Abstract
Background
Metabolic dysfunction associated steatotic liver disease (MASLD), closely linked to excessive lipogenesis, induces chronic liver disease. MASLD often cause other metabolic diseases, such as cardiovascular disease, diabetes and obesity. However, the mechanism of N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) mRNA modification in lipogenesis of MASLD has not been fully elucidated. This study investigated the role of NAT10 in lipogenesis targeting mRNA ac4C modification.Methods
The expression of NAT10 in mouse liver was assessed after a 12-week high-fat diet. In addition, the expression of NAT10 also was detected after AML12 hepatocytes cells were treated with 150 µmol/L palmitic acid (PA). The ac4C mRNA modification was performed by dot blotting. Oil red O staining and the mRNA expression of Srebf1, Acaca and Fasn were used to assess lipogenesis in AML12 cells with NAT10 overexpression or knockdown. acRIP-PCR and NAT10 RIP-PCR were used to verify the Srebf1 and Scap mRNA ac4C modification by NAT10. Furthermore, the liver lipogenesis was evaluated by AAV-mediated target knockdown of NAT10 in mouse liver and treating a specific inhibitor, Remodelin.Results
This study revealed that NAT10 is significantly upregulated in liver lipogenesis after a 12-week high-fat diet. NAT10 and ac4C mRNA modification were also drastically increased in AML12 cells after treated with 150 µmol/L PA. Silencing of NAT10 notably inhibited the lipogenesis in AML12 cells and AAV-mediated target knockdown of NAT10 in mouse liver. The acRIP-PCR and NAT10-RIP-PCR revealed that NAT10 ac4C modified Srebf1 and Scap mRNA, the critical modulator of liver lipogenesis, to regulate liver lipogenesis. Besides, Remodelin strongly inhibited liver lipogenesis, including liver TG, serum ALT, AST, TG and TC level and glucose metabolism.Conclusions
NAT10 mediates ac4C modification of Srebf1 and Scap mRNA, thereby affecting lipogenesis in the liver. This study provided a new target for the treatment of MASLD.Free full text
NAT10 promotes liver lipogenesis in mouse through N4-acetylcytidine modification of Srebf1 and Scap mRNA
Abstract
Background
Metabolic dysfunction associated steatotic liver disease (MASLD), closely linked to excessive lipogenesis, induces chronic liver disease. MASLD often cause other metabolic diseases, such as cardiovascular disease, diabetes and obesity. However, the mechanism of N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) mRNA modification in lipogenesis of MASLD has not been fully elucidated. This study investigated the role of NAT10 in lipogenesis targeting mRNA ac4C modification.
Methods
The expression of NAT10 in mouse liver was assessed after a 12-week high-fat diet. In addition, the expression of NAT10 also was detected after AML12 hepatocytes cells were treated with 150 µmol/L palmitic acid (PA). The ac4C mRNA modification was performed by dot blotting. Oil red O staining and the mRNA expression of Srebf1, Acaca and Fasn were used to assess lipogenesis in AML12 cells with NAT10 overexpression or knockdown. acRIP-PCR and NAT10 RIP-PCR were used to verify the Srebf1 and Scap mRNA ac4C modification by NAT10. Furthermore, the liver lipogenesis was evaluated by AAV-mediated target knockdown of NAT10 in mouse liver and treating a specific inhibitor, Remodelin.
Results
This study revealed that NAT10 is significantly upregulated in liver lipogenesis after a 12-week high-fat diet. NAT10 and ac4C mRNA modification were also drastically increased in AML12 cells after treated with 150 µmol/L PA. Silencing of NAT10 notably inhibited the lipogenesis in AML12 cells and AAV-mediated target knockdown of NAT10 in mouse liver. The acRIP-PCR and NAT10-RIP-PCR revealed that NAT10 ac4C modified Srebf1 and Scap mRNA, the critical modulator of liver lipogenesis, to regulate liver lipogenesis. Besides, Remodelin strongly inhibited liver lipogenesis, including liver TG, serum ALT, AST, TG and TC level and glucose metabolism.
Conclusions
NAT10 mediates ac4C modification of Srebf1 and Scap mRNA, thereby affecting lipogenesis in the liver. This study provided a new target for the treatment of MASLD.
Supplementary Information
The online version contains supplementary material available at 10.1186/s12944-024-02360-1.
Background
The metabolic dysfunction associated steatotic liver disease (MASLD) has been extensively studied recently and named in 2023 [1]. The incidence rate of MASLD is about 39% worldwide, which induces liver cirrhosis, liver dysfunction and hepatocellular carcinoma [2, 3]. In addition, MASLD is strongly associated with most of the metabolic diseases, such as, obesity, diabetes, cardiovascular disease and other diseases [2, 4]. The pathological mechanism of MASLD include dysfunction of lipid uptake, transportation of fatty acids, hepatic lipogenesis and β-oxidation [5, 6], and among these, increased liver lipogenesis is one of critical pathological mode for MASLD that lead to excessive lipid deposition in the liver [7, 8]. There is a large number of studies to explore the molecular aspects contributing to MASLD [5, 9, 10], however, no studies have been reported the involvement of N4-acetylcytidine (ac4C) mRNA modification in MASLD.
