Abstract Metabolic dysfunction–associated steatohepatitis is well accepted as a potential precursor of hepatocellular carcinoma. Previously, we reported that fibroblast growth factor 21 (FGF21) revealed a novel anti-inflammatory activity via inhibiting the TLR4–IL-17A signaling, which could be a potential anticarcinogenetic mechanism to prevent to MASH-HCC transition. Here, we set out to determine whether FGF21 has a major impact on Kupffer cells’ (KCs) ability during MASH-HCC transition. We found aberrant hepatic FGF21 and KC pool in human MASH-HCC. Lack of FGF21 up-regulated ALOX15, which converted the oxidized fatty acids to induce excessive KC death and mobilization of monocyte-derived macrophages (MoMFs) for KC replacement. Lack of FGF21 oversupplied free fatty acids for sphingosine-1-phosphate (S1P) cascade synthesis to mediate MASH-HCC transition via S1P-YAP signaling and cross-talk between tumor cells and macrophages. In conclusion, lack of FGF21 accelerated MASH-HCC transition via the S1P-AP signaling. Compromised MoMFs could present as tumor-associated macrophage phenotype rendering tumor immune microenvironment for MASH-HCC transition. __________________________________________________________________ FGF21 could prevent MASH-HCC transition via preservation of KC pool homeostasis and inhibition of S1P-S1PR2-YAP signaling. INTRODUCTION Metabolic dysfunction–associated steatohepatitis (MASH), formerly known as nonalcoholic steatohepatitis (NASH), a severe form of metabolic dysfunction–associated steatotic liver disease (MASLD; formerly known as NAFLD), is well accepted as a potential precursor of hepatocellular carcinoma (HCC) ([42]1). Using a Markov model to forecast MASLD progression, MASH is predicted to increase 27%, while the incidence of HCC will increase 137% by 2030 ([43]2). MASH is becoming a main cause for end-stage liver disease, in particular malignancy, as a major clinical and economic burden ([44]3). Although MASH was first documented more than 30 years ago ([45]4), its pathogenesis/carcinogenesis is not fully elucidated. Because early and effective treatment may determine the long-term MASH prognosis ([46]5), there is an unmet need to identify the factor(s) that can be targeted therapeutically to prevent MASH progression. Previously, we reported that fibroblast growth factor 21 (FGF21) revealed a novel anti-inflammatory activity via inhibiting the Toll-like receptor 4 (TLR4)–interleukin-17A (IL-17A) signaling, which could be a potential anticarcinogenetic mechanism to prevent to MASH-HCC transition ([47]6). Regarding the tumor microenvironment in MASH liver, studies have emphasized the pathophysiological importance of TLR4 from hepatic resident Kupffer cells (KCs) ([48]7–[49]9) and stellate cells ([50]10). Of note, both hepatic resident KCs and infiltrated immune cells [monocyte-derived macrophages (MoMFs), natural killer (NK) cells, neutrophils, NKT cells, and T cells] are known as inflammation/immune mediators that contribute to MASH-HCC transition. KCs account for approximately 15% of the total liver cell population and are the largest population constituting 80 to 90% of the total number of tissue-resident macrophages in whole body. The maintenance of KC pool has been thought to involve two mechanisms, (i) replenishment by KC self-renewal and (ii) recruitment from bone marrow–derived monocytes (BMDMs) ([51]11). In MASH scenario, underlying self-renewal of hepatic resident KCs could be impaired, while the KC pool was gradually seeded by the MoMFs, which could be partly immature or less efficiently than the hepatic resident KCs, but exhibited a pro-inflammatory effect to exacerbate liver damage ([52]12). In MASH, metabolism of KCs/MoMFs can be reprogrammed to fulfill carcinogenetic processes, i.e., decreased glycolysis and fatty acid (FA) oxidation but increased FA synthesis ([53]13). KCs/MoMFs represent as one of the most abundant innate immune cells, while macrophage polarization has been associated with tumor growth, metastasis, angiogenesis, and poor prognosis ([54]14). Aberrant signaling from immature macrophages could be deleterious, but little is known about the KCs/MoMFs’ metabolic changes, which may determine functional fate of the macrophages during HCC development. FGF21, an endocrine FGF primarily produced in liver ([55]15, [56]16), elicits metabolic benefits protect liver against metabolic stress caused by MASH ([57]17). This major endocrine action of FGF21 results in a combination of effects, including control of lipolysis, clearance of excessive free FAs (FFAs), enhanced expenditure of the stored lipid energy by mitochondrial substrate oxidation, and catabolism and uncoupling, which negatively regulates hepatic or tissue steatosis and adiposity ([58]18, [59]19). Pharmacological application of FGF21 holds great promise as an effective therapeutic means for treating obesity, diabetes, and MASH ([60]20–[61]29). However, no previous study has been performed to investigate the role of FGF21 played on KCs/MoMFs, in particular, maintenance of the KC pool during MASH-HCC transition. Here, we set out to determine whether FGF21 has a major and long-lasting impact on KCs/MoMFs’ ability that in turn alters the response to lipotoxicity during MASH-HCC transition. Our findings reveal that compromised function of macrophages contribute to MASH progression to HCC, while FGF21 plays a critical role to maintain the KC pool to prevent MASH-HCC transition. RESULTS Aberrant hepatic FGF21 and KC pool in human MASH-HCC In the specimens of patients with HCC with medical history of MASH, the pathological features of MASH-HCC were confirmed by hematoxylin and eosin (H&E) and Oil Red O staining. Much weaker staining of FGF21 was found in the tumor region in comparison with the adjacent benign region ([62]Fig. 1A). Consistently, significantly decreased protein and mRNA levels of FGF21 were found in the tumor tissues compared to the adjacent benign tissues ([63]Fig. 1B). Serological levels of FGF21 were determined by enzyme-linked immunosorbent assay (ELISA) in the postresection patients with HCC and followed up to 9 months. In comparison with preoperation, 8 of 10 patients (no relapse) showed increases of postresection FGF21 levels, while only 3 of 10 patients (relapse) showed increases of postresection FGF21 levels ([64]Fig. 1C). In 156 