Open Access

Ferroptosis: A potential therapeutic target in autoimmune disease (Review)

  • Authors:
    • Liang Shen
    • Xiaohan Wang
    • Changlin Zhai
    • Yunqing Chen
  • View Affiliations

  • Published online on: June 15, 2023     https://doi.org/10.3892/etm.2023.12067
  • Article Number: 368
  • Copyright: © Shen 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 distinct type of regulated cell death characterized by iron overload and lipid peroxidation. Ferroptosis is regulated by numerous factors and controlled by several mechanisms. This cell death type has a relationship with the immune system, which may be regulated by damage‑associated molecular patterns. Ferroptosis participates in the progression of autoimmune diseases, including autoimmune hepatitis, rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, multiple sclerosis, Parkinson's Disease, psoriasis and insulin‑dependent diabetes mellitus. The present review summarizes the role of ferroptosis in autoimmune disorders and discusses ferroptosis as a potential therapeutic target for autoimmune disease.

1. Introduction

Since being named in 2012 by Dixon et al (1), ferroptosis has been probed in a wide range of pathologies and proposed as a novel therapeutic strategy for numerous diseases, including cancer (2,3), ischemia-reperfusion injury (4,5) and neurodegenerative disorder (6,7). Autoimmune disease, particularly rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), insulin-dependent diabetes mellitus (IDDM) and inflammatory bowel disease (IBD) (8,9), are heterogeneous with regard to prevalence, manifestation and pathogenesis. Accumulating evidence in recent times has shown an association of ferroptosis with the pathogenesis and development of autoimmune diseases (10-12). The present review aimed to summarize the association between ferroptosis and autoimmune disease, focusing on potential mechanisms and therapeutic strategies.

2. Overview of ferroptosis

Ferroptosis was proposed in 2012 as a distinctive type of non-apoptotic cell death (1) characterized by iron overload and lipid peroxidation. This differs from other forms of regulated cell death. The primary feature of ferroptosis is mitochondrial shrinkage, which occurs alongside an increase in mitochondrial membrane density and degeneration of mitochondrial crista but with no changes in morphology of the nucleus (1).

Several pathways, including the metabolism of iron, acid and lipid, have been implicated in ferroptosis (13). Excessive cytosolic Fe2+ catalyzes the Fenton reaction and activates iron-dependent metabolic enzymes, leading to production of highly reactive hydroxyl radicals and oxidized polyunsaturated fatty acids (PUFAs), which results in the promotion of the accumulation of lipid reactive oxygen species (ROS) and ferroptosis. The metabolism of amino acids, especially the system Xc/glutathione (GSH)/GSH peroxidase 4 (GPX4) axis, is key to eliminating lipid ROS, with GPX4 regarded as a key regulator of ferroptosis. Additionally, GPX4-independent pathways, such as the NADPH/ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 and the GTP cyclohydrolase-1/tetrahydrobiopterin/dihydrobiopterin axes, have also been implicated in the ferroptosis process in the past few years (14-16) (Fig. 1).

3. Association between ferroptosis and the immune system

Two types of immunity exist in the body, innate and adaptive. The innate immune system detects invading pathogens, while the adaptive immune system promotes a specific and long-lasting protection against infection. Innate immune cells mainly include dendritic cells, macrophages and neutrophils, while the adaptive immune system generally contains T and B lymphocytes and nature killer (NK) cells (17).

In the last few years, more evidence has revealed a close association between ferroptosis and the immune system (18,19). Notably, autoimmune disease is initiated and propagated by the activation of self-antigen-reactive T cells (20), pointing to the crucial role of T cells in autoimmunity. In a study investigating ferroptosis in immunity, both antigen-specific CD8+ and CD4+ T cells failed to expand and protect infection in T cell-specific GPX4-deficient mice (TΔGpx4/ΔGpx4), whereas GPX4-deficient T cells rapidly accumulated lipid peroxides and underwent ferroptosis in vitro (21). Ferroptosis was found to be involved in immunotherapy-activated CD8+ T cells, with increased ferroptosis contributing to the anti-tumor efficacy of immunotherapy (22). Recently, the homeostasis of follicular helper T cells, a specialized subset of CD4+ T cells, was also shown to be regulated by ferroptosis (23).

Key features of ferroptosis, iron overload and lipid peroxidation, participate in immunity. Iron overload increases oxidative stress and DNA damage in T cells, leading to immune dysfunction (24), while lipid peroxidation is associated with intracellular ROS in regulatory T cells (25). Ferroptosis affects the viability of B cells, with Muri et al (26) demonstrating that GPX4 is key to the development, maintenance and responses of B1 and marginal zone B cells via suppression of ferroptosis. Moreover, the ferroptosis inducer erastin increases lipid peroxidation and promotes peripheral blood mononuclear cell proliferation and differentiation into B and NK cells (27).

In addition to its links to the adaptive immune system, ferroptosis also plays a key role in innate immunity. In tumor cells, exogenous circularly polarized magnetic field-induced ferroptosis leads to the maturation of dendritic cells (28) and in immune-competent mice, ferroptosis promotes phenotypical maturation of bone-marrow-derived dendritic cells (29). With regard to other innate immune cells, ferroptosis is associated with the infiltration of macrophages and neutrophils (30), while also regulating polarization of macrophages (31) and the recruitment of neutrophils (32).

4. Damage-associated molecular patterns (DAMPs)

Although the pathology of autoimmune disease is complex, it is hypothesized that inflammation serves a key role in autoimmunity (33). DAMPs, endogenous molecules released by damaged tissue or dying cells, have been proved to be detrimental in inflammatory response and lead to the development of inflammatory disorders (34,35). Autoimmune diseases, such as SLE (36) and IBD (37), are among the inflammatory disorders initiated by DAMPs. In the host, DAMPs either activate innate immune cells, leading to release of various cytokines and chemokines and activation of adaptive immune responses, or stimulate adaptive immune cells directly (35).

As a key part of regulated cell death, ferroptosis can stimulate the release of DAMPs. Adenosine triphosphate (ATP) and high mobility group box 1 (HMGB1), two well-characterized DAMPs, are released along during ferroptosis in murine fibrosarcoma MCA205 or glioma GL261(29) and p53 R273H-expressing non-small cell lung cancer cells (38). Cotreatment with erastin and celastrol initiates expression of heat shock proteins (HSPs) (39). Using the immunoprecipitation assay, an interaction between HSP90 and GPX4 has been demonstrated in a model of acute kidney injury (AKI) (40). Another DAMP, calreticulin, also participates in ferroptosis. In the head and neck squamous cell carcinoma, Zhao et al (41) found ferroptosis reverses immunosuppressive microenvironments by releasing calreticulin and HMGB1, while Van Loenhout et al (42) demonstrated that auranofin and plasma-treated PBS mixture-induced ferroptosis led to a significant increase in calreticulin, ATP and HMGB1. The aforementioned reports point to a close link between ferroptosis and DAMPs, which may partly explain the mechanism of ferroptosis-mediated autoimmunity.
Autoimmune diseases are also associated with cytokines and chemokines. In a mouse AKI model, ferrostatin-1 was shown to prevent upregulation of IL-33(43). Additionally, liproxstatin-1 alleviates radiation-induced lung fibrosis via the downregulation of TGF-β1(44). Ferroptosis, therefore, could have an intimate relationship with autoimmune disease.

