High expression level of TMEM147 protein is associated with poor survival in HCC.
To elucidate the relationship between TMEM147 and HCC progression, we first searched the GEO database and mined TCGA transcriptome datasets to compare the levels of TMEM147 between HCC tissues and normal tissues on the GAPIA website and found that TMEM147 mRNA was significantly upregulated in HCC tissues (Fig. 1A and 1B).
We measured TMEM147 expression in 72 HCC tissues and 28 non-tumor liver tissues, and found that the TMEM147 protein exhibited a high staining rate of 83.3% (60/72) in HCC tissues, which was markedly higher than the staining rate of 10.7% (3/28) in normal liver tissues (Fig. 1C), and the protein expression level of TMEM147 in the HCC cohort was significantly correlated with patient survival (Fig. 1I). Detailed clinicopathological characteristics are shown in additional file 1: Table S1. Likewise, qRT-PCR in 24 pairs of HCC and peritumoral tissues revealed that the relative mRNA expression of TMEM147 was significantly upregulated in HCC tissues compared to that in adjacent non-tumorous tissues (Fig. 1D). Accordingly, Western blot analysis was conducted on 24 pairs of HCC tissues, and the results revealed that TMEM147 protein levels were higher in HCC tissues than in non-malignant samples (Fig. 1F and S1A). In agreement with these results, the mRNA and protein expression levels of TMEM147 in HCC cell lines were significantly higher than those in normal liver cells (Fig. 1E and 1G).
Moreover, Kaplan–Meier survival curve analysis of TCGA datasets demonstrated that HCC patients with increased TMEM147 protein expression had poorer overall five-year survival (Fig. 1H).
TMEM147 boosts tumor cell proliferation and metastasis
To investigate the effect of TMEM147 on HCC, we constructed lentivirus vectors to upregulate and silence TMEM147 in Huh7 and HepG2 cells; Western blot and qRT-PCR were applied to confirm the overexpression and knockdown efficiency of TMEM147 (Fig S2A and S2B). According to growth curve analysis and colony formation assay, deletion of TMEM147 significantly decreased HCC cell proliferation, whereas TMEM147 overexpression significantly increased HCC cell proliferation (Fig. 2A and 2B).
Next, we sought to determine the role of TMEM147 in HCC metastasis, migration, and invasion by performing Transwell and wound healing assays in vitro were performed. The results showed that overexpression of TMEM147 promoted the migration and invasion of Huh7 and HepG2 cells (Fig. 2C and S2C), whereas TMEM147 knockdown exerted the opposite effect (Fig. 2D and S2D). Collectively, these results indicate that elevated TMEM147 protein level in HCC boosts tumor cell proliferation and metastasis.
To evaluate the tumorigenic function of TMEM147 in vivo, a subcutaneous xenograft tumor model was established. Consistent with our in vitro data, the tumor volume and weight in mice implanted with Huh7- TMEM147 cells were markedly higher than those in the control group; and those in HepG2-shTMEM147 mice were drastically lower than those in the control group (Fig. 2E). To determine whether TMEM147 affects HCC metastasis in vivo, stably transfected cells (Huh7-TMEM147 and controls) were injected into the tail vein to establish a lung metastasis model. Six weeks after the intervention, the number and volume of lung metastases decreased in the TMEM147 silencing group, but increased in the TMEM147 overexpression group. (Fig. 2F). In summary, these results provided further evidence that TMEM147 is involved in the exacerbation of HCC growth and metastasis.
TMEM147 facilitates 27HC expression by activating DHCR7 to accelerate the growth and metastasis of HCC
To identify the target genes regulated by TMEM147, we knocked down TMEM147 in Huh7 cells. RNA-seq analysis revealed that the transcriptome profiles of TMEM147-silenced cells were distinct from those of the control cells (Fig. 3A). Gene ontology (GO) analysis showed that the differentially expressed genes were significantly enriched in cholesterol metabolism pathways (Fig. 3B). Several key enzymes in this pathway were further assessed, including methylsterol monooxygenase 1 (MSMO1), emopamil binding protein (EBP), Hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1), type 2 isopentenyl diphosphate isomerase (IDI2), DHCR7, farnesyl-diphosphate farnesyltransferase (FDFT1), and squalene epoxidase (SQLE); DHCR7 was identified as the most differentially expressed (Fig. 3C and S3A), implying that the expression of DHCR7 may be modulated by TMEM147.
