Open Access

Regulation of gene transcription of B lymphoma Mo‑MLV insertion region 1 homolog (Review)

  • Authors:
    • Meizhen Zhou
    • Qichao Xu
    • Deqiang Huang
    • Lingyu Luo
  • View Affiliations

  • Published online on: April 5, 2021     https://doi.org/10.3892/br.2021.1428
  • Article Number: 52
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

B lymphoma Mo‑MLV insertion region 1 homolog (Bmi‑1) is a core protein component of the polycomb repressive complex 1 that inhibits cell senescence and maintains the self‑renewal ability of stem cells via downregulation of p16Ink4a and p19Arf expression. Bmi‑1 serves an important role in hematopoietic stem cell maintenance and neurodevelopment during embryonic development, and it has been shown to enhance tumorigenesis by promoting cancer stem cell self‑renewal and epithelial to mesenchymal transition. Emerging evidence suggests that Bmi‑1 overexpression is closely related to the development and progression of various types of cancer, and that downregulation of Bmi‑1 expression can inhibit the proliferation, invasion and metastasis of cancer cells. It is therefore important to elucidate the mechanisms underlying the regulation of Bmi‑1 expression both under normal growth conditions and in malignant tissues. In the present review, the current body of knowledge pertaining to the transcriptional and post‑transcriptional regulation of the BMI‑1 gene is discussed, and the potential mechanisms by which Bmi‑1 is dysregulated in various types of cancer are highlighted. Bmi‑1 expression is primarily controlled via transcriptional regulation, and is regulated by the transcription https://www.ushuaia.pl/hyphen/?ln=en factors of the Myc family, including Myb, Twist1, SALL4 and E2F‑1. Post‑transcriptionally, regulation of Bmi‑1 expression is inhibited by several microRNAs and certain small‑molecule drugs. Thus, regulatory transcriptional factors are potential therapeutic targets to reduce Bmi‑1 expression in cancer cells. Thus, the present review provides an up‑to‑date review on the regulation of BMI‑1 gene expression at the transcriptional and post‑transcriptional level.

1. Introduction

The polycomb group (PcG) is a class of highly conserved transcriptional repressors. They are divided into two core protein complexes: Polycomb repressive complex (PRC)1 and PRC2. Both PRC1 and PRC2 serve an important role in the maintenance of the inhibition state of chromatins by polycomb proteins. PRC2 binds to the target gene during the initial stage of transcription and recruits the PRC1 complex to bind to the target gene, which maintains the repressed state of the gene (1).

The PRC1 complex core proteins include RING1B (also referred to as RNF2), RING1A (also referred to as RING1), B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), EDR1 (also referred to as HPH1) and CBX4 (also referred to as HPC2). Among these components, Bmi-1 serves a pivotal role in the PRC1 complex (1). The binding of Bmi-1 to chromatin along with other PcG proteins of the PRC1 complex leads to histone H3K27 methylation, which results in continued silencing of the Ink4a/Arf locus. Decreased levels of p16Ink4a and p19Arf lead to activation of nuclear E2F and downregulation of p53(2), which in turn promotes cell proliferation and self-renewal of cancer stem cells (Fig. 1). In addition, Bmi-1 inhibits the expression of E-cadherin by interacting with epithelial to mesenchymal transition (EMT) regulatory molecules, such as Twist1, Wnt, Snail and β-catenin to induce EMT, whereas inhibition of Bmi-1 expression leads to EMT reversal and decreased cell migratory ability (3). BMI-1 is a highly conserved gene with rare mutations. It serves as a central node of various oncogenes and plays an important role in cell proliferation and tumorigenesis. Multiple signaling pathways, including N-Myc (MYCN), c-Myc, (MYC) (4), twist (3), Akt (5), and MAPK upregulate BMI-1 expression. Under normal growth conditions, BMI-1 expression is maintained within physiological levels through a feed-back loop that involves the PcG family members, PRC1 and PRC2(6). However, BMI-1 expression is upregulated in malignant cells, partly due to stimulation by oncogenes, such as E2F-1 and c-MYC, and this allows for the maintenance of an undifferentiated state of the cells. BMI-1 overexpression is a biomarker of malignant tumors and is closely related to tumor malignancy, invasion, metastasis and prognosis (7). Therefore, inhibition of BMI-1 expression, restoration of p16Ink4a and p19Arf levels, and induction of cellular senescence are novel potential therapeutic targets for anti-cancer targeted therapy (8).

Bmi-1 is a short-lived protein, and its expression levels are controlled by various mechanisms. Bmi-1 expression is primarily controlled by transcriptional and post-transcriptional regulation (1,8). Transcriptional regulation of eukaryotic genes involves DNA methylation, histone modification, chromatin remodeling and transcription factors. Post-transcriptional regulation is predominantly achieved through regulation of RNA, which includes RNA processing and maturation, RNA transport and subcellular localization, mRNA translation and mRNA degradation (9). In this review, the recent advances in the understanding of transcriptional (Table I) and post-transcriptional regulation (Table II) of BMI-1 expression are summarized.

Table I

Factors that regulate BMI-1 expression at the transcriptional level.

Table I

Factors that regulate BMI-1 expression at the transcriptional level.

