• Users Online: 175
  • Print this page
  • Email this page

 
Table of Contents
ORIGINAL ARTICLE
Year : 2023  |  Volume : 66  |  Issue : 4  |  Page : 276-283

Knockdown of neurotrophin receptor-interacting melanoma-associated antigen homolog inhibits acute myeloid leukemia cell growth via the ERK pathway


1 Department of Hematology, First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang Uygur Autonomous Region, China
2 Department of Hematological, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, Guangxi, China

Date of Submission19-Dec-2022
Date of Decision06-Mar-2023
Date of Acceptance23-Mar-2023
Date of Web Publication04-Jul-2023

Correspondence Address:
Dr. Beili Chen
Department of Hematological, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Guilin, Guangxi Zhuang Autonomous Region, Guangxi 541001
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/cjop.CJOP-D-22-00162

Rights and Permissions
  Abstract 


Neurotrophin receptor-interacting melanoma-associated antigen homolog (NRAGE), a type II melanoma-associated antigen, plays a critical role in cell processes that are involved in the tumorigenesis of various cancers. However, the effect of NRAGE on acute myeloid leukemia (AML) is rarely reported. The expression of NRAGE in AML tissues and the survival rates between different AML groups were obtained from the GEPIA tool. Human AML cell lines were cultured and transfected with siRNA targeting NRAGE. The ability of AML cells to proliferate and cell cycle were examined. Western blotting was performed to detect the activity of the extracellular signal-regulated kinase (ERK) signaling pathway in AML cells. NRAGE expression was enhanced in AML tissues relative to control tissues, and the high NRAGE expression in AML patients is associated with a poor prognosis. The capacity of AML cells to survive and proliferate was significantly decreased and its cell cycle was arrested at the G1 phase after NRAGE was silenced. Furthermore, silencing NRAGE induced the inactivation of the ERK signaling pathway. Furthermore, supplement of tert-Butylhydroquinone, an ERK activator, improved the reduced ability of AML cell survival and proliferation as well as cell cycle arrest induced by NRAGE knockdown. In this study, NRAGE was identified as a tumor promoter in AML, which had an effect on cell proliferation, cell survival, and cell cycle through the ERK signaling pathway in AML cells.

Keywords: Acute myeloid leukemia, cell cycle, cell growth, extracellular signal-regulated kinase signaling pathway, neurotrophin receptor-interacting melanoma-associated antigen homolog


How to cite this article:
Zhang H, Wu G, Chen B. Knockdown of neurotrophin receptor-interacting melanoma-associated antigen homolog inhibits acute myeloid leukemia cell growth via the ERK pathway. Chin J Physiol 2023;66:276-83

How to cite this URL:
Zhang H, Wu G, Chen B. Knockdown of neurotrophin receptor-interacting melanoma-associated antigen homolog inhibits acute myeloid leukemia cell growth via the ERK pathway. Chin J Physiol [serial online] 2023 [cited 2023 Sep 26];66:276-83. Available from: https://www.cjphysiology.org/text.asp?2023/66/4/276/380364




  Introduction Top


Acute myeloid leukemia (AML) is a type of leukemia characterized by abnormal myeloid differentiation and the accumulation of leukemia stem cell precursors in the hemopoietic system.[1],[2] As the most frequent acute leukemia in adults (accounting for 80% in all cases), its incidence is increasing with age which ranges from 1.3/100,000 in people under 65 years to 12.2/100,000 in people over 65 years.[1],[3] At present, conventional chemotherapy, allogeneic hematopoietic cell transplantation, and chimeric antigen receptor-T-cell therapy have made some improvements in the treatment response and prognosis for AML, but the treatment effect is still generally poor.[4],[5],[6] Therefore, the treatment options of AML remain restricted, and more molecular studies are required to better understand the disease and find new therapeutic targets.

