Open Access

Association between B‑Myb proto‑oncogene and the development of malignant tumors (Review)

  • Authors:
    • Yuelei Jin
    • Gangqiao Qi
    • Guang Chen
    • Chen Wang
    • Xiaoyan Fan
  • View Affiliations

  • Published online on: January 4, 2021     https://doi.org/10.3892/ol.2021.12427
  • Article Number: 166
  • Copyright: © Jin 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‑Myb is a critical transcription factor in regulating cell cycle. Dysregulated expression of B‑Myb promotes tumor formation and development. B‑Myb is a proto‑oncogene ubiquitously expressed in proliferating cells, which maintains normal cell cycle progression. It participates in cell apoptosis, tumorigenesis and aging. In addition, B‑Myb is overexpressed in several malignant tumors, including breast cancer, lung cancer and hepatocellular carcinoma, and is associated with tumor development. B‑Myb expression is also associated with the prognosis of patients with malignant tumors. Both microRNAs and E2F family of transcription factors (E2Fs) contribute to the function of B‑Myb. The present review highlights the association between B‑Myb and malignant tumors, and offers a theoretical reference for the diagnosis and treatment of malignant tumors.

Introduction

B-Myb, also known as MYB proto-oncogene like 2 (MYBl2), is a transcription factor that belongs to the Myb gene family, including A-Myb and c-Myb (1). MYB was the first discovered family member and is the mammalian homolog of the retroviral v-Myb oncogene that causes acute leukemia in birds and can transform hematopoietic cells (2). A-Myb is predominantly expressed in germ cells and primordial lymphocytes (3). c-Myb is highly expressed in cells of the hematopoietic system, with low expression in epithelial cells of specific tissues, such as the colon and brain (3,4). B-Myb is ubiquitously expressed in mammalian cells with high proliferative ability (1,4,5), and B-Myb is a physiological regulator of cell cycle progression, cell survival and cell differentiation (6). However, several experimental studies have demonstrated that B-Myb is highly expressed in different types of human malignant tumors, including breast cancer (7), lung cancer (8), hepatocellular carcinoma (9), endometrial cancer (10), prostate cancer (11) and ovarian cancer (12). Both in vivo and in vitro experiments have demonstrated that high B-Myb expression promotes colony formation, cell cycle progression, migration and invasion of cancer cells (13). B-Myb also participates in the occurrence of epithelial-to-mesenchymal transition (EMT) in malignant tumor, inhibits cancer cell apoptosis and results in a poor prognosis (14). However, the molecular mechanisms underlying the regulation of B-Myb in the development of malignant tumors remain unclear.

The present review highlights the association between B-Myb proto-oncogene and the development of malignant tumors based on available studies, aiming to provide insight into the molecular mechanisms underlying B-Myb-induced development of malignant tumors.

Role of B-Myb in the development of malignant tumors

B-Myb expression in malignant tumors

B-Myb is expressed at high levels in several human malignant tumor tissues (Table I). For example, there are five molecular subtypes of breast cancer, basal-like, human epidermal growth factor receptor 2 positive (HER2+)/estrogen receptor negative (ER-), luminal A, luminal B and normal-like (7). Microarray analyses of B-Myb in breast cancer tissues have demonstrated that its expression levels significantly differ among the five subtypes, with the highest level in basal-like tumors (7). Furthermore, overexpression of B-Myb has been observed in 83% of primary tumors and all cell lines of non-small cell lung cancer (NSCLC) (8), and B-Myb has also been demonstrated to modulate cell cycle and proliferation (15). Frau et al (9) assessed B-Myb mRNA and protein expression levels across different stages of hepatocarcinogenesis and demonstrated that B-Myb expression levels were substantially higher in precancerous lesions, early proliferative nodules, advanced proliferative nodules and hepatocellular carcinoma compared with normal liver tissues. In addition, the highest levels of B-Myb were observed in hepatocellular carcinoma, among the four different stages. Nakajima et al (16) reported amplification of the B-Myb gene copy number in 36/66 cases of primary hepatocellular carcinoma.

Table I.

Role of B-Myb in malignant tumor development.

Table I.

Role of B-Myb in malignant tumor development.

Tumor typeB-Myb expression levelPatient prognosisB-Myb expression treatmentIn vitro effect on cell phenotypeIn vivo effect on nude mice(Refs.)
Breast cancerHighPoorInterferenceInhibit cell cycle progression, cell proliferation, and cell migration and invasion.Inhibit tumor formation.(7,32)
Lung cancerHighPoorOverexpressionPromote tumorigenesis, cell proliferation and cell cycle progression.Promote tumor formation.(8,13,78)
InterferenceInhibit cell proliferation, cell cycle progression, and cell migration and invasion.Inhibit tumor formation.
Hepatocellular carcinomaHighPoorOverexpressionPromote tumorigenesis, cell proliferation and cell cycle progression.(9,34)
InterferenceInhibit cell proliferation and cell cycle progression.
Colorectal cancerHighPoorInterferenceInhibit cell proliferation, cell cycle progression and cell migration and invasion.(18)
Esophageal squamous-cell carcinomaHighPoorOverexpressionPromote cell cycle progression and cell proliferation.(19,20)
InterferenceInhibit cell proliferation.Inhibit tumor formation.
Gallbladder cancerHighPoorOverexpressionPromote cell cycle progression and cell proliferation.Promote tumor formation.(29)
InterferenceInhibit cell cycle progression and cell proliferation.Inhibit tumor formation.
GliomaHighPoorOverexpressionPromote cell proliferation. (25)
InterferenceInhibit cell cycle progression and cell proliferation.
Prostate cancerHigh(11,17)
Renal cell carcinomaHigh(26)
Ovarian cancerHigh(12,31)
NeuroblastomaHighPoor(47)
FibrosarcomaHigh (30)
Endometrial cancerHigh(10)
Primary leukemiaHigh(28)

[i] -, not available.

