Open Access

Ferroptosis and renal fibrosis: A new target for the future (Review)

  • Authors:
    • Han Yin Zhang
    • Meng Cheng
    • Lei Zhang
    • Yi Ping Wang
  • View Affiliations

  • Published online on: November 17, 2022     https://doi.org/10.3892/etm.2022.11712
  • Article Number: 13
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ferroptosis is a type of non‑apoptotic controlled cell death triggered by oxidative stress and iron‑dependent lipid peroxidation. Ferroptosis is regulated by signalling pathways that are associated with metabolism, including glutathione peroxidase 4 dysfunction, the cystine/glutamate antiporter system, lipid peroxidation and inadequate iron metabolism. Ferroptosis is associated with renal fibrosis; however, further research is required to understand the specific molecular mechanisms involved. The present review aimed to discuss the known molecular mechanisms of ferroptosis and outline the biological reactions that occur during renal fibrosis that may be associated with ferroptosis. Further investigation into the association between ferroptosis and renal fibrosis may lead to the development of novel treatment methods.

1. Introduction

Regulated cell death (RCD) is a key biological mechanism in the body that is required for healthy development, homeostasis maintenance and disease prevention. RCD primarily includes apoptosis, necroptosis, autophagy and ferroptosis (1). Ferroptosis is fuelled by oxidative stress and iron-dependent lipid peroxidation, which differs from apoptosis, necroptosis and autophagy morphologically, biochemically and genetically (2). It is characterized by increased intracellular free iron and accumulation of toxic lipid peroxides, leading to cell death (3,4). Previous research has demonstrated that renal fibrosis, which is defined by the breakdown of healthy kidney architecture, fibroblast proliferation and excessive extracellular matrix deposition, is a common pathological state of almost all types of chronic and progressive kidney disorder (5,6). Ferroptosis is closely associated with the pathological process of numerous renal diseases and plays a key role in numerous fibrotic diseases; however, the specific mechanisms underlying development of renal fibrosis remain to be fully elucidated. Ferroptosis serves a key role in the development of renal fibrosis, and a comprehensive understanding of this involvement may identify novel targets and approaches for the development of disease prevention and therapy.

2. Ferroptosis

Ferroptosis is an iron-dependent and lipotoxic RCD. Erastin is a cell-permeable substance that was discovered by Dolma et al (7) in 2003 using a high-content screening assay. In that study, it was demonstrated that erastin selectively inhibits genetically engineered cells with oncogenic RAS mutations without harming healthy cells. In 2012, Dixon et al (2) named erastin-induced iron-dependent non-apoptotic RCD as ‘ferroptosis’. Ferroptosis is characterized by high levels of lipid peroxidation at the cytoplasmic membrane and/or intracellular locations, such as the mitochondria, endoplasmic reticulum or lysosome, and is a caspase-independent type of cell death (2,8-10). Morphologically, ferroptosis is characterized by decreased mitochondrial density, decreased or absent mitochondrial cristae and rupture of the outer mitochondrial membrane. Moreover, these features are accompanied by intact membranes, normal nuclear size and non-condensed chromatin (11). Biochemically, ferroptosis is characterized by accumulation of reactive oxygen species (ROS), increase of lipid peroxides and the deposition of intracellular iron ions (2). Ferroptosis is regulated by factors associated with metabolism, including glutathione peroxidase 4 (GPX4) dysfunction, the cystine (Cys)/glutamate (Glu) antiporter system (system Xc-1), lipid peroxidation and iron metabolism dysfunction (12). It is also associated with signalling pathways, such as p53(13), ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (COQ10)/NAD(P)H (14), PI3K/Akt/mTOR (15), sequestosome 1 (p62)/Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (NRF2) (16) and autophagy-related (ATG) 5/ATG7/nuclear receptor coactivator (NCOA) 1(17).

GPX4

The antioxidant enzyme GPX4 is a member of the GPXs family and a key target in the regulation of ferroptosis (18). GPX4 interrupts the lipid peroxidation chain reaction by reducing complex hydroperoxides (including phospholipid hydroperoxides and cholesterol hydroperoxides) to their corresponding Sub-units (19). GPX4 is a selenoprotein with selenocysteine (Sec) in its active site, and its active state requires the catalysis of glutathione (GSH). GPX4 activity is reduced or inactivated when GSH is depleted, and GPX4 also converts GSH to glutathione disulfide (GSSG), thereby reducing esterified oxidized fatty acids and cholesterol hydroperoxide, and reducing lipid hydroperoxide (L-OOH) to a nontoxic lipid hydroxy derivative (L-OH), thus resisting oxidative damage (13). During GPX4 maturation, Sec-tRNA is mediated by mevalonate, one of the key regulatory factors in positive regulation of the pathway product, isopentenyl pyrophosphate (13). Both erastin and RAS-selective lethal 3 (RSL3) compounds induce ferroptosis via inactivation of GPX4, according to Dixon et al (20). A recent study also demonstrated that erastin upregulates the expression of ATF3, which inhibits expression of GPX4, in addition to inactivating GPX4 and inhibiting its expression (21).