overweight [body mass index (BMI) ≥ 25 kg/m^2] patients with HCC from The Cancer Genome Atlas (TCGA) database, 108 patients were graded as G1–2, while 48 patients were graded as G3–4, according to the National Comprehensive Cancer Network severity scale. Analysis of survival rate indicated that low expression of FGF21 had a negative impact on the survival of high-BMI patients with advanced disease (G3–4) ([65]Fig. 1D). Cumulative survival of the overweight patients with HCC was further analyzed to associate with immune cells, including macrophage, neutrophil, dendritic cell, CD4^+ cell, CD8^+ cell, and B cell. By splitting 50% of the patients with high level of cell subpopulations and 50% with low level of cell subpopulations, the Kaplan-Meier curve showed a poor outcome for the patient with high level of macrophages (fig. S1A). When CD68 (a marker highly expressed by circulating macrophages) was normalized by FGF21, high expression of CD68 was negatively correlated with the percent survival in the overweight patients with HCC (fig. S1B). By coexpression analysis, a negative correlation was also found between CD68 and FGF21 (fig. S1C). To study the tumor immune microenvironment (TIME) in which both KCs and MoMFs were distributed in liver, imaging mass cytometry was performed in the human MASH-HCC tissues because this high-plex imaging allowed visualizing multiple markers to obtain a precise picture of cell subpopulations in the tissue architecture. To provide a detailed inventory of landscape of KC pool, we characterized the macrophages in tumor and adjacent benign regions of MASH-HCC samples using an optimized eight-marker panel for Hyperion Imaging. The cell subpopulations were identified in the regions of tumor and adjacent benign ([66]Fig. 1E) for spatial distribution analyses. On the basis of the expression of lineage markers, the subpopulation included CD68^+ cells, CD68^+CD163^+ cells, and Cd68^+CD163^+CD14^+ cells. Extensively distributed CD68^+ macrophages were found in the regions either tumor or adjacent benign ([67]Fig. 1F), while relatively increased CD68^+CD163^+ macrophages (potential M2 polarization) and CD68^+CD163^+CD14^+ cells (MoMFs) were found in the tumor regions in comparison with that in the adjacent benign regions ([68]Fig. 1, G to I), which suggested the alternatively activated M2 macrophages and KC pool reseeding by MoMFs ([69]12) in TIME. Together, either decreased hepatic FGF21 or replacement of hepatic resident KCs by MoMFs negatively affected the outcome of patients with MASH-HCC. As the major metabolic benefits of FGF21 result in controlling lipid metabolism, it evoked the interest to explore the effect of FGF21 on the lipid metabolic signaling of KC pool in MASH liver. Fig. 1. Aberrant hepatic FGF21 and KC pool in human MASH-HCC. [70]Fig. 1. [71]Open in a new tab (A) Representative images of MASH-HCC histology by H&E and Oil Red O staining and immunohistochemistry detection of GF21 protein distribution in the regions of tumor (white dash circle) tissue and adjacent benign tissue. (B) Protein and mRNA levels of FGF21 by Western blotting and qPCR in the tumor and adjacent tissues. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (C) Serum FGF21 protein levels by ELISA in the postresection patients with HCC (no relapse and relapse). (D) A cohort of 156 overweight (BMI ≥ 25 kg/m^2) patients with HCC from TCGA database were subgrouped into G1–2 patients and G3–4 patients to analyze the survival rate, based on high/low expression of FGF21. (E) Representative images of Hyperion Imaging in paraffin-embedding tissues of patients with HCC. (F to H) Cell subpopulations of CD68^+ macrophages, CD68^+CD163^+ macrophages, and CD68^+CD163^+CD14^+ macrophages in tumor and adjacent tissues. (I) the ratio of CD68^+CD163^+CD14^+ macrophages in CD68^+ macrophages in tumor and adjacent tissues. *P < 0.05 and **P < 0.01. KC pool and FA metabolism in FGF21KO-MASH mice To study the effect of FGF21 on lipid metabolic signaling in KCs upon MASH insult, two MASH mouse models were established in FGF21 knockout (KO) mice and wild-type (WT) littermates by feeding a high-fat methionine/choline-deficient diet (HFMCD) or a Western-style diet (WSHFD; high fat and high fructose). The mice with HFMCD showed high severity of MASH, while the mice with WSHFD displayed the MASH characteristics with clinical relevance, particularly lipid metabolic disorders (fig. S2). The WSHFD-MASH model was therefore used for further studies. The result of whole-body calorimetry showed significant decreases of respiratory exchange ratio and energy expenditure in the FGF21KO-MASH mice (fig. S3A), while rhFGF21 treatment attenuated the changes (fig. S3B), suggesting that lack of FGF21 caused switch from glucose to FA for energy generation in the MASH liver. Macrophages were isolated from liver tissue for RNA sequencing (RNA-seq). Volcano plot showed that FGF21KO-MASH mice (versus WT-CD controls) had great numbers of differentially expressed genes (DEGs), including Dd93, Itgax, Mmp12, Gpnmb, Cd9, Fam20c, Emp1, Fabp5, Atp6v0d2, Anxa1, and Clec4e, which were uniformly expressed across all the macrophages ([72]Fig. 2A). The gene signatures of MoMF were up-regulated, but the gene signatures of KC were down-regulated in the FGF21-MASH mice ([73]Fig. 2B). These characteristics of gene expression and signature were agreed with the previous report in which MoMFs could gradually seed the KC pool in response to the death of embryo-derived KCs in MASH ([74]12). Running Enrichment Score showed that the gene signatures of FA metabolism were positively associated with the up-regulated genes of MoMF but negatively associated with genes of tissue-resident KC such as Timd4 and Cd207 genes in the FGF21KO-MASH mice ([75]Fig. 2C), suggesting that FA metabolism could play a critical role for seeding MoMFs to maintain the KC pool. As hepatic FA deposition has been reported to contribute to approximately 60% of hepatic lipid accumulation ([76]30), the enzymes for FA metabolism were therefore analyzed by quantitative polymerase chain reaction (qPCR). Increases of FA synthesis (FASN, ACC1, ACC2, SREBP1c, and PPAR-r), FA update (Slc27a1, Slc27a2, Slc27a3, and CD36), TG synthesis (Dgat1 and Acat1), and export (Mttp and Apoa1) but decrease of FA oxidation (Cpt1-a, Acox1, Scacd, Mcad, and Ppar-γ) were found