Hostname: page-component-84b7d79bbc-rnpqb Total loading time: 0 Render date: 2024-07-26T04:43:20.893Z Has data issue: false hasContentIssue false

Identification of DUSP7 as an RNA Marker for Prognostic Stratification in Acute Myeloid Leukemia: Evidence from Large Population Cohorts

Published online by Cambridge University Press:  01 January 2024

Xin Gao*
Affiliation:
Anhui Medical College, Hefei, China
*
Correspondence should be addressed to Xin Gao; gaoxin@ahyz.edu.cn

Abstract

Background. The problem of prognostic stratification in acute myeloid leukemia (AML) patients still has limitations. Methods. The expression profile data and clinical features of AML patients were obtained from multiple publicly available sources, including GSE71014, TCGA-LAML, and TARGET-AML. Single-cell analysis was performed using the TISCH project. All the analysis was conducted in the R software. Results. In our study, three public AML cohorts, GSE71014, TARGET-AML, and TCGA-AML, were selected. Then, we identified the prognosis-related molecules through bioinformatic analysis. Finally, the DUSP7 was noticed as a risk factor for AML patients, which has not been reported previously. Biological enrichment analysis and immune-related analysis were performed to illustrate the role of DUSP7 in AML. Single-cell analysis indicated that the DUSP7 was widely distributed in various cells, especially in monocyte/macrophages and malignant. Following this, a prognosis model based on DUSP7-derived genes was constructed, which showed a good prognosis prediction ability in all cohorts. Conclusions. Our results preliminarily reveal the role and potential mechanism of DUSP7 in AML, providing direction for future research.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © 2023 Xin Gao.

1. Introduction

Acute myeloid leukemia (AML) is a highly heterogeneous malignant clonal disease that seriously affects human health [Reference De Kouchkovsky and Abdul-Hay1]. The pathogenesis of AML is extremely complex, and researchers have not yet fully elucidated it [Reference Bewersdorf and Abdel-Wahab2]. It is now recognized that chromosomal karyotype abnormalities and gene mutation reproducibility play an important initiating role in the occurrence and development of diseases [Reference Shi, Ma and Liu3]. By classifying these two factors, risk stratification can be performed on most patients. Based on the different risk stratification of patients, corresponding treatment plans can be adopted, which significantly improves the five-year survival rate of AML patients [Reference Newell and Cook4]. At the same time, with the development of chemotherapy, hematopoietic stem cell transplantation, gene targeting therapy, biological immunotherapy, and other treatment methods, the complete remission rate and 5-year disease-free survival rate of AML patients have improved, but most patients still have drug resistance and relapse, leading to poor prognosis [Reference Dohner, Estey and Amadori5]. These issues suggest that clinical doctors need to further explore the new pathogenesis of AML and improve the existing prognostic evaluation system.

At present, studies have found that the factors affecting the prognosis of AML patients not only include general clinical characteristics such as age and FAB typing, but also include molecular genetics, cytogenetics, and mutations of specific molecules [Reference George, Kantarjian, Baran, Krocker and Rios6]. For instance, Falini et al. noticed that the NPM1 mutation could assist in the genetic typing of AML, thereby indicating clinical prognosis and treatment choices [Reference Falini, Brunetti, Sportoletti and Martelli7]. Nowadays, many gene mutations have been linked to AML patients’ prognosis, such as MLL, HOX, RAR-α, CBF, NPM1, WNT, RUNX1, WT1, RB, PU.1, p53, MYC, MPL, JUNB, GATA-1, FOS, FES, N-RAS, CEBPA, KIT, and FLT3 [Reference Grove and Vassiliou8Reference Prada-Arismendy, Arroyave and Röthlisberger10]. Some mutations have been involved in the risk stratification of AML, such as NPM1, RUNX1, c-KIT, RUNX1-RUNX1T1, CEBPA, PML-RARα, BCR-ABL1, ASXL1, TP53, FLT3-ITD, and MLL [Reference Creutzig, Van Den Heuvel-Eibrink and Gibson11, Reference Dohner, Estey and Grimwade12]. However, many cases do not have the above gene mutations, especially 50% of AML patients with normal cytogenetics (CN-AML). These patients lack characteristic chromosome changes and clear molecular markers, and it is difficult to evaluate their prognosis [Reference Martelli, Sportoletti, Tiacci, Martelli and Falini13]. Numerous pieces of evidence suggest that gene expression levels are of great significance for the prognostic evaluation of AML [Reference Bauer, Vaxevanis and Heimer14]. As early as 1996, Bergmann et al. observed that the expression of WT-1 can serve as a prognostic and recurrence marker for AML [Reference Bergmann, Maurer and Weidmann15]. Paschka et al.’s study of 196 patients with CN-AML also supported the above viewpoint, finding that WT-1 mutation is an independent risk factor for CN-AML [Reference Paschka, Marcucci and Ruppert16]. Recent studies have shown that gene expression levels of CPT1A, TGFβ1, VEGF, CSRP2, and others are all associated with the prognosis of AML [Reference Shi, Fu, Jia, He, Fu and Wang17Reference Wang, Zhang and Liu19]. As is well known, the prognosis of diseases is influenced by multiple factors, and the relationships between these factors are complex. Therefore, traditional prognostic evaluation systems are no longer able to meet the requirements. With the rise of big data, bioinformatics, and artificial intelligence, their clinical applications in medicine are becoming increasingly widespread and showing great potential [Reference Ren, Zhang and Chen20Reference Zhang, Zhou and Zhang22].