The mitogen-activated protein kinase (MAPK) signaling pathway performs a pivotal role in signal transduction, which is related to numerous cellular biological processes.[7] Upon activation, MAPKs coordinate and regulate various cellular activities such as cell proliferation, cell motility, cell survival, and cell apoptosis.[8],[9] Extracellular signal-regulated kinase (ERK) is a crucial member of the MAPK family. Moreover, dysregulation of the ERK signaling pathway has been reported to be correlated with malignant phenotypes of AML cells.[10]

Melanoma-associated antigen (MAGE) is a member of cancer/testis antigen family comprising more than fifty kinds of proteins.[11] The MAGE family can be classified into two groups based on various gene architectures and tissue-specific expression patterns: MEGA-1 and MEGA-2.[11] Aberrant activation and expression of MAGEs were found in various cancers.[12],[13] Neurotrophin receptor-interacting melanoma-associated antigen homolog (NRAGE, also called as MAGED1) is one of the MAGE families.[11] Prior studies have shown that NRAGE can promote cancer in various tumors.[14] NRAGE facilitates cancer invasion and chemoresistance, acting as a tumor promoter in gastric cancer.[15] NRAGE expression was significantly higher in hepatocellular carcinoma tissues, which accelerated the malignant progress of hepatocellular carcinoma.[16] Furthermore, NRAGE significantly promotes the radiation resistance of esophageal cancer cells.[17] However, the effect of NRAGE on AML remains unidentified.

In this report, the expression of NRAGE in AML and the survival analysis with NRAGE subgroups were analyzed by bioinformatic approach. The capability of AML cell viability and apoptosis and cell cycle were also assessed. Moreover, the activity of the ERK signaling pathway in AML cells was also detected. The present research attempted to investigate the influence of NRAGE on cellular functions of AML cells, which might provide new ideas for the treatment of AML.


  Materials and Methods Top


Bioinformatic analysis

The transcript levels of NRAGE in tumor samples and control samples from 173 AML patients and 70 controls were obtained, and then, the survival analysis with NRAGE subgroup in AML patients was analyzed using the Gene Expression Profiling Interactive Analysis (GEPIA), which is a web-based tool (http://gepia.cancer-pku.cn/index.html) to deliver fast and customizable gene expression datasets based on the TCGA and GTEx database.[18] Transcript per million normalization was used to measure gene or transcript expression levels of RNA sequencing data.

Cell culture and transfection

In this study, human bone marrow stromal cell line (HS-5) and four human AML cell lines (OCI-AML-5, KG-1, HL-60, and MOLM-14) which were all obtained from American Type Culture Collection (Manassas, VA, USA) were cultured in RPMI 1640 medium containing 10% FBS (Thermo Fisher Scientific, Waltham, Massachusetts, USA) with 1% penicillin and streptomycin (Thermo Fisher Scientific) at 37°C with 5% CO2 in a humidified incubator.

KG-1 and HL-60 cells were transfected with NRAGE-short interference RNA (siRNA) and negative control (NC)-siRNA (Thermo Fisher Scientific) as previously described.[19] When cells were 60%–80% confluence, cells were incubated with Lipofectamine transfection reagent (Thermo Fisher Scientific) for 24 h according to the manufacturer's protocol. After transfection, the ability of cell survival and cell proliferation was examined. Besides, AML cells were treated with tert-Butylhydroquinone (tBHQ) (MedChemExpress, Shanghai, China) at a dose of 5 μM for 15 min to activate the ERK signaling pathway.

Colony formation assay

Cells were inoculated into 12-well plates and cultured for 2 weeks at 37°C. Then, cells were fixed with methanol and stained with crystal violet for 10 min. After staining, cell colonies were photographed and counted.

Cell viability assay

Cell viability was assessed using a Cell Counting Kit-8 (CCK-8) assay (Sigma, St. Louis, MO, USA) as directed by the manufacturer.[20] A total of 5 × 103 cells were seeded into 96-well plates and then 10 μL CCK-8 solution was added for 2-h incubation. At a wavelength of 450 nm, optical density values were determined using a microplate reader (Bio-Rad, Richmond, VA, USA).

