Mitochondrial dynamics play vital roles in the tumorigenicity and malignancy of various types of cancers by promoting the tumor-initiating potential of cancer cells, suggesting that targeting crucial factors that drive mitochondrial dynamics may lead to promising anticancer therapies. In the current study, we report that overexpression of mitochondrial fission factor (MFF), which is upregulated significantly in liver cancer–initiating cells (LCIC), promotes mitochondrial fission and enhances stemness and tumor-initiating capability in non-LCICs. MFF-induced mitochondrial fission evoked mitophagy and asymmetric stem cell division and promoted a metabolic shift from oxidative phosphorylation to glycolysis that decreased mitochondrial reactive oxygen species (ROS) production, which prevented ROS-mediated degradation of the pluripotency transcription factor OCT4. CRISPR affinity purification in situ of regulatory elements showed that T-box transcription factor 19 (TBX19), which is overexpressed uniquely in LCICs compared with non-LCICs and liver progenitor cells, forms a complex with PRMT1 on the MFF promoter in LCICs, eliciting epigenetic histone H4R3me2a/H3K9ac-mediated transactivation of MFF. Targeting PRMT1 using furamidine, a selective pharmacologic inhibitor, suppressed TBX19-induced mitochondrial fission, leading to a profound loss of self-renewal potential and tumor-initiating capacity of LCICs. These findings unveil a novel mechanism underlying mitochondrial fission–mediated cancer stemness and suggest that regulation of mitochondrial fission via inhibition of PRMT1 may be an attractive therapeutic option for liver cancer treatment.

Significance:

These findings show that TBX19/PRMT1 complex–mediated upregulation of MFF promotes mitochondrial fission and tumor-initiating capacity in liver cancer cells, identifying PRMT1 as a viable therapeutic target in liver cancer.

Mitochondria function as energy stations for adenosine triphosphate and reactive oxygen species (ROS) production, which are required for proliferating cells, including stem cells and cancer cells (1–3). To allow cells to adapt to various stressful circumstances, mitochondria form a dynamic network via the opposing processes of fission and fusion, which divide or fuse the two lipid bilayers that surround mitochondria, resulting in a decrease or increase in energy production, respectively (3, 4). Recently, it has been demonstrated that mitochondrial fission contributes to strengthening the self-renewal capacity in both induced pluripotent stem (iPS) cells and cancer-initiating cells (CIC) via the asymmetric allocation of healthy and oxidatively damaged mitochondria (5, 6). Importantly, inhibition of mitochondrial fission using mdivi-1 suppresses the stem-like traits of CICs, suggesting that mitochondrial fission may be a promising target for cancer therapy (6, 7). However, because of its critical role in normal stem cells and CICs, targeting mitochondrial fission directly would disrupt somatic stem cell activity, resulting in the loss of tissue homeostasis and cause developmental defects. Therefore, it is of great clinical value to unveil the aberrant molecular control of mitochondrial fission in CICs that is distinct from that of normal stem cells, which may provide a new paradigm to develop more effective treatment strategies for cancers.

In response to metabolic demands and stressors, mitochondrial fission aids in allocating mitochondrial contents, enabling asymmetric division, and eliminating dysfunctional mitochondria via mitophagy (3, 8, 9). During the fission process, mitochondrial fission protein dynamin–related protein 1 (DRP1) is recruited from the cytosol to mitochondria by interaction with different adapters located on the outer mitochondrial membrane, such as mitochondrial fission factor (MFF) and FIS1 (10–12), and forms a ring-like structure around mitochondria, which subsequently initiates mitochondrial fission via its GTPase activity (13, 14). Recently, multiple studies have documented that mitochondrial fission is involved in asymmetric cell division, pluripotency, and the fate of stem cells. It is reported that mitochondria become spherical shaped during iPS reprogramming (15), whereas silencing DRP1 impairs both mitochondrial fragmentation and generation of iPS cell colonies (13). Clearance of dysfunctional mitochondria by mitophagy can avoid senescence and prevent stem cell exhaustion (16–18), and inhibition of fission by mdivi-1 disrupts the asymmetric apportioning mitochondrial and caused loss of stem cell properties in the progeny cells (16).

Notably, mitochondrial fission regulators also play vital roles in self-renewal and tumor-initiating capability of CICs (5, 7, 19). For instance, CDK5-mediated DRP1 hyperactivation promotes mitochondrial fission and enhances tumorigenicity in brain tumor–initiating cells (BTIC; ref. 7). AMPK/FIS1-mediated mitochondrial fission/mitophagy is crucial for self-renewal of human AML stem cells, and BRD4/MFF axis–induced mitochondrial fission enables asymmetric division and the self-renewal ability in prostate CICs (5, 19). These studies indicate that mitochondrial fission may be involved in tumorigenesis.

Transcription factor TBX19, characterized by a highly conserved DNA binding motif (T-box), is reported to be upregulated in colorectal cancer and pituitary corticotroph tumors and KRAS-mutant cells (20, 21). However, the clinical significance and biological role of TBX19 in cancer remain largely unknown. Herein, we revealed that TBX19 formed complex with PRMT1 and epigenetically upregulated MFF in liver cancer–initiating cells (LCIC), resulting in increased mitochondrial fission and mitophagy, and enhanced tumor-initiating capability. Pharmacologic inhibition of PRMT1 impaired mitochondrial fission and inhibited the tumor-forming ability of LCICs. Thus, our results revealed the underlying mechanism of mitochondrial fission in LCICs, which provided a new paradigm to develop promising therapeutic strategies for liver cancer treatment.

Cell lines and cell culture

The hepatocellular carcinoma (HCC) cell lines, including Hep3B, SNU449 were purchased from the ATCC and cultured in DMEM (GIBCO) supplemented with 10% FBS (GIBCO), 100 U/mL penicillin and 100 μg/mL streptomycin (Invitrogen). EpCAM+/CD133+ LCICs were cultured in ultra-low attachment plates with RPMI-1640 medium, supplemented with EGF (20 ng/mL), bFGF (20 ng/mL), 1 × B27 and 1× antibiotic/antimycotic, in 4% O2 and 5% CO2. All the cell lines were tested for Mycoplasma contamination and were authenticated by short tandem repeat fingerprinting at Medicine Laboratory of Forensic Medicine, Department of Sun Yat-Sen University (Guangzhou, China).

Patient information and tissue specimens

Two clinical human liver cancer tissues were diagnosed and obtained from Sun Yat-sen University Cancer Center according to the Declaration of Helsinki, and each patient signed a written informed consent for all the procedures. The study protocols were approved by the Institutional Research Ethics Committee of Sun Yat-sen University Cancer Center for the use of these clinical materials for research purposes.

Isolation of LCICs

The EpCAM+/CD133+ LCICs were isolated from Hep3B and SNU449 cell lines and two clinical HCC primary tissues as previously described (22, 23). Briefly, liver cancer specimens were immediately digested with collagenase and DNase to obtain cell suspensions, and further washed and adjusted to a concentration of 2 × 107 cells/mL. These cells were incubated with antibodies against EpCAM (1:100; ab237396, Abcam) and CD133 (1:100; ab252126, Abcam) and sorted by FACS to isolate EpCAM+/CD133+ LCIC and EpCAMCD133 non-LCIC populations.

Streptavidin affinity purification of dCas9-captured DNA and proteins

Streptavidin affinity purification of dCas9-Captured DNA and proteins was performed following the published protocol (24). Briefly, EpCAM+/CD133+ LCICs and EpCAM/CD133 non-LCICs (5 × 107) transfected with FB-dCas9 plasmid and MFF promoter sgRNAs were harvested and treated with 1% formaldehyde to cross-link the proteins to DNA. The cells were lysed in 1 mL Lysis buffer 1 (1 mol/L HEPES-KOH, 1 mol/L NaCl, 0.5 mol/L EDTA, 50% glycerol, 10% NP-40, 10% Triton X-100, and pH 7.5), and rotated for 15 minutes at 4°C, then centrifuged at 2,300 × g for 5 minutes at 4°C to isolate the nuclei, which were suspended in 1 mL of lysis buffer 2 (1 mol/L Tris-HCl, 1 mol/L NaCl, 0.5 mol/L EDTA, 0.5 mol/L EGTA, and pH 8.0). Cell lysates were centrifuged at 2,300 × g for 5 minutes at 4°C to isolate precipitate and suspended in 1 mL of lysis buffer 3 (1 mol/L Tris-HCl, 1 mol/L NaCl, 0.5 mol/L EDTA, 0.5 mol/L EGTA, 5% Na-Deoxycholate, 5% N-lauroylsarcosine, and pH 8.0). The cells were subjected for sonication to shear chromatin fragments and centrifuged at 12,000 × g for 10 minutes at 4°C. Supernatant was then incubated with 50 μL of Streptavidin T1 Dynabeads (Thermo-Fisher Scientific) at 4°C overnight. After overnight incubation, Dynabeads were washed twice with 1 mL of Wash buffer (1 mol/L HEPES-KOH, 5 mol/L LiCl, 0.5 mol/L EDTA, 5% Na-Deoxycholate, 10% NP-40, and pH 8.0), and twice with 1 mL of TE buffer (1 mol/L Tris-HCl, 0.5 mol/L EDTA, 1 mol/L NaCl, and pH 8.0). The proteins were separated by SDS-PAGE and analyzed by Western blot and IP-MS analysis (Shanghai Applied Protein Technology Co. Ltd.).