In our study, three public AML cohorts, GSE71014, TARGET-AML, and TCGA-AML, were selected. Then, we identified the prognosis-related molecules through bioinformatic analysis. Finally, the DUSP7 was noticed as a risk factor for AML patients, which has not been reported previously. Biological enrichment analysis, single-cell analysis, and immune-related analysis were performed to illustrate the role of DUSP7 in AML. Following this, a prognosis model based on DUSP7-derived genes was constructed, which showed a good prognosis prediction ability in all cohorts.

2. Methods

2.1. Data Collection Process

The expression profile data and clinical features of AML patients were obtained from multiple publicly available sources. For TCGA-LAML (access time 22/3/2023) and TARGET-AML (access time 22/3/2023), the expression profile of each AML patient was downloaded in a “STAR-Count” form and then extracted into a merge matrix through R code. The human genomic reference file obtained from the Ensembl project (https://www.ensembl.org/) was utilized for probe annotation. Clinical information was downloaded in the “bcr-xml” form and organized using Perl code. For GSE71014, the expression profile of patients was downloaded from the direct link “Series Matrix File(s)” of the website https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE71014. Probe annotation was conducted by the GPL10558 platform. For all downloaded data, data quality control and standardization processing are carried out before data analysis. The interaction data of multiple proteins were downloaded from the STRING database and then visualized through Cytoscape software [Reference Shannon, Markiel and Ozier23].

2.2. Differentially Expressed Genes (DEGs) Analysis

DEGs analysis was performed using the limma package with the specific threshold (|logFC| > 1 and P value <0.05) [Reference Ritchie, Phipson and Wu24].

2.3. Prognosis Analysis

For a specific gene list, the expression profile of these genes was firstly combined with the corresponding survival information. Next, the molecules remarkably correlated with AML patients’ survival were identified using the univariate Cox regression analysis (P < 0.05). Furthermore, LASSO regression analysis was performed for variable optimization. Ultimately, multivariate Cox regression analysis was used to screen independent prognosis markers and model construction. The TCGA-AML cohort was set as the training cohort. The GSE71014 and TARGET-LAML cohorts were set as the validation cohort. Kaplan–Meier (KM) survival curves were used to compare the prognosis difference in different groups. The prediction ability of specific variables on patients’ survival was performed using the receiver operating characteristic (ROC) curves.

2.4. Biological Enrichment Analysis

Evaluation of biological function was performed using specific R packages. Clusterprofiler package was used for the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis [Reference Yu, Wang, Han and He25]. Gene set enrichment analysis (GSEA) was conducted based on the Hallmark gene set [Reference Subramanian, Tamayo and Mootha26].

2.5. Sing-Cell Analysis

The single-cell data of GSE116256, GSE135851, GSE154109, PBMC_8K, PBMC_30K, and PBMC_60K were analyzed in the TISCH project [Reference Sun, Wang and Han27].

2.6. Tissue Microenvironment Analysis

Estimate package was used to quantify the stromal score, immune score, and estimate score of the tissue microenvironment, respectively, representing the corresponding cell components [Reference Becht, Giraldo and Lacroix28]. The microenvironment of bone marrow was quantified using the single sample GSEA (ssGSEA) algorithm [Reference Hanzelmann, Castelo and Guinney29].

2.7. Statistical Analysis

All the statistical analyses were performed in the R software. Analysis with a statistic P value <0.05 was regarded as statistically significant. For data with different distributions, different analysis methods are used according to statistical requirements.

3. Results

The flowchart of the whole study is shown in Figure 1.

FIGURE 1: The flowchart of the whole study.

3.1. Identification of the Molecules Significantly Correlated with AML Patients’ Survival

Firstly, after a comprehensive search, we have finally identified three AML datasets with complete transcriptional profiles and clinical information, including GSE71014, TCGA-LAML, and TARGET-AML (Figure 2(a)). Then, we performed univariate Cox regression analysis to identify the prognosis-related molecules in these three cohorts (Supplementary file 1–3). By taking intersections, we noticed that 169 molecules were common risk factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts, while 36 molecules were common protective factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts (Figures 2(b) and 2(c)). The PPI network of the above prognosis-related molecules is shown in Figure 2(d).

FIGURE 2: Identification of prognosis-related molecules in three AML cohorts. (a) Three AML cohorts were identified, including GSE71014, TARGET-AML, and TCGA-LAML; (b) 169 molecules were common risk factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts; (c) 36 molecules were common protective factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts; (d) PPI network of prognosis-related molecules.

