Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases
Abstract
:1. Introduction
2. Forms of Programmed Cell Death
2.1. Apoptosis
2.2. Necroptosis
2.3. Pyroptosis
2.4. Ferroptosis
2.5. Cuproptosis
2.6. Other Forms of PCD
2.7. Characteristics of Different PCD Subroutines
3. PCD in NDDs
3.1. PCD in AD
3.2. PCD in PD
3.3. PCD in HD
3.4. PCD in ALS, SMA and MS
3.5. PCD in TBI and Stroke
3.6. Therapeutic Strategies Targeting PCD Signaling Pathways in NDDs
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
- Peng, F.; Liao, M.; Qin, R.; Zhu, S.; Peng, C.; Fu, L.; Chen, Y.; Han, B. Regulated cell death (RCD) in cancer: Key pathways and targeted therapies. Signal Transduct. Target. Ther. 2022, 7, 286. [Google Scholar] [CrossRef] [PubMed]
- Tong, X.; Tang, R.; Xiao, M.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Yu, X.; Shi, S. Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J. Hematol. Oncol. 2022, 15, 174. [Google Scholar] [CrossRef] [PubMed]
- Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef]
- Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef]
- Liu, X.; Zhuang, L.; Gan, B. Disulfidptosis: Disulfide stress-induced cell death. Trends Cell Biol. 2024, 34, 327–337. [Google Scholar] [CrossRef]
- Chen, L.; Min, J.; Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 2022, 7, 378. [Google Scholar] [CrossRef]
- Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
- Collaborators, G.B.D.N. Global, regional, and national burden of neurological disorders, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef]
- Jia, L.; Du, Y.; Chu, L.; Zhang, Z.; Li, F.; Lyu, D.; Li, Y.; Li, Y.; Zhu, M.; Jiao, H.; et al. Prevalence, risk factors, and management of dementia and mild cognitive impairment in adults aged 60 years or older in China: A cross-sectional study. Lancet Public Health 2020, 5, e661–e671. [Google Scholar] [CrossRef]
- Rawlins, M.D.; Wexler, N.S.; Wexler, A.R.; Tabrizi, S.J.; Douglas, I.; Evans, S.J.; Smeeth, L. The Prevalence of Huntington’s Disease. Neuroepidemiology 2016, 46, 144–153. [Google Scholar] [CrossRef] [PubMed]
- Feldman, E.L.; Goutman, S.A.; Petri, S.; Mazzini, L.; Savelieff, M.G.; Shaw, P.J.; Sobue, G. Amyotrophic lateral sclerosis. Lancet 2022, 400, 1363–1380. [Google Scholar] [CrossRef] [PubMed]
- Mercuri, E.; Sumner, C.J.; Muntoni, F.; Darras, B.T.; Finkel, R.S. Spinal muscular atrophy. Nat. Rev. Dis. Primers 2022, 8, 52. [Google Scholar] [CrossRef] [PubMed]
- Owens, B. Multiple sclerosis. Nature 2016, 540, S1. [Google Scholar] [CrossRef]
- Zille, M.; Ikhsan, M.; Jiang, Y.; Lampe, J.; Wenzel, J.; Schwaninger, M. The impact of endothelial cell death in the brain and its role after stroke: A systematic review. Cell Stress 2019, 3, 330–347. [Google Scholar] [CrossRef]
- Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int. J. Stroke 2022, 17, 18–29. [Google Scholar] [CrossRef]
- Moujalled, D.; Strasser, A.; Liddell, J.R. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 2021, 28, 2029–2044. [Google Scholar] [CrossRef]
- Kuan, C.Y.; Roth, K.A.; Flavell, R.A.; Rakic, P. Mechanisms of programmed cell death in the developing brain. Trends Neurosci. 2000, 23, 291–297. [Google Scholar] [CrossRef]
- Fogarty, L.C.; Flemmer, R.T.; Geizer, B.A.; Licursi, M.; Karunanithy, A.; Opferman, J.T.; Hirasawa, K.; Vanderluit, J.L. Mcl-1 and Bcl-xL are essential for survival of the developing nervous system. Cell Death Differ. 2019, 26, 1501–1515. [Google Scholar] [CrossRef]
- Arbour, N.; Vanderluit, J.L.; Le Grand, J.N.; Jahani-Asl, A.; Ruzhynsky, V.A.; Cheung, E.C.; Kelly, M.A.; MacKenzie, A.E.; Park, D.S.; Opferman, J.T.; et al. Mcl-1 is a key regulator of apoptosis during CNS development and after DNA damage. J. Neurosci. 2008, 28, 6068–6078. [Google Scholar] [CrossRef]
- Ke, F.F.S.; Vanyai, H.K.; Cowan, A.D.; Delbridge, A.R.D.; Whitehead, L.; Grabow, S.; Czabotar, P.E.; Voss, A.K.; Strasser, A. Embryogenesis and Adult Life in the Absence of Intrinsic Apoptosis Effectors BAX, BAK, and BOK. Cell 2018, 173, 1217–1230.e17. [Google Scholar] [CrossRef] [PubMed]
- Gorman, A.M. Neuronal cell death in neurodegenerative diseases: Recurring themes around protein handling. J. Cell Mol. Med. 2008, 12, 2263–2280. [Google Scholar] [CrossRef] [PubMed]
- Roberts, J.Z.; Crawford, N.; Longley, D.B. The role of Ubiquitination in Apoptosis and Necroptosis. Cell Death Differ. 2022, 29, 272–284. [Google Scholar] [CrossRef]
- Puthalakath, H.; Strasser, A. Keeping killers on a tight leash: Transcriptional and post-translational control of the pro-apoptotic activity of BH3-only proteins. Cell Death Differ. 2002, 9, 505–512. [Google Scholar] [CrossRef]
- Chen, L.; Willis, S.N.; Wei, A.; Smith, B.J.; Fletcher, J.I.; Hinds, M.G.; Colman, P.M.; Day, C.L.; Adams, J.M.; Huang, D.C. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol. Cell 2005, 17, 393–403. [Google Scholar] [CrossRef]
- Moldoveanu, T.; Czabotar, P.E. BAX, BAK, and BOK: A Coming of Age for the BCL-2 Family Effector Proteins. Cold Spring Harb. Perspect. Biol. 2020, 12, a036319. [Google Scholar] [CrossRef]
- Batandier, C.; Leverve, X.; Fontaine, E. Opening of the mitochondrial permeability transition pore induces reactive oxygen species production at the level of the respiratory chain complex I. J. Biol. Chem. 2004, 279, 17197–17204. [Google Scholar] [CrossRef]
- Danial, N.N.; Korsmeyer, S.J. Cell death: Critical control points. Cell 2004, 116, 205–219. [Google Scholar] [CrossRef] [PubMed]
- Wang, X. The expanding role of mitochondria in apoptosis. Genes. Dev. 2001, 15, 2922–2933. [Google Scholar]
- Slee, E.A.; Adrain, C.; Martin, S.J. Serial killers: Ordering caspase activation events in apoptosis. Cell Death Differ. 1999, 6, 1067–1074. [Google Scholar] [CrossRef]
- Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef] [PubMed]
- Pinkaew, D.; Chattopadhyay, A.; King, M.D.; Chunhacha, P.; Liu, Z.; Stevenson, H.L.; Chen, Y.; Sinthujaroen, P.; McDougal, O.M.; Fujise, K. Fortilin binds IRE1alpha and prevents ER stress from signaling apoptotic cell death. Nat. Commun. 2017, 8, 18. [Google Scholar] [CrossRef] [PubMed]
- Song, S.; Tan, J.; Miao, Y.; Li, M.; Zhang, Q. Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress. J. Cell Physiol. 2017, 232, 2977–2984. [Google Scholar] [CrossRef]
- Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
- Susin, S.A.; Lorenzo, H.K.; Zamzami, N.; Marzo, I.; Snow, B.E.; Brothers, G.M.; Mangion, J.; Jacotot, E.; Costantini, P.; Loeffler, M.; et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999, 397, 441–446. [Google Scholar] [CrossRef] [PubMed]
- Joza, N.; Susin, S.A.; Daugas, E.; Stanford, W.L.; Cho, S.K.; Li, C.Y.; Sasaki, T.; Elia, A.J.; Cheng, H.Y.; Ravagnan, L.; et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature 2001, 410, 549–554. [Google Scholar] [CrossRef]
- Strasser, A.; Jost, P.J.; Nagata, S. The many roles of FAS receptor signaling in the immune system. Immunity 2009, 30, 180–192. [Google Scholar] [CrossRef]
- Shalini, S.; Dorstyn, L.; Dawar, S.; Kumar, S. Old, new and emerging functions of caspases. Cell Death Differ. 2015, 22, 526–539. [Google Scholar] [CrossRef]
- Fulda, S.; Debatin, K.M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef]
