Abstract graphic file with name ao4c01215_0018.jpg Previous studies have demonstrated the regulatory roles of Transmembrane protein 147 (TMEM147) in various diseases, including cancer. However, systematic pan-cancer analyses investigating the role of TMEM147 in diagnosis, prognosis, and immunological prediction are lacking. An analysis of data from The Cancer Genome Atlas (TCGA) revealed differential TMEM147 expression across various types of cancer as well as within immune and molecular cancer subtypes. Moreover, high TMEM147 expression was associated with poor disease-specific survival (DSS), overall survival (OS), and progression-free interval (PFI) across cancers, suggesting its potential as a prognostic biomarker. Our study further revealed a significant correlation between TMEM147 expression and T helper cell and Tcm cell infiltration in most cancer types. In the case of liver hepatocellular carcinoma (LIHC), the effect of TMEM147 on prognosis varied among different clinical subtypes. Additionally, functional enrichment analysis revealed an association between TMEM147 and metabolic pathways. Finally, experiments on the MIHA cell line and four LIHC cell lines confirmed the role of TMEM147 in promoting liver cancer cell proliferation, further confirming the clinical value of TMEM147 in liver cancer diagnosis. Our findings suggest that TMEM147 may serve as a diagnostic and prognostic biomarker across cancers while also playing a significant role in LIHC. Introduction Membrane proteins account for a significant proportion of total cellular proteins; these proteins are anchored to cell membranes and play crucial roles in regulating intercellular interactions and cellular interactions with the surrounding environment. Due to their inter- and intracellular biological signaling functions, numerous membrane proteins have been implicated in cancer development, cancer cell metastasis, and immune-related disorders.^[36]1 These poorly characterized transmembrane proteins belong to the Transmembrane protein (TMEM) family, which includes a diverse range of transmembrane proteins that are found in various cellular membranes, such as endoplasmic reticulum (ER), mitochondrial, and lysosomal membranes. TMEM proteins are involved in many biological processes,^[37]2 and TMEM proteins have been demonstrated to be involved in tumor metastasis. For instance, TMEM48 contributes to the invasive metastasis of lung cancer cells,^[38]3 while TMEM206 enhances the metastasis and invasion of colon cancer cells by activating the AKT and FAK pathways.^[39]4 Additionally, TMEM176 expression has been shown to be increased in lymphoma, breast cancer, liver cancer, and other malignancies compared to normal tissues.^[40]5 These findings collectively indicate that TMEM proteins are promising potential biomarkers and prognostic indicators in cancer. TMEM147, which is a member of the TMEM family, is a transmembrane protein that includes seven transmembrane domains. TMEM147 has a molecular mass of 25 kDa and predominantly localizes to the ER membrane and the nuclear envelope. Moreover, TMEM147 is an integral component of the nicalin-TMEM147-NOMO protein complex.^[41]6,[42]7 Based on published studies, dysregulation of TMEM147 expression may contribute to the pathogenesis of various diseases via diverse mechanisms. A clinical study demonstrated that TMEM147 deficiency leads to the upregulation of CKAP4 (CLIMP-63) and RTN4 (NOGO), accompanied by ER redirection. Children with TMEM147 deficiency present consistent clinical features, such as coarse facial features, developmental delays, intellectual disability, and behavioral problems. These findings suggest that biallelic loss-of-function TMEM147 variants cause syndromic intellectual disability, ER translocations, and nuclear tissue dysfunction.^[43]6 TMEM147 is a membrane receptor that interacts with galactose lectin, thereby influencing both host innate immune responses and parasite immune responses by regulating cellular proliferation and phagocytosis. Additionally, TMEM147 mediates the transcription and production of nitric oxide, transforming growth factor β1 (TGF-β1), and interleukin 10 (IL-10).^[44]8 Furthermore, knockdown of TMEM147 substantially decreases the abundance of the nuclear envelope protein lamin B receptor (LBR) and causes its mislocalization within the ER. Additionally, TMEM147 physically interacts with DHCR7, which is an enzyme that is crucial for cholesterol biosynthesis; thus, TMEM147 is an important novel regulator of intracellular cholesterol homeostasis.^[45]9 Moreover, TMEM147 is a major activator of NF-κB in chondrocytes and is expressed at elevated levels in chondrocytes from individuals with rheumatoid arthritis (RA); these findings suggest its involvement in the pathogenesis of RA.^[46]10 Furthermore, TMEM147 exhibits differential expression patterns in colon cancer and osteosarcoma, making it a potential tumor biomarker.