System Xc-

System Xc- is an antioxidant system located on the cell membrane and an amino acid antiporter that is broadly distributed in the phospholipid bilayer. It is a heterodimer consisting of two subunits and primarily consists of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2(22). System Xc- regulates the 1:1 ratio of Cys and Glu entering and leaving cells (23). The primary antioxidant in cells is the reducing agent GSH and GSH biosynthesis is limited by cysteine (2,24). Cys is reduced to cysteine in the cell and then synthesized into GSH. GSH impacts intracellular redox homeostasis and activates GPX4. Inhibiting activity of system Xc- reduces the uptake of Cys, therefore impacting the synthesis of GSH. In turn, this decreases activity of GPX4 and the antioxidant capacity of cells and accumulates ROS, ultimately leading to oxidative damage and ferroptosis (25,26). Erastin impairs cellular antioxidant defenses by inhibiting system Xc- mediated cystine uptake, thereby promoting the accumulation of ROS and thus ferroptosis (7). Embryonic fibroblasts derived from SLC7A11 knockout mice have ferroptotic cell death due to SLC7A11 gene deletion (27). And deletion of SLC7A11 gene in mice can lead to ferroptotic-like impairment (28). Chang et al (29) found that the increase of SLC7A11 significantly inhibited the occurrence of ferroptosis. This shows that system Xc- plays an important role in the occurrence of ferroptosis.

Lipid peroxidation

Lipid metabolism is key in the process of ferroptosis and is a typical free radical chain reaction. Polyunsaturated fatty acids (PUFAs) are involved in almost all pathways of ferroptosis because they are susceptible to lipid peroxidation due to the presence of easily extractable hydrogen atoms at bis-allylic carbon positions (30). Any free radical that can extract hydrogen atoms from oxidizable substrates can initiate the lipid peroxidation process, and the abundance and location of intracellular lipid peroxidizable substrates determines the extent of lipid peroxidation and ferroptosis (8). Free PUFAs are substrates for synthesis of lipid signal transduction mediators, and these are esterified into membrane phospholipids during lipid metabolism and oxidized into ferroptosis signals (13). Lipidomic analysis has revealed that phosphatidylethanolamines (PEs) are key membrane phospholipids that drive ferroptosis by oxidizing phospholipid hydroperoxides [arachidonic acid (AA) and adrenic acid (AdA)-hydroperoxides-PE] via a non-enzymatic process (13,16). Acyl-CoA Synthetase is a long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyl-transferase 3 (LPCAT3) are two enzymes involved in the biosynthesis and remodeling of PE, which activate PUFA and affect its transmembrane properties (12). Therefore, blocking expression of ACSL4 and LPCAT3 inhibits the esterification of AA or AdA to PE and decreases accumulation of intracellular lipid peroxidative substrates, thereby inhibiting ferroptosis (13). As enzymatic effectors, lipoxygenases, of which free PUFAs are the preferred substrates, mediate the peroxidative reaction of ferroptosis (31). SLC38A1 is a regulator of glutamate uptake and metabolism in lipid peroxidation (32). Yang et al (33) found that lncRNA ZFAS1 could regulate the expression of SLC38A1 through miR-150-5p and activate the conversion of fibroblasts into myofibroblasts in lung tissue with the development of cytosolic iron death.

Iron metabolism dysfunction

Iron can form iron (Fe3+) ions and ferrous (Fe2+) ions, which are one of the essential trace elements for the human body and are involved in a variety of biological processes (like the formation of hemoglobin from proteins, the transport of oxygen, and the formation of enzymes necessary for the human body) (34). One of the primary characteristics of ferroptosis is accumulation of iron ions (2). Under normal conditions, intracellular iron is balanced by the iron transport system, while extracellular iron is taken into the cell by transferrin (TF) and transferrin receptor (TFR) and stored and transported as ferritin complexes (mainly ferritin) (35). Ferritin is mainly composed of ferritin light chain (FTL) and ferritin heavy chain (FTH1), which has iron oxidase activity and can catalyze the conversion of Fe2+ to Fe3+, promote iron binding to ferritin and reduce free iron levels (36). Ferroportin 1 (FPN1) is the only protein known to control iron export in mammalian cells and serves an important role in iron metabolism (FPN1 is a target molecule of hepcidin. FPN1, in the action of hepcidin, controls the amount of dietary iron, circulating iron and stored iron released into the plasma by altering its distribution across the cell membrane to maintain iron homeostasis in the body.) (2,3). Fe2+ is the form involved in the reaction during iron death, and when the body's iron metabolism is upset, intracellular iron stores are reduced and excess Fe2+ is involved in the Fenton reaction catalyzing the production of large amounts of ROS and hydroxyl radicals, which leads to the occurrence of ferroptosis (24). Alvarez et al (37) demonstrated that iron-sulphur cluster biosynthetic enzyme could resist the onset of iron death by inhibiting the elevation of intracellular iron levels. Fang et al (38) and Chang et al (29) observed that heme oxygenase 1 (HO-1) induces ferroptosis via promotion of heme decomposition to release Fe2+ ions. Serine/threonine protein kinase ataxia-telangiectasia mutated protein, a crucial DNA damage response regulator, promotes ferroptosis by preventing nuclear translocation of metal regulatory transcription factor 1, a transcription factor that triggers production of FTH1 and FTL to decrease iron toxicity (39).

Other molecular mechanisms

Results of a recent study demonstrated that p53 is essential for inducing ferroptosis (13). p53 responds to different stress signals via the coordination of specific cellular responses and the corresponding cell cycle arrest and apoptosis play important roles in inhibiting development of cancer (13). It was found that p53 could inhibit the uptake of System Xc- to Cys by suppressing the expression of SLC7A11, which resulted in a large decrease in GSH production and affected the activity of GPX4, leading to reduced antioxidant capacity and ROS accumulation, thus promoting cellular iron death (40). The communication between mitochondria and other organelles is aided by voltage-dependent anion channels (VDACs), channel proteins located in the outer mitochondrial membranous layer (41). Yagoda et al (42) demonstrated that erastin alters the permeability of the mitochondrial outer membrane via direct binding to VDAC2/3, thereby decreasing the oxidation rate of NADH and inducing ferroptosis. FSP1 is a potent ferroptosis-resistance factor (43). Doll et al (14) demonstrated that myristoylation of FSP1 inhibits lipid peroxidation via NAD(P)H reduction of COQ10, thereby inhibiting ferroptosis. Moreover, methionine is converted to Cys under oxidative stress via the sulphur transfer pathway to create GSH, which exerts its antioxidant action (44). In addition, the PI3K/Akt/mTOR (15), p62/Keap1/NRF2(16) and ATG5/ATG7/NCOA4(17) signalling pathways serve regulatory roles in the occurrence of ferroptosis.

