Research Paper Volume 15, Issue 7 pp 2503—2524

NCAPG as a novel prognostic biomarker in numerous cancers: a meta-analysis and bioinformatics analysis

Jie Lin1, *, , Gangyi Li2, *, , Yanping Bai1, , Yingjun Xie1, ,

  • 1 Department of Hepatobiliary and Pancreatic Surgery, Jilin University Second Hospital, Jilin 130000, China
  • 2 Department of Corneal Refraction, Jilin University Second Hospital, Jilin 130000, China
* Equal contribution

Received: September 12, 2022       Accepted: March 21, 2023       Published: March 29, 2023      

https://doi.org/10.18632/aging.204621
How to Cite

Copyright: © 2023 Lin et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Background: Identification of effective biomarkers for cancer prognosis is a primary research challenge. Recently, several studies have reported the relationship between NCAPG and the occurrence of various tumors. However, none have combined meta-analytical and bioinformatics approaches to systematically assess the role of NCAPG in cancer.

Methods: We searched four databases, namely, PubMed, Web of Science, Embase, and the Cochrane Library, for relevant articles published before April 30, 2022. The overall hazard ratio or odds ratio and 95% confidence intervals were calculated to assess the relationship between NCAPG expression and cancer survival prognosis or clinical characteristics. Furthermore, the aforementioned results were validated using the GEPIA2, Kaplan-Meier plotter, and PrognoScan databases.

Results: The meta-analysis included eight studies with 1096 samples. The results showed that upregulation of NCAPG was correlated with poorer overall survival (hazard ratio = 2.90, 95% confidence interval = 2.06–4.10, P < 0.001) in the cancers included in the study. Subgroup analysis showed that in some cancers, upregulation of NCAPG was correlated with age, distant metastasis, lymph node metastasis, TNM stage, relapse, differentiation, clinical stage, and vascular invasion. These results were validated using the GEPIA2, UALCAN, and PrognoScan databases. We also explored the processes of NCAPG methylation and phosphorylation.

Conclusion: Dysregulated NCAPG expression is associated with the clinical prognostic and pathological features of various cancers. Therefore, NCAPG can serve as a human cancer therapeutic target and a new potential prognostic biomarker.

Introduction

Since the turn of the century, the incidence and mortality of cancer have increased significantly compared to that in previous years [1]. Although various treatment options, such as surgery, radiation, and chemotherapy, are available, the overall survival rates of patients with cancer have not improved significantly [2]. Therefore, identification of new tumor biomarkers and therapeutic targets can better guide clinical tumor treatment.

Non-SMC condensin I complex subunit G (NCAPG) is a subunit of the condensin complex, which condenses and stabilizes chromosomes during mitosis and meiosis [3]. Knockdown of NCAPG significantly reduced the viability of hepatocellular carcinoma cells by regulating Bax, cleaved caspase-3, E-cadherin, N-cadherin, cyclin A1, CDK2, and Bcl-2, and the expression of HOXB9 induces apoptosis and cell cycle arrest in the DNA synthesis phase [4]. In addition, upregulation of NCAPG can activate multiple signaling pathways to promote cell proliferation and anti-apoptotic activity and regulate DNA replication and mismatch repair in different cancer types [57].

Studies have shown that NCAPG is overexpressed in several tumors and associated with clinical features of cancer, such as tumor proliferation, metastasis, invasion, and patient survival [5, 810]. However, its role in various types of tumors remains controversial. For example, NCAPG is overexpressed in hepatocellular carcinoma [11] and glioma [12] but underexpressed in out-of-niche primary tumor cells of multiple myeloma and acute myeloid leukemia [13, 14]. Therefore, we performed a meta-analysis to explore the relationship between NCAPG upregulation and the clinical characteristics of cancer, analyze the prognostic value of NCAPG for cancer patients, and validate its role by bioinformatics methods.

Materials and Methods

Literature search

Two authors independently searched four databases, namely, Pubmed, Embase, Web of Science, and the Cochrane Library for studies published before April 30, 2022. The following search terms were used: (“Neoplasms” OR “Carcinoma” OR “Prognosis” OR “Diagnosis” OR “Survival”) AND (“non-SMC condensin I complex subunit G” OR “NCAPG”).

Inclusion criteria

The following inclusion criteria were considered when screening the databases: (1) the original literature was in English; (2) cancers with abnormal NCAPG expression were investigated; (3) high and low NCAPG expression was delineated; and (4) HR and 95% CIs of the OS can be obtained or calculated from the survival curve.

Exclusion criteria

The exclusion criteria were as follows: (1) reviews, publication letters, retracted literature, and case reports; (2) insufficient data; (3) bioinformatics analysis; and (4) studies not relevant to NCAPG.

Data extraction

Data extraction was performed by two investigators for all included studies and submitted to a third researcher to resolve disagreements. The following data were extracted according to the inclusion criteria: first author, publication date, country of origin, cancer type, number of cases, follow-up time, measurement method of NCAPG expression, outcome measures, HR and 95% CIs for OS.

Quality assessment

The quality of the literature was evaluated using the Newcastle–Ottawa Scale. The evaluation was conducted independently by two investigators, and when disagreements arose, a third investigator participated in the discussion. The total score was 9 points, and a score of ≥6 points indicated high-quality research [15].

Validation of the bioinformatics database

The GEPIA2.0 database (http://gepia.cancer-pku.cn/index.html) is a platform for sequencing and expression data, that includes most tumors and normal tissue samples [16]. Moreover, this study used the “Expression DIY” module to explore the differences between NCAPG transcripts from cancer tissue samples and normal tissue samples. In addition, we downloaded tumor transcription samples from the TCGA database (https://portal.gdc.cancer.gov/) [17] and used R (survival and timeROC packages) to perform a cox regression and ROC analysis of the survival rate. Next, we used the Kaplan-Meier Plotter database (https://kmplot.com/analysis/) to analyze the effect of the NCAPG gene on the survival rate in different cancers for additional data supplementation [18]. To validate the prognostic tumor status of NCAPG, we utilized the PrognoScan database to verify the survival information of this gene in multiple cancer datasets. We used the UALCAN database (http://ualcan.path.uab.edu/index.html) [19] to validate clinical information on NCAPG expression in tumors and explore the methylation and phosphorylation of NCAPG in these tumors.

Molecular role and functional enrichment analysis of NCAPG

GeneMANIA (http://www.genemania.org) is an online tool for protein-protein interactions that helped in identifying genes with similar functions relative to that of the NCAPG gene in this study [20, 21]. We used the STRING database (https://string-db.org/) for GO and KEGG pathway analyses of NCAPG [22]. Finally, we used the multiMiR package (version 4.12) to identify competitive endogenous RNA of NCAPG and constructed a NCAPG network of competitive endogenous RNA interactions of target genes through Cytoscape software (version 3.8.2, https://cytoscape.org/) [23].

Data processing and statistical analysis

The K-M curves of the included studies were processed by Enguage Digitizer 11.3 software to obtain HR values and 95% CIs. Further data analysis was performed using Review Manager 5.3 software. Survival outcomes were calculated by logarithmic HR values and their standard errors. In addition, the correlation between NCAPG upregulation and clinicopathological parameters of cancers (age, gender, degree of differentiation, TNM stage, metastasis, vascular invasion) was assessed by calculating the ORs and 95% CIs. Cochran’s Q test and I2 test assessed the heterogeneity to determine the effect model. A fixed model was used if the included studies had no significant heterogeneity (I2 < 50%, P > 0.1), while a random model was used otherwise. A sensitivity analysis of the included studies was performed using STATA 12.0 software to assess the stability of the results. Publication bias was evaluated using Begg’s rank correlation and Egger’s linear regression. At P < 0.05, publication bias was observed. If publication bias was present, then the trim-and-fill method was used to further assess the stability of the pooled results.

Data availability statement

All relevant data is contained within the article: The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding authors.

