Abstract Background & Aims The role of infiltrating neutrophils in hepatocellular carcinoma (HCC) is modulated by cellular metabolism, specifically lipid homeostasis. Throughout the progression of HCC, alterations in lipid metabolism are intricately linked with regulation of neutrophil function and the release of neutrophil extracellular traps (NETs). However, how much the protumor effect of a high-fat diet (HFD) depends on NETs and the potential interplay between NETs and other leukocytes in HCC remains uncertain. Methods In this study, the molecular mechanism of NET release and the potential beneficial effects of PPARα agonists on the HCC microenvironment were explored through proteomics, metabolomics, tissue microarray, immunofluorescence, flow cytometry, western blot, and dual-luciferase reporter gene assays (n = 6 per group). Results Our study demonstrated a notable inhibition of PPARα signaling in HCC. Furthermore, the disruption of PPARα-mediated lipid metabolism was responsible for the release of NETs. The presence of a HFD was observed to induce mitochondrial impairment in neutrophils, leading to the activation of cGAS-STING by oxidized mitochondrial DNA (Ox-mtDNA). Consequently, this activation triggered the release of NETs containing Ox-mtDNA through the enhancement of NLRP3-GSDMD-N in a NF-κB-dependent manner. Moreover, the release of NETs within HCC tissues effectively isolated cytotoxic leukocytes in the outer regions of HCC. Conclusions Our study not only provides insight into the relationship between lipid metabolism disorders and NETs’ tumor-promoting function, but also provides an important strategic reference for multi-target or combined immunotherapy of HCC. Impact and implications: We have identified PPARα and its agonists as therapeutic targets for controlling the neutrophil extracellular traps associated with high lipid metabolism. Results from preclinical models suggest that PPARα can limit mitochondrial oxidative stress, inhibit cGAS-STING-NF-κB signaling, and limit the release of neutrophil extracellular traps, thereby increasing the contact of anti-tumor leukocytes and hepatocellular cancer cells and limiting tumor growth. Keywords: hepatocellular carcinoma, neutrophil extracellular traps, PPARα, oxidative stress, cGAS-STING, NF-κB Graphical abstract [41]Image 1 [42]Open in a new tab Highlights: * • HFD induces the release of NETs through Ox-mtDNA. * • HFD activates the cGAS-STING-NF-κB axis by inducing mitochondrial ROS. * • PPARα inhibits the expression of GSDMD to prevent the release of NETs. * • PPARα promotes cytotoxic leukocyte contact with HCC cells by inhibiting release of NETs. Introduction Hepatocellular carcinoma (HCC) is influenced by various factors, with fatty liver disease resulting from high-fat intake being a significant contributing factor. Disruption of glucose metabolism in normal hepatocytes induced by hyperlipidemia resembles the metabolic pattern observed in HCC cells.[43]^1 Additionally, a high-fat diet (HFD) can induce chronic endoplasmic reticulum stress, exacerbating hepatic steatosis and potentially leading to the development of HCC.[44]^2 In recent years, the field of tumor immune microenvironment research has witnessed a surge, leading to the identification of numerous leukocytes as potential targets for regulating HCC progression. Unlike other malignant tumors, HCC mainly occurs in a chronic inflammatory environment.[45]^3 Neutrophils are numerous and heterogeneous in the HCC microenvironment. The timely recruitment of neutrophils is controversial in tumorigenesis, showing different functional characteristics after being converted into tumor-associated neutrophils. The anti-tumor N1 phenotype has the ability to kill tumor cells and stimulate T-cell immunity, while the N2 phenotype can promote tumor development by inhibiting T-cell responses and upregulating angiogenic factors.[46]^4 Therefore, a more precise understanding of the subtype characteristics and functions of tumor-associated neutrophils in HCC progression is urgent. Significant advances have been made in understanding the role of neutrophils in the progression of metabolic dysfunction-associated steatotic liver disease (MASLD)-associated HCC. The progression from steatosis to MASLD in HFD-fed mice is facilitated by the overexpression of CXCL1 in the liver, mediated by neutrophil-derived reactive oxygen species (ROS) and stress kinases.[47]^5 Additionally, the promotion of regulatory T cell (Treg) differentiation through metabolic reprogramming of naive CD4^+ T cells by NETs presents a potential therapeutic target for alleviating HCC progression in patients with MASLD.[48]^6 Previous research has demonstrated the presence of elevated levels of NETs in the serum of individuals afflicted with hepatitis and HCC.[49]^7 Additionally, a substantial influx of neutrophils and the release of NETs have been observed within the livers of obese mouse models with HCC.[50]^6 Nevertheless, the mechanisms by which a HFD environment triggers the release of NETs, as well as the impact of this process on the progression of HCC through interactions with leukocytes within the microenvironment, remain unexplored in the literature. It is worth mentioning that PPARα is closely related to MASLD.