3. Ferroptosis and renal fibrosis

Oxidative stress

When the redox system is damaged, ROS and reactive nitrogen species are excessively produced and oxidative stress occurs (45). Well-established mechanisms of oxidative stress-mediated renal injury include production of ROS and the ensuing disruption of the antioxidant system, which result in apoptosis, ferroptosis and necrosis (46,47). There are numerous causes of renal fibrosis, including oxidative stress (48). Specific inhibitors, including ferrostatin-1 (Fer-1), which is characterized by lipid-dependent peroxidation, prevent ferroptosis (2). Roxadustat is an emerging therapeutic option for treatment of anaemia in patients with chronic kidney disease (CKD). It is an oral inhibitor of hypoxia-inducible factor (HIF) prolyl hydroxylase, which stimulates erythropoiesis and regulates iron metabolism. Using a folic acid-induced kidney injury model, Li et al (49) demonstrated that roxadustat pre-treatment decreases ferroptosis and inhibits inflammation by stabilizing HIF-1α and activating the nuclear factor erythroid 2-related factor 2 (Nrf2) signalling pathway, thereby inhibiting renal fibrosis. Ide et al (50) demonstrated that lipid peroxidation induces ferroptotic stress and ferroptosis. Following injury, renal proximal tubule (PT) cells may exhibit a pro-inflammatory state and genes involved in high ferroptosis stress may trigger accumulation of inflammatory PT cells, thereby enhancing inflammation and fibrosis. Feng et al (51) demonstrated that diabetes increases HIF-1α and HO-1 in the kidney of mice, resulting in increased lipid peroxidation due to increased ROS production and tubular iron deposition. This leads to renal tubular damage and fibrosis in mice. In a diabetic mouse model, inhibition of ferroptosis prevents lipid peroxidation by decreasing ROS production in the kidneys and reducing iron deposition in the renal tubules (51). This alleviates diabetes mellitus, renal tubular injury and renal fibrosis progression in mice (51). Nobiletin (Nob), an important active flavonoid found in citrus fruits, exhibits antioxidant, anti-inflammatory, antifibrotic and antiapoptotic properties (52). Lo et al (53) demonstrated that Nob partially decreases oxidative stress and ferroptosis or apoptosis in unilateral ureteral obstruction (UUO) mice, which decreases inflammatory responses and subsequently inhibits development of renal fibrosis. In conclusion, the inhibition of ferroptosis via regulation of oxidative stress may represent a novel method for treatment of renal fibrosis.

Inflammation

Inflammation is an immune response to exogenous or endogenous injury and contributes to the maintenance of tissue homeostasis under stressful conditions (54). A high inflammatory burden is associated with kidney damage. Results of a clinical study on type 2 diabetes demonstrated that the ratio of C-reactive protein expression to serum albumin is increased in patients with diabetic nephropathy compared with those without diabetic nephropathy (55). Using an adenine-induced mouse model of aging, the model group demonstrated increased levels of extensive tubular damage and fibrosis, as well as increased inflammatory responses, compared with groups of control (56). Inflammation is the main pathogenesis of diabetic kidney injury (DKI), and the monocyte to lymphocyte ratio (MLR) is considered a marker of inflammatory disease. microalbuminuria (MA) is the Microalbuminuria (MA) is the last reversible stage of DKI treatment, and in type 2 diabetic patients, MLR expression levels are significantly higher in the MA group compared to the normoalbuminuria (NA) group (57). Monocyte chemoattractant protein-1, macrophage colony-stimulating factor and neopterin levels are markedly increased in patients with chronic renal disease compared with controls (58). Moreover, numerous inflammatory cytokines, such as neuregulin (59), kidney injury molecule-1 (KIM-1) (60) and omentin (61) are associated with degree of kidney damage. In addition, interleukin (IL)-10 exerts anti-inflammatory effects. In a renal ischemia-reperfusion injury model, IL-10 knockout mice demonstrated decreased levels of renal function, upregulation of renal injury biomarkers, such as KIM-1, and increased expression of certain pro-inflammatory cytokines, compared with the control group (62). Therefore, renal damage is associated with inflammation.

Damage to renal tissue induces the inflammatory and fibrotic processes that aid in regeneration and repair (63). Results of previous studies demonstrated that macrophages are a potential therapeutic target for renal injury and fibrosis and play a significant role in the pathophysiology of kidney disease (64-66). Notably, renal fibrosis may be reversed as different subpopulations of macrophages in the kidney can either promote or inhibit deposition of extracellular matrix in the kidney (64). Inflammatory cell infiltration is a key characteristic of renal fibrosis (67). Renal tubular injury is considered a proinflammatory driving force in fibrosis. Following renal tubular injury, renal tubular epithelial cells (TECs) produce immune responses and release inflammatory mediators. The aggravation of inflammation leads to cell death, while cell death also has a strong pro-inflammatory effect, further worsening tubular injury, and continued inflammation and injury can lead to tubulointerstitial fibrosis (68). In conclusion, renal fibrosis and inflammation are associated with renal damage.