5. Ferroptosis and autoimmune disease

Autoimmune diseases are complicated and characterized by the development of specific autoantibodies and the presence of autoreactive T cells, leading to the impairment of sustained immune responses and organs (45). Recent studies have highlighted the association between ferroptosis and autoimmune disease (Table I).

Table I

Role of ferroptosis in autoimmune disease.

Table I

Role of ferroptosis in autoimmune disease.

Diseaseeffect Ferroptosis(Refs.)
Autoimmune hepatitisPromote(47,48)
Rheumatoid arthritisInconsistent(51-54)
Systemic lupus erythematosusPromoting(60)
Inflammatory bowel diseasePromoting(63-65)
Multiple sclerosisPromoting(70,71)
Parkinson's diseasePromoting(76-79)
PsoriasisPromoting(81,82)
Insulin-dependent diabetes mellitusPromoting(88)

6. Ferroptosis and autoimmune hepatitis (AIH)

AIH is an immune-mediated inflammatory liver disorder characterized by histological abnormality, as well as elevated aspartate aminotransferase, alanine aminotransferase and total IgG and the presence of autoantibodies (46).

In concanavalin A (ConA)-induced hepatitis, redox-active iron accumulation and malondialdehyde (MDA) are detected in the hepatic tissues of mice. Moreover, the expression of GPX4 and system xc- is markedly decreased in the liver of ConA-treated mice and is accompanied by the downregulation of caveolin-1(47). In LO2 hepatocyte cell line, the overexpression of caveolin-1 results in the upregulation of the expression of system xc-, suggesting that cavelolin-1 protects against ConA-induced AIH by inhibiting ferroptosis. In another study, levels of cyclooxygenase2 and acyl-coenzyme A synthase long-chain family member 4 (ACSL4) were shown to be upregulated in the liver tissue of S100-induced AIH model mice, while the levels of GPX4 and ferritin heavy chain 1 (FTH1) are downregulated (48). In addition, GPX4 knockdown via adeno-associated virus injection aggravates severity of S100-induced AIH. The aforementioned studies suggested that ferroptosis is a possible mediator of AIH.

7. Ferroptosis and RA

RA is the most common autoimmune inflammatory arthritis in adults and is characterized by chronic destructive synovitis and multisystem disorder (49,50).

In the rheumatoid synovium and synovial fluid of patients with RA, the levels of iron accumulation and lipid peroxidation increase, while in a collagen-induced arthritis (CIA) mouse model, selectively targeting fibroblasts in vivo to induce ferroptosis attenuates arthritis progression, indicating that ferroptosis inducers serve as candidates for RA treatment (51). Sulfasalazine, a U.S. Food and Drug Administration-approved RA drug, is an effective inducer of ferroptosis (52). As shown by Ling et al (53), the expression of ACSL4 declines, while the expression of FTH1, GPX4, and cystine/glutamate antiporter solute carrier family 7 member 11 are increased in the RA synovium of CIA model mouse compared with healthy control (53). Luo and Zhang (54) showed that in a lipopolysaccharide-induced synovitis cell model, MDA levels are increased, whereas GPX4 levels are decreased, representing an increase in ferroptosis in human synoviocytes; inhibition of ferroptosis may be a new therapeutic strategy for synovitis (54). Further studies are required to identify the exact role of ferroptosis in RA.

8. Ferroptosis and SLE

SLE is a multisystem autoimmune disease characterized by formation of autoantibodies, deposition of immune complexes and inflammation that primarily presents in women of reproductive age. The pathogenetic mechanisms of SLE are complex and this disorder is prone to relapse and remissions, leading to considerable morbidity and mortality (55,56).

Previous studies have examined the association between iron metabolism and SLE (57-59). Li et al (60) investigated the direct association between ferroptosis and SLE; the study demonstrated ferroptosis of neutrophils in lupus-prone mice and patients with SLE and hypothesized that neutrophil ferroptosis as an essential driver of neutropenia in SLE and treatment using specific ferroptosis inhibitors may ameliorate SLE severity and symptoms (60).

9. Ferroptosis and IBD

IBD, including Crohn's disease (CD) and ulcerative colitis (UC), is a complex chronic inflammation disorder that arises due to dysregulated immune response (61). Smoking, diet, lifestyle and behavior, as well as gut microbiota, are all key contributors to disease pathogenesis (62).

Mayr et al (63) found GPX4 activity is impaired and lipid peroxidation is augmented in small intestinal epithelial cells (IECs) of patients with CD. They also found that PUFA exposure induces lipid peroxidation and cytokine production by GPX4 small-interfering RNA IECs, while the genetic deletion of ACSL4 abrogates PUFA-induced cytokine production, suggesting that inflammatory cytokine production in IECs of patients with CD is driven by ferroptotic mechanisms (63). Xu et al (64) demonstrated significantly downregulated GPX4 and notably upregulated ACSL4 expression in the colonic biopsy specimens of patients with CD. In addition, MDA content and prostaglandin-endoperoxide synthase 2 (PTGS2) levels are higher in colon samples from mice with trinitrobenzene sulfonic acid-induced colitis, pointing to a close association between ferroptosis and CD (64).

Ferroptosis has also been investigated in UC, with Xu et al (65) reporting its involvement in IEC death in UC (65). The aforementioned study found several ferroptosis-associated genes to be remarkably down- or upregulated in human colonic biopsy samples from patients with UC, while PTGS2 is elevated and GPX4 diminished in colonic IECs from experimental colitis mice. Preventing ferroptosis through inhibiting the Nrf2/heme oxygenase-1 signaling pathway may be a valuable approach to inhibit progression of UC in dextran sulfate sodium (DSS)-induced experimental colitis mice (66,67). The aforementioned findings suggested that ferroptosis is involved in IBD and could serve as a therapeutic target.

10. Ferroptosis and MS

MS is considered an autoimmune disorder of the central nervous system that is characterized by inflammation, demyelination and degeneration (68,69). mRNA levels of GPX4 in the brain of patients with MS are decreased, while the levels of GPX4 mRNA and protein are decreased and lipid peroxidation is enhanced in an experimental autoimmune encephalomyelitis mouse model (70). Jhelum et al (71) investigated the underlying mechanism of cuprizone (CZ), a copper chelator, used to induce oligodendrocyte (OL) cell loss and demyelination, revealing that CZ treatment resulted in an increase in mRNA expression of nuclear receptor coactivator 4, transferrin receptor 1 and PTGS2, as well as lipid peroxidation, and a decrease in the expression of GPX4 and system xc- in the brain tissue of experimental mice. Additionally, the CZ-induced loss of OL and demyelination was prevented by ferrostatin-1(71). These results indicated that ferroptosis is a potential therapeutic target for MS.

