Home A lipid metabolism-related gene model reveals the prognosis and immune microenvironment of cutaneous melanoma
Article Open Access

A lipid metabolism-related gene model reveals the prognosis and immune microenvironment of cutaneous melanoma

  • Congcong Zhang and Hao Chen EMAIL logo
Published/Copyright: August 20, 2024

Abstract

Objectives

Lipid metabolic dysregulation plays a critical role in the biological behavior of skin cutaneous melanoma (SKCM). Hence, we aimed to identify lipid metabolism-related genes (LMGs) and possible prognostic models for SKCM, perform prognostic risk assessment, and predict possible effective therapies.

Methods

SKCM patient data were downloaded from The Cancer Genome Atlas (TCGA) and used as the training set; GSE65904 was used as the validation dataset. A prognostic risk model was established by multivariate Cox regression analysis and the LASSO algorithm. The samples in training and validation sets were grouped into high- and low-risk groups, respectively, in accordance with the risk model, and risk score (RS) distribution and survival ROC curve were obtained. The ‘limma’ package in R3.6.1 Version 3.34.7 was used to filter significant differentially expressed genes (DEGs) in the training set between the high- and low-risk groups. For DEGs, functional enrichment and immune infiltration analyses were used to reveal potential disease mechanisms and responses to immunotherapy. The expression level of LMGs involved in the prognostic risk was verified by diverse methods.

Results

A predictive model comprising four LMGs, including ADH4, ALDH7A1, HADH, and HADHA, was established to predict SKCM patient survival. Functional enrichment has revealed enriched immune-associated pathways. Different immune microenvironments were identified by immune infiltration analysis. HPA immunohistochemical analysis, Real-time PCR analysis, and Western blotting revealed the upregulation of HADH and HADHA and the downregulation of ADH4 and ALDH7A1 in melanoma tissues or cell lines compared to normal skin tissues and melanocyte cells.

Conclusions

LMGs, including ADH4, ALDH7A1, HADH, and HADHA involved in the predictive model may play a critical role in the biological behaviors and therapeutic response of melanoma. The model we constructed may serve as a prospective biological marker to predict the prognosis and therapeutic response of melanoma patients.

Introduction

The devastating malignant tumor skin cutaneous melanoma (SKCM) is responsible for a large proportion of skin cancer-related deaths [1]. Approximately one-third of patients with advanced melanoma have already developed distant metastasis at diagnosis [2]. Each year, over 232,100 people develop SKCM worldwide, and 55,500 patients die [3]. Recently, immune checkpoint inhibitors and molecular targeted drugs have been shown to effectively treat unresectable or metastatic melanoma, and the 5-year survival rate has significantly improved [4]. However, only a minimal number of patients see benefits from these treatments owing to the small applicable population, the side effects, and drug resistance [4]. Tumor mutational burden, major histocompatibility complex, weakened tumor-infiltrating lymphocyte function, and T-cell exhaustion are major factors influencing the efficacy of immune checkpoint inhibitors [5]. Therefore, constructing a risk stratification model for prognosis prediction and developing personalized targeted or immune therapy is critical.

Lipids are important to cell membrane components and function as energy sources and signaling molecules [6]. Increasing evidence has shown that lipid metabolism disorders are connected with tumorigenesis, tumor progression, and treatment [6]. Liu et al. indicated that lipids in breast cancer tissue are relevant to the function and polarity of macrophages, tumor progression, and patient survival [7]. In addition, increased expression of enzymes involved in lipid synthesis is significantly linked to unfavorable outcomes in lung cancer patients [8]. In melanoma, studies have shown that lipid metabolic dysregulation can promote cell growth and metastasis, and targeting lipid metabolism could be an applicable treatment for melanoma [9]. Furthermore, lipid metabolism – related gene (LMG) prognostic signatures have been developed using public datasets for lung adenocarcinoma, breast cancer, and osteosarcoma [10], [11], [12]. The LMG risk score can be used to determine the effect of immunization therapy on lung cancer [13]. However, the association between LMGs and the prognosis of cutaneous melanoma patients remains elusive.

In this study, in order to identify possible prognostic models and predict possible effective therapies for cutaneous melanoma, we first determined the relative expression levels of LMGs by analyzing SKCM patient sample data. In addition, we analyzed specific LMGs and constructed a prognostic model. Enriched pathway, TME immune infiltration, immunotherapy response, and drug sensitivity characteristics were also investigated.

Materials and methods

Major strategies of this study

A workflow demonstrating the strategies employed in this study is shown in Figure 1.

Figure 1: 
The workflow diagram utilized in the present research.
Figure 1:

The workflow diagram utilized in the present research.

Data gathering and preprocessing

Human SKCM tumors gene expression data (n=471) were extracted from The Cancer Genome Atlas (TCGA) database. Normal tissue sample data (n=812) were obtained from the GTEx repository (http://commonfund.nih.gov/GTEx/). The above data were merged with the shared genes expression levels. The combined expression spectrum was normalized by the normalize between arrays algorithm function in the ‘limma’ package of R3.6.1 Version 3.34.7 (https://bioconductor.org/packages/release/bioc/html/limma.html) [13]. The above data were used as the training set in the present research.

Additionally, we downloaded the GSE65904 dataset [14] from the NCBI GEO (http://www.ncbi.nlm.nih.gov/geo/) database [15], [16], [17]. The above information was incorporated into the GPL10558 platform (Illumina HumanHT-12 V4.0 expression beadchip). This dataset contained information on 214 SKCM samples and the survival data of 210 patients. This dataset served as the validation dataset in this analysis.

Determination of differentially expressed genes related to lipid metabolism

LMGs were extracted from MSigDB (http://www.gsea-msigdb.org/gsea/msigdb/cards/KEGG_FATTY_ACID_METABOLISM.html) in the Gene Set Enrichment Analysis (GSEA) database [18] (http://software.broadinstitute.org/gsea/downloads.jsp). Standardized TCGA-GTEX expression data were used to determine the relevant genes expression levels in the samples.

Differentially expressed genes (DEGs) connected with lipid metabolism between SKCM and normal samples were determined using the ‘limma’ package of R3.6.1 Version 3.34.7 [14], ensuring the false discovery rate (FDR) lower than 0.05. A heatmap was used to display significant DEGs.

Generation of genes related to overall survival and lipid metabolism

Version 2.41-1 of the ‘survival’ package in R3.6.1 language [19] (http://bioconductor.org/packages/survivalr/) was adopted to filtrate the DEGs related to lipid metabolism in TCGA SKCM samples with clinical prognostic source. Then, survival-related LMGs with p <0.05 were generated by univariate Cox hazards regression analysis.

