5. Experimental Section
Patient Samples Collection: Twenty-one patients with AKI and acute tubular necrosis verified by renal biopsy from Xinqiao Hospital of Army Medical University were enrolled. Fifteen para-carcinoma tissues of renal nephrectomy samples from patients with kidney cancer were used as control (Table S1, Supporting Information). Informed consent was obtained from all participants, and all procedures involving human subjects were approved by the Ethics Committee of Xinqiao Hospital, Army Medical University, following the Declaration of Helsinki guidelines. Mouse Models: SREBP1c-/- mice (C57BL/6J background) were obtained from Jackson Laboratory (B6; 129S6-Srebf1tm1jdh, stock number: 0 04365 Bar Harbor, ME, USA) and littermate wild-type (WT) mice served as controls. The YME1L1 kidney tubule-specific knock-in mice (YME1L1KI) were obtained from GemPharmatech (Nanjing, China), with littermate WT mice used as controls. As previously described, the Yme1l1-Wpre-PolyA gene fragment carrying the cadherin 16 (Cdh16) promoter was inserted into the Hipp11 (H11) locus of C57BL/6J mice using CRISPR-Cas9 gene editing technology.[37] F0 positive mice were crossed with C57BL/6J mice to create stably inherited heterozygous positive mice, which were further bred to produce homozygous positive mice with renal tubule specific knock-in of YME1L1. C57BL/6J mice were purchased from Huafu Kang Biotechnology (Beijing, China). To construct exogenous YME1L1 overexpression mice, 50 μL of adenovirus expressing only GFP (mock control) or YME1L1-GFP (≈1011 plaque-forming units mL−1, Baiouni Biotechnology, Chongqing, China) was injected into the tail vein of mice as previously described.[38] Cisplatin-induced AKI models were established as previously reported.[9d] In brief, 8-week-old male WT C57BL/6J, YME1L1KI and SREBP1c-/- mice were injected intraperitoneally with either saline or 25 mg kg−1 cisplatin (MCE, Monmouth Junction, NJ, USA). Mice were euthanized after 3 days, and blood and kidney specimens were collected. IRI-induced AKI mouse model was created by performing a 30 min clamping of bilateral renal arteries to induce ischemia, followed by 24 h of reperfusion.[9c] FA-induced AKI mouse model was established by intraperitoneal injection of 250 mg kg−1 FA or vehicle, followed by euthanasia 24 h later.[9d] The AKI-to-CKD mouse model was established as previously reported,[39] with two injections of cisplatin (15 mg kg−1) on days 0 and 14, followed by euthanasia and collection of blood and kidney specimens on day 28. All animal procedures were approved by the Animal Experimentation Ethics Committee at Army Medical University (No. AMUWEC20224523). Cell Culture and Treatment: HK-2 cells were purchased from the American Tissue Culture Collection (ATCC, Manassas, VA, USA) and maintained in DMEM/F12 medium (Meilunbio, Dalian, China) containing 10% fetal bovine serum (FBS, Corning, Corning, NY, USA) at 37 °C in a humidified environment. To construct HR model of HK-2 cells, the cells were placed in 94% N2, 1% O2, and 5% CO2 atmosphere with glucose and serum-free DMEM/F12 medium hypoxia treatment for 24 h. Subsequently, DMEM/F12 medium containing glucose and 10% FBS was replaced, and reoxygenation was continued in 95% air and 5% CO2, and cells were collected at 2, 4, and 6 h for analysis. To construct a cisplatin-induced HK-2 