FactorsFunction(Refs.)
c-MycIncreases Bmi-1 expression by binding to the E-box sequence in the BMI-1 promoter(18,19)
N-MycIncreases Bmi-1 expression by binding to the E-box sequence in the BMI-1 promoter(22,23)
c-MybIncreases Bmi-1 expression by binding 5'-flanking region nucleotides +3 to +8(31)
Twist1Increases Bmi-1 expression by binding the E-box sequence in intron 1 of BMI-1 gene(3)
SALL4Binds to the -450 to -1 region of the BMI-1 promoter and increases Bmi-1 transcription(36)
E2F-1Combines with the E2F-1 binding site in BMI-1 promoter to increase BMI-1 transcriptional activity(38,40)
Hedgehog signalHedgehog downstream protein GLI1 binds to BMI-1 promoter and increases Bmi-1 transcription(44)
FoxM1cPromotes Bmi-1 expression by increasing c-Myc expression(28)
Estrogen receptor αInteracts with the BMI-1 promoter upstream element at -327 to -172 bp to activate BMI-1 transcription(45)
Sp1Binds to the region from +181 to +214 within the BMI-1 promoter and increases BMI-1 transcriptional activity(46)
Nrf2Promotes BMI-1 transcription via an unknown mechanism(49)
Id1Promotes Bmi-1 expression by increasing c-Myc expression(49)
Mel-18Suppresses Bmi-1 expression via inhibition of c-Myc expression(20,26)
HDACiIndirectly inhibits BMI-1 promoter activity(50)
KLF4Binds to the BMI-1 promoter sequence, -233 to 0, to suppress BMI-1 transcription(47)
Copper sulfateInhibits Bmi-1 expression via an unknown mechanism(70)

[i] Bmi-1, B lymphoma Mo-MLV insertion region 1 homolog; E2F, E2 promoter binding factor; SALL4, Sal-like protein 4; Id1, inhibitor of differentiation and DNA binding; HDACi, histone deacetylase inhibitor; KLF4, Krüppel-like factor 4.

Table II

Factors that regulate BMI-1 expression at the post-transcriptional level.

Table II

Factors that regulate BMI-1 expression at the post-transcriptional level.

FactorsCell or tissue types(Refs.)
miR-15aGastric tumor tissues(58)
 Prostate tumor tissues(71)
miR-16Ovarian cancer tissues(53,66)
miR-30dProstate cancer(61)
miR-30e*Tumor-associated macrophages in gastrointestinal cancer(61)
miR-34aBrain tumor; breast cancer(54,72)
miR-128Brain tumor(54)
miR-135aPancreatic ductal adenocarcinoma(56)
miR-141Human diploid fibroblasts(60)
miR-183Gastric cancer(57)
miR-194Endometrial cancer cell lines(59)
miR-200bProstate cancer(62)
miR-200cMelanoma(65)
miR-203Leukemia stem cells(63)
miR-218Colon cancer(55)
miR-221prostate cancer(71)
miR-302MCF7, HepG2 Cell lines(73)
miR-320aNasopharyngeal carcinoma.(60)
miR-452Colorectal cancer; glioma(74,75)
miR-495Mesenchymal stem cells(76)
NVP-LDE-225Prostate cancer(32)
PTC-209Colorectal cancer(68)

[i] miR, microRNA.

2. Bmi-1 protein structure

The BMI-1 gene was identified as a common provirus-binding site during induction of B cell lymphoma using Moroni's murine leukemia virus in transgenic mice (10). The human BMI-1 gene is very similar to its mouse homolog and is located in the short arm 13 region of chromosome 10 (10p13). The cDNA length of human BMI-1 gene is 3,203 bp with 86% similarity with the mouse gene sequence. The human BMI-1 gene consists of 10 exons, which encode a protein containing 326 amino acids with 98% amino acid sequence homology with mouse Bmi-1(11). Bmi-1 protein structure is divided into the amino terminus, the central region and the carboxyl terminus (Fig. 2). The amino terminus has a Ring Finger domain consisting of a cysteine-rich zinc finger motif and C3HC4(12). The central region is a conserved helix-turn-helix-turn-helix-turn (HTHTHT) domain; the carboxyl terminus is considered a peptide sequence that is rich in proline (P), glutamic acid (E), serine (S), and threonine (T) (PEST) sequence. The nuclear localization signal sequence (NLS)1 (KRRR) and NLS2 (KRMK) are both located between the three regions (13). Each region has a different function, and the function of the loop finger domain is to bind to DNA and exert transcriptional regulation (14), which is also required for the activation of E3 ubiquitin ligase activity (15). The HTHTHT domain is the key structure involved in the gene transcriptional repressor function of Bmi-1(14); the NLS consists of a short-chain amino acids that is involved in the nuclear localization of the Bmi-1 protein. The carboxy-terminal PEST region is enriched with proline, glutamic acid, serine and threonine, and is involved in the rapid intracellular degradation of Bmi-1(16).

3. Transcriptional regulation of the BMI-1 gene

Transcriptional regulation is the primary mechanism by which BMI-1 expression is controlled. The transcription factors that regulate BMI-1 include members of the Myc family, Myb, Twist1, SALL4 and E2F-1 (Fig. 3).

Myc family

The Myc family is a group of important oncogenes, including MYC, MYCN, L-Myc, S-Myc and B-Myc. Of these, MYC and MYCN are involved in BMI-1 transcription (4). MYC plays an important role in cell proliferation, differentiation and apoptosis, and is abnormally expressed in several types of cancer (17). In murine lymphoma and human malignant glioma, both MYC and BMI-1 are highly expressed, and exhibit a positive correlation with each other (18,19). The MYC protein is a transcription factor of the basic helix-loop-helix leucine zipper family. MYC forms a functional DNA-binding complex with Max, another member of the same family, and this complex specifically recognizes the E-box sequence (CACGTG) of the BMI-1 gene promoter, thereby increasing the expression of BMI-1 at the transcriptional level (4,19,20).