The sterol regulatory element-binding proteins (SREBPs) are the core regulators of liver lipogenesis [11]. SREBPs include three subtypes: SREBP-1a, SREBP-1c, and SREBP-2 [12], among them, SREBP-1c is the predominant subtype in liver [13]. SREBPs binding to the SREBF chaperone (SCAP) and insulin induced genes (INSIGs) further form a SREBPs/SCAP/INSIGs complex, and SREBPs are transported to nucleus and activate the transcription process of acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), and stearoyl-CoA desaturase-1 (SCD1) of lipogenesis [14].
Post-transcriptional modification of mRNA is extensively studied, and the most common expression patterns include N6-methyladenine (m6A), 5-ethylcytosine (m5C), N1-methyladenine (m1A) and N7-methylguanosine (m7G) and others [15–17]. In previous studies, ac4C-modified tRNAs and 18 S rRNAs were reported [18, 19], but in 2018, Arango et al. found that ac4C is also present in mRNA [20]. N-acetyltransferase-like protein 10 (NAT10) plays an activator or inhibitor as the “writer” of ac4C, and further affect mRNA stability and translation efficiency [21–23]. In addition, ac4C modification motifs are mainly enriched in the protein coding sequence (CDS) of mRNA, a small portion of motifs are enriched in 5’-untranslated region (5’-UTR) and 3’-UTR [24]. NAT10 acetylates the N4 nucleoside ac4C of mRNA cytosine in the target gene during the translation of mRNA, which affects the spatial conformation of its cytosine and further affects the binding to ornithine, thereby significantly increases or inhibits the structural stability of mRNA, and thus improves or decreases the translation efficiency [25, 26]. So far, it’s evident that NAT10 regulates different diseases including myocardial remodeling, oocyte maturation, tumor progression via mRNA ac4C modification [27–31].
In a liver lipogenesis related study, Zhang et al. found that knockout of NAT10 lead to decreased hepatic steatosis in male offspring of high fat-diet induced mother mice [32]. In addition, knockdown of NAT10 drastically inhibit cellular lipid accumulation after human HepG2 cell were treated with palmitic acid (PA) [32]. In molecular level, by using NAT10-RNA immunoprecipitation (RIP)-PCR silencing of NAT10 inhibited Cd36/FATP2 mRNA stability to prevent hepatic lipid uptake and steatosis [32]. Moreover, silencing of NAT10 blocked free fatty acid (FFA) metabolism in human MCF7 breast cancer cells via ac4C modification of ELOVL fatty acid elongase 6 (ELOVL6), Acyl-CoA synthetase long chain family member 1 (ACSL1), Acyl-CoA synthetase long chain family member 3 (ACSL3), Acyl-CoA synthetase long chain family member 4 (ACSL4), Acyl-CoA dehydrogenase short/branched chain (ACADSB) and Acetyl-CoA acetyltransferase 1 (ACAT1) mRNA [33]. The NAT10 inhibitor, Remodelin was also reported to participate in impeding mitochondrial lipid metabolism in cancer cells [34]. Although Zhang et al.‘s research has shown that NAT10 can interact with Cd36/FATP2 mRNA via NAT10-RIP-PCR, in HepG2 and MCF7 cells cancer cells [32, 33], whether NAT10 can directly regulate liver lipogenesis via mRNA ac4C modification was not verified.
In this study, NAT10 was found significantly upregulated in high-fat diet mice liver. NAT10 positively regulated lipogenesis in mice AML12 hepatocytes cells and mice liver lipogenesis via ac4C modified Srebf1 and Scap mRNA. This study may provide a new treatment target for MASLD.
Materials and methods
Culture of AML12 and HepG2 c ells and PA or Remodelin treatment
The AML12 and HepG2 cell were purchased from China National Collection of Authenticated Cell Cultures. AML12 cells were maintained in Gibco’s DMEM/F-12 (Shanghai, China) media enriched with 10% fetal bovine serum (FBS, Sigma, F8687, Australia), ITS Liquid Media Supplement (Sigma, I3146, MO, USA) and 40 ng/ml Dexamethasone (Sigma, D4902, Shanghai, China). In addition, AML12 cells were treated with 150 µmol/L PA (Sigma, P0500, Shanghai, China) or 20 µmol/L Remodelin (Beyotime, SD1168, Shanghai, China) for 48 h. HepG2 cells were cultured in Gibco’s DMEM (Shanghai, China) media containing 10% FBS.
