Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 14, 2024; 30(34): 3856-3861
Published online Sep 14, 2024. doi: 10.3748/wjg.v30.i34.3856
Overview of ferroptosis and pyroptosis in acute liver failure
Ya-Wen Sun, Bo-Wen Zhao, Hai-Fang Li, Guang-Xiao Zhang, College of Life Sciences, Shandong Agricultural University, Tai’an 271018, Shandong Province, China
ORCID number: Ya-Wen Sun (0009-0009-5244-2322); Bo-Wen Zhao (0009-0009-9186-8895); Hai-Fang Li (0000-0002-2827-5925); Guang-Xiao Zhang (0009-0001-6209-9048).
Co-first authors: Ya-Wen Sun and Bo-Wen Zhao.
Author contributions: Sun YW and Zhao BW wrote the original draft, and revised the manuscript; Zhang GX wrote the original draft; Li HF supervised, conceived, verified, reviewed, and edited the manuscript. All authors were involved in the critical review of the results and have contributed to reading and approving the final manuscript. Sun YW and Zhao BW contributed equally to this work as co-first authors. The reasons for designating Sun YW and Zhao BW as co-first authors are twofold. First, the review was prepared as a collaborative effort with Sun YW and Zhao BW contributing equally to literature searching, draft writing, and manuscript revising. The designation of co-first authors authorship reflects the distribution of responsibilities and burdens associated with the time and effort required to complete the review and ensure effective communication and management of post-submission matters. Second, Sun YW and Zhao BW are skilled in different fields, which promotes the most comprehensive and in-depth discussion of the review topic, ultimately enriching reader understanding by offering various expert perspectives.
Conflict-of-interest statement: The authors declare that they have no conflicts of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hai-Fang Li, PhD, Associate Professor, College of Life Sciences, Shandong Agricultural University, No. 61 Daizong Street, Tai'an 271018, Shandong Province, China. hfli1228@163.com
Received: March 24, 2024
Revised: August 14, 2024
Accepted: August 16, 2024
Published online: September 14, 2024
Processing time: 170 Days and 1.6 Hours

Abstract

In this editorial, we comment on the article by Zhou et al published in a recent issue. We specifically focus on the crucial roles of ferroptosis and pyroptosis in acute liver failure (ALF), a disease with high mortality rates. Ferroptosis is the result of increased intracellular reactive oxygen species due to iron accumulation, glutathione (GSH) depletion, and decreased GSH peroxidase 4 activity, while pyroptosis is a procedural cell death mediated by gasdermin D which initiates a sustained inflammatory process. In this review, we describe the characteristics of ferroptosis and pyroptosis, and discuss the involvement of the two cell death modes in the onset and development of ALF. Furthermore, we summarize several interfering methods from the perspective of ferroptosis and pyroptosis for the alleviation of ALF. These observations might provide new targets and a theoretical basis for the treatment of ALF, which are also crucial for improving the prognosis of patients with ALF.

Key Words: Acute liver failure; Ferroptosis; Pyroptosis; Glutathione peroxidase 4; Gasdermin D

Core Tip: In this review we describe the characteristics of ferroptosis and pyroptosis, and discuss the involvement of the two cell death modes in the onset and development of acute liver failure (ALF). Furthermore, we summarize several interfering methods from the perspective of ferroptosis and pyroptosis for the alleviation of ALF. These observations might provide new targets and a theoretical basis for the treatment of ALF, which is also crucial for improving the prognosis of patients with ALF.



INTRODUCTION

Acute liver failure (ALF) is a severe clinical syndrome characterized by massive hepatocyte necrosis and acute liver damage in a short period of time with a mortality rate as high as 30%[1]. When ALF occurs, liver function declines sharply, which fails to fulfill the organ’s basic physiological actions. Clinically, the main manifestations of ALF are jaundice, ascites, and hepatic encephalopathy[2]. The common causes of ALF include pharmacological liver injury, hepatitis virus, ischemia, and autoimmunity, which account for nearly 80% of the ALF cases[3]. For example, acetaminophen overdose triggers the formation of NOD-like receptor protein 3 (NLRP3) inflammasome, resulting in pyroptosis and ultimately leading to ALF[4]. In addition, acute hepatitis B (AHB) virus infection is the main cause of ALF in many countries[5]. A recent study indicated that 6.6% of patients with AHB developed ALF[6].

