Psmd14-mediated deubiquitination of carm1 facilitates the proliferation and metastasis of hepatocellular carcinoma by inducing the transcriptional activation of fermt1

Nature

Psmd14-mediated deubiquitination of carm1 facilitates the proliferation and metastasis of hepatocellular carcinoma by inducing the transcriptional activation of fermt1"


Play all audios:

Loading...

ABSTRACT Hepatocellular carcinoma (HCC) is a highly potent malignancy. The enzyme coactivator-associated arginine methyltransferase 1 (CARM1) is highly expressed in different types of


cancer. However, the precise levels of expression, clinical significance, biological functions, and molecular mechanisms of CARM1 in HCC, particularly related to the downstream genes


regulated by CARM1 through histone arginine methylation, remain unclear. In this study, we presented findings from the TCGA database and clinical samples, which collectively demonstrated the


overexpression of CARM1 in HCC. Additionally, we found that the upregulation of CARM1 was mediated by PSMD14-induced deubiquitination. CARM1 promoted the proliferation and metastasis of HCC


cells in vitro and in vivo. Mechanistic investigations further revealed that FERMT1 is a downstream gene of CARM1, and CARM1 activates the transcription of FERMT1 through the dimethylation


of arginine 17 on histone 3 (H3R17me2). Additionally, administering SGC2085, a CARM1 inhibitor, effectively suppressed the malignant behaviors of HCC cells. To summarize, our findings


provided strong evidence that CARM1 can serve as a key oncoprotein; thus, it holds promise as a therapeutic target for HCC. SIMILAR CONTENT BEING VIEWED BY OTHERS SMYD3 PROMOTES


HEPATOCELLULAR CARCINOMA PROGRESSION BY METHYLATING S1PR1 PROMOTERS Article Open access 23 July 2021 PRMT1-MEDIATED METHYLATION OF ME2 PROMOTES HEPATOCELLULAR CARCINOMA GROWTH BY INHIBITING


UBIQUITINATION Article Open access 11 November 2024 ABCF1-K430-LACTYLATION PROMOTES HCC MALIGNANT PROGRESSION VIA TRANSCRIPTIONAL ACTIVATION OF HIF1 SIGNALING PATHWAY Article 03 January 2025


INTRODUCTION Coactivator-associated arginine methyltransferase 1 (CARM1), also known as protein arginine methyltransferase 4 (PRMT4), was originally identified as a protein that binds to


the p160 steroid receptor coactivator GRIP1 [1]. CARM1 has an N-terminal pleckstrin homology-like domain (PH-like), a C-terminal transactivation domain, and a central catalytic domain


containing four conserved PRMT motifs [2]. It regulates various cellular processes, including transcriptional regulation, pre-mRNA splicing, the cell cycle, and DNA damage repair


[3,4,5,6,7]. CARM1 methylates arginine 17 (H3R17) and H3R26 of histone 3 and also forms transcriptional complexes with RNA polymerase II complex component 1 (PAF1), androgen receptor (AR),


estrogen receptor (ER), and histone acetyltransferase P300 to activate downstream gene expression [8,9,10]. CARM1 also methylates non-histone proteins, regulating their functions. For


example, CARM1 activates aerobic glycolysis and promotes tumor development by methylating the key glycolytic enzyme PKM2 [11]. It facilitates the growth and metastasis of breast cancer by


methylating arginine 1064 of the chromatin remodeling factor BAF155 [12]. Additionally, CARM1 enhances sensitivity to chemotherapeutic drugs in breast cancer cells by methylating MED12 [13].


However, the expression level, clinical significance, biological function, and molecular mechanism of CARM1 in hepatocellular carcinoma (HCC), particularly the downstream genes regulated by


CARM1 through histone arginine methylation, remain unclear. Studies have found that CARM1 can be regulated by post-translational modifications. CARM1 is phosphorylated on several residues,


such as T131, which is phosphorylated by glycogen synthase kinase 3 beta to prevent proteasomal degradation [14]. Some post-translational modifications of CARM1 are important [15]. For


example, phosphorylation of serine 217 of CARM1 inhibits the methylation activity of PRMT4 [16, 17]; O-acetylglycosylation modulates the substrate specificity of CARM1 [18]. CARM1 undergoes


methylation, affecting its transcriptional activation and pre-mRNA splicing functions [15]; nuclear AMPK regulates the ubiquitination-mediated degradation of CARM1 through the E3 ligase SKP2


[19]. The ubiquitin-proteasome system is one of the main pathways for degrading proteins. Under the sequential actions conducted by several enzymes, including activating (E1), conjugating


(E2), and ligating (E3) enzymes, ubiquitin (Ub) is attached to the lysine residue of substrate proteins through monoubiquitination and polyubiquitination [20]. Ubiquitination can be reversed


by deubiquitinating enzymes (DUBs), and dysregulation of DUBs can cause various diseases, including cancer [21]. The deubiquitinase PSMD14, the 26S proteasome non-ATPase regulatory subunit


1, belongs to the JAMM domain protease family [22]. High expression levels of PSMD14 also predict poor prognosis in various types of cancer [22, 23]. PSMD14 plays important roles in various


biological processes, such as proliferation, metastasis, DNA damage response, immune tolerance, and multidrug resistance [22,23,24,25]. In HCC, PSMD14 stabilizes E2F1, which upregulates the


expression of survivin and FOXM1, thereby facilitating the growth of HCC [26]. Additionally, PSMD14 deubiquitinates the TGF-β receptor and caveolin-1 to facilitate HCC metastasis [27]. In


another study, we revealed that PSMD14 deubiquitinates and stabilizes GRB2 to promote the growth and metastasis of HCC [28]. By conducting mass spectrometric analysis, we identified CARM1 as


a potential PSMD14-interacting protein. However, the regulatory relationship between PSMD14 and CARM1 remains unclear. In this study, we investigated the clinical significance of CARM1


upregulation in HCC tissue samples and determined the oncogenic role of CARM1 in HCC. Our findings revealed that PSMD14 interacts with CARM1, which prevents its degradation. We also found


that FERMT1 serves as a downstream gene of CARM1-mediated histone methylation in HCC, influencing the proliferation and metastasis of HCC. Additionally, the CARM1 inhibitor SGC2085 strongly


inhibited HCC. Our findings revealed a novel PSMD14-CARM1-FERMT1 axis that facilitates the progression of HCC. MATERIALS AND METHODS TISSUE COLLECTION We collected 66 pairs of HCC tissues


and their adjacent normal liver tissues from the Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University. These tissues were obtained from patients who underwent


curative hepatectomy from 2016 to 2020. The collected samples were immediately frozen with liquid nitrogen and stored at –80 °C. All patients provided informed consent and none underwent


prior treatments such as chemotherapy, targeted therapy, or interventional therapy. The study was approved by the ethics committee of Zhongshan Hospital of Xiamen University. CELL CULTURE


PLC/PRF/5, Huh7, SK-Hep-1, and HEK293T cells were purchased from Cellcock Company (Guangzhou China). These cell lines were compared with those in the STR database for authentication. All


cell lines were cultured with high-glucose Dulbecco’s modified Eagle medium (DMEM, Gibco) supplemented with 10% fetal bovine serum (FBS, Gibco) and 1% penicillin/streptomycin (Gibco) at 37 


°C in a humidified incubator with 5% CO2. IMMUNOHISTOCHEMISTRY (IHC) Tissues were fixed with 10% neutral formalin, embedded in paraffin, and cut into sections (4 μm thick) for IHC staining.


