Abstract Podocyte injury is a critical event in the pathogenesis of diabetic nephropathy (DN). Hyperglycemia, oxidative stress, inflammation, and other factors contribute to podocyte damage in DN. In this study, we demonstrate that signaling regulatory protein alpha (SIRPα) plays a pivotal role in regulating the metabolic and immune homeostasis of podocytes. Deletion of SIRPα in podocytes exacerbates, while transgenic overexpression of SIRPα alleviates, podocyte injury in experimental DN mice. Mechanistically, SIRPα downregulation promotes pyruvate kinase M2 (PKM2) phosphorylation, initiating a positive feedback loop that involves PKM2 nuclear translocation, NF-κB activation, and oxidative stress, ultimately impairing aerobic glycolysis. Consistent with this mechanism, shikonin ameliorates podocyte injury by reducing PKM2 nuclear translocation, preventing oxidative stress and NF-κB activation, thereby restoring aerobic glycolysis. Keywords: SIRPα, Podocyte injury, PKM2, Oxidative stress, Aerobic glycolysis, NF-κB Graphical abstract [41]Image 1 [42]Open in a new tab High light * • Podocyte SIRPα is downregulated in DN, and whereas transgenic SIRPα reduces podocyte damage, its deletion exacerbates it. * • SIRPα deficiency promoted PKM2 nuclear translocation, which triggers NF-κB activation and oxidative stress. * • Shikonin relieves SIRPα-deficient podocyte damage by preventing PKM2 nuclear translocation. __________________________________________________________________ Abbreviations Abbreviations Definition DN Diabetic nephropathy SIRPα signaling regulatory protein alpha PKM2 pyruvate kinase M2 WT Wild type CKO Podocyte-specific SIRPα-knockout KO Primary podocytes from podocyte-specific SIRPα-knockout TG Podocyte-specific SIRPα-transgenic STZ streptozotocin HPC human podocyte cell line PK Pyruvate kinase HG High glucose OCR Oxygen consumption rate FCCP fluorocarbonyl cyanide phenylhydrazone GBM Glomerular basement membrane GSEA Gene Set Enrichment Analysis NOX4 NADPH oxidase 4 TEMPO Mito-TEMPO TEM Transmission electron microscope PAS Periodic Acid-Schiff OE-SIRPα Overexpressing SIRPα pPKM2 Phosphorylated PKM2 (Tyr105) [43]Open in a new tab 1. Introduction Diabetic nephropathy (DN) is one of the most severe microvascular complications of diabetes mellitus and a leading cause of end-stage renal disease (ESRD)[[44]1] [[45][1], [46][2]]. Podocyte injury is a central mechanism in the progression of DN. Chronic hyperglycemia exacerbates oxidative stress, leading to increased production of reactive oxygen species (ROS), mitochondrial dysfunction, and activation of inflammatory pathways [[47][3], [48][4], [49][5], [50][6], [51][7], [52][8]]. Numerous studies suggest that maintaining a high level of ATP is essential for preserving the filtration structure and function of podocytes. Both mitochondrial function and glycolysis are critical in meeting the high ATP demand in podocytes [[53][8], [54][9], [55][10]]. Additionally, podocytes contain components of both innate and adaptive immune systems, enabling them to actively participate in inflammatory responses [[56]11]. However, the interaction between metabolic dysregulation and inflammatory activation in podocyte injury under hyperglycemic conditions remains poorly understood. Signal regulatory protein alpha (SIRPα), a member of the immunoglobulin superfamily, is predominantly expressed in leukocytes and plays a critical role in modulating various inflammatory responses [[57][12], [58][13], [59][14]]. The cytoplasmic tail of SIRPα contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs), which recruit and activate SHP-1/2, resulting in the dephosphorylation of intracellular phosphorylated substrates and the regulation of various downstream signaling pathways [[60]15]. Recent studies have demonstrated that SIRPα is also expressed in podocytes [[61]16,[62]17]. However, while these studies focus on the role of SIRPα in maintaining the normal structure of podocytes, there is a notable lack of research on the function of SIRPα under pathological conditions such as DN. In this study, we identified the reduction of SIRPα as an independent risk factor for renal function decline in patients with DN. We further validated the protective effects of podocyte-specific transgenic SIRPα against podocyte injury and elucidated the underlying protective mechanism, which involves the regulation of PKM2 nuclear translocation. 2. Materials and METHODS Ethics approval The collection of renal biopsy samples was approved by the National Clinical Research Center of Jinling Hospital (Nanjing, China). All animal experiments were conducted in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals. 2.1. Clinical and pathological information All individuals underwent renal biopsies between 2013 and 2023. Baseline clinical features were collected within one month following the biopsy for each patient. Kidney samples were obtained from the Renal Biobank at the National Clinical Research Center for Kidney Diseases. 2.2. Animals Podocyte-specific SIRPα-knockout (CKO) mice were generated as described in our previous study [[63]18]. Podocyte-specific SIRPα-transgenic (TG) mice were obtained from GemPharmatech. Podocyte-specific SIRPα-transgenic (TG) mice were obtained from GemPharmatech. SIRPα-transgenic flox homozygous mice were created using the CRISPR-Cas9 system. To generate SIRPα-transgenic mice, SIRPα-transgenic flox homozygous mice were mated with NPHS2-Cre mice. SIRPα-transgenic flox homozygous mice were created using the CRISPR-Cas9 system. To generate SIRPα-transgenic mice, SIRPα-transgenic flox homozygous mice were mated with NPHS2-Cre mice. For the construction of the DN model, Eight-week-old male mice were intraperitoneally injected with streptozotocin (STZ) (50 mg/kg, Sigma, S0130, St. Louis, MO, USA) for five consecutive days [[64]19]. Blood glucose levels were checked one week later, and mice with stable blood glucose levels exceeding 11.3 mmol/L after one month were included in the follow-up study. Blood glucose, urine protein, and urine creatinine levels were measured weekly. For treatment, DN model mice received intraperitoneal injections of shikonin (MCE, HY-N0822, New Jersey, USA) at a dosage of 2 mg/kg body weight every other day, beginning at 20 weeks of age [[65][20], [66][21], [67][22]]. At 24 weeks of age, mice (n = 6) were anesthetized with isoflurane, and samples were collected. All animal experiments were conducted in accordance with the guidelines established by the Animal Ethics Committee. 