[51]^8^,[52]^9 PPARα is highly expressed in the liver and is widely involved in various biological activities such as energy metabolism, oxidative stress, and inflammation. In recent years, studies have shown that PPARα hinders fatty liver disease by regulating lipid metabolism, improving liver lipid accumulation, accelerating fatty acid oxidation, and inhibiting inflammatory responses.[53]^9 It also inhibited HCC by increasing antioxidant enzymes and reducing oxidative stress.[54]^8 Therefore, whether PPARα can be activated to limit the progression of MASLD-related HCC, reduce inflammatory responses, and enhance immune infiltration of the tumor microenvironment (TME) are key topics of this study. The primary constituent of NETs, known as NETs-DNA, has been associated with tumor growth, functioning as a chemotactic factor that attracts tumor cells to distant metastatic sites.[55]^10 Consequently, the targeted manipulation of NETs holds great potential as an effective therapeutic approach against tumors. However, the precise origin of DNA within NETs remains uncertain, with both mitochondrial DNA (mtDNA) and nuclear DNA being considered significant contributors to the reticular structure.[56]^11 The ribonucleoprotein immune complex induces the upregulation of ROS in low-density granulocytes, leading to oxidative stress-induced mitochondrial damage, mtDNA is then released to form NETs.[57]^12 Nevertheless, the precise molecular mechanism underlying mtDNA extrusion, the specific irritants in the TME that induce mitochondrial hypopolarization, and the potential inhibitory effects of drugs or small molecule compounds that enhance mitochondrial function on NET release remain elusive. This study employed an HFD-HCC mouse model to investigate the correlation between HFD and NETs, as well as the involvement of NETs in HCC. The findings of this study were as follows: 1) HFD led to oxidative stress of neutrophils’ mtDNA and contributed to the release of NETs, with PPARα playing a regulatory role in this process; 2) oxidized mtDNA activated the pro-inflammatory response mediated by cGAS-STING signaling, resulting in increased expression of NLRP3-GSDMD-N and facilitating the release of the network structure; 3) the presence of NETs impeded the infiltration of anti-tumor leukocytes into the TME. Our study presented novel evidence elucidating the development of HFD-associated NETs and the underlying mechanism by which they impeded immune infiltration. Furthermore, we proposed that the pharmacological regulation of PPARα could serve as a promising strategy for HCC immunotherapy. Materials and methods Reagents All antibodies, chemicals, small molecule inhibitors, drugs, commercial kits and primers used are displayed in [58]Tables S1-S4. Animals C57BL/6J (female, 6 weeks old, 18∼20 g) were purchased from Shanghai Model Organisms Center, Inc. Mice were housed four per cage in individually ventilated cage systems in specific pathogen-free grade animal room at the Animal Center of Fujian Medical University. Mouse were kept under a 12/12 h light/dark cycle. The animal experiments in this study were approved by the Experimental Animal Ethics Committee of Fujian Medical University (approval number IACUC FJMU2023-Y-0841) and were carried out according to the Institutional Animal Care and Use Committee guidelines. Statistical analysis All data analysis was performed with GraphPad Prism V.8 or SPSS V.19, and data are presented as mean ± standard deviation. When comparing two samples, the independent sample t test was used if the data were normally distributed and the variances were homogeneous. If the data were normally distributed with unequal variance, Wilcoxon rank sum test was used. European-style clustering was employed to examine and evaluate the resemblances among various samples. Flow cytometry data were analyzed using FlowJo software V10. p <0.05 was used as the criterion for statistical significance. Other detailed methods are provided in the supplementary information. Results Impaired PPARα signaling was correlated with dysregulation of lipid metabolism in HCC To explore the relative contribution of the etiology or complications of HCC in the release of NETs, ELISA was employed to assess the correlation between the expression indicator of NETs, specifically citrullinated histone H3 (CitoH3), and various clinical indicators (such as HBV infection, cirrhosis, liver cyst, focal nodular hyperplasia, and MASLD). Notably, our finding indicated that only two factors (HBV infection and MASLD) were responsible for the aberrant expression of CitoH3 in patients ([59]Fig. 1A). Specifically, HBV infection hindered the expression of CitoH3, whereas MASLD enhanced its expression ([60]Fig. 1A). Previous research has documented the inhibition of NETs by HBV infection.