Real-time quantitative reverse transcription PCR (RT-qPCR)
RNAs were isolated from AML12 cells or liver tissues using Triquick reagent (Solarbio, R1100, Beijing, China), and subsequently were reverse transcribed to cDNA (Solarbio, RP1100, Beijing, China). qPCR with 2×SYBR Green PCR Mastermix (Solarbio, SR1110, Beijing, China) was used to detect relative mRNA expression, normalized to Gapdh expression. Primer sequences are provided in Supplementary Table 1.
Oil red O staining and FFA quantification
After AML12 cells were incubated with 150 µmol/L PA for 48 h, the cells were first washed with PBS and fixed with 4% paraformaldehyde, followed by multiple washes with water. Next, the cells were then washed with water for 3 times, incubated with diluted Oil red O (Beyotime, C0157S, Shanghai, China) for 10 min, and were washed with water again. The photographs were taken with microscope (Leica DM IL LED Fluo, Wetzlar, Germany). The liver tissue slices were added to diluted Oil red O, and incubated for 20 min, decolorized with 70% alcohol, and then washed with water again. Next, the liver slices were further stained using hematoxylin. The cellular FFA content were detected with FFA content assay kit (Solarbio, BC0590, Beijing, China).
Immunoblotting
The AML12 cells or liver tissue protein samples were extracted with ProteoPrep® total protein isolation kit (Sigma, MO, USA), and the content was detected using the BCA quantitative analysis kit (Sigma, Shanghai, China). Subsequently, approximate 100 µg protein was separated by SDS-PAGE (70 V, 30 min then 120 V, 90 min) and transferred (300 mA, 60 min) and transferred to PVDF membranes (Millipore, MA, USA). The membrane was then blocked with 5% non-fat milk for 2 h, and incubated in diluted primary antibody including NAT10 (13365-1-AP), SREBP-1 (14088-1-AP), ACC1 (21923-1-AP), FASN (10624-2-AP) and β-actin (66009-1-Ig) [all provided by Proteintech, Wuhan, China], SCAP (Abcam, ab308060, MA, USA) and Cd36 (Abcam, ab255331, MA, USA). The next day, the membranes were incubated with HRP-labeled secondary antibodies for 1 h, and after washed for 3 times, chemiluminescence reaction was performed by using BeyoECL plus kit (Beyotime, P0018S, Beijing, China). Images were captured using the GelDoc XR Biorad system (Bio-Rad, CA, USA) for further analysis.
mRNA ac4C dot blot
The mRNA was isolated from about 10 µg of total RNA using Dynabeads™ mRNA purification kit (Invitrogen, CA, USA), and mRNA was denatured at 95 °C for 5 min, and was placed at ice immediately for 1 min. Next, the mRNA was added to PVDF membranes (Millipore, MA, USA), the membrane was cross-linked for 3 times at ultraviolet 254 nm, and subsequently were blocked with 5% non-fat milk containing 0.1% PBST. The membrane was incubated with anti-ac4C antibody (Abcam, ab252215, MA, USA) overnight at 4 °C. The membrane was washed for 3 times and incubated using an anti-rabbit IgG-HRP (Sigma, MO, USA) for 1.5 h at room temperature, and then washed for 3 times. The membrane was incubated with ECL (Beyotime, P0018S, Shanghai, China) and 0.02% methylene blue (Solarbio, M8030, Beijing, China) was used to stain membrane. After that, membrane was washed with RNase-free water (Solarbio, R1600, Beijing, China) for 1.5 h, and the membrane was photographed with ChemiDoc imaging system (Bio-Rad, CA, USA).
Hepatocyte-targeted mouse adeno-associated virus (AAV) shRNA-NAT10 plasmid construction
The overexpressed NAT10 (OE-NAT10, NM_153126. 4), OE-Srebf1, OE-Scap and overexpressed negative control plasmid (OE-NC), siRNA NAT10 (siRNA-NAT10) and the siRNA negative control plasmid (siRNA-NC) was constructed by Shanghai Sanggong Biotechnology Co., Ltd, China. Hepatocyte-targeted AAV-shRNA-NAT10 and AAV-shRNA-NC plasmids were designed and constructed by Shanghai Gene Chemistry Co., Ltd, China. The NAT10 target sequence was 5’-GCTGGATTTGTTCCTGTCTAT-3’, and element sequences was pAAV-ApoE/hAATp-EGFP-MIR155(MCS)-SV40 PolyA. The cells were seeded in six-well palate and when cells growth to 80–90% confluence and then were transfected by 50nM siRNA-NAT10 and siRNA-NC.
