Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2022 Dec 14;17(12):e0279010. doi: 10.1371/journal.pone.0279010

Aspirin mediates protection from diabetic kidney disease by inducing ferroptosis inhibition

Ziyu Wu 1,2, Dan Li 1, Dingyuan Tian 1, Xuejun Liu 1,*, Zhongming Wu 1,3,*
Editor: Aldrin V Gomes4
PMCID: PMC9749971  PMID: 36516169

Abstract

Diabetic kidney disease (DKD) progression can be predicted by abnormalities in the tubulointerstitial lining, and their treatment may be useful for preventing the disease. DKD is a progressive disease that contributes to renal tubular cell death, but its underlying mechanisms remain unclear. Ferroptosis is a novel term linked to lipid hydroperoxidation, and it plays an important role in the pathogenesis of DKD. Overexpression of cyclooxygenase-2 (COX2), an enzyme of the proximal tubule, causes cellular redox damage in DKD. It remains unknown whether COX2 exacerbates tubular damage by accelerating ferroptosis in the kidneys of diabetic mice. HK-2 cells cultured in high glucose exhibited ferroptosis, which was inhibited by ferroptosis inhibitors. Additionally, alterations in the sensors of ferroptosis metabolism, such as glutathione peroxidase 4 (GPX4) activity, lipid hydroperoxidation, reduced glutathione (GSH) levels and changes in mitochondrial morphology, were observed in high glucose-cultured HK-2 cells. Diabetic mice manifested tubular injury and deranged renal physiological indices, which were mitigated by ferrostatin-1 (Fer-1). Importantly, these perturbations were ameliorated by downregulating COX2. In addition, the increased COX2 was observed to be elevated in the daibetic kindney. To explore the relevance of COX2 to ferroptosis, HK-2 cells that knocked down from COX2 exhibited decreased ferroptosis sensitivity under high glucose conditions. In RSL-3-treated HK-2 cells, ferroptosis was improved by downregulating COX2 by treatment with aspirin, which was confirmed in high glucose-cultured HK-2 cells. Furthermore, the ferroptosis changes were also suppressed by decreasing COX2 in diabetic mice treated with aspirin, which retarded DKD progression. In conclusion, our results demonstrated that ferroptosis in renal tubular cells contributes to DKD development and that diabetes-related ferroptosis was inhibited through the downregulation of COX2 by aspirin, thus retarding the progression of DKD. Our findings support a renoprotective mechanism by which aspirin inhibits COX2 activation, identify COX2 as a potential target of ferroptosis, and establish that ferroptosis in renal tubular cells is an integral process in the pathogenesis of DKD regulated by COX2 expression profiles.

1. Introduction

Diabetic kidney disease (DKD) is one of the leading causes of end-stage renal disease (ESRD) and is epidemic around the world. It is estimated that the total diabetes prevalence will reach 33% of the US adult population by 2050 [1]. DKD is characterized by proteinuria and is generally attributed to glomerular abnormalities [2]. It has been reported that blocking the renin-angiotensin system can retard the progression of kidney disease and supplies partial renoprotection. However, approximately 30% of patients with diabetes on angiotensin receptor blockers (ARBs) or angiotensin-converting enzyme inhibitors (ACEi) progress to ESRD [3], and there is a large unmet need to develop more effective treatments for DKD patients. Although the exact pathogenesis of DKD is still unresolved, ferroptosis-related renal tubular injury is suggested to be associated with DKD. Therefore, in addition to developing additional therapies to lessen proteinuria, protecting proximal tubular epithelial cells (PTECs) from ferroptosis may provide an approach to ameliorate outcomes in patients with DKD.

Tubular cell death is the major early step in the pathogenesis of DKD and is accompanied by inflammation induced by damage-associated molecular patterns and chemokines of dead and dying cells. Altered tubular cell death signals are also implicated during DKD. The best parameter of kidney disease functional progression is tubulointerstitial disease [4]. Tubular abnormalities may precede the onset of early glomerulopathy in DKD [5]. The pathogenesis of DKD, which may initiate tubular epithelial degeneration and injury, is still poorly understood. Recently, the renoprotective effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors have confirmed PTECs as a target of therapy [6]. Many factors, including ischemia, hypoxia, mechanical stretch, ROS and inflammatory cytokines, can cause tubular cell death [710]. However, the relationship between fibrosis and tubular cell death during the progression of DKD is still unclear. Further exploring the pathogenesis of tubulointerstitial disease will provide new treatment strategies for DKD.

Ferroptosis is a novel nonapoptotic cell death that was discovered to be iron-dependent in 2012 and induced by redox imbalance redox homeostasis, which is characterized by detoxification of lipid oxidation products and free radicals [11]. It is caused by the loss of antioxidant capacity, redox-active iron accumulation and intracellular lipid hydroperoxide accumulation[12]. The pathogenesis of ferroptosis can be inhibited by glutathione peroxidase 4 (GPX4) [13]. The products of lipid peroxidation involve reactive aldehydes (e.g., 4-hydroxynonenal (4HNE) and malondialdehyde (MDA)), which increase during ferroptosis [14]. In the presence of ferroptosis, the process of System Xc- was disrupted, depleting sufficient GSH production to prevent GPX4 from exerting its antioxidant capacity [15,16]. Because renal tubular cells are enriched in polyunsaturated fatty acids (PUFAs) [17], the kidney is susceptible to ferroptosis induced by lipid hydroperoxides. It has been demonstrated that ferroptosis pathogenesis is the cause of folic acid-induced AKI and IRI-induced ATN [18,19], but its role in DKD remains to be investigated. These results suggest that inhibiting ferroptosis in injured PTECs may become a new treatment for kidney diseases.

Cyclooxygenase-2 (COX2) is a specific enzyme of the renal proximal tubule that was recognized several years ago [20]. COX2 has recently attracted greater attention because COX2 expression occurs early in the disease process by physiological stimuli or inflammatory cytokines [2124]. It catabolizes arachidonic acid to various prostanoids, including thromboxane and prostaglandins, and it plays an important role in renal tubular damage. COX2 is a cyclooxygenase superfamily protein that is an inducible enzyme at low levels in most tissues and is significantly increased in the proximal tubule of the injured kidney [25,26]. Moreover, upregulated COX2 expression also induces increased production of ROS and aggravates renal tubular damage in many pathological conditions [27]. However, its role in the pathogenesis of ferroptosis remains to be explored.

Aspirin is a nonspecific inhibitor of COX2 activation. Various recent clinical trials have indicated that aspirin lessens the risk of cardiovascular disease and improves renal disease progression in diabetes [28,29]. Notably, a clinical observational study demonstrated that aspirin markedly reduced proteinuria in DKD patients [30]. While the role of COX2 expression in aspirin-mediated cardioprotection is still controversial, its role in renoprotection remains unclear. Thus, showing the mechanism of aspirin-mediated renoprotection may supply a novel target for the pathogenesis of DKD. Studies have demonstrated that aspirin can protect cells from the production of thromboxane and reduce platelet hypersensitivity to inhibit the progression of diabetic complications, but there is still debate about its exact mechanism [31]. In addition, inhibited COX2 expression in tissues may be involved in aspirin-mediated renoprotection. However, the mechanism by which COX2 regulates ferroptosis has not been elucidated. We now show that aspirin can markedly lessen proteinuria and retard diabetic kidney disease progression by inhibiting the protein COX2. Because aspirin is a nonspecific inhibitor of COX2 activation, this research aimed to explore the underlying mechanisms of aspirin in the treatment of tubulointerstitial injury in diabetic mice. Therefore, we hypothesized that inhibiting ferroptosis prevents PTEC damage by downregulating COX2 expression signaling, and this mechanism may illustrate how aspirin mediates renoprotection.

Due to the presence of COX2 changes and ferroptosis markers on the renal tubules, we explored the COX2 pathobiology and ferroptosis of tubulointerstitial injury in diabetic kidney disease. Our research shows that inhibiting ferroptosis has renoprotective properties by downregulating COX2 signaling in PTECs, which illustrates the mechanism of aspirin-mediated renoprotection in DKD. This study is expected to supply novel avenues for treating DKD.

2. Material and methods

2.1. Cell culture studies

In a humidified environment, human proximal tubular epithelial cell line (HK-2) cells were cultured with 5% CO2 at 37°C. They were maintained in Dulbecco’s Modified Eagle Media (DMEM; Gibco, Carlsbad, CA, USA), consisting of 10% fetal bovine serum (FBS; Biological Industries, Cromwell, CT, USA) and 100 U/mL streptomycin and penicillin (Solarbio, Beijing, China) in Ctrl group. In HG group, cells were maintained in DMEM containing 30 mmol/L glucose supplemented with100 U/mL streptomycin and penicillin and 10% FBS. In Mannitol group, cells were maintained in DMEM with 5.5 mmol/L glucose, 24.5 mmol/L mannitol, 100 U/mL streptomycin and penicillin and 10% FBS. Mannitol group was applied to control cell osmolality. Ferrostatin-1 (Fer-1, Selleck, Houston, TX, USA, 400nM) and Aspirin (Solarbio, Beijing, China, 400uM) were used to treat the cells. In additional, COX2-siRNA (Sangon Bitotech, Cat. SR310776, 50 nM) in cells transfected with Lipofectamine 2000 regent (US Everbright Inc, Cat.L7003) was used to knockdown studies. In Fer-1 group, AS group and DMSO group, cells were respectively cultured in 30 mmol/L glucose medium supplemented with 400nM Fer-1, 400 μM Aspirin and 0.1% DMSO for 48 h. In Si-COX2 group, cells were respectively treated with COX2-siRNA transfections and cultured in 5.5 mmol/L glucose or 30 mmol/L glucose medium for 48 h. In RSL-3 group, RSL-3 + AS group and DMSO group, cells were respectively cultured in 5.5 mmol/L glucose medium supplemented with 100nM RSL-3, 100nM RSL-3 plus 400 μM aspirin and 0.1% DMSO for 48 h. And in Si-COX2 + RSL-3 group and Si-COX2 + DMSO group, cells were treated with COX2-siRNA transfections and maintained in 5.5 mmol/L glucose medium supplemented with 100nM RSL-3 and 0.1% DMSO for 48 h.

2.2. Animal model system

All animal research experimental protocols were conducted adhered to the National Institutes of Health Guide for the Care and Use of Laboratory Animals. The ethical committee of Tianjin Medical University approved this study. These mice were on eight weeks old male DBA/2J background (n = 36, HFK Bioscience, Beijing, China). They were randomized one of the six groups: control normal mice group (NC); diabetic mice group (DM); diabetic mice group (Fer-1),who intraperitoneal injected Fer-1 (Selleck, Houston, TX, USA); diabetic mice group (vehicle-P),who intraperitoneal injected 1% dimethyl sulfoxide (DMSO); diabetic mice group (As),who intragastric administrated Aspirin (Solarbio, Beijing, China); diabetic mice group (vehicle-G),who intragastric administrated 0.5% sodium carboxymethyl cellulose (Na-CMC; Solarbio, Beijing, China). Diabetes models were induced with 5 consecutive days of a single intraperitoneal injection of streptozotocin 40 mg/kg (dissolved in 0.1 M citrate buffer, pH 4.5; SigmaAldrich, St Louis, MO, USA). Control mice only was injected the same volume of citrate buffer. In the Fer-1 or vehicle-P groups, the diabetic mice were treated respectively with Fer-1 (2.5 μmol/kg, dissolved in 1% DMSO) or 1% DMSO during the duration of treatment for 12-week every day. And in the AS and vehicle-G groups, the diabetic mice were treated respectively with aspirin (50 mg/kg, dissolved in 0.5% Na-CMC) or 0.5% Na-CMC for 12-week every day. Plasma glucose were measured from tail vein blood. Mice with random plasma glucose concentrations >300 mg/dL (16.7 mmol/L) for 3 days after the last injection of streptozotocin were used for further study. All mice (3–5 mice per cage) were exposed to standard laboratory conditions (12 h on/off; lights on at 7:00 a.m.) and were housed at 10% humidity with an ambient temperature (22–24°C) ad free access to food and water. Mice body weight and blood glucose levels were measured at week 0, 2, 4, 8 and 12. Mice were respectively housed in metabolic cages for 24 h, and urine samples were gathered and urine output was recorded at week 0 and 12. Urinary creatinine concentrations and Urinary albumin were assayed by the Mouse Cr ELISA Kit (Fankewei, Shanghai, China) and Mouse uALb ELISA Kit (Fankewei, Shanghai, China). Then urinary albumin/creatinine ratio (ACR) and urinary albumin were calculated. All mice were euthanized with 60 mg/ml of pentobarbital sodium at the end of the experimental period. Kidneys were harvested at the time of sacrifice and stored for examination.