Serial sections were deparaffinized, hydrated, and incubated in 3% H2O2 for 20 min at room temperature and then stained with anti-PSMD14 (1:3200, A9608, ABclonal) and anti-CARM1 (1:1000,


NB200-342, Novus) at 4 °C overnight. After washing three times in phosphate-buffered saline (PBS) for 5 min each, secondary antibodies (goat anti-rabbit) were added to the slides, which were


subsequently incubated for 1 h at room temperature. The sections were visualized with diaminobenzidine (DAB; Maixin Biotechnology, Fuzhou, China) and counterstained with hematoxylin (Maixin


Biotechnology) for 3–5 min at room temperature. To avoid bias, two pathologists who were blinded to the clinical data examined the IHC-stained slides separately. They randomly counted 100


cells in the microscopic field at 200× magnification. All sections were classified into five ranges depending on the percentage of cells with positive staining: 0 = negative, 1 = 0–25%, 2 = 


26–50%, 3 = 51–75%, and 4 = 76–100%. Moreover, we grouped all sections into four levels based on the staining intensity as follows: 0 = negative, 1 = pale yellow, 2 = medium yellow, and 3 =


tawny. The final score was the product of the proportion multiplied by the intensity score; the final score ranged from 0 to 12, where 0 to 6 was considered to indicate low expression and 7


to 12 was considered to indicate high expression. WESTERN BLOTTING Cellular protein was extracted using radioimmunoprecipitation assay (RIPA) lysis buffer (Beyotime; Beijing, China)


containing a protease inhibitor (APExBIO). Proteins were quantified using a bicinchoninic acid (BCA) kit (Thermo Fisher, US) and then boiled in an SDS-PAGE loading buffer (Solabio). Next,


10% SDS-PAGE was used to separate the protein samples, which were transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore). The PVDF membranes were blocked with 5% non-fat milk


(Bio-Rad, US) for at least 1 h at room temperature and then incubated with primary antibodies overnight at 4 °C, followed by incubation with anti-rabbit-HRP or anti-mouse-HRP secondary


antibodies. The target proteins were immediately visualized via enhanced chemiluminescence (ECL; Millipore). The following antibodies were used: anti-PSMD14 (4197 s, Cell Signaling),


anti-CARM1 (NB200-342, Novus), anti-β-actin (3700, Cell Signaling), anti-FLAG (F1804, Sigma), anti-HA (3724, Cell Signaling), anti-ubiquitin (60396, Cell Signaling), anti-GFP (ab32146,


Abcam), and anti-IgG (2729, Cell Signaling). RNA EXTRACTION AND QUANTITATIVE REAL-TIME PCR (QRT-PCR) The standard TRIzol (Invitrogen, US) protocol was used for extracting total RNA, followed


by reverse transcription using the One-Step gDNA Removal and cDNA Synthesis Kit (TransGen, Beijing, China). Then, qRT-PCR was performed in a Lightcycler 96 Real-Time PCR System (Roche,


Switzerland) using Taq Pro Universal SYBR qPCR Master Mix (Vazyme). The results were normalized to those of GAPDH and presented as the mean of at least three independent experiments. The


primers used were as follows: GAPDH-Forward: GGTGTGAACCATGAGAAGTATGA, GAPDH-Reverse: GAGTCCTTCCACGATACCAAAG; CARM1-Forward: GAAGGAGATTTGCACAGGATAGA, CARM1-Reverse: GACAGCCACACGGTCATTAT;


PSMD14-Forward: CTTAGACTTGGAGGAGGTATGC, PSMD14-Reverse: CAGTGCCAGGGAAGAGATATAG. CONSTRUCTION OF STABLE OVEREXPRESSION AND KNOCKDOWN CELLS For the purpose of CARM1 overexpression, pLVX-Puro,


and pLVX-Puro-CARM1-FLAG (containing 3×FLAG tags) plasmids were purchased from Public Protein/Plasmid Library Corporation (Nanjing; Jiangsu Province, China). In order to carry out knockdown,


pLV [shRNA]-mCherry-U6, pLV [shRNA]-mCherry-U6 > CARM1[shRNA], and pLV [shRNA]-mCherry-U6 > CARM1[shRNA] plasmids were purchased from VectorBuilder (Guangzhou; Guangdong Province,


China). The lentiviral particles were packed in HEK293T cells following the standard protocol. The medium was collected after 48 h of transfection and added to the target cells with


polybrene (8 μg/mL) for 48 h. Stable cells were selected with puromycin (Beyotime, Beijing, China). The shRNA sequences used are as follows: shCARM1-1-Forward:


gatccGCAGCCATGAAGATGTGTGTTTCAAGAGAACACACATCTTCATGGCTGTTTTTTg; shCARM1-1-Reverse: aattcAAAAAACAGCCATGAAGATGTGTGTTCTCTTGAAACACACATCTTCATGGCTGCg; shCARM1-2-Forward:


gatccGTGGCCAAGTCTGTCAAGTATTCAAGAGATACTTGACAGACTTGGCCATTTTTTg; shCARM1-2-Reverse: aattcAAAAAATGGCCAAGTCTGTCAAGTATCTCTTGAATACTTGACAGACTTGGCCACg. TRANSWELL ASSAY The migration and invasion


abilities of cells were detected by Transwell assays using 24-well transwell chambers (8 μm pore size; Corning Incorporated; New York, NY, US) coated with or without Matrigel (BD


bioscience). The cells were mixed with 200 μL of serum-free DMEM and seeded in the upper chambers, and the lower layer was supplemented with 200 μL of DMEM supplemented with 10% FBS. After


incubation, 500 μL of 4% paraformaldehyde was used to fix the cells on the bottom surface of the chambers for 30 min at room temperature. These cells were subsequently stained with 1%


crystal violet for 30 min at room temperature. Finally, 10 random fields were selected for imaging and quantification under a microscope. CELL PROLIFERATION ASSAY To detect the proliferation


of HCC cells, cell counting kit-8 (CCK-8) assays (Dojindo; Beijing; China) were performed. HCC cells (2000 cells per well) were seeded in 96-well plates in 100 μL of medium. The absorbance


was measured at 450 nm after the cells were incubated with 10 μL of CCK-8 solution at 37 °C for 1.5 h. The CCK-8 assay for each cell line was independently repeated three times.


IMMUNOFLUORESCENCE (IF) STAINING ASSAY The cells were seeded on glass coverslips in six-well plates, fixed with 1% formaldehyde for 10 min, and permeabilized with 0.2% Triton X-100 in PBS


for 10 min at room temperature. The cells were washed twice with PBS, and 5% BSA in PBS was used to block them for 1 h. Then, the cells were incubated with primary antibodies at 4 °C


overnight, washed three times in PBS, and incubated with secondary antibodies for 1 h at room temperature. The cell nuclei were counterstained with 500 nM DAPI (Sigma). Finally, photographs


were taken using a confocal microscope (Zeiss). COIMMUNOPRECIPITATION (CO-IP) ASSAY The cells were collected and lysed with IP lysis buffer (Beyotime, Beijing, China). After extracting total


proteins, Dynabeads Protein G (Invitrogen; Beijing; China) and the appropriate antibodies were added to the cell lysis buffer, and the incubation was prolonged overnight at 4 °C on a


rocking platform. After the beads were washed thrice, the proteins immunoprecipitated with a loading buffer were boiled and evaluated by Western blotting analysis. CHROMATIN


IMMUNOPRECIPITATION ASSAY (CHIP) AND HIGH-THROUGHPUT SEQUENCING A ChIP assay was performed following the instructions provided by the manufacturer using a Magna ChIP™ A/G Chromatin


Immunoprecipitation Kit (Millipore). Briefly, the cells were cross-linked in a 1% formaldehyde solution at room temperature for 10 min. To terminate the cross-linking process, glycine was


added, and the mixture was incubated for 5 min. The samples were washed with ice-cold PBS, scraped, and resuspended in PBS. After washing, the cell pellets were lysed with a cell lysis


buffer. The lysates were incubated on ice for 10 min and centrifuged at 5000 rpm for 5 min at 4 °C to pellet the nuclei. The pellets formed were lysed using a nuclear lysis buffer containing


50 mM Tris-HCl (pH 8.0), 10 mM EDTA, 1% SDS, 1 mM PMSF, and a proteinase inhibitor cocktail. Then, the lysates were diluted with an IP dilution buffer. The nuclear lysates were sonicated,


and the debris was removed through centrifugation. For immunoprecipitation, CARM1 antibodies (Novus), H3R17me2 antibodies (Active Motif), or IgG antibodies (Millipore) were mixed with the


nuclear lysates. The coprecipitated DNA was purified, and the expression levels of the target genes were quantified by qRT-PCR. The sequences of the primers used were as follows:


FERMT1-A-Forward: ATTAGCCAGGTGTGGTGGCAC, FERMT1-A-Reverse: GGAGTGCAATGGTGTGATCTC; FERMT1-B-Forward: GCTAGTCTTGAACTCCTGACC, FERMT1-B-Reverse: GACCGTGGGACTAAAAATGAA; FERMT1-C-Forward:


GTACGGTCCTGTTGTGTCCTC, FERMT1-C-Reverse: ATCTTGGCGCTGTGAGGAGAG; FERMT1-D-Forward: AGTGCACCGGGCACGCCGCGC, FERMT1-D-Reverse: GCTCAGCTAAGGCTGCGGCAG. High-throughput DNA sequencing libraries


were prepared using the VAHTS Universal DNA Library Prep Kit for Illumina V3 (Catalog No. ND607, Vazyme). Library fragments in the 200-500 base pair range were enriched, quantified, and


sequenced using the Novaseq 6000 sequencer (Illumina) with the PE150 protocol. Data analysis was conducted by SeqHealth Technology Co., Ltd. (Wuhan, China). RNA SEQUENCING Total RNA was


extracted from shCON and shCARM1 Huh7 cells using TRIzol Reagent (Invitrogen, cat. no. 15596026) according to the method described by Chomczynski et al. (DOI: 10.1006/abio.1987.9999). DNA


digestion was performed post-RNA extraction using DNase I. RNA quality was assessed by measuring the A260/A280 ratio with a Nanodrop™ OneC spectrophotometer (Thermo Fisher Scientific Inc.).


