Abstract Renal cell carcinoma is characterized by a poor prognosis. Recently, renal cell carcinoma has been recognized as a metabolic disease associated with fatty acid metabolic reprogramming, although in-depth studies on this topic are still lacking. We found that fatty acid metabolism reprogramming in renal cell carcinoma is primarily characterized by high expression of FABP1. FABP1 + tumors significantly impact survival and display distinct differentiation trajectories compared to other tumor subclusters. They show elevated expression of angiogenesis and cell migration signals, with PLG-PLAT-mediated interactions with endothelial cells notably enhanced. Spatial transcriptomics show a prominent co-localization of FABP1 + tumors with endothelial cells, and their spatial distribution closely aligns with that of PLAT + endothelial cells. FABP1 + tumors exhibit a unique pattern in spatial transcriptomics, enriched in Extracellular Matrix and angiogenesis-related pathways. Through receptor-ligand interaction analysis, a novel PLG-PLAT functional axis was found between tumor epithelial cells and endothelial cells. Based on results of experiments, we infer that FABP1 + tumors can promote plasmin-related tumor angiogenesis by triggering the PLG-PLAT signaling axis. Finally, utilizing preclinical models, we suggest that targeting the FABP1-PLG-PLAT axis may serve as promising strategy enhancing the sensitivity of Tyrosine Kinase Inhibitor therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-025-02377-9. Keywords: Renal cell carcinoma, Fatty acid metabolism, Multi-Omics, FABP1, Tumor angiogenesis Introductions Renal cell carcinoma (RCC), a prevalent malignancy originating from the kidney epithelium, was associated with 431,288 new diagnoses and 179,368 fatalities globally in 2020 [[38]1]. The clear-cell variant of renal cell carcinoma (ccRCC) is the most common type, accounting for roughly 75% of all RCC cases [[39]2]. Patients with ccRCC typically experience a worse prognosis compared to those with other histological types of RCC[[40]3]. RCC has been considered a metabolic disease. Oncogenic drivers—including mutations, epigenetic silencing, and constitutive activation of signaling nodes—reprogram tumor metabolism by hijacking bioenergetic and biosynthetic pathways. Hallmarks include exaggerated glycolytic flux ("Warburg effect"), disrupted TCA cycle intermediates, glutamine anaplerosis, and impaired oxidative phosphorylation, culminating in ATP depletion [[41]4, [42]5]. Critically, these adaptations are coupled with hypoxia-inducible factor (HIF)-driven pseudohypoxia and glutathione-mediated redox buffering, enabling RCC survival under nutrient scarcity and oxidative duress [[43]6]. The association between patients with RCC and obesity suggests that fatty acid metabolic reprogramming plays a critical role in renal cancer [[44]7]. Previous studies have identified certain molecules and pathways associated with fatty acid metabolic reprogramming in RCC; however, a comprehensive understanding of tumor cell metabolic reprogramming remains insufficient [[45]8, [46]9]. Recently, scRNA-sequencing have enabled the investigation of tumorigenesis at the cellular level, while the integration of spatial transcriptomics allows for more systematic spatial characterization of tumor microenvironment interactions. In this study, we explored the fatty acid metabolism reprogramming pattern in RCC through scRNA-seq combined with multi-omics analysis, identifying its biological features as well as interactions with other cells and spatial patterns. Additionlly, based on the results of biological experiments, a PLG-PLAT functional axis was identified between FABP1 + tumor cells and vascular endothelial cell. Plasmin, a key component of the fibrinolytic system, is a serine protease derived from its precursor plasminogen [[47]10]. The two most important plasminogen activators (PAs) in vivo are tissue-type plasminogen activator (t-PA, also known as PLAT) and urokinase-type plasminogen activator (u-PA). t-PA is primarily synthesized and secreted by vascular endothelial cells and is continuously released into the bloodstream, widely distributed in various tissues throughout the body [[48]11]. Previous study have reported that plasmin activates TGF-β, thereby promoting tumor growth and metastasis [[49]12]. As a critical signaling molecule in vascular biological behavior, investigating the dysregulation of the PLG-PLAT axis during tumor progression may provide important insights for cancer treatment. Through this integrated analysis combining single-cell multi-omics with functional biological validation, we will provide valuable insights to aid in the treatment of RCC. Methods Data availability The single-cell sequencing data and spatial transcriptomics sequencing (ST-seq) data used in this study were obtained from the Gene Expression Omnibus (GEO) database, while the bulk-RNA data came from the TCGA database. The bulk RNA data were obtained from the TCGA-KIRC dataset, comprising 541 tumor tissues and 72 normal tissues. The single-cell data were obtained from [50]GSE210038 [[51]13], [52]GSE237429 and [53]GSE242299 [[54]14]. We utilized ST-seq from Series [55]GSE175540 for the analysis of spatial positioning [[56]15] (Supplement Table 1). Patient sample This research was approval from the Ethics Committees of First Affiliated Hospital of Fujian Medical University and adhered to established ethical standards throughout (Table S2). The expression levels of FABP1 in RCC samples were assessed via qRT-PCR. Survival and correlation analyses were performed based on the expression levels. Patients were diagnosed independently by three pathologists. Written informed consents were obtained from all patients before collection of tissues, which were used for and IHC. Cell culture and reagents The human RCC cell strains 786-O and OSRC-2 were sourced from the American Type Culture Collection (Manassas, VA, USA) and propagated following suggested guidelines. All cells were confirmed to be free from mycoplasma contamination and were validated using short tandem repeat (STR) profiling before experimentation. The cells were maintained in MEM (Gibco) supplemented with 10% FBS (Gibco) and incubated at 37 ℃ in a 5% CO₂ humidified environment. For the hypoxia treatment, cells were cultured under 1% O[2] at hypoxia station (Don Whitley Scientific:H35 hypoxystation, UK). Lentivirus constructs and transfection To knock down PLAT, the PLAT-shRNA sequence was cloned into the pLVX-shRNA2 interference vector, while an empty vector served as the negative control. The complete coding sequence of PLG was inserted into the pcDNA3.1 expression vector through digestion with KpnI and BamHI restriction endonucleases. For cellular delivery, the recombinant pcDNA3.1-PLG construct was introduced into target cells employing Lipo Plus transfection reagent under optimized conditions. For lentiviral particle generation, HEK293T cells were co-transfected with the essential packaging plasmids RRE, REV, and VSVG. To establish stable gene expression, target cells were exposed to the custom-produced lentiviral particles for a 24-hour transduction period. Following viral infection, polyclonal cell populations with stable integration were isolated via puromycin-based antibiotic selection. Cell lines expressing luciferase-green fluorescent protein (GFP) were generated by transducing cells with a lentivirus based on the pLEX vector and encoded the luciferase-GFP fusion gene. CRISPR/Cas9 knockout (KO) To generate FABP1 knockout (KO) cell lines, we first validated the functional importance of intronic complementary sequences and their adjacent downstream regions. The gene editing procedure was performed following conventional CRISPR-Cas9 protocols. Specifically, custom-designed single-guide RNAs (sgRNAs) were cloned into the LentiCRISPRv2 backbone (item #52961, puromycin resistance). Lentiviral particles were then produced by co-transfecting HEK293T cells with the LentiCRISPRv2 construct along with packaging plasmids pVSVg (item #8454) and psPAX2 (item #12260). The resulting viral supernatant was used to transduce renal cell carcinoma (RCC) cells, achieving stable FABP1 ablation. scRNA-seq data processing Single-cell data analysis was performed using Seurat v5.0.1. Initial quality control involved filtering cells based on the following criteria: 1) more than 3 expressed cells, 2) fewer than 200 total genes, 3) mitochondrial/ribosomal genes comprising over 20% of total gene expression, 4) fewer than 200 or more than 10,000 detected RNA features, and 5) total molecule counts below 1,000. The "NormalizeData" function was employed to perform library size normalization and obtain normalized counts. Specifically, the "LogNormalize" method adjusted the gene expression measurements for each cell based on total expression, multiplied by a default scaling factor of 10,000, and log-transformed the results. The top 2,000 highly variable genes were identified in the dataset using the "FindVariableFeatures" function. To remove batch effects in the scRNA-Seq data, we utilized "harmony".The 30 top principal components were calculated based on the gene expression profiles of the highly variable genes. We selected an appropriate resolution for the "FindNeighbors" and "FindClusters" functions to perform cell clustering. The "RunUMAP" function was used to visualize the initial clustering. We annotated the cells using the following canonical marker genes: endothelial cells (PECAM1, VWF), neutrophils (NAMPT, FCGR3B), dendritic cells (CD1A, HLA-DRA), T cells (CD3D, CD8A, CD4), NK cells (NKG7, GNLY, CD160), epithelial cells (EPCAM, KRT8, KRT18), fibroblasts (ACTA2, LUM, DCN), mesangial cells (ACTA2, PDGFRB), B cells (CD79A), Mast (TPSAB1, MS4A2, KIT) monocytes/macrophages (CD68, CD14). Using the "FindAllMarkers" function in Seurat, we identified marker genes for each cluster, focusing only on positive markers relative to all other cells. The copy number variation (CNV) analysis The CNV evaluation of the epithelial cell subcluster in the tumor group was conducted using the "infercnv" R package (version 1.18.1) on the chromosome [[57]16]. We selected T cells and epithelial cells from normal adjacent tissue as references, setting the parameters denoise =