2.3. Morphological evaluation and tubular damage scoring

Paraffin-embedded tissue sections (4 μm thick) were heated and deparaffinized with xylene. The sections were rehydrated by gradually decreasing ethanol concentrations. Following the slides were stained sequentially by Hematoxylin (3 min) and Eosin (30 sec). After distilled water rinsed a short, the slides were dehydrated by ethanol, cleared in xylene, mounted with covers lip and assessed. The slides were mounted with neutral balsam at last. The degree of renal injury, including renal tubular injury, mesangial matrix expansion and interstitial fibrosis was analyzed by Image-Pro Plus 6.0 software. HE-stained sections were applied to evaluate disruption of tubular epithelium and dilation of tubule. For each mice, two investigators assessed100 tubules from 10 different views. Seven parameters were graded as follows: brush border loss(0 or 1),flattening of tubular epithelial cell (0 or 1), vacuolization of cytoplasm (0 or 1), bleb formation of cell membrane (0 or 2), obstruction of tubular lumen (0 or 2), edema of interstitium (0 or 1), and cell necrosis (0 or 2). Cumulative values in the range 0 (unaffected) - 10 (severely damaged) were considered an indication of tubular damage extent. For Periodic acid-Schiff’s (PAS), dehydrated slides were incubated with Periodic Acid Solution for 10 min. Then distilled water washed and Schiff Solution immersed in the slides for 30 min. Following distilled water rinsing, Hematoxylin stained the slides for 3 min. After distilled water washing again, Bluing Reagent TM was used for 30 sec, then slides were rewashed, dehydrated, cleared in xylene and mounted with covers lip. The slides were assessed for the mesangial matrix expansion by optical microscopy. Measuring the relative number of pixels (pink or red areas) divided by the total area of the glomerulus to evaluate semi-quantitatively Mesangial matrix expansion. Randomly selected 20 stained glomeruli each slide (3 mice/group) were analyzed. The extent of renal fibrotic areas were assessedby Masson staining with estimating the relative number of pixels (blue).

2.4. Immunohistochemical (IHC) studies

Paraffin tissue sections of kidneys(4 μm thick) were deparaffinized and rehydrated. Following heat-induced epitope retrieval, phosphate buffer saline (PBS) washed the sections for 3 times. To block the background staining, all sections were incubated with 3% BSA in PBS at room temperature for 1 hour. The sections are immersed respectively in COX2 antibody (A1253; ABclonal;1:100),GPX4 antibody (A11243;ABclonal,1:100), FTH-1 antibody (DF6278; Affinity;1:100), and SLC7A11 antibody(DF12509;Affinity;1:100) overnight at 4°C and washed with PBS. HRP conjugated secondary antibody (Vector Laboratories) were incubated in the sections at room temperature for 1 hour. Then rinsing with PBS, to develop the peroxidase reaction, productdiamino-benzidine (DAB) solution was used for 20–35 seconds. All sections were following washed immediately with distilled water and counter stained with Hematoxylin, dehydrated in graded series of ethanols, mounted with covers lip and assessed by optical microscopy.

2.5. Immunofluorescence (IMF) studies

Variously treated HK-2 cells were fixed in 4% paraformaldehyde. Then they were permeabilized by 0.25% Triton X-100 in PBS buffer. 2% BSA in PBST was appplied to block the background staining. After primary antibodies were used to incubated with the cells overnight at 4°C: GPX4 antibody (A11243; ABclonal,1:100). Then the cells were rinsed with PBS and the secondary antibodies goat anti-rabbit IgG were incubated for 1 h and counter stained with 4,6-diamidino-2-phenylindole (DAPI; Solarbio, Beijing, China) at the room temperature. Subsequently, being rinsed with PBS for 3 times, the cells were then mounted with covers lip and observed by fluorescence microscopy.

2.6. Transmission electron microscopy of cells

HK-2 cells pellets were fixed with 2.5% glutaraldehyde (Alfa Aesar, Ward Hill, MA, USA) in 0.1 M phosphate buffer saline (pH 7.4) at 4°C for 3 h. After incubated with 1% aqueous osmium tetraoxide and 0.1 M phosphate buffer saline (pH 7.4) at room temperature for 2 h, dehydrated in graded series of ethanols (50%–100%), embedded in epoxy resin monomer and cured at 60°C for48 h. Ultra-thin sections (50 nm) were stained with uranyl acetate and lead citrate. Two samples were prepared for each group, and each sample were randomly selected five fields of view and observed by transmission electron microscopy (HT7700-SS; HITACHI, Tokyo, Japan).

2.7. Western blotting analysis

Immuno-blotting were applied to assess various samples protein expression. The tissues were diced into 1 mm3 and homogenized in RIPA lysis buffer (Solarbio, Beijing, China). Furthermore the HK-2 cells were lysed with RIPA lysis buffer. Centrifuge the homogenate at 10,000ⅹg for 5 minutes and collect the supernatant. After each of the samples were adjusted the concentration (100 μg/100 μl), the SDS loading buffer were mixed with equal amounts of protein (20 μg), heated for 10 min at 100°C, followed by centrifugation and vortexing to remove undissolved fragment, ice-cooled and subjected to 7.5,10 or 12.5% SDS-PAGE. After the proteins was separated on SDS-PAGE, they were transferred to PVDF membranes by electro-blotting procedures. Following the membranes were blocked with 0.1% tris buffered saline tween that contained 5% skimmed milk at room temperature for 1 h, members were individually incubated with each primary antibodies overnight at 4°C: GPX4 antibody (A11243;ABclonal,1:1000), FTH-1 antibody (DF6278; Affinity;1:1000), and SLC7A11 antibody(DF12509;Affinity;1:1000), TFR-1 antibody (DF214039;Affinity; 1:1,000), COX2 antibody (A1253; ABclonal;1:1000), and GAPDH antibody (60004-1-Ig;Proteintech;1:5,000). They were rinsed with TBST buffer and incubated with the normal isotype-matched horseradish peroxide-labeled IgG (1:1000) for 60 min at room temperature. Following another wash with TBST, the ECL chemiluminescence reagent (Amersham Biosciences) was applied to visualize the immunoblots, and densitometry was performed by Image J software v1.49.

2.8. Quantitative real-time PCR (qRT-PCR)

The gene expression in various samples were evaluated by Real-time PCR. HK-2 cells and 10 mg of kidney tissue were used to isolate RNA by using the HP Total RNA Kit (Omega Bio-Tek, Norcross, GA, USA). The RNA was reverse transcribed into cDNA using M-MuLV First Strand cDNA Synthesis Kit (Sangon Biotech, Shanghai, China). For quantitative PCR (qPCR), reaction mixture, containing 1 μmol/L of forward and reverse primer,100 ng cDNA, 2 μl of nuclease-free water and 1 ⅹ Fast SYBR Green Master Mix, in a total volume of 20 μl was prepared. The C1000 Touch Thermocycler CFX96 Real-Time System (Bio-Rad, Hercules, CA, USA) was performed to PCR. The mRNA levels of different samples was calculate byRelative CT values compared with GAPDH. The primers used in the research were listed in S1 Table.

2.9. Transfections

The cells were transfected with 50 nM of siRNA (Sangon Bitotech, Shanghai, China) individually against COX2(Si-COX2) and control siRNA (Si-Ctrl), performing with lipofectamine RNAimax (US Everbright Inc, Cat.L7003) to generate COX2 knockdown studies. Target sequences for preparing siRNAs of human COX2 are listed in S1 Table. 250 μL of Opti-MEM serum-free media (Gibco, Carlsbad, CA, USA)was combined with 5 μL of 20 μM siRNAs in one tube. 250 μL of Opti-Mem media was combined with 5 μL of lipofectamine 2000 in another tube. They were equilibrated for 5 min at regular temperature. Two tubes were gently mixed and incubated for 20 min at 37°C, transferred into 6-well plate combined with 1.5 mL of Opti-MEM serum-free media. Performing Quantitative real-time PCR to confirm the siRNA induced gene silencing.

2.10. Assessment of intracellular ROS in HK-2 cells

2′,7′ -dichlorofluorescin diacetate (DCHF-DA, Solarbio, Beijing, China) was applied to evaluate intracellular ROS level. The cells were stained with DCHF-DA at 37°C for 30 min. we rinsed excess dye out with phosphate buffer saline in the dark. Following fluorescence intensity of cells were observed by fluorescence microscopy with excitation at 488 nm and emission at 525 nm.

2.11. GSH levels of kidney tissues and cells assay

About 20mg kidney tissues or ~2 x 106 cells with 90–95% confluency per culture dish (55 cm2) were applied to evaluate the GSH levels. The tissues were homogenized with 200 μl GSH Extraction Buffer. Besides the cells were lysed in 50 μl GSH Extraction Buffer. Then the samples were shortly centrifuged and vortexed at 8,000 x g for 10 min. 20 μl upernatants or standards samples were added into the respective wells of 96 well-plate. GSH was detected by Micro Reduced Glutathione GSH Assay Kit (Solarbio,Beijing, China).When the reaction period was over, colorimetric reading were made by using a Microplate Reader (BIO-RAD) at the wavelength of 412 nm. At last, the GSH levels of the samples was calculated by comparing the optical density of the samples to the standard curve.

2.12. Iron assay

The iron levels were detected by the Iron Assay Kit (BioAssay, Hayward, CA, USA), transfer the working reagents and collected supernatant to a 96-well plate. Flow cytometric reading was made at the wavelength of 590 nm.

2.13. CCK8 assay

According to the manufacturer’s instructions, the viability of cell was detected by applying the Cell Counting Kit-8 (Dojindo, Kumamoto, Japan). HK-2 cells were seeded at a density of 3000 cells per well into the 96-well plate (~3,000 cells/well). The cells were administrated with different concentrations of the compounds to indicate times. After treatment, the growth medium was removed and cells were cultured with 10 μL work reagent at each well and incubated at 37°C for 2 h. A microplate reader (Synergy HT, Bio-Tek, United States) detected the absorbance at 450 nm. Orbitally shake the 96-well plate for 10 min to completely dissolve the formazan. Then measure the absorbance at 490 nm by a microplate reader (Synergy HT, Bio-Tek, United States).

2.14. MDA and 4-HNE assay

10 mg tissues of kidney were sliced and rinsed with cold saline in each group. Immediately homogenize the tissue by shaking the homogenizer with cold saline. Otherwise, the ultrasonic cell disrupter homogenized the cells. To detect the MDA concentrations of tissues and cells,the supernatant was collected after centrifugation for 10 min. The concentrations of MDA were measured by Micro Malondialdehyde Assay Kit (Solarbio, Beijing, China). Then transfer the working reagent and collect the supernatant into a 96-well plate. To calculate the MDA concentration,the optical density was detected at the wavelength of 600, 532 and 450 nm. After tissues of kidney were homogenized, the supernatant was collected after centrifugation. In addition, the ultrasonic cell disrupter homogenized the cells. 4-HNE concentrations was measured by 4-HNE ELISA Kit (Fankewei, Shanghai, China). Microplate reader detected the optical density of the supernatant at the wavelength of 450 nm. And 4-HNE concentrations of the supernatant were then obtained by comparing the optical density of the samples to the standard curve.

