Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

RETRACTED ARTICLE: K-ras-ERK1/2 down-regulates H2A.XY142ph through WSTF to promote the progress of gastric cancer

verfasst von: Chao Dong, Jing Sun, Sha Ma, Guoying Zhang

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

Histone H2AX phosphorylation at the site of Tyr-142 can participates in multiple biological progressions, which is including DNA repair. Ras pathway is closely involved in human cancers. Our study investigated the effects of Ras pathway via regulating H2AX.Y142ph.

Methods

Gastric cancer cell line SNU-16 and MKN1 cells were transfected with Ras for G12D and T35S site mutation. The phosphorylation of H2A.XY142 and ERK1/2, WSTF and MDM2 was detected by western blot. Cell viability, cell colonies and migration was analyzed by MTT assay, soft-agar colony formation assay, and Transwell assay, respectively. The expression of Ras pathway related downstream factors, EYA3 and WSTF was detected by qRT-PCR. The relationship between Ras and downstream factors were detected by ChIP. The cell cycle progression was measured by flow cytometry.

Results

RasG12D/T35V transection decreased the phosphorylation of H2A.XY142 and activated phosphorylation of ERK-1/2. H2A.XY142 inhibited cell viability, colonies and migration. H2A.XY142ph altered the expression of Ras downstream factors. CHIP assay revealed that RasG12D/T35V could bind to the promoters of these Ras pathway downstream factors. Silence of EYA3 increased H2A.XY142ph and inhibited cell viability, migration and percent cells in S stage. Furthermore, silence of EYA3 also changed the downstream factors expression. WSTF and H2A.XY142ph revealed the similar trend and MDM2 on the opposite.

Conclusion

Ras/ERK signal pathway decreased H2A.XY142ph and promoted cell growth and metastasis. This Ras regulation process was down-regulated by the cascade of MDM2-WSTF-EYA3 to decrease H2A.XY142ph in SNU-16 cells.
Hinweise
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s12885-022-10052-1.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12885-022-10052-1"
Abkürzungen
ChIP
Chromatin immunoprecipitation
ERK
Extracellular regulated protein kinases
FBS
Fetal bovine serum
GC
Ras-like (Ral), Gastric cancer
HRP
Horseradish peroxidase
PI3K
Phosphatidylinositol 3′-kinase
RT-PCR
Reverse transcription polymerase chain reaction
RUNX3
Runt-related transcription factor 3
WSTF
Williams-Beuren syndrome transcription factor

Highlights

1.
H2A.XY142ph is down-regulated by K-Ras-ERK1/2 in SNU-16 cells;
 
2.
H2A.XY142ph restrains SNU-16 cell growth;
 
3.
H2A.XY142ph down-regulates Ras downstream factors;
 
4.
Silence of EYA3 up-regulates H2A.XY142ph;
 
5.
Ras-ERK1/2 induces WSTF degradation to down-regulate H2A.XY142ph;
 
6.
Ras-ERK1/2 down-regulates WSTF via MDM2.
 

Backgroud

Increasing evidence suggests that abnormal Ras pathway is closely related with the progress of human cancer, but the exact epigenetic regulation mechanism is not clear [1, 2]. K-RasG12 is an oncogenic gene which is widely observed in human cancers [3]. In addition, the main downstream factors of Ras signaling included extracellular regulated protein kinases (ERK) 1/2, phosphatidylinositol 3′-kinase (PI3K), and Ras-like (Ral) 2 guanine nucleotide exchange factors (RalGEFs) [46]. However, the detailed information and the underlying mechanisms how Ras signal pathways involved are still not well understood and studied.
It is well-known that eukaryotic DNA is wrapped by histone octamers which was made of four different kinds of histones, H2A, H2B, H3 and H4. Importantly, the post-transcriptionally modification of histone N-terminal tail can regulate chromatin organization and DNA utilization processes, including transcription [7]. With the development of science and technology, the deciphering of histone code and its biological functions has received increasing attention. A substantial amount of studies point out that histone modification is subjected to a wide range of tumor [8]. For example, a study from Yang et al. found that histone modification is involved in regulating tumorigenesis of gastric cancer (GC) [9]. The silencing or removing H2A.X, a histone variant, was involved in cellular DNA repair and robust growth [10]. Interestingly, the roles of histone modification received considerable attention in GC [11]. For example, hypoxia silences runt-related transcription factor 3 (RUNX3) by epigenetic histone modification in the progression of GC [12]. Histone deacetylases expression is an independent prognostic marker in GC [13]. These finding sindicated that histone modification exert paramount important role in GC.
H2A.X is the damage-related histone variant, which is identified by the C-terminal tyrosyl residue, Tyr-142. The reason for that is Tyr-142 could be phosphorylated by an atypical kinase, Williams-Beuren syndrome transcription factor (WSTF) and inducing phosphorylation of H2A.X into H2A.XY142ph [14, 15]. High level of Tyr142 phosphorylation regulates several biological progressions, including apoptosis and DNA repair [14]. Meanwhile, the effects of dephosphorylation for the eyes absent homolog (EYA) 1/3 also address novel insight into this process [16]. Therefore, we investigated the role of Ras-ERK pathway in cell viability, colonies and migration via the regulation of H2A.XY142ph. Finally, we investigated the underlying mechanisms in gastric cancer cell line SNU-16 and MKN1 cells.

