Abstract Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and the production of autoantibodies. Previous studies have indicated an association between high-salt diets (HSD) and an increased risk of RA, yet the underlying mechanisms remain unclear. Macrophage pyroptosis, a pro-inflammatory form of cell death, plays a pivotal role in RA. In this study, we demonstrate that HSD exacerbates the severity of arthritis in collagen-induced arthritis (CIA) mice, correlating with macrophage infiltration and inflammatory lesions. Given the significant alterations observed in macrophages from CIA mice subjected to HSD, we specifically investigate the impact of HSD on macrophage responses in the inflammatory milieu of RA. In our in vitro experiments, pretreatment with NaCl enhances LPS-induced pyroptosis in RAW.264.7 and THP-1 cells through the p38 MAPK/NF-κB signaling pathway. Subsequent experiments reveal that Slc6a12 inhibitors and SGK1 silencing inhibit sodium-induced activation of macrophage pyroptosis and the p38 MAPK/NF-κB signaling pathway, whereas overexpression of the SGK1 gene counteracts the effect of sodium on macrophages. In conclusion, our findings verified that high salt intake promotes the progression of RA and provided a detailed elucidation of the activation of macrophage pyroptosis induced by sodium transportation through the Slc6a12 channel. Keywords: High-salt diet, rheumatoid arthritis, macrophages, pyroptosis, SGK1, Slc6a12 Introduction Rheumatoid arthritis (RA) is a chronic systemic disorder defined by synovial inflammation and proliferation, concurrent with the generation of autoantibodies, culminating in the deterioration of cartilage and bone, and consequent disability [69]^1. It is hypothesized that the onset and progression of RA result from a multifaceted interplay of genetic, environmental, and life style determinants [70]^2^,[71]^3. This condition preferentially afflicts the geriatric female demographic, accounting for approximately 1% of the global population. Manifesting primarily through inflammation in small and medium-sized joints, RA substantially erodes the quality of patients' life [72]^4^,[73]^5. Further to this, sustained inflammation poses risks to additional organs, including the cardiovascular system and kidneys. Despite this, extant therapeutic strategies largely center on ameliorating inflammatory symptoms rather than providing comprehensive RA mitigation, and are frequently associated with a spectrum of adverse effects [74]^6. Contemporary understanding posits that the pathogenesis of bone and cartilage erosion in RA is predominantly due to the interactions involving both the innate and adaptive immune cohorts. Noteworthy is the aberrant activation of macrophages, innate immune cells, during the incipient phases of the disease [75]^7. RA-afflicted synovia witness a pronounced influx of these macrophages, believed to be sourced from both circulating monocytes and sessile tissue-resident populations. Monocyte-derived macrophages are progeny of hematopoietic stem cells located in the bone marrow, whereas resident macrophages trace their lineage back to embryonic development stages in the fetal liver and yolk sac. The phenotypic plasticity of macrophages is considerable, allowing them to adapt their functional stance in response to the evolving extracellular milieu [76]^8. Upon environmental cueing, they shift towards a pro-inflammatory stance, unleashing a cascade of inflammatory cytokines, including IL-1β, IL-12, IL-6, TNF-α and chemokines, which collectively conscript additional immune effector T and B cells to the site, thus exacerbating the inflammatory milieu within the joint space [77]^9. Pyroptosis, a lytic and inflammatory form of programmed cell death, primarily documented in relation to infectious disease pathologies, has recently garnered attention for its role in autoimmune diseases, including RA [78]^10^,[79]^11. Investigations have underscored the occurrence of macrophage pyroptosis in RA, implicating it as a driver in the disease's pathogenesis. This cell death pathway is distinguished by a swift breach of the cell membrane accompanied by the discharge of inflammatory mediators, most notably IL-1β and IL-18 [80]^12. The process is initiated when macrophages recognize exogenous danger cues such as lipopolysaccharide (LPS). This detection precipitates the assembly of the NLRP3 inflammasome, a multiprotein complex involving