11. Ferroptosis and Parkinson's disease (PD)

PD is one of the most common types of neurodegenerative disorder (72) and has also been proposed as an autoimmune disease (73). Numerous studies have examined the association between ferroptosis and PD (7,74,75).

In 2016, Do Van et al (7) reported the role of ferroptosis in PD for the first time, finding ferroptosis components in PD neuropathology. Moreover, the aforementioned study found dopaminergic neuronal loss is inhibited by ferroptosis-specific inhibitors ferostatin-1 and liproxstatin-1 and that modulation of the ferroptotic signaling cascade is a possible target for drug candidates for PD. Ferroptosis occurs in the pathology of PD and they share several hallmarks, including iron overload, lipid peroxidation and decreased GSH levels (76-78). Recently, Zuo et al (79) demonstrated that paraquat, a neurotoxin that increases the risk of PD, significantly induces iron accumulation in the cytoplasm and mitochondria of SH-SY5Y human neuroblastoma cells via the ferritinophagy pathway; however, ferritinophagy-mediated ferroptosis is significantly ameliorated by ferrostatin-1, pointing to the inhibition of ferroptosis as a potential new strategy for the prevention of neurotoxicity or PD (79). Reagents targeting ferroptosis could be used in the treatment of PD in the future.

12. Ferroptosis and psoriasis

Psoriasis is a chronic immune-mediated inflammatory skin disease characterized by hyperproliferation of keratinocytes and excessive infiltration of immune cells. Currently, it is considered a systemic disease associated with metabolic, arthritic and cardiovascular comorbidities (80).

A previous study showed a significant reduction in GPX4 and elevation in Nrf2 downstream targets in psoriatic skin lesions compared with samples from healthy patients (81). Additionally, the mRNA levels of ACSL4, PTGS2 and TFR are much higher in psoriasis lesions than in healthy controls. Furthermore, in an imiquimod (IMQ)-induced mouse model of psoriasis, immunohistochemical analysis uncovered notably increased ACSL4 levels and markedly decreased GPX4 levels in the basal epidermal layer and ferrostatin-1 treatment attenuated IMQ-induced psoriasis-like dermatitis (82). Ferroptosis is, therefore, a potential physiological mechanism for eliminating inflammatory response in psoriasis.

13. Ferroptosis and IDDM

IDDM is a chronic disorder stemming from autoimmune damage of pancreatic β cells (83). While ferroptosis is involved in cell death of the myocardium and renal tubules during diabetes (84-87), the role of ferroptosis in the death of β cells is unknown. In 2018, Bruni et al reported massive ferroptosis in pancreatic islets isolated from IDDM patients, whereas the transplantable number of islet equivalents increased following addition of ferrostatin (88). Along with evidence that ferroptosis is induced in rat pancreatic β cells after exposure to tert-butyl hydroperoxide (89), ferroptosis can be considered a possible mode of β cell destruction. However, more studies are required to determine the link between ferroptosis and β cell death.

14. Ferroptosis as a therapeutic target for autoimmune disease

As aforementioned, there is an association between ferroptosis and autoimmune disease. Therefore, targeting ferroptosis is a promising therapeutic option for autoimmune disease. Ferroptosis can primarily be inhibited by iron chelators and lipophilic antioxidants (1). The present review summarizes anti-ferroptosis agents and their potential benefits in the treatment of autoimmune disorder (Table II).

Table II

Therapeutic options for autoimmune disease.

Table II

Therapeutic options for autoimmune disease.

ReagentMechanismDisease(Refs.)
DeferoxamineIron chelationRA, PD(90,94)
DeferiproneIron chelationMS, PD, IBD(67,95,96)
Ferrostatin-1Peroxidation inhibitionAIH, IBD, PD(7,48,64)
Liproxstatin-1Peroxidation inhibitionIBD, PD(7,67)
SeleniumPeroxidation inhibitionMS, IBD, PD, psoriasis(102-104,107)
N-acetylcysteinePeroxidation inhibitionRA, SLE(108,109)
PolyphenolPeroxidation inhibitionRA, IBD, IDDM, PD(111-116)
α-tocopherolPeroxidation inhibitionRA, MS, PD(100-102)

[i] RA, rheumatoid arthritis; PD, Parkinson's disease; MS, multiple sclerosis; IBD, inflammatory bowel disease; SLE, systemic lupus erythematosus; IDDM, insulin-dependent diabetes mellitus; AIH, autoimmune hepatitis.

15. Iron chelators

Deferoxamine (DFO) has been investigated in the treatment of several types of autoimmune disease. In patients with RA, DFO prevents synovial injury (90) and improves anemia (91). A pilot study showed that patients with MS tolerate a short course of DFO therapy relatively well (92), however, no effect on disease progression has been noted (93). In addition to its effect against RA and MS, DFO has been found to ameliorate motor defects and pathology in a PD rat model (94). Deferiprone (DFP), another iron chelator, suppresses disease activity in a mouse model of MS (95). Additionally, DFP reportedly improves motor performance of patients with PD in a phase II clinical trial (96). DFP can ameliorate DSS-induced UC in a mouse model by suppressing ferroptosis (67). The aforementioned studies indicate that iron chelators are promising therapeutic options for autoimmune disease. However, larger clinal trials are needed to determine the value of iron chelators in the therapy of autoimmune disease.

16. Lipophilic antioxidants

Ferrostatin-1 and liproxstatin-1 are well-known inhibitors of ferroptosis. Numerous studies have investigated these ferroptosis inhibitors in autoimmune diseases, including AIH, IBD and PD (48,97). To the best of our knowledge, however, examinations have yet to be conducted in models other than experimental mouse models. Hence, clinical trials must be performed to explore their roles in patients.

Vitamin E is a key lipid soluble antioxidant that can suppress ferroptosis by inhibiting 15-lipoxygenase (98). Reports show that supplementation with vitamin E relieves joint pain in patients with RA (99). In patients with SLE, vitamin E is said to suppress autoantibody production. Moreover, vitamin E improves functional capacity and gait parameters in patients with relapsing-remitting MS (100) and improves clinical signs and metabolic status in patients with PD (101). Furthermore, supplementation with vitamin E is a feasible option for the management of patients with severe forms of psoriasis as it decreases the markers of oxidative stress (102). Vitamin E treatment, therefore, may be a therapeutic option for autoimmune diseases.

Selenium, an essential trace element with antioxidant properties, has been assessed as a potential treatment for autoimmune diseases. Supplementation with selenium relieves inflammatory reaction in patients with MS (103), IBD (104) and psoriasis (102). However, it has shown no significant clinical benefit against RA (105,106). In preclinical studies, selenium decreases loss of dopamine and slows the progression of neurodegeneration during PD (107).

N-acetylcysteine, a pharmaceutical drug with an anti-ferroptosis property, has been investigated in the treatment of RA and SLE. In patients with RA, the oral administration of N-acetylcysteine relieves severity of joint pain and improves physical performance (108). In patients with SLE, N-acetylcysteine inhibits lupus disease activity (109).