2.3. Urinary protein and creatinine Mouse urine was collected using a metabolic cage, and samples were centrifuged to remove cells, cell debris, and other contaminants. Urinary albumin was measured with the Mouse Urinary Albumin Assay Kit (Wako, 291–92703, Osaka, Japan), and urinary creatinine was measured with a creatinine kit (Wako, 290–65901, Osaka, Japan). 2.4. Transmission electron microscope (TEM) Renal tissues were collected, cut into 1 mm³ pieces, and fixed with 2.5 % glutaraldehyde at 4 °C overnight. After fixation, the tissues were washed three times with phosphate-buffered saline and post-fixed with 1 % osmium tetroxide at 4 °C for 2 hours. The tissues were then dehydrated in a graded series of acetone and ethanol before being embedded in epoxy resin. Sections of 80–90 nm thickness were stained with 5 % uranyl acetate and 0.1 % lead citrate. Electron micrographs were obtained and analyzed using a Hitachi 7500 transmission electron microscope. 2.5. RT-qPCR Total RNA was extracted from tissues or cells using Trizol reagent (Takara, 9109, Kusatsu, Japan). The cDNA was synthesized using the HiScript III All-in-one RT SuperMix Perfect for qPCR (Vazyme, R333-01, Nanjing, China). Gene expression was measured using SYBR Green real-time quantitative PCR (ChamQ Universal SYBR qPCR Master Mix, Vazyme, Q711-02, Nanjing, China), with mRNA levels normalized to β-actin expression. Primer sequences are listed in [68]Supplementary Table 1. 2.6. Histological analysis For Periodic Acid-Schiff (PAS) staining (Beyotime, C0142S, Shanghai, China) and immunohistochemical assays, renal tissues were embedded in paraffin and sectioned at a thickness of 2 μm. For immunofluorescence assays, renal tissues were embedded in optimal cutting temperature compound (OCT, Tissue-Tek, 4583, Osaka, Japan) and sectioned at a thickness of 4 μm. Details of antibodies used are listed in [69]Supplementary Table 2. DAPI (1:4000, Beyotime, C1002, Shanghai, China) was used to stain nuclei. Stained tissues were imaged using an Olympus FluoView 3000 confocal microscope (Olympus, Tokyo, Japan) and analyzed with FV3000-ASW software (version 1.3c; Olympus). Fluorescence intensity was quantified using ImageJ. 2.7. Western blot analyses Proteins were extracted using RIPA lysis buffer (Beyotime, P0013B, Shanghai, China) supplemented with 100 μg/mL PMSF (Beyotime, ST507, Shanghai, China) and a phosphatase inhibitor cocktail (Thermo, 78442, Waltham, US). The extracted proteins were separated by SDS-PAGE, transferred to PVDF membranes (Sigma, IPVH00010, St. Louis, MO, US), and then blocked with BSA. Membranes were incubated with primary and secondary antibodies, as detailed in [70]Supplementary Table 2. 2.8. Extraction and culture of primary mouse podocytes Primary mouse podocytes were isolated according to the established protocol[[71]23] [[72][23], [73][24]]. In brief, mice were anesthetized with isoflurane. Sequentially, 10 mL PBS, 2 mL Perfusion Solution 1 (containing 50 μL/mL Dynabeads [Dynabeads M450 tosylactivated; Invitrogen, 14013, Carlsbad, CA, US] in PBS), and 2 mL Perfusion Solution 2 (containing 50 μL/mL Dynabeads, 1 mg/mL Collagenase [Sigma, C6885, St. Louis, MO, US], 0.5 mg/mL Pronase E [Sigma, P6911, St. Louis, MO, US], and 50 U/mL DNase I [Sigma, D5025, St. Louis, MO, US] in PBS) were perfused at a rate of 5 mL/min. The perfused kidneys were cut into 1 mm³ pieces and transferred to a 2 mL centrifuge tube. Subsequently, 1 mL digestion solution (1 mg/mL Collagenase, 0.5 mg/mL Pronase E, and 50 U/mL DNase I in PBS) was added, and the tube was centrifuged at 300 rpm for 5 min. The digested solution was filtered through a 100 μm sieve, followed by magnetic separation to isolate glomeruli. Glomeruli were then washed five times with 1 mL PBS containing 10 % FBS and 3 % Penicillin/Streptomycin. Finally, glomeruli were resuspended and plated at a density of 300 glomeruli per 10 cm^2 on type I collagen-coated culture plates or flasks for further experiments. Cells were incubated with high glucose (48 hours, 30 mM, Sigma, 49163, St. Louis, MO, US) [[74]25], shikonin (1 μM, 24 hours, MCE, HY-N0822, New Jersey, US) [[75]26], mito-TEMPO (10 μM, 24 hours, MCE, HY-112879, New Jersey, US) [[76]27], or GLX351322 (10 μM, 24 hours, MCE, HY-100111, New Jersey, US) [[77]28]. 2.9. Detection of pyruvate kinase (PK) activity A total of 1 × 10^6 podocytes from each group were homogenized in 200 μL of normal saline (0.9 % NaCl). After centrifugation, the protein concentration of the supernatant was determined. Pyruvate kinase activity was measured using the Pyruvate Kinase Activity Assay Kit (Elabscience, E-BC-K611-M, Shanghai, China). Activity was calculated as the amount of enzyme required to consume 1 mmol of NADH per minute per gram of cellular protein at room temperature, defining one unit of pyruvate kinase activity. 2.10. Detection of total ROS ROS levels were quantified using a Total ROS Assay Kit (Invitrogen, 88-5930-74, Carlsbad, CA, US). Briefly, dilute the ROS assay buffer (500 × ) with dimethyl sulfoxide (DMSO). Prepare at least 5 × 10^5 cells per group. Incubate the cells with the ROS assay buffer in a 37 °C incubator with 5 % CO₂ for 60 min. After staining, centrifuge and resuspend the cells in phosphate-buffered saline (PBS), then immediately analyze on a flow cytometer (Beckman-Coulter Cyan ADP, Brea, CA). A total of 1 × 10^4 single cells were analyzed for each group, and the average fluorescence intensity was recorded. 