^[47]2,[48]11 Despite reports on the role of TMEM147 in diverse physiopathological processes, the functional characteristics of TMEM147 remain poorly understood; in particular, systemic analyses across cancers are lacking. By integrating data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases, our recent study revealed differential expression of TMEM147 in more than 20 distinct types of cancer. This differential expression was evident even among various molecular or immune subtypes within the same type of cancer, suggesting that TMEM147 may serve as a pan-cancer biomarker. Furthermore, analyses of the associations between TMEM147 expression and overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) revealed that elevated TMEM147 expression is associated with unfavorable prognosis. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and GSEA analyses were performed to determine shared biological pathways and to conduct enrichment analysis of the differentially expressed genes. Subsequently, we focused on liver hepatocellular carcinoma (LIHC) and revealed correlations between TMEM147 expression and various clinical characteristics. Finally, experiments on liver cell lines were conducted to confirm the role of TMEM147 in liver cancer. The current investigation of the role of TMEM147 across cancers will undoubtedly provide additional evidence to facilitate subsequent clinical translation. Materials and Methods Data Preprocessing and Gene Expression Analysis Transcriptional RNA sequencing (RNA-Seq) expression profiles and the corresponding clinical information for 33 types of cancer were obtained from the TCGA database sharing data portal ([49]https://portal.gdc.cancer.gov/) and the Genotype-Tissue Expression (GTEx) database ([50]https://commonfund.nih.gov/GTEx) from UCSC Xena ([51]https://xenabrowser.net/datapages/). The data were transformed to transcripts per kilobase million (TPM) via the Toil process.^[52]12 R software v3.6.3 was used for statistical analysis, and the ggplot2 package was used for visualization. The Wilcoxon signed rank test was used to analyze differences among all the transcriptomic data, and P < 0.05 was considered to indicate statistical significance (ns, P ≥ 0.05; *P < 0.05; **P < 0.01; ***P < 0.001). Diagnostic Value Analysis Receiver operating characteristic (ROC) curves were used to assess the diagnostic value of TMEM147 in 33 types of cancer, and the pROC package was used for analysis. The area under the curve (AUC) ranged from 0.5 to 1.0; 0.5–0.7 indicated low accuracy, 0.7–0.9 indicated medium accuracy, and 0.9 or higher indicated high accuracy. Analysis of TMEM147 Expression in Different Immunological and Molecular Subtypes of Human Cancers The TISIDB database ([53]http://cis.hku.hk/TISIDB/index.php), which is a web portal for information on tumor and immune system interactions that integrates multiple heterogeneous data types, was used to determine the relationships between TMEM147 expression and immune or molecular subtypes across cancers. Results with a P value <0.05 were considered statistically significant, and they are shown in [54]Results. Interaction Network Analysis and Functional Clustering Analysis The String ([55]https://string-db.org/) database was used to construct a protein–protein interaction (PPI) network of TMEM147. The network was constructed by searching for the protein name of TMEM147 in Homo sapiens and setting the main parameters to medium confidence and active interaction sources. Cytoscape (v3.9.1) was used for visualization. GO and KEGG enrichment analyses were conducted with the clusterProfiler package (v3.14.3), and the results were visualized with the ggplot2 package.^[56]13 The KEGG pathways and GO_BP terms were examined using the gene set enrichment analysis (GSEA) function module. Prognosis Analysis The relationships between TMEM147 and OS and between DSS and the PFI were investigated using Kaplan–Meier plots. The Survminer package (v0.4.9) was used for visualization, and the survival package was used for statistical analysis of survival data.^[57]14 All the data were obtained from the TCGA database in HTSeq-FPKM format and transformed into the TPM format. Clinical Correlation Analysis in LIHC Data from the TCGA-LIHC database in the level 3 HTSeq-fragments per kilobase per million (FPKM) format were downloaded from the TCGA database, converted to the transcripts per million reads (TPM) format, and then analyzed after log2 transformation. Ggplot2 (v3.3.3) was used for visualization. Different clinical characteristics related to LIHC, such as age, weight, AFP (ng/mL), and Child–Pugh score, were included. Coexpression Analysis and Enrichment Analysis of TMEM147 in LIHC Genes that were coexpressed with TMEM147 were identified by R based on the expression profiles of the TCGA cohort. The top 50 genes that were positively and negatively related to TMEM147 in LIHC were identified, and they are shown. Ggplot2 was used for visualization. Then, we used all of the genes that were coexpressed with TMEM147 in LIHC for enrichment analysis. Analysis of Immune Cell Infiltration across Cancer Types and in LIHC The markers of 24 kinds of immune cells were confirmed^[58]15 using the ssGSEA immune infiltration algorithm and GSVA package to determine the immune profiles across cancer types and in LIHC.