Open Access

Jumonji AT‑rich interactive domain 1B promotes the growth of pancreatic tumors via the phosphatase and tensin homolog/protein kinase B signaling pathway

Retraction in: /10.3892/ol.2023.13660

  • Authors:
    • Xudong Shen
    • Guilian Cheng
    • Liming Xu
    • Wei Wu
    • Zhengrong Chen
    • Peng Du
  • View Affiliations

  • Published online on: May 2, 2018     https://doi.org/10.3892/ol.2018.8618
  • Pages: 267-275
  • Copyright: © Shen 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

Jumonji AT‑rich interactive domain 1B (JARID1B) has been revealed to remove methyl residues from methylated lysine 4 on histone H3 (H3K4) and has also been reported to be associated with the progression of numerous types of tumor. However, its roles and mechanisms in pancreatic cancer (PC) remain unknown. The present study demonstrated that JARID1B is elevated in PC and is associated with the growth of pancreatic tumors. Overexpression of JARID1B significantly promoted the proliferation in vitro and tumor formation in vivo of PC cells. Furthermore, silencing the expression of JARID1B in other PC cells revealed opposite effects. Further research revealed that JARID1B exerted its function through modulation of H3K4me3 at the phosphatase and tensin homolog (PTEN) gene promoter which was associated with inactive PTEN transcription. To the best of our knowledge, the present study was the first to demonstrate that JARID1B promotes the growth of PC and that targeting JARID1B may be a useful strategy to suppress the progression of PC.

Introduction

Pancreatic cancer (PC) is one of the leading causes of cancer-associated mortality in developed and developing countries and one of the most lethal malignant neoplasms in the world (14). Based upon the estimates of GLOBOCAN 2012, >331,000 mortalities per year were caused by PC, accounting for 4% of all mortalities, and PC was considered to be the seventh leading cause of cancer-associated mortality in males and females (1). With regards to treatment, surgery is the only option with the potential to cure PC; however, as a result of the concealed pathogenesis, rapid progress and high metastatic rate, a limited number patients with PC (15–20%) are candidates for radical surgery at the time of disease diagnosis (5). Less than 30% of the patients who undergo surgery and adjuvant chemotherapy survive >5 years following treatment (5). Life expectancy is decreased by up to 98% in patients with PC compared with healthy patients (6).

The causes of PC remain unknown, but certain risk factors have been reported, including cigarette smoking, a family history of PC, excessive alcohol consumption, diabetes mellitus, obesity, dietary factors and a lack of physical exercise (710). The present study focused on the Jumonji AT-rich interactive domain 1B (JARID1B) gene, which is a member of JmjC domain-containing protein family. JARID1B specifically removes the methyl residues from tri-, di- and mono-methylated lysine 4 on histone H3 (H3K4) that are associated with gene activation (11,12). JARID1B acts as a transcriptional repressor due to its intrinsic histone demethylase activity. Trimethylation at H3K4 is an important histone modification associated with the activation of transcribed genes, including PTEN (13), and JARID1B specifically demethylates H3K4me3 (tri-methylated histone H3 at lysine 4) to a transcriptionally inactive state that will repress the activation of target genes (14).

Expression of JARID1B has been reported be elevated in a number of different types of cancer, including breast cancer, bladder cancer, lung cancer, colorectal cancer, prostate cancer and malignant melanoma and is required for the proliferation of cancer cells and tumor growth (11,1522). It has been reported that the depletion of JARID1B inhibited the proliferation of breast cancer cells and restrained tumor growth in xenografts (23) and a syngeneic mouse mammary tumor model (24). Similar results were obtained in lung, bladder and colorectal tumors (1921). To the best of our knowledge, the present study was the first to reveal that the expression of JARID1B was elevated in PC, and that this was responsible for the inhibition of cell proliferation and tumor growth. Furthermore, it was also previously revealed that JARID1B is associated with the inactivation of phosphatase and tensin homolog (PTEN) in hepatocellular carcinoma (13) which was verified in the present study. The results of the present study reveal a novel function of JARID1B in PC and may provide perspective in order to develop novel therapeutic interventions for PC.