Mitochondrial morphology analysis

Quantification of mitochondrial morphology followed as reported previously (25). Briefly, cells from more than 10 randomly selected fields were imaged and analyzed per sample from three independent experiments, and it was done by 2 researchers who were blinded to genotype and treatment of the samples. Cells were divided into three categories based on mitochondrial morphology stained by MitoTracker probe dye: “Tubulated” is with a majority of mitochondria in a cell forming interconnected and elongated morphology with mitochondrial length >10 μm; “Intermediate” is with mixed short tubular mitochondria with mitochondrial length <10 μm in a cell; “Fragmented” is with a majority of punctiform mitochondria in a cell.

In vivo tumorigenesis experiments

All of the animal procedures were complied with all relevant ethical regulations for animal testing and research and the ethical approval was approved by the Sun Yat-sen University Animal Care Committee. Different numbers (5 × 105, 1 × 104, 5 × 103, 1 × 103, 5 × 102, 1 × 102) of indicated LCICs and non-LCICs were inoculated with Matrigel (final concentration, 25%) subcutaneously into the inguinal folds of NOD/Shi-scid/IL-2Rγnull (NOG) mice (5–6 weeks old; CREA Japan Inc.). Tumorigenic cell frequency (TIC frequency) was calculated by Extreme Limiting Dilution Analysis (ELDA) using publically available website tool (http://bioinf.wehi.edu.au/software/elda/), and displayed by 95% confidence intervals for the frequency in each population group. The statistical P value was obtained using a χ2 test.

In vivo orthotopic liver injection model

Tumors were initiated by implantation of 5 × 105 or 1 × 102 EpCAM+/CD133+ LCIC and EpCAM/CD133 non-LCIC populations derived from patients, coated with Matrigel and media in a 1:1 ratio, into liver of NOG mice (5–6 weeks old; CREA Japan Inc.). Orthotopic liver injection was performed using a 25‐μL syringe with a cemented 22‐gauge needle. The NOG mice were anesthetized with 50 mg/kg of pentobarbital and a small incision was made in the abdomen. The left lobe of the liver was gently exposed and the re-suspended cells were carefully injected into the lobe. After injection, the abdominal wound was sutured. For drug treatment, vehicle or furamidine (20 mg/kg) were injected intraperitoneally into mice. The mice were sacrificed when the mice become moribund and tumors were excised and paraffin embedded.

Statistical analysis

All values are expressed as the mean ± standard deviation of individual samples. n represents the number of independent experiments performed on different mice or different batches of cells or different clinical tissues. Statistical analysis was performed using the Student two-tailed t test and one or two-way ANOVA. Survival curves were plotted by the Kaplan–Meier method and compared by the log-rank test. The P values of <0.05 were considered as statistically significant. Statistical analysis was performed using GraphPad Prism 8.

Additional information is provided in Supplementary Materials and Methods.

LCICs display increased mitochondrial fission

To examine the impact of mitochondrial dynamics on LCIC function, we isolated EpCAM+/CD133+ LCICs and matched EpCAM/CD133 non-LCIC populations from two patient-derived HCC tissues as previously reported (26–28). An in vitro tumorsphere formation assay further confirmed that the EpCAM+/CD133+ LCICs exhibited higher sphere-forming ability than the EpCAM/CD133 non-LCICs (Supplementary Fig. S1A). However, we did not observe significant growth differences between 2D-cultured LCICs and non-LCICs (Supplementary Fig. S1B), suggesting that the differences of tumorsphere formation were caused by stemness. We further validated the mitochondrial morphology of LCICs and non-LCICs using three different methods, including mitochondrial red fluorescent probe (MitoTracker probe), mitochondrial marker (TOM20), and transmission electron microscopy (TEM). As shown in Fig. 1A and Supplementary Fig. S1C and S1D, mitochondria in matched LCICs displayed more a fragmented and less tubular morphology, whereas non-LCICs exhibited elongated tubular mitochondria with longer cristae and higher cristae density. Meanwhile, expression of total DRP1 and phosphorylated DRP1 (p-DRP1, S616) was found increased on mitochondria and colocalized with TOM20 in LCICs compared with non-LCICs (Fig. 1B–D; Supplementary Fig. S1E). Consistently, the LCICs isolated from liver cancer cell lines Hep3B and SNU449 also exhibited shorter, rounded mitochondria in LCICs as compared with elongated, tubular structures in non-LCICs (Fig. 1A and B; Supplementary Fig. S1A and S1C–S1E). Taken together, these results suggest that LCICs display increased mitochondrial fission.

Figure 1.

Upregulated MFF induces mitochondrial fission in LCICs. A, The mitochondria morphologies were visualized using MitoTracker probe in LCIC and non-LCIC tumor cells isolated from two patient-derived HCC tissues and two HCC cell lines, respectively. Scale bars, top, 5 μm; bottom, 1 μm. B, Representative confocal images of TOM20 (green) or p-DRP1 (S616) (red). Scale bars, top, 5 μm; bottom, 2 μm. C, Real-time analysis of 58 mitochondrial dynamics–related mediators showing that MFF was highly upregulated in two patient-derived LCICs compared with two matched non-LCICs. The expression of mRNA was normalized to GAPDH. D, IB analysis of MFF, p-DRP1 (S616), and total DRP1 levels in mitochondria from two patients HCC tissues and two HCC cell line–derived LCICs. TOM20 served as the protein loading control. E, IB analysis of MFF expression in vector-control and MFF-silenced LCICs. GAPDH served as the loading control. F, Representative confocal images of TOM20 (green) or p-DRP1 (S616) (red; scale bars, top, 5 μm; bottom, 2 μm) in the indicated cells. G, Different numbers of the indicated cells were inoculated subcutaneously into the inguinal folds of NOG mice. Representative images of tumors formed by 5 × 104 and 5 × 102 of the indicated cells are shown. TIC frequency was calculated on the basis of the ELDA analysis. H, TCGA analysis comparing relapse-free survival in patients with HCC (n = 345; P = 0.009) and the well-differentiated (grade I; n = 54; P = 0.012) group with low and high MFF expression levels. Each error bar in A represents the mean ± SD of three independent experiments. ***, P < 0.001.

Figure 1.

Upregulated MFF induces mitochondrial fission in LCICs. A, The mitochondria morphologies were visualized using MitoTracker probe in LCIC and non-LCIC tumor cells isolated from two patient-derived HCC tissues and two HCC cell lines, respectively. Scale bars, top, 5 μm; bottom, 1 μm. B, Representative confocal images of TOM20 (green) or p-DRP1 (S616) (red). Scale bars, top, 5 μm; bottom, 2 μm. C, Real-time analysis of 58 mitochondrial dynamics–related mediators showing that MFF was highly upregulated in two patient-derived LCICs compared with two matched non-LCICs. The expression of mRNA was normalized to GAPDH. D, IB analysis of MFF, p-DRP1 (S616), and total DRP1 levels in mitochondria from two patients HCC tissues and two HCC cell line–derived LCICs. TOM20 served as the protein loading control. E, IB analysis of MFF expression in vector-control and MFF-silenced LCICs. GAPDH served as the loading control. F, Representative confocal images of TOM20 (green) or p-DRP1 (S616) (red; scale bars, top, 5 μm; bottom, 2 μm) in the indicated cells. G, Different numbers of the indicated cells were inoculated subcutaneously into the inguinal folds of NOG mice. Representative images of tumors formed by 5 × 104 and 5 × 102 of the indicated cells are shown. TIC frequency was calculated on the basis of the ELDA analysis. H, TCGA analysis comparing relapse-free survival in patients with HCC (n = 345; P = 0.009) and the well-differentiated (grade I; n = 54; P = 0.012) group with low and high MFF expression levels. Each error bar in A represents the mean ± SD of three independent experiments. ***, P < 0.001.

Close modal

Upregulated MFF induces mitochondrial fission in LCICs

To determine the mechanism of increased mitochondrial fission in LCICs, we examined 58 mitochondrial dynamics–related mediators, including 39 genes involved in mitochondrial fission and 19 genes involved in mitochondrial fusion. Real-time PCR analyses revealed that expression of MFF, but not DRP1, FIS1, MID49 and MID51, was highly and consistently upregulated in four LCICs isolated from patient-derived HCC tissues and HCC cell lines compared with that in four matched non-LCICs (Fig. 1C; Supplementary Fig. S1F). Immunoblotting (IB) analysis further confirmed that mitochondrial MFF was also markedly increased in LCICs (Fig. 1D). Importantly, MFF-downregulated LCICs not only displayed elongated, tubular mitochondrial structures and reduced total and phosphorylated DRP1 on mitochondria, but also showed decreased the tumor-forming potential (Fig. 1E–G; Supplementary Fig. S2A and S2B). Correspondingly, MFF-overexpressing non-LCICs exhibited more a fragmented mitochondrial morphology and increased total/phosphorylated DRP1 on mitochondrial (Supplementary Fig. S2C and S2D). Overexpression of MFF significantly enhanced the tumorsphere formation ability of non-LCICs, and obviously increased the percentage of the side-population (SP) cells (SP+ population), which was a subpopulation of cells based on SP assay that may represent a universal TIC-like phenotype (Supplementary Fig. S2E and S2F). Therefore, these results indicate that MFF-mediated mitochondrial fission enhances LCIC traits.