RNA integrity was further confirmed by 1.5% agarose gel electrophoresis. The quantity of qualified RNA was determined using a Qubit 3.0 fluorometer and the Qubit™ RNA Broad Range Assay Kit


(Life Technologies, Q10210). For RNA sequencing, 2 μg of total RNA was used for library preparation with the KCTM Stranded mRNA Library Prep Kit for Illumina® (Catalog No. DR08402, Wuhan


Seqhealth Co., Ltd., China) according to the manufacturer’s instructions. PCR products corresponding to 200–500 bp were enriched, quantified, and sequenced on a NovaSeq 6000 sequencer


(Illumina) using the PE150 protocol. RNA-Seq data analysis was performed by Seqhealth Technology Co., Ltd. (Wuhan, China). IN VIVO DEUBIQUITINATION ASSAY HA-ubiquitinated FLAG-CARM1 or


endogenous CARM1 was immunoprecipitated by FLAG antibodies (Sigma) or CARM1 antibodies (Novus), respectively, under denaturing conditions (1% SDS, 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM


EDTA) and incubated at 95 °C for 5 minutes. Subsequently, the FLAG-CARM1 and CARM1 proteins were purified by anti-FLAG M2 magnetic beads (Sigma-Aldrich) and anti-CARM1 antibodies (Novus)


conjugated with Protein G magnetic beads (Thermo Fisher), respectively. Then incubated at 4 °C overnight with gentle rotation to allow specific binding. The eluted proteins were subjected to


immunoblotting with antibodies specific to HA or ubiquitin. IN VITRO DEUBIQUITINATION ASSAY HEK293T cells expressing HA-Ubi-FLAG-CARM1 were lysed in lysis buffer (50 mM Tris-HCl, pH 7.5,


150 mM NaCl, 1% NP-40, 1 mM EDTA, and protease inhibitor cocktail). HA-Ubi-FLAG-CARM1 was immunoprecipitated using protein G-agarose beads (Thermo Fisher Scientific) pre-incubated with


anti-FLAG antibody (Sigma-Aldrich) at 4 °C for 4 h with gentle rotation. Similarly, GFP-tagged wild-type (WT) or mutant PSMD14 protein complexes were isolated from HEK293T cells expressing


the respective constructs. Cell lysates were prepared and incubated with protein G-agarose beads pre-bound to an anti-GFP antibody (Thermo Fisher) under the same conditions as FLAG-CARM1.


Next, the protein complexes were mixed in a reaction buffer containing 50 mM Tris (pH 7.5), 10 mM MgCl2, 1 mM dithiothreitol (DTT), 100 mM NaCl, and 1 mM ATP. The reaction mixture was


incubated at 37 °C for 1 h with gentle agitation. Following incubation, FLAG-CARM1 was re-purified using protein G-agarose beads pre-bound with anti-FLAG antibody. Subsequently, SDS-PAGE and


immunoblotting were performed. XENOGRAFT MOUSE MODELS A total of 2 × 106 cells with or without CARM1 knockdown were injected subcutaneously into the backs of nude mice (6-week-old male


BALB/c athymic nude mice). After 1 week, the volume of the tumor was measured using the formula 1/2 × length × width2 every 3 days with Vernier calipers. After 28 days, the tumors were


harvested, weighed, and used for further analysis. To inject SGC2085, when the tumors were about 200 mm3, the mice were randomly divided into two groups (_n_ = 4 mice per group). Then,


SGC2085 (10 mg/kg, diluted in sterile water containing 30% PEG300, 5% Tween-80, and 10% DMSO) or an equal volume of solvent was injected intraperitoneally every four days. After six


injections, the tumors were harvested and weighed. All animal experiments were approved by the Animal Care and Use Committee of Xiamen University. STATISTICAL ANALYSIS All statistical


analyses were performed using SPSS 20 (SPSS, Inc., Chicago, IL). The data were presented as the mean ± standard deviation (SD), and three independent replicates were performed. The


differences between groups were determined by conducting Student’s _t_-tests or ANOVA. The Chi-square test was conducted to analyze the relationships between CARM1 expression and clinical


features. The Kaplan-Meier survival test and log-rank test were performed to analyze the relationship between the expression of PSMD14 or CARM1 and the prognosis of HCC patients. All


differences among and between groups were considered to be statistically significant at _P_ < 0.05. RESULTS OVEREXPRESSION OF CARM1 INDICATES POOR CLINICAL OUTCOMES IN HCC PATIENTS A


study reported that CARM1, an oncogene, is dysregulated in several types of cancer [29]. However, the expression pattern of CARM1 and its clinical significance in HCC tissues remain elusive.


First, we determined the differential expression of CARM1 in 35 paired HCC and corresponding adjacent normal tissues. The results of Western blotting assays showed that the expression level


of CARM1 was significantly greater in HCC tissues than in corresponding adjacent normal tissues (Fig. 1A). For further validation, IHC staining of CARM1 was performed in 66 HCC tissues and


paired adjacent normal tissues. CARM1 was upregulated in 71.2% (47/66) of HCC tissues compared to its expression in adjacent normal tissues (Fig. 1B). To determine the clinical significance


of CARM1 overexpression in HCC patients, a correlation analysis between CARM1 expression and clinicopathological features was performed, and the results showed that CARM1 expression was


significantly related to tumor size (_P_ = 0.0443), the level of differentiation (_P_ = 0.0239), and satellite foci (_P_ = 0.0272). However, CARM1 expression was not associated with other


factors, including age, sex, AFP level, PVTT, HBV DNA level, or liver cirrhosis (Table 1). Moreover, the results of the Kaplan-Meier survival analysis revealed that patients with high


expression of CARM1 in HCC had a lower overall rate than those with low CARM1 expression (Fig. 1C). Additionally, the UALCAN online tool (http://ualcan.path.uab.edu/index.html) and TIMER 2.0


were used to validate the reliability of our results. Consistent with our findings, the CARM1 expression levels were higher in primary HCC tissues (Fig. 1D and Supplementary Fig. 1A). A


higher tumor grade and advanced cancer stage were associated with higher expression of CARM1 (Supplementary Fig. 1B, C). The Kaplan-Meier analysis of the data in the TCGA database using the


Kaplan-Meier plotter online tool (https://ualcan.path.uab.edu/cgi-bin/TCGA-survival) showed that patients with high CARM1 expression exhibited markedly shorter overall survival than those


with low CARM1 expression (_P_ = 0.035) (Fig. 1E). The data from our cohort and the TCGA database indicated that CARM1 is upregulated in HCC tissues and associated with the malignant


progression of HCC. PSMD14 INTERACTS WITH AND STABILIZES CARM1 Next, we investigated the regulatory mechanism of the overexpression of CARM1 in HCC. By conducting mass spectrometry analysis,


in our previous study, we identified 710 proteins potentially associated with the deubiquitinase PSMD14 [28]. CARM1 was identified as one of the main PSMD14-interacting proteins, indicating


that PSMD14 might regulate CARM1. Through double IF labeling for CARM1 and PSMD14, we found that CARM1 and PSMD14 were co-localized (Fig. 2A). The interaction between endogenous CARM1 and