Material and methods

Cell culture

Human GC cell line SNU-16 and MKN1 cells were purchased from Shanghai Institute for Biological Science (Shanghai, China). Cells were maintained at 37 °C, 5% CO2 in RPMI-1640 (Gibco Laborato-ties, Grand Island, NY) with 100 units/ml penicillin, 100 μg/ml streptomycin and 10% fetal bovine serum (FBS, Life Science, UT, USA).

Plasmid construction and siRNA

Empty-pEGFP-N1 vector, pEGFP-K-RasWT, pEGFP-K-RasG12V/T35S plasmids were transfected in cells. The transfection with specific gene with HA-tag was used for screening out the target factor through western blot. pEGFP-K-RasG12V/T35S plasmids were obtained by site-directed mutagenesis. SiRNAs (Shanghai GenePharma, Shanghai, China) refers to using interference RNA to silence the goal RNA (Mouse double minute 2 homolog (MDM2) or EYA3). The pEGFP-H2A.XY142A construct was constructed using the TaKaRa MutanBEST Kit (#D401) (TaKaRa, Shiga, Japan) as recommend by the manufacturer.

Transfection

Cells at the density of 5 × 105 per well were cultured in 6-well plates for 12 h in the darkness. Then the cells were transfected with plasmids or siRNA using Lipofectamine 2000 (Invitrogen, Carlsbad, CA). Then qRT-PCR and western blot were used to determine the transfection efficiency after 48 h of transfection.

Cell viability

MTT (Sigma-Aldrich, St Louis, MO, USA) was used for the detecting cell viability. After cells were cultured for 48 h, 20 μl 5 mg/mL MTT was administrated to each well. Cells were cultured for 4 h. Afterward, we used 100 μl dimethyl sulfoxide (Sigma-Aldrich, St Louis, MO, USA) to lyse formazan crystal. The value was obtained at 570 nm by a multiwell spectrophotometer (Emax; Molecular Devices, Sunnyvale, CA).

Reverse transcription polymerase chain reaction (RT-PCR)

RT-PCR method was referred to what described in the report [17]. Total RNA was obtained from SNU-16 cells using TRIzol (Invitrogen) reagent. DNase-I-treated total RNA was supplied for first-strand cDNA synthesis by M-MuLV reverse transcriptase (Fermentas, York, UK) and oligo-dT primers (Invitrogen). QuantiTect SYBR Green PCR Kit (Qiagen, Hilden, Germany) was used to amplify the target sequence. GAPDH was an internal control for detecting RNA expression based on triplicate experiments.

Soft-agar colony formation assay

Soft-agar assay was performed to measure the cell colonies ability [18]. The cells suspended in full culture medium with 0.35% low-melting agarose, then cells were transferred into solidified 0.6% agarose in six-well culture plates (1 × 103 cells/well). The number of the colonies was counted 3 weeks later using microscopically (40 ×).

Transwell migration assay

Cell migration was evaluated by using a modified two-chamber migration Transwell (Corning Costa, NY, US) with a pore size of 8 μm. 100 μl (around 2 × 105 cells/ml) cell suspension without serum was added to upper Transwell. 600 μl complete medium was added in the lower compartment. Cells were maintained for 24 h at 37 °C, 5% CO2. After incubation, cells at the upper surface of the filter were removed by a cotton swab, and the filter was fixed with methanol for 5 min. Cells at the lower surface of the filter were stained by 0.1% Giemsa (Sigma-Aldrich) for 15 min. Cells were counted by 100 × microscope.

Western blot analysis

Protein was obtained using RIPA lysis buffer (Cat. No:R0010, Solarbio, Beijing, China) supplemented with protease inhibitors (Thermo Fisher Scientific, Rockford, IL). The BCA™ Protein Assay Kit (Pierce, Appleton, WI, USA) was used for determining protein concentrations. Western blot system was established using a Bio-Rad Bis-Tris Gel system following the manufacturer’s instructions. Primary antibodies were prepared in 5% blocking buffer and diluted according to the product instruction. These primary antibodies were incubated in membrane and maintained at 4 °C overnight at recommended concentration. Then secondary antibody incubated with horseradish peroxidase (HRP) conjugated secondary antibody. Captured the signals, and Image Lab™ Software (Bio-Rad, Shanghai, China) quantified the intensity of the bands.

Flow cytometric analysis of cell cycle distribution

SNU-16 cells were cultured until reach 75–80% confluence, and then cells were washed by PBS to remove the non-adherent cells. Collected cells were all adherent cells and fixed with cold 70% ethanol. Cells were washed with PBS again. After that, cells were stained with 4, 6-diamidino-2-phenylindole (DAPI) (Partec, Germany) and cultured in the darkness for 30 min. Flow cytometry was used for detecting cell cycle distribution. The percentage of cells in different cell cycle stages was calculated.

Chromatin immunoprecipitation (ChIP)

Cells were fixed in 1% formaldehyde, then lysed, and sonicated cells. Then 5 mg chromatin was immunoprecipitated with Dynabeads. After that, purified DNA was used for PCR amplification at the CYR61, IGFBP3, WNT16B, NT5E, GDF15, CARD16 promoter. The detailed process could refer to the literature [19].

Statistical analysis

Data was analyzed by Graphpad 6.0 statistical software (GraphPad, San Diego, CA, USA). Data were present as mean + SD. The statistical analyses were performed using the Student’s t-test or one way ANOVA followed by Duncan post-hoc of multiple comparisons. A P value of < 0.05 was considered significant (* P < 0.05, ** P < 0.01 or ***P < 0.001).

