L, Losi L, Leyvraz S, Benhattar J. Different effects of BORIS/CTCFL on stemness gene expression, sphere formation and cell survival in epithelial cancer stem cells. PLoS ONE. 2015;10(7):e0132977.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Garikapati KR, Patel N, Makani VKK, Cilamkoti P, Bhadra U, Bhadra MP. Down-regulation of BORIS/CTCFL efficiently regulates cancer stemness and metastasis in MYCN amplified neuroblastoma cell line by modulating Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 2017;484(1):93–9.

    Article  CAS  PubMed  Google Scholar 

  41. Asano T, Hirohashi Y, Torigoe T, Mariya T, Horibe R, Kuroda T, et al. Brother of the regulator of the imprinted site (BORIS) variant subfamily 6 is involved in cervical cancer stemness and can be a target of immunotherapy. Oncotarget. 2016;7(10):11223.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Yamada R, Takahashi A, Torigoe T, Morita R, Tamura Y, Tsukahara T, et al. Preferential expression of cancer/testis genes in cancer stem-like cells: proposal of a novel sub-category, cancer/testis/stem gene. HLA. 2013;81(6):428–34.

    CAS  Google Scholar 

  43. Alberti L, Renaud S, Losi L, Leyvraz S, Benhattar J. High expression of hTERT and stemness genes in BORIS/CTCFL positive cells isolated from embryonic cancer cells. PLoS ONE. 2014;9(10):e109921.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Monk M, Hitchins M, Hawes S. Differential expression of the embryo/cancer gene ECSA(DPPA2), the cancer/testis gene BORIS and the pluripotency structural gene OCT4, in human preimplantation development. Mol Hum Reprod. 2008;14(6):347–55.

    Article  CAS  PubMed  Google Scholar 

  45. Woloszynska-Read A, Zhang W, Yu J, Link PA, Mhawech-Fauceglia P, Collamat G, et al. Coordinated cancer germline antigen promoter and global DNA hypomethylation in ovarian cancer: association withBORIS/CTCF expression ratio and advanced stage. Clin Cancer Res. 2011;17(8):2170–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen K, Huang W, Huang B, Wei Y, Li B, Ge Y, et al. BORIS, brother of the regulator of imprinted sites, is aberrantly expressed in hepatocellular carcinoma. Genet Test Mol Biomarkers. 2013;17(2):160–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhao R, Chen K, Zhou J, He J, Liu J, Guan P, et al. The prognostic role of BORIS and SOCS3 in human hepatocellular carcinoma. Medicine. 2017;96(12):e6420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cheema Z, Hari-Gupta Y, Kita GX, Farrar D, Seddon I, Corr J, et al. Expression of the cancer-testis antigen BORIS correlates with prostate cancer. Prostate. 2014;74(2):164–76.

    Article  CAS  PubMed  Google Scholar 

  49. Okabayashi K, Fujita T, Miyazaki J, Okada T, Iwata T, Hirao N, et al. Cancer-testis antigen BORIS is a novel prognostic marker for patients with esophageal cancer. Cancer Sci. 2012;103(9):1617–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kujundžić RN, Grbeša I, Ivkić M, Krušlin B, Konjevoda P, Trošelj KG. Possible prognostic value of BORIS transcript variants ratio in laryngeal squamous cell carcinomas—a pilot study. Pathol Oncol Res. 2014;20(3):687–95.

    Article  Google Scholar 

  51. Jones TA, Ogunkolade BW, Szary J, Aarum J, Mumin MA, Patel S, et al. Widespread expression of BORIS/CTCFL in normal and cancer cells. PLoS ONE. 2011;6(7):e22399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rosa-Garrido M, Ceballos L, Alonso-Lecue P, Abraira C, Delgado MD, Gandarillas A. A cell cycle role for the epigenetic factor CTCF-L/BORIS. PLoS ONE. 2012;7(6):e39371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hines WC, Bazarov AV, Mukhopadhyay R, Yaswen P. BORIS (CTCFL) is not expressed in most human breast cell lines and high grade breast carcinomas. PLoS ONE. 2010;5(3):e9738.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Link PA, Zhang W, Odunsi K, Karpf AR. BORIS/CTCFL mRNA isoform expression and epigenetic regulation in epithelial ovarian cancer. Cancer Immun. 2013;13:6.

