Abstract Melanoma is an aggressive type of skin cancer that arises from melanocytes, the cells responsible for producing skin pigment. In contrast to non-melanoma skin cancers like basal cell carcinoma and squamous cell carcinoma, melanoma is more invasive. Melanoma was distinguished by its rapid progression, high metastatic potential, and significant resistance to conventional therapies. Although it accounted for a small proportion of skin cancer cases, melanoma accounts for the majority of deaths caused by skin cancer due to its ability to invade deep tissues, adapt to diverse microenvironments, and evade immune responses. These unique features highlighted the challenges of treating melanoma and underscored the importance of advanced tools, such as single-cell sequencing, to unravel its biology and develop personalized therapeutic strategies. Thus, we conducted a single-cell analysis of the cellular composition within melanoma tumor tissues and further subdivided melanoma cells into subpopulations. Through analyzing metabolic pathways, stemness genes, and transcription factors (TFs) among cells in different phases (G1, G2/M, and S) as well as between primary and metastatic foci cells, we investigated the specific mechanisms underlying melanoma metastasis. We also revisited the cellular stemness and temporal trajectories of melanoma cell subpopulations, identifying the core subpopulation as C0 SOD3 + Melanoma cells. Our findings revealed a close relationship between the pivotal C0 SOD3 + Melanoma cells subpopulation and oxidative pathways in metastatic tumor tissues. Additionally, we analyzed prognostically relevant differentially expressed genes (DEGs) within the C0 SOD3 + Melanoma cells subpopulation and built a predictive model associated with melanoma outcomes. We selected the gene IGF1 with the highest coefficient (coef) value for further analysis, and experimentally validated its essential function in the proliferation and invasive metastasis of melanoma. In immune infiltration analysis, we discovered the critical roles played by M1/M2 macrophages in melanoma progression and immune evasion. Furthermore, the development and progression of malignant melanoma were closely associated with various forms of programmed cell death (PCD), including apoptosis, autophagic cell death, ferroptosis, and pyroptosis. Melanoma cells often resisted cell death mechanisms, maintaining their growth by inhibiting apoptosis and evading autophagic cell death. Meanwhile, the induction of ferroptosis and pyroptosis was thought to trigger immune responses that helped suppress melanoma dissemination. A deeper understanding of the relationship between melanoma and PCD pathways provided a critical foundation for developing novel targeted therapies, with the potential to enhance melanoma treatment efficacy. These findings contributed to the development of novel prognostic models for melanoma and shed light on research directions concerning melanoma metastasis mechanisms and therapeutic targets. Keywords: Melanoma, Tumor metastasis, Oxidative phosphorylation, Macrophages, Single-cell RNA sequencing, Programmed cell death Introduction Melanoma, a malignancy originating from melanocytes, stands as one of the most prevalent and severe form of cutaneous carcinoma [[36]1–[37]3]. Often triggered by exposure to ultraviolet radiation, its incidence has demonstrated a persistent increase in recent years, with an estimated global annual incidence of 300,000 cases and mortality rates reaching up to 60,000, imposing substantial burdens on both public health and healthcare systems [[38]3, [39]4]. Presently, surgical excision remains the cornerstone of melanoma management, exhibiting favorable prognostic outcomes in the absence of metastatic spread [[40]5]. However, unfortunately, melanoma frequently progressed to form aggressive metastatic tumor tissues, leading to the demise of a majority of afflicted individuals [[41]6]. Patients with metastatic disease typically faced a median survival duration of merely 6–9 months, with a grim 5-year survival rate below 5% [[42]7], indicative of the limited efficacy of surgical interventions at this stage, which are also prone to relapse [[43]8]. Tumor invasion and metastasis served as critical determinants of disease progression, recurrence, and mortality in melanoma patients [[44]9]. Hence, there is a pressing need to delve deeper into the biological underpinnings of melanoma invasion and metastasis, as well as to identify pertinent pathways or immunotherapeutic targets, in order to ameliorate the prognosis of individuals afflicted with melanoma. Macrophages serve as effector cells in regulating the body's immune response, exhibiting high plasticity and categorizing into M1 macrophages, which promote inflammation progression, and M2 macrophages, which facilitate tissue repair. They participate in shaping the tumor microenvironment (TME) and play crucial roles in the initiation, proliferation, and metastasis of tumors [[45]10]. Specifically, M1 macrophages are involved in inhibiting tumor growth, whereas M2 macrophages contribute to its progression [[46]11, [47]12]. The redox balance is fundamental for cell survival and normal functioning, with oxidative processes in tumor cells often surpassing those in normal cells [[48]13, [49]14]. Oxidative phosphorylation, a pivotal pathway in this context, generates reactive oxygen species (ROS), during metabolism, which pose a risk of damaging nuclear and mitochondrial DNA, thereby increasing cancer susceptibility [[50]15, [51]16]. Furthermore, oxidative phosphorylation itself plays a certain promoting role in tumor metastasis [[52]17]. However, the specific mechanisms of macrophages and oxidative phosphorylation in the process of melanoma metastasis remain relatively understudied. Based on this, our study investigated the functional relevance and specific mechanisms of various melanoma cell subpopulations at the cellular level. Furthermore, we examined the correlation between the primary and metastatic tumor tissues of Melanoma cells, considering cell staging and metabolism. Through analysis of cellular stemness and pseudo-temporal trajectories, we identified the crucial subpopulation, and designed a prognostic model connected to them. In addition, we explored the interplay with immune function. These investigations aimed to provide a deeper insight into the mechanisms driving melanoma metastasis, with the ultimate goal of designing effective and specific immune therapies for melanoma and developing prognostic models. Materials and methods Data download and processing The single-cell sample data from 8 melanoma patients, including primary and metastatic tumor tissues, were sourced from the NCBI Gene Expression Omnibus (GEO) database (Accession No. [53]GSE189889). RNA-Seq data and clinical information of melanoma cells were retrieved from The Cancer Genome Atlas (TCGA) database. The data were normalized using R software (version 4.3.0). Since the data were sourced from public databases, no ethical review was required. Quality control The R package “DoubletFinder” [[54]18, [55]19] was used to purify, filter, and remove doublets and other low-quality data from the single-cell data obtained from the GEO database, which included 9 melanoma tumor tissue samples. The selection criteria were as follows: (1) 200 < number of expressed genes per cell (nFeature) < 6000; (2) 500 < total number of transcriptional genes per cell (nCount) < 80,000; (3) proportion of red blood cell genes < 5%; (4) proportion of mitochondrial genes < 25%. Dimensionality reduction, clustering, and cell annotation Single-cell data were normalized through the “NormalizeData” function in Seurat [[56]20–[57]22]. Identified highly variable genes with the Find Variable Features function, focusing on the top 2000 based on variance and standard deviation [[58]23, [59]24]. Conducted gene centering and standardization using the “ScaleData” function, and evaluated cell cycle effects with the Cell Cycle Features function. Performed principal component analysis (PCA) on the standardized highly variable genes with the PCA function. Then, addressed batch effects using the Harmony algorithm and executed clustering with “FindNeighbors” and “FindClusters” [[60]25]. Obtained cell annotations for different subpopulations from the Cell Marker database, refining based on relevant literature [[61]26, [62]27]. Cell annotation and tumor tissue preference analysis Then, we reclustered tumor cells and identified subpopulations based on marker genes. Differential expression analysis between subpopulations and between primary and metastatic samples was conducted using the “FindAllMarkers” function [[63]28, [64]29]. To assess the cancer preference of melanoma cell subpopulations, odds ratios were calculated using the calculation method. Subpopulation cell activity and metabolism analysis The “scMetabolism” package in the R package was used to screen and analyze the metabolic pathways of different phases, primary and metastatic tumor tissues and subpopulations. The metabolic pathways of enrichment were visualized using violin and UMAP plots. Subpopulation enrichment analysis We identified differentially expressed genes (DEGs) [[65]30] among subpopulation cells, with criteria including detection in at least 25% of cells, false discovery rate (FDR) < 0.05, and | logFCfilter |> 1. The DEGs before and after filtering were displayed for each subpopulation. Based on the DEGs among subpopulation cells, we performed Gene Ontology (GO) [[66]31–[67]33] enrichment analysis for each subpopulation by using “Cluserprofiler” package. Cell trajectory analysis Utilized the R package CytoTRACE to predict the differentiation states of melanoma subpopulations. Combined this with slingshot (v2.6.0) and monocle2 to infer cellular lineages and pseudotemporal variations [[68]34–[69]36]. Integrated synchronous principal and branching curves using the “getCurves” function to extract smoothed trajectory curves. Subpopulation cell stemness and transcription factors (TFs) The AUCell method was used to identify cells with active gene expression by analyzing single-cell RNA-seq data. This approach evaluated gene expression profiles by taking gene sets as input and assessing the activity of each gene set within individual cells. In this research, AUCell was specifically employed to assess the stemness characteristics of different tumor cell subpopulations. We utilized pySCENIC (v0.10.0) in Python 3.7 to infer regulatory networks at the single-cell level and to cluster tumor cell subpopulations [[70]37]. Target genes of TFs were identified using “GRNBoost” and further refined through DNA motif analysis. The AUCell method evaluated regulon activities, identifying the top 5 TFs with the significant changes in expression. Clinical implications and prognostic evaluation of the C0 SOD3 + melanoma cells subpopulation Developed predictive models for prognostication in patients with melanoma [[71]38]. Excluded incomplete clinical data and merged the intersection genes with the standardized clinical dataset. Conducted single-variable Cox regression analysis using the “coxph” function from the survival package in R, and validated the results with least absolute shrinkage and selection operator (LASSO)-penalized Cox regression [[72]39]. Identified prognostic differential genes through multivariable Cox regression analysis. Calculated the risk score for each sample (risk score = ΣXλ × coefλ, where X represents the relative expression levels of prognostic-related genes and coef represents the coefficients and stratified samples into high-risk and low-risk groups based on the median score. Employed PCA to examine sample distribution, and visualized the expression profiles of prognostic genes in each risk group using a heatmap. Highlighted coef associated with DEGs for further experimental validation. Illustrated the survival status of high-risk and low-risk groups over time using Kaplan–Meier curves [[73]40–[74]42], and assessed the specificity and sensitivity with time-dependent receiver operating characteristic (ROC) curves 's area under the curve (AUC) [[75]43–[76]45]. Explored the link between risk scores and prognostic gene expression, and incorporated age and melanoma staging information to refine the prognostic model, modeling it with the “rms” package in R [[77]46]. Developed a nomogram chart by combining the risk score with clinically relevant factors, serving as a predictive tool for melanoma patient prognosis. Validated the accuracy using ROC curves. Immunological and enrichment analysis Utilized the “Xcell” and “CIBERSORT” algorithms to analyze immune cell infiltration in high-risk and low-risk groups. Investigated correlations between risk scores, prognostic differential genes, and immune cells. Employed the R package “ESTIMATE” to calculate immune scores, stromal scores, and overall scores for the TME, compared Tumor Immune Dysfunction and Exclusion (TIDE) scores between the groups, and visualized results with violin plots. Filtered DEGs using the “limma” package with criteria of |log2 Fold Change|> 1 and FDR < 0.05. Conducted Gene Set Enrichment Analysis (GSEA) on the filtered genes [[78]47]. Cell culture The SK-MEL2 and A375 cell lines were sourced from the American Type Culture Collection (ATCC) and cultured in F12K and PRMI1640 media (Gibco BRL, USA), supplemented with 10% fetal bovine serum (Gibco BRL, USA) and 1% penicillin/streptomycin, under standard conditions (37 °C, 5% CO2, 95% humidity). Cell transfection Small interfering RNA (siRNA) constructs targeting IGF1 knockdown were obtained from GenePharma (Suzhou, China). Transfection was performed according to the Lipofectamine 3000 RNAiMAX protocol (Invitrogen, USA). Cells were seeded at 50% confluence in 6-well plates and transfected with negative control (si-NC) or knockdown siRNAs (Si-IGF1-1 and Si-IGF1-2). Each transfection utilized Lipofectamine 3000 RNAiMAX reagent. Cell viability assay Cell viability of transfected SK-MEL2 and A375 cells was evaluated using the CCK-8 assay. Cells were seeded at 5 × 10^3 per well in a 96-well plate and incubated for 24 h. Afterward, 10 μL of CCK-8 reagent (A311-01, Vazyme) was added per well and incubated for 2 h at 37 °C in the dark [[79]48]. Absorbance was measured at 450 nm on days 1, 2, 3, and 4 using a microplate reader (A33978, Thermo), and the average OD values were plotted. Western blot analysis Cells were cultured and transfected until reaching 70% confluence. Cell lysates were prepared using RIPA buffer, clarified by centrifugation at 12,000 rpm for 15 min, and subjected to SDS-PAGE. Proteins were transferred onto PVDF membranes, blocked with 5% BSA at room temperature for 1.5 h, incubated with primary antibodies overnight at 4 °C, followed by corresponding secondary antibodies for 1 h. Protein bands were visualized using ECL Western Blot [[80]49] Substrate. Quantitative real-time polymerase chain reaction (qRt-PCR) The process of RNA extraction was performed using Trizol reagent, followed by reverse transcription with the PrimeScript™ Kit. qRt-PCR was then conducted with SYBR Green premix for detecting amplification [[81]50]. EdU proliferation assay Transfected SK-MEL2 and A375 cells were seeded at a density of 5 × 10^3 cells per well in 6-well plates and cultured overnight. Cells were treated with 10 mM EdU solution in serum-free medium for 2 h at 37 °C. After fixation