Western blotting

The collected cells were lysed in the RIPA Lysis and Extraction Buffer (Thermo Fisher Scientific). A BCA protein detection kit (Thermo Fisher Scientific) was utilized to evaluate the protein concentrations of supernatant.[21] Protein was loaded, separated, and then transferred to PVDF membranes. After blocking, the blots were incubated with primary antibodies. Second antibodies (Thermo Fisher Scientific) were added for 1-h incubation. Following that, ECL was applied to develop the blots. The primary antibodies used in Western blotting are present in [Table 1].
Table 1: Primary antibodies used in Western blotting

Click here to view


Cell apoptosis assay

Cell apoptosis was detected using an Annexin Apoptosis Detection Kit (Thermo Fisher Scientific) according to the manufacturer's protocol.[22] Briefly, cells were incubated with 5 μL Annexin V reagent and 5 μL propidium iodide reagent for 5 min in the dark. Then, cells were enumerated and data were analyzed using FlowJo software (TreeStar, Ashland, OR, USA).

Cell cycle analysis

Cells were collected and fixed with 70% cold ethanol at 4°C overnight. Then, cells were stained with 1 mL of prepared DNA staining solution (Thermo Fisher Scientific) and 10 μL permeabilization solution (Thermo Fisher Scientific) in the dark at 37°C for 30 min. Following that, cells were enumerated and then data were analyzed using FlowJo software.

Statistical analysis

In this study, the GraphPad Prism software (version 8.2, San Diego, CA, USA) was used for all data analysis and visualization. The mean ± standard deviation was employed to express the data. To ascertain the statistical difference, the student's t-test or one-way analysis of variance was applied. Kaplan–Meier plotter was applied to assess the survival rates between different NRAGE expression groups. The hazard ratio of 95% confidence interval was displayed in the graph, and the log-rank P < 0.05 was considered statistically different.


  Results Top


The increased expression of NRAGE is correlated with a poor prognosis in AML patients

Rising research reports that NRAGE plays a vital role in multiple cancers.[14],[15],[16],[17] We first explored the expression level of NRAGE in AML patients using public database. As shown in [Figure 1]a, GEPIA platform indicated that the expression of NRAGE in AML tissues was higher than that in control tissues. Further analysis was carried out to compare the relationship between NRAGE expression and the survival rates of AML patients. Moreover, AML patients with high NRAGE expression exhibited a poor prognosis in relation to those with low NRAGE levels [Figure 1]b. A set of 100 samples was divided into the low NRAGE expression group and the high NRAGE expression group. In [Table 2], we compared clinicopathological characteristics between various groups. However, there was no difference in age or gender between the two groups [Table 2]. In comparison to human bone marrow stromal cell line (HS-5), the protein levels of NRAGE were also elevated in human AML cell lines (OCI-AML-5, KG-1, HL-60, and MOLM-14), especially KG-1 and HL-60, which were then chosen for further cell experiment [Figure 1]c. These data imply that NRAGE is highly expressed in AML tissues and is linked to overall survival in AML patients.
Figure 1: The increased expression of NRAGE is linked to a poor prognosis in AML patients. (a) The transcript level of NRAGE in AML tissues (n = 173) and normal tissues (n = 70) was obtained from GEPIA tool. (b) Kaplan–Meier curve showed the survival rates of the high NRAGE expression group (n = 53) and low NRAGE expression group (n = 53). (c) Western blotting showed the protein levels of NRAGE in human AML cell lines (OCI-AML-5, KG-1, HL-60, and MOLM-14) (n = 3) and human bone marrow stromal cell line (HS-5) (n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001 versus the normal tissue group or HS-5 cells. NRAGE: Neurotrophin receptor-interacting melanoma-associated antigen homolog, AML: Acute myeloid leukemia.