in the FGF21KO-MASH mice ([77]Fig. 2D). Because FASN and CD36 were noticed among the FA enzymes, their protein levels were further determined by Western blotting. High levels of FASN and CD36 were found in FGF21KO-MASH mice in comparison with other three groups, with statistical significance ([78]Fig. 2E). Restoration of FGF21 with rhFGF21 injection (125 μg/kg body weight) daily for 4 weeks resulted in significantly decreased FASN and CD36 ([79]Fig. 2F) in consistent with the alleviation of MASH, evidenced by the histological changes of steatohepatitis ([80]Fig. 2F) and decreased NAFLD activity score (NAS; fig. S3C). We next asked whether lack of FGF21 could increase FA synthesis and transport. The in vitro studies were performed using FL83B cells (a benign mouse cell line of hepatocyte) and FL83B-FGF21KD cells in which FGF21 gene was knocked down (KD) by short hairpin RNA. Significantly increased FASN and CD36 were found when the FL83B-FGF21KD cells were challenged with palmitic acid (PA) at 100 μM for 48 hours ([81]Fig. 2G), while PA-induced steatosis and increases of FASN and CD36 were significantly attenuated by rhFGF21 treatment (fig. S3, D and E). To elucidate whether lack of FGF21 could liberate FAs from adipose tissue, we measured the protein phosphorylation of hormone-sensitive lipase (HSL), which had broad substrate specificity to catalyze the hydrolysis of triacylglycerol, diacylglycerol, and 1(3)-monoacylglycerol ([82]31). Significantly increased phosphorylation of HSL at Ser563 and Ser660, two major sites for the hydrolytic activity for triacylglycerol, was found in the adipose tissues from FGF21KO-MASH mice ([83]Fig. 2H). Significantly increased phosphorylation of HSL at Ser563 and Ser660 was also found in the FGF21KO mice compared to the WT mice ([84]Fig. 2H). All these results indicated the critical roles of FGF21 played in controlling of adipose tissue hydrolysis to liberate FAs, FA de novo synthesis, and FA accumulation in liver. To characterize the specificity of FA metabolites in FGF21KO-MASH mice, long-chain FAs (LCFAs) were analyzed. Two specific oxidized FAs, Hydroxyoctadecadienoic acids (HODE) and 15-hydroxyeicosatetraenoic acids (15-HETE), which are converted by Alox15 from arachidonic acid and linoleic acid ([85]32, [86]33), were noticed in the liver tissue of FGF21KO-MASH mice ([87]Fig. 2I). Although the oxidized FAs such as HODE and HETE have been reported for MASH development ([88]34), there is no study to report the Alox15 signaling in MASH progression. We thus investigated Alox15 expression in FG21KO-MASH mice thereby to understand FGF21-Alox15 signaling in disease progression and carcinogenetic transformation. Fig. 2. KC pool and FA metabolism in FGF21KO-MASH mice. [89]Fig. 2. [90]Open in a new tab (A) Volcano plot of DEGs (log2 fold change >1.5) by RNA-seq analysis in the isolated macrophages between FGF21KO-MASH mice and WT-CD controls. (B) Heatmap of the signature genes for monocytes and KCs by RNA-seq analysis between FGF21KO-MASH mice and WT-CD controls. (C) Running Enrichment Score of transcripts of genes of KCs/macrophages associated with fatty acid (FA) metabolism. (D) Heatmap of enzymes for FA metabolism by qPCR. Protein levels of FASN and CD36 by Western blotting in: (E) the WT/FGF21KO mice with WSHFD/CD diets, (F) the FGF21KO-MASH mice with rhFGF21 treatment compared to vehicle controls and WT-CD controls, and (G) FL83B/FL83B-FGF21KD cells treated with palmitic acid (PA), while 1% bovine serum albumin (BSA) was used as treatment control. (H) Protein levels of HSL and phosphorylated HSL at S563 and S660 by Western blotting in the WT/FGF21KO mice with WSHFD/CD diets. (I) Heatmap of long-chain FAs (LCFAs) detected by Waters ACQUITY UPLC Systems coupled with Waters Xevo TQ-S micro triple quadrupole mass spectrometer in the WT/FGF21KO mice with WSHFD/CD diets. 21KO, FGF21KO. *P < 0.05, **P < 0.01, and ***P < 0.001. Up-regulated ALOX15 signaling in KCs from FGF21KO-MASH mice In the WSHFD-MASH model, significantly increased ALOX15 protein level was found in FGF21KO-MASH mice in comparison with other three groups ([91]Fig. 3A). The ALOX15 expression was almost undetected in the isolated hepatocytes but abundantly expressed in the isolated macrophages from liver ([92]Fig. 3B). Consistently, positive ALOX15 staining was distributed in the regions where KCs were located (fig. S4A) and coexpression of F4/80 and ALOX15 was confirmed by immunofluorescent staining in the isolated macrophages ([93]Fig. 3C). To determine whether the oxidized FA could induce KC death, an in vitro study was performed using the isolated mouse KCs from WT and FGF21KO mice. As expected, 13(S)-HODE challenging caused significant increases of cleaved caspase-3 (C-casp3) and FAS, while treatment with N-acetyl-l-cysteine (NAC), a commonly used antioxidant against reactive oxygen species, attenuated levels of C-casp3 and FAS ([94]Fig. 3D). In FGF21KO-MASH mice, pronounced cell death was also found, evident as significantly increased apoptotic cells (fig. S4B), while the increased C-casp3 and FAS in FGF21KO-MASH mice were attenuated by restoration of FGF21 ([95]Fig. 3E). We next asked whether MoMFs could be mobilized for KC replacement. The study of thioglycollate medium–induced peritoneal macrophages showed much high mobilization ability of MoMFs and up-regulated monocyte chemoattractant protein-1/C-C motif chemokine ligand 2 (MCP-1/CCL2), the key chemokine to regulate migration and infiltration of monocytes/macrophages), from the FGF21KO mice (versus WT) (fig. S4, C and D). All these results suggested that overexpression of ALOX15 caused increases of oxidized FAs, which induced excessive KC death, while mobilization ability of MoMFs was increased in FGF21KO mice. As the ALOX15-converted oxidized FAs could accelerate MASH progression ([96]35), we wondered whether knockout of ALOX15 would prevent MASH development/progression. ALOX15KO mice were therefore chosen to establish MASH models by feeding WSHFD or HFMCD. As expected, lack of ALOX15 protected the MASH insult, evidenced by decreased NAS, which was counted by the decrease of inflammation other than steatosis or ballooning ([97]Fig. 3F), confirming that ALOX15 signaling contributed to inflammation for MASH progression. To investigate whether ALOX15 signaling could affect the balance of KCs and MoMFs