Due to increased permeability and rupture of the cell membrane during ferroptosis, associated contents, including damage-associated molecular pattern (DAMP) may be released, causing an inflammatory response and activation of the innate immune response. However, the specific mechanism requires further investigation (69). Necroinflammation associated with ferroptosis is observed acute kidney injury model in mice, as well as in GPX4-deficient knockout mouse models (70,71). In the kidneys of GPX4 knockout mice induced by tamoxifen, a large number of renal tubular cells died, and the release of cellular debris, mitochondria and even nuclei from ruptured cells into the tubular lumen could be observed at the histological level; this may be associated with DAMP (72). In ferroptotic tissues, F4/80 immunofluorescent staining has demonstrated that macrophages are markedly activated (73), releasing pro-inflammatory substances, thus triggering inflammatory responses.

However, to the best of our knowledge, the interaction between ferroptosis and inflammation in renal fibrosis remains unclear. Tectorigenin, a compound derived from the iris plant Belamcanda chinensis, is an active ingredient used in Traditional Chinese Medicine (74). Tectorigenin exhibits numerous pharmacological activities, such as anti-inflammatory and antioxidant properties, liver protection and diabetes control (75,76). Li et al (77) demonstrated that tectorigenin inhibits ferroptosis and fibrosis induced by external stimuli in primary TECs. Moreover, Fer-1, a ferroptosis inhibitor, inhibits the pro-fibrotic effect of TGF-β1-stimulated TECs, suggesting that tectorigenin may alleviate renal fibrosis via inhibition of ferroptosis in TECs (77). Results of a recent study also demonstrated that Fer-1 attenuates oxalate-induced TEC damage and renal fibrosis via inhibition of ferroptosis (78). Zhang et al (79) demonstrated that ferroptosis of TECs may be induced following UUO in mice, while liproxstatin-1 (Lip-1), a ferroptosis inhibitor, inhibits downregulation of GPX4 expression and ferroptosis in TECs and attenuates expression of pro-fibrotic factors in UUO mice. These results suggested that Lip-1 alleviates renal fibrosis in UUO mice via inhibition of ferroptosis in TECs. Moreover, Luo et al (80) demonstrated that obesity induces ferroptosis in the kidney while Fer-1 inhibits the development of high-fat diet-induced inflammation and fibrosis in renal tissue. Tocilizumab is an emerging interleukin-6 (IL6) receptor-targeting drug Yang et al (81) demonstrated that tocilizumab attenuates renal fibrosis in mice via inhibition of ferroptosis. Renal fibrosis is a common pathological process in diabetic nephropathy (82). Results of previous studies have demonstrated that expression levels of ACSL4 are increased in diabetic nephropathy mice and expression levels of GPX4 are decreased (83). Using the ACSL4 inhibitor rosiglitazone, both ferroptosis and production of pro-inflammatory cytokines are inhibited in TECs, preventing development of diabetic nephropathy (83). Zhou et al (84) confirmed that inhibiting ferroptosis in TECs reduces interstitial inflammation and renal fibrosis. Collectively, these results suggested that attenuating cellular inflammation development via inhibition of ferroptosis may be a novel approach for the treatment of renal fibrosis.

Autophagy

Autophagy refers to self-phagocytosis of cells, which removes misfolded proteins and damaged organelles in cells awaiting degradation, thereby maintaining cell homeostasis (85). Autophagy is a self-protection mechanism of eukaryotic cells (85). A previous study demonstrated that changes in autophagy activity are associated with renal fibrosis (86). The regulatory function of autophagy in fibrosis is associated with coordinated regulation of tubular cell death, interstitial inflammation and, in particular, production of pro-fibrotic secretory proteins (87). Results of a previous study demonstrated that, as a relatively recently discovered regulatory mode of cell death, ferroptosis differs from other regulatory modes such as autophagy, apoptosis, necrosis (2). Nonetheless, a more recent study demonstrated that ferroptosis and autophagy exhibit common regulators such as SLC7A11, GPX4, Nrf2 and heat shock protein β-1(88). The autophagy-related protein beclin 1 (BECN1) inhibits the function of system Xc- via formation of the BECN1/SLC7A11 complex and induces ferroptosis under the action of erastin and RSL3(84). Ferritinophagy is a type of cell-selective autophagy mediated by NCOA4. To facilitate the movement of intracellular ferritin to autophagy lysosomes and liberate free iron, NCOA4 functions as a selective autophagy receptor and binds to FTH1 of ferritin (89). Overexpression of NCOA4 increases ferritin degradation in cancer cells and fibroblasts, thereby promoting ferroptosis (90). Wang et al (91) demonstrated that expression of NCOA4 is increased in a 5/6 nephrectomy-induced CKD rat model Following the addition of ferroptosis inducer cisplatin or the ferroptosis inhibitor desferrioxamine mesylate, expression of NCOA4 is enhanced or attenuated, respectively. This treatment alters the progression of renal fibrosis. Therefore, ferritinophagy may induce ferroptosis in CKD and promote development of renal fibrosis. Consequently, inhibition of ferroptosis via regulation of autophagy may act as a novel therapeutic method in treatment of renal fibrosis.

4. Conclusion

Renal fibrosis is a common pathological state in almost all chronic and progressive kidney diseases, but effective measures for its clinical prevention and treatment are still not available. Ferroptosis is a novel regulatory cell death modality, and by summarizing the association between renal fibrosis and ferroptosis, we found that ferroptosis is involved in various biological processes such as oxidative stress, inflammation, and autophagy during renal fibrosis (Fig. 1). However, the specific molecular mechanism is still unclear, and further research is needed to investigate the role of ferroptosis in the development of renal fibrosis and to explore effective and highly targeted therapeutic measures against ferroptosis to provide new targets and more valuable therapeutic approaches for renal fibrosis research.