Results

Literature selection

A flow chart depicting the literature screening process is shown in Figure 1. A total of 340 articles were screened, and 180 duplicate articles were excluded by searching PubMed, EMBASE, the Cochrane Library, and Web of Science. After scrutinizing the titles and abstracts, 133 more studies were excluded. As eight studies did not record the hazard ratios (HRs) and/or Kaplan-Meier (K-M) curves for overall survival (OS), 11 articles were excluded from bioinformatics analysis. Finally, we included eight studies in the meta-analysis [5, 6, 8, 9, 12, 2426], all of which were cohort studies, and all outcome measures were OS.

Flow chart of literature screening for this meta-analysis.

Figure 1. Flow chart of literature screening for this meta-analysis.

Study characteristics and quality assessment

The characteristics of the included studies are presented in Table 1. All the studies were conducted in China, and 1096 patients were recruited. The types of cancer included non-small cell lung cancer (NSCLC), glioma, breast cancer, gastric cancer, and hepatocellular carcinoma. All included studies assessed the association between OS and NCAPG expression, with follow-ups ranging from 80 to 140 months. All but one study reported clinicopathological parameters. All studies measured NCAPG expression via immunohistochemical staining, except for one study that performed RNA sequencing instead. Newcastle–Ottawa Scale scores were ≥6, indicating that the included studies were of moderate to high quality.

Table 1. Characteristics of included studies.

NameYearRegionEthnicTypeSample size (high/low)FollowUp (months)MethodOutcomeHR estimation methodHR (95% CI)NOS
Jiang2020ChinaAsianBC103 (35/68)120IHCOSK-M7.57 (3.13, 18.29)7
Sun2022ChinaAsianNSCLC156 (84/72)140IHCOS, CPREP2.35 (1.3, 4.27)7
Sun2020ChinaAsianGC135 (71/64)120IHCOS, CPREP2.03 (1.23, 3.35)7
Wang2022ChinaAsianNSCLC60 (28/32)80IHCOS, CPK-M3.45 (1.26, 9.49)6
Wang2019ChinaAsianHCC70 (35/35)120RNA-SeqOS, CPK-M2.34 (1.08, 5.07)7
Wu2021ChinaAsianNSCLC292 (164/128)120IHCOS, CPREP2.05 (1.35, 3.11)7
Zheng2022ChinaAsianGlioma140 (47/93)120IHCOS, CPK-M9.34 (3.75, 23.26)7
Zhou2022ChinaAsianNSCLC140 (70/70)100IHCOS, CPREP2.32 (1.41, 3.81)6

Correlation of NCAPG expression with OS and cancer type

Eight studies reported that NCAPG upregulation was associated with tumors. Therefore, a pooled analysis of the eight studies was performed. As shown in Figure 2, a random model was used because of the absence of obvious heterogeneity (I2 = 56%, P = 0.02). The pooled results suggested that high NCAPG expression rates were correlated with worse OS in patients with different cancers (HR = 2.90, 95% confidence interval (CI) = 2.06–4.10, P < 0.00001).

Forest plot of the pooled OS for subgroup analysis.

Figure 2. Forest plot of the pooled OS for subgroup analysis.

Due to the existence of heterogeneity, a subgroup analysis was performed to further explore the impact of high NCAPG expression on survival in different types of cancer (Figure 2). Four studies [5, 9, 25, 26] investigated cancers of the respiratory system, two studies [8, 24] investigated cancers of the digestive system, and the remaining two studies investigated other types of cancers, namely, glioma [12] and breast cancer [6]. According to the forest plot, upregulation of NCAPG expression in cancer tissues was related to worse OS regardless of the group (respiratory cancer, HR = 2.27, 95% CI = 1.73–2.98, P < 0.00001; digestive cancer, HR = 2.12, 95% CI = 1.39–3.22, P = 0.0005; breast cancer, HR = 7.57, 95% CI = 3.13–18.30, P < 0.00001; glioma, HR = 9.34, 95% CI = 3.75–23.26, P < 0.00001).

Correlation of NCAPG expression with clinicopathological parameters

As all studies included in this meta-analysis reported clinicopathological parameters, we analyzed the high expression of NCAPG and these parameters. As shown in Figure 3, the upregulation of NCAPG was not significantly correlated with gender (male vs. female, odds ratio (OR) = 1.20, 95% CI = 0.92–1.56, P = 0.19, fixed model; Figure 3A), age (young vs. old, OR = 0.68, 95% CI = 0.44–1.06, P = 0.09, random model; Figure 3B), vascular invasion (yes vs. no, OR = 2.23, 95% CI = 0.81–6.11, P = 0.12, random model; Figure 3D), differentiation (well differentiated vs. poorly differentiated, OR = 1.20, 95% CI = 0.50–2.90, P = 0.68, fixed model; Figure 3E), TNM stage (III–IV vs. I–II, OR = 1.64, 95% CI = 0.92–2.92, P = 0.09, random model; Figure 3G), or T classification (T3+T4 vs. T1+T2, OR = 1.68, 95% CI = 0.83–3.40, P = 0.15, random model; Figure 3J), although it was correlated with distant metastasis (yes vs. no, OR = 5.65, 95% CI 2.60–12.26, P < 0.0001, fixed model; Figure 3C), lymph node metastasis (yes vs. no, OR = 2.08, 95% CI = 1.54–2.80, P < 0.00001, fixed model; Figure 3F), relapse (yes vs. no, OR = 2.62, 95% CI = 1.02–6.70, P = 0.04, random model; Figure 3H), and clinical stage (III–IV vs. I–II, OR = 2.23, 95% CI = 1.44–3.44, P = 0.0003, fixed model; Figure 3I). The results of database validation of the clinicopathological characteristics and NCAPG expression data (Supplementary Figure 1) are consistent with most of our previous meta-analysis results.

Forest plot of the relationship between high NCAPG expression and clinicopathological parameters. (A) gender, (B) age, (C) distant metastasis, (D) vascular invasion, (E) differentiation, (F) lymph node metastasis, (G) TNM stage, (H) relapse, (I) clinical stage, (J) T classification.

Figure 3. Forest plot of the relationship between high NCAPG expression and clinicopathological parameters. (A) gender, (B) age, (C) distant metastasis, (D) vascular invasion, (E) differentiation, (F) lymph node metastasis, (G) TNM stage, (H) relapse, (I) clinical stage, (J) T classification.

Sensitivity analysis and publication bias

To verify the robustness of the results, we performed a sensitivity analysis (Figure 4). Removal of one or more articles did not significantly affect the results, indicating that the results were relatively stable. Begg’s and Egger’s tests indicated publication bias for OS and distant metastasis (Table 2, Supplementary Figure 2). We analyzed the stability of the study further using the trim-and-fill method. For OS, the results indicated that the estimated number of missing studies was 0 and the adjusted HR was 2.90 (95% CI = 2.06–4.10, P < 0.001), which indicated that upregulation of NCAPG was associated with poorer OS, suggesting that our result is reliable. In addition, for distant metastases, two studies were estimated to be missing and the adjusted OR was 4.85 (95% CI = 2.48–9.58, P < 0.001), which indicated a reliable result.

Sensitivity analysis. (A) OS, (B) gender, (C) age, (D) distant metastasis, (E) vascular invasion, (F) differentiation, (G) lymph node metastasis, (H) TNM stage, (I) relapse, (J) clinical stage, (K) T classification.

Figure 4. Sensitivity analysis. (A) OS, (B) gender, (C) age, (D) distant metastasis, (E) vascular invasion, (F) differentiation, (G) lymph node metastasis, (H) TNM stage, (I) relapse, (J) clinical stage, (K) T classification.

Table 2. Results of Begg’s and Egger’s tests for publication bias.