PSMD14 was confirmed via co-IP experiments (Fig. 2B). As PSMD14 functions as a deubiquitinase to modulate protein stability, we examined whether PSMD14 can regulate CARM1 through this


mechanism. Decreasing PSMD14 expression did not affect the expression level of CARM1 mRNA; however, the results of Western blotting analyses showed that PSMD14 knockdown resulted in a


substantial decrease in CARM1 protein levels in HCC cells (Fig. 2C, D). In contrast, the overexpression of PSMD14 upregulated the expression of the CARM1 protein but did not increase its


mRNA level (Fig. 2E, F). To determine whether CARM1 is stabilized by PSMD14, we conducted cycloheximide (CHX) pulse-chase assays to measure the degradation rates of endogenous CARM1 under


the effect of PSMD14 knockdown in HCC cells. CARM1 degradation was strongly accelerated in PSMD14-knockdown cells compared to that in control cells (Fig. 2G). These results suggested that


PSMD14 interacts with CARM1 and prevents its degradation. PSMD14 DEUBIQUITINATES CARM1 Next, the mechanism of PSMD14-mediated stability of CARM1 was investigated. To determine whether PSMD14


stabilizes CARM1 via deubiquitination, we performed an in vivo deubiquitination assay to test the level of CARM1 ubiquitination in control and PSMD14-knockdown Huh7 cells. The level of


ubiquitinated CARM1 increased substantially after the depletion of PSMD14 (Fig. 3A). Similarly, Capzimin, a specific inhibitor of PSMD14 [30], also led to a sharp increase in the CARM1


ubiquitination level (Fig. 3B), further suggesting that PSMD14 deubiquitinated CARM1. The JAMM domain of PSMD14 plays a key role in maintaining its deubiquitinating enzymatic activity [31].


We constructed a plasmid expressing a GFP-tagged PSMD14 mutant with deletion of the JAMM motif (named GFP-PSMD14-MUT). Compared to wild-type PSMD14, mutant PSMD14 failed to decrease the


ubiquitination level of exogenous CARM1 (Fig. 3C). By conducting an in vitro deubiquitination assay, we also found that PSMD14 directly deubiquitinated CARM1. Only purified GFP-PSMD14-WT but


not GFP-PSMD14-MUT deubiquitinated CARM1 in vitro (Fig. 3D). These results indicated the indispensable role of the deubiquitinating activity of PSMD14 in the regulation of CARM1. Next, we


investigated the polyubiquitin modifications on the CARM1 protein that are specifically influenced by deubiquitination mediated by PSMD14. FLAG-CARM1, GFP-PSMD14, and various ubiquitins (WT,


K11-, K48-, or K63-only ubiquitin-HA) were separately transfected into HEK293T cells. After transfecting individual plasmids into cells, we found that ectopic PSMD14 substantially decreased


K48-linked and K63-linked ubiquitin on CARM1 but did not affect K11-linked ubiquitin on CARM1 (Fig. 3E). To elucidate the underlying mechanism by which PSMD14 deubiquitinates CARM1, we used


UbPred, an online tool for predicting the ubiquitination sites of substrates. Five potential lysine ubiquitination sites on human CARM1 (K227, K363, K444, K462, and K470) were predicted.


Based on the predicted ubiquitination sites, we constructed a full-length CARM1 plasmid and two C-terminal truncated CARM1 plasmids (1–287 aa and 1–453 aa) with N-terminal FLAG tags (Fig.


3F). We then transiently transfected the GFP-PSMD14 plasmid along with the above three derived CARM1 plasmids separately into HEK293T cells. The resulting proteins were coimmunoprecipitated


with anti-GFP antibodies and immunoblotted with anti-FLAG antibodies. FL and Δ2, but not Δ1, were pulled down by GFP-PSMD14, indicating that the ubiquitination sites of CARM1 might reside at


K363 and/or K444 (Fig. 3G). Hence, we mutated C-proximal K363 and K444 to arginine separately in FLAG-tagged CARM1 plasmids and transiently transfected them into HEK293T cells. After


immunoprecipitation with an anti-FLAG antibody, compared to wild-type CARM1, FLAG-CARM1K363R, but not FLAG-CARM1K444R, reduced the ubiquitination of CARM1 (Fig. 3H). These results indicated


that PSMD14 deubiquitinates and stabilizes CARM1 and that the residue K363 is the only site implicated in the PSMD14-mediated ubiquitination of CARM1. CARM1 PROTEIN EXPRESSION IS POSITIVELY


CORRELATED WITH PSMD14 IN HCC TISSUES Our previous study revealed an oncogenic role of PSMD14 in HCC [28]. In this study, we examined whether CARM1 participates in this process. CARM1 was


ectopically expressed following PSMD14 silencing in Huh7 and SK-Hep-1 cells. The results of the CCK8 assays revealed that CARM1 overexpression compensated for the inhibition of proliferation


caused by knocking down PSMD14 (Fig. 4A). Similarly, colony formation assays revealed that restoring CARM1 expression considerably reversed the impaired colony formation effects induced by


PSMD14 knockdown in HCC cells (Fig. 4B). The results of the transwell assays revealed that the ability of silencing PSMD14 to suppress migration and invasion was restored through the


overexpression of CARM1 (Fig. 4C). Thus, these findings suggested that CARM1 acts as a downstream effector of PSMD14 to promote the proliferation, migration, and invasion of HCC. Based on


our finding that PSMD14 stabilizes CARM1 expression in HCC cells, we further analyzed the pathologic correlation of PSMD14 with CARM1 expression in HCC tissues. IHC staining of PSMD14 and


CARM1 in the same 66 HCC and matched adjacent normal tissues was performed. In HCC tissues with elevated PSMD14 expression, high expression of CARM1 was recorded. In contrast, in HCC with a


decrease in PSMD14 expression, a corresponding decrease in CARM1 expression was recorded (Fig. 4D, E, _P_ = 0.001, _R_ = 0.430). IHC staining for PSMD14 and CARM1 was scored based on


intensity, and a positive correlation was found between PSMD14 and CARM1 expression in the same HCC tissues (Fig. 4F, _R_ = 0.4283, _P_ = 0.0003). Additionally, the group characterized by


high levels of PSMD14 and CARM1 presented a more unfavorable prognosis than the group characterized by low expression of PSMD14 and CARM1 (Fig. 4G). Our results confirmed the clinical


relevance of PSMD14-mediated regulation of CARM1 in HCC, suggesting that the upregulation of CARM1 and PSMD14 may hold promise as prognostic indicators for patients with HCC. CARM1 PROMOTES


THE GROWTH OF HCC To determine the functional significance of CARM1 in HCC, we generated stable cell lines with CARM1 knockdown using Huh7 and PLC/PRF/5 cells (Fig. 5A). We conducted CCK8


and colony formation assays to evaluate the effect of CARM1 knockdown on cell proliferation. Compared to the proliferation of the control cells, the proliferation of the CARM1-knockdown


cells was strongly inhibited (Fig. 5B, C), whereas CARM1 overexpression improved this ability (Supplementary Fig. 2A, B). Similarly, knockout of CARM1 expression resulted in significant


suppression of cellular proliferation (Supplemental Fig. 2C–E). To further elucidate the in vivo pro-proliferative effect of CARM1, control and CARM1-knockdown Huh7 cells were subcutaneously


injected into nude mice. Tumors formed by CARM1-knockdown cells exhibited significantly decelerated growth and smaller mean volumes and weights than those formed by control cells (Fig.