3.2. DUSP7 Is Associated with Poor Prognosis of AML Patients

Then, based on the identified risk genes, we performed LASSO regression analysis to optimize variables (Figures 3(a) and 3(b)). Following this, multivariate Cox regression analysis was performed, and we noticed that the SOCS2, DUSP7, and DDIT4 were significantly correlated with patients’ prognosis (P < 0.001) (Figure 3(c)). Considering the DUSP7 has not been reported in AML, we selected it for further analysis. KM survival curves in GSE71014, TCGA-LAML, and TARGET-AML cohorts all showed that the AML patients with high DUSP7 expression might have a poor prognosis (Figure 3(d), GSE71014, HR = 2.64, P = 0.009; Figure 3(e), TARGET-AML, HR = 1.72, P < 0.001; Figure 3(f), TCGA-LAML, HR = 2.27, P < 0.001).

FIGURE 3: DUSP7 is associated with poor prognosis of AML patients. (a, b) LASSO regression analysis; (c) multivariate Cox regression analysis of prognosis-related molecules; (d) KM survival curves of DUSP7 in GSE71014 cohort; (e) KM survival curves of DUSP7 in TARGET-AML cohort; (f) KM survival curves of DUSP7 in TCGA-LAML cohort.

3.3. Biological Enrichment of DUSP7 in AML and Single-Cell Analysis

Then, we tried to explore the potential biological function of DUSP7 in AML. Limma package was used to screen the DEGs in patients with high and low DUSP7 expression (Figure 4(a)). The heatmap of these DEGs is shown in Figure 4(b). KEGG analysis showed that the molecules upregulated in patients with high DUSP7 levels were mainly enriched in transcriptional misregulation in cancer, systemic lupus erythematosus, salivary secretion, renin-angiotensin system, and relaxin signaling pathway (Figure 4(c)). For GO analysis, the terms of response to fungus, neutrophil-mediated cytotoxicity, neutrophil degranulation, and neutrophil activation-related pathways were enriched (Figure 4(d)). As for the downregulated genes, the terms of viral protein interaction, cytokine and cytokine receptor, tuberculosis, and transcriptional misregulation in cancer were enriched by the KEGG analysis (Figure 4(e)). For GO analysis, these downregulated genes were enriched in the regulation of regulatory T-cell differentiation, mononuclear cell proliferation, lymphocyte proliferation, leukocyte proliferation, and leukocyte cell-cell adhesion (Figure 4(f)). Based on the GSEA, we found that the IL6_JAK_STAT3 signaling, interferon alpha response, bile acid metabolism, coagulation, and UV response DN were the top five pathways DUSP7 involved in (Figure 5(a)). Single-cell analysis indicated that the DUSP7 was widely distributed in various cells, especially in monocyte/macrophages and malignant (Figure 5(b)).

FIGURE 4: Biological enrichment analysis of DUSP7. (a) DEGs analysis in patients with high and low DUSP7 expression; (b) the heatmap of the DEGs; (c) KEGG analysis of the molecules upregulated in patients with high DUSP7 expression; (d) GO analysis of the molecules upregulated in patients with high DUSP7 expression; (e) KEGG analysis of the molecules downregulated in patients with high DUSP7 expression; (f) GO analysis of the molecules downregulated in patients with high DUSP7 expression.

FIGURE 5: Single-cell analysis of DUSP7. (a) GSEA of DUSP7 based on hallmark gene set; (b–g) single-cell analysis of DUSP7 in multiple cohorts.

3.4. Tissue Microenvironment Analysis

Then, we tried to explore the effect of DUSP7 on the AML microenvironment. Interestingly, we found that the DUSP7 was positively correlated with the immune score, stromal score, and estimate score according to the Estimate package (Figure 6(a), immune score, cor = 0.367, P < 0.001; Figure 6(b), stromal score, cor = 0.204, P = 0.012; Figure 6(c), estimate score, cor = 0.327, P < 0.001). Next, the ssGSEA algorithm was used to quantify the tissue microenvironment of AML (Figure 6(d)). Correlation analysis showed that DUSP7 was positively correlated with B cells, NK CD56 dim cells, Tem, Th17 cells, macrophages, and Treg (Figure 6(e)).

FIGURE 6: Immune-related analysis of DUSP7. (a) Correlation between DUSP7 and immune score quantified by Estimate package; (b) correlation between DUSP7 and stromal score quantified by Estimate package; (c) correlation between DUSP7 and estimate score quantified by Estimate package; (d) tissue microenvironment of AML was quantified by ssGSEA algorithm; (e) correlation between DUSP7 and quantified cells.