in the KC pool, cytometry by time-of-flight (CyTOF) was performed to precisely analyze the KC and MoMF subpopulations. KCs were defined as F4/80^hiTim4^+CX3CR1^− cells, while MoMFs were defined as CD11b^+Tim4^−CX3CR1^+ cells. In the WSHFD-MASH model, significantly decreased KCs but significantly increased MoMFs were found in both WT-MASH mice and FGF21KO-MASH mice compared to WT-CD control mice. Treatment with either PD146176 (an inhibitor of ALox15) or rhFGF21 significantly attenuated the imbalance of KCs/MoMFs in KC pool ([98]Fig. 3G), suggesting that inhibition of ALox15 or restoration of FGF21 could preserve homeostasis of KC pool against MASH insulant. To further understand that lack of FGF21 could render inflammation/immune microenvironment contributing to MASH progression, RNA-seq data were analyzed to associate the metabolic pathway with immune response. The Kyoto Encyclopedia of Genes and Genomes heatmap of genes in metabolic pathway showed great changes in FGF21KO-MASH mice compared with WT-CD mice (fig. S4E), while up-regulated genes of immune response signaling and cytokine-cytokine interaction signaling were found in consistent with the up-regulated genes in metabolic pathway ([99]Fig. 3H). Together, FGF21 was essential against ALOX15-converted oxidized FAs to keep homeostasis of KC pool, while lack of FGF21 could render inflammation/immune microenvironment to accelerate MASH-HCC transition. Fig. 3. Up-regulated ALOX15 signaling in KCs from FGF21KO-MASH mice. [100]Fig. 3. [101]Open in a new tab (A) Protein levels of Alox15 by Western blotting in the liver tissues of WT/FGF21KO mice with WSHFD/CD diets. (B) Alox15 mRNA levels by qPCR analysis in the isolated hepatocytes and KCs and the protein levels of Alox15 by Western blotting in KCs from the WT/FGF21KO mice with WSHFD/CD diets. (C) Immunofluorescent staining using the antibodies of anti-ALOX15 and anti-F4/80 to detect the positive ALOX15 macrophages. Green, ALOX15-positive staining cells; red, F4/80-positive staining cells; blue, 4′,6-diamidino-2-phenylindole (DAPI) staining to detect the nuclei as a counterstain. (D) Protein levels of FAS and cleaved caspase-3 by Western blotting in the isolated mouse KCs challenged with 13-HODE and treated with N-acetyl-l-cysteine (NAC). (E) Protein levels of FAS and cleaved caspase-3 by Western blotting in the liver tissues from FGF21KO-MASH mice with rhFGF21 treatment compared to vehicle controls or WT-CD controls. (F) Histology-based NAFLD active score (NAS) in the WT/ALOX15KO mice with HFMCD/WSHFD diets. (G) CyTOF analysis in the isolated macrophages from mice to detect the KCs defined as F4/80^hiTim4^+CX3CR1^− cells and the MoMFs defined as CD11b^+ Tim4^−CX3CR1^+ cells in KC pool. The FGF21KO-MASH mice were treated daily with PD146176 or rhFGF21 for 4 weeks. (H) Analysis of the RNA-seq data for the mRNA fold changes of immune response signaling pathway and cytokine-cytokine interaction signaling. 21KO, FGF21KO. *P < 0.05 and **P < 0.01. Lack of FGF21 accelerates MASH-HCC transition To validate the role of FGF21 against MASH-HCC transition, four MASH-HCC models were established in WT and FGF21KO mice with diethylnitrosamine (DEN) treatment at 2 weeks old, following with (i) control diet (CD), (ii) HFMCD, (iii) WSHFD, and (iv) HFD ([102]Fig. 4A). On the basis of the appearance of tumor nodule by ultrasound (fig. S5A), the mice were euthanized at 16 weeks for DEN + HFMCD, 26 weeks for DEN + WSHFD, 28 weeks for DEN + HFD, and 36 weeks for DEN + CD ([103]Fig. 4A). Histological examination by H&E staining, Oil Red O, and Sirius Red staining confirmed the MASH, which was characterized as steatohepatitis (wildly distributed lipid drops, infiltration of inflammatory cells in the acinar zone, and hepatocyte ballooning) and HCC (showed the cytological features of cancerous cells ranging from well to poorly differentiated distributed in parenchyma showing an abnormal hepatic architecture) (fig. S5, B and C). The MASH-HCC in FGF21KO mice showed an aggressive tumor growth pattern, evident as the significantly increased number and maximal diameter of tumor nodule compared to the MASH-HCC in WT mice ([104]Fig. 4B). NAS system was applied in combination of histological and serological findings to further evaluate the characteristics of MASH-HCC. Significantly increased NAS was found in FGF21KO-MASH-HCC mice (versus WT-MASH-HCC mice) induced by either DEN + HFMCD or DEN + WSHFD ([105]Fig. 4C). Consistently, significantly increased levels of alanine transaminase (ALT) and hepatic/serum TG were found in the FGF21KO-MASH-HCC mice in comparison with WT-MASH-HCC (fig. S5D). Restoration of FGF21 was performed in the FGF21KO mice with rhFGF21 injection (125 μg/kg body weight) daily for 4 weeks in both DEN + HFMCD model and DEN + WSHFD model. Although tumor nodules were detected in the mice with rhFGF21 treatment, the tumor number was smaller and tumor size was lower, with statistical significance compared to the vehicle controls ([106]Fig. 4D). Then, we compare the KC/MoMF subpopulations between MASH mice and MASH-HCC mice. By CyTOF analysis, significantly decreased KCs but significantly increased MoMFs were found in both MASH mice and MASH-HCC mice compared to the CD control mice. No statistical significance was found between MASH mice and MASH-HCC mice; however, decreased KCs but increased MoMFs were found in the FGF21KO-MASH mice (versus WT-MASH mice) and FGF21KO-MASH-HCC mice (versus WT-MASH-HCC mice) with statistical significance ([107]Fig. 4E), suggesting that lack of FGF21 worsened the KCs’ depletion and the KC pool could be seeded by MoMFs during MASH-HCC transition. To characterize the specificity of FA metabolites in MASH-HCC mice, LCFAs were analyzed. Consistently, markedly increased ALOX15-converting productions such as HODE and arachidonic acid, a precursor of bioactive lipid mediators (prostaglandin production), were found in the liver tissues from FGF21KO-MASH-HCC mice ([108]Fig. 4F). Significantly increased protein levels of ALOX15 and the key lipolytic enzymes, cytosolic phospholipase A 2 (cPLA2), which catalyzes the hydrolysis of phospholipids to arachidonic acid (cleave and release FAs), and monoacylglycerol lipase (MAGL), which hydrolyzes intracellular TG to FAs and glycerol, were found in the FGF21KO-MASH-HCC mice ([109]Fig. 4G), suggesting the