cell model, HK-2 cells were exposed to varying concentrations of cisplatin for 24 h, after which the cells were collected for subsequent analysis. Cell Transfection: HK-2 cells were transfected with YME1L1 or SREBP1c overexpression plasmids (Youbio, Hunan, China) or siRNA (YME1L1, OPA1, METTL3, IGF2BP1, IGF2BP2, IGF2BP3) using Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA). Cells were transfected for 24 h, and then treated with cisplatin for 24 h. The siRNAs targeting YME1L1, OPA1, METTL3, IGF2BP1, IGF2BP2, and IGF2BP3 (listed in Table S6, Supporting Information) were synthesized by Biomics (Jiangsu, China). The knockout of EGR1, FOSB, and SREBP1c in HK-2 cells was achieved through CRISPR/Cas9 genome editing.[40] The sgRNA sequences (listed in Table S7, Supporting Information) were inserted into the pRPCas9(BB)-2A-Puro vector. Then the plasmids containing the sgRNA (VectorBuilder, Guangzhou, China) were transfected into HK-2 cells. After 48 h, the cells were screened with puromycin (1 μg mL−1). Knockdown efficiency was confirmed by Western blot. Mouse Primary Tubule Isolation and Cell Culture: Mouse primary renal tubule cells culture was performed as previously described.[41] Briefly, WT C57BL/6J mice and SREBP1c-/- mice (4 weeks old) were used for primary renal tubule isolation. The kidneys were harvested, and the renal cortex was minced and digested for 8 min via 0.1% collagenase II (Sigma-Aldrich, St. Louis, MO, USA). The resulting tissue homogenate was then filtered twice through a 70 μm filter (BD Falcon, Franklin Lakes, NJ, USA). The samples were collected and centrifuged at 1000 g to obtain pellets containing renal tubules. Primary renal tubule cells were placed in DMEM/F12 medium containing 10% FBS. Flow Assay for Mouse Primary RTECs: Primary renal tubular cells were extracted from the renal cortex of both control and cisplatin-induced AKI mice. These cells were then labeled with Lotus tetragonolobus lectin (LTL), a marker for proximal tubular epithelial cells. Subsequently, cells were fixed and stained with primary antibody against YME1L1 (PA5-24808, Invitro- caspase-3 and cleaved caspase-9 expression (e) (n = 3), TEM observation (f, scale bar, 1 μm (top) and 0.5 μm (bottom)), determination of relative ATP levels (g) (n = 8), and qPCR analysis of ACADM and NDUFV1 (h and i) (n = 3). (j) Representative images of IHC staining of SREBP1 in the kidney biopsies from AKI patients (n = 21) and Para-carcinoma tissues of renal nephrectomy samples from patients with kidney cancer (n = 15). Scale bar, 50 μm. k,l) Correlation analysis between SREBP1 staining intensity and renal function. Data are shown as means ± SD and were analyzed by one-way ANOVA (a-e and g-i) or Spearman’s rank correlation test (k and l). *P < 0.05, **P < 0.01, and ***P < 0.001. Adv. Sci. 2024, 2412233 2412233 (12 of 17) © 2024 The Author(s). Advanced Science published by Wiley-VCH GmbH nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C gen) and PE-conjugated secondary antibody. The cells were analyzed with a Gallios flow cytometer (Beckman Coulter, Brea, CA, USA). qRT-PCR: Total RNA was extracted using Trizol reagent (T9424, Sigma-Aldrich), followed by reverse transcription using the reverse transcription kit (MCE, Monmouth Junction, NJ, IUSA) and qPCR analysis using a SYBR Green qPCR kit (MCE, Monmouth Junction, NJ, USA). Total