[61]^13 Consequently, we aimed to investigate the association between MASLD and NETs. To elucidate the link between aberrant lipid metabolism and NETs release, we conducted a comparative analysis of the proteome and the metabolome of HCC. The proteomic data revealed a significant correlation between impaired PPAR signaling and the development of mouse HCC ([62]Fig. 1B). Additionally, metabolomics indicated disruptions in various lipid metabolic signals, particularly triglycerides, within patient cancerous tissues ([63]Fig. 1C). Furthermore, we found that the inhibition of PPAR signaling contributed to the dysregulation of lipid metabolism, and PPAR signaling was the second most influential pathway ([64]Fig. 1D and [65]Table S6). Moreover, our study indicated that, alongside the identification of nine differentially expressed proteins through proteomics analysis ([66]Figs 1E and [67]S1-9), the expression of an additional set of 16 downstream proteins of PPAR signaling, including PPARα, was observed to be suppressed in patient cancerous tissues ([68]Fig. 1F and [69]Fig. S10-25). However, the expression of another 21 PPAR signaling pathway-related proteins, including ME1, did not exhibit significant changes in patient HCC tissues ([70]Fig. S26). Fig. 1. [71]Fig. 1 [72]Open in a new tab Impaired PPARα signaling was correlated with dysregulation of lipid metabolism in HCC. (A) ELISA showing the concentration of CitoH3 within patient HCC (n = 79). (B) KEGG pathway enrichment analysis examining the differentially expressed proteins between HCC and para-cancer tissues in the mouse models (n = 3). PPAR signaling pathway was marked in red. (C) A network map illustrating the differential lipid metabolites between HCC and para-cancer tissues in patients (n = 20). The KEGG pathway was represented by blue circles, while the red circles indicated the metabolites enriched in the corresponding pathway. (D) KEGG pathway enrichment analysis investigating the differential metabolites between HCC and para-cancer tissues in patients (n = 20). (E) The proteomic identification of mice was confirmed through patient tissue microarray, revealing differential expression levels of PPAR signaling pathway-associated proteins between HCC and para-cancerous tissues in patients (n = 20). (F) The expression of the other 16 PPAR signaling pathway-related proteins in HCC and para-cancerous tissues was examined using tissue microarray (n = 20). (G) KEGG enrichment analysis determining the distribution of differentially expressed PPAR signaling pathway-related proteins in the three subtypes of PPAR and their downstream targets, with repressed genes indicated by the color blue. Mean ± SD. Statistical significance evaluated by Student’s t test (A, E-F). ∗p <0.05, ∗∗p <0.01. CitoH3, citrullinated histone H3; FNH, focal nodular hyperplasia; HCC, hepatocellular carcinoma; KEGG, Kyoto Encyclopedia of Genes and Genomes; MASLD, metabolic dysfunction-associated steatotic liver disease; NETs, neutrophilic extracellular traps. There exist three distinct subtypes of PPAR, namely PPARα, PPARβ/δ, and PPARγ, each exhibiting distinct functional roles.[73]^8^,[74]^9 Based on the patient metabolome data ([75]Fig. 1C), it was evident that HCC was characterized by notable alterations in fatty acid synthesis, extension, absorption, degradation, and disruption of unsaturated fatty acid synthesis. Consequently, we proposed that compromised PPARα signaling played a crucial role in HCC. We found that proteins involved in PPARα signaling were inhibited in HCC ([76]Fig. 1G). Therefore, low PPARα expression associated with high lipid metabolism might be the key to inhibiting release of NETs. PPARα inhibited release of NETs in HCC The absence of PPARα hinders the release of pro-inflammatory cytokines, thereby impairing the functionality of Tregs and macrophages.[77]^14 However, the extent to which PPARα is implicated in the functionality of neutrophils remains inadequately understood. Consequently, we examined the correlation between PPARα and the release of NETs in HCC. Patient HCC were categorized into high and low PPARα signaling groups, predicated on the expression levels of 25 PPARα-associated genes ([78]Fig. 2A). Our study indicated that the release of NETs was inhibited in the high PPARα signaling group ([79]Fig. 2B). Moreover, the low PPARα signaling group demonstrated a significantly elevated occurrence of lymph node metastasis, distant metastasis, and vascular invasion ([80]Fig. 2C,D and [81]Fig. S27). Fig. 2. [82]Fig. 2 [83]Open in a new tab PPARα inhibited NET release in HCC. (A) HCC tissues obtained from a cohort of 12 patients were stratified into two distinct groups, namely high and low PPARα signaling, using the expression levels of 25 PPARα-associated genes (n = 6). (B) Immunofluorescence investigating the presence of NETs in HCC tissues of patients (n = 3). Blue, DAPI; green, CitoH3; red, MPO. The co-localization of these three markers indicated NETs. The NETs were marked using a white box. (C) Baseline table from TCGA database were utilized to conduct a comparative analysis of vascular invasion in HCC (n = 205). Red indicates statistical significance. (D) Baseline table was employed to compare TNM staging and vascular invasion in patients with HCC from our center (n = 24). Red indicates statistical significance. (E) Immunofluorescence revealing the presence of NETs in tumor tissues of primary HCC mice (n = 6). (F) Immunofluorescence visualizing the presence of NETs in HCC tissues from patients (n = 6). (G) Immunofluorescence showing the presence of NETs in tumor tissues of primary HCC mice (n = 6). (H) Flow cytometry demonstrating the expression of NET-related markers in primary HCC-infiltrating neutrophils from mice (n = 6). (I) Flow cytometry showing the expression of NET-related markers in HCC-infiltrating neutrophils from patients (n = 6). Mean ± SD. European-style clustering was employed to examine and evaluate the resemblances among various samples (A). Statistical significance evaluated by Student’s t test (B, E-I). ∗p <0.05, ∗∗p <0.01. CitoH3, citrullinated histone H3; HCC, hepatocellular carcinoma; HFD, high-fat diet; KEGG, Kyoto Encyclopedia of Genes and