Furthermore, analysis of The Cancer Genome Atlas (TCGA) revealed that increased MFF expression correlated significantly with HCC grade and associated reversely with relapse-free survival of patients with HCC (n = 345; P = 0.009), even in HCC grade I group (n = 54; P = 0.012; Supplementary Fig. S2G; Fig. 1H), providing further evidence that MFF overexpression contributed to the initiation and progression of liver cancer.

TBX19 transcriptionally upregulates MFF

To investigate the molecular mechanism of MFF upregulation in LCICs, the CRISPR affinity purification in situ of regulatory elements (CAPTURE) approach (24) and quantitative mass spectrometry were used to identify the trans-regulatory factors targeting MFF in LCICs (Fig. 2A). Compared with those in the non-LCICs, we found that the levels of 12 potential trans-regulatory factors were increased on the MFF promoter in LCICs (Fig. 2A). However, qPCR analysis revealed that MFF mRNA expression was only significantly decreased in TBX19-, or PRMT1-, or RNA polymerase II (RNAP II)–, or TFIID-silenced LCICs (Fig. 2B). Furthermore, chromatin immunoprecipitation (ChIP) assays showed that the enrichment of TBX19 and PRMT1 on the MFF promoter was increased in LCICs compared with that in non-LCICs (Fig. 2C; Supplementary Fig. S3A). Moreover, IB analysis showed that expression levels of TBX19, PRMT1, and MFF were elevated overtly in all four LCICs derived from two clinical HCC tissues and in Hep3B, SNU449 cell lines compared with that in non-LCICs and liver progenitor cells (LPCs#1, LPCs#2; Fig. 2D). All these results suggested that TBX19 and PRMT1 might regulate MFF expression in LCICs. Indeed, overexpressing TBX19 in non-LCICs dramatically increased expression of mitochondrial MFF and total/phosphorylated DRP1. However, we did not observe the inducing effect of TBX19 on MFF upregulation in LPCs (Fig. 2E). Taken together, our data indicate that TBX19 transcriptionally upregulates MFF in LCICs.

Figure 2.

TBX19 transcriptionally upregulates MFF and induces mitochondrial fission and promotes mitophagy in LCICs. A, Schematic of the CRISPR CAPTURE approach following the quantitative mass spectrometry to identify the trans-regulatory factors targeting MFF in LCICs. B, Real-time analysis of the mRNA expression of MFF in TBX19-, PRMT1-, RNAPII-, or TFIID-silenced LCICs. The mRNA expression level was normalized to GAPDH. C, ChIP assay analysis of the enrichment of TBX19 and PRMT1 on the MFF promoter in LCICs compared with non-LCICs. D, IB analysis of TBX19, PRMT1, and MFF levels in two liver progenitor cells, LCICs, and non-LCICs derived from two patient-derived HCC tissues and two HCC cell lines. GAPDH served as the loading control. E, IB analysis of expression of mitochondrial MFF, p-DRP1 (S616), total DRP1, and total MFF in the indicated cells. TOM20 served as the mitochondrial loading control, and GAPDH served as the total loading control. F, Mitochondrial morphology was visualized using MitoTracker probe in the indicated cells. Scale bars, top, 5 μm; bottom, 2 μm. G, Left, a diagram showing that the Mito./Nuc. area ratio was quantified by segregating the DAPI and MitoTracker channels and subsequent quantification of the area of each channel. Middle and right, representative images of mitochondria and quantification of Mito./Nuc. area ratio in control and TBX19-overexpressing non-LCICs. Scale bars, 5 μm. H, Quantification of mtDNA content in control and TBX19-overexpressing non-LCICs. I, TEM images of mitochondria (top) and quantification (bottom) of mitochondria engulfed in autophagosome in the indicated cells. Scale bars, 1 μm. M, mitochondria. J, Representative images (left; scale bars, 5 μm) and quantification (right) of the colocalization of LC3B and MitoTracker probe in the indicated cells determined by Manders overlap coefficients (MOC) using ImageJ software. The values on the y-axis mean the proportion of MitoTracker/LC3B colocalization in the total MitoTracker fluorescence region. K, Representative confocal images of anti-TOM20 (green) or p-DRP1 (S616) (red; scale bars, 5 μm) and quantification of colocalization of p-DRP1 and TOM20 in the indicated cells determined by MOC using ImageJ software. The values on the y-axis mean the proportion of TOM20/p-DRP1 colocalization in the total TOM20 fluorescence region. Each error bar in B and C and GK represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 2.

TBX19 transcriptionally upregulates MFF and induces mitochondrial fission and promotes mitophagy in LCICs. A, Schematic of the CRISPR CAPTURE approach following the quantitative mass spectrometry to identify the trans-regulatory factors targeting MFF in LCICs. B, Real-time analysis of the mRNA expression of MFF in TBX19-, PRMT1-, RNAPII-, or TFIID-silenced LCICs. The mRNA expression level was normalized to GAPDH. C, ChIP assay analysis of the enrichment of TBX19 and PRMT1 on the MFF promoter in LCICs compared with non-LCICs. D, IB analysis of TBX19, PRMT1, and MFF levels in two liver progenitor cells, LCICs, and non-LCICs derived from two patient-derived HCC tissues and two HCC cell lines. GAPDH served as the loading control. E, IB analysis of expression of mitochondrial MFF, p-DRP1 (S616), total DRP1, and total MFF in the indicated cells. TOM20 served as the mitochondrial loading control, and GAPDH served as the total loading control. F, Mitochondrial morphology was visualized using MitoTracker probe in the indicated cells. Scale bars, top, 5 μm; bottom, 2 μm. G, Left, a diagram showing that the Mito./Nuc. area ratio was quantified by segregating the DAPI and MitoTracker channels and subsequent quantification of the area of each channel. Middle and right, representative images of mitochondria and quantification of Mito./Nuc. area ratio in control and TBX19-overexpressing non-LCICs. Scale bars, 5 μm. H, Quantification of mtDNA content in control and TBX19-overexpressing non-LCICs. I, TEM images of mitochondria (top) and quantification (bottom) of mitochondria engulfed in autophagosome in the indicated cells. Scale bars, 1 μm. M, mitochondria. J, Representative images (left; scale bars, 5 μm) and quantification (right) of the colocalization of LC3B and MitoTracker probe in the indicated cells determined by Manders overlap coefficients (MOC) using ImageJ software. The values on the y-axis mean the proportion of MitoTracker/LC3B colocalization in the total MitoTracker fluorescence region. K, Representative confocal images of anti-TOM20 (green) or p-DRP1 (S616) (red; scale bars, 5 μm) and quantification of colocalization of p-DRP1 and TOM20 in the indicated cells determined by MOC using ImageJ software. The values on the y-axis mean the proportion of TOM20/p-DRP1 colocalization in the total TOM20 fluorescence region. Each error bar in B and C and GK represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

TBX19 overexpression induces mitochondrial fission and promotes mitophagy

IF staining using the MitoTracker probe showed that the mitochondria became more fragmented and less tubular in TBX19-overexpressing non-LCICs (Fig. 2F; Supplementary Fig. S3B), suggesting that TBX19 contributed to mitochondrial fission. Consistently, the mitochondrial/nuclear ratio (Mito./Nuc. area ratio) and mitochondrial mtDNA, accompanied by mitochondrial ROS, were decreased in TBX19-overexpressing non-LCICs (Fig. 2G and H; Supplementary Fig. S3C). These phenomena were also found in LCICs compared with that in matched non-LCICs (Supplementary Fig. S3D and S3E). These results suggested that TBX19 overexpression might promote mitochondrial clearance.

Previously, it was reported that mitophagy, representing selective mitochondrial autophagy, plays a critical role in mitochondrial clearance and maintenance of stem cells and cancer stem cells (CSC; refs. 29, 30). We therefore examined whether TBX19 overexpression induced mitophagy. TEM analysis showed that mitochondria in TBX19-overexpressing non-LCICs displayed increased numbers of mitochondrial engulfed in autophagosomes (Fig. 2I). Correspondingly, IF staining showed that upregulation of TBX19 significantly increased LC3B-positive signals and the colocalization of LC3B with the MitoTracker probe in LCICs (Fig. 2J). Therefore, these results indicate that TBX19 overexpression promotes mitophagy. However, the inducing effect of TBX19 overexpression on mitochondrial fission was significantly abrogated by MFF ablation (Fig. 2K; Supplementary Fig. S3F), demonstrating that TBX19-mediated mitochondrial fission functions through upregulation of MFF.