the NOD-like receptor thermal protein domain associated protein (NLRP3), the apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and procaspase-1. Subsequent activation triggers the release of IL-1β and IL-18 following their cleavage by caspase-1. Beyond eliciting an inflammatory response, macrophage pyroptosis fosters the aberrant clonal expansion of fibroblast-like synoviocytes and the secretion of matrix metalloproteinases, thus magnifying tissue damage and promoting irreversible bone erosion in RA [81]^13^,[82]^14. Salt is a common seasoning that enhances the flavor of food. The World Health Organization recommends that sodium chloride intake should be less than 5 grams per person per day [83]^15. However, statistics indicate that the per capita salt intake in most countries is between 9-12 grams or even higher, far exceeding the recommended amount. Excessive salt intake not only increases the risk of hypertension and cardiovascular disease, but recent studies have also demonstrated that a high-salt diet can lead to abnormal changes in the immune system, thereby increasing the risk of multiple sclerosis and systemic lupus erythematosus [84]^16. The impact of high-salt diet on the progression of RA is still inconclusive. A clinical study from Spain indicated that individuals with high sodium intake had a 50% increased risk of developing RA [85]^17. In this study, we have engineered a murine model to investigate the influence of dietary salt on collagen-induced arthritis (CIA) by employing DBA/1 mice. The model consisted of administering a high-salt diet to evaluate its impact on joint inflammation. Our observations reveal that mice fed with such a diet exhibited exacerbated joint destruction in the context of CIA. Further molecular investigations disclosed an upregulation of serum and glucocorticoid-regulated kinase 1 (SGK1) activity in response to elevated NaCl levels. This upregulation appeared to precipitate the activation of the mitogen-activated protein kinase (MAPK) p38 signaling pathway, which, in turn, potentiated the pyroptotic death of macrophages, thereby intensifying joint inflammation. These findings underscore the potential peril associated with high salt consumption as an aggravating factor for RA. Consequently, we propose adopting a low-salt diet as a strategic measure for individuals contending with RA. Additionally, our results nominate SGK1 as a promising molecular target for the development of innovative RA therapeutic strategies. Materials and Methods Clinical data The clinical data were sourced from patients admitted to the rheumatology department of the Second People's Hospital of Hefei between January 2022 and April 2023. The majority of patients included were aged between 35 and 85 years old (n=100), with a diagnosis of either RA or osteoarthritis (OA). Patients with a history of hypertension or heart disease, and those who required treatment with drugs containing sodium or potassium, were excluded from the study. Patients with RA were diagnosed using the 2010 classification criteria established by the American College of Rheumatology and the Federation of European Societies of Rheumatology. The diagnostic criteria for OA were in accordance with the Bone and Joint Diagnosis and Treatment Guidelines updated by the Orthopaedic Branch of the Chinese Medical Association in 2018. Specimens In this study, we utilized human knee synovium tissues from RA (n = 5) and OA (n = 5) patients undergoing total joint replacement in the Department of Orthopaedics of Hefei Second People's Hospital. All patients provided written informed consent prior to the experiment, and the study protocol was approved by the Biomedical Ethics Committee of Anhui Medical University (Approval No. 83230267). Animals Seven to eight-week-old male DBA/1 mice (average weight 18±2g) were procured from Gem Pharmatec (Nanjing, China) and were maintained in a Specific Pathogen Free (SPF) animal house, under conditions of 25°C and a 12-hour light-dark cycle. In the experiment, cage bias was taken into consideration. Instead of being fed in a separate ventilated cage, they were all kept in the same open environment. This study was approved by the Experimental Animal Ethics Committee of Anhui Medical University (Approval No. 20220454). HSD treatment The mice were randomly assigned to either the NSD (Normal mice fed standard diet and provided regular drinking water) or HSD (High-salt