3.5. Establishment of a Prognosis Model Based on the DUSP7-Derived Molecules

We then identified the top 100 molecules positively and negatively correlated with DUSP7 in AML patients (Figures 7(a) and 7(b)). Univariate Cox regression analysis was performed to identify the prognosis-related genes (Figure 7(c)). LASSO regression analysis was then conducted to reduce data dimensions and optimize variables (Figures 7(d) and 7(e)). Multivariate Cox regression analysis was applied to construct a prognosis model based on the molecules identified by LASSO regression analysis, including DOC2B, TGIF1, TWIST1, SNORA38, HPS6, and PARP3 (Figure 7(f)). The risk score of each patient was calculated with the formula of “Risk score = DOC2B ∗ − 0.229 + TGIF1 ∗ − 0.443 + TWIST1 ∗ − 0.382 + SNORA 38 ∗ − 0.260 + HPS6 ∗ 0.514 + PARP3 ∗ 0.385”. We noticed a higher proportion of death cases in high-risk patients (Figure 8(a)). KM survival curves indicated that high-risk patients might have a worse prognosis (Figure 8(b), HR = 3.70, P < 0.001). ROC curves indicated that the 1-year AUC = 0.809, 3-year AUC = 0.851, and 5-year AUC = 0.935 (Figures 8(c)8(e)). In the validation cohorts (GSE71014 and TARGET-AML), our model also showed a good prognosis prediction ability (Figure 8(f), HR = 2.34, P = 0.017, 1-year AUC = 0.687, 3-year AUC = 0.707, 5-year AUC = 0.897; Figure 8(g), HR = 2.57, P < 0.001, 1-year AUC = 0.677, 3-year AUC = 0.696, 5-year AUC = 0.686).

FIGURE 7: Construction of a prognosis model based on DUSP7-derived genes in AML. (a) The top 100 genes positively correlated with DUSP7 in AML; (b) the top 100 genes negatively correlated with DUSP7 in AML; (c) univariate Cox regression analysis of the genes significantly correlated with DUSP7 in AML; (d)-(e): LASSO regression analysis; (f) multivariate Cox regression analysis.

FIGURE 8: Evaluation of the prognosis model. (a) The overview of the prognosis model; (b) KM survival curve of patients in high- and low-risk groups; (c–e) ROC curves of 1, 3, and 5 years in TCGA-LAML cohort; (f) evaluation of our model in GSE71014; (g) evaluation of our model in TARGET-AML cohort.

4. Discussion

AML is a highly heterogeneous hematological malignancy caused by dysgenesis of myeloid hematopoietic stem/progenitor cells, characterized by abnormal clonal proliferation and differentiation arrest of primitive and immature myeloid cells in the bone marrow and peripheral blood [Reference Liu30Reference Cao, Shu and Wang32]. AML has complex cytogenetic and epigenetic mutations, including abnormal DNA methylation, resulting in high heterogeneity in disease diagnosis, development, and prognosis [Reference Schoofs and Müller-Tidow33]. With the continuous research on AML, it is found that abnormal DNA methylation of special gene promoters affects the change of its expression level, especially tumor suppressor, which leads to the occurrence and development of disease [Reference Cammenga34]. Although various molecular targeted therapies for AML have emerged one after another, increasing the possibility of selecting treatment options for AML, its 5-year survival rate is still very poor, especially for elderly patients with high-risk factors [Reference Rauch, Ellegast and Widmer35]. Therefore, an in-depth exploration of the pathogenesis of AML has important scientific significance and clinical application value for disease prevention, diagnosis, specific treatment, and prognosis stratification.

In our study, three public AML cohorts, GSE71014, TARGET-AML, and TCGA-AML, were selected. Then, we identified the prognosis-related molecules through bioinformatic analysis. Finally, the DUSP7 was noticed as a risk factor for AML patients, which has not been reported previously. Biological enrichment analysis, single-cell analysis, and immune-related analysis were performed to illustrate the role of DUSP7 in AML. Following this, a prognosis model based on DUSP7-derived genes was constructed, which showed a good prognosis prediction ability in all cohorts.

DUSP7, whose full name is dual specificity phosphatase 7, has been reported involved in various pathophysiological processes, especially in cancers [Reference Vasilaki, Morikawa and Koinuma36Reference Xu, Song, Gokulnath, Vulugundam and Xiao38]. For instance, Li et al. found that DUSP7 could affect the dephosphorylation of PEA15 and resistance to breast cancer [Reference Li, Wang, Xiong, Guo, Zhu and Gu39]. Guo et al. have proved that DUSP7 is extremely important for the correct chromosome arrangement during cell division through a series of experiments at the cellular level [Reference Guo, Ramirez and Garcia40]. Moreover, Tischer and Schuh also supported this viewpoint at the animal level [Reference Tischer and Schuh41]. It is worth noting that over half of AML patients are accompanied by chromosomal abnormalities, and the promoting effect of DUSP7 on AML seems understandable, but the relevant mechanisms still need further exploration.