    PubMed  PubMed Central  Google Scholar 

  55. Ogunkolade BW, Jones TA, Aarum J, Szary J, Owen N, Ottaviani D, et al. BORIS/CTCFL is an RNA-binding protein that associates with polysomes. BMC Cell Biol. 2013;14(1):52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Velázquez-Hernández N, Reyes-Romero M, Barragán-Hernández M, Guerrero-Romero F, Rodríguez-Moran M, Aguilar-Duran M, et al. BORIS and CTCF are overexpressed in squamous intraepithelial lesions and cervical cancer. Genet Mol Res. 2015;14(2):6094–100.

    Article  PubMed  CAS  Google Scholar 

  57. Ghochikyan A, Mkrtichyan M, Loukinov D, Mamikonyan G, Pack SD, Movsesyan N, et al. Elicitation of T cell responses to histologically unrelated tumors by immunization with the novel cancer-testis antigen, brother of the regulator of imprinted sites. J Immunol. 2007;178(1):566–73.

    Article  CAS  PubMed  Google Scholar 

  58. Freitas M, Malheiros S, Stavale JN, Biassi TP, Zamuner FT, de Souza Begnami M, et al. Expression of cancer/testis antigens is correlated with improved survival in glioblastoma. Oncotarget. 2013;4(4):636–46.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Soltanian S, Dehghani H, Matin MM, Bahrami AR. Expression analysis of BORIS during pluripotent, differentiated, cancerous, and non-cancerous cell states. Acta Biochim Biophys Sin. 2014;46(8):647–58.

    Article  CAS  PubMed  Google Scholar 

  60. Mastracci TL, Shadeo A, Colby SM, Tuck AB, O’Malley FP, Bull SB, et al. Genomic alterations in lobular neoplasia: a microarray comparative genomic hybridization signature for early neoplastic proliferation in the breast. Genes Chromosomes Cancer. 2006;45(11):1007–17.

    Article  CAS  PubMed  Google Scholar 

  61. de Necochea-Campion R, Ghochikyan A, Josephs SF, Zacharias S, Woods E, Karimi-Busheri F, et al. Expression of the epigenetic factor BORIS (CTCFL) in the human genome. J Transl Med. 2011;9:213.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Rasko JE, Klenova EM, Leon J, Filippova GN, Loukinov DI, Vatolin S, et al. Cell growth inhibition by the multifunctional multivalent zinc-finger factor CTCF. Cancer Res. 2001;61(16):6002–7.

    CAS  PubMed  Google Scholar 

  63. Dougherty CJ, Ichim TE, Liu L, Reznik G, Min WP, Ghochikyan A, et al. Selective apoptosis of breast cancer cells by siRNA targeting of BORIS. Biochem Biophys Res Commun. 2008;370(1):109–12.

    Article  CAS  PubMed  Google Scholar 

  64. Kosaka-Suzuki N, Suzuki T, Pugacheva EM, Vostrov AA, Morse HC 3rd, Loukinov D, et al. Transcription factor BORIS (brother of the regulator of imprinted sites) directly induces expression of a cancer-testis antigen, TSP50, through regulated binding of BORIS to the promoter. J Biol Chem. 2011;286(31):27378–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Recillas-Targa F, De La Rosa-Velazquez IA, Soto-Reyes E, Benitez-Bribiesca L. Epigenetic boundaries of tumour suppressor gene promoters: the CTCF connection and its role in carcinogenesis. J Cell Mol Med. 2006;10(3):554–68.