5D–F). Furthermore, Ki-67 is a proliferative activity marker of tumors and is often used to compare the proliferative capacity of tumors [32]. IHC staining of Ki-67 in xenografts revealed a


notable reduction in the expression of Ki-67 in the CARM1-knockdown group compared to that in the control group (Fig. 5G). These results indicated that CARM1 promotes HCC proliferation in


vivo and in vitro. CARM1 FACILITATES HCC METASTASIS To investigate whether CARM1 influences HCC tumor metastasis, we first examined whether CARM1 affects the migratory capacity of HCC cells


by conducting a transwell assay. We found that CARM1 knockdown or knockout significantly impaired the migration of HCC cells (Fig. 6A and Supplementary Fig. 2F). Similarly, cell invasion was


attenuated following the knockdown or knockout of CARM1 (Fig. 6A and Supplementary Fig. 2F). To assess the effect of CARM1 on tumor metastasis in vivo, we intravenously injected HCC cells


with or without CARM1 knockdown into the tail vein of nude mice. After 2 months, the lungs were extracted, showing a remarkable decrease in the quantity of pulmonary metastatic nodules in


the CARM1-knockdown group compared to the control group (Fig. 6B, C). To summarize, these findings showed that CARM1 promoted HCC cell migration and invasion in vitro, as well as tumor


metastasis in vivo. ACTIVATION OF FERMT1 TRANSCRIPTION BY CARM1-METHYLATED H3R17 AFFECTS HCC PROGRESSION As a histone arginine methyltransferase, CARM1 catalyzes the dimethylation of H3R17


(H3R17me2) to activate the transcription of downstream genes. However, the target genes regulated by CARM1-mediated H3R17me2 modification in HCC remain unknown. Therefore, to elucidate the


mechanisms by which CARM1 regulates target genes in HCC, transcriptomic RNA sequencing combined with anti-CARM1 ChIP-sequencing (ChIP-seq) was performed (Fig. 7A, B). Integration of the


transcriptome and ChIP-seq data revealed significant regulatory associations between CARM1 binding events and gene expression changes. We identified 553 differentially expressed genes that


exhibited concurrent CARM1 binding within their regulatory regions, and their expression levels were closely related to CARM1 (Fig. 7C). Moreover, these 231 genes were evaluated by the Kyoto


Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. These common genes were significantly enriched in cancer-related pathways, such as metabolism, Hippo signaling and cell


adhesion molecules (Supplemental Fig. 3A). Among these genes, we selected FERM domain containing kindlin 1 (FERMT1) as a candidate target gene of CARM1 for further investigation. FERMT1 is


involved in integrin signaling and the linkage of the actin cytoskeleton to the extracellular matrix, which plays an important role in enhancing tumor progression [33]. Additionally, FERMT1


was found to be involved in the regulation of the EMT pathway [34]. The expression of the FERMT1 gene was most significantly altered after knocking down CARM1, and it was found to be


strongly bound by CARM1 (Supplementary Fig. 3B). TCGA analysis via the TIMER 2.0 and GEPIA2 online tools revealed a positive correlation between FERMT1 and CARM1 expression in HCC tissues


(Supplemental Fig. 3C, D). We conducted a qRT-PCR assay and found that knocking down CARM1 significantly decreased FERMT1 expression (Fig. 7D). To confirm the binding of CARM1 to the FERMT1


promoter region, we performed ChIP assays using four distinct pairs of ChIP primers at the FERMT1 promoter locus (Fig. 7E). Using an antibody specifically targeting CARM1, we revealed the


presence of CARM1 at the promoter region of FERMT1 in Huh7 cells. Our findings indicated that CARM1 bound to amplicons A, B, and D of FERMT1 but did not bind to amplicon C. The most


proximate binding occurred within segment B (Fig. 7F). Next, we evaluated the effect of CARM1 on H3R17me2 modification. The results revealed that H3R17me2 decreased after CARM1 was knocked


down (Fig. 7G). Moreover, suppression of CARM1 expression considerably decreased H3R17me2 levels within the FERMT1 promoter, as determined by ChIP assays with an anti-H3R17me2 antibody,


followed by qRT-PCR analysis (Fig. 7H). These results suggested that CARM1 activates the transcription of FERMT1 via H3R17me2. To determine whether FERMT1 functions downstream of CARM1,


rescue experiments were conducted (Supplemental Fig. 3E). The results of CCK-8 and transwell assays revealed that overexpression of FERMT1 abolished the suppression of proliferation,


migration, and invasion caused by CARM1 knockdown in Huh7 cells (Fig. 7I, J). These results revealed that CARM1 activates the transcription of FERMT1 via H3R17me2, which is critical for the


oncogenic role of CARM1 in the progression of HCC. CARM1 INHIBITOR RESTRAINS HCC A study showed that SGC2085, a potent and selective inhibitor, can inhibit CARM1 activity [35]. Based on our


findings that CARM1 functions as an oncogene in HCC, we speculated that SGC2085 might have an inhibitory effect on HCC cells. To test our hypothesis, HCC cells were exposed to different


concentrations of SGC2085. Notably, the administration of SGC2085 considerably inhibited proliferation, migration, and invasion in a dose-dependent manner (Fig. 8A, B). Moreover,


administering SGC2085 substantially suppressed tumor growth in vivo, as confirmed by the outcomes of the xenograft model (Fig. 8C–E). To summarize, these findings supported the therapeutic


potential of the CARM1 inhibitor SGC2085 on HCC. DISCUSSION HCC is the predominant form of primary liver cancer, constituting about 90% of cases. With 910,000 newly diagnosed cases and


830,000 deaths, it is the sixth most prevalent cancer and the third leading cause of mortality [36]. Surgical resection, liver transplantation, radiotherapy, chemotherapy, and molecular


targeted therapy have been widely used as primary approaches in the clinical management of HCC. However, owing to its undetectable onset, early symptoms and signs of HCC often remain hidden.


By the time symptoms manifest and patients seek medical attention, the disease often progresses to an advanced stage, depriving them of the opportunity for curative surgery. Moreover,


postoperative recurrence and metastasis significantly affect the treatment outcomes and overall survival of HCC patients [37]. Therefore, understanding the underlying molecular mechanisms of


HCC occurrence and progression can help identify effective biomarkers for early diagnosis and prognosis prediction, as well as discover novel drug targets to overcome therapeutic barriers


in the treatment of HCC. By analyzing our cohort and TCGA database, we revealed a significant increase in CARM1 expression in HCC tissue samples. Moreover, the correlation analysis between


CARM1 expression and clinicopathological features revealed that CARM1 overexpression was strongly associated with the presence of satellite foci, increased serum HBV and AFP levels, and a


poor prognosis. Consistent with our findings, a recent investigation revealed that CARM1 expression is notably correlated with survival and various clinical parameters, including


alpha-fetoprotein levels, tumor size, satellite nodules, and microvascular invasion [38]. These findings provided additional support for the potential of CARM1 as a prognostic indicator in


patients with HCC. Post-translational modifications, including phosphorylation, O-acetylglycosylation, and methylation, regulate CARM1 function [15,16,17,18]. Several studies have reported


the critical role of ubiquitination in modulating CARM1 stability. For example, the E3 ligase S-phase kinase-associated protein 2 (SKP2) interacts with CARM1 to facilitate the


ubiquitin-mediated degradation of CARM1 [19]. Tripartite motif-containing protein 28 (TRIM28) physically interacts with CARM1, decreasing its ubiquitination and protecting it from


proteasome-mediated degradation [38]. However, whether deubiquitinase is responsible for regulating CARM1 stability is still unclear. The PSMD14 protein plays a crucial role in various


biological processes, including DNA damage repair, cellular differentiation, and multidrug resistance [24, 39, 40]. Moreover, abnormal overexpression of PSMD14 contributes to tumor growth


and metastasis. PSMD14 increases the stability of the Snail protein, a transcription factor associated with epithelial-mesenchymal transition (EMT), thus inducing metastasis in esophageal


cancer cells [41]. In HCC, PSMD14 stabilizes E2F1, which upregulates survivin and FOXM1 expression, thus facilitating the growth of HCC [26]. PSMD14 also influences HCC metastasis by


deubiquitinating the TGF-β receptor and caveolin-1 [28]. Our previous study revealed that GRB2 is a target of PSMD14-mediated deubiquitination, which is critical for the progression of HCC.


In this study, we identified CARM1 as a novel substrate of PSMD14. CARM1 was important for PSMD14-enhanced proliferation, migration, and invasion in HCC cells. The interaction between PSMD14


and CARM1 led to a reduction in CARM1 ubiquitination and protected it from degradation. Notably, residue K363 of CARM1 exclusively plays a crucial role in this process. We also found that


PSMD14 efficiently deubiquitinates CARM1 by removing the K48-linked and K63-linked polyubiquitin chains, and this finding was similar to previous studies, revealing that PSMD14 is


responsible for removing the K48-linked and K63-linked ubiquitin chains of target proteins [23, 42]. K48-linked and K63-linked polyubiquitin chains act as discerning signs of the proteasome,


enabling efficient processing of substrates within its confines [42]. Our investigation also revealed a synchronized increase in the expression of PSMD14 and CARM1 in clinical HCC samples


and its significance for the prognosis of patients, suggesting strong pathological implications for the regulation of CARM1 by PSMD14 in the progression of HCC. Along with its involvement in


transcription regulation, pre-mRNA splicing, and the cell cycle, CARM1 plays various roles in different cellular processes, including DNA end resection, the replication stress response,


cell autophagy, tumor immunity, and tumor metabolism [7, 11, 43,44,45]. In HCC, CARM1 promotes malignant behaviors by activating the AKT/mTOR signaling pathway [38]. This study revealed that


CARM1 functions as an oncogene to facilitate HCC growth and metastasis. However, another study revealed that glucose starvation-induced CARM1 expression induces GAPDH hypermethylation to


suppress glycolysis in liver cancer cells, indicating a tumor-suppressive role of CARM1 [46]. The contradictory effects of CARM1 may arise from the distinct metabolic environments of tumor


cells, potentially involving different primary pathways of influence. FERMT1 is a novel adhesion protein belonging to the kindlin family and consists of ezrin, radisin, moesin, and a


pleckstrin homology domain. It is a mutant gene closely associated with Kindler syndrome [47]. It plays an important role in regulating cell cycle arrest, apoptosis, EMT, motility,


invasiveness, and angiogenesis in cancer. FERMT1 activates the NF-κB, Wnt/β-catenin, and PI3K/AKT signaling pathways and is associated with Nod-like receptor family protein 3 [48,49,50,51].