Materials and methods

Clinical specimens and cell culture

The present study was conducted with the approval of the Institutional Ethical Review Board of the Second Affiliated Hospital of Soochow University (Suzhou, Jiangsu, China). Between June 2014 and April 2015, a total of 42 paired (25 male and 17 female, from 37 to 62 years old, mean 50.6 years old) PC specimens and adjacent tissue samples, frozen in liquid nitrogen, were obtained from the pathology laboratory at the Second Affiliated Hospital of Soochow University. None of the patients had received any antitumor treatments prior to biopsy. PC cell lines H6c7, MIA PaCa-2, AsPC-1, BxPC-3, Su86.86, UACC462 were obtained from the American Type Culture Collection (Manassas, VA, USA) and cultured in adaptive culture medium (Sigma-Aldrich; Merck KgaA, Darmstadt, Germany) according to ATCC and cultured at 37°C in 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from freshly-frozen samples or cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Total RNA was reverse transcribed at 42°C for 60 min using a First Strand cDNA Synthesis kit (Invitrogen; Thermo Fisher Scientific, Inc.). qPCR reactions were conducted using Platinum SYBR Green qPCR SuperMix-UDG reagents (Invitrogen; Thermo Fisher Scientific, Inc.) on the PRISM 7900HT system (Applied Biosystems; Thermo Fisher Scientific, Inc.) followed the following primers: JARID1B forward primer 5′-AGAGGCTGAATGAGCTGGAG-3′ and reverse primer 5′-TGGCAATTTTGGTCCATTTT-3′; GAPDH forward primer 5′-ATCACTGCCACCCAGAAGAC-3′ and reverse primer 5′-ATGAGGTCCACCACCCTGTT-3′. The thermocycling conditions were as follows: 95°C for 10 min, followed by 40 cycles of 95°C for 10 sec, 50°C for 30 sec and 72°C for 20 sec. All reactions were performed in triplicate and reactions without reverse transcriptase were used as negative controls. GAPDH was used as an endogenous control and the 2−ΔΔCq method was used to calculate the relative expression levels (25).

Western blot analysis

Western-blot assay was used to analyze the expressions of candidate proteins in indicated cell lines. Cells were cultured in 100 mm dishes and radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Haimen, China) containing protein inhibitor cocktail (cat. no. P9599; Sigma-Aldrich; Merck KGaA) was input at 70–90% density. Whole protein was extracted by centrifugation (13,000 × g) for 20 min at 4°C. And the concentration of proteins was measured by NanoDrop (NanoDrop2000; Thermo Fisher Scientific, Inc.). Samples containing 30–50 ug of protein were separated by 10% SDS-PAGE. Proteins were transferred to polyvinylidene fluoride membranes (Merck KGaA) which was blocked in 5% bovine serum albumin (Beijing Seasky Bio Technology, Co., Ltd., Beijing, China) or 5% skim milk at room temperature for 1 h. Then the membrane was probed overnight at 4°C in 5% bovine serum albumin (Beijing Seasky Bio Technology, Co., Ltd.) with primary antibodies (Cell Signaling Technology, Inc., Danvers, MA, USA) against Actin (1:2,000; cat. no. CST 3700S), anti-JARID1B (1:1,000; cat. no. CST 3273T), anti-PTEN (1:1,000; cat. no. CST 9188T), anti-protein kinase B (Akt) (1:1,000; cat. no. CST 2920S), anti-p-Akt (1:1,000; cat. no. CST 4060T), anti-P13K (1:1,000; cat. no. CST 4249T), anti-p-P13K (1:1,000; cat. no. CST 4228T), anti-p53 (1:1,000; cat. no. CST 2527T), anti-p21 (1:1,000; cat. no. CST 2947T), anti-p27 (1:1,000; cat. no. CST 3686T), anti-H3K4me3 (1:1,000; cat. no. CST 9751T), anti-K3K9me3 (1:1,000; cat. no. CST 4658T), anti-H3K27me3 (1:1,000; cat. no. CST 9728T) followed by washing in Tris-buffered saline with Tween-20 (TBST; 0.02M Tris PH 7.6, 0.8% NaCl, 01% Tween-20) and incubated in TBST containing HRP secondary antibodies (1:10,000; cat. no. ZB2301; ZSGB-BIO) for 1 h at room temperature. Following washing in TBST again, the Chemiluminescent HRP substrate (cat. no. P90720; Merck KGaA) was added, according to the manufacturer's protocol, and the fluorescence was assessed using FluorChem E (version 4.1.3; Proteinsimple, FE0444). All antibodies were purchased from Cell Signaling Technology.