Polyphenols are natural antioxidants that prevent ferroptosis owing to their ROS scavenging property (110). Resveratrol, a well-studied polyphenol, decreases disease activity score assessment for 28 joints in patients with RA (111), decreases the clinical colitis activity index score and improves quality of life in patients with UC (112,113) and exerts antidiabetic and antioxidant effects in patients with IDDM (114). Other polyphenols, such as pomegranate juice could alleviate disease activity of patients with RA (115), and licorice could improve symptoms in patients with PD (116).

Although the aforementioned experiments posited lipophilic antioxidants as having potential role in the therapy of autoimmune disease, larger and more in-depth studies are required to determine the exact impact of various polyphenols in the treatment of autoimmune disorder.

17. Conclusion

The present review summarized research on ferroptosis in autoimmune disorders and discussed ferroptosis as a promising therapeutic target. Although autoimmune diseases are heterogeneous in manifestation, there are commonalities between these disorders with respect to ferroptosis. Among these key commonalities is inflammation (117,118). As an important part of regulated cell death, ferroptosis stimulates release of DAMPs and inflammatory cytokines, leading to activation of immune response and eventually promoting the development of autoimmune disease.

Recently, the role of ferroptosis in autoimmune diseases has been reviewed. Fan et al (11) highlighted crosstalk between ferroptosis and different immune cells and discussed the role of ferroptosis in autoimmune disease and Lai et al (10) also discussed how ferroptosis contributes to the pathogenesis of SLE, RA and IBD. However, the autoimmune diseases included in the aforementioned reviews are relatively limited and did not summarize the association between ferroptosis and autoimmune response. Hence, the present review is more comprehensive and may provide more information about the association between ferroptosis and autoimmune disease.

Even though recent evaluations have investigated ferroptosis in autoimmune disorders, the association between this cell death type and autoimmune diseases is relatively undeveloped. Therefore, more studies are required to determine the association between ferroptosis and autoimmune disease, including Graves' disease, Hashimoto thyroiditis, coeliac disease, Addison disease and autoimmune myocarditis and polyendocrine syndrome type 2. Additionally, although numerous ferroptosis-related reagents have been investigated in the treatment of various autoimmune diseases, the reported efficacy pertains mainly to basic studies, patient sample sizes and follow-up periods were relatively limited. Hence, larger clinical trials must be performed to highlight and confirm the treatment values of ferroptosis-associated regents.

In summary, ferroptosis plays a critical role in the pathogenesis of autoimmune diseases and is a promising therapeutic target for autoimmune diseases.

Acknowledgements

Figures were produced by Figdraw (www.figdraw.com), we express our gratitude.

Funding

Funding: The present study was supported by the Zhejiang Provincial Science Foundation of China (grant no. LGF21H020006), the Pioneer Innovation Team of Jiaxing Institute of Atherosclerotic Diseases (grant no. XFCX-DMYH), Program of the First Hospital of Jiaxing (grant no. 2021-YA-001), the Jiaxing Key Laboratory of Arteriosclerotic Diseases (grant no. 2020-dmzdsys) and Jiaxing Key Laboratory of Virus-mediated Infectious Diseases (grant no. 2021-bdzdsys).

Availability of data and materials

Not applicable.

Authors' contributions

LS and XW drafted the manuscript. CZ edited the manuscript. YC designed the study and revised the manuscript. 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 

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

2 

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

3 

Lee JY, Nam M, Son HY, Hyun K, Jang SY, Kim JW, Kim MW, Jung Y, Jang E, Yoon SJ, et al: Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc Natl Acad Sci USA. 117:32433–32442. 2020.PubMed/NCBI View Article : Google Scholar

4 

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

5 

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. Natl Acad Sci USA. 116:2672–2680. 2019.PubMed/NCBI View Article : Google Scholar

6 

Park MW, Cha HW, Kim J, Kim JH, Yang H, Yoon S, Boonpraman N, Yi SS, Yoo ID and Moon JS: NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer's diseases. Redox Biol. 41(101947)2021.PubMed/NCBI View Article : Google Scholar

7 

Do Van B, Gouel F, Jonneaux A, Timmerman K, Gelé P, Pétrault M, Bastide M, Laloux C, Moreau C, Bordet R, et al: Ferroptosis, a newly characterized form of cell death in Parkinson's disease that is regulated by PKC. Neurobiol Dis. 94:169–178. 2016.PubMed/NCBI View Article : Google Scholar

8 

Hayter SM and Cook MC: Updated assessment of the prevalence, spectrum and case definition of autoimmune disease. Autoimmun Rev. 11:754–765. 2012.PubMed/NCBI View Article : Google Scholar

9 

Cho JH and Gregersen PK: Genomics and the multifactorial nature of human autoimmune disease. N Engl J Med. 365:1612–1623. 2011.PubMed/NCBI View Article : Google Scholar

10 

Lai B, Wu CH, Wu CY, Luo SF and Lai JH: Ferroptosis and autoimmune diseases. Front Immunol. 13(916664)2022.PubMed/NCBI View Article : Google Scholar

11 

Fan J, Jiang T and He D: Emerging insights into the role of ferroptosis in the pathogenesis of autoimmune diseases. Front Immunol. 14(1120519)2023.PubMed/NCBI View Article : Google Scholar

12 

Ohl K, Rauen T and Tenbrock K: Dysregulated neutrophilic cell death in SLE: A spotlight on ferroptosis. Signal Transduct Target Ther. 6(392)2021.PubMed/NCBI View Article : Google Scholar

13 

Hu H, Chen Y, Jing L, Zhai C and Shen L: The link between ferroptosis and cardiovascular diseases: A novel target for treatment. Front Cardiovasc Med. 8(710963)2021.PubMed/NCBI View Article : Google Scholar

14 

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

15 

Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, et al: GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 6:41–53. 2020.PubMed/NCBI View Article : Google Scholar

16 

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

17 

Iwasaki A and Medzhitov R: Control of adaptive immunity by the innate immune system. Nat Immunol. 16:343–353. 2015.PubMed/NCBI View Article : Google Scholar

18 

Yang Y, Wang Y, Guo L, Gao W, Tang TL and Yan M: Interaction between macrophages and ferroptosis. Cell Death Dis. 13(355)2022.PubMed/NCBI View Article : Google Scholar

19 

Stockwell BR and Jiang X: A physiological function for ferroptosis in tumor suppression by the immune system. Cell Metab. 30:14–15. 2019.PubMed/NCBI View Article : Google Scholar

20 

Akirav EM, Ruddle NH and Herold KC: The role of AIRE in human autoimmune disease. Nat Rev Endocrinol. 7:25–33. 2011.PubMed/NCBI View Article : Google Scholar

21 

Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW and Kopf M: T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 212:555–568. 2015.PubMed/NCBI View Article : Google Scholar

22 

Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, et al: CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 569:270–274. 2019.PubMed/NCBI View Article : Google Scholar

23 

Yao Y, Chen Z, Zhang H, Chen C, Zeng M, Yunis J, Wei Y, Wan Y, Wang N, Zhou M, et al: Selenium-GPX4 axis protects follicular helper T cells from ferroptosis. Nat Immunol. 22:1127–1139. 2021.PubMed/NCBI View Article : Google Scholar