Establishment and validation of risk model

The LASSO algorithm in the R3.6.1 language ‘lars’ package version 1.2 [20] (https://cran.r-project.org/web/packages/lars/index.html) was applied to screen the optimal combinations of LMGs among those significantly associated prognosis and perform survival regression analysis. Then, the risk score (RS) model was established using the LASSO prognostic coefficient of each component in the optimized LMG composition and target gene expression level, and the RS calculation equation was presented below:

RS = Coef genes × Exp genes

(Coefgenes represents the prognostic coefficient of the target gene under LASSO analysis, and Expgenes represents the corresponding gene expression level.)

RS values were calculated for the TCGA training set and the GSE65904 validation dataset, and the samples were divided into high (RS score≥RS median value) and low (RS score<RS median value) risk sample groups. The Kaplan‒Meier curve method in the ‘survival’ survival package in R 3.6.1 version 2.41-1 [19] was used to assess the interrelation between the actual survival prognosis and the grouping conditions.

The clinical factors of samples in the TCGA training dataset in different risk groups were analyzed according to the high- and low-risk sample groups. Furthermore, the clinical information was compared utilizing the accurate intergroup Fisher test in R3.6.1.

Determination of differentially expressed genes related to risk groups and functional analyses

The TCGA training set samples were grouped into high- and low-risk groups in accordance with the previously determined RSs. Significant DEGs between the two groups were filtered using the ‘limma’ package in R3.6.1 Version 3.34.7, and the standards were FDR <0.05 and |log2FC| >0.5. The significant DEGs were then analyzed by Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses to evaluate the genes with FDR values <0.05 using the Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/) [20, 21].

Immune analysis

CIBERSORT (https://cibersort.stanford.edu/index.php) [22] was employed to evaluate diverse immune cell ratios in TCGA SKCM samples, and the comparison of immune cell distributions between the two risk groups was conducted. In addition, the correlation between the RS characteristic values according to the RS model and immune cells was carried out using the COR function in R3.6.1. The TIDE (Tumor Immune Dysfunction and Exclusion) database (http://tide.dfci.harvard.edu/) [23] was utilized to predict the response of each sample to immunotherapy, denoted by the TIDE score, and the TIDE score of each patient was determined. Then, a statistical analysis was conducted using a t-test to assess the variance in TIDE scores between the RS groups. Furthermore, the ‘estimate’ package in R3.6.1 (http://127.0.0.1:29606/library/estimate/html/estimateScore.html) [24] facilitated the calculation the stromal, immune, and ESTIMATE scores and tumor purity. Finally, the Kruskal–Wallis test in R3.6.1 was utilized to contrast the risk group estimated scores.

Analysis of drug sensitivity

Genomics of Drug Sensitivity in Cancer (GDSC) database (https://www.cancerrxgene.org/) [25] data were combined with patient data, and ‘pRRophetic’ in R3.6.1 [26] (http://127.0.0.1:22402/doc/html/Search?objects=1&port=22402) was utilized to assess each patient’s susceptibility to diverse chemical medicines. IC50 values, representing drug susceptibility, were compared between the risk groups for diverse drugs using the Kruskal–Wallis test.

HPA immunohistochemical analysis

The Human Protein Atlas (HPA) database (https://www.proteinatlas.org/) [27, 28] was utilized to obtain immunohistochemical information on the proteins encoded by the screened genes in melanoma and normal tissue.

Cell lines and culture

Primary normal human epidermal melanocytes were separated from human prepuce tissues (human tissues were obtained from the patients of Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College) refer to our laboratory protocols. This study was approved by the ethics committee of Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College (2023017). Specifically, fresh foreskin tissue was soaked in iodophor for 5 min and washed with phosphorate buffered solution (PBS, 0.01 mol/L) 3 times. The subcutaneous tissue was separated, and the remaining tissue was cut into strips and added to a centrifuge tube containing dermal–epidermal separation solution (catalog number: 4942078001; Roche, Switzerland) at 4 °C overnight. The epidermis was isolated from the dermis with sterile ophthalmic tweezers, and the epidermis tissue was collected and cut into pieces. 0.25 % Trypsin-EDTA (catalog number: 25200056; Gibco, Canada) was applied to digest the tissues at 37 °C for 5 min. Complete medium (catalog number: 2201; Sciencell, United States) added with 10 % fetal bovine serum was added to neutralize the trypsin. Then, 100 µm and 70 µm filters were used to remove the excess ingredients, and the cell suspension was centrifuged (Centrifuge model: KDC-40; Zhongke Zhongjia company, Anhui, China) at 250×g at room temperature for 4 min. Finally, the cell precipitate was collected and resuspended in a melanocyte medium. The M14, A375, A2058, and SK-MEL-28 melanoma cell lines were restored at Hospital for skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College (the M14, A375 and SK-MEL-28 cell lines were obtained from human skin tissues, and A2058 was obtained from human lymph node, all the four cell lines have correct STR identification and have no mycoplasma contamination). All melanoma cells were cultivated in Dulbecco’s Modified Eagle Medium (DMEM) (catalog number: C11995500BT; Gibco, China) added with 110 mg/L L-glutamine and 4,500 mg/L D-glucose combined with 1 % penicillin and streptomycin and 10 % fetal bovine serum. The above cells were incubated in cell incubators.

Real-time PCR analysis

RNAiso Plus (catalog number: 9108; Takara, Dalian, China) was applied to isolate total RNA according to its specification from primary human normal melanocytes (MCs) and melanoma cell lines (M14, SK-MEL-28, A2058, and A375) at passages 5–10. HiScript II Q RT SuperMix (catalog number: R22301; Vazyme Biotech, Nanjing, China) was utilized to synthesize cDNA from these RNAs. The obtained cDNA was subsequently served as a template for PCR magnification on a LightCycler® 480 Instrument II (Roche, Basel, Switzerland) using ChamQ SYBR qPCR Master Mix (catalog number: Q33102; Vazyme Biotech, Nanjing, China). The 2−ΔΔCt method was utilized for the calculation of gene expression levels which were then uniformization against GAPDH. The primers used are enumerated in Supplementary Table 1.