To confirm the role of TMEM147 in the regulation of DHCR7, the expression of DHCR7 in HCC cells was detected after TMEM147 was knocked down or overexpressed. TMEM147 overexpression increased DHCR7 transcription and protein levels, whereas DHCR7 levels were significantly reduced in the absence of TMEM147 (Fig. 3D and 3E).
CEs, the intracellular storage forms of excess cholesterol, are of central importance to cholesterol homeostasis; their formation is a measure of the availability of cellular free cholesterol, and CE levels are consistent with sterol reductase activity.[31] ELISA was employed to quantify the cholesteryl esters in cell lysates and subcutaneous tumors from mice. The results indicate that the total levels of CEs are greatly reduced after TMEM147 knockdown and are significantly upregulated after TMEM147 overexpression (Fig. 3F). A study showed that, in breast cancer, cholesterol mediates the metastatic effects via its oxysterol metabolite, 27-hydroxycholesterol.[28] Therefore, we next detected the content of 27HC in cells and the cell culture medium by ELISA assay. As anticipated, the content of 27HC was consistent with CEs and TMEM147 (Fig. 3G).
CYP27A1 is the enzyme responsible for the rate-limiting step in 27HC biosynthesis, and we found that the protein and mRNA levels of CYP27A1 were closely correlated with TMEM147 levels (Fig. 3H and S3B). To explore the role of DHCR7 in the tumor-promoting function of TMEM147, the effect of DHCR7 knockdown on the tumorigenic potential of TMEM147, RNA lentivirus stably transfected into Huh7 and HepG2 cells was examined (Fig S3C and S3D). Downregulation of DHCR7 expression markedly suppressed the proliferative, migratory, and invasive capabilities of HCC cells, whereas the addition of exogenous 27HC partially restored the inhibitory effect of DHCR7 silencing. The function of 27HC was impeded by the CYP27A1 inhibitor, GW297X (Fig. 3I, S3E, 3 J, and S3F).
A xenograft model showed that DHCR7 knockdown inhibited the growth of subcutaneous tumors, whereas 27HC supplementation reversed the inhibitory effect of DHCR7 downregulation on tumor growth. Additionally, DHCR7 knockdown decreases lung metastasis in a mouse model. However, 27HC supplementation neutralized the inhibition of lung metastasis by DHCR7 knockdown, whereas the injection of the CYP27A1 inhibitor GW297X in cells overexpressing TMEM147 restrained tumor progression and metastasis, and supplementation with 27HC offset the effect of GW297X (Fig S3G and S3H). Collectively, the in vitro and in vivo results suggest that DHCR7 is involved in the oncogenic effect of TMEM147 in HCC and that the function of DHCR7 depends on increased 27HC levels.
TMEM147-mediated promotion of DHCR7 expression is dependent on the transcription factor STAT2
In Huh7 and HepG2 cell lines, protein binding to TMEM147 was analyzed by mass spectrometry using CO-IP. It was found that the target gene STAT2, which encodes a transcription factor was closely bound to TMEM147 in both cell lines (Fig. 4A). The interaction between TMEM147 and STAT2 was further confirmed by CO-IP. IP assays confirmed that STAT2 existed in complexes and was precipitated with an antibody against Flag-TMEM147, unlike with the control IgG (Fig. 4B, upper). The binding of endogenous STAT2 to TMEM147 was validated by an IP assay using an antibody against STAT2 (Fig. 4B, lower). Using JASPAR and UCSC database analyses, we found that the DHCR7 promoter contained putative binding sites for the transcription factor STAT2 (Fig. 4C). We speculated that TMEM147 might regulate DHCR7 expression through the transcription factor STAT2.