important role of bioactive FA mediators in contribution to the MASH-HCC transition. To investigate the role of FGF21 on FAs release, FFA levels were determined in three MASH-HCC models (DEN + CD, DEN + HFMCD, and DEN + WSHFD). Significant increases of FFA in the serum were found in FGF21KO mice (versus WT mice) in all three MASH-HCC models ([110]Fig. 4H), suggesting that lack of FGF21 caused increase of FFA release during MASH-HCC transition. Previous studies revealed that cPLA2 was regulated by sphingolipids ([111]36). Sphingosine-1-phosphate (S1P), one of the most active sphingolipids, had been reported to promote liver fibrosis and cancer ([112]37). Thus, S1P levels in both serum and liver tissue were investigated. As expected, significantly increased S1P levels were noticed in FGF21KO mice (versus WT mice) in all three MASH-HCC models ([113]Fig. 4I). Together, these results indicated that lack of FGF21 accelerated MASH-HCC transition, while abnormal FA metabolism and increased S1P were noticed in the FGF21KO mice during MASH-HCC transition. Fig. 4. Lack of FGF21 accelerates MASH-HCC transition. [114]Fig. 4. [115]Open in a new tab (A) Schematic diagram for establishing MASH-HCC models and the gross anatomy of tumor mass in four MASH-HCC models with CD, HFMCD, WSHFD, and HFD, respectively. (B) Maximal diameter of tumor nodules and number of tumor nodules in four MASH-HCC models. (C) Histology-based NAS in the MASH-HCC mice with HFMCD and WSHFD diets. (D) Schematic diagram for rhFGF21 treatment in the MASH-HCC mice with HFMCD and WSHFD diets. (E) CyTOF analysis of the KCs (F4/80^hiTim4^+CX3CR1^− cells) and MoMFs (CD11b^+Tim4^−CX3CR1^+ cells) from the MASH mice and MASH-HCC mice, as well as the control mice. (F) LCFAs in the FGF21KO-MASH-HCC mice compared to normal WT controls. (G) Protein levels of cPLA2, Alox15, and MAGL by Western blotting in the liver tissues of FGF21KO-MASH-HCC mice compared to normal WT controls. (H) The serum FFA levels in FGF21KO-HCC mice in comparison with the WT-HCC mice, with three diets (CD, HFMCD, and WSHFD). (I) S1P levels in serum and liver tissue from FGF21KO-HCC mice in comparison with the WT-HCC mice, with three diets (CD, HFMCD, and WSHFD). 21KO, FGF21KO. *P < 0.05, **P < 0.01, and ***P < 0.001. S1P-S1PR2-YAP signaling mediated MSAH-HCC transition in FGF21KO mice Given that the FFA and S1P levels were increased in FGF21KO-MASH-HCC mice, we next asked whether lack of FGF21 would activate the S1P signaling to promote the HCC initiation ([116]37). We established premalignant models (preHCC) in FGF21KO mice and WT littermate, with DEN-HFMCD for 14 weeks or with DEN-WSHFD for 24 weeks. In preHCC mice, no tumor was detected but the preHCC mice were characterized by increased plasma ALT activity, increased α-fetoprotein level, and elevated hepatic TG content (fig. S6A). The premalignant models recapitulated the tumor microenvironment for MASH-HCC transition; thus, we could get insight into the metabolic features of FA/S1P and carcinogenetic signaling. A qPCR assay was performed to profile gene expression for the enzyme cascade of S1P synthesis. Fourfold changes (FGF21KO-preHCC mice versus WT-preHCC mice) of up-regulated gene expression were found, including the enzymes for FFA uptake [CD36 and FABP (1, 3, and 4)] and FA de novo synthesis (ACC1 and FASN), S1P cascade synthesis [SPTLC1, CERS (1, 5, and 6), DES2, and GCS (ceramide de novo pathway), ASMase, and NSMase (sphingomyelin pathway)], sphingosine synthesis [SGPP1, ACER (1–3), and ASAH1], and S1P synthesis (SPKK1) ([117]Fig. 5A). This result suggested the oversupplied FFA and the substrates for S1P cascade synthesis in FGF21KO mice during MASH-HCC transition. Consistently, significant increase of phosphorylated SPHK1 protein along with the increased S1P level was found in liver tissues from FGF21KO-preHCC mice, while treatment with rhFGF21 (125 μg/kg body weight, daily for 4 weeks) significantly attenuated the increased S1P level in liver tissues from FGF21KO-preHCC mice ([118]Fig. 5B). Previously, we reported that YAP had a negative impact on the survival of patients with HCC ([119]6). In the FGF21KO-preHCC mice, decrease of phosphorylated YAP ([120]Fig. 5C) and increased expression of YAP-targeting genes (Ctgf and Cyr61) (fig. S6B) were found, suggesting that lack of FGF21 could initiate the SPHK1/S1P-YAP signaling. To address this point, in vitro studies were performed. Challenged with PA, significantly increased S1P levels ([121]Fig. 5D) and phosphorylated SPHK1 ([122]Fig. 5E) were found in both FL83B-FGF21KD cells and Hepal-6-FGF21KD cells. Treatment with rhFGF21 attenuated the increased levels of S1P and phosphorylated SPHK1 ([123]Fig. 5E). To validate that phosphorylated YAP was induced by S1P, the Hepal-6 and two human HCC cell lines, Huh7 and Hep3B, were treated with S1P. Time/dose-dependent effects were found to be associated with YAP phosphorylation (fig. S6C), while S1P-induced YAP nuclear translocation in Hepal-6 cells was detected by fluorescent staining (fig. S6D). The SPHK1/S1P-YAP signaling transduction was further studied. Inhibition of SPHK1 by SKI-II attenuated the decreased levels of phosphorylated YAP in Hepal-6 cells with PA challenging ([124]Fig. 5F), while inhibition of the S1P receptor 2 (S1PR2) by JTE013 attenuated the decreased levels of phosphorylated YAP in the Hepal-6 cells with S1P challenging ([125]Fig. 5G). We reasoned that such a S1P-S1PR2-YAP signal would be potential to favor the MASH-HCC transition in FGF21KO mice. To test this hypothesis, the expression of S1P receptors was determined, and the significantly increased mRNA and protein levels of S1PR2 were found in the FGF21KO-DEN-WSHFD mice (fig. S6, E and F). This result encouraged us to further investigate S1P-S1PR2-YAP signaling in contribution to tumor growth in vitro and in vivo. Inhibition of S1PR2 by JTE013 or inhibition of YAP by verteporfin significantly prevented the colony growth in the Hepal-6 cells with S1P challenging ([126]Fig. 5H), while significantly suppressing tumor growth was consistent with the increased phosphorylation of YAP in the tumor tissues from in MASH-HCC mice (DEN-WSHFD model) with JTE013 treatment ([127]Fig. 5, I to J). Together, all these results suggested a negative feedback of FGF21 on the S1P-S1PR2-YAP signaling. The next question is whether S1P signaling