High expression of COMMD7 is an adverse prognostic factor in acute myeloid leukemia

Kongfei Li, Lieguang Chen, Hua Zhang, Lu Wang, Keya Sha, Xiaohong Du, Daiyang Li, Zhongzheng Zheng, Renzhi Pei, Ying Lu, Hongyan Tong, Kongfei Li, Lieguang Chen, Hua Zhang, Lu Wang, Keya Sha, Xiaohong Du, Daiyang Li, Zhongzheng Zheng, Renzhi Pei, Ying Lu, Hongyan Tong

Abstract

Acute myeloid leukemia (AML) is a frequent malignancy in adults worldwide; identifying preferable biomarkers has become one of the current challenges. Given that COMMD7 has been reported associated with tumor progression in various human solid cancers but rarely reported in AML, herein, RNA sequencing data from TCGA and GTEx were obtained for analysis of COMMD7 expression and differentially expressed gene (DEG). Furthermore, functional enrichment analysis of COMMD7-related DEGs was performed by GO/KEGG, GSEA, immune cell infiltration analysis, and protein-protein interaction (PPI) network. In addition, the clinical significance of COMMD7 in AML was figured out by Kaplan-Meier Cox regression and prognostic nomogram model. R package was used to analyze incorporated studies. As a result, COMMD7 was highly expressed in various malignancies, including AML, compared with normal samples. Moreover, high expression of COMMD7 was associated with poor prognosis in 151 AML samples, as well as subgroups with age >60, NPM1 mutation-positive, FLT3 mutation-negative, and DNMT3A mutation-negative, et al. (P < 0.05). High COMMD7 was an independent prognostic factor in Cox regression analysis; Age and cytogenetics risk were included in the nomogram prognostic model. Furthermore, a total of 529 DEGs were identified between the high- and the low- expression group, of which 92 genes were up-regulated and 437 genes were down-regulated. Collectively, high expression of COMMD7 is a potential biomarker for adverse outcomes in AML. The DEGs and pathways recognized in the study provide a preliminary grasp of the underlying molecular mechanisms of AML carcinogenesis and progression.

Keywords: COMMD7; R packages; The Cancer Genome Atlas; acute myeloid leukemia.