Silencing TBX19 inhibited mitochondrial fission

Consistently, we found that silencing TBX19 in LCICs significantly decreased the expression of mitochondrial MFF and total/phosphorylated DRP1 (Fig. 3A), and also converted the LCIC mitochondrial morphology into elongated, tubular structures in LCICs (Fig. 3B). Importantly, the TBX19 downregulation induced increasing of mitochondrial content in LCICs was significantly rescued by exogenous MFF transduction, as indicated by reduced ratio of Mito./Nuc. area and mtDNA/nDNA (Fig. 3C,E). Therefore, these results provided further evidence that TBX19/MFF axis induced mitochondrial fission.

Figure 3.

Silencing TBX19 inhibited mitochondrial fission. A, IB analysis of expression of mitochondrial MFF, p-DRP1 (S616), total DRP1, and total MFF in the indicated cells. TOM20 served as the mitochondrial loading control, and GAPDH served as the total loading control. B, Mitochondrial morphology was visualized using MitoTracker probe in the indicated cells. Scale bars, top, 5 μm; bottom, 2 μm. C, IB analysis of MFF expression in the indicated cells. GAPDH was used as protein loading control. D, Immunofluorescence images of mitochondria (left) and quantification of Mito./Nuc. area ratio (right) in the indicated cells. Scale bars, 5 μm. E, Quantification of mtDNA content in the indicated cells. Each error bar in B, D, and E represents the mean ± SD of three independent experiments. *, P < 0.05; ***, P < 0.001.

Figure 3.

Silencing TBX19 inhibited mitochondrial fission. A, IB analysis of expression of mitochondrial MFF, p-DRP1 (S616), total DRP1, and total MFF in the indicated cells. TOM20 served as the mitochondrial loading control, and GAPDH served as the total loading control. B, Mitochondrial morphology was visualized using MitoTracker probe in the indicated cells. Scale bars, top, 5 μm; bottom, 2 μm. C, IB analysis of MFF expression in the indicated cells. GAPDH was used as protein loading control. D, Immunofluorescence images of mitochondria (left) and quantification of Mito./Nuc. area ratio (right) in the indicated cells. Scale bars, 5 μm. E, Quantification of mtDNA content in the indicated cells. Each error bar in B, D, and E represents the mean ± SD of three independent experiments. *, P < 0.05; ***, P < 0.001.

Close modal

Overexpression of TBX19 enhances tumor-initiating cell traits in liver cancer cells

Mitochondrial fission is reported to be involved in self-renewal and tumor-initiating capability of CICs (5–7), which prompted us to examine the effect of TBX19 on the properties of LCICs. Overexpression of TBX19 significantly enhanced the tumorsphere formation ability of non-LCICs, whereas silencing TBX19 significantly decreased the tumor-initiating cell (TIC) traits of LCICs, as indicated by fewer and smaller tumorsphere formation (Fig. 4A; Supplementary Fig. S4A–S4C). Importantly, an in vivo limiting dilution assay showed that TBX19-overexpressing non-LCICs exhibited a significantly higher tumor incidence and tumor-forming capacity, whereas TBX19-silenced LCICs displayed a markedly lower capacity to form tumors (Fig. 4B). SP assay, a flow cytometry method based on the dye efflux properties of ABC transporters was further performed to examine the effect of TBX19 on SP+ population, a subpopulation of cancer cells that have been identified in cancers and display increased capacity of self-renewal and tumorigenicity (31, 32). As shown in Supplementary Fig. S4D, silencing TBX19 significantly reduced the percentage of SP+ population. In line with the notion that mitochondrial fission–mediated asymmetric stem cell division leads to functional inheritance of stem cells and CSCs (16). We also observed that the frequency of asymmetric cell division was significantly increased in TBX19-overexpressing non-LCICs but reduced in TBX19-silenced LCICs (Supplementary Fig. S4E and S4F). These results demonstrate that TBX19 contributes to the tumor-forming and self-renewal capabilities of HCC cells.

Figure 4.

TBX19 overexpression enhances the TIC traits of non-LCICs. A, Representative images (top) and quantification (bottom) of tumorspheres formed by vector and TBX19-transduced non-LCICs (scale bars, 50 μm). B, Representative images of tumors formed by the indicated cells in NOG mice (top); TIC frequency was calculated on the basis of the ELDA analysis (bottom). C, Quantification of tumorspheres formed by TBX19-silenced Hep3B/and SNU449/LCICs cells transfected with vector or MFF. D, IB analysis of DRP1 expression in vector-control and DRP1-silenced TBX19-overexpressing non-LCICs. GAPDH served as the loading control. E, The quantification of tumorspheres and the percentage of SP+ population in vector-control and DRP1-silenced TBX19-overexpressing non-LCICs. F, Quantification of the OCR and ECAR in the indicated cells. G, Mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator. H, IB analysis of the levels of TBX19, oxidized OCT4, total OCT4, and NANOG in the indicated cells. The level of oxidized OCT4 was determined in the indicated cells incubated with biotin-maleimide to label reduced thiols that were further immunoprecipitated by streptavidin–sepharose beads, and then IB analysis was performed using anti-OCT4 antibody. GAPDH served as the loading control. Each error bar in A, C, and EG represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 4.

TBX19 overexpression enhances the TIC traits of non-LCICs. A, Representative images (top) and quantification (bottom) of tumorspheres formed by vector and TBX19-transduced non-LCICs (scale bars, 50 μm). B, Representative images of tumors formed by the indicated cells in NOG mice (top); TIC frequency was calculated on the basis of the ELDA analysis (bottom). C, Quantification of tumorspheres formed by TBX19-silenced Hep3B/and SNU449/LCICs cells transfected with vector or MFF. D, IB analysis of DRP1 expression in vector-control and DRP1-silenced TBX19-overexpressing non-LCICs. GAPDH served as the loading control. E, The quantification of tumorspheres and the percentage of SP+ population in vector-control and DRP1-silenced TBX19-overexpressing non-LCICs. F, Quantification of the OCR and ECAR in the indicated cells. G, Mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator. H, IB analysis of the levels of TBX19, oxidized OCT4, total OCT4, and NANOG in the indicated cells. The level of oxidized OCT4 was determined in the indicated cells incubated with biotin-maleimide to label reduced thiols that were further immunoprecipitated by streptavidin–sepharose beads, and then IB analysis was performed using anti-OCT4 antibody. GAPDH served as the loading control. Each error bar in A, C, and EG represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

MFF is prerequisite for TBX19-induced TIC traits in liver cancer cells

However, TBX19 overexpression–induced tumorsphere formation in non-LCICs was significantly impaired by MFF knockdown (Fig. 4A; Supplementary Fig. S4A). Meanwhile, the inhibitory effects of TBX19 silencing on LCIC traits and asymmetric cell division were rescued by ectopic expression of MFF (Fig. 4C; Supplementary Fig. S4F and S4G). These results indicate that MFF is a prerequisite for TBX19-induced TIC traits in liver cancer cells. Consistent with the effect of silencing MFF, inhibition of DRP1 using siRNAs or mdivi-1 or treated with the mitophagy inhibitor chloroquine or a pharmacologic inhibitor of DRP1 significantly abolished the stimulating effect of TBX19 on TIC traits, as indicated by the decreased SP and number of tumorspheres (Fig. 4D and E; Supplementary Fig. S4H–S4J). Therefore, these data provided further evidence that the TBX19/MFF axis–induced mitochondrial fission contributed to the LCIC traits.

The TBX19/MFF axis upregulates OCT4 via metabolic reprogramming

Mitochondrial fission is necessary for both pluripotential and metabolic reprogramming (10), in which a metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis has been found to be beneficial for stem cells, including CSCs, for self-renew and maintenance of stem cell states (33). Consistently, overexpression of TBX19 significantly suppressed OXPHOS in non-LCICs, as indicated by a reduced oxygen consumption rate (OCR), but increased the glycolytic activity, as demonstrated by the elevated extracellular acidification rate (ECAR; Fig. 4F; Supplementary Fig. S5A), indicating that the metabolic profile of TBX19-overexpressing non-LCICs shifted from OXPHOS to glycolysis. The mitochondrial ROS level also decreased significantly in TBX19-overexpressing non-LCICs, but increased in TBX19-silenced LCICs (Fig. 4G; Supplementary Fig. S5B). Importantly, silencing MFF dramatically abolished the effects of TBX19 on the OXPHOS/glycolysis switch and reversed the change in the mitochondrial ROS level (Fig. 4F,G; Supplementary Fig. S5A and S5B), suggesting that TBX19/MFF-induced mitochondrial fission promoted metabolic reprogramming from OXPHOS to glycolysis.