Analysis typeBegg’s testEgger’s test
ZPtP
OS2.600.0093.130.020
Gender (male vs. female)0.900.368−1.080.328
Age (young vs. old)0.620.536−1.160.291
Distant metastasis (yes vs. no)1.700.0896.990.020
Vascular invasion (yes vs. no)0.001.000NANA
Differentiation (well vs. poor)1.040.29612.420.051
Lymph node metastasis (yes vs. no)0.240.8061.150.332
TNM stage (III−IV vs. I−II)0.001.000NANA
Relapse (yes vs. no)0.001.0001.590.357
Clinical stage (III−IV vs. I−II)0.001.0000.790.573
T classification (T3+T4 vs. T1+T2)1.220.2212.270.108
If P < 0.05, the results are in bold. Abbreviation: NA: Not available.

Validation of NCAPG expression against public databases

We evaluated the NCAPG expression levels and performed a survival analysis in various cancers using the GEPIA2.0 database to validate our results. The findings showed that, compared with normal tissues, the expression of NCAPG was significantly upregulated in tumors, including BRCA, GBM, LIHC, LUAD, LUSC, and STAD (Figure 5A). We next performed a univariate Cox survival analysis on the above tumors, and it showed that the OS of LIHC and LUAD was related to NCAPG (Figure 5B). The progression-free survival (PFS) of BRCA, LIHC, LUAD, and STAD was related to NCAPG (Figure 5C). The relapse-free survival (RFS) of LIHC, LUSC, and STAD was related to NCAPG (Figure 5D), and the disease-specific survival (DSS) of LIHC and LUAD was related to NCAPG (Figure 5E). We then performed receiver operating characteristic (ROC) survival analysis of the data on the above tumors (Supplementary Figure 3), and the area under the curve of GBM, LIHC, and STAD was greater than 0.7. Therefore, we concluded that NCAPG may be a good prognostic indicator of various cancers. In addition, we analyzed the survival prognosis of the above tumors using the Kaplan-Meier plotter database based on the median cutoff value of NCAPG expression (including probes 218663_at and 218663_s_at) in cancer. The results showed that NCAPG expression can be used to predict OS in breast cancer (P < 0.01; Figure 6A), RFS (P < 0.01; Figure 6B), PPS (P < 0.01; Figure 6C), DMFS (P < 0.01; Figure 6D), liver cancer, PFS, PFS, and DSS (P < 0.01; Figure 6E), lung cancer (P < 0.01; Figure 6F), FP (P < 0.01; Figure 6G), and PPS (P < 0.01; Figure 6H), gastric cancer (P < 0.01; Figure 6I), FP (P < 0.01; Figure 6J,), and PPS (P < 0.01; Figure 6K). Moreover, the findings were validated against multiple cancer datasets using the PrognoScan database. We collected 29 datasets on breast and lung cancers. As shown in Table 3, NCAPG expression in these tumors significantly affected prognosis-related indicators, such as OS, DSS, RFS, and PFS (P < 0.05).

(A) Expression levels of NCAPG in cancer tissues and normal tissues in GEPIA2. From left to right are gastric cancer (STAD), lung cancer (LUAD), liver cancer (LIHC), glioma (GBM) and breast cancer (BRCA). The red box represents the expression level of NCAPG in cancer tissues; the gray box represents the expression level of NCAPG in normal tissues, the screening criteria were log2FC|>1 and P B) OS of BRCA, GBM, LIHC, LUAD, LUSC and STAD, (C) PFS of BRCA, GBM, LIHC, LUAD, LUSC and STAD, (D) DFS of BRCA, LIHC, LUAD, LUSC and STAD, (E) BRCA, GBM, LIHC, LUAD, LUSC and STAD's DSS.

Figure 5. (A) Expression levels of NCAPG in cancer tissues and normal tissues in GEPIA2. From left to right are gastric cancer (STAD), lung cancer (LUAD), liver cancer (LIHC), glioma (GBM) and breast cancer (BRCA). The red box represents the expression level of NCAPG in cancer tissues; the gray box represents the expression level of NCAPG in normal tissues, the screening criteria were log2FC|>1 and P < 0.01, (B) OS of BRCA, GBM, LIHC, LUAD, LUSC and STAD, (C) PFS of BRCA, GBM, LIHC, LUAD, LUSC and STAD, (D) DFS of BRCA, LIHC, LUAD, LUSC and STAD, (E) BRCA, GBM, LIHC, LUAD, LUSC and STAD's DSS.

(A) OS of NCAPG (218663-s-at) in BRCA (n = 1879), OS of NCAPG (218663-at) in BRCA (n = 1879), (B) RFS of NCAPG (218663-s-at) in BRCA (n = 4929), RFS of NCAPG (218663-at) in BRCA (n = 4929), (C) PPS of NCAPG (218663-s-at) in BRCA (n = 458), PPS of NCAPG (218663-at) in BRCA (n = 458)(D) DMPS of NCAPG (218663-s-at) in BRCA (n = 2765), DMPS of NCAPG (218663-at) in BRCA (n = 2765), (E) OS (n = 364), PFS (n = 316), PFS (n = 370)and DSS (n = 362)of NCAPG in LIHC, (F) OS of NCAPG (218663-s-at) in lung cancer (n = 1925), OS of NCAPG (218663-at) in lung cancer (n = 1925), (G) FP of NCAPG (218663-s-at) in lung cancer (n = 982), FP of NCAPG (218663-at) in lung cancer (n = 982), (H) PPS of NCAPG (218663-s-at) in lung cancer (n = 344), PPS of NCAPG (218663-at) in lung cancer (n = 344), (I) OS of NCAPG (218663-s-at) in STAD (n = 592), OS of NCAPG (218663-at) in STAD (n = 592), (J) FP of NCAPG (218663-s-at) in STAD (n = 358), FP of NCAPG (218663-at) in STAD (n = 358), (K) PPS of NCAPG (218663-s-at) in STAD (n = 221), PPS of NCAPG (218663-at) in STAD (n = 221).

Figure 6. (A) OS of NCAPG (218663-s-at) in BRCA (n = 1879), OS of NCAPG (218663-at) in BRCA (n = 1879), (B) RFS of NCAPG (218663-s-at) in BRCA (n = 4929), RFS of NCAPG (218663-at) in BRCA (n = 4929), (C) PPS of NCAPG (218663-s-at) in BRCA (n = 458), PPS of NCAPG (218663-at) in BRCA (n = 458)(D) DMPS of NCAPG (218663-s-at) in BRCA (n = 2765), DMPS of NCAPG (218663-at) in BRCA (n = 2765), (E) OS (n = 364), PFS (n = 316), PFS (n = 370)and DSS (n = 362)of NCAPG in LIHC, (F) OS of NCAPG (218663-s-at) in lung cancer (n = 1925), OS of NCAPG (218663-at) in lung cancer (n = 1925), (G) FP of NCAPG (218663-s-at) in lung cancer (n = 982), FP of NCAPG (218663-at) in lung cancer (n = 982), (H) PPS of NCAPG (218663-s-at) in lung cancer (n = 344), PPS of NCAPG (218663-at) in lung cancer (n = 344), (I) OS of NCAPG (218663-s-at) in STAD (n = 592), OS of NCAPG (218663-at) in STAD (n = 592), (J) FP of NCAPG (218663-s-at) in STAD (n = 358), FP of NCAPG (218663-at) in STAD (n = 358), (K) PPS of NCAPG (218663-s-at) in STAD (n = 221), PPS of NCAPG (218663-at) in STAD (n = 221).

Table 3. NCAPG-related cancer GEO database classifications.