Establishment of cell lines

Human JARID1B shRNA was cloned into a pSuper-puro vector (GENEWIZ, South Plainfield, NJ, USA) while JARID1B mRNA was cloned into a pBabe-puro vector (GENEWIZ). A total of 10 µg plasmid was transfected into Phoenix packaging cells by X-tremeGENE HP (Roche Applied Science, Penzberg, Germany) and retrovirus supernatants containing pSuper and pSuper-sh.JARID1B, pBabe and pBabe-JARID1B were collected and filtered. AsPC-1 was incubated in retrovirus supernatants contaning pBabe and pBabe-JARID1B, but UACC-462 was incubated in retrovirus supernatants containing pSuper and pSuper-shJARID1B 1 or 2. A total of 4 µg/ml polybrene (Sigma-Aldrich; Merck KGaA) was added to accelerate the transfection. Then AsPC-1-pBabe-JARID1B, UACC-462-pSuper-shJARID1B 1 or 2 and control cells were subsequently selected by puromycin (2 µg/ml) as previously described (26).

Proliferation assay

An MTT assay was used to detect the proliferative rate of AsPC-1-pBabe-JARID1B, UACC-462-pSuper-shJARID1B 1 or 2 and control cell lines. A total of 1,000 cells were plated into a 96-well plate and were cultured at 37°C. MTT was added at 70% density and the 96-well plate was incubated for 4 h, followed by the addition of 150 µl dimethyl sulfoxide in order to dissolve the purple formazan. The optical density values at 492 nm were measured using Multiskan 3 at 12, 24, 48 and 72 h.

In vivo tumor growth model

A total of 20 Male BALB/c nude mice (weight ~15 g) aged 4–6 weeks were purchased from the Hunan Slac Jingda Laboratory Animal Company, Ltd. (Changsha, China) and provided with sterile food and water in the specific pathogen free (SPF) laboratory animal room (22°C, 40% humidity, noise ≤60 dB, clean pass box). All animals were used in accordance with institutional guidelines and the current experiments were approved by the Animal Care and Use Ethics Committe. For the tumor growth assay, AsPC-1-pBabe-JARID1B, UACC-462-pSuper-shJARID1B 2 and control cells were resuspended in phosphate-buffered saline and 4×106 cells (100 µl) were subcutaneously injected into the axilla of the nude mice. Six weeks later, the mice were sacrificed using 1 mg/kg chloral hydrate, and the tumors were dissected and weighed. Animal handling and research protocols were approved by the Animal Care and Use Ethics Committee.

Chromatin immunoprecipitation (ChIP)-qPCR

A ChIP kit was purchased from EMD Millipore (Billerica, MA, USA) and ChIP experiments were performed as previously described (27). Immunoprecipitated DNA was analyzed on the ABI PRISM 7900HT sequence detection system. The PTEN promoter primers of −1,235 to −1,072 bp (forward primer, 5′-CGCCCAGCTCCTTTTCCC-3′; reverse primer, 5′-CTGCCGCCGATTCTTAC-3′) and 260 to 512 bp (forward primer, 5′-GCTCGCACCCAGAGCTAC-3′; reverse primer, 5′-GGAGAGAACTGAGC-3′) used for detection of promoters after ChIP are available upon request.

