Skip to main content
Journal of Oncology logoLink to Journal of Oncology
. 2022 Aug 25;2022:7204415. doi: 10.1155/2022/7204415

Prognostic Signature and Therapeutic Value Based on Membrane Lipid Biosynthesis-Related Genes in Breast Cancer

Yingkun Xu 1, Yudi Jin 2, Shun Gao 1, Yuan Wang 1, Chi Qu 1, Yinan Wu 1, Nan Ding 1, Yuran Dai 1, Linshan Jiang 1, Shengchun Liu 1,
PMCID: PMC9436593  PMID: 36059802

Abstract

There is a need to improve diagnostic and therapeutic approaches to enhance the prognosis of breast cancer, the most common malignancy worldwide. Membrane lipid biosynthesis is a hot biological pathway in current cancer research. It is unclear whether membrane lipid biosynthesis is involved in the prognosis of BRCA. With LASSO regression, a 14-gene prediction model was constructed using data from the TCGA-BRCA cohort. The prediction model includes GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, HACD1, SGPP1, PRKD2, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3. BRCA patients from the TCGA-BRCA cohort were divided into two risk subgroups based on the model. Kaplan–Meier survival curves showed that patients with lower risk scores had significantly improved overall survival (P=2.49e − 09). In addition, risk score, age, stage, and TNM classification were used to predict mortality in BRCA patients. In addition, the 14 genes in the risk model were analyzed for gene variation, methylation level, drug sensitivity, and immune cell infiltration, and the miRNA-mRNA network was constructed. Afterward, the THPA website then analyzed the protein expression of 14 of these risk model genes in normal and pathological BRCA tissues. In conclusion, the membrane lipid biosynthesis-related risk model and nomogram can be used to predict BRCA clinical prognosis.

1. Introduction

Breast cancer is the most common cancer in women and one of the leading causes of cancer death [13]. Breast cancer mainly refers to malignant tumors in breast epithelial tissue, with a high clinical cure rate early. In contrast, advanced breast cancer is highly aggressive, malignant, and has sizeable histological heterogeneity [4, 5]. In the actual diagnosis and treatment process, the clinical symptoms of early breast cancer are not obvious, which is easy to cause misdiagnosis and missed diagnosis, leading to breast cancer patients missing the best time for treatment [6, 7]. At the same time, there is still no effective treatment for breast cancer patients with metastases [8]. With the continuous development of molecular biology, gene therapy has become a new direction in breast cancer treatment. However, breast cancer is a complex disease, and its occurrence and development are affected by many molecular factors. Therefore, studying the pathogenesis of breast cancer is of great significance to its treatment.

Cancer cells, as mutated cells, are the source of cancer. Unlike normal cells, cancer cells have three characteristics of infinite proliferation, transformation, and easy transfer [9]. They can proliferate indefinitely and destroy normal cells and tissues. In addition to dividing out of control, cancer cells can invade surrounding normal tissues locally and even metastasize to other body parts via the circulatory system or lymphatic system. By remodeling their metabolism, cancer cells provide ATP and macromolecules necessary for cell growth, division, and survival. To meet their rapid proliferation needs, tumor cells exhibit metabolic characteristics different from normal tissue cells [10, 11]. Although cancer types and etiologies are different, there are certain similarities in the changes in metabolic pathways in cancer cells. Glucose, pentose phosphate, and glutamine metabolic pathways adapt to cancer cells' rapid growth and proliferation. Recent studies have shown that membrane lipid biosynthesis is also altered in cancer cells [1214]. In cancer development, changes in membrane lipid biosynthesis play an essential role in providing biomembrane macromolecules and signaling molecules for the occurrence and development of cancer [1518]. Alterations in membrane lipid biosynthesis in tumor tissues provide macromolecular precursors necessary for their abnormal survival and growth. This suggests that clinical actions targeting this critical metabolism could be a highly effective therapeutic approach [1923].

As a computer science and high-throughput sequencing technology developed, bioinformatic analysis can provide researchers with more opportunities to explore the genetic risk, regulatory mechanism, and protein function of cancer. As a highly representative bioinformatics database, the TCGA database is generated by large-scale gene sequencing, which can quantitatively study and analyze the changes in gene expression during tumor occurrence and development [24, 25]. Therefore, in this study, we performed a series of in-depth explorations of membrane lipid biosynthesis in BRCA based on the TCGA database using multiple bioinformatic analyses. Our findings suggest that targeting membrane lipid biosynthesis has excellent potential for BRCA prevention, treatment, and control.

2. Materials and Methods

2.1. Data Acquisition and Selection of Membrane Lipid Biosynthesis-Related Genes

This study downloaded BRCA mRNA expression and clinical information from TCGA (https://portal.gdc.cancer.gov/). A total of 1,109 tumor samples and 113 normal samples with clinical and expression data were obtained. Expression data were normalized using log2 (TPM + 1) transformation. Transcript data and human profiles are matched and sorted by Perl to get comprehensive and accurate mRNA gene expression data using information from an integrated database. Gene IDs are converted to gene names for subsequent analysis. Membrane lipid biosynthesis-related genes were obtained from the Gene Set Enrichment Analysis (GSEA) database (https://www.gsea-msigdb.org/gsea/index.jsp) [26, 27]. The standard name is GOBP membrane lipid biosynthetic process. The systematic name is M16158. Differential expression analysis was carried out on TCGA data using the “limma” R package. Univariate Cox analysis was used to identify membrane lipid biosynthesis-related genes with prognostic values (P < 0.05). The TCGA cohort was evaluated for membrane lipid biosynthesis-related genes with predictive value.

2.2. Construction and Validation of a Prognostic Signature of Membrane Lipid Biosynthesis-Related Genes

A predictive model was constructed in BRCA using LASSO regression analysis to minimize the use of overlapping membrane lipid biosynthesis-related gene risk values. Statistics and machine learning use LASSO regression to identify the most critical factors and improve the accuracy of statistical models. LASSO is a popular machine learning algorithm widely used in medical research [25, 28, 29]. In our study, risk scores were obtained for all patients and then divided into high- or low-risk subgroups based on the median risk score of the TCGA cohort. The “survival,” “survminer,” and “timeROC” packages in R were used to perform KM survival analysis and ROC analysis based on OS to estimate the predictive accuracy of the two sets of gene signatures.

2.3. Prognostic Independence of Prognostic Signature from Routine Clinicopathological Features and Generation of the Corresponding Nomogram

To further evaluate the gene prognostic signature's independent predictive value, it was examined using univariate and multivariate Cox regression analysis to see if it was affected by other clinical traits such as age, gender, stage, and TNM stage. Corresponding nomograms were drawn using the R package “RMS” based on risk scores and other routine clinicopathological features.