Results

H2A.XY142ph was down-regulated by Ras-ERK1/2 pathway

Ras/ERK signal pathway was often found to be closely related with GC [20]. In our study, SNU-16 and MKN1 cells were respectivly transfected with empty-pEGFP-N1 vector, pEGFP-K-RasWT and pEGFP-K-RasG12V/T35S plasmids. Results showed that RasG12V/T35S decreased the expression of H2A.XY142ph level (P < 0.01, Fig. 1a). In addition, we measured the effects of RasG12V/T35S activated phosphorylation of ERK1/2 (Fig. 1b). To confirm this suggestion, another cell line MKN1 was used and similar results were also observed in this cell line as what we describe in SNU-16 cells (P < 0.01, Fig. 1c-d), which suggested that RasG12V/T35S played a role as a switch for the phosphorylation of ERK.

H2A.XY142ph restrained Ras pathway on GC cell phenotype

Histone modification influenced cell growth and cell metastasis [21]. In the current study, we constructed H2A.XY142A plasmids to mimic the situation of the phosphorylation of H2A.XY142. The mimicked H2A.XY142A plasmids were co-transfected with RasG12V/T35S plasmids into SNU-16 cells and MKN1 cells. Hence, we examined the effects of phosphorylation of histone H2A in GC cell viability, cell colony ability and cell migration. Firstly, SNU-16 cell was transfected with H2A.XY142A.With the increasing concentration, the expression of phosphorylation of H2A.XY142 was inhibited in a dose-dependent manner (Fig. 2a). Moreover, results showed that Ras/ERK pathway significantly increased cell viability (P < 0.001) while H2A.XY142A decreased cell viability to some extent in SNU-16 cells (P < 0.001, Fig. 2b). This result suggested that Ras/ERK has the ability to increase cell viability while H2A.XY142A decreased cell viability, which indicating H2A.XY142A suppressed cell growth in GC. In addition, the number of colonies (P < 0.01, Fig. 2c) and cell migration (P < 0.001, Fig. 2d) revealed the similar trend by RasG12V/T35S pathway in SNU-16 cells. Similar results were also observed in MKN1 cell line (P < 0.05 or P < 0.01, Fig. 2e-h). Taken together, we inferred that the phosphorylation of H2A.XY142 was involved in the progression of GC cells.

H2A.XY142ph down-regulated the downstream factors of Ras pathway

Ras/ERK pathway was a complex and exact regulation pathway which was modulated by diverse downstream factors [22]. We explored the expression of these important downstream factors CYR61 [23], IGFBP3 [24], WNT16B [25], NT5E [26], GDF15 [27], CARD16 [28], which are involved in the tumor cell growth and cell metastasis. Then, we found that RasG12V/T35S increased the expression of WNT16B (P < 0.05), NT5E (P < 0.01) while decreased the expression of IGFBP3 (P < 0.01), GDF15 (P < 0.01) and CARD16 (P < 0.01). Meanwhile, we found that co-transfection with RasG12V/T35S and H2A.XY142A decreased the expression of CYR61 (P < 0.05), IGFBP (P < 0.001), WNT16B (P < 0.05) and increased the expression of NT5E (P < 0.05), GDF15 (P < 0.05) and CARD16 (P < 0.05) as relative to RasG12V/T35S group, which suggested that H2A.XY142A was down-regulated these downstream genes of RasG12V/T35S pathway. In addition, the ChIP assay results showed RasG12V/T35S decreased expression of these genes (P < 0.05, P < 0.01 or P < 0.001, Fig. 3b), this suggested that H2A.XY142ph could down-regulate gene expression via directly binding to these gene’s promoters. In conclusion, H2A.XY142ph could down-regulate the downstream transcription progression of the downstream factors.

Knockdown of EYA3 up-regulated H2A.XY142ph expression and recovered the GC cell phenotype

Previous study reported that the effects of dephosphorylation of histone H2A by the EYA1/3 also address significant influence in human cancers [16]. Thereafter, knock-down EYA3 expression to investigate whether upregulation of phosphorylation of H2A.XY142 could modulate cell phenotype was performed. qRT-PCR and western blot was used to determine the transfection efficiency. We used two interference miRNAs to silence the expression of EYA3, which named si-EYA3–1 and EYA3–2, respectively. Result in Fig. 4a showed that si-EYA3–1 and si-EYA3–2 both decreased EYA3 expression in mRNA level and adding si-EYA3–1 and si-EYA3–2 both up-regulated the phosphorylation of H2A.XY142. After that, we detected the effects of si-EYA3–1/2 on cell viability, cell migration, cell cycle stage and cell relative mRNA levels. Results showed that si-EYA3–1/2 decreased cell viability (P < 0.05), cell migration (P < 0.05) and cell percent in S stage as compared with RasG12V/T35S alone (Fig. 4b-d). Furthermore, EYA3–1/2 also altered the expression of the downstream factors of RasG12D/T35V pathway compared with RasG12V/T35S alone in GC cells (P < 0.05, P < 0.01 or P < 0.001, Fig. 4e), which suggesting that EYA3 also participated in the regulation of the downstream factors of RasG12V/T35S pathway.