Statistical analysis

Statistical analysis data are presented as the mean ± standard deviation Associations between JAEID1B and PTEN, Akt, P13K, p53, p21 and p27 expression in PC tissues was assessed using Spearman's rank correlation. Comparisons between different groups were undertaken using one-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference. Statistical analysis was performed using SPSS 11.0 software for Windows (SPSS, Inc., Chicago, IL, USA).

Results

JARID1B is highly expressed in PC

In order to investigate whether JARID1B was associated with the progression of PC, the expression levels of JARID1B in 42 patient PC samples RT-qPCR. As demonstrated in Fig. 1A and B, JARID1B was significantly overexpressed in 42 PC tissues compared with the normal adjacent tissues. To further confirm the elevated expression of JARID1B in PC, the expression levels of JARID1B in distinct PC cell lines were determined. The results revealed that JARID1B was significantly overexpressed in PC cell lines including MIA PaCa-2, AsPC-1, BxPC-3, Su86.86 and UACC-462 compared with expression in the control cell line H6c7 (Fig. 1C). Furthermore, the expression levels of JARID1B in these cell lines described above were verified using western blot analysis (Fig. 1D). The results revealed that JARID1B was overexpressed in PC.

JARID1B promotes the proliferative capacity of PC cells in vitro and in vivo

In order to further identify the role of JARID1B in PC cells, the pancreatic cell lines exhibiting overexpression or silencing of JARID1B were established using a lentiviral vector and the expression levels of JARID1B were examined by western blot analysis and RT-qPCR. As demonstrated in Fig. 2A, the expression levels of JARID1B in UACC-462 cells with three different shRNAs were examined by western blot analysis and JARID1B shRNA2 was the most effective. Similar results were observed in RT-qPCR (Fig. 2B). Overexpression of JARID1B in the AsPC-1 cell line was observed in western blot analysis (Fig. 2C) and RT-qPCR (Fig. 2D).

Based on the established cell lines, the effect of JARID1B silencing on the proliferation of PC cancer cells was subsequently determined using an MTT assay. The results revealed that silencing of JARID1B significantly inhibited the proliferation of UACC-462 cells (Fig. 3A), while overexpression of JARID1B promoted the proliferation of AsPC-1 cells (Fig. 3B).

In order to further confirm the effect of JARID1B on the proliferation of PC cells, xenografts were used to reveal whether JARID1B affected the proliferation rate of pancreatic tumors. AsPC-1-pBabe-JARID1B, UACC-462-pSuper-shJARID1B 2 and control cells were injected subcutaneously into the axilla of nude mice. As demonstrated in Fig. 4, silencing of JARID1B significantly inhibited the proliferation of pancreatic tumors formed in nude mice (Fig. 4A), which were smaller in size (Fig. 4B) and lighter in weight (Fig. 4C). However, overexpression of JARID1B significantly promoted the proliferation of pancreatic tumors formed by the injection of AsPC-1 cells (Fig. 5A), in terms of tumor size (Fig. 5B) and weight (Fig. 5C). The aforementioned results demonstrated that JARID1B may promote the proliferation of PC cells in vitro and tumorigenesis in vivo.