24 

Shaw J, Chakraborty A, Nag A, Chattopadyay A, Dasgupta AK and Bhattacharyya M: Intracellular iron overload leading to DNA damage of lymphocytes and immune dysfunction in thalassemia major patients. Eur J Haematol. 99:399–408. 2017.PubMed/NCBI View Article : Google Scholar

25 

Anupam K, Kaushal J, Prabhakar N and Bhatnagar A: Effect of redox status of peripheral blood on immune signature of circulating regulatory and cytotoxic T cells in streptozotocin induced rodent model of type I diabetes. Immunobiology. 223:586–597. 2018.PubMed/NCBI View Article : Google Scholar

26 

Muri J, Thut H, Bornkamm GW and Kopf M: B1 and marginal zone B cells but not follicular B2 cells require Gpx4 to prevent lipid peroxidation and ferroptosis. Cell Rep. 29:2731–2744.e4. 2019.PubMed/NCBI View Article : Google Scholar

27 

Wang D, Xie N, Gao W, Kang R and Tang D: The ferroptosis inducer erastin promotes proliferation and differentiation in human peripheral blood mononuclear cells. Biochem Biophys Res Commun. 503:1689–1695. 2018.PubMed/NCBI View Article : Google Scholar

28 

Yu B, Choi B, Li W and Kim DH: Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat Commun. 11(3637)2020.PubMed/NCBI View Article : Google Scholar

29 

Efimova I, Catanzaro E, Van der Meeren L, Turubanova VD, Hammad H, Mishchenko TA, Vedunova MV, Fimognari C, Bachert C, Coppieters F, et al: Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 8(e001369)2020.PubMed/NCBI View Article : Google Scholar

30 

Zhang Z, Qiu X, Yan Y, Liang Q, Cai Y, Peng B, Xu Z and Xia F: Evaluation of ferroptosis-related gene AKR1C1 as a novel biomarker associated with the immune microenvironment and prognosis in breast cancer. Int J Gen Med. 14:6189–6200. 2021.PubMed/NCBI View Article : Google Scholar

31 

Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, Zeh HJ, Kang R, Wang J and Tang D: Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 16:2069–2083. 2020.PubMed/NCBI View Article : Google Scholar

32 

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

33 

Hall SW and Cooke A: Autoimmunity and inflammation: Murine models and translational studies. Mamm Genome. 22:377–389. 2011.PubMed/NCBI View Article : Google Scholar

34 

Roh JS and Sohn DH: Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 18(e27)2018.PubMed/NCBI View Article : Google Scholar

35 

Gong T, Liu L, Jiang W and Zhou R: DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 20:95–112. 2020.PubMed/NCBI View Article : Google Scholar

36 

Schaper F, Westra J and Bijl M: Recent developments in the role of high-mobility group box 1 in systemic lupus erythematosus. Mol Med. 20:72–79. 2014.PubMed/NCBI View Article : Google Scholar

37 

Boyapati RK, Dorward DA, Tamborska A, Kalla R, Ventham NT, Doherty MK, Whitfield PD, Gray M, Loane J, Rossi AG, et al: Mitochondrial DNA is a pro-inflammatory damage-associated molecular pattern released during active IBD. Inflamm Bowel Dis. 24:2113–2122. 2018.PubMed/NCBI View Article : Google Scholar

38 

Freire Boullosa L, Van Loenhout J, Flieswasser T, De Waele J, Hermans C, Lambrechts H, Cuypers B, Laukens K, Bartholomeus E, Siozopoulou V, et al: Auranofin reveals therapeutic anticancer potential by triggering distinct molecular cell death mechanisms and innate immunity in mutant p53 non-small cell lung cancer. Redox Biol. 42(101949)2021.PubMed/NCBI View Article : Google Scholar

39 

Liu M, Fan Y, Li D, Han B, Meng Y, Chen F, Liu T, Song Z, Han Y, Huang L, et al: Ferroptosis inducer erastin sensitizes NSCLC cells to celastrol through activation of the ROS-mitochondrial fission-mitophagy axis. Mol Oncol. 15:2084–2105. 2021.PubMed/NCBI View Article : Google Scholar

40 

Chen C, Wang D, Yu Y, Zhao T, Min N, Wu Y, Kang L, Zhao Y, Du L, Zhang M, et al: Legumain promotes tubular ferroptosis by facilitating chaperone-mediated autophagy of GPX4 in AKI. Cell Death Dis. 12(65)2021.PubMed/NCBI View Article : Google Scholar

41 

Zhao YY, Lian JX, Lan Z, Zou KL, Wang WM and Yu GT: Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis. 29:933–941. 2023.PubMed/NCBI View Article : Google Scholar

42 

Van Loenhout J, Freire Boullosa L, Quatannens D, De Waele J, Merlin C, Lambrechts H, Lau HW, Hermans C, Lin A, Lardon F, et al: Auranofin and cold atmospheric plasma synergize to trigger distinct cell death mechanisms and immunogenic responses in glioblastoma. Cells. 10(2936)2021.PubMed/NCBI View Article : Google Scholar

43 

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

44 

Li X, Duan L, Yuan S, Zhuang X, Qiao T and He J: Ferroptosis inhibitor alleviates radiation-induced lung fibrosis (RILF) via down-regulation of TGF-β1. J Inflamm (Lond). 16(11)2019.PubMed/NCBI View Article : Google Scholar

45 

Zhernakova A, Withoff S and Wijmenga C: Clinical implications of shared genetics and pathogenesis in autoimmune diseases. Nat Rev Endocrinol. 9:646–659. 2013.PubMed/NCBI View Article : Google Scholar

46 

Mack CL, Adams D, Assis DN, Kerkar N, Manns MP, Mayo MJ, Vierling JM, Alsawas M, Murad MH and Czaja AJ: Diagnosis and management of autoimmune hepatitis in adults and children: 2019 Practice guidance and guidelines from the American association for the study of liver diseases. Hepatology. 72:671–722. 2020.PubMed/NCBI View Article : Google Scholar

47 

Deng G, Li Y, Ma S, Gao Z, Zeng T, Chen L, Ye H, Yang M, Shi H, Yao X, et al: Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med. 148:151–161. 2020.PubMed/NCBI View Article : Google Scholar

48 

Zhu L, Chen D, Zhu Y, Pan T, Xia D, Cai T, Lin H, Lin J, Jin X, Wu F, et al: GPX4-regulated ferroptosis mediates S100-induced experimental autoimmune hepatitis associated with the Nrf2/HO-1 signaling pathway. Oxid Med Cell Longev. 2021(6551069)2021.PubMed/NCBI View Article : Google Scholar

49 

Singh JA, Saag KG, Bridges SL Jr, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, et al: 2015 American college of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 68:1–26. 2016.PubMed/NCBI View Article : Google Scholar

50 

Fraenkel L, Bathon JM, England BR, St Clair EW, Arayssi T, Carandang K, Deane KD, Genovese M, Huston KK, Kerr G, et al: 2021 American college of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 73:1108–1123. 2021.PubMed/NCBI View Article : Google Scholar