Acknowledgements

Not applicable.

Funding

Funding: The present study was funded by the National Natural Science Foundation of China (grant no. 82004165) and the Collaborative Public Relations Project Plan of Chinese and Western Medicine of Major and Difficult Diseases in Anhui Province (grant no. 2021-70).

Availability of data and materials

Not applicable.

Authors' contributions

HYZ, MC, LZ and YPW were responsible for the conceptualization of the present review. HYZ and MC were responsible for the original draft preparation. LZ and YPW were responsible for reviewing and editing the manuscript. LZ and YPW were responsible for funding acquisition. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhang X and Li X: Abnormal iron and lipid metabolism mediated ferroptosis in kidney diseases and its therapeutic potential. Metabolites. 12(58)2022.PubMed/NCBI View Article : Google Scholar

2 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012.PubMed/NCBI View Article : Google Scholar

3 

Dixon SJ and Stockwell BR: The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 10:9–17. 2014.PubMed/NCBI View Article : Google Scholar

4 

Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, Liuyang ZY, Roisman L, Zhang ST, Ayton S, et al: Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 22:1520–1530. 2017.PubMed/NCBI View Article : Google Scholar

5 

Lu X, Rudemiller NP, Ren J, Wen Y, Yang B, Griffiths R, Privratsky JR, Madan B, Virshup DM and Crowley SD: Opposing actions of renal tubular- and myeloid-derived porcupine in obstruction-induced kidney fibrosis. Kidney Int. 96:1308–1319. 2019.PubMed/NCBI View Article : Google Scholar

6 

Humphreys BD: Mechanisms of renal fibrosis. Annu Rev Physiol. 80:309–326. 2018.PubMed/NCBI View Article : Google Scholar

7 

Dolma S, Lessnick SL, Hahn WC and Stockwell BR: Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 3:285–296. 2003.PubMed/NCBI View Article : Google Scholar

8 

Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB and Jiang X: Role of mitochondria in ferroptosis. Mol Cell. 73:354–363.e3. 2019.PubMed/NCBI View Article : Google Scholar

9 

Lee YS, Lee DH, Choudry HA, Bartlett DL and Lee YJ: Ferroptosis-induced endoplasmic reticulum stress: Cross-talk between ferroptosis and apoptosis. Mol Cancer Res. 16:1073–1076. 2018.PubMed/NCBI View Article : Google Scholar

10 

Hirayama T, Miki A and Nagasawa H: Organelle-specific analysis of labile Fe(ii) during ferroptosis by using a cocktail of various colour organelle-targeted fluorescent probes. Metallomics. 11:111–117. 2019.PubMed/NCBI View Article : Google Scholar

11 

Lin X, Ping J, Wen Y and Wu Y: The mechanism of ferroptosis and applications in tumor treatment. Front Pharmacol. 11(1061)2020.PubMed/NCBI View Article : Google Scholar

12 

Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017.PubMed/NCBI View Article : Google Scholar

13 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11(88)2020.PubMed/NCBI View Article : Google Scholar

14 

Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, et al: FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 575:693–698. 2019.PubMed/NCBI View Article : Google Scholar

15 

Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020.PubMed/NCBI View Article : Google Scholar

16 

Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017.PubMed/NCBI View Article : Google Scholar

17 

Wen J, Chen H, Ren Z, Zhang P, Chen J and Jiang S: Ultrasmall iron oxide nanoparticles induced ferroptosis via beclin1/ATG5-dependent autophagy pathway. Nano Converg. 8(10)2021.PubMed/NCBI View Article : Google Scholar

18 

Galaris D, Barbouti A and Pantopoulos K: Iron homeostasis and oxidative stress: An intimate relationship. Biochim Biophys Acta Mol Cell Res. 1866(118535)2019.PubMed/NCBI View Article : Google Scholar

19 

Wang S, Luo J, Zhang Z, Dong D, Shen Y, Fang Y, Hu L, Liu M, Dai C, Peng S, et al: Iron and magnetic: New research direction of the ferroptosis-based cancer therapy. Am J Cancer Res. 8:1933–1946. 2018.PubMed/NCBI

20 

Dixon SJ, Winter GE, Musavi LS, Lee ED, Snijder B, Rebsamen M, Superti-Furga G and Stockwell BR: Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem Biol. 10:1604–1609. 2015.PubMed/NCBI View Article : Google Scholar

21 

Dai C, Chen X, Li J, Comish P, Kang R and Tang D: Transcription factors in ferroptotic cell death. Cancer Gene Ther. 27:645–656. 2020.PubMed/NCBI View Article : Google Scholar

22 

Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, et al: The cystine/glutamate antiporter system x(c)(-) in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 18:522–555. 2013.PubMed/NCBI View Article : Google Scholar

23 

Bannai S: Exchange of cystine and glutamate across plasma membrane of human fibroblasts. J Biol Chem. 261:2256–2263. 1986.PubMed/NCBI

24 

Gao M, Monian P, Quadri N, Ramasamy R and Jiang X: Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 59:298–308. 2015.PubMed/NCBI View Article : Google Scholar

25 

Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, Liao P, Zhou J, Zhang Q, Dow A, et al: Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9:1673–1685. 2019.PubMed/NCBI View Article : Google Scholar

26 

Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021.PubMed/NCBI View Article : Google Scholar

27 

Sato H, Shiiya A, Kimata M, Maebara K, Tamba M, Sakakura Y, Makino N, Sugiyama F, Yagami K, Moriguchi T, et al: Redox imbalance in cystine/glutamate transporter-deficient mice. J Biol Chem. 280:37423–37429. 2005.PubMed/NCBI View Article : Google Scholar