Western blotting

Total protein from MC, M14, A375, A2058, and SK-MEL-28 cells was obtained using RIPA lysis buffer. After centrifugation, the supernatant was measured with a BCA kit (catalog number: KGB2101; Keygen Biotech, Nanjing, China) for soluble protein content. In accordance with the protein concentration, the final protein solution was calculated by the principle of equal mass and equal volume, and RIPA lysate was used to supplement the lack of volume. Then 1/4 volume of 5× protein loading buffer (catalog number: P0285; Beyotime, Shanghai, China) was added to each specimen. All the final mixtures were bathed in boiling water for 6 min and centrifuged at low speed for further experiments. Protein mixtures were separated by SDS‒PAGE (catalog number: SLE015; Smart-lifesciences, Changzhou, China) and transferred to a PVDF membrane. Then, the membranes were blocked in EveryBlot Blocking Buffer (catalog number: 12010020; Bio-Rad, USA) for 5 min at room temperature and then incubated with primary antibodies (dilution ratio, 1:1,000; Proteintech, Wuhan, China; antibody codes: ADH4 16474-1-AP, ALDH7A1 10368-1-AP, HADH 19828-1-AP, and HADHA 10758-1-AP.) for 15 h at 4 °C. After being washed in TBST for 30 min, these membranes were immersed in the corresponding secondary diluting antibodies (HRP Goat Anti-rabbit IgG, dilution ratio, 1:2,000; MedChemExpress, Shanghai, China) for an appropriate time at room temperature. A chemiluminescence kit (catalog number: P0018; Beyotime, Shanghai, China) was applied for protein detection.

Statistical analysis

Each set of experiments was conducted in triplicate at least. GraphPad Prism9 and Image J version 1.8.0 software were used to get the result of the experiment analysis and visualization. Student’s t-test was used to analyze the differences between the two groups. “*”, “**” and “***” respectively showed p <0.05, p <0.01, and p <0.001. “ns” showed no obvious difference (p>0.05).

Results

Data preprocessing and identification of differentially expressed genes

A total of 1,283 samples (812 normal and 471 cutaneous melanoma samples) and 60,370 genes were obtained. A total of 42 LMGs were obtained, all of which met the criteria of significant differential expression in the above two groups. The DEGs involved in lipid metabolism are shown in a heatmap (Figure 2A) and volcano map (Figure 2B). Univariate Cox regression analysis of the 42 genes is shown in a forest plot (Figure 2C).

Figure 2: 
Identification of differentially expressed genes and these associated with prognosis. (A) Heatmap showed the expression of LMGs in TCGA SKCM tumor and GTEX normal group. (B) DEG volcano plot; red dots represent upregulated genes; blue dots represent downregulated genes. (C) Forest plot showed the univariate Cox regression of the LMGs. The red, blue and black squares respectively represented genes associated with risk, protection and non-significant prognosis. (D) KM curve with significant prognostic significance. Blue and red curves respectively represented low and high expression levels of genes. (E) Expression levels of prognostic significantly related LMGs in TCGA SKCM tumors and GTEX normal control samples, *** indicates p<0.001.
Figure 2:

Identification of differentially expressed genes and these associated with prognosis. (A) Heatmap showed the expression of LMGs in TCGA SKCM tumor and GTEX normal group. (B) DEG volcano plot; red dots represent upregulated genes; blue dots represent downregulated genes. (C) Forest plot showed the univariate Cox regression of the LMGs. The red, blue and black squares respectively represented genes associated with risk, protection and non-significant prognosis. (D) KM curve with significant prognostic significance. Blue and red curves respectively represented low and high expression levels of genes. (E) Expression levels of prognostic significantly related LMGs in TCGA SKCM tumors and GTEX normal control samples, *** indicates p<0.001.

Identification of differentially expressed genes related to prognosis

A total of 10 DEGs with significant prognostic correlations were obtained. Based on the median gene expression level, the samples were classified into high- and low-expression groups, and the KM curve of 10 DEGs was constructed (Figure 2D). The expression levels in tumor and normal samples are displayed (Figure 2E).

The downregulation of ACAA2, ALDH7A1, CPT2, HADHA, HADHB, ADH7, and HADH expression and the upregulation of ACSL5, ACSL4, and ADH4 expression were speculated to be related to improved overall survival. In addition, the expression of genes such as ACAA2, ADH4, ADH7, and ALDH7A1 was predicted to be greater in normal tissue than in tumor tissue.

Establishment of lipid metabolism – related Gene Risk Model

The LASSO algorithm was applied to obtain the optimal gene combination for the 10 genes significantly correlated with prognosis (Figure 3). The following four optimized genes were obtained: ADH4, ALDH7A1, HADH, and HADHA. The RS computational equation was calculated according to the LASSO regression coefficients and four genes as follows:

RS = ( 0.02277991 ) * Exp ADH 4 + ( 0.03256654 ) * Exp ALDH 7 A 1 + ( 0.0513173 ) * Exp HADH + ( 0.06442325 ) Exp HADHA

Figure 3: 
Establishment of the lipid metabolism-related Gene Risk Model. (A) LASSO parameter diagram. (B) The location where the horizontal and vertical black lines cross was the location where the optimal parameters (−4.113, 0.247), and the corresponding gene was the optimal gene combination.
Figure 3:

Establishment of the lipid metabolism-related Gene Risk Model. (A) LASSO parameter diagram. (B) The location where the horizontal and vertical black lines cross was the location where the optimal parameters (−4.113, 0.247), and the corresponding gene was the optimal gene combination.

Validation of the Gene Risk Model

The RS and survival time distributions of patients in the TCGA and GSE65904 datasets are shown in Figure 4A and B (upper and middle figures). The ROC curve results are shown in Figure 4A and B (bottom figure). The KM curve of each dataset is shown in Figure 4C and D, and. These results revealed a link between the actual prognosis and the two risk groups in both datasets.

Figure 4: 
Validation of the Gene Risk Model. RS distribution (top), survival time status (middle) and ROC curves (bottom) of TCGA training (A) set and GSE65904 validation set (B). The figures in brackets showed the corresponding ROC curves and sensitivity. TCGA training set (C) and GSE65904 validation set (D) samples on the basis of the RS prediction model and prognostic KM curve. Blue and red curves respectively represented low- and high-risk samples.
Figure 4:

Validation of the Gene Risk Model. RS distribution (top), survival time status (middle) and ROC curves (bottom) of TCGA training (A) set and GSE65904 validation set (B). The figures in brackets showed the corresponding ROC curves and sensitivity. TCGA training set (C) and GSE65904 validation set (D) samples on the basis of the RS prediction model and prognostic KM curve. Blue and red curves respectively represented low- and high-risk samples.

Functional assessment

Samples in TCGA were split into high- and low-risk groups in accordance with the RS of the risk model. According to the ‘limma’ package, a total of 2,612 significant DEGs were identified between these two groups, followed by GO and KEGG analyses. A collection of functional categories relevant to immunity was highlighted in the GO terms MF (molecular function), CC (cellular component), and BP (biological process) (Figure 5). In the BP category, “immune response,” “chemokine-mediated signaling pathways,” and “T-cell activation” were the enriched terms. In the CC category, “external side of plasma membrane,” “extracellular region,” “plasma membrane,” and “receptor complex” were also enriched. In the MF category, “receptor binding,” “transmembrane signaling receptor activity,” and “cytokine activity” were the most enriched terms.