Biological enrichment analysis showed that the IL6_JAK_STAT3 signaling, interferon alpha response, bile acid metabolism, coagulation, and UV response DN were the top five pathways DUSP7 was involved in. This suggests the importance of metabolism and immunity in AML. AML cells have an atypical metabolic phenotype, characterized by increased mitochondrial mass, and more dependence on oxidative phosphorylation and fatty acid oxidation for survival. Tcheng et al. indicated that AML has a special fatty acid metabolism manner and level [Reference Tcheng, Roma and Ahmed42]. Moreover, in a retrospective study involving 84 patients, researchers found that IFN-α can effectively reduce the recurrence rate of AML patients [Reference Jiang, Liu and Kong43]. Also, based on the long-term results of two registered studies, IFN-α can prevent recurrence and improve the survival rate [Reference Shen, Zhang and Xu44]. Therefore, the correlation between DUSP7 and IFN-α response can partially explain the impact of DUSP7 on the prognosis of AML patients. However, further research is needed to confirm the relationship between DUSP7 and IFN-α response.

Immune-related analysis showed that the DUSP7 was positively correlated with B cells, NK CD56 dim cells, Tem, Th17 cells, macrophages, and Treg. Some studies have begun to focus on the role of immune cells in AML. Goswami et al. found that after chemotherapy, AML patients experienced irreversible immune damage to their B cells [Reference Goswami, Prince and Biancotto45]. Meanwhile, researchers also found that regulatory B cells have clinical indicative significance in AML [Reference Lv, Wang and Liu46, Reference Zahran, Shibl and Rayan47]. Moore et al. found that the AML progression could be suppressed by LC3-associated phagocytosis medicated by bone marrow macrophages [Reference Moore, Mistry and Hellmich48]. Liu et al. demonstrated that chenodeoxycholic acid can inhibit M2 macrophage polarization through lipid peroxidation [Reference Liu, Wei and Jia49]. Moreover, Treg is believed to play a role in helping leukemia cells evade immune surveillance, thereby promoting AML progression [Reference Mansour, Zayed, Said and Latif50, Reference Swatler, Turos-Korgul and Brewinska-Olchowik51]. Our results suggest that DUSP7 may affect AML progression by affecting the recruitment of local immune cells.

The continuous development of bioinformatics and the massive data in the era of big data have improved the convenience of data secondary mining. Although our results are based on reliable data and algorithms, some limitations cannot be ignored. Firstly, most patients are of Western ethnicity. There are specific differences in genomics among populations of different ethnicities; therefore, racial bias will reduce the universality of our conclusions. Secondly, it cannot be denied that bioinformatics analysis cannot fully reflect the true situation in the body. Therefore, most conclusions only have indicative significance and still require subsequent experimental verification.

Data Availability

All raw data can be obtained from the corresponding author based on reasonable requirements.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Acknowledgments

This study was funded by the Key Funding Project for Teaching and Research of Anhui Provincial Department of Education (2022jyxm796); Anhui Medical College Curriculum Ideological and Political Integration General Practitioner Training Innovation Team Funding Project (YZ2020TD005).

Supplementary Materials

Supplementary file 1: results of univariate Cox regression analysis in GSE71014. Supplementary file 2: results of univariate Cox regression analysis in TARGET-LAM. Supplementary file 3: results of univariate Cox regression analysis in TCGA-LAML. (Supplementary Materials)