Another member of the Myc family, MYCN, is frequently upregulated in human neuroblastoma (21). MYCN protein has been shown to be associated with Bmi-1 expression in orthotopic neuroblastoma cell lines and tumor samples. In addition, MYCN protein expression was shown to increase the transcriptional activity of BMI-1 gene by binding to the BMI-1 promoter E-box region (22). Overexpression of MYCN promotes tumorigenesis and proliferation of neuroblastoma cells by directly targeting and upregulating BMI-1 (23).

Other factors regulate BMI-1 indirectly through the Myc gene family. Mel-18 protein is a member of the PcG family that possesses close structural similarities with the Bmi-1 protein (24); however, functionally, it cannot replace the role of Bmi-1 in PRC1(6). Mel-18 inhibits transcriptional expression of c-Myc and prevents c-Myc from binding to the BMI-1 gene promoter (4,25), which indirectly inhibits BMI-1 transcription. Mel-18 is considered a tumor suppressor due to its ability to inhibit the transcription of the MYC and BMI-1 genes, thereby inhibiting the proliferation and inducing apoptosis of cancer cells (20). Induction of overexpression of Mel-18 to downregulate the expression of BMI-1 gene was shown to attenuate the malignant attributes of breast cancer cells (26). Furthermore, transcription factor FoxM1c was shown to indirectly increase the expression of BMI-1 via c-Myc (27,28).

MYB

c-Myb is a member of the MYB transcription factor family. It promotes the expression of gut stem cell genes, particularly Lgr5, which affects the self-renewal capacity of intestinal and hematopoietic stem cells (29). In B-lymphocytic leukemia cells, c-Myb binds to the E-box element in the proximal regulatory region of the miR-155 host gene promoter, facilitating its transcription (30). Waldron et al (31) found that c-Myb binds directly to the +3 to +8 nucleotide sequence of the BMI-1 5' flanking region to increase BMI-1 transcription, and that c-Myb and Bmi-1 are required for human and mouse p190 BCR/ABL leukemogenesis.

Twist1

Twist1 belongs to the family of basic helix-loop-helix transcription factors. It promotes EMT by inhibiting E-cadherin expression (32). Mechanistically, Twist1 binds directly to the E-cadherin promoter to inhibit its expression, and it also directly binds to the E-box site of the -732 to -727 nucleotide sequence in intron 1 of the BMI-1 promoter to initiate the transcriptional upregulation of the BMI-1 gene (5). Twist1-mediated suppression of E-cadherin and upregulation of Bmi-1 leads to disruption of the tight junction between cells, thereby increasing tumor cell metastasis (33,35).

Sal-like protein 4 (SALL4)

SALL4 is a more recently identified zinc finger transcription factor that plays an important role in the maintenance of pluripotency of embryonic stem cells and the self-renewal capacity of hematopoietic stem cells (36). Significant upregulation of SALL4 and Bmi-1 expression has been reported in patients with myeloid leukemia (37). Additionally, high expression levels of these two genes were shown to be associated with the expansion of hematopoietic progenitor cells. This suggests that the expression of SALL4 and Bmi-1 is a prognostic biomarker of acute leukemia. Results of luciferase reporter assays by Yang et al (36) showed that the BMI-1 gene is a target of SALL4, and, increased expression of SALL4 was found to upregulate the activity of the BMI-1 promoter. Further analysis of the binding sites revealed that the SALL4 protein binds to a functional site in the -450 to -1 nucleotide sequence of the BMI-1 promoter to initiate transcription of the BMI-1 gene.

E2 promoter binding factor (E2F)-1

E2F-1 is a member of the E2F transcription factor family. E2F-1 is involved in cell cycle progression and regulates cell viability via both p53-independent and p53-dependent pathways (38). E2F-1 initiates transcription of the BMI-1 gene and upregulates BMI-1 expression by directly binding to the E2F binding site in the BMI-1 gene promoter; interestingly, when the cell is in the cell cycle or differentiation phase, BMI-1 is not regulated by E2F-1(39). In androgen-deficient prostate cancer cells, the IκB kinase α (IKKα)-E2F1-Bmi-1 cascade is activated. In these cells, activated IKKα phosphorylates E2F-1 to promote E2F-1 nuclear localization, whereby E2F-1 binds to the co-activator CREB binding protein (histone H3 acetyltransferase) and recruits the target genes, including BMI-1, thereby resulting in activation of BMI-1 (40).

Hedgehog signaling

Hedgehog signaling is a major regulator of vertebrate embryonic development, as it is involved in stem cell maintenance and cell differentiation and proliferation. Abnormal activation of the Hedgehog signaling pathway was shown to be associated with the development of lung, prostate and breast cancer (41). The primary components of the Hedgehog signaling pathway include patched, Smoothened and glioma-associated oncogene (GLI) (42). In a study by Liu et al (43), addition of sonic Hedgehog to activate the Hedgehog signaling pathway increased the expression of Bmi-1, whereas inhibition of the Hedgehog signaling pathway using cyclopamine resulted in downregulated expression of Bmi-1. Wang et al (44) found that GLI1 binds to the promoter of the BMI-1 gene, and that the BMI-1 transcription level changes in accordance with the increase or decrease in GLI1 expression.