Click here to view
Table 2: The clinical characteristics of acute myeloid leukemia patients between the different NRAGE expression groups

Click here to view


Silencing NRAGE inhibits AML cell survival and growth

We further investigated the effect of NRAGE on the capacity of AML cells. KG-1 and HL-60 cells were transfected with siNRAGE to downregulate the expression of NRAGE. After transfection, NRAGE expression was dramatically lower in the siNRAGE groups compared to the siNC group [Figure 2]a. Furthermore, the ability of AML cell survival and proliferation was examined by both CCK-8 assay and colony formation assay. We observed that NRAGE knockdown reduced AML cell viability using a CCK-8 assay [Figure 2]b. Furthermore, colony formation assay demonstrated that silencing NRAGE weakened the proliferation ability of AML cells [Figure 2]c. As a result, silencing NRAGE downregulates the capacity of AML cell survival and growth.
Figure 2: Silencing NRAGE inhibits AML cell survival and growth. (a-c) KG-1 and HL-60 cells were transfected with siNRAGE or siNC. (a) Western blotting showed the protein levels of NRAGE in the siNC and siNRAGE groups (n = 3). (b) CCK-8 assay showed cell viability of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). (c) Colony formation assay showed the ability of cell proliferation of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). *P < 0.05, **P < 0.01, and ***P < 0.001 versus the siNC group. NRAGE: Neurotrophin receptor-interacting melanoma-associated antigen homolog, AML: Acute myeloid leukemia, CCK-8: Cell Counting Kit-8.

Click here to view


Silencing NRAGE induces AML cell cycle arrest at G1 phase

We next explored the impacts of NRAGE on AML cell cycle progression, and the cell cycle phase distribution of AML cells with or without NRAGE knockdown was examined by flow cytometry. NRAGE knockdown led to AML cell cycle arrest at G1 phase compared to the siNC group [Figure 3]a. Cyclin D1 and cyclin-dependent kinase 4 (CDK4) are vital players in the G1-S cell cycle transition which are overexpressed in multiple cancers.[23] Silencing NRAGE decreased both cyclin D1 and CDK4 production in AML cells [Figure 3]b and [Figure 3]c. Altogether, these data suggest that NRAGE knockdown results in AML cell cycle arrest by regulating cyclin D1 and CDK4 expression.
Figure 3: Silencing NRAGE induces AML cell cycle arrest at G1 phase. (a-c) KG-1 and HL-60 cells were transfected with siNRAGE or siNC. (a) Flow cytometry showed the cell cycle phase distribution of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). (b) Western blotting showed the protein levels of cyclin D1 of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). (c) Western blotting showed the protein levels of CDK4 of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). *P < 0.05, **P < 0.01 and ***P < 0.001 versus the siNC group. NRAGE: Neurotrophin receptor-interacting melanoma-associated antigen homolog, AML: Acute myeloid leukemia, CDK4: Cyclin-dependent kinase 4.

Click here to view


NRAGE controls AML cell apoptosis through regulating Bcl-2 expression

Whether NRAGE had a role in AML cell apoptosis needs to be identified, and flow cytometry and Western blotting were applied to detect the ability of AML cell apoptosis. Flow cytometry indicated that the number of apoptotic AML cells was significantly increased after the depletion of NRAGE [Figure 4]a. Prior research has evidenced that B-cell lymphoma-2 (Bcl-2) is crucial for the survival of AML cells.[24] Western blotting was used to determine the protein levels of Bcl-2 and Bcl-2-associated X protein (Bax) in order to confirm the hypothesis that NRAGE modulates AML cell apoptosis by Bcl-2. Comparing the siNRAGE group to the siNC group, we observed that the levels of Bax were higher and the levels of Bcl-2 were lower [Figure 4]b. These results indicate that NRAGE has a regulatory role in AML cell apoptosis through regulating Bcl-2 expression.
Figure 4: NRAGE controls AML cell apoptosis through regulating Bcl-2 expression. (a and b) KG-1 and HL-60 cells were transfected with siNRAGE or siNC. (a) Flow cytometry showed the number of apoptotic KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). (b) Western blotting showed the protein levels of Bcl-2 and Bax of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). **P < 0.01 and ***P < 0.001 versus the siNC group. NRAGE: Neurotrophin receptor-interacting melanoma-associated antigen homolog, AML: Acute myeloid leukemia, Bcl-2: B-cell lymphoma-2, Bax: Bcl-2-associated X protein.