2.11. Seahorse assay Oxygen consumption rate (OCR) was measured using an XF96 extracellular flux analyzer (Seahorse Bioscience) with the Seahorse XF Cell Mito Stress Test Kit (Agilent, 103015-100, Santa Clara, CA, US) Following previously published protocols [[78]4], cells were seeded on XF96 cell culture plates at a density of 1.5 × 10⁴ cells per well (near 90 % confluence) on the first day. Since podocytes cannot proliferate, they were cultured for 3–5 days to ensure stable attachment to the plate. On the day before the assay, hydrate the probe plate by adding calibration solution (Agilent, 103059-000, Santa Clara, CA, US) and incubate overnight at 37 °C without CO₂. On the experiment day, wash the cell culture plate per the manufacturer's instructions, then add analysis medium (Agilent, 103576-100, Santa Clara, CA, US) containing 1 mmol/L sodium pyruvate (Agilent, 103578-100), 2 mmol/L glutamine (Agilent, 103579-100), and 10 mmol/L glucose (Agilent, 103579-100). Stabilize the cell plates for 1 h at 37 °C in a CO₂-free incubator. After baseline measurements, sequentially inject oligomycin (2 μM, Agilent, 103015-100), FCCP (2 μM, Agilent, 103015-100), and rotenone/antimycin A (1.5 μM, Agilent, 103015-100) into each well. Measure and analyze OCR on the Agilent Seahorse XF Pro analyzer (US). Finally, calibrate OCR data by counting cells stained with the live-cell dye Hoechst (Thermo, 62249, Carlsbad, CA, US). 2.12. mRNA sequencing (RNA-seq) analysis Primary podocytes were isolated from wild-type (n = 3) and podocyte-specific SIRPα knockout mice (n = 3). After 7 days of standard culture, total RNA was extracted using Trizol reagent (Thermo, 15596018, Carlsbad, CA, US) according to the manufacturer's protocol. RNA quantity and purity were assessed using the Bioanalyzer 2100 and RNA 6000 Nano Lab Chip Kit (Agilent, 5067-1511, US). Only high-quality RNA samples with an RNA Integrity Number (RIN) greater than 7.0 were used for sequencing library construction. Gene expression analysis between the two groups was conducted using DESeq2 software, and genes with a false discovery rate (FDR) of less than 0.05 and an absolute fold change ≥2 were classified as differentially expressed ([79]https://www.ncbi.nlm.nih.gov/geo/, [80]GSE274089). Differentially expressed genes were then subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. To investigate the signaling pathways regulated by SIRPα deficiency in podocytes, we selected pathways under the signal transduction category in the KEGG secondary classification. The top 8 pathways, ranked by enrichment scores, are presented in [81]Fig. 5A. Additionally, to ensure key molecules were not overlooked, genes with expression levels greater than 1 and a fold change ≥1.5 were selected for Gene Set Enrichment Analysis (GSEA). Oxidative stress, a biological process associated with mitochondrial damage, was chosen from the Gene Ontology (GO) enrichment set, and differentially expressed genes involved in oxidative stress were identified based on P < 0.05. Lastly, pyruvate metabolism-related genes enriched in KEGG were highlighted, and a heat map was generated ([82]Supplementary Fig. 4). Fig. 5. [83]Fig. 5 [84]Open in a new tab Deficiency of SIRPα results in formation a positive feedback loop between nuclear PKM2, NF-κB activation, and oxidative stress. (A) KEGG secondary classification of signal pathway, sorted by enrichment scores (fc > 1.5, P < 0.05). (B, C) Proteins levels of SIRPα, pP65, P65, and IL-6 were detected by Western blotting (n = 3). (D, E) Levels of Nox4 mRNA in podocytes (n = 3). (F–I) Podocytes isolated from CKO mice were treated with (KO-Shikonin) or without 1 μM shikonin (KO-Ctrl) for 24 hours (n = 3). PKM2 distribution (F, Scale bar, 15 μm), protein level of pP65, P65 and IL-6 (G), total ROS production (H), mRNA level of Nox4 (I) in podocytes were determined. (J–L) NOX4 inhibitor GLX351322 (GLX, 10 μM, 24 hours) and Mito-TEMPO (10 μM, 24 hours) were used to inhibit the production or clearance of ROS in podocytes. PKM2 distribution (J, Scale bar, 15 μm), total ROS production (K), and protein levels of pP65, P65 and IL-6 were evaluated (L). (M, N) The primary CKO podocytes were transfected with control siRNA (siCtrl) or IL-6 siRNA (siIL-6). The nuclear distribution of PKM2 (M, Scale bar, 15 μm), and total ROS production were determined (n = 3) (N, Scale bar, 15 μm). 2.13. Statistical analysis Data were analyzed using GraphPad Prism 9.4. Differences between two groups were assessed using Student's t-test, while differences among multiple groups were evaluated using one-way or two-way ANOVA followed by Tukey's post hoc test. Each group of mice included six subjects, and all experiments were conducted at least three times. P value of <0.05 (∗), <0.01 (∗∗), <0.001 (∗∗∗) and <0.0001 (∗∗∗∗) were considered statistically significant. 3. Results 3.1. Podocyte-specific deletion of SIRPα promotes podocyte injury in experimental diabetic nephropathy (DN) mice SIRPα protein levels in podocytes progressively decrease with disease advancement in both DN patients and experimental DN mice ([85]Supplementary Figs. 1A and B). Similarly, SIRPα expression was found to gradually decline in podocytes exposed to serum from DN patients over time ([86]Supplementary Fig. 1C). When podocytes were treated with high glucose (HG) and various cytokines, including TGF-β, TNF-α, IL-6, IL-17, and IFN-γ, SIRPα levels were further reduced ([87]Supplementary Figs. 1D–F). To investigate the role of SIRPα under DN conditions, previously constructed homozygous SIRPα-flox knockout mice were crossed with NPHS2-Cre mice to generate podocyte-specific SIRPα knockout mice [[88]18] ([89]Supplementary Fig. 2A). The successful deletion of SIRPα in podocytes was confirmed ([90]Supplementary Figs. 2B–D), and a STZ-induced DN model was established ([91]Fig. 1A). Compared to their WT littermates, CKO mice exhibited an increased risk of podocyte injury, including elevated urinary protein levels ([92]Fig. 1B), glomerular hypertrophy ([93]Fig. 1C), podocyte loss ([94]Fig. 1D), and mitochondrial damage ([95]Fig. 1E). In streptozotocin (STZ) -induced diabetic nephropathy mice, the urinary albumin-to-creatinine ratio ([96]Fig. 1B) was significantly higher in the CKO-STZ group compared to the WT-STZ group, indicating worsened renal dysfunction. Pathological analysis revealed that podocyte-specific SIRPα knockdown exacerbated STZ-induced podocyte injury and accelerated kidney disease progression. In CKO-STZ mice, this was characterized by an enlarged glomerular area ([97]Fig. 1C), increased podocyte loss ([98]Fig. 1D), thickened glomerular basement membrane (GBM), widened podocyte foot processes, more extensive podocyte foot process effacement, and increased mitochondrial