Genomes; MASLD, metabolic dysfunction-associated steatotic liver disease; NETs, neutrophilic extracellular traps; TCGA, the Cancer Genome Atlas; TNM, tumor-node-metastasis. We then explored the relationship between hyper-lipid metabolism and NETs. We performed cluster analysis based on the expression of 25 PPARα-associated genes in primary HCC tissues of patients and mice, and divided them into high and low PPARα signaling groups ([84]Figs S28-29). A higher abundance of NETs was observed in HCC tissues from mice fed a HFD or patients with MASLD compared to those from mice on a normal diet or patients without MASLD ([85]Fig. 2E,F). Conversely, in mice fed a HFD, the release of NETs was suppressed in the group with high PPARα signaling compared to low PPARα signaling ([86]Fig. 2E,F). In addition, bezafibrate, an agonist of PPARα, or norathyriol, an inhibitor, were used to determine the effect of PPARα on the release of NETs, and it was found that bezafibrate inhibited the release of NETs, while norathyriol did the opposite ([87]Fig. 2G). Futhermore, we examined the expression of four markers of NETs,[88]^10 including PAD4, mitochondrial ROS, CitoH3, and 8-OHdG in neutrophils infiltrating HCC from patients and mice ([89]Fig. S30). We found that HFD or MASLD combined with low PPARα signaling resulted in increased expression of the four markers ([90]Fig. 2H,I). In comparison to the group consuming the HFD/MASLD and low PPARα signaling, the HFD/MASLD and high PPARα signaling group exhibited suppressed expression of these markers ([91]Fig. 2H,I). Hence, activating PPARα was a feasible strategy to inhibit NETs related to high lipid metabolism. PPARα mitigated HFD-associated NETs through the attenuation of mitochondrial oxidative stress Considering PPARα inhibited the production of mitochondria ROS induced by a HFD ([92]Fig. 2H,I), we further investigated whether PPARα hinder the release of NETs by suppressing mitochondrial DNA (mtDNA) oxidation. We found that overexpression of Ppara downregulated mitochondrial ROS levels in neutrophils infiltrating mouse HCC, while knockout upregulated them ([93]Fig. 3A). Our findings demonstrated that neutrophils generated higher levels of mitochondrial ROS when exposed to a HFD, while the opposite effect was observed in the low-fat diet (LFD) group ([94]Fig. 3B). The reduction of HFD-induced mitochondrial ROS was observed when treated with two PPARα agonists (bezafibrate or fenofibrate) ([95]Fig. 3B). Moreover, we found that overexpression of Ppara increased mitochondrial membrane potential, while knockout decreased it ([96]Fig. 3C). HFD resulted in lower mitochondrial membrane potential, whereas low lipidemia led to higher levels ([97]Fig. 3D). The reduction in mitochondrial membrane potential induced by HFD was suppressed by PPARα agonists, and this effect was similarly observed in the groups fed normal-fat diet and LFD ([98]Fig. 3D). Given that the loss of mitochondrial membrane potential is associated with PKC-mediated mitochondrial ROS,[99]^15 we conducted an experiment where we added the PKC inhibitor chelerythrine-chloride to neutrophils. Our results showed a reduction in HFD-mediated mitochondrial polarization, while chelerythrine-chloride did the opposite ([100]Fig. S31A). In addition, we introduced rotenone, an inductor of mitochondrial ROS,[101]^16 to neutrophils infiltrating HFD-HCC that were treated with PPARα agonists. Our findings indicated that PPARα agonists resulted in an increase in the mitochondrial polarization that was inhibited by rotenone ([102]Fig. S31B). Fig. 3. [103]Fig. 3 [104]Open in a new tab PPARα mitigated HFD-associated NETs through the attenuation of mitochondrial oxidative stress. (A,B) MitoSOX™ Green assessing the levels of mitochondrial ROS in mouse neutrophils (n = 3). Effects of overexpression or knockout of Ppara in vitro (A). Role of HFD, LFD or the PPARa agonists (bezafibrate or fenofibrate) in tumor tissues of primary HCC mice (B). (C,D) JC-1 assessing the extent of mitochondrial membrane potential in mouse neutrophils (n = 3). Effects of overexpression or knockout of Ppara in vitro (C). Role of HFD, LFD or the PPARa agonists (bezafibrate or fenofibrate) in tumor tissues of primary HCC mice (D). (E) Immunofluorescence visualizing the presence of NETs in tumor tissues of primary HCC mice (n = 6). Blue, DAPI; green, CitoH3; red, MPO. The co-localization of these three markers indicated NETs. The NETs were marked using a white box. (F) Immunofluorescence showing the presence of NETs in tumor tissues of primary HCC mice (n = 6). Mean ± SD. Statistical significance evaluated by Student’s t test (A-F). ∗p <0.05, ∗∗p <0.01. B, bezafibrate; CitoH3, citrullinated histone H3; F, fenofibrate; HCC, hepatocellular carcinoma; HFD, high-fat diet; LFD, low-fat diet; NETs, neutrophilic extracellular traps; R, rotenone; ROS, reactive oxygen species. We further explored the potential of mitochondrial ROS in the release of NETs. Rotenone and feprazone, an inhibitor of mitochondrial ROS, were used to determine the effect of mitochondrial ROS on the release of NETs, and it was found that rotenone induced the release of NETs, while feprazone did the opposite ([105]Fig. 3E). Furthermore, our study revealed that HFD augmented the release of NETs in HCC ([106]Fig. 3F). Notably, this effect was counteracted by PPARα agonists ([107]Fig. 3F). To determine whether low levels of mitochondrial oxidative stress were necessary