2.4. Prognostic Value and Gene Variation of Risk Model Genes Associated with Membrane Lipid Biosynthesis in Pan-Cancer

In this study, we first targeted the membrane lipid biosynthesis-related risk model genes GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, HACD1, SGPP1, PRKD2, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3 were analyzed for OS, CNV, SNV, methylation, and immune cell infiltration in pan-cancer via Gene Set Cancer Analysis (GSCA) database (https://bioinfo.life.hust.edu.cn/GSCA/#/) [30]. We then used this database to drill down to the percentage of cancers, in which mRNA expression of membrane lipid biosynthesis-related risk model genes had a potential impact (activation or inhibition) on biological pathways. Finally, we used this database to construct a miRNA regulatory network for membrane lipid biosynthesis-related risk model genes.

2.5. Anticancer Drug Sensitivity Analysis of Risk Model Genes Associated with Membrane Lipid Biosynthesis

This study integrated the drug sensitivity and gene expression profiling data from Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Therapeutics Response Portal (CTRP). The correlation between small molecule/drug sensitivity (IC50) and the expression of 14 membrane lipid biosynthesis-related risk model genes was analyzed by Spearman correlation. GDSC was developed by the Sanger Institute in the United Kingdom to collect the sensitivity and response of tumor cells to drugs. Raw data from GDSC comes from 75,000 experiments describing the response of about 200 anticancer drugs in more than 1,000 tumor cells. The oncogene mutation information in this database comes from the COSMIC database, including oncogene point mutations, gene amplification and loss, tissue types, and expression profiles (https://www.cancerrxgene.org/) [3133]. In addition, CTRP, a database of 70,000 cancer cell line compounds that link susceptibility to genetic or lineage characteristics, serves as a public resource for researchers worldwide (https://portals.broadinstitute.org/ctrp.v2.1/) [3436]. This important resource drives the discovery of potential cancer drugs that match the patient population and maximize the likelihood that patients will benefit from them. Such data are therefore crucial for the discovery of potential tumor therapeutic targets.

2.6. Gene Set Enrichment Analysis and Protein Expression Analysis of Membrane Lipid Biosynthesis-Related Risk Model Genes in BRCA

Gene Set Enrichment Analysis (GSEA) is a method for enrichment analysis of target genes, which can be used to test the correlation between target genes and gene sets of known functions. This study used the GTBA database to explore the relationship between membrane lipid biosynthesis-related risk model genes and the HALLMARK pathway (https://guotosky.vip:13838/GTBA/). The Human Protein Atlas (THPA) database is based on proteomics, transcriptomics, and systems biology data, which can map tissues, cells, organs, etc. This database includes protein expression in tumors and normal tissues (https://www.proteinatlas.org/) [37, 38]. Therefore, in this study, we used this database to explore the expression of risk model genes related to membrane lipid synthesis in breast cancer and normal breast tissue.

2.7. Tumor Immune Estimation Resource (TIMER) Database

A comprehensive database of immune infiltration in various cancer types, the TIMER database offers multiple methods of assessing immune infiltration levels, providing researchers with the ability to generate high-quality graphs online to explore immunomics, clinical manifestations, and tumor genomes. This study used this database to assess the association between membrane lipid biosynthesis-related risk model genes and immune cell infiltration in BRCA.

2.8. Statistical Analysis

Statistical analysis was performed using R 4.1.2 and Strawberry Perl software. The non-parametric rank test (Wilcox test) was used to analyze the expression differences of membrane lipid biosynthesis-related genes in breast cancer and normal breast tissue samples. The Kaplan–Meier analysis of gene expression was used to analyze the relationship between breast cancer survival and gene expression, and the significance of the breast cancer risk model was assessed using univariate and multivariate Cox regression analysis. The relationship between gene expression and clinicopathological characteristics was analyzed by χ2 test.

3. Results

3.1. Gene Expression and Protein Interaction of Membrane Lipid Biosynthesis-Related Molecules in BRCA

It is well known that the process of carcinogenesis is often accompanied by abnormal expression of gene mRNA. These abnormally expressed genes are of more interest to cancer researchers than those that are not [9, 39]. To explore whether there are differences in the expression of membrane lipid biosynthesis-related genes in BRCA, we extracted the mRNA expression data of membrane lipid biosynthesis-related genes from the TCGA database. We used the R language to draw a corresponding heatmap (Figure 1(a)). We can intuitively see that the mRNA expression of the vast majority of membrane lipid biosynthesis-related genes is significantly different in BRCA. Then, to explore the interaction of the protein molecules encoded by membrane lipid biosynthesis-related genes, we used the String website to draw the corresponding PPI network (Figure 1(b)). The PPI network revealed extensive interactions of membrane lipid biosynthesis-related molecules. To further explore the relationship, we used the MCODE algorithm of the Metascape website to perform cluster analysis on the PPI network to identify subnetworks, that is, potential protein complexes (Figures 1(c) and 1(d)) (Supplementary Material Table S1). The results showed that the top three subnetworks were sphingolipid metabolism, membrane lipid metabolic process, and fatty acid elongation. Finally, the results of univariate Cox regression analysis showed that GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, PIGU, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3 play an inhibitory role in the malignant progression of BRCA, in contrast, HACD1, SGPP1, and PRKD2 play an inhibitory role in BRCA malignant progression (Figure 1(e)).

Figure 1.

Figure 1

Expression and interaction of membrane lipid biosynthesis-related molecules in BRCA. (a) Heat map showing the mRNA expression of membrane lipid biosynthesis-related genes in BRCA. (b) Protein-protein interaction network showing interactions of proteins encoded by genes involved in membrane lipid biosynthesis. (c, d) Protein interaction network diagrams show clustering analysis results based on the MCODE algorithm. (e) The forest plot shows the univariate Cox analysis of membrane lipid biosynthesis-related genes in BRCA. P < 0.05, ∗∗P < 0.01, and ∗∗∗P < 0.001.

3.2. Construction of Risk Models Associated with Membrane Lipid Biosynthesis in BRCA

To fully explore the prognostic value of membrane lipid biosynthesis-related genes in BRCA, we used LASSO regression curve analysis to establish a prognostic risk model in BRCA, which included GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, HACD1, SGPP1, PRKD2, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3 (Figures 2(a) and 2(b)). LASSO regression curve analysis is often used in medical modeling [28, 29]. Based on the calculation formula of the risk model related to membrane lipid biosynthesis, we can divide breast cancer patients into high-risk and low-risk groups. The drawn survival curve shows that the survival rate of breast cancer patients in the high-risk group is significantly lower than that in the low-risk group. cancer patients (P=2.49e − 09) (Figure 2(c)). To test the predictive accuracy of this model of membrane lipid biosynthesis-related risk, we performed a ROC curve analysis, which showed that the five-year AUC was equal to 0.69, the seven-year AUC was equal to 0.722, and the ten-year AUC was equal to 0.729 (Figures 2(d)2(f)). This indicates that this membrane lipid biosynthesis-related risk model has good predictive accuracy. The formula for calculating the risk model is as follows.