In the absence of gene amplification, malignant tumors of the prostate exhibit elevated B-Myb expression levels, and B-Myb expression is markedly higher in metastatic prostate cancer compared with non-metastatic prostate cancer (11,17). Based on a study involving 180 patients with colorectal cancer, B-Myb mRNA and protein expression levels were notably higher in cancer tissues compared with adjacent normal tissues, and B-Myb expression was positively associated with tumor size and clinical stage (18).

Qin et al (19) demonstrated that B-Myb is amplified in esophageal cancer, based on whole-genome sequencing (10 pairs) and whole-exome sequencing (57 pairs) of esophageal cancer tissues and matched adjacent normal tissues selected from high-incidence areas of esophageal cancer in China. Additionally, Qin et al (20) reported that B-Myb protein was expressed at higher levels in esophageal squamous cell carcinoma (ESCC) tissues compared with adjacent normal tissues in a Chinese population of 107 patients with ESCC, based on immunohistochemistry.

Among patients with neuroblastoma, individuals with malignant metastasis and poor prognosis express B-Myb at significantly elevated levels; this phenomenon suggests that B-Myb expression is associated with the risk of developing neuroblastoma (21). Notable, inhibition of B-Myb expression prevents the proliferation of normal human cells and cancer cells (2124). In addition, B-Myb expression levels are significantly higher in glioma tissues compared with adjacent normal tissues and are positively associated with the grade of glioma, based on the results of reverse transcription-PCR (RT-PCR) and western blot analyses from 79 patients with glioma (25).

In renal cell carcinoma, metastatic tumor tissues highly express B-Myb when metastasis occurs in primary tumors negative for B-Myb expression (26). B-Myb is also amplified in high-grade bladder cancer (27). In addition, high B-Myb expression has been implicated in leukemias (28), gallbladder cancer (29), fibrosarcoma (30), ovarian cancer (12) and aggressive T-cell lymphoma (31).

Biological function of B-Myb in malignant tumor development

Promotion of cancer cell proliferation

Increasing evidence suggests that B-Myb is overexpressed in different types of human cancer, including breast cancer (32), cervical cancer (33), colorectal cancer (18), liver cancer (34), leukemia cells (28) and lung cancer (35). In these types of cancer, B-Myb promotes cell proliferation and/or cell cycle progression (18,34). Thomas et al (36) observed a positive correlation between B-Myb mRNA expression and Ki-67 proliferation index in breast cancer. In addition, the breast cancer cell line, MDA-MB-231, exhibits remarkably decreased abilities to form colonies, migrate and invade following knockdown of B-Myb with short-hairpin RNA (37). Flow cytometric analysis demonstrated that the cell cycle is arrested at S and G2/M phases, while in vivo experiments indicated that both the rate of tumor formation and the weight of tumor mass are significantly lower in breast cancer compared with the control group (32).

Jin et al (13) demonstrated that overexpression of B-Myb promotes the proliferation of NSCLC cells; both extracellular regulated MAP kinase (ERK) and phosphorylated-protein kinase B (Akt) signaling pathways participate in the modulation of NSCLC by B-Myb. Liang et al (29) reported that B-Myb expression is upregulated in gallbladder cancer tissues, which in turn facilitates the proliferation of gallbladder cancer cells by facilitating cell cycle progression through the S and G2/M phases (30).

In ESCC, an EdU-retention assay demonstrated that downregulation of B-Myb expression decreases the DNA synthesis ability of EC9706 cells, while a Cell Counting Kit-8 assay demonstrated that overexpression of B-Myb also promotes the proliferation of KYSE510 cells (20). These findings suggest that B-Myb can promote the proliferation and DNA synthesis of ESCC cells. Based on a colony formation assay, overexpression of B-Myb in the low-grade glioma cell line, Hs683, considerably increased the number of colonies, whereas knockout of B-Myb in the high-grade glioma cell line, U251, decreased the number of colonies (25). Subsequently, a MTT assay demonstrated that the proliferative ability of glioma U251 cells is enhanced following transfection with small interfering (si)RNAs (25).

Promotion of EMT

B-Myb has been demonstrated to play a role in EMT, a process whereby epithelial cells lose their polarity, migrate and increase polarity (38). In breast cancer cells, downregulation of B-Myb expression can recover the expression of the epithelial marker, E-cadherin and promote the formation of intercellular adhesion, in addition to inhibiting cell invasion, anchorage-dependent growth and tumor formation (32). Conversely, overexpression of B-Myb can decrease E-cadherin expression and increase the expression of mesenchymal markers (14). In addition, it has been demonstrated that B-Myb can upregulate the expression of Snail, a key regulator of EMT, thereby mediating the promotion of EMT and cancer cell invasion (14).

The role of B-Myb in colorectal cancer invasion and metastasis has also been proven and is associated with EMT (18). For example, EMT inhibits B-Myb activity in colorectal cancer cells, thereby upregulating the expression of the epithelial marker, E-cadherin and downregulating the expression of the mesenchymal marker, Vimentin and matrix metalloproteinase 9 (MMP9) (18). Based on a western blot assay on the expression of EMT markers in glioma cells, interference with B-Myb expression inhibits the protein expression levels of N-cadherin, Vimentin, MMP2 and MMP9, while upregulating the protein levels of E-cadherin and zinc finger E-box binding homeobox 1 (25). Taken together, these findings suggest that B-Myb plays an important role in the promotion of EMT in several malignant tumors, thereby facilitating cancer cell infiltration and metastasis.