DNA from RTECs and HK-2 cells was extracted using a DNA extraction kit (Takara, Shiga, Japan) as previously described,[42] and mtDNA copy number was analyzed using qPCR. Primers for human and mouse are provided in Tables S4 and S5 (Supporting Information). Western Blot: Total proteins were lysed with Ripa Lysis Buffer (Beyotime, Shanghai, China). Western blot was performed using primary antibodies against YME1L1 (11510-1-AP), Mfn1 (13798-1-AP), Fis1 (10956- 1-AP), METTL14 (26158-1-AP), IGF2BP1 (22803-1-AP), METTL3 (15073-1- AP), IGF2BP2 (11601-1-AP), IGF2BP3 (14642-1-AP), EGR1 (22008-1-AP) from Proteintech (Wuhan, China); Mfn2 (ab124733), OPA1 (ab90857), Drp1 (ab56788), SREBP1 (ab28481), Fibronectin (ab268020) from Abcam (MA, USA); FOSB (sc-398595) from Santa Cruz; 𝛼-SMA (48938S), Cleaved caspase-3 (9664S), Cleaved caspase-9 (20750S) from CST (Danvers, MA, USA) or 𝛽-actin (AF0003) from Beyotime as described previously.[43] Dot Blot: Total RNA was isolated with TRIzol. The RNA concentration was adjusted to 100 ng μL−1, and 2 μL of the quantified RNA solution was applied to a nylon membrane. The membranes were cross-linked under UV light and blocked with PBS-T containing 5% BSA. Subsequently, the membrane was incubated overnight using m6A antibody (ab284130, Abcam). After that, it was incubated with the secondary antibody, and the signal was detected using ECL chemiluminescence reagents (17 047, Zenbio, Chengdu, China). Finally, the total RNA level was assessed by methylene blue staining at 0.02%. ATP Measurement: Cells or mouse kidney tissues were lysed and centrifuged, and ATP levels were quantified using the ATP Assay Kit (S0026, Beyotime) following the provided instructions. ATP concentrations were calculated based on the standard curve, and protein content was measured to standardize ATP levels (nmol) per milligram of total protein. The relative ATP levels in each sample were then analyzed in comparison to the control group. Cell Mito Stress Test: The OCR was measured by using the Seahorse XF HS Mini Analyzer (Agilent Technologies, Santa Clara, CA, USA). HK-2 cells or primary RTECs were plated in XF8 culture plates (13 022, Agilent Technologies) and treated with cisplatin (20 μm) for 24 h once they reached a density of 2 × 104 cells per well. The Seahorse XFp Cell Mito Stress Test Kit (103 010, Agilent Technologies) was employed to measure OCR. Following three baseline respiration readings without any additions, 1.5 μm oligomycin was introduced, followed by the addition of 2 μm FCCP and Rotenone & antimycin A (0.5 μm Rot/AA) for further measurements. OCR was expressed as units of picomoles (pmol) per minute, and data were normalized against the number of cells. Mitochondrial Membrane Potential and ROS Assay: HK-2 cells or primary RTECs were treated with JC-1 (C2003S, Beyotime) or the fluorescent probe DCFH-DA (S0033M, Beyotime), and subsequently analyzed by using a flow cytometer. Apoptosis Assay: Apoptosis analysis was performed following the manufacturer’s guidelines (559 763, BD biosciences). HK-2 cells or primary RTECs were incubated with the fluorescent probe Annexin V and 7-AAD for 20 min and subsequently analyzed by using a flow cytometer. TUNEL staining was conducted with a TUNEL assay kit (Roche, Mannheim, Germany) and visualized using confocal microscopy (Zeiss, Oberkochen, Germany). Detection of Scr and BUN: Mouse Scr and BUN levels were measured using a creatinine assay kit (C011-2; njjcbio, China) and a urea nitrogen measurement kit (C013-2; njjcbio), respectively, according to the protocol specified by the manufacturer. Transmission Electron Microscopy: HK-2 cells, primary RTECs, and mouse renal tissues were fixed in glutaraldehyde and 2% osmium tetroxide for 1 h at 37 °C, then dehydrated, embedded, sectioned ultrathin, and visualized using TEM (JEM-1400PLUS, Japan).