2.15. Statistical analysis

Student’s t-test was used for the difference analysis of two groups, and one-way ANOVA analysis was performed for multiple group comparison. p less than 0.05 was considered significant. Data were shown as mean ±SD. GraphPad Prism 8.0 software (GraphPad, La Jolla, CA, USA) and Microsoft Excel 2013 were used for all calculations.

3. Results

3.1 Ferroptosis is an essential part of high glucose-cultured HK-2 cells

Under treatment with various media with different glucose concentrations (5.5, 10, 15, 20, 25, 30, 35, 40, 45 and 50 mmol/L) for 48 h, the viability of the cells was measured by CCK8 assay. HK-2 cells cultivated in medium containing 30 mmol/L glucose had a higher viability than those cultivated in medium containing 5.5 mmol/L glucose (Fig 1A). DCHF-DA staining (2’,7’-dichlorofluorescein diacetate) showed that high glucose-cultured HK-2 cells generated more ROS than the control cells. The Ctrl group cells exhibited a minimal degree of fluorescence, and cells cultured in high glucose alone generated the greatest background fluorescence. The generation of ROS was significantly reduced by cotreatment with 400 nm Fer-1 (Fig 1B). To highlight the role of ferroptosis, our study observed the dynamics of intracellular iron, i.e., ferrum and antioxidant stress-related protein, i.e., solute carrier family 7 member (SLC7A11), glutathione peroxidase 4 (GPX4) with iron metabolism-related protein, i.e., heavy chain of ferritin (FTH1), transferrin receptor 1 (TFR-1). Western blot analysis was applied to indicate that COX2 and TFR-1 protein expression was increased, and the expression of SLC7A11, GPX4 and FTH-1 were decreased in HG group cells compared with the Ctrl group (Fig 1C). Additionally, quantitative real-time PCR results revealed that the mRNA expression levels of GPX4, SLC7A11, FTH-1 and TFR-1 and the corresponding proteins were changed in parallel. Thus, the changes in ferroptosis-related genes were reversed in the Fer-1 group (Fig 1D–1G). The intracellular GSH levels were assayed by a Micro Reduced Glutathione GSH Assay Kit. The research demonstrated that GSH was also clearly decreased in HG group cells (Fig 1H). Simultaneously, the data showed the end products of lipid peroxidation; a micro malondialdehyde assay kit was used to assess the level of malondialdehyde (MDA), and the production of MDA was increased by 50% (Fig 1I) compared with that in the Ctrl group. Another products of lipid peroxidation, 4-HNE(Fig 1J) was dramatically elevated in HG group cells. Furthermore, the concentration of iron (Fig 1K) was increased by 80% in HG group cells compared to Ctrl group cells. However, Fer-1 significantly inhibited the production of MDA and enhanced the intracellular GSH levels and iron concentration in high glucose-cultured HK-2 cells. Thus, the above mentioned findings demonstrated that the pathogenesis of ferroptosis was caused by high glucose in vitro, and we further explored its biology in vivo.

Fig 1. Ferroptosis is a critical component of high glucose-cultured HK-2 cells.

Fig 1

(A) CCK8 was used to detect the viability of HK-2 cells cultured in different concentrations of glucose medium for 48 h. (B) DCHF-DA staining revealed the generation of ROS (green) in high glucose-cultured HK-2 cells compared to the control cells, which was partially reduced by Fer-1 treatment for 48 h. (C) Western blot analysis of GPX4, SLC7A11, FTH-1, COX2 and TFR-1 protein expression in cells in the Ctrl and HG groups. (D-G) GPX4 (D), SLC7A11 (E), FTH-1 (F) and TFR-1 (G) mRNA levels in cells in the Ctrl, HG, Fer-1, DMSO and mannitol groups.(H-K) GSH (H), MDA (I),4-HNE(J) and iron concentrations (K) of cells in the Ctrl, HG, Fer-1, DMSO and mannitol groups. Data are expressed as the mean ± SD. **p < 0.05 vs. Ctrl group; ***p < 0.05 vs. HG group.

3.2 Ferroptosis inhibition attenuates the renal pathological changes of kidney fibrosis in diabetic kidney disease

We investigated whether ferroptosis inhibition can have the renoprotective effects of Fer-1 treatments in DKD. To evaluate the effects of Fer-1 on an experimental model of streptozotocin (STZ)-induced type 1 diabetes, five consecutive injections of low-dose streptozotocin (STZ) (40 mg/kg/day intraperitoneally (IP)) in 8-week-old male DBA/2J mice resulted in the development of diabetes (Fig 2A). To elucidate the pathogenesis of ferroptosis in diabetic kidney disease, diabetic mice were treated with Fer-1 for 12 weeks. All mice were monitored after STZ injection for 3 months before sacrifice. The body weight was gently increased in the NC group, while it was progressively decreased in the Fer-1, DM and vehicle-P groups (Fig 2B). Meanwhile, random blood glucose progressively increased and attained 25 mmol/L in the Fer-1, DM and vehicle-P groups at week 12, but it was retained at approximately 7 mmol/L in the NC group (Fig 2C). To evaluate renal function, the renal dysfunction of diabetic mice was defined by high levels of the 24-h urine volume (Fig 2D), urinary albuminuria (Fig 2E) and ACRs (Fig 2F) in the Fer-1, DM and Vehicle-P groups. In particular, compared with the normal mice, the 24-h urine volume and urinary albuminuria were elevated by 30 and 100 times in the DM group. Diabetic mice had significantly higher kidney weights than NC mice (Fig 2G). These four quotas of the mice in the Fer-1 group were higher than those of the NC group, while they were clearly lower than those indicators of the DM group. HE staining showed changes in renal morphology, including tubular epithelial degeneration and brush border loss, with dilatation of the tubular lumen in diabetic mice. These tubular injuries were clearly attenuated by Fer-1 treatment (Fig 2H). PAS staining revealed clear mesangial expansion and a loss of brush borders with more dilated cortical proximal tubules and numerous exfoliated tubular cells compared with the Ctrl group (Fig 2H). Masson staining revealed basement membrane atrophy, cortical proximal tubule thickening, and relative collagen deposition in the lumen of the DM group. Moreover, this renal damage was remarkably reversed in diabetic mice treated with Fer-1 (Fig 2H). Of note, the scores of tubular damage and KIM-1 and NGAL mRNA were increased in the kidneys of diabetic mice. These changes were alleviated by Fer-1 (Fig 2I and 2J), demonstrating that notable renal functional deterioration induced by high glucose is attenuated by the inhibition of ferroptosis.

Fig 2. Inhibition of ferroptosis by Fer-1 ameliorated the pathological changes of kidney fibrosis in diabetic kidney disease.

Fig 2

(A) Schematic diagram. Five doses of STZ (40 mg/kg/day IP) were injected to show the induction of diabetes in DM and Fer-1 mice. (B-G) Body weight (B), blood glucose (C), 24-h urine volume (D), urinary albuminuria (E), albumin-to-creatinine ratio ACR (F) and kidney weight/body weight (G) were monitored in the NC, Fer-1, DM and vehicle-P groups. (H) HE staining, PAS staining and Masson staining of kidneys in nondiabetic and diabetic control Fer-1 mice. The relative area of fibrosis (%) and relative collagen (%) were assessed by the ImageJ program. (I-J) The increased KIM-1 and NGAL mRNA expression was relieved by Fer-1 in diabetic mice. Data are the mean ± SEM. **p < 0.05 vs. NC group; ***p < 0.05 vs. DM group.

3.3 Renal ferroptosis and COX2 expression are observed in diabetic mice

It is believed that both SLC7A11 and GPX4 are involved in antioxidative stress. In this research, SLC7A11 and GPX4 were expressed at lower levels in the renal proximal tubules of diabetic mice by immunohistochemical staining and semiquantification analyses (Fig 3A). The protein and mRNA expression levels of SLC7A11 and GPX4 in the DM group were clearly decreased compared with those in the NC group (Fig 3B–3D). This study also showed that the iron metabolism-related genes TFR-1 and FTH-1 were changed in diabetic mice. FTH-1 mRNA expression was dramatically decreased, and TFR-1 and PTGS2 were significantly increased in diabetic mice (Fig 3E–3G). Meanwhile, immunohistochemical staining showed that FTH-1 staining was weaker and COX2 staining was stronger in renal sections of the DM group than in the control group (Fig 3A), which was consistent with the corresponding protein expression changes (Fig 3B). The obtained data showed that GSH was also clearly decreased in the kidneys of diabetic mice (Fig 3H), indicating that the capacity for peroxidation repair was significantly reduced. Moreover, the obtained results showed that MDA (Fig 3I) and 4-HNE (Fig 3J), which are the end products of lipid peroxidation, were dramatically elevated in diabetic mice. Furthermore, the iron concentration in the kidneys of diabetic mice was approximately four times higher than that of normal mice, demonstrating iron overload in the kidneys of the DM group (Fig 3K). The decreased GSH levels were improved, and the increased kidney iron content and MDA and 4-HNE levels were blocked by Fer-1 treatment in diabetic mice. Interestingly, these results illustrated that the abovementioned ferroptosis-related changes were significantly reversed by the ferroptosis inhibitor Fer-1 in diabetic mice.

Fig 3. Ferroptosis-related changes in the kidney are observed in diabetic mice.

Fig 3

(A) Immunohistochemical staining results and semiquantification analyses of GPX4, SLC7A11, FTH-1 and COX2 in the kidneys of nondiabetic, diabetic control and diabetic treated with Fer-1 mice. (B) Western blot analysis and quantification of GPX4, SLC7A11, FTH-1, TFR-1 and COX2 in the kidneys of nondiabetic, diabetic and diabetic mice treated with Fer-1. (C-G) The mRNA expression levels of GPX4 (C), SLC7A11 (D), FTH-1 (E), TFR-1 (F) and PTGS2 (G) in kidney tissue in mice. (H-K) GSH (H), MDA (I), 4-HNE (J) and iron concentrations (K) were detected in the kidney tissue of mice. Data are the mean ± SEM. **p < 0.05 vs. NC group; ***p < 0.05 vs. DM group.

3.4 High glucose promotes ROS generation, and COX2 gene disruption alleviates high glucose-induced ferroptosis

We examined the effect of aspirin-mediated renoprotection in high glucose-cultured HK-2 cells treated with aspirin under high glucose conditions for 48 h. DCHF-DA staining was used to evaluate intracellular ROS generation. After being treated with high amounts of glucose for 48 h, HK-2 cells were significantly more likely to stain positive for DCHF-DA (green fluorescence). The high glucose-induced increase in DCHF-DA staining was partially weakened by aspirin treatment (Fig 4A). High expression of COX2 protein and mRNA was observed in cells cultured under 30 mmol/L glucose for 48 h. To explain the relevance of COX2 in ferroptosis in vitro, aspirin was used in the experiment. COX2 gene disruption was confirmed by Western blotting analysis and qRT‒PCR. After 48 h, COX2 expression considerably increased in high glucose-cultured HK-2 cells, which was attenuated by aspirin administration (Fig 4B). In cells cultured in high-glucose medium with aspirin, the protein expression of SLC7A11, GPX4, and FTH-1 was clearly upregulated, while TFR-1 was decreased compared to cells cultured in high-glucose medium (Fig 4B). These genes exhibited parallel mRNA expression (Fig 4C–4G). In high glucose-cultured HK-2 cells, both Fer-1 treatment and aspirin restored GPX4, SLC7A11, FTH-1 and TFR-1 expression. These results demonstrate that COX2 hyperactivation is associated with ferroptosis in high glucose-cultured HK-2 cells. Normally, untreated control cells contained only a minimal amount of MDA and 4-HNE. The cells treated with high glucose showed the maximal level compared to NC group cells. Interestingly, aspirin treatment markedly attenuated the level compared to that in HG group cells (Fig 4I and 4J). The obtained data showed that GSH was also clearly decreased and that the levels of iron were higher in HG group cells. However, the changes were ameliorated by Fer-1 and aspirin in high-glucose medium (Fig 4H and 4K). The abovementioned data showed that the inhibition of COX2 ameliorated the ferroptosis sensitivity of HK-2 cells under high glucose conditions. Moreover, the mRNA levels of KIM-1 and NGAL were significantly increased in high glucose-cultured HK-2 cells, and these changes were alleviated by Fer-1 and aspirin (Fig 4L and 4M).