Previous studies have established that the primary mode of cell death involved in ALF is apoptosis[7]. As a form of cell-autonomous programmed death, apoptosis triggers a series of reactions by activating the caspase family of proteins, leading to intracellular protein degradation and the elimination of damaged or no longer needed cells[8]. Rutherford et al[9] demonstrated that apoptosis was markedly activated in ALF, with notable elevations of apoptosis markers such as tumor necrosis factor-alpha, hepatocyte growth factor (HGF), interleukin-6, and receptor interacting protein kinase 3 (RIPK3). Notably, the elevated levels of HGF and RIPK3 could serve as a confirmatory diagnostic marker and a poorer clinical prognosis for patients suffering from ALF, respectively[10].

Studies over the past 10 years have garnered widespread attention related to two emerging modes of cell death, ferroptosis and pyroptosis, in ALF pathogenesis. Ferroptosis is a form of cell death that relies on iron ions (Fe2+), characterized by the peroxidation of polyunsaturated fatty acids on the cell membrane[11]. Fe2+ promotes the generation of lipid peroxides, leading to excessive reactive oxygen species (ROS) production, which further disrupts the integrity of the cell membrane and triggers cell death[12,13]. It is worth noting that GSH peroxidase 4 (GPX4) functions as a crucial enzyme in the ferroptosis pathway, which catalyzes the decomposition of lipid peroxides. When GPX4 activity is inhibited, lipid peroxides accumulate in the cell membrane, ultimately culminating in the onset of ferroptosis[14]. Pyroptosis is a form of cell death with inflammatory characteristics, typically manifested by the formation of numerous small vesicular structures known as pyroptotic bodies[15]. Pyroptotic bodies mediate the cleavage of gasdermin D (GSDMD) by caspase proteins to release its N-terminal fragment. The N-terminus of GSDMD is the main effector of pyroptosis, which is released and inserted into the cell membrane to form holes, leading to rupture of the cell membrane and the release of cell contents, ultimately triggering the onset of pyroptosis[16,17]. These released substances can activate immune cells and trigger severe inflammatory responses[18]. Additionally, the released proinflammatory mediators may further initiate a sustained inflammatory process and create a vicious cycle[19]. The intense inflammatory response may aggravate liver tissue damage and promote the further development of ALF.

Ferroptosis and pyroptosis represent two different modes of cell death, with an increasing body of evidence indicating a potential correlation between them. The release of damage-associated molecular patterns (DAMPs) from plasma membrane pores may be a common feature of ferroptosis, pyroptosis, and necroptosis[20]. Furthermore, the released DAMPs triggered by ferroptosis may promote pyroptosis and necroptosis. GPX4 is a key enzyme in ferroptosis and has also been demonstrated to be implicated in the negative modulation of pyroptosis[21]. Interestingly, GPX4 has been observed to induce a range of other forms of cell death, including apoptosis, autophagy, necroptosis, and pyroptosis[22]. This suggests the possibility of a complex interconnection between ferroptosis and other forms of programmed cell death. It has been demonstrated that iron-activated ROS can induce pyroptosis through the Tom20-Bax-caspase-GSDME pathway, and GPX4 can inhibit the activity of caspase-1, thereby preventing the process of pyroptosis[23]. Additionally, GPX4 functions as a regulator of NOD-, LRR-, and NLRP3-mediated pyroptosis in kidney injury[24]. The interaction between pyroptosis and ferroptosis in epilepsy may be associated with toll-like receptor 4 (TLR4)-mediated neuroinflammation[25]. Ferroptosis has been demonstrated to induce neuroinflammation and the release of a considerable number of inflammatory factors. Among these factors, IL-1β can activate the HMGB1/TLR4 signaling pathway, thereby increasing the production of NLRP3 and IL-1β. This process serves to amplify the inflammatory response by inducing pyroptosis.