[42] HE and Masson Staining: The kidney tissues were sectioned to a thickness of 3 μm and stained with hematoxylin and eosin, as well as Masson trichrome, for light microscopic analysis. Immunofluorescence: Primary RTECs were fixed with 4% paraformaldehyde, then blocked in 2% BSA. Cells were incubated overnight with primary antibody against YME1L1 (ab234744, Abcam), followed by an FITCconjugated secondary antibody. Kidney tissue cryosections (4 μm thick) underwent similar fixation, permeabilization, blocking, and overnight incubation with primary antibodies against YME1L1 (11510-1-AP, Proteintech), Fibronectin (ab268020, Abcam), and 𝛼-SMA (48938S, CST), followed by secondary antibodies and DAPI. Visualization was performed using a confocal microscope. Immunohistochemistry: Kidney biopsy tissues from AKI patients and control sections underwent antigen repair and were incubated with antiSREBP1 (ab28481, Abcam) or anti-YME1L1 (ab234744, Abcam) antibodies. Visualization was performed using a DAB kit (ZSGB-BIO, Beijing, China). As previously stated,[44] tubular staining was scored by two independent researchers on a scale of 0-4: score 0 indicated no specific staining; score 1 represented less than 25% of the area with specific staining; score 2 indicated 25% to 50%; score 3 corresponded to 50% to 75%; and score 4 indicated more than 75% of the area. Construction of Reporter Plasmids: According to the predicted binding sites of SREBP1 in the YME1L1 promoter region, corresponding primers (Table S8, Supporting Information)) were designed to amplify different lengths of the YME1L1 promoter. The fragments including YME1L1-2000 (−2000 to +200), YME1L1-1000 (−1000 to +200), YME1L1-700 (−700 to +200), YME1L1-400 (−400 to +200), and YME1L1-150 (−150 to +200) were individually inserted into pGL3 basic vector following digestion with KpnI and XhoI. The recombinant reporter plasmids were designated as pGL3-YME1L1-P1 through pGL3-YME1L1-P5, respectively. Point mutations were introduced into the SREBP1 binding element of pGL3-YME1L1M3 (CCGAGAACCCGACGCTGTAC, Underlines represent mutated bases) with pGL3-YME1L1-P4 (−400 to +200) serving as the positive control. Dual Luciferase Reporter Analysis: Recombinant reporter plasmids and subsequent point mutation plasmids were constructed based on the promoter region of YME1L1. The recombinant plasmids and Renilla plasmids, along with the pGL3-basic vector, were co-transfected into HK-2 cells alongside either the pCDNA3.1 vector or an SREBP1c overexpression plasmid. Luciferase activity was measured using a dual luciferase reporter assay kit (Promega, Madison, WI, USA), and it was normalized using fluorescence activity with Renilla activity. Chromatin Immunoprecipitation (ChIP): ChIP assays were conducted using kits from Invitrogen.