Figure 5: 
Histogram of GO and KEGG analysis. BP (A), CC (B), MF (C), and KEGG (D) signaling pathways with significant DEGs among risk groups. The vertical axis showed the name of the item, the horizontal axis showed the number of genes, and the color of the column represented significance, the red color represented the most significance and the green color represented lesser significance.
Figure 5:

Histogram of GO and KEGG analysis. BP (A), CC (B), MF (C), and KEGG (D) signaling pathways with significant DEGs among risk groups. The vertical axis showed the name of the item, the horizontal axis showed the number of genes, and the color of the column represented significance, the red color represented the most significance and the green color represented lesser significance.

Pathway maps representing reaction networks and molecular interactions were included in the KEGG analysis. The enriched immune pathways included “cytokine–cytokine receptor interaction,” “JAK-STAT signaling pathways,” “natural killer-mediated cytotoxicity,” “T-cell receptor signaling pathway,” and “NF-κB signaling pathways” (Figure 5). Hence, enrichment of immune-related cascades was acquired between the two groups (the top 10 were chosen according to the order of the FDR).

Immune infiltration analysis

CIBERSORT was used to extract the following six forms of immune cells with obvious variances in distribution between the two groups: CD4+ activated memory T cells, CD8+ T cells, M2 macrophages, activated mast cells, resting NK cells, and eosinophils (Figure 6A).

Figure 6: 
Immune infiltration analysis. (A) Immune cells distribution with apparent discrepancy in distribution among the two risk groups. (B) The relationship between the immune cells distribution and the Risk Score characteristic values based on RS model (left: horizontal distribution in different risk groups and right: scatter plot of correlation with Risk Score characteristic values.). (C) The estimated scores of different risk groups. * represents p<0.05, ** represents p<0.01, and *** represents p<0.001.
Figure 6:

Immune infiltration analysis. (A) Immune cells distribution with apparent discrepancy in distribution among the two risk groups. (B) The relationship between the immune cells distribution and the Risk Score characteristic values based on RS model (left: horizontal distribution in different risk groups and right: scatter plot of correlation with Risk Score characteristic values.). (C) The estimated scores of different risk groups. * represents p<0.05, ** represents p<0.01, and *** represents p<0.001.

In comparison with those in the high-risk group, the low-risk group exhibited a higher presence of CD8+ T cells and CD4+ activated memory T cells, but a lower abundance of resting NK cells, M2 macrophages, eosinophils, and activated mast cells. In R3.6.1, the RS characteristic values according to the RS model were negatively related to the distribution of CD4+ activated memory T cells (p=0.0001499) and CD8+ T cells (p=0.01181) but positively related to the distributions of resting NK cells (p=0.02363), M2 macrophages (p=0.007852), and activated mast cells (p=0.002235). Furthermore, the group at lower risk exhibited decreased tumor purity but elevated levels of immune, stromal, and ESTIMATE scores (Figure 6B).

Comparison of immune response and drug sensitivity

A total of 148 non-responder and 311 responder samples were predicted, revealing that the proportion of responder samples in the group at lower risk was greater than that in the group at higher risk (Figure 7A). Furthermore, patients in the group at lower risk exhibited increased sensitivity to AZD7762, camptothecin, gefitinib, gemcitabine, nilotinib, rapamycin, and vinblastine (Figure 7B).

Figure 7: 
Comparison of immune response and drug sensitivity analysis. (A) Distribution of TIDE prediction of immune response in the two risk groups. (B) Contrast of the IC50 values of various drugs between different risk groups by Kruskal–Wallis test.
Figure 7:

Comparison of immune response and drug sensitivity analysis. (A) Distribution of TIDE prediction of immune response in the two risk groups. (B) Contrast of the IC50 values of various drugs between different risk groups by Kruskal–Wallis test.

Validation of the genes by RT‒qPCR, western blotting, and HPA immunohistochemical analysis

The RT-qPCR and Western blot confirmed that ADH4 and ALDH7A1 expression were downregulated in the M14, A375, A2058, and SK-MEL-28 cell lines, but upregulated in the MC cell line, whereas HADH was increased in the M14, A2058, and SK-MEL-28 cell lines, and HADHA was increased in the SK-MEL-28 and A375 cell lines (Figure 8). These differences were significant (p<0.05). The HPA immunohistochemical results supported the downregulation of ADH4 and ALDH7A1 expression and the upregulation of HADH and HADHA expression in melanoma.

Figure 8: 
Gene expression validated by RT-qPCR, Western blot and HPA immunohistochemical analysis. (A) RT-qPCR revealed the relative mRNAs expression levels of ADH4, ALDH7A1, HADH and HADHA. Data are shown as mean±standard deviations, n=3; *p<0.05; **p<0.01; ***p<0.001; ns means p≧0.05. (B) Western blot and their quantification revealed the relative protein levels of ADH4, ALDH7A1, HADH and HADHA. (C) The protein products expression of ADH4, ALDH7A1, HADH and HADHA were obtained by HPA immunohistochemical analysis.
Figure 8:

Gene expression validated by RT-qPCR, Western blot and HPA immunohistochemical analysis. (A) RT-qPCR revealed the relative mRNAs expression levels of ADH4, ALDH7A1, HADH and HADHA. Data are shown as mean±standard deviations, n=3; *p<0.05; **p<0.01; ***p<0.001; ns means p≧0.05. (B) Western blot and their quantification revealed the relative protein levels of ADH4, ALDH7A1, HADH and HADHA. (C) The protein products expression of ADH4, ALDH7A1, HADH and HADHA were obtained by HPA immunohistochemical analysis.

Discussion

Recently, the impact of lipids on the immunotherapy of tumors has attracted increasing attention. Lipids in the TME are used as the original energy source and are critical regulators in immune and tumor cells; furthermore, lipid metabolism impacts the immune response [29]. Activated lipid metabolism enhances antigen expression in melanoma, increasing tumor sensitivity to immunotherapy [30]. Furthermore, changes in lipid metabolism in aged fibroblasts have been reported to induce cell resistance to targeted therapy in age-dependent melanoma [31]. Therefore, lipid metabolism plays a vital and complex role in TME and melanoma treatment. In this present study, 10 lipid metabolism-related DEGs associated with prognosis were identified in SKCM. A new predictive model comprising four LMGs was established and confirmed in external cohorts. Functional enrichment was used to reveal the immune-related pathways involved. Furthermore, drug sensitivity was analyzed.