Cell transfection
When AML12 cells were grown in approximately 80% confluency, the cells were transfected with OE-NAT10, OE-Srebf1, OE-Scap, OE-NC, siRNA-NAT10 and siRNA-NC with Lipo 5000 (Solarbio, L3200, Beijing, China) according to operation manual. After 48 h, the cells were treated with or without 150 µmol/L PA.
ac4C-specific RNA immunoprecipitation (RIP) PCR (acRIP-PCR) and NAT10-RIP-PCR
After AML12 cells transfected with siRNA-NAT10 or siRNA-NC for 48 h, the total RNA was isolated and mRNA was purified using a mRNA purification kit (Invitrogen, CA, USA). The acRIP (Millipore, 17–700, MA, USA) was performed. Briefly, the mRNA was incubated with anti-ac4C antibody (Abcam, ab252215, MA, USA) and anti-IgG antibody (Abcam, ab172730, MA, USA) using Protein G magnetic beads. The ac4C modified mRNA was collected and further were amplified using RT-qPCR. NAT10-RIP-PCR method was performed according to acRIP-PCR. The Srebf1 and Scap primers were designed according to ac4C modification sites predicted by PACES software (http://www.rnanut.net/paces/). The primer sequences are listed in Supplementary Table 1.
RNA decay assay
After AML12 cells were transfected with siRNA-NAT10 and siRNA-NC for 48 h, the two group of cells were harvested after 5 µg/mL Actinomycin D (Selleck, S8964, TX, USA) treatment for 0, 2, 4, 6 h. The total RNA was extracted and reverse transcribed, and then the Srebf1 and Scap expression were amplified using qPCR.
Dual-luciferase reporter assay
According to Srebf1 and Scap mRNA ac4C motif site predicted by PACES software, the pGL3-basic-Srebf1-wild type (Srebf1-WT), pGL3-basic-Scap-wild type (Scap-WT), pGL3-basic-Srebf1-mutation (Srebf1-MUT, mutation sequence: CTGCTGGCAAGTCTC) and pGL3-basic-Scap-mutation (Scap-MUT, mutation sequence: CGACCGAATACCCGA) plasmid were constructed. Approximately 1 µg plasmid was transfected into HEK293T and evaluation of firefly luciferase and renilla luciferase was carried out using a dual-luciferase reporter assay kit (Solarbio, D0011, Beijing, China).
In vivo study
To explore NAT10 and mRNA ac4C expression in liver tissue following a high-fat diet in mice, 6-weeks-old male mice were fed by chow diet (D12450J) (n=5) and high-fat diet (D12492), all feed were provided by Research Diets, NJ, USA (n=5) for 12 weeks, the liver tissue were collected at 12th week. In addition, for AAV mediated NAT10 hepatocyte-targeted silencing experiment, 6-weeks-old male C57BL/6J mice were injected with hepatocyte-targeted AAV-shRNA-NAT10 (4×1011v.g, n=5) or AAV-shRNA-NC (4×1011v.g, n=5) through tail vein injection, and then subsequently were fed with a high-fat diet for 12 weeks. Additionally, two group of mice were injected again using same dose at 4th week. Furthermore, for Remodelin gavage experiment, male 6-weeks-old C57BL/6J mice were gavage with DMSO (n=5) and Remodelin (100 mg/kg/day, n=5), respectively, and two groups also were fed with high-fat diet for 12 weeks. All animal procedures were conducted in accordance with ethical guidelines approved by the Animal Ethics Committee of The Third Xiangya Hospital of Central South University, Changsha, China, (Approval NO: 2023-S027).
Hematoxylin and eosin (H&E) staining
The liver tissues were fixed with 4% paraformaldehyde for overnight at 4 °C, and the liver tissue were paraffin imbedded and cut into 5µM sections. After deparaffinized and rehydrated, the specimens were stained with H&E staining kit (Solarbio, G1120, Beijing, China). Microscopic images were captured using a Leica DM IL LED Fluo microscope (Wetzlar, Germany).
Biochemical analysis
The alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity were assayed by corresponding assay kits (BC1555 and BC1565, Solarbio, Beijing, China) along with the triglyceride (TG) and cholesterol (TC) assay kits (BC0625 and BC1985, Solarbio, Beijing, China). 100 mg of liver tissue were added 1mL of a mixture containing n-heptane and isopropyl alcohol at a volumetric ratio of 1:1, and were centrifuged at 10,000 rmp at 4. The supernatant was used to detect liver TG content.
Glucose tolerance test (GTT) and insulin tolerance test (ITT)
For AAV mediated NAT10 hepatocytes targeted silencing experiment and Remodelin gavage experiment, after 12 weeks, mice were subjected to a fasting period of 12 h, the mice were then intraperitoneally injected with 1.5 g/kg of glucose and their blood glucose levels were monitored at various time points −0, 30, 60, 90, and 120 min post-injection. In addition, for ITT, after mice were fasted for 12 h, they were injected with 1 IU/kg of insulin (Novo Nordisk, Copenhagen, Denmark), the blood glucose levels were measured at the same intervals as the GTT experiment. Area under the curve (AUC) for GTT and ITT were calculated by Graphpad Prism 10 software.