Figure 2.

Figure 2

Construction of risk models using membrane lipid biosynthesis-related prognostic genes in BRCA. (a) LASSO regression of OS-related genes. (b) Cross‐validation for tuning the parameter selection. (c) Based on this membrane lipid biosynthesis-related risk model, the Kaplan–Meier curve in BRCA showed that the overall survival rate of the low-risk group was significantly higher than that of the high-risk group (P=2.49e − 09). Among them, blue represents the low-risk group and red represents the high-risk group. (d–f) The AUC of the prediction of 5-, 7-, and 10‐year survival rates of BRCA.

Risk model = 0.004049719 GPAA1 + 0.119845071 PIGF + 0.002817462 ST3GAL1 + 0.019867802 ST6GALNAC4 + 0.013021966 PLPP2 + 0.007145373 ELOVL1 + 0.00779386 VAPB + 0.000841801  CERS2 + 0.045125517 SGMS2 + 0.001158905 ALDH3B2 + 0.002111798 HACD3 − 0.116447762 HACD1 − 0.040517734 SGPP1 − 0.023673124 PRKD2.

3.3. Independent Predictive Value of Risk Models Associated with Membrane Lipid Biosynthesis and Construction of Corresponding Predictive Survival Nomogram in BRCA

To gain an in-depth understanding of the correlation between membrane lipid biosynthesis-related risk models and clinical clinicopathological characteristics, we obtained the corresponding BRCA clinical data through the TCGA official website and performed a correlation analysis. The results are presented in the form of a heatmap. The results showed a significant correlation between this membrane lipid biosynthesis-related risk model and stage, age, and fustat in breast cancer patients (Figure 3(a)). We subsequently performed univariate and multivariate Cox regression analyses to explore the role of this membrane lipid biosynthesis-related risk model and clinicopathological features in breast cancer progression. Univariate Cox regression analysis showed that age, stage, T, M, N, and risk score were risk factors for the malignant progression of breast cancer (Figure 3(b)). Multivariate Cox regression analysis showed that age and risk score were independent risk factors for the malignant progression of breast cancer (Figure 3(c)). Finally, based on the risk model associated with membrane lipid biosynthesis, we drew corresponding nomograms to predict breast cancer patients 5-, 7-, and 10-year survival (Figure 3(d)). The above results indicate that this membrane lipid biosynthesis-related risk model has solid clinical relevance and clinical application value in BRCA.

Figure 3.

Figure 3

Based on this risk model associated with membrane lipid biosynthesis, univariate and multivariate Cox regression curve analysis was performed and a corresponding nomogram was generated in BRCA. (a) Heatmap showing correlations between membrane lipid biosynthesis-related risk models and clinicopathological features of breast cancer patients. (b, c) Forest plots showing the results of univariate Cox analysis and multivariate Cox analysis. (d) Based on this membrane lipid biosynthesis-related risk model, a corresponding nomogram was drawn to predict breast cancer patients' 5-, 7-, and 10-year survival.

3.4. Gene Expression and Prognostic Value of Risk Model Genes Associated with Membrane Lipid Biosynthesis in Pan-Cancer

To further understand the significance of membrane lipid biosynthesis-related risk model genes in pan-cancer, first, we explored the mRNA expression differences of these fourteen risk model genes in pan-cancer and presented them in the form of a heat map. The results showed that these fourteen risk model genes were widely differentially expressed in LUSC, LUAD, BRCA, LIHC, and THCA (Figure 4(a)). Subsequently, we performed a biological pathway correlation analysis for these fourteen risk model genes. The results showed that its mRNA expression was negatively correlated with the inhibition of DNA Damage, EMT, and Hormone AR pathways. In contrast, its mRNA expression was positively associated with activation of Apoptosis, Cell Cycle, and EMT pathways (Figure 4(b)). Furthermore, to closely link the clinic, we explored the predictive value of risk model genes associated with membrane lipid biosynthesis in pan-cancer. The results showed that these fourteen risk model genes were significantly associated with DSS, OS, and PFS in patients with multiple types of cancer (Figure 4(c)). We then performed a Gene set variation analysis (GSVA) on these fourteen risk model genes to explore the differences in multiple tumors of the membrane lipid biosynthesis-related risk model genes. GSVA is a non-parametric, unsupervised algorithm that transforms gene expression data, from an expression matrix featuring a single gene to an expression matrix featuring a specific set of genes. GSVA quantified gene enrichment results, making the subsequent statistical analysis more convenient. Boxplots show that the GSVA scores of these fourteen risk model genes tended to increase in various tumors, including BLCA, BRCA, ESCA, LUAD, and THCA (Figure 4(d)). The biological pathway correlation analysis showed a negative correlation between GSVA score and DNA Damage (Figure 4(e)). The above results suggest that this membrane lipid biosynthesis-related risk model gene may play a similar biological function in pan-cancer and provide meaningful clues for future cancer research.

Figure 4.

Figure 4

Differences in expression, prognosis, and biological pathway correlation of risk model genes associated with membrane lipid biosynthesis in pan-cancer. (a)–(b) Heatmaps showing differential mRNA expression of these membrane lipid biosynthesis-related risk model genes in pan-cancer and their correlation with biological pathways. (c) Heat map showing DFI, DSS, OS, and PFS of these membrane lipid biosynthesis-related risk model genes in pan-cancer. (d, e) These show how the GSVA scores of these membrane lipid biosynthesis-related risk model genes differ in pan-cancer and their correlation with biological pathways. The red represents the activated biological pathway, and the blue represents the inhibited biological pathway.

3.5. Genetic Variation of Risk Model Genes Associated with Membrane Lipid Biosynthesis in Pan-Cancer

Variations in the human genome play an essential role in inherited diseases. In addition to DNA point mutations, the genome involves variation in large DNA sequences, including microduplications and microdeletions of submicroscopic structures. Therefore, we further explored the genetic variation of risk model genes related to membrane lipid biosynthesis in pan-cancer. In the SNV variant analysis results, we found that PRKD2, SGPP1, and GPAA1 have widespread high CNVs in UCEC, SKCM, COAD, and STAD. The results also showed that the top 10 SNV variants were PRKD2, ALDH3B2, GPAA1, ST3GAL1, SGPP1, CERS2, SGMS2, HACD1, PLPP2, and HACD3. Among them, PRKD2 accounted for as high as 22% of SNVs in pan-cancer (Figures 5(a)5(d)). Since CNVs are an essential part of genomic variation, we then explored the CNVs of membrane lipid biosynthesis-related risk model genes in pan-cancer. The results showed that CERS2, VAPB, GPAA1, and ST3GAL1 were widespread CNVs in pan-cancer (Figure 5(e)). In LUAD, LUSC, OV, BRCA, and UCS, CNVs of GPAA1, VAPB, ERS2, PRKD2, and ELOVL1 showed a positive correlation with their mRNA expression (Figure 5(f)).