Fig 4. High glucose-induced ferroptosis was alleviated by treatment with aspirin.

Fig 4

(A) DCHF-DA staining revealed ROS (green) generation in high glucose-cultured HK-2 cells compared to control cells, which was partially alleviated by AS treatment. (B) Western blot analysis of GPX4, SLC7A11, FTH-1, TFR-1 and COX2 protein expression in cells in the Ctrl, HG, Fer-1, DMSO and AS groups. (C-G) GPX4 (C), SLC7A11 (D), FTH-1 (E), TFR-1 (F) and PTGS2 (G) mRNA expression in cells in the Ctrl, HG and AS groups. (H-K) GSH (H), MDA (I),4-HNE(J) and iron concentrations (K) of cells in the Ctrl, HG and AS groups. (L-M) The KIM-1 (L) and NGAL (M) increased mRNA levels induced by high glucose were relieved by treatment with AS. Data are the mean ± SD. **p < 0.05 vs. Ctrl group; ***p < 0.05 vs. HG group.

3.5 Transfection of COX2-siRNA inhibits ferroptosis in high glucose-cultured HK-2 cells

Ferroptosis is a newly discovered iron-dependent form of nonapoptotic cell death; when GSH is depleted or GPX4 is inhibited, ferroptosis can be accelerated. To explore the underlying mechanism of ferroptosis induced by COX2, we examined the genes associated with ferroptosis. Immunofluorescence microscopy showed intense fluorescence correlated with GPX4 expression in control HK-2 cells. Of note, decreased fluorescence was detected in high glucose-cultured HK-2 cells compared to the controls. Interestingly, GPX4-related levels were partially restored following the transfection of COX2-siRNA in high glucose-cultured cells (Fig 5A). Furthermore, we examined the GSH concentration and GPX4 activity in HK-2 cells. Western blotting analysis showed that GPX4 expression was decreased in high glucose-cultured HK-2 cells (Fig 5B). Similarly, FTH1 was significantly decreased in high glucose-cultured HK-2 cells. This contention led to the investigation of FTH1 and GPX4 degradation in ferroptosis in HK-2 cells cultured in high glucose. However, COX2-siRNA treatment reversed the expression to levels similar to those in control cells (Fig 5B). The mRNA expression of these genes displayed parallel results, and they revealed that transfection of COX2-siRNA significantly affected FTH1 and GPX4 degradation in HK-2 cells after high glucose treatment (Fig 5C–5G). To explore whether COX2 is a target of ferroptosis, we detected the levels of GSH. The obtained results indicated that the GSH levels were decreased after high glucose treatment, which was partially restored by COX2-siRNA transfection. Understandably, this study has demonstrated that COX2 gene disruption can block such depletion of GSH (Fig 5H). Thus, higher levels of MDA and 4-HNE were also observed in high glucose-treated HK-2 cells, and this decrease was ameliorated in high glucose-cultured cells transfected with COX2-siRNA (Fig 5I and 5J). Ferroptosis and lipid hydroperoxidation require free iron, and the obtained data further showed that the intracellular free iron concentration was significantly increased in high glucose-cultured HK-2 cells, which was attenuated by COX2 gene disruption for 48 h (Fig 5K). Meanwhile, the mRNA levels of KIM-1 and NGAL increased in high glucose-cultured HK-2 cells. Furthermore, these changes were also attenuated by COX2-siRNA transfection (Fig 5L and 5M).

Fig 5. Knockdown of COX2 ameliorated the ferroptosis sensitivity of cells cultured in high glucose.

Fig 5

(A) Immunofluorescence analysis with GPX4 antibody (red) and nuclear staining with DAPI (blue) in the HK-2 cells of the Si-Ctrl group, Si-COX2 group, HG+Si-Ctrl group and HG+Si-COX2 group. (B) Western blotting showed the protein expression of COX2, GPX4, SLC7A11, FTH-1 and TFR-1 in the cells of each group. G) GPX4 (C), SLC7A11 (D), FTH-1 (E), TFR-1 (F) and PTGS2 (G) mRNA expression in cells in each group. (H-J) GSH (H), MDA (I), 4-HNE(J)and iron concentrations (K) of cells in each group. (K-L) KIM-1 (L) and NGAL (M) increased mRNA expression was alleviated by transfection of COX2-siRNA in high glucose. Data are expressed as the mean ± SD. **p < 0.05 vs. Si-Ctrl group; ***p < 0.05 vs. HG+Si-Ctrl group.

3.6 RSL-3-induced ferroptosis was ameliorated by Fer-1 and aspirin

It has been reported that RSL3 is a GPX4 inhibitor. We utilized RSL3 to cause ferroptosis in HK-2 cells in order to further explore the role of COX2 in ferroptosis. Compared with the Ctrl group, RSL-3-triggered ROS accumulation exhibited stronger fluorescence intensity, while the high degree of fluorescence was strongly alleviated by aspirin (Fig 6A). Furthermore, the viability of cells treated with RSL-3 was assessed by CCK8 assay. Our study showed that the RSL-3 concentration increased as fluorescence intensity was clearly decreased. When the RLS-3 concentration was over 0.1 μM, it clearly decreased. As HK-2 cells were treated with different concentrations of RSL-3 for 48 h, cell viability was partially improved by 400 nm Fer-1 or 400 μm aspirin treatment (Fig 6B). The cell viability measurements in HK-2 cells treated with RSL-3 100 nm for 48 h were partially improved by treatment with different concentrations of Fer-1 or aspirin (Fig 6C). Meanwhile, treatment with Fer-1 or aspirin inhibited the COX2-increased expression of RSL-3-treated cells (Fig 6D). Decreased mRNA expression of SLC7A11, GPX4 and FTH-1 and increased expression of PTGS2 and TFR-1 caused by ferroptosis were clearly reversed by Fer-1 or aspirin (Fig 6E–6I). In addition, our study showed that GSH was also clearly decreased in RSL-3-treated cells (Fig 6J). Moreover, lipid peroxidation product accumulation, MDA (Fig 6K), 4-HNE(Fig 6L)and concentrations of iron (Fig 6M) under ferroptosis conditions were also attenuated after Fer-1 or aspirin treatment. Otherwise, the mRNA levels of KIM-1 and NGAL increased in RSL-3-treated HK-2 cells. Interestingly, these changes were also mitigated by Fer-1 or aspirin (Fig 6N and 6O). Overall, these data demonstrated that aspirin could alleviate the pathogenesis of ferroptosis by downregulating COX2 expression.

Fig 6. Cellular ferroptosis induced by RSL-3 was alleviated by Fer-1 and aspirin.

Fig 6

(A)DCHF-DA staining revealed the generation of ROS (green) in the HK-2 cells cultured in 100 nmol/L RSL-3 medium compared to the control cells, and it was partially reduced by aspirin. (B) Cell viability was measured in HK-2 cells cultured with different concentrations of RSL-3 for 48 h, and it was partially improved by 400 nm Fer-1 or 400 μm aspirin treatment. (C) Cell viability measurements in HK-2 cells treated with RSL-3 100 nm for 48 h were partially improved by treatment with different concentrations of Fer-1 or AS. (D) Western blot analysis of SLC7A11, GPX4, FTH-1, TFR-1 and COX2 protein expression of cells in Ctrl, DMSO, RSL-3 100 nm, RSL-3 100 nm+Fer-1 400 nm, RSL-3 100 nm+AS400 μm and AS 400 μm groups. (E-I) GPX4 (E), SLC7A11 (F), FTH-1 (G), TFR-1 (H) and PTGS2 (I) mRNA expression in cells of each group. (J-M) GSH (J), MDA (K), 4-HNE(L)and iron concentrations (M) in the cells of each group. (N-O) The increased mRNA expression of KIM-1 (N) and NGAL (O) induced by RSL-3 was relieved by treatment with Fer-1 or aspirin. Data are expressed as the mean ± SD. **p < 0.05 vs. Ctrl group; ***p < 0.05 vs. RSL-3 group.

3.7 Transfection of COX2-siRNA alleviates RSL-3-induced ferroptosis in HK-2 cells

To further verify that transfection of COX2-siRNA may protect against ferroptosis-induced RSL-3 in HK-2 cells, intense GPX4 fluorescence in HK-2 cells was revealed by immunofluorescence microscopy. Notably, decreased fluorescence was detected in RSL-3-treated HK-2 cells compared to the controls. In contrast, GPX4-related levels were partially recovered by the transfection of COX2-siRNA in RSL-3-treated HK-2 cells (Fig 7A). Furthermore, CCK8 assays showed that RSL-3 induced massive cell death through the pathogenesis of ferroptosis after 48 h. Interestingly, RSL-3-induced ferroptosis-specific cell death was attenuated by COX2 gene disruption. The CCK8 assay demonstrated that HK-2 cell death induced by RSL-3 was alleviated by COX2-siRNA treatment (Fig 7B). Thus, the high expression of COX2 protein (Fig 7C) and mRNA (Fig 7H) was also detected in cells cultured with 100 nm RSL3 for 48 h. We also detected that ferroptosis-related protein expression was reversed by transfection of COX2-siRNA. Our study showed that SLC7A11, GPX4, and FTH-1 protein expression was clearly increased and TFR-1 was decreased in cells transfected with COX2-siRNA (Fig 7C). The mRNA expression of these genes showed parallel results (Fig 7D–7G). The obtained data revealed that GSH was also clearly decreased in RSL-3-treated cells. However, the changes were ameliorated by COX2 knockdown in RSL-3-treated cells (Fig 7I). Simultaneously, pretreatment with transfection of COX2-siRNA reduced the concentrations of MDA (Fig 7J), 4-HNE (Fig 7K)and iron (Fig 7L), which implied that transfection of COX2-siRNA could improve the states of lipid peroxidation and iron overload induced by cells cultured in 100 nm RSL-3 medium. In addition, the mRNA levels of KIM-1 and NGAL increased in RSL-3-treated HK-2 cells. Interestingly, this pathogenesis was also impaired by transfection with COX2-siRNA (Fig 7M and 7N). Overall, the abovementioned research indicated that the inhibition of COX2 can ameliorate the ferroptosis sensitivity of RSL-3-treated HK-2 cells.

Fig 7. Cellular ferroptosis-induced RSL-3 was inhibited by transfection with COX2-siRNA.

Fig 7

(A) Immunofluorescence analysis with GPX4 antibody (red) and nuclear staining with DAPI (blue) of HK-2 cells in the Si-Ctrl, Si-COX2, RSL-3+Si-Ctrl, RSL-3+Si-COX2 and DMSO+Si-Ctrl groups. (B) The cell viability measurements in HK-2 cells treated with RSL-3 100 nm for 48 h were partially improved by transfection of COX2-siRNA. (C) Western blotting results of COX2, GPX4, SLC7A11, FTH-1 and TFR-1 in cells in each group. (D-H) The mRNA expression of GPX4 (D), SLC7A11 (E), FTH-1 (F), TFR-1 (G) and PTGS2 (H) in each group of cells. (I-L) GSH (I), MDA (J), 4-HNE(K) and iron concentrations (L) of HK-2 cells in the Si-Ctrl, Si-COX2, RSL-3+Si-Ctrl and RSL-3+Si-COX2 groups. (M-N) KIM-1 (M) and NGAL (N) increased mRNA expression induced by RSL-3 and were relieved by transfection of COX2-siRNA. Data are the mean ± SEM. **p < 0.05 vs. Si-Ctrl group; ***p < 0.05 vs. Si-Ctrl+ RSL-3 group.