[42] HK-2 cells treated with cisplatin or control for 24 h were cross-linked with 1% formaldehyde, lysed, and sonicated to fragment DNA. Immunoprecipitation was performed with 2 μg of SREBP1 antibody (14088-1-AP, Proteintech). The isolated DNA was then subjected to PCR and qPCR amplification with primers designed to encompass the SREBP1 binding sites (−214 to −205). Additionally, primers targeting regions without SREBP1 binding sites were used as a negative control, while total DNA (Input) acted as a positive control (The primers are listed in Table S9, Supporting Information). RNA Immunoprecipitation: The RIP assay was assessed via a kit from Millipore (Burlington, MA, USA) following the manufacturer’s were collected for qPCR analysis of SREBP1c expression (i) (n = 3) and Western blot analysis of SREBP1 and IGF2BP1/2/3 expression (j) (n = 3). k) RIP assay was conducted in HK-2 cells treated with control or CP for 24 h. Agarose electrophoresis and qPCR analysis showed the direct binding between IGF2BP3 protein and SREBP1c mRNA (n = 3). l) HK-2 cells were transfected with siIGF2BP3 or siNC, and treated with Actd for the indicated time in the absence or presence of CP. Cells were collected for qPCR analysis of SREBP1c. ns: no significance, *P < 0.05, ***P < 0.001 versus Actd-siIGF2BP3 group with CP treatment (n = 3). Data are shown as means ± SD and were analyzed by two-way ANOVA (a, h, and l), two-tailed unpaired Student’s t-test (e and k) or one-way ANOVA (d, f, g, i, and j). ns: no significance. *P < 0.05, **P < 0.01, ***P < 0.001. Adv. Sci. 2024, 2412233 2412233 (14 of 17) © 2024 The Author(s). Advanced Science published by Wiley-VCH GmbH nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C guidelines.[45] HK-2 cells were exposed to either control conditions or cisplatin for 24 h. Cells were lysed by adding 150 μl RIP lysis buffer. Subsequently, 5 μg of anti-mouse beads containing antibodies specific for IgG or IGF2BP3 (14642-1-AP, Proteintech) were incubated for 6 h. Next, the RNA-protein complexes were treated with proteinase K to extract the RNA. SREBP1c mRNA was assayed using qPCR to quantify its interaction with IGF2BP3. Bioinformatics Analysis: The microarray data set (GSE87025)[46] was retrieved from the Gene Expression Omnibus (GEO) database. Differential expression analysis was then conducted using limma package (version 3.50.3).[47] Genes were considered differentially expressed if they met the criteria of |fold change| > 1 and an adjusted p-value (FDR < 0.01). The JASPAR database (http://jaspar.genereg.net) was used to predict the binding site of SREBP1 in the YME1L1 promoter region.[42] GO analysis was performed by Metascape[48] (http://metascape.org) and clusterprofiler. Mouse kidney single-cell RNA sequencing database (GSE197266) were obtained from the NCBI GEO database.[49] Analysis was conducted using the R package Seurat (version 3.1.1). Quality control for the dataset were as follows: gene counts between 500 and 4000, NUMI counts greater than 500 and less than 15000; exclusion of cells with high complexity (log10GenesPerUMI) ≤ 0.8; and removal of cells with a mitochondrial ratio exceeding 10%. Harmoniously integrate matrices for all samples and eliminate batch effects between different samples.[50] Statistical Analysis: Data were presented as mean± SD. One-way analysis of variance (ANOVA) was used for comparisons among multiple groups, while two-tailed unpaired Student’s t-test was employed for twogroup comparisons. The correlation was examined by Spearman’s rank correlation test. Statistical analysis was performed using GraphPad Prism 8.0 (GraphPad Software Inc, La Jolla, CA, USA). P < 0.05 was considered statistically significant. Supporting Information Supporting Information is available from the Wiley Online Library or from the author.