Statistics analysis
The data analysis was conducted using GraphPad Prism 10 software. A Student’s t-test was used to compare data from two groups, while one-way analysis of variance (ANOVA) was used to analyze and compare multiple groups. The significance level was set at P<0.05 to determine statistically significant differences among the groups.
Results
NAT10 is elevated in liver tissue of high-fat diet mice
To investigate the NAT10 expression in liver, the NAT10 level were analyzed after mice were fed chow diet (n=5) or high-fat diet (n=5) for 12 weeks, the results revealed the Nat10 mRNA and protein expression were drastically increased (Fig. 1A-B). In addition, ac4C dot blot revealed that the total mRNA ac4C modification of liver tissue was also strongly upregulated after mice were fed high-fat diet (Fig. 1C). Furthermore, after AML12 cells were treated with 150µmol/L PA, the Nat10 mRNA, protein and total mRNA ac4C modification also were apparently increased (Fig. 1D-F). The results elucidated that high-fat diet elevated liver NAT10 expression and mRNA ac4C level. The results indicated that NAT10 may play an important role on liver lipogenesis.
NAT10 radically induces AML12 cell lipogenesis
To further explore the role of NAT10 on lipogenesis in vitro, NAT10 was overexpressed or silenced in AML12 cells for 48 h, and were treated with 150µmol/L PA, and Oil red O staining results showed that NAT10 significantly promoted lipid accumulation in AML12 cells, and conversely, silencing of NAT10 strongly inhibited lipogenesis in AML12 cells (Fig. 2A). The cellular content of FFA was greatly enhanced or reduced in NAT10 overexpressed or silenced in AML12 cells respectively after 150µmol/L PA treatment (Fig. 2B). Additionally, some critical molecules of hepatocyte lipogenesis, such as, Srebf1, acetyl-CoA carboxylase alpha (Acaca), and Fasn mRNA and protein, including Cd36, were also positively regulated by NAT10, especially Srebf1 mRNA and precursor and mature SREBP-1 were increased or decreased even without PA intervention (Fig. 2C-F). Furthermore, knockdown of NAT10 suppressed mRNA ac4C modification, which was revealed by using ac4C dot blot (Fig. 2G). In addition, Oil red O staining displayed that a specific inhibitor of NAT10, Remodelin, strongly repressed AML12 cell lipogenesis (Fig. 2H), decreased cellular FFA contents (Fig. 2I), and inhibited Srebf1, Acaca, Fasn mRNA and protein, including Cd36 expression (Fig. 2J-K). The results demonstrated that NAT10 notably promoted lipogenesis in AML12 cells.
NAT10-mediated ac4C Srebf1 mRNA modification promotes AML12 lipogenesis
To further explore the molecular mechanism of NAT10 regulated lipogenesis, some important genes who participate in hepatocyte lipogenesis, were evaluated by PACES website prediction. Srebf1 mRNA was predicted as a target gene ac4C modified by NAT10 (score: 0.3281) (Fig. 3A). The prediction site was found located at 1939 to1953 of mRNA sequence, belonging to CDS. The motif sequence “CTGCTCCAGCGTCTC” is a characteristic ac4C motif “CXXCXXCXX” structure (Fig. 3B). To verify whether Srebf1 mRNA was ac4C modified by NAT10, after silencing NAT10, acRIP-PCR was used to detect the Srebf1 mRNA ac4C modification level. The results proved that knockdown of NAT10 downregulated Srebf1 mRNA ac4C modification level (Fig. 3C). Moreover, NAT10-RIP-PCR also confirmed that NAT10 was able to bind to Srebf1 mRNA and silencing of NAT10 decreased Srebf1 mRNA (Fig. 3D). Besides, the RNA decay assay exhibited that silencing of NAT10 significantly decreased Srebf1 mRNA half-life time (Fig. 3E). Next, to explore whether NAT10 can directly bind to Srebf1 mRNA ac4C modification site, dual-luciferase reporter assay elucidated that mutated Srebf1 mRNA decrease the combination with NAT10, which strongly supported the fact that NAT10 can bind to the motif of Srebf1 mRNA (Fig. 3F). Furthermore, overexpression or inhibition of NAT10 increased or decreased with or without PA-induced the precursor and mature SREBP-1 protein expression (Fig. 3G). The results illustrated that NAT10 induced lipogenesis in AML12 cells via ac4C modification of Srebf1 mRNA. In addition, mice CCAAT/enhancer binding protein beta (Cebpb) (score: 0.4322), Fasn (score: 0.5839) and human SREBF1 (score: 0.2318) and FASN (score: 0.6972) were predicted as possibly ac4C modified, using PACES software. However, despite high prediction score, acRIP-PCR verified that NAT10 did not affect their mRNA ac4C modification (acRIP-PCR of human SREBF1 and FASN were performed in HepG2 cells) (Fig. 3H-K). In addition, the rescue experiment showed overexpression of Srebf1 was able to partially increase FASN and Cd36 protein expression after AML12 was knockdown of NAT10 (Fig. 3L).