damage ([99]Fig. 1E). Due to the significant mitochondrial damage observed in podocytes, we further examined the mitochondrial ultrastructure and number in primary podocytes isolated from WT and CKO mice. The CKO podocytes exhibited shorter mitochondrial network length ([100]Fig. 1F), mitochondrial swelling, and cristae disruption ([101]Fig. 1G), all indicative of more severe mitochondrial damage compared to WT podocytes. In summary, these findings underscore the critical role of SIRPα deficiency in promoting podocyte injury, with a particular emphasis on mitochondrial dysfunction. Fig. 1. [102]Fig. 1 [103]Open in a new tab Reduction of SIRPα in Podocytes of Diabetic Nephropathy. (A) Schematic representation of wild type (WT-STZ) and CKO mice (CKO-STZ) induced by streptozotocin (STZ). (B) Analysis of the urinary albumin-to-creatinine ratio in WT-STZ and CKO-STZ mice. (C, D) Periodic Acid-Schiff (PAS) staining and WT1 immunohistochemistry (scale bar, 25 μm). Glomerular area was quantified based on PAS staining (C) (n = 6, 50 glomeruli randomly counted per mouse). Podocyte number was quantified based on WT1 immunohistochemistry results (D) (n = 6, 50 glomeruli randomly counted per mouse). (E) Ultrastructural analysis of podocytes in WT-STZ and CKO-STZ mice. Glomerular basement membrane (GBM) thickness, podocyte foot process width (GBM length/number of foot processes), percentage of podocyte foot process fusion (foot process fusion length/GBM length), and damaged mitochondria in podocytes were quantitatively analyzed based on podocyte ultrastructure (n = 6, 3 fields randomly selected per mouse). (F, G) Mitochondrial morphology in primary mouse podocytes (MitoTracker, scale bar, 15 μm) and mitochondrial ultrastructure in mouse podocytes. The mitochondrial network was reconstructed using ImageJ, and the mean mitochondrial branch length was measured (F) (n = 5). Mitochondrial ultrastructure was quantified, and mitochondrial area was analyzed at the same order of magnitude (G). 3.2. Podocyte-specific transgenic SIRPα alleviates podocyte injury in experimental diabetic nephropathy (DN) mice We further investigated whether restoring SIRPα expression in podocytes could alleviate podocyte injury and slow the progression of experimental DN. To achieve this, podocyte-specific SIRPα-transgenic (TG) mice were generated using the nephrin promoter, and their successful construction was verified ([104]Supplementary Figs. 2E–H). TG mice exhibited a significantly lower urinary albumin/creatinine ratio compared to wild-type (WT) mice ([105]Supplementary Fig. 3A). To assess the protective effects of transgenic SIRPα under pathological conditions, STZ was administered. In STZ-induced TG mice (TG-STZ), the urinary albumin/creatinine ratio ([106]Fig. 2A), glomerular area ([107]Fig. 2B), podocyte loss ([108]Fig. 2C), glomerular basement membrane (GBM)thickness, foot process width, foot process fusion percentage, and the extent of mitochondrial damage ([109]Fig. 2D and E) were significantly reduced compared to STZ-induced WT mice (WT-STZ). There were no significant differences in baseline pathological histology and podocyte ultrastructure between WT and TG mice under non-pathogenic conditions ([110]Fig. 2B–E). There was no significant difference in the ultrastructure or quantity of mitochondria between primary podocytes from WT and TG mice ([111]Fig. 2F). Fig. 2. [112]Fig. 2 [113]Open in a new tab Podocyte-Specific Transgenic SIRPα protects against hyperglycemia-induced podocyte injury. (A) Urinary albumin-to-creatinine ratio in WT, WT-STZ, TG, and TG-STZ mice (n = 6). (B) PAS staining of renal paraffin sections (scale bar, 25 μm). Glomerular area was quantified (n = 6, 50 glomeruli randomly counted per mouse). (C) Podocyte number was quantitatively analyzed based on WT1 immunohistochemistry (n = 6, 50 glomeruli randomly counted per mouse; scale bar, 25 μm). (D) Ultrastructural analysis of podocytes, including glomerular basement membrane (GBM) thickness, podocyte foot process width (GBM length/number of foot processes), percentage of podocyte foot process fusion (foot process fusion length/GBM length). (E) Damaged mitochondria in podocytes were quantitatively assessed (n = 6, 3 fields randomly selected per mouse). (F) Mitochondrial morphology in primary mouse podocytes (MitoTracker, scale bar, 15 μm) and mitochondrial ultrastructure. The mitochondrial network was reconstructed using ImageJ, and the mean mitochondrial branch length was measured (n = 5). To further elucidate the protective role of SIRPα in podocytes under DN conditions, we assessed the expression levels of podocyte-specific genes and proteins. In vitro, under stimulation with cytokines (TNF-α, IL-6, IL-17, IFN-γ, and TGF-β) and high glucose (HG), TG podocytes consistently exhibited stable expression of SIRPα ([114]Supplementary Figs. 3B and C). Moreover, the levels of nephrin and synaptopodin were significantly higher in TG podocytes compared to WT podocytes ([115]Supplementary Figs. 3D and E). In vivo, diabetic TG podocytes displayed sustained high expression of SIRPα ([116]Supplementary Figs. 3F and G). WT-STZ podocytes showed a hyperglycemia-induced reduction in nephrin and synaptopodin levels, a decline that was notably mitigated in TG-STZ podocytes ([117]Supplementary Figs. 3F and G). Taken together, these findings suggest that transgenic expression of SIRPα in podocytes provides protection against podocyte injury under diabetic conditions. 