    Article  CAS  PubMed  Google Scholar 

  66. Bhan S, Negi SS, Shao C, Glazer CA, Chuang A, Gaykalova DA, et al. BORIS binding to the promoters of cancer testis antigens, MAGEA2, MAGEA3, and MAGEA4, is associated with their transcriptional activation in lung cancer. Clin Cancer Res. 2011;17(13):4267–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Makovski A, Yaffe E, Shpungin S, Nir U. Intronic promoter drives the BORIS-regulated expression of FerT in colon carcinoma cells. J Biol Chem. 2012;287(9):6100–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Nguyen P, Bar-Sela G, Sun L, Bisht KS, Cui H, Kohn E, et al. BAT3 and SET1A form a complex with CTCFL/BORIS to modulate H3K4 histone dimethylation and gene expression. Mol Cell Biol. 2008;28(21):6720–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Glazer CA, Smith IM, Ochs MF, Begum S, Westra W, Chang SS, et al. Integrative discovery of epigenetically derepressed cancer testis antigens in NSCLC. PLoS ONE. 2009;4(12):e8189.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Yasumoto S, Kunimura C, Kikuchi K, Tahara H, Ohji H, Yamamoto H, et al. Telomerase activity in normal human epithelial cells. Oncogene. 1996;13(2):433–9.

    CAS  PubMed  Google Scholar 

  71. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266(5193):2011–5.

    Article  CAS  PubMed  Google Scholar 

  72. Shay JW, Wright WE, editors. Role of telomeres and telomerase in cancer. Seminars in cancer biology. Amsterdam: Elsevier; 2011.

  73. Guilleret I, Benhattar J. Demethylation of the human telomerase catalytic subunit (hTERT) gene promoter reduced hTERT expression and telomerase activity and shortened telomeres. Exp Cell Res. 2003;289(2):326–34.

    Article  CAS  PubMed  Google Scholar 

  74. Renaud S, Bosman FT, Benhattar J. Implication of the exon region in the regulation of the human telomerase reverse transcriptase gene promoter. Biochem Biophys Res Commun. 2003;300(1):47–54.

    Article  CAS  PubMed  Google Scholar 

  75. Meeran SM, Patel SN, Tollefsbol TO. Sulforaphane causes epigenetic repression of hTERT expression in human breast cancer cell lines. PLoS ONE. 2010;5(7):e11457.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Guilleret I, Yan P, Grange F, Braunschweig R, Bosman FT, Benhattar J. Hypermethylation of the human telomerase catalytic subunit (hTERT) gene correlates with telomerase activity. Int J Cancer. 2002;101(4):335–41.

    Article  CAS  PubMed  Google Scholar 

  77. Zendman AJ, Ruiter DJ, Van Muijen GN. Cancer/testis-associated genes: identification, expression profile, and putative function. J Cell Physiol. 2003;194(3):272–88.

    Article  CAS  PubMed  Google Scholar 

  78. Woloszynska-Read A, James SR, Song C, Jin B, Odunsi K, Karpf AR. BORIS/CTCFL expression is insufficient for cancer-germline antigen gene expression and DNA hypomethylation in ovarian cell lines. Cancer Immun. 2010;10:6.

    PubMed  PubMed Central  Google Scholar 

  79. Gaykalova D, Vatapalli R, Glazer CA, Bhan S, Shao C, Sidransky D, et al. Dose-dependent activation of putative oncogene SBSN by BORIS. PLoS ONE. 2012;7(7):e40389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Yawata T, Nakai E, Park KC, Chihara T, Kumazawa A, Toyonaga S, et al. Enhanced expression of cancer testis antigen genes in glioma stem cells. Mol Carcinog. 2010;49(6):532–44.

    Article  CAS  PubMed  Google Scholar 

  81. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11.