28 

Badgley MA, Kremer DM, Maurer HC, DelGiorno KE, Lee HJ, Purohit V, Sagalovskiy IR, Ma A, Kapilian J, Firl CEM, et al: Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 368:85–89. 2020.PubMed/NCBI View Article : Google Scholar

29 

Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH and Chang WC: Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 416:124–137. 2018.PubMed/NCBI View Article : Google Scholar

30 

Gan B: Mitochondrial regulation of ferroptosis. J Cell Biol. 220(e202105043)2021.PubMed/NCBI View Article : Google Scholar

31 

Kuhn H, Banthiya S and van Leyen K: Mammalian lipoxygenases and their biological relevance. Biochim Biophys Acta. 1851:308–330. 2015.PubMed/NCBI View Article : Google Scholar

32 

Qureshi T, Sørensen C, Berghuis P, Jensen V, Dobszay MB, Farkas T, Dalen KT, Guo C, Hassel B, Utheim TP, et al: The glutamine transporter Slc38a1 regulates GABAergic neurotransmission and synaptic plasticity. Cereb Cortex. 29:5166–5179. 2019.PubMed/NCBI View Article : Google Scholar

33 

Yang Y, Tai W, Lu N, Li T, Liu Y, Wu W, Li Z, Pu L, Zhao X, Zhang T and Dong Z: lncRNA ZFAS1 promotes lung fibroblast-to-myofibroblast transition and ferroptosis via functioning as a ceRNA through miR-150-5p/SLC38A1 axis. Aging (Albany NY). 12:9085–9102. 2020.PubMed/NCBI View Article : Google Scholar

34 

Chen X, Yu C, Kang R and Tang D: Iron metabolism in ferroptosis. Front Cell Dev Biol. 8(590226)2020.PubMed/NCBI View Article : Google Scholar

35 

Doll S and Conrad M: Iron and ferroptosis: A still ill-defined liaison. IUBMB Life. 69:423–434. 2017.PubMed/NCBI View Article : Google Scholar

36 

Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021.PubMed/NCBI View Article : Google Scholar

37 

Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, Sabatini DM, Birsoy K and Possemato R: NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 551:639–643. 2017.PubMed/NCBI View Article : Google Scholar

38 

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019.PubMed/NCBI View Article : Google Scholar

39 

Chen PH, Wu J, Ding CC, Lin CC, Pan S, Bossa N, Xu Y, Yang WH, Mathey-Prevot B and Chi JT: Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 27:1008–1022. 2020.PubMed/NCBI View Article : Google Scholar

40 

Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015.PubMed/NCBI View Article : Google Scholar

41 

Mazure NM: VDAC in cancer. Biochim Biophys Acta Bioenerg. 1858:665–673. 2017.PubMed/NCBI View Article : Google Scholar

42 

Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface JJ, et al: RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 447:864–868. 2007.PubMed/NCBI View Article : Google Scholar

43 

Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019.PubMed/NCBI View Article : Google Scholar

44 

McBean GJ: The transsulfuration pathway: A source of cysteine for glutathione in astrocytes. Amino Acids. 42:199–205. 2012.PubMed/NCBI View Article : Google Scholar

45 

Sedeek M, Nasrallah R, Touyz RM and Hébert RL: NADPH oxidases, reactive oxygen species, and the kidney: Friend and foe. J Am Soc Nephrol. 24:1512–1518. 2013.PubMed/NCBI View Article : Google Scholar

46 

Guerrero-Hue M, García-Caballero C, Palomino-Antolín A, Rubio-Navarro A, Vázquez-Carballo C, Herencia C, Martín-Sanchez D, Farré-Alins V, Egea J, Cannata P, et al: Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death. FASEB J. 33:8961–8975. 2019.PubMed/NCBI View Article : Google Scholar

47 

Linkermann A, Chen G, Dong G, Kunzendorf U, Krautwald S and Dong Z: Regulated cell death in AKI. J Am Soc Nephrol. 25:2689–2701. 2014.PubMed/NCBI View Article : Google Scholar

48 

Su H, Wan C, Song A, Qiu Y, Xiong W and Zhang C: Oxidative stress and renal fibrosis: Mechanisms and therapies. Adv Exp Med Biol. 1165:585–604. 2019.PubMed/NCBI View Article : Google Scholar

49 

Li X, Zou Y, Xing J, Fu YY, Wang KY, Wan PZ and Zhai XY: Pretreatment with roxadustat (FG-4592) attenuates folic acid-induced kidney injury through antiferroptosis via Akt/GSK-3 β/Nrf2 pathway. Oxid Med Cell Longev. 2020(6286984)2020.PubMed/NCBI View Article : Google Scholar

50 

Ide S, Kobayashi Y, Ide K, Strausser SA, Abe K, Herbek S, O'Brien LL, Crowley SD, Barisoni L, Tata A, et al: Ferroptotic stress promotes the accumulation of pro-inflammatory proximal tubular cells in maladaptive renal repair. Elife. 10(e68603)2021.PubMed/NCBI View Article : Google Scholar

51 

Feng X, Wang S, Sun Z, Dong H, Yu H, Huang M and Gao X: Ferroptosis enhanced diabetic renal tubular injury via HIF-1α/HO-1 pathway in db/db mice. Front Endocrinol (Lausanne). 12(626390)2021.PubMed/NCBI View Article : Google Scholar

52 

Liu B, Deng Q, Zhang L and Zhu W: Nobiletin alleviates ischemia/reperfusion injury in the kidney by activating PI3K/AKT pathway. Mol Med Rep. 22:4655–4662. 2020.PubMed/NCBI View Article : Google Scholar