3.8 COX2 gene inhibition prevents renal tubular injury in diabetic kidney disease

The diabetic mice received intragastric treatment with aspirin and 0.5% Na-CMC. HE staining revealed that aspirin ameliorated the renal morphological changes in diabetic mice, including tubular epithelial degeneration with brush border loss and dilatation of the tubular lumen. PAS staining demonstrated mitigated tubular epithelial disruption, and Masson staining revealed reduced relative collagen deposition and thickening of the basement membrane in aspirin-treated diabetic mice (Fig 8A). This study showed that the levels of blood glucose (Fig 8B) and body weight (Fig 8C) were not different in the DM, AS and vehicle-G groups. Aspirin had no effect on the blood glucose and body weights of diabetic mice, which indicated that aspirin did not affect the regulation of blood glucose in diabetes. Polyuria was clearly ameliorated by aspirin in diabetic mice. The average 24-h urine volume was approximately 14 mL in the DM group but approximately 6 mL in the AS group (Fig 8D). Compared to diabetic mice, urinary albuminuria in the AS group (Fig 8E) was almost 2 times lower than that in diabetic mice, the ACR in the AS group (Fig 8F) decreased by almost 50%, and the kidney weight (Fig 8G) decreased from 9.93 to 7.9 mg/g. Even though these exponents of renal injury were still higher in the AS group than in the normal group, they were clearly ameliorated compared with those in diabetic mice. Moreover, compared to the diabetic mice, decreased NGAL and KIM-1 mRNA levels were detected in aspirin-treated diabetic mice, demonstrating alleviation of renal pathological damages in diabetic mice (Fig 8H and 8I).

Fig 8. Aspirin ameliorates renal injury in diabetic mice.

Fig 8

(A) HE staining, PAS staining and Masson staining of kidneys in nondiabetic and diabetic control AS mice were assessed. Relative collagen (%) and relative area of fibrosis (%) were measured by the Image J program. (B-G) Physiological parameters, including blood glucose (B), body weight (C), 24-h urine volume (D), urinary albuminuria (E), albumin-to-creatinine ratio ACR (F) and kidney weight/body weight (G) were measured. (H-I) KIM-1 (H) and NGAL (I) increased mRNA expression induced by diabetes and were relieved by aspirin treatment. Data are the mean ± SEM. **p < 0.05 vs. NC group; ***p < 0.05 vs. DM group.

3.9 COX2 gene inhibition attenuates ferroptosis in diabetic kidney disease

Immunohistochemical staining showed that GPX4 and COX2 were primarily expressed in renal tubular epithelium. Notably, changes in COX2 expression were opposite compared to those detected for GPX4 in diabetic mice. The obtained data showed that COX2 expression was markedly increased in diabetic mice (Fig 9A). Both the protein and mRNA levels of GPX4 and COX2 were explored to evaluate ferroptosis in vivo. Western blotting analysis and RT‒PCR studies showed that GPX4 expression was decreased in diabetic mice. GPX4 expression was significantly increased in aspirin-treated diabetic mice. In contrast to GPX4, COX2 expression in diabetic mice was increased, while a decrease was noted in aspirin-treated diabetic mice (Fig 9B). Subsequently, the transcription conditions of FTH1 and TFR-1 in the kidneys were assessed by RT‒PCR analyses. In diabetic kidneys, decreased FTH1 and increased TFR-1 mRNA levels were detected, suggesting that there may be a high intracellular free iron concentration (Fig 9E, 9F and 9K). Interestingly, in AS group mice, GPX4, SLC7A11 and FTH-1 protein expression were significantly enhanced, and COX2 expression was clearly decreased in the renal sections compared with the DM group (Fig 9B). The mRNA expression of the genes was similar to the corresponding immunohistochemical staining (Fig 9C–9G). The GSH levels were clearly lower in diabetic mice than in controls, whereas the GSH levels were increased in AS mice, indicating that the ability of the kidneys to resist oxidative stress was ameliorated in AS group mice (Fig 9H). To assess lipid hydroperoxidation during the process of diabetic-induced injury, the extent of 4-HNE and MDA were observed. The levels of MDA and 4-HNE were notably increased in the kidneys of diabetic mice. Both of them decreased by approximately 50% in aspirin-treated diabetic mice compared to diabetic mice (Fig 9I and 9J). Moreover, we observed that the iron accumulation decreased by almost 65% in the kidneys of AS group mice and was ameliorated (Fig 9K). Ultrastructural analysis showed that the changes in mitochondrial morphology in HK-2 cells exposed to high glucose, including increased membrane density with shrunken mitochondria and mitochondrial ridge decrease or even disappearance, were alleviated by aspirin. The mitochondrial ridge remained visible, and the status of increased membrane density and mitochondrial shrinkage was weakened in the AS group (Fig 9L). Overall, this study revealed that the mitochondrial morphological pathogenesis caused by diabetic ferroptosis can be ameliorated by aspirin. The biomarkers of diabetic ferroptosis were observed in HK-2 cells cultured with high glucose, which was significantly improved by aspirin. The abovementioned data demonstrated that aspirin significantly improved renal injuries in diabetic mice. Therefore, our research illustrated that the pharmacological mechanism of aspirin ameliorates ferroptosis of the kidney in diabetic mice via inhibition of COX2, as shown in Fig 9M.

Fig 9. Aspirin ameliorates ferroptosis indices in the diabeitc kidney.

Fig 9

(A) GPX4, SLC7A11, FTH-1 and COX2 of the kidneys in nondiabetic, diabetic control and diabetic treated with AS mice were assessed based on immunohistochemical staining results and semiquantification analyses. (B) Western blot analysis of GPX4, SLC7A11, FTH-1, TFR-1 and COX2 in the kidneys of nondiabetic, diabetic control and diabetic mice treated with AS. (C-G) GPX4 (C), SLC7A11 (D), FTH-1 (E), TFR-1 (F) and PTGS2 (G) mRNA expression in the kidney tissue of mice were measured. (H-K) The GSH (G), MDA (H), 4-HNE (I) and iron concentrations (J) in the kidney tissue were analyzed. (L) The mitochondrial morphology of cells was detected by transmission electron microscopy in the Ctrl, HG and AS groups. (M) Schematic model: COX2/GPX4-mediated ferroptosis in the process of diabetic kidney. Data are the mean ± SEM. **p < 0.05 vs. NC group; ***p < 0.05 vs. DM group.

4. Discussion

DKD is a potentially life-threatening microvascular complication of diabetes mellitus (DM) [32]. DKD is closely related to several types of cell death, including apoptosis, necroptosis and ferroptosis. These studies showed that tubular cell death may be linked to tubular pathological changes and renal fibrosis in prolonged DKD. The majority of them have been thoroughly investigated except for ferroptosis, which is a more recent discovery [33,34]. The term ferroptosis is typically characterized by reduced antioxidant defense, iron overload and lipid peroxidation [3537]. The essential function of cell metabolism, especially phospholipid peroxidation, in ferroptosis depends on iron. It has been reported that system xc-, GSH synthesis, and glutathione peroxidase 4 (GPX4) can protect cells against death induced by oxidative stress conditions, especially that caused by inhibition of system xc- activity [3842]. GPX4 is a selenoprotein of antioxidant enzymes and the major enzyme capable of catalyzing the reduction of PLOOHs in mammals. Furthermore, GPX4 is essential for the survival of proximal kidney tubular cells, and the inducible depletion of GPX4 causes massive ferroptosis of renal tubular epithelial cells. Ferroptosis is essential in the genetic model of inducible whole-body depletion of Gpx4 and in folic acid-induced acute kidney injury (AKI) [18,19].

Although the role of ferroptosis in tubular injury in DKD has been less researched, it has been demonstrated that iron homeostasis is disrupted in the kidneys of diabetic mice [43]. Through the Fenton reaction, iron can directly produce excessive ROS, thus causing further increasing oxidative damage. Because iron is a critical component of cell viability and death, cellular iron homeostasis must be carefully managed. Our studies showed that TFR-1 expression was clearly increased, and FTH-1 expression was significantly decreased at both the protein and mRNA levels, indicating that iron homeostasis is imbalanced in diabetes. The evident release of free iron, which is a typical characteristic of ferroptosis, has been observed in high glucose-cultured HK-2 cells. We demonstrated that diabetic nephropathy was significantly attenuated by Fer-1. This study also showed that the increased mRNA levels of KIM-1 and NGAL in high glucose-cultured HK-2 cells were attenuated by Fer-1, indicating that ferroptosis plays an important role in cellular injury. Overall, these data demonstrated that ferroptosis inhibition is operative in DKD.

Even though tubular injury plays an important role in DKD pathogenesis, there is currently no cell-specific therapy for the condition. Evidence continues to accumulate showing the link between kidney disease and disturbances of tubular cell ferroptosis, and such insight will open up potential for developing new therapeutic approaches for DKD patients. Our study demonstrated the biological functions of COX2 in tubular cells. Importantly, COX2 in the kidney was markedly upregulated in DKD, indicating that COX2 is a potential target for DKD. Our results showed that tubular cells secrete COX2 under diabetic conditions. This research was designed to explore the biological functions of COX2 in tubular cells. We found that downregulating COX2 significantly lessened urinary albumin excretion and tubular damage in aspirin-treated diabetic mice, demonstrating that COX2 may be an essential target molecule for tubular damage.

COX2 is a proximal tubular-specific enzyme. Similarly, this study revealed that ferroptosis occurred in damaged PTECs. Our work showed that downregulation of COX2 alleviates tubular redox injury in DKD, but its role in ferroptosis remains enigmatic. ROS accumulation induced by hyperglycemia supplies oxidative stress. GPX4 and GSH were reduced in diabetic kidneys, which further exacerbated the oxidative stress response. As COX2 expression increased, it was examined in the kidneys of diabetic mice, which was in agreement with previously reported studies [20,25,26]. Functionally, our study verified one unappreciated role of COX2 in mediating renal tubular cell ferroptosis. In this study, we demonstrated whether diabetic ferroptosis could be suppressed by the downregulation of COX2 expression, thereby showing that COX2 is a potential target for the progression of DKD. First, our study demonstrated that the sensitivity of HK-2 cells to ferroptosis was decreased by the specific knockdown of COX2 under high glucose conditions. When COX2 was suppressed, TFR-1 expression was decreased, while its target gene FTH-1 expression was increased. As a result of silencing COX2, cells grown in high glucose medium have an enhanced antioxidant capacity, as well as GPX4 and SLC7A11 expression. Knockdown of COX2 diminished free iron accumulation in cells, increased resistance to oxidative stress and weakened lipid peroxidation occurrence, which induced ferroptosis and further decreased cell injury under high glucose conditions. In addition, as a biomarker of ferroptosis sensitivity, the intracellular levels of GSH were lower in high glucose-cultured HK-2 cells and diabetic mice even without aspirin treatment, suggesting that downregulation of COX2 expression can inhibit the occurrence of diabetic ferroptosis.

It is difficult to investigate the process of ferroptosis due to the lack of specific markers. Accordingly, ferroptosis is characterized by a pronounced relentless process of lipid hydroperoxidation and iron overload [37]. Lipid hydroperoxides are the executors of ferroptosis, which causes massive cell death [44], while increased intracellular free iron can catalyze ferroptosis [45]. MDA and 4-HNE are the end products of lipid hydroperoxides [14]. They were used to evaluate the extent of lipid hydroperoxidation in this research. Under high glucose conditions in vivo and in vitro, our studies showed that the increase in MDA and 4-HNE levels were dampened by COX2 gene disruption. In addition, the Iron Assay Kit assay revealed increased intracellular free iron levels, which were attenuated by COX2 gene disruption in HK-2 cells cultured under high glucose conditions. Ferroptosis is important in HK-2 cells cultured with high glucose. We hypothesized that COX2 can modulate the pathogenesis of ferroptosis. This study revealed that downregulation of COX2 was beneficial to cellular homeostasis during damage to HK-2 cells and DKD. In this study, RSL-3 directly suppressed GPX4 signaling in cultured HK2 cells, and we demonstrated that the COX2 expression profile can modulate significant ferroptosis-specific cell death induced by RSL-3.