for PPARα agonists to inhibit the release of NETs, we promoted ROS production with rotenone and found that rotenone further exacerbated the release of NETs, while PPARα agonists mitigated its effects ([108]Fig. 3F). Therefore, we concluded that PPARα inhibited the release of NETs by suppressing the level of mitochondrial ROS in neutrophils and increasing mitochondrial membrane potential. PPARα mitigated HFD-associated NETs by impairing the release of mtDNA We observed that overexpression of Ppara inhibited the expression of 8-OHdG and TOM20 on the cell membrane in neutrophils infiltrating mouse HCC, while knockout induced their expression ([109]Fig. 4A). Furthermore, our findings indicated that HFD promoted mitochondrial mobilization to the cell membrane, whereas LFD had the opposite effect ([110]Fig. 4B). However, PPARα agonists mitigated HFD-induced mitochondrial mobilization, as observed in both conventionally fed and LFD groups ([111]Fig. 4B). To mitigate the potential bias associated with two-dimensional fluorescence, we investigated the aggregation of mitochondria on the membrane of nonpermeable cells. Ppara knockout was found to exhibit strong inducement of mitochondrial outer membrane proteins TOM20 and HSP60, and vice versa ([112]Fig. 4C). Within the HFD group, we observed the translocation of TOM20 and HSP60 to the surface of nonpermeable cells, which was diminished by PPARα agonists ([113]Fig. 4D,E and [114]Fig. S32). To eliminate the potential occurrence of neutrophils containing mitochondria binding to the cell surface, which enhance the staining of TOM20 on the membrane, we subjected neutrophils to treatment with DNaseI. The level of TOM20 on the cell surface was unaffected by DNaseI, indicating that it was unlikely to be a consequence of neutrophils being deposited on the cell surface as an external trapping mechanism ([115]Fig. S33). To examine the detrimental effects of mitochondrial ROS, we employed 8-OHdG to assess whether PPARα agonists mitigate DNA oxidation induced by HFD. The HCC-infiltrating neutrophils with high lipid association exhibited significant 8-OHdG staining, while the neutrophils associated with LFD showed low levels ([116]Fig. 4B). PPARα agonists not only decreased 8-OHdG expression in HFD-stimulated cells, but also reduced 8-OHdG levels to negligible amounts in the hypolipidemia group ([117]Fig. 4B). Co-localization of TOM20 with 8-OHdG suggested that the majority of oxidation occurs on mtDNA rather than nuclear DNA ([118]Fig. 4B). To establish the origin of the released oxidized DNA as being from mitochondria, we isolated mitochondria from neutrophils and assessed their expression of 8-OHdG. As anticipated, PPARα agonists resulted in a reduction of 8-OHdG expression ([119]Fig. 4F). Moreover, this inhibitory effect was reversed by rotenone ([120]Fig. 4G). To ascertain the impact of PPARα-mediated lipid metabolism on nuclear DNA damage, nuclear protein was extracted and analyzed. In the HFD group, it was observed that high lipid metabolism induced the expression of 8-OHdG, which was inhibited by PPARα agonists ([121]Fig. S34A). Conversely, in the LFD group, although the expression of 8-OHdG was suppressed, PPARα agonists did not exhibit any protective effects on nuclear DNA ([122]Fig. S34A). These findings suggested that PPARα exclusively influenced nuclear DNA damage caused by HFD intake, without participating in the regulation of nuclear DNA effects associated with LFD consumption. It was noteworthy that the protective impact of PPARα agonists on nuclear DNA in the HFD group was impeded by the ROS agonist GSK5182 ([123]Fig. S34B). Hence, it appeared that HFD induced mitochondrial membrane mobilization, and PPARα inhibited this process to regulate neutrophil function. Fig. 4. [124]Fig. 4 [125]Open in a new tab PPARα mitigated HFD-associated NETs by impairing the release of mtDNA. (A,B) Immunofluorescence illustrating the expression and localization of 8-OHdG (green) and TOM20 (red) of neutrophils infiltrated by HCC in mice (n = 3). Effects of overexpression or knockout of Ppara in vitro (A). Role of HFD, LFD or the PPARa agonists (Bezafibrate or Fenofibrate) in tumor tissues of primary HCC mice (B). (C-E) Flow cytometry analyzing the expression of TOM20 and HSP60 in nonpermeable neutrophils infiltrated by mouse HCC (n = 3). Effects of overexpression or knockout of Ppara in vitro (C). Role of HFD, LFD or the PPARa agonists (bezafibrate or fenofibrate) in tumor tissues of primary HCC mice (D-E). (F,G) Western blot demonstrating mitochondrial 8-OHdG expression in infiltrating neutrophils of mouse primary HCC (n = 3). Effects of PPARα agonists (F). Role of PPARα agonists and rotenone (G). Mean ± SD. Statistical significance evaluated by Student’s t test (A-G). ∗p <0.05, ∗∗p <0.01. B, bezafibrate; F, fenofibrate; HCC, hepatocellular carcinoma; HFD, high-fat diet; LFD, low-fat diet; mtDNA, mitochondrial DNA; NETs, neutrophilic extracellular traps; R, rotenone. PPARα inhibited cGAS-STING signaling and induced disruption of NETs Considering cGAS-STING is a receptor for DNA,[126]^17 we investigated the impact of PPARα and lipid metabolism on cGAS-STING signaling. Our findings revealed the level of p-STING/STING decreased after Ppara overexpression in neutrophils infiltrating mouse HCC, accompanied by a robust decline of downstream inflammatory cytokines p-IRF3/IRF3, IFN-α, IFN-β, IL-6, TNF-α, and ISG15 ([127]Fig. 