Conflict of interest statement

CONFLICTS OF INTEREST: The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
The higher expression of COMMD7 was showed in AML compared with normal samples. (A) Expression level of COMMD7 in paired normal and pan-cancer samples. (B) Expression level of COMMD7 in paired normal and AML samples. Analysis between two groups: Wilcoxon Rank sum test; NS: P 0.05 or higher; *P < 0.05; **P < 0.01; ***P < 0.001.
Figure 2
Figure 2
A total of 529 DEGs were identified as being statistically significant between COMMD7 high-expressed and low-expressed groups. (A) Volcano plot of differentially expressed genes, including 92 up-regulated and 437 down-regulated genes. Normalized expression levels were shown in descending order from green to red. (B) Heat map of the 10 differentially expressed RNAs, including 5 up-regulated genes and 5 down-regulated genes. The X-axis represents the samples, while the Y-axis denotes the differentially expressed RNAs. Green and red tones represented down-regulated and up-regulated genes, respectively.
Figure 3
Figure 3
GO/KEGG enrichment analysis of DEGs between high- and low- COMMD7 expression in TCGA-LAML patients. (A) Enriched GO terms in the “biological process” category; (B) Enriched GO terms in the “molecular function” category. (C) Enriched GO terms in the “cellular component” category; (D) KEGG pathway annotations. The X-axis represented the proportion of DEGs, and the Y-axis represented different categories. The different colors indicate different properties, and the different sizes represent the number of DEGs.
Figure 4
Figure 4
Enrichment plots from the gene set enrichment analysis (GSEA). (AL) ES, enrichment score; NES, normalized ES; ADJ P-val, adjusted P-value.
Figure 5
Figure 5
The expression of COMMD7 was associated with immune infiltration in the AML microenvironment. (A), The forest plots showed a positive correlation between COMMD7 and 13 immune cells, and a negative correlation between COMMD7 and 11 immune cell subsets. The size of dots showed the absolute value of Spearman r. (B) Correlation between the relative enrichment score of NK CD56(bright) cells and the expression level (TPM) of COMMD7. (C) Infiltration of NK CD56(bright) cells between low- and high-COMMD7 expressed.
Figure 6
Figure 6
The PPI network of COMMD7-related DEGs and the most significant module. (A) The PPI network of DEGs was constructed using Cytoscape. (B) The most significant module was obtained from PPI network with 42 nodes and 150 edges.
Figure 7
Figure 7
Association between COMMD7 expression and clinical features and cytogenetic risks. (A) The diagnostic efficacy of COMMD7 in acute myelogenous leukemia analyzed by ROC. (BH) Association between COMMD7 expression and BM blasts (20%), WBC counts (20 × 109), FAB classification, cytogenetics risk, NPM1 mutation, FLT3 mutation, and IDH1 R132 mutation analyzed by using Wilcoxon Rank SUM test.
Figure 8
Figure 8
High expression of COMMD7 was associated with poor OS in AML patients. (A) Kaplan-Meier curves in all AML patients. (B) Kaplan-Meier curves in AML patients with BM blasts > 20%. (C) Kaplan-Meier curves in AML patients with PB blasts ≤ 70%. (D) Kaplan-Meier curves in AML patients with age ≥ 60. (EL) Kaplan-Meier curves in subgroups with FLT3 mutation-negative, IDH1 R132 mutation-positive, IDH1 R140 mutation-positive, R172 mutation-positive, NPM1 mutation-positive, RAS mutation-positive, RUX1 mutation-negative, and DNMT3A mutation-negative in AML patients.
Figure 9
Figure 9
Forest plot showed that COMMD7 predicted poor prognosis in the subgroup of WBC count (>20 × 109/L) (HR = 2.062, P = 0.030), BM blasts (>20%) (HR = 1.897, P = 0.024), PB blasts (>70%) (HR = 2.435, P = 0.007), FLT3 mutation negative (HR = 3.330, p = 0.009), and NPM1 mutation positive (HR = 2.345, P < 0.001).
Figure 10
Figure 10
A prognostic predictive model of COMMD7 in AML. (A) Nomogram for predicting the probability of 1-, 3-, 5-year OS for AML. (B) Calibration plot of the nomogram for predicting the probability of OS at 1, 3, and 5 years.