Other factors

In addition to the above transcription factors, several other factors may regulate BMI-1. Estrogen receptor α activates the transcriptional activity of the BMI-1 gene by interacting with the -327 to -172 bp nucleotide sequence upstream of the BMI-1 promoter, thereby increasing BMI-1 gene expression (45). The transcription factor Sp1 binds to the +181 to +214 region of the BMI-1 promoter and increases BMI-1 gene expression (46). Krüppel-like factor 4 (KLF4) is a zinc finger protein that is normally expressed in the intestines and skin, and plays an important role in the regulation of stem cells. Yu et al (47) found that KLF4 binds directly to the -233 to 0 sequence of the BMI-1 gene promoter and inhibits the transcriptional activity of BMI-1, thereby reducing the expression of Bmi-1. The binding site of KLF4 to the BMI-1 gene is different from the binding site of c-Myc to the BMI-1 promoter. Redox sensing factor Nrf2 was shown to increase the expression of BMI-1 at the transcriptional level, thereby promoting the proliferation of cancer stem cells and inhibiting cancer cell apoptosis (48). The helix-loop-helix inhibitor of differentiation and DNA binding facilitates tumorigenesis by increasing the expression of Bmi-1 via c-Myc (49). Bommi et al (50) found that histone deacetylase inhibitors (HDACi) inhibit BMI-1 gene transcription in breast cancer cells via an indirect mechanism. In certain cancer cell lines, c-Myc is the target gene of HDACi, whereas in breast cancer cells, the inhibitory effect of HDACi on BMI-1 gene expression is not dependent on downregulation of c-Myc; however, the precise mechanism is not clear. Thus, there are various transcription factors that bind the promoter of BMI-1 and regulate BMI-1 gene expression at the transcriptional level.

4. Regulation of Bmi-1 at the post-transcriptional level

Post-transcriptional regulation primary involves the regulation of RNA and is divided into the following steps in chronological order: RNA processing and maturation, RNA transport and subcellular localization, mRNA translation and mRNA degradation. MicroRNAs (miRNAs) block gene expression primarily by preventing mRNA translation and/or promoting mRNA degradation (51). miRNAs are non-coding, short-stranded RNAs, which typically consist of 18-22 nucleotides. An miRNA complements the 3'-untranslated region (UTR) of its target mRNA and directs RNA-induced silencing complex to a specific region of the mRNA, thereby inhibiting mRNA translation or promoting mRNA degradation (8).

Bmi-1 expression is inhibited by several miRNAs, including miR-135a, miR-141, miR-183, miR-15a, miR-194, miR-203, miR-200b, miR-320a, miR-200c, miR-16, miR-495, miR-221, miR-30d, miR-128a, miR-34a, miR-452, miR-302 and miR-30e (51-67). For example, the expression of miR-218 in cancer tissues is lower than that in the paratumoral normal tissues, whereas the expression of Bmi-1 in cancer tissues is higher than that in the paratumoral normal tissues. An inverse correlation between Bmi-1 expression and miR-128 has been demonstrated in glioma and rectal cancer cells. Results of luciferase reporter assays showed that miR-128 inhibits Bmi-1 protein expression by binding to the 476 to 488 region of BMI-1 3'-UTR (54). several transcription factors and cytokines affect the expression levels of Bmi-1 by altering the expression of miRNAs. For example, Zeb1 was shown to downregulate Bmi-1 expression by inducing upregulation of the expressions of miR-203 and miR-183(32).

5. Conclusion and future prospects

The polycomb family protein member Bmi-1 acts as an oncogene and maintains the undifferentiated state of malignant tumor cells. Bmi-1 expression levels are closely related to the degree of malignancy, invasion and metastasis, and is a biomarker of adverse prognosis in cancer patients. As a pivotal node of multiple signaling pathways, Bmi-1 regulates the function of several downstream transcription factors and cytokines. Therefore, inhibition of Bmi-1 expression is a promising strategy for anticancer drug development. It has been shown that NVP-LDE-225 (Erismodegib) inhibits Bmi-1 expression by inducing upregulation of miR-128(68). In addition, PTC-209 is a small molecule drug that specifically inhibits Bmi-1 expression at the post-transcriptional level by binding to the 5' and 3' non-coding regions of BMI-1 mRNA (69). Transcriptional and post-transcriptional regulation are the primary means of regulation of Bmi-1 expression. Therefore, regulatory factors are potential therapeutic targets to reduce Bmi-1 expression in cancer cells.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by the National Nature Science Foundation of China (grant no. 82060450), Nature Science Foundation of Jiangxi province of China (grant nos. 20192BAB205072 and 20181BAB205050), and The Education Department of Jiangxi Province of China (grant no. 160032).

Availability of data and materials

Not applicable.

Authors' contributions

MZ and QX searched the literature, reviewed the articles and collected the relevant data from the selected papers. DH and LL wrote the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare no competing interests.

References

1 

Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, Morrison SJ and Clarke MF: Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 423:302–305. 2003.PubMed/NCBI View Article : Google Scholar

2 

Park IK, Morrison SJ and Clarke MF: Bmi1, stem cells, and senescence regulation. J Clin Invest. 113:175–179. 2004.PubMed/NCBI View Article : Google Scholar

3 

Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, Huang CH, Kao SY, Tzeng CH, Tai SK, et al: Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat Cell Biol. 12:982–992. 2010.PubMed/NCBI View Article : Google Scholar : Erratum in: Nat Cell Biol 21, 533, 2019.