In the article by Zhou et al[26] entitled ‘Silent information regulator sirtuin 1 ameliorates acute liver failure (ALF) via the p53/glutathione peroxidase 4/gasdermin D axis’, the authors demonstrated that ferroptosis and pyroptosis are key modes of hepatocyte death in the progression of ALF. Therefore, it is crucial to conduct a comprehensive analysis of the regulatory roles of ferroptosis and pyroptosis in ALF. This will not only enhance our understanding of the pathological process of liver failure but also potentially provide theoretical support for the development of new treatment strategies.

MECHANISMS OF FERROPTOSIS AND PYROPTOSIS IN ALF

The mechanisms underlying the occurrence and development of ALF have been described in previous publications[3,10,11,27]. Here, we mainly focus on the roles of ferroptosis and pyroptosis in ALF.

The involvement of ferroptosis in ALF

Ferroptosis has been shown to play important roles in several pathological processes associated with liver diseases[12,14], including ALF[28], which is regulated by lipid peroxidation and iron accumulation[29]. The key process in ferroptosis is the Fenton reaction, in which Fe2+ converts lipid peroxides to ROS[30]. As a key regulator of ferroptosis, GPX4 is the only GSH peroxidase isoform capable of regulating lipid peroxidation and ROS levels[31], which could catalyze the decomposition of toxic peroxides depending on the conversion of GSH to oxidized GSH[32]. Additionally, the glutamate/cystine antitransport system (System Xc) is another important regulator of ferroptosis, the inhibition of which would promote the ferroptosis process[33]. It has been indicated that p53 could induce ferroptosis by inhibiting System Xc activity and reducing cystine entry into the cell[34]. Moreover, solute carrier family 7 member 11 (SLC7A11) has been proved to be a crucial upstream regulator of ferroptosis[35], the down-regulation of which inhibits the cysteine metabolic pathway, leading to a reduction in intracellular cystine levels and an indirect inhibition of GPX4 activity[36]. Jiang et al[34] found that p53-mediated transcriptional repression of SLC7A11 is critical for ROS-induced ferroptosis. Another study revealed that G3BP stress granule assembly factor 1 (G3BP1) inhibits the entry of p53 protein into the nucleus and reduces SLC7A11 transcription and hepatocyte ferroptosis during ALF[37]. Also, hepatitis B virus X protein exacerbates ALF by promoting iron-mediated cell death through EZH2/H3K27me3-mediated inhibition of SLC7A11[38]. In addition, nuclear factor erythroid 2-related factor 2 (Nrf2) is also an important regulator of ferroptosis[39], and its activation inhibits H2O2-induced cellular ferroptosis by modulating the GPX4 pathway[14]. Collectively, these findings suggest that ferroptosis plays a significant role in the development of ALF.

The involvement of pyroptosis in ALF

Pyroptosis, another cell death mode, also triggers significant hepatocyte death in liver failure[40]. In the pathogenesis of ALF, GSDMD can be cleaved by caspase-1 into inflammatory GSDMD-N in the hepatocyte cytoplasm[41]. GSDMD-N recognizes phospholipids and inserts into the cell membrane, facilitating membrane pore formation and pyroptotic cell death[42]. Additionally, GSDMD-N up-regulates the release of inflammatory mediators from macrophages recruited by monocyte chemotactic protein 1 and its receptor CC chemokine receptor-2[43]. This leads to an amplified inflammatory response, which in turn exacerbates ALF. NLRP3 could stimulate pyroptosis by inducing caspase-1 and GSDMD activation[18]. Furthermore, HMGB1 induces the formation of NLRP3 inflammasome by activating the TLR4/MyD88/NF-κB signaling pathway, thereby causing pyroptosis[44]. Studies have shown that IL-10 inhibits NLRP3 expression[45], suggesting a potential role for IL-10 in alleviating ALF. CD38, a type II transmembrane protein, is an important NAD-dependent enzyme, and it has been shown that CD38 stimulated TLR4-NLRP3-GSDMD-mediated pyroptosis and aggravated liver injury[46]. The occurrence of pyroptosis is influenced by multiple factors, which in turn impacts the progression of ALF, suggesting that the inhibition of pyroptosis could potentially alleviate ALF.