diet containing 4% added NaCl and drinking water with 1% NaCl) groups [86]^18, with 20 mice in each group. Prior to the experiment, the mice in the HSD group were acclimated to the high-salt diet for one week. After seven days, ten mice from each group were randomly selected to establish the collagen-induced arthritis (CIA) model. The mice were then divided into four experimental groups, with ten mice per group: NSD + normal group (NSD), HSD + normal group (HSD), NSD + CIA model group (NSD + CIA), and HSD + CIA model group (HSD+CIA). Throughout the duration of the experiment, the body weight, dietary intake, and water consumption of each group were measured at two-day intervals. CIA mouse model Fully emulsified chicken type II collagen (Chondrex, cat#20011, USA) was combined with complete Freund's adjuvant (Chondrex, cat#7023, USA) containing 5 mg/mL inactivated Mycobacterium tuberculosis. On day 0, 8-week-old male DBA/1 mice received a subcutaneous injection of 100 μL of the emulsified collagen at the tail root. On day 21, a second injection of 100 μL was administered to enhance immunity. The clinical severity of arthritis was evaluated between 29 and 49 days after the initial immunization. Evaluation of arthritis To assess the severity of CIA, two independent observers, unaware of the experimental protocol, conducted a comprehensive evaluation. Beginning on the 29th day after the initial immunization, the mice's clinical scores were measured three times every two days, including arthritis index, paw swelling number, and body weight. The criteria for arthritis index scoring were as follows: 0 = normal; 1 = erythema and slight swelling of the ankle joint; 2 = erythema and slight swelling from ankle to metatarsal or metacarpal joints; 3 = erythema and moderate swelling of ankle to metatarsophalangeal or metacarpal joints; 4 = erythema and severe swelling of ankle to toe joints. The paw swelling number of mice in each group was also recorded every 2 days from day 0, with each mouse's paw consisting of 1 ankle joint and 5 knuckles. A single spot of redness was scored as 1 point, with each mouse having a maximum score of 24. Histological analysis of joint and spleen To ascertain the histopathological hallmarks, we employed Hematoxylin and Eosin (H&E) staining. Hind limbs and spleens excised from the subjects were initially preserved in 4% paraformaldehyde for a duration of 24 hours. After the fixation process, hind limbs underwent a decalcification protocol for 30 days, with the decalcifying solution replenished biweekly. In contrast, spleens were promptly processed for paraffin embedding. We assessed inflammatory manifestations and the extent of bone erosion within the knee and ankle joints by adopting a systematic approach that incorporated an analysis of synovial hypertrophy, infiltration of inflammatory cells, pannus formation, and the integrity of bone and cartilage. This assessment was performed independently by a duo of experienced investigators to ensure objectivity. In addition to the joint evaluation, a separate pair of independent researchers conducted H&E staining on spleen tissue samples, whereby they quantified alterations based on parameters which included changes in spleen morphology, the prevalence of germinal centers, and the degree of leukocytic infiltration. Safranin O/Fast green staining Following the removal of wax, tissue sections were immersed in safranin O for three minutes, then briefly rinsed in distilled water for one minute. Fast green staining ensued for two minutes, succeeded by another distilled water rinse. Differentiation was achieved with a one-minute treatment in 1% acetic acid. Progressive dehydration was conducted using 95% ethanol, followed by clearance in xylene. The morphological alterations of the samples were then examined post-mounting with neutral balsam. Mouse primary peritoneal macrophages isolation and culture Following euthanasia by cervical dislocation, the corpses of mice were sanitized through a five-minute submersion in 75% ethanol. The abdominal integument was retracted to reveal the peritoneal cavity. A cooled phosphate-buffered saline (PBS) solution was injected into the splenic cavity, succeeded by gentle abdominal massaging for five minutes to dislodge immune cells. Peritoneal exudate was then aspirated using a syringe armed with a 21-gauge needle. The resultant fluid was centrifuged at 4°C at 400×g for ten minutes