Inhibition of cancer cell apoptosis

B-Myb inhibits cancer cell apoptosis potentially through multiple pathways. First, B-Myb may perform its anti-apoptotic function by positively regulating the expression of the anti-apoptotic gene, Clusterin (30), also known as apolipoprotein J (39). A study demonstrated that inhibition of Clusterin gene expression promotes the apoptosis of fibrosarcoma cells (30). Secondly, B-Myb may inhibit apoptosis by positively regulating the expression of the anti-apoptotic gene, Bcl-2 (40), a critical regulator of apoptosis (41). In support of these pathways, the anti-apoptotic function of B-Myb has been observed in different cancer cell lines, including colorectal cancer (18) and liver cancer (34).

Ren et al (18) reported that Bcl-2 protein expression is downregulated in SW480 colorectal cancer cells via interference with B-Myb expression. Calvisi et al (34) demonstrated that B-Myb exerts an anti-apoptotic function, and interference with B-Myb expression via transfection with siRNA induces apoptosis in four different hepatocellular carcinoma cell lines. In glioma U251 cells transfected with B-Myb siRNA, Zhang et al (25) detected an increase in the percentage of apoptotic cells by Annexin V-FITC/PI and hochest 3342 staining. Furthermore, Zhang et al (25) assessed the effects of B-Myb silencing on apoptosis-related proteins, including caspase-3/9, Bcl/Bax, PTEN and P53. Western blot analysis demonstrated that B-Myb silencing decreases Bcl-2 expression, while increasing the expression levels of Bax, PTEN and P53, and activates caspase-3/9 activity. Taken together, these results suggest that downregulation of B-Myb can induce apoptosis in glioma cells.

Enhancement of drug resistance

The role of B-Myb in tumor resistance to therapy is associated with its pro-apoptotic function (42,43). Overexpression of B-Myb promotes the expression of the anti-apoptotic gene, Bcl-2 in IL2-dependent murine CTLL-2 cells, thereby increasing the therapeutic resistance to drugs, including doxorubicin, ceramide and dexamethasone (42). Similarly, Levenson et al (43) demonstrated that B-Myb expression is notably upregulated in fibrosarcoma cells with therapeutic resistance induced by chemotherapeutic drugs that inhibit DNA synthesis, including hydroxyurea, cysteine, etoposide and adriamycin. Furthermore, B-Myb regulates the expression of the anti-apoptotic gene, Apolipoprotein J/Clusterin in neuroblastoma cells to resist apoptosis caused by doxycycline (30).

Sottile et al (44) reported that individuals with B-Myb overexpression or MYCN amplification are more sensitive to therapy with camptothecins (irinotecan and topotecan) among patients with neuroblastoma. B-Myb is a downstream target of MYCN; MYCN amplification promotes B-Myb overexpression, while B-Myb overexpression in turn promotes an upregulation of MYCN expression, thus these two factors regulate each other (21). Camptothecins selectively downregulates B-Myb and MYCN expression, while upregulation of B-Myb decreases the killing effect of camptothecins, suggesting that B-Myb is an important target of camptothecins (44). In addition, B-Myb is overexpressed in cetuximab-resistant NSCLC, suggesting that B-Myb overexpression is associated with cetuximab resistance (45).

Effect on patient prognosis

High B-Myb expression is associated with tumor growth and poor prognosis of patients, making it a potential clinical marker for poor prognosis (20,46). Based on microRNA (miRNA/miR) prediction and RT-PCR analyses, B-Myb may have a negative regulatory association with miR-30 family members, and biochemical relapse-free survival time is shortened in patients with acute myeloid leukemia highly overexpressing B-Myb; thus, B-Myb can be a predictive marker for the prognosis of patients with acute myeloid leukemia (46). In addition, B-Myb expression is elevated in ESCC tissues and negatively associated with postoperative overall survival in patients with ESCC, as revealed by a Kaplan-Meier analysis (20).

A prognostic analysis of breast cancer and its different subtypes revealed that the B-Myb high expression group has a worse prognosis compared with the low expression group (7). High B-Myb expression also increases the risk of poor prognosis, decreases the differentiation ability of cells, and promotes tumor development in neuroblastoma (47). In HepG2 and HuH7 hepatocellular carcinoma cell lines, overexpression of B-Myb increases the cell proliferative ability and facilitates G1-S and G2-M transitions, whereas interference of B-Myb with siRNA results in cell cycle arrest at G0-G1 and G2-M phases (9).

Among patients with colorectal cancer, the 5-year survival rate is notably lower in individuals with high B-Myb expression than those with low B-Myb expression; B-Myb expression and clinical stage of the tumor can be used as independent prognostic factors of colorectal cancer (18). With regards to the prognosis and survival of 79 patients with glioma, Kaplan-Meier analysis and log-rank test results indicated that high B-Myb expression is negatively associated with survival, and is a poor prognostic factor in patients with glioma (25).

Role of B-Myb in the diagnosis and treatment of malignant tumors effect

B-Myb plays a role in the clinical diagnosis and treatment of malignant tumors. B-Myb can be used as a biological marker of cervical cancer to make up for the limitations of conventional cytology for cervical intraepithelial neoplasia and cervical cancer diagnosis in routine cervical cytology testing (43,44). Astbury et al (48) measured MYBL2 expression levels in cervical cancer cell lines, cervical intraepithelial neoplasia and cervical glandular epithelium using genomics and proteomics technology, and assessed the potential of B-Myb as a biomarker. Previous studies (47) have demonstrated that patients with neuroblastoma, with MYBL2 overexpression, are more sensitive to camptothecin therapy (44). Camptothecin drugs can selectively downregulate the expression of B-Myb. Conversely, upregulating B-Myb can decrease the killing effect of camptothecin drugs (44). Collectively, these results suggest that B-Myb is an important target of camptothecin drugs (44).