DatasetCancer typePrognostic factorCox P-valueln (HR)HR (95% CI)
GSE5287Bladder cancerOverall Survival0.0434720.5553051.74 (1.02–2.99)
GSE5287Bladder cancerOverall Survival0.3286030.2236761.25 (0.80–1.96)
GSE13507Bladder cancerOverall Survival0.0003996360.3803111.46 (1.19–1.81)
GSE13507Bladder cancerDisease Specific Survival7.89E-050.6989042.01 (1.42–2.85)
GSE12417-GPL96Blood cancerOverall Survival0.294458−0.1538150.86 (0.64–1.14)
GSE12417-GPL96Blood cancerOverall Survival0.391317−0.1213010.89 (0.67–1.17)
GSE12417-GPL570Blood cancerOverall Survival0.924132−0.01554830.98 (0.71–1.36)
GSE12417-GPL570Blood cancerOverall Survival0.8955520.02059931.02 (0.75–1.39)
GSE5122Blood cancerOverall Survival0.852157−0.0252550.98 (0.75–1.27)
GSE5122Blood cancerOverall Survival0.980633−0.004892071.00 (0.67–1.48)
GSE8970Blood cancerOverall Survival0.116843−0.3178770.73 (0.49–1.08)
GSE8970Blood cancerOverall Survival0.14972−0.2742130.76 (0.52–1.10)
GSE4475Blood cancerOverall Survival0.0095096−0.4649770.63 (0.44–0.89)
E-TABM-346Blood cancerOverall Survival0.865305−0.06302350.94 (0.45–1.94)
E-TABM-346Blood cancerOverall Survival0.9518710.01745841.02 (0.58–1.79)
E-TABM-346Blood cancerEvent Free Survival0.707167−0.1261660.88 (0.46–1.70)
E-TABM-346Blood cancerEvent Free Survival0.9986530.0004386281.00 (0.60–1.66)
GSE16131-GPL96Blood cancerOverall Survival0.863640.02962221.03 (0.73–1.44)
GSE16131-GPL96Blood cancerOverall Survival0.774936−0.04759060.95 (0.69–1.32)
GSE2658Blood cancerDisease Specific Survival0.08358940.4105551.51 (0.95–2.40)
GSE2658Blood cancerDisease Specific Survival0.000394760.3823431.47 (1.19–1.81)
GSE4271Brain cancerOverall Survival0.001742770.587751.80 (1.25–2.60)
GSE7696Brain cancerOverall Survival0.9938690.001192411.00 (0.74–1.36)
GSE7696Brain cancerOverall Survival0.5676860.09198791.10 (0.80–1.50)
GSE16581Brain cancerOverall Survival0.8445170.1411271.15 (0.28–4.72)
GSE16581Brain cancerOverall Survival0.5428490.6582951.93 (0.23–16.10)
GSE19615Breast cancerDistant Metastasis Free Survival0.3758690.3465811.41 (0.66–3.05)
GSE19615Breast cancerDistant Metastasis Free Survival0.533930.2790451.32 (0.55–3.18)
GSE12276Breast cancerRelapse Free Survival0.0001050430.4277981.53 (1.24–1.90)
GSE6532-GPL570Breast cancerDistant Metastasis Free Survival0.1154570.3148441.37 (0.93–2.03)
GSE6532Breast cancerDistant Metastasis Free Survival0.03613860.3848261.47 (1.03–2.11)
GSE6532-GPL570Breast cancerRelapse Free Survival0.1154570.3148441.37 (0.93–2.03)
GSE6532Breast cancerRelapse Free Survival0.03613860.3848261.47 (1.03–2.11)
GSE9195Breast cancerDistant Metastasis Free Survival0.4348690.2616181.30 (0.67–2.50)
GSE9195Breast cancerDistant Metastasis Free Survival0.1658060.5102741.67 (0.81–3.43)
GSE9195Breast cancerRelapse Free Survival0.2247450.3973021.49 (0.78–2.83)
GSE9195Breast cancerRelapse Free Survival0.4997710.2026341.22 (0.68–2.21)
GSE12093Breast cancerDistant Metastasis Free Survival0.01226970.9236432.52 (1.22–5.19)
GSE11121Breast cancerDistant Metastasis Free Survival0.009946040.5749411.78 (1.15–2.75)
GSE1378Breast cancerRelapse Free Survival0.7550550.06398281.07 (0.71–1.59)
GSE1379Breast cancerRelapse Free Survival0.7284360.08049051.08 (0.69–1.71)
GSE2034Breast cancerDistant Metastasis Free Survival0.002498380.5156441.67 (1.20–2.34)
GSE1456Breast cancerOverall Survival0.0007869641.068032.91 (1.56–5.43)
GSE1456Breast cancerDisease Specific Survival0.0006485690.9839012.67 (1.52–4.71)
GSE1456Breast cancerRelapse Free Survival0.0002796831.150843.16 (1.70–5.88)
GSE7378Breast cancerDisease Free Survival0.03350110.6542871.92 (1.05–3.52)
GSE7378Breast cancerDisease Free Survival0.07093170.5546411.74 (0.95–3.18)
E-TABM-158Breast cancerDistant Metastasis Free Survival0.6811730.08734891.09 (0.72–1.66)
E-TABM-158Breast cancerOverall Survival0.203745−0.2276070.80 (0.56–1.13)
E-TABM-158Breast cancerRelapse Free Survival0.361575−0.171990.84 (0.58–1.22)
E-TABM-158Breast cancerDisease Specific Survival0.0899647−0.3991620.67 (0.42–1.06)
E-TABM-158Breast cancerOverall Survival0.361575−0.171990.84 (0.58–1.22)
E-TABM-158Breast cancerDistant Metastasis Free Survival0.7572490.06667031.07 (0.70–1.63)
E-TABM-158Breast cancerRelapse Free Survival0.203745−0.2276070.80 (0.56–1.13)
E-TABM-158Breast cancerDisease Specific Survival0.0351901−0.4545090.63 (0.42–0.97)
GSE3494Breast cancerDisease Specific Survival0.001638430.5905311.80 (1.25–2.61)
GSE4922Breast cancerDisease Free Survival4.93E-050.8085442.24 (1.52–3.32)
GSE2990Breast cancerDistant Metastasis Free Survival0.07598890.3518491.42 (0.96–2.10)
GSE2990Breast cancerRelapse Free Survival0.1057990.5006211.65 (0.90–3.03)
GSE2990Breast cancerRelapse Free Survival0.1233930.2394491.27 (0.94–1.72)
GSE2990Breast cancerDistant Metastasis Free Survival0.002397210.8959662.45 (1.37–4.37)
GSE2990Breast cancerDistant Metastasis Free Survival0.07965030.4120161.51 (0.95–2.39)
GSE2990Breast cancerRelapse Free Survival0.10580.3130351.37 (0.94–2.00)
GSE2990Breast cancerDistant Metastasis Free Survival0.1420130.5563771.74 (0.83–3.67)
GSE2990Breast cancerRelapse Free Survival0.003699610.726962.07 (1.27–3.38)
GSE7390Breast cancerDistant Metastasis Free Survival0.175050.1439451.15 (0.94–1.42)
GSE7390Breast cancerOverall Survival0.06546230.2115011.24 (0.99–1.55)
GSE7390Breast cancerRelapse Free Survival0.2173780.1402861.15 (0.92–1.44)
GSE7390Breast cancerDistant Metastasis Free Survival0.2797840.1524571.16 (0.88–1.54)
GSE7390Breast cancerRelapse Free Survival0.1339690.1271091.14 (0.96–1.34)
GSE7390Breast cancerOverall Survival0.2032240.1913661.21 (0.90–1.63)
GSE12945Colorectal cancerDisease Free Survival0.3903240.6262821.87 (0.45–7.81)
GSE12945Colorectal cancerOverall Survival0.1154020.7894132.20 (0.82–5.88)
GSE12945Colorectal cancerDisease Free Survival0.672657−0.7737450.46 (0.01–16.70)
GSE12945Colorectal cancerOverall Survival0.168871.334583.80 (0.57–25.43)
GSE17536Colorectal cancerDisease Specific Survival0.589054−0.1305160.88 (0.55–1.41)
GSE17536Colorectal cancerDisease Specific Survival0.8283920.04287821.04 (0.71–1.54)
GSE17536Colorectal cancerOverall Survival0.8567830.03907471.04 (0.68–1.59)
GSE17536Colorectal cancerOverall Survival0.2621510.1985861.22 (0.86–1.73)
GSE17536Colorectal cancerDisease Free Survival0.109121−0.5067030.60 (0.32–1.12)
GSE17536Colorectal cancerDisease Free Survival0.268629−0.2635130.77 (0.48–1.23)
GSE14333Colorectal cancerDisease Free Survival0.0827771−0.4274420.65 (0.40–1.06)
GSE14333Colorectal cancerDisease Free Survival0.021572−0.4441430.64 (0.44–0.94)
GSE17537Colorectal cancerOverall Survival0.9089530.03578761.04 (0.56–1.91)
GSE17537Colorectal cancerOverall Survival0.6165030.1361261.15 (0.67–1.95)
GSE17537Colorectal cancerDisease Free Survival0.7132610.1258221.13 (0.58–2.22)
GSE17537Colorectal cancerDisease Free Survival0.335550.2944751.34 (0.74–2.44)
GSE17537Colorectal cancerDisease Specific Survival0.1429370.7411752.10 (0.78–5.66)
GSE17537Colorectal cancerDisease Specific Survival0.09899510.7897852.20 (0.86–5.63)
GSE22138Eye cancerDistant Metastasis Free Survival0.4404030.1747461.19 (0.76–1.86)
GSE22138Eye cancerDistant Metastasis Free Survival0.09423720.7461582.11 (0.88–5.05)
GSE2837Head and neck cancerRelapse Free Survival0.160158−0.6445510.52 (0.21–1.29)
jacob-00182-CANDFLung cancerOverall Survival0.4269860.1978761.22 (0.75–1.99)
jacob-00182-CANDFLung cancerOverall Survival0.1848010.2372981.27 (0.89–1.80)
jacob-00182-HLMLung cancerOverall Survival0.5659960.09199481.10 (0.80–1.50)
jacob-00182-HLMLung cancerOverall Survival0.5721480.08133851.08 (0.82–1.44)
jacob-00182-MSKLung cancerOverall Survival0.08654120.2679831.31 (0.96–1.78)
jacob-00182-MSKLung cancerOverall Survival0.04145680.3673991.44 (1.01–2.06)
GSE13213Lung cancerOverall Survival0.005241860.36751.44 (1.12–1.87)
GSE31210Lung cancerRelapse Free Survival3.00E-050.6232341.86 (1.39–2.50)
GSE31210Lung cancerOverall Survival0.004043870.5972411.82 (1.21–2.73)
jacob-00182-UMLung cancerOverall Survival0.1586490.1658381.18 (0.94–1.49)
jacob-00182-UMLung cancerOverall Survival0.3136680.09156181.10 (0.92–1.31)
GSE3141Lung cancerOverall Survival0.2512460.2220221.25 (0.85–1.82)
GSE3141Lung cancerOverall Survival0.4397870.1860231.20 (0.75–1.93)
GSE14814Lung cancerOverall Survival0.3476440.2757491.32 (0.74–2.34)
GSE14814Lung cancerDisease Specific Survival0.2130610.408741.50 (0.79–2.86)
GSE14814Lung cancerDisease Specific Survival0.1632010.630311.88 (0.77–4.56)
GSE14814Lung cancerOverall Survival0.3394290.3947371.48 (0.66–3.34)
GSE8894Lung cancerRelapse Free Survival0.1949850.1380561.15 (0.93–1.41)
GSE8894Lung cancerRelapse Free Survival0.08871640.166561.18 (0.98–1.43)
GSE4573Lung cancerOverall Survival0.1023690.4581031.58 (0.91–2.74)
GSE4573Lung cancerOverall Survival0.2384140.3564871.43 (0.79–2.58)
GSE17710Lung cancerRelapse Free Survival0.2573060.3428581.41 (0.78–2.55)
GSE17710Lung cancerRelapse Free Survival0.2032910.3894131.48 (0.81–2.69)
GSE17710Lung cancerOverall Survival0.1688220.4352781.55 (0.83–2.87)
GSE17710Lung cancerOverall Survival0.1279730.4890011.63 (0.87–3.06)
GSE9891Ovarian cancerOverall Survival0.04581450.1716561.19 (1.00–1.41)
DUKE-OCOvarian cancerOverall Survival0.190803−0.1326140.88 (0.72–1.07)
DUKE-OCOvarian cancerOverall Survival0.774339−0.03934880.96 (0.73–1.26)
GSE26712Ovarian cancerOverall Survival0.104731−0.2039390.82 (0.64–1.04)
GSE26712Ovarian cancerOverall Survival0.493253−0.1359510.87 (0.59–1.29)
GSE26712Ovarian cancerDisease Free Survival0.817429−0.04128680.96 (0.68–1.36)
GSE26712Ovarian cancerDisease Free Survival0.112295−0.1816270.83 (0.67–1.04)
GSE17260Ovarian cancerOverall Survival0.3740230.1257221.13 (0.86–1.50)
GSE17260Ovarian cancerProgression Free Survival0.2493240.1230871.13 (0.92–1.39)
GSE14764Ovarian cancerOverall Survival0.203350.2973581.35 (0.85–2.13)
GSE14764Ovarian cancerOverall Survival0.3300250.2687341.31 (0.76–2.25)
GSE19234Skin cancerOverall Survival0.004852851.285663.62 (1.48–8.85)
GSE30929Soft tissue cancerDistant Recurrence Free Survival0.0001345060.4667651.59 (1.26–2.03)