- Vitale, I.; Pietrocola, F.; Guilbaud, E.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostini, M.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; et al. Apoptotic cell death in disease-Current understanding of the NCCD 2023. Cell Death Differ. 2023, 30, 1097–1154. [Google Scholar] [CrossRef]
- Green, D.R. Apoptotic pathways: Paper wraps stone blunts scissors. Cell 2000, 102, 1–4. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef]
- Yan, J.; Wan, P.; Choksi, S.; Liu, Z.G. Necroptosis and tumor progression. Trends Cancer 2022, 8, 21–27. [Google Scholar] [CrossRef]
- Tummers, B.; Green, D.R. Caspase-8: Regulating life and death. Immunol. Rev. 2017, 277, 76–89. [Google Scholar] [CrossRef] [PubMed]
- Yuan, J.; Amin, P.; Ofengeim, D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat. Rev. Neurosci. 2019, 20, 19–33. [Google Scholar] [CrossRef]
- Laurien, L.; Nagata, M.; Schunke, H.; Delanghe, T.; Wiederstein, J.L.; Kumari, S.; Schwarzer, R.; Corona, T.; Kruger, M.; Bertrand, M.J.M.; et al. Autophosphorylation at serine 166 regulates RIP kinase 1-mediated cell death and inflammation. Nat. Commun. 2020, 11, 1747. [Google Scholar] [CrossRef]
- Vandenabeele, P.; Galluzzi, L.; Vanden Berghe, T.; Kroemer, G. Molecular mechanisms of necroptosis: An ordered cellular explosion. Nat. Rev. Mol. Cell Biol. 2010, 11, 700–714. [Google Scholar] [CrossRef] [PubMed]
- Degterev, A.; Hitomi, J.; Germscheid, M.; Ch’en, I.L.; Korkina, O.; Teng, X.; Abbott, D.; Cuny, G.D.; Yuan, C.; Wagner, G.; et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 2008, 4, 313–321. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wang, H.; Wang, Z.; He, S.; Chen, S.; Liao, D.; Wang, L.; Yan, J.; Liu, W.; Lei, X.; et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 2012, 148, 213–227. [Google Scholar] [CrossRef]
- Murphy, J.M.; Czabotar, P.E.; Hildebrand, J.M.; Lucet, I.S.; Zhang, J.G.; Alvarez-Diaz, S.; Lewis, R.; Lalaoui, N.; Metcalf, D.; Webb, A.I.; et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 2013, 39, 443–453. [Google Scholar] [CrossRef]
- Samson, A.L.; Zhang, Y.; Geoghegan, N.D.; Gavin, X.J.; Davies, K.A.; Mlodzianoski, M.J.; Whitehead, L.W.; Frank, D.; Garnish, S.E.; Fitzgibbon, C.; et al. MLKL trafficking and accumulation at the plasma membrane control the kinetics and threshold for necroptosis. Nat. Commun. 2020, 11, 3151. [Google Scholar] [CrossRef] [PubMed]
- Patel, S.; Webster, J.D.; Varfolomeev, E.; Kwon, Y.C.; Cheng, J.H.; Zhang, J.; Dugger, D.L.; Wickliffe, K.E.; Maltzman, A.; Sujatha-Bhaskar, S.; et al. RIP1 inhibition blocks inflammatory diseases but not tumor growth or metastases. Cell Death Differ. 2020, 27, 161–175. [Google Scholar] [CrossRef] [PubMed]
- Strilic, B.; Yang, L.; Albarran-Juarez, J.; Wachsmuth, L.; Han, K.; Muller, U.C.; Pasparakis, M.; Offermanns, S. Tumour-cell-induced endothelial cell necroptosis via death receptor 6 promotes metastasis. Nature 2016, 536, 215–218. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Yin, C.; Boyd, D.F.; Quarato, G.; Ingram, J.P.; Shubina, M.; Ragan, K.B.; Ishizuka, T.; Crawford, J.C.; Tummers, B.; et al. Influenza Virus Z-RNAs Induce ZBP1-Mediated Necroptosis. Cell 2020, 180, 1115–1129.e1113. [Google Scholar] [CrossRef]
- Upton, J.W.; Kaiser, W.J.; Mocarski, E.S. DAI/ZBP1/DLM-1 Complexes with RIP3 to Mediate Virus-Induced Programmed Necrosis that Is Targeted by Murine Cytomegalovirus vIRA. Cell Host Microbe 2019, 26, 564. [Google Scholar] [CrossRef]
- Kaiser, W.J.; Sridharan, H.; Huang, C.; Mandal, P.; Upton, J.W.; Gough, P.J.; Sehon, C.A.; Marquis, R.W.; Bertin, J.; Mocarski, E.S. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J. Biol. Chem. 2013, 288, 31268–31279. [Google Scholar] [CrossRef]
- Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef]
- Yang, D.; Liang, Y.; Zhao, S.; Ding, Y.; Zhuang, Q.; Shi, Q.; Ai, T.; Wu, S.Q.; Han, J. ZBP1 mediates interferon-induced necroptosis. Cell Mol. Immunol. 2020, 17, 356–368. [Google Scholar] [CrossRef]
- Zhou, J.; Qiu, J.; Song, Y.; Liang, T.; Liu, S.; Ren, C.; Song, X.; Cui, L.; Sun, Y. Pyroptosis and degenerative diseases of the elderly. Cell Death Dis. 2023, 14, 94. [Google Scholar] [CrossRef]
- Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and diseases. Signal Transduct. Target. Ther. 2021, 6, 128. [Google Scholar] [CrossRef]
- Rao, Z.; Zhu, Y.; Yang, P.; Chen, Z.; Xia, Y.; Qiao, C.; Liu, W.; Deng, H.; Li, J.; Ning, P.; et al. Pyroptosis in inflammatory diseases and cancer. Theranostics 2022, 12, 4310–4329. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Zhang, Z.; Ruan, J.; Pan, Y.; Magupalli, V.G.; Wu, H.; Lieberman, J. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 2016, 535, 153–158. [Google Scholar] [CrossRef]
- Coll, R.C.; Schroder, K.; Pelegrin, P. NLRP3 and pyroptosis blockers for treating inflammatory diseases. Trends Pharmacol. Sci. 2022, 43, 653–668. [Google Scholar] [CrossRef] [PubMed]
- Elias, E.E.; Lyons, B.; Muruve, D.A. Gasdermins and pyroptosis in the kidney. Nat. Rev. Nephrol. 2023, 19, 337–350. [Google Scholar] [CrossRef] [PubMed]
- Deng, M.; Tang, Y.; Li, W.; Wang, X.; Zhang, R.; Zhang, X.; Zhao, X.; Liu, J.; Tang, C.; Liu, Z.; et al. The Endotoxin Delivery Protein HMGB1 Mediates Caspase-11-Dependent Lethality in Sepsis. Immunity 2018, 49, 740–753.e747. [Google Scholar] [CrossRef] [PubMed]
- Aglietti, R.A.; Estevez, A.; Gupta, A.; Ramirez, M.G.; Liu, P.S.; Kayagaki, N.; Ciferri, C.; Dixit, V.M.; Dueber, E.C. GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc. Natl. Acad. Sci. USA 2016, 113, 7858–7863. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Yang, D.; He, Y.; Munoz-Planillo, R.; Liu, Q.; Nunez, G. Caspase-11 Requires the Pannexin-1 Channel and the Purinergic P2X7 Pore to Mediate Pyroptosis and Endotoxic Shock. Immunity 2015, 43, 923–932. [Google Scholar] [CrossRef]
- Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef]
- Chai, Q.; Yu, S.; Zhong, Y.; Lu, Z.; Qiu, C.; Yu, Y.; Zhang, X.; Zhang, Y.; Lei, Z.; Qiang, L.; et al. A bacterial phospholipid phosphatase inhibits host pyroptosis by hijacking ubiquitin. Science 2022, 378, eabq0132. [Google Scholar] [CrossRef]
- Wang, Y.; Gao, W.; Shi, X.; Ding, J.; Liu, W.; He, H.; Wang, K.; Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 2017, 547, 99–103. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Xia, S.; Zhang, Z.; Wu, H.; Lieberman, J. Channelling inflammation: Gasdermins in physiology and disease. Nat. Rev. Drug Discov. 2021, 20, 384–405. [Google Scholar] [CrossRef]
- Hou, J.; Zhao, R.; Xia, W.; Chang, C.W.; You, Y.; Hsu, J.M.; Nie, L.; Chen, Y.; Wang, Y.C.; Liu, C.; et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat. Cell Biol. 2020, 22, 1264–1275. [Google Scholar] [CrossRef]
- Orning, P.; Weng, D.; Starheim, K.; Ratner, D.; Best, Z.; Lee, B.; Brooks, A.; Xia, S.; Wu, H.; Kelliher, M.A.; et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 2018, 362, 1064–1069. [Google Scholar] [CrossRef]
- Sarhan, J.; Liu, B.C.; Muendlein, H.I.; Li, P.; Nilson, R.; Tang, A.Y.; Rongvaux, A.; Bunnell, S.C.; Shao, F.; Green, D.R.; et al. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc. Natl. Acad. Sci. USA 2018, 115, E10888–E10897. [Google Scholar] [CrossRef] [PubMed]
- Zhao, A.; Kirkby, M.; Man, S.M. Streptococcus makes the cut: Gasdermin A-induced pyroptosis. Cell Host Microbe 2022, 30, 410–412. [Google Scholar] [CrossRef]