51 

Wu J, Feng Z, Chen L, Li Y, Bian H, Geng J, Zheng ZH, Fu X, Pei Z, Qin Y, et al: TNF antagonist sensitizes synovial fibroblasts to ferroptotic cell death in collagen-induced arthritis mouse models. Nat Commun. 13(676)2022.PubMed/NCBI View Article : Google Scholar

52 

Floros KV, Cai J, Jacob S, Kurupi R, Fairchild CK, Shende M, Coon CM, Powell KM, Belvin BR, Hu B, et al: MYCN-amplified neuroblastoma is addicted to iron and vulnerable to inhibition of the system Xc-/glutathione axis. Cancer Res. 81:1896–1908. 2021.PubMed/NCBI View Article : Google Scholar

53 

Ling H, Li M, Yang C, Sun S, Zhang W, Zhao L, Xu N, Zhang J, Shen Y, Zhang X, et al: Glycine increased ferroptosis via SAM-mediated GPX4 promoter methylation in rheumatoid arthritis. Rheumatology (Oxford). 61:4521–4534. 2022.PubMed/NCBI View Article : Google Scholar

54 

Luo H and Zhang R: Icariin enhances cell survival in lipopolysaccharide-induced synoviocytes by suppressing ferroptosis via the Xc-/GPX4 axis. Exp Ther Med. 21(72)2021.PubMed/NCBI View Article : Google Scholar

55 

Gordon C, Amissah-Arthur MB, Gayed M, Brown S, Bruce IN, D'Cruz D, Empson B, Griffiths B, Jayne D, Khamashta M, et al: The British Society for Rheumatology guideline for the management of systemic lupus erythematosus in adults. Rheumatology (Oxford). 57:e1–e45. 2018.PubMed/NCBI View Article : Google Scholar

56 

Tsokos GC, Lo MS, Costa Reis P and Sullivan KE: New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 12:716–730. 2016.PubMed/NCBI View Article : Google Scholar

57 

Indrakanti DL, Alvarado A, Zhang X, Birmingham DJ, Hinton A and Rovin BH: The interleukin-6-hepcidin-hemoglobin circuit in systemic lupus erythematosus flares. Lupus. 26:200–203. 2017.PubMed/NCBI View Article : Google Scholar

58 

Chen W, Li W, Zhang Z, Tang X, Wu S, Yao G, Li K, Wang D, Xu Y, Feng R, et al: Lipocalin-2 exacerbates lupus nephritis by promoting Th1 cell differentiation. J Am Soc Nephrol. 31:2263–2277. 2020.PubMed/NCBI View Article : Google Scholar

59 

Scindia Y, Wlazlo E, Ghias E, Cechova S, Loi V, Leeds J, Ledesma J, Helen C and Swaminathan S: Modulation of iron homeostasis with hepcidin ameliorates spontaneous murine lupus nephritis. Kidney Int. 98:100–115. 2020.PubMed/NCBI View Article : Google Scholar

60 

Li P, Jiang M, Li K, Li H, Zhou Y, Xiao X, Xu Y, Krishfield S, Lipsky PE, Tsokos GC and Zhang X: Glutathione peroxidase 4-regulated neutrophil ferroptosis induces systemic autoimmunity. Nat Immunol. 22:1107–1117. 2021.PubMed/NCBI View Article : Google Scholar

61 

Ananthakrishnan AN: Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 12:205–217. 2015.PubMed/NCBI View Article : Google Scholar

62 

Guan Q: A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019(7247238)2019.PubMed/NCBI View Article : Google Scholar

63 

Mayr L, Grabherr F, Schwärzler J, Reitmeier I, Sommer F, Gehmacher T, Niederreiter L, He GW, Ruder B, Kunz KTR, et al: Dietary lipids fuel GPX4-restricted enteritis resembling Crohn's disease. Nat Commun. 11(1775)2020.PubMed/NCBI View Article : Google Scholar

64 

Xu J, Liu S, Cui Z, Wang X, Ning T, Wang T, Zhang N, Xie S, Min L, Zhang S, et al: Ferrostatin-1 alleviated TNBS induced colitis via the inhibition of ferroptosis. Biochem Biophys Res Commun. 573:48–54. 2021.PubMed/NCBI View Article : Google Scholar

65 

Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, Zheng F and Wu B: Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 11(86)2020.PubMed/NCBI View Article : Google Scholar

66 

Chen Y, Wang J, Li J, Zhu J, Wang R, Xi Q, Wu H, Shi T and Chen W: Astragalus polysaccharide prevents ferroptosis in a murine model of experimental colitis and human Caco-2 cells via inhibiting NRF2/HO-1 pathway. Eur J Pharmacol. 911(174518)2021.PubMed/NCBI View Article : Google Scholar

67 

Chen Y, Zhang P, Chen W and Chen G: Ferroptosis mediated DSS-induced ulcerative colitis associated with Nrf2/HO-1 signaling pathway. Immunol Lett. 225:9–15. 2020.PubMed/NCBI View Article : Google Scholar

68 

Montalban X, Gold R, Thompson AJ, Otero-Romero S, Amato MP, Chandraratna D, Clanet M, Comi G, Derfuss T, Fazekas F, et al: ECTRIMS/EAN guideline on the pharmacological treatment of people with multiple sclerosis. Eur J Neurol. 25:215–237. 2018.PubMed/NCBI View Article : Google Scholar

69 

Hemmer B, Archelos JJ and Hartung HP: New concepts in the immunopathogenesis of multiple sclerosis. Nat Rev Neurosci. 3:291–301. 2002.PubMed/NCBI View Article : Google Scholar

70 

Hu CL, Nydes M, Shanley KL, Morales Pantoja IE, Howard TA and Bizzozero OA: Reduced expression of the ferroptosis inhibitor glutathione peroxidase-4 in multiple sclerosis and experimental autoimmune encephalomyelitis. J Neurochem. 148:426–439. 2019.PubMed/NCBI View Article : Google Scholar

71 

Jhelum P, Santos-Nogueira E, Teo W, Haumont A, Lenoël I, Stys PK and David S: Ferroptosis mediates cuprizone-induced loss of oligodendrocytes and demyelination. J Neurosci. 40:9327–9341. 2020.PubMed/NCBI View Article : Google Scholar

72 

Stewart DA: NICE guideline for Parkinson's disease. Age Ageing. 36:240–242. 2007.PubMed/NCBI View Article : Google Scholar

73 

Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, Liong C, McMurtrey C, Hildebrand WH, Mao X, et al: T cells from patients with Parkinson's disease recognize α-synuclein peptides. Nature. 546:656–661. 2017.PubMed/NCBI View Article : Google Scholar

74 

Thapa K, Khan H, Kanojia N, Singh TG, Kaur A and Kaur G: Therapeutic insights on ferroptosis in Parkinson's disease. Eur J Pharmacol. 930(175133)2022.PubMed/NCBI View Article : Google Scholar