Molecular role and functional enrichment analysis results of NCAPG

We used the GeneMANIA database for the protein-molecular interaction analysis of NCAPG and its related molecules, such as NCAPG2, NCAPH, and SMC4 (Figure 7A). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional and pathway enrichment analyses of NCAPG were performed using the STRING database. The most abundant GO terms were nuclear division, cell division, and cell cycle process (Table 4). In addition, the KEGG pathway analysis confirmed that these co-expressed genes were significantly involved in the p53 signaling pathway, cell cycle, and cellular senescence (Table 4). These results indicate that NCAPG is involved in the biological pathways of cancer. Additionally, we used the multiMiR package to identify NCAPG-related miRNAs and lncRNAs (the screening criterion for miRNAs was a predicted cutoff of 500,000, and the screening criteria for lncRNAs were lnc_mi$pancancerNum>10 and lnc_mi$clipExpNum>4) that may interact with NCAPG (Figure 7B). We identified 20 miRNAs and 13 lncRNAs, which could provide direction for future experimental designs. DNA methylation directly affects the occurrence and progression of cancers. We used the UALCAN database to investigate the DNA methylation of NCAPG. Our results showed that NCAPG methylation levels were significantly reduced in BRCA, GBM, LIHC, LUAD, and LUSC tissues compared to normal tissues (Figure 8A8F), which may explain the difference in NCAPG expression between BRCA, GBM, LIHC, LUAD, and LUSC tissues and normal tissues. Post-translational modification is a key molecular mechanism underlying NCAPG activation. Therefore, we examined the changes in NCAPG phosphorylation levels between tumor tissues and normal tissues. The Clinical Proteomic Tumor Analysis Consortium database includes four cancers: BRCA, GBM, LIHC, and LUAD. Compared with the normal samples, the phosphorylation levels at 674/973/975/984 of NCAPG were higher in BRCA, GBM, LIHC, and LUAD, respectively (Figure 8G). The specific results are shown in Figure 8H8M. Since the p53 signaling pathway is highly enriched, we also explored the phosphorylation of NCAPG in this pathway, and the results are shown in Figure 8N8R.

Table 4. Functional enrichment analysis and pathway enrichment analysis of NCAPG genes.