NAT10 ac4C modified Scap mRNA
In addition, another critical hepatocyte lipogenesis factor, mouse Scap, was also predicted as a target gene, ac4C modified by NAT10 (score: 0.2613) (Fig. 4A). The motif sequence, CGACGACGCCCCCGA, is located from 2807 to 2821 of Scap mRNA, also positioned at CDS (Fig. 4B). acRIP-PCR revealed that silencing of NAT10 inhibited Scap mRNA ac4C modification (Fig. 4C). NAT10 also was verified that it can bind to Scap mRNA and silencing of NAT10 decrease Scap mRNA via NAT10-RIP-PCR (Fig. 4D). In addition, inhibition of NAT10 attenuated Scap mRNA stability and mRNA and protein level expression (Fig. 4E-G). Furthermore, NAT10 was validated to directly bind to Scap mRNA ac4C modification through dual-luciferase reporter assay (Fig. 4H). The rescue experiment also showed that upregulated Scap was able to partially improve FASN and Cd36 protein expression after NAT10 knockdown in AML12 cells (Fig. 4I). The results indicated that NAT10 regulated Scap mRNA ac4C modification in AML12 cells.
NAT10 promoted liver lipogenesis via Srebf1 and Scap mRNA ac4C modification in vivo
Liver-specific knockout of NAT10 has been shown to reduce liver steatosis in male offspring mice after maternal high-fat diet [30]. However, there is no direct evidence that NAT10 participate in liver lipogenesis via mRNA ac4C modification. To explore whether NAT10 regulate liver lipogenesis in vivo, hepatocyte-targeted AAV-shRNA-NAT10 (n=5) and AAV-shRNA-NC (n=5) were injected through tail vein of mice at 0 and 4th week, while simultaneously fed a high-fat diet for 12 weeks (Fig. 5A). The results showed that although body weight was not significantly different in two groups, liver size was smaller and lighter in weight in AAV-shRNA-NAT10 than AAV-shRNA-NC group (Fig. 5B-D). In addition, the pathological staining like H&E and Oil red O staining showed that silencing of NAT10 inhibited liver lipid droplet formation (Fig. 5E-F). Moreover, TG content of liver, serum ALT, AST, TC and TG concentration were decreased in AAV-shRNA-NAT10 (Fig. 5G-K). Besides, GTT results showed that blood glucose level in AAV-shRNA-NC was similar to AAV-shRNA-NAT10 group except for at 30 min, however, there was no difference in AUC of GTT, ITT and AUC of ITT. These results indicated normal glucose tolerance and insulin sensitivity (Fig. 5L-O). In molecular level, acRIP-PCR results revealed that silencing of NAT10 decreased Srebf1 and Scap mRNA ac4C modification (Fig. 5P, Q). In addition, knockdown of NAT10 reduced Srebf1, Scap, Acaca and Fasn mRNA and protein expression in liver (Fig. 5R, S). The results suggested that NAT10 inhibited liver lipogenesis through the downregulation of Srebf1 and Scap mRNA ac4C modification.
Remodelin significantly inhibits high-fat diet induced liver lipogenesis through Srebf1 and Scap mRNA ac4C modification
To test Remodelin, the inhibitor of NAT10, whether can inhibit liver lipogenesis after mice were fed with high-fat diet. The mice were gavage with Remodelin (100 mg/kg/day, n=5) or DMSO (n=5) and high-fat dieted for 12 weeks (Fig. 6A). The results elucidated that Remodelin significantly inhibited mice weight, liver size and weight (Fig. 6B-D). H&E and Oil red O staining showed that Remodelin almost completely inhibited liver lipid droplets form (Fig. 6E-F). In addition, liver TG, serum ALT, AST, TC and TG were also apparently decreased in Remodelin treated group compared to DMSO treated group (Fig. 6G-K). In glucose metabolism, Remodelin also improved glucose tolerance compared to DMSO group (Fig. 6L, M). Moreover, in ITT experiment, Remodelin decreased blood glucose level via exogenous insulin in 0, 30, 60 min, although there was no difference in AUC of ITT, illustrating increased insulin sensitivity (Fig. 6N, O). Furthermore, acRIP-PCR showed that Remodelin also inhibited Srebf1 and Scap mRNA ac4C modification in liver (Fig. 6P, Q). In addition, Remodelin inhibited Srebf1, Scap, Acaca and Fasn mRNA and protein expression in liver (Fig. 6R, S). The results showed that Remodelin decreased the liver lipogenesis via ac4C modification of Srebf1 and Scap mRNA. The molecular mechanism of NAT10 regulation of liver lipogenesis is depicted in Fig. 7.