The predictive RS model comprised four LMGs (ADH4, ALDH7A1, HADH, and HADHA). Among them, alcohol dehydrogenase 4 (ADH4), the pivotal member of the ADH family, participates in the metabolism of biogenic amines, steroids, and hydroxyl fatty acids [32]. In addition, ADH4 can efficiently reduce aldehydes generated from lipid peroxidation and thus protect against the toxic effects of aldehydes [33]. Noticeably, ADH4 expression is significantly reduced in hepatocellular carcinoma tissues and is an independent prognostic factor [34]. Our study findings illustrated that an increase in ADH4 expression may improve the survival rates of patients with melanoma by reducing the toxic effect of aldehydes on immune cells.

The enzyme aldehyde dehydrogenase 7 family member A1 (ALDH7A1) catalyzes the lipid peroxidation of fatty aldehydes, which is necessary for ATP synthesis and the maintenance of a high oxygen consumption rate (OCR) [35]. The function of ALDH7A1 in cancer prognosis is unclear. ALDH7A1 is highly expressed in prostate and pancreatic cancer, and patients with elevated ALDH7A1 expression in non-small cell lung cancer have lower recurrence-free survival rates, however, the opposite trend has been observed in oral cancer [36], [37], [38]. It has been noted in previous studies that ALDH7A1 expression level is decreased in nodular melanoma, normal skin, and superficial spreading melanoma, possibly due to epigenetic regulation [39]. In our study, ALDH7A1 expression was downregulated in melanoma cell lines in comparison with that in MCs, whereas higher expression in SKCM was related to a reduced overall survival time.

The gene HADH encodes the short-chain-L-3-hydroxyacyl-CoA dehydrogenase in mitochondria, which is involved in the critical process of the β-oxidation of fatty acids [40]. In acute myeloid leukemia and colon cancer, high HADH levels have been linked to unfavorable outcomes [40, 41]. Hydroxyaryl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) is the alpha subunit of mitochondrial trifunctional protein. HADHA is accountable for the catalysis of long-chain 3-hydroxyacyl-CoA dehydrogenase and enoyl-CoA hydratase activities. In addition to fatty acid β-oxidation, HADHA is involved in additional metabolic processes including lipid biosynthesis, ketone formation, and ketone hydrolysis [42]. HADHA expression, which was found to be greater in SKCM samples than in normal samples, was related to poor prognosis. HADH and HADHA are key genes involved in mitochondrial metabolism, and mitochondria play crucial roles in the differentiation, maintenance, and functional decline of CD8+ T cells [43]. Therefore, the harmful effects of HADH and HADHA on the survival rates of melanoma patients may be associated with changes in mitochondrial metabolism.

TME constituents can render tumors resistant to immunization therapy [44]. The proportion of infiltrating immunocytes has been confirmed to regulate tumor susceptibility to immunization therapy [45]. The cancer-associated fibroblast population in the TME is related to immune dysregulation and melanoma immunotherapy [46]. The components of the extracellular matrix influence the initiation, activation, and migration of effector infiltration and the function of NK cells, ultimately affecting tumor response to immunotherapy [47, 48].

One study showed that reprogramming lipid metabolism in melanoma could inhibit effector T cells during aging, thus enhancing tumor immunotherapy efficacy [49]. Ferroptosis, typified by iron-dependent lipid peroxide aggregation, is regulated by CD8+ T cells and promote the efficacy of melanoma immunotherapy [50]. However, research has demonstrated that lipid metabolism in the TME has immunosuppressive effects, thus impairing immunotherapy efficacy. Long-chain fatty acid accumulation in the TME decreases the cytotoxicity of effector T cells [51]. Tumor-infiltrating immunosuppressive CD4+ T cells, Tregs, mainly rely on fatty acids to exert immunosuppressive effects. In addition, the function of dendritic cells, including their antigen-presenting ability and antitumor effects, is impaired by high lipid content [52]. The precise role of lipid metabolism reprogramming in the tumor immune response is unclear. The immune response includes protumor and antitumor immune responses, both of which involve diverse immune cells, such as macrophages, myeloid-derived suppressor cells, natural killer (NK) cells, and T cells. T cells in the TME could play a crucial role in the antitumor immune response. CD8+ T cells directly remove tumor cells by differentiating into cytotoxic T lymphocytes (CTLs) [53]. CD4+ T cells directly bring on tumor cell destruction through IFN-γ-dependent mechanisms [54]. The numbers of activated CD4+ memory T cells and CD8+ T cells were increased in the low-risk group. Furthermore, the upregulation of ADH4 expression was reported to promote the infiltration of auxiliary T cells and mast cell neutrophils in tumors [55]. The levels of immune effectors, including T cells, CD8+ T cells, and Tregs, exhibit a negative correlation with ALDH7A1 mRNA levels in multiple cancers, including melanoma [56]. In the TME, HADH and HADHA are key genes involved in mitochondrial metabolism, which is necessary for CD8+ T-cell regulation [43]. Therefore, these differences in immune cells may explain why patients in the low-risk group had a better prognosis and a better immune response than those in the high-risk group. In addition, we found a close association between immune function and gene signatures by analyzing the functions of genes and pathways enriched between the two risk groups. In particular, drug sensitivity analysis may promote the development of personalized medicine. In conclusion, our work provides further evidence of the relevance of tumor lipid metabolism to the tumor immune landscape.

Our work provides the a new predictive model of four LMGs in cutaneous melanoma, and we also investigated the features of enriched pathways, TME immune infiltration, immunotherapy response, and drug sensitivity. Additionally, we validated the mRNA and protein levels of the four LMGs in various ways to demonstrate the reliability of the predictive model. Finally, we drew a conclusion that the four LMGs including ADH4, ALDH7A1, HADH, and HADHA may play a vital role in the biological behaviors and therapeutic response of melanoma, and the genes in the model could potentially guide the prediction of prognosis and combination therapy involving immune. However, the present study has several limitations. First, this was a retrospective study using a database with inaccuracies. Second, the underlying mechanisms of LMGs in melanoma and their correlation with tumor immune status and prognosis still need to be verified in further investigations.


Corresponding author: Dr. Hao Chen, Department of Pathology, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 12# Jiangwangmiao Road, Nanjing 210042, China, E-mail:

Funding source: National Key R&D Program of China

Award Identifier / Grant number: 2022YFC3601800

Funding source: CAMS Innovation Fund for Medical Sciences and Clinical Translational Project

Award Identifier / Grant number: 2023‐I2M‐C&T‐B‐110

Acknowledgments

We thank LetPub (www.letpub.com.cn) for its linguistic assistance during the preparation of this manuscript.

  1. Research ethics: This study was approved by the ethics committee of Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College (2023017).