could affect the macrophages to rend an immunocompromised microenvironment in liver. Fig. 5. S1P-S1PR2-YAP signaling mediated MSAH-HCC transition in FGF21KO mice. [128]Fig. 5. [129]Open in a new tab (A) Schematic diagram of S1P biosynthetic cascade enzymes with the fold changes (FGF21KO-preHCC versus WT-preHCC mice). ND, no detection; NC, no change; red, >2-fold up-regulated; blue, <2-fold down-regulated. (B) The S1P levels and protein levels of phosphorylated SPHK1 in liver issues from WT/FGF21KO-preHCC mice compared to that from WT/FGF21KO mice. (C) The protein levels of phosphorylated YAP and YAP in WT/FGF21KO-preHCC mice. (D) S1P content in FL83B/FL83B-FGF21KD cells and Hepal-6/Hepal-6-FGF21KD cells challenged with PA and treated with rhFGF21. (E) The protein levels of phosphorylated SPHK1 in FL83B/FL83B-FGF21KD cells and Hepal-6/Hepal-6-FGF21KD cells challenged with PA and treated with rhFGF21. (F) The protein levels of phosphorylated YAP in Hepal-6/Hepal-6-FGF21KD cells challenged with PA or PA + S1P and treated with SKI-II, an inhibitor of SPHK1. (G) The protein levels of phosphorylated YAP in Hepal-6/Hepal-6-FGF21KD cells challenged with S1P and treated with inhibitors of S1P receptors (JTE013 for S1PR2 and W146 for S1PR1). (H) Colony-forming assay to detect the number of cell colonies in Hepal-6 cells challenged with S1P and treated with JTE013 or verteporfin, an inhibitor of YAP. (I) Schematic diagram for JTE013 treatment in the MASH-HCC mice with WSHFD diets. (J) The protein levels of phosphorylated YAP in MASH-HCC mice with JTE013 treatment compared to vehicle controls. 21KO, FGF21KO. *P < 0.05, **P < 0.01, and ***P < 0.001. Tumor-associated macrophage polarization mediated NSAH-HCC transition in FGF21KO mice As an important component of TIME, KCs are believed to be essential to control the MASH insults. However, it is not fully elucidated how the TIME contributed to MSAH-HCC transition via impaired macrophages ([130]38). To provide insight into the transcriptome of macrophages/KCs during MASH-HCC transition, the DEN-WSHFD–induced MASH-HCC, WSHFD-induced MASH, and normal controls were selected to perform RNA-seq in the isolated KCs/MoMFs. The top 1000 highly variant genes are shown in heatmap (fig. S7A). Defined as false discovery rate < 0.05 and fold change ≥ 2, the volcano plots showed 1826 DEGs (MASH versus MASH-HCC) (fig. S7B). Standardized by normal controls, there are 1296 overlapping DEGs between 2773 DEGs (MASH versus controls) and 2468 DEGs (MASH-HCC versus controls). Among the 1296 overlapping DEGs, 680 up-regulated, 584 down-regulated, and 32 DEGs reversed ([131]Fig. 6A), suggesting that these shared genes could be critical for comprised function of macrophages. By pathway enrichment analysis (MASH versus MASH-HCC), peroxisome proliferator–activated receptor (PPAR) signaling pathway and inflammation/cytokine pathway were noticed in the top 15 pathways ([132]Fig. 6B). It is well known that all PPAR family members are sensitive to FAs, while PPARβ/δ is not only expressed in macrophages but also presented in liver ([133]39). Further analysis indicated that PPARβ/δ signaling was up-regulated in MASH-HCC compared to MASH ([134]Fig. 6C). IL-10 was found as one of the top five up-regulated cytokines in the inflammation/cytokine pathway ([135]Fig. 6D). Although the role of IL-10 in carcinogenesis is currently controversial, the macrophage-produced IL-10 has been reported to importantly contribute to TIME, in particular, macrophage polarization into tumor-associated macrophages (TAMs) ([136]40). In the tumor tissue from FGF21KO-MASH-HCC mice, M2 polarization was noticed by significantly increased F4/80^+CD206^+ cells by fluorescent staining ([137]Fig. 6E), while TAM phenotype was demonstrated by qPCR analysis ([138]Fig. 6F). As the mutual interactions between tumor cells and stromal microenvironment could contribute to phenotypically polarization of TAMs, BMDMs of mouse were therefore isolated to demonstrate whether cross-talk between macrophages and tumor cells could be mediated by PPARβ/δ and IL-10 signaling. Treated with the culture medium of Hepal-6 cells (HCM) challenged with S1P, the M0 BMDMs switched their expression profile, showing TAM phenotype (M2d), along with significantly increased protein expressions of PPARβ/δ and IL-10 ([139]Fig. 6G). To elucidate the mechanism of S1P in regulating the TAM polarization, we investigated the expressions of IL-4 and phospho–signal transducers and activators of transcription 6 (p-STAT6), which were reported as an important pathway to activate the PPAR-δ/IL-10 signaling for macrophage M2 polarization by FA ([140]41). Treatment with HCM-S1P induced p-STAT6 translocation to nucleus (fig. S7C), while the increased protein levels of IL-4 and p-STAT6 were significantly attenuated by inhibition of SPHK1 ([141]Fig. 6H), suggesting the potential mechanism of S1P mediated TAM polarization by PPAR-δ/IL-10 activation via IL-4/STAT6 pathway. To determine whether the oversupply of FA could contribute to the TIME in term of TAM polarization, the BMDMs were treated with HCM, HCM + PA, and HCM + S1P. Significantly increased lipid accumulation by Oil Red O staining and significantly increased S1P level by ELISA were detected in the BMDMs with HCM + PA treatment and HCM + S1P treatment (fig. S7, D and E). Both HCM + PA treatment and HCM + S1P treatment induced phenotypically polarization of TAMs in BMDMs, while use of sulfosuccinimidyl oleate (SSO), an inhibitor of CD36 to prevent FA uptake, attenuated the polarization of TAMs (fig. S7F). Together, up-regulation of PPAR-δ and IL-10 in KCs/MoMFs of MASH-HCC mice could be the potential signaling pathway of TAM polarization. S1P induced PPAR-δ/IL-10 activation via IL-4/STAT6 pathway to mediate TAM polarization. Oversupply of FA contributed, at least partly, to the increase of S1P production to render TIME for MSAH-HCC transition. Fig. 6. TAM polarization mediated NSAH-HCC transition in FGF21KO mice. [142]Fig. 6. [143]Open in a new tab (A) The volcano plots for the profile of DEGs in isolated macrophages/KCs from the liver tissues (MASH versus controls and MASH-HCC versus control) and the overlapping DEGs. (B) Fifteen enriched pathways were identified in MASH and MASH-HCC. (C and D) Up-regulated PPARδ in the lipid metabolism pathway and up-regulated IL10 in the