GO IDTerm descriptionOntologyCountFalse discovery rate
GO:0000280Nuclear divisionBP111.26E-16
GO:0140014Mitotic nuclear divisionBP101.26E-16
GO:0030261Chromosome condensationBP85.29E-16
GO:0051301Cell divisionBP117.60E-15
GO:0000070Mitotic sister chromatid segregationBP84.14E-13
GO:0007076Mitotic chromosome condensationBP68.07E-13
GO:0022402Cell cycle processBP117.24E-12
GO:0010032Meiotic chromosome condensationBP51.78E-11
GO:1903046Meiotic cell cycle processBP61.29E-07
GO:0051276Chromosome organizationBP91.48E-07
GO:0007077Mitotic nuclear envelope disassemblyBP33.34E-05
GO:0022414Reproductive processBP85.10E-05
GO:0051304Chromosome separationBP30.00033
GO:0140013Meiotic nuclear divisionBP40.00049
GO:1905448Positive regulation of mitochondrial atp synthesis coupled electron transportBP20.0023
GO:0051987Positive regulation of attachment of spindle microtubules to kinetochoreBP20.0029
GO:0031145Anaphase-promoting complex-dependent catabolic processBP30.0046
GO:0045132Meiotic chromosome segregationBP30.0051
GO:0035404Histone-serine phosphorylationBP20.0082
GO:0055015Ventricular cardiac muscle cell developmentBP20.0082
GO:0034501Protein localization to kinetochoreBP20.01
GO:0051782Negative regulation of cell divisionBP20.0139
GO:0000086G2/M transition of mitotic cell cycleBP30.0145
GO:0007292Female gamete generationBP30.0145
GO:0051383Kinetochore organizationBP20.0145
GO:0007051Spindle organizationBP30.0162
GO:0007093Mitotic cell cycle checkpointBP30.0197
GO:0045931Positive regulation of mitotic cell cycleBP30.0197
GO:0060045Positive regulation of cardiac muscle cell proliferationBP20.0247
GO:0000226Microtubule cytoskeleton organizationBP40.0261
GO:0010971Positive regulation of g2/m transition of mitotic cell cycleBP20.0261
GO:0018105Peptidyl-serine phosphorylationBP30.0261
GO:0010389Regulation of g2/m transition of mitotic cell cycleBP30.0338
GO:1901991Negative regulation of mitotic cell cycle phase transitionBP30.0412
GO:0000796Condensin complexCC63.01E-14
GO:0000793Condensed chromosomeCC83.53E-11
GO:0000799Nuclear condensin complexCC42.14E-09
GO:0000794Condensed nuclear chromosomeCC64.97E-09
GO:0000797Condensin core heterodimerCC37.47E-07
GO:0000779Condensed chromosome, centromeric regionCC51.06E-06
GO:0098687Chromosomal regionCC61.80E-06
GO:0005694ChromosomeCC92.71E-06
GO:0000228Nuclear chromosomeCC86.95E-06
GO:0043232Intracellular non-membrane-bounded organelleCC113.32E-05
GO:0000307Cyclin-dependent protein kinase holoenzyme complexCC30.00021
GO:0097125Cyclin b1-cdk1 complexCC20.00021
GO:0005813CentrosomeCC50.00087
GO:0032991Protein-containing complexCC100.0011
GO:0005634NucleusCC110.0019
GO:0005819SpindleCC40.0026
GO:0072686Mitotic spindleCC30.0029
GO:0000922Spindle poleCC30.0064
GO:0030496MidbodyCC30.0083
GO:0000780Condensed nuclear chromosome, centromeric regionCC20.0092
GO:0005829CytosolCC90.0132
GO:0005654NucleoplasmCC80.0161
GO:0005876Spindle microtubuleCC20.018
GO:0035173Histone kinase activityMF30.00041
KEGG IDTerm descriptionCountFalse discovery rate
hsa04115p53 signaling pathway30.0029
hsa04914Progesterone-mediated oocyte maturation30.0032
hsa04110Cell cycle30.0043
hsa04114Oocyte meiosis30.0043
hsa04218Cellular senescence30.005
hsa05170Human immunodeficiency virus 1 infection30.0101
Abbreviations: BP: Biological Process; CC: Cellular Component; ME: Molecular Function; KEGG: Kyoto Encyclopedia of Genes and Genomes.
Network analysis between NCAPG and target genes (A) PPI network for KIF23 was constructed in Gene MANIA, Different colors of the network edge indicate the bioinformatics methods applied: physical interaction, co-expression, predicted, co-localization, pathway, genetic interaction and shared protein domains. Abbreviation: PPI: protein–protein interaction. (B) The relationship between NCAPG and non-coding RNA, the red square represents the target gene NCAPG, the blue oval represents miRNA, and the yellow triangle represents lncRNA.

Figure 7. Network analysis between NCAPG and target genes (A) PPI network for KIF23 was constructed in Gene MANIA, Different colors of the network edge indicate the bioinformatics methods applied: physical interaction, co-expression, predicted, co-localization, pathway, genetic interaction and shared protein domains. Abbreviation: PPI: protein–protein interaction. (B) The relationship between NCAPG and non-coding RNA, the red square represents the target gene NCAPG, the blue oval represents miRNA, and the yellow triangle represents lncRNA.

DNA methylation features of NCAPG in BRCA (A), GBM (B), LIHC (C), LUAD (D), LUSC (E) and STAD (F). Phosphorylation of NCAPG in several selected cancers according to the CPTAC database. (G) The schematic diagram and phosphorylation sites of the NCAPG protein are shown. The phosphorylation of NCAPG at S674, S973, S975 and 984 in BRCA (H–J), S674 in GBM (K), S674 in LIHC (L), S674 in LUAD (M). The P53 pathway phosphorylation of NCAPG at S674, S973, S975 and 984 in BRCA (N–P), S674 in GBM (Q), S674 in LUAD (R), from the UALCAN database. *p **p ***p

Figure 8. DNA methylation features of NCAPG in BRCA (A), GBM (B), LIHC (C), LUAD (D), LUSC (E) and STAD (F). Phosphorylation of NCAPG in several selected cancers according to the CPTAC database. (G) The schematic diagram and phosphorylation sites of the NCAPG protein are shown. The phosphorylation of NCAPG at S674, S973, S975 and 984 in BRCA (HJ), S674 in GBM (K), S674 in LIHC (L), S674 in LUAD (M). The P53 pathway phosphorylation of NCAPG at S674, S973, S975 and 984 in BRCA (NP), S674 in GBM (Q), S674 in LUAD (R), from the UALCAN database. *p < 0:05, **p < 0:01, and ***p < 0:001, Abbreviation: ns: No statistical significance.

Discussion

NCAPG expression was initially found to be correlated with the prognosis of liver cancer. Later on, the expression of NCAPG was often closely associated with the survival outcome and clinical pathology of patients with diseases such as NSCLC, renal clear cell carcinoma, breast cancer, and gastric cancer [27]. To better verify and summarize the value of this gene and avoid the errors caused by small samples or small queues, we used meta-methods and bioinformatics jointly.

First, in this study, the results of the meta-analysis showed that high expression of NCAPG is associated with poor prognosis, suggesting its role as a proto-oncogene in cancer. Second, eight studies were included in this meta-analysis. Our results suggest that cancer patients with upregulated NCAPG expression have a 2.90-fold worse OS than those with low expression. We also performed a subgroup analysis according to the different systems. The results showed that NCAPG might be a potential prognostic marker for cancers of the respiratory, digestive, and other systems. Third, from the perspective of bioinformatics, univariate cox regression analysis showed that NCAPG was a bad prognostic factor for LIHC, LUAD, and STAD. We also verified this inference using the Kaplan-Meier plotter database and found that the high expression of NCAPG was related to the poor prognosis of BRCA, LIHC, LUAD, and STAD. We calculated the relationship between the expression of NCAPG and the annual survival rate of the cancer (Supplementary Figure 3). The results showed that NCAPG could predict the survival and prognosis of GBM, LIHC, and STAD.

Additionally, we assessed the association between NCAPG expression and clinicopathological parameters. The pooled results showed that the upregulation of NCAPG was not associated with age, sex, vascular invasion, differentiation, TNM stage, and T classification but was associated with distant metastasis, lymph node metastasis, relapse, and clinical stage. The results of the sensitivity and publication bias analyses demonstrated the reliability of the results. To our knowledge, this is the first meta-analysis to demonstrate the prognostic value of NCAPG in cancer.