Figure 5.

Figure 5

Variation of risk model genes associated with membrane lipid biosynthesis in pan-cancer. (a–d) These show the SNVs of these membrane lipid biosynthesis-related risk model genes in pan-cancer. (e) Heatmap showing the CNVs of these membrane lipid biosynthesis-related risk model genes in pan-cancer. (f) Heatmap showing the correlation between CNV and mRNA expression of risk model genes.

3.6. Methylation and Drug Sensitivity of Risk Model Genes Associated with Membrane Lipid Biosynthesis in Pan-Cancer

Epigenetic studies are the characteristics of heritable changes outside the DNA sequence. All cells of a living individual have essentially the same DNA. Still, different organs and tissues have different functions and maintain their specific cellular identity over multiple cell divisions, which are primarily thought to be determined by the appearance of mediated by genetic information [9, 40]. Since methylation is essential to epigenetic research, we explored the methylation differences of membrane lipid biosynthesis-related genes in various tumors [41, 42]. We presented them in the form of heat maps. The results showed that ALDH3B2, ST3GAL1, and SGMS2 genes had noticeable methylation differences in BRCA, KIRC, and LUSC (Figures 6(a) and 6(b)). Because mutations in the cancer genome can affect the efficacy of clinical treatments, responses to anticancer drugs vary widely across different therapeutic targets. Therefore, we used data from GDSC and CTRP databases to explore the sensitivity of membrane lipid biosynthesis-related genes to various anticancer drugs. The results showed that SGMS2, PLPP2, HACD1, ELOVL1, ALDH3B2, and VAPB significantly correlated with the sensitivity of various anticancer drugs. Meanwhile, there were significant negative correlations between PRKD2, ST6GALNAC4, and HACD1 sensitivities to multiple anticancer drugs (Figures 6(c) and 6(d)). We believe our study provides valuable data for future drug target development.

Figure 6.

Figure 6

Correlations between methylation, drug sensitivity, and mRNA expression of risk model genes associated with membrane lipid biosynthesis. (a) Heatmap showing differences in methylation levels of these membrane lipid biosynthesis-related risk model genes across multiple tumors. (b) Heat map showing the correlation between methylation levels and mRNA expression of these membrane lipid biosynthesis-related risk model genes. (c, d) Heatmaps showing correlations between these membrane lipid biosynthesis-related risk model genes and susceptibility to multiple anticancer drugs in the GDSC and CTRP databases.

3.7. Immune Cell Infiltration of Membrane Lipid Biosynthesis-Related Risk Model Genes in BRCA

We all know that tumor tissue is not simply composed of tumor cells, it is composed of various types of cells, including stromal cells, fibroblasts, immune cells, etc. These cells constitute the tumor microenvironment [43, 44]. In recent years, we have paid more attention to the role of immune cells in this microenvironment [4547]. Immune cells include many kinds, such as B cells and T cells. Different immune cells play different roles in the process of tumorigenesis, and the composition of immune cells of various tumors also has its characteristics. Because of the importance of immune cell infiltration in tumor progression, based on the Timer database, we examined six types of immune cell infiltration and this membrane lipid biosynthesis-related gene in BRCA. The results showed a significant positive correlation between ST3GAL1, SGPP1, VAPB, and SGMS2 and CD8+ T Cell infiltration (Figures 7(a)7(n)).

Figure 7.

Figure 7

Scatter plot showing the correlation between membrane lipid biosynthesis-related risk model genes and immune cell infiltration in BRCA. (a–n) GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, HACD1, SGPP1, PRKD2, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3; the data come from TIMER database.

3.8. Biological Pathway Enrichment Analysis and Protein Expression of Membrane Lipid Biosynthesis-Related Risk Model Genes in BRCA

In this study, we performed GSEA on the HALLMARK gene set for membrane lipid biosynthesis-related risk model genes in BRCA (Figures 8(a) and 8(b)) [26, 48]. GSEA results showed that these risk model genes were associated with abnormal activation of PI3K AKT MTOR SIGNALING, PROTEIN SECRETION, and MTORC1 SIGNALING, HEME METABOLISM, and OXIDATIVE PHOSPHORYLATION. Subsequently, we used immunohistochemical data from the THPA database to validate our previous findings to explore the expression of risk model genes associated with membrane lipid biosynthesis in mammary and normal breast tissues. Since ST6GALNAC4 and HACD1 have not been included in the THPA database, we provide the immunohistochemical results for ALDH3B2, CERS2, ELOVL1, GPAA1, HACD3, PIGF, PLPP2, PRKD2, SGMS2, SGPP1, ST3GAL1, and VAPB. The results showed that the protein expression levels confirmed our previous findings at the mRNA level, confirming our findings were correct (Figures 8(c)8(n)). These data may provide a solid basis for further research into these risk model genes and pave the way for future interventions.

Figure 8.

Figure 8

Pathway enrichment and protein expression analysis of membrane lipid biosynthesis-related risk model genes in BRCA. (a, b) These show pathway enrichment analysis in BRCA for these membrane lipid biosynthesis-related risk model genes and their corresponding quantitative histograms. (c–n) Protein expression of membrane lipid biosynthesis-related risk model genes ALDH3B2, CERS2, ELOVL1, GPAA1, HACD3, PIGF, PLPP2, PRKD2, SGMS2, SGPP1, ST3GAL1, and VAPB in breast cancer and normal breast tissue; the picture data come from THPA database.

3.9. Construction of a Risk Model Genes-Related miRNA Interaction Network for Membrane Lipid Biosynthesis

MicroRNAs (miRNAs) are a class of endogenous non-coding RNAs with a length of about 22 nt, which regulate gene expression by complementary binding to target gene transcripts [49, 50]. In recent years, studies have found that miRNAs are closely related to the occurrence of cancer, and miRNAs are involved in regulating multiple aspects of tumors, including transcription, cell cycle regulation, apoptosis, angiogenesis, tumor invasion, and invasion metastasis. miRNAs can directly act as oncogenes or tumor suppressor genes to affect the occurrence and growth of tumors. To further explore the regulatory relationship between membrane lipid biosynthesis-related risk genes and miRNAs, we used the GSCALite database to map the miRNA-mRNA regulation network associated with membrane lipid biosynthesis-related risk genes (Figure 9). Among them, we can find a regulatory relationship between a variety of miRNAs and the SGMS2 gene, which suggests that we can target these miRNAs to regulate SGMS2 in the future, thereby affecting the progression of BRCA.