Strikingly, the levels of the pluripotency factors OCT4 and NANOG, which are key regulatory genes that initiate stem cell characteristics in liver cancer (23, 34–36), increased significantly in the TBX19-overexpressing non-LCICs, but were decreased in the TBX19-silenced LCICs. However, silencing MFF abrogated the inductive effect of TBX19 on OCT4 and NANOG levels (Fig. 4H; Supplementary Fig. S5C and S5D). Interestingly, TBX19 and MFF had no impact on the OCT4 mRNA level (Supplementary Fig. S5D), suggesting that the TBX19/MFF axis–mediated OCT4 upregulation might act via posttranscriptional regulation. We found that expression of oxidized OCT4 decreased in TBX19-overexpressing non-LCICs and increased in TBX19-silenced LCICs (Fig. 4H; Supplementary Fig. S5C). Meanwhile, treatment with cell-permeable glutathione (GSH), which phenocopied MFF overexpression, also resulted in a reduction in oxidized OCT4 levels (Fig. 4H; Supplementary Fig. S5C). Therefore, these results demonstrate that the TBX19/MFF axis–mediated metabolic switch from OXPHOS to glycolysis results in mitochondrial ROS reduction and prevents ROS-mediated OCT4 degradation.

TBX19 overexpression correlates with poor prognosis and therapeutic failure in HCC

Analysis of two public HCC datasets (TCGA-LIHC and GSE25097) revealed that TBX19 was significantly upregulated in HCC tissues (Supplementary Fig. S6A and S6B), and patients with higher TBX19-expressing HCC had poorer overall survival and a higher rate of HCC recurrence than those with lower TBX19-expressed HCC (P = 0.001, P < 0.001; P < 0.001, P < 0.001; Supplementary Fig. S6C and S6D). Notably, increased TBX19 levels also correlated with shorter progression-free and disease-specific survival in HCC patient, as well as shorter progression and relapse-free survival in patients treated with sorafenib (P < 0.001, P < 0.001; P < 0.001, P = 0.010; Supplementary Fig. S6E and S6F). Taken together, these results indicate that TBX19 upregulation is associated with worse prognosis and leads to therapeutic failure in HCC.

PRMT1 contributes epigenetically to TBX19-mediated MFF upregulation

Our previous dCas9 CAPTURE/MS analysis showed that PRMT1 was also enriched on the MFF promoter and silencing PRMT1 resulted in a reduction in MFF expression (Fig. 2A–C; Supplementary Fig. S3A). Although silencing PRMT1 significantly abrogated the inductive effect of TBX19 on the mRNA and protein levels of MFF in non-LCICs, overexpressing PRMT1 did not increase MFF expression at mRNA and protein levels in TBX19-silenced LCICs (Fig. 5A and B). These results suggested that PRMT1-mediated MFF upregulation was dependent on TBX19. Consistent with this hypothesis, ChIP assays showed that overexpressing TBX19 in non-LCICs significantly increased, but silencing TBX19 in LCICs decreased, enrichment of PRMT1 on MFF promoter (Fig. 5C; Supplementary Fig. S7A). However, dysregulation of PRMT1 has no impact on the association of TBX19 with MFF promoter. Furthermore, reciprocal coimmunoprecipitation (co-IP) analysis and far-Western blotting analyses showed that TBX19 directly interacted with PRMT1 (Fig. 5D and E), and co-IP assays using a series of truncated forms of TBX19 indicated that the N-terminal domain of TBX19 mediated the protein–protein interaction with PRMT1 (Fig. 5F). Taken together, these results demonstrate that TBX19 recruits PRMT1 to the MFF promoter.

Figure 5.

PRMT1 contributes epigenetically to TBX19-mediated MFF upregulation. A, Real-time analysis of the mRNA expression of MFF in the indicated cells. The mRNA expression was normalized to GAPDH. B, IB analysis of MFF and PRMT1 protein levels in the indicated cells. GAPDH served as a loading control. C, ChIP assay analysis of enrichment of PRMT1 in TBX19-silenced LCICs and in TBX19-overexpressing non-LCICs (top) and enrichment of TBX19 in PRMT1-silenced LCICs and in PRMT1-overexpressing non-LCICs (bottom). D, Reciprocal co-IP analysis revealed an interaction of TBX19 with PRMT1 endogenously and exogenously. E, Far-Western blotting analysis showed that Flag-tagged TBX19 interacted with recombinant His-PRMT1 (rPRMT1). F, Schematic illustration of the full-length and truncated TBX19 protein (top); co-IP assays were performed using anti-HA antibody in the cells transfected with HA-tagged PRMT1 and Flag-tagged TBX19 or truncated TBX19 fragments (bottom). G, ChIP assay analysis of the enrichment of H4R3me2a, H2AR3me2a, and H4R17me1 on the MFF promoter in the TBX19-overexpressing LCICs transfected with control or PRMT1-siRNA, or treated with vehicle or the PRMT1 inhibitor furamidine. H, ChIP assay analysis of the enrichment of H3K9ac, TFIID, and RNAPII on the MFF promoter in the TBX19-overexpressing LCICs transfected with control or PRMT1-siRNA, or treated with vehicle or the PRMT1 inhibitor furamidine. The heatmap represented by pseudocolors was generated using the ChIP–qPCR values, arrayed from green (no enrichment) to red (maximal enrichment), to demonstrate the histone methylarginine code surrounding the promoter of MFF. Each error bar in A, C, and H represents the mean ± SD of three independent experiments. **, P < 0.01; ***, P < 0.001.

Figure 5.

PRMT1 contributes epigenetically to TBX19-mediated MFF upregulation. A, Real-time analysis of the mRNA expression of MFF in the indicated cells. The mRNA expression was normalized to GAPDH. B, IB analysis of MFF and PRMT1 protein levels in the indicated cells. GAPDH served as a loading control. C, ChIP assay analysis of enrichment of PRMT1 in TBX19-silenced LCICs and in TBX19-overexpressing non-LCICs (top) and enrichment of TBX19 in PRMT1-silenced LCICs and in PRMT1-overexpressing non-LCICs (bottom). D, Reciprocal co-IP analysis revealed an interaction of TBX19 with PRMT1 endogenously and exogenously. E, Far-Western blotting analysis showed that Flag-tagged TBX19 interacted with recombinant His-PRMT1 (rPRMT1). F, Schematic illustration of the full-length and truncated TBX19 protein (top); co-IP assays were performed using anti-HA antibody in the cells transfected with HA-tagged PRMT1 and Flag-tagged TBX19 or truncated TBX19 fragments (bottom). G, ChIP assay analysis of the enrichment of H4R3me2a, H2AR3me2a, and H4R17me1 on the MFF promoter in the TBX19-overexpressing LCICs transfected with control or PRMT1-siRNA, or treated with vehicle or the PRMT1 inhibitor furamidine. H, ChIP assay analysis of the enrichment of H3K9ac, TFIID, and RNAPII on the MFF promoter in the TBX19-overexpressing LCICs transfected with control or PRMT1-siRNA, or treated with vehicle or the PRMT1 inhibitor furamidine. The heatmap represented by pseudocolors was generated using the ChIP–qPCR values, arrayed from green (no enrichment) to red (maximal enrichment), to demonstrate the histone methylarginine code surrounding the promoter of MFF. Each error bar in A, C, and H represents the mean ± SD of three independent experiments. **, P < 0.01; ***, P < 0.001.

Close modal

The arginine methyltransferase PRMT1 plays roles in transcriptional regulation via diverse histone methylation modifications, such as histone H2AR3, H4R17, and H4R3 (37–39). ChIP assays revealed that overexpressing TBX19 significantly increased level of H4R3me2a, but not H4R17me1a and H2AR3me2a, on the MFF promoter. Meanwhile, either silencing PRMT1 or treatment with the PRMT1 special inhibitor furamidine drastically abolished the inductive effect of TBX19 on H4R3me2a expression (Fig. 5G). The histone H4R3me2a modification could induce histone H3 acetylation on Lys9/Lys14 or histone H4 acetylation on Lys8/Lys12, resulting in the general transcriptional machinery binding to promoters and initiating transcription (40, 41). In line with these reports, we observed that TBX19 overexpression significantly increased the enrichment of H3K9ac, RNAPII, and TFIID on the MFF promoter, which was abrogated by PRMT1 knockdown or furamidine treatment (Fig. 5H). Consistent with the effect of PRMT1 silencing, furamidine treatment also resulted in a reduction of MFF in LCICs (Supplementary Fig. S7B and S7C). Therefore, these results suggest that PRMT1 contributes epigenetically to TBX19-mediated MFF upregulation.

PRMT1 promotes TBX19-mediated mitochondrial fission

Consistent with the results that PRMT1 enhanced TBX19-mediated MFF upregulation, overexpressing PRMT1 in non-LCICs significantly promoted, but silencing PRMT1 reduced, mitochondrial fragmentation and colocalization of LC3B with mitochondria (Fig. 6A and B; Supplementary Fig. S8A–S8C). These results indicated that PRMT1 was also involved in mitochondrial fission and mitophagy. Correspondingly, PRMT1-overexpressing non-LCICs displayed increased asymmetric stem cell division, but reduced mitochondrial ROS stress (Fig. 6C and D). However, overexpressing PRMT1 in TBX19-silenced non-LCICs or overexpressing TBX19 in PRMT1-silenced non-LCICs failed to promote mitochondrial fission and mitophagy (Fig. 6A and B; Supplementary Fig. S8A–S8E). These results further demonstrate that TBX19 and PRMT1 form a complex that has a collaborative effect on mitochondrial fission.