The regulation of deacetylases on ferroptosis and pyroptosis in ALF

In the study by Zhou et al[26], the authors suggested that SIRT1 activation reduced hepatic injury and inflammatory responses by reducing the p53 acetylation level and further suppressing the expression of ferroptosis and pyroptosis-related proteins GPX4 and GSDMD. SIRT1 is one of the important deacetylases, which could remove acetyl groups from histones and non-histone proteins. As protein acetylation is a crucial post-translational modification, we here review the regulation of deacetylases on ferroptosis and pyroptosis, and further on ALF.

SIRT1, which regulates the onset and progression of ALF by affecting ferroptosis and pyroptosis pathways[47] has received significant attention in recent years. The inhibitory effect of SIRT1-mediated deacetylation on ferroptosis is exerted through two major pathways: The deacetylation of p53 to suppress p53-dependent downregulation of SLC7A11[48], and the other pathway is the deacetylation of Nrf2 to promote Nrf2-mediated upregulation of GPX4[49]. In addition, SIRT1 could reduce the activation of NLRP3 inflammasome induced by IL-1β, thereby inhibiting pyroptosis[50]. Besides SIRT1, other deacetylases, such as HDAC6 and HDAC2 also have crucial actions in the regulation of ferroptosis and pyroptosis. For instance, studies have demonstrated that inhibition of HDAC6 expression reduced the expression of NLRP3 inflammatory vesicles and NF-κB, thereby affecting pyroptosis and ferroptosis in ALF, respectively[51,52]. Furthermore, downregulation of HDAC2 alleviates cell pyroptosis in ALF by regulating the acetylation level of the ULK1 K68 site[53].

In summary, it can be concluded that deacetylases could alleviate ALF by inhibiting both the ferroptosis and pyroptosis process. These results may provide new therapeutic targets for alleviating ALF.

CONCLUSION

ALF is a severe liver disease in clinical practice, which is accompanied by the rapid death of massive hepatocytes and multiple inflammatory outbreaks. Ferroptosis and pyroptosis are important cell death modes in ALF. Ferroptosis is the result of increased intracellular ROS due to iron accumulation, GSH depletion, and decreased GPX4 activity[54]. Pyroptosis is a procedural cell death mediated by the gasdermin family, which is dependent on the cleavage of GSDMD to the active GSDMD-N fragment by the NLRP3-induced caspases, and further triggering cell membrane perforation and an inflammatory storm[42]. Therefore, suppressing the ferroptosis and pyroptosis process has great potential in the treatment of ALF.

To date, some agents with iron chewing or GPX4 regulating effects have shown ferroptosis-inhibitory functions. As iron ion accumulation is an essential prerequisite for ferroptosis, iron chewing agents can be used to reduce the accumulation and deposition of iron in the organ and further mitigate the ferroptosis procedure. Iron chelators such as deferoxamine, deferiprone and deferasirox have been proved to weaken lipid peroxidation by restricting the Fenton reaction, thereby inhibiting ferroptosis and alleviating ALF development[55]. GPX4 is a critical regulator of ferroptosis, thus enhancing the expression or the activity of GPX4 could inhibit this cell death mode. It was indicated that selenium supplementation could increase the level of GPX4 and inhibit ferroptosis as selenium is required for the synthesis of GPX4[56]. Multiple studies have shown that SIRT1 alleviates the inhibitory effects of p53 on GPX4 and SLC7A11 by reducing the level of p53 acetylation, which is a potential target for the treatment of ferroptosis-related diseases[57,58].

To date, a significant number of inhibitors and drugs targeting pyroptosis-related proteins have shown therapeutic potential in preclinical studies. As the activation of NLRP3 serves as the initiating point of pyroptosis, NLRP3-specific inhibitors have emerged as promising therapeutic targets for the treatment of pyroptosis, with MCC950 and the natural compound kaempferol currently undergoing clinical trials[59]. A study also pointed out that metformin, a "magic drug for diabetes treatment", could reduce inflammatory responses and cellular pyroptosis by inhibiting the NF-κB-NLRP3 pathway[60]. In addition, caspase inhibitors such as Vx765 and Z-YVAD-FMK block the binding of caspases to GSDMD and further inhibit pyroptosis[61]. Research has shown that the tricarboxylic acid cycle intermediate fumarate acts as a pyroptosis inhibitor by succinylating GSDMD to prevent its binding to caspases[62].