Figure 9.

Figure 9

miRNA regulatory networks associated with risk model genes related to membrane lipid biosynthesis. Among them, blue represents miRNA and yellow represents genes.

4. Discussion

Globally, cancer is the leading cause of death and a significant barrier to extending life [51]. With the aging population and the rapid development of society, the cancer burden in countries worldwide is overgrowing [52, 53]. It is worth noting that in 2020, breast cancer in women surpassed lung cancer as the most common cancer worldwide for the first time, with an estimated 2,261,419 new cases. Meanwhile, breast cancer is the fifth leading cause of cancer death globally, with 684,996 deaths. In recent years, the morbidity and mortality of female breast cancer worldwide have increased sharply, and the disease burden has also increased, becoming a significant global public health problem [54]. In recent years, the biological role of membrane lipid biosynthesis-related genes in cancer has attracted more and more attention from cancer researchers. Therefore, in this study, we used “glmnet” and “survival” extension packages to perform LASSO regression curve analysis. We utilized genes related to membrane lipid biosynthesis to construct a prognosis-related risk model in BRCA. The risk model contains fourteen genes, namely, GPAA1, PIGF, ST3GAL1, ST6GALNAC4, PLPP2, ELOVL1, HACD1, SGPP1, PRKD2, VAPB, CERS2, SGMS2, ALDH3B2, and HACD3.

GPAA1 is a crucial subunit of glycosylphosphatidylinositol transferase, responsible for binding GPI anchors to precursor proteins, but not involved in GPI anchor synthesis. Previous studies have shown that GPAA1 is abnormally expressed and amplified in various malignant tumors, such as liver, breast, gastric, and colorectal [5558]. Since GPAA1 is an enzyme, its chemical activity is vulnerable to the intervention of various physicochemical factors or inhibitors, so the development of its inhibitors is less complex and more drug-producing. Therefore, we believe that targeting GPAA1-mediated synthesis of GPI-anchored proteins is a potential tumor-targeted therapeutic option [55]. Furthermore, PIGF is a homodimeric glycoprotein belonging to the VEGF subfamily. It exists in two isoforms, PIGF-1 and PIGF-2, the latter having a heparin-binding domain. Like VEGF, it is a potent angiogenic factor. Tumor growth promotes the proliferation of tumor blood vessels by interacting with its receptors, thereby affecting tumor growth, infiltration, and metastasis [59, 60]. And ST3GAL1 regulates cancer progression by affecting the activity of the TGF-β signaling pathway in glioblastoma and breast cancer [61, 62].

Additionally, SGPP1 controls multiple cellular functions, including differentiation, proliferation, metastasis, cytoskeleton remodeling, senescence, and apoptosis [63]. SGPP1 is a tumor suppressor gene that plays a crucial role in cancer invasion and metastasis [6466]. In gastric cancer tissues, the expression of SGPP1 was down-regulated compared with adjacent tissues and cancer-free tissues. It was confirmed that the low expression of SGPP1 was positively correlated with the distant metastasis of lymph nodes and gastric cancer. SGPP1 downregulation can promote cell migration to epidermal growth factor (EGF), while SGPP1 overexpression can decrease chemotaxis to EGF [67]. There is evidence that PRKD2 regulates fundamental biological processes, including signal transduction, membrane trafficking, cell survival, migration, differentiation, and proliferation [6871]. PRKD2 affects drug resistance in various tumors such as breast cancer and leukemia by regulating tumor cell proliferation, apoptosis, metastasis, and invasion [72]. As in glioblastoma, PRKD2 can promote CDKN1A gene overexpression through p53-dependent or independent pathways and integrated extracellular signal regulation, promoting cellular senescence and affecting cell sensitivity [73]. Furthermore, SGMS2 is a key regulator involved in ceramide and sphingomyelin homeostasis. A previous study showed that high expression of SGMS2 is associated with breast cancer metastasis. SGMS2 promotes cancer cell invasion by enhancing TGF-β/smad signaling to initiate epithelial-mesenchymal transition [74].

Meanwhile, several other risk model genes were identified in this study (ST6GALNAC4, PLPP2, ELOVL1, HACD1, VAPB, CERS2, ALDH3B2, and HACD3) have not yet been explored in depth in breast cancer. In the future, we should focus on these genes' biological functions and roles in the malignant progression of breast cancer. This study still has some limitations, mainly divided into the following three points. First, the data of BRCA patients used in this study were extracted from TCGA, and data from more databases are still needed for validation; second, although the newly proposed risk model has apparent clinical significance, its underlying mechanisms are still not precise. Therefore, we plan to explore further the role of this risk model in single-center or multicenter clinical samples in the future. Third, due to experimental conditions and time constraints, we have not yet validated the biological functions of risk model genes in BRCA. Therefore, we hope to plan to explore the biological processes of these risk models in BRCA through in vivo and in vitro experiments in the future.

5. Conclusions

This study deeply explored the potential biological functions and application values of membrane lipid biosynthesis-related genes in various tumors, including breast cancer. Surprisingly, we successfully constructed a fourteen-gene risk model in BRCA using these lipid biosynthesis-related genes. Based on this risk model, we can easily divide BRCA patients into high-risk and low-risk groups. In addition, to facilitate future practical applications, we also draw a nomogram corresponding to this risk. Therefore, we believe that this study guides the precision treatment of breast cancer by interpreting genomic data and lays the foundation for future scientific research and molecular typing of BRCA.

Acknowledgments

The authors thank The Cancer Genome Atlas (TCGA) for providing publicly available data. In addition, Yingkun Xu is particularly grateful to Shipeng Guo of Chongqing Medical University for his assistance in the progress of this research. This research was funded by the National Natural Science Foundation of China (Grant no. 81772979), the Key Research and Development Project of Chongqing's Technology Innovation and Application Development Special Big Health Field (Grant no. CSTC2021jscx-gksb-N0027), and the Doctoral Research Innovation Project of the First Affiliated Hospital of Chongqing Medical University (Grant no. CYYY-BSYJSCXXM-202213).