Ras-ERK1/2 induced WSTF degradation to decrease H2A.XY142ph expression

WSTF makes mutant H2A.X phosphorylated in Tyr 142, and the activity of WSTF exert indispensable functions in regulating multiple events [29]. Further experiments were performed to explore the exact mechanism how WSTF affects the phosphorylation of H2A.XY142. Result from Fig. 5a showed that no obviously difference was observed in the expression of WSTF and EYA3 in the group pEGFP-N1 and RasG12V/T35S. This suggested that WSTF was not played part in the transcription level. Afterwards, we found that the expression of EYA3 in protein level was no difference (Fig. 5b) while WSTF was significantly decreased (Fig. 5c) in the group RasG12V/T35S. Combined the result of Fig. 5b and c, we inferred that the effects of WSTF induced by RasG12V/T35S was in translation level. Interestingly, the result in Fig. 5d confirmed our inference, WSTF and H2A.XY142ph revealed the same trend reacted in group RasG12V/T35S. Further result showed that RasG12V/T35S could bind to the promoters of genes to reduce the input levels of WSTF in downstream factors of RasG12V/T35S pathway (Fig. 5e). Moreover, the proteasome inhibitor MG132 administration made the changes of WSTF expression by RasG12V/T35S disappeared, which indicated high inhibition for WSTF (Fig. 5f). Afterwards, we found that without MG132 administration, the phosphorylation level of H2A.XY142ph was decreased with the delaying of the transfection time (24, 48, 51, 54 and 60 h) in SNU-16 cells (Fig. 5g). Similar, under the same treatment, we found that the accumulated levels of WSTF was decreasing with the increasing of the transfection time (Fig. 5h). On the opposite, with the MG132 supplement, the decreased phosphorylation level of H2A.XY142ph by RasG12V/T35S was vanished. Instead the phosphorylation level of H2A.XY142ph was increased after administration of MG132 in different time treatment (0, 3, 6 and 12 h) (Fig. 5i). Taken together, these findings indicated that RasG12V/T35S pathway affected the phosphorylation of H2A.XY142ph was through WSTF.

Ras-ERK1/2 down-regulated WSTF via MDM2

It is well validated that histone modification was mediated by MDM2 in various cells [30]. Further experiments were performed to explore the mechanism about whether MDM2 was involved in the regulation of WSTF on H2A.XY142ph. We co-transfected RasG12V/T35S and WSTF with tagged HA into cells. The result of western blot showed that WSTF expression was decreased with the increasing of MDM2 (Fig. 6a). Further result showed without transfecting WSTF, the expression of WSTF was still decreased with the increasing of MDM2, which indicated that there might be a negative relationship between MDM2 and WSTF (Fig. 6b). Furthermore, we co-transfected RasG12V/T35S and WSTF with tag HA, the expression of WSTF was inhibited when transfected MDM2-His while WSTF expression was enhanced when transfected MDM2-MU (Fig. 6c), which strongly suggested that there was closely negative relationship between MDM2 and WSTF expression. Moreover, As shown in Fig. 6d, RasG12V/T35S and detected the expression of WSTF got the similar result as Fig. 6c, which confirmed the strong negative association between MDM2 and WSTF. We thereafter determined the relationship between MDM2 and H2A.XY142 ph and result in Fig. 6e. The result showed that RasG12V/T35S inhibited H2AX.Y142ph while up-regulated MDM2 expression. Then si-MDM2 to silence MDM2 (Fig. 6f) and we found that si-MDM2 could enhance the expression of H2A.XY142ph, which confirmed the result in Fig. 6e. Then through these experiments, we concluded the cascade reaction might be RasG12V/T35S positively regulated MDM2 expression, and then MDM2 negatively regulated WSTF, and WSTF positively regulate H2A.XY142 ph.

