Abstract Acute lung injury is a serious complication of sepsis with high morbidity and mortality. Pyroptosis is a proinflammatory form of programmed cell death that leads to immune dysregulation and organ dysfunction during sepsis. We previously found that adenosine deaminase acting on double-stranded RNA 1 (ADAR1) plays regulatory roles in the pathology of sepsis, but the mechanism of ADAR1 in sepsis-induced pyroptosis and lung injury remains unclear. Here, we mainly investigated the regulatory effects and underlying mechanism of ADAR1 in sepsis-induced lung injury and pyroptosis of pulmonary macrophages through RNA sequencing of clinical samples, caecal ligation and puncture (CLP)-induced septic mouse models, and in vitro cellular experiments using RAW264.7 cells with lipopolysaccharide (LPS) stimulation. The results showed that pyroptosis was activated in peripheral blood mononuclear cells (PBMCs) from patients with sepsis. In the CLP-induced septic mouse model, pyroptosis was mainly activated in pulmonary macrophages. LPS-stimulated RAW264.7 cells showed significantly increased activation of the NLRP3 inflammasome. ADAR1 was downregulated in PMBCs of patients with sepsis, and overexpression of ADAR1 alleviated CLP-induced lung injury and NLRP3 inflammasome activation. Mechanistically, the regulatory effects of ADAR1 on macrophage pyroptosis were mediated by the miR-21/A20/NLRP3 signalling cascade. ADAR1 attenuated sepsis-induced lung injury and hindered the activation of pyroptosis in pulmonary macrophages in sepsis through the miR-21/A20/NLRP3 axis. Our study highlights the role of ADAR1 in protecting pulmonary macrophages against pyroptosis and suggests targeting ADAR1/miR-21 signalling as a therapeutic opportunity in sepsis-related lung injury. Keywords: sepsis, macrophage, ADAR1, A20, pyroptosis, lung injury Introduction Sepsis is acknowledged as one of the main causes of death in the emergency room, with high incidence and mortality rates globally over many years [47]^1. Sepsis is a multifaceted disease with evolving definitions. The latest study states that sepsis is a dysregulated host response to infection that can result in life-threatening organ failure [48]^2. Sepsis is characterized by increased inflammation as well as immune suppression, and the imbalance between them leads to cellular dysfunction and even multiple organ failure. Clinically, six organ systems, the cardiovascular, respiratory, renal, neurological, haematological, and hepatic systems, are frequently examined in patients with sepsis [49]^3. Among them, sepsis-induced pulmonary dysfunction, which results in tachypnoea and acute respiratory distress syndrome (ARDS), is intimately associated with hypoxia and metabolic acidosis [50]^2. ARDS is a serious complication of sepsis with high morbidity and mortality [51]^4. Although the application of lung-protective ventilation has improved the prognosis of ARDS, additional research examining the mechanism and pathogenesis of sepsis-related acute lung injury (ALI) is also important to develop more efficient and novel approaches for ALI treatment [52]^5. Numerous cell death processes, such as apoptosis, necrosis, autophagy, and pyroptosis, are always activated as sepsis progresses [53]^3. Pyroptosis is an inflammatory programmed cell death that is mediated by caspases-1/4/5/11 [54]^6^, [55]^7. The Nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome mediates caspase-1 activation and the secretion of the proinflammatory cytokines interleukin (IL)-1β and IL-18 [56]^8^-[57]^10. Inflammatory caspases cleave the gasdermin D (GSDMD) substrate, which is important for pore formation on cell membranes, cytokine release, and ultimately pyroptosis [58]^11. Bacterial infection is a crucial incentive for the assembly and activation of the inflammasome in the host response [59]^12. Pyroptosis is needed for defence against bacterial infection to minimize tissue damage during sepsis. However, overactivated pyroptosis can result in septic shock, multiple organ dysfunction syndrome, or an increased risk of secondary infection [60]^8. Studies have revealed that pannexin-1 and P2X7 signalling triggers pyroptosis of bone marrow-derived macrophages through LPS-induced activation of caspase-11, suggesting possible therapeutic options for patients with bacterial sepsis [61]^13. Additionally, Xue et