    Article  CAS  PubMed  Google Scholar 

  82. Sell S. Stem cell origin of cancer and differentiation therapy. Crit Rev Oncol Hematol. 2004;51(1):1–28.

    Article  PubMed  Google Scholar 

  83. Soltanian S, Matin MM. Cancer stem cells and cancer therapy. Tumour Biol. 2011;32(3):425–40.

    Article  PubMed  Google Scholar 

  84. Clarke MF, Fuller M. Stem cells and cancer: two faces of eve. Cell. 2006;124(6):1111–5.

    Article  CAS  PubMed  Google Scholar 

  85. Santagata S, Ligon KL, Hornick JL. Embryonic stem cell transcription factor signatures in the diagnosis of primary and metastatic germ cell tumors. Am J Surg Pathol. 2007;31(6):836–45.

    Article  PubMed  Google Scholar 

  86. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.

    Article  CAS  PubMed  Google Scholar 

  87. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  CAS  PubMed  Google Scholar 

  88. Schoenhals M, Kassambara A, De Vos J, Hose D, Moreaux J, Klein B. Embryonic stem cell markers expression in cancers. Biochem Biophys Res Commun. 2009;383(2):157–62.

    Article  CAS  PubMed  Google Scholar 

  89. Monk M, Holding C. Human embryonic genes re-expressed in cancer cells. Oncogene. 2001;20(56):8085–91.

    Article  CAS  PubMed  Google Scholar 

  90. Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, van Roozendaal KE, et al. POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res. 2003;63(9):2244–50.

    CAS  PubMed  Google Scholar 

  91. Li XL, Eishi Y, Bai YQ, Sakai H, Akiyama Y, Tani M, et al. Expression of the SRY-related HMG box protein SOX2 in human gastric carcinoma. Int J Oncol. 2004;24(2):257–63.

    CAS  PubMed  Google Scholar 

  92. Tsukamoto T, Mizoshita T, Mihara M, Tanaka H, Takenaka Y, Yamamura Y, et al. Sox2 expression in human stomach adenocarcinomas with gastric and gastric-and-intestinal-mixed phenotypes. Histopathology. 2005;46(6):649–58.

    Article  CAS  PubMed  Google Scholar 

  93. Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, Chang HY. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell. 2008;2(4):333–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kang J, Shakya A, Tantin D. Stem cells, stress, metabolism and cancer: a drama in two Octs. Trends Biochem Sci. 2009;34(10):491–9.

    Article  CAS  PubMed  Google Scholar 

  95. Cole MD, Henriksson M. 25 years of the c-Myc oncogene. Semin Cancer Biol. 2006;16(4):241.

    Article  PubMed  Google Scholar 

  96. Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40(5):499–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Nagata S, Hirano K, Kanemori M, Sun LT, Tada T. Self-renewal and pluripotency acquired through somatic reprogramming to human cancer stem cells. PLoS ONE. 2012;7(11):e48699.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med. 2006;355(12):1253–61.

    Article  CAS  PubMed  Google Scholar 

  99. Schatton T, Frank NY, Frank MH. Identification and targeting of cancer stem cells. BioEssays. 2009;31(10):1038–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, Taguchi T, et al. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res. 2009;15(12):4234–41.

    Article  CAS  PubMed  Google Scholar 

  101. Dean M. ABC transporters, drug resistance, and cancer stem cells. J Mammary Gland Biol Neoplasia. 2009;14(1):3–9.

    Article  PubMed  Google Scholar 

  102. Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P, et al. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Can Res. 2009;69(4):1302–13.

    Article  CAS  Google Scholar 

  103. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60.

    Article  CAS  PubMed  Google Scholar 

  104. Duong H-Q, Hwang JS, Kim HJ, Kang HJ, Seong Y-S, Bae I. Aldehyde dehydrogenase 1A1 confers intrinsic and acquired resistance to gemcitabine in human pancreatic adenocarcinoma MIA PaCa-2 cells. Int J Oncol. 2012;41(3):855–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Thomas M, Coyle K, Sultan M, Vaghar-Kashani A, Marcato PP. Chemoresistance in cancer stem cells and strategies to overcome resistance. Chemotherapy. 2014;3(125):2.