^[59]16 Cell Culture The cell lines that were used in this study included the MIHA, BEL-7402, SMMC 7721, HepG2, and Huh-7 cell lines. MIHA cells were cultured in 1640 medium (Gibco, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco, USA). The remaining cell lines were cultured in DMEM (Gibco, USA) supplemented with 10% FBS. All the cell cultures were maintained in a humidified incubator at 37 °C with 5% CO[2]. Subculturing was routinely performed when the cells reached 80–90% confluence using trypsin-EDTA (Gibco, USA). Cell viability was assessed during each passage using the trypan blue exclusion assay (Gibco, USA). Real-Time PCR and Western Blotting Real-time PCR and western blotting were used to analyze gene expression. For real-time PCR, cDNA was synthesized by reverse transcription, followed by amplification with gene-specific primers. The sequences of the synthetic primers are shown in [60]Table [61]1 . Western blotting analysis included protein extraction, protein separation by SDS–PAGE, protein transfer to PVDF membranes, and protein detection using primary antibodies against the proteins of interest. The utilization of these established methodologies ensured the reliable and accurate evaluation of gene expression in our study. Table 1. Sequences of Synthetic Primers and siRNAs. gene sequence Homo-TMEM147 forward: 5′- CGGGGCATTGAGTTTGACTG-3′; reverse: 3′- AGACGCGACGATGTAATGGA-5′ siRNA-1 forward: 5′-CCUACUUCAUCACCUACAATT-3′; reverse: 3′- UUGUAGGUGAUGAAGUAGGTT-5′ siRNA-2 forward: 5′-GCAGACCUGAUAGGUCUAATT-3′; reverse: 3′- UUAGACCUAUCAGGUCUGCTT-5′ siRNA-3 forward: 5′- CCACUGCUGAGCUUAUUAUTT-3′; reverse: 3′- AUAAUAAGCUCAGCAGUGGTT-5′ siRNA-4 forward: 5′- GUGUCUACAAGGCCUUUGUTT-3′; reverse: 3′- ACAAAGGCCUUGUAGACACTT-5′ [62]Open in a new tab Cell Colony Formation Assay HepG2 and Huh-7 cells were cultured in DMEM supplemented with 10% FBS and 1% penicillin–streptomycin. siRNAs were purchased from Biosune, and the sequences of the siRNAs are shown in [63]Table [64]1 . The experiment was performed in six-well plates, and 4% paraformaldehyde and a 0.1% crystal violet staining solution were used in this experiment. Huh-7 and HepG2 cells were seeded at a density of 2 × 10^4 or 2 × 10^5 cells per well, respectively, and cultured for 16 h. The cells were then transfected with siRNAs for 24 h using Lipofectamine 2000. The cells were subsequently fixed with 4% paraformaldehyde, stained with 0.1% crystal violet, and observed under a microscope. CCK-8 Assay Following siRNA transfection, the cells were incubated for 24, 72, 96, or 120 h. At each time point, the culture medium was replaced with fresh DMEM supplemented with CCK-8 reagent (Cell Counting Kit-8), and the cells were incubated for an additional 2 h at 37 °C in a humidified incubator. After the incubation period, the absorbance of the formazan dye, which is directly proportional to the number of viable cells, was measured at 450 nm using a microplate reader. EdU Assay After 72 h of siRNA transfection, the proliferation of the cells was assessed using the Click-iT EdU Alexa Fluor 488 Imaging Kit. Briefly, the cells were labeled with EdU, which is a nucleoside analog that is incorporated into newly synthesized DNA during active DNA replication. The cells were incubated with EdU labeling medium for 2 h at 37 °C. Following incubation, the cells were fixed with 4% paraformaldehyde and permeabilized with 0.3% Triton X-100. The incorporated EdU was then detected by a Click-iT reaction with Alexa Fluor 488 dye. Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (DAPI). Results TMEM147 Expression Levels Were Increased across Various Cancer Tissues The expression profiles of TMEM147 across many cancer types in the TCGA database were analyzed. As shown in [65]Figure [66]1 A, among all 18 types of cancer that were investigated, TMEM147 was significantly more highly expressed in 13 types of cancer—bladder urothelial carcinoma (BLCA), breast invasive carcinoma (BRCA), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), esophageal carcinoma (ESCA), kidney renal papillary cell carcinoma (KIRP), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), prostate adenocarcinoma (PRAD), rectal adenocarcinoma (READ), stomach adenocarcinoma (STAD), and uterine corpus endometrial carcinoma (UCEC)—than in cancer-adjacent normal tissues. Data from the GTEx database were used as controls, and TMEM147 was also found to be highly expressed in 27 out of 33 cancer types ([67]Figure [68]1B). Figure 1. [69]Figure 1 [70]Open in a new tab Expression level of the TMEM147 gene in tumor tissues and normal tissues. (A) TMEM147 expression in tumor tissues and adjacent normal tissues from the TCGA database; (B) TMEM147 expression in tumor tissues and normal tissues from the TCGA database when data from the GTEx database were used as controls (*P < 0.05, ** P < 0.01, *** P < 0.001). TMEM147 Expression in Different Molecular or Immune Subtypes across Types of Cancer To further explore the potential role of TMEM147 across cancers, we used data from the TISIDB database and found that TMEM147 was differentially expressed in different molecular subtypes of nine cancers and in different immune subtypes of 10 cancers. Among the