  2. Informed consent: Written informed consent has been obtained from the patients involved in this study.

  3. Author contributions: All authors contributed to the study conception and design. Material preparation and data collection were performed by Congcong Zhang and Hao Chen. Data analysis was performed by Congcong Zhang. The first draft of the manuscript was written by Congcong Zhang and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

  4. Competing interests: There are no conflicts of interest.

  5. Research funding: CAMS Innovation Fund for Medical Sciences and Clinical Translational Project (2023‐I2M‐C&T‐B‐110); National Key R&D Program of China (2022YFC3601800).

  6. Data availability: The article/supplementary materials include the original contributions proposed in the study. For further inquiries, please contact the corresponding author directly.

References

1. Garbe, C, Amaral, T, Peris, K, Hauschild, A, Arenberger, P, Basset-Seguin, N, et al.. European consensus-based interdisciplinary guideline for melanoma. Part 1: diagnostics: update 2022. Eur J Cancer 2022;170:236–55. https://doi.org/10.1016/j.ejca.2022.03.008.Search in Google Scholar PubMed

2. Luke, JJ, Flaherty, KT, Ribas, A, Long, GV. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol 2017;14:463–82. https://doi.org/10.1038/nrclinonc.2017.43.Search in Google Scholar PubMed

3. Schadendorf, D, van Akkooi, A, Berking, C, Griewank, K, Gutzmer, R, Hauschild, A, et al.. Melanoma. Lancet 2018;392:971–84. https://doi.org/10.1016/s0140-6736(18)31559-9.Search in Google Scholar PubMed

4. Thornton, J, Chhabra, G, Singh, CK, Guzmán-Pérez, G, Shirley, CA, Ahmad, N. Mechanisms of immunotherapy resistance in cutaneous melanoma: recognizing a shapeshifter. Front Oncol 2022;12:880876. https://doi.org/10.3389/fonc.2022.880876.Search in Google Scholar PubMed PubMed Central

5. Caraban, BM, Aschie, M, Deacu, M, Cozaru, GC, Pundiche, MB, Orasanu, CI, et al.. A narrative review of current knowledge on cutaneous melanoma. Clin Pract 2024;14:214–41. https://doi.org/10.3390/clinpract14010018.Search in Google Scholar PubMed PubMed Central

6. Bian, X, Liu, R, Meng, Y, Xing, D, Xu, D, Lu, Z, et al.. Lipid metabolism and cancer. J Exp Med 2021;218:e20201606. https://doi.org/10.1084/jem.20201606.Search in Google Scholar PubMed PubMed Central

7. Liu, Z, Gao, Z, Li, B, Li, J, Ou, Y, Yu, X, et al.. Lipid-associated macrophages in the tumor-adipose microenvironment facilitate breast cancer progression. OncoImmunology 2022;11:2085432. https://doi.org/10.1080/2162402x.2022.2085432.Search in Google Scholar

8. Eltayeb, K, La, MS, Tiseo, M, Alfieri, R, Fumarola, C. Reprogramming of lipid metabolism in lung cancer: an overview with focus on EGFR-mutated non-small cell lung cancer. Cells 2022;11:413. https://doi.org/10.3390/cells11030413.Search in Google Scholar PubMed PubMed Central

9. Pellerin, L, Carrié, L, Dufau, C, Nieto, L, Ségui, B, Levade, T, et al.. Lipid metabolic reprogramming: role in melanoma progression and therapeutic perspectives. Cancers 2020;12:3147. https://doi.org/10.3390/cancers12113147.Search in Google Scholar PubMed PubMed Central

10. Jiang, A, Chen, X, Zheng, H, Liu, N, Ding, Q, Li, Y, et al.. Lipid metabolism-related gene prognostic index (LMRGPI) reveals distinct prognosis and treatment patterns for patients with early-stage pulmonary adenocarcinoma. Int J Med Sci 2022;19:711–28. https://doi.org/10.7150/ijms.71267.Search in Google Scholar PubMed PubMed Central

11. Gong, M, Liu, X, Yang, W, Song, H, Zhao, X, Ai, X, et al.. Identification of a lipid metabolism-associated gene signature predicting survival in breast cancer. Int J Gen Med 2021;14:9503–13. https://doi.org/10.2147/ijgm.s343426.Search in Google Scholar

12. Qian, H, Lei, T, Hu, Y, Lei, P. Expression of lipid-metabolism genes is correlated with immune microenvironment and predicts prognosis in osteosarcoma. Front Cell Dev Biol 2021;9:673827. https://doi.org/10.3389/fcell.2021.673827.Search in Google Scholar PubMed PubMed Central

13. Wu, XN, Su, D, Mei, YD, Xu, MQ, Zhang, H, Wang, ZY, et al.. Identified lung adenocarcinoma metabolic phenotypes and their association with tumor immune microenvironment. Cancer Immunol Immunother 2021;70:2835–50. https://doi.org/10.1007/s00262-021-02896-6.Search in Google Scholar PubMed PubMed Central

14. Ritchie, ME, Phipson, B, Wu, D, Hu, Y, Law, CW, Shi, W, et al.. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43:e47. https://doi.org/10.1093/nar/gkv007.Search in Google Scholar PubMed PubMed Central

15. Clough, E, Barrett, T. The gene expression omnibus database. Methods Mol Biol 2016;1418:93–110. https://doi.org/10.1007/978-1-4939-3578-9_5.Search in Google Scholar PubMed PubMed Central

16. Cirenajwis, H, Ekedahl, H, Lauss, M, Harbst, K, Carneiro, A, Enoksson, J, et al.. Molecular stratification of metastatic melanoma using gene expression profiling: prediction of survival outcome and benefit from molecular targeted therapy. Oncotarget 2015;6:12297–309. https://doi.org/10.18632/oncotarget.3655.Search in Google Scholar PubMed PubMed Central

17. Cabrita, R, Lauss, M, Sanna, A, Liu, B, He, S, Tu, C, et al.. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020;577:561–5. https://doi.org/10.1038/s41586-019-1914-8.Search in Google Scholar PubMed

18. Liberzon, A, Birger, C, Thorvaldsdóttir, H, Ghandi, M, Mesirov, JP, Tamayo, P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25. https://doi.org/10.1016/j.cels.2015.12.004.Search in Google Scholar PubMed PubMed Central

19. Wang, P, Wang, Y, Hang, B, Zou, X, Mao, JH. A novel gene expression-based prognostic scoring system to predict survival in gastric cancer. Oncotarget 2016;7:55343–51. https://doi.org/10.18632/oncotarget.10533.Search in Google Scholar PubMed PubMed Central

20. Goeman, JJ. L1 penalized estimation in the Cox proportional hazards model. Biom J 2010;52:70–84. https://doi.org/10.1002/bimj.200900028.Search in Google Scholar PubMed

21. Huang, DW, Sherman, BT, Lempicki, RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 2009;37:1–13. https://doi.org/10.1093/nar/gkn923.Search in Google Scholar PubMed PubMed Central