Discussion
In a recent study, Zhang et al. revealed that knockout of NAT10 in maternal mice inhibited male offspring liver hepatic steatosis, and in human HepG2, downregulation of NAT10 prevented lipid accumulation via NAT10 bound to Cd36/FATP2 mRNA via NAT10-RIP-PCR [32]. However, there is no direct evidence to show that NAT10 regulates liver adipogenesis by ac4C modified downstream genes. In MCF7 cells, a human breast cancer cells, knockdown of NAT10 inhibited FFA metabolism targeting ac4C modified ELOVL6, ACSL1, ACSL3, ACSL4, ACADSB and ACAT1 mRNA [33]. In all, NAT10 has been verified to positively regulate lipogenesis in HepG2 and MCF7 cells. However, HepG2 and MCF7 cells both belong to cancer cells, and whether NAT10 is involved in regulating lipogenesis in normal liver cells has not been studied yet.
To find the direct evidence of NAT10 regulation of liver lipogenesis via mRNA ac4C modification, the expression of NAT10 was evaluated in liver after mice were fed high-fat diet, the results showed that NAT10 also was induced, and mRNA ac4C modification level was elevated in mice liver. In addition, NAT10 and mRNA ac4C modification also was upregulated after AML12 cells were treated with 150 µmol/L PA for 48 h. Thus, the results implied that NAT10 may act as a critical factor that regulate liver lipogenesis through mRNA ac4C modification. Although NAT10 has been reported to regulate FFA metabolism in cancer cells [33], whether NAT10 participate in regulating others genes of lipogenesis, has not been studied well. Some critical genes of lipogenesis were analyzed via PACES software, and found mice Srebf1, Scap, Cebpb, Fasn and human SREBF1 and FASN are possible genes which were ac4C modified by NAT10. The results verified that Srebf1 and Scap mRNA were two target genes which were ac4C modified by NAT10. However, although there was high prediction score of mice Cebpb, Fasn and human SREBF1 and FASN, the acRIP-PCR verified that these genes are not ac4C modified by NAT10. In addition, as a downstream target gene of Srebf1, in this study, Fasn mRNA was downregulated after silencing of NAT10. This inhibitory effect was directly related to the reduction of Srebf1, rather than being modified by ac4C mRNA modification of NAT10.
SREBP-1 is a critical and central modulator that participate in liver lipogenesis, when there is low sterol level in hepatocytes, SCAP-mediated SREBP-1 is transported from ER to Golgi, and release an amnio-terminal domain to nucleus and further activate the downstream genes involved in lipogenesis [9, 35, 36]. It’s evident that promoting SREBP-1 or SCAP can activate liver lipogenesis, while silencing SREBP-1 or SCAP can inhibit liver lipogenesis [35, 37, 38]. In this study, both of mice Srebf1 and Scap mRNA were validated as two target genes, which were ac4C modified by NAT10, and the ac4C motifs are all located at CDS. Previous studies also reported that NAT10 positively regulate target gene’s mRNA, of which motifs are located in CDS stability and mRNA expression level [20, 39–41]. In this study, NAT10 ac4C modified mice Srebf1 and Scap mRNA and also positive regulation of their expression. Additionally, NAT10 bind to Cd36/FATP2 mRNA to affect lipid uptake in HepG2 cells and ac4C modified ELOVL6, ACSL1, ACSL3, ACSL4, ACADSB and ACAT1 mRNA to affect mitochondrial FFA metabolism in cancer cells [32–34]. However, although human ELOVL6 mRNA was ac4C modified, mouse Elovl6 mRNA was not been predicted in presence of ac4C modification motifs. In this study, NAT10 can ac4C modify mice Srebf1 and Scap mRNA, therefore, the results revealed that NAT10 can regulate liver lipogenesis through ac4C modification of multiple genes.
Remodelin, the only developed NAT10 inhibitor, has been widely applied to study NAT10’s molecular function, Remodelin was found to improve Hutchinson-Gilford Progeria Syndrome cell phenotype [42], and also Remodelin was found able to prevent tumor procession [27, 43–45]. Moreover, Remodelin also was found not toxic for cell and mice experiments [40, 46–48]. Therefore, Remodelin may be used for a potential small molecular compound to treat MASLD. But it is noteworthy that in this study, some mice’s hair partially turned white after Remodelin gavage treatment for 12 weeks. From here, the potential systemic effects and long-term safety of Remodelin is needed to be explored further. In a recent study, Liu et al. found Panobinostat had a more efficient affinity with NAT10 and inhibitory effect than Remodelin, as an anti-tumor drug, Panobinostat may not be suitable for treatment of MASLD [49]. So, developing new NAT10 inhibitor in vivo and treatment of MASLD is needed by further research.