molecular subtypes, TMEM147 was expressed at the highest levels in the CIMP-high subtype (P = 2.99 × 10^–04) of ACC ([71]Figure [72]2 A), in the basal subtype (P = 6.61× 10^–15) of BRCA ([73]Figure [74]2B), in the classical subtype (P = 2.19 × 10^–03) of HNSC ([75]Figure [76]2C), in the C2b subtype (P = 2.19 × 10^–03) of KIRP ([77]Figure [78]2D), in the classic-like subtype (P = 8.2 × 10^–55) of LGG ([79]Figure [80]2E), in the iCluster subtype (P = 1.03 × 10^–02) of HIHC ([81]Figure [82]2F), in the ortical admixture subtype (P = 2.33 × 10^–03) of PCPG ([83]Figure [84]2G), in the HM-SNV subtype (P = 3.74 × 10^–05) of STAD ([85]Figure [86]2H), and in the CN-HIGH subtype (P = 1.89 × 10^–03) of UCEC ([87]Figure [88]2I). Figure 2. [89]Figure 2 [90]Open in a new tab Correlations between TMEM147 expression and molecular subtypes across TCGA tumors. (A) ACC; (B) BRCA; (C) HNSC; (D) KIRP; (E) LGG; (F) LIHC; (G) PCPG; (H) STAD; (I) UCEC. Furthermore, regarding the immune subtypes, the expression of TMEM147 was related to different immune subtypes (C1: wound healing; C2: IFN-γ-dominant; C3: inflammatory; C4: lymphocyte-depleted; C5: immunologically quiescent; C6: TGF-b-dominant) of 10 cancers, including BRCA ([91]Figure [92]3 A), KIRC ([93]Figure [94]3B), KIRP ([95]Figure [96]3C), LIHC ([97]Figure [98]3D), LIAD ([99]Figure [100]3E), PRAD ([101]Figure [102]3F), LGG ([103]Figure [104]3G), STAD ([105]Figure [106]3H), TGCT ([107]Figure [108]3I), and SKCM ([109]Figure [110]3J). Figure 3. [111]Figure 3 [112]Open in a new tab Correlations between TMEM147 expression and immune subtypes across tumors from the TCGA database. (A) BRCA; (B) KIRC; (C) KIRP; (D) LIHC; (E) LUAD; (F) PRAD; (G) LGG; (H) STAD; (I) TGCT; (J) SKCM. High Diagnostic Value of TMEM147 across Cancers To confirm the diagnostic value of TMEM147 across cancers, we generated ROC curves using TCGA data. The ROC curves showed high predictive values (ROC > 0.7) of TMEM147 in 15 types of cancer, including READ (AUC = 0.941; CI = 0.916–0.966), STAD (AUC = 0.786; CI = 0.750–0.822), DLBC (AUC = 0.955; CI = 0.933–0.977), LUSC (AUC = 0.809; CI = 0.780–0.838), PRAD (AUC = 0.817; CI = 0.780–0.855), UCEC (AUC = 0.841; CI = 0.794–0.887), BLCA (AUC = 0.877; CI = 0.813–0.941), CHOL (AUC = 1.000; CI = 1.000–1.000), LUAD (AUC = 0.778; CI = 0.747–0.808), LIHC (AUC = 0.908; CI = 0.880–0.937), COAD (AUC = 0.934; CI = 0.911–0.958), BRCA (AUC = 0.915; CI = 0.898–0.932), ESCA (AUC = 0.837; CI = 0.685–0.990), LAML (AUC = 0.933; CI = 0.981–1.000), and PAAD (AUC = 0.936; CI = 0.908–0.964) ([113]Figure [114]4 A–O). Figure 4. [115]Figure 4 [116]Open in a new tab Receiver operating characteristic (ROC) curve for TMEM147 expression across cancers. (A) READ; (B) STAD; (C) DLBC; (D) LUSC; (E) PRAD; (F) UCEC; (G) BLCA; (H) CHOL; (I) LUAD; (J) LIHC; (K) COAD; (L) BRCA; (M) ESCA; (N) LAML; (O) PAAD. TMEM147 Expression Was Correlated with the Prognosis of Patients with Different Cancers To determine the prognostic role of TMEM147 across cancers, Kaplan–Meier analysis was performed, and the results showed that higher expression of TMEM147 predicted a lower probability of survival among patients with LICH ([117]Figure [118]5 A–C), LGG ([119]Figure [120]5D–F), ACC ([121]Figure [122]5G–I), and MESO ([123]Figure [124]5J–L). The TMEM147 expression level was correlated with the OS, DSS, and PFI of patients with these types of cancer. Figure 5. [125]Figure 5 [126]Open in a new tab Correlations between TMEM147 expression and cancer patient prognosis (OS, DSS, and PFI). (A–C) LIHC; (D–F) LGG; (G–I) ACC; (J–L) MESO. TMEM147 Expression Was Correlated with Immune Cell Infiltration in Tumors Infiltrating immune cells can recognize and attack cancer cells to suppress tumor growth and metastasis. Activation of the immune system helps to control tumor development and results in better prognosis. In our study, we investigated the relationship between TMEM147 expression and infiltrating immune cells. The heatmap showed that in most tumors, the expression of TMEM147 was negatively correlated with the infiltration of T helper cells and Tcm cells ([127]Figure [128]6 A). Immune checkpoint-related genes play a role in regulating the antitumor immune response and affect the immune escape and progression of tumors in patients with liver cancer. Typically, immune checkpoint molecules balance the immune response and prevent overactivated immune cells from damaging normal tissues. We studied the relationships between the expression of six immune checkpoint genes (CD274, CTLA4, HAVCR2, LAG3, PDCD1, PDCD1LG2, and TIGIT) and TMEM147. Our results showed that TMEM147 expression was almost negatively correlated with immune checkpoint-related gene expression in BLCA, OV, SKCM, and THCA and positively correlated with immune checkpoint gene expression in LGG and LIHC, suggesting that TMEM147 is related to immune escape in cancer ([129]Figure [130]6B). Figure 6. [131]Figure 6 [132]Open in a new tab Correlation analysis of TMEM147 expression and immune cell infiltration across cancers. (A) Heatmap of the correlation of TMEM147 expression with 24 types of tumor-infiltrating immune cells. (B) Correlation analysis