References

De Kouchkovsky, I. and Abdul-Hay, M., “Acute myeloid leukemia: a comprehensive review and 2016 update,Blood Cancer Journal, vol. 6, no. 7, p. e441, 2016.CrossRefGoogle ScholarPubMed
Bewersdorf, J. P. and Abdel-Wahab, O., “Translating recent advances in the pathogenesis of acute myeloid leukemia to the clinic,Genes & Development, vol. 36, no. 5-6, pp. 259277, 2022.CrossRefGoogle ScholarPubMed
Shi, L. H., Ma, P., Liu, J. S. et al., “Current views of chromosomal abnormalities in pediatric acute myeloid leukemia (AML),European Review for Medical and Pharmacological Sciences, vol. 21, no. 4, pp. 2530, 2017.Google ScholarPubMed
Newell, L. F. and Cook, R. J., “Advances in acute myeloid leukemia,BMJ, vol. 375, p. n2026, 2021.CrossRefGoogle ScholarPubMed
Dohner, H., Estey, E. H., Amadori, S. et al., “Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet,Blood, vol. 115, no. 3, pp. 453474, 2010.CrossRefGoogle Scholar
George, B., Kantarjian, H., Baran, N., Krocker, J. D., and Rios, A., “TP53 in acute myeloid leukemia: molecular aspects and patterns of mutation,International Journal of Molecular Sciences, vol. 22, no. 19, Article ID 10782, 2021.CrossRefGoogle ScholarPubMed
Falini, B., Brunetti, L., Sportoletti, P., and Martelli, M. P., “NPM1-mutated acute myeloid leukemia: from bench to bedside,Blood, vol. 136, no. 15, pp. 17071721, 2020.CrossRefGoogle ScholarPubMed
Grove, C. S. and Vassiliou, G. S., “Acute myeloid leukaemia: a paradigm for the clonal evolution of cancer?,Disease Models & Mechanisms, vol. 7, no. 8, pp. 941951, 2014.CrossRefGoogle ScholarPubMed
Meyer, S. C. and Levine, R. L., “Translational implications of somatic genomics in acute myeloid leukaemia,The Lancet Oncology, vol. 15, no. 9, pp. e382e394, 2014.CrossRefGoogle ScholarPubMed
Prada-Arismendy, J., Arroyave, J. C., and Röthlisberger, S., “Molecular biomarkers in acute myeloid leukemia,Blood Reviews, vol. 31, no. 1, pp. 6376, 2017.CrossRefGoogle ScholarPubMed
Creutzig, U., Van Den Heuvel-Eibrink, M. M., Gibson, B. et al., “Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel,Blood, vol. 120, no. 16, pp. 31873205, 2012.CrossRefGoogle ScholarPubMed
Dohner, H., Estey, E., Grimwade, D. et al., “Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel,Blood, vol. 129, no. 4, pp. 424447, 2017.CrossRefGoogle ScholarPubMed
Martelli, M. P., Sportoletti, P., Tiacci, E., Martelli, M. F., and Falini, B., “Mutational landscape of AML with normal cytogenetics: biological and clinical implications,Blood Reviews, vol. 27, no. 1, pp. 1322, 2013.CrossRefGoogle ScholarPubMed
Bauer, M., Vaxevanis, C., Heimer, N. et al., “Expression, regulation and function of microRNA as important players in the transition of MDS to secondary AML and their cross talk to RNA-binding proteins,International Journal of Molecular Sciences, vol. 21, no. 19, p. 7140, 2020.CrossRefGoogle ScholarPubMed
Bergmann, L., Maurer, U., and Weidmann, E., “Wilms tumor gene expression in acute myeloid leukemias,Leukemia and Lymphoma, vol. 25, no. 5-6, pp. 435443, 1997.CrossRefGoogle ScholarPubMed
Paschka, P., Marcucci, G., Ruppert, A. S. et al., “Wilms' tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study,Journal of Clinical Oncology, vol. 26, no. 28, pp. 45954602, 2008.CrossRefGoogle ScholarPubMed
Shi, J., Fu, H., Jia, Z., He, K., Fu, L., and Wang, W., “High expression of CPT1A predicts adverse outcomes: a potential therapeutic target for acute myeloid leukemia,EBioMedicine, vol. 14, pp. 5564, 2016.CrossRefGoogle ScholarPubMed
Jabari, M., Allahbakhshian Farsani, M., Salari, S., Hamidpour, M., Amiri, V., and Mohammadi, M. H., “Hypoxia-inducible factor1-Α (HIF1α) and vascular endothelial growth factor-A (VEGF-A) expression in de novo AML patients,Asian Pacific Journal of Cancer Prevention, vol. 20, no. 3, pp. 705710, 2019.CrossRefGoogle ScholarPubMed
Wang, S., Zhang, Y., Liu, Y. et al., “Inhibition of CSRP2 promotes leukemia cell proliferation and correlates with relapse in adults with acute myeloid leukemia,OncoTargets and Therapy, vol. 13, Article ID 12549, 12560 pages, 2020.CrossRefGoogle ScholarPubMed
Ren, X., Zhang, T., Chen, X. et al., “Early-life exposure to bisphenol A and reproductive-related outcomes in rodent models: a systematic review and meta-analysis,Aging (Albany NY), vol. 12, no. 18, Article ID 18099, 18126 pages, 2020.Google ScholarPubMed
Zhang, D., Xu, X., Wei, Y. et al., “Prognostic role of DNA damage response genes mutations and their association with the sensitivity of olaparib in prostate cancer patients,Cancer Control, vol. 29, Article ID 107327482211294, 2022.CrossRefGoogle ScholarPubMed
Zhang, T., Zhou, X., Zhang, X. et al. “Gut microbiota may contribute to the postnatal male reproductive abnormalities induced by prenatal dibutyl phthalate exposure,Chemosphere, vol. 287, Article ID 132046, 2022.10.1016/j.chemosphere.2021.132046CrossRefGoogle Scholar
Shannon, P., Markiel, A., Ozier, O. et al., “Cytoscape: a software environment for integrated models of biomolecular interaction networks,Genome Research, vol. 13, no. 11, pp. 24982504, 2003.CrossRefGoogle ScholarPubMed
Ritchie, M. E., Phipson, B., Wu, D. et al., “Limma powers differential expression analyses for RNA-sequencing and microarray studies,Nucleic Acids Research, vol. 43, no. 7, p. e47, 2015.CrossRefGoogle ScholarPubMed
Yu, G., Wang, L. G., Han, Y., and He, Q. Y., “clusterProfiler: an R package for comparing biological themes among gene clusters,OMICS: A Journal of Integrative Biology, vol. 16, no. 5, pp. 284287, 2012.CrossRefGoogle Scholar
Subramanian, A., Tamayo, P., Mootha, V. K. et al “Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles,Proceedings of the National Academy of Sciences of the U S A, vol. 102, no. 43, Article ID 15545, 15550 pages, 2005.CrossRefGoogle ScholarPubMed