Figure 6.

PRMT1 promotes TBX19-mediated mitochondrial fission. A, Representative images of mitochondrial morphology and LC3B (left), and quantification of the mitochondrial length (right) in the indicated cells. Scale bars, 5 μm. B, Quantification of the colocalization of LC3B and MitoTracker probe in the indicated cells determined by Manders overlap coefficients using ImageJ software. The values on the y-axis mean the proportion of MitoTracker/LC3B colocalization in the total MitoTracker fluorescence region. C, The frequency of asymmetric and symmetric cell division was quantified in the indicated cells. D, Mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator. E, Representative images (left) and quantification (right) of tumorspheres formed by control and PRMT1-silenced LCICs. Scale bars, 50 μm. F, Representative images of tumors formed by the indicated cells in NOG mice (left) and the frequency of TICs in the indicated cells (right). Each error bar in AE represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 6.

PRMT1 promotes TBX19-mediated mitochondrial fission. A, Representative images of mitochondrial morphology and LC3B (left), and quantification of the mitochondrial length (right) in the indicated cells. Scale bars, 5 μm. B, Quantification of the colocalization of LC3B and MitoTracker probe in the indicated cells determined by Manders overlap coefficients using ImageJ software. The values on the y-axis mean the proportion of MitoTracker/LC3B colocalization in the total MitoTracker fluorescence region. C, The frequency of asymmetric and symmetric cell division was quantified in the indicated cells. D, Mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator. E, Representative images (left) and quantification (right) of tumorspheres formed by control and PRMT1-silenced LCICs. Scale bars, 50 μm. F, Representative images of tumors formed by the indicated cells in NOG mice (left) and the frequency of TICs in the indicated cells (right). Each error bar in AE represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Consistent with the inhibitory effect of PRMT1 downregulation on mitochondrial fission, silencing PRMT1 drastically decreased the tumorsphere formation capability of LCICs (Fig. 6E; Supplementary Fig. S8F). Importantly, in vivo limiting dilution and serial transplantation assays revealed a significant reduction in tumor incidence and tumor-forming capacity in PRMT1-silenced LCICs compared with that of the control cells (Fig. 6F), which further supported the notion that PRMT1 contributes to the tumor-forming and self-renewal capabilities of HCC cells.

Pharmacologic blocking of PRMT1 inhibited the tumorigenicity of LCICs

Finally, we examined whether pharmacologic inhibition of PRMT1 could reduce the tumorigenesis capability of LCICs and prevented tumor progression. As shown in Fig. 7A–D, treatment with PRMT1 inhibitor, furamidine, dramatically converted the fragmented mitochondria in clinical HCC tissue-derived LCICs into tubular and prolonged shape, accompanied by a reduction of asymmetric division and mitochondrial oxidative stress. Furthermore, tumorsphere formation assay revealed that furamidine-treated LCICs formed obviously smaller and fewer tumorspheres than vehicle-treated LCICs (Fig. 7E). Strikingly, furamidine treatment completely inhibited the tumor formation by 1 × 102 LCICs and significantly reduced the tumor incidence by 5 × 105 LCICs (Fig. 7F). In addition, IF staining and IHC showed less MFF expression and TOM20/LC3B colocalization in furamidine-treated LCICs and tumors, suggesting that furamidine could suppress mitochondrial fission and mitophagy in vivo (Fig. 7G). Collectively, our results demonstrate that pharmacologic blocking PRMT1 could decrease the recurrence of liver cancer through inhibition of mitochondrial fission and mitophagy.

Figure 7.

Pharmacologic blocking of PRMT1 inhibits tumorigenicity of LCICs. A, IB analysis of MFF and PRMT1 expression in the indicated cells. GAPDH was used as protein loading control. B, Representative images of the morphology of mitochondria (left) and quantification of the mitochondrial length (right) in patient-derived LCICs treated with vehicle or the PRMT1 inhibitor furamidine. Scale bars, top, 5 μm; bottom, 2 μm. C, The frequency of asymmetric and symmetric cell division was quantified in patient-derived LCICs treated with vehicle or furamidine. D, The relative mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator in patient-derived LCICs treated with vehicle or furamidine. E, Representative images (left) and quantification (right) of tumorspheres formed by vehicle or furamidine-treated LCICs. Scale bars, 50 μm. F, Representative images of tumors (top) and the incidence of tumors (bottom) formed by patient-derived LCICs in NOG mice treated with vehicle or furamidine. G, Representative images (top) and quantification (bottom) of MFF, TOM20, and LC3B in vehicle or furamidine-treated LCIC-formed tumors analyzed by IHC and immunofluorescence assays. Each error bar in BE and G represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 7.

Pharmacologic blocking of PRMT1 inhibits tumorigenicity of LCICs. A, IB analysis of MFF and PRMT1 expression in the indicated cells. GAPDH was used as protein loading control. B, Representative images of the morphology of mitochondria (left) and quantification of the mitochondrial length (right) in patient-derived LCICs treated with vehicle or the PRMT1 inhibitor furamidine. Scale bars, top, 5 μm; bottom, 2 μm. C, The frequency of asymmetric and symmetric cell division was quantified in patient-derived LCICs treated with vehicle or furamidine. D, The relative mitochondrial ROS level was measured in the indicated cells stained with MitoSOX red mitochondrial superoxide indicator in patient-derived LCICs treated with vehicle or furamidine. E, Representative images (left) and quantification (right) of tumorspheres formed by vehicle or furamidine-treated LCICs. Scale bars, 50 μm. F, Representative images of tumors (top) and the incidence of tumors (bottom) formed by patient-derived LCICs in NOG mice treated with vehicle or furamidine. G, Representative images (top) and quantification (bottom) of MFF, TOM20, and LC3B in vehicle or furamidine-treated LCIC-formed tumors analyzed by IHC and immunofluorescence assays. Each error bar in BE and G represents the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

CICs are implicated in tumor initiation and progression, which are responsible for high therapeutic failure and recurrence in multiple types of cancers (42). Recently, mitochondrial fission has been reported to enable self-renewal of CICs in multiple human cancers (10, 15), and treatment with mdivi-1, which inhibits the GTPase activity of mitochondrial fission protein DRP1, was sufficient to suppress the propagation of stem-like cells in CICs, such as in brain tumors, prostate cancer, and nasopharyngeal carcinoma (6, 7). However, iPSCs also lost pluripotency when exposed to mdivi-1 treatment, indicating the drawback of mitochondrial fission target therapy when applied in cancer treatment, in which it might also impair normal stem cells (43). Therefore, to identify the pivotal factor that drives CIC-specific mitochondrial fission might be of great clinical value. In the present study, we identified MFF as the major candidate that promoted mitochondrial fission in LCICs, enabling self-renewal and enhanced tumorigenic capability of LCICs. Mechanistically, we identified the TBX19/PRMT1 axis as the central hub for upregulating MFF expression in LCICs, which allowed the application of the PRMT1 inhibitor, furamidine, to restrict the self-renewal and tumorigenesis capacity of LCICs. More importantly, the expression levels of TBX19 and PRMT1 were much higher in LCICs than in non-LCICs and normal liver stem cells, which might eliminate the simultaneous inhibition effect of furamidine for somatic stem cells. Therefore, our study reveals the distinct molecular mechanism of mitochondrial fission in LCICs, suggesting an efficient regimen for liver cancer treatment.

Fission-related proteins required for mitochondrial fission have been discovered in mammals (3, 10). For instance, DRP1 activation–mediated mitochondrial fragmentation is crucial for iPS cell reprogramming (13). Silencing FIS1 inhibited mitochondrial fission–mediated mitochondrial elongation in senescent cells and AML stem cells (19, 44). Downregulation of mitochondrial fission accessory proteins MIEF1/2 leads to mitochondrial elongation via blockage the interaction of DRP1 with mitochondria (45, 46). In our study, we examined 39 genes involved in mitochondrial fission in LCICs compared with non-LCICs, which revealed the indispensable role of MFF in regulating mitochondrial fission, whereas the expression level of other factors, including DRP1, FIS1, and MIEF1/2, showed no significant changes. Moreover, overexpression of MFF increased, whereas suppression of MFF reduced, mitochondrial segregation, asymmetric stem cell division, and mitophagy in LCICs. Importantly, it has been validated using in vivo and in vitro assays that stable MFF-depleted Hep3B and SNU449 CICs had disrupted mitochondrial fission and reduced tumor-initiating capability. Therefore, our study not only demonstrates the crucial role of MFF in mitochondrial fission, but also provides a basis for the discovery of molecular targets for mitochondrial fission–driven LCICs.