    Google Scholar 

  107. Teng Y, Wang X, Wang Y, Ma D. Wnt/β-catenin signaling regulates cancer stem cells in lung cancer A549 cells. Biochem Biophys Res Commun. 2010;392(3):373–9.

    Article  CAS  PubMed  Google Scholar 

  108. Vermeulen L, Felipe De Sousa EM, Van Der Heijden M, Cameron K, De Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010;12(5):468–76.

    Article  CAS  PubMed  Google Scholar 

  109. Cai C, Zhu X. The Wnt/β-catenin pathway regulates self-renewal of cancer stem-like cells in human gastric cancer. Mol Med Rep. 2012;5(5):1191–6.

    CAS  PubMed  Google Scholar 

  110. Weng AP, Ferrando AA, Lee W, Morris JP, Silverman LB, Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306(5694):269–71.

    Article  CAS  PubMed  Google Scholar 

  111. Liu S, Dontu G, Mantle ID, Patel S, Ahn N-S, Jackson KW, et al. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006;66(12):6063–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hu Y, Fu L. Targeting cancer stem cells: a new therapy to cure cancer patients. Am J Cancer Res. 2012;2(3):340.

    PubMed  PubMed Central  Google Scholar 

  113. Chen K, Huang Y-H, Chen J-L. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin. 2013;34(6):732.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol. 2013;15(4):338.

    Article  CAS  PubMed  Google Scholar 

  115. Moghbeli M, Moghbeli F, Forghanifard MM, Abbaszadegan MR. Cancer stem cell detection and isolation. Med Oncol. 2014;31(9):69.

    Article  PubMed  CAS  Google Scholar 

  116. Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8(5):486–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Salerno M, Avnet S, Bonuccelli G, Eramo A, De Maria R, Gambarotti M, et al. Sphere-forming cell subsets with cancer stem cell properties in human musculoskeletal sarcomas. Int J Oncol. 2013;43(1):95–102.

    Article  PubMed  Google Scholar 

  118. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401.

    Article  CAS  PubMed  Google Scholar 

  119. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445(7123):111–5.

    Article  CAS  PubMed  Google Scholar 

  120. Hirschmann-Jax C, Foster A, Wulf G, Nuchtern J, Jax T, Gobel U, et al. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci USA. 2004;101(39):14228–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Chiba T, Kita K, Zheng YW, Yokosuka O, Saisho H, Iwama A, et al. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology. 2006;44(1):240–51.

    Article  CAS  PubMed  Google Scholar 

  122. Kondo T, Setoguchi T, Taga T. Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci USA. 2004;101(3):781–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Liu Q, Chen K, Liu Z, Huang Y, Zhao R, Wei L, et al. BORIS up-regulates OCT4 via histone methylation to promote cancer stem cell-like properties in human liver cancer cells. Cancer Lett. 2017;403:165–74.

    Article  CAS  PubMed  Google Scholar 

  124. Vicente-Dueñas C, Barajas-Diego M, Romero-Camarero I, González-Herrero I, Flores T, Sánchez-García I. Essential role for telomerase in chronic myeloid leukemia induced by BCR-ABL in mice. Oncotarget. 2012;3(3):261.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Marian CO, Wright WE, Shay JW. The effects of telomerase inhibition on prostate tumor-initiating cells. Int J Cancer. 2010;127(2):321–31.

    CAS  PubMed  Google Scholar 

  126. Marian CO, Cho SK, Mcellin BM, Maher EA, Hatanpaa KJ, Madden CJ, et al. The telomerase antagonist, imetelstat, efficiently targets glioblastoma tumor-initiating cells leading to decreased proliferation and tumor growth. Clin Cancer Res. 2010;16(1):154–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Babaie Y, Herwig R, Greber B, Brink TC, Wruck W, Groth D, et al. Analysis of Oct4-dependent transcriptional networks regulating self-renewal and pluripotency in human embryonic stem cells. Stem Cells. 2007;25(2):500–10.