Some small RNAs, such as hsa_circ_0000554 and miR-24, can regulate FERMT1 expression [52, 53]. Here, we identified FERMT1 as a downstream gene of CARM1 and found that FERMT1 is important for


CARM1-induced malignant behaviors. Although CARM1 can methylate both H3R17 and H3R26, it preferentially methylates H3R17 [54]. Therefore, we estimated H3R17me2 levels at the FERMT1 promoter


and found that CARM1 knockdown significantly decreased H3R17me2 levels. CARM1 forms transcriptional complexes with PAF1, AR, ER, or P300 to activate downstream gene expression [8,9,10].


Further investigations are needed to identify the specific transcription factor involved in the CARM1-mediated activation of FERMT1 transcription. As CARM1 is overexpressed and functions as


an oncogene in various types of tumors, targeting CARM1 may help in cancer treatment. For example, the CARM1-specific inhibitors TP-064 and EZM2302 have demonstrated their effectiveness as


promising therapeutic interventions for multiple myeloma [55, 56]. In this study, we found that administering SGC2085 markedly inhibited the proliferation, migration, and invasion of HCC


cells in vitro and strongly suppressed tumor growth in vivo, suggesting that targeting CARM1 may be a promising treatment for HCC. In the last five years, immune checkpoint inhibitors have


revolutionized the management of HCC. The inhibition of CARM1 induces high antitumor efficacy in cytotoxic T-cells and tumor cells [45]. The combination of the CARM1 inhibitor EZM2302 and an


anti-PD-1 antibody exerted promising synergistic effects on a preclinical model of lung cancer [57]. These findings suggested that the combination of CARM1 inhibitors and immunotherapy may


generate synergistic effects in the treatment of HCC. CONCLUSION To summarize, our findings revealed that CARM1 is stabilized by PSMD14-mediated deubiquitination and activates FERMT1


transcription through H3R17me2 modification (Fig. 9), indicating a key role in the progression of HCC. Thus, directing therapeutic efforts toward CARM1 may be a promising strategy to treat


HCC. DATA AVAILABILITY The data substantiating the findings of this study can be obtained from the corresponding authors upon reasonable inquiry. REFERENCES * 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. Article  CAS  PubMed  Google Scholar  * Suresh S, Huard S, Dubois T.


CARM1/PRMT4: making its mark beyond its function as a transcriptional coactivator. Trends Cell Biol. 2021;31:402–17. Article  CAS  PubMed  Google Scholar  * Gao G, Hausmann S, Flores NM,


Benitez AM, Shen J, Yang X, et al. The NFIB/CARM1 partnership is a driver in preclinical models of small-cell lung cancer. Nat Commun. 2023;14:363. Article  CAS  PubMed  PubMed Central 


Google Scholar  * Larsen SC, Sylvestersen KB, Mund A, Lyon D, Mullari M, Madsen MV, et al. Proteome-wide analysis of arginine monomethylation reveals widespread occurrence in human cells.


Sci Signal. 2016;9:rs9. Article  PubMed  Google Scholar  * Li WJ, He YH, Yang JJ, Hu GS, Lin YA, Ran T, et al. Profiling PRMT methylome reveals roles of hnRNPA1 arginine methylation in RNA


splicing and cell growth. Nat Commun. 2021;12:1946. Article  CAS  PubMed  PubMed Central  Google Scholar  * Kim EJ, Liu P, Zhang S, Donahue K, Wang Y, Schehr JL, et al. BAF155 methylation


drives metastasis by hijacking super-enhancers and subverting anti-tumor immunity. Nucleic Acids Res. 2021;49:12211–33. Article  CAS  PubMed  PubMed Central  Google Scholar  * Genois MM,


Gagné JP, Yasuhara T, Jackson J, Saxena S, Langelier MF, et al. CARM1 regulates replication fork speed and stress response by stimulating PARP1. Mol Cell. 2021;81:784–800.e788. Article  CAS


  PubMed  PubMed Central  Google Scholar  * Wu J, Xu W. Histone H3R17me2a mark recruits human RNA polymerase-associated factor 1 complex to activate transcription. Proc Natl Acad Sci USA.


2012;109:5675–80. Article  CAS  PubMed  PubMed Central  Google Scholar  * Lee E, Madar A, David G, Garabedian MJ, Dasgupta R, Logan SK. Inhibition of androgen receptor and beta-catenin


activity in prostate cancer. Proc Natl Acad Sci USA. 2013;110:15710–5. Article  CAS  PubMed  PubMed Central  Google Scholar  * Al-Dhaheri M, Wu J, Skliris GP, Li J, Higashimato K, Wang Y, et


al. CARM1 is an important determinant of ERalpha-dependent breast cancer cell differentiation and proliferation in breast cancer cells. Cancer Res. 2011;71:2118–28. Article  CAS  PubMed 


PubMed Central  Google Scholar  * Liu F, Ma F, Wang Y, Hao L, Zeng H, Jia C, et al. PKM2 methylation by CARM1 activates aerobic glycolysis to promote tumorigenesis. Nat Cell Biol.


2017;19:1358–70. Article  CAS  PubMed  PubMed Central  Google Scholar  * Wang L, Zhao Z, Meyer MB, Saha S, Yu M, Guo A, et al. CARM1 methylates chromatin remodeling factor BAF155 to enhance


tumor progression and metastasis. Cancer Cell. 2014;25:21–36. Article  PubMed  PubMed Central  Google Scholar  * Wang L, Zeng H, Wang Q, Zhao Z, Boyer TG, Bian X, et al. MED12 methylation by


CARM1 sensitizes human breast cancer cells to chemotherapy drugs. Sci Adv. 2015;1:e1500463. Article  PubMed  PubMed Central  Google Scholar  * Li X, Lai Y, Li J, Zou M, Zou C. Oxidative


stress destabilizes protein arginine methyltransferase 4 via glycogen synthase kinase 3β to impede lung epithelial cell migration. Am J Physiol-Cell Physiol. 2017;313:C285–C294. Article 


PubMed  PubMed Central  Google Scholar  * Kuhn P, Chumanov R, Wang Y, Ge Y, Burgess RR, Xu W. Automethylation of CARM1 allows coupling of transcription and mRNA splicing. Nucleic Acids Res.


2011;39:2717–26. Article  CAS  PubMed  Google Scholar  * Feng Q, He B, Jung SY, Song Y, Qin J, Tsai SY, et al. Biochemical control of CARM1 enzymatic activity by phosphorylation. J Biol


Chem. 2009;284:36167–74. Article  CAS  PubMed  PubMed Central  Google Scholar  * Higashimoto K, Kuhn P, Desai D, Cheng X, Xu W. Phosphorylation-mediated inactivation of


coactivator-associated arginine methyltransferase 1. Proc Natl Acad Sci USA. 2007;104:12318–23. Article  CAS  PubMed  PubMed Central  Google Scholar  * Charoensuksai P, Kuhn P, Wang L,


Sherer N, Xu W. O-GlcNAcylation of co-activator-associated arginine methyltransferase 1 regulates its protein substrate specificity. Biochem J. 2015;466:587–99. Article  CAS  PubMed  Google


Scholar  * Li C, Yu L, Xue H, Yang Z, Yin Y, Zhang B, et al. Nuclear AMPK regulated CARM1 stabilization impacts autophagy in aged heart. Biochem Biophys Res Commun. 2017;486:398–405. Article


  CAS  PubMed  Google Scholar  * Swatek KN, Komander D. Ubiquitin modifications. Cell Res. 2016;26:399–422. Article  CAS  PubMed  PubMed Central  Google Scholar  * Sun T, Liu Z, Yang Q. The


role of ubiquitination and deubiquitination in cancer metabolism. Mol Cancer. 2020;19:146. Article  PubMed  PubMed Central  Google Scholar  * Jing C, Duan Y, Zhou M, Yue K, Zhuo S, Li X, et


al. Blockade of deubiquitinating enzyme PSMD14 overcomes chemoresistance in head and neck squamous cell carcinoma by antagonizing E2F1/Akt/SOX2-mediated stemness. Theranostics.