References

    1. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, et al.. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med. 2016; 374:2209–21. 10.1056/NEJMoa1516192
    1. Lindsley RC, Mar BG, Mazzola E, Grauman PV, Shareef S, Allen SL, Pigneux A, Wetzler M, Stuart RK, Erba HP, Damon LE, Powell BL, Lindeman N, et al.. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015; 125:1367–76. 10.1182/blood-2014-11-610543
    1. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, Levine RL, Lo-Coco F, Naoe T, et al.. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017; 129:424–47. 10.1182/blood-2016-08-733196
    1. Burstein E, Hoberg JE, Wilkinson AS, Rumble JM, Csomos RA, Komarck CM, Maine GN, Wilkinson JC, Mayo MW, Duckett CS. COMMD proteins, a novel family of structural and functional homologs of MURR1. J Biol Chem. 2005; 280:22222–32. 10.1074/jbc.M501928200
    1. You N, Li J, Gong Z, Huang X, Wang W, Wang L, Wu K, Zheng L. COMMD7 functions as molecular target in pancreatic ductal adenocarcinoma. Mol Carcinog. 2017; 56:607–24. 10.1002/mc.22520
    1. You N, Li J, Huang X, Wu K, Tang Y, Wang L, Li H, Mi N, Zheng L. COMMD7 promotes hepatocellular carcinoma through regulating CXCL10. Biomed Pharmacother. 2017; 88:653–57. 10.1016/j.biopha.2017.01.046
    1. Esposito E, Napolitano G, Pescatore A, Calculli G, Incoronato MR, Leonardi A, Ursini MV. COMMD7 as a novel NEMO interacting protein involved in the termination of NF-κB signaling. J Cell Physiol. 2016; 231:152–61. 10.1002/jcp.25066
    1. Zheng L, You N, Huang X, Gu H, Wu K, Mi N, Li J. COMMD7 Regulates NF-κB Signaling Pathway in Hepatocellular Carcinoma Stem-like Cells. Mol Ther Oncolytics. 2018; 12:112–23. 10.1016/j.omto.2018.12.006
    1. Zheng L, Deng CL, Wang L, Huang XB, You N, Tang YC, Wu K, Liang P, Mi N, Li J. COMMD7 is correlated with a novel NF-κB positive feedback loop in hepatocellular carcinoma. Oncotarget. 2016; 7:32774–84. 10.18632/oncotarget.9047
    1. Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, Pfeil J, Narkizian J, Deran AD, Musselman-Brown A, Schmidt H, Amstutz P, Craft B, et al.. Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. 2017; 35:314–16. 10.1038/nbt.3772
    1. Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, Zhu J, Haussler D. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020; 38:675–78. 10.1038/s41587-020-0546-8
    1. Li K, Luo H, Luo H, Zhu X. Clinical and prognostic pan-cancer analysis of m6A RNA methylation regulators in four types of endocrine system tumors. Aging (Albany NY). 2020; 12:23931–44. 10.18632/aging.104064
    1. Wang JD, Zhou HS, Tu XX, He Y, Liu QF, Liu Q, Long ZJ. Prediction of competing endogenous RNA coexpression network as prognostic markers in AML. Aging (Albany NY). 2019; 11:3333–47. 10.18632/aging.101985
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15:550. 10.1186/s13059-014-0550-8
    1. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012; 16:284–87. 10.1089/omi.2011.0118
    1. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, Angell H, Fredriksen T, Lafontaine L, Berger A, Bruneval P, Fridman WH, Becker C, et al.. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013; 39:782–95. 10.1016/j.immuni.2013.10.003
    1. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ, Mering CV. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019; 47:D607–D613. 10.1093/nar/gky1131
    1. Demchak B, Hull T, Reich M, Liefeld T, Smoot M, Ideker T, Mesirov JP. Cytoscape: the network visualization tool for GenomeSpace workflows. F1000Res. 2014; 3:151. 10.12688/f1000research.4492.2
    1. Bandettini WP, Kellman P, Mancini C, Booker OJ, Vasu S, Leung SW, Wilson JR, Shanbhag SM, Chen MY, Arai AE. MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: a clinical validation study. J Cardiovasc Magn Reson. 2012; 14:83. 10.1186/1532-429X-14-83
    1. Isidro-Sánchez J, Akdemir D, Montilla-Bascón G. Genome-Wide Association Analysis Using R. Methods Mol Biol. 2017; 1536:189–207. 10.1007/978-1-4939-6682-0_14
    1. Falini B, Brunetti L, Sportoletti P, Martelli MP. NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood. 2020; 136:1707–21. 10.1182/blood.2019004226