Although several candidates could be selected to regulate the critical steps in ferroptosis and pyroptosis, we still face many problems in clinics. For example, many existing inhibitors may interfere with normal metabolism pathways due to their nonspecific activity. The poor absorption rate of some drugs is another problem which needs to be resolved. With advances in science and technology, the use of big data and artificial intelligence methods to screen existing FDA-approved drugs may identify new ferroptosis and pyroptosis modulators. Existing studies have revealed that there is crosstalk between ferroptosis and pyroptosis through complex feedbacks, and the specific mechanism of the two cell death modes is still largely unknown, and the in-depth study of these pathways may provide targets and a theoretical basis for the development of ferroptosis and pyroptosis-targeted drugs.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Wang Y S-Editor: Qu XL L-Editor: Webster JR P-Editor: Zhang XD

References
1.  Lemmer P, Sowa JP, Bulut Y, Strnad P, Canbay A. Mechanisms and aetiology-dependent treatment of acute liver failure. Liver Int. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Vento S, Cainelli F. Acute liver failure in low-income and middle-income countries. Lancet Gastroenterol Hepatol. 2023;8:1035-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Reference Citation Analysis (0)]
3.  Stravitz RT, Lee WM. Acute liver failure. Lancet. 2019;394:869-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 435]  [Article Influence: 87.0]  [Reference Citation Analysis (0)]
4.  Li L, Shan S, Kang K, Zhang C, Kou R, Song F. The cross-talk of NLRP3 inflammasome activation and necroptotic hepatocyte death in acetaminophen-induced mice acute liver injury. Hum Exp Toxicol. 2021;40:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
5.  Bernal W, Wendon J. Acute liver failure. N Engl J Med. 2013;369:2525-2534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 736]  [Cited by in F6Publishing: 764]  [Article Influence: 69.5]  [Reference Citation Analysis (2)]
6.  Du WJ, Liu L, Sun C, Yu JH, Xiao D, Li Q. Prodromal fever indicates a high risk of liver failure in acute hepatitis B. Int J Infect Dis. 2017;57:98-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
7.  Bechmann LP, Marquitan G, Jochum C, Saner F, Gerken G, Canbay A. Apoptosis versus necrosis rate as a predictor in acute liver failure following acetaminophen intoxication compared with acute-on-chronic liver failure. Liver Int. 2008;28:713-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
8.  Sahoo G, Samal D, Khandayataray P, Murthy MK. A Review on Caspases: Key Regulators of Biological Activities and Apoptosis. Mol Neurobiol. 2023;60:5805-5837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 21]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
9.  Rutherford AE, Hynan LS, Borges CB, Forcione DG, Blackard JT, Lin W, Gorman AR, Shaikh OS, Reuben A, Harrison E, Reddy KR, Le WM, Chung RT; ALF Study Group. Serum apoptosis markers in acute liver failure: a pilot study. Clin Gastroenterol Hepatol. 2007;5:1477-1483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
10.  Kondo T, Fujimoto K, Fujiwara K, Yumita S, Ishino T, Ogawa K, Nakagawa M, Iwanaga T, Koroki K, Kanzaki H, Inoue M, Kobayashi K, Kiyono S, Nakamura M, Kanogawa N, Ogasawara S, Nakamoto S, Chiba T, Kato J, Fujiwara K, Kato N. Potential of circulating receptor-interacting protein kinase 3 levels as a marker of acute liver injury. Sci Rep. 2023;13:14043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Yamada N, Karasawa T, Kimura H, Watanabe S, Komada T, Kamata R, Sampilvanjil A, Ito J, Nakagawa K, Kuwata H, Hara S, Mizuta K, Sakuma Y, Sata N, Takahashi M. Ferroptosis driven by radical oxidation of n-6 polyunsaturated fatty acids mediates acetaminophen-induced acute liver failure. Cell Death Dis. 2020;11:144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 173]  [Article Influence: 43.3]  [Reference Citation Analysis (0)]
12.  Xing G, Meng L, Cao S, Liu S, Wu J, Li Q, Huang W, Zhang L. PPARα alleviates iron overload-induced ferroptosis in mouse liver. EMBO Rep. 2022;23:e52280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 45]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
