Rnf115 deficiency alleviates lipopolysaccharide/D-galactosamine-induced acute liver injury in mice
Rnf115 knockout mice were generated by CRISPR/Cas9-mediated genome editing. We designed two targets ending in NGG on both sides of exon 2 (59 bp), which caused a frameshift and led to the translational termination of RNF115 (amino acids 1–34; Fig. S1a). The Rnf115 knockout was confirmed by performing PCR of mouse tail genomic DNA (Fig. S1b) and immunoblotting and quantitative RT-PCR analysis of bone marrow-derived macrophages (BMDMs; Fig. S1c and S1d). The resulting Rnf115−/− mice did not exhibit spontaneous phenotypes compared with age-matched Rnf115+/+ littermate controls. Flow cytometry data indicated that no significant difference in the proportion and number of T and B cells, macrophages, or neutrophils in different tissues between Rnf115+/+ and Rnf115−/− mice.
Next, we investigated the effects of Rnf115 knockout mice in LPS/D-GalN-induced ALI. Survival analysis showed that 85.7% of Rnf115+/+ mice were dead 6 h after intraperitoneal administration of LPS/D-GalN, whereas only 28.6% of Rnf115−/− mice were dead at 8 h post treatment. Approximately 71.4% of Rnf115−/− mice were alive at 12 h post treatment (Fig. 1a). Examination of the gross morphology of the liver showed that Rnf115+/+ livers displayed hemorrhaging and congestion, whereas Rnf115−/− livers exhibited partial hemorrhaging at 5 h (Fig. 1b). The levels of serum aminotransaminase (ALT) and aspartate transaminase (AST) in Rnf115−/− mice at 5 h post treatment were significantly lower than those in Rnf115+/+ mice (Fig. 1c). Data from Hematoxylin and eosin (H&E) staining suggested that compared with Rnf115+/+ mice, Rnf115−/− liver displayed less hemorrhaging and structural disorders, which were consistent with the decreased levels of ALT and AST in LPS/D-Gal-treated Rnf115−/− mice (Fig. 1d). These results suggest that the deletion of Rnf115 alleviated the severity of ALI in mice.
Next, we performed reciprocal Rnf115 gain-of-function experiments using an AAV8-Rnf115 construct or AAV8-Null control. At 4 weeks following the AAV8-Rnf115 injection, the LPS/D-GalN-induced ALI mouse model was generated. As shown in Fig. 1e, compared with AAV8-Null-injected Rnf115−/− mice, Rnf115−/− mice in the AAV8-Rnf115 group showed more severe hemorrhaging and congestion in the liver and disrupted liver architecture (Fig. S2). Simultaneously, the recovery of Rnf115 expression in Rnf115−/− mice increased the levels of serum ALT and AST (Fig. 1f), indicating that RNF115 promotes LPS/D-GaIN-induced ALI in mice.
Rnf115 knockout attenuates inflammatory response in ALI mice
LPS is a common endotoxin that binds and activates TLR4 and the downstream cascade signaling pathways in macrophages, leading to the release of pro-inflammatory cytokines such as TNF-α and IL-6, which promote inflammatory impairment [27]. To determine the effect of Rnf115 deficiency on induction of the inflammatory response, serum samples and liver tissues were harvested at 5 h following LPS/D-GaIN administration. Our findings showed that the levels of inflammatory cytokines (TNF-α and IL-6) and chemokine MCP1/CCL2 was significantly downregulated in the serum of Rnf115−/− mice compared with those in Rnf115+/+ mice (Fig. 2a). Consistent with these findings, the levels of these cytokines in the liver tissue were lower in Rnf115−/− mice than in Rnf115+/+ mice (Fig. 2b). Moreover, the deletion of Rnf115 significantly decreased the activity of hepatic myeloperoxidase (MPO; Fig. 2c). Furthermore, we analyzed the degree of inflammatory cell infiltration using flow cytometry and immunohistochemistry, which indicated that the proportion of CD11b+CD14+ monocytes and CD11b+Ly6G+ neutrophils was significantly lower in Rnf115−/− mice than in Rnf115+/+ mice, both at 3 h and 5 h after LPS/D-GalN treatment (Fig. 2d and Fig. S3). Immunohistochemical staining further confirmed the lower neutrophil infiltration in the livers of Rnf115−/− mice treated with LPS/D-GalN (Fig. 2e). Collectively, these findings demonstrate that Rnf115 knockout attenuated LPS/D-GaIN-induced ALI, which was associated with a decrease in the hepatic inflammation response.
Rnf115 knockout upregulates hepatocyte autophagy and attenuates mitochondrial damage and apoptosis.