JARID1B regulates PTEN expression through the demethylation of H3K4me3

It has been demonstrated that JARID1B may affect the demethylation of H3K4me3 and that the activation of PTEN is regulated by H3K4me3 (13). In order to determine whether JARID1B regulates the tumorigenesis of PC through the PTEN/Akt signaling pathway, the expression levels of proteins participating in the PTEN/Akt signaling pathway were examined. As demonstrated in Fig. 6, silencing of JARID1B significantly elevated the expression of PTEN in UACC-462 cells (Fig. 6A), while overexpression of JARID1B inhibited the expression of PTEN in AsPC-1 cells (Fig. 6B). It has been reported that PTEN, as well as p53, p27 and p21, is able to regulate the phosphoinositide 3-kinase (PI3K)/Akt pathway in a number of types of tumor cell (28,29). Therefore, whether or not JARID1B-mediated inhibition of PTEN may affect the expression of the aforementioned proteins was subsequently investigated. Notably, silencing of JARID1B followed by ectopic expression of PTEN significantly inhibited the level of phosphorylated PI3K and Akt but increased the level of p53, p27 and p21 (Fig. 6A), while overexpression of JARID1B induced the opposite effect (Fig. 6B). In further research, silencing of JARID1B significantly increased the mRNA level of PTEN and p53 (Fig. 6C), while overexpression of JARID1B decreased the mRNA level of PTEN and p53 (Fig. 6D). Western blot analysis and RT-qPCR revealed that JARID1B did not affect the level of P13K and Akt (Fig. 6A-D). These data revealed that JARID1B may inhibit the transcription of PTEN and p53. In order to further confirm the correlation between JARID1B and PTEN or p53, Spearman's rank correlation analysis was performed. As demonstrated in Fig. 7, the mRNA level of JARID1B was inversely correlated with the mRNA levels of PTEN (Fig. 7A) and p53 (Fig. 7D), while no association was observed between JARID1B and P13K, Akt, p27 or p21 (Fig. 7B, C, E and F). Based upon these aforementioned data, JARID1B inhibited the expression of PTEN and p53 at the RNA level.

Subsequently, the manner in which JARID1B inhibited the expression of PTEN was investigated. It has previously been demonstrated that JARID1B regulates the methylation of H3K4 (11). To begin with, the expression levels of H3K4me3 with silencing or overexpression of JARID1B were investigated. Silencing of JARID1B significantly increased the level of H3K4me3 (Fig. 8A), while overexpression of JARID1B decreased the level of H3K4me3 (Fig. 8B). However, silencing and overexpression of JARID1B did not affect the levels of H3K9me3 or H3K27me3 (Fig. 8A and B). Due to the fact that H3K4me3 is associated with the active transcription of a number of genes, including PTEN, whether or not JARID1B expression was associated with the H3K4me3 modification at the promoter of PTEN in PC cells was investigated. Quantitative chromatin immunoprecipitation (qChIP) assays were performed in AsPC-1-pBabe-JARID1B, UACC-462-pSuper-shJARID1B and their corresponding control cells. Silencing of JARID1B was revealed to be associated with increased H3K4me3 levels at the region −1,235 to −1,072 bp (Fig. 8C1) and 260 to 512 bp (Fig. 8C2) in the PTEN gene regions (Fig. 8D). Furthermore, less occupancy of those PTEN gene regions by H3K4me3 was detected in AsPC-1 cells with overexpression of JARID1B (Fig. 8E). Taken together, these results indicated that JARID1B induced inactive transcription of PTEN due to decreased levels of H3K4me3 in the PTEN gene.

Discussion

PC has been reported to be the most lethal malignant neoplasm, with a difficult diagnosis and a high rate of relapse. The results of the present study demonstrated that JARID1B was aberrantly expressed in patients with PC and that elevated JARID1B significantly promoted the cell and tumor proliferation of PC, while silencing of JARID1B had the opposite effect. With regards to the mechanism of JARID1B-promoted cell and tumor proliferation, it was revealed that PTEN serves an essential role in these processes. JARID1B may regulate the methylation of H3K4 in order to affect the activation of PTEN, which ultimately promotes the progression of PC. Therefore, JARID1B may be a novel target for the diagnosis of PC.

In recent years, a number of novel insights into the pathogenesis of PC have been reported. Extracellular signal-regulated kinase promoted the degradation of FBXW7 through phosphorylation, which resulted in the inhibition of the tumor suppressor function of FBXW7 and eventually promoted the occurrence of PC (30). Ataxia telangiectasia group D complementing gene (gene-ATDC), which serves an essential role from the early development of invasive cancer to metastatic cancer, was also reported to be associated with 90% of the proliferation of PC (31). Patients with high gene-ATDC levels often experience very early metastasis, resulting in a <30% survival rate in patients with early PC (32). Another study reported that >95% of the patients with PC exhibited KRAS mutations (32), while a different study revealed that miRNA-21 serves a key role in the drug resistance of PC (33). These data may aid doctors in better understanding the pathogenesis of PC. The present study revealed the novel role of JARID1B in the proliferation of PC, which may further improve the pathogenesis of PC and may serve as a novel therapeutic target for PC.