Sun, D., Wang, J., Han, Y. et al., “TISCH: a comprehensive web resource enabling interactive single-cell transcriptome visualization of tumor microenvironment,Nucleic Acids Research, vol. 49, no. 1, pp. D1420d1430, 2021.CrossRefGoogle Scholar
Becht, E., Giraldo, N. A., Lacroix, L. et al., “Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression,Genome Biology, vol. 17, no. 1, p. 218, 2016.CrossRefGoogle ScholarPubMed
Hanzelmann, S., Castelo, R., and Guinney, J., “GSVA: gene set variation analysis for microarray and RNA-seq data,BMC Bioinformatics, vol. 14, no. 1, p. 7, 2013.CrossRefGoogle ScholarPubMed
Liu, H., “Emerging agents and regimens for AML,Journal of Hematology & Oncology, vol. 14, no. 1, p. 49, 2021.10.1186/s13045-021-01062-wCrossRefGoogle ScholarPubMed
Willier, S., Rothämel, P., Hastreiter, M. et al., “CLEC12A and CD33 coexpression as a preferential target for pediatric AML combinatorial immunotherapy,Blood, vol. 137, no. 8, pp. 10371049, 2021.10.1182/blood.2020006921CrossRefGoogle ScholarPubMed
Cao, Z., Shu, Y., Wang, J. et al., “Super enhancers: pathogenic roles and potential therapeutic targets for acute myeloid leukemia (AML),Genes & Diseases, vol. 9, no. 6, pp. 14661477, 2022.CrossRefGoogle ScholarPubMed
Schoofs, T. and Müller-Tidow, C., “DNA methylation as a pathogenic event and as a therapeutic target in AML,Cancer Treatment Reviews, vol. 37, no. 1, pp. S13S18, 2011.CrossRefGoogle ScholarPubMed
Cammenga, J., “Gatekeeper pathways and cellular background in the pathogenesis and therapy of AML,Leukemia, vol. 19, no. 10, pp. 17191728, 2005.CrossRefGoogle ScholarPubMed
Rauch, P. J., Ellegast, J. M., Widmer, C. C. et al., “MPL expression on AML blasts predicts peripheral blood neutropenia and thrombocytopenia,Blood, vol. 128, no. 18, pp. 22532257, 2016.CrossRefGoogle ScholarPubMed
Vasilaki, E., Morikawa, M., Koinuma, D. et al., “Ras and TGF-β signaling enhance cancer progression by promoting the ΔNp63 transcriptional program,Science Signaling, vol. 9, no. 442, p. ra84, 2016.CrossRefGoogle ScholarPubMed
Chen, H. F., Chuang, H. C., and Tan, T. H., “Regulation of dual-specificity phosphatase (DUSP) ubiquitination and protein stability,International Journal of Molecular Sciences, vol. 20, no. 11, p. 2668, 2019.CrossRefGoogle ScholarPubMed
Xu, G. E., Song, J., Gokulnath, P., Vulugundam, G., and Xiao, J., “DUSP7: an unusual player in adverse cardiac remodeling,Journal of Cardiovascular Translational Research, vol. 15, no. 6, pp. 12171218, 2022.CrossRefGoogle ScholarPubMed
Li, L., Wang, N., Xiong, Y., Guo, G., Zhu, M., and Gu, Y., “Transcription factor FOSL1 enhances drug resistance of breast cancer through DUSP7-mediated dephosphorylation of PEA15,Molecular Cancer Research, vol. 20, no. 4, pp. 515526, 2022.CrossRefGoogle ScholarPubMed
Guo, X., Ramirez, I., Garcia, Y. A. et al., “DUSP7 regulates the activity of ERK2 to promote proper chromosome alignment during cell division,Journal of Biological Chemistry, vol. 296, Article ID 100676, 2021.CrossRefGoogle ScholarPubMed
Tischer, T. and Schuh, M., “The phosphatase Dusp7 drives meiotic resumption and chromosome alignment in mouse oocytes,Cell Reports, vol. 17, no. 5, pp. 14261437, 2016.CrossRefGoogle ScholarPubMed
Tcheng, M., Roma, A., Ahmed, N. et al., “Very long chain fatty acid metabolism is required in acute myeloid leukemia,Blood, vol. 137, no. 25, pp. 35183532, 2021.CrossRefGoogle ScholarPubMed
Jiang, H., Liu, X. H., Kong, J. et al., “Interferon-α as maintenance therapy can significantly reduce relapse in patients with favorable-risk acute myeloid leukemia,Leukemia and Lymphoma, vol. 62, no. 12, pp. 29492956, 2021.CrossRefGoogle ScholarPubMed
Shen, M. Z., Zhang, X. H., Xu, L. P. et al., “Preemptive interferon-α therapy could protect against relapse and improve survival of acute myeloid leukemia patients after allogeneic hematopoietic stem cell transplantation: long-term results of two registry studies,Frontiers in Immunology, vol. 13, Article ID 757002 2022.Google ScholarPubMed
Goswami, M., Prince, G., Biancotto, A. et al., “Impaired B cell immunity in acute myeloid leukemia patients after chemotherapy,Journal of Translational Medicine, vol. 15, no. 1, p. 155, 2017.CrossRefGoogle ScholarPubMed
Lv, Y., Wang, H., and Liu, Z., “The role of regulatory B cells in patients with acute myeloid leukemia,Medical Science Monitor, vol. 25, pp. 30263031, 2019.CrossRefGoogle ScholarPubMed
Zahran, A. M., Shibl, A., Rayan, A. et al., “Increase in polymorphonuclear myeloid-derived suppressor cells and regulatory T-cells in children with B-cell acute lymphoblastic leukemia,Scientific Reports, vol. 11, no. 1, Article ID 15039, 2021.CrossRefGoogle ScholarPubMed
Moore, J. A., Mistry, J. J., Hellmich, C. et al., “LC3-associated phagocytosis in bone marrow macrophages suppresses acute myeloid leukemia progression through STING activation,The Journal of Clinical Investigation, vol. 132, no. 5, Article ID e153157, 2022.CrossRefGoogle ScholarPubMed
Liu, J., Wei, Y., Jia, W. et al. “Chenodeoxycholic acid suppresses AML progression through promoting lipid peroxidation via ROS/p38 MAPK/DGAT1 pathway and inhibiting M2 macrophage polarization,Redox Biology, vol. 56, Article ID 102452, 2022.CrossRefGoogle ScholarPubMed
Mansour, I., Zayed, R. A., Said, F., and Latif, L. A., “Indoleamine 2,3-dioxygenase and regulatory T cells in acute myeloid leukemia,Hematology, vol. 21, no. 8, pp. 447453, 2016.CrossRefGoogle ScholarPubMed
Swatler, J., Turos-Korgul, L., Brewinska-Olchowik, M. et al., “4-1BBL-containing leukemic extracellular vesicles promote immunosuppressive effector regulatory T cells,Blood Advances, vol. 6, pp. 18791894, 2022.CrossRefGoogle ScholarPubMed
Figure 0