of TMEM147 expression and immune checkpoint expression in pan-cancer tissues. Gene Functional Enrichment Analysis of TMEM147 across Cancers To investigate the mechanism by which TMEM147 functions during tumorigenesis, PPI network analysis was performed by utilizing the STRING database and Cytoscape. As shown in [133]Figure [134]7 A, an interaction network of 50 TMEM147-binding proteins that were identified by experimental evidence was visualized. GO enrichment analysis of the 50 TMEM147-binding proteins was conducted ([135]Figure [136]7B), and the results showed that 19 pathways were enriched, including the structural constituent of ribosome, rRNA binding, protein transporter activity, protein transmembrane transporter activity, ubiquitin-protein transferase regulator activity, 5S rRNA binding, ribosome, cytosolic part, ribosomal subunit, large ribosomal subunit, cytosolic ribosome, cytosolic large ribosomal subunit, protein targeting, protein localization to endoplasmic reticulum, establishment of protein localization to membrane, protein targeting to membrane, cotranslational protein targeting to the membrane, mRNA catabolic process, nuclear-transcribed mRNA catabolic process, translational initiation, viral gene expression, viral transcription, nuclear-transcribed mRNA catabolic process, and nonsense-mediated decay ([137]Figure [138]7C). KEGG pathway enrichment analysis revealed that the most highly enriched pathways were ribosome, phagosome, Vibrio cholerae infection, and protein export. Figure 7. [139]Figure 7 [140]Open in a new tab PPI network, GO analysis, and KEGG analysis of 50 TMEM147-binding proteins. (A) PPI network; (B) visual network of GO and KEGG analyses; (C) GO analysis; (D) KEGG analysis. High TMEM147 Expression Was Correlated with Poor Prognosis in Patients with Different Clinical Characteristics According to the results of the pan-cancer study described above, TMEM147 was differentially expressed in liver cancer and may be used as a biomarker for liver cancer; therefore, we conducted a more in-depth study on LIHC. The higher expression of TMEM147 was associated with worse OS in patients with the following characteristics: age ≤ 60 years ([141]Figure [142]8 A); N stage: N0 ([143]Figure [144]8B); M stage: M0 ([145]Figure [146]8C); AFP (ng/mL): 400 ([147]Figure [148]8D); sex: male ([149]Figure [150]8E); tumor status: positive ([151]Figure [152]8F); height ≥ 170 ([153]Figure [154]8G); race: Asian ([155]Figure [156]8H); histological grade: G3 and G4 ([157]Figure [158]8I). The higher expression of TMEM147 was associated with worse DSS in patients with the following characteristics: tumor status: positive ([159]Figure [160]9A); race: Asian ([161]Figure [162]9B); pathological M stage: M0 ([163]Figure [164]9C); pathological N stage: N0 ([165]Figure [166]9D); sex: male ([167]Figure [168]9E); histological type: hepatocellular carcinoma ([169]Figure [170]9F). The higher expression of TMEM147 was associated with worse PFI in patients with the following characteristics: M stage: M0 ([171]Figure [172]10A); tumor status: positive ([173]Figure [174]10B); race: Asian ([175]Figure [176]10C); pathological N stage: N0 ([177]Figure [178]10D); age ≤ 60 years ([179]Figure [180]10E); height ≥ 170 ([181]Figure [182]10F). Figure 8. [183]Figure 8 [184]Open in a new tab Associations between TMEM147 expression and OS in different clinical subgroups of patients with LIHC. (A) Age > 60; (B) N stage: N0; (C) M stage: M0; (D) AFP (ng/mL): >400; (E) sex: male; (F) tumor status: positive; (G) height: >170; (H) race: Asian; (I) histological grade: G3 and G4. Figure 9. [185]Figure 9 [186]Open in a new tab Associations between TMEM147 expression and DSS in different clinical subgroups of LIHC patients. (A) Tumor status: positive; (B) race: Asian; (C) pathological M stage: M0; (D) pathological N stage: N0; (E) sex: male; (F) histological type: hepatocellular carcinoma. Figure 10. [187]Figure 10 [188]Open in a new tab Associations between TMEM147 expression and the PFI in different clinical subgroups of LIHC patients. (A) M stage: M0; (B) tumor status: positive; (C) race: Asian. (D) pathological N stage: N0. (E) Age ≤ 60 years; (F) height ≥ 170. TMEM147 Expression Was Correlated with the Clinicopathological Characteristics of LICH Patients The associations between the expression of TMEM147 and the clinical characteristics of LICH patients were explored, and the results are shown in [189]Table [190]2 . AFP (ng/mL) (P < 0.001), BMI (P = 0.019), weight (P = 0.002), OS (P = 0.002), T stage (P = 0.005), pathological stage (P = 0.004), and histological grade (P < 0.001) were significantly related to TMEM147 expression. Table 2. Clinical Characteristics of LIHC Patients. characteristic low expression of TMEM147 high expression of TMEM147 P value n 187 187 tumor status, n (%) 0.163 tumor-free 110 (31%) 92 (25.9%) with tumor 71 (20%) 82 (23.1%) AFP (ng/mL), n (%) <0.001 ≤400 123 (43.9%) 92 (32.9%) >400 20 (7.1%) 45 (16.1%) BMI, n (%) 0.019 ≤25 77 (22.8%) 100 (29.7%) >25 91 (27%) 69 (20.5%) weight, n (%) 0.002 ≤70 78 (22.5%) 106 (30.6%) >70 96 (27.7%) 66 (19.1%) OS event, n (%) 0.002 alive 137 (36.6%) 107 (28.6%) dead 50 (13.4%) 80 (21.4%) T stage, n (%) 0.005 T1 108 (29.1%) 75 (20.2%) T2 41 (11.1%) 54 (14.6%) T3 30 (8.1%) 50 (13.5%) T4 6 (1.6%) 7 (1.9%) pathological stage, n (%) 0.004 stage I 103 (29.4%) 70 (20%) stage II 38 (10.9%) 49 (14%) stage III 34 (9.7%) 51 (14.6%) stage IV 1 (0.3%) 4 (1.1%) histological grade, n (%) <0.001 G1 35 (9.5%) 20 (5.4%) G2 102 (27.6%) 76 (20.6%) G3 43 (11.7%) 81 (22%) G4 5 (1.4%) 7 (1.9%) age, median (IQR) 62 (54, 69) 60 (50, 68) 0.123 [191]Open in a new tab Moreover, the results of Welch one-way ANOVA revealed that TMEM147 was highly expressed in the tumor-positive group ([192]Figure [193]11 A), in the AFP > 400 ng/mL group ([194]Figure [195]11B), in the BMI ≤ 25 group ([196]Figure [197]11C), in the weight ≤70 kg group ([198]Figure [199]11D), in the Dead group ([200]Figure [201]11E), in the T2 stage group ([202]Figure [203]11F), and in the G2, G3, and G4 groups ([204]Figure [205]11G) compared with the control group. Figure 11. [206]Figure 11 [207]Open in a new tab Associations between TMEM147 expression and different clinical characteristics in LIHC patients. (A) Tumor status; (B) AFP; (C) BMI; (D) weight; (E) OS; (F) T stage; (G) pathological stage; (H) histological grade. ns, P ≥ 0.05; *P < 0.05; **P < 0.01; ***P < 0.001. Analysis of Molecules that Were Correlated with TMEM147 in LIHC Patients Using data from the TCGA database, we identified 50 genes that were most highly positively or negatively related to TMEM147 in LIHC. [208]Figure [209]12 A shows the gene coexpression heatmap of the top 50 positively correlated genes, and the scatter plot shows that CNPAS1 (r = 0.740) ([210]Figure [211]12B), RFXANK (r = 0.742) ([212]Figure [213]12C), SMG9 (r = 0.718) ([214]Figure [215]12D), EIF3K (r = 0.712) ([216]Figure [217]12E), TOMM40 (r = 0.730) ([218]Figure [219]12F), SNRPD2 (r = 0.768) ([220]Figure [221]12G), PPP2R1A (r = 0.745) ([222]Figure [223]12H), GSK3A (r = 0.719) ([224]Figure [225]12I), TMM50 (r = 0.744) ([226]Figure [227]12J), and UBE2M ([228]Figure [229]12K) (r = 0.750) were the top 10 positively related genes. Moreover, the heatmap of negatively related genes ([230]Figure [231]13A) revealed the top 10 genes, which included ABAT (r = −0.617) ([232]Figure [233]13B), C8A (r = −0.614) ([234]Figure [235]13C), DMGDH (r = −0.587) ([236]Figure [237]13D), EHHADH (r = −0.600) ([238]Figure [239]13E), CYP8B1 (r = −0.581) ([240]Figure [241]13F), F11 (r = −0.584) ([242]Figure [243]13G), PFKFB1 (r = −0.565) ([244]Figure [245]14H), GLYATL1 (r = −0.587) ([246]Figure [247]13I), ACSM2A (r = −0.603) ([248]Figure [249]13J), and GYS2 (r = −0.647) ([250]Figure [251]13K). Figure 12. [252]Figure 12 [253]Open in a new tab Top 50 genes that were positively correlated with TMEM147 expression in LIHC. (A) Gene coexpression heatmap of the top 50 genes that were positively correlated with TMEM147 in LIHC; (B–K) correlation analysis of the top 10 genes that were positively correlated with TMEM147 in the heatmap. Figure 13. [254]Figure 13 [255]Open in a new tab Top 50 genes that were negatively correlated with TMEM147 expression in LIHC. (A) Gene coexpression heatmap of the top 50 genes that were negatively correlated with TMEM147 in LIHC; (B–K) correlation analysis of the top 10 genes that were negatively correlated with TMEM147 in the heatmap. Figure 14. [256]Figure 14 [257]Open in a new tab PPI network construction and GO and KEGG analyses of DEGs between LIHC samples with high TMEM147 expression and LIHC samples with low TMEM147 expression. (A) Volcano map of DEGs (red: upregulated; blue: downregulated); (B) GSEA; (C, D) GO and KEGG analyses of DEGs. Differentially Expressed Genes (DEGs) Between LIHC Samples with High TMEM147 Expression and LIHC Samples with Low TMEM147 Expression To elucidate gene expression profiles, ggplot2 was used to identify sets of genes that were significantly up- and downregulated. Using |log (fold change)| > 1 and adjusted P value <0.05 as cutoff criteria, a total of 3818 upregulated genes and 1408 downregulated genes were identified ([258]Figure [259]14 A). Then, we conducted GSEA and GO and KEGG enrichment analyses ([260]Figure [261]14B–D) of the DEGs, and the GSEA results revealed that genes that are related to arachidonic acid metabolism, butanoate metabolism, drug metabolism, cytochrome p450, drug metabolism, other enzymes, fatty acid metabolism, and metabolism of xenobiotics by cytochrome p450 were enriched ([262]Figure [263]14B). GO analysis revealed that the fatty acid catabolic process, fatty acid oxidation, lipid oxidation, regulation of chromosome segregation, lipid modification, membrane lipid metabolic process, branched-chain amino acid metabolic process, lipid catabolic process, sphingolipid metabolic process, membrane lipid biosynthetic process, and fatty acid metabolic process were enriched in BP, the chromosomal region was enriched in CC, and transferase activity, transferring acyl groups, coenzyme binding, and fatty acid binding were enriched in MF ([264]Figure [265]14C). The KEGG pathway enrichment showed that the DEGs were mainly related to propanoyl-CoA metabolism, propanoyl-CoA, beta-oxidation, and fatty acid degradation ([266]Figure [267]14D). Analysis of the Association between TMEM147 Expression and Immune Cell Infiltration in LIHC To better confirm the association between immune cell infiltration and TMEM147 expression in LIHC, the ssGSEA immuno-infiltration algorithm in GSVA was used. The Wilcoxon rank sum test showed that TH2 cells, TFH cells, and NK CD56 bright cells were significantly positively correlated with the expression of TMEM147 (P < 0.001) ([268]Figure [269]15 A). The scatter plots in [270]Figure [271]15B show that Th2 cells (r = 0.318, P < 0.001), TFH cells (r = 0.285, P < 0.001), and NK CD56 bright cells (r = 0.280, P < 0.001) show this positive correlation, but no significant associations were observed for macrophages, activated dendritic cells (aDCs), or Th1 cells. Moreover, Th17 cells, Tcm cells, neutrophils, Tregs, DCs, and cytotoxic cells were significantly negatively associated with TMEM147 expression ([272]Figure [273]16A). The scatter plots in [274]Figure [275]16B show this negative correlation of Th17 cells (r = 0.275, P < 0.001), Tcm cells (r = 0.260, P < 0.001), neutrophils (r = 0.217, P < 0.001), Tregs (r = 0.185, P < 0.001), DCs (r = 0.182, P < 0.001), and cytotoxic cells (r = 0.144, P = 0.005) with TMEM147 expression. Figure 15. [276]Figure 15 [277]Open in a new tab Analysis of the association of immune cell infiltration with TMEM147 expression in LIHC revealed 6 immune cell types that were positively related to TMEM147 expression. (A) Enrichment scores of TMEM147 expression in Th2 cells, TFH cells, NK CD56 bright cells, macrophages, aDCs, and Th1 cells. Correlation between TMEM147 expression and Th2 cells, TFH cells, NK CD56 bright cells, macrophages, aDCs, and Th1 cells (B); ns, P ≥ 0.05; *P < 0.05; **P < 0.01; ***P < 0.001. Figure 16. [278]Figure 16 [279]Open in a new tab Analysis of the association of immune cell infiltration with TMEM147 expression in LIHC revealed 6 immune cell types that were positively related to TMEM147 expression. (A) Enrichment scores of TMEM147 expression in Th17 cells, Tcm cells, neutrophils, Tregs, DCs, and cytotoxic cells. (B) Correlation between TMEM147 expression and Th17 cells, Tcm cells, neutrophils, Tregs, DCs, and cytotoxic cells; ns, P ≥ 0.05; *P < 0.05; **P < 0.01; ***P < 0.001. TMEM147 Promoted Cell Proliferation in Liver Cancer PCR and western blotting experiments revealed that TMEM147 was overexpressed in liver cancer cells, with higher expression levels observed in HepG2 and Huh-7 cells ([280]Figure [281]17 A,B). Subsequent investigations were performed using the HepG2 and Huh-7 cell lines. Four siRNAs were designed and used to knock down TMEM147 expression in the cells, and the results demonstrated that siRNA-4 most effectively knocked down TMEM147 expression in HepG2 cells, while siRNA-3 had the greatest effect on Huh-7 cells ([282]Figure [283]17C). CCK-8 and colony formation assays indicated a significant reduction in cell proliferation after TMEM147 knockdown ([284]Figure [285]17D,E). Moreover, EDU experiments demonstrated that downregulation of TMEM147 reduced DNA replication in cells, thereby affecting cell proliferation ([286]Figure [287]17F). Figure 17. [288]Figure 17 [289]Open in a new tab TMEM147 promoted cell proliferation in liver cancer. (A, B) TMEM147 is overexpressed in liver cancer cells. (C) siRNA was used to knock down TMEM147 expression in HepG2 and Huh-7 liver cancer cells. (D) The results of the CCK-8, (E) colony formation, and (F) EdU assays were obtained. Discussion Cancer is a major public health problem worldwide, and it is the second leading cause of death in the United States.^[290]17 Researchers have continuously worked to develop treatments for cancer. Currently, there are many methods and approaches for treating tumors, such as surgery, radiotherapy, chemotherapy, biological therapy, and immunotherapy; each of these methods is efficacious and has its own advantages. However, investigations of tumor biomarkers are still very limited, and further research is warranted. The TMEM family is a group of proteins that span the entire width of the lipid bilayer and are permanently anchored in membranes. Many TMEMs act as channels, allowing the transport of specific substances across biological membranes.^[291]18 Because they allow specific substances to be transported through biofilms, facilitate communication between intracellular and extracellular spaces, and perform other unknown biological functions, TMEM family members play important roles in the migration, metastasis, and progression of a variety of tumors.^[292]19,[293]20 TMEM45A and TMEM205 have been implicated in tumor progression and invasion but also in tumor chemoresistance.^[294]21 Few studies have systematically explored the expression of TMEM147, which is a member of the TMEM family, across cancers via bioinformatics. This study aimed to investigate the role of TMEM147 across cancers and to use LIHC as a specific example to illustrate the potential value of TMEM147. In the present study, by using high-throughput RNA sequencing data, we found that TMEM147 is differentially expressed across cancers, and ROC curves of TMEM147 expression in 15 kinds of cancer revealed a high AUC, indicating that TMEM147 might be a potential diagnostic biomarker. The heterogeneity or intrinsic diversity of cancer is the main reason why cancers can adapt to pharmacological pressures and then evolve;^[295]22 for instance, LIHC is a heterogeneous disease at both the molecular and histological levels.