al. found that miR-21 mediated nuclear factor kappa B (NF-κB) signalling and protein A20-mediated NLRP3 inflammasome-regulated caspase-1 activation, both of which were positive regulators of LPS-induced sepsis and pyroptosis [62]^14. A prospective cohort study revealed increased peripheral blood mononuclear cell (PBMC) pyroptosis in patients with sepsis, which was related to disease severity and patient mortality; however, the mechanisms of pyroptosis in sepsis and its related organ disorders are complex, so in-depth exploration of the regulatory molecules and mechanisms of septic pyroptosis is important to develop targeted interventions and improve clinical outcomes [63]^15^, [64]^16. The regulation of cell death is greatly influenced by RNA modification [65]^17. The RNA editing enzyme known as adenosine deaminase acting on double-stranded RNA 1 (ADAR1) is widely known for converting adenosine residues into inosine (A-to-I) in double-stranded RNAs [66]^18. Studies have revealed that ADAR1 regulates innate immunity and cell death mechanisms such as pyroptosis, apoptosis, necrosis, and PANoptosis [67]^19^, [68]^20. These processes are crucial for host survival, development, and immunity against infections. Additionally, ADAR1 can modify the precursors of miRNA as well as the mature miRNA sequence during the production of microRNAs, which suppresses the expression of downstream target genes [69]^21. Our previous study demonstrated that ADAR1 regulated macrophage polarization by binding to miR-21 precursors and editing miR-21 biogenesis [70]^22. ADAR1 was also found to alleviate sepsis-induced inflammation and intestinal injury in mice by interfering with miR-30a synthesis and altering the miR-30a/SOCS3 axis [71]^23. Collectively, ADAR1 regulates the development of sepsis, although it is still unknown whether ADAR1 has an impact on the organ failure and cell death pathways induced by sepsis. Through clinical samples, animal models, and in vitro experiments, we investigated the regulatory effects and underlying mechanism of ADAR1 on sepsis-induced lung injury and pulmonary macrophage pyroptosis, offering new insights and fundamental evidence for the therapeutic intervention of sepsis-related lung injury and diseases. Methods Ethic statement All procedures during the study were approved by the Ethical Committee of Xijing Hospital of the Fourth Military Medical University (approved number: KY20212172-C-1). The human study complied with the principles of the Helsinki Declaration, and all participants signed informed consent forms. The experiments relating to mouse models were performed according to the Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of the Fourth Military Medical University (approved number: 20190305). Patients and PBMC sample collection We mainly enrolled patients with sepsis who presented to the emergency department and intensive care unit in Xijing Hospital from 07.2021 to 07.2022. Patients with sepsis were diagnosed with a SOFA score ≥2, combined with at least one infection site, and were managed following the newly updated international guidelines [72]^24^, [73]^25. Patients who were <18 or >80 years old or pregnant, had other blood diseases, cancer, or autoimmune diseases, and had incomplete clinical data were excluded. There were 52 patients with sepsis and 36 healthy volunteers included in the study. We collected blood samples from these patients within 24 h of admission. For PBMC isolation, blood samples were added to a Lymphocyte Separation Tube for Human Peripheral Blood (DAKEWE BIOTECH, Shenzhen, China) and centrifuged at 400 g and room temperature for 20 minutes. Plasma (supernatant) samples were collected and stored at -80 °C for further use. PBMC layers were isolated and centrifuged at 400 g and 4 °C for 10 minutes. The precipitate was added to red blood cell lysis buffer (Yeasen, Shanghai, China) and centrifuged again, followed by washing with PBS and further centrifugation three times. The final precipitate was kept as PBMC samples in liquid nitrogen. RNA sequencing analysis RNA sequencing analysis of PBMCs in the study was conducted by Gene Denovo Biotechnology Co. (Guangzhou, China). Four PBMC samples from patients with sepsis and 4 from healthy subjects were randomly selected to perform mRNA transcriptome sequencing. Briefly, RNA quality was detected by an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA, USA) and RNase-free agarose gel electrophoresis after total RNA extraction. Oligo (dT) beads were used to enrich mRNAs, which were fragmented into short fragments and reverse-transcribed into cDNAs. Double-strand cDNAs were purified using a QiaQuick PCR extraction kit (Qiagen, Venlo, Netherland), end repaired, poly (A) added, and ligated to the Illumina sequencing adapter. The products were sequenced using an Illumina NovaSeq 6000. Differentially expressed genes (DEGs) were screened by the parameters of fold change ≥ 2 and p value ≤ 0.05. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to perform pathway enrichment analysis of the screened DEGs. Bioinformatics analysis Expression data of ADARs in different cell types were obtained from The Human Protein Atlas website ([74]https://www.proteinatlas.org/) [75]^26^, [76]^27. Single-cell RNA sequencing data ([77]GSE207651) were obtained from the GEO database ([78]https://www.ncbi.nlm.nih.gov/geo/) and analysed using the Seurat R package (version 3.0.1). Cell types were clustered and annotated as previously described [79]^28. Luminex assay Serum levels of cytokines (IL-1β, IL-6, TNF-α, IL-10, and IL-4) in 29 patients with sepsis and 6 healthy volunteers were detected by a Luminex assay using a Human XL Cytokine Luminex Performance Panel Premixed Kit (#FCSTM18-30, R&D Systems, Minneapolis, MN, USA) according to the manufacturer's instructions. Outlier data were removed during statistical analysis. Scanning electron microscopy (SEM) Cell samples were rinsed with PBS, followed by fixation with electron microscope fixation liquid (#CR2202015, Servicebio, Wuhan, China) for 30 min in the dark. The slides were preserved at 4 °C. After dehydration and drying, the samples were sputtered with gold for 30 s by a sputter coater and further detected under a scanning electron microscope (Hitachi SU8100, Tokyo, Japan). Animal model establishment Male C57BL/6 mice (weight: 20-25 g, age: 8-10 weeks) were purchased from the Animal Center of the Fourth Military Medical University. Before modelling, all mice were adaptively cultured under normal conditions for one week. An animal sepsis model was established through caecal ligation and puncture (CLP) as previously described [80]^29. Briefly, mice were anaesthetized by inhalation of isoflurane. A midline incision (1 cm) on the lower abdomen was made, and then, the free end of the caecum was gently removed from the abdominal cavity to carry out caecal ligation by a 3/0 silk suture at the middle and two punctures with a 21-gauge needle, followed by gently squeezing a small amount of faeces to keep the puncture sites open. The caecum was carefully returned to the original position, and the abdomen was closed. A similar operation without CLP was performed on the mice in the sham group. A subcutaneous injection of normal saline (1 ml) was given for fluid resuscitation after surgery. A total of 1×10^8 PFU of ADAR1-overexpressing adenovirus (dissolved in 200 μl of PBS) was administered to septic mice through tail vein injection. Lung tissue samples were collected at 0 (sham), 6, 24, 48, and 72 h after surgery. Mouse ADAR1 overexpression adenovirus (Ad-O/E-ADAR1, [81]NM_001146296.1) was cloned and inserted into the pENT/CMV vector and prepared through a commercial service from GenePharma Co. Ltd. (Shanghai, China). Subsequent animal experiments were performed at 24 h after CLP administration. Complete blood count One hundred microlitres of peripheral blood was taken from the mice and transferred into a disposable capillary tube (#KJ002, KangJianMedical Apparatus, China). The tube was gently tapped to ensure optimal contact between the blood and anticoagulant coating on the inner wall of the tube. The tube was allowed to stand at room temperature for 5 minutes and then gently tapped again prior to utilizing the fully automated blood cell DF-3000Vet Analyzer (Beijing, China). Determination of bacterial load in blood and lungs Blood samples from the orbital cavity and lung tissues of mice in three groups, namely, sham, CLP, and CLP+OE-A, were obtained. Blood (100 μl) was collected from the orbital cavity of each group of mice and transferred into a sterile anticoagulant tube (#G4811-1.5ML, Servicebio). The bacterial count was determined by plating 100 μl of undiluted blood onto plate count agar