Regulatory mechanisms of B-Myb in malignant tumor

B-Myb regulation by miRNAs

miRNAs are a group of small non-coding single stranded RNAs that have been extensively observed in animals and plants, typically 18–22 nucleotides in length (49). Both misregulation and mutation of miRNAs can cause tumorigenesis, and they contribute to the function of oncogenes through targeted downregulation of tumor suppressor genes or activation of oncogene transcription factors (50,51). B-Myb is mainly regulated by the miR-29 and miR-30 families (52). In breast cancer cell lines, miRNA binds to the 3′-untranslated region of B-Myb and thereby inhibits B-Myb expression (53). Additionally, miR-29a expression is negatively correlated with B-Myb expression, and cyclin A2 and cyclin D1 expression is positively correlated with B-Myb expression (54). These results indicate that miR-29a suppresses tumor growth by downregulating B-Myb expression (54).

Geng et al (55) reported that miR-30a expression is significantly downregulated in NSCLC tissues compared with adjacent normal tissues, and B-Myb is a target gene of miR-30a based on a double-luciferase reporter gene assay; miR-30a can suppress proliferation and growth of NSCLC through targeted inhibition of B-Myb expression. Li et al (56) demonstrated a close association between B-Myb overexpression and low levels of miR-30a, miR-30b and miR-30c in 291 patients with acute myeloid, based on RT-qPCR analysis of B-Myb, miR-29 family and miR-30 family genes. Li et al (56) reported that the long non-coding RNA, LINC01139, upregulates B-Myb by competitively binding to the miR-30 family, thereby promoting the progression of hepatocellular carcinoma.

B-Myb is also regulated by other miRNAs during tumor development

Zauli et al (28) demonstrated that the cell cycle is arrested at G1 following downregulation of B-Myb expression in primary leukemia cells and P53 wild-type myeloid and lymphoblastic cells; miR-34a plays a pivotal regulatory role in this process. Lee et al (57) reported that inhibition of miR-34a recovers B-Myb expression, while miR-34a mimics downregulates B-Myb expression in HCT116 colorectal cancer cells. In addition, Li et al (56) and Yu et al (58) have concluded that the expression of G1/S-related genes, Ezh2 and B-Myb, are suppressed by miR-34c overexpression in the cancer cell line cultured from a mouse model of tubal high-grade serous ovarian carcinoma, leading to cell cycle arrest at G1 phase and induction of apoptosis.

Wang et al (59) demonstrated that post-transcriptional regulation of MALAT1 is regulated by miR-101 and miR-217 in ESCC. Specifically, post-transcriptional silencing of MALAT1 significantly inhibits ESCC cell proliferation by arresting the G2/M phase, while the migratory and invasive abilities of ESCC cells decrease following overexpression of miR-101 and miR-217. This may be due to MALAT1-mediated upregulation of P21 and P27 expression and inhibition of B-Myb expression (60). In addition, Chen et al (61) indicated that miR-143-3p negatively regulates B-Myb in breast cancer cells, and modulates cancer cell proliferation and apoptosis (62).

B-Myb regulation by E2Fs

The E2F transcription factor family plays a crucial role in the regulation of cell cycle progression, DNA replication and apoptosis (63). The E2F family can be divided into two groups, E2F1-3a are transcription factors that activate the cell cycle and mainly encode genes that promote the progression of G1 to S phase, while E2F3b and E2F4-8 mainly promote cells to exit the cell replication cycle and facilitate cell differentiation (64,65). B-Myb is a typical cell cycle regulator that is rarely expressed in the G0 phase (66). When external growth factor-mediated signal pathways, such as ERK1/2, are activated, they promotes the release and activation of E2F1-3 from pRB through CCND1; these factors drive their target genes to encode G1/S-related cytokines, and B-Myb expression is also induced by E2F1-3 (4,67,68). The induction of B-Myb expression at the end of the G1 phase is due to a substantial increase in gene transcription, suggesting that B-Myb may be a gene regulated by the E2F transcription factors (62).

The promoters of both human and murine B-Myb genes contain completely conserved E2F-binding sites, which is key for B-Myb to participate in the transcription regulation of the cell cycle (69,70). A chromatin immunoprecipitation assay of NIH3T3 in fibroblasts revealed that both E2F4/P107 and E2F4/P130 are associated with the B-Myb promoter in the G0 phase, while E2F4/P107 is associated with the B-Myb promoter in the early G1 phase (69). In human malignant glioma T98G cells, E2F1 and E2F3 are associated with the B-Myb promoter in the late G1 phase (71). B-Myb transcriptionally activates its own promoter through the SP1-binding site adjacent to the transcription initiation site (72). SP1 is coupled with E2F1 to promote transcriptional activation of the B-Myb promoter (73,74). It is suggested that E2F1 and E2F3 play a role in promoting B-Myb transcription in at least some cells (75).