Abbreviations

BRCA:

Breast cancer

TCGA:

The Cancer Genome Atlas

GSCA:

Gene set cancer analysis

THPA:

The Human Protein Atlas

ImmuCellAI:

Immune cell abundance identifier

GDSC:

Genomics of drug sensitivity in cancer

CTRP:

Cancer therapeutics response portal

LASSO:

Least absolute shrinkage and selection operator

GSEA:

Gene set enrichment analysis

GPAA1:

Glycosylphosphatidylinositol anchor attachment 1

PIGF:

Phosphatidylinositol glycan anchor biosynthesis class F

ST3GAL1:

ST3 beta-galactoside alpha-2,3-sialyltransferase 1

ST6GALNAC4:

ST6 N-acetylgalactosaminide alpha-2,6-sialyltransferase 4

PLPP2:

Phospholipid phosphatase 2

ELOVL1:

ELOVL fatty acid elongase 1

HACD1:

3-hydroxyacyl-CoA dehydratase 1

SGPP1:

Sphingosine-1-phosphate phosphatase 1

PRKD2:

Protein kinase D2

VAPB:

VAMP-associated proteins B and C

CERS2:

Ceramide synthase 2

SGMS2:

Sphingomyelin synthase 2

ALDH3B2:

Aldehyde dehydrogenase 3 family member B2

HACD3:

3-hydroxyacyl-CoA dehydratase 3

EGF:

Epidermal growth factor.

Data Availability

The data used to support the findings of this study are available from the corresponding author upon request.

Ethical Approval

Ethical approval was not required.

Consent

Consent is not applicable.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors' Contributions

Shengchun Liu and Yingkun Xu designed the research methods and analyzed the data. Yingkun Xu, Yudi Jin, and Shun Gao participated in data collection. Yuan Wang, Chi Qu, and Yinan Wu drafted the manuscript. Nan Ding, Yuran Dai, and Linshan Jiang revised the manuscript. All authors approved the release version and agreed to be responsible for all aspects of the work.

Supplementary Materials

Supplementary Materials

Table S1. Cluster analysis results of membrane lipid biosynthesis-related genes under the MCODE algorithm.