75 

Zhang P, Chen L, Zhao Q, Du X, Bi M, Li Y, Jiao Q and Jiang H: Ferroptosis was more initial in cell death caused by iron overload and its underlying mechanism in Parkinson's disease. Free Radic Biol Med. 152:227–234. 2020.PubMed/NCBI View Article : Google Scholar

76 

Dexter DT, Wells FR, Lees AJ, Agid F, Agid Y, Jenner P and Marsden CD: Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson's disease. J Neurochem. 52:1830–1836. 1989.PubMed/NCBI View Article : Google Scholar

77 

Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A, Jenner P and Marsden CD: Basal lipid peroxidation in substantia nigra is increased in Parkinson's disease. J Neurochem. 52:381–389. 1989.PubMed/NCBI View Article : Google Scholar

78 

Sofic E, Lange KW, Jellinger K and Riederer P: Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson's disease. Neurosci Lett. 142:128–130. 1992.PubMed/NCBI View Article : Google Scholar

79 

Zuo Y, Xie J, Li X, Li Y, Thirupathi A, Zhang J, Yu P, Gao G, Chang Y and Shi Z: Ferritinophagy-mediated ferroptosis involved in paraquat-induced neurotoxicity of dopaminergic neurons: Implication for neurotoxicity in PD. Oxid Med Cell Longev. 2021(9961628)2021.PubMed/NCBI View Article : Google Scholar

80 

Parisi R, Iskandar IYK, Kontopantelis E, Augustin M, Griffiths CEM and Ashcroft DM: Global Psoriasis Atlas. National, regional, and worldwide epidemiology of psoriasis: Systematic analysis and modelling study. BMJ. 369(m1590)2020.PubMed/NCBI View Article : Google Scholar

81 

Arbiser JL, Bonner MY, Ward N, Elsey J and Rao S: Selenium unmasks protective iron armor: A possible defense against cutaneous inflammation and cancer. Biochim Biophys Acta Gen Subj. 1862:2518–2527. 2018.PubMed/NCBI View Article : Google Scholar

82 

Shou Y, Yang L, Yang Y and Xu J: Inhibition of keratinocyte ferroptosis suppresses psoriatic inflammation. Cell Death Dis. 12(1009)2021.PubMed/NCBI View Article : Google Scholar

83 

Atkinson MA and Maclaren NK: The pathogenesis of insulin-dependent diabetes mellitus. N Engl J Med. 331:1428–1436. 1994.PubMed/NCBI View Article : Google Scholar

84 

Zang H, Wu W, Qi L, Tan W, Nagarkatti P, Nagarkatti M, Wang X and Cui T: Autophagy inhibition enables Nrf2 to exaggerate the progression of diabetic cardiomyopathy in mice. Diabetes. 69:2720–2734. 2020.PubMed/NCBI View Article : Google Scholar

85 

Ni T, Huang X, Pan S and Lu Z: Inhibition of the long non-coding RNA ZFAS1 attenuates ferroptosis by sponging miR-150-5p and activates CCND2 against diabetic cardiomyopathy. J Cell Mol Med. 25:9995–10007. 2021.PubMed/NCBI View Article : Google Scholar

86 

Li S, Zheng L, Zhang J, Liu X and Wu Z: Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic Biol Med. 162:435–449. 2021.PubMed/NCBI View Article : Google Scholar

87 

Kim S, Kang SW, Joo J, Han SH, Shin H, Nam BY, Park J, Yoo TH, Kim G, Lee P and Park JT: Characterization of ferroptosis in kidney tubular cell death under diabetic conditions. Cell Death Dis. 12(160)2021.PubMed/NCBI View Article : Google Scholar

88 

Bruni A, Bornstein S, Linkermann A and Shapiro AMJ: Regulated cell death seen through the lens of islet transplantation. Cell Transplant. 27:890–901. 2018.PubMed/NCBI View Article : Google Scholar

89 

Krümmel B, Plötz T, Jörns A, Lenzen S and Mehmeti I: The central role of glutathione peroxidase 4 in the regulation of ferroptosis and its implications for pro-inflammatory cytokine-mediated beta-cell death. Biochim Biophys Acta Mol Basis Dis. 1867(166114)2021.PubMed/NCBI View Article : Google Scholar

90 

Fudman EJ, Till GO and Fox IH: Deferoxamine induced decreases of lipid peroxides in rheumatoid arthritis. J Rheumatol. 14:686–691. 1987.PubMed/NCBI

91 

Salvarani C, Baricchi R, Lasagni D, Boiardi L, Piccinini R, Brunati C, Macchioni P and Portioli I: Effects of desferrioxamine therapy on chronic disease anemia associated with rheumatoid arthritis. Rheumatol Int. 16:45–48. 1996.PubMed/NCBI View Article : Google Scholar

92 

Lynch SG, Peters K and LeVine SM: Desferrioxamine in chronic progressive multiple sclerosis: A pilot study. Mult Scler. 2:157–160. 1996.PubMed/NCBI View Article : Google Scholar

93 

Lynch SG, Fonseca T and LeVine SM: A multiple course trial of desferrioxamine in chronic progressive multiple sclerosis. Cell Mol Biol (Noisy-le-grand). 46:865–869. 2000.PubMed/NCBI

94 

Febbraro F, Andersen KJ, Sanchez-Guajardo V, Tentillier N and Romero-Ramos M: Chronic intranasal deferoxamine ameliorates motor defects and pathology in the α-synuclein rAAV Parkinson's model. Exp Neurol. 247:45–58. 2013.PubMed/NCBI View Article : Google Scholar

95 

Sweeney ME, Slusser JG, Lynch SG, Benedict SH, Garcia SL, Rues L and LeVine SM: Deferiprone modulates in vitro responses by peripheral blood T cells from control and relapsing-remitting multiple sclerosis subjects. Int Immunopharmacol. 11:1796–1801. 2011.PubMed/NCBI View Article : Google Scholar

96 

Devos D, Moreau C, Devedjian JC, Kluza J, Petrault M, Laloux C, Jonneaux A, Ryckewaert G, Garçon G, Rouaix N, et al: Targeting chelatable iron as a therapeutic modality in Parkinson's disease. Antioxid Redox Signal. 21:195–210. 2014.PubMed/NCBI View Article : Google Scholar

97 

Yang B, Hou S, Huang S, Li H and Li Y: Ferroptosis inhibitor regulates the disease progression of systematic lupus erythematosus mice model through Th1/Th2 ratio. Curr Mol Med: May 25, 2022 (Epub ahead of print).