Overexpression of JARID1B occurs in a wide variety of cancer types, but the function of JARID1B is not fully understood (1522). It was recently observed that the demethylation of H3K4 regulated by JARID1B contributed to the silencing of retinoblastoma target genes in senescent cells, presumably through closing the chromatin in which the silencing of retinoblastoma trigger genes was involved (34). In esophageal cancer, JARID1B promoted cell proliferation and tumor growth following an effect of JARID1B on the activation of PTEN (35). The association between JARID1B and PTEN has also been demonstrated in previous studies (13,36,37). The present study also revealed that JARID1B promoted the cell proliferation and tumor growth of PC by regulating the activation of the PTEN gene through demethylation of H3K4me3, which further confirmed the regulatory association between JARID1B and PTEN.

In recent years, good diagnostic markers, drug targets and therapeutic strategies remain insufficient for the successful treatment of PC. The results of the present study identified the role and mechanism of JARID1B in the cell proliferation and tumor growth of PC, which may contribute toward the diagnosis and therapeutic strategy of PC patients.

Acknowledgements

Not applicable.

Funding

No funding received.

Availability of data and materials

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

Authors' Contributions

PD was responsible for the conception and design of the present study. XS, GC and PD developed the methodology. XS, GC, LX, WW and ZC undertook the acquisition of data. XS, GC, LX, WW and ZC were responsible for the analysis and interpretation of data. XS and GC wrote and undertook any manuscript revisions and PD was responsible for study supervision and any administrative, technical, or material support.

Ethics approval and consent to participate

The present study was conducted with the approval of the Institutional Ethical Review Board of the Second Affiliated Hospital of Soochow University (Suzhou, Jiangsu, China).

Consent to publish

The study participants provided their consent for the publication of this data.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, Real FX, Van Laethem JL and Heinemann V: Addressing the challenges of pancreatic cancer: Future directions for improving outcomes. Pancreatology. 15:8–18. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA, Silverman DT, Ji BT, Gallinger S, Holly EA, et al: Pancreatitis and pancreatic cancer risk: A pooled analysis in the international pancreatic cancer Case-control consortium (PanC4). Ann Oncol. 23:2964–2970. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Tumas J, Kvederaviciute K, Petrulionis M, Kurlinkus B, Rimkus A, Sakalauskaite G, Cicenas J and Sileikis A: Metabolomics in pancreatic cancer biomarkers research. Med Oncol. 33:1332016. View Article : Google Scholar : PubMed/NCBI

6 

Carrato A, Falcone A, Ducreux M, Valle JW, Parnaby A, Djazouli K, Alnwick-Allu K, Hutchings A, Palaska C and Parthenaki I: A systematic review of the burden of pancreatic cancer in europe: Real-world impact on survival, quality of life and costs. J Gastrointest Cancer. 46:201–211. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Peto R: The fraction of cancer attributable to lifestyle and environmental factors in the UK in 2010. Br J Cancer. 105 Suppl 2:S12011. View Article : Google Scholar : PubMed/NCBI