22. Chen, B, Khodadoust, MS, Liu, CL, Newman, AM, Alizadeh, AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol 2018;1711:243–59. https://doi.org/10.1007/978-1-4939-7493-1_12.Search in Google Scholar PubMed PubMed Central

23. Ding, C, Shan, Z, Li, M, Chen, H, Li, X, Jin, Z. Characterization of the fatty acid metabolism in colorectal cancer to guide clinical therapy. Mol Ther Oncolytics 2021;20:532–44. https://doi.org/10.1016/j.omto.2021.02.010.Search in Google Scholar PubMed PubMed Central

24. Hu, D, Zhou, M, Zhu, X. Deciphering immune-associated genes to predict survival in clear cell renal cell cancer. BioMed Res Int 2019;2019:2506843. https://doi.org/10.1155/2019/2506843.Search in Google Scholar PubMed PubMed Central

25. Yang, W, Soares, J, Greninger, P, Edelman, EJ, Lightfoot, H, Forbes, S, et al.. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res 2013;41:D955–61. https://doi.org/10.1093/nar/gks1111.Search in Google Scholar PubMed PubMed Central

26. Zhu, Y, Meng, X, Ruan, X, Lu, X, Yan, F, Wang, F. Characterization of neoantigen load subgroups in gynecologic and breast cancers. Front Bioeng Biotechnol 2020;8:702. https://doi.org/10.3389/fbioe.2020.00702.Search in Google Scholar PubMed PubMed Central

27. Pontén, F, Jirström, K, Uhlen, M. The Human Protein Atlas – a tool for pathology. J Pathol 2008;216:387–93. https://doi.org/10.1002/path.2440.Search in Google Scholar PubMed

28. Miura, K, Ishida, K, Fujibuchi, W, Ito, A, Niikura, H, Ogawa, H, et al.. Differentiating rectal carcinoma by an immunohistological analysis of carcinomas of pelvic organs based on the NCBI literature survey and the Human Protein Atlas database. Surg Today 2012;42:515–25. https://doi.org/10.1007/s00595-012-0167-z.Search in Google Scholar PubMed

29. Yu, W, Lei, Q, Yang, L, Qin, G, Liu, S, Wang, D, et al.. Contradictory roles of lipid metabolism in immune response within the tumor microenvironment. J Hematol Oncol 2021;14:187. https://doi.org/10.1186/s13045-021-01200-4.Search in Google Scholar PubMed PubMed Central

30. Harel, M, Ortenberg, R, Varanasi, SK, Mangalhara, KC, Mardamshina, M, Markovits, E, et al.. Proteomics of melanoma response to immunotherapy reveals mitochondrial dependence. Cell 2019;179:236–50. https://doi.org/10.1016/j.cell.2019.08.012.Search in Google Scholar PubMed PubMed Central

31. Alicea, GM, Rebecca, VW, Goldman, AR, Fane, ME, Douglass, SM, Behera, R, et al.. Changes in aged fibroblast lipid metabolism induce age-dependent melanoma cell resistance to targeted therapy via the fatty acid transporter FATP2. Cancer Discov 2020;10:1282–95. https://doi.org/10.1158/2159-8290.cd-20-0329.Search in Google Scholar

32. Höög, JO, Strömberg, P, Hedberg, JJ, Griffiths, WJ. The mammalian alcohol dehydrogenases interact in several metabolic pathways. Chem Biol Interact 2003;143–144:175–81. https://doi.org/10.1016/s0009-2797(02)00225-9.Search in Google Scholar PubMed

33. Belin, AC, Westerlund, M, Anvret, A, Lindqvist, E, Pernold, K, Ogren, SO, et al.. Modeling Parkinson’s disease genetics: altered function of the dopamine system in Adh4 knockout mice. Behav Brain Res 2011;217:439–45. https://doi.org/10.1016/j.bbr.2010.11.023.Search in Google Scholar PubMed

34. Wei, RR, Zhang, MY, Rao, HL, Pu, HY, Zhang, HZ, Wang, HY. Identification of ADH4 as a novel and potential prognostic marker in hepatocellular carcinoma. Med Oncol 2012;29:2737–43. https://doi.org/10.1007/s12032-011-0126-3.Search in Google Scholar PubMed

35. Lee, JS, Lee, H, Woo, SM, Jang, H, Jeon, Y, Kim, HY, et al.. Aldh7a1 overall survival of pancreatic ductal adenocarcinoma is doubled by deletion in the KPC mouse. Theranostics 2021;11:3472–88. https://doi.org/10.7150/thno.53935.Search in Google Scholar PubMed PubMed Central

36. van den Hoogen, C, van der Horst, G, Cheung, H, Buijs, JT, Pelger, RC, van der Pluijm, G. The aldehyde dehydrogenase enzyme 7A1 is functionally involved in prostate cancer bone metastasis. Clin Exp Metastasis 2011;28:615–25. https://doi.org/10.1007/s10585-011-9395-7.Search in Google Scholar PubMed PubMed Central

37. Tan, M, Meng, J, Sun, X, Fu, X, Wang, R. EPS8 supports pancreatic cancer growth by inhibiting BMI1 mediated proteasomal degradation of ALDH7A1. Exp Cell Res 2021;407:112782. https://doi.org/10.1016/j.yexcr.2021.112782.Search in Google Scholar PubMed

38. Lu, HJ, Chuang, CY, Chen, MK, Su, CW, Yang, WE, Yeh, CM, et al.. ALDH7A1The impact of variants in oral cancer development and prognosis. Aging 2022;14:4556–71. https://doi.org/10.18632/aging.204099.Search in Google Scholar PubMed PubMed Central

39. Rose, AE, Poliseno, L, Wang, J, Clark, M, Pearlman, A, Wang, G, et al.. Integrative genomics identifies molecular alterations that challenge the linear model of melanoma progression. Cancer Res 2011;71:2561–71. https://doi.org/10.1158/0008-5472.can-10-2958.Search in Google Scholar

40. Wei, J, Xie, Q, Liu, X, Wan, C, Wu, W, Fang, K, et al.. Identification the prognostic value of glutathione peroxidases expression levels in acute myeloid leukemia. Ann Transl Med 2020;8:678. https://doi.org/10.21037/atm-20-3296.Search in Google Scholar PubMed PubMed Central

41. Ren, J, Feng, J, Song, W, Wan, C, Wu, W, Fang, K, et al.. Development and validation of a metabolic gene signature for predicting overall survival in patients with colon cancer. Clin Exp Med 2020;20:535–44. https://doi.org/10.1007/s10238-020-00652-1.Search in Google Scholar PubMed