with 4% paraformaldehyde and permeabilization with glycine (2 mg/ml) and 0.5% Triton X-100, cells were incubated with 1X Apollo and 1X Hoechst 33,342 for 30 min at room temperature. Cell proliferation was quantitatively analyzed under a fluorescence microscope. Wound healing assay Transfected SK-MEL2 and A375 cells were grown in 6-well plates until 95% confluence. A straight scratch was made using a sterile pipette tip, followed by washing with PBS. The medium was replaced, and cells were incubated further. Images of the scratch were taken at 0 and 48 h to measure wound width. Transwell assay Prior to the experiment, cells were serum-starved for 24 h in serum-free medium. After coating with Matrigel (BD Biosciences, USA), cell suspensions were added to the upper chambers of Costar Transwell plates, while serum-containing medium was added to the lower chambers. Cells were incubated in a humidified chamber for 48 h. Following incubation, cells were fixed with 4% paraformaldehyde, stained with crystal violet, and assessed for invasive capability. Statistical analysis R and Python were employed for database data analysis, while experimental data were analyzed with GraphPad Prism version 8.0.1. Two-tailed p-values were applied in all analyses, with statistical significance determined as p-values less than 0.05. Results Single-cell analysis of melanoma tumor tissues Figure [82]1 illustrated the overall workflow of this study. At the beginning, we obtained single-cell data from 9 melanoma samples ([83]GSM5708993-[84]GSM5709001) representing tumors from 8 patients, including 5 primary and 4 metastatic tumor tissues. Employing rigorous data filtration and dimensionality reduction clustering techniques, we subdivided the melanoma tumor tissue into 34 subpopulations (Fig. [85]2A). Investigation into the origins and metastatic attributions of these cells revealed heterogeneous distribution patterns across the subpopulations, indicating diverse cellular compositions within the TME. The study mainly focused on primary acral melanoma tissues (AM2, AM3, AM4, AM6, AM8-Primary) and metastatic acral melanoma tissues (AM1, AM5, AM7, AM8-Mets, Mets indicated Metastatic). Notably, two types of samples were extracted from a single patient (Fig. [86]2B). Subsequent annotation and classification of the 34 clusters, delineated 9 distinct cellular populations within the melanoma tissue (Fig. [87]2C, D). These populations comprised Melanoma cells (18,083), NK-T cells (10,548), Endothelial cells (ECs) (1418), B-Plasma cells (1295), Myeloid cells (1535), Fibroblasts (1651), Pericytes (552), Epithelial cells (EPCs) (225), and Mast cells (79). Notably, melanoma cells constituted the largest cluster, serving as the predominant cellular entity within the melanoma TME, followed by NK-T cells, indicative of their immunological relevance. To investigate cellular migratory capabilities, we analyzed the distribution and proportions of cellular origins in primary and metastatic tumor tissues across the major cell populations (Fig. [88]3A–C). Cell cycle distribution (Fig. [89]3D–F) revealed a prominent G2/M phase predominance in EPCs, indicative of heightened proliferative activity. These findings underscored the necessity for further subtyping of melanoma cells to elucidate mechanisms underlying tumor progression and metastasis. Fig. 1. [90]Fig. 1 [91]Open in a new tab Workflow of single cell sequencing analysis for melanoma. We used single-cell sequencing technology, combined with bulk sequencing, in vitro animal experiments, and a series of analyses to comprehensively investigate melanoma. The aim was to identify novel therapeutic targets and improve patient prognosis Fig. 2. [92]Fig. 2 [93]Open in a new tab Clustering of melanoma tumor tissues. A Following single-cell clustering, a total of 34 clusters were identified from tumors and surrounding tissues of 8 melanoma patients. B These samples originated from 9 specimens of 8 patients. It could be divided into 5 primary acral melanoma tissue and 4 metastatic acral melanoma tissues. C, D Annotation of different cell populations based on distinct cell surface markers, including Melanoma cells, NK-T cells, ECs, B-Plasma cells, Myeloid cells, Fibroblasts, Pericytes, EPCs, and Mast cells Fig. 3. [94]Fig. 3 [95]Open in a new tab The distribution of melanoma cell populations and the cell cycle phases. A–C The UMAP plots illustrated the spatial distribution of different cells within primary and metastatic melanoma tissues. Pie charts were used to depict the proportions of primary and metastatic melanoma tissues from different groups within each cell type. D–F The UMAP plots illustrated the spatial distribution of various cells within primary and metastatic melanoma tissues. Pie charts were used to describe the proportions of primary and metastatic melanoma tissues from different cell types across different cell cycle phases (G1, G2/M, S) Subpopulation and primary-metastasis analysis of melanoma cells Further exploration was done to understand the relationship between heterogeneity and subpopulations in melanoma patients. Through further clustering of melanoma cells, we subdivided them into 8 subpopulations (Fig. [96]4A). Investigation into the cellular origins of these subpopulations revealed their derivation (Fig. [97]4B). According to the expression levels of cell-specific genes, we reclassified them into 8 subpopulations, including C0 SOD3 + Melanoma cells, C1 MTRNR2L12 + Melanoma cells, C2 PMEL + Melanoma cells, C3 IGF2 + Melanoma cells, C4 COMP + Melanoma cells, C5 CXCL9 + Melanoma cells, C6 THBS2 + Melanoma cells, C7 PECAM1 + Melanoma cells (Fig. [98]4C). To delve deeper into distribution of subpopulations and metastatic relationships of each subpopulation, we evaluated the cell cycle phases and primary metastatic status of each subpopulation. Remarkably, the majority of cells in the C0 SOD3 + Melanoma cells subpopulation was found to be in the G1 phase (Fig. [99]4D–F). In addition, the proportion of metastatic tumor tissues in the C0 SOD3 + Melanoma cells subpopulation was relative higher (Fig. [100]4G–I). Fig. 4. [101]Fig. 4 [102]Open in a new tab Comprehensive distribution landscape of melanoma cell subpopulations. A The melanoma cells underwent dimensionality reduction and clustering, resulting in the identification of 8 distinct subpopulations. B The 8 subpopulations were derived from different melanoma tissues, primarily categorized into primary and metastatic tissues. C The melanoma cells were distinguished into 8 subpopulations based on the different characteristic genes. D–F The UMAP plots illustrated the distribution characteristics of the 8 subpopulations across different cell cycle phases. Pie charts depicted the proportions of each phase within the various subpopulations. G–I The UMAP plots showed the distribution characteristics of the 8 subpopulations based on their tissue origins. Pie charts displayed the proportions of primary and metastatic melanoma tissues within the different subpopulations To further investigate the interplay between cell cycle phases and primary metastasis among different subpopulations, we visualized the expression of key genes across all subpopulations (Fig. [103]5A). Strikingly, the key genes expression profile of the C0 SOD3 + Melanoma cells subpopulation closely mirrored that of metastatic tumor tissues, suggesting an association between the C0 SOD3 + Melanoma cells subpopulation and tumor metastasis. Analysis of the cell cycle in each subpopulations revealed that the C0 SOD3 + Melanoma cells subpopulation showed distinct differences compared to other subpopulations, exhibiting a considerably higher proportion of cells in the G1 phase, reaching 50.7%, while