References

  • 1.Siegel R. L., Miller K. D., Fuchs H. E., Jemal A. Cancer statistics, 2022. CA: A Cancer Journal for Clinicians . 2022;72(1):7–33. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
  • 2.Harbeck N., Gnant M. Breast cancer. The Lancet . 2017;389(10074):1134–1150. doi: 10.1016/s0140-6736(16)31891-8. [DOI] [PubMed] [Google Scholar]
  • 3.Wilkinson L., Gathani T. Understanding breast cancer as a global health concern. British Journal of Radiology . 2022;95(1130) doi: 10.1259/bjr.20211033.20211033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yeo S. K., Guan J. L. Breast cancer: multiple subtypes within a tumor? Trends in Cancer . 2017;3(11):753–760. doi: 10.1016/j.trecan.2017.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Grabinski V. F., Brawley O. W. Disparities in breast cancer. Obstetrics & Gynecology Clinics of North America . 2022;49(1):149–165. doi: 10.1016/j.ogc.2021.11.010. [DOI] [PubMed] [Google Scholar]
  • 6.Lin N. U., Claus E., Sohl J., Razzak A. R., Arnaout A., Winer E. P. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer . 2008;113(10):2638–2645. doi: 10.1002/cncr.23930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wahba H. A., El-Hadaad H. A. Current approaches in treatment of triple-negative breast cancer. Cancer Biology and Medicine . 2015;12(2):106–116. doi: 10.7497/j.issn.2095-3941.2015.0030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lebert J. M., Lester R., Powell E., Seal M., McCarthy J. Advances in the systemic treatment of triple-negative breast cancer. Current Oncology . 2018;25(11):S142–s150. doi: 10.3747/co.25.3954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discovery . 2022;12(1):31–46. doi: 10.1158/2159-8290.cd-21-1059. [DOI] [PubMed] [Google Scholar]
  • 10.Cree I. A. Cancer biology. Methods in Molecular Biology . 2011;731:1–11. doi: 10.1007/978-1-61779-080-5_1. [DOI] [PubMed] [Google Scholar]
  • 11.Lim K. L., Teoh H. K., Choong P. F., Teh H. X., Cheong S. K., Kamarul T. Reprogramming cancer cells: overview & current progress. Expert Opinion on Biological Therapy . 2016;16(7):941–951. doi: 10.1517/14712598.2016.1174211. [DOI] [PubMed] [Google Scholar]
  • 12.Li T., Le A. Glutamine metabolism in cancer. Advances in Experimental Medicine and Biology . 2018;1063:13–32. doi: 10.1007/978-3-319-77736-8_2. [DOI] [PubMed] [Google Scholar]
  • 13.Peng Y., Yang H., Li S. The role of glycometabolic plasticity in cancer. Pathology, Research & Practice . 2021;226 doi: 10.1016/j.prp.2021.153595.153595 [DOI] [PubMed] [Google Scholar]
  • 14.Zheng M., Wang W., Liu J., Zhang X., Zhang R. Lipid metabolism in cancer cells. Advances in Experimental Medicine and Biology . 2021;1316:49–69. doi: 10.1007/978-981-33-6785-2_4. [DOI] [PubMed] [Google Scholar]
  • 15.Skotland T., Kavaliauskiene S., Sandvig K. The role of lipid species in membranes and cancer-related changes. Cancer and Metastasis Reviews . 2020;39(2):343–360. doi: 10.1007/s10555-020-09872-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Nicolson G. L. Cell membrane fluid-mosaic structure and cancer metastasis. Cancer Research . 2015;75(7):1169–1176. doi: 10.1158/0008-5472.can-14-3216. [DOI] [PubMed] [Google Scholar]
  • 17.Agarwala P. K., Aneja R., Kapoor S. Lipidomic landscape in cancer: actionable insights for membrane-based therapy and diagnoses. Medicinal Research Reviews . 2022;42(2):983–1018. doi: 10.1002/med.21868. [DOI] [PubMed] [Google Scholar]
  • 18.Szlasa W., Zendran I., Zalesińska A., Tarek M., Kulbacka J. Lipid composition of the cancer cell membrane. Journal of Bioenergetics and Biomembranes . 2020;52(5):321–342. doi: 10.1007/s10863-020-09846-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zalba S., Ten Hagen T. L. Cell membrane modulation as adjuvant in cancer therapy. Cancer Treatment Reviews . 2017;52:48–57. doi: 10.1016/j.ctrv.2016.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Escribá P. V., Busquets X., Inokuchi J. i., et al. Membrane lipid therapy: modulation of the cell membrane composition and structure as a molecular base for drug discovery and new disease treatment. Progress in Lipid Research . 2015;59:38–53. doi: 10.1016/j.plipres.2015.04.003. [DOI] [PubMed] [Google Scholar]
  • 21.Kopecka J., Trouillas P., Gašparović A. Č., Gazzano E., Assaraf Y. G., Riganti C. Phospholipids and cholesterol: inducers of cancer multidrug resistance and therapeutic targets. Drug Resistance Updates . 2020;49 doi: 10.1016/j.drup.2019.100670.100670 [DOI] [PubMed] [Google Scholar]
  • 22.Butler L. M., Mah C. Y., Machiels J., et al. Lipidomic profiling of clinical prostate cancer reveals targetable alterations in membrane lipid composition. Cancer Research . 2021;81(19):4981–4993. doi: 10.1158/0008-5472.can-20-3863. [DOI] [PubMed] [Google Scholar]
  • 23.D’Angelo G., Moorthi S., Luberto C. Role and function of sphingomyelin biosynthesis in the development of cancer. Advances in Cancer Research . 2018;140:61–96. doi: 10.1016/bs.acr.2018.04.009. [DOI] [PubMed] [Google Scholar]
  • 24.Li W., Gao Y., Jin X., et al. Comprehensive analysis of N6-methylandenosine regulators and m6A-related RNAs as prognosis factors in colorectal cancer. Molecular Therapy—Nucleic Acids . 2022;27:598–610. doi: 10.1016/j.omtn.2021.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Xu Y., Peng Y., Shen M., et al. Construction and validation of angiogenesis-related prognostic risk signature to facilitate survival prediction and biomarker excavation of breast cancer patients. Journal of Oncology . 2022;2022:21. doi: 10.1155/2022/1525245.1525245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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 United States of America . 2005;102(43):15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mootha V. K., Lindgren C. M., Eriksson K. F., et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genetics . 2003;34(3):267–273. doi: 10.1038/ng1180. [DOI] [PubMed] [Google Scholar]
  • 28.Wu G., Xu Y., Zhang H., et al. A new prognostic risk model based on autophagy-related genes in kidney renal clear cell carcinoma. Bioengineered . 2021;12(1):7805–7819. doi: 10.1080/21655979.2021.1976050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Xu Y., Shu D., Shen M., et al. Development and validation of a novel PPAR signaling pathway-related predictive model to predict prognosis in breast cancer. Journal of Immunology Research . 2022;2022:22. doi: 10.1155/2022/9412119.9412119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Liu C. J., Hu F. F., Xia M. X., Han L., Zhang Q., Guo A. Y. GSCALite: a web server for gene set cancer analysis. Bioinformatics . 2018;34(21):3771–3772. doi: 10.1093/bioinformatics/bty411. [DOI] [PubMed] [Google Scholar]
  • 31.Yang W., Soares J., Greninger P., et al. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Research . 2012;41:D955–D961. doi: 10.1093/nar/gks1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Iorio F., Knijnenburg T. A., Vis D. J., et al. A landscape of pharmacogenomic interactions in cancer. Cell . 2016;166(3):740–754. doi: 10.1016/j.cell.2016.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Garnett M. J., Edelman E. J., Heidorn S. J., et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature . 2012;483(7391):570–575. doi: 10.1038/nature11005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rees M. G., Seashore-Ludlow B., Cheah J. H., et al. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nature Chemical Biology . 2016;12(2):109–116. doi: 10.1038/nchembio.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Seashore-Ludlow B., Rees M. G., Cheah J. H., et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discovery . 2015;5(11):1210–1223. doi: 10.1158/2159-8290.cd-15-0235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Basu A., Bodycombe N. E., Cheah J. H., et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell . 2013;154(5):1151–1161. doi: 10.1016/j.cell.2013.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Uhlén M., Fagerberg L., Hallström B. M., et al. Proteomics tissue-based map of the human proteome. Science . 2015;347(6220) doi: 10.1126/science.1260419.1260419 [DOI] [PubMed] [Google Scholar]
  • 38.Thul P. J., Åkesson L., Wiking M., et al. A subcellular map of the human proteome. Science . 2017;356 doi: 10.1126/science.aal3321. [DOI] [PubMed] [Google Scholar]
  • 39.Hanahan D., Weinberg R. A. The hallmarks of cancer. Cell . 2000;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 40.Nirmaladevi R., Paital B., Jayachandran P., Padma P. R., Nirmaladevi R. Epigenetic alterations in cancer. Frontiers in Bioscience . 2020;25(6):4847–1109. doi: 10.2741/4847. [DOI] [PubMed] [Google Scholar]
  • 41.Klutstein M., Nejman D., Greenfield R., Cedar H. DNA methylation in cancer and aging. Cancer Research . 2016;76(12):3446–3450. doi: 10.1158/0008-5472.can-15-3278. [DOI] [PubMed] [Google Scholar]