Click here to view


The capacity of AML cell growth and apoptosis is modulated by NRAGE through the ERK signaling pathway

Emerging evidence reveals that the ERK/MAPK signaling pathway modulates AML cell proliferation and G1/S transition.[25] We investigate whether NRAGE has an involvement in cell proliferation, cell survival, and cell cycle through the ERK/MAPK signaling pathway. After NRAGE was knockdown, the phosphorylation of ERK was inhibited in AML cells [Figure 5]a. tBHQ, an ERK activator, was then applied to activate the ERK signaling pathway.[26] We observed that tBHQ treatment restored the reduced cell viability in AML cells with siNRAGE transfection [Figure 5]b. Moreover, the levels of cell cycle progression regulators (cyclin D1 and CDK4) were enhanced by tBHQ treatment in the siNRAGE group [Figure 5]c and [Figure 5]d. tBHQ treatment also decreased the levels of Bax in AML cells, which was increased by NRAGE knockdown [Figure 5]d. Moreover, the downregulated levels of Bcl-2 in the siNRAGE group were upregulated by tBHQ treatment [Figure 5]d. These findings suggest that NRAGE regulates the activation of the ERK signaling pathway, thereby controlling AML cell growth and cell cycle.
Figure 5: The capacity of AML cell growth and apoptosis is modulated by NRAGE through the ERK signaling pathway. (a) KG-1 and HL-60 cells were transfected with siNRAGE or siNC. Western blotting showed the protein levels of phosphorylated ERK of KG-1 and HL-60 cells in the siNC and siNRAGE groups (n = 3). (b-d) KG-1 and HL-60 cells were transfected with siNRAGE or siNC and treated with the ERK activator, tBHQ. (b) CCK-8 assay showed cell viability of KG-1 and HL-60 cells in the siNC, siNRAGE, and siNRAGE with tBHQ treatment groups (n = 3). (c) Western blotting showed the protein levels of cyclin D1 of KG-1 and HL-60 cells in the siNC, siNRAGE, and siNRAGE with tBHQ treatment groups (n = 3). (d) Western blotting showed the protein levels of CDK4, Bcl-2, and Bax of KG-1 and HL-60 cells in the siNC, siNRAGE, and siNRAGE with tBHQ treatment groups (n = 3). *P < 0.05, **P < 0.01, and ***P < 0.001 versus the siNC group. #P < 0.05, ##P < 0.01 and ###P < 0.001 versus the siNRAGE group. NRAGE: Neurotrophin receptor-interacting melanoma-associated antigen homolog, AML: Acute myeloid leukemia, CCK-8: Cell Counting Kit-8, CDK4: Cyclin-dependent kinase 4, Bcl-2: B-cell lymphoma-2, Bax: Bcl-2-associated X protein, tBHQ: Tert-Butylhydroquinone.

Click here to view



  Discussion Top


AML is a genetically heterogeneous disorder of blood system. Increasing evidence shows a variety of genetic and epigenetic alterations in oncogenes and tumor suppressors in patients with AML.[27],[28],[29] Despite considerable advances in understanding the molecular landscape of AML and its effect on disease pathophysiology and the invention of new drugs, standard medical interventions have not changed significantly over the past decades.[4],[5],[6] In this study, we observed that NRAGE functioned as a tumor promoter in AML, regulating cell survival, cell apoptosis, and cell cycle through the ERK1/2 signaling pathway.