3.3. SIRPα rescues high glucose-induced oxidative stress and increases glycolytic flux in podocytes The findings mentioned above underscore the critical role of SIRPα in maintaining mitochondrial integrity and function. To further elucidate the underlying molecular mechanisms, we examined the role of SIRPα in these processes. Gene Set Enrichment Analysis (GSEA) of RNA sequencing data ([118]https://www.ncbi.nlm.nih.gov/geo/, [119]GSE274089) from primary podocytes of WT and CKO mice revealed that numerous genes associated with the oxidative stress response, linked to mitochondrial damage, were upregulated ([120]Fig. 3A). Genes exhibiting a fold change greater than 1.5 (P < 0.05) are detailed in [121]Supplementary Fig. 4A. Further validation confirmed that the expression of Il-6, Cd36, and Dgkk ([122]Fig. 3A), and the level of mito-ROS ([123]Supplementary Fig. 5A) and total ROS ([124]Fig. 3B) were significantly upregulated, while mitochondrial respiratory capacity (Oxygen consumption rate, OCR) and ATP levels ([125]Fig. 3C) were markedly decreased in CKO podocytes compared to WT podocytes (WT, KO). These findings indicate pronounced mitochondrial damage and dysfunction in CKO podocytes. In the context of DN, mitochondrial dysfunction and impaired glycolytic flux are believed to play a pivotal role in disease progression [[126]29,[127]30], leading us to investigate the relationship between mitochondrial dysfunction and aerobic glycolysis. As depicted in [128]Supplementary Fig. 4B, genes associated with pyruvate metabolism were downregulated in CKO (KO) podocytes. Further validation demonstrated that the expression levels of Ppargc1a, Ldha, Pfkl, Pgk1, Pdk1, and Slc4a4 were significantly reduced ([129]Fig. 3D). Additionally, pyruvate kinase activity in CKO podocytes was found to be decreased compared to WT podocytes ([130]Fig. 3E). Thus, both mitochondrial respiration and glycolytic flux are disrupted in CKO podocytes. Fig. 3. [131]Fig. 3 [132]Open in a new tab SIRPα rescues high glucose-induced oxidative stress and increases glycolytic flux in podocytes. (A) Gene Set Enrichment Analysis (GSEA) enrichment profiles of oxidative stress (NES = 1.80, FDR<0.01, P < 0.001). RT-qPCR was used to verify the expression of related genes (n = 6). (B) The total ROS levels in primary podocytes from WT and CKO mice (n = 3). (C) The OCR and ATP levels in podocytes from WT and CKO mice (n = 6). (D) The mRNA levels of pyruvate metabolism-related genes were detected (n = 6). (E) The pyruvate kinase activity in podocytes from WT and CKO mice (n = 3). Mito-TEMPO (10 μM, 24 hours) was used to clearance of ROS. (F–H) Levels of mito-ROS (F), OCR and ATP (G), pyruvate kinase activity (H) in podocytes. (I–M) TG and WT podocytes were treated with 30 mM glucose for 48 h. Oxidative stress related genes (I), total ROS (J, n=3), OCR and ATP levels (K), pyruvate metabolism related genes (L, n = 6), and pyruvate kinase activity (M, n = 3) in HG-induced WT and TG podocytes were detected. ROS are both a product and an indicator of cellular oxidative stress[[133]31], [[134][31], [135][32]]. Consequently, we investigated the relationship between ROS production and the disruptions in glycolysis and mitochondrial respiration observed in CKO podocytes. Treatment with the mitochondrial-targeted antioxidant Mito-TEMPO (TEMPO) was used to scavenge mito-ROS, thereby reducing ROS levels ([136]Fig. 3F), resulted in a significant increase in oxygen consumption rate (OCR) and ATP levels in CKO podocytes ([137]Fig. 3G). Additionally, pyruvate kinase activity was also enhanced in CKO podocytes following TEMPO treatment ([138]Fig. 3H). These findings suggest that inhibiting mito-ROS can induce mitochondrial respiration and glycolytic flux in CKO podocytes. We subsequently explored the impact of transgenic SIRPα on mitochondrial respiration and glycolysis in podocytes under high glucose (HG) conditions. In HG-induced TG podocytes (TG + HG), the expression of oxidative stress-related genes, including Dgkk, Cd36, Sod1, and Il-6, was downregulated compared to HG-induced WT podocytes (WT + HG) ([139]Fig. 3I). Additionally, TG + HG podocytes exhibited reduced mito-ROS ([140]Supplementary Fig. 5B) and total ROS production ([141]Fig. 3J), accompanied by increased OCR and ATP levels ([142]Fig. 3K), relative to WT + HG podocytes. Furthermore, glycolysis-related genes, such as Ppargc1a and Ldha, were significantly upregulated ([143]Fig. 3L). Pyruvate kinase activity (PK) was also elevated in TG + HG podocytes compared to WT + HG podocytes ([144]Fig. 3M). Collectively, these findings underscore the crucial role of SIRPα in maintaining mitochondrial respiration and glycolytic flux in podocytes under hyperglycemic conditions. 3.4. SIRPα deficiency facilitates PKM2 nuclear translocation by encouraging PKM2 phosphorylation SIRPα exerts its regulatory effects by recruiting the protein tyrosine phosphatase SHP1 through its immunosuppressive ITIM motif, which modulates the phosphorylation of various downstream kinases [[145]33]. To explore the molecular mechanisms by which SIRPα influences oxidative stress, mitochondrial respiration, and glycolytic flux, we constructed a FLAG-tagged SHP1 mutant plasmid by mutating the Cysteine 453 to serine 453. This mutation inhibited SHP1's dephosphorylation ability without impairing its capacity to bind substrates[[146]18] [[147][18], [148][34]]. Thus, overexpressing this mutant plasmid enriches downstream target molecules. Proteins bound to the mutant SHP1 were then isolated via FLAG antibody immunoprecipitation for mass spectrometry analysis. By integrating RNA-seq findings on oxidative stress and pyruvate metabolism pathways with mass spectrometry results ([149]Fig. 3), we identified that PKM2, ENO1, ACACB, HK2, and RANBP2 proteins were associated with oxidative phosphorylation and pyruvate metabolism ([150]Supplementary Fig. 6). PKM2, a well-known rate-limiting enzyme catalyzing the final step of glycolysis, also possesses protein kinase activity. Phosphorylation of PKM2 at tyrosine 105 regulates diverse biological processes, including cell inflammation and ROS production[[151]35] [[152][35], [153][36]]. Thus, PKM2 was selected as a candidate target protein of SIRPα for further validation. We subsequently confirmed the binding interaction between SIRPα, PKM2, and SHP1 ([154]Fig. 4A and B). Knockdown or overexpression of SIRPα resulted in increased or decreased phosphorylation of PKM2, respectively ([155]Fig. 4C and D). Stimulation with serum from DN patients led to a reduction in SIRPα expression while simultaneously increasing phosphorylated PKM2 in HPC ([156]Fig. 4E). Immunofluorescence staining of renal tissue sections further revealed a significant increase in nuclear pPKM2 in podocytes from DN patients ([157]Fig. 4F). Fig. 4. [158]Fig. 4 [159]Open in a new tab SIRPα deficiency facilitates PKM2 nuclear translocation by promoting PKM2 phosphorylation. (A) Co-immunoprecipitation of SHP1 with SIRPα and PKM2. (B) Co-immunoprecipitation of PKM2 with