Discussion

The alternation of epigenetic modifications might be a key reason in the progress of cancer, including gastric cancer [12]. Epigenetic modifications include diverse forms, such as the methylation of cytosines on DNA, and N-terminal of the histone proteins (H2A, H2B, H3 and H4). Importantly, because its main reason causing aberrant gene damage, histone modification which included acetylation, methylation, phosphorylation and ubiquitylation of the specific histones receives great attentions worldwide now. It also becomes a hallmark of human cancer progress [31]. In the other hand, it is well-known that Ras pathway is involved in diverse human cancers, such as colorectal cancer [32] and colon cancer [33]. Importantly, Ras/ERK is the effective approach in the regulation of cell proliferation and cell invasion in GC [34]. In our study, we investigated the effects of H2A.XY142ph, RasG12V/T35S pathway and the underlying mechanisms in gastric cancer cell SNU16 and MKN1 cells.
H2A.X is the histone H2A family variant, and the H2AX C-terminal domain could be phosphorylated by tyrosine 142 by the WSTF remodeling factor kinase [29]. The expression of H2A.XY142ph was related to DNA damage. In our study, we found that site mutation of G12 V and T35S in Ras pathway decreased the expression of H2A.XY142ph, which indicated that RasG12V/T35S might worked through down-regulated H2A.XY142ph. In addition, further result also proved that RasG12V/T35S was a switch of phosphorylation of ERK1/2, which suggested that Ras/ERK might play a vital role in regulation of H2A.XY142ph.
Cell viability, cell colonies and cell migration was three important factors for judging cell growth and cell metastasis. Ras/ERK signaling pathway was often found activated in multiple human cancers [35], we explored the effects of Ras on GC cells SNU-16 and MKN1 cells. In our study, experiments explored whether RasG12V/T35S and H2A.XY142ph worked in cell viability, number of colonies and cell migration. Result showed that Ras/ERK pathway could enhance cell viability, cell colonies and cell migration, this consistent with the previous studies that activation of ERK pathway promoted GC cell growth [36]. On the other hand, H2A.XY142ph reversed the results, which suggested that phosphorylation of H2A.X might regulate GC cell growth and metastasis. Meanwhile, histone modification exerts crucial functions in the progress of cancer. For example, pancreatic cancer cell growth and metastasis was modulated by histone modification of P27, P53 and Bax [21]. We therefore inferred that histone modification of H2A.XY142ph could affect cell growth might also through regulating the downstream related genes.
Next, results revealed that Ras/ERK down-regulated downstream factors, CYR61, IGFBP3, WNT16B, NT5E, GDF15 and CARD16. Meanwhile, phosphorylation of H2A.X exert the opposite functions as compared with Ras/ERK pathway, this suggested that phosphorylation of H2AX might play important role in modulating Ras/ERK pathway. CHIP assay result showed that Ras/ERK significantly decreased all the gene expression, which confirmed that H2A.XY142ph could up-regulated the downstream factors.
EYA phosphatases are responsible for the phosphorylation of H2A.X on the C-terminal of tyrosyl residue, and EYA2 and EYA3 were proved to be for specificity for Tyr-142 of H2A.X [15]. Results showed that silence of EYA3 increased H2A.XY142ph, further results demonstrated that silence of EYA3 decreased cell viability, cell migration, the percentage of S stage and alter the downstream factors expression in RNA level. These result showed that downregulation of EYA3 led to enhancement of phosphorylation and reduced DNA-damage dephosphorylation of Tyr-142 of H2A.X in vivo [15]. These findings suggested that silence of EYA3 revealed the similar trend with H2A.XY142ph.
In the beginning, we knew that H2A.XY142ph was phosphorylated by WSTF [14, 15]. Then we asked how WSTF was involved in the phosphorylation progress? After, we detected the role of WSTF in SNU-16 cells. We found that no different was found in the RNA level between RasG12V/T35S pathway with the control, which indicated that the effects of WSTF were not in the transcription level. Then we found that WSTF showed that similar trend under transfection with Ras, and further CHIP assay showed that RasG12V/T35S could bind to the promoters of genes to reduce the input levels of WSTF in downstream factors of Ras pathway. Further result showed that activity of Ras-ERK1/2 induced WSTF degradation to decrease H2A.XY142ph expression.
In the last, we studied the mechanism how WSTF was regulated in the progression. MDM2 displays important role in histone ubiquitylation and transcriptional repression [30]. Result demonstrated that Ras/ERK degraded WSTF through upregulation of MDM2. MDM2 revealed negative relationship with H2A.XY142ph expression. Taken together, the whole cascade process might be MDM2 negatively regulate WSTF, WSTF positively regulated EYA3, and EYA3 positively regulated H2A.X Y142ph, which was down-regulated by RasG12V/T35S.