Nakajima et al (16) demonstrated that knockout of E2F1 in JHH-5 hepatocellular carcinoma cells decreases the expression levels of B-MYB, CCNE1, MYC, TK1 and RRM1. The kinetics of B-Myb interaction with G2-regulated promoters coincides with the activation of the genes; a decrease in RNAi-regulated B-Myb expression can inhibit cyclin B1 and cell division cycle 2 (cdc2) expression, arrest cell cycle at G2/M, and increase apoptosis (76). The interaction of B-Myb with the cdc2 promoter is dependent on the complete E2F-binding site, and the B-Myb gene is regulated by E2F at G1/S transition, thereby modulating the target genes associated with G1/S and G2/M transitions, including cdc2, cyclin A2 and cyclin B1 (77).

Conclusions

B-Myb, a classic oncogene, promotes the development of malignant tumors (5). B-Myb is highly expressed in several tumors, and its expression is associated with the clinicopathological characteristics of tumors (13,33,78). High B-Myb expression severely affects the prognosis of patients, with a relatively low 5-year survival rate (7). In vitro studies have demonstrated that B-Myb promotes cell cycle progression, proliferation, invasion and migration of cancer cells (35). In vivo experiments have also reported that B-Myb facilitates tumor formation (35). Both miRNAs (46,47,51) and E2Fs (16) contribute to the function of B-Myb by regulating its expression. miR-29, miR-30, miR-34, amiR-101 and miR-217 all participate in the regulation of B-Myb, thus affecting cell functions, including senescence, proliferation, invasion and metastasis (Fig. 1). E2Fs interact with B-Myb through promoter elements, which in turn activates target genes involved in G1/S and G2/M transitions, thereby promoting cell cycle progression (16). Following research advances on the molecular mechanisms of malignant tumor development, it may be possible to apply B-Myb to the diagnosis and treatment of patients with tumors. Determining the role of B-Myb in tumor development will provide novel tumor markers, while starting a novel chapter of potential targeted intervention therapy of malignant tumors.

Acknowledgements

Not applicable.

Funding

The present review was supported in part by a grant-in-aid from the Scientific Research Fund of Zhejiang Provincial Education Department (grant no. Y202045646 to XF), the Scientific Research Project of Taizhou Municipal Science and Technology Bureau (grant no. 20ywb100 to XF and grant no. 20ywb101 to YJ), and Students Science and Technology Innovation Activity Plan of Zhejiang Province (also known as ‘New Seedling Talent Plan’; grant no. 2020R468017 to XF). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

YJ, GQ, GC, CW and XF made substantial contributions to conception and design, or acquisition of data, or analysis and interpretation of data. XF and YJ drafted the initial manuscript and critically revised it for important intellectual content. GC and CW designed the study, and critically revised the article. XF approved the final version to be published.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Oh IH and Reddy EP: The myb gene family in cell growth, differentiation and apoptosis. Oncogene. 18:3017–3033. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Lipsick JS and Wang DM: Transformation by v-Myb. Oncogene. 18:3047–3055. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Toscani A, Mettus RV, Coupland R, Simpkins H, Litvin J, Orth J, Hatton KS and Reddy EP: Arrest of spermatogenesis and defective breast development in mice lacking A-myb. Nature. 386:713–717. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Sala A: B-MYB, a transcription factor implicated in regulating cell cycle, apoptosis and cancer. Eur J Cancer. 41:2479–2484. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Iness AN, Felthousen J, Ananthapadmanabhan V, Sesay F, Saini S, Guiley KZ, Rubin SM, Dozmorov M and Litovchick L: The cell cycle regulatory DREAM complex is disrupted by high expression of oncogenic B-Myb. Oncogene. 38:1080–1092. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Musa J, Aynaud MM, Mirabeau O, Delattre O and Grünewald TG: MYBL2 (B-Myb): A central regulator of cell proliferation, cell survival and differentiation involved in tumorigenesis. Cell Death Dis. 8:e28952017. View Article : Google Scholar : PubMed/NCBI

7 

Thorner AR, Hoadley KA, Parker JS, Winkel S, Millikan RC and Perou CM: In vitro and in vivo analysis of B-Myb in basal-like breast cancer. Oncogene. 28:742–751. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Long EM, Long MA, Tsirigotis M and Gray DA: Stimulation of the murine Uchl1 gene promoter by the B-Myb transcription factor. Lung Cancer. 42:9–21. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Frau M, Ladu S, Calvisi DF, Simile MM, Bonelli P, Daino L, Tomasi ML, Seddaiu MA, Feo F and Pascale RM: Mybl2 expression is under genetic control and contributes to determine a hepatocellular carcinoma susceptible phenotype. J Hepatol. 55:111–119. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Krakstad C, Tangen IL, Hoivik EA, Halle MK, Berg A, Werner HM, Ræder MB, Kusonmano K, Zou JX, Øyan AM, et al: ATAD2 overexpression links to enrichment of B-MYB-translational signatures and development of aggressive endometrial carcinoma. Oncotarget. 6:28440–28452. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Bar-Shira A, Pinthus JH, Rozovsky U, Goldstein M, Sellers WR, Yaron Y, Eshhar Z and Orr-Urtreger A: Multiple genes in human 20q13 chromosomal region are involved in an advanced prostate cancer xenograft. Cancer Res. 62:6803–6807. 2002.PubMed/NCBI

12 

Tanner MM, Grenman S, Koul A, Johannsson O, Meltzer P, Pejovic T, Borg A and Isola JJ: Frequent amplification of chromosomal region 20q12-q13 in ovarian cancer. Clin Cancer Res. 6:1833–1839. 2000.PubMed/NCBI

13 

Jin Y, Zhu H, Cai W, Fan X, Wang Y, Niu Y, Song F and Bu Y: B-Myb is up-regulated and promotes cell growth and motility in non-small cell lung cancer. Int J Mol Sci. 18:8602017. View Article : Google Scholar