13.  Wu J, Xue R, Wu M, Yin X, Xie B, Meng Q. Nrf2-Mediated Ferroptosis Inhibition Exerts a Protective Effect on Acute-on-Chronic Liver Failure. Oxid Med Cell Longev. 2022;2022:4505513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 20]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
14.  Luo J, Song G, Chen N, Xie M, Niu X, Zhou S, Ji Y, Zhu X, Ma W, Zhang Q, Yu D. Ferroptosis contributes to ethanol-induced hepatic cell death via labile iron accumulation and GPx4 inactivation. Cell Death Discov. 2023;9:311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  Hurtado-Navarro L, Angosto-Bazarra D, Pelegrín P, Baroja-Mazo A, Cuevas S. NLRP3 Inflammasome and Pyroptosis in Liver Pathophysiology: The Emerging Relevance of Nrf2 Inducers. Antioxidants (Basel). 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
16.  Wang K, Ding J, Shao F. Determination of Gasdermin Pores. Methods Mol Biol. 2023;2696:149-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2573]  [Cited by in F6Publishing: 3879]  [Article Influence: 431.0]  [Reference Citation Analysis (0)]
18.  Gaul S, Leszczynska A, Alegre F, Kaufmann B, Johnson CD, Adams LA, Wree A, Damm G, Seehofer D, Calvente CJ, Povero D, Kisseleva T, Eguchi A, McGeough MD, Hoffman HM, Pelegrin P, Laufs U, Feldstein AE. Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis. J Hepatol. 2021;74:156-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 286]  [Article Influence: 95.3]  [Reference Citation Analysis (0)]
19.  Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol. 2023;14:1279264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
20.  Riegman M, Sagie L, Galed C, Levin T, Steinberg N, Dixon SJ, Wiesner U, Bradbury MS, Niethammer P, Zaritsky A, Overholtzer M. Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat Cell Biol. 2020;22:1042-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 234]  [Article Influence: 58.5]  [Reference Citation Analysis (0)]
21.  Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, Cao L, Xie M, Ran Q, Kroemer G, Wang H, Billiar TR, Jiang J, Tang D. Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe. 2018;24:97-108.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 382]  [Article Influence: 63.7]  [Reference Citation Analysis (0)]
22.  Fan R, Sui J, Dong X, Jing B, Gao Z. Wedelolactone alleviates acute pancreatitis and associated lung injury via GPX4 mediated suppression of pyroptosis and ferroptosis. Free Radic Biol Med. 2021;173:29-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 51]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
23.  Wang X, Liu K, Gong H, Li D, Chu W, Zhao D, Wang X, Xu D. Death by histone deacetylase inhibitor quisinostat in tongue squamous cell carcinoma via apoptosis, pyroptosis, and ferroptosis. Toxicol Appl Pharmacol. 2021;410:115363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
24.  Gu X, Wang Y, He Y, Zhao B, Zhang Q, Li S. MiR-1656 targets GPX4 to trigger pyroptosis in broilers kidney tissues by activating NLRP3 inflammasome under Se deficiency. J Nutr Biochem. 2022;105:109001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
25.  Feng F, Luo R, Mu D, Cai Q. Ferroptosis and Pyroptosis in Epilepsy. Mol Neurobiol. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
26.  Zhou XN, Zhang Q, Peng H, Qin YJ, Liu YH, Wang L, Cheng ML, Luo XH, Li H. Silent information regulator sirtuin 1 ameliorates acute liver failure via the p53/glutathione peroxidase 4/gasdermin D axis. World J Gastroenterol. 2024;30:1588-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Vento S, Cainelli F. Acute liver failure. Lancet. 2020;395:1833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
28.  Cheng H, Shi Y, Li X, Jin N, Zhang M, Liu Z, Liang Y, Xie J. Human umbilical cord mesenchymal stem cells protect against ferroptosis in acute liver failure through the IGF1-hepcidin-FPN1 axis and inhibiting iron loading. Acta Biochim Biophys Sin (Shanghai). 2024;56:280-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
29.  Tang D, Kroemer G. Ferroptosis. Curr Biol. 2020;30:R1292-R1297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 307]  [Article Influence: 102.3]  [Reference Citation Analysis (0)]