- Zhou, Z.; He, H.; Wang, K.; Shi, X.; Wang, Y.; Su, Y.; Wang, Y.; Li, D.; Liu, W.; Zhang, Y.; et al. Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science 2020, 368, aaz7548. [Google Scholar] [CrossRef]
- Liu, Y.; Fang, Y.; Chen, X.; Wang, Z.; Liang, X.; Zhang, T.; Liu, M.; Zhou, N.; Lv, J.; Tang, K.; et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci. Immunol. 2020, 5, aax7969. [Google Scholar] [CrossRef] [PubMed]
- Kong, Q.; Xia, S.; Pan, X.; Ye, K.; Li, Z.; Li, H.; Tang, X.; Sahni, N.; Yi, S.S.; Liu, X.; et al. Alternative splicing of GSDMB modulates killer lymphocyte-triggered pyroptosis. Sci. Immunol. 2023, 8, eadg3196. [Google Scholar] [CrossRef] [PubMed]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
- Bogdan, A.R.; Miyazawa, M.; Hashimoto, K.; Tsuji, Y. Regulators of Iron Homeostasis: New Players in Metabolism, Cell Death, and Disease. Trends Biochem. Sci. 2016, 41, 274–286. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.Y.; Han, Y.; Li, W.N.; Xu, R.H.; Ju, H.Q. Tumor iron homeostasis and immune regulation. Trends Pharmacol. Sci. 2024, 45, 145–156. [Google Scholar] [CrossRef]
- Galy, B.; Conrad, M.; Muckenthaler, M. Mechanisms controlling cellular and systemic iron homeostasis. Nat. Rev. Mol. Cell Biol. 2024, 25, 133–155. [Google Scholar] [CrossRef] [PubMed]
- Roemhild, K.; von Maltzahn, F.; Weiskirchen, R.; Knuchel, R.; von Stillfried, S.; Lammers, T. Iron metabolism: Pathophysiology and pharmacology. Trends Pharmacol. Sci. 2021, 42, 640–656. [Google Scholar] [CrossRef]
- Yambire, K.F.; Rostosky, C.; Watanabe, T.; Pacheu-Grau, D.; Torres-Odio, S.; Sanchez-Guerrero, A.; Senderovich, O.; Meyron-Holtz, E.G.; Milosevic, I.; Frahm, J.; et al. Impaired lysosomal acidification triggers iron deficiency and inflammation in vivo. eLife 2019, 8, e51031. [Google Scholar] [CrossRef]
- Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef]
- Fang, X.; Ardehali, H.; Min, J.; Wang, F. The molecular and metabolic landscape of iron and ferroptosis in cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 7–23. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Fang, Z.M.; Yi, X.; Wei, X.; Jiang, D.S. The interaction between ferroptosis and inflammatory signaling pathways. Cell Death Dis. 2023, 14, 205. [Google Scholar] [CrossRef]
- Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef]
- Zheng, J.; Conrad, M. The Metabolic Underpinnings of Ferroptosis. Cell Metab. 2020, 32, 920–937. [Google Scholar] [CrossRef]
- Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef] [PubMed]
- Mao, C.; Liu, X.; Zhang, Y.; Lei, G.; Yan, Y.; Lee, H.; Koppula, P.; Wu, S.; Zhuang, L.; Fang, B.; et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021, 593, 586–590. [Google Scholar] [CrossRef] [PubMed]
- Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019, 575, 688–692. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Kang, R.; Kroemer, G.; Tang, D. Broadening horizons: The role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 2021, 18, 280–296. [Google Scholar] [CrossRef]
- Qiu, S.; Zhong, X.; Meng, X.; Li, S.; Qian, X.; Lu, H.; Cai, J.; Zhang, Y.; Wang, M.; Ye, Z.; et al. Mitochondria-localized cGAS suppresses ferroptosis to promote cancer progression. Cell Res. 2023, 33, 299–311. [Google Scholar] [CrossRef]
- Liang, D.; Feng, Y.; Zandkarimi, F.; Wang, H.; Zhang, Z.; Kim, J.; Cai, Y.; Gu, W.; Stockwell, B.R.; Jiang, X. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell 2023, 186, 2748–2764.e2722. [Google Scholar] [CrossRef] [PubMed]
- Mancias, J.D.; Wang, X.; Gygi, S.P.; Harper, J.W.; Kimmelman, A.C. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 2014, 509, 105–109. [Google Scholar] [CrossRef]
- Ajoolabady, A.; Aslkhodapasandhokmabad, H.; Libby, P.; Tuomilehto, J.; Lip, G.Y.H.; Penninger, J.M.; Richardson, D.R.; Tang, D.; Zhou, H.; Wang, S.; et al. Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends Endocrinol. Metab. 2021, 32, 444–462. [Google Scholar] [CrossRef]
- Liu, J.; Kuang, F.; Kroemer, G.; Klionsky, D.J.; Kang, R.; Tang, D. Autophagy-Dependent Ferroptosis: Machinery and Regulation. Cell Chem. Biol. 2020, 27, 420–435. [Google Scholar] [CrossRef]
- Wu, Z.; Geng, Y.; Lu, X.; Shi, Y.; Wu, G.; Zhang, M.; Shan, B.; Pan, H.; Yuan, J. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl. Acad. Sci. USA 2019, 116, 2996–3005. [Google Scholar] [CrossRef]
- Yang, M.; Chen, P.; Liu, J.; Zhu, S.; Kroemer, G.; Klionsky, D.J.; Lotze, M.T.; Zeh, H.J.; Kang, R.; Tang, D. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci. Adv. 2019, 5, eaaw2238. [Google Scholar] [CrossRef] [PubMed]
- Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef] [PubMed]
- Rouault, T.A. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat. Chem. Biol. 2006, 2, 406–414. [Google Scholar] [CrossRef] [PubMed]
- Protchenko, O.; Baratz, E.; Jadhav, S.; Li, F.; Shakoury-Elizeh, M.; Gavrilova, O.; Ghosh, M.C.; Cox, J.E.; Maschek, J.A.; Tyurin, V.A.; et al. Iron Chaperone Poly rC Binding Protein 1 Protects Mouse Liver from Lipid Peroxidation and Steatosis. Hepatology 2021, 73, 1176–1193. [Google Scholar] [CrossRef]
- Paradkar, P.N.; Zumbrennen, K.B.; Paw, B.H.; Ward, D.M.; Kaplan, J. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol. Cell Biol. 2009, 29, 1007–1016. [Google Scholar] [CrossRef]
- Chang, L.C.; Chiang, S.K.; Chen, S.E.; Yu, Y.L.; Chou, R.H.; Chang, W.C. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018, 416, 124–137. [Google Scholar] [CrossRef]
- Tang, Z.; Ju, Y.; Dai, X.; Ni, N.; Liu, Y.; Zhang, D.; Gao, H.; Sun, H.; Zhang, J.; Gu, P. HO-1-mediated ferroptosis as a target for protection against retinal pigment epithelium degeneration. Redox Biol. 2021, 43, 101971. [Google Scholar] [CrossRef]
- Yu, Y.; Jiang, L.; Wang, H.; Shen, Z.; Cheng, Q.; Zhang, P.; Wang, J.; Wu, Q.; Fang, X.; Duan, L.; et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood 2020, 136, 726–739. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Guo, M.; Shen, M.; Kong, D.; Zhang, F.; Shao, J.; Tan, S.; Wang, S.; Chen, A.; Cao, P.; et al. The BRD7-P53-SLC25A28 axis regulates ferroptosis in hepatic stellate cells. Redox Biol. 2020, 36, 101619. [Google Scholar] [CrossRef]
- Li, N.; Wang, W.; Zhou, H.; Wu, Q.; Duan, M.; Liu, C.; Wu, H.; Deng, W.; Shen, D.; Tang, Q. Ferritinophagy-mediated ferroptosis is involved in sepsis-induced cardiac injury. Free Radic. Biol. Med. 2020, 160, 303–318. [Google Scholar] [CrossRef]
- Brown, C.W.; Amante, J.J.; Chhoy, P.; Elaimy, A.L.; Liu, H.; Zhu, L.J.; Baer, C.E.; Dixon, S.J.; Mercurio, A.M. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev. Cell 2019, 51, 575–586.e574. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.S.; Kim, K.J.; Gaschler, M.M.; Patel, M.; Shchepinov, M.S.; Stockwell, B.R. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, E4966–E4975. [Google Scholar] [CrossRef] [PubMed]
- Fang, X.; Wang, H.; Han, D.; Xie, E.; Yang, X.; Wei, J.; Gu, S.; Gao, F.; Zhu, N.; Yin, X.; et al. Ferroptosis as a target for protection against cardiomyopathy. Proc. Natl. Acad. Sci. USA 2019, 116, 2672–2680. [Google Scholar] [CrossRef]
- Tuo, Q.Z.; Lei, P.; Jackman, K.A.; Li, X.L.; Xiong, H.; Li, X.L.; Liuyang, Z.Y.; Roisman, L.; Zhang, S.T.; Ayton, S.; et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol. Psychiatry 2017, 22, 1520–1530. [Google Scholar] [CrossRef]