As shown in Fig. 3, TMEM147 altered the mRNA and protein expression of DHCR7. Western blotting revealed that STAT2 protein expression levels did not significantly correlate with those of TMEM147 in HCC cells. However, transcription 2 (STAT2) at tyrosine residue 690 (Tyr690) and phosphorylated-STAT2(Tyr690) changed upon TMEM147 treatment (Fig. 4D). Consistent with this observation, a chromatin immunoprecipitation assay (ChIP) showed that the STAT2-bound DHCR7 promoter was enriched in TMEM147 overexpressed cells but diminished in TMEM147-knocked down cells (Fig. 4E). The luciferase reporter assay showed that STAT2 overexpression significantly increased the luciferase activity of reporters containing wild-type binding sites compared to that of NC-vector cells (Fig. 4F). However, no significant change in luciferase activity was observed upon binding of STAT2 to the mutant DHCR7 promoter (Fig. 4F). To determine whether STAT2 influenced DHCR7 expression, we transfected STAT2 into HCC cells. We found that STAT2 significantly promoted the expression levels of DHCR7 in HCC cells; in contrast, elimination of STAT2 significantly decreased DHCR7 expression (Fig. 4G). When TMEM147 was overexpressed, si-STAT2 inhibited DHCR7 expression, indicating that TMEM147’s promoting function of DHCR7 depended on STAT2 (Fig. 4H). A moderate correlation between TMEM147 and STAT2, and DHCR7 and STAT2 was detected in human HCC (Fig. 4I), suggesting that TMEM147 promotes the phosphorylation of STAT2, enhances its transcriptional activity, and then increases the expression of DHCR7. To assess the effect of STAT2 on HCC, we transfected si-STAT2 into HCC cells and found si-STAT2 significantly suppressed HCC cell proliferation, invasion, and migration (Fig. 4J and 4K).
TMEM147 induces ferroptosis by promoting 27HC/GPX4 pathway in HCC.
Previous studies have shown that chronic exposure to 27HC increases lipid accumulation in cells and enhances the resistance of breast cancer cells to ferroptosis, whereas GPX4 inhibition reverses this effect.[20] To explore the effects of TMEM147 and 27HC on ferroptosis, we measured the levels of GSH, which plays an essential role in the repair of oxidative damage. As shown in Fig. 5A, erastin and RSL3 significantly decreased GSH levels in HCC cells, and TMEM147 overexpression rescued the decreased GSH levels caused by erastin and RSL3. When GW297X was added, the GSH value decreased significantly. Correspondingly, ferrous ion (Fe2+) and MDA levels were reduced when TMEM147 was overexpressed but increased in cells with TMEM147 overexpression subjected to GW297X treatment (Fig S5A and S5B). MMP increased over time, with a maximum at approximately 8 h after erastin treatment (Fig. 5B); however, MMP was overwhelmingly suppressed by TMEM147, and completely rescued by GW297X. Correspondingly, as shown in Fig. 5C, TMEM147 reduced ROS production via 27HC, whereas ROS generation was significantly enhanced in GW297X treated cells.
TEM analysis further revealed that erastin and RSL3 treated HCC cells contained shrunken mitochondria with elevated membrane density, a typical morphological feature of ferroptosis. TMEM147 overexpression protected cells from ferroptosis, whereas treatment with GW297X aggravated ferroptosis. This indicates that TMEM147 confers ferroptosis resistance by promoting 27HC. (Fig. 5D)
In parallel with our observation in vitro, in comparison with erastin group, tumor size and weight were significantly increased by TMEM147 overexpression, whereas GW297X pretreatment markedly reversed this TMEM147-mediated oncogenic effect (Fig. 5E). Tissues from the control and GW297X injected tumors, exhibited increased levels of 4-hydroxy-2-noneal (4HNE, a biomarker of lipid peroxidation), whereas TMEM147 overexpression reduced the levels of 4HNE, as determined by IHC (Fig. 5F). These in vivo results further support the idea that the resistance to ferroptosis induced by TMEM147 is mainly dependent on 27HC.