8 

Hidalgo M: Pancreatic cancer. N Engl J Med. 362:1605–1617. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Ezzati M, Henley SJ, Lopez AD and Thun MJ: Role of smoking in global and regional cancer epidemiology: Current patterns and data needs. Int J Cancer. 116:963–971. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Willett WC: Diet and cancer. Oncologist. 5:393–404. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, Ma Y, Yu Y, Lin H, Chen AP and Chen CD: JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc Natl Acad Sci USA. 104:19226–19231. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Scibetta AG, Santangelo S, Coleman J, Hall D, Chaplin T, Copier J, Catchpole S, Burchell J and Taylor-Papadimitriou J: Functional analysis of the transcription repressor PLU-1/JARID1B. Mol Cell Biol. 27:7220–7235. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Tang B, Qi G, Tang F, Yuan S, Wang Z, Liang X, Li B, Yu S, Liu J, Huang Q, et al: JARID1B promotes metastasis and epithelial-mesenchymal transition via PTEN/AKT signaling in hepatocellular carcinoma cells. Oncotarget. 6:12723–12739. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Benevolenskaya EV: Histone H3K4 demethylases are essential in development and differentiation. Biochem Cell Biol. 85:435–443. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Barrett A, Madsen B, Copier J, Lu PJ, Cooper L, Scibetta AG, Burchell J and Taylor-Papadimitriou J: PLU-1 nuclear protein, which is upregulated in breast cancer, shows restricted expression in normal human adult tissues: A new cancer/testis antigen? Int J Cancer. 101:581–588. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Lu PJ, Sundquist K, Baeckstrom D, Poulsom R, Hanby A, Meier-Ewert S, Jones T, Mitchell M, Pitha-Rowe P, Freemont P and Taylor-Papadimitriou J: A novel gene (PLU-1) containing highly conserved putative DNA/chromatin binding motifs is specifically up-regulated in breast cancer. J Biol Chem. 274:15633–15645. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Y, Tsunoda T, Field HI, Kelly JD, Neal DE, Yamaue H, et al: Overexpression of the JmjC histone demethylase KDM5B in human carcinogenesis: Involvement in the proliferation of cancer cells through the E2F/RB pathway. Mol Cancer. 9:592010. View Article : Google Scholar : PubMed/NCBI

19 

Li X, Su Y, Pan J, Zhou Z, Song B, Xiong E and Chen Z: Connexin 26 is down-regulated by KDM5B in the progression of bladder cancer. Int J Mol Sci. 14:7866–7879. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Shen X, Zhuang Z, Zhang Y, Chen Z, Shen L, Pu W, Chen L and Xu Z: JARID1B modulates lung cancer cell proliferation and invasion by regulating p53 expression. Tumour Biol. 36:7133–7142. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Ohta K, Haraguchi N, Kano Y, Kagawa Y, Konno M, Nishikawa S, Hamabe A, Hasegawa S, Ogawa H, Fukusumi T, et al: Depletion of JARID1B induces cellular senescence in human colorectal cancer. Int J Oncol. 42:1212–1218. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Roesch A, Vultur A, Bogeski I, Wang H, Zimmermann KM, Speicher D, Körbel C, Laschke MW, Gimotty PA, Philipp SE, et al: Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell. 23:811–825. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Catchpole S, Spencer-Dene B, Hall D, Santangelo S, Rosewell I, Guenatri M, Beatson R, Scibetta AG, Burchell JM and Taylor-Papadimitriou J: PLU-1/JARID1B/KDM5B is required for embryonic survival and contributes to cell proliferation in the mammary gland and in ER+ breast cancer cells. Int J Oncol. 38:1267–1277. 2011.PubMed/NCBI