Although the role of aspirin-mediated organ protection has been greatly emphasized, the exact role of aspirin has long been controversial. Diabetic mice developed albuminuria and tubular injury, showing that this model is likely to be an ideal experimental model to study tubular injury-related kidney damage induced by hyperglycemia. Aspirin can ameliorate albuminuria in this model. In our study, aspirin treatment under diabetic conditions decreased COX2 levels and showed renoprotection. Thus, elevated GSH levels may be an aspirin-mediated renoprotective effect in diabetic mice. Systemic improvements, such as GSH, are likely to have a protective effect against tubular loss and renal fibrosis.

Ordinarily, the key antioxidases GPX4 and GSH catalyze noxious lipid hydroperoxides into harmless lipid alcohols [12]. Thus, GPX4 inhibitors and system Xc- antagonists (related to GSH depletion) can act as inducers of ferroptosis [11]. Subsequently, under high glucose conditions, we demonstrated that COX2 expression could be downregulated by aspirin, which resulted in SLC7A11, GPX4, FTH-1 and TFR-1 expression being also altered, and COX2 knockdown ameliorated the process of ferroptosis in cells. To confirm the effect of aspirin on ferroptosis, we applied it to cells induced by RSL-3 and detected that COX2 expression decreased and the biomarkers of ferroptosis improved. When aspirin was used to treat diabetic mice, we observed that the increase in COX2 was inhibited in mice because the pathogenesis of ferroptosis was also ameliorated. In diabetic mice, aspirin also mitigated pathological kidney damage. Thus, our study demonstrated that aspirin can upregulate SLC7A11 and GPX4 expression by suppressing COX2. Moreover, the antioxidative stress ability was improved in the kidneys of diabetic mice. Consequently, the iron deposition in the kidneys of diabetic mice was reduced by increasing FTH-1 and downregulating TFR-1. Because iron metabolism stabilized and antioxidant capacity was improved in diabetic mice, ferroptosis of the kidneys was mitigated. Herein, a significant decrease in “GPX4 activity” was observed in diabetic mice, while it was enhanced in diabetic mice treated with aspirin. The abovementioned findings showed that the depletion of GSH and oxidative stress caused by high glucose were ameliorated by downregulating COX2. Here, our study shows that aspirin can effectively inhibit COX-2-mediated ferroptosis in vitro and reduce subsequent renal injury responses in vivo. Accordingly, inhibition of ferroptosis caused by downregulation of COX2 could alleviate the progression of DKD. An explanation for the nephroprotective effects of aspirin was provided by our study, i.e., inhibition of ferroptosis by downregulating COX2.

Therefore, this study demonstrated a novel link between COX2 inhibition, ferroptosis, and DKD progression. While there are multiple mechanisms by which aspirin exerts renoprotective effects, this study indicated that cellular ferroptosis plays a major role in the progression of DKD, while alleviating ferroptosis by inhibiting the COX2 enzyme can potentially attenuate the progression and development of DKD. As a result of these novel insights into DKD pathogenesis, therapeutic strategies that alleviate renal tubular injury can be developed and provide a new pharmacological target for preventing and treating DKD.

Supporting information

S1 Table

(DOCX)

S1 Raw images

(PDF)

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

This work was supported by the National Natural Science Foundation of China (32171339), the Key Program of Natural Science Foundation of Tianjin (19JCZDJC36900), Fujian Provincial health technology project (2019-1-22) and the Science and Technology Self-financing Project of Tianjin Municipal Commission of Health (ZC20143).

References

  • 1.James P Boyle, Theodore J Thompson, Edward W Gregg, Lawrence E Barker, David F Williamson. Projection of the year 2050 burden of diabetes in the US adult population: dynamic modeling of incidence, mortality, and prediabetes prevalence.Popul Health Metr,2010,8:29. doi: 10.1186/1478-7954-8-29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Paola Fioretto, Michael Mauer. Histopathology of diabetic nephropathy.Semin Nephrol,2007, 27(2):195–207. doi: 10.1016/j.semnephrol.2007.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Robert N Foley, Allan J Collins. End-stage renal disease in the United States: an update from the United States Renal Data System. J Am Soc Nephrol, 2007, 18(10):2644–8. doi: 10.1681/ASN.2007020220 [DOI] [PubMed] [Google Scholar]
  • 4.Joseph V Bonventre. Can we target tubular damage to prevent renal function decline in diabetes? Semin Nephrol, 2012, 32(5):452–62. doi: 10.1016/j.semnephrol.2012.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jefferson J A, Shankland S J, Pichler R H. Proteinuria in diabetic kidney disease: a mechanistic viewpoint. Kidney Int, 2008, 74(1):22–36. doi: 10.1038/ki.2008.128 [DOI] [PubMed] [Google Scholar]
  • 6.Perkovic V., Jardine M.J., Neal B., Bompoint S., Heerspink H.J.L., et al. CREDENCE Trial Investigators (2019). Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med, 2019,380(24):2295–2306. doi: 10.1056/NEJMoa1811744 [DOI] [PubMed] [Google Scholar]
  • 7.Akosua Vilaysane, Justin Chun, Mark E Seamone, Wenjie Wang, Rick Chin, et al. The NLRP3 inflammasome promotes renal inflammation and contributes to CKD. J Am Soc Nephrol, 2010, 21(10):1732–44. doi: 10.1681/ASN.2010020143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Xiao X, Du C, Yan Z, Shi Y, Duan H, et al. Inhibition of necroptosis attenuates kidney inflammation and interstitial fibrosis induced by unilateral ureteral obstruction. Am J Nephrol, 2017, 46(2):131–138. doi: 10.1159/000478746 [DOI] [PubMed] [Google Scholar]
  • 9.Bing Yang, Shanshan Lan, Mélanie Dieudé, Jean-Paul Sabo-Vatasescu, Annie Karakeussian-Rimbaud, et al. Caspase-3 is a pivotal regulator of microvascular rarefaction and renal fifibrosis after ischemia reperfusion injury. J Am Soc Nephrol, 2018, 29(7):1900–1916. doi: 10.1681/ASN.2017050581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Mitsuru Imamura, Jong-Seok Moon, Kuei-Pin Chung, Kiichi Nakahira, Thangamani Muthukumar, et al. RIPK3 promotes kidney fifibrosis via AKT-dependent ATP citrate lyase. JCI Insight, 2018, 3(3):e94979. doi: 10.1172/jci.insight.94979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Daolin Tang, Xin Chen, Rui Kang, Guido Kroemer. Ferroptosis: molecular mechanisms and health implications. Cell Res, 2021, 31(2):107–125. doi: 10.1038/s41422-020-00441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Xuejun Jiang, Brent R Stockwell, Marcus Conrad. Ferroptosis: mechanisms, biology, and role in disease. Nat Rev Mol Cell Biol, 2021, 22(4):266–282. doi: 10.1038/s41580-020-00324-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brent R Stockwell, José Pedro Friedmann Angeli, Hülya Bayir, Ashley I Bush, Marcus Conrad, et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell, 2017, 171(2):273–85. doi: 10.1016/j.cell.2017.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Antonio Ayala, Mario F. Munoz, Sandro Argüelles. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2- nonenal. Oxidative Medicine and Cellular Longevity, 2014, 2014: 360438, doi: 10.1155/2014/360438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jiashuo Zheng, Marcus Conrad. The Metabolic Underpinnings of Ferroptosis. Cell Metab, 2020, 32(6):920–937. doi: 10.1016/j.cmet.2020.10.011 [DOI] [PubMed] [Google Scholar]
  • 16.Tobias M Seibt, Bettina Proneth, Marcus Conrad. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med, 2019, 133:144–152. doi: 10.1016/j.freeradbiomed.2018.09.014 [DOI] [PubMed] [Google Scholar]
  • 17.Rajesh K Dutta, Vinay K Kondeti, Isha Sharma, Navdeep S Chandel, Susan E Quaggin, et al. Beneficial Effects of Myo-Inositol Oxygenase Deficiency in Cisplatin Induced AKI. J Am Soc Nephrol, 2017, 28(5):1421–1436. doi: 10.1681/ASN.2016070744 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Diego Martin-Sanchez, Olga Ruiz-Andres, Jonay Poveda, Carrasco Susana, Pablo Cannata-Ortiz, et al. Ferroptosis, but Not Necroptosis,Is Important in Nephrotoxic Folic Acid-Induced AKI. J Am Soc Nephrol, 2017, 28(1):218–229. doi: 10.1681/ASN.2015121376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jose Pedro Friedmann Angeli, Manuela Schneider, Bettina Proneth, Yulia Y Tyurina, Vladimir A Tyurin, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol, 2014,16(12):1180–91. doi: 10.1038/ncb3064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Komers R, Lindsley J N, Oyama T T, Schutzer W E, Reed J F, et al. Immunohistochemical and functional correlations of renal cyclooxygenase-2 in experimental diabetes. J Clin Invest, 2001,107(7):889–98. doi: 10.1172/JCI10228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Huifang Cheng, Suwan Wang, Young-Il Jo, Chuan-Ming Hao, Mingzhi Zhang, et al. Overexpression of cyclooxygenase-2 predisposes to podoyte injury. J Am Soc Nephrol, 2007,18(2):551–9. doi: 10.1681/ASN.2006090990 [DOI] [PubMed] [Google Scholar]
  • 22.Huifang Cheng, Xiaofeng Fan, Youfei Guan, Gilbert W Moeckel, Roy Zent, et al. Distinct roles for basal and induced COX-2 in podocyte injury. J Am Soc Nephrol, 2009, 20(9):1953–62. doi: 10.1681/ASN.2009010039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Huifang Cheng, Xiaofeng Fan, Gilbert W Moeckel, Raymond C Harris. Podocyte COX-2 exacerbates diabetic nephropathy by increasing podocyte (pro)renin receptor expression. J Am Soc Nephrol, 2011, 22(7):1240–51. doi: 10.1681/ASN.2010111149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Keyvan Amirshahrokhi.Thalidomide reduces glycerol-induced acute kidney injury by inhibition of NF-κB, NLRP3 inflammasome, COX-2 and inflammatory cytokines. Cytokine, 2021, 144:155574. doi: 10.1016/j.cyto.2021.155574 [DOI] [PubMed] [Google Scholar]
  • 25.Rikke Nørregaard, Tae-Hwan Kwon, Jørgen Frøkiær. Physiology and pathophysiology of cyclooxygenase-2 and prostaglandin E2 in the kidney. Kidney Res Clin Pract, 2015, 34(4):194–200. doi: 10.1016/j.krcp.2015.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Syed S Quadri, Silas A Culver, Caixia Li, Helmy M Siragy .Interaction of the renin angiotensin and cox systems in the kidney. Front Biosci (Schol Ed), 2016, 8:215–26. doi: 10.2741/s459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jia-Feng Chang, Jih-Chen Yeh, Chun-Ta Ho, Shih-Hao Liu, Chih-Yu Hsieh, et al. Targeting ROS and cPLA2/COX2 Expressions Ameliorated Renal Damage in Obese Mice with Endotoxemia. Int J Mol Sci, 2019, 20(18):4393. doi: 10.3390/ijms20184393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Michele Sacco, Fabio Pellegrini, Maria C Roncaglioni, Fausto Avanzini, Gianni Tognoni, et al. Primary prevention of cardiovascular events with low-dose aspirin and vitamin E in type 2 diabetic patients: results of the Primary Prevention Project (PPP) trial. Diabetes Care,2003,26(12):3264–72. doi: 10.2337/diacare.26.12.3264 [DOI] [PubMed] [Google Scholar]
  • 29.Wilson Y Leung, Wing-yee So, Derek Stewart, Augustine Lui, Peter C Tong, et al. Lack of benefits for prevention of cardiovascular disease with aspirin therapy in type 2 diabetic patients—a longitudinal observational study. Cardiovasc Diabetol, 2009,8:57. doi: 10.1186/1475-2840-8-57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Eduardo G Camargo, Letícia S Weinert, Joel Lavinsky, Jorge L Gross, Sandra P Silveiro. The effect of aspirin on the antiproteinuric properties of enalapril in microalbuminuric type 2 diabetic patients: a randomized, double-blind, placebo-controlled study. Diabetes Care, 2007, 30(7):e66. doi: 10.2337/dc07-0187 [DOI] [PubMed] [Google Scholar]
  • 31.Fabio M Pulcinelli, Luigi M Biasucci, Silvia Riondino, Simona Giubilato, Andrea Leo, et al. COX-1 sensitivity and thromboxane A2 production in type 1 and type 2 diabetic patients under chronic aspirin treatment. Eur Heart J, 2009, 30(10):1279–86. doi: 10.1093/eurheartj/ehp097 [DOI] [PubMed] [Google Scholar]
  • 32.Arici Mustafa, Chana Ravinder, Lewington Andrew, Brown Jez, Nigel John Brunskill. Stimulation of proximal tubular cell apoptosis by albumin-bound fatty acids mediated by peroxisome proliferator activated receptor-gamma. J Am Soc Nephrol, 2003, 14(1):17–27. doi: 10.1097/01.asn.0000042167.66685.ea [DOI] [PubMed] [Google Scholar]
  • 33.Yue Wang, Ran Bi, Fei Quan, Qiuhua Cao, Yanting Lin, et al. Ferroptosis involves in renal tubular cell death in diabetic nephropathy. Eur J Pharmacol, 2020, 888:173574. doi: 10.1016/j.ejphar.2020.173574 [DOI] [PubMed] [Google Scholar]
  • 34.Seonghun Kim, Shin-Wook Kang, Jeongho Joo, Seung Hyeok Han, Huiyoon Shin, et al. Characterization of ferroptosis in kidney tubular cell death under diabetic conditions. Cell Death Dis,2021,12(2):160. doi: 10.1038/s41419-021-03452-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sally E Wenzel, Yulia Y Tyurina, Jinming Zhao, Claudette M St Croix, Haider H Dar, et al. PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals. Cell, 2017, 171(3):628–641.e26. doi: 10.1016/j.cell.2017.09.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jennifer Yinuo Cao, Scott J Dixon. Mechanisms of ferroptosis. Cell Mol Life Sci, 2016,73(11–12):2195–209. doi: 10.1007/s00018-016-2194-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Scott J Dixon, Kathryn M Lemberg, Michael R Lamprecht, Rachid Skouta, Eleina M Zaitsev, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell, 2012, 149(5):1060–72. doi: 10.1016/j.cell.2012.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Shi Z Z, Osei-Frimpong J, Kala G, Kala S V, Barrios R J, et al. Glutathione synthesis is essential for mouse development but not for cell growth in culture. Proc Natl Acad Sci U S A, 2000, 97(10):5101–6. doi: 10.1073/pnas.97.10.5101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Levi J Yant, Qitao Ran, Lin Rao, Holly Van Remmen, Toru Shibatani, et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic Biol Med, 2003, 34(4):496–502. doi: 10.1016/s0891-5849(02)01360-6 [DOI] [PubMed] [Google Scholar]
  • 40.Banjac A, Perisic T, Sato H, Seiler A, Bannai S, et al. The cystine/cysteine cycle: a redox cycle regulating susceptibility versus resistance to cell death. Oncogene, 2008, 27(11):1618–28. doi: 10.1038/sj.onc.1210796 [DOI] [PubMed] [Google Scholar]
  • 41.Pankaj Kumar Mandal, Alexander Seiler, Tamara Perisic, Pirkko Kölle, Ana Banjac Canak, et al. System x(c)− and thioredoxin reductase 1 cooperatively rescue glutathione deficiency. J Biol Chem, 2010, 285(29):22244–53. doi: 10.1074/jbc.M110.121327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Alexander Seiler, Manuela Schneider, Heidi Förster, Stephan Roth, Eva K Wirth, et al. Glutathione Peroxidase 4 Senses and Translates Oxidative Stress into 12/15- Lipoxygenase Dependent- and AIF-Mediated Cell Death. Cell Metab, 2008, 8(3):237–48. doi: 10.1016/j.cmet.2008.07.005 [DOI] [PubMed] [Google Scholar]
  • 43.Mika Matsumoto, Naoko Sasaki, Takeshi Tsujino, Hirokuni Akahori, Yoshiro Naito, et al. Iron restriction prevents diabetic nephropathy in otsuka long evans tokushima fatty rat. Ren Fail, 2013,35(8):1156–62. doi: 10.3109/0886022X.2013.819729 [DOI] [PubMed] [Google Scholar]
  • 44.Michael M Gaschler, Brent R Stockwell. Lipid peroxidation in cell death. Biochem Biophys Res Commun, 2017, 482(3):419–425. doi: 10.1016/j.bbrc.2016.10.086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Minghui Gao, Prashant Monian, Nosirudeen Quadri, Ravichandran Ramasamy, Xuejun Jiang. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell, 59(2):298–308. doi: 10.1016/j.molcel.2015.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Aldrin V Gomes