To further elucidate the mechanism of ferroptosis triggered by TMEM147, the iron metabolism-related proteins GPX4, HMOX1, SLC7A11, NCOA4, NRF2, and CP were analyzed. GPX4 was markedly altered by TMEM147 and GW297X (Fig. 5G and S5C). The expression of GPX4 in HCC cells was assessed by qRT-PCR and western blot analysis, and it was found that GPX4 expression was downregulated upon the knockdown of TMEM147. Indeed, we demonstrated that the TMEM147-dependent GPX4 downregulation was dramatically rescued by the addition of exogenous 27HC. (Fig. 5H and 5I). In summary, these results suggested that TMEM147 promotes GPX4 expression via 27HC to reduce the resistance of HCC cells to ferroptosis.
HCC cell-derived 27HC enhances lipid metabolism in macrophages and drives M2 polarization
Previous studies have shown that pretreatment of macrophages with 27HC can inhibit T cell function .[30] However, it remains unclear whether tumor-associated macrophages, which are important immune cells in the tumor microenvironment, are also regulated by 27HC. To further substantiate this hypothesis, the following experiments were conducted.
THP-1 induced macrophages were incubated with CM gathered from HCC cells (Fig. 6A), and the expression of TAM markers arginase 1 (ARG1) and CD206 was further analyzed. CM from TMEM147 overexpression cells or the addition of exogenous 27HC, but not the control medium, increased the expression of the M2 markers ARG1 and CD206, whereas GW297X deterred this conversion (Fig. 6B and S6A). These results suggest that endogenous 27HC mediates M2 macrophage polarization. The results of cell immunofluorescence and flow cytometry further confirmed that tumor-derived 27HC was responsible for cancer CM-induced M2 macrophage polarization (Fig. 6C and 6D).
In animal studies, IHC staining for the M2 marker ARG1 showed that the accumulation of M2 macrophages was greater in MΦs cultured with the CM obtained from TMEM147 overexpression cells or with exogenous 27HC (Fig. 6E).
Pan Su et.al. reported that the reprogramming of fatty acid oxidative metabolism plays a crucial role in M2 polarization of TMA. Macrophages from both human and murine tumor tissues are enriched with lipids owing to increased lipid uptake by macrophages.[32] In our study, as expected, compared with control MΦs, MΦs cultured with CM collected from TMEM147 overexpression cells or added exogenous 27HC were enriched with more lipids (Fig. 6F), genes involved in fatty acid β-oxidation including FAO rate-limiting enzymes CPT1A and PPARγ, were significantly increased (Fig. 6B and S6A). In metabolic assay experiments, TAMs co-cultured with TMEM147 overexpression cells and exogenous 27HC supplementation displayed enhanced mitochondrial OCR and markedly increased spare respiratory capacity (SRC), in contrast to TMEM147 knockdown cells co-cultured with GW297X-treated macrophages (Fig. 6G and 6H). Thus, we validated that 27HC promotes the accumulation of lipids and fatty acid-oxidation in TAMs, which are crucial for M2 macrophage polarization.
In summary, we highlighted the importance of HCC cell-derived 27HC in enhancing the lipid metabolism of macrophages and promoting M2 polarization.
27HC- induced polarized macrophages potentiate migration of HCC cells
As cancer cell-derived 27HC can promote the M2 polarization of TAMs, we hypothesized that TAMs with increased M2 polarization could further promote the migration of HCC cells. CM-induced polarized macrophages were co-cultured with HCC cells (Fig. 7A), and the invasion and migration of HCC cells in each group were consistent with M2 polarization of macrophages (Fig. 7B and 7C). In vivo, we injected co-cultured cells through the tail vein of nude mice and monitored lung metastatic nodules (Fig. 7D), which showed that polarized macrophages induced by 27HC could significantly exacerbate the metastasis of HCC cells.