    1. Kunchala P, Kuravi S, Jensen R, McGuirk J, Balusu R. When the good go bad: Mutant NPM1 in acute myeloid leukemia. Blood Rev. 2018; 32:167–83. 10.1016/j.blre.2017.11.001
    1. Patel SS, Kuo FC, Gibson CJ, Steensma DP, Soiffer RJ, Alyea EP 3rd, Chen YA, Fathi AT, Graubert TA, Brunner AM, Wadleigh M, Stone RM, DeAngelo DJ, et al.. High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML. Blood. 2018; 131:2816–25. 10.1182/blood-2018-01-828467
    1. Webersinke G, Kranewitter W, Deutschbauer S, Zach O, Hasenschwandtner S, Wiesinger K, Erdel M, Marschon R, Böhm A, Tschurtschenthaler G. Switch of the mutation type of the NPM1 gene in acute myeloid leukemia (AML): relapse or secondary AML? Blood Cancer J. 2014; 4:e221. 10.1038/bcj.2014.42
    1. Pepper M, Tan B. Acute myeloid leukemia with NPM1 and FLT3 ITD mimicking acute promyelocytic leukemia. Blood. 2020; 136:1467. 10.1182/blood.2020007198
    1. Cocciardi S, Dolnik A, Kapp-Schwoerer S, Rücker FG, Lux S, Blätte TJ, Skambraks S, Krönke J, Heidel FH, Schnöder TM, Corbacioglu A, Gaidzik VI, Paschka P, et al.. Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation. Nat Commun. 2019; 10:2031. 10.1038/s41467-019-09745-2
    1. Gruszka AM, Valli D, Alcalay M. Wnt Signalling in Acute Myeloid Leukaemia. Cells. 2019; 8:1403. 10.3390/cells8111403
    1. Simon M, Grandage VL, Linch DC, Khwaja A. Constitutive activation of the Wnt/beta-catenin signalling pathway in acute myeloid leukaemia. Oncogene. 2005; 24:2410–20. 10.1038/sj.onc.1208431
    1. Sanchez-Correa B, Campos C, Pera A, Bergua JM, Arcos MJ, Bañas H, Casado JG, Morgado S, Duran E, Solana R, Tarazona R. Natural killer cell immunosenescence in acute myeloid leukaemia patients: new targets for immunotherapeutic strategies? Cancer Immunol Immunother. 2016; 65:453–63. 10.1007/s00262-015-1720-6
    1. Montaldo E, Vacca P, Moretta L, Mingari MC. Development of human natural killer cells and other innate lymphoid cells. Semin Immunol. 2014; 26:107–13. 10.1016/j.smim.2014.01.006
    1. Michel T, Poli A, Cuapio A, Briquemont B, Iserentant G, Ollert M, Zimmer J. Human CD56bright NK Cells: An Update. J Immunol. 2016; 196:2923–31. 10.4049/jimmunol.1502570
    1. Klein E, Vánky F, Vose BM. Natural killer and tumor recognizing lymphocyte activity in tumor patients. Haematologia (Budap). 1978-1979; 12:107–12.
    1. Bruno A, Focaccetti C, Pagani A, Imperatori AS, Spagnoletti M, Rotolo N, Cantelmo AR, Franzi F, Capella C, Ferlazzo G, Mortara L, Albini A, Noonan DM. The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer. Neoplasia. 2013; 15:133–42. 10.1593/neo.121758
    1. Bruno A, Bassani B, D'Urso DG, Pitaku I, Cassinotti E, Pelosi G, Boni L, Dominioni L, Noonan DM, Mortara L, Albini A. Angiogenin and the MMP9-TIMP2 axis are up-regulated in proangiogenic, decidual NK-like cells from patients with colorectal cancer. FASEB J. 2018; 32:5365–77. 10.1096/fj.201701103R
    1. Poli A, Michel T, Thérésine M, Andrès E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology. 2009; 126:458–65. 10.1111/j.1365-2567.2008.03027.x
    1. Rocca YS, Roberti MP, Arriaga JM, Amat M, Bruno L, Pampena MB, Huertas E, Loria FS, Pairola A, Bianchini M, Mordoh J, Levy EM. Altered phenotype in peripheral blood and tumor-associated NK cells from colorectal cancer patients. Innate Immun. 2013; 19:76–85. 10.1177/1753425912453187
    1. Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, Castellano R, Gonçalves A, André P, Romagné F, Thibault G, Viens P, Birnbaum D, Bertucci F, et al.. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 2011; 121:3609–22. 10.1172/JCI45816
    1. Bhatia A, Kumar Y. Cellular and molecular mechanisms in cancer immune escape: a comprehensive review. Expert Rev Clin Immunol. 2014; 10:41–62. 10.1586/1744666X.2014.865519
    1. Konjević GM, Vuletić AM, Mirjačić Martinović KM, Larsen AK, Jurišić VB. The role of cytokines in the regulation of NK cells in the tumor environment. Cytokine. 2019; 117:30–40. 10.1016/j.cyto.2019.02.001
    1. Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr Top Microbiol Immunol. 2010; 341:37–58. 10.1007/82_2010_20

Source: PubMed

3
Abonnere