53 

Lo YH, Yang SF, Cheng CC, Hsu KC, Chen YS, Chen YY, Wang CW, Guan SS and Wu CT: Nobiletin alleviates ferroptosis-associated renal injury, inflammation, and fibrosis in a unilateral ureteral obstruction mouse model. Biomedicines. 10(595)2022.PubMed/NCBI View Article : Google Scholar

54 

Yang L and Xia H: TRIM proteins in inflammation: From expression to emerging regulatory mechanisms. Inflammation. 44:811–820. 2021.PubMed/NCBI View Article : Google Scholar

55 

Bilgin S, Kurtkulagi O, Atak BM, Duman TT, Kahveci G, Khalid A and Aktas G: Does C-reactive protein to serum albumin ratio correlate with diabEtic nephropathy in patients with Type 2 dIabetes MEllitus? The CARE TIME study. Prim Care Diabetes. 15:1071–1074. 2021.PubMed/NCBI View Article : Google Scholar

56 

Jung SW, Kim DJ, Kim YG, Moon JY, Jeong KH and Lee SH: Renal aging resembles a continuum between normal and diseased kidneys that potentiates inflammatory response to injury. J Gerontol A Biol Sci Med Sci. 76:385–392. 2021.PubMed/NCBI View Article : Google Scholar

57 

Kocak MZ, Aktas G, Duman TT, Atak BM, Kurtkulagi O, Tekce H, Bilgin S and Alaca B: Monocyte lymphocyte ratio as a predictor of diabetic kidney injury in type 2 diabetes mellitus; The MADKID study. J Diabetes Metab Disord. 19:997–1002. 2020.PubMed/NCBI View Article : Google Scholar

58 

Musiał K and Zwolińska D: New markers of cell migration and inflammation in children with chronic kidney disease. Biomarkers. 24:295–302. 2019.PubMed/NCBI View Article : Google Scholar

59 

Kocak MZ, Aktas G, Atak BM, Duman TT, Yis OM, Erkus E and Savli H: Is neuregulin-4 a predictive marker of microvascular complications in type 2 diabetes mellitus? Eur J Clin Invest. 50(e13206)2020.PubMed/NCBI View Article : Google Scholar

60 

Kin Tekce B, Tekce H, Aktas G and Sit M: Evaluation of the urinary kidney injury molecule-1 levels in patients with diabetic nephropathy. Clin Invest Med. 37:E377–E383. 2014.PubMed/NCBI View Article : Google Scholar

61 

Tekce H, Tekce BK, Aktas G, Alcelik A and Sengul E: Serum omentin-1 levels in diabetic and nondiabetic patients with chronic kidney disease. Exp Clin Endocrinol Diabetes. 122:451–456. 2014.PubMed/NCBI View Article : Google Scholar

62 

Sakai K, Nozaki Y, Murao Y, Yano T, Ri J, Niki K, Kinoshita K, Funauchi M and Matsumura I: Protective effect and mechanism of IL-10 on renal ischemia-reperfusion injury. Lab Invest. 99:671–683. 2019.PubMed/NCBI View Article : Google Scholar

63 

Black LM, Lever JM and Agarwal A: Renal inflammation and fibrosis: A double-edged sword. J Histochem Cytochem. 67:663–681. 2019.PubMed/NCBI View Article : Google Scholar

64 

Tang PM, Nikolic-Paterson DJ and Lan HY: Macrophages: Versatile players in renal inflammation and fibrosis. Nat Rev Nephrol. 15:144–158. 2019.PubMed/NCBI View Article : Google Scholar

65 

Meng XM, Wang S, Huang XR, Yang C, Xiao J, Zhang Y, To KF, Nikolic-Paterson DJ and Lan HY: Inflammatory macrophages can transdifferentiate into myofibroblasts during renal fibrosis. Cell Death Dis. 7(e2495)2016.PubMed/NCBI View Article : Google Scholar

66 

Wei J, Xu Z and Yan X: The role of the macrophage-to-myofibroblast transition in renal fibrosis. Front Immunol. 13(934377)2022.PubMed/NCBI View Article : Google Scholar

67 

Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Tao J, Sharma K, Pullman J, et al: Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat Med. 21:37–46. 2015.PubMed/NCBI View Article : Google Scholar

68 

Liu BC, Tang TT, Lv LL and Lan HY: Renal tubule injury: A driving force toward chronic kidney disease. Kidney Int. 93:568–579. 2018.PubMed/NCBI View Article : Google Scholar

69 

Wen Q, Liu J, Kang R, Zhou B and Tang D: The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 510:278–283. 2019.PubMed/NCBI View Article : Google Scholar

70 

Von Mässenhausen A, Tonnus W and Linkermann A: Cell death pathways drive necroinflammation during acute kidney injury. Nephron. 140:144–147. 2018.PubMed/NCBI View Article : Google Scholar

71 

Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. 2014.PubMed/NCBI View Article : Google Scholar

72 

Martin-Sanchez D, Ruiz-Andres O, Poveda J, Carrasco S, Cannata-Ortiz P, Sanchez-Niño MD, Ruiz Ortega M, Egido J, Linkermann A, Ortiz A and Sanz AB: Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI. J Am Soc Nephrol. 28:218–229. 2017.PubMed/NCBI View Article : Google Scholar

73 

Shah R, Shchepinov MS and Pratt DA: Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 4:387–396. 2018.PubMed/NCBI View Article : Google Scholar

74 

Ha le M, Que do TN, Huyen do TT, Long PQ and Dat NT: Toxicity, analgesic and anti-inflammatory activities of tectorigenin. Immunopharmacol Immunotoxicol. 35:336–340. 2013.PubMed/NCBI View Article : Google Scholar