- Yuan, H.; Li, X.; Zhang, X.; Kang, R.; Tang, D. CISD1 inhibits ferroptosis by protection against mitochondrial lipid peroxidation. Biochem. Biophys. Res. Commun. 2016, 478, 838–844. [Google Scholar] [CrossRef]
- Kim, E.H.; Shin, D.; Lee, J.; Jung, A.R.; Roh, J.L. CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett. 2018, 432, 180–190. [Google Scholar] [CrossRef]
- Gunshin, H.; Mackenzie, B.; Berger, U.V.; Gunshin, Y.; Romero, M.F.; Boron, W.F.; Nussberger, S.; Gollan, J.L.; Hediger, M.A. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 1997, 388, 482–488. [Google Scholar] [CrossRef] [PubMed]
- Terzi, E.M.; Sviderskiy, V.O.; Alvarez, S.W.; Whiten, G.C.; Possemato, R. Iron-sulfur cluster deficiency can be sensed by IRP2 and regulates iron homeostasis and sensitivity to ferroptosis independent of IRP1 and FBXL5. Sci. Adv. 2021, 7, abg4302. [Google Scholar] [CrossRef]
- Wang, N.; Ma, H.; Li, J.; Meng, C.; Zou, J.; Wang, H.; Liu, K.; Liu, M.; Xiao, X.; Zhang, H.; et al. HSF1 functions as a key defender against palmitic acid-induced ferroptosis in cardiomyocytes. J. Mol. Cell Cardiol. 2021, 150, 65–76. [Google Scholar] [CrossRef]
- Zhu, S.; Zhang, Q.; Sun, X.; Zeh, H.J., 3rd; Lotze, M.T.; Kang, R.; Tang, D. HSPA5 Regulates Ferroptotic Cell Death in Cancer Cells. Cancer Res. 2017, 77, 2064–2077. [Google Scholar] [CrossRef]
- Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
- Hua, J.; Gao, Z.; Zhong, S.; Wei, B.; Zhu, J.; Ying, R. CISD1 protects against atherosclerosis by suppressing lipid accumulation and inflammation via mediating Drp1. Biochem. Biophys. Res. Commun. 2021, 577, 80–88. [Google Scholar] [CrossRef] [PubMed]
- Shui, S.; Zhao, Z.; Wang, H.; Conrad, M.; Liu, G. Non-enzymatic lipid peroxidation initiated by photodynamic therapy drives a distinct ferroptosis-like cell death pathway. Redox Biol. 2021, 45, 102056. [Google Scholar] [CrossRef] [PubMed]
- Wenzel, S.E.; Tyurina, Y.Y.; Zhao, J.; St Croix, C.M.; Dar, H.H.; Mao, G.; Tyurin, V.A.; Anthonymuthu, T.S.; Kapralov, A.A.; Amoscato, A.A.; et al. PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals. Cell 2017, 171, 628–641.e626. [Google Scholar] [CrossRef] [PubMed]
- Jha, J.C.; Banal, C.; Chow, B.S.; Cooper, M.E.; Jandeleit-Dahm, K. Diabetes and Kidney Disease: Role of Oxidative Stress. Antioxid. Redox Signal 2016, 25, 657–684. [Google Scholar] [CrossRef]
- Xie, Y.; Zhu, S.; Song, X.; Sun, X.; Fan, Y.; Liu, J.; Zhong, M.; Yuan, H.; Zhang, L.; Billiar, T.R.; et al. The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity. Cell Rep. 2017, 20, 1692–1704. [Google Scholar] [CrossRef]
- Nieto-Fontarigo, J.J.; Gonzalez-Barcala, F.J.; San Jose, E.; Arias, P.; Nogueira, M.; Salgado, F.J. CD26 and Asthma: A Comprehensive Review. Clin. Rev. Allergy Immunol. 2019, 56, 139–160. [Google Scholar] [CrossRef]
- Yang, Y.; Luo, M.; Zhang, K.; Zhang, J.; Gao, T.; Connell, D.O.; Yao, F.; Mu, C.; Cai, B.; Shang, Y.; et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat. Commun. 2020, 11, 433. [Google Scholar] [CrossRef]
- Liang, Z.; Wu, Q.; Wang, H.; Tan, J.; Wang, H.; Gou, Y.; Cao, Y.; Li, Z.; Zhang, Z. Silencing of lncRNA MALAT1 facilitates erastin-induced ferroptosis in endometriosis through miR-145-5p/MUC1 signaling. Cell Death Discov. 2022, 8, 190. [Google Scholar] [CrossRef]
- Kang, Y.P.; Mockabee-Macias, A.; Jiang, C.; Falzone, A.; Prieto-Farigua, N.; Stone, E.; Harris, I.S.; DeNicola, G.M. Non-canonical Glutamate-Cysteine Ligase Activity Protects against Ferroptosis. Cell Metab. 2021, 33, 174–189.e177. [Google Scholar] [CrossRef]
- Suzuki, S.; Venkatesh, D.; Kanda, H.; Nakayama, A.; Hosokawa, H.; Lee, E.; Miki, T.; Stockwell, B.R.; Yokote, K.; Tanaka, T.; et al. GLS2 Is a Tumor Suppressor and a Regulator of Ferroptosis in Hepatocellular Carcinoma. Cancer Res. 2022, 82, 3209–3222. [Google Scholar] [CrossRef] [PubMed]
- Hayano, M.; Yang, W.S.; Corn, C.K.; Pagano, N.C.; Stockwell, B.R. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 2016, 23, 270–278. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Zhang, H. CHAC1 exacerbates LPS-induced ferroptosis and apoptosis in HK-2 cells by promoting oxidative stress. Allergol. Immunopathol. (Madr.) 2023, 51, 99–110. [Google Scholar] [CrossRef]
- Wang, Z.; Chen, X.; Liu, N.; Shi, Y.; Liu, Y.; Ouyang, L.; Tam, S.; Xiao, D.; Liu, S.; Wen, F.; et al. A Nuclear Long Non-Coding RNA LINC00618 Accelerates Ferroptosis in a Manner Dependent upon Apoptosis. Mol. Ther. 2021, 29, 263–274. [Google Scholar] [CrossRef]
- Yamane, D.; Hayashi, Y.; Matsumoto, M.; Nakanishi, H.; Imagawa, H.; Kohara, M.; Lemon, S.M.; Ichi, I. FADS2-dependent fatty acid desaturation dictates cellular sensitivity to ferroptosis and permissiveness for hepatitis C virus replication. Cell Chem. Biol. 2022, 29, 799–810.e794. [Google Scholar] [CrossRef]
- Klasson, T.D.; LaGory, E.L.; Zhao, H.; Huynh, S.K.; Papandreou, I.; Moon, E.J.; Giaccia, A.J. ACSL3 regulates lipid droplet biogenesis and ferroptosis sensitivity in clear cell renal cell carcinoma. Cancer Metab. 2022, 10, 14. [Google Scholar] [CrossRef] [PubMed]
- Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef]
- Liu, J.; Kang, R.; Tang, D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022, 289, 7038–7050. [Google Scholar] [CrossRef]
- Shen, L.; Zhang, J.; Zheng, Z.; Yang, F.; Liu, S.; Wu, Y.; Chen, Y.; Xu, T.; Mao, S.; Yan, Y.; et al. PHGDH Inhibits Ferroptosis and Promotes Malignant Progression by Upregulating SLC7A11 in Bladder Cancer. Int. J. Biol. Sci. 2022, 18, 5459–5474. [Google Scholar] [CrossRef]
- Cao, F.; Luo, A.; Yang, C. G6PD inhibits ferroptosis in hepatocellular carcinoma by targeting cytochrome P450 oxidoreductase. Cell Signal 2021, 87, 110098. [Google Scholar] [CrossRef]
- Fang, X.; Zhang, J.; Li, Y.; Song, Y.; Yu, Y.; Cai, Z.; Lian, F.; Yang, J.; Min, J.; Wang, F. Malic Enzyme 1 as a Novel Anti-Ferroptotic Regulator in Hepatic Ischemia/Reperfusion Injury. Adv. Sci. (Weinh) 2023, 10, e2205436. [Google Scholar] [CrossRef] [PubMed]
- Zhu, W.; Liu, D.; Lu, Y.; Sun, J.; Zhu, J.; Xing, Y.; Ma, X.; Wang, Y.; Ji, M.; Jia, Y. PHKG2 regulates RSL3-induced ferroptosis in Helicobacter pylori related gastric cancer. Arch. Biochem. Biophys. 2023, 740, 109560. [Google Scholar] [CrossRef]
- Hassannia, B.; Vandenabeele, P.; Vanden Berghe, T. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef]
- Garcia-Bermudez, J.; Baudrier, L.; Bayraktar, E.C.; Shen, Y.; La, K.; Guarecuco, R.; Yucel, B.; Fiore, D.; Tavora, B.; Freinkman, E.; et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 2019, 567, 118–122. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef]
- Liu, Y.; Gu, W. p53 in ferroptosis regulation: The new weapon for the old guardian. Cell Death Differ. 2022, 29, 895–910. [Google Scholar] [CrossRef] [PubMed]
- Lin, Z.; Song, J.; Gao, Y.; Huang, S.; Dou, R.; Zhong, P.; Huang, G.; Han, L.; Zheng, J.; Zhang, X.; et al. Hypoxia-induced HIF-1alpha/lncRNA-PMAN inhibits ferroptosis by promoting the cytoplasmic translocation of ELAVL1 in peritoneal dissemination from gastric cancer. Redox Biol. 2022, 52, 102312. [Google Scholar] [CrossRef]
- Xie, X.; Tian, L.; Zhao, Y.; Liu, F.; Dai, S.; Gu, X.; Ye, Y.; Zhou, L.; Liu, X.; Sun, Y.; et al. BACH1-induced ferroptosis drives lymphatic metastasis by repressing the biosynthesis of monounsaturated fatty acids. Cell Death Dis. 2023, 14, 48. [Google Scholar] [CrossRef]