14 

Tao D, Pan Y, Jiang G, Lu H, Zheng S, Lin H and Cao F: B-Myb regulates snail expression to promote epithelial-to-mesenchymal transition and invasion of breast cancer cell. Med Oncol. 32:4122015. View Article : Google Scholar : PubMed/NCBI

15 

Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L and Powell CA: Non-small-cell lung cancer molecular signatures recapitulate lung developmental pathways. Am J Pathol. 163:1949–1960. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Nakajima T, Yasui K, Zen K, Inagaki Y, Fujii H, Minami M, Tanaka S, Taniwaki M, Itoh Y, Arii S, et al: Activation of B-Myb by E2F1 in hepatocellular carcinoma. Hepatol Res. 38:886–895. 2008.PubMed/NCBI

17 

Skotheim RI, Monni O, Mousses S, Fossa SD, Kallioniemi OP, Lothe RA and Kallioniemi A: New insights into testicular germ cell tumorigenesis from gene expression profiling. Cancer Res. 62:2359–2364. 2002.PubMed/NCBI

18 

Ren F, Wang L, Shen X, Xiao X, Liu Z, Wei P, Wang Y, Qi P, Shen C, Sheng W and Du X: MYBL2 is an independent prognostic marker that has tumor-promoting functions in colorectal cancer. Am J Cancer Res. 5:1542–1552. 2015.PubMed/NCBI

19 

Qin HD, Liao XY, Chen YB, Huang SY, Xue WQ, Li FF, Ge XS, Liu DQ, Cai Q, Long J, et al: Genomic characterization of esophageal squamous cell carcinoma reveals critical genes underlying tumorigenesis and poor prognosis. Am J Hum Genet. 98:709–727. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Qin H, Li Y, Zhang H, Wang F, He H, Bai X and Li S: Prognostic implications and oncogenic roles of MYBL2 protein expression in esophageal squamous-cell carcinoma. OncoTargets Ther. 12:1917–1927. 2019. View Article : Google Scholar

21 

Gualdrini F, Corvetta D, Cantilena S, Chayka O, Tanno B, Raschellà G and Sala A: Addiction of MYCN amplified tumours to B-MYB underscores a reciprocal regulatory loop. Oncotarget. 1:278–288. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Sala A, Casella I, Bellon T, Calabretta B, Watson RJ and Peschle C: B-myb promotes S phase and is a downstream target of the negative regulator p107 in human cells. J Biol Chem. 271:9363–9367. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Arsura M, Introna M, Passerini F, Mantovani A and Golay J: B-myb antisense oligonucleotides inhibit proliferation of human hematopoietic cell lines. Blood. 79:2708–2716. 1992. View Article : Google Scholar : PubMed/NCBI

24 

Sala A and Calabretta B: Regulation of BALB/c 3T3 fibroblast proliferation by B-myb is accompanied by selective activation of cdc2 and cyclin D1 expression. Proc Natl Acad Sci USA. 89:10415–10419. 1992. View Article : Google Scholar : PubMed/NCBI

25 

Zhang X, Lv QL, Huang YT, Zhang LH and Zhou HH: Akt/FoxM1 signaling pathway-mediated upregulation of MYBL2 promotes progression of human glioma. J Exp Clin Cancer Res. 36:1052017. View Article : Google Scholar : PubMed/NCBI

26 

Sakai N, Kubota Y and Shuin T: Statistically significant expression of B-myb in clinically advanced human renal-cell carcinomas. Int J Oncol. 2:419–423. 1993.PubMed/NCBI

27 

Nord H, Segersten U, Sandgren J, Wester K, Busch C, Menzel U, Komorowski J, Dumanski JP, Malmström PU and Díaz de Ståhl T: Focal amplifications are associated with high grade and recurrences in stage Ta bladder carcinoma. Int J Cancer. 126:1390–1402. 2010.PubMed/NCBI

28 

Zauli G, Voltan R, di Iasio MG, Bosco R, Melloni E, Sana ME and Secchiero P: miR-34a induces the downregulation of both E2F1 and B-Myb oncogenes in leukemic cells. Clin Cancer Res. 17:2712–2724. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Liang HB, Cao Y, Ma Q, Shu YJ, Wang Z, Zhang F, Ye YY, Li HF, Xiang SS, Song XL, et al: MYBL2 is a potential prognostic marker that promotes cell proliferation in gallbladder cancer. Cell Physiol Biochem. 41:2117–2131. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Cervellera M, Raschella G, Santilli G, Tanno B, Ventura A, Mancini C, Sevignani C, Calabretta B and Sala A: Direct transactivation of the anti-apoptotic gene apolipoprotein J (clusterin) by B-MYB. J Biol Chem. 275:21055–21060. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Mao X, Orchard G, Lillington DM, Russell-Jones R, Young BD and Whittaker SJ: Amplification and overexpression of JUNB is associated with primary cutaneous T-cell lymphomas. Blood. 101:1513–1519. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Tao D, Pan Y, Lu H, Zheng S, Lin H, Fang H and Cao F: B-myb is a gene implicated in cell cycle and proliferation of breast cancer. Int J Clin Exp Pathol. 7:5819–5827. 2014.PubMed/NCBI