75 

Lee HU, Bae EA and Kim DH: Hepatoprotective effect of tectoridin and tectorigenin on tert-butyl hyperoxide-induced liver injury. J Pharmacol Sci. 97:541–544. 2005.PubMed/NCBI View Article : Google Scholar

76 

Pan CH, Kim ES, Jung SH, Nho CW and Lee JK: Tectorigenin inhibits IFN-gamma/LPS-induced inflammatory responses in murine macrophage RAW 264.7 cells. Arch Pharm Res. 31:1447–1456. 2008.PubMed/NCBI View Article : Google Scholar

77 

Li J, Yang J, Zhu B, Fan J, Hu Q and Wang L: Tectorigenin protects against unilateral ureteral obstruction by inhibiting Smad3-mediated ferroptosis and fibrosis. Phytother Res. 36:475–487. 2022.PubMed/NCBI View Article : Google Scholar

78 

Xie J, Ye Z, Li L, Xia Y, Yuan R, Ruan Y and Zhou X: Ferrostatin-1 alleviates oxalate-induced renal tubular epithelial cell injury, fibrosis and calcium oxalate stone formation by inhibiting ferroptosis. Mol Med Rep. 26(256)2022.PubMed/NCBI View Article : Google Scholar

79 

Zhang B, Chen X, Ru F, Gan Y, Li B, Xia W, Dai G, He Y and Chen Z: Liproxstatin-1 attenuates unilateral ureteral obstruction-induced renal fibrosis by inhibiting renal tubular epithelial cells ferroptosis. Cell Death Dis. 12(843)2021.PubMed/NCBI View Article : Google Scholar

80 

Luo Y, Chen H, Liu H, Jia W, Yan J, Ding W, Zhang Y, Xiao Z and Zhu Z: Protective effects of ferroptosis inhibition on high fat diet-induced liver and renal injury in mice. Int J Clin Exp Pathol. 13:2041–2049. 2020.PubMed/NCBI

81 

Yang L, Guo J, Yu N, Liu Y, Song H, Niu J and Gu Y: Tocilizumab mimotope alleviates kidney injury and fibrosis by inhibiting IL-6 signaling and ferroptosis in UUO model. Life Sci. 261(118487)2020.PubMed/NCBI View Article : Google Scholar

82 

Zhang Y, Jin D, Kang X, Zhou R, Sun Y, Lian F and Tong X: Signaling pathways involved in diabetic renal fibrosis. Front Cell Dev Biol. 9(696542)2021.PubMed/NCBI View Article : Google Scholar

83 

Wang Y, Bi R, Quan F, Cao Q, Lin Y, Yue C, Cui X, Yang H, Gao X and Zhang D: Ferroptosis involves in renal tubular cell death in diabetic nephropathy. Eur J Pharmacol. 888(173574)2020.PubMed/NCBI View Article : Google Scholar

84 

Zhou L, Xue X, Hou Q and Dai C: Targeting ferroptosis attenuates interstitial inflammation and kidney fibrosis. Kidney Dis (Basel). 8:57–71. 2021.PubMed/NCBI View Article : Google Scholar

85 

Mancias JD, Wang X, Gygi SP, Harper JW and Kimmelman AC: Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 509:105–109. 2014.PubMed/NCBI View Article : Google Scholar

86 

Zhang Z, Yao Z, Wang L, Ding H, Shao J, Chen A, Zhang F and Zheng S: Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy. 14:2083–2103. 2018.PubMed/NCBI View Article : Google Scholar

87 

Livingston MJ, Ding HF, Huang S, Hill JA, Yin XM and Dong Z: Persistent activation of autophagy in kidney tubular cells promotes renal interstitial fibrosis during unilateral ureteral obstruction. Autophagy. 12:976–998. 2016.PubMed/NCBI View Article : Google Scholar

88 

Li W, Feng G, Gauthier JM, Lokshina I, Higashikubo R, Evans S, Liu X, Hassan A, Tanaka S, Cicka M, et al: Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J Clin Invest. 129:2293–2304. 2019.PubMed/NCBI View Article : Google Scholar

89 

Masaldan S, Clatworthy SAS, Gamell C, Meggyesy PM, Rigopoulos AT, Haupt S, Haupt Y, Denoyer D, Adlard PA, Bush AI and Cater MA: Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 14:100–115. 2018.PubMed/NCBI View Article : Google Scholar

90 

Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ III, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016.PubMed/NCBI View Article : Google Scholar

91 

Wang J, Wang Y, Liu Y, Cai X, Huang X, Fu W, Wang L, Qiu L, Li J and Sun L: Ferroptosis, a new target for treatment of renal injury and fibrosis in a 5/6 nephrectomy-induced CKD rat model. Cell Death Discov. 8(127)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2023
Volume 25 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang HY, Cheng M, Zhang L and Wang YP: Ferroptosis and renal fibrosis: A new target for the future (Review). Exp Ther Med 25: 13, 2023
APA
Zhang, H.Y., Cheng, M., Zhang, L., & Wang, Y.P. (2023). Ferroptosis and renal fibrosis: A new target for the future (Review). Experimental and Therapeutic Medicine, 25, 13. https://doi.org/10.3892/etm.2022.11712
MLA
Zhang, H. Y., Cheng, M., Zhang, L., Wang, Y. P."Ferroptosis and renal fibrosis: A new target for the future (Review)". Experimental and Therapeutic Medicine 25.1 (2023): 13.
Chicago
Zhang, H. Y., Cheng, M., Zhang, L., Wang, Y. P."Ferroptosis and renal fibrosis: A new target for the future (Review)". Experimental and Therapeutic Medicine 25, no. 1 (2023): 13. https://doi.org/10.3892/etm.2022.11712