NRAGE is a type II melanoma-associated antigen that is required for numerous cell processes, such as cell survival and cell apoptosis.[14] NRAGE has been identified as a cancer-related protein with complex functions in various cancers.[14] Previous studies reported that NRAGE acted as a tumor suppressor in melanoma, pancreatic cancer, and breast cancer.[30],[31] Chu et al. reported that overexpression of NRAGE inhibited the mRNA expression and activity of matrix metalloproteinase 2 (MMP2), thus suppressing metastasis of melanoma and pancreatic cancer. Paradoxically, conflicting research has shown that NRAGE could function as a tumor promoter, which facilitates the tumorigenesis of hepatocellular carcinoma, esophageal carcinoma, and gastric cancer.[15],[16],[17] Prior research found that NRAGE depletion negatively regulated Bcl-2 and p-ERK and upregulated ZO-1 and p27 expression levels which then acted as a tumor promoter in gastric cancer by facilitating cancer invasion and chemoresistance. Furthermore, NRAGE subcellular localization is related to radiation resistance of esophageal carcinoma cells by regulating epithelial-mesenchymal transition process. However, the role of NRAGE in AML is barely identified. In the present study, an increase in the expression of NRAGE in AML tissue samples was observed. Moreover, NRAGE upregulation was linked to a poor survival of AML patients. Moreover, silencing NRAGE inhibited the ability of AML cell survival and cell proliferation and induced AML cell cycle arrest. Therefore, we identified NRAGE as a tumor promoter in AML patients.

The Bcl-2 family proteins have a central role in the mitochondrial apoptotic pathway.[32] The death effector, Bax, promotes cell apoptosis by activating mitochondrial membrane potential, while the antiapoptotic protein, Bcl-2, negatively regulates Bax to inhibit cell apoptosis.[33] We observed that silencing NRAGE reduced the levels of Bcl-2, whereas Bax expression was enhanced by NRAGE knockdown in AML cells, suggesting that NRAGE regulated cell survival through Bcl-2 family proteins.

Cyclins and CDKs are important proteins that are essential for the regulation and expression of the large number of components required for the passage through the cell cycle.[23] Cyclin D1 and CDK4 are the important G1/S checkpoint of the cell cycle.[34] In this study, we found that knockdown of NRAGE induced cell cycle arrest and decreased cyclin D1 and CDK4 expression, demonstrating that NRAGE controlled cell cycle through regulating the production of cyclin D1 and CDK4.

There are four specific cascades – ERK, Jun amino-terminal kinases, p38-MAPK, and ERK5 sharing the MAPK signaling pathway.[35] The ERK/MAPK signaling pathway is reported to be closely correlated with cellular biological processes such as cell proliferation, cell apoptosis, and cell cycle that were dysregulated in various cancers.[8],[36] In leukemia, progenitor cells were revealed to have abnormally activated ERK/MAPK signaling pathways, and decreasing ERK activity prevented primary AML cells from proliferating and resulted in cell apoptosis.[37] Furthermore, the improper MAPK signaling pathway activity is a poor predictor of overall survival in patients with AML.[38] In this report, silencing NRAGE inactivated the ERK signaling pathway. Moreover, in AML cells with NRAGE knockdown, tBHQ, an ERK activator, could restore the impaired ability of AML cell proliferation and survival as well as cell cycle arrest. Taken together, these data imply that NRAGE possesses a role in AML cell function through the ERK signaling pathway.


  Conclusion Top


In the present study, an increase of NRAGE expression was observed in AML tissues, and high NRAGE expression showed a poor prognosis in AML patients. Moreover, NRAGE has a regulatory role in the capability of AML cell survival and proliferation as well as cell cycle through the ERK signaling pathway. Thus, we identified NRAGE as a tumor promoter in AML, which might be a therapeutic target for AML treatment in the future.

Availability of data and materials

All data generated or analyzed during this study are included in this published article. The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

All authors contributed to the study conception and design. Material preparation and the experiments were performed by Hongxia Zhang. Data collection and analysis were performed by Guangsheng Wu. The first draft of the manuscript was written by BeiLi Chen and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Financial support and sponsorship

This work was supported by the Shihezi University 2021 Corps financial science and technology projects (Grant No. 2021CA002) and Shihezi University 2021 independent funding support for university-level research projects (Grant No. ZZZC202186).