Conclusions

In conclusion, our result demonstrated the RasG12V/T35S and H2A.XY142ph in regulating GC cell growth. The underlying mechanisms are also explored. We firstly found that Ras/ERK pathway could promote GC cell growth while H2A.XY142ph inhibited cell growth. Our study proved the importance of histone modification to some extent. At the same time, we provided novel insight in the relationship between histone modification and the treatment of GC.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12885-022-10052-1"
Literatur
1.
Zurück zum Zitat Hollestelle A, Elstrodt F, Nagel JH, Kallemeijn WW, Schutte M. Phosphatidylinositol-3-OH kinase or RAS pathway mutations in human breast cancer cell lines. Mol Cancer Res. 2007;5(2):195–201.CrossRefPubMed Hollestelle A, Elstrodt F, Nagel JH, Kallemeijn WW, Schutte M. Phosphatidylinositol-3-OH kinase or RAS pathway mutations in human breast cancer cell lines. Mol Cancer Res. 2007;5(2):195–201.CrossRefPubMed
2.
Zurück zum Zitat Zhao YY, Yu L, Liu BL, He XJ, Zhang BY. Downregulation of P-gp, Ras and p-ERK1/2 contributes to the arsenic trioxide-induced reduction in drug resistance towards doxorubicin in gastric cancer cell lines. Mol Med Rep. 2015;12(5):7335–43.CrossRefPubMedPubMedCentral Zhao YY, Yu L, Liu BL, He XJ, Zhang BY. Downregulation of P-gp, Ras and p-ERK1/2 contributes to the arsenic trioxide-induced reduction in drug resistance towards doxorubicin in gastric cancer cell lines. Mol Med Rep. 2015;12(5):7335–43.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Voisset E, Oeztuerk-Winder F, Ruiz EJ, Ventura JJ. p38alpha negatively regulates survival and malignant selection of transformed bronchioalveolar stem cells. PLoS One. 2013;8(11):e78911.CrossRefPubMedPubMedCentral Voisset E, Oeztuerk-Winder F, Ruiz EJ, Ventura JJ. p38alpha negatively regulates survival and malignant selection of transformed bronchioalveolar stem cells. PLoS One. 2013;8(11):e78911.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Niu Z, Zheng L, Wu S, Mu H, Ma F, Song W, Zhu H, Wu J, He X, Hua J. Ras/ERK1/2 pathway regulates the self-renewal of dairy goat spermatogonia stem cells. Reproduction (Cambridge, England). 2015;149(5):445–52.CrossRef Niu Z, Zheng L, Wu S, Mu H, Ma F, Song W, Zhu H, Wu J, He X, Hua J. Ras/ERK1/2 pathway regulates the self-renewal of dairy goat spermatogonia stem cells. Reproduction (Cambridge, England). 2015;149(5):445–52.CrossRef
5.
Zurück zum Zitat Yang HW, Shin MG, Lee S, Kim JR, Park WS, Cho KH, Meyer T, Heo WD. Cooperative activation of PI3K by Ras and Rho family small GTPases. Mol Cell. 2012;47(2):281–90.CrossRefPubMedPubMedCentral Yang HW, Shin MG, Lee S, Kim JR, Park WS, Cho KH, Meyer T, Heo WD. Cooperative activation of PI3K by Ras and Rho family small GTPases. Mol Cell. 2012;47(2):281–90.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Cooper JM, Bodemann BO, White MA. The RalGEF/Ral pathway: evaluating an intervention opportunity for Ras cancers. Enzymes. 2013;34 Pt. B:137–56.CrossRefPubMed Cooper JM, Bodemann BO, White MA. The RalGEF/Ral pathway: evaluating an intervention opportunity for Ras cancers. Enzymes. 2013;34 Pt. B:137–56.CrossRefPubMed
7.
Zurück zum Zitat Lennartsson A, Ekwall K. Histone modification patterns and epigenetic codes. Biochim Biophys Acta. 2009;1790(9):863–8.CrossRefPubMed Lennartsson A, Ekwall K. Histone modification patterns and epigenetic codes. Biochim Biophys Acta. 2009;1790(9):863–8.CrossRefPubMed
8.
Zurück zum Zitat Hattori N, Ushijima T. Compendium of aberrant DNA methylation and histone modifications in cancer. Biochem Biophys Res Commun. 2014;455(1–2):3–9.CrossRefPubMed Hattori N, Ushijima T. Compendium of aberrant DNA methylation and histone modifications in cancer. Biochem Biophys Res Commun. 2014;455(1–2):3–9.CrossRefPubMed
9.
Zurück zum Zitat Yang WY, Gu JL, Zhen TM. Recent advances of histone modification in gastric cancer. J Cancer Res Ther. 2014;10 Suppl:240–5.CrossRefPubMed Yang WY, Gu JL, Zhen TM. Recent advances of histone modification in gastric cancer. J Cancer Res Ther. 2014;10 Suppl:240–5.CrossRefPubMed
10.
Zurück zum Zitat Weyemi U, Redon CE, Choudhuri R, Aziz T, Maeda D, Boufraqech M, Parekh PR, Sethi TK, Kasoji M, Abrams N, et al. The histone variant H2A.X is a regulator of the epithelial-mesenchymal transition. Nat Commun. 2016;7:10711.CrossRefPubMedPubMedCentral Weyemi U, Redon CE, Choudhuri R, Aziz T, Maeda D, Boufraqech M, Parekh PR, Sethi TK, Kasoji M, Abrams N, et al. The histone variant H2A.X is a regulator of the epithelial-mesenchymal transition. Nat Commun. 2016;7:10711.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Wang GG, Allis CD, Chi P. Chromatin remodeling and cancer, part I: covalent histone modifications. Trends Mol Med. 2007;13(9):363–72.CrossRefPubMed Wang GG, Allis CD, Chi P. Chromatin remodeling and cancer, part I: covalent histone modifications. Trends Mol Med. 2007;13(9):363–72.CrossRefPubMed
12.