- Ouyang, S.; Li, H.; Lou, L.; Huang, Q.; Zhang, Z.; Mo, J.; Li, M.; Lu, J.; Zhu, K.; Chu, Y.; et al. Inhibition of STAT3-ferroptosis negative regulatory axis suppresses tumor growth and alleviates chemoresistance in gastric cancer. Redox Biol. 2022, 52, 102317. [Google Scholar] [CrossRef]
- Wang, L.; Liu, Y.; Du, T.; Yang, H.; Lei, L.; Guo, M.; Ding, H.F.; Zhang, J.; Wang, H.; Chen, X.; et al. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 2020, 27, 662–675. [Google Scholar] [CrossRef]
- He, F.; Zhang, P.; Liu, J.; Wang, R.; Kaufman, R.J.; Yaden, B.C.; Karin, M. ATF4 suppresses hepatocarcinogenesis by inducing SLC7A11 (xCT) to block stress-related ferroptosis. J. Hepatol. 2023, 79, 362–377. [Google Scholar] [CrossRef] [PubMed]
- Li, J.Y.; Ren, C.; Wang, L.X.; Yao, R.Q.; Dong, N.; Wu, Y.; Tian, Y.P.; Yao, Y.M. Sestrin2 protects dendrite cells against ferroptosis induced by sepsis. Cell Death Dis. 2021, 12, 834. [Google Scholar] [CrossRef]
- Gao, R.; Kalathur, R.K.R.; Coto-Llerena, M.; Ercan, C.; Buechel, D.; Shuang, S.; Piscuoglio, S.; Dill, M.T.; Camargo, F.D.; Christofori, G.; et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol. Med. 2021, 13, e14351. [Google Scholar] [CrossRef]
- Tsvetkov, P.; Coy, S.; Petrova, B.; Dreishpoon, M.; Verma, A.; Abdusamad, M.; Rossen, J.; Joesch-Cohen, L.; Humeidi, R.; Spangler, R.D.; et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 2022, 375, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
- Xie, J.; Yang, Y.; Gao, Y.; He, J. Cuproptosis: Mechanisms and links with cancers. Mol. Cancer 2023, 22, 46. [Google Scholar] [CrossRef]
- Chen, X.; Cai, Q.; Liang, R.; Zhang, D.; Liu, X.; Zhang, M.; Xiong, Y.; Xu, M.; Liu, Q.; Li, P.; et al. Copper homeostasis and copper-induced cell death in the pathogenesis of cardiovascular disease and therapeutic strategies. Cell Death Dis. 2023, 14, 105. [Google Scholar] [CrossRef]
- Lutsenko, S. Copper trafficking to the secretory pathway. Metallomics 2016, 8, 840–852. [Google Scholar] [CrossRef] [PubMed]
- Tang, D.; Chen, X.; Kroemer, G. Cuproptosis: A copper-triggered modality of mitochondrial cell death. Cell Res. 2022, 32, 417–418. [Google Scholar] [CrossRef] [PubMed]
- Lutsenko, S.; Bhattacharjee, A.; Hubbard, A.L. Copper handling machinery of the brain. Metallomics 2010, 2, 596–608. [Google Scholar] [CrossRef]
- Tsvetkov, P.; Detappe, A.; Cai, K.; Keys, H.R.; Brune, Z.; Ying, W.; Thiru, P.; Reidy, M.; Kugener, G.; Rossen, J.; et al. Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat. Chem. Biol. 2019, 15, 681–689. [Google Scholar] [CrossRef]
- Zhu, X.; Boulet, A.; Buckley, K.M.; Phillips, C.B.; Gammon, M.G.; Oldfather, L.E.; Moore, S.A.; Leary, S.C.; Cobine, P.A. Mitochondrial copper and phosphate transporter specificity was defined early in the evolution of eukaryotes. eLife 2021, 10, e64690. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.L.; Yu, C.X.; Song, L.J. Programmed cell death in hepatic fibrosis: Current and perspectives. Cell Death Discov. 2023, 9, 449. [Google Scholar] [CrossRef] [PubMed]
- Hunter, D.R.; Haworth, R.A.; Southard, J.H. Relationship between configuration, function, and permeability in calcium-treated mitochondria. J. Biol. Chem. 1976, 251, 5069–5077. [Google Scholar] [CrossRef] [PubMed]
- Izzo, V.; Bravo-San Pedro, J.M.; Sica, V.; Kroemer, G.; Galluzzi, L. Mitochondrial Permeability Transition: New Findings and Persisting Uncertainties. Trends Cell Biol. 2016, 26, 655–667. [Google Scholar] [CrossRef]
- Vanden Berghe, T.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 135–147. [Google Scholar] [CrossRef]
- Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.H.; Lee, M.S. Autophagy--a key player in cellular and body metabolism. Nat. Rev. Endocrinol. 2014, 10, 322–337. [Google Scholar] [CrossRef]
- Denton, D.; Kumar, S. Autophagy-dependent cell death. Cell Death Differ. 2019, 26, 605–616. [Google Scholar] [CrossRef]
- Denton, D.; Shravage, B.; Simin, R.; Mills, K.; Berry, D.L.; Baehrecke, E.H.; Kumar, S. Autophagy, not apoptosis, is essential for midgut cell death in Drosophila. Curr. Biol. 2009, 19, 1741–1746. [Google Scholar] [CrossRef]
- Liu, S.; Yao, S.; Yang, H.; Liu, S.; Wang, Y. Autophagy: Regulator of cell death. Cell Death Dis. 2023, 14, 648. [Google Scholar] [CrossRef]
- Wang, M.; Kaufman, R.J. Protein misfolding in the endoplasmic reticulum as a conduit to human disease. Nature 2016, 529, 326–335. [Google Scholar] [CrossRef]
- Gubas, A.; Dikic, I. ER remodeling via ER-phagy. Mol. Cell 2022, 82, 1492–1500. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Duan, B.; Zhang, Y.; Zhang, X.; Xia, B. Excessive ER-phagy mediated by the autophagy receptor FAM134B results in ER stress, the unfolded protein response, and cell death in HeLa cells. J. Biol. Chem. 2019, 294, 20009–20023. [Google Scholar] [CrossRef] [PubMed]
- Meyer, N.; Henkel, L.; Linder, B.; Zielke, S.; Tascher, G.; Trautmann, S.; Geisslinger, G.; Munch, C.; Fulda, S.; Tegeder, I.; et al. Autophagy activation, lipotoxicity and lysosomal membrane permeabilization synergize to promote pimozide- and loperamide-induced glioma cell death. Autophagy 2021, 17, 3424–3443. [Google Scholar] [CrossRef]
- Onishi, M.; Yamano, K.; Sato, M.; Matsuda, N.; Okamoto, K. Molecular mechanisms and physiological functions of mitophagy. EMBO J. 2021, 40, e104705. [Google Scholar] [CrossRef] [PubMed]
- Ajoolabady, A.; Chiong, M.; Lavandero, S.; Klionsky, D.J.; Ren, J. Mitophagy in cardiovascular diseases: Molecular mechanisms, pathogenesis, and treatment. Trends Mol. Med. 2022, 28, 836–849. [Google Scholar] [CrossRef]
- Reef, S.; Zalckvar, E.; Shifman, O.; Bialik, S.; Sabanay, H.; Oren, M.; Kimchi, A. A short mitochondrial form of p19ARF induces autophagy and caspase-independent cell death. Mol. Cell 2006, 22, 463–475. [Google Scholar] [CrossRef]
- Shoji-Kawata, S.; Sumpter, R.; Leveno, M.; Campbell, G.R.; Zou, Z.; Kinch, L.; Wilkins, A.D.; Sun, Q.; Pallauf, K.; MacDuff, D.; et al. Identification of a candidate therapeutic autophagy-inducing peptide. Nature 2013, 494, 201–206. [Google Scholar] [CrossRef]
- Liu, Y.; Shoji-Kawata, S.; Sumpter, R.M., Jr.; Wei, Y.; Ginet, V.; Zhang, L.; Posner, B.; Tran, K.A.; Green, D.R.; Xavier, R.J.; et al. Autosis is a Na+,K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc. Natl. Acad. Sci. USA 2013, 110, 20364–20371. [Google Scholar] [CrossRef]
- Nah, J.; Zablocki, D.; Sadoshima, J. Autosis: A New Target to Prevent Cell Death. JACC Basic. Transl. Sci. 2020, 5, 857–869. [Google Scholar] [CrossRef]
- Xue, Q.; Kang, R.; Klionsky, D.J.; Tang, D.; Liu, J.; Chen, X. Copper metabolism in cell death and autophagy. Autophagy 2023, 19, 2175–2195. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.; Wang, X. Lysosome biogenesis: Regulation and functions. J. Cell Biol. 2021, 220, e202102001. [Google Scholar] [CrossRef] [PubMed]
- Aits, S.; Jaattela, M. Lysosomal cell death at a glance. J. Cell Sci. 2013, 126, 1905–1912. [Google Scholar] [CrossRef]
- Andrabi, S.A.; Dawson, T.M.; Dawson, V.L. Mitochondrial and nuclear cross talk in cell death: Parthanatos. Ann. N. Y. Acad. Sci. 2008, 1147, 233–241. [Google Scholar] [CrossRef]
- Robinson, N.; Ganesan, R.; Hegedus, C.; Kovacs, K.; Kufer, T.A.; Virag, L. Programmed necrotic cell death of macrophages: Focus on pyroptosis, necroptosis, and parthanatos. Redox Biol. 2019, 26, 101239. [Google Scholar] [CrossRef]