5A). Moreover, a HFD led to a significant rise in cGAS-STING-dependent type I IFN signaling, while the opposite was observed for LFD ([128]Fig. 5A). PPARα agonists exhibited notable antagonistic effects on cGAS-STING signaling, resulting in the suppression of the cGAS-STING signal to an imperceptible level in the LFD group ([129]Fig. 5A). We found that the blocking effect of Ppara overexpression on cGAS-STING signaling was mitigated by rotenone ([130]Fig. 5B). As expected, rotenone upregulated HFD-induced pro-inflammatory signaling, and this positive effect was damaged by PPARα agonists ([131]Fig. 5B). In addition, CL656, an agonist of STING, and omaveloxolone, an inhibitor, were used to determine the effect of STING on the release of NETs, and it was found that CL656 induced the release of NETs, while omaveloxolone did the opposite ([132]Fig. 5C). Furthermore, our findings indicated that elevated levels of STING promoted the release of NETs in HCC, and this positive effect was disrupted by PPARα agonists ([133]Fig. 5D). In short, PPARα prevented the formation of NETs by inhibiting cGAS-STING signaling induced by mitochondrial oxidized DNA. Fig. 5. [134]Fig. 5 [135]Open in a new tab PPARα inhibited cGAS-STING signaling and induced disruption of NETs. (A,B) Western blot showing the expression of proteins related to cGAS-STING signaling in infiltrating neutrophils of mouse primary HCC (n = 3). Effect of overexpression, knockout of Ppara or PPARα agonists (A). Role of overexpression of Ppara, PPARα agonists and rotenone (B). (C) Immunofluorescence visualizing the presence of NETs in tumor tissues of mouse primary HCC mice (n = 6). Blue, DAPI; green, CitoH3; red, MPO. The co-localization of these three markers indicated NETs. The NETs were marked using a white box. (D) Immunofluorescence demonstrating the presence of NETs in tumor tissues of mouse primary HCC mice (n = 6). Mean ± SD. Statistical significance evaluated by Student’s t test (A-D). ∗p <0.05, ∗∗p <0.01. B, bezafibrate; C, CL656; CitoH3, citrullinated histone H3; F, fenofibrate; HCC, hepatocellular carcinoma; HFD, high-fat diet; LFD, low-fat diet; NETs, neutrophilic extracellular traps; R, rotenone; ROS, reactive oxygen species. PPARα inhibited NF-κB signaling depending on cGAS-STING signaling Given that NF-κB works in conjunction with IRF3 to facilitate the production of type I IFN in response to stimulation by double-stranded DNA.[136]^18 We hypothesized that the inhibition of NF-κ signaling by PPARα was reliant on Ox-mtDNA-cGAS-STING signaling. We found that overexpression of Ppara inhibited phosphorylation of IKKα/β and p65 in neutrophils infiltrating mouse HCC, while knockdown promoted it ([137]Fig. 6A). High levels of IKKα/β and p65 phosphorylation were observed in the HFD group, whereas a significant inhibition of their phosphorylation was observed in the LFD group ([138]Fig. 6A). Additionally, PPARα agonists demonstrated antagonism of the NF-κ pathway ([139]Fig. 6A). To ascertain the potential pro-inflammatory function of Ox-mtDNA, we subjected HCC-infiltrating neutrophils obtained from HFD mice treated with PPARα agonists to rotenone. Our findings revealed a significant upregulation of phosphorylation levels in IKKα/β and p65 upon rotenone administration, which was impaired by overexpression of Ppara or PPARα agonists ([140]Fig. 6B). To ascertain the dependence of PPARα inhibition of the NF-κ pathway on cGAS-STING signaling, we administered CL656 to neutrophils and observed that elevated STING signaling augmented NF-κ signaling, whereas overexpression of Ppara or PPARα agonists impeded this favorable impact ([141]Fig. 6C). Given that both Ox-mtDNA and cGAS-STING signaling contribute to the robust NF-κ signaling induced by heightened lipid metabolism, we further investigated whether NF-κ signaling serves as a crucial link in the downregulation of NET release by PPARα. To this end, NF-κ activator 1, an agonist of NF-κ, and JSH-23, an inhibitor, were utilized to determine the effect of NF-κ on the release of NETs, and it was found that NF-κ activator 1 induced the release of NETs, while JSH-23 did the opposite ([142]Fig. 6D). Moreover, we found that elevated levels of NF-κB enhanced the release of NETs in HCC, while PPARα agonists disrupted this favorable outcome ([143]Fig. 6E). Therefore, PPARα downregulated the release of NETs through the NF-kB pathway induced by cGAS-STING signaling. Fig. 6. [144]Fig. 6 [145]Open in a new tab PPARα inhibited NF-κB signaling depending on cGAS-STING signaling. (A-C) Western blot showing the expression of proteins related to the NF-κB signaling in infiltrating neutrophils of mouse primary HCC (n = 3). Effect of overexpression, knockout of Ppara or PPARα agonists (A). Role of overexpression of Ppara, PPARα agonists and rotenone (B). Influence of overexpression of Ppara or PPARα agonists and CL656 (C). (D) Immunofluorescence visualizing the presence of NETs in tumor tissues of mouse primary HCC (n = 6). Blue, DAPI; green, CitoH3; red, MPO. The co-localization of these three markers indicated NETs. The NETs were marked using a white box. (E) Immunofluorescence showing the presence of NETs in tumor tissues of mouse primary HCC mice (n = 6). Mean ± SD. Statistical significance evaluated by Student’s t test (A-E). ∗p <0.05, ∗∗p <0.01. B, bezafibrate; C, CL656; CitoH3, citrullinated histone H3; F, fenofibrate; HCC, hepatocellular carcinoma; HFD, high-fat diet; LFD, low-fat diet; NA, NF-κ activator 1; NETs, neutrophil extracellular traps; R, rotenone. PPARα suppressed NLRP3-GSDMD-N depending on the Ox-mtDNA-cGAS-STING-NF-κ-NETs axis It has been reported that GSDMD is cleaved to form an active N-terminal fragment with a size of about 32 kD, which binds to the membrane and perforates.