Keywords
AKI, Mitochondrial dysfunction, SREBP1c, YME1L1 Received: October 1, 2024 Revised: November 29, 2024 Published online: [1] a) R. Bellomo, J. A. Kellum, C. Ronco, Lancet 2012, 380, 756; b) A. S. Levey, M. T. James, Ann. Intern. Med. 2017, 167, ITC66; c) L. Gao, X. Zhong, J. Jin, J. Li, X. M. Meng, Signal Transduction Targeted Ther. 2020, 5, 9. [2] Y. Hu, C. Yang, T. Amorim, M. Maqbool, J. Lin, C. Li, C. Fang, L. Xue, A. Kwart, H. Fang, M. Yin, A. J. Janocha, D. Tsuchimoto, Y. Nakabeppu, X. Jiang, A. Mejia-Garcia, F. Anwer, J. Khouri, X. Qi, Q. Y. Zheng, J. S. Yu, S. Yan, T. LaFramboise, K. C. Anderson, L. C. Herlitz, N. C. Munshi, J. Lin, J. Zhao, Cancer Res. 2021, 81, 713. [3] a) L. Xu, J. Guo, D. G. Moledina, L. G. Cantley, Nat. Commun. 2022, 13, 4892; b) S. Sears, L. Siskind, J. Am. Soc. Nephrol. 2021, 32, 1559; c) F. Wang, J. Ding, Kidney Dis. (Basel) 2021, 7, 10. [4] P. Bhargava, R. G. Schnellmann, Nat. Rev. Nephrol. 2017, 13, 629. [5] a) C. Tang, J. Cai, X. M. Yin, J. M. Weinberg, M. A. Venkatachalam, Z. Dong, Nat. Rev. Nephrol. 2021, 17, 299; b) X. Yu, M. Xu, X. Meng, S. Li, Q. Liu, M. Bai, R. You, S. Huang, L. Yang, Y. Zhang, Z. Jia, A. Zhang, Sci. Transl. Med. 2020, 12, aay7591. c) H. M. Kang, S. H. Ahn, P. Choi, Y. A. Ko, S. H. Han, F. Chinga, A. S. Park, J. Tao, K. Sharma, J. Pullman, E. P. Bottinger, I. J. Goldberg, K. Susztak, Nat. Med. 2015, 21, 37. [6] Y. Ruan, H. Li, K. Zhang, F. Jian, J. Tang, Z. Song, Cell Death Dis. 2013, 4, 896. [7] Y. J. Lee, G. H. Kim, S. I. Park, J. H. Lim, J. Cell. Mol. Med. 2020, 24, 899. [8] K. Ando, T. Yokochi, A. Mukai, G. Wei, Y. Li, S. Kramer, T. Ozaki, Y. Maehara, A. Nakagawara, Mol. Carcinog. 2019, 58, 1134. [9] a) V. Volarevic, B. Djokovic, M. G. Jankovic, C. R. Harrell, C. Fellabaum, V. Djonov, N. Arsenijevic, J. Biomed. Sci. 2019, 26, 25; b) Y. Xu, H. Ma, J. Shao, J. Wu, L. Zhou, Z. Zhang, Y. Wang, Z. Huang, J. Ren, S. Liu, X. Chen, J. Han, J. Am. Soc. Nephrol. 2015, 26, 2647; c) L. Yang, T. Y. Besschetnova, C. R. Brooks, J. V. Shah, J. V. Bonventre, Nat. Med. 2010, 16, 535; d) A. Ortiz, H. Husi, L. GonzalezLafuente, L. Valiño-Rivas, M. Fresno, A. B. Sanz, W. Mullen, A. Albalat, S. Mezzano, T. Vlahou, H. Mischak, M. D. Sanchez-Niño, J. Am. Soc. Nephrol. 2017, 28, 823. [10] Y. Wang, J. Viscarra, S. J. Kim, H. S. Sul, Nat. Rev. Mol. Cell Biol. 2015, 16, 678. [11] L. He, Q. Wei, J. Liu, M. Yi, Y. Liu, H. Liu, L. Sun, Y. Peng, F. Liu, M. A. Venkatachalam, Z. Dong, Kidney Int. 2017, 92, 1071. [12] a) J. S. Guo, J. Ma, X. H. Zhao, J. F. Zhang, K. L. Liu, L. T. Li, Y. X. Qin, F. H. Meng, L. Y. Jian, Y. H. Yang, X. Y. Li, Adv. Sci. (Weinh) 2024, 11, 2402450; b) X. Wu, H. Liu, J. Wang, S. Zhang, Q. Hu, T. Wang, W. Cui, Y. Shi, H. Bai, J. Zhou, L. Han, L. Li, T. Zhao, Y. Wu, J. Luo, D. Feng, W. Guo, S. Ge, Y. Qu, Cell Death Differ. 2024; c) Y. C. Tsai, T. H. Hsieh, Y. R. Liao, M. T. Tsai, T. P. Lin, D. Y. Lee, J. Park, D. Kim, K. Susztak, S. F. Yang, C. C. Lin, S. Y. Li, J. Am. Soc. Nephrol. 