- Chen, F.; Zhu, S.; Kang, R.; Tang, D.; Liu, J. ATP6V0D1 promotes alkaliptosis by blocking STAT3-mediated lysosomal pH homeostasis. Cell Rep. 2023, 42, 111911. [Google Scholar] [CrossRef] [PubMed]
- Song, X.; Zhu, S.; Xie, Y.; Liu, J.; Sun, L.; Zeng, D.; Wang, P.; Ma, X.; Kroemer, G.; Bartlett, D.L.; et al. JTC801 Induces pH-dependent Death Specifically in Cancer Cells and Slows Growth of Tumors in Mice. Gastroenterology 2018, 154, 1480–1493. [Google Scholar] [CrossRef]
- Holze, C.; Michaudel, C.; Mackowiak, C.; Haas, D.A.; Benda, C.; Hubel, P.; Pennemann, F.L.; Schnepf, D.; Wettmarshausen, J.; Braun, M.; et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat. Immunol. 2018, 19, 130–140. [Google Scholar] [CrossRef]
- Liu, X.; Nie, L.; Zhang, Y.; Yan, Y.; Wang, C.; Colic, M.; Olszewski, K.; Horbath, A.; Chen, X.; Lei, G.; et al. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat. Cell Biol. 2023, 25, 404–414. [Google Scholar] [CrossRef]
- Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
- Karmakar, M.; Minns, M.; Greenberg, E.N.; Diaz-Aponte, J.; Pestonjamasp, K.; Johnson, J.L.; Rathkey, J.K.; Abbott, D.W.; Wang, K.; Shao, F.; et al. N-GSDMD trafficking to neutrophil organelles facilitates IL-1beta release independently of plasma membrane pores and pyroptosis. Nat. Commun. 2020, 11, 2212. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.T.; Wei, S.; Nguyen, T.H.; Jo, Y.; Zhang, Y.; Park, W.; Gariani, K.; Oh, C.M.; Kim, H.H.; Ha, K.T.; et al. Mitochondria-associated programmed cell death as a therapeutic target for age-related disease. Exp. Mol. Med. 2023, 55, 1595–1619. [Google Scholar] [CrossRef] [PubMed]
- Remijsen, Q.; Vanden Berghe, T.; Wirawan, E.; Asselbergh, B.; Parthoens, E.; De Rycke, R.; Noppen, S.; Delforge, M.; Willems, J.; Vandenabeele, P. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2011, 21, 290–304. [Google Scholar] [CrossRef]
- Yipp, B.G.; Petri, B.; Salina, D.; Jenne, C.N.; Scott, B.N.; Zbytnuik, L.D.; Pittman, K.; Asaduzzaman, M.; Wu, K.; Meijndert, H.C.; et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat. Med. 2012, 18, 1386–1393. [Google Scholar] [CrossRef] [PubMed]
- Vorobjeva, N.; Dagil, Y.; Pashenkov, M.; Pinegin, B.; Chernyak, B. Protein kinase C isoforms mediate the formation of neutrophil extracellular traps. Int. Immunopharmacol. 2023, 114, 109448. [Google Scholar] [CrossRef]
- Overholtzer, M.; Mailleux, A.A.; Mouneimne, G.; Normand, G.; Schnitt, S.J.; King, R.W.; Cibas, E.S.; Brugge, J.S. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 2007, 131, 966–979. [Google Scholar] [CrossRef]
- Hamann, J.C.; Surcel, A.; Chen, R.; Teragawa, C.; Albeck, J.G.; Robinson, D.N.; Overholtzer, M. Entosis Is Induced by Glucose Starvation. Cell Rep. 2017, 20, 201–210. [Google Scholar] [CrossRef] [PubMed]
- Durgan, J.; Tseng, Y.Y.; Hamann, J.C.; Domart, M.C.; Collinson, L.; Hall, A.; Overholtzer, M.; Florey, O. Mitosis can drive cell cannibalism through entosis. eLife 2017, 6, e27134. [Google Scholar] [CrossRef]
- Durgan, J.; Florey, O. Cancer cell cannibalism: Multiple triggers emerge for entosis. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 831–841. [Google Scholar] [CrossRef]
- Martins, I.; Raza, S.Q.; Voisin, L.; Dakhli, H.; Law, F.; De Jong, D.; Allouch, A.; Thoreau, M.; Brenner, C.; Deutsch, E.; et al. Entosis: The emerging face of non-cell-autonomous type IV programmed death. Biomed. J. 2017, 40, 133–140. [Google Scholar] [CrossRef]
- Casares, N.; Pequignot, M.O.; Tesniere, A.; Ghiringhelli, F.; Roux, S.; Chaput, N.; Schmitt, E.; Hamai, A.; Hervas-Stubbs, S.; Obeid, M.; et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 2005, 202, 1691–1701. [Google Scholar] [CrossRef] [PubMed]
- Green, D.R.; Ferguson, T.; Zitvogel, L.; Kroemer, G. Immunogenic and tolerogenic cell death. Nat. Rev. Immunol. 2009, 9, 353–363. [Google Scholar] [CrossRef]
- Bedoui, S.; Herold, M.J.; Strasser, A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat. Rev. Mol. Cell Biol. 2020, 21, 678–695. [Google Scholar] [CrossRef] [PubMed]
- Ding, J.; Wang, K.; Liu, W.; She, Y.; Sun, Q.; Shi, J.; Sun, H.; Wang, D.C.; Shao, F. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 2016, 535, 111–116. [Google Scholar] [CrossRef]
- Mishima, E.; Conrad, M. Nutritional and Metabolic Control of Ferroptosis. Annu. Rev. Nutr. 2022, 42, 275–309. [Google Scholar] [CrossRef]
- de Bont, C.M.; Boelens, W.C.; Pruijn, G.J.M. NETosis, complement, and coagulation: A triangular relationship. Cell Mol. Immunol. 2019, 16, 19–27. [Google Scholar] [CrossRef]
- Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chetelat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef] [PubMed]
- Graff-Radford, J.; Yong, K.X.X.; Apostolova, L.G.; Bouwman, F.H.; Carrillo, M.; Dickerson, B.C.; Rabinovici, G.D.; Schott, J.M.; Jones, D.T.; Murray, M.E. New insights into atypical Alzheimer’s disease in the era of biomarkers. Lancet Neurol. 2021, 20, 222–234. [Google Scholar] [CrossRef]
- Jack, C.R., Jr.; Knopman, D.S.; Jagust, W.J.; Shaw, L.M.; Aisen, P.S.; Weiner, M.W.; Petersen, R.C.; Trojanowski, J.Q. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9, 119–128. [Google Scholar] [CrossRef]
- Jucker, M.; Walker, L.C. Alzheimer’s disease: From immunotherapy to immunoprevention. Cell 2023, 186, 4260–4270. [Google Scholar] [CrossRef]
- Wesseling, H.; Mair, W.; Kumar, M.; Schlaffner, C.N.; Tang, S.; Beerepoot, P.; Fatou, B.; Guise, A.J.; Cheng, L.; Takeda, S.; et al. Tau PTM Profiles Identify Patient Heterogeneity and Stages of Alzheimer’s Disease. Cell 2020, 183, 1699–1713.e1613. [Google Scholar] [CrossRef] [PubMed]
- Stefanoska, K.; Gajwani, M.; Tan, A.R.P.; Ahel, H.I.; Asih, P.R.; Volkerling, A.; Poljak, A.; Ittner, A. Alzheimer’s disease: Ablating single master site abolishes tau hyperphosphorylation. Sci. Adv. 2022, 8, eabl8809. [Google Scholar] [CrossRef] [PubMed]
- Karran, E.; De Strooper, B. The amyloid hypothesis in Alzheimer disease: New insights from new therapeutics. Nat. Rev. Drug Discov. 2022, 21, 306–318. [Google Scholar] [CrossRef]
- Kumari, S.; Dhapola, R.; Reddy, D.H. Apoptosis in Alzheimer’s disease: Insight into the signaling pathways and therapeutic avenues. Apoptosis 2023, 28, 943–957. [Google Scholar] [CrossRef]
- Zhang, Y.; McLaughlin, R.; Goodyer, C.; LeBlanc, A. Selective cytotoxicity of intracellular amyloid beta peptide1-42 through p53 and Bax in cultured primary human neurons. J. Cell Biol. 2002, 156, 519–529. [Google Scholar] [CrossRef]
- Della Torre, L.; Beato, A.; Capone, V.; Carannante, D.; Verrilli, G.; Favale, G.; Del Gaudio, N.; Megchelenbrink, W.L.; Benedetti, R.; Altucci, L.; et al. Involvement of regulated cell deaths in aging and age-related pathologies. Ageing Res. Rev. 2024, 95, 102251. [Google Scholar] [CrossRef] [PubMed]
- Ajoolabady, A.; Lindholm, D.; Ren, J.; Pratico, D. ER stress and UPR in Alzheimer’s disease: Mechanisms, pathogenesis, treatments. Cell Death Dis. 2022, 13, 706. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; McQuade, T.; Siemer, A.B.; Napetschnig, J.; Moriwaki, K.; Hsiao, Y.S.; Damko, E.; Moquin, D.; Walz, T.; McDermott, A.; et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 2012, 150, 339–350. [Google Scholar] [CrossRef]
- Salvadores, N.; Moreno-Gonzalez, I.; Gamez, N.; Quiroz, G.; Vegas-Gomez, L.; Escandon, M.; Jimenez, S.; Vitorica, J.; Gutierrez, A.; Soto, C.; et al. Abeta oligomers trigger necroptosis-mediated neurodegeneration via microglia activation in Alzheimer’s disease. Acta Neuropathol. Commun. 2022, 10, 31. [Google Scholar] [CrossRef]
- Dong, Y.; Yu, H.; Li, X.; Bian, K.; Zheng, Y.; Dai, M.; Feng, X.; Sun, Y.; He, Y.; Yu, B.; et al. Hyperphosphorylated tau mediates neuronal death by inducing necroptosis and inflammation in Alzheimer’s disease. J. Neuroinflammation 2022, 19, 205. [Google Scholar] [CrossRef]
- Halle, A.; Hornung, V.; Petzold, G.C.; Stewart, C.R.; Monks, B.G.; Reinheckel, T.; Fitzgerald, K.A.; Latz, E.; Moore, K.J.; Golenbock, D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat. Immunol. 2008, 9, 857–865. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Xu, P.; Shan, J.; Sun, W.; Ji, X.; Chi, T.; Liu, P.; Zou, L. Interaction between hyperphosphorylated tau and pyroptosis in forskolin and streptozotocin induced AD models. Biomed. Pharmacother. 2020, 121, 109618. [Google Scholar] [CrossRef]
- Tan, M.S.; Tan, L.; Jiang, T.; Zhu, X.C.; Wang, H.F.; Jia, C.D.; Yu, J.T. Amyloid-beta induces NLRP1-dependent neuronal pyroptosis in models of Alzheimer’s disease. Cell Death Dis. 2014, 5, e1382. [Google Scholar] [CrossRef]
- Wang, Q.; Sun, J.; Chen, T.; Song, S.; Hou, Y.; Feng, L.; Fan, C.; Li, M. Ferroptosis, Pyroptosis, and Cuproptosis in Alzheimer’s Disease. ACS Chem. Neurosci. 2023, 14, 3564–3587. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Gao, X.; Bai, X.; Yao, S.; Chang, Y.Z.; Gao, G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl. Neurodegener. 2022, 11, 39. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, A.; Shin, R.W.; Hasegawa, K.; Naiki, H.; Sato, H.; Yoshimasu, F.; Kitamoto, T. Iron (III) induces aggregation of hyperphosphorylated tau and its reduction to iron (II) reverses the aggregation: Implications in the formation of neurofibrillary tangles of Alzheimer’s disease. J. Neurochem. 2002, 82, 1137–1147. [Google Scholar] [CrossRef]
- Ward, R.J.; Zucca, F.A.; Duyn, J.H.; Crichton, R.R.; Zecca, L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014, 13, 1045–1060. [Google Scholar] [CrossRef]
- Smith, D.P.; Ciccotosto, G.D.; Tew, D.J.; Fodero-Tavoletti, M.T.; Johanssen, T.; Masters, C.L.; Barnham, K.J.; Cappai, R. Concentration dependent Cu2+ induced aggregation and dityrosine formation of the Alzheimer’s disease amyloid-beta peptide. Biochemistry 2007, 46, 2881–2891. [Google Scholar] [CrossRef]
- Kitazawa, M.; Hsu, H.W.; Medeiros, R. Copper Exposure Perturbs Brain Inflammatory Responses and Impairs Clearance of Amyloid-Beta. Toxicol. Sci. 2016, 152, 194–204. [Google Scholar] [CrossRef]
- Singh, I.; Sagare, A.P.; Coma, M.; Perlmutter, D.; Gelein, R.; Bell, R.D.; Deane, R.J.; Zhong, E.; Parisi, M.; Ciszewski, J.; et al. Low levels of copper disrupt brain amyloid-beta homeostasis by altering its production and clearance. Proc. Natl. Acad. Sci. USA 2013, 110, 14771–14776. [Google Scholar] [CrossRef]
- Du, H.; Guo, L.; Fang, F.; Chen, D.; Sosunov, A.A.; McKhann, G.M.; Yan, Y.; Wang, C.; Zhang, H.; Molkentin, J.D.; et al. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer’s disease. Nat. Med. 2008, 14, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
- Du, H.; Yan, S.S. Mitochondrial permeability transition pore in Alzheimer’s disease: Cyclophilin D and amyloid beta. Biochim. Biophys. Acta 2010, 1802, 198–204. [Google Scholar] [CrossRef] [PubMed]
- Boland, B.; Kumar, A.; Lee, S.; Platt, F.M.; Wegiel, J.; Yu, W.H.; Nixon, R.A. Autophagy induction and autophagosome clearance in neurons: Relationship to autophagic pathology in Alzheimer’s disease. J. Neurosci. 2008, 28, 6926–6937. [Google Scholar] [CrossRef] [PubMed]
- Bordi, M.; Berg, M.J.; Mohan, P.S.; Peterhoff, C.M.; Alldred, M.J.; Che, S.; Ginsberg, S.D.; Nixon, R.A. Autophagy flux in CA1 neurons of Alzheimer hippocampus: Increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy 2016, 12, 2467–2483. [Google Scholar] [CrossRef]
- Strosznajder, J.B.; Jesko, H.; Strosznajder, R.P. Effect of amyloid beta peptide on poly(ADP-ribose) polymerase activity in adult and aged rat hippocampus. Acta Biochim. Pol. 2000, 47, 847–854. [Google Scholar] [CrossRef]
- Kauppinen, T.M.; Suh, S.W.; Higashi, Y.; Berman, A.E.; Escartin, C.; Won, S.J.; Wang, C.; Cho, S.H.; Gan, L.; Swanson, R.A. Poly(ADP-ribose)polymerase-1 modulates microglial responses to amyloid beta. J. Neuroinflammation 2011, 8, 152. [Google Scholar] [CrossRef]
- Pietronigro, E.C.; Della Bianca, V.; Zenaro, E.; Constantin, G. NETosis in Alzheimer’s Disease. Front. Immunol. 2017, 8, 211. [Google Scholar] [CrossRef]
- Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
- Tolosa, E.; Garrido, A.; Scholz, S.W.; Poewe, W. Challenges in the diagnosis of Parkinson’s disease. Lancet Neurol. 2021, 20, 385–397. [Google Scholar] [CrossRef]
- Burre, J.; Sharma, M.; Sudhof, T.C. Cell Biology and Pathophysiology of alpha-Synuclein. Cold Spring Harb. Perspect. Med. 2018, 8, a024091. [Google Scholar] [CrossRef]
- Morris, H.R.; Spillantini, M.G.; Sue, C.M.; Williams-Gray, C.H. The pathogenesis of Parkinson’s disease. Lancet 2024, 403, 293–304. [Google Scholar] [CrossRef]
- Shahmoradian, S.H.; Lewis, A.J.; Genoud, C.; Hench, J.; Moors, T.E.; Navarro, P.P.; Castano-Diez, D.; Schweighauser, G.; Graff-Meyer, A.; Goldie, K.N.; et al. Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes. Nat. Neurosci. 2019, 22, 1099–1109. [Google Scholar] [CrossRef] [PubMed]
- Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, A.; Michel, P.P.; Troadec, J.D.; Mouatt-Prigent, A.; Faucheux, B.A.; Ruberg, M.; Agid, Y.; Hirsch, E.C. Is Bax a mitochondrial mediator in apoptotic death of dopaminergic neurons in Parkinson’s disease? J. Neurochem. 2001, 76, 1785–1793. [Google Scholar] [CrossRef]
- Tatton, N.A. Increased caspase 3 and Bax immunoreactivity accompany nuclear GAPDH translocation and neuronal apoptosis in Parkinson’s disease. Exp. Neurol. 2000, 166, 29–43. [Google Scholar] [CrossRef] [PubMed]
- Blandini, F.; Mangiagalli, A.; Cosentino, M.; Marino, F.; Samuele, A.; Rasini, E.; Fancellu, R.; Martignoni, E.; Riboldazzi, G.; Calandrella, D.; et al. Peripheral markers of apoptosis in Parkinson’s disease: The effect of dopaminergic drugs. Ann. N. Y. Acad. Sci. 2003, 1010, 675–678. [Google Scholar] [CrossRef]
- Mogi, M.; Kondo, T.; Mizuno, Y.; Nagatsu, T. p53 protein, interferon-gamma, and NF-kappaB levels are elevated in the parkinsonian brain. Neurosci. Lett. 2007, 414, 94–97. [Google Scholar] [CrossRef]
- Tanaka, Y.; Engelender, S.; Igarashi, S.; Rao, R.K.; Wanner, T.; Tanzi, R.E.; Sawa, A.; Dawson, V.L.; Dawson, T.M.; Ross, C.A. Inducible expression of mutant alpha-synuclein decreases proteasome activity and increases sensitivity to mitochondria-dependent apoptosis. Hum. Mol. Genet. 2001, 10, 919–926. [Google Scholar] [CrossRef]
- Gordon, R.; Albornoz, E.A.; Christie, D.C.; Langley, M.R.; Kumar, V.; Mantovani, S.; Robertson, A.A.B.; Butler, M.S.; Rowe, D.B.; O’Neill, L.A.; et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci. Transl. Med. 2018, 10, aah4066. [Google Scholar] [CrossRef]
- Weindel, C.G.; Martinez, E.L.; Zhao, X.; Mabry, C.J.; Bell, S.L.; Vail, K.J.; Coleman, A.K.; VanPortfliet, J.J.; Zhao, B.; Wagner, A.R.; et al. Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell 2022, 185, 3214–3231.e3223. [Google Scholar] [CrossRef]