4 

Adhikary S and Eilers M: Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol. 6:635–645. 2005.PubMed/NCBI View Article : Google Scholar

5 

Kim J, Hwangbo J and Wong PK: p38 MAPK-Mediated Bmi-1 down-regulation and defective proliferation in ATM-deficient neural stem cells can be restored by Akt activation. PLoS One. 6(e16615)2011.PubMed/NCBI View Article : Google Scholar

6 

Cao R, Tsukada Y and Zhang Y: Role of Bmi-1 and Ring1A in H2A ubiquitylation and Hox gene silencing. Mol Cell. 20:845–854. 2005.PubMed/NCBI View Article : Google Scholar

7 

Bhattacharyya J, Mihara K, Ohtsubo M, Yasunaga S, Takei Y, Yanagihara K, Sakai A, Hoshi M, Takihara Y and Kimura A: Overexpression of BMI-1 correlates with drug resistance in B-cell lymphoma cells through the stabilization of survivin expression. Cancer Sci. 103:34–41. 2012.PubMed/NCBI View Article : Google Scholar

8 

Cao L, Bombard J, Cintron K, Sheedy J, Weetall ML and Davis TW: BMI1 as a novel target for drug discovery in cancer. J Cell Biochem. 112:2729–2741. 2011.PubMed/NCBI View Article : Google Scholar

9 

Venkatesh S and Workman JL: Histone exchange, chromatin structure and the regulation of transcription. Nat Rev Mol Cell Biol. 16:178–189. 2015.PubMed/NCBI View Article : Google Scholar

10 

Siddique HR, Parray A, Tarapore RS, Wang L, Mukhtar H, Karnes RJ, Deng Y, Konety BR and Saleem M: BMI1 polycomb group protein acts as a master switch for growth and death of tumor cells: Regulates TCF4-transcriptional factor-induced BCL2 signaling. PLoS One. 8(e60664)2013.PubMed/NCBI View Article : Google Scholar

11 

Alkema MJ, Wiegant J, Raap AK, Berns A and van Lohuizen M: Characterization and chromosomal localization of the human proto-oncogene BMI-1. Hum Mol Genet. 2:1597–1603. 1993.PubMed/NCBI View Article : Google Scholar

12 

Freemont PS, Hanson IM and Trowsdale J: A novel cysteine-rich sequence motif. Cell. 64:483–484. 1991.PubMed/NCBI View Article : Google Scholar

13 

Dimri GP, Martinez JL, Jacobs JJL, Keblusek P, Itahana K, Van Lohuizen M, Campisi J, Wazer DE and Band V: The Bmi-1 oncogene induces telomerase activity and immortalizes human mammary epithelial cells. Cancer Res. 62:4736–4745. 2002.PubMed/NCBI

14 

Cohen KJ, Hanna JS, Prescott JE and Dang CV: Transformation by the Bmi-1 oncoprotein correlates with its subnuclear localization but not its transcriptional suppression activity. Mol Cell Biol. 16:5527–5535. 1996.PubMed/NCBI View Article : Google Scholar

15 

Buchwald G, van der Stoop P, Weichenrieder O, Perrakis A, van Lohuizen M and Sixma TK: Structure and E3-ligase activity of the Ring-Ring complex of polycomb proteins Bmi1 and Ring1b. EMBO J. 25:2465–2474. 2006.PubMed/NCBI View Article : Google Scholar

16 

Rogers S, Wells R and Rechsteiner M: Amino acid sequences common to rapidly degraded proteins: The PEST hypothesis. Science. 234:364–368. 1986.PubMed/NCBI View Article : Google Scholar

17 

Hoffman B and Liebermann DA: Apoptotic signaling by c-MYC. Oncogene. 27:6462–6472. 2008.PubMed/NCBI View Article : Google Scholar

18 

Jacobs JJL, Scheijen B, Voncken JW, Kieboom K, Berns A and van Lohuizen M: Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 13:2678–2690. 1999.PubMed/NCBI View Article : Google Scholar

19 

Cenci T, Martini M, Montano N, D'Alessandris QG, Falchetti ML, Annibali D, Savino M, Bianchi F, Pierconti F, Nasi S, et al: Prognostic relevance of c-Myc and BMI1 expression in patients with glioblastoma. Am J Clin Pathol. 138:390–396. 2012.PubMed/NCBI View Article : Google Scholar

20 

Guo WJ, Datta S, Band V and Dimri GP: Mel-18, a polycomb group protein, regulates cell proliferation and senescence via transcriptional repression of Bmi-1 and c-Myc oncoproteins. Mol Biol Cell. 18:536–546. 2007.PubMed/NCBI View Article : Google Scholar

21 

Weiss WA, Aldape K, Mohapatra G, Feuerstein BG and Bishop JM: Targeted expression of MYCN causes medulloblastoma in transgenic mice. EMBO J. 16:2985–2995. 1997.PubMed/NCBI View Article : Google Scholar

22 

Ochiai H, Takenobu H, Nakagawa A, Yamaguchi Y, Kimura M, Ohira M, Okimoto Y, Fujimura Y, Koseki H, Kohno Y, et al: Bmi1 is a MYCN target gene that regulates tumorigenesis through repression of KIF1Bβ and TSLC1 in neuroblastoma. Oncogene. 29:2681–2690. 2010.PubMed/NCBI View Article : Google Scholar

23 

Huang R, Cheung NKV, Vider J, Cheung IY, Gerald WL, Tickoo SK, Holland EC and Blasberg RG: MYCN and MYC regulate tumor proliferation and tumorigenesis directly through BMI1 in human neuroblastomas. FASEB J. 25:4138–4149. 2011.PubMed/NCBI View Article : Google Scholar

24 

Ishida A, Asano H, Hasegawa M, Koseki H, Ono T, Yoshida MC, Taniguchi M and Kanno M: Cloning and chromosome mapping of the human Mel-18 gene which encodes a DNA-binding protein with a new ‘RING-finger’ motif. Gene. 129:249–255. 1993.PubMed/NCBI View Article : Google Scholar

25 

Tetsu O, Ishihara H, Kanno R, Kamiyasu M, Inoue H, Tokuhisa T, Taniguchi M and Kanno M: mel-18 negatively regulates cell cycle progression upon B cell antigen receptor stimulation through a cascade leading to c-myc/cdc25. Immunity. 9:439–448. 1998.PubMed/NCBI View Article : Google Scholar