[146]^19 Our findings indicated that the expression of NLRP3 and GDSMD-N induced by HFD was suppressed by PPARα agonists in neutrophils infiltrating mouse HCC, but this inhibition was hindered by rotenone, CL656, and NF-κ activator 1 ([147]Fig. S35A). The JASPAR database predicted that p65 adhered to the promoter of NLRP3 ([148]Fig. S35B,C), and our study confirmed that p65 bound to the -187 ∼ -178 “gggaaccccc” of the mouse NLRP3 promoter ([149]Fig. S35D). Furthermore, HFD induced p65 binding to the NLRP3 promoter, while PPARα agonists inhibited this interaction ([150]Fig. S35E). PPARα impeded HCC depending on Ox-mtDNA-cGAS-STING-NF-κ-NETs We next investigated whether PPARα impeded HCC growth by inhibiting the release of NETs in vivo. Our study revealed that HFD resulted in the growth of tumors and a higher hemorrhagic ascites rate ([151]Fig. 7). Elimination of NETs through Dnase I impeded this cancer-promoting effect ([152]Fig. 7A-F). Furthermore, PPARα agonists were found to hinder the growth of HCC and hemorrhagic ascites induced by HFD ([153]Fig. 7A-F). However, their effectiveness was compromised by PMA, which triggers the release of NETs, as well as by rotenone, CL656, and NF-κ activator 1 ([154]Fig. 7A-F). Fig. 7. [155]Fig. 7 [156]Open in a new tab PPARα impeded HCC depending on Ox-mtDNA-cGAS-STING-NF-κ-NETs. (A) A representative diagram illustrating the growth of HCC in the abdominal cavity in a mouse model of peritoneal growth (n = 6). (B) The tumor volume for (A) (n = 6). (C) The tumor weight for (A) (n = 6). (D) The hemorrhagic ascites for (A) (n = 6). (E) A representative diagram showing HCC growth in situ in mice (n = 6). (F) The tumor volume for (E) (n = 6). Mean ± SD. Statistical significance evaluated by Student’s t test (B–C, F). ∗p <0.05, ∗∗p <0.01. B, bezafibrate; C, CL845; F, fenofibrate; HCC, hepatocellular carcinoma; HFD, high-fat diet; NA, NF-κ activator 1; NETs, neutrophil extracellular traps; R, rotenone. NETs inhibited contact between HCC cells and cytotoxic cells To investigate the tumor immunomodulatory effect of NETs, we investigated the spatial localization of HCC cells, NETs, and cytotoxic cells (CD4^+ T cells, CD8^+ T cells and NK cells) within mouse HCC tissues. Our findings revealed that NETs increased physical separation between the cytotoxic cells and HCC cells ([157]Fig. 8A-D). We observed that HFD promoted the release of NETs, isolating cytotoxic cells away from HCC cells ([158]Fig. 8A-D). Conversely, PPARα agonists diminished the spatial separation between cytotoxic cells and HCC cells by inhibiting the release of NETs ([159]Fig. 8A-D). Hence, NETs converted the TME from “hot” to “cold” by capturing T cells and NK cells and isolating them from tumor cells. Fig. 8. [160]Fig. 8 [161]Open in a new tab NETs inhibited contact between HCC cells and cytotoxic cells. (A-C) Immunofluorescence showing the quantitative and spatial arrangement of cytotoxic cells, including CD4^+ T cells (A), CD8^+ T cells (B), and NK cells (C), along with HCC cells and NETs in tumor tissues of mouse primary HCC mice. Blue, DAPI; green, CK18; red, CitoH3; yellow, CD8/CD4/NK1.1. The arrows indicated that NETs capture cytotoxic cells (n = 6). (D) Quantization graph for A-C (n = 6). (E) The mechanism diagram. Mean ± SD. Statistical significance evaluated by Student’s t test (D). ∗p <0.05, ∗∗p <0.01. CitoH3, citrullinated histone H3; HCC, hepatocellular carcinoma; NETs, neutrophil extracellular traps. Discussion Prior research has demonstrated the involvement of mitochondrial ROS-mediated cytotoxicity in liver injury induced by HFD.[162]^20 Through an examination of mitochondria ROS during the release of NETs, we have discerned three novel functions of mitochondria in the context of HFD. Firstly, the inflammatory nature of a HFD led to mitochondrial impairment, while mitochondrial ROS sustained the release of NETs. Secondly, Ox-mtDNA, possessing potent pro-inflammatory and interferon attributes, activated the DNA receptor cGAS-STING and triggered NLRP3-GSDMD-N, thereby augmenting membrane permeability to facilitate the release of mtDNA in an NF-κB-dependent fashion. Lastly, the inhibition of PPARα signaling in HCC tissues was pronounced, and PPARα agonists demonstrated a significant ability to suppress the release of oxidative stress-induced NETs. The identification of NETs along with the systemic inhibition of the Ox-mtDNA-cGAS-STING-NF-κB-NLRP3-GSDMD-N axis, provided robust evidence to support our findings that the progression of HCC was exacerbated through NET-mediated mechanisms. The correlation between the abundance of neutrophils and the extent of steatosis, as well as their association with the development of fatty hepatocytes, has been established.[163]^21 The elevated neutrophil-to-lymphocyte ratio observed in the peripheral blood of individuals is regarded as a non-invasive indicator of both non-alcoholic steatohepatitis and the severity of hepatic fibrosis.[164]^22 Inhibiting neutrophil infiltration has been shown to effectively protect mice from HFD-induced steatohepatitis.