To further explore the relationship between NCAPG expression and the clinicopathological features of different cancers, a subgroup analysis was performed. We found that NCAPG overexpression was significantly correlated with positive lymph node metastasis in gastric cancer and NSCLC, TNM stage in hepatocellular carcinoma, age in glioma, differentiation in hepatocellular carcinoma and glioma, and vascular invasion in gastric cancer. Next, we verified the clinicopathological features of cancers with NCAPG expression using the UALCAN database (Supplementary Figure 1). The results showed that the expression of NCAPG was related to the age, lymph node metastasis, and stage of tumor patients.

How NCAPG can precisely regulate oncogenes remains unknown, but some studies have proved that it may be related to the following mechanisms. NCAPG induces epigenetic changes of tumors through a variety of signal pathways and molecules. In lung cancer, NCAPG expression activates TGF-β signaling pathway [5]. In breast cancer, it is related to the SRC/STAT3 signaling pathway [6] and p53 signaling pathway [28]. In colorectal cancer, it is associated with Wnt/β-catenin signaling pathway [29]. In hepatocellular carcinoma, it is related to activation of PI3K/AKT signaling pathway [7]. In cardiac adenocarcinoma, Wnt/β-catenin signaling pathway [30] and PI3K/AKT signaling pathway are involved [31]. In endometrial carcinoma, it is related to Wnt/β-catenin signaling pathway [32]. In oral squamous cell carcinoma, it is related to miR-378a-3p-mediated GSK-3β/β-catenin signaling [33]. In prostate cancer, it interacts with miR-99a-3p [34]. In bladder cancer, it is related to NF-κB signaling pathway [35]. In ovarian cancer, it is related to p38 MAPK signaling pathway [36].

We also analyzed the GO and KEGG pathways of NCAPG. The richest GO terms were nuclear division, cell division, and cell cycle processes, all of which are related to cancer cell proliferation. In addition, KEGG pathway analysis confirmed that NCAPG-related genes were involved in the p53 signaling pathway, cell senescence, and cell cycle, which are involved in the mechanism underlying carcinogenesis. We also explored the changes in phosphorylation and methylation of NCAPG in these tumors. Finally, we identified the miRNAs and lncRNAs related to NCAPG.

This study has a few limitations. First, a total of eight studies were included in this study, which is a relatively small sample size and may affect the accuracy of the results; therefore, additional research is needed to confirm the findings. Second, it is necessary to fully verify and clarify the role and mechanism of NCAPG in cancer through cell models in vitro and in vivo. Furthermore, some studies used the K-M curve to extract the HR of OS, which may have had an impact on the results and led to publication bias. Finally, due to the different methods used to evaluate NCAPG expression and different cutoff value standards, statistical errors may have been introduced.

Conclusion

In conclusion, our study is the first to systematically address the prognostic and clinical significance of the NCAPG gene in cancer. We provide meta-analytical and bioinformatic evidence that NCAPG acts as an oncogenic mRNA with great potential as a biological prognostic marker for cancer. However, this study had certain limitations, and more basic experiments are needed to verify these conclusions.

Supplementary Materials

Supplementary Figures

Abbreviations

BC: Breast cancer; NSCLC: Non-small cell lung cancer; GC: Gastric cancer; HCC: Hepatocellular carcinoma; IHC: Immunohistochemistry; RNA-Seq: RNA sequence; CP: Clinicopathological parameters; CI: Confidence interval; K-M: Kaplan-Meier curve; REP: Reported; NOS: Newcastle–Ottawa Scale; BRCA: Breast cancer; GBM: Glioblastoma; LIHC: Liver cancer; LUAD: Lung cancer; LUSC: Lung squamous cell carcinoma; STAD: Stomach adenocarcinoma; OS: Over survival; PFS: Progression Free Survival; DFS: Disease Free Survival; DSS: Disease Specific Survival; RFS: Recurrence Free Survival; PPS: Post-progression Survival; DMPS: Disease Median Progression Survival; FP: Free Progression; CeRNA: Competing endogenous RNAs; P: Phosphorylation; Ser: Serine; H8: HEAT 8; H9: HEAT 9; PR: Polar Residues; BAR: Basic and Acidic Residues.

Author Contributions

Yingjun Xie (XYJ): Conceptualization and Methodology, Administrative support: Jie Lin (LJ): Provide learning materials or patients, Writing-Original draft preparation. Gangyi Li (LGY): Data Collection and Analysis, Writing-Original draft preparation. Yanping Bai (BYP): Data analysis and interpretation.

Conflicts of Interest

The authors declare no conflicts of interest related to this study.

Ethical Statement

This study was registered in PROSPERO (registration number CRD42022333964).

Funding

Special Medical and Health Personnel Program of Jilin Province Finance Department (2019SCZT015), Health Technology Innovative Program of Jilin Province (2018J053).