26 

Guo WJ, Zeng MS, Yadav A, Song LB, Guo BH, Band V and Dimri GP: Mel-18 acts as a tumor suppressor by repressing Bmi-1 expression and down-regulating Akt activity in breast cancer cells. Cancer Res. 67:5083–5089. 2007.PubMed/NCBI View Article : Google Scholar

27 

Liu-Bryan R and Terkeltaub R: Chondrocyte innate immune myeloid differentiation factor 88-dependent signaling drives procatabolic effects of the endogenous Toll-like receptor 2/Toll-like receptor 4 ligands low molecular weight hyaluronan and high mobility group box chromosomal protein 1 in mice. Arthritis Rheum. 62:2004–2012. 2010.PubMed/NCBI View Article : Google Scholar

28 

Li SKM, Smith DK, Leung WY, Cheung AM, Lam EW, Dimri GP and Yao KM: FoxM1c counteracts oxidative stress-induced senescence and stimulates Bmi-1 expression. J Biol Chem. 283:16545–16553. 2008.PubMed/NCBI View Article : Google Scholar

29 

Cheasley D, Pereira L, Lightowler S, Vincan E, Malaterre J and Ramsay RG: Myb controls intestinal stem cell genes and self-renewal. Stem Cells. 29:2042–2050. 2011.PubMed/NCBI View Article : Google Scholar

30 

Vargova K, Curik N, Burda P, Basova P, Kulvait V, Pospisil V, Savvulidi F, Kokavec J, Necas E, Berkova A, et al: MYB transcriptionally regulates the miR-155 host gene in chronic lymphocytic leukemia. Blood. 117:3816–3825. 2011.PubMed/NCBI View Article : Google Scholar

31 

Waldron T, De Dominici M, Soliera AR, Audia A, Iacobucci I, Lonetti A, Martinelli G, Zhang Y, Martinez R, Hyslop T, et al: c-Myb and its target Bmi1 are required for p190BCR/ABL leukemogenesis in mouse and human cells. Leukemia. 26:644–653. 2012.PubMed/NCBI View Article : Google Scholar

32 

Martin A and Cano A: Tumorigenesis: Twist1 links EMT to self-renewal. Nat Cell Biol. 12:924–925. 2010.PubMed/NCBI View Article : Google Scholar

33 

Yang MH, Hsu DSS, Wang HW, Wang HJ, Lan HY, Yang WH, Huang CH, Kao SY, Tzeng CH, Tai SK, et al: Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat Cell Biol. 12:982–992. 2010.PubMed/NCBI View Article : Google Scholar

34 

Wu KJ: Direct activation of Bmi1 by Twist1: Implications in cancer stemness, epithelial-mesenchymal transition, and clinical significance. Chang Gung Med J. 34:229–238. 2011.PubMed/NCBI

35 

Wu CY, Hung JJ and Wu KJ: Linkage between Twist1 and Bmi1: Molecular mechanism of cancer metastasis/stemness and clinical implications. Clin Exp Pharmacol Physiol. 39:668–673. 2012.PubMed/NCBI View Article : Google Scholar

36 

Yang J, Chai L, Liu F, Fink LM, Lin P, Silberstein LE, Amin HM, Ward DC and Ma Y: Bmi-1 is a target gene for SALL4 in hematopoietic and leukemic cells. Proc Natl Acad Sci USA. 104:10494–10499. 2007.PubMed/NCBI View Article : Google Scholar

37 

Vander Griend DJ, D'Antonio J, Gurel B, Antony L, Demarzo AM and Isaacs JT: Cell-autonomous intracellular androgen receptor signaling drives the growth of human prostate cancer initiating cells. Prostate. 70:90–99. 2010.PubMed/NCBI View Article : Google Scholar

38 

Irwin M, Marin MC, Phillips AC, Seelan RS, Smith DI, Liu W, Flores ER, Tsai KY, Jacks T, Vousden KH, et al: Role for the p53 homologue p73 in E2F-1-induced apoptosis. Nature. 407:645–648. 2000.PubMed/NCBI View Article : Google Scholar

39 

Nowak K, Kerl K, Fehr D, Kramps C, Gessner C, Killmer K, Samans B, Berwanger B, Christiansen H and Lutz W: BMI1 is a target gene of E2F-1 and is strongly expressed in primary neuroblastomas. Nucleic Acids Res. 34:1745–1754. 2006.PubMed/NCBI View Article : Google Scholar

40 

Ammirante M, Kuraishy AI, Shalapour S, Strasner A, Ramirez-Sanchez C, Zhang W, Shabaik A and Karin M: An IKKα-E2F1-BMI1 cascade activated by infiltrating B cells controls prostate regeneration and tumor recurrence. Genes Dev. 27:1435–1440. 2013.PubMed/NCBI View Article : Google Scholar

41 

Gupta S, Takebe N and Lorusso P: Targeting the Hedgehog pathway in cancer. Ther Adv Med Oncol. 2:237–250. 2010.PubMed/NCBI View Article : Google Scholar

42 

Briscoe J and Thérond PP: The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol. 14:416–429. 2013.PubMed/NCBI View Article : Google Scholar

43 

Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, Suri P and Wicha MS: Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 66:6063–6071. 2006.PubMed/NCBI View Article : Google Scholar

44 

Wang X, Venugopal C, Manoranjan B, McFarlane N, O'Farrell E, Nolte S, Gunnarsson T, Hollenberg R, Kwiecien J, Northcott P, et al: Sonic hedgehog regulates Bmi1 in human medulloblastoma brain tumor-initiating cells. Oncogene. 31:187–199. 2012.PubMed/NCBI View Article : Google Scholar