Homeostasis of mitochondrial dynamics is required for cellular reprogramming and differentiation in both stem cells and CSCs (3, 10). However, the mechanism regulating this requirement has not been unraveled. In this study, we found that mitochondrial fission modulated by the TBX19/MFF axis switched the metabolic process from OXPHOS to glycolysis and decreased mitochondrial ROS production. The reduction of ROS resulted in decreased oxidation modification of pluripotency factor OCT4 degradation, which in turn led to upregulation of OCT4 protein levels and its downstream target NANOG, and therefore sustained stemness and augmented the tumorigenicity of LCICs. These findings were consistent with a previous report that the ROS level correlates inversely with OCT4 expression in human embryonic stem cells (47). Therefore, our study shed light on the mechanism of mitochondrial fission and stemness maintenance in LCICs.

Recent studies have provided conflicting evidence regarding effects of mitochondrial fission versus fusion in cancer stem-like cells. For instance, it has been reported that several types of CSCs, such as BTICs, prostate CSCs, and leukemic stem cells, exhibited elevated mitochondrial fission activity and displayed a significantly more fragmented mitochondrial morphology compared with the more differentiated non-CSCs (5, 7, 19). They further demonstrated that inhibition of mitochondrial fission by silencing DRP1 not only decreased tumorsphere formation but also reduced expression of stemness markers, such as NANOG and Oct4 (7). Meanwhile, mitochondrial fission is also found to be associated with pro-survival mitophagy to clear defective mitochondria compromised by the mitochondrial stress generated from oncogenic transformation (19). Consistently, we also found that TBX19/PRMT1/MFF axis–mediated mitochondrial fission sustained the stemness and enhanced tumor-initiating capability of LCICs. However, it was also reported that unbalanced and abnormal activation of mitochondrial fission by aberrant MFF expression drive the impairment of mitochondrial metabolic function, leading to inhibition of breast CSCs (BCSC) propagation (48). Lee and colleagues (49) found that the chemotherapy-resistant BCSCs exhibited increased fusion activity and elongated mitochondrial network, which enhanced mitochondrial oxidative phosphorylation (mtOXPHOS) of BCSCs for survival after neoadjuvant chemotherapy. These results indicated that BCSCs may depend on mitochondrial fusion to adapt the demand of elevated mitochondrial biogenesis and metabolism. Therefore, mitochondria dynamics–associated CSCs might be highly context dependent and might also vary in different tumor environment and various states of the requirement. Moreover, a recent study of drosophila neural stem cell–derived tumor model reported that uncontrolled mitochondrial fusion is the rate-limiting step for irreversible immortalization of neural stem cells during tumor formation (50), which was distinct different from BTICs that depend on fission to maintain the TIC traits (7). Taken together, the opposite effects of mitochondrial fission and fusion on cancer stem-like cells might be caused by the response to distinct environment and meet the specific functional requirement.

In summary, our study provides strong evidence to support the hypothesis that the TBX19/PRMT1/MFF axis–induced mitochondrial fission evokes mitophagy and asymmetric stem cell division, and promotes the metabolic shift from OXPHOS to glycolysis in LCICs. This results in a reduction of mitochondrial ROS and the prevention of ROS-mediated OCT4 degradation. Understanding the precise role of TBX19 in the self-renewal of LCICs and the regulation of mitochondrial fission will not only advance our knowledge of liver cancer pathogenesis, but also will allow the development of new therapeutic strategies against liver cancer.

No disclosures were reported.

M. Tang: Data curation, validation. M. Yang: Data curation, validation. G. Wu: Validation, methodology. S. Mo: Methodology. X. Wu: Software. S. Zhang: Validation, methodology. R. Yu: Methodology. Y. Hu: Methodology. Y. Xu: Methodology. Z. Li: Methodology. X. Liao: Methodology. J. Li: Writing–original draft, project administration, writing–review and editing. L. Song: Writing–original draft, project administration, writing–review and editing.