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Pelcovits A, Niroula R. Acute myeloid leukemia: A review. R I Med J (2013) 2020;103:38-40.  Back to cited text no. 1
    
2.
Chopra M, Bohlander SK. The cell of origin and the leukemia stem cell in acute myeloid leukemia. Genes Chromosomes Cancer 2019;58:850-8.  Back to cited text no. 2
    
3.
De Kouchkovsky I, Abdul-Hay M. 'Acute myeloid leukemia: A comprehensive review and 2016 update'. Blood Cancer J 2016;6:e441.  Back to cited text no. 3
    
4.
Kassim AA, Savani BN. Hematopoietic stem cell transplantation for acute myeloid leukemia: A review. Hematol Oncol Stem Cell Ther 2017;10:245-51.  Back to cited text no. 4
    
5.
Floren M, Gillette JM. Acute myeloid leukemia: Therapy resistance and a potential role for tetraspanin membrane scaffolds. Int J Biochem Cell Biol 2021;137:106029.  Back to cited text no. 5
    
6.
Medinger M, Heim D, Halter JP, Lengerke C, Passweg JR. Diagnosis and therapy of acute myeloid leukemia. Ther Umsch 2019;76:481-6.  Back to cited text no. 6
    
7.
Morrison DK. MAP kinase pathways. Cold Spring Harb Perspect Biol 2012;4:a011254.  Back to cited text no. 7
    
8.
Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res 2015;35:600-4.  Back to cited text no. 8
    
9.
Ling S, Zhang L, Qian Y, Liu Z, Mao Z, Zhang Q. Curcumol inhibits PDGF-BB-induced proliferation and migration of airway smooth muscle cells by suppressing ERK/CREB pathway. Allergol Immunopathol (Madr) 2022;50:17-24.  Back to cited text no. 9
    
10.
Scholl C, Gilliland DG, Fröhling S. Deregulation of signaling pathways in acute myeloid leukemia. Semin Oncol 2008;35:336-45.  Back to cited text no. 10
    
11.
Sang M, Wang L, Ding C, Zhou X, Wang B, Wang L, et al. Melanoma-associated antigen genes – An update. Cancer Lett 2011;302:85-90.  Back to cited text no. 11
    
12.
Li S, Shi X, Li J, Zhou X. Pathogenicity of the MAGE family. Oncol Lett 2021;22:844.  Back to cited text no. 12
    
13.
Schäfer P, Paraschiakos T, Windhorst S. Oncogenic activity and cellular functionality of melanoma associated antigen A3. Biochem Pharmacol 2021;192:114700.  Back to cited text no. 13
    
14.
Zhang G, Zhou H, Xue X. Complex roles of NRAGE on tumor. Tumour Biol 2016;37:11535-40.  Back to cited text no. 14
    
15.
Jiang X, Jiang X, Yang Z. NRAGE confers poor prognosis and promotes proliferation, invasion, and chemoresistance in gastric cancer. Gene 2018;668:114-20.  Back to cited text no. 15
    
16.
Shimizu D, Kanda M, Sugimoto H, Sueoka S, Takami H, Ezaka K, et al. NRAGE promotes the malignant phenotype of hepatocellular carcinoma. Oncol Lett 2016;11:1847-54.  Back to cited text no. 16
    
17.
Chang X, Xue X, Zhang Y, Zhang G, Zhou H, Yang Y, et al. The role of NRAGE subcellular location and epithelial-mesenchymal transition on radiation resistance of esophageal carcinoma cell. J Cancer Res Ther 2018;14:46-51.  Back to cited text no. 17
    
18.
Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res 2017;45:W98-102.  Back to cited text no. 18
    
19.
Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, et al. Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res 2020;8:436-50.  Back to cited text no. 19
    
20.
Papachristou F, Anninou N, Koukoulis G, Paraskakis S, Sertaridou E, Tsalikidis C, et al. Differential effects of cisplatin combined with the flavonoid apigenin on HepG2, Hep3B, and Huh7 liver cancer cell lines. Mutat Res Genet Toxicol Environ Mutagen 2021;866:503352.  Back to cited text no. 20
    
21.
Olson BJSC. Assays for determination of protein concentration. Curr Protoc Pharmacol 2016;73:A.3A.1-A.3A.32.  Back to cited text no. 21
    