SHP1. (C) Phosphorylation of PKM2 was induced in SIRPα knockdown podocytes (SIRPα siRNA). (D) Phosphorylation of PKM2 was inhibited in SIRPα overexpressing podocytes (OE-SIRPα). (E) Protein levels of SIRPα, pPKM2, PKM2 and podocin in HPC treated with serum from healthy persons and DN patients (n = 5). (F) The expression and distribution of SIRPα (red), and pPKM2 (green) were determined by immunofluorescence staining in renal tissues of DN patients. Scale bar, 25 μm. Three experiments were repeated independently. (G) The expression and distribution relationship between SIRPα and PKM2 were determined in primary podocytes from WT, TG, CKO mice with or without 30 mM HG treatment. Nucleus (blue, DAPI), F-actin (red), and PKM2 (green). Three experiments were repeated independently. (H) SIRPα, pPKM2, and PKM2 were measured in podocytes obtained from WT, TG, CKO, WT-STZ, TG-STZ, and CKO-STZ mice (Scale bar, 15 μm, n = 6). The level of pPKM2 was calculated using Image Pro Plus 6.0. To determine whether the nuclear translocation of PKM2 is regulated by SIRPα, we isolated podocytes from WT, CKO, and TG mice, and subjected them to HG stimulation. CKO podocytes exhibited higher basal levels of PKM2 nuclear translocation compared to WT podocytes ([160]Fig. 4G). Upon HG stimulation, both WT and CKO podocytes showed increased PKM2 nuclear translocation, with the translocation in CKO podocytes being more pronounced than in WT podocytes ([161]Fig. 4G). In contrast, TG podocytes were resistant to HG-induced PKM2 nuclear translocation. Notably, under normal conditions, PKM2 was highly enriched in the foot processes of both WT and TG podocytes, while this enrichment was absent in CKO podocytes. Under HG conditions, the PKM2 enrichment in the foot processes of WT podocytes was lost; however, TG podocytes partially resisted this HG-induced effect. Similarly, phosphorylated PKM2 levels were elevated in the glomeruli of diabetic CKO mice compared to WT mice, whereas these levels were decreased in diabetic TG mice compared to WT mice ([162]Fig. 4H). Collectively, these findings demonstrate that hyperglycemia induces PKM2 phosphorylation and nuclear translocation in podocytes via SIRPα. 3.5. SIRPα deficiency facilitates formation of positive feedback loop between PKM2 nuclear translocation, NF-κB activation, and oxidative stress To elucidate the signal transduction pathway underlying PKM2 nuclear translocation, we performed KEGG secondary classification analysis based on the RNA-seq data from WT and CKO podocytes. The analysis identified the NF-κB pathway as the top-ranked pathway according to enrichment score ([163]Fig. 5A). Further validation confirmed that NF-κB activation was significantly higher in CKO podocytes compared to WT podocytes ([164]Fig. 5B), while transgenic SIRPα effectively resisted HG-induced NF-κB activation ([165]Fig. 5C). In addition, proinflammatory genes Nlrp3, Casp1, Il1b, Il18 were also upregulated ([166]Supplementary Figs. 7A and B). As a co-transcription factor, NF-κB not only promotes the expression of various cytokines [[167]37,[168]38], but also upregulates the expression of several members of the NOX family, thereby enhancing oxidative stress and inflammatory responses [[169][39], [170][40], [171][41]]. Given that NADPH oxidase 4 (NOX4) is the most abundant NOX isoform in the kidney, we examined Nox4 expression and found it to be upregulated in CKO podocytes compared to WT podocytes ([172]Fig. 5D). Additionally, Nox4 levels were lower in HG-induced TG podocytes compared to HG-induced WT podocytes ([173]Fig. 5E). Shikonin, a known PKM2 inhibitor, has demonstrated a preventive effect on kidney diseases; however, the underlying mechanism remains incompletely understood [[174][42], [175][43], [176][44]]. Larissa et al. demonstrated that shikonin can reverse the nuclear translocation of PKM2 and restore glycolytic flux in neuronal cells, thereby reestablishing the healthy characteristics of neurons [[177]45]. In this study, we found that shikonin inhibited PKM2 nuclear translocation ([178]Fig. 5F), NF-κB activation ([179]Fig. 5G), total ROS production ([180]Supplementary Fig. 7C and Fig. 5H), and Nox4 expression ([181]Fig. 5I), while it increased pyruvate kinase activity ([182]Supplementary Fig. 7D), mitochondrial respiration, and ATP production ([183]Supplementary Fig. 7E) in CKO podocytes. Similar effects were observed with the NOX4 inhibitor GLX351322 (GLX) and the superoxide scavenger Mito-TEMPO (TEMPO) ([184]Fig. 5J–L, [185]Supplementary Figs. 7F–J). Interestingly, the treatment of SIRPα KO podocytes with mito-TEMPO also resulted in the downregulation of IL-6 and NOX4 ([186]Fig. 5L, [187]Supplementary Fig. 7J. Additionally, interfering with IL-6 expression inhibited PKM2 nuclear translocation ([188]Supplementary Fig. 7K, Fig. 5M) and total ROS production ([189]Supplementary Fig. 7L and Fig. 5N), while enhanced pyruvate kinase activity ([190]Supplementary Fig. 7M) and reduced mito-ROS ([191]Supplementary Fig. 7N). We further validated the effects of PKM2 knockdown in primary podocytes with SIRPα knockout. As shown in [192]Supplementary Fig. 8, PKM2 knockdown partially counteracted the activation of the NF-κB signaling pathway induced by SIRPα knockout ([193]Supplementary Figs. 8A–C), but it did not reduce overall ROS levels ([194]Supplementary Fig. 8D) or mitochondrial ROS production ([195]Supplementary Fig. 8E). The differing effects of PKM2 knockdown on ROS production and NF-κB signaling pathway activation may be due to a reduction in PKM2 levels in both the nucleus and cytoplasm. Notably, neither SIRPα knockout nor shikonin treatment affected PKM2 expression levels ([196]Fig. 4, [197]Fig. 6E); instead, they promoted the nuclear translocation of PKM2, resulting in decreased cytoplasmic PKM2 and increased nuclear PKM2. Taken together, these findings suggest the that there may be a positive feedback loop between PKM2 nuclear translocation, NF-κB activation, and ROS production, with SIRPα playing a regulatory role in this loop. Fig. 6. [198]Fig. 6 [199]Open in a new tab Shikonin alleviates hyperglycemia induced podocyte injury in CKO mice via inhibiting PKM2 nuclear translocation. (A) Schematic diagram of shikonin administration on STZ-induced DN experimental