45 

Wang H, Liu H, Li X, Zhao J, Zhang H, Mao J, Zou Y, Zhang H, Zhang S, Hou W, et al: Estrogen receptor α-coupled Bmi1 regulation pathway in breast cancer and its clinical implications. BMC Cancer. 14(122)2014.PubMed/NCBI View Article : Google Scholar

46 

Wang HB, Liu GH, Zhang H, Xing S, Hu LJ, Zhao WF, Xie B, Li MZ, Zeng BH, Li Y, et al: Sp1 and c-Myc regulate transcription of BMI1 in nasopharyngeal carcinoma. FEBS J. 280:2929–2944. 2013.PubMed/NCBI View Article : Google Scholar

47 

Yu T, Chen X, Zhang W, Colon D, Shi J, Napier D, Rychahou P, Lu W, Lee EY, Weiss HL, et al: Regulation of the potential marker for intestinal cells, Bmi1, by β-catenin and the zinc finger protein KLF4: Implications for colon cancer. J Biol Chem. 287:3760–3768. 2012.PubMed/NCBI View Article : Google Scholar

48 

Jia Y, Chen J, Zhu H, Jia ZH and Cui MH: Aberrantly elevated redox sensing factor Nrf2 promotes cancer stem cell survival via enhanced transcriptional regulation of ABCG2 and Bcl-2/Bmi-1 genes. Oncol Rep. 34:2296–2304. 2015.PubMed/NCBI View Article : Google Scholar

49 

Qian T, Lee JY, Park JH, Kim HJ and Kong G: Id1 enhances RING1b E3 ubiquitin ligase activity through the Mel-18/Bmi-1 polycomb group complex. Oncogene. 29:5818–5827. 2010.PubMed/NCBI View Article : Google Scholar

50 

Bommi PV, Dimri M, Sahasrabuddhe AA, Khandekar J and Dimri GP: The polycomb group protein BMI1 is a transcriptional target of HDAC inhibitors. Cell Cycle. 9:2663–2673. 2010.PubMed/NCBI View Article : Google Scholar

51 

Slaby O, Svoboda M, Michalek J and Vyzula R: MicroRNAs in colorectal cancer: Translation of molecular biology into clinical application. Mol Cancer. 8(102)2009.PubMed/NCBI View Article : Google Scholar

52 

Bhattacharyya J, Mihara K, Yasunaga S, Tanaka H, Hoshi M, Takihara Y and Kimura A: BMI-1 expression is enhanced through transcriptional and posttranscriptional regulation during the progression of chronic myeloid leukemia. Ann Hematol. 88:333–340. 2009.PubMed/NCBI View Article : Google Scholar

53 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004.PubMed/NCBI View Article : Google Scholar

54 

Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G, Raychaudhury A, Newton HB, Chiocca EA and Lawler S: Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 68:9125–9130. 2008.PubMed/NCBI View Article : Google Scholar

55 

He X, Dong Y, Wu CW, Zhao Z, Ng SS, Chan FK, Sung JJ and Yu J: MicroRNA-218 inhibits cell cycle progression and promotes apoptosis in colon cancer by downregulating BMI1 polycomb ring finger oncogene. Mol Med. 18:1491–1498. 2013.PubMed/NCBI View Article : Google Scholar

56 

Dang Z, Xu WH, Lu P, Wu N, Liu J, Ruan B, Zhou L, Song WJ and Dou KF: MicroRNA-135a inhibits cell proliferation by targeting Bmi1 in pancreatic ductal adenocarcinoma. Int J Biol Sci. 10:733–745. 2014.PubMed/NCBI View Article : Google Scholar

57 

Xu L, Li Y, Yan D, He J and Liu D: MicroRNA-183 inhibits gastric cancer proliferation and invasion via directly targeting Bmi-1. Oncol Lett. 8:2345–2351. 2014.PubMed/NCBI View Article : Google Scholar

58 

Wu C, Zheng X, Li X, Fesler A, Hu W, Chen L, Xu B, Wang Q, Tong A, Burke S, et al: Reduction of gastric cancer proliferation and invasion by miR-15a mediated suppression of Bmi-1 translation. Oncotarget. 7:14522–14536. 2016.PubMed/NCBI View Article : Google Scholar

59 

Dong P, Kaneuchi M, Watari H, Hamada J, Sudo S, Ju J and Sakuragi N: MicroRNA-194 inhibits epithelial to mesenchymal transition of endometrial cancer cells by targeting oncogene BMI-1. Mol Cancer. 10(99)2011.PubMed/NCBI View Article : Google Scholar

60 

Dimri M, Carroll JD, Cho JH and Dimri GP: microRNA-141 regulates BMI1 expression and induces senescence in human diploid fibroblasts. Cell Cycle. 12:3537–3546. 2013.PubMed/NCBI View Article : Google Scholar

61 

Sugihara H, Ishimoto T, Watanabe M, Sawayama H, Iwatsuki M, Baba Y, Komohara Y, Takeya M and Baba H: Identification of miR-30e* regulation of Bmi1 expression mediated by tumor-associated macrophages in gastrointestinal cancer. PLoS One. 8(e81839)2013.PubMed/NCBI View Article : Google Scholar

62 

Yu J, Lu Y, Cui D, Li E, Zhu Y, Zhao Y, Zhao F and Xia S: miR-200b suppresses cell proliferation, migration and enhances chemosensitivity in prostate cancer by regulating Bmi-1. Oncol Rep. 31:910–918. 2014.PubMed/NCBI View Article : Google Scholar