22.
Crowley LC, Marfell BJ, Scott AP, Waterhouse NJ. Quantitation of apoptosis and necrosis by annexin V binding, propidium iodide uptake, and flow cytometry. Cold Spring Harb Protoc 2016;2016. [doi: 10.1101/pdb.prot087288].  Back to cited text no. 22
    
23.
Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: A changing paradigm. Nat Rev Cancer 2009;9:153-66.  Back to cited text no. 23
    
24.
Pan R, Hogdal LJ, Benito JM, Bucci D, Han L, Borthakur G, et al. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer Discov 2014;4:362-75.  Back to cited text no. 24
    
25.
Lu J, Chen S, Tan H, Huang Z, Li B, Liu L, et al. Eukaryotic initiation factor-2, gamma subunit, suppresses proliferation and regulates the cell cycle via the MAPK/ERK signaling pathway in acute myeloid leukemia. J Cancer Res Clin Oncol 2021;147:3157-68.  Back to cited text no. 25
    
26.
Chen T, Qiao X, Cheng L, Liu M, Deng Y, Zhuo X. LGR4 silence aggravates ischemic injury by modulating mitochondrial function and oxidative stress via ERK signaling pathway in H9c2 cells. J Mol Histol 2021;52:363-71.  Back to cited text no. 26
    
27.
Prada-Arismendy J, Arroyave JC, Röthlisberger S. Molecular biomarkers in acute myeloid leukemia. Blood Rev 2017;31:63-76.  Back to cited text no. 27
    
28.
Bullinger L, Döhner K, Döhner H. Genomics of acute myeloid leukemia diagnosis and pathways. J Clin Oncol 2017;35:934-46.  Back to cited text no. 28
    
29.
Cai SF, Levine RL. Genetic and epigenetic determinants of AML pathogenesis. Semin Hematol 2019;56:84-9.  Back to cited text no. 29
    
30.
Chu CS, Xue B, Tu C, Feng ZH, Shi YH, Miao Y, et al. NRAGE suppresses metastasis of melanoma and pancreatic cancer in vitro and in vivo. Cancer Lett 2007;250:268-75.  Back to cited text no. 30
    
31.
Du Q, Zhang Y, Tian XX, Li Y, Fang WG. MAGE-D1 inhibits proliferation, migration and invasion of human breast cancer cells. Oncol Rep 2009;22:659-65.  Back to cited text no. 31
    
32.
Dietrich JB. Apoptosis and anti-apoptosis genes in the Bcl-2 family. Arch Physiol Biochem 1997;105:125-35.  Back to cited text no. 32
    
33.
Gaumer S, Guénal I, Brun S, Théodore L, Mignotte B. Bcl-2 and Bax mammalian regulators of apoptosis are functional in Drosophila. Cell Death Differ 2000;7:804-14.  Back to cited text no. 33
    
34.
Montalto FI, De Amicis F. Cyclin D1 in cancer: A molecular connection for cell cycle control, adhesion and invasion in tumor and stroma. Cells 2020;9:2648.  Back to cited text no. 34
    
35.
Lee Y, Kim YJ, Kim MH, Kwak JM. MAPK cascades in guard cell signal transduction. Front Plant Sci 2016;7:80.  Back to cited text no. 35
    
36.
Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med 2020;19:1997-2007.  Back to cited text no. 36
    
37.
Janssen M, Schmidt C, Bruch PM, Blank MF, Rohde C, Waclawiczek A, et al. Venetoclax synergizes with gilteritinib in FLT3 wild-type high-risk acute myeloid leukemia by suppressing MCL-1. Blood 2022;140:2594-610.  Back to cited text no. 37
    
38.
He M, Jia Y, Wang Y, Cai X. Dysregulated MAPK signaling pathway in acute myeloid leukemia with RUNX1 mutations. Expert Rev Hematol 2022;15:769-79.  Back to cited text no. 38
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5]
 
 
    Tables

  [Table 1], [Table 2]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Materials and Me...
Results
Discussion
Conclusion
References
Article Figures
Article Tables

 Article Access Statistics
    Viewed648    
    Printed33    
    Emailed0    
    PDF Downloaded40    
    Comments [Add]    

Recommend this journal