33 

Martin CM, Astbury K, Kehoe L, O'Crowley JB, O'Toole S and O'Leary JJ: The use of MYBL2 as a novel candidate biomarker of cervical cancer. Methods Mol Biol. 1249:241–251. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Calvisi DF, Simile MM, Ladu S, Frau M, Evert M, Tomasi ML, Demartis MI, Daino L, Seddaiu MA, Brozzetti S, et al: Activation of v-Myb avian myeloblastosis viral oncogene homolog-like2 (MYBL2)-LIN9 complex contributes to human hepatocarcinogenesis and identifies a subset of hepatocellular carcinoma with mutant p53. Hepatology. 53:1226–1236. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Iltzsche F, Simon K, Stopp S, Pattschull G, Francke S, Wolter P, Hauser S, Murphy DJ, Garcia P, Rosenwald A and Gaubatz S: An important role for Myb-MuvB and its target gene KIF23 in a mouse model of lung adenocarcinoma. Oncogene. 36:110–1121. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Thomas C, Robinson C, Dessauvagie B, Wood B, Sterrett G, Harvey J and Amanuel B: Expression of proliferation genes in formalin-fixed paraffin-embedded (FFPE) tissue from breast carcinomas. Feasibility and relevance for a routine histopathology laboratory. J Clin Pathol. 70:25–32. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Wolter P, Hanselmann S, Pattschull G, Schruf E and Gaubatz S: Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy. Oncotarget. 8:11160–11172. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Diepenbruck M and Christofori G: Epithelial-mesenchymal transition (EMT) and metastasis: Yes, no, maybe? Curr Opin Cell Biol. 43:7–13. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R and Wang CY: Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol. 7:909–915. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Lang G, Gombert WM and Gould HJ: A transcriptional regulatory element in the coding sequence of the human Bcl-2 gene. Immunology. 114:25–36. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Lee HJ, Lee EK, Seo YE, Shin YH, Kim HS, Chun YH, Yoon JS, Kim HH, Han MY, Kim CK, et al: Roles of Bcl-2 and caspase-9 and −3 in CD30-induced human eosinophil apoptosis. J Microbiol Immunol Infect. 50:145–152. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Grassilli E, Salomoni P, Perrotti D, Franceschi C and Calabretta B: Resistance to apoptosis in CTLL-2 cells overexpressing B-Myb is associated with B-Myb-dependent bcl-2 induction. Cancer Res. 59:2451–2456. 1999.PubMed/NCBI

43 

Levenson VV, Davidovich IA and Roninson IB: Pleiotropic resistance to DNA-interactive drugs is associated with increased expression of genes involved in DNA replication, repair, and stress response. Cancer Res. 60:5027–5030. 2000.PubMed/NCBI

44 

Sottile F, Gnemmi I, Cantilena S, D'Acunto WC and Sala A: A chemical screen identifies the chemotherapeutic drug topotecan as a specific inhibitor of the B-MYB/MYCN axis in neuroblastoma. Oncotarget. 3:535–545. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Iida M, Brand TM, Campbell DA, Li C and Wheeler DL: Yes and Lyn play a role in nuclear translocation of the epidermal growth factor receptor. Oncogene. 32:759–767. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Fuster O, Llop M, Dolz S, García P, Such E, Ibáñez M, Luna I, Gómez I, López M, Cervera J, et al: Adverse prognostic value of MYBL2 overexpression and association with microRNA-30 family in acute myeloid leukemia patients. Leuk Res. 37:1690–1696. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Raschella G, Cesi V, Amendola R, Negroni A, Tanno B, Altavista P, Tonini GP, De Bernardi B and Calabretta B: Expression of B-myb in neuroblastoma tumors is a poor prognostic factor independent from MYCN amplification. Cancer Res. 59:3365–338. 1999.PubMed/NCBI

48 

Astbury K, McEvoy L, Brian H, Spillane C, Sheils O, Martin C and O'Leary JJ: MYBL2 (B-MYB) in cervical cancer: putative biomarker. Int J Gynecologic Cancer. 21:206–212. 2011. View Article : Google Scholar

49 

Komiya R: Biogenesis of diverse plant phasiRNAs involves an miRNA-trigger and Dicer-processing. J Plant Res. 130:17–23. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, et al: Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 99:15524–15529. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M and Croce CM: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Martinez I, Cazalla D, Almstead LL, Steitz JA and DiMaio D: miR-29 and miR-30 regulate B-Myb expression during cellular senescence. Proc Natl Acad Sci USA. 108:522–527. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Martinez I and Dimaio D: B-Myb, cancer, senescence, and microRNAs. Cancer Res. 71:5370–5373. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Wu Z, Huang X, Huang X, Zou Q and Guo Y: The inhibitory role of Mir-29 in growth of breast cancer cells. J Exp Clin Cancer Res. 32:982013. View Article : Google Scholar : PubMed/NCBI

55 

Geng GJ, Yang YT, Jiang J, Yu XY and Fa XE: MicroRNA-30a suppresses non-small-cell lung cancer by targeting Myb-related protein B. Exp Ther Med. 15:1633–1639. 2018.PubMed/NCBI

56 

Li ZB, Chu HT, Jia M and Li L: Long noncoding RNA LINC01139 promotes the progression of hepatocellular carcinoma by upregulating MYBL2 via competitively binding to miR-30 family. Biochem Biophys Res Commun. 525:581–588. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Lee YJ, Kang YR, Lee SY, Jin Y, Han DC and Kwon BM: Ginkgetin induces G2-phase arrest in HCT116 colon cancer cells through the modulation of bMyb and miRNA34a expression. Int J Oncol. 51:1331–1342. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Yu Z, Kim J, He L, Creighton CJ, Gunaratne PH, Hawkins SM and Matzuk MM: Functional analysis of miR-34c as a putative tumor suppressor in high-grade serous ovarian cancer. Biol Reprod. 91:1132014. View Article : Google Scholar : PubMed/NCBI