medium (#HBPM002, Hopebio, China), followed by incubation at 37 °C. Subsequently, the colonies were counted. Furthermore, the lungs were excised and maintained under sterile conditions. A small volume (100 μl) of tissue homogenate was diluted 10-fold in sterilized phosphate-buffered saline (PBS), inoculated onto plate count agar medium, and incubated overnight at 37 °C. Afterwards, the bacterial colony count was determined [82]^30^, [83]^31. Haematoxylin and eosin (HE) staining and scoring Lung tissues were embedded in paraffin and cut into 5 μm-thick slices, followed by staining with haematoxylin and eosin dyes (Servicebio). The slices were observed under an upright microscope (Nikon Eclipse E100, Tokyo, Japan). The disease score and lung injury score of tissues were blindly evaluated by an experienced pathologist based on six random fields of vision of each HE-stained slice according to previously published scoring criteria with a slight modification. The disease clinical scoring system contained 5 aspects: appearance (4 points), behaviour changes at resting state (3 points) and after stimulation (3 points), respiratory clinical signs (3 points), and dehydration status (5 points) [84]^32. Lung injury scoring mainly included 4 aspects: pulmonary interstitial oedema, alveolar oedema, inflammatory infiltration, and alveolar haemorrhage, with each criterion scored according to severity (4 points in total for each, higher points indicated more severe damage) [85]^33. The lung injury scores were calculated by the sum of these criteria. Cell culture and transfection The murine RAW264.7 macrophage line was purchased from the Cell Culture Center, Chinese Academy of Medical Sciences (Beijing, China). The cells were cultured in high-glucose DMEM (Servicebio) with 10% foetal bovine serum (Zeta Life, Menlo Park, CA, USA) and 1% penicillin‒streptomycin (Biosharp, Anhui, China) at 37℃ under 5% CO[2]. RAW264.7 cells (1×10^6) were treated with 1 μg/ml lipopolysaccharide (LPS, Corning, Wilmington, NC, USA) to mimic the septic environment in vitro (6, 12, and 24 h). For cell transfection, RAW264.7 cells were seeded on 60 mm-cell dishes and cultured with serum-free Opti-MEM (Gibco, Brooklyn, NY, USA) 1 h before transfection when cell confluence reached 70-80%. Then, the cells were transfected with ADAR1-overexpression adenovirus (GenePharma), ADRA1-siRNA (RiboBio, Guangzhou, China), A20-siRNA (RiboBio), miR-21 mimic, miR-21 inhibitor (RiboBio) and their negative controls through Zeta transfection reagents (Zeta Life). Further LPS treatment was performed 24 h after transfection. The cells were collected for further experiments 48 h after transfection. Immunofluorescence (IF) staining Immunofluorescence staining was performed on both lung tissue and cellular slices through routine operation. For tissue staining, after dewaxing, rehydrating, antigen repairing, and blocking, paraffin-embedded tissue slices were incubated with diluted primary antibodies of anti-F4/80 (#GB11027, Servicebio), anti-Cytokeratin 7 (CK7, #GB11225, Servicebio), and/or anti-ADAR1 (#SC-73408, Santa Cruz), anti-cleaved-GSDMD (#AF4013, Affinity, Jiangsu, China), TUNEL (#GDP1043, Servicebio), anti-iNOS (#GB11119, Servicebio), or anti-CD163 (#GB11340-1, Servicebio) at 4 °C overnight, followed by incubation with their corresponding secondary antibodies at room temperature for 1 h in the dark. Between double staining procedures, 488-TSA (Servicebio) incubation and reantigen repair were conducted. DAPI (#G1012, Servicebio) was used for nuclear counterstaining. The slices were mounted with antifluorescence quenching sealing reagents (Boster, Wuhan, China) and captured under a fluorescence microscope (Life Technologies, Carlsbad, USA). For cellular staining, the primary antibodies used were anti-cleaved GSDMD (#AF4013, Affinity) and anti-caspase-1 (#GB11383, Servicebio). Mean fluorescence intensity (MFI) in a random field of view was measured using ImageJ software. Preparation and induction of bone marrow-derived macrophages Bone marrow-derived macrophages (BMDMs) were prepared following previously established protocols [86]^34. BMDMs were stimulated with LPS (1 mg/mL) to induce M1 polarization, and after 24 hours, the cells were harvested for further experimentation. Flow cytometry analysis Flow cytometry was adopted to assess molecular expression alterations