other subpopulations showed more uniform distributions across G2/M and S phases (Fig. [104]5B, C). Furthermore, our analysis of the cell proportions involved in primary and metastatic tissues across different subpopulations indicated that within the C0 SOD3 + Melanoma cells subpopulation, metastatic tumor tissue cells represented the higher proportion, accounting for 78.1% (Fig. [105]5D, E). Further analysis of Ro/e preference analysis across subpopulations revealed that within all subpopulations, the C0 SOD3 + Melanoma cells subpopulation exhibited the highest Ro/e value of metastatic tumor tissue cells and G1 phase cells (Fig. [106]5F, G). Hence, we hypothesized that certain life activities or metabolic pathways of the C0 SOD3 + Melanoma cells subpopulation during the G1 phase could be associated with tumor metastasis, potentially influencing other subpopulations through specific metabolic pathways to promote tumor proliferation and metastasis. Fig. 5. [107]Fig. 5 [108]Open in a new tab Phase and distribution of subpopulations within primary and metastatic tumor tissues. A The bubble diagram showed the expression levels of different marker genes across the 8 subpopulations and various tissues. Pie charts illustrated the cell cycle proportions for different cell subpopulations and tissues, while violin plots presented the G2/M Score, S Score, and nCount-RNA levels for different cell subpopulations and tissues. B, C The bar graphs showed the proportion of each subpopulations in the different phases. D, E The bar graphs showed the proportion of each subpopulations in the primary and metastatic groups. The proportion of primary and metastatic melanoma tissues within subpopulations C0 to C7. F Ro/e values of 8 melanoma cell subpopulations in metastatic and primary tumor tissues G Ro/e values of 8 melanoma cell subpopulations were assessed across different cell cycle phases (G1, G2/M, and S phases) Metabolic pathways and activity analysis of subpopulations To better understand the mechanisms of melanoma cell subpopulations in the context of staging and metastasis, we assessed the metabolic pathways across distinct subpopulations, cell cycle phases, as well as primary and metastatic tumor tissues (Fig. [109]6A–C). We observed that the pathways of oxidative phosphorylation, glutathione metabolism, and glycolysis/gluconeogenesis consistently ranked high across different groups. To further explore and compare the scores of these pathways across different subpopulations, cell cycle phases, and primary and metastatic tumor tissue cells, we visualized them using UMAP and violin plots (Fig. [110]6D–O). Notably, there was a significant upregulation in the expression of these three metabolic pathways in the C0 SOD3 + Melanoma cells subpopulation. In addition, we found that the above three metabolic pathways exhibited higher expression levels in the G1 phase and metastatic tumor tissues. This suggested a potential correlation between these pathways and the activity and metastasis of melanoma tumor cells. Interestingly, the expression levels nFeature-RNA and nCount-RNA were high in the C0 SOD3 + Melanoma cells subpopulation. Metastatic tumor tissue cells and those in the G1 phase also exhibited significantly higher expression compared to other cell types and cell cycle phases (Fig. [111]6P–W). We hypothesized that the C0 SOD3 + Melanoma cells subpopulation might influence the activity and metastasis of tumor cells through oxidative-related pathways and potentially promote tumor proliferation by affecting other subpopulations' cells via differentiation or signaling pathways. Fig. 6. [112]Fig. 6 [113]Open in a new tab Cell metabolic pathway analysis landscape. A–C Top 20 metabolic pathways analyzed across different subpopulations, phases, as well as primary and metastatic tumor tissues. D–O UMAP and violin plots depicted the distribution and scores of oxidative phosphorylation, glutathione metabolism, glycolysis/gluconeogenesis pathways in different subpopulations, phases, as well as primary and metastatic tumor tissues. P–S UMAP and violin plots showed nCount-RNA distribution and scores across different subpopulations, phases, as well as primary and metastatic tumor tissues. T–W UMAP and Violin plots displayed nFeature-RNA distribution and scores across different subpopulations, phases, as well as primary and metastatic tumor tissues. (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, "ns" was used to say that there was no significant difference) Enrichment analysis of subpopulations Then, we computed the DEGs between each subpopulation of melanoma cells and showcased the top 5 upregulated and downregulated genes for each comparison (Fig. [114]7A–H). Subsequently, we proceeded with GO enrichment analysis on these DEGs (Fig. [115]7I–P). We discovered that the enriched pathways associated with the C0 SOD3 + Melanoma cells subpopulation included “extracellular matrix organization”, “extracellular structure organization”, “external encapsulating structure”, and “cell-substrate adhesion organization”. Existing studies had shown that the “extracellular matrix organization” pathway was closely related to cancer cell metastasis [[116]51]. Therefore, we inferred that the C0 SOD3 + Melanoma cells subpopulation was intricately linked to melanoma metastasis and might achieve this through pathways involving extracellular matrix and tissue adhesion. Fig. 7. [117]Fig. 7 [118]Open in a new tab Enrichment analysis of melanoma cell subpopulations. A–H DEGs within each melanoma cell subpopulations. I–P Functional enrichment analysis of GO-BP terms for differential genes within each melanoma cell subpopulation Cell trajectory analysis of the subpopulations For a clearer insight into the developmental trajectories of melanoma cell subpopulations, we used pseudotime analysis to infer their progression. We utilized CytoTRACE to predict the cellular stemness of each subpopulation of melanoma cells (Fig. [119]8A, B). We observed that the C0 SOD3 + Melanoma cells subpopulation exhibited the strongest cellular stemness, followed by the C2 PMEL + Melanoma cells subpopulations. Additionally, we utilized slingshot to elucidate the temporal ordering of subpopulations (Fig. [120]8C, D), which revealed four distinct trajectories corresponding to various cellular differentiation pathways. Notably, all four trajectories originated from the C0 SOD3 + Melanoma cells subpopulation, suggesting that this subpopulation exhibited higher level of cellular stemness and functions as the primary source for the differentiation of the other subpopulations. Subsequently, we validated these results through monocle analysis and examined the temporal expression order of stemness-associated genes (Fig. [121]8E). Additionally, we analyzed the subpopulation trajectories calculated by monocle (Fig. [122]8F, G), revealing that C0 SOD3 + Melanoma cells was likely positioned at the initial phase of the temporal order, consistent with the results obtained through slingshot and CytoTRACE. Through our investigations, we identified the C0 SOD3 + Melanoma cells subpopulation as the key subpopulation, characterized by its strong cellular stemness, metabolic activity, and high correlation with metastasis. Fig. 8. [123]Fig. 8 [124]Open in a new tab Pseudotime analysis of subpopulation cells. A Using CytoTRACE, predicted ordering was performed on the 8 subpopulations, with higher scores indicating greater differentiation potential. B Distribution characteristics of predicted order and corresponding cell subpopulations of melanoma cells. C, D Prediction of time trajectories for subpopulations using slingshot, revealing 4 distinct trajectories, with UMAP plots illustrating each trajectory. (Lineage1:C0 → C1 → C2 → C3; Lineage2:C0 → C1 → C4; Lineage3:C0 → C1 → C5; Lineage4:C0 → C1 → C7. “ → ” represented the direction of pseudotemporal trajectory differentiation. E Dynamic expression trend of different stemness genes over time based on monocle analysis. F, G Violin and UMAP plots illustrated the pseudotime expression levels of different melanoma cell subpopulations Stemness and TFs analysis To further investigate the crucial role played by tumor development, cell proliferation, metastatic behavior, and drug resistance, we investigated the stemness-associated genes of each cell subpopulation, different cell cycle phases, as well as primary and metastatic tumor tissues. (Fig. [125]9A–D). We observed significantly higher cellular stemness in the C0 SOD3 + Melanoma cells subpopulation, G1 phase, and metastatic tumor tissues. Moreover, we examined the expression patterns of stemness-associated genes across subpopulations, cell cycle phases, as well as primary and metastatic tumor tissues (Fig. [126]9E–H). Notably, TWIST1, MYC, HLFA, CD44, exhibited significant expression across C0 SOD3 + Melanoma cells subpopulation, and their expression levels were visualized using UMAP plots (Fig. [127]9I–L). Furthermore, stemness-associated genes showed pronounced expression in the C0 SOD3 + Melanoma cells subpopulation, G1 phase, and metastatic tumor tissues, suggesting their involvement in the activity of the C0 SOD3 + Melanoma cells subpopulation and potentially in the metastatic process. Fig. 9. [128]Fig. 9 [129]Open in a new tab Stemness analysis of cells. A–C The AUC value of cell stemness within each subpopulation, phase, as well as in the primary and metastatic tumor tissues. D The UMAP plot demonstrated the AUC density distribution characteristics of stemness across the cell subpopulations. E The expression levels of stemness genes in the 8 subpopulations, as well as in primary and metastatic melanoma tissues, were analyzed. F–H Expression levels of stemness genes within each subpopulation of cells, across different phases, as well as within primary and metastatic tumor tissues. I–L Expression distribution characteristic of stemness genes TWIST1, MYC, CD44, and HIF1A within UMAP plots To delve deeper into the specific mechanisms underlying tumor metastasis, we investigated the TFs associated with primary and metastatic tumor tissues, computed and selected the top 5 TFs, and visualized them using UMAP plots (Fig. [130]10A–N). We found that TFs related to metastatic tumor tissues were predominantly expressed in the region associated with the C0 SOD3 + Melanoma cells subpopulation, while TFs related to primary tissues were distributed among various subpopulations. Additionally, we observed that TCF7L2 ranked highly in specificity scores within melanoma metastasis samples. Consequently, we conducted an in-depth analysis of this TF, focusing on its visualization across different melanoma subpopulations, cell cycle phases, and various classifications (Fig. [131]10O–R). The results indicated that TCF7L2 exhibited expression levels in the C0 SOD3 + Melanoma cells subpopulation and metastasis samples. Notably, TCF7L2 was linked to the advancement of various cancers, particularly gastric cancer, where it played a key role in regulating upstream and downstream genes during transcription, enhancing tumor cell drug resistance, and significantly promoting apoptosis and metastasis. Moreover, the presence of TCF7L2 was often observed in cases where gastric cancer patients had poor prognoses [[132]52].Therefore, we might hypothesize that TCF7L2 played an integral part in the metastatic process of melanoma. This TF could serve as a potential therapeutic target. Fig. 10. [133]Fig. 10 [134]Open in a new tab TFs analysis. A, H TFs distribution characteristics of primary and metastatic melanoma tissues. B, I The scatter diagrams displayed the specificity scores of TFs within regulons in primary and metastatic melanoma tissues. C–G, J–N UMAP plots showed distribution of top 5 TFs in primary and metastatic tumor tissues. O UMAP plot displayed the distribution of TCF7L2 within subpopulations. P–R Violin plots depicted expression levels of TCF7L2 within each different subpopulations, cell cycle phases, as well as primary and metastatic tumor tissues. (****P < 0.0001. "ns" was used to say that there was no significant difference) Construction and analysis of the prognostic model To further explore the patients' prognostic status, we performed an intersection analysis between the DEGs from the TCGA database and the signature genes of the C0 SOD3 + Melanoma cells subpopulation in melanoma. Using univariate Cox regression analysis, we identified 38 risk genes associated with prognosis (Fig. [135]11A). The stability and reliability of these genes were confirmed through LASSO Cox regression analysis (Fig. [136]11B, C). Furthermore, through multivariable Cox regression analysis, we ultimately identified 15 prognostic genes and performed a coef analysis. The figure presented the coef values of these prognostic genes (Fig. [137]11D). Given that IGF1 had relatively higher coef values, we hypothesized its important role in melanoma metastasis and progression. Consequently, we selected this gene for further experimental validation. Among them, IGF1, CD81, FBLN1, GPC3, and APCDD1 were classified as high-risk genes, while APOD, PLSCR4, MGP, MRPS6, GADD45A, SERPING1, SPRY1, MT2A, RGS16, and IFITM1 were categorized as low risk genes. Risk scores were used to categorize patients into high-risk and low-risk groups (Fig. [138]11E), and their survival status over time was assessed (Fig. [139]11F). We observed a concentration of deceased patients in the high-risk group, with a significant decrease in the number of survivors over time. Additionally, we displayed the expression levels of prognostic genes in the high-risk and low-risk groups using a heatmap (Fig. [140]11G). The ROC curve's area under the curve values at 1-year, 3-years and 5-years were 0.69, 0.69, and 0.74, respectively (Fig. [141]11H), indicating the stable and reliable predictive capability of our model. Kaplan–Meier survival curves (Fig. [142]11I) revealed a substantial difference in survival rates between the high-risk and low-risk groups, with a statistically significant p-value of less than 0.0001. We also explored the correlation between genes and risk scores, highlighting genes with strong correlations (Fig. [143]11J, K). The expression levels of APCDD1, IGF1, CD81, and FBLN1 were positively correlated with risk, while MRPS6, PLSCR4, SERPING1, and IFITM1 were negatively correlated with risk. In other words, higher expression levels of APCDD1, IGF1, CD81, and FBLN1 were associated with an poor prognosis. Fig. 11. [144]Fig. 11 [145]Open in a new tab Independent prognostic analysis. A Univariate regression analysis uncovered 38 genes connected to prognosis. (HR < 1: protective factors, HR > 1: risk factors). B The coef spectrum distribution of 15 prognostic genes as determined by LASSO regression analysis. C Parameter selection in optimal cross-validation LASSO regression. D The coef values of genes associated with prognosis. E Sorting patients into high-risk and low-risk groups according to risk scores. F Scatter diagram showed the temporal trends in the occurrence of death and alive events. G The heatmap visualized the expression levels of prognostic genes in high-risk and low-risk groups. H Time-dependent ROC curves with AUC values of 0.69, 0.69, and 0.74 for 1-year, 3-years, and 5-years survival, respectively. I Kaplan–Meier survival analysis curves for high-risk and low-risk groups. J Correlation analysis between genes and risk scores. K Scatter diagrams illustrated the correlation