^[296]23 Heterogeneity is a major obstacle to the development of effective targeted therapies, and studying the heterogeneity of tumors can help clinicians implement individualized and differential treatments for patients.^[297]24,[298]25 The subtype analysis in our study revealed that TMEM147 was differentially expressed in many molecular and immune subtypes of cancer, which may provide directions for treatment in the context of tumor heterogeneity. Notably, TMEM147 is closely correlated with both molecular and immune subtypes in BRCA, KIRP, LGG, LIHC, and STAD. Furthermore, Kaplan–Meier survival curves confirmed the high prognostic value of TMEM147 among patients with LIHC, LGG, ACC, and MESO. This result shows that TMEM147 could be a biomarker for the prognosis of patients with different kinds of cancer. To further understand the role of TMEM147 across cancers, enrichment analysis of 50 TMEM147-binding proteins was performed. The results revealed that ribosome-related functions and membrane protein-related functions, which are associated with the transmembrane protein activity of TMEM147, were highly enriched. These results demonstrated the potential role of TMEM147 across cancers. Among all malignant diseases, liver cancer is the fourth most common cause of cancer-related deaths worldwide and ranks sixth in terms of incidence.^[299]26 Viral hepatitis, alcoholism, and fungal toxin exposure are major risk factors for the development of liver cancer, but in recent years, obesity, insulin resistance, and type 2 diabetes-associated fatty liver disease (NAFLD) have been recognized as triggers of liver cancer, especially in developed countries.^[300]27 Despite its prevalence and high mortality rate, reliable treatments to reduce mortality are still scarce,^[301]28 and predictive biomarkers are urgently needed to inform the selection of molecular and immune-based treatments in addition to surgery, liver transplantation, and local ablative and intra-arterial therapies.^[302]29 In addition to poor prognosis of patients with different kinds of cancers, multiple clinical subgroups of patients with LIHC who also had high TMEM147 expression had poor DSS and PFI. Moreover, the expression of TMEM147 differed according to different clinical characteristics. More importantly, in the functional analysis, the KEGG, GO, and GSEA analyses revealed enrichment of metabolism-related pathways, such as fatty acid metabolism, arachidonic acid metabolism, lipid modification, and membrane lipid metabolic processes. Abnormal metabolic processes, including abnormal levels of lipid metabolism, can affect tumor metastasis, recurrence, and drug resistance and induce tumor immune evasion by influencing the antitumor response of immune cells.^[303]30 In the tumor microenvironment, T cells, macrophages, DCs, NK cells, and other immune cells can regulate immune checkpoints, resulting in the inhibition of tumor cell elimination.^[304]31 The liver is a major regulatory organ that manages metabolism throughout the body and maintains metabolic homeostasis; once metabolic homeostasis is disrupted, a variety of cancers, including LIHC, may occur.^[305]32 Previous studies have shown that disorders of glucose metabolism,^[306]33 lipid metabolism,^[307]34 and nucleotide metabolism^[308]35 may lead to the occurrence of liver cancer via different pathways. Understanding the changes that occur in the metabolic profile during hepatocarcinogenesis is important for elucidating the pathogenic mechanisms of cancer and identifying new diagnostic and prognostic biomarkers. In our study, analysis of the correlation between TMEM147 expression and immune cell infiltration in LIHC revealed that TMEM147 was positively related to Th2 cells, TFH cells, and NK CD56 bright cells and negatively related to Th17 cells, Tcm cells, neutrophils, Tregs, DCs, and cytotoxic cells. Experiments that were performed with the normal human liver cell line MIHA and four LIHC cell lines (BEL-7402, SMMC-7721, HepG2, and Huh7 cells) confirmed the role of TMEM147 in promoting liver cancer cell proliferation, further confirming the clinical value of TMEM147 in the diagnosis of liver cancer. In conclusion, this is the first study to investigate the oncogenic role of TMEM147 across cancers as well as its carcinogenic role in liver cancer. The identification of the diagnostic and prognostic significance of TMEM147 across cancers, and further exploration of its importance in LIHC, can provide new insights for the comprehensive understanding of the key role of TMEM147 in tumor promotion and inhibition, provide a comprehensive basis for in-depth verification of its function in molecular biology experiments, and even provide integrated analysis results for the development of future cancer treatments for use in the clinic. Acknowledgments