inflammation/cytokine pathway were identified. (E) Immunofluorescent staining using the antibodies of anti-F4/80 and anti-CD206 to detect the M2 polarization of macrophages in FGF21KO-MASH-HCC mice, FGF21KO-MASH, and WT control mice. Red, F4/80-positive cells; green, CD206-positive cells; blue, positive DAPI staining to detect the nuclei as a counterstain. (F) qPCR detection for the markers of M2/TAM polarization in the isolated macrophages/KCs from FGF21KO-MASH-HCC mice, FGF21KO-MASH mice, and WT control mice. (G) Schematic diagram of mouse BMDMs treated with HCM, HCM + S1P, and Dulbecco’s modified Eagle’s medium (DMEM). qPCR detection for the markers of M2/TAM polarization in BMDMs induced by HCM and HCM + S1P. The protein levels of PPARβ/δ and IL-10 in BMDMs with treatments of HCM and HCM + S1P. (H) The mRNA fold changes of IL-4 by qPCR and the protein levels of IL-4 and phosphorylated STAT6 in BMDMs treated with HCM + S1P and HCM + S1P + SKI-II. BMDMs, bone marrow–derived macrophages. HCM, culture medium of Hepal-6 cells. *P < 0.05, **P < 0.01, and ***P < 0.001. DISCUSSION Liver is the major target of lipotoxicity. Hepatic lipotoxicity resulting from an excessive FFA influx has been accepted as a crucial cellular event clearly involved in the progression from MASH to HCC ([144]42, [145]43). As the largest population of tissue-resident macrophages, KCs have been reported to be essential for controlling the inflammatory process in MASH liver ([146]44); however, accumulation of “harmful lipids” has a long-lasting impact on KCs and, in turn, alters the liver immune microenvironment contributing to MASH-HCC transition. FGF21, as a liver safeguard ([147]45), has been widely reported to alleviate the hepatic fat stress ([148]46–[149]48), but the action of FGF21 on macrophage heterogeneity in particular for their distinct ontogeny (embryonic KCs versus BMDMs/MoMFs) is largely unknown. In this study, we report that lack of FGF21KO renders liver immunosuppressive microenvironment via compromised macrophages during MASH-HCC transition. The major signaling components mediated alteration of KC homeostasis and carcinogenetic transition are shown as a schematic diagram in [150]Fig. 7. Fig. 7. Schematic diagram of working hypothesis. [151]Fig. 7. [152]Open in a new tab The major signaling components mediated alteration of KC homeostasis and carcinogenetic transition polarization mediated NSAH-HCC transition being studied. HFMCD, high-fat methionine/choline-deficient diet; WSHFD, Western-style diet (high-fat and high-fructose); M1, macrophage M1 polarization; M2, macrophage M2 polarization; TAM, tumor-associated macrophage. Previously, TLR-4 signaling and the essential role of KCs in the development of MASH were reported ([153]49). KCs could promote hepatic steatosis via IL-1β–dependent suppression of PPAR-α activity ([154]50). Recruitment of KCs via CC-chemokine receptor 2 contributed to the progression of MASH by recruiting BMDMs ([155]51). KCs are known to self-renew locally to maintain themselves in the long term, while steatohepatitis may cause exhaustion of KC pool, resulting in mobilization of MoMFs for KC replacement ([156]12). However, the impact of FAs on KC during MASH-HCC transition remains undefined. Here, we asked first whether the lipid metabolites such as oxidized FAs in MASH liver could be the leading cause of KC death to affect the homeostasis of KC pool. We found that lack of FGF21 causes severe aberrant FA signaling, leading to not only liberation of FAs from adipose tissue but also increased FA de novo synthesis. Up-regulating Alox15 expression in KCs, other than hepatocytes, resulted in the increases of HODE and HETE to induce KC death. Increase of CD11b^+ Tim4^−CX3CR1^+ cells in FGF21KO-MASH liver indicated the replacement of KC pool by MoMFs. ALOX15 signaling affecting KC homeostasis for the MASH development was further supported by following data: (i) NAS was significantly decreased in ALOX15KO mice and (i) inhibition of ALox15 significantly attenuated the increased MoMFs in MASH liver. Supported by multiple lines of evidence, our findings indicated that aberrant FA metabolism via KC-ALOX15 signaling played a causal role for mobilization of MoMFs to replenish KC pool in FGF21KO-MASH liver. Of note, FGF21 may directly inhibit the ALOX15 activity to save KCs. Further study is needed to address this important issue. We next investigated the cross-talk between macrophages and tumor cells, in particular the S1P-mediated cross-talking before and during MASH-HCC transition. Although existing data indicate the carcinogenetic role of S1P/SPHK signaling in HCC, it is largely unknown how the S1P-mediated macrophage polarization contributes to the TIME for MASH-HCC transition. Here, we found that oversupplied FAs and the substrates for S1P cascade synthesis and activated SPHK1/S1P-YAP signaling in the preHCC-FGF21KO liver. RNA-seq analysis in the isolated KCs/MoMFs indicated that about 50% shared genes (MASH versus MASH-HCC), which could be critical for comprised function of macrophages. On the basis of pathway enrichment analysis, up-regulation of PPARβ/δ and IL-10 was noticed. PPAR-δ/IL-10 activation via IL-4/STAT6 pathway was identified as the key signals to mediate the cross-talking between tumor cells and macrophages. Up-regulation of PPAR-δ and IL-10 in macrophages/KCs by S1P induction could be potential mechanism of TAM polarization, and this was supported by the following: (i) M2d polarization was found in the BMDMs treated with HCM + S1P via activation PPAR-δ/IL-10 signaling, and (ii) blockage of FA supply attenuated S1P production thereby prevented M2d polarization of BMDMs. All these findings demonstrated that S1P production mediated not only the YAP signaling to initiate carcinogenetic cells but also TAM polarization, contributing to the TIME for MASH-HCC transition. In conclusion, FGF21 is essential against ALOX15-converted oxidized FAs and thereby preserve homeostasis of KC pool in liver. Lack of FGF21 accelerates MASH-HCC transition via the SPHK1/S1P-S1PR2-YAP signaling. Compromised macrophages presented as TAM phenotype contribute to TIME for MASH-HCC transition. MATERIALS AND METHODS Human studies To study FGF21 expression (by immunohistochemistry, qPCR, and Western blotting) and cell subpopulations in KC pool (by Hyperion Imaging), the