    Article  CAS  PubMed  Google Scholar 

  128. Murakami S, Ninomiya W, Sakamoto E, Shibata T, Akiyama H, Tashiro F. SRY and OCT4 are required for the acquisition of cancer stem cell-like properties and are potential differentiation therapy targets. Stem Cells. 2015;33(9):2652–63.

    Article  CAS  PubMed  Google Scholar 

  129. Kumar SM, Liu S, Lu H, Zhang H, Zhang PJ, Gimotty PA, et al. Acquired cancer stem cell phenotypes through Oct4-mediated dedifferentiation. Oncogene. 2012;31(47):4898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wang XQ, Ongkeko WM, Chen L, Yang ZF, Lu P, Chen KK, et al. Octamer 4 (Oct4) mediates chemotherapeutic drug resistance in liver cancer cells through a potential Oct4–AKT–ATP-binding cassette G2 pathway. Hepatology. 2010;52(2):528–39.

    Article  CAS  PubMed  Google Scholar 

  131. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci. 2003;100(7):3983–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Damaschke NA, Yang B, Blute ML, Lin CP, Huang W, Jarrard DF. Frequent disruption of chromodomain helicase DNA-binding protein 8 (CHD8) and functionally associated chromatin regulators in prostate cancer. Neoplasia. 2014;16(12):1018–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Joosse S, Müller V, Steinbach B, Pantel K, Schwarzenbach H. Circulating cell-free cancer-testis MAGE-A RNA, BORIS RNA, let-7b and miR-202 in the blood of patients with breast cancer and benign breast diseases. Br J Cancer. 2014;111(5):909–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Gonzalez DM, Medici D. Signaling mechanisms of the epithelial–mesenchymal transition. Sci Signal. 2014;7(344):re8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Jang G-B, Kim J-Y, Cho S-D, Park K-S, Jung J-Y, Lee H-Y, et al. Blockade of Wnt/β-catenin signaling suppresses breast cancer metastasis by inhibiting CSC-like phenotype. Sci Rep. 2015;5:12465.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Malanchi I, Peinado H, Kassen D, Hussenet T, Metzger D, Chambon P, et al. Cutaneous cancer stem cell maintenance is dependent on β-catenin signalling. Nature. 2008;452(7187):650–3.

    Article  CAS  PubMed  Google Scholar 

  137. Zeng YA, Nusse R. Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell. 2010;6(6):568–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Cai W-Y, Wei T-Z, Luo Q-C, Wu Q-W, Liu Q-F, Yang M, et al. The Wnt–β-catenin pathway represses let-7 microRNA expression through transactivation of Lin28 to augment breast cancer stem cell expansion. J Cell Sci. 2013;126(13):2877–89.

    Article  CAS  PubMed  Google Scholar 

  139. Dodge ME, Lum L. Drugging the cancer stem cell compartment: lessons learned from the hedgehog and Wnt signal transduction pathways. Annu Rev Pharmacol Toxicol. 2011;51:289–310.

    Article  CAS  PubMed  Google Scholar 

  140. Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, et al. EMT and tumor metastasis. Clin Transl Med. 2015;4(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Mani SA, Guo W, Liao M-J, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Es-haghi M, Soltanian S, Dehghani H. Perspective: cooperation of Nanog, NF-κΒ, and CXCR4 in a regulatory network for directed migration of cancer stem cells. Tumor Biol. 2016;37(2):1559–65.

    Article  CAS  Google Scholar 

  143. Bartkowiak K, Wieczorek M, Buck F, Harder SN, Moldenhauer J, Effenberger KE, et al. Two-dimensional differential gel electrophoresis of a cell line derived from a breast cancer micrometastasis revealed a stem/progenitor cell protein profile. J Proteome Res. 2009;8(4):2004–14.