30.  Proneth B, Conrad M. Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 2019;26:14-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 214]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
31.  Wang Y, Zheng L, Shang W, Yang Z, Li T, Liu F, Shao W, Lv L, Chai L, Qu L, Xu Q, Du J, Liang X, Zeng J, Jia J. Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 2022;29:2190-2202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 131]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
32.  Ursini F, Maiorino M. Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic Biol Med. 2020;152:175-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 783]  [Article Influence: 195.8]  [Reference Citation Analysis (0)]
33.  Liu P, Yuan J, Feng Y, Chen X, Wang G, Zhao L. Ferroptosis contributes to isoflurane-induced neurotoxicity and learning and memory impairment. Cell Death Discov. 2021;7:72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
34.  Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, Gu W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1173]  [Cited by in F6Publishing: 1967]  [Article Influence: 218.6]  [Reference Citation Analysis (0)]
35.  Lin F, Chen W, Zhou J, Zhu J, Yao Q, Feng B, Feng X, Shi X, Pan Q, Yu J, Li L, Cao H. Mesenchymal stem cells protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in acute liver injury. Cell Death Dis. 2022;13:271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 71]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
36.  Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 951]  [Article Influence: 237.8]  [Reference Citation Analysis (0)]
37.  Li W, Li W, Li X, Wang L, Wang Y. Effect of P53 nuclear localization mediated by G3BP1 on ferroptosis in acute liver failure. Apoptosis. 2023;28:1226-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
38.  Liu GZ, Xu XW, Tao SH, Gao MJ, Hou ZH. HBx facilitates ferroptosis in acute liver failure via EZH2 mediated SLC7A11 suppression. J Biomed Sci. 2021;28:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
39.  Bellezza I, Giambanco I, Minelli A, Donato R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res. 2018;1865:721-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 565]  [Cited by in F6Publishing: 1008]  [Article Influence: 168.0]  [Reference Citation Analysis (0)]
40.  Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, Hoffman HM, Feldstein AE. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology. 2014;59:898-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 687]  [Cited by in F6Publishing: 698]  [Article Influence: 69.8]  [Reference Citation Analysis (0)]
41.  He Y, Hara H, Núñez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci. 2016;41:1012-1021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1358]  [Cited by in F6Publishing: 1875]  [Article Influence: 234.4]  [Reference Citation Analysis (0)]
42.  Zheng Z, Bian Y, Zhang Y, Ren G, Li G. Metformin activates AMPK/SIRT1/NF-κB pathway and induces mitochondrial dysfunction to drive caspase3/GSDME-mediated cancer cell pyroptosis. Cell Cycle. 2020;19:1089-1104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 123]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
43.  Li H, Zhao XK, Cheng YJ, Zhang Q, Wu J, Lu S, Zhang W, Liu Y, Zhou MY, Wang Y, Yang J, Cheng ML. Gasdermin D-mediated hepatocyte pyroptosis expands inflammatory responses that aggravate acute liver failure by upregulating monocyte chemotactic protein 1/CC chemokine receptor-2 to recruit macrophages. World J Gastroenterol. 2019;25:6527-6540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (1)]
44.  Hou W, Wei X, Liang J, Fang P, Ma C, Zhang Q, Gao Y. HMGB1-Induced Hepatocyte Pyroptosis Expanding Inflammatory Responses Contributes to the Pathogenesis of Acute-on-Chronic Liver Failure (ACLF). J Inflamm Res. 2021;14:7295-7313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
45.  Wang J, Ren H, Yuan X, Ma H, Shi X, Ding Y. Interleukin-10 secreted by mesenchymal stem cells attenuates acute liver failure through inhibiting pyroptosis. Hepatol Res. 2018;48:E194-E202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 64]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