References

  • 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68:394–424. https://doi.org/10.3322/caac.21492 [PubMed]
  • 2. Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L. Current Challenges in Cancer Treatment. Clin Ther. 2016; 38:1551–66. https://doi.org/10.1016/j.clinthera.2016.03.026 [PubMed]
  • 3. Murphy LA, Sarge KD. Phosphorylation of CAP-G is required for its chromosomal DNA localization during mitosis. Biochem Biophys Res Commun. 2008; 377:1007–11. https://doi.org/10.1016/j.bbrc.2008.10.114 [PubMed]
  • 4. Gu C, Wang Z, Zhou N, Li G, Kou Y, Luo Y, Wang Y, Yang J, Tian F. Mettl14 inhibits bladder TIC self-renewal and bladder tumorigenesis through N6-methyladenosine of Notch1. Mol Cancer. 2019; 18:168. https://doi.org/10.1186/s12943-019-1084-1 [PubMed]
  • 5. Wu Y, Lin Y, Pan J, Tu X, Xu Y, Li H, Chen Y. NCAPG promotes the progression of lung adenocarcinoma via the TGF-β signaling pathway. Cancer Cell Int. 2021; 21:443. https://doi.org/10.1186/s12935-021-02138-w [PubMed]
  • 6. Jiang L, Ren L, Chen H, Pan J, Zhang Z, Kuang X, Chen X, Bao W, Lin C, Zhou Z, Huang D, Yang J, Huang H, et al. NCAPG confers trastuzumab resistance via activating SRC/STAT3 signaling pathway in HER2-positive breast cancer. Cell Death Dis. 2020; 11:547. https://doi.org/10.1038/s41419-020-02753-x [PubMed]
  • 7. Gong C, Ai J, Fan Y, Gao J, Liu W, Feng Q, Liao W, Wu L. NCAPG Promotes The Proliferation Of Hepatocellular Carcinoma Through PI3K/AKT Signaling. Onco Targets Ther. 2019; 12:8537–52. https://doi.org/10.2147/OTT.S217916 [PubMed]
  • 8. Sun DP, Lin CC, Hung ST, Kuang YY, Hseu YC, Fang CL, Lin KY. Aberrant Expression of NCAPG is Associated with Prognosis and Progression of Gastric Cancer. Cancer Manag Res. 2020; 12:7837–46. https://doi.org/10.2147/CMAR.S248318 [PubMed]
  • 9. Zhou Y, Fan Y, Mao Y, Lou M, Liu X, Yuan K, Tong J. NCAPG is a prognostic biomarker of immune infiltration in non-small-cell lung cancer. Biomark Med. 2022; 16:523–35. https://doi.org/10.2217/bmm-2021-1090 [PubMed]
  • 10. Sang L, Wu X, Yan T, Naren D, Liu X, Zheng X, Zhang N, Wang H, Li Y, Gong Y. The m6A RNA methyltransferase METTL3/METTL14 promotes leukemogenesis through the mdm2/p53 pathway in acute myeloid leukemia. J Cancer. 2022; 13:1019–30. https://doi.org/10.7150/jca.60381 [PubMed]
  • 11. Liu W, Liang B, Liu H, Huang Y, Yin X, Zhou F, Yu X, Feng Q, Li E, Zou Z, Wu L. Overexpression of non-SMC condensin I complex subunit G serves as a promising prognostic marker and therapeutic target for hepatocellular carcinoma. Int J Mol Med. 2017; 40:731–8. https://doi.org/10.3892/ijmm.2017.3079 [PubMed]
  • 12. Zheng G, Han T, Hu X, Yang Z, Wang J, Wen Z, Li H, Wang H. NCAPG Promotes Tumor Progression and Modulates Immune Cell Infiltration in Glioma. Front Oncol. 2022; 12:770628. https://doi.org/10.3389/fonc.2022.770628 [PubMed]
  • 13. Liang ML, Hsieh TH, Ng KH, Tsai YN, Tsai CF, Chao ME, Liu DJ, Chu SS, Chen W, Liu YR, Liu RS, Lin SC, Ho DM, et al. Downregulation of miR-137 and miR-6500-3p promotes cell proliferation in pediatric high-grade gliomas. Oncotarget. 2016; 7:19723–37. https://doi.org/10.18632/oncotarget.7736 [PubMed]
  • 14. Cohen Y, Gutwein O, Garach-Jehoshua O, Bar-Haim A, Kornberg A. The proliferation arrest of primary tumor cells out-of-niche is associated with widespread downregulation of mitotic and transcriptional genes. Hematology. 2014; 19:286–92. https://doi.org/10.1179/1607845413Y.0000000125 [PubMed]
  • 15. Hu SP, Ge MX, Gao L, Jiang M, Hu KW. LncRNA HCP5 as a potential therapeutic target and prognostic biomarker for various cancers: a meta-analysis and bioinformatics analysis. Cancer Cell Int. 2021; 21:686. https://doi.org/10.1186/s12935-021-02404-x [PubMed]
  • 16. 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. https://doi.org/10.1093/nar/gkx247 [PubMed]
  • 17. Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, Ceccarelli M, Bontempi G, Noushmehr H. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016; 44:e71. https://doi.org/10.1093/nar/gkv1507 [PubMed]
  • 18. Győrffy B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Comput Struct Biotechnol J. 2021; 19:4101–9. https://doi.org/10.1016/j.csbj.2021.07.014 [PubMed]
  • 19. Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVS, Varambally S. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia. 2017; 19:649–58. https://doi.org/10.1016/j.neo.2017.05.002 [PubMed]
  • 20. Franz M, Rodriguez H, Lopes C, Zuberi K, Montojo J, Bader GD, Morris Q. GeneMANIA update 2018. Nucleic Acids Res. 2018; 46:W60–4. https://doi.org/10.1093/nar/gky311 [PubMed]
  • 21. Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, Franz M, Grouios C, Kazi F, Lopes CT, Maitland A, Mostafavi S, Montojo J, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010; 38:W214–20. https://doi.org/10.1093/nar/gkq537 [PubMed]
  • 22. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, Jensen LJ, von Mering C. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021; 49:D605–12. https://doi.org/10.1093/nar/gkaa1074 [PubMed]
  • 23. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, Jensen LJ, von Mering C. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017; 45:D362–8. https://doi.org/10.1093/nar/gkw937 [PubMed]
  • 24. Wang Y, Gao B, Tan PY, Handoko YA, Sekar K, Deivasigamani A, Seshachalam VP, OuYang HY, Shi M, Xie C, Goh BKP, Ooi LL, Man Hui K. Genome-wide CRISPR knockout screens identify NCAPG as an essential oncogene for hepatocellular carcinoma tumor growth. FASEB J. 2019; 33:8759–70. https://doi.org/10.1096/fj.201802213RR [PubMed]
  • 25. Wang X, Tian X, Sui X, Li X, Zhao X, Han K, Sun L, Dong Y. Increased expression of NCAPG (Non-SMC condensing I complex subunit G) is associated with progression and poor prognosis of lung adenocarcinoma. Bioengineered. 2022; 13:6113–25. https://doi.org/10.1080/21655979.2022.2035124 [PubMed]
  • 26. Sun H, Zhang H, Yan Y, Li Y, Che G, Zhou C, Nicot C, Ma H. NCAPG promotes the oncogenesis and progression of non-small cell lung cancer cells through upregulating LGALS1 expression. Mol Cancer. 2022; 21:55. https://doi.org/10.1186/s12943-022-01533-9 [PubMed]
  • 27. Cai X, Gao J, Shi C, Guo WZ, Guo D, Zhang S. The role of NCAPG in various of tumors. Biomed Pharmacother. 2022; 155:113635. https://doi.org/10.1016/j.biopha.2022.113635 [PubMed]
  • 28. Dong M, Xu T, Cui X, Li H, Li X, Xia W. NCAPG upregulation mediated by four microRNAs combined with activation of the p53 signaling pathway is a predictor of poor prognosis in patients with breast cancer. Oncol Lett. 2021; 21:323. https://doi.org/10.3892/ol.2021.12585 [PubMed]
  • 29. Shi Y, Ge C, Fang D, Wei W, Li L, Wei Q, Yu H. NCAPG facilitates colorectal cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition by activating the Wnt/β-catenin signaling pathway. Cancer Cell Int. 2022; 22:119. https://doi.org/10.1186/s12935-022-02538-6 [PubMed]
  • 30. Zhang X, Zhu M, Wang H, Song Z, Zhan D, Cao W, Han Y, Jia J. Overexpression of NCAPG inhibits cardia adenocarcinoma apoptosis and promotes epithelial-mesenchymal transition through the Wnt/β-catenin signaling pathway. Gene. 2021; 766:145163. https://doi.org/10.1016/j.gene.2020.145163 [PubMed]
  • 31. Zhang X, Wang H, Han Y, Zhu M, Song Z, Zhan D, Jia J. NCAPG Induces Cell Proliferation in Cardia Adenocarcinoma via PI3K/AKT Signaling Pathway. Onco Targets Ther. 2020; 13:11315–26. https://doi.org/10.2147/OTT.S276868 [PubMed]
  • 32. Liu C, Yan Y, Di F, Li W, Yin X, Dong L. Inhibition of NCAPG expression inactivates the Wnt/β-catenin signal to suppresses endometrial cancer cell growth in vitro. Environ Toxicol. 2021; 36:2512–20. https://doi.org/10.1002/tox.23364 [PubMed]
  • 33. Li J, Sun S, Li J, Zhao X, Li Z, Sha T, Cui Z. NCAPG, mediated by miR-378a-3p, regulates cell proliferation, cell cycle progression, and apoptosis of oral squamous cell carcinoma through the GSK-3β/β-catenin signaling. Neoplasma. 2021; 68:1201–11. https://doi.org/10.4149/neo_2021_210421N544 [PubMed]
  • 34. Arai T, Okato A, Yamada Y, Sugawara S, Kurozumi A, Kojima S, Yamazaki K, Naya Y, Ichikawa T, Seki N. Regulation of NCAPG by miR-99a-3p (passenger strand) inhibits cancer cell aggressiveness and is involved in CRPC. Cancer Med. 2018; 7:1988–2002. https://doi.org/10.1002/cam4.1455 [PubMed]
  • 35. Tang F, Yu H, Wang X, Shi J, Chen Z, Wang H, Wan Z, Fu Q, Hu X, Zuhaer Y, Liu T, Yang Z, Peng J. NCAPG promotes tumorigenesis of bladder cancer through NF-κB signaling pathway. Biochem Biophys Res Commun. 2022; 622:101–7. https://doi.org/10.1016/j.bbrc.2022.07.007 [PubMed]
  • 36. Yu H, Zou D, Ni N, Zhang S, Zhang Q, Yang L. Overexpression of NCAPG in ovarian cancer is associated with ovarian cancer proliferation and apoptosis via p38 MAPK signaling pathway. J Ovarian Res. 2022; 15:98. https://doi.org/10.1186/s13048-022-01030-z [PubMed]