63 

Zhang Y, Zhou SY, Yan HZ, Xu DD, Chen HX, Wang XY, Wang X, Liu YT, Zhang L, Wang S, et al: miR-203 inhibits proliferation and self-renewal of leukemia stem cells by targeting survivin and Bmi-1. Sci Rep. 6(19995)2016.PubMed/NCBI View Article : Google Scholar

64 

Qi X, Li J, Zhou C, Lv C and Tian M: MicroRNA-320a inhibits cell proliferation, migration and invasion by targeting BMI-1 in nasopharyngeal carcinoma. FEBS Lett. 588:3732–3738. 2014.PubMed/NCBI View Article : Google Scholar

65 

Liu S, Tetzlaff MT, Cui R and Xu X: miR-200c inhibits melanoma progression and drug resistance through down-regulation of BMI-1. Am J Pathol. 181:1823–1835. 2012.PubMed/NCBI View Article : Google Scholar

66 

Bhattacharya R, Nicoloso M, Arvizo R, Wang E, Cortez A, Rossi S, Calin GA and Mukherjee P: miR-15a and miR-16 control Bmi-1 expression in ovarian cancer. Cancer Res. 69:9090–9095. 2009.PubMed/NCBI View Article : Google Scholar

67 

Venkataraman S, Alimova I, Fan R, Harris P, Foreman N and Vibhakar R: MicroRNA 128a increases intracellular ROS level by targeting Bmi-1 and inhibits medulloblastoma cancer cell growth by promoting senescence. PLoS One. 5(e10748)2010.PubMed/NCBI View Article : Google Scholar

68 

Nanta R, Kumar D, Meeker D, Rodova M, Van Veldhuizen PJ, Shankar S and Srivastava RK: NVP-LDE-225 (Erismodegib) inhibits epithelial-mesenchymal transition and human prostate cancer stem cell growth in NOD/SCID IL2Rγ null mice by regulating Bmi-1 and microRNA-128. Oncogenesis. 2(e42)2013.PubMed/NCBI View Article : Google Scholar

69 

Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C, Davis T, Cao L, Baiazitov R, Du W, Sydorenko N, et al: Self-renewal as a therapeutic target in human colorectal cancer. Nat Med. 20:29–36. 2014.PubMed/NCBI View Article : Google Scholar

70 

Li Y, Hu J, Guan F, Song L, Fan R, Zhu H, Hu X, Shen E and Yang B: Copper induces cellular senescence in human glioblastoma multiforme cells through downregulation of Bmi-1. Oncol Rep. 29:1805–1810. 2013.PubMed/NCBI View Article : Google Scholar

71 

Xuan HQ, Xue W, Pan JH, Sha JJ, Dong BJ and Huang YR: Downregulation of miR-221,-30d, and-15a contributes to pathogenesis of prostate cancer by targeting Bmi-1. Biochemistry (Mosc). 80:276–283. 2015.PubMed/NCBI View Article : Google Scholar

72 

Guo JL, Li WP, Shi HL, Xie XH, Li LS, Tang HL, Wu MQ, Kong Y, Yang L, Gao J, et al: Synergistic effects of curcumin with emodin against the proliferation and invasion of breast cancer cells through upregulation of miR-34a. Mol Cell Biochem. 382:103–111. 2013.PubMed/NCBI View Article : Google Scholar

73 

Lin SL, Chang DC, Ying SY, Leu D and Wu DTS: MicroRNA miR-302 inhibits the tumorigenecity of human pluripotent stem cells by coordinate suppression of the CDK2 and CDK4/6 cell cycle pathways. Cancer Res. 70:9473–9482. 2010.PubMed/NCBI View Article : Google Scholar

74 

Li TT, Jian XY, He H, Lai QH, Li XZ, Deng DL, Liu TF, Zhu JH, Jiao HL, Ye YP, et al: MiR-452 promotes an aggressive colorectal cancer phenotype by regulating a Wnt/β-catenin positive feedback loop. J Exp Clin Cancer Res. 37(238)2018.PubMed/NCBI View Article : Google Scholar

75 

Liu LP, Chen K, Wu JH, Shi L, Hu B, Cheng SY, Li MF and Song LB: Downregulation of miR-452 promotes stem-like traits and tumorigenicity of gliomas. Clin Cancer Res. 19:3429–3438. 2013.PubMed/NCBI View Article : Google Scholar

76 

Li X, Song Y, Liu D, Zhao J, Xu J, Ren J, Hu Y, Wang Z, Hou Y and Zhao G: MiR-495 promotes senescence of mesenchymal stem cells by targeting Bmi-1. Cell Physiol Biochem. 42:780–796. 2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2021
Volume 14 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou M, Xu Q, Huang D and Luo L: Regulation of gene transcription of B lymphoma Mo‑MLV insertion region 1 homolog (Review). Biomed Rep 14: 52, 2021
APA
Zhou, M., Xu, Q., Huang, D., & Luo, L. (2021). Regulation of gene transcription of B lymphoma Mo‑MLV insertion region 1 homolog (Review). Biomedical Reports, 14, 52. https://doi.org/10.3892/br.2021.1428
MLA
Zhou, M., Xu, Q., Huang, D., Luo, L."Regulation of gene transcription of B lymphoma Mo‑MLV insertion region 1 homolog (Review)". Biomedical Reports 14.6 (2021): 52.
Chicago
Zhou, M., Xu, Q., Huang, D., Luo, L."Regulation of gene transcription of B lymphoma Mo‑MLV insertion region 1 homolog (Review)". Biomedical Reports 14, no. 6 (2021): 52. https://doi.org/10.3892/br.2021.1428