In addition, although targeted knockdown of NAT10 in liver showed to decrease liver size and lipogenesis including liver TG, serum TG, TC, ALT and AST, there was no difference in body weight and glucose metabolism. However, Remodelin strongly prevented liver lipogenesis, hepatic damage and decreased glucose tolerance. Remodelin treatment was more efficient than targeted knockdown of NAT10 in liver, possible reason is that the inhibitory effect of Remodelin on NAT10 is stronger than targeted liver knockout of NAT10. In addition, the efficient and widespread systemic inhibition of NAT10 by Remodelin, especially in adipose tissue, may further improve metabolism.
Study strengths and limitations
This study revealed that NAT10 can regulate mice liver lipogenesis via ac4C modified Srebf1 and Scap mRNA. Also, the knockdown of NAT10, as well as the administration of Remodelin, an effective NAT10 inhibitor, successfully inhibited liver lipogenesis in vivo. However, in this study, acRIP-seq was not used to comprehensively examine the ac4C-modified mRNAs involved in lipogenesis in AML12 and HepG2 cells. It was observed that some mice exhibited partial whitening of their hair following Remodelin treatment, suggesting that the safety profile of NAT10 inhibitors such as Remodelin requires further assessment.
Conclusions
In summary, in this study provides direct evidence for NAT10 regulated mice liver lipogenesis in vitro and in vivo, and knockdown of NAT10 decreased ac4C modified Srebf1 and Scap mRNA, and further decreasing their mRNA stability. Additionally, Remodelin or development of new NAT10 inhibitor may be a promising method to treat for MASLD. In all, the present study will provide a new intervention target for the treatment of MASLD.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Abbreviations
AAV | Adeno-associated virus |
ac4C | N4-acetylcytidine |
Acaca | Acetyl-CoA carboxylase alpha |
ACADSB | Acyl-CoA dehydrogenase short/branched chain |
ACAT1 | Acetyl-CoA acetyltransferase 1 |
ACC1 | Acetyl-CoA carboxylase 1 |
ACSL1 | Acyl-CoA synthetase long chain family member 1 |
ACSL3 | Acyl-CoA synthetase long chain family member 3 |
ACSL4 | Acyl-CoA synthetase long chain family member 4 |
ALT | Alanine aminotransferase |
AST | Aspartate aminotransferase |
AUC | Area under the curve |
CDS | Coding sequence |
Cebpb | CCAAT/enhancer binding protein beta |
DMSO | Dimethyl sulfoxide |
ELOVL6 | ELOVL fatty acid elongase 6 |
Fasn | Fatty acid synthase |
FBS | Fetal bovine serum |
FFA | Free fatty acids |
GTT | Glucose tolerance test |
H&E | Hematoxylin and eosin |
INSIGs | Insulin induced genes |
ITT | Insulin tolerance test |
m1A | N1-methyladenine |
m5C | 5-ethylcytosine |
m6A | N6-methyladenine |
m7G | N7-methylguanosine |
MASLD | Metabolic dysfunction associated steatotic liver disease |
NAT10 | N-acetyltransferase 10 |
PA | Palmitic acid |
RIP | RNA immunoprecipitation |
SCAP | SREBF chaperone |
SCD1 | Stearoyl-CoA desaturase-1 |
SREBPs | Sterol regulatory element-binding proteins |
Srebf1 | Sterol regulatory element binding transcription factor 1 |
TC | Total cholesterol |
TG | Triglyceride |
UTR | Untranslated region |
Author contributions
All authors were involved in the development of the primary manuscript and revision. ZQW-performed the experiments, data analysis, manuscript drafting. XXW- performed the experiments, data analysis. AK-wrote the manuscript, LP-performed the experiments, XYS-performed the experiments, XY-performed the experiments, KC-performed the experiments, data analysis, manuscript drafting, supervision, planning, data interpretation. All authors reviewed the manuscript.
Funding
This study received funding from the Hunan Provincial Health Commission for the Health Research Project (grant number: W20243129).
Declarations
The animal experiments conducted at The Third Xiangya Hospital of Central South University, Changsha, China adhered to strict ethical guidelines set forth by the Ethical Committee for Animal Experiments. Approval for the experiments was granted under Approval NO: 2023-S027.
Not applicable.
The authors declare no competing interests.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
Articles from Lipids in Health and Disease are provided here courtesy of BMC
Funding
Funders who supported this work.
Health Research Project of Hunan Provincial Health Commission (1)
Grant ID: W20243129