    Article  CAS  PubMed  Google Scholar 

  144. Willipinski-Stapelfeldt B, Riethdorf S, Assmann V, Woelfle U, Rau T, Sauter G, et al. Changes in cytoskeletal protein composition indicative of an epithelial–mesenchymal transition in human micrometastatic and primary breast carcinoma cells. Clin Cancer Res. 2005;11(22):8006–14.

    Article  CAS  PubMed  Google Scholar 

  145. Zampieri M, Ciccarone F, Palermo R, Cialfi S, Passananti C, Chiaretti S, et al. The epigenetic factor BORIS/CTCFL regulates the NOTCH3 gene expression in cancer cells. Biochim Biophys Acta. 2014;1839(9):813–25.

    Article  CAS  PubMed  Google Scholar 

  146. Fan X, Khaki L, Zhu TS, Soules ME, Talsma CE, Gul N, et al. NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells. 2010;28(1):5–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Gerby B, Clappier E, Armstrong F, Deswarte C, Calvo J, Poglio S, et al. Expression of CD34 and CD7 on human T-cell acute lymphoblastic leukemia discriminates functionally heterogeneous cell populations. Leukemia. 2011;25(8):1249–58.

    Article  CAS  PubMed  Google Scholar 

  148. Harrison H, Farnie G, Howell SJ, Rock RE, Stylianou S, Brennan KR, et al. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Can Res. 2010;70(2):709–18.

    Article  CAS  Google Scholar 

  149. Sansone P, Storci G, Giovannini C, Pandolfi S, Pianetti S, Taffurelli M, et al. p66Shc/Notch-3 interplay controls self-renewal and hypoxia survival in human stem/progenitor cells of the mammary gland expanded in vitro as mammospheres. Stem Cells. 2007;25(3):807–15.

    Article  CAS  PubMed  Google Scholar 

  150. Schwarzenbach H, Eichelser C, Steinbach B, Tadewaldt J, Pantel K, Lobanenkov V, et al. Differential regulation of MAGE-A1 promoter activity by BORIS and Sp1, both interacting with the TATA binding protein. BMC Cancer. 2014;14(1):796.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Lee J-H, Cook JR, Yang Z-H, Mirochnitchenko O, Gunderson SI, Felix AM, et al. PRMT7, a new protein arginine methyltransferase that synthesizes symmetric dimethylarginine. J Biol Chem. 2005;280(5):3656–64.

    Article  CAS  PubMed  Google Scholar 

  152. Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293(5532):1074–80.

    Article  CAS  PubMed  Google Scholar 

  153. Sun L, Huang L, Nguyen P, Bisht KS, Bar-Sela G, Ho AS, et al. DNA methyltransferase 1 and 3B activate BAG-1 expression via recruitment of CTCFL/BORIS and modulation of promoter histone methylation. Cancer Res. 2008;68(8):2726–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Suzuki T, Kimura A, Nagai R, Horikoshi M. Regulation of interaction of the acetyltransferase region of p300 and the DNA-binding domain of Sp1 on and through DNA binding. Genes Cells. 2000;5(1):29–41.

    Article  CAS  PubMed  Google Scholar 

  155. Safe S, Abdelrahim M. Sp transcription factor family and its role in cancer. Eur J Cancer. 2005;41(16):2438–48.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

SS and HD wrote the review. Both authors read and approved the final manuscript.

Acknowledgements

The authors would like to thank all the members of their research teams who have contributed to findings on cancer and germ cells in the past 5 years.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Funding/support

The research in the laboratory of HD is funded by Ferdowsi University of Mashhad, Iran National Science Foundation, Council for Stem Cell Sciences and Technologies, and National Institute for Medical Research Development of Iran. The research in the laboratory of SS is funded by Shahid Bahonar University of Kerman.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hesam Dehghani.

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Soltanian, S., Dehghani, H. BORIS: a key regulator of cancer stemness. Cancer Cell Int 18, 154 (2018). https://doi.org/10.1186/s12935-018-0650-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-018-0650-8

Keywords