in specific cells of human PBMCs and RAW264.7 cells in different groups. Human PE-conjugated CD14 antibody (#12-0149-42, eBioscience) was used to label monocytes of PBMCs for 30 min on ice in the dark. After three washes with FACS buffer, the cells were treated with BD Cytofix/Cytoperm Fixation and Permeabilization Solution (BD Biosciences) for 30 min and 1× BD Perm/Was buffer for 15 min. Anti-cleaved-GSDMD (#ab227821, human, Abcam, Shanghai, China) was further incubated with the cells for 30 min, followed by the addition of Alexa Fluor™ 488-conjugated goat anti-rabbit IgG (#A11008, Thermo Fisher Scientific, Waltham, MA, USA). For murine RAW264.7 cell detection, APC-conjugated anti-F4/80 (#MF48005, eBioscience, San Diego, CA, USA) was used to label macrophages. FITC-conjugated anti-CD11c (#11-0114-81, eBioscience) and PE-conjugated anti-CD206 (#12-2061-80, eBioscience) were used to label M1 and M2 polarization. Cell pyroptosis was evaluated by anti-cleaved GSDMD (#ab255603, mice, Abcam) and the following flow cytometry secondary antibody: Alexa Fluor™ 488-conjugated goat anti-rabbit IgG (#A11008, Thermo Fisher Scientific). The cell death states of BMDMs and RAW264.7 cells were determined using the FITC Annexin V Apoptosis Detection Kit I (#556547, BD Pharmingen, San Diego, CA) according to the manufacturer's instructions. In brief, cells (1×10^6) were resuspended in 100 μl of 1× binding buffer, followed by the addition of 5 μl of Annexin V-FITC and 10 μl of PI. Subsequently, the samples were incubated for 30 minutes at a temperature of 4 °C in darkness, and the fluorescence signals were detected using a COULTER EPICS XL flow cytometer. In addition, for determination of the CD4^+ and CD8^+ T-cell subsets within the mononuclear cell suspensions prepared from the blood of mice, the levels of CD4^+, CD8^+, and CD4^+/CD8^+ were measured using a flow cytometer. Mouse APC-conjugated anti-CD19 (#17-0193-82, eBioscience) and PE-Cyanine7-conjugated anti-CD3 (#25-0032-82, eBioscience) were used to label B and T lymphocytes. T-cell subsets were analysed by FITC-conjugated anti-CD4 (#11-0042-83, eBioscience) and PE-conjugated anti-CD8 (#12-0081-82, eBioscience) [87]^35. Cell samples were analysed using a NovoCyte flow cytometer and NovoExpress software (ACEA Biosciences, San Diego, CA, USA). RNP immunoprecipitation (RIP) The association between ADAR1 and miR-21 was studied in our previous study. Here, we performed RIP experiments again to verify the binding effects of ADAR1 on pre-miR-21 using RAW264.7 cells as previously described [88]^22^, [89]^36. A total of 20 million RAW264.7 cells were collected and lysed and precipitated with 30 mg of anti-ADAR1 (#SC-73408, Santa Cruz) or anti-IgG antibodies (#2729, CST) at room temperature for 4 h. IP products were then reverse transcribed, and quantitative PCR analysis was performed to assess the expression of pre-miR-21 and the internal reference. Dual luciferase reporter assay A pmirGLO Dual-Luciferase miRNA Target Expression Vector (GenePharma) containing wild-type (WT) or mutant (MUT) 3'UTR of A20 (sequences are shown in the figure) was constructed and cotransfected with miR-21-5p mimic or mimic-NC into RAW264.7 cells using Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA). Twenty-four hours after transfection, the relative luciferase activity of the cells was determined by an Infinite M1000 multimode microplate reader (Tecan, Männedorf, Switzerland) via a dual-luciferase reporter assay system (Promega, Madison, WI, USA). Quantitative reverse transcription (qRT)-PCR Total RNA from human PBMCs, murine lung tissues, or RAW264.7 cells was extracted using TRIzol reagent (Invitrogen) and then reverse-transcribed into cDNA by a PrimeScript RT Reagent Kit (TaKaRa, Beijing, China) according to the manufacturer's instructions. Moreover, the miRNA qRT‒PCR Stater Kit (#C107R-1, RiboBio) was used for RT and qPCR for miRNA. Further quantitative PCR was performed using cDNA templates, primers, PCR Mix (#11141ES60, Yeasen), and SYBR Green Mix (#11201ES08, Yeasen). Specific primer pairs were synthesized by Tsingke Biotechnology (Beijing, China). The primer sequences are shown in [90]Supplementary Table 1. The primer pairs of miR-21 and premiR-21 were synthesized by RiboBio. GAPDH and U6 RNAs were used as internal references for coding genes and miRNAs, respectively. Final relative