human liver tissue samples were collected from 19 patients with MESH-HCC along with corresponding adjacent benign tissues. To study the serological levels of FGF21 (by ELISA), the human serum samples were collected from 20 patients with MESH-HCC tissue who had undergone HCC nodule resection and were followed up to 9 months. The liver tissue samples and serum samples were from the patients who were recruited between 2010 and 2020 from the James Graham Brown Cancer Center Bio-Repository at the University of Louisville. A microscope examination of the cellular composition of hepatic tissue confirmed the diagnoses of MASH-HCC and benign on these liver tissues, reviewed by two pathologists independently, blinded to the subject’s clinical history. All patients and their legally authorized representatives had given informed consent for the release of information and use of available tissue samples. The human sample collection procedures at the University of Louisville for this study were approved by the Institutional Review Board for Human Study of the University of Louisville (IRB no. 2.496). Gene Expression Profiling Interactive Analysis 2.0, an online tool, was used for survival analyses based on TCGA database. In the TCGA database, 156 overweight (BMI ≥ 25 kg/m^2) patients with HCC were graded as G1–G4 according to the National Comprehensive Cancer Network severity scale. The gene expression higher than median was defined as high expression group, while the gene expression lower than median was defined as low expression group. Using a log-rank test for hypothesis evaluation, survival rate of patients graded as G1–F2 and G3–G4 were analyzed on the basis of high/low gene expression levels of FGF21. To explore the correlation between survivals and six immune cells (B cells, CD4^+ T cells, CD8^+ T cells, neutrophils, macrophages, and dendritic cells), the split percentage of patients is set as 50% of the patients with high/low level of cell subpopulations to generate the Kaplan-Meier curve. CD68 was further normalized by FGF21 expression to estimate the survival probability over time. The Pearson’s correlation analysis shows that the expression levels of CD68 (x axis) and FGF21 (y axis) are negative correlation, as indicated by the correlation coefficient of −0.109 and P value of 3.52 × 10^−2. The result was analyzed by TIMER2.0 database. Animals and in vivo studies Male FGF21 KO (FGF21KO) mice with C57 BL/6J background were granted by S. Kliewer (University of Texas Southwestern Medical Center). C57BL/6J (strain no. 000664) WT mice and 12/15-LOX (Alox15) KO mice (strain no. 002778) were purchased from Jackson Laboratories (Bar Harbor, ME). All animals were housed four per cage, given rodent chow and tap water ad libitum, and maintained at 22°C and on a 12-hour light/dark cycle. The WT C57BL/6J mice were crossed to FGF21KO or Alox15KO mice to generate heterozygous F1 offspring. The F1 heterozygous mice were crossed to generate homozygous FGF21^−/− mice, homozygous Alox15^−/− mice, and WT littermates, which were used for establishment of animal models. Two MASH model were established by feeding (i) HFMCD (A06071302, l-amino acid diet with 60% kcal fat, 0.1% methionine, and no added choline, Research Diet, Inc., NJ) or (ii) WSHFD (D16030909, 60% kcal fat and 19% kcal fructose, Research Diets Inc., NJ). At 3 months, the mice were euthanized for advanced MASH models according to our previous report ([157]52). Three MASH-HCC models were established by administration of DEN in mice aged 2 weeks at 40 mg/kg body weight (i.p.) and fed with the following: (i) HFMCD, (ii) WSHFD, and (iii) high-fat diet (D12492, 60% kcal% fat, Research Diets Inc., NJ). The diet (D12450B, 10% kcal% fat, Research Diets, Inc. NJ) was used as CD. Ultrasound was performed to monitor the tumor nodules to determine HCC growth “windows” in all three MASH-HCC models. Treatments in animals were as follows: recombinant human FGF21 (rhFGF21) (no. 100-42; PeproTech) was injected subcutaneously every day at 125 μg/kg body weight. JTE013 (no. 1866, Axon Medchem) was injected intraperitoneally every day at 4 mg/kg body weight. PD146176 (no. NC1641198, Cayman Chemical) was injected subcutaneously every day at 5 mg/kg body weight. All animal procedures were approved by the Institutional Animal Care and Use Committee of the University of Louisville, which is certified by the American Association for Accreditation of Laboratory Animal Care (IACUC no. 19528). Cell lines and in vitro studies The cell lines for in vitro studies include a mouse hepatocyte line FL83B (ATCC CRL-2390) and a mouse hepatoma cell line Hepal-6 (ATCC CRL-1830), as well as two human HCC cell lines, Huh7 (GBTC-099H) and Hep3B (ATCC HB-8064). FL83B cells were cultured in the F12K medium (ATCC), Hepal-6, Huh7, and Hep3B cells in Dulbecco’s modified Eagle’s medium, with 10 to 15% fetal bovine serum. Establishment of Hepa1–6–FGF21KD cell and FL83B-FGF21KD cell and isolation of macrophages are described in the Supplementary Materials. Palmitate (PA) media (no. P9767, Sigma-Aldrich) was made by dissolving 2% bovine serum albumin (no. A1311, US Biologicals) in cell culture medium, and the 100 μM PA working solution was prepared from a high concentration (20 mM) stock PA solution made by dH[2]O heated to 70°C. The in vitro treatments were the following: rhFGF21 (no. 100-42; PeproTech) at 100 ng/ml, 13-HODE (no. 38600, Cayman Chemical) at 1 μM, NAC (no. A9165, Sigma-Aldrich) at 50 mM, S1P (no. 567727, Sigma-Aldrich) at 10 μM, SKI-II (no. S5696, Sigma-Aldrich) at 15 μM, W146 (no. 10009109, Cayman Chemical) at 10 μM, JTE013 (no. 1866, Axon Medchem) at 10 μM, and SSO at 50 μM to treat cells. All the antibodies and primers are listed in the Supplementary Materials. Statistical analysis Statistical analysis and graphics were performed by using GraphPad Prism 7.00 software (San Diego, California). Statistical significance was determined by analysis of variance (ANOVA). The post hoc Tukey’s test was used for analysis of any differences between groups. The collected data from experiments were presented as mean ±SD. For in vitro studies, experiments were performed with a minimum of triplicate samples and triplicate repetition of experiments. Group difference was considered significant for P < 0.05 (*), P < 0.01 (**), and P < 0.001 (***). Acknowledgments