[165]^23 In this study, it was observed that HFD contributed to the generation of mitochondria ROS that induced damage to mtDNA in neutrophils. The presence of depolarized mitochondria plays a crucial role in facilitating the release of NETs. Significantly, PPARα agonists demonstrated effective mitigation of externalized Ox-mtDNA, suppression of inflammatory cytokine production, and prevention of membrane damage in neutrophils through a mechanism reliant on the cGAS-STING-NF-κB-NLRP3-GSDMD-N axis. Furthermore, these agonists alleviated the extrusion of mtDNA, a crucial constituent of NETs, into surrounding cells. The observed increase in neutrophil granuloprotein,[166]^24 pro-inflammatory cytokines, and type I IFN[167]^25 within the microenvironment of HCC closely resembled the induction outcomes observed in NETs. Furthermore, it is worth noting that additional mechanisms contributing to chronic inflammation in HCC synergistically interacted with the dysregulation of neutrophils, thereby intensifying the inflammatory response.[168]^26 Consequently, mitigating the innate immune response triggered by NETs emerges as a crucial approach to attenuate the inflammatory milieu within HCC, diminish the inflammatory harm inflicted upon HCC cells, and counteract the malignant conversion of hepatocytes as well as the development of metastases. The findings of our study indicated that PPARα agonists could decrease the type I IFN response and hinder the activation of NF-κB signaling, which was responsible for promoting pro-inflammatory responses in HCC associated with MASLD. These results provided evidence that the antagonistic impact of PPARα on cGAS-STING signaling serves as a mechanism to mitigate the release of NETs, which were mediated by disorders in lipid metabolism. Additionally, these findings shed light on the interplay between PPARα and innate immunity. Recent evidence indicated that the pathogenesis of various malignancies, including HCC, is influenced by abnormal release of NETs and/or impaired clearance.[169]^10^,[170]^11^,[171]^27 This assertion was substantiated by mouse studies, which demonstrated that inhibiting the release of NETs enhances tumor invasion,[172]^7 macrophage polarization,[173]^28 and infiltration of Tregs.[174]^6 Nevertheless, the impact of NETs on the functionality of other leukocytes, their involvement in tumor-leukocyte interactions, and their potential hindrance of cytotoxic leukocyte infiltration remain inadequately understood. This study demonstrated that NETs possessed the ability to impede intracellular apoptosis by establishing a physical blockade between malignant cells and leukocytes via their distinctive web-like architecture, thereby capturing and enclosing infiltrating T cells and NK cells. In conclusion, the deficiency of PPARα in HCC associated with HFD triggered the activation of the cGAS-STING-NF-κB-NLRP3-GSDMD-N axis, resulting in heightened NET release. The pivotal function of NETs lies in impeding the localization and elimination of HCC cells by cytotoxic leukocytes. Hence, PPARα agonists to obstruct this axis represent a promising therapeutic approach for MASLD-related HCC ([175]Fig. 8E). Abbreviations CitoH3, citrullinated histone H3; HCC, hepatocellular carcinoma; HFD, high-fat diet; LFD, low-fat diet; mtDNA, mitochondrial DNA; MASLD, metabolic dysfunction-associated steatotic liver disease; NETs, neutrophil extracellular traps; ORF, open reading frame; Ox-mtDNA, oxidized mitochondrial DNA; ROS, reactive oxygen species; TMA, tissue microarray; TME; tumor microenvironment; Treg, regulatory T cell. Financial support This study was supported by the Postdoctoral Supporting Fund of Fujian Medical University Union Hospital (2023XH022), the Startup Fund for Scientific Research of the Fujian Medical University (2023QH2020), the Joint Funds for the Innovation of Science and Technology, Fujian Province (2021Y9067), the National Natural Science Foundation of China (82372936) and the High-Level Medical Care Construction Foundation of Fujian Province ([2021]76). Authors’ contributions BLP, YLC and NHT designed the study. BLP, ZZ and DJY conducted animal experiments, processed data and wrote the manuscript. BLP, ZZ, DJY, XXZ, YXY and YL conducted the cell experiments and molecular biology experiments. HJH and XRC analyzed the pathological and clinical data. YLC and NHT supervised the project and reviewed the manuscript. All authors read and approved the final manuscript. Data availability statement The raw mass spectrometry data that support the findings of this study are deposited (OMIX006828, https://ngdc.cncb.ac.cn/omix/select-edit/OMIX006828; OMIX006841, [176]https://ngdc.cncb.ac.cn/omix/select-edit/OMIX006841). Conflict of interest The authors of this study declare that they do not have any conflict of interest. Please refer to the accompanying ICMJE disclosure forms for further details. Footnotes Author names in bold designate shared co-first authorship Supplementary data to this article can be found online at [177]https://doi.org/10.1016/j.jhepr.2024.101228. Contributor Information Yanling Chen, Email: chenyanling@fjmu.edu.cn. Nanhong Tang, Email: fztnh@fjmu.edu.cn. Supplementary data The following are the Supplementary data to this article: Multimedia component 1 [178]mmc1.pdf^ (10.3MB, pdf) Multimedia component 2 [179]mmc2.docx^ (99.5KB, docx) Multimedia component 3 [180]mmc3.pdf^ (536.3KB, pdf) Multimedia component 4 [181]mmc4.pdf^ (18.7MB, pdf) References