of 8 prognostic genes with risk scores Furthermore, we investigated the association between clinical variables like age, tumor stages, and prognosis (Fig. [146]12A–D). In addition, we performed risk assessments based on patients' risk groups, race, age, and tumor stages. Forest map revealed some independent risk factors (P < 0.05) (Fig. [147]12E). Based on the prognostic genes and clinical factors, we constructed a nomogram chart to predict the survival rates of melanoma patients at 1-year, 3-years, and 5-years (Fig. [148]12F). The accuracy of our nomogram chart model was validated using ROC curve, showing AUC values of 0.72, 0.79, and 0.79 at 1-year, 3-years, and 5-years, respectively (Fig. [149]12G), indicating its high specificity and sensitivity. Fig. 12. [150]Fig. 12 [151]Open in a new tab Clinical relevance analysis. A–D Comparative analysis of risk scores between different prognostic factor groups. E The forest plot demonstrated the results of multivariate cox regression analysis integrating risk scores and clinical factors (age, race and tumor clinical stage T, M and N). F Nomogram showed the prediction of 1-year, 3-years, and 5-year of OS based on race, tumor clinical stage (T, M, and N), age, and risk score. G Time-dependent ROC curves with AUC values of 0.72, 0.79, and 0.79 for 1-year, 3-years, and 5-years survival, respectively Immuno-related and enriched functional analysis Additionally, we carried out an in-depth analysis to explore the role of immune infiltration in disease progression. We conducted an analysis of immune infiltration in both high-risk and low-risk groups (Fig. [152]13A, B) and identified immune cell populations with higher proportions (Fig. [153]13C), where macrophages constituted a significant portion. Further comparison of immune cell compositions between the two groups (Fig. [154]13D) revealed a higher proportion of M1 macrophages in the low-risk group and a higher proportion of M2 macrophages in the high-risk group. Additionally, M1 macrophages exhibited a negative correlation with the risk score, while M2 macrophages showed a positive correlation (Fig. [155]13E). In summary, we hypothesized that macrophages might be involved in the proliferation and metastasis of melanoma and thus affect the prognosis of patients, but the roles played by M1 and M2 macrophages were different. Fig. 13. [156]Fig. 13 [157]Open in a new tab Immune-related analysis. A The heatmap displayed the differences in immune infiltration expression of various factors between the high-risk and low-risk groups. B Differential proportions of immune cells between high-risk and low-risk groups. C Comparison of immune cell infiltration and estimated proportions. D Differential estimated proportions of immune cells between high-risk and low-risk groups. E Correlation analysis between risk scores and different immune cells. F Heatmap analysis of the correlation between gene expression, risk score, OS, and immune cell infiltration. G–I Differential immune scores, stromal scores, and ESTIMATE scores of TME in high-risk and low-risk groups. J Violin plot displayed TIDE expression levels in high-risk and low-risk groups. (*P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001.) We also explored the interplay between prognostic genes, risk, OS and immune cells (Fig. [158]13F) and found an association between the OS and both M1 and M2 macrophages, displaying consistent with previous findings, M1 macrophages were positively correlated with OS, while M2 macrophages were negatively correlated with OS. M2 macrophages might have been associated with tumor progression. Regarding TME-related scores, the low-risk group exhibited higher scores for stromal, immune, and estimate scores (Fig. [159]13G–I). While TIDE scores were also higher in the low-risk group, suggesting that patients in the high-risk group might have a poorer response to treatment with immune checkpoint inhibitors (Fig. [160]13J). Furthermore, GSEA revealed notable enrichment of pathways connected to "epidermis development", “skin development” and "keratinization" in the positive expression trend group, which were associated with skin metabolism. Conversely, pathways related to "positive regulation of leukocyte cell–cell adhesion", "regulation of lymphocyte mediated immunity," and "adaptive immune response" showed a negative expression trend (Fig. [161]14A–F). Fig. 14. [162]Fig. 14 [163]Open in a new tab GSEA depicted enriched functional pathways. A–C Enrichment analysis of pathways associated with positive trend expression. D–F Pathways enrichment analysis related to pathways with negative expression trends In vitro experiment of IGF1 Based on analysis of prognostic genes, we selected IGF1 as the target gene for our experiments. We conducted experiments using two melanoma cell lineages, employing methods to compare negative control and knockdown infection groups. In the cell viability assay (Fig. [164]15A, B), CCK-8 tests indicated significantly reduced cell viability after knockdown. In colony formation assays (Fig. [165]15C, D), the colony numbers formed by IGF1 knockdown cells was notably lower in contrast to the negative control group. Cell proliferation assays (Fig. [166]15E, F) showed a decreased cell count in both melanoma cell lines following IGF1 knockdown compared to the negative control group. In the wound healing assays (Fig. [167]15G, H), the width of scratches made on IGF1 knockdown cells was significantly wider than those on the negative control group after 48 h. In the Transwell assays, the migration of both cell lines treated with IGF1 knockdown showed significantly smaller stained areas against the negative control group (Fig. [168]15I, J). Similarly, in the invasion assay (Fig. [169]15K, L), the two melanoma cell lines with IGF1 knockdown exhibited markedly reduced invasion compared to the negative control group. Therefore, through these experiments, we found that knockdown of IGF1 in melanoma cells led to decreased proliferation, migration, and invasion capabilities, indicating that IGF1 probably played a positive role in the proliferation, migration, and invasion of melanoma, thereby promoting its progression. Fig. 15. [170]Fig. 15 [171]Open in a new tab In vitro assays of prognostic gene IGF1 on proliferation and invasion metastasis. A, B CCK-8 assays revealed significantly reduced cell viability after IGF1 knockout. C, D Colony formation assays demonstrated a notable decrease in colony numbers in cells with IGF1 knockdown against the negative control group. E, F EdU staining results indicated suppressed proliferation of SK-MEL2 and A375 cells upon IGF1 knockdown. G, H Wound healing assays revealed markedly decelerated migration of SK-MEL2 and A375 cells following IGF1 knockdown. I, J Transwell assays showed significantly decreased migration of SK-MEL2 and A375 cells upon IGF1 knockdown. K, L Transwell assays demonstrated a significant reduction in invasion of SK-MEL2 and A375 cells following IGF1 knockdown Discussion Melanoma is a fast-growing and aggressive tumor, often linked to a poor prognosis. Its heterogeneity and the complexity of the TME posed significant challenges for treatment [[172]53]. In recent years, single-cell sequencing technology, as an innovative tool, provided precise insights into tumor heterogeneity and the dynamic changes of the microenvironment. By analyzing gene expression, immune characteristics, and metabolic states at the single-cell level, this technology enabled the identification of key pathological cell populations and their functional properties, offering a scientific basis for developing personalized therapeutic strategies. This study leveraged single-cell sequencing technology to comprehensively investigate