98 

Hinman A, Holst CR, Latham JC, Bruegger JJ, Ulas G, McCusker KP, Amagata A, Davis D, Hoff KG, Kahn-Kirby AH, et al: Vitamin E hydroquinone is an endogenous regulator of ferroptosis via redox control of 15-lipoxygenase. PLoS One. 13(e0201369)2018.PubMed/NCBI View Article : Google Scholar

99 

Edmonds SE, Winyard PG, Guo R, Kidd B, Merry P, Langrish-Smith A, Hansen C, Ramm S and Blake DR: Putative analgesic activity of repeated oral doses of vitamin E in the treatment of rheumatoid arthritis. Results of a prospective placebo controlled double blind trial. Ann Rheum Dis. 56:649–655. 1997.PubMed/NCBI View Article : Google Scholar

100 

Aristotelous P, Stefanakis M, Pantzaris M, Pattichis CS, Calder PC, Patrikios IS, Sakkas GK and Giannaki CD: The effects of specific omega-3 and omega-6 polyunsaturated fatty acids and antioxidant vitamins on gait and functional capacity parameters in patients with relapsing-remitting multiple sclerosis. Nutrients. 13(3661)2021.PubMed/NCBI View Article : Google Scholar

101 

Taghizadeh M, Tamtaji OR, Dadgostar E, Daneshvar Kakhaki R, Bahmani F, Abolhassani J, Aarabi MH, Kouchaki E, Memarzadeh MR and Asemi Z: The effects of omega-3 fatty acids and vitamin E co-supplementation on clinical and metabolic status in patients with Parkinson's disease: A randomized, double-blind, placebo-controlled trial. Neurochem Int. 108:183–189. 2017.PubMed/NCBI View Article : Google Scholar

102 

Kharaeva Z, Gostova E, De Luca C, Raskovic D and Korkina L: Clinical and biochemical effects of coenzyme Q(10), vitamin E, and selenium supplementation to psoriasis patients. Nutrition. 25:295–302. 2009.PubMed/NCBI View Article : Google Scholar

103 

Odinak MM, Bisaga GN and Zarubina IV: New approaches to antioxidant therapy in multiple sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova. (Suppl):S72–S75. 2002.PubMed/NCBI(In Russian).

104 

Miroliaee AE, Esmaily H, Vaziri-Bami A, Baeeri M, Shahverdi AR and Abdollahi M: Amelioration of experimental colitis by a novel nanoselenium-silymarin mixture. Toxicol Mech Methods. 21:200–208. 2011.PubMed/NCBI View Article : Google Scholar

105 

Peretz A, Siderova V and Nève J: Selenium supplementation in rheumatoid arthritis investigated in a double blind, placebo-controlled trial. Scand J Rheumatol. 30:208–212. 2001.PubMed/NCBI View Article : Google Scholar

106 

Tarp U, Overvad K, Thorling EB, Graudal H and Hansen JC: Selenium treatment in rheumatoid arthritis. Scand J Rheumatol. 14:364–368. 1985.PubMed/NCBI View Article : Google Scholar

107 

Zafar KS, Siddiqui A, Sayeed I, Ahmad M, Salim S and Islam F: Dose-dependent protective effect of selenium in rat model of Parkinson's disease: Neurobehavioral and neurochemical evidences. J Neurochem. 84:438–446. 2003.PubMed/NCBI View Article : Google Scholar

108 

Batooei M, Tahamoli-Roudsari A, Basiri Z, Yasrebifar F, Shahdoust M, Eshraghi A, Mehrpooya M and Ataei S: Evaluating the effect of oral N-acetylcysteine as an adjuvant treatment on clinical outcomes of patients with rheumatoid arthritis: A randomized, double blind clinical trial. Rev Recent Clin Trials. 13:132–138. 2018.PubMed/NCBI View Article : Google Scholar

109 

Lai ZW, Hanczko R, Bonilla E, Caza TN, Clair B, Bartos A, Miklossy G, Jimah J, Doherty E, Tily H, et al: N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: A randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 64:2937–2946. 2012.PubMed/NCBI View Article : Google Scholar

110 

Zheng K, Dong Y, Yang R, Liang Y, Wu H and He Z: Regulation of ferroptosis by bioactive phytochemicals: Implications for medical nutritional therapy. Pharmacol Res. 168(105580)2021.PubMed/NCBI View Article : Google Scholar

111 

Khojah HM, Ahmed S, Abdel-Rahman MS and Elhakeim EH: Resveratrol as an effective adjuvant therapy in the management of rheumatoid arthritis: A clinical study. Clin Rheumatol. 37:2035–2042. 2018.PubMed/NCBI View Article : Google Scholar

112 

Samsami-Kor M, Daryani NE, Asl PR and Hekmatdoost A: Anti-inflammatory effects of resveratrol in patients with ulcerative colitis: A randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 46:280–285. 2015.PubMed/NCBI View Article : Google Scholar

113 

Samsamikor M, Daryani NE, Asl PR and Hekmatdoost A: Resveratrol supplementation and oxidative/anti-oxidative status in patients with ulcerative colitis: A randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 47:304–309. 2016.PubMed/NCBI View Article : Google Scholar

114 

Movahed A, Raj P, Nabipour I, Mahmoodi M, Ostovar A, Kalantarhormozi M and Netticadan T: Efficacy and safety of resveratrol in type 1 diabetes patients: A two-month preliminary exploratory trial. Nutrients. 12(161)2020.PubMed/NCBI View Article : Google Scholar

115 

Ghavipour M, Sotoudeh G, Tavakoli E, Mowla K, Hasanzadeh J and Mazloom Z: Pomegranate extract alleviates disease activity and some blood biomarkers of inflammation and oxidative stress in rheumatoid arthritis patients. Eur J Clin Nutr. 71:92–96. 2017.PubMed/NCBI View Article : Google Scholar

116 

Petramfar P, Hajari F, Yousefi G, Azadi S and Hamedi A: Efficacy of oral administration of licorice as an adjunct therapy on improving the symptoms of patients with Parkinson's disease, A randomized double blinded clinical trial. J Ethnopharmacol. 247(112226)2020.PubMed/NCBI View Article : Google Scholar

117 

Lin D, Zhang M, Luo C, Wei P, Cui K and Chen Z: Targeting ferroptosis attenuates inflammation, fibrosis, and mast cell activation in chronic prostatitis. J Immunol Res. 2022(6833867)2022.PubMed/NCBI View Article : Google Scholar

118 

Zhao T, Yang Q, Xi Y, Xie Z, Shen J, Li Z, Li Z and Qin D: Ferroptosis in rheumatoid arthritis: A potential therapeutic strategy. Front Immunol. 13(779585)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2023
Volume 26 Issue 2

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
Shen L, Wang X, Zhai C and Chen Y: Ferroptosis: A potential therapeutic target in autoimmune disease (Review). Exp Ther Med 26: 368, 2023
APA
Shen, L., Wang, X., Zhai, C., & Chen, Y. (2023). Ferroptosis: A potential therapeutic target in autoimmune disease (Review). Experimental and Therapeutic Medicine, 26, 368. https://doi.org/10.3892/etm.2023.12067
MLA
Shen, L., Wang, X., Zhai, C., Chen, Y."Ferroptosis: A potential therapeutic target in autoimmune disease (Review)". Experimental and Therapeutic Medicine 26.2 (2023): 368.
Chicago
Shen, L., Wang, X., Zhai, C., Chen, Y."Ferroptosis: A potential therapeutic target in autoimmune disease (Review)". Experimental and Therapeutic Medicine 26, no. 2 (2023): 368. https://doi.org/10.3892/etm.2023.12067