to precipitate macrophages. These cells were then re-suspended in culture medium and enumerated. The macrophages were seeded in Roswell Park Memorial Institute (RPMI) 1640 medium, procured from VivaCell in Shanghai, enhanced with 10% fetal bovine serum from WISENT, based in Nanjing, and buffered with an antibiotic-antimycotic cocktail comprising 1% penicillin and 1% streptomycin, both sourced from Beyotime in Shanghai. Following adherence to culture vessels, the cells were appropriated for experimental evaluations. Cell culture and treatment RAW264.7 cell line (Procell Life Science & Technology Co., Ltd, Wuhan, China) in 10% fetal bovine serum (WISENT, China was cultured in medium (High sugar DMEM) (VivaCell, Shanghai, China) and placed in an incubator at 37°C and 5% CO[2]. Mononuclear leukemia cell line (THP-1 cells) (Procell Life Science & Technology Co., Ltd, Wuhan, China) in 15% fetal bovine serum (WISENT, Nanjing, China) was cultured in medium (RPMI 1640) (VivaCell, Shanghai, China) and placed in an incubator at 37°C and 5% CO[2]. Prior to the experiment, THP-1 monocytes were transformed into M0 adherents after being treated with 100 nM phorbol12-myristate 13-acetate (PMA) (GLPBIO, CA, USA) on a 6-well plate for 48h. LPS (1μg/mL) (Solarbio, Beijing, China) in combination with ATP(5μM) (Sigma, USA) induces pyroptosis in Raw264.7 cells, whereas in THP-1-derived macrophages, pyroptosis is induced using only LPS. Slc6a12 inhibitor (BPDBA) (MCE, Nj, USA) was added to the six-well plate with a final concentration of 20μM, and the cells were stimulated for 24h. NaCl pretreatment was 50 mM for 12h. Western blotting (WB) The total protein was separated by 10% fast polyacrylamide (PAGE) gel (YEASEN, Shanghai, China), and the isolated protein was transferred to PVDF membrane (Millipore, MA USA). Sealed with 5% skim milk at room temperature for 2h, then with specific primary antibody at 4°C. Rabbit anti-β-actin (Cat#: AF7018, Affinity) and rabbit anti-phospho-SGK1 (Ser422) (Cat#: AF3001, Affinity) were purchased from Affinity Biosciences (Cincinnati, OH, USA). Rabbit anti-phospho-p38 MAPK (Thr180/Tyr182) (Cat#: 4511) and rabbit anti-p38 MAPK (Cat#: 8690) antibodies were purchased from Cell signaling technology (MA, USA). Anti-NF-κB p65 recombinant rabbit monoclonal antibody (Cat#: ET1603-12), anti-phospho-NF-κB p65(S529) recombinant rabbit monoclonal antibody (Cat#: ET1604-27) and anti-Gasdermin D (N terminal) recombinant rabbit monoclonal antibody (Cat#: HA721144) were purchased from HUABIO Co., LTD (Hangzhou, China). Rabbit anti-IL-18 (Cat#: [87]R24693) and rabbit anti-cleaved-Caspase-1 (Cat#: 341030) antibodies were purchased from ZEN-BIOSCIENCE (Chengdu, China). Rabbit anti-IL-1beta (Cat#: 66737-1-Ig), rabbit anti-SGK1 (Cat#: 28454-1-AP) and mouse anti-Slc6a12 (Cat#: 67700-1-Ig) antibodies were purchased from Proteintech group (Wuhan, China). Rabbit anti-NLRP3 (Cat#: WL02635) antibody was purchased by WANLEIBIO (Shenyang, China). After washing with PBS for 3 times, it was incubated with secondary antibody (Elabscience, Wuhan, China) at room temperature for 2h. After washing with TPBS, bands were observed with enhanced chemiluminescence reagent (Affinity, USA) by fully automated chemiluminescence imaging system (Tanon 5200, China). Immunohistochemical staining Mice was sacrificed on Day49, the knee joints were fixed and decalcified, dehydrated with fractional alcohol, embedded in paraffin, and sectioned for 5µm. The tissue sections were placed in an oven at 55°C overnight and immersed in xylene dewaxing and gradient ethanol. Washed with PBS 3 times for 15 minutes, washed with PBS 3 times, the slices were boiled in a large antigen repair solution at 100°C and cooled to room temperature. Wash with PBS 3 times, seal with endogenous peroxidase for 10 minutes, rinse with PBS 3 times, seal and serum for 15 minutes. The primary antibody F4/80 (Cell signaling technology, MA, USA) was then incubated overnight at 4°C. The next day, biotin-labeled secondary antibody was incubated at room temperature for 20 minutes, washed with PBS for 3 times, labeled with horseradish peroxidase marker, washed with PBS for 3 times, colored with DAB, stained with hematoxylin, dehydrated with gradient alcohol, and mounted with neutral adhesive. Finally, the tissue staining results were observed in the digital slice scanning system Pannoramic MIDI (Pannoramic MIDI II, Hungary, China). Immunofluorescence