2021;11:2655–69. Article  CAS  PubMed  PubMed Central  Google Scholar  * Seo D, Jung SM, Park JS, Lee J, Ha J, Kim M, et al. The deubiquitinating enzyme PSMD14 facilitates tumor growth and


chemoresistance through stabilizing the ALK2 receptor in the initiation of BMP6 signaling pathway. EBioMedicine. 2019;49:55–71. Article  CAS  PubMed  PubMed Central  Google Scholar  * Butler


LR, Densham RM, Jia J, Garvin AJ, Stone HR, Shah V, et al. The proteasomal de-ubiquitinating enzyme POH1 promotes the double-strand DNA break response. EMBO J. 2012;31:3918–34. Article  CAS


  PubMed  PubMed Central  Google Scholar  * Liu Y, Zhang L, Wang B, Yang Z, Xu G, Ma A, et al. Requirement for POH1 in differentiation and maintenance of regulatory T cells. Cell Death


Differ. 2019;26:751–62. Article  PubMed  Google Scholar  * Wang B, Ma A, Zhang L, Jin WL, Qian Y, Xu G, et al. POH1 deubiquitylates and stabilizes E2F1 to promote tumour formation. Nat


Commun. 2015;6:8704. Article  CAS  PubMed  PubMed Central  Google Scholar  * Wang B, Xu X, Yang Z, Zhang L, Liu Y, Ma A, et al. POH1 contributes to hyperactivation of TGF-beta signaling and


facilitates hepatocellular carcinoma metastasis through deubiquitinating TGF-beta receptors and caveolin-1. EBioMedicine. 2019;41:320–32. Article  PubMed  PubMed Central  Google Scholar  *


Lv J, Zhang S, Wu H, Lu J, Lu Y, Wang F, et al. Deubiquitinase PSMD14 enhances hepatocellular carcinoma growth and metastasis by stabilizing GRB2. Cancer Lett. 2020;469:22–34. Article  CAS 


PubMed  Google Scholar  * Yang S, Zhang J, Chen D, Cao J, Zheng Y, Han Y, et al. CARM1 promotes gastric cancer progression by regulating TFE3 mediated autophagy enhancement through the


cytoplasmic AMPK-mTOR and nuclear AMPK-CARM1-TFE3 signaling pathways. Cancer Cell Int. 2022;22:102. Article  CAS  PubMed  PubMed Central  Google Scholar  * Li J, Yakushi T, Parlati F,


Mackinnon AL, Perez C, Ma Y, et al. Capzimin is a potent and specific inhibitor of proteasome isopeptidase Rpn11. Nat Chem Biol. 2017;13:486–93. Article  CAS  PubMed  PubMed Central  Google


Scholar  * Nabhan JF, Ribeiro P. The 19 S proteasomal subunit POH1 contributes to the regulation of c-Jun ubiquitination, stability, and subcellular localization. J Biol Chem.


2006;281:16099–107. Article  CAS  PubMed  Google Scholar  * Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA. Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol.


2010;11:174–83. Article  CAS  PubMed  Google Scholar  * Rognoni E, Ruppert R, Fässler R. The kindlin family: functions, signaling properties and implications for human disease. J Cell Sci.


2016;129:17–27. Article  CAS  PubMed  Google Scholar  * Liu CC, Cai DL, Sun F, Wu ZH, Yue B, Zhao SL, et al. FERMT1 mediates epithelial-mesenchymal transition to promote colon cancer


metastasis via modulation of β-catenin transcriptional activity. Oncogene. 2017;36:1779–92. Article  CAS  PubMed  Google Scholar  * Ferreira de Freitas R, Eram MS, Smil D, Szewczyk MM,


Kennedy S, Brown PJ, et al. Discovery of a potent and selective coactivator associated arginine methyltransferase 1 (CARM1) inhibitor by virtual screening. J Med Chem. 2016;59:6838–47.


Article  CAS  PubMed  Google Scholar  * Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7–30. Article  PubMed  Google Scholar  * Forner A, Llovet JM, Bruix


J. Hepatocellular carcinoma. Lancet. 2012;379:1245–55. Article  PubMed  Google Scholar  * Du P, Luo K, Li G, Zhu J, Xiao Q, Li Y, et al. PRMT4 promotes hepatocellular carcinoma progression


by activating AKT/mTOR signaling and indicates poor prognosis. Int J Med Sci. 2021;18:3588–98. Article  CAS  PubMed  PubMed Central  Google Scholar  * Ma Z, Gao X, Zhao W, Li Y, Li C, Li C.


Relationship between expression of Pad1 homologue and multidrug resistance of idiopathic nephrotic syndrome. Pediatr Int. 2009;51:732–5. Article  CAS  PubMed  Google Scholar  * Schwarz T,


Sohn C, Kaiser B, Jensen ED, Mansky KC. The 19S proteasomal lid subunit POH1 enhances the transcriptional activation by Mitf in osteoclasts. J Cell Biochem. 2010;109:967–74. Article  CAS 


PubMed  PubMed Central  Google Scholar  * Zhu R, Liu Y, Zhou H, Li L, Li Y, Ding F, et al. Deubiquitinating enzyme PSMD14 promotes tumor metastasis through stabilizing SNAIL in human


esophageal squamous cell carcinoma. Cancer Lett. 2018;418:125–34. Article  CAS  PubMed  Google Scholar  * Sun T, Liu Z, Bi F, Yang Q. Deubiquitinase PSMD14 promotes ovarian cancer


progression by decreasing enzymatic activity of PKM2. Mol Oncol. 2021;15:3639–58. Article  CAS  PubMed  PubMed Central  Google Scholar  * Shin HJ, Kim H, Oh S, Lee JG, Kee M, Ko HJ, et al.


AMPK-SKP2-CARM1 signalling cascade in transcriptional regulation of autophagy. Nature. 2016;534:553–7. Article  CAS  PubMed  PubMed Central  Google Scholar  * Karakashev S, Fukumoto T, Zhao


B, Lin J, Wu S, Fatkhutdinov N, et al. EZH2 Inhibition sensitizes CARM1-high, homologous recombination proficient ovarian cancers to PARP inhibition. Cancer Cell. 2020;37:157–167.e156.


Article  CAS  PubMed  PubMed Central  Google Scholar  * Kumar S, Zeng Z, Bagati A, Tay RE, Sanz LA, Hartono SR, et al. CARM1 inhibition enables immunotherapy of resistant tumors by dual


action on tumor cells and T cells. Cancer Discov. 2021;11:2050–71. Article  CAS  PubMed  PubMed Central  Google Scholar  * Zhong XY, Yuan XM, Xu YY, Yin M, Yan WW, Zou SW, et al. CARM1


methylates GAPDH to regulate glucose metabolism and is suppressed in liver cancer. Cell Rep. 2018;24:3207–23. Article  CAS  PubMed  Google Scholar  * Has C, Castiglia D, del Rio M, Diez MG,


Piccinni E, Kiritsi D, et al. Kindler syndrome: extension of FERMT1 mutational spectrum and natural history. Hum Mutat. 2011;32:1204–12. Article  CAS  PubMed  Google Scholar  * Wang X, Chen


Q. FERMT1 knockdown inhibits oral squamous cell carcinoma cell epithelial-mesenchymal transition by inactivating the PI3K/AKT signaling pathway. BMC Oral Health. 2021;21:598. Article  CAS 


PubMed  PubMed Central  Google Scholar  * Liu CC, Cai DL, Sun F, Wu ZH, Yue B, Zhao SL, et al. FERMT1 mediates epithelial-mesenchymal transition to promote colon cancer metastasis via


modulation of beta-catenin transcriptional activity. Oncogene. 2017;36:1779–92. Article  CAS  PubMed  Google Scholar  * Li L, Li P, Zhang W, Zhou H, Guo E, Hu G, et al. FERMT1 contributes to


the migration and invasion of nasopharyngeal carcinoma through epithelial-mesenchymal transition and cell cycle arrest. Cancer Cell Int. 2022;22:70. Article  PubMed  PubMed Central  Google


Scholar  * Fan H, Zhang S, Zhang Y, Liang W, Cao B. FERMT1 promotes gastric cancer progression by activating the NF-kappaB pathway and predicts poor prognosis. Cancer Biol Ther.