46.  Zhang H, Du Y, Guo Y, Wang Z, Li H, Lv Z, Zeng L, Chen Y, Xie Z, Li R. TLR4-NLRP3-GSDMD-Mediated Pyroptosis Plays an Important Role in Aggravated Liver Injury of CD38(-/-) Sepsis Mice. J Immunol Res. 2021;2021:6687555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, Yang X, Fei J, Hao X, Zhao Y, Gui L, Ding X. Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 2021;17:2703-2717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 179]  [Article Influence: 59.7]  [Reference Citation Analysis (1)]
48.  Liu Q, Liu Y, Li Y, Hong Z, Li S, Liu C. PUM2 aggravates the neuroinflammation and brain damage induced by ischemia-reperfusion through the SLC7A11-dependent inhibition of ferroptosis via suppressing the SIRT1. Mol Cell Biochem. 2023;478:609-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
49.  Li C, Wu Z, Xue H, Gao Q, Zhang Y, Wang C, Zhao P. Ferroptosis contributes to hypoxic-ischemic brain injury in neonatal rats: Role of the SIRT1/Nrf2/GPx4 signaling pathway. CNS Neurosci Ther. 2022;28:2268-2280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 26]  [Reference Citation Analysis (0)]
50.  Ma Z, Tang P, Dong W, Lu Y, Tan B, Zhou N, Hao J, Shen J, Hu Z. SIRT1 alleviates IL-1β induced nucleus pulposus cells pyroptosis via mitophagy in intervertebral disc degeneration. Int Immunopharmacol. 2022;107:108671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 34]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
51.  Chen Q, Wang Y, Jiao F, Cao P, Shi C, Pei M, Wang L, Gong Z. HDAC6 inhibitor ACY1215 inhibits the activation of NLRP3 inflammasome in acute liver failure by regulating the ATM/F-actin signalling pathway. J Cell Mol Med. 2021;25:7218-7228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
52.  Chen Q, Zhang Q, Cao P, Shi C, Zhang L, Wang L, Gong Z. NOD2-mediated HDAC6/NF-κb signalling pathway regulates ferroptosis induced by extracellular histone H3 in acute liver failure. J Cell Mol Med. 2022;26:5528-5538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
53.  Wang Y, Chen Q, Jiao F, Shi C, Pei M, Wang L, Gong Z. Histone deacetylase 2 regulates ULK1 mediated pyroptosis during acute liver failure by the K68 acetylation site. Cell Death Dis. 2021;12:55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
54.  Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2184]  [Cited by in F6Publishing: 2533]  [Article Influence: 844.3]  [Reference Citation Analysis (0)]
55.  Guo G, Yang W, Sun C, Wang X. Dissecting the potential role of ferroptosis in liver diseases: an updated review. Free Radic Res. 2023;57:282-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
56.  Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021;31:107-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1311]  [Cited by in F6Publishing: 1597]  [Article Influence: 532.3]  [Reference Citation Analysis (0)]
57.  Herskovits AZ, Guarente L. SIRT1 in neurodevelopment and brain senescence. Neuron. 2014;81:471-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 359]  [Article Influence: 35.9]  [Reference Citation Analysis (0)]
58.  Zeng J, Guo J, Huang S, Cheng Y, Luo F, Xu X, Chen R, Ma G, Wang Y. The roles of sirtuins in ferroptosis. Front Physiol. 2023;14:1131201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
59.  Li T, Zheng G, Li B, Tang L. Pyroptosis: A promising therapeutic target for noninfectious diseases. Cell Prolif. 2021;54:e13137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
60.  Zhang Y, Zhang H, Li S, Huang K, Jiang L, Wang Y. Metformin Alleviates LPS-Induced Acute Lung Injury by Regulating the SIRT1/NF-κB/NLRP3 Pathway and Inhibiting Endothelial Cell Pyroptosis. Front Pharmacol. 2022;13:801337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 22]  [Reference Citation Analysis (0)]
61.  Chen M, Rong R, Xia X. Spotlight on pyroptosis: role in pathogenesis and therapeutic potential of ocular diseases. J Neuroinflammation. 2022;19:183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 33]  [Reference Citation Analysis (0)]
62.  Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, Li S, Wang B, Nemmara VV, Wilson R, Jiang Z, Khalighinejad F, Muneeruddin K, Shaffer SA, Dutta R, Ionete C, Pesiridis S, Yang S, Thompson PR, Fitzgerald KA. Succination inactivates gasdermin D and blocks pyroptosis. Science. 2020;369:1633-1637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 340]  [Article Influence: 85.0]  [Reference Citation Analysis (0)]