21 Apr 2022

PONE-D-21-40203Aspirin Mediates Protection from Diabetic Kidney Disease by Inducing ferroptosis InhibitionPLOS ONE

Dear Dr. Wu,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

While both reviewers enjoyed reading the manuscript and found it meaningful, some concerns need to be addressed. These concerns include the detection of mitochondrial specific ROS, the rationale for the amount of aspirin used, poor quality western blot images, and grammatical errors in the manuscript.

Please submit your revised manuscript by Jun 05 2022 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Aldrin V. Gomes, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels. 

  

In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions

3. PLOS requires an ORCID iD for the corresponding author in Editorial Manager on papers submitted after December 6th, 2016. Please ensure that you have an ORCID iD and that it is validated in Editorial Manager. To do this, go to ‘Update my Information’ (in the upper left-hand corner of the main menu), and click on the Fetch/Validate link next to the ORCID field. This will take you to the ORCID site and allow you to create a new iD or authenticate a pre-existing iD in Editorial Manager. Please see the following video for instructions on linking an ORCID iD to your Editorial Manager account: https://www.youtube.com/watch?v=_xcclfuvtxQ

4. In your Methods section, please provide additional information on the animal research and ensure you have included details on : (1) methods of sacrifice (2) methods of anesthesia and/or analgesia, and (2) efforts to alleviate suffering

5. Thank you for submitting the above manuscript to PLOS ONE. During our internal evaluation of the manuscript, we found significant text overlap between your submission and the following previously published works, some of which you are an author.

https://pubmed.ncbi.nlm.nih.gov/31586053/

https://www.nature.com/articles/s41418-021-00755-6

https://linkinghub.elsevier.com/retrieve/pii/S1550413121001704

https://www.nature.com/articles/s41422-020-00441-1?code=c0e202d9-78be-496a-b93a-01b8059a8703&error=cookies_not_supported

https://www.sciencedirect.com/science/article/abs/pii/S0891584920315975?via%3Dihub

https://www.jci.org/articles/view/129903

https://www.cell.com/cell-metabolism/fulltext/S1550-4131(20)30552-0?_returnURL=https%3A%2F%2Flinkinghub.elsevier.com%2Fretrieve%2Fpii%2FS1550413120305520%3Fshowall%3Dtrue

We would like to make you aware that copying extracts from previous publications, especially outside the methods section, word-for-word is unacceptable. In addition, the reproduction of text from published reports has implications for the copyright that may apply to the publications.

Please revise the manuscript to rephrase the duplicated text, cite your sources, and provide details as to how the current manuscript advances on previous work. Please note that further consideration is dependent on the submission of a manuscript that addresses these concerns about the overlap in text with published work.

We will carefully review your manuscript upon resubmission, so please ensure that your revision is thorough.

5. Thank you for stating the following in your Competing Interests section:  

"No"

Please complete your Competing Interests on the online submission form to state any Competing Interests. If you have no competing interests, please state "The authors have declared that no competing interests exist.", as detailed online in our guide for authors at http://journals.plos.org/plosone/s/submit-now 

 This information should be included in your cover letter; we will change the online submission form on your behalf.

6. In your Data Availability statement, you have not specified where the minimal data set underlying the results described in your manuscript can be found. PLOS defines a study's minimal data set as the underlying data used to reach the conclusions drawn in the manuscript and any additional data required to replicate the reported study findings in their entirety. All PLOS journals require that the minimal data set be made fully available. For more information about our data policy, please see http://journals.plos.org/plosone/s/data-availability.

"Upon re-submitting your revised manuscript, please upload your study’s minimal underlying data set as either Supporting Information files or to a stable, public repository and include the relevant URLs, DOIs, or accession numbers within your revised cover letter. For a list of acceptable repositories, please see http://journals.plos.org/plosone/s/data-availability#loc-recommended-repositories. Any potentially identifying patient information must be fully anonymized.

Important: If there are ethical or legal restrictions to sharing your data publicly, please explain these restrictions in detail. Please see our guidelines for more information on what we consider unacceptable restrictions to publicly sharing data: http://journals.plos.org/plosone/s/data-availability#loc-unacceptable-data-access-restrictions. Note that it is not acceptable for the authors to be the sole named individuals responsible for ensuring data access.

We will update your Data Availability statement to reflect the information you provide in your cover letter

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: I Don't Know

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: No

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This is a detailed research article that unraveled the role of ferroptosis and its inhibition in diabetic kidney disease(DKD) using in-vitro HK-2 cells and male diabetic mice model. Although interesting, informative, and thorough, a number of concerns remain.

1. In figure 4, mitochondrial ROS generation was supposedly accessed via DCHF-DA staining. How true is this? DCHF-DA ( 2′ ,7′ -dichlorofluorescein diacetate) can be used to detect total intracellular ROS but certainly not mitochondrial specific ROS. I would suggest the authors revise this section and other sections where mitochondrial ROS was mentioned.

2. The authors mentioned using Aspirin, a non-specific inhibitor of COX-2 activation, at 400uM in cells and 50mg/kg in diabetic mice, but there was no rationale on why those concentration were chosen in their experiments. I would suggest they provide some pharmacological reference backing the concentration if its a low, medium, or high concentration.

3. The manuscript focused on about ~5 protein (GPX4, SLC7A11, FTH-1, TFR-1, COX2) expression. Were other antioxidant and oxidative stress protein looked into and what was their results? It would make sense to look into other pathways and include in the supplemental in order to inform readers. (Plus supplemental is missing as I wasn't able to locate it while reviewing).

4. Involvement of mitochondria should be looked into. Since mitochondria are major sources of ROS within the cell and vulnerable to oxidative stress it would make more sense to investigate their involvement in this Fe2+ overload model of DKD. I would suggest the authors include mitochondrial ROS/superoxide, morphological changes, mitochondrial permeability transition, and oxphos protein expression if possible.

5. Western blotting images could be improved especially SLC7A11 protein expression.

6. English expression needs major expression. I would suggest that grammatical errors in the manuscript be corrected.

Reviewer #2: Reviewers' comments:

In the present study, Wang et al. found that ferroptosis in renal tubular cells was involved in the development of diabetic kidney disease, and COX2 is a potential target for ferroptosis. They also demonstrated the beneficial effect of aspirin by inhibiting the diabetes-related ferroptosis. The study is well designed and of promising practical guidance. The manuscript is well written and clearly presented. However, there are some issues that the authors should address and they are as follows:

1. How is the level of 4-HNE in high glucose incubated HK2 cells and together with aspirin? It would be better to have consistent results with the animal model.

2. In Fig 4B, COX2 level is increased with DMSO treatment, is there any explanation for this result?

3. The author showed the high glucose increased the cell death in the manuscript and the beneficial effect of aspirin in the animal model, how is the effect of aspirin to the high glucose induced cell death?

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2022 Dec 14;17(12):e0279010. doi: 10.1371/journal.pone.0279010.r002

Author response to Decision Letter 0


4 Aug 2022

、I ensure that my manuscript meets PLOS ONE's style requirements, including those for file naming.

2、My blot/gel image data are in Supporting Information.