Using transmission electron microscopy, we analyzed the liver mitochondrial morphology in mice with ALI. As seen in Fig. 3a, Rnf115+/+ mice showed several swollen mitochondria in the liver tissue, whereas Rnf115−/− mice showed fewer damaged mitochondria. Next, we performed a terminal deoxynucleotidyl transferase-mediated dUDP nick-end labeling (TUNEL) assay to investigate hepatocyte apoptosis in LPS/D-GaIN-induced ALI. The results showed a significantly lower proportion of apoptotic hepatocytes in Rnf115−/− mice than in Rnf115+/+ mice (Fig. 3b and 3c). These data suggest that Rnf115 deficiency decreases mitochondrial damage and hepatocyte apoptosis in mice treated with LPS/D-GaIN.
Accumulating evidence suggests that enhanced autophagy protects against liver injury [11–14]. Next, we assessed cell autophagy in Rnf115+/+ and Rnf115−/− mice with ALI. Western blotting analysis showed that the accumulation of Lc3b-II in the liver was higher in Rnf115−/− mice with ALI than in Rnf115+/+ mice (Fig. 3d). However, restoration of Rnf115 expression in mouse liver achieved through AAV8-Rnf115 injection decreased the elevated Lc3b-II levels in Rnf115−/− mice (Fig. 3e). The expression pattern of BECN1/Beclin-1 protein detected by immunohistochemistry was similar to that of Lc3b-II (Fig. 3f). Furthermore, we injected 3-methyladenine (3-MA, an inhibitor of PIK3C3 complex and autophagy) into Rnf115−/− mice to inhibit autophagy. The serum AST and ALT levels in these mice were comparable with those in Rnf115+/+ mice (Fig. 3g and h). Moreover, the levels of P62/SQSTM1 in Rnf115−/− mice were lower than those in Rnf115+/+ mice, and the 3-MA treatment increased the expression of P62/SQSTM1 in Rnf115−/− mice (Fig. 3g), suggesting that 3-MA-inhibted autophagy promotes liver injury induced by LPS/D-GaIN. These findings indicate that Rnf115 deficiency-mediated autophagy contributes to the suppression of liver inflammation and maintains mitochondrial homeostasis in the context of ALI.
RNF115 interacts with LC3B and negatively regulates its protein homeostasis
RNF115 acts as an E3 ubiquitin ligase with a wide range of substrates [18]; furthermore, in the present study, Rnf115 knockout increased the levels of Lc3b. Therefore, we investigated whether LC3B is a substrate of RNF115. Co-immunoprecipitation (CO-IP) experiments showed that the T7-LC3B protein was present in the GFP-RNF115 immunoprecipitates and in a dose-dependent manner (Fig. 4a). In reciprocal co-immunoprecipitation assays, FLAG-RNF115 was detected in the GFP-LC3B immunoprecipitants (Fig. 4b), demonstrating that the two proteins interacted in a complex in cells. Pull-down experiments showed that the GST-LC3B protein bound to both eukaryotic-expressed GFP-RNF115 (Fig. 4c) and prokaryotic-expressed His-RNF115 (Fig. 4d).
It is known that LC3-binding proteins have the LC3-interacting region (LIR) motif [28]. We found that the RNF115 protein contained three LIR motifs: FAEL71 − 74, WDHL75 − 78, and WLEL258 − 261, respectively (Fig. 4e). Therefore, we constructed three mutant LIR domains to determine which RNF115 mutant failed to bind LC3B. These RNF115 mutants were as follows: RNF115-LIR1mt for F71/L74A, RNF115-LIR2mt for W75/L78A, RNF115-LIR3mt for W258/L261A, and RNF115-LIR1/2/3mt for F71/L74/W75/L78/W258/L261A. The pull-down results showed that GST-LC3B interacted with RNF115-LIR2mt and RNF115-LIR3mt but failed to bind both RNF115-LIR1mt and RNF115-LIR1/2/3mt (Fig. 4f), indicating that RNF115-LIR1 is required to bind LC3B. We also studied other members of the LC3/GABARAP family. The results revealed that GST-LC3A and GST-LC3C also interacted with GFP-RNF115, whereas GABARAP, GABARAP-L1, and GABARAP-L2 did not bind GFP-RNF115 (Fig. 4g and 4h).