2024, 35, 1312. [13] Z. Ni, P. Sun, J. Zheng, M. Wu, C. Yang, M. Cheng, M. Yin, C. Cui, G. Wang, L. Yuan, Q. Gao, Y. Li, Cancer Res. 2022, 82, 1789. [14] A. Zuk, J. V. Bonventre, Annu. Rev. Med. 2016, 67, 293. [15] Y. Yang, S. Liu, H. Gao, P. Wang, Y. Zhang, A. Zhang, Z. Jia, S. Huang, Free Radical Biol. Med. 2020, 152, 821. [16] S. J. Allison, Nat. Rev. Nephrol. 2015, 11, 197. [17] T. Chiba, K. D. Peasley, K. R. Cargill, K. V. Maringer, S. S. Bharathi, E. Mukherjee, Y. Zhang, A. Holtz, N. Basisty, S. D. Yagobian, B. Schilling, E. S. Goetzman, S. Sims-Lucas, J. Am. Soc. Nephrol. 2019, 30, 2384. Adv. Sci. 2024, 2412233 2412233 (16 of 17) © 2024 The Author(s). Advanced Science published by Wiley-VCH GmbH nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense [18] A. V. Birk, S. Liu, Y. Soong, W. Mills, P. Singh, J. D. Warren, S. V. Seshan, J. D. Pardee, H. H. Szeto, J. Am. Soc. Nephrol. 2013, 24, 1250. [19] T. MacVicar, Y. Ohba, H. Nolte, F. C. Mayer, T. Tatsuta, H. G. Sprenger, B. Lindner, Y. Zhao, J. Li, C. Bruns, M. Krüger, M. Habich, J. Riemer, R. Schwarzer, M. Pasparakis, S. Henschke, J. C. Brüning, N. Zamboni, T. Langer, Nature 2019, 575, 361. [20] J. Cesnekova, M. Rodinova, H. Hansikova, J. Zeman, L. Stiburek, Int. J. Mol. Sci. 2018, 19, 3930. [21] a) D. C. Chan, Annu. Rev. Pathol. 2020, 15, 235; b) T. Wai, J. GarcíaPrieto, M. J. Baker, C. Merkwirth, P. Benit, P. Rustin, F. J. Rupérez, C. Barbas, B. Ibañez, T. Langer, Science 2015, 350, aad0116. [22] D. Dorotea, D. Koya, H. Ha, Front. Pharmacol. 2020, 11, 265. [23] X. Wen, Y. Zeng, L. Liu, H. Zhang, W. Xu, N. Li, X. Jia, J. Ethnophar- macol. 2012, 142, 144. [24] D. Wang, Y. Luo, X. Wang, D. J. Orlicky, K. Myakala, P. Yang, M. Levi, Int. J. Mol. Sci. 2018, 19, 137. [25] a) A. Chen, X. Chen, S. Cheng, L. Shu, M. Yan, L. Yao, B. Wang, S. Huang, L. Zhou, Z. Yang, G. Liu, Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2018, 1863, 538; b) T. T. P. Nguyen, D. Y. Kim, Y. G. Lee, Y. S. Lee, X. T. Truong, J. H. Lee, D. K. Song, T. K. Kwon, S. H. Park, C. H. Jung, C. Moon, T. F. Osborne, S. S. Im, T. I. Jeon, Mol. Cell 2021, 81, 3820; c) A. Bist, P. E. Fielding, C. J. Fielding, Proc. Natl. Acad. Sci. USA 1997, 94, 10693; d) S. Su, H. Tian, X. Jia, X. Zhu, J. Wu, Y. Zhang, Y. Chen, Z. Li, Y. Zhou, J. Cell. Mol. Med. 2020, 24, 10063. [26] C. Tan, J. Gu, T. Li, H. Chen, K. Liu, M. Liu, H. Zhang, X. Xiao, Int. J. Mol. Med. 2021, 47, 19. [27] R. Lan, H. Geng, P. K. Singha, P. Saikumar, E. P. Bottinger, J. M. Weinberg, M. A. Venkatachalam, J. Am. Soc. Nephrol. 2016, 27, 3356. [28] S. van der Rijt, J. C. Leemans, S. Florquin, R. H. Houtkooper, A. Tammaro, Nat. Rev. Nephrol. 2022, 18, 588. [29] H. Shimano, R. Sato, Nat. Rev. Endocrinol. 2017, 13, 710. [30] M. Naito, K. Bomsztyk, R. A. Zager, Am. J. Pathol. 2009, 174, 54. [31] a) Z. Song, Y. Xia, L. Shi, H. Zha, J. Huang, X. Xiang, H. Li, H. Huang, R. Yue, H. Wang, J. Zhu, Cell. Mol. Biol. Lett. 2024, 29, 31; b) Z. J. Fu, Z. Y. Wang, L. Xu, X. H. Chen, X. X. Li, W. T. Liao, H. K. Ma, M. D. Jiang, T. T. Xu, J. Xu, Y. Shen, B. Song, P. J. Gao, W. Q. Han, W. Zhang, Redox Biol. 2020, 36, 101671; c) K. A. Nath, J. P. Grande, A. J. Croatt, S. Likely, R. P. Hebbel, H. Enright, Kidney Int. 1998, 53, 100. [32] a) W. Zhu, M. Chen, Y. Wang, Y. Chen, Y. Zhang, Y. Wang, P. Liu, P. Li, Chem. Biol. Interact. 