42. Wu, X, Feng, R, Wang, X, Guo, F, Liu, W. Roles of hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha, a lipid metabolism enzyme, in Wilms tumor patients. J Cancer Res Ther 2021;17:1281–5. https://doi.org/10.4103/jcrt.jcrt_1388_21.Search in Google Scholar PubMed

43. Sainero-Alcolado, L, Liaño-Pons, J, Ruiz-Pérez, MV, Arsenian-Henriksson, M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ 2022;29:1304–17. https://doi.org/10.1038/s41418-022-01022-y.Search in Google Scholar PubMed PubMed Central

44. Datta, M, Coussens, LM, Nishikawa, H, Hodi, FS, Jain, RK. Reprogramming the tumor microenvironment to improve immunotherapy: emerging strategies and combination therapies. Am Soc Clin Oncol Educ Book 2019;39:165–74. https://doi.org/10.1200/edbk_237987.Search in Google Scholar PubMed PubMed Central

45. Zhang, Y, Zhang, Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol 2020;17:807–21. https://doi.org/10.1038/s41423-020-0488-6.Search in Google Scholar PubMed PubMed Central

46. Wong, PF, Wei, W, Gupta, S, Smithy, JW, Zelterman, D, Kluger, HM, et al.. Multiplex quantitative analysis of cancer-associated fibroblasts and immunotherapy outcome in metastatic melanoma. J Immunother Cancer 2019;7:194. https://doi.org/10.1186/s40425-019-0675-0.Search in Google Scholar PubMed PubMed Central

47. Rossi, GR, Trindade, ES, Souza-Fonseca-Guimaraes, F. Tumor microenvironment-associated extracellular matrix components regulate NK cell function. Front Immunol 2020;11:73. https://doi.org/10.3389/fimmu.2020.00073.Search in Google Scholar PubMed PubMed Central

48. He, Y, Liu, T, Dai, S, Xu, Z, Wang, L, Luo, F. Tumor-associated extracellular matrix: how to be a potential aide to anti-tumor immunotherapy? Front Cell Dev Biol 2021;9:739161. https://doi.org/10.3389/fcell.2021.739161.Search in Google Scholar PubMed PubMed Central

49. Liu, X, Hartman, CL, Li, L, Albert, CJ, Si, F, Gao, A, et al.. Reprogramming lipid metabolism prevents effector T cell senescence and enhances tumor immunotherapy. Sci Transl Med 2021;13:eaaz6314. https://doi.org/10.1126/scitranslmed.aaz6314.Search in Google Scholar PubMed

50. Wang, W, Green, M, Choi, JE, Gijón, M, Kennedy, PD, Johnson, JK, et al.. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019;569:270–4. https://doi.org/10.1038/s41586-019-1170-y.Search in Google Scholar PubMed PubMed Central

51. Manzo, T, Prentice, BM, Anderson, KG, Raman, A, Schalck, A, Codreanu, GS, et al.. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med 2020;217:e20191920. https://doi.org/10.1084/jem.20191920.Search in Google Scholar PubMed PubMed Central

52. Gao, F, Liu, C, Guo, J, Sun, W, Xian, L, Bai, D, et al.. Radiation-driven lipid accumulation and dendritic cell dysfunction in cancer. Sci Rep 2015;5:9613. https://doi.org/10.1038/srep09613.Search in Google Scholar PubMed PubMed Central

53. Jiang, X, Xu, J, Liu, M, Xing, H, Wang, Z, Huang, L, et al.. Adoptive CD8 T cell therapy against cancer: challenges and opportunities. Cancer Lett 2019;462:23–32. https://doi.org/10.1016/j.canlet.2019.07.017.Search in Google Scholar PubMed

54. Togashi, Y, Shitara, K, Nishikawa, H. Regulatory T cells in cancer immunosuppression-implications for anticancer therapy. Nat Rev Clin Oncol 2019;16:356–71. https://doi.org/10.1038/s41571-019-0175-7.Search in Google Scholar PubMed

55. Xu, W, Ding, H, Zhang, M, Liu, L, Yin, M, Weng, Z, et al.. The prognostic role of fatty acid metabolism-related genes in patients with gastric cancer. Transl Cancer Res 2022;11:3593–609. https://doi.org/10.21037/tcr-22-761.Search in Google Scholar PubMed PubMed Central

56. Stoll, G, Kremer, M, Bloy, N, Joseph, A, Castedo, M, Meurice, G, et al.. Metabolic enzymes expressed by cancer cells impact the immune infiltrate. Oncoimmunology 2019;8:e1571389. https://doi.org/10.1080/2162402X.2019.1571389.Search in Google Scholar PubMed PubMed Central


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/oncologie-2024-0202).


Received: 2024-04-24
Accepted: 2024-07-23
Published Online: 2024-08-20

© 2024 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Review Articles
  3. Autophagy and radiotherapy in esophageal cancer: modulating treatment sensitivity and overcoming challenges
  4. Functions of CAFs in microenvironment of non-small cell lung cancer: based on updated hallmarks of cancer
  5. Cisplatin-induced pyroptosis: a double-edged sword in cancer treatment
  6. Radiotherapy directed to inferior vena cava tumor thrombus among patients with renal cell carcinoma: an illustrative case and review of the literature
  7. Research Articles
  8. A lipid metabolism-related gene model reveals the prognosis and immune microenvironment of cutaneous melanoma
  9. Boanmycin induces apoptosis and overcomes venetoclax resistance in acute myeloid leukemia
  10. Identification of GNB1 as a downstream effector of the circRNA-0133711/miR-145-5p axis involved in breast cancer proliferation and metastasis
  11. Clinical characteristics and prognosis of lung metastases from unknown primary cancer sites
  12. Integrating bulk-RNA and single-cell analysis reveals heterogeneous expression of cuproptosis-related sorafenib-resistant genes in hepatocellular carcinoma
  13. Exploring the mechanism of genistein in treating hepatocellular carcinoma through network pharmacology and molecular docking
  14. Investigating the potential mechanisms of Litsea cubeba essential oil for anti-melanoma through experimental validation, network pharmacology, and molecular docking analysis
  15. Exploring the anti-lung cancer mechanism of Ganoderma lucidum and its relationship with the level of immune cell infiltration based on network pharmacology and molecular docking
  16. Constructing a prognostic model for colon cancer patients on basis of coagulation genes enriched in cancer-associated fibroblasts to guide personalized immunotherapy
  17. Rapid Communication
  18. Evaluation of HER2 immunohistochemistry-positive and immunohistochemistry-negative FISH amplification breast cancers using next-generation sequencing
  19. Short Commentary
  20. Unveiling the unexplored: shedding light on a novel aspect of colorectal carcinoma
Downloaded on 12.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2024-0202/html
Scroll to top button