the TME and cellular heterogeneity in melanoma [[173]54]. The aim was to elucidate the mechanisms of key pathological cell populations. The findings of this research not only contributed to optimizing treatment strategies and improving patient prognosis but also advanced the clinical application of single-cell technology in melanoma treatment, providing theoretical and data support for the development of personalized therapeutic approaches. In tumor types, metastasis and dissemination often signify poor prognosis for patients, with approximately 90% of cancer-related deaths attributed to metastasis [[174]55]. Melanoma, as one of the most malignant forms of skin neoplasm, lacked effective treatment options once metastasized, owing to the incomplete elucidation of its underlying mechanisms and molecular targets [[175]56]. Therefore, investigating and refining the mechanisms and treatment targets of melanoma metastasis, as well as identifying new therapeutic directions, was paramount. Hence, in this study, we selected key subpopulations associated with metastasis, explored their intrinsic metabolic pathways, constructed prognostic models relevant to them, and investigated their relationship with immunity. Our analysis of metabolic pathways revealed that oxidative-related pathways, particularly oxidative phosphorylation, ranked highest across all subpopulations. This pathway showed the strongest expression in the C0 SOD3 + Melanoma cells subpopulation and was notably elevated in metastatic tumor tissues compared to primary tumor tissues. Hence, we hypothesized that oxidative-related pathways, particularly oxidative phosphorylation, played a crucial role in melanoma metastasis. Literature review revealed the vital importance of oxidative phosphorylation in tumor biology, as inhibiting this pathway's activity could reduce the anti-tumor effect and cell proliferation of melanoma cells [[176]57–[177]59]. Intriguingly, certain studies suggested that tumor stem cells with high metastatic potential were more dependent on the oxidative phosphorylation process, and inhibiting this pathway could improve tumor recurrence [[178]60, [179]61], aligning with our findings of significantly elevated oxidative phosphorylation pathway expression in metastatic cells. Current studies [[180]62] had demonstrated that inhibiting oxidative phosphorylation could suppress ARNT and PDK1-deficient melanoma cell migration and invasion. Additionally, the oxidative phosphorylation process was accompanied by ROS production, and further studies had [[181]63] found that carbon dots-induced ROS promoted melanoma cell growth and invasiveness. What is more, other studies had indicated that ROS exert inhibitory effects on melanoma dissemination and metastasis [[182]64, [183]65]. We believed that this discrepancy could be correlated with ROS, a hypothesis that necessitates further experimental investigation and confirmation. Subsequently, to further investigate the evolutionary trajectory of melanoma subpopulations, we employed pseudotime analysis. We observed that, regardless of lineage, the C0 SOD3 + Melanoma cells subpopulation consistently emerged as the initial subpopulation with the highest predicted ordering by CytoTRACE. Thus, compared to other subpopulations, the C0 SOD3 + Melanoma cells subpopulation was more primitive, with lower differentiation and higher proliferation capacity, indicative of higher malignancy and tumor-promoting potential. The unique characteristics of the C0 SOD3 + The melanoma cell subpopulation indicated that it might play a crucial role in melanoma treatment. Furthermore, when combined with the trajectory analysis provided by Slingshot, which traced the developmental pathways of melanoma subpopulations, our findings further emphasized the central role of the C0 SOD3 + Melanoma cells subpopulation in the early stages of melanoma progression. Slingshot’s reconstruction of the lineage relationships reinforced the notion that this subpopulation might have been pivotal in initiating malignant transformation, making it an ideal candidate for targeted therapeutic strategies. Stemness analysis revealed that the C0 SOD3 + Melanoma cells subpopulation exhibited a higher stemness score, indicating a more undifferentiated state. Higher stemness scores are typically associated with increased self-renewal capacity and the potential for differentiation into various cell types. This suggests that the C0 SOD3 + Melanoma cells subpopulation not only maintains a primitive state but also contributes to tumor heterogeneity and progression. Cells with elevated stemness are often linked to poor prognosis, as they can drive metastasis and resist conventional therapies. Therefore, targeting the stemness characteristics of the C0 SOD3 + Melanoma cells subpopulation could provide new therapeutic strategies for melanoma treatment. In the analysis of stemness gene expression, key genes such as TWIST1, MYC, CD44, and HIF1A were predominantly expressed. TWIST1 was a TF involved in the epithelial-mesenchymal transition (EMT) process [[184]66], playing a crucial role in the initiation and metastasis of melanomas [[185]67]. During EMT, TWIST1 suppressed EPCs characteristics while inducing the expression of mesenchymal markers, thereby endowing cancer cells with enhanced migratory and invasive capabilities. TWIST1 was implicated in the progression of various cancers, such as breast cancer, lung cancer, and gastric cancer, often correlating with aggressive tumor behavior, metastasis, drug resistance, and poor prognosis [[186]68–[187]70]. MYC was a well-known oncogene that played a pivotal role in cancer cell invasion and migration, thus promoting tumor progression [[188]71]. It was involved in the expression of multiple oncogenic non-coding RNAs, and when aberrantly expressed, MYC disrupted normal signaling pathways, leading to crosstalk between oncogenic pathways and possibly inducing replication stress, which could, in turn, lead to cancer development [[189]72, [190]73].CD44 was a transmembrane glycoprotein that acted as an adhesion molecule on the cell surface, promoting cell-extracellular matrix interactions. Its adhesive properties and ability to degrade hyaluronic acid made it a key driver of tumor invasion and metastasis [[191]74]. Additionally, CD44 was recognized as a cancer stem cell marker in various tumors [[192]75], closely associated with tumor recurrence, metastasis, and chemoresistance. While CD44 had both oncogenic and tumor-suppressive roles, in this study, we focused on its oncogenic effects. By binding to its ligands, such as hyaluronic acid, CD44 strengthened the connection between tumor cells and the extracellular matrix, thereby promoting tumor cell invasiveness and metastatic potential [[193]76]. HIF1A was a TF upregulated under hypoxic conditions, regulating cellular adaptation to hypoxia, which was especially critical in the TME. Hypoxia was a key element in tumor survival and growth, and the TME was frequently hypoxic. Activation of HIF1A helped tumor cells adapt to low oxygen conditions, promoting immune evasion by releasing exosome-mediated signaling, thus contributing to tumor progression [[194]77, [195]78].In summary, these genes and proteins regulated tumor cell proliferation, survival, migration, and metastasis through distinct mechanisms. They were not only key drivers of tumor progression but also potential therapeutic targets. To gain a comprehensive understanding of melanoma patient prognosis, we constructed a prognostic model to analyze genes that might contribute to poor prognosis, to intervene and improve patient prognosis. Based on current data, the IGF1 gene scored the highest coef values. Existing studies suggested