mice. WT and CKO mice were given STZ at 8 weeks of age for constructing experimental DN model, and shikonin was administrated every two days. After four weeks of administration, biochemical and pathological indicators of the mice were measured to evaluate the effect of shikonin on renal disease progression. (B) Levels of urinary albumin/creatinine (n = 6). (C) PAS staining of renal sections and glomerular area was counted (n = 6, and 50 glomeruli were counted randomly per mouse, Scale bar, 25 μm). (D) Immunohistochemical staining of WT1 to calculate the number of podocytes (n = 6, and 50 glomeruli were counted randomly per mouse, Scale bar, 25 μm). (E) The expression and nuclear distribution (blue, DAPI) relationship between SIRPα (purple), Nephrin (red), and PKM2 (green) were determined through immunofluorescence staining. Scale bar, 15 μm. (F) The protein levels of pP65, P65 and IL-6 in renal cortex of mice. 3.6. Shikonin alleviates hyperglycemia-induced podocyte injury in CKO mice via inhibiting PKM2 nuclear translocation Finally, we investigated the in vivo effects of shikonin on alleviating podocyte injury caused by SIRPα deficiency. Shikonin was administered to STZ-induced WT and CKO mice every two days ([200]Fig. 6A). Notably, shikonin significantly mitigated the increase in urinary albumin-to-creatinine ratio ([201]Fig. 6B), glomerular area ([202]Fig. 6C), and podocyte loss ([203]Fig. 6D) that were induced by SIRPα deletion. Importantly, shikonin markedly inhibited the nuclear translocation of PKM2 in podocytes ([204]Fig. 6E). Additionally, shikonin strongly suppressed NF-κB activation and IL-6 expression in the glomeruli of STZ-induced CKO mice ([205]Fig. 6F). These findings suggest that shikonin inhibits PKM2 nuclear translocation, NF-κB activation, and IL-6 expression, thereby protecting against podocyte damage associated with SIRPα deficiency. 4. Discussion This study demonstrated that SIRPα acts as a molecular integrator of mitochondrial dysfunction, inflammatory response, and aerobic glycolysis in podocytes through the regulation of PKM2 nuclear translocation. Oxidative phosphorylation (OXPHOS) and glycolysis are two key mechanisms by which cells convert glucose into energy. However, there is no consensus on the predominant metabolic pathway in podocytes. For instance, some studies indicate that kidneys are unaffected by the podocyte-specific deletion of genes involved in mitochondrial production and dynamics [[206]46]. Conversely, other studies have shown that mitochondrial respiration accounts for approximately 77 % of podocyte respiration[[207]47] [[208][47], [209][48]]. Consistent with our findings, Qi et al. [[210]25] reported that a reduction in podocyte-specific PKM2 leads to mitochondrial dysfunction and a decrease in glycolytic flux, whereas PKM2 enzyme activity enhancers reverse mitochondrial dysfunction and increase glycolytic flux in podocytes. These contradictions may result from the use of different experimental settings, animal models, cell types, or disease models. Some studies have hypothesized that OXPHOS provides energy for the cytoplasm of podocytes, while glycolysis supplies energy for the foot processes of these cells [[211]49]. Due to their distinct architecture, mitochondria in podocytes are unevenly distributed [[212]49]. This study also offers direct experimental evidence supporting this hypothesis: under physiological conditions, PKM2 is enriched in the foot processes of podocytes ([213]Fig. 4G); under HG conditions, SIRPα deletion promotes PKM2 nuclear translocation. Nuclear PKM2 can function as a transcriptional co-activator of glycolysis-related genes [[214][50], [215][51], [216][52]]. While we observed that SIRPα deletion-induced PKM2 nuclear translocation did not promote the expression of glycolytic genes, it inhibited their expression. This might be due to the fact that podocytes are terminally differentiated cells, and their pathological changes under DN conditions differ fundamentally from those of tumor cells. We also revealed that SIRPα regulates NF-κB activation by controlling PKM2 nuclear translocation. The activation of NF-κB by nuclear PKM2 has been reported in tumor cells and cancer-associated fibroblasts[[217]53] [[218][53], [219][54]]. However, the underlying mechanism remains unclear, although Gu et al. [[220]55] reported that nuclear PKM2 prolongs the retention of NF-κB p65 in the nucleus. Moreover, podocytes themselves share many innate and acquired immune features[[221]29] [[222][29], [223][56], [224][57]] [[225][29], [226][56], [227][57]]. Therefore, the decrease in podocyte SIRPα in DN might represent a critical link between metabolic imbalance and inflammation activation. It is well-established that the dimeric form of PKM2 has low enzymatic activity and cannot generate pyruvate at a normal rate, leading to an accumulation of upstream glycolytic intermediates [[228]30]. Thus, under DN conditions, SIRPα reduction-induced dimerization and nuclear translocation of PKM2, along with the excessive glucose utilization, may result in the accumulation of numerous by-products [[229]52]. Multiple studies highlight the importance of PKM2 in regulating podocyte metabolism to maintain structure and function[[230]4] [[231][4], [232][30], [233][58]] [[234][4], [235][30], [236][58]], as well as the significance of PKM2's pyruvate kinase activity in DN [[237]59]. However, the regulatory mechanisms of PKM2 pyruvate kinase activity in DN remain unclear. Our findings indicate that the reduced enzymatic activity of PKM2 in podocytes under DN conditions is due to downregulated SIRPα expression, which promotes PKM2 nuclear translocation. In our study, both SIRPα knockout and shikonin treatment promoted PKM2 nuclear translocation without altering PKM2 expression levels, resulting in decreased cytoplasmic PKM2 and increased nuclear PKM2. This observation explains why PKM2 knockdown counteracted NF-κB pathway activation induced by SIRPα knockout without reducing overall or mitochondrial ROS production. Regarding ROS production, Yuan et al. reported that PKM2 knockdown in podocytes not only disrupts glycolysis but also impairs mitochondrial function, reducing ATP levels in podocytes [[238]60]. Nox4, a major ROS