2023, 385, 110711; b) G. Lee, Y. Y. Kim, H. Jang, J. S. Han, H. Nahmgoong, Y. J. Park, S. M. Han, C. Cho, S. Lim, J. R. Noh, W. K. Oh, C. H. Lee, S. Kim, J. B. Kim, Cell Metab. 2022, 34, 702. [33] L. Sun, N. Halaihel, W. Zhang, T. Rogers, M. Levi, J. Biol. Chem. 2002, 277, 18919. [34] Y. C. Lin, M. S. Wu, Y. F. Lin, C. R. Chen, C. Y. Chen, C. J. Chen, C. C. Shen, K. C. Chen, C. C. Peng, Int. J. Mol. Sci. 2019, 20, 1570. [35] S. Oerum, V. Meynier, M. Catala, C. Tisne, Nucleic Acids Res. 2021, 49, 7239. [36] D. Wu, C. B. Spencer, L. Ortoga, H. Zhang, C. Miao, Redox Biol. 2024, 74, 103194. [37] Z. Zhang, L. Zhou, Q. Liu, Y. Zheng, X. Tan, Z. Huang, M. Guo, X. Wang, X. Chen, S. Liang, W. Li, K. Song, K. Yan, J. Li, Q. Li, Y. Zhang, S. Yang, Z. Cai, M. Dai, Q. Xian, Z. L. Shi, K. Xu, K. Lan, Y. Chen, Emerging Microbes Infect. 2024, 13, 2353302. [38] Y. Pan, S. Jiang, Q. Hou, D. Qiu, J. Shi, L. Wang, Z. Chen, M. Zhang, A. Duan, W. Qin, K. Zen, Z. Liu, Diabetes 2018, 67, 717. [39] a) S. I. Landau, X. Guo, H. Velazquez, R. Torres, E. Olson, R. GarciaMilian, G. W. Moeckel, G. V. Desir, R. Safirstein, Kidney Int. 2019, 95, 797; b) X. Guo, L. Xu, H. Velazquez, T. M. Chen, R. M. Williams, D. A. Heller, B. Burtness, R. Safirstein, G. V. Desir, J. Am. Soc. Nephrol. 2022, 33, 342. [40] R. Graf, X. Li, V. T. Chu, K. Rajewsky, Cell Rep. 2019, 26, 1098. [41] J. Xiong, L. Ran, Y. Zhu, Y. Wang, S. Wang, Y. Wang, Q. Lan, W. Han, Y. Liu, Y. Huang, T. He, Y. Li, L. Liu, J. Zhao, K. Yang, Theranostics 2022, 12, 5069. [42] Y. Huang, J. Zhou, S. Wang, J. Xiong, Y. Chen, Y. Liu, T. Xiao, Y. Li, T. He, Y. Li, X. Bi, K. Yang, W. Han, Y. Qiao, Y. Yu, J. Zhao, Theranostics 2020, 10, 7384. [43] Y. Liu, X. Bi, J. Xiong, W. Han, T. Xiao, X. Xu, K. Yang, C. Liu, W. Jiang, T. He, Y. Yu, Y. Li, J. Zhang, B. Zhang, J. Zhao, Mol. Ther. 2019, 27, 1051. [44] Y. Fan, W. Xiao, K. Lee, F. Salem, J. Wen, L. He, J. Zhang, Y. Fei, D. Cheng, H. Bao, Y. Liu, F. Lin, G. Jiang, Z. Guo, N. Wang, J. C. He, J. Am. Soc. Nephrol. 2017, 28, 2007. [45] J. N. Wang, F. Wang, J. Ke, Z. Li, C. H. Xu, Q. Yang, X. Chen, X. Y. He, Y. He, X. G. Suo, C. Li, J. T. Yu, L. Jiang, W. J. Ni, J. Jin, M. M. Liu, W. Shao, C. Yang, Q. Gong, H. Y. Chen, J. Li, Y. G. Wu, X. M. Meng, Sci. Transl. Med. 2022, 14, abk2709. [46] L. Markó, E. Vigolo, C. Hinze, J. K. Park, G. Roël, A. Balogh, M. Choi, A. Wübken, J. Cording, I. E. Blasig, F. C. Luft, C. Scheidereit, K. M. Schmidt-Ott, R. Schmidt-Ullrich, D. N. Müller, J. Am. Soc. Nephrol. 2016, 27, 2658. [47] M. E. Ritchie, B. Phipson, D. Wu, Y. Hu, C. W. Law, W. Shi, G. K. Smyth, Nucleic Acids Res. 2015, 43, 47. [48] Y. Zhou, B. Zhou, L. Pache, M. Chang, A. H. Khodabakhshi, O. Tanaseichuk, C. Benner, S. K. Chanda, Nat. Commun. 2019, 10, 1523. [49] Y. Wang, Y. Li, Z. Chen, Y. Yuan, Q. Su, K. Ye, C. Chen, G. Li, Y. Song, H. Chen, Y. Xu, Cell Death Dis. 2022, 13, 693. [50] I. Korsunsky, N. Millard, J. Fan, K. Slowikowski, F. Zhang, K. Wei, Y. Baglaenko, M. Brenner, P. R. Loh, S. Raychaudhuri, Nat. Methods 2019, 16, 1289. Adv. Sci. 2024, 2412233 2412233 (17 of 17) © 2024 The Author(s). Advanced Science published by Wiley-VCH GmbH nloaded from https://onlinelibrary.w iley.com /doi/10.1002/advs.202412233 by IN A SP - N E PA L , W iley O nline L ibrary on [25/12/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense