Data retrieval and processing
We downloaded RNA sequencing data for gene expression of the TCGA-LIHC cohort (in Fragments Per Kilobase Million, FPKM values) and the corresponding clinical information for 371 HCC samples from the XENA. We also downloaded the expression matrix file and clinical information for 115 HCC samples from the GSE76427 cohort (platform GPL10558) from the GEO database. After taking the intersection of genes, we merged the samples from TCGA-LIHC and GSE76427 to construct a meta-cohort. The clinical information of patients in TCGA-LIHC and GES76427 cohort were summarized in Table S1.The batch effects were eliminated by utilizing the "ComBat" function from the "SVA" package.[20]. PCA was used to validate the distribution of the expression profiles of TCGA-LIHC and GSE76427 before and after batch effects(Figure S1B). Samples that lacked complete survival data were excluded, resulting in a meta-cohort comprising 478 HCC samples and 17117 genes.
Acquisition and exploration of the lactate metabolism gene set
We downloaded the lactate metabolism gene set from the GSEA portal in January 2022[21]. We utilized the Gene Set Cancer Analysis (GSCA) database to analyze the mRNA expression profile of 14 Lactate metabolism genes (LMGs) thereby decoding the difference of survival prognosis, clinical staging, and pathway enrichment among them. The "survival" and "survminer" packages were utilized to identify the optimal cutoff point for LMGs in the meta-cohort through the "surv_cutpoint" function. Survival analysis was conducted on LMGs in the meta-cohort. Using the "igraph" R package, a network composed of 13 prognosis-related LMGs was visualized, and the attributes of nodes in the network were determined based on the source of genes and univariate Cox regression analysis. The correlation between genes defined the attributes of the edges.
Cluster analysis
We performed unsupervised clustering using the "ConsensusClusterPlus" R package based on 14 LMGs[22]. The clustering was conducted using the pam algorithm with the Euclidean distance measure, and we resampled 80% of the samples and repeated the process 50 times. The within-group correlation was found to be strong, while the between-group correlation was moderate for k = 2. Subsequently, we performed unsupervised clustering again, based on 163 LMRPGs. The cophenetic coefficient was utilized to determine the optimal rank. Typically, when the value of the cophenetic correlation coefficient begins to decrease, it is considered the optimal factorization rank. Here, the results indicated that the sample partitioning is more stable and reasonable while K = 3. We used the "pheatmap" R package to create a clustering heatmap, illustrating the expression of LMGs, clinical-pathological features, and their relationships with the clustering. Additionally, we conducted survival analysis to compare the overall survival (OS) differences among patients of different subtypes. To get information on immune cell genes, we obtained the "immune.xlsx" file from the literature[23]. Using the "GSVA" R package, we performed ssGSEA enrichment analysis on the integrated cohort samples and generated boxplots depicting the infiltration of 28 different immune cell types among the various patient subgroups
Gene set variation analysis and pathway enrichment analysis
We obtained the h.all.v7.4.symbols.gmt files as reference gene sets from the Molecular Signatures Database (MsigDB) (http://software.broadinstitute.org/gsea/msigdb/). We performed Gene Set Variation Analysis (GSVA) on different subtypes using the "GSVA" R package and generated a heatmap to visualize the analysis results[24]. Additionally, we conducted enrichment analysis on differentially expressed genes (DEGs) and visualized the results using the "ClusterProfiler" package.
Construction of the lactate metabolism score model
We developed a lactate metabolism scoring scheme to quantify the lactate metabolism level in individual patients. Specifically, we conducted principal component analysis (PCA) on LMRPGs expression profiles and then extracted principal components 1 and 2 as the signature scores. Finally, we defined the lactate metabolism score using a formula similar to previous studies: lactate metabolism score = ∑(PC1i + PC2i)[25, 26].
The "ggalluvial" package was utilized for generating Sankey diagrams to visualize the flow comparisons of HCC patients across different clusters, gene clusters, scores, and outcome events. Stacked bar charts, depicting the clinical pathological characteristics of different lactate metabolism score groups, were created to illustrate the percentages of the various components. Copy number variation and single nucleotide variation data of HCC patients were obtained from cBioPortal(cBioPortal for Cancer Genomics) to examine the differences in mutation landscapes between distinct subtypes. The gene mutation waterfall plot was generated using the "maftools" R package. The "spearman" approach was utilized to assess the correlation between immune cells, immune checkpoints, and lactate metabolism scores, visualized respectively through scatter plots and lollipop charts.
Immunotherapy and potential drug therapy
“ESTIMATE” was utilized to assess the immune scores and tumor purity in individuals with HCC. The ICI treatment score data for LIHC was obtained from The Cancer Imaging Archive (TCIA) database. Based on the LMScore, the benefits of PD-1 and CTLA-4 targeted therapies in patients of distinct subtypes were predicted. The TIDE scores for immune dysfunction and exclusion in HCC patients were downloaded from the TIDE database, and the immune evasion and immune exclusion of the two groups were analyzed[27]. MSI can lead to ineffective mismatch repair of DNA, resulting in a series of genetic mutations. The accumulation of these genetic mutations can cause tumor cells to produce a greater number of mutated antigens, triggering a stronger immune response from the immune system. Additionally, the "pRRophetic" package was employed to assess the therapeutic sensitivity and provide potential therapeutic drugs. (p < 0.001)[28].
Collection of liver cancer patient specimens
Tissue samples were collected from liver cancer patients who underwent surgical resection at the First Affiliated Hospital of Zhengzhou University. All specimens were diagnosed as hepatocellular carcinoma. All patients signed informed consent forms.
Cell culture and transfection
The liver cancer cell lines including HUH7, HepG2, HCCLM3, MHCC-97H, and Hep3B were all obtained from the Henan Organ Transplantation Research Center[29] and were cultured using Dulbecco's modified Eagle's medium (DMEM; HyClone/Thermo Fisher Scientific Inc,Switzerland)with 10% fetal bovine serum(FBS; LONSA SCIENCE S.R.L,Switzerland),1% streptomycin and penicillin. HCC cells were transfected with lipo8000 transfection reagent(Beyotime, China). Short hairpin RNA s(shRNA) targeting TTC36 were purchased from PPL company(China), with the following target sequences: (The sequence of TTC36-OE and TTC36-vector was provided in the supplemental document.)
TTC36-sh1, CGAGAAGAAGATGAAGTTT;
TTC36-sh2,GGCCATCTTCAACCCTGACAC;
TTC36-sh3,TCAAGCACAGCTGGAACAGTC;
TTC36-NC, GTTCTCCGAACGTGTCACGTT.
RT-qPCR
Cellular RNA was isolated using TRIzol(Invitrogen, USA), and cDNA was synthesized from the extracted RNA using a cDNA synthesis kit(Vazyme, China). Target gene primers(TsingkeBiotechnologyCo, Ltd., China), SYBR(Vazyme, China), and cDNA were mixed to prepare a 10µl reaction system. The 2-ΔΔCT method was used to calculate the expression level of RNA, with 18s serving as the reference gene. Primer sequences are here:
TTC36-F2: 5′-GGACTCCAAATGATCAGGCAGTG-3′,
TTC36-R2: 5′-GAGGTCCAATCCAACAATGTCTCC-3′;
18S-F: 5′-CGCTTCCTTACCTGGTTGAT-3′,
18S-R: 5′-GAGCGACCAAAGGAACGATA-3′.
Western blotting
Cellular total protein was extracted using RIPA buffer(Solarbio, China), and quantified with the BCA protein assay(seven, China). Electrophoresis was conducted on a 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS‐PAGE). Proteins were transferred onto a PVDF(millipore, USA) membrane at room temperature for a duration of 1 hour, using a constant current of 300 mA. Since the TTC36 protein has an approximate molecular weight of 25 kDa, the PVDF membrane was carefully cut around the corresponding 25 kDa marker. Following one-hour blocking step with skim milk, the PVDF membrane was incubated with the primary antibody at 4℃overnight. The membrane was subsequently washed with TBST and then incubated with a diluted secondary antibody at room temperature for one hour. Protein bands were visualized using an ECL((NCM Biotech, China)) detection reagent and detected with AmershamTM ImageQuantTM 800 (Amersham, UK) and Odyssey® Dlx Imaging System (LI‐COR Biosciences, USA). ImageJ software (NIH) was used to analyze the densitometric data. Antibodies against the following proteins were used as primary antibodies: GAPDH (Proteintech; 10494-1-AP), TTC36 (Abcam; ab122507).The following secondary antibodies were used: HRP‐conjugated anti‐rabbit IgG and anti‐mouse IgG (Cell Signaling Technology, USA).
EDU assay
Cells were seeded in a 96-well plate and incubated overnight in a cell culture incubator after EdU (5-ethynyl-2'-deoxyuridine) labeling. According to the instructions, the EdU detection was performed using the Edu kit (#C0071S, Shanghai Biotech, China). A 1:1000 dilution of the EdU solution was prepared using DMEM culture medium, and then added to the corresponding wells of the pre-planned 96-well plate, 100µl per well. The 96-well plate was incubated in a 37°C cell culture incubator for 4 hours, and then fixed with 4% paraformaldehyde at room temperature for 15 minutes. After completing the corresponding preparation work according to the instructions, cells were added with 50 µl of reaction solution for 30 minutes, followed by nuclear staining with Hoechst for 15 minutes. During the waiting period, we place the aluminum foil-wrapped 96-well plate in a dark area. Cell proliferation was imaged using a 20x fluorescence microscope.
Cell invasion and migration assays
The cells were seeded into six-well plates and cultured overnight. When cells reached 90% confluence, a single line of cells was scraped off in each well using a 100µl plastic pipette tips. Photographs were taken at 0 and 24 hours to record the degree of wound healing. HCC cell line was inoculated in upper Transwell chambers(Corning Costar, USA) with 200µl of DMEM culture medium without serum. The lower chambers were then filled with 500 µl of complete culture medium supplemented with 10% fetal bovine serum. Matrigel(Corning, USA) was applied to the bottom of the upper Transwell chambers to evaluate cell invasion ability. In order to assess cell migration ability, matrigel was not required on the bottom surface of the chambers. After a 24-hour incubation period, the chambers were extracted, fixed with 4% paraformaldehyde for 15 minutes, rinsed, and stained with crystal violet for 30 minutes. Five random fields were chosen for image capture, and cell counting was performed under a microscope.
Lactate measurement.
The transfected cells grown in a 6-well plate were digested using pancreatin(Solarbio, China). After centrifugation at 1000 RPM for 5 minutes, the supernatant was carefully discarded, leaving behind the pellet. The cell pellet was then dissolved in PBS(Solarbio, China) and kept for further use. Subsequently, the cells were disrupted using an ultrasonic homogenizer to obtain the cell lysate. The intracellular lactate levels in the transfected cells were determined using a lactate assay kit (Bestbio, China) and a BCA protein assay kit.
Statistical analysis
Data cleaning, statistical analysis, and visualization work were all conducted using R 4.2.3 and GraphPad Prism 9 software. Spearman correlation analysis was utilized to analyze the relationship between two continuous variables. The Overall Survival (OS) discrepancies between distinct subtypes were evaluated with Kaplan-Meier approach and Log-rank statistical analysis. When comparing two continuous variables, t-test or Wilcoxon rank-sum test was employed to evaluate differences. Pearson chi-square test or Fisher's exact test was used to compare categorical variables. The Cox proportional hazards model along with a 95% confidence interval (95% CI) was employed to assess the hazard ratio (HR). All statistical tests were two-sided, and a p-value below 0.05 was considered statistically significant.