2020;21:815–25. Article  CAS  PubMed  PubMed Central  Google Scholar  * Yan Q, Chen T, Yang H, Yu H, Zheng Y, He T, et al. The Effect of FERMT1 regulated by miR-24 on the growth and


radiation resistance of esophageal cancer. J Biomed Nanotechnol. 2019;15:621–31. Article  CAS  PubMed  Google Scholar  * Wang J, Chen Y, Wu R, Lin Y. Circular RNA hsa_circ_0000554 promotes


progression and elevates radioresistance through the miR-485-5p/fermitin family members 1 axis in esophageal cancer. Anticancer Drugs. 2021;32:405–16. Article  CAS  PubMed  Google Scholar  *


Jacques SL, Aquino KP, Gureasko J, Boriack-Sjodin PA, Porter Scott M, Copeland RA, et al. CARM1 preferentially methylates H3R17 over H3R26 through a random kinetic mechanism. Biochemistry.


2016;55:1635–44. Article  CAS  PubMed  Google Scholar  * Inoue F, Sone K, Toyohara Y, Tanimoto S, Takahashi Y, Kusakabe M, et al. Histone arginine methyltransferase CARM1 selective inhibitor


TP-064 induces apoptosis in endometrial cancer. Biochem Biophys Res Commun. 2022;601:123–8. Article  CAS  PubMed  Google Scholar  * Drew AE, Moradei O, Jacques SL, Rioux N, Boriack-Sjodin


AP, Allain C, et al. Identification of a CARM1 inhibitor with potent in vitro and in vivo activity in preclinical models of multiple myeloma. Sci Rep. 2017;7:17993. Article  PubMed  PubMed


Central  Google Scholar  * Zhang LX, Gao J, Long X, Zhang PF, Yang X, Zhu SQ, et al. The circular RNA circHMGB2 drives immunosuppression and anti-PD-1 resistance in lung adenocarcinomas and


squamous cell carcinomas via the miR-181a-5p/CARM1 axis. Mol Cancer. 2022;21:110. Article  PubMed  PubMed Central  Google Scholar  Download references FUNDING This work was supported by


grants from the National Natural Science Foundation of China (No. 81871961), the Key Project of the Natural Science Foundation of Fujian Province (Nos. 2022J02056, 2023D001), and the Medical


and Health Key Project of Xiamen (No. 3502Z20191106). AUTHOR INFORMATION Author notes * These authors contributed equally: Jing Lu, Huita Wu, Ping Zhan. AUTHORS AND AFFILIATIONS * Xiamen


Translational Medical Key Laboratory of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen


University, School of Medicine, Xiamen University, Xiamen, Fujian Province, China Jing Lu, Huita Wu, Ping Zhan, Yuyan Lu, Qinliang Fang, Changhong Luo, Chengrong Xie & Zhenyu Yin *


Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China Jing Lu * Department of Oncology, Zhongshan


Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China Huita Wu * Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Liver Diseases, Xiamen


Hospital of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Xiamen, Fujian Province, China Fuqiang Wang & Jing Wen Authors * Jing Lu View author publications You


can also search for this author inPubMed Google Scholar * Huita Wu View author publications You can also search for this author inPubMed Google Scholar * Ping Zhan View author publications


You can also search for this author inPubMed Google Scholar * Yuyan Lu View author publications You can also search for this author inPubMed Google Scholar * Qinliang Fang View author


publications You can also search for this author inPubMed Google Scholar * Changhong Luo View author publications You can also search for this author inPubMed Google Scholar * Fuqiang Wang


View author publications You can also search for this author inPubMed Google Scholar * Jing Wen View author publications You can also search for this author inPubMed Google Scholar *


Chengrong Xie View author publications You can also search for this author inPubMed Google Scholar * Zhenyu Yin View author publications You can also search for this author inPubMed Google


Scholar CONTRIBUTIONS JL, HW, and PZ performed most of the experiments. YL, QF, CL, and JW made significant contributions to the data analysis. FW, JW, C,X and ZY made significant


contributions to the manuscript drafting, and revision, finalizing the version for publication, endorsing the chosen journal, and assuming responsibility for all aspects of the work.


CORRESPONDING AUTHORS Correspondence to Fuqiang Wang, Jing Wen, Chengrong Xie or Zhenyu Yin. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. ETHICS


APPROVAL AND CONSENT TO PARTICIPATE The Ethics Committee of Zhongshan Hospital of Xiamen University granted approval for the study (No. 2024-555), and informed consent was obtained from the


participating patients. All methods were performed in accordance with the relevant guidelines and regulations. The Animal Care and Use Committee of Xiamen University approved the animal


study, which adhered to the fundamental principles of animal experimentation. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in


published maps and institutional affiliations. Edited by Stephen Tait SUPPLEMENTARY INFORMATION SUPPLEMENTARY FIGURE LEGENDS SUPPLEMENTARY FIGURE 1 SUPPLEMENTARY FIGURE 2 SUPPLEMENTARY


FIGURE 3 ORIGINAL IMAGES OF WESTERN BLOTTING RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,


sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative


Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated


otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds


the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Reprints and


permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Lu, J., Wu, H., Zhan, P. _et al._ PSMD14-mediated deubiquitination of CARM1 facilitates the proliferation and metastasis of hepatocellular


carcinoma by inducing the transcriptional activation of FERMT1. _Cell Death Dis_ 16, 141 (2025). https://doi.org/10.1038/s41419-025-07416-3 Download citation * Received: 19 July 2024 *


Revised: 13 January 2025 * Accepted: 31 January 2025 * Published: 27 February 2025 * DOI: https://doi.org/10.1038/s41419-025-07416-3 SHARE THIS ARTICLE Anyone you share the following link


with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt


content-sharing initiative


Trending News

Patient mutations alter atrx targeting to pml nuclear bodies

ABSTRACT ATRX is a SWI/SNF-like chromatin remodeling protein mutated in several X-linked mental retardation syndromes. G...

Location, location, location: trump has the best spot in american politics

History can weigh heavily on a filmmaker, and that is what happens with “Amelia,” a disappointing rendering of the remar...

Rear viewing system for safe reversing - farmers weekly

26 OCTOBER 2001 ------------------------- REAR VIEWING SYSTEM FOR SAFE REVERSING SPALDINGS has now launched a full colou...

Gang invades guerrero town, terrorizes residents for not paying extortion

An armed gang known as Los Cuernudos stormed the community of Barrio Lozano in Guerrero’s Costa Grande region Sunday, te...

How to be happy: 5 easy ways to increase your happiness

Memorial Day Sale! Join AARP for just $11 per year with a 5-year membership Join now and get a FREE gift. Expires 6/4  G...

Latests News

Psmd14-mediated deubiquitination of carm1 facilitates the proliferation and metastasis of hepatocellular carcinoma by inducing the transcriptional act

ABSTRACT Hepatocellular carcinoma (HCC) is a highly potent malignancy. The enzyme coactivator-associated arginine methyl...

The first front line: aspen area physicians support patients, navigate covid-19 crisis

* CHIEF OF STAFF DR. CATHERINE BERNARD STANDS IN AN EXAMINATION ROOM IN THE ASPEN VALLEY HOSPITAL EMERGENCY DEPARTMENT O...

Neuronal expression of the transcription factor gli1 using the tα1 α-tubulin promoter is neuroprotective in an experimental model of parkinson's disea

ABSTRACT Nigrostriatal neurons degenerate during Parkinson's disease. Experimentally, neurotoxins such as 6-hydroxy...

Kenyan doctors mourn death of young colleague from covid-19

Kenyan doctors mourn death of young colleague from COVID-19 | WTVB | 1590 AM · 95.5 FM | The Voice of Branch County Clos...

Something went wrong, sorry. :(

Droits de douane : la France craint une situation « intenable » pour ses principales filières d’exportation vers les Eta...

Top