59 

Wang X, Li M, Wang Z, Han S, Tang X, Ge Y, Zhou L, Zhou C, Yuan Q and Yang M: Silencing of long noncoding RNA MALAT1 by miR-101 and miR-217 inhibits proliferation, migration, and invasion of esophageal squamous cell carcinoma cells. J Biol Chem. 290:3925–3935. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Zhang Y, Tang X, Shi M, Wen C and Shen B: MiR-216a decreases MALAT1 expression, induces G2/M arrest and apoptosis in pancreatic cancer cells. Biochem Biophys Res Commun. 483:816–822. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Chen J and Chen X: MYBL2 is targeted by miR-143-3p and regulates breast cancer cell proliferation and apoptosis. Oncol Res. 26:913–922. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Lam EW, Robinson C and Watson RJ: Characterization and cell cycle-regulated expression of mouse B-myb. Oncogene. 7:1885–1890. 1992.PubMed/NCBI

63 

de Bruin A, Maiti B, Jakoi L, Timmers C, Buerki R and Leone G: Identification and characterization of E2F7, a novel mammalian E2F family member capable of blocking cellular proliferation. J Biol Chem. 278:42041–42049. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Wong JV, Dong P, Nevins JR, Mathey-Prevot B and You L: Network calisthenics: Control of E2F dynamics in cell cycle entry. Cell Cycle. 10:3086–3094. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Zhu W, Giangrande PH and Nevins JR: Temporal control of cell cycle gene expression mediated by E2F transcription factors. Cell Cycle. 4:633–636. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Robinson C, Light Y, Groves R, Mann D, Marias R and Watson R: Cell-cycle regulation of B-Myb protein expression: Specific phosphorylation during the S phase of the cell cycle. Oncogene. 12:1855–1864. 1996.PubMed/NCBI

67 

Mowla SN, Lam EW and Jat PS: Cellular senescence and aging: The role of B-MYB. Aging Cell. 13:773–779. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Inoue K and Fry EA: Novel molecular markers for breast cancer. Biomark Cancer. 8:25–42. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Lam EW, Bennett JD and Watson RJ: Cell-cycle regulation of human B-myb transcription. Gene. 160:277–281. 1995. View Article : Google Scholar : PubMed/NCBI

70 

Lam EW and Watson RJ: An E2F-binding site mediates cell-cycle regulated repression of mouse B-myb transcription. EMBO J. 12:2705–2713. 1993. View Article : Google Scholar : PubMed/NCBI

71 

Lukas J, Petersen BO, Holm K, Bartek J and Helin K: Deregulated expression of E2F family members induces S-phase entry and overcomes p16INK4A-mediated growth suppression. Mol Cell Biol. 16:1047–1057. 1996. View Article : Google Scholar : PubMed/NCBI

72 

Sala A, Saitta B, De Luca P, Cervellera MN, Casella I, Lewis RE, Watson R and Peschle C: B-MYB transactivates its own promoter through SP1-binding sites. Oncogene. 18:1333–1339. 1999. View Article : Google Scholar : PubMed/NCBI

73 

Müller H, Bracken AP, Vernell R, Moroni MC, Christians F, Grassilli E, Prosperini E, Vigo E, Oliner JD and Helin K: E2Fs regulate the expression of genes involved in differentiation, development, proliferation, and apoptosis. Genes Dev. 15:267–285. 2001. View Article : Google Scholar : PubMed/NCBI

74 

Infante A, Laresgoiti U, Fernandez-Rueda J, Fullaondo A, Galán J, Díaz-Uriarte R, Malumbres M, Field SJ and Zubiaga AM: E2F2 represses cell cycle regulators to maintain quiescence. Cell Cycle. 7:3915–3927. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Joaquin M and Watson RJ: Cell cycle regulation by the B-Myb transcription factor. Cell Mol Life Sci. 60:2389–2401. 2003. View Article : Google Scholar : PubMed/NCBI

76 

Santilli G, Schwab R, Watson R, Ebert C, Aronow BJ and Sala A: Temperature-dependent modification and activation of B-MYB: Implications for cell survival. J Biol Chem. 280:15628–15634. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Zhu W, Giangrande PH and Nevins JR: E2Fs link the control of G1/S and G2/M transcription. EMBO J. 23:4615–4626. 2004. View Article : Google Scholar : PubMed/NCBI

78 

Fan X, Wang Y, Jiang T, Cai W, Jin Y, Niu Y, Zhu H and Bu Y: B-Myb mediates proliferation and migration of non-small-cell lung cancer via suppressing IGFBP3. Int J Mol Sci. 19:14792018. View Article : Google Scholar

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jin Y, Qi G, Chen G, Wang C and Fan X: Association between B‑<em>Myb</em> proto‑oncogene and the development of malignant tumors (Review). Oncol Lett 21: 166, 2021
APA
Jin, Y., Qi, G., Chen, G., Wang, C., & Fan, X. (2021). Association between B‑<em>Myb</em> proto‑oncogene and the development of malignant tumors (Review). Oncology Letters, 21, 166. https://doi.org/10.3892/ol.2021.12427
MLA
Jin, Y., Qi, G., Chen, G., Wang, C., Fan, X."Association between B‑<em>Myb</em> proto‑oncogene and the development of malignant tumors (Review)". Oncology Letters 21.2 (2021): 166.
Chicago
Jin, Y., Qi, G., Chen, G., Wang, C., Fan, X."Association between B‑<em>Myb</em> proto‑oncogene and the development of malignant tumors (Review)". Oncology Letters 21, no. 2 (2021): 166. https://doi.org/10.3892/ol.2021.12427