Zurück zum Zitat Lee SH, Kim J, Kim WH, Lee YM. Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene. 2009;28(2):184–94.CrossRefPubMed Lee SH, Kim J, Kim WH, Lee YM. Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene. 2009;28(2):184–94.CrossRefPubMed
13.
Zurück zum Zitat Weichert W, Roske A, Gekeler V, Beckers T, Ebert MP, Pross M, Dietel M, Denkert C, Rocken C. Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol. 2008;9(2):139–48.CrossRefPubMed Weichert W, Roske A, Gekeler V, Beckers T, Ebert MP, Pross M, Dietel M, Denkert C, Rocken C. Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol. 2008;9(2):139–48.CrossRefPubMed
14.
Zurück zum Zitat Liu Y, Long YH, Wang SQ, Li YF, Zhang JH. Phosphorylation of H2A.X(T)(yr39) positively regulates DNA damage response and is linked to cancer progression. FEBS J. 2016;283(24):4462–73.CrossRefPubMed Liu Y, Long YH, Wang SQ, Li YF, Zhang JH. Phosphorylation of H2A.X(T)(yr39) positively regulates DNA damage response and is linked to cancer progression. FEBS J. 2016;283(24):4462–73.CrossRefPubMed
15.
Zurück zum Zitat Krishnan N, Jeong DG, Jung SK, Ryu SE, Xiao A, Allis CD, Kim SJ, Tonks NK. Dephosphorylation of the C-terminal tyrosyl residue of the DNA damage-related histone H2A.X is mediated by the protein phosphatase eyes absent. J Biol Chem. 2009;284(24):16066–70.CrossRefPubMedPubMedCentral Krishnan N, Jeong DG, Jung SK, Ryu SE, Xiao A, Allis CD, Kim SJ, Tonks NK. Dephosphorylation of the C-terminal tyrosyl residue of the DNA damage-related histone H2A.X is mediated by the protein phosphatase eyes absent. J Biol Chem. 2009;284(24):16066–70.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Stucki M. Histone H2A.X Tyr142 phosphorylation: a novel sWItCH for apoptosis? DNA repair. 2009;8(7):873–6.CrossRefPubMed Stucki M. Histone H2A.X Tyr142 phosphorylation: a novel sWItCH for apoptosis? DNA repair. 2009;8(7):873–6.CrossRefPubMed
17.
Zurück zum Zitat Bustin SA. Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J Mol Endocrinol. 2002;29(1):23–39.CrossRefPubMed Bustin SA. Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J Mol Endocrinol. 2002;29(1):23–39.CrossRefPubMed
18.
Zurück zum Zitat Horibata S, Vo TV, Subramanian V, Thompson PR, Coonrod SA. Utilization of the soft agar Colony formation assay to identify inhibitors of Tumorigenicity in breast Cancer cells. J Vis Exp. 2015;20(99):e52727. Horibata S, Vo TV, Subramanian V, Thompson PR, Coonrod SA. Utilization of the soft agar Colony formation assay to identify inhibitors of Tumorigenicity in breast Cancer cells. J Vis Exp. 2015;20(99):e52727.
19.
Zurück zum Zitat Schulz S, Haussler S. Chromatin immunoprecipitation for ChIP-chip and ChIP-seq. Methods Mol Biol. 2014;1149:591–605.CrossRefPubMed Schulz S, Haussler S. Chromatin immunoprecipitation for ChIP-chip and ChIP-seq. Methods Mol Biol. 2014;1149:591–605.CrossRefPubMed
20.
Zurück zum Zitat Chen H, Zhao JY, Qian XC, Cheng ZY, Liu Y, Wang Z. RASAL1 attenuates gastric carcinogenesis in nude mice by blocking RAS/ERK signaling. Asian Pac J Cancer Prev. 2015;16(3):1077–82.CrossRefPubMed Chen H, Zhao JY, Qian XC, Cheng ZY, Liu Y, Wang Z. RASAL1 attenuates gastric carcinogenesis in nude mice by blocking RAS/ERK signaling. Asian Pac J Cancer Prev. 2015;16(3):1077–82.CrossRefPubMed
21.
Zurück zum Zitat Jiao F, Hu H, Yuan C, Jin Z, Guo Z, Wang L, Wang L. Histone deacetylase 3 promotes pancreatic cancer cell proliferation, invasion and increases drug-resistance through histone modification of P27, P53 and Bax. Int J Oncol. 2014;45(4):1523–30.CrossRefPubMed Jiao F, Hu H, Yuan C, Jin Z, Guo Z, Wang L, Wang L. Histone deacetylase 3 promotes pancreatic cancer cell proliferation, invasion and increases drug-resistance through histone modification of P27, P53 and Bax. Int J Oncol. 2014;45(4):1523–30.CrossRefPubMed
22.
Zurück zum Zitat Harada H, Omi M, Sato T, Nakamura H. Pea3 determines the isthmus region at the downstream of Fgf8-Ras-ERK signaling pathway. Develop Growth Differ. 2015;57(9):657–66.CrossRef Harada H, Omi M, Sato T, Nakamura H. Pea3 determines the isthmus region at the downstream of Fgf8-Ras-ERK signaling pathway. Develop Growth Differ. 2015;57(9):657–66.CrossRef
23.
Zurück zum Zitat Xu W, Chou CL, Israel DD, Hutchinson AJ, Regan JW. PGF(2alpha) stimulates FP prostanoid receptor mediated crosstalk between Ras/Raf signaling and Tcf transcriptional activation. Biochem Biophys Res Commun. 2009;381(4):625–9.CrossRefPubMedPubMedCentral Xu W, Chou CL, Israel DD, Hutchinson AJ, Regan JW. PGF(2alpha) stimulates FP prostanoid receptor mediated crosstalk between Ras/Raf signaling and Tcf transcriptional activation. Biochem Biophys Res Commun. 2009;381(4):625–9.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Guo L, Costanzo-Garvey DL, Smith DR, Zavorka ME, Venable-Kang M, MacDonald RG, Lewis RE. Cell non-autonomous regulation of hepatic IGF-1 and neonatal growth by kinase suppressor of Ras 2 (KSR2). Sci Rep. 2016;6:32093.CrossRefPubMedPubMedCentral Guo L, Costanzo-Garvey DL, Smith DR, Zavorka ME, Venable-Kang M, MacDonald RG, Lewis RE. Cell non-autonomous regulation of hepatic IGF-1 and neonatal growth by kinase suppressor of Ras 2 (KSR2). Sci Rep. 2016;6:32093.