This work was supported by National Key Research and Development Program of China (2020YFA0509400 to L. Song), Natural Science Foundation of China (81830082 and 82030078 to J. Li, 82003128 to G. Wu, 82072609 to L. Song, and 81773106 to L. Song), Guangzhou Science and Technology Plan Projects (201803010098 to J. Li), and China Postdoctoral Science Foundation (2019M653220 to G. Wu).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Zhao
J
,
Zhang
J
,
Yu
M
,
Xie
Y
,
Huang
Y
,
Wolff
DW
, et al
Mitochondrial dynamics regulates migration and invasion of breast cancer cells
.
Oncogene
2013
;
32
:
4814
24
.
2.
Simula
L
,
Nazio
F
,
Campello
S
. 
The mitochondrial dynamics in cancer and immune-surveillance
.
Semin Cancer Biol
2017
;
47
:
29
42
.
3.
Fu
W
,
Liu
Y
,
Yin
H
. 
Mitochondrial dynamics: biogenesis, fission, fusion, and mitophagy in the regulation of stem cell behaviors
.
Stem Cells Int
2019
;
2019
:
9757201
.
4.
Westermann
B
. 
Mitochondrial fusion and fission in cell life and death
.
Nat Rev Mol Cell Biol
2010
;
11
:
872
84
.
5.
Civenni
G
,
Bosotti
R
,
Timpanaro
A
,
Vazquez
R
,
Merulla
J
,
Pandit
S
, et al
Epigenetic control of mitochondrial fission enables self-renewal of stem-like tumor cells in human prostate cancer
.
Cell Metab
2019
;
30
:
303
18
.
6.
Zhou
TJ
,
Zhang
SL
,
He
CY
,
Zhuang
QY
,
Han
PY
,
Jiang
SW
, et al
Downregulation of mitochondrial cyclooxygenase-2 inhibits the stemness of nasopharyngeal carcinoma by decreasing the activity of dynamin-related protein 1
.
Theranostics
2017
;
7
:
1389
406
.
7.
Xie
Q
,
Wu
Q
,
Horbinski
CM
,
Flavahan
WA
,
Yang
K
,
Zhou
W
, et al
Mitochondrial control by DRP1 in brain tumor–initiating cells
.
Nat Neurosci
2015
;
18
:
501
10
.
8.
Caino
MC
,
Seo
JH
,
Aguinaldo
A
,
Wait
E
,
Bryant
KG
,
Kossenkov
AV
, et al
A neuronal network of mitochondrial dynamics regulates metastasis
.
Nat Commun
2016
;
7
:
13730
.
9.
Huang
Q
,
Zhan
L
,
Cao
H
,
Li
J
,
Lyu
Y
,
Guo
X
, et al
Increased mitochondrial fission promotes autophagy and hepatocellular carcinoma cell survival through the ROS-modulated coordinated regulation of the NFKB and TP53 pathways
.
Autophagy
2016
;
12
:
999
1014
.
10.
Seo
BJ
,
Yoon
SH
,
Do
JT
. 
Mitochondrial dynamics in stem cells and differentiation
.
Int J Mol Sci
2018
;
19
:
3893
.
11.
Otera
H
,
Wang
C
,
Cleland
MM
,
Setoguchi
K
,
Yokota
S
,
Youle
RJ
, et al
Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells
.
J Cell Biol
2010
;
191
:
1141
58
.
12.
Hoppins
S
,
Lackner
L
,
Nunnari
J
. 
The machines that divide and fuse mitochondria
.
Annu Rev Biochem
2007
;
76
:
751
80
.
13.
Prieto
J
,
Leon
M
,
Ponsoda
X
,
Sendra
R
,
Bort
R
,
Ferrer-Lorente
R
, et al
Early ERK1/2 activation promotes DRP1-dependent mitochondrial fission necessary for cell reprogramming
.
Nat Commun
2016
;
7
:
11124
.
14.
Kashatus
JA
,
Nascimento
A
,
Myers
LJ
,
Sher
A
,
Byrne
FL
,
Hoehn
KL
, et al
Erk2 phosphorylation of Drp1 promotes mitochondrial fission and MAPK-driven tumor growth
.
Mol Cell
2015
;
57
:
537
51
.
15.
Choi
HW
,
Kim
JH
,
Chung
MK
,
Hong
YJ
,
Jang
HS
,
Seo
BJ
, et al
Mitochondrial and metabolic remodeling during reprogramming and differentiation of the reprogrammed cells
.
Stem Cells Dev
2015
;
24
:
1366
73
.
16.
Katajisto
P
,
Dohla
J
,
Chaffer
CL
,
Pentinmikko
N
,
Marjanovic
N
,
Iqbal
S
, et al
Stem cells. asymmetric apportioning of aged mitochondria between daughter cells is required for stemness
.
Science
2015
;
348
:
340
3
.
17.
Garcia-Prat
L
,
Martinez-Vicente
M
,
Perdiguero
E
,
Ortet
L
,
Rodriguez-Ubreva
J
,
Rebollo
E
, et al
Autophagy maintains stemness by preventing senescence
.
Nature
2016
;
529
:
37
42
.
18.
Ito
K
,
Turcotte
R
,
Cui
J
,
Zimmerman
SE
,
Pinho
S
,
Mizoguchi
T
, et al
Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance
.
Science
2016
;
354
:
1156
60
.
19.
Pei
S
,
Minhajuddin
M
,
Adane
B
,
Khan
N
,
Stevens
BM
,
Mack
SC
, et al
AMPK/FIS1-mediated mitophagy is required for self-renewal of human AML stem cells
.
Cell stem cell
2018
;
23
:
86
100
.
20.
Ando
J
,
Saito
M
,
Imai
JI
,
Ito
E
,
Yanagisawa
Y
,
Honma
R
, et al
TBX19 is overexpressed in colorectal cancer and associated with lymph node metastasis
.
Fukushima J Med Sci
2017
;
63
:
141
51
.
21.
Chen
YF
,
Wang
JY
,
Wu
CH
,
Chen
FM
,
Cheng
TL
,
Lin
SR
. 
Detection of circulating cancer cells with K-ras oncogene using membrane array
.
Cancer Lett
2005
;
229
:
115
22
.
22.
Chen
CL
,
Tsukamoto
H
,
Liu
JC
,
Kashiwabara
C
,
Feldman
D
,
Sher
L
, et al
Reciprocal regulation by TLR4 and TGF-beta in tumor-initiating stem-like cells
.
J Clin Invest
2013
;
123
:
2832
49
.
23.
Chen
CL
,
Kumar
DBU
,
Punj
V
,
Xu
J
,
Sher
L
,
Tahara
SM
, et al
NANOG metabolically reprograms tumor-initiating stem-like cells through tumorigenic changes in oxidative phosphorylation and fatty acid metabolism
.
Cell Metab
2016
;
23
:
206
19
.
24.
Liu
X
,
Zhang
Y
,
Chen
Y
,
Li
M
,
Zhou
F
,
Li
K
, et al
In situ capture of chromatin interactions by biotinylated dCas9
.
Cell
2017
;
170
:
1028
43
.
25.
Wu
MJ
,
Chen
YS
,
Kim
MR
,
Chang
CC
,
Gampala
S
,
Zhang
Y
, et al
Epithelial–mesenchymal transition directs stem cell polarity via regulation of mitofusin
.
Cell Metab
2019
;
29
:
993
1002
.
26.
Flores-Tellez
TN
,
Villa-Trevino
S
,
Pina-Vazquez
C
. 
Road to stemness in hepatocellular carcinoma
.
World J Gastroenterol
2017
;
23
:
6750
76
.
27.
Zhu
Z
,
Hao
X
,
Yan
M
,
Yao
M
,
Ge
C
,
Gu
J
, et al
Cancer stem/progenitor cells are highly enriched in CD133+CD44+ population in hepatocellular carcinoma
.
Int J Cancer
2010
;
126
:
2067
78
.
28.
Yamashita
T
,
Budhu
A
,
Forgues
M
,
Wang
XW
. 
Activation of hepatic stem cell marker EpCAM by wnt-beta-catenin signaling in hepatocellular carcinoma
.
Cancer Res
2007
;
67
:
10831
9
.
29.
Lee
J
,
Liu
K
,
Stiles
B
,
Ou
JJ
. 
Mitophagy and hepatic cancer stem cells
.
Autophagy
2018
;
14
:
715
6
.
30.
Liu
K
,
Lee
J
,
Kim
JY
,
Wang
L
,
Tian
Y
,
Chan
ST
, et al
Mitophagy controls the activities of tumor suppressor p53 to regulate hepatic cancer stem cells
.
Mol Cell
2017
;
68
:
281
92
.
31.
Golebiewska
A
,
Brons
NH
,
Bjerkvig
R
,
Niclou
SP
. 
Critical appraisal of the side population assay in stem cell and cancer stem cell research
.
Cell stem cell
2011
;
8
:
136
47
.
32.
Behbod
F
,
Vivanco
MD
. 
Side population
.
Methods Mol Biol
2015
;
1293
:
73
81
.
33.
Lisowski
P
,
Kannan
P
,
Mlody
B
,
Prigione
A
. 
Mitochondria and the dynamic control of stem cell homeostasis
.
EMBO Rep
2018
;
19
:
e45432
.
34.
Zhu
P
,
Wang
Y
,
He
L
,
Huang
G
,
Du
Y
,
Zhang
G
, et al
ZIC2-dependent OCT4 activation drives self-renewal of human liver cancer stem cells
.
J Clin Invest
2015
;
125
:
3795
808
.
35.
Qian
YW
,
Chen
Y
,
Yang
W
,
Fu
J
,
Cao
J
,
Ren
YB
, et al
p28(GANK) prevents degradation of Oct4 and promotes expansion of tumor-initiating cells in hepatocarcinogenesis
.
Gastroenterology
2012
;
142
:
1547
58
.
36.
Shan
J
,
Shen
J
,
Liu
L
,
Xia
F
,
Xu
C
,
Duan
G
, et al
Nanog regulates self-renewal of cancer stem cells through the insulin-like growth factor pathway in human hepatocellular carcinoma
.
Hepatology
2012
;
56
:
1004
14
.
37.
Fulton
MD
,
Brown
T
,
Zheng
YG
. 
Mechanisms and inhibitors of histone arginine methylation
.
Chem Rec
2018
;
18
:
1792
807
.
38.
Tang
J
,
Frankel
A
,
Cook
RJ
,
Kim
S
,
Paik
WK
,
Williams
KR
, et al
PRMT1 is the predominant type I protein arginine methyltransferase in mammalian cells
.
J Biol Chem
2000
;
275
:
7723
30
.
39.
Chen
D
,
Ma
H
,
Hong
H
,
Koh
SS
,
Huang
SM
,
Schurter
BT
, et al
Regulation of transcription by a protein methyltransferase
.
Science
1999
;
284
:
2174
7
.
40.
Li
X
,
Hu
X
,
Patel
B
,
Zhou
Z
,
Liang
S
,
Ybarra
R
, et al
H4R3 methylation facilitates beta-globin transcription by regulating histone acetyltransferase binding and H3 acetylation
.
Blood
2010
;
115
:
2028
37
.
41.
Wang
H
,
Huang
ZQ
,
Xia
L
,
Feng
Q
,
Erdjument-Bromage
H
,
Strahl
BD
, et al
Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor
.
Science
2001
;
293
:
853
7
.
42.
Clevers
H
. 
The cancer stem cell: premises, promises, and challenges
.
Nat Med
2011
;
17
:
313
9
.
43.
Vazquez-Martin
A
,
Cufi
S
,
Corominas-Faja
B
,
Oliveras-Ferraros
C
,
Vellon
L
,
Menendez
JA
. 
Mitochondrial fusion by pharmacological manipulation impedes somatic cell reprogramming to pluripotency: new insight into the role of mitophagy in cell stemness
.
Aging
2012
;
4
:
393
401
.
44.
Mai
S
,
Klinkenberg
M
,
Auburger
G
,
Bereiter-Hahn
J
,
Jendrach
M
. 
Decreased expression of Drp1 and Fis1 mediates mitochondrial elongation in senescent cells and enhances resistance to oxidative stress through PINK1
.
J Cell Sci
2010
;
123
:
917
26
.
45.
Zhao
J
,
Liu
T
,
Jin
S
,
Wang
X
,
Qu
M
,
Uhlen
P
, et al
Human MIEF1 recruits Drp1 to mitochondrial outer membranes and promotes mitochondrial fusion rather than fission
.
EMBO J
2011
;
30
:
2762
78
.
46.
Palmer
CS
,
Osellame
LD
,
Laine
D
,
Koutsopoulos
OS
,
Frazier
AE
,
Ryan
MT
. 
MiD49 and MiD51, new components of the mitochondrial fission machinery
.
EMBO Rep
2011
;
12
:
565
73
.
47.
Marsboom
G
,
Zhang
GF
,
Pohl-Avila
N
,
Zhang
Y
,
Yuan
Y
,
Kang
H
, et al
Glutamine metabolism regulates the pluripotency transcription factor OCT4
.
Cell Rep
2016
;
16
:
323
32
.
48.
Sanchez-Alvarez
R
,
De Francesco
EM
,
Fiorillo
M
,
Sotgia
F
,
Lisanti
MP
. 
Mitochondrial fission factor (MFF) inhibits mitochondrial metabolism and reduces breast cancer stem cell (CSC) activity
.
Front Oncol
2020
;
10
:
1776
.
49.
Lee
KM
,
Giltnane
JM
,
Balko
JM
,
Schwarz
LJ
,
Guerrero-Zotano
AL
,
Hutchinson
KE
, et al
MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells via regulation of mitochondrial oxidative phosphorylation
.
Cell Metab
2017
;
26
:
633
47
.
50.
Bonnay
F
,
Veloso
A
,
Steinmann
V
,
Kocher
T
,
Abdusselamoglu
MD
,
Bajaj
S
, et al
Oxidative metabolism drives immortalization of neural stem cells during tumorigenesis
.
Cell
2020
;
182
:
1490
507
.