24 

Yamane K, Tateishi K, Klose RJ, Fang J, Fabrizio LA, Erdjument-Bromage H, Taylor-Papadimitriou J, Tempst P and Zhang Y: PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol Cell. 25:801–812. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Wen M, Kwon Y, Wang Y, Mao JH and Wei G: Elevated expression of UBE2T exhibits oncogenic properties in human prostate cancer. Oncotarget. 6:25226–25239. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Wang Y, Wen M, Kwon Y, Xu Y, Liu Y, Zhang P, He X, Wang Q, Huang Y, Jen KY, et al: CUL4A induces epithelial-mesenchymal transition and promotes cancer metastasis by regulating ZEB1 expression. Cancer Res. 74:520–531. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Wang H, Quah SY, Dong JM, Manser E, Tang JP and Zeng Q: PRL-3 down-regulates PTEN expression and signals through PI3K to promote epithelial-mesenchymal transition. Cancer Res. 67:2922–2926. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Kim SR, Lee KS, Park SJ, Min KH, Lee KY, Choe YH, Lee YR, Kim JS, Hong SJ and Lee YC: PTEN down-regulates IL-17 expression in a murine model of toluene diisocyanate-induced airway disease. J Immunol. 179:6820–6829. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Ji S, Qin Y, Shi S, Liu X, Hu H, Zhou H, Gao J, Zhang B, Xu W, Liu J, et al: ERK kinase phosphorylates and destabilizes the tumor suppressor FBW7 in pancreatic cancer. Cell Res. 25:561–573. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Wang L, Yang H, Abel EV, Ney GM, Palmbos PL, Bednar F, Zhang Y, Leflein J, Waghray M, Owens S, et al: ATDC induces an invasive switch in KRAS-induced pancreatic tumorigenesis. Genes Dev. 29:171–183. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Liou GY, Döppler H, Necela B, Edenfield B, Zhang L, Dawson DW and Storz P: Mutant KRAS-induced expression of ICAM-1 in pancreatic acinar cells causes attraction of macrophages to expedite the formation of precancerous lesions. Cancer Discov. 5:52–63. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Ma Y, Wu Q, Li X, Gu X, Xu J and Yang J: Pancreatic cancer: From bench to bedside. Ann Transl Med. 4:4582016. View Article : Google Scholar : PubMed/NCBI

34 

Chicas A, Kapoor A, Wang X, Aksoy O, Evertts AG, Zhang MQ, Garcia BA, Bernstein E and Lowe SW: H3K4 demethylation by Jarid1a and Jarid1b contributes to retinoblastoma-mediated gene silencing during cellular senescence. Proc Natl Acad Sci USA. 109:8971–8976. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Kano Y, Konno M, Ohta K, Haraguchi N, Nishikawa S, Kagawa Y, Hamabe A, Hasegawa S, Ogawa H, Fukusumi T, et al: Jumonji/Arid1b (Jarid1b) protein modulates human esophageal cancer cell growth. Mol Clin Oncol. 1:753–757. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Lu W, Liu S, Li B, Xie Y, Adhiambo C, Yang Q, Ballard BR, Nakayama KI, Matusik RJ and Chen Z: SKP2 inactivation suppresses prostate tumorigenesis by mediating JARID1B ubiquitination. Oncotarget. 6:771–788. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Yuan P, Ito K, Perez-Lorenzo R, Del Guzzo C, Lee JH, Shen CH, Bosenberg MW, McMahon M, Cantley LC and Zheng B: Phenformin enhances the therapeutic benefit of BRAF(V600E) inhibition in melanoma. Proc Natl Acad Sci USA. 110:18226–18231. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 16 Issue 1

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
Shen X, Cheng G, Xu L, Wu W, Chen Z and Du P: Jumonji AT‑rich interactive domain 1B promotes the growth of pancreatic tumors via the phosphatase and tensin homolog/protein kinase B signaling pathway Retraction in /10.3892/ol.2023.13660. Oncol Lett 16: 267-275, 2018
APA
Shen, X., Cheng, G., Xu, L., Wu, W., Chen, Z., & Du, P. (2018). Jumonji AT‑rich interactive domain 1B promotes the growth of pancreatic tumors via the phosphatase and tensin homolog/protein kinase B signaling pathway Retraction in /10.3892/ol.2023.13660. Oncology Letters, 16, 267-275. https://doi.org/10.3892/ol.2018.8618
MLA
Shen, X., Cheng, G., Xu, L., Wu, W., Chen, Z., Du, P."Jumonji AT‑rich interactive domain 1B promotes the growth of pancreatic tumors via the phosphatase and tensin homolog/protein kinase B signaling pathway Retraction in /10.3892/ol.2023.13660". Oncology Letters 16.1 (2018): 267-275.
Chicago
Shen, X., Cheng, G., Xu, L., Wu, W., Chen, Z., Du, P."Jumonji AT‑rich interactive domain 1B promotes the growth of pancreatic tumors via the phosphatase and tensin homolog/protein kinase B signaling pathway Retraction in /10.3892/ol.2023.13660". Oncology Letters 16, no. 1 (2018): 267-275. https://doi.org/10.3892/ol.2018.8618