source in renal tissue, is primarily located in the mitochondria of the renal cortex [[239]61]. Shanmugasundaram et al. demonstrated that ATP directly binds to NOX4, negatively regulating NOX4 activity and ROS production [[240]62]. Thus, PKM2 knockdown reduces ATP production in podocytes, relieving ATP's inhibitory effect on NOX4, thereby enhancing ROS production and offsetting the effects of decreased NOX4 expression. It has been reported that in macrophages, nuclear PKM2 recognizes the HIF-1α and IL-1β promoters, contributing to inflammation[[241]63] [[242][63], [243][64]]. Yang et al. found that lipopolysaccharides enhance PKM2 binding to the STAT3 promoter, thereby promoting TNF-α and IL-1β expression in colorectal cancer cells [[244]65]. These findings suggest that nuclear PKM2 plays a crucial role in activating inflammatory responses. Consistent with these studies, our results indicate that PKM2 nuclear translocation induced by SIRPα deficiency is a significant factor in podocyte inflammation in DN. Specifically, we found that SIRPα reduction promotes PKM2 phosphorylation and pathological nuclear translocation, which regulates the transcription and expression of downstream genes, including NOX4, IL-6, and IL-1β, via PKM2's protein kinase and co-transcriptional functions. NOX4, IL-1β, and IL-6 are also key players in cellular oxidative stress. For instance, IL-6 regulates mitophagy and impacts mitochondrial function [[245]66], while IL-1β can increase cellular ROS by upregulating NOX4 expression [[246]67]. NOX4 is particularly associated with mitochondrial function and oxidative stress[[247]68] [[248][68], [249][69]]. Karen B. et al. demonstrated that NOX4 is primarily localized in renal cortex mitochondria in diabetic mice [[250]61], where it modulates cellular redox states. Elevated NOX4 expression leads to mitochondrial dysfunction and increased mitophagy [[251]70]. Our findings suggest that high NOX4 expression in diabetic podocytes is a major contributor to ROS production, promoting inflammatory activation and oxidative stress in podocytes. This aligns with reported roles of NOX4, IL-6, and IL-1β in DN progression [[252][71], [253][72], [254][73], [255][74]]. Targeting PKM2 enzyme activity has been shown to regulate renal function [[256]30,[257]58,[258]75]. Interestingly, both the PKM2 inhibitor shikonin [[259]76,[260]77] and the PKM2 activator TEPP-46 [[261]78] exhibit protective effects on the kidneys. This may be because these regulators have functions beyond directly modulating PKM2 enzyme activity, or their regulatory roles may differ depending on cell type and environmental conditions. Shikonin, a natural drug extract, exhibits anti-inflammatory and antioxidant properties by modulating multiple molecular pathways. For instance, it selectively inhibits the activation of NLRP3 and AIM2 inflammasomes[[262]79] [[263][79], [264][80]], reduces oxidative stress and inflammation in tubular cells by downregulating the NOX4/PTEN pathway, and decreases the expression of downstream pro-inflammatory cytokines IL-1β and IL-6 [[265]81]. Additionally, shikonin can inhibit chloride channel proteins [[266]20], potentially interfering with chloride-related IL-1β production and secretion pathways, which subsequently suppresses IL-6 expression [[267]82]. Moreover, it prevents the nuclear translocation of PKM2, enhances glycolysis, and promotes PKM2 tetramer formation in neuronal cells [[268]45]. We found that shikonin reverses PKM2 nuclear translocation, thereby restoring mitochondrial function and promoting podocyte glycolysis. Our study identified a novel function of shikonin, distinguishing it from a mere enzyme activity inhibitor: it regulates the subcellular localization of PKM2 in podocytes. Whether shikonin directly modulates PKM2 enzyme activity or exerts its effects through regulating PKM2 nuclear translocation may depend on the specific cell type and extracellular environment. Therefore, our study offers new insights into the anti-inflammatory mechanisms of shikonin in DN. In summary, reduced SIRPα levels in DN podocytes stimulate PKM2 phosphorylation, which subsequently forms a positive feedback loop involving PKM2 nuclear translocation, NF-κB activation, and oxidative stress. This loop promotes mitochondrial dysfunction and inhibits glycolytic flux. Shikonin disrupts the PKM2 nuclear translocation induced by SIRPα deficiency. SIRPα serves as a crucial molecule linking oxidative stress, inflammatory activation, and glycolysis in podocytes, and it may represent a potential therapeutic target for modulating PKM2 nuclear translocation. CRediT authorship contribution statement Yang Chen: Writing – original draft, Visualization, Validation, Project administration, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Mingchao Zhang: Visualization, Validation, Investigation, Formal analysis, Data curation. Ruoyu Jia: Validation, Data curation. Bin Qian: Visualization, Formal analysis. Chenyang Jing: Data curation. Caihong Zeng: Resources. Dihan Zhu: Writing – review & editing. Zhihong Liu: Writing – review & editing, Supervision. Ke Zen: Writing – review & editing, Supervision, Conceptualization. Limin Li: Writing – original draft, Investigation, Funding acquisition, Data curation, Conceptualization. Funding This work was supported by grants from the National Natural Science Foundation of China (32170897). Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Limin Li reports financial support was provided by the National Natural Science Foundation of China (32170897). Limin Li reports a relationship with the National Natural Science Foundation of China (32170897) that includes: funding grants. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Footnotes ^Appendix A Supplementary data to this article can be found online at [269]https://doi.org/10.1016/j.redox.2024.103439. Contributor Information Zhihong Liu, Email: liuzhihong@nju.edu.cn. Ke Zen, Email: kzen@nju.edu.cn. Limin Li, Email: liminli@cpu.edu.cn. Appendix A. Supplementary data The following are the Supplementary data to this article. Multimedia component 1 [270]mmc1.pdf^ (2MB, pdf) Multimedia component 2 [271]mmc2.docx^ (5.3MB, docx) Data availability Data will be made available on request. References