  • 42.Morgan A. E., Davies T. J., Mc Auley M. T. The role of DNA methylation in ageing and cancer. Proceedings of the Nutrition Society . 2018;77(4):412–422. doi: 10.1017/s0029665118000150. [DOI] [PubMed] [Google Scholar]
  • 43.Hinshaw D. C., Shevde L. A. The tumor microenvironment innately modulates cancer progression. Cancer Research . 2019;79(18):4557–4566. doi: 10.1158/0008-5472.can-18-3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Soysal S. D., Tzankov A., Muenst S. E. Role of the tumor microenvironment in breast cancer. Pathobiology . 2015;82(3-4):142–152. doi: 10.1159/000430499. [DOI] [PubMed] [Google Scholar]
  • 45.Mao X., Xu J., Wang W., et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Molecular Cancer . 2021;20(1):p. 131. doi: 10.1186/s12943-021-01428-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tanaka A., Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Research . 2017;27(1):109–118. doi: 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Burugu S., Asleh-Aburaya K., Nielsen T. O. Immune infiltrates in the breast cancer microenvironment: detection, characterization and clinical implication. Breast Cancer . 2017;24(1):3–15. doi: 10.1007/s12282-016-0698-z. [DOI] [PubMed] [Google Scholar]
  • 48.Reimand J., Isserlin R., Voisin V., et al. Pathway enrichment analysis and visualization of omics data using g:profiler, GSEA, cytoscape and enrichmentmap. Nature Protocols . 2019;14(2):482–517. doi: 10.1038/s41596-018-0103-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lee Y. S., Dutta A. MicroRNAs in cancer. Annual Review of Pathology: Mechanisms of Disease . 2009;4(1):199–227. doi: 10.1146/annurev.pathol.4.110807.092222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hayes J., Peruzzi P. P., Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends in Molecular Medicine . 2014;20(8):460–469. doi: 10.1016/j.molmed.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • 51.Bray F., Laversanne M., Weiderpass E., Soerjomataram I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer . 2021;127(16):3029–3030. doi: 10.1002/cncr.33587. [DOI] [PubMed] [Google Scholar]
  • 52.Xia C., Dong X., Li H., et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chinese Medical Journal . 2022;135(5):584–590. doi: 10.1097/cm9.0000000000002108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hulvat M. C. Cancer incidence and trends. Surgical Clinics of North America . 2020;100(3):469–481. doi: 10.1016/j.suc.2020.01.002. [DOI] [PubMed] [Google Scholar]
  • 54.Sung H., Ferlay J., Siegel R. L., et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians . 2021;71(3):209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
  • 55.Zhang X. X., Ni B., Li Q., et al. GPAA1 promotes gastric cancer progression via upregulation of GPI-anchored protein and enhancement of ERBB signalling pathway. Journal of Experimental & Clinical Cancer Research . 2019;38(1):p. 214. doi: 10.1186/s13046-019-1218-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ho J. C., Cheung S. T., Patil M., Chen X., Fan S. T. Increased expression of glycosyl-phosphatidylinositol anchor attachment protein 1 (GPAA1) is associated with gene amplification in hepatocellular carcinoma. International Journal of Cancer . 2006;119(6):1330–1337. doi: 10.1002/ijc.22005. [DOI] [PubMed] [Google Scholar]
  • 57.Wu G., Guo Z., Chatterjee A., et al. Overexpression of glycosylphosphatidylinositol (GPI) transamidase subunits phosphatidylinositol glycan class T and/or GPI anchor attachment 1 induces tumorigenesis and contributes to invasion in human breast cancer. Cancer Research . 2006;66(20):9829–9836. doi: 10.1158/0008-5472.can-06-0506. [DOI] [PubMed] [Google Scholar]
  • 58.Chen G., Li S. Y., Cai H. Y., Zuo F. Y. Enhanced expression and significance of glycosylphosphatidylinositol anchor attachment protein 1 in colorectal cancer. Genetics and Molecular Research . 2014;13(1):499–507. doi: 10.4238/2014.january.21.19. [DOI] [PubMed] [Google Scholar]
  • 59.Song Y., Tang C., Yin C. Combination antitumor immunotherapy with VEGF and PIGF siRNA via systemic delivery of multi-functionalized nanoparticles to tumor-associated macrophages and breast cancer cells. Biomaterials . 2018;185:117–132. doi: 10.1016/j.biomaterials.2018.09.017. [DOI] [PubMed] [Google Scholar]
  • 60.Pang V., Bates D. O., Leach L. Regulation of human feto-placental endothelial barrier integrity by vascular endothelial growth factors: competitive interplay between VEGF-A(165)a, VEGF-A(165)b, PIGF and VE-cadherin. Clinical Science . 2017;131(23):2763–2775. doi: 10.1042/cs20171252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chong Y. K., Sandanaraj E., Koh L. W. H., et al. ST3GAL1-associated transcriptomic program in glioblastoma tumor growth, invasion, and prognosis. Journal of the National Cancer Institute . 2016;108(2) doi: 10.1093/jnci/djv326.djv326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yeo H. L., Fan T. C., Lin R. J., et al. Sialylation of vasorin by ST3Gal1 facilitates TGF-β1-mediated tumor angiogenesis and progression. International Journal of Cancer . 2019;144(8):1996–2007. doi: 10.1002/ijc.31891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.He J., Zhao H., Liu X., et al. Sevoflurane suppresses cell viability and invasion and promotes cell apoptosis in colon cancer by modulating exosome-mediated circ-HMGCS1 via the miR-34a-5p/SGPP1 axis. Oncology Reports . 2020;44(6):2429–2442. doi: 10.3892/or.2020.7783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bao Y., Chen Z., Guo Y., et al. Tumor suppressor microRNA-27a in colorectal carcinogenesis and progression by targeting SGPP1 and Smad2. PLoS One . 2014;9(8) doi: 10.1371/journal.pone.0105991.e105991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Huang X., Taeb S., Jahangiri S., et al. miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Research . 2013;73(23):6972–6986. doi: 10.1158/0008-5472.can-13-1657. [DOI] [PubMed] [Google Scholar]
  • 66.Zhang B., Gao S., Bao Z., Pan C., Tian Q., Tang Q. MicroRNA-656-3p inhibits colorectal cancer cell migration, invasion, and chemo-resistance by targeting sphingosine-1-phosphate phosphatase 1. Bioengineered . 2022;13(2):3810–3826. doi: 10.1080/21655979.2022.2031420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Le Stunff H., Mikami A., Giussani P., et al. Role of sphingosine-1-phosphate phosphatase 1 in epidermal growth factor-induced chemotaxis. Journal of Biological Chemistry . 2004;279(33):34290–34297. doi: 10.1074/jbc.m404907200. [DOI] [PubMed] [Google Scholar]
  • 68.Martínez-Jiménez F., Muiños F., Sentís I., et al. A compendium of mutational cancer driver genes. Nature Reviews Cancer . 2020;20(10):555–572. doi: 10.1038/s41568-020-0290-x. [DOI] [PubMed] [Google Scholar]
  • 69.Pusapati G. V., Krndija D., Armacki M., et al. Role of the second cysteine-rich domain and Pro275 in protein kinase D2 interaction with ADP-ribosylation factor 1, trans-golgi network recruitment, and protein transport. Molecular Biology of the Cell . 2010;21(6):1011–1022. doi: 10.1091/mbc.e09-09-0814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Matthews S. A., Liu P., Spitaler M., et al. Essential role for protein kinase D family kinases in the regulation of class II histone deacetylases in B lymphocytes. Molecular and Cellular Biology . 2006;26(4):1569–1577. doi: 10.1128/mcb.26.4.1569-1577.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rey O., Yuan J., Rozengurt E. Intracellular redistribution of protein kinase D2 in response to G-protein-coupled receptor agonists. Biochemical and Biophysical Research Communications . 2003;302(4):817–824. doi: 10.1016/s0006-291x(03)00269-9. [DOI] [PubMed] [Google Scholar]
  • 72.Azoitei N., Cobbaut M., Becher A., Van Lint J., Seufferlein T. Protein kinase D2: a versatile player in cancer biology. Oncogene . 2018;37(10):1263–1278. doi: 10.1038/s41388-017-0052-8. [DOI] [PubMed] [Google Scholar]
  • 73.Bernhart E., Damm S., Heffeter P., et al. Silencing of protein kinase D2 induces glioma cell senescence via p53-dependent and -independent pathways. Neuro-Oncology . 2014;16(7):933–945. doi: 10.1093/neuonc/not303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zheng K., Chen Z., Feng H., et al. Sphingomyelin synthase 2 promotes an aggressive breast cancer phenotype by disrupting the homoeostasis of ceramide and sphingomyelin. Cell Death & Disease . 2019;10(3):p. 157. doi: 10.1038/s41419-019-1303-0. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Materials

Table S1. Cluster analysis results of membrane lipid biosynthesis-related genes under the MCODE algorithm.

Data Availability Statement

The data used to support the findings of this study are available from the corresponding author upon request.


Articles from Journal of Oncology are provided here courtesy of Wiley

RESOURCES