FIGURE 1: The flowchart of the whole study.

Figure 1

FIGURE 2: Identification of prognosis-related molecules in three AML cohorts. (a) Three AML cohorts were identified, including GSE71014, TARGET-AML, and TCGA-LAML; (b) 169 molecules were common risk factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts; (c) 36 molecules were common protective factors in GSE71014, TCGA-LAML, and TARGET-AML cohorts; (d) PPI network of prognosis-related molecules.

Figure 2

FIGURE 3: DUSP7 is associated with poor prognosis of AML patients. (a, b) LASSO regression analysis; (c) multivariate Cox regression analysis of prognosis-related molecules; (d) KM survival curves of DUSP7 in GSE71014 cohort; (e) KM survival curves of DUSP7 in TARGET-AML cohort; (f) KM survival curves of DUSP7 in TCGA-LAML cohort.

Figure 3

FIGURE 4: Biological enrichment analysis of DUSP7. (a) DEGs analysis in patients with high and low DUSP7 expression; (b) the heatmap of the DEGs; (c) KEGG analysis of the molecules upregulated in patients with high DUSP7 expression; (d) GO analysis of the molecules upregulated in patients with high DUSP7 expression; (e) KEGG analysis of the molecules downregulated in patients with high DUSP7 expression; (f) GO analysis of the molecules downregulated in patients with high DUSP7 expression.

Figure 4

FIGURE 5: Single-cell analysis of DUSP7. (a) GSEA of DUSP7 based on hallmark gene set; (b–g) single-cell analysis of DUSP7 in multiple cohorts.

Figure 5

FIGURE 6: Immune-related analysis of DUSP7. (a) Correlation between DUSP7 and immune score quantified by Estimate package; (b) correlation between DUSP7 and stromal score quantified by Estimate package; (c) correlation between DUSP7 and estimate score quantified by Estimate package; (d) tissue microenvironment of AML was quantified by ssGSEA algorithm; (e) correlation between DUSP7 and quantified cells.

Figure 6

FIGURE 7: Construction of a prognosis model based on DUSP7-derived genes in AML. (a) The top 100 genes positively correlated with DUSP7 in AML; (b) the top 100 genes negatively correlated with DUSP7 in AML; (c) univariate Cox regression analysis of the genes significantly correlated with DUSP7 in AML; (d)-(e): LASSO regression analysis; (f) multivariate Cox regression analysis.

Figure 7

FIGURE 8: Evaluation of the prognosis model. (a) The overview of the prognosis model; (b) KM survival curve of patients in high- and low-risk groups; (c–e) ROC curves of 1, 3, and 5 years in TCGA-LAML cohort; (f) evaluation of our model in GSE71014; (g) evaluation of our model in TARGET-AML cohort.

Supplementary material: File

Gao and Liu supplementary material

Supplementary files 1-3
Download Gao and Liu supplementary material(File)
File 6.3 MB