that IGF1 was closely associated with drug resistance, and targeting the IGF1 gene could enhance drug sensitivity to some extent, leading to improved therapeutic outcomes for patients [[196]79]. IGF1 was often associated with poor prognosis in various cancers, such as breast cancer and Wilms tumor [[197]80, [198]81]. It contributed significantly to the initiation, progression, and prognosis of many cancers by activating different signaling pathways through the IGF1 receptor, promoting tumor cell proliferation, survival, invasion, and metastasis. For example, in colorectal cancer, IGF1 exerted its effects on downstream genes via the PI3K/AKT/HIF1α pathway, enhancing tumor cell proliferation and inhibiting apoptosis, thereby increasing cancer cell invasiveness and metastatic potential [[199]82].In conclusion, while research on IGF1 in melanoma was limited, based on these findings, we could infer that the IGF1 gene played a significant role in melanoma. The IGF1 gene was poised to be a critical target for improving patient prognosis. Therefore, to verify the role of IGF1 in tumor cells, we conducted experiments to support our conclusions. Consistent with the aforementioned findings, in assays such as cell viability, wound healing assays, and invasion tests, our experiments exhibited that IGF1 was instrumental in promoting the proliferation, invasion, and migration of melanoma cells. These experimental results further reinforced that IGF1 was a promising therapeutic target for improving patient prognosis. Therefore, more attention should be given to the IGF1 gene, and by targeting this gene, inhibiting its activity could potentially lead to improved patient outcomes. In our investigation of immune-related phenomena, we observed a relatively high infiltration of macrophages, among which both M1 and M2 macrophages exhibited significant correlations with risk scores, albeit in opposing directions. Typically, M1 and M2 macrophages could interconvert, with M1 primarily associated with inflammation and exerting anti-tumor activity, while M2 was associated with tissue repair and often promoted tumor progression and invasion [[200]83–[201]85]. Consistent with our findings, M1 was positively associated with patient prognosis, while M2 was indicative of a poorer prognosis. Further literature review revealed that M1 macrophages infiltrating the immune microenvironment could inhibit tumor metastasis through phagocytosis and immune regulation [[202]86–[203]88], while M2 macrophages could promote tumor progression and metastasis through mechanisms such as secreting CCL5, upregulating CRYAB expression, and activating the ERK1/2/Fra-1/slug signaling pathway [[204]89–[205]91]. Studies had shown that inhibiting M2 macrophage polarization could effectively suppress melanoma metastasis [[206]92]. Intriguingly, existing research had [[207]93] found that activation of oxidative-related pathways could lead to immune suppression and polarization of macrophages from M1 to M2. Mouton [[208]94] also discovered that M2 macrophage activity and conversion were highly dependent on the tricarboxylic acid cycle and oxidative phosphorylation. Additionally, ROS, besides participating in tumor formation, could induce macrophage polarization toward M2 and promote the production of IFN and IL-6 by macrophages, leading to a suppressive tumor immune microenvironment [[209]95, [210]96], ultimately resulting in M2 macrophage polarization and consistent immune-related functions, thereby increasing the likelihood of tumor escape and melanoma metastasis. Therefore, we proposed that the activation of oxidative phosphorylation and related oxidative pathways in the C0 SOD3 + Melanoma cells subpopulation and metastatic tumor tissue cells might induce M2 macrophage polarization, thereby triggering a suppressive immune microenvironment and enhancing the likelihood of melanoma metastasis. We conducted an in-depth study on melanoma using data obtained through single-cell sequencing technology. We analyzed the heterogeneous subpopulations of cells within melanoma tumors, providing high-resolution insights into the distinct functions of diverse cell groups within the TME, particularly the key mechanisms involved in tumor metastasis. This cell-level information aid in identifying critical subpopulations related to malignant metastasis and prognosis, such as the C0 SOD3 + subpopulation identified in the study. Apart from examining tumor cells, we extended our analysis to the distinct roles of M1 and M2 macrophages in the TME, revealing their contributions to melanoma progression and investigating their associations with prognostic genes. This offered potential targets for the development of novel immunotherapies [[211]97]. In melanoma, PCD pathways had critical roles in disease progression and therapeutic resistance [[212]98]. Melanoma cells frequently evaded apoptosis and autophagy, which allowed sustained tumor growth and resistance to conventional therapies. However, inducing ferroptosis and pyroptosis showed promise in stimulating anti-tumor immune responses, potentially limiting melanoma dissemination. By disrupting redox balance and releasing pro-inflammatory factors, these PCD pathways facilitated immune activation and reshaped the TME. Targeting PCD in melanoma thus provided innovative strategies to overcome therapeutic resistance and improve patient outcomes. However, there were certain limitations to our study. Firstly, the dataset was limited. Although the study analyzed single-cell samples from 8 melanoma patients, the sample size was relatively constrained, which might restrict the generalizability of the results, particularly for different types or stages of melanoma. Additionally, our experimental focus was primarily on in vitro cellular studies, with a lack of in vivo animal model experiments. Therefore, it is essential to conduct a more in-depth exploration. Conclusion Our study delved into the detailed mechanisms of melanoma metastasis at the single-cell level, exploring the roles of oxidative stress pathways and the immune system therein. Firstly, we identified the critical subpopulation, C0 SOD3 + Melanoma cell subpopulation, which played a pivotal role in disease progression. These cells exhibited antioxidant properties and immune modulation capabilities, contributing to tumor plasticity and resistance to oxidative stress. Targeting this subpopulation could have disrupted key pathways that facilitated melanoma progression and metastasis. Therefore, understanding the functional characteristics of C0 subpopulation provided valuable insights for developing novel therapeutic strategies against melanoma. Additionally, we constructed a prognostic model associated with these findings. Thus, key prognostic genes were identified, and for this purpose, we analyzed them. What is more, our findings showed that the IGF1 gene was crucial for prognosis, with its expression associated with patient outcomes. Furthermore, experimental validation confirmed the critical role of the IGF1 in the proliferation and invasive metastasis of melanoma. These discoveries contribute to a deeper understanding of the underlying mechanisms of melanoma metastasis, offering new therapeutic avenues and targets for the treatment of melanoma. Besides, they also provide a basis for creating new prognostic models for melanoma diagnosis and prognosis. Furthermore, the outcomes of this study demonstrate crucial clinical implications, particularly for guiding therapy and improving prognostic predictions. Understanding melanoma cell heterogeneity and the role of immune infiltration in disease progression can lead to more personalized treatments and better outcome predictions. Acknowledgements