staining The RAW264.7 cells were adhered to a 24-well plate with a 1.5 mm polylysine coating. After pretreatment with NaCl (50 mM) for 12 hours, the RAW264.7 cells were stimulated with LPS and ATP. After 24 hours, the cells were fixed with acetone for 15 minutes at room temperature, followed by infiltration with PBS + 0.1% Triton X-100 for 15 minutes. Subsequently, the cells were washed three times with PBS and blocked with 5% BSA at room temperature for 1 hour to prevent non-specific antibody binding. Primary antibody staining was carried out overnight using NLRP3 antibody (HUABIO, Hangzhou, China) dissolved in 5% BSA at a 1:200 ratio. A secondary antibody conjugated with Alexa Fluor 488 (715-545-151, Jackson Immunol Research) was applied according to the manufacturer's instructions. The secondary antibody was incubated with the cells for 1 hour, followed by three washes with PBS. The cells were then incubated with DAPI for 10 minutes, washed three times to remove water stains, and finally, an anti-fluorescence quencher was applied to seal the slide cover. Images were obtained using a laser scanning confocal microscope (Leica SP8, Germany) and saved for analysis. Scanning electron microscopy The RAW264.7 cells were seeded onto a 1.5 mm polylysine coated surface of a 24-well plate. Consistent with previous studies [88]^19, Prior to inducing pyroptosis, RAW264.7 cells were pretreated with NaCl (50 mM) for 12 hours. After 24 hours, the cells were fixed in 2% glutaraldehyde solution (prepared in PBS) for 2 hours. Subsequently, the samples were dehydrated using a series of ethanol gradients ranging from 20% to 100% (20%, 40%, 60%, 80%, and 100%). Each ethanol concentration was maintained for approximately 30 minutes. Following dehydration, the samples were subjected to vacuum-assisted drying with 100% acetone, followed by critical point drying. Finally, the samples were gold-plated and observed under a GeminiSEM300 (Zeiss, Germany) scanning electron microscope. Flow cytometry Mouse paws were sectioned into 3-4 mm tissue pieces and placed in 1.5mL EP tubes. Hank's equilibrium salt solution (1mL), type II collagenase (1g/mL, 2μL), and CaCl2 solution (3mM, 5μL) were added. The mixture was then incubated at 37°C for 4 hours. After centrifugation and three washes with PBS, the cells were obtained through nylon sieving to yield a dispersed cell suspension. The treated single-cell suspension (1×10^6/100 μL) was incubated in the dark at 4°C for 30 minutes with a fluorescent antibody. Subsequently, the cells were centrifuged at 300 g, washed three times with PBS, and resuspended in 300 μL of PBS for cell sorting observation. The isolated cells were stained with the following antibodies: CD45-FITC (553080, BD Pharmingen), CD19-PE (553786, BD Pharmingen), CD3-PE (553063, BD Pharmingen), CD4-FITC (553650, BD Pharmingen), CD11b-FITC (101206, biolgend), Ly6G-PE (551461, BD Pharmingen), and Ly6C-APC (553129, BD Pharmingen). ELISA for cytokine measurements RAW264.7 macrophages were seeded in 6-well plates at a density of 5×10^5 cells/mL. Prior to inducing pyroptosis, the cells were pre-incubated with NaCl (50 mM) for 12 hours. Subsequently, the cell culture supernatant was centrifuged at 4°C and 1500 rpm for 15 minutes to discard any particles. The levels of secreted IL-1β and IL-18 were quantified using a commercially available mouse IL-1β and IL-18 ELISA kit (MLBIO, Shanghai, China), following the manufacturer's instructions. For measurement, mouse serum samples were appropriately diluted. Quantitative real-time PCR (RT-qPCR) Total RNA was extracted from mouse paw tissues and cells using TRIzol. A 20 μL total system of 5× reverse transcriptase was used for cDNA synthesis. The cDNA was subsequently diluted four times, and PCR amplification was conducted according to the manufacturer's instructions. The PCR reaction volume was set to 20 μL, and the amplification conditions were as follows: initial denaturation at 95°C for 5 minutes, followed by 40 cycles of denaturation at 95°C for 10 seconds, annealing at 60°C for 35 seconds, and extension at 72°C for 15 seconds. A melting curve analysis was performed by heating the samples to 95°C for 15 seconds, cooling to 60°C for 60 seconds, and then heating again to 95°C for 15 seconds. The expression of target genes was quantitatively analyzed using the 2-^ΔΔCT method, with β-actin or GAPDH genes used as internal references. The primer sequences for