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Johnson LM, Price DK, Figg WD. Treatment-induced secretion of WNT16B promotes tumor growth and acquired resistance to chemotherapy: implications for potential use of inhibitors in cancer treatment. Cancer Biol Ther. 2013;14(2):90–1.CrossRefPubMedPubMedCentral Johnson LM, Price DK, Figg WD. Treatment-induced secretion of WNT16B promotes tumor growth and acquired resistance to chemotherapy: implications for potential use of inhibitors in cancer treatment. Cancer Biol Ther. 2013;14(2):90–1.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Wang N, Xiang X, Chen K, Liu P, Zhu A. Targeting of NT5E by miR-30b and miR-340 attenuates proliferation, invasion and migration of gallbladder carcinoma. Biochimie. 2018;146:56–67.CrossRefPubMed Wang N, Xiang X, Chen K, Liu P, Zhu A. Targeting of NT5E by miR-30b and miR-340 attenuates proliferation, invasion and migration of gallbladder carcinoma. Biochimie. 2018;146:56–67.CrossRefPubMed
27.
Zurück zum Zitat Li C, Wang J, Kong J, Tang J, Wu Y, Xu E, Zhang H, Lai M. GDF15 promotes EMT and metastasis in colorectal cancer. Oncotarget. 2016;7(1):860–72.CrossRefPubMed Li C, Wang J, Kong J, Tang J, Wu Y, Xu E, Zhang H, Lai M. GDF15 promotes EMT and metastasis in colorectal cancer. Oncotarget. 2016;7(1):860–72.CrossRefPubMed
28.
Zurück zum Zitat Karasawa T, Kawashima A, Usui F, Kimura H, Shirasuna K, Inoue Y, Komada T, Kobayashi M, Mizushina Y, Sagara J, et al. Oligomerized CARD16 promotes caspase-1 assembly and IL-1beta processing. FEBS Open Bio. 2015;5:348–56.CrossRefPubMedPubMedCentral Karasawa T, Kawashima A, Usui F, Kimura H, Shirasuna K, Inoue Y, Komada T, Kobayashi M, Mizushina Y, Sagara J, et al. Oligomerized CARD16 promotes caspase-1 assembly and IL-1beta processing. FEBS Open Bio. 2015;5:348–56.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Xiao A, Li H, Shechter D, Ahn SH, Fabrizio LA, Erdjument-Bromage H, Ishibe-Murakami S, Wang B, Tempst P, Hofmann K, et al. WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity. Nature. 2009;457(7225):57–62.CrossRefPubMed Xiao A, Li H, Shechter D, Ahn SH, Fabrizio LA, Erdjument-Bromage H, Ishibe-Murakami S, Wang B, Tempst P, Hofmann K, et al. WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity. Nature. 2009;457(7225):57–62.CrossRefPubMed
30.
Zurück zum Zitat Minsky N, Oren M. The RING domain of Mdm2 mediates histone ubiquitylation and transcriptional repression. Mol Cell. 2004;16(4):631–9.CrossRefPubMed Minsky N, Oren M. The RING domain of Mdm2 mediates histone ubiquitylation and transcriptional repression. Mol Cell. 2004;16(4):631–9.CrossRefPubMed
31.
Zurück zum Zitat Lippman Z, Gendrel AV, Colot V, Martienssen R. Profiling DNA methylation patterns using genomic tiling microarrays. Nat Methods. 2005;2(3):219–24.CrossRefPubMed Lippman Z, Gendrel AV, Colot V, Martienssen R. Profiling DNA methylation patterns using genomic tiling microarrays. Nat Methods. 2005;2(3):219–24.CrossRefPubMed
32.
Zurück zum Zitat Rui Y, Wang C, Zhou Z, Zhong X, Yu Y. K-Ras mutation and prognosis of colorectal cancer: a meta-analysis. Hepatogastroenterology. 2015;62(137):19–24.PubMed Rui Y, Wang C, Zhou Z, Zhong X, Yu Y. K-Ras mutation and prognosis of colorectal cancer: a meta-analysis. Hepatogastroenterology. 2015;62(137):19–24.PubMed
33.
Zurück zum Zitat Goel S, Huang J, Klampfer L. K-Ras, intestinal homeostasis and colon cancer. Curr Clin Pharmacol. 2015;10(1):73–81.CrossRefPubMed Goel S, Huang J, Klampfer L. K-Ras, intestinal homeostasis and colon cancer. Curr Clin Pharmacol. 2015;10(1):73–81.CrossRefPubMed
34.
Zurück zum Zitat Chen H, Cheng ZY, Pan Y, Wang Z, Liu Y, Zhang JQ. RASAL1 influences the proliferation and invasion of gastric cancer cells by regulating the RAS/ERK signaling pathway. Hum Cell. 2014;27(3):103–10.CrossRefPubMed Chen H, Cheng ZY, Pan Y, Wang Z, Liu Y, Zhang JQ. RASAL1 influences the proliferation and invasion of gastric cancer cells by regulating the RAS/ERK signaling pathway. Hum Cell. 2014;27(3):103–10.CrossRefPubMed
35.
Zurück zum Zitat Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014;13(12):928–42.CrossRefPubMed Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014;13(12):928–42.CrossRefPubMed
36.
Zurück zum Zitat Qu JL, Qu XJ, Zhao MF, Teng YE, Zhang Y, Hou KZ, Jiang YH, Yang XH, Liu YP. Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig Liver Dis. 2009;41(12):875–80.CrossRefPubMed Qu JL, Qu XJ, Zhao MF, Teng YE, Zhang Y, Hou KZ, Jiang YH, Yang XH, Liu YP. Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig Liver Dis. 2009;41(12):875–80.CrossRefPubMed
Metadaten
Titel
RETRACTED ARTICLE: K-ras-ERK1/2 down-regulates H2A.XY142ph through WSTF to promote the progress of gastric cancer
verfasst von
Chao Dong
Jing Sun
Sha Ma
Guoying Zhang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5750-x

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.