Next, we investigated the biological significance of RNF115-LC3 interaction. Our results showed that RNF115 overexpression significantly decreased the LC3B protein levels with or without cell autophagic state (Fig. 5a). We then tested whether LC3B reduction in RNF115-overexpressing cells may be attributed to increased proteasomal or lysosomal degradation. As shown in Fig. 5b, the decreased LC3B was not restored in cells treated with Baf.A1 (lysosomal inhibitor; Fig. 5b, Lane 4 vs. Lane 2). In contrast, treatment with MG132 (proteasomal inhibitor) largely restored the LC3B expression in RNF115-overexpressing cells (Fig. 5b, Lane 6 vs. Lane 2), indicating that RNF115 overexpression leads to increased proteasomal degradation of LC3B. The same experiment was performed to assess LC3A and LC3C levels, and the results showed that RNF115 overexpression did not affect their levels with or without BafA1 and MG132 treatment (Fig. 5c and 5d).
Next, we analyzed the half-life of the LC3 protein using the protein translation inhibitor cycloheximide (CHX, 100 µg/mL). As shown in Fig. 5e, RNF115 overexpression promoted the decay of LC3B, conversely, RNF115 failed to affect the half-life of LC3A and LC3C (Fig. 5f and S4), indicating that RNF115 negatively regulates LC3B stabilization.
Rnf115 knockout inhibits LPS-stimulated macrophage activation
Liver macrophages play an important role in inflammatory damage in ALI. Because Rnf115 expression is silenced in all cells of Rnf115−/− mice, we wanted to investigate whether macrophages are involved in Rnf115-mediated effects. Rnf115+/+or Rnf115−/− mice were intravenously injected with clodronate liposomes, and hepatic F4/80+ macrophages were analyzed by immunohistochemistry at 48 h post treatment. As shown in Fig. S5a, liposome administration significantly decreased the number of F4/80+ macrophages in mouse liver, indicating effective clearance of mouse macrophages. Subsequently, mice were subjected to LPS/D-GalN stimulation for 5 h. As illustrated in Fig. S5b, depletion of macrophages reduced the difference in the extent of liver injury between Rnf115+/+ and Rnf115−/− mice, demonstrating that macrophage elimination increased LPS-stimulated inflammatory damage in Rnf115–/– mice. These results suggest that macrophages were required for the decease in liver injury in LPS/D-GalN-treated Rnf115 KO mice. Therefore, we further analyzed the biological activities of Rnf115−/− macrophages.
BMDMs isolated from the bone marrow of Rnf115+/+ and Rnf115−/− mice were used as experimental cells. qRT-PCR results showed that the mRNA levels of Tnf-α, Il-1β, Cd80, and Nos2 were significantly lower in Rnf115−/− BMDMs than that in Rnf115+/+ BMDMs after stimulation with LPS (Fig. 6a). Moreover, flow cytometry data (Fig. S5c and 6b) and western blotting (Fig. 6c and 6d) also showed that the protein levels of Cd80 and iNOS were lower in Rnf115−/− BMDMs. These findings suggest that Rnf115 KO reduced the activation of M1 macrophages and the inflammatory response to LPS.
LPS-stimulated macrophages activate the NF-κB and MAPK pro-inflammatory signaling pathways to induce the release of inflammatory cytokines. To investigate the potential mechanism underlying RNF115-regulated macrophage inactivation, Rnf115+/+ and Rnf115–/– BMDMs were stimulated with 100 ng/mL LPS for the indicated times. The levels of total and phosphorylated NF-κB p65 and MAPKs were then assessed by immunoblotting. Compared with LPS-treated Rnf115+/+, LPS-treated Rnf115–/– BMDMs showed a significant decrease in the levels of total and phosphorylated NF-κB p65 (Fig. 6e and 6f) and phosphorylated Jnk (Fig. 6g and 6h). However, Rnf115 KO did not affect the activity of ERK1/2 and p38MAPK (Fig. S6).
Because the nuclear translocation of NF-κB p65 is a key indicator of the activation of NF-κB signaling pathway, we next examined the distribution of the NF-κB p65 subunit using confocal microscopy. The fluorescence intensity and nuclear distribution of NF-κB p65 subunit were significantly lower in Rnf115–/– macrophages than that in Rnf115+/+ macrophages (Fig. S7). Using the nucleoplasm separation kit, we further demonstrated that the nuclear accumulation of p65 protein in Rnf115−/− BMDMs was significantly lower than that in Rnf115+/+ BMDMs after LPS stimulation (Fig. 6i). Collectively, the above data indicate that for LPS stimulation, Rnf115 KO limits the NF-κB and Jnk signaling pathways, consequently inhibiting macrophage activation and the release of inflammatory cytokines. These results indicate that Rnf115 deficiency-mediated inactivation of macrophages contributes to the suppression of liver inflammatory injury in the context of ALI.