3、I ensure that I have an ORCID iD and that it is validated in Editorial Manager.

4、All mice were euthanized with 60 mg/ml of pentobarbital sodium at the end of the experimental period.

5、I have revised the manuscript to rephrase the duplicated text.

6、All authors have declared that no competing interests that pertain to this work.This information have be included in my cover letter.

Reviewer #1

Q1:DCHF-DA ( 2′ ,7′ -dichlorofluorescein diacetate) can be used to detect total intracellular ROS, the expression in the text has been modified by us.

Q2:The viability of cells treating with RSL-3 was assessed by CCK8. Our study showed that as RSL-3 concentration increased as it was obviously decreased . When RLS-3 concentration was over 0.1 μM, it decreased obviously. As the HK-2 cells was treated with different concentrations of RSL-3 for 48 h, the cell viability was partially recused by Fer-1 400nm or AS 400um treatment. In Fer-1 group and AS group , cells were respectively cultured in 30 mmol/L glucose medium supplemented with 400nM Fer-1 and 400 μM Aspirin for 48 h .

Q3:In this study, we mainly explored the relationship between aspirin and ferroptosis, so we did not examine other antioxidant and oxidative stress protein.

Q4:Theultrastructural analysis displayed that the changes of mitochondria morphological in the HK-2 cells cultured with high glucose, containing increased membrane density with shrunken mitochondria and mitochondrial ridge decrease or even vanishing, were alleviated by aspirin. The mitochondrial ridge remains obviously visible , the status of increased membrane density and mitochondrial shrinkage were weakened in AS group (Fig 9L). The research implied that the mitochondrial morphological pathogenesis caused by diabetic ferroptosis could be ameliorated by aspirin.

Q5:I have fixed the grammar mistakes in the article.

Reviewer #2

Q1:In this study, the end products of lipid peroxidation ,MDA,was detected in the cells and animal.

Q2:In Fig 4B, COX2 level is increased with DMSO treatment. Because cells were respectively cultured in 30 mmol/L glucose medium supplemented with 0.1% DMSO for 48 h.

Q3: In cells being cultured in high-glucose medium with Aspirin,the protein expression of SLC7A11,GPX4, FTH-1 were obviously upregulated while TFR-1 was decreased compared to cells under high-glucose medium ((Fig 4B), these genes exhibited the parallel mRNA expression (Fig 4C-4F).Aspirin treatment markedly attenuated the MDA level of HG group cell(Fig 4I). The GSH was also obviously decreased and the levels of iron was higher in HG group cells,the changes were ameliorated by aspirin in high-glucose medium (Fig 4H,4J).Moreover, the mRNA levels of KIM-1 and NGAL significantly increased in high glucose-cultured HK-2 cells,and these changes were alleviated by aspirin.(Fig 4K-4L)

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Aldrin V Gomes

15 Sep 2022

PONE-D-21-40203R1A spirin Mediates Protection from Diabetic Kidney Disease by Inducing ferroptosis InhibitionPLOS ONE

Dear Dr. Wu,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

The article requires some improvements in the abstract and text as suggested by the reviewers.

Please ensure that your decision is justified on PLOS ONE’s publication criteria and not, for example, on novelty or perceived impact.

For Lab, Study and Registered Report Protocols: These article types are not expected to include results but may include pilot data. 

==============================

Please submit your revised manuscript by Oct 30 2022 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Aldrin V. Gomes, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: (No Response)

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This is an improved version of the detailed research article that unraveled the role of ferroptosis and its inhibition in diabetic kidney disease(DKD) using in-vitro HK-2 cells and male diabetic mice model. The experiments in this manuscript were well executed and the rationale behind the experiments support the hypothesis and final conclusion, Although interesting, informative, and thorough, I would suggest a major revision of the Introduction section as it happens to contain a lot of typographical and grammatical errors that cannot be overlooked.

Due to the standard of this journal, I would recommend this manuscript to be accepted only after if the author are able to fix the grammatical errors in the manuscript.

Some examples of concerns that needs to be rectified but not limited to:

1. Abstract, line 4 and 5: Ferroptosis is a novelty the term linked to lipid hydroperoxidation, and it takes an important part in the pathogenesis of DKD.

Suggestion: It should read "Ferroptosis is a novelty term linked to lipid hydroperoxidation, and it plays an important part/role in the pathogenesis of DKD."

2. "DN models" were mentioned in the manuscript multiple times but I believe it should be DM. Please clarify the meaning of DN.

3. Introduction, line 28: Ferroptosis is a novel nonapoptotic cell death that is discovered iron-dependent form in 2012, induced by a redox imbalance redox homeostasis, which is characterized by detoxify lipid oxidation product and free radicals [11].

4. "These suggesting that by inhibiting ferroptosis in injury PTECs may become a new treatment for kidney"

Suggestion: It should be suggest; ... injured PTECs

5. "COX2 as an cyclooxygenase super-family protein that is an inducible enzyme at low levels in most tissues and

significantly increased in the proximal tubule of the injured kidney [25,26]"

Suggestion: This statement needs to be changed/reworded

6. Results, 3.1, line 22: "Simultaneously, the data showed that the end products of lipid peroxidation, Micro Malondialdehyde Assay Kit was used to assess the level of Malondialdehyde (MDA), the production of MDA was increased by 50% (Fig 1I) compared with the Ctrl group.

Suggestion: This sentence is confusing and would be better rewritten in a succinct manner.

7. "We hypothesized that RSL3 was applied to cause ferroptosis in HK-2 cells in order to further explore the role of COX2 in ferroptosis." This word hypothesized isn't needed in this and might be better in a version similar to "We utilized RSL3 to cause ferroptosis in HK-2 cells in order to further explore the role of COX2 in ferroptosis"

8. Furthermore, the viability of cells treating with RSL-3 was assessed by CCK8.

Suggestion: It should read the viability of cells treated with with RSL-3 was assessed by CCK8.

9. Moreover, the lipid peroxidation product accumulation,MDA(Fig 6K) and high the concentrations of iron (Fig 6L) under ferroptosis conditions were also attenuated after Fer-1 or aspirin treatment.

The word "high the concentrations of iron" is confusing in this sentence.

Please endeavor to check for mistakes in order to allow better comprehension of this article and captivate the readers.

Reviewer #2: The author answered most of the questions, but the author didn't address the 4-HNE level in high glucose incubated HK2 cells.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2022 Dec 14;17(12):e0279010. doi: 10.1371/journal.pone.0279010.r004

Author response to Decision Letter 1


25 Oct 2022

Reviewer #1: This article contain some typographical and grammatical errors that cannot be overlooked.

Thank you for your kind suggestion.We have described these sentence in clearer manner in the revised manuscript, as follows:

1. Abstract, line 4 and 5: Ferroptosis is a novelty the term linked to lipid hydroperoxidation, and it takes an important part in the pathogenesis of DKD.

Reply: Ferroptosis is a novel term linked to lipid hydroperoxidation, and it plays an important role in the pathogenesis of DKD.

2."DN models" were mentioned in the manuscript multiple times but I believe it should be DM. Please clarify the meaning of DN.

Reply:Thank you for your nice advice.We have removed the DN models from the article.

3. Introduction, line 28: Ferroptosis is a novel nonapoptotic cell death that is discovered iron-dependent form in 2012, induced by a redox imbalance redox homeostasis, which is characterized by detoxify lipid oxidation product and free radicals [11].

Reply:Ferroptosis is a novel nonapoptotic cell death that was discovered to be iron-dependent in 2012 and induced by redox imbalance redox homeostasis, which is characterized by detoxification of lipid oxidation products and free radicals [11].

4. "These suggesting that by inhibiting ferroptosis in injury PTECs may become a new treatment for kidney"

Reply:These results suggest that inhibiting ferroptosis in injured PTECs may become a new treatment for kidney diseases.

5. "COX2 as an cyclooxygenase super-family protein that is an inducible enzyme at low levels in most tissues and significantly increased in the proximal tubule of the injured kidney [25,26]"

Reply:COX2 is a cyclooxygenase superfamily protein that is an inducible enzyme at low levels in most tissues and is significantly increased in the proximal tubule of the injured kidney [25,26].

6. Results, 3.1, line 22: "Simultaneously, the data showed that the end products of lipid peroxidation, Micro Malondialdehyde Assay Kit was used to assess the level of Malondialdehyde (MDA), the production of MDA was increased by 50% (Fig 1I) compared with the Ctrl group.

Reply:Simultaneously, the data showed the end products of lipid peroxidation; a micro malondialdehyde assay kit was used to assess the level of malondialdehyde (MDA), and the production of MDA was increased by 50% (Fig. 1I) compared with that in the Ctrl group.

7."We hypothesized that RSL3 was applied to cause ferroptosis in HK-2 cells in order to further explore the role of COX2 in ferroptosis."

Reply:We utilized RSL3 to cause ferroptosis in HK-2 cells in order to further explore the role of COX2 in ferroptosis.

8. Furthermore, the viability of cells treating with RSL-3 was assessed by CCK8.

Reply:Furthermore, the viability of cells treated with RSL-3 was assessed by CCK8 assay.

9. Moreover, the lipid peroxidation product accumulation,MDA(Fig 6K) and high the concentrations of iron (Fig 6L) under ferroptosis conditions were also attenuated after Fer-1 or aspirin treatment.

Reply:Moreover, lipid peroxidation product accumulation, MDA (Fig. 6K) and concentrations of iron (Fig. 6L) under ferroptosis conditions were also attenuated after Fer-1 or aspirin treatment.

Reviewer #2: The author answered most of the questions, but the author didn't address the 4-HNE level in high glucose incubated HK2 cells.

Thank you for your kind suggestion. The products of lipid peroxidation involve reactive aldehydes (e.g. 4-hydroxynonenal (4HNE) and malondialdehyde (MDA)), which increase during ferroptosis . In this study, we have added experiments on 4-HNE levels of the cells.The products of lipid peroxidation, 4-HNE(Fig. 1J) was dramatically elevated in in HG group cells.Normally, untreated control cells contained only a minimal amount of 4-HNE. The cells treated with high glucose showed the maximal level compared to NC group cells. Interestingly, aspirin treatment markedly attenuated the level compared to that in HG group cells (Fig. 4J). Moreover,the higher levels of 4-HNE were also observed in high glucose-treated HK-2 cells, and this decrease was ameliorated in high glucose-cultured cells transfected with COX2-siRNA (Fig. 5J). The lipid peroxidation product accumulation, 4-HNE(Fig.6L) under ferroptosis induced by cells cultured in 100 nm RSL-3 medium was also attenuated after Fer-1 or aspirin treatment.Simultaneously, pretreatment with transfection of COX2-siRNA reduced the concentrations of 4-HNE(Fig.7K), which implied that transfection of COX2-siRNA could improve the states of lipid peroxidation induced by cells cultured in 100 nm RSL-3 medium.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 2

Aldrin V Gomes

29 Nov 2022

A spirin Mediates Protection from Diabetic Kidney Disease by Inducing ferroptosis Inhibition

PONE-D-21-40203R2

Dear Dr. Wu,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Aldrin V. Gomes, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: (No Response)

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This is an improved revision of the manuscript. However, minor typographical error I picked up was in results section

3.1:Another products of lipid peroxidation, 4-HNE(Fig. 1J) was dramatically "elevated in in" HG group cells. It should be "elevated in". Another minor but optional recommendation was the NC group abbreviation used in the mouse model during this study. I'm aware its the control group but it might be better if NC meaning is written in the full form at least once to inform the readers what that group is exactly.

Reviewer #2: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

**********

Acceptance letter

Aldrin V Gomes

4 Dec 2022

PONE-D-21-40203R2

Aspirin Mediates Protection from Diabetic Kidney Disease by Inducing ferroptosis Inhibition

Dear Dr. Wu:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Aldrin V. Gomes

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Table

    (DOCX)

    S1 Raw images

    (PDF)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Attachment

    Submitted filename: Response to Reviewers.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLOS ONE are provided here courtesy of PLOS

    RESOURCES