- Iannielli, A.; Bido, S.; Folladori, L.; Segnali, A.; Cancellieri, C.; Maresca, A.; Massimino, L.; Rubio, A.; Morabito, G.; Caporali, L.; et al. Pharmacological Inhibition of Necroptosis Protects from Dopaminergic Neuronal Cell Death in Parkinson’s Disease Models. Cell Rep. 2018, 22, 2066–2079. [Google Scholar] [CrossRef] [PubMed]
- Amin, P.; Florez, M.; Najafov, A.; Pan, H.; Geng, J.; Ofengeim, D.; Dziedzic, S.A.; Wang, H.; Barrett, V.J.; Ito, Y.; et al. Regulation of a distinct activated RIPK1 intermediate bridging complex I and complex II in TNFalpha-mediated apoptosis. Proc. Natl. Acad. Sci. USA 2018, 115, E5944–E5953. [Google Scholar] [CrossRef] [PubMed]
- Castellani, R.J.; Siedlak, S.L.; Perry, G.; Smith, M.A. Sequestration of iron by Lewy bodies in Parkinson’s disease. Acta Neuropathol. 2000, 100, 111–114. [Google Scholar] [CrossRef]
- Golts, N.; Snyder, H.; Frasier, M.; Theisler, C.; Choi, P.; Wolozin, B. Magnesium inhibits spontaneous and iron-induced aggregation of alpha-synuclein. J. Biol. Chem. 2002, 277, 16116–16123. [Google Scholar] [CrossRef]
- Dong-Chen, X.; Yong, C.; Yang, X.; Chen-Yu, S.; Li-Hua, P. Signaling pathways in Parkinson’s disease: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2023, 8, 73. [Google Scholar] [CrossRef]
- Dudzik, C.G.; Walter, E.D.; Millhauser, G.L. Coordination features and affinity of the Cu(2)+ site in the alpha-synuclein protein of Parkinson’s disease. Biochemistry 2011, 50, 1771–1777. [Google Scholar] [CrossRef] [PubMed]
- Martin, L.J.; Semenkow, S.; Hanaford, A.; Wong, M. Mitochondrial permeability transition pore regulates Parkinson’s disease development in mutant alpha-synuclein transgenic mice. Neurobiol. Aging 2014, 35, 1132–1152. [Google Scholar] [CrossRef]
- Hardy, J.; Lewis, P.; Revesz, T.; Lees, A.; Paisan-Ruiz, C. The genetics of Parkinson’s syndromes: A critical review. Curr. Opin. Genet. Dev. 2009, 19, 254–265. [Google Scholar] [CrossRef]
- Kim, S.; Wong, Y.C.; Gao, F.; Krainc, D. Dysregulation of mitochondria-lysosome contacts by GBA1 dysfunction in dopaminergic neuronal models of Parkinson’s disease. Nat. Commun. 2021, 12, 1807. [Google Scholar] [CrossRef]
- Kam, T.I.; Mao, X.; Park, H.; Chou, S.C.; Karuppagounder, S.S.; Umanah, G.E.; Yun, S.P.; Brahmachari, S.; Panicker, N.; Chen, R.; et al. Poly(ADP-ribose) drives pathologic alpha-synuclein neurodegeneration in Parkinson’s disease. Science 2018, 362, aat8407. [Google Scholar] [CrossRef]
- Jimenez-Sanchez, M.; Licitra, F.; Underwood, B.R.; Rubinsztein, D.C. Huntington’s Disease: Mechanisms of Pathogenesis and Therapeutic Strategies. Cold Spring Harb. Perspect. Med. 2017, 7, a024240. [Google Scholar] [CrossRef] [PubMed]
- Tabrizi, S.J.; Estevez-Fraga, C.; van Roon-Mom, W.M.C.; Flower, M.D.; Scahill, R.I.; Wild, E.J.; Munoz-Sanjuan, I.; Sampaio, C.; Rosser, A.E.; Leavitt, B.R. Potential disease-modifying therapies for Huntington’s disease: Lessons learned and future opportunities. Lancet Neurol. 2022, 21, 645–658. [Google Scholar] [CrossRef] [PubMed]
- Rub, U.; Vonsattel, J.P.; Heinsen, H.; Korf, H.W. The Neuropathology of Huntington s disease: Classical findings, recent developments and correlation to functional neuroanatomy. Adv. Anat. Embryol. Cell Biol. 2015, 217, 1–146. [Google Scholar]
- Zhang, Y.; Ona, V.O.; Li, M.; Drozda, M.; Dubois-Dauphin, M.; Przedborski, S.; Ferrante, R.J.; Friedlander, R.M. Sequential activation of individual caspases, and of alterations in Bcl-2 proapoptotic signals in a mouse model of Huntington’s disease. J. Neurochem. 2003, 87, 1184–1192. [Google Scholar] [CrossRef]
- Leon, R.; Bhagavatula, N.; Ulukpo, O.; McCollum, M.; Wei, J. BimEL as a possible molecular link between proteasome dysfunction and cell death induced by mutant huntingtin. Eur. J. Neurosci. 2010, 31, 1915–1925. [Google Scholar] [CrossRef]
- Vis, J.C.; Schipper, E.; de Boer-van Huizen, R.T.; Verbeek, M.M.; de Waal, R.M.; Wesseling, P.; ten Donkelaar, H.J.; Kremer, B. Expression pattern of apoptosis-related markers in Huntington’s disease. Acta Neuropathol. 2005, 109, 321–328. [Google Scholar] [CrossRef] [PubMed]
- Erekat, N.S. Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin. Anat. 2022, 35, 65–78. [Google Scholar] [CrossRef]
- Zhang, Y.; Leavitt, B.R.; van Raamsdonk, J.M.; Dragatsis, I.; Goldowitz, D.; MacDonald, M.E.; Hayden, M.R.; Friedlander, R.M. Huntingtin inhibits caspase-3 activation. EMBO J. 2006, 25, 5896–5906. [Google Scholar] [CrossRef]
- Bossy-Wetzel, E.; Petrilli, A.; Knott, A.B. Mutant huntingtin and mitochondrial dysfunction. Trends Neurosci. 2008, 31, 609–616. [Google Scholar] [CrossRef]
- Choo, Y.S.; Johnson, G.V.; MacDonald, M.; Detloff, P.J.; Lesort, M. Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum. Mol. Genet. 2004, 13, 1407–1420. [Google Scholar] [CrossRef]
- Sawa, A.; Wiegand, G.W.; Cooper, J.; Margolis, R.L.; Sharp, A.H.; Lawler, J.F., Jr.; Greenamyre, J.T.; Snyder, S.H.; Ross, C.A. Increased apoptosis of Huntington disease lymphoblasts associated with repeat length-dependent mitochondrial depolarization. Nat. Med. 1999, 5, 1194–1198. [Google Scholar] [CrossRef]
- Zhu, S.; Zhang, Y.; Bai, G.; Li, H. Necrostatin-1 ameliorates symptoms in R6/2 transgenic mouse model of Huntington’s disease. Cell Death Dis. 2011, 2, e115. [Google Scholar] [CrossRef] [PubMed]
- Paldino, E.; D’Angelo, V.; Sancesario, G.; Fusco, F.R. Pyroptotic cell death in the R6/2 mouse model of Huntington’s disease: New insight on the inflammasome. Cell Death Discov. 2020, 6, 69. [Google Scholar] [CrossRef]
- Chen, L.; Hambright, W.S.; Na, R.; Ran, Q. Ablation of the Ferroptosis Inhibitor Glutathione Peroxidase 4 in Neurons Results in Rapid Motor Neuron Degeneration and Paralysis. J. Biol. Chem. 2015, 290, 28097–28106. [Google Scholar] [CrossRef]
- Mi, Y.; Gao, X.; Xu, H.; Cui, Y.; Zhang, Y.; Gou, X. The Emerging Roles of Ferroptosis in Huntington’s Disease. Neuromolecular Med. 2019, 21, 110–119. [Google Scholar] [CrossRef]
- Xiao, G.; Fan, Q.; Wang, X.; Zhou, B. Huntington disease arises from a combinatory toxicity of polyglutamine and copper binding. Proc. Natl. Acad. Sci. USA 2013, 110, 14995–15000. [Google Scholar] [CrossRef] [PubMed]
- Wold, M.S.; Lim, J.; Lachance, V.; Deng, Z.; Yue, Z. ULK1-mediated phosphorylation of ATG14 promotes autophagy and is impaired in Huntington’s disease models. Mol. Neurodegener. 2016, 11, 76. [Google Scholar] [CrossRef] [PubMed]
- Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3, 17071. [Google Scholar] [CrossRef]
- Chia, R.; Chio, A.; Traynor, B.J. Novel genes associated with amyotrophic lateral sclerosis: Diagnostic and clinical implications. Lancet Neurol. 2018, 17, 94–102. [Google Scholar] [CrossRef]
- Arseni, D.; Hasegawa, M.; Murzin, A.G.; Kametani, F.; Arai, M.; Yoshida, M.; Ryskeldi-Falcon, B. Structure of pathological TDP-43 filaments from ALS with FTLD. Nature 2022, 601, 139–143. [Google Scholar] [CrossRef]
- Munsat, T.L.; Davies, K.E. International SMA consortium meeting. (26-28 June 1992, Bonn, Germany). Neuromuscul. Disord. 1992, 2, 423–428. [Google Scholar] [CrossRef]
- Wirth, B. Spinal Muscular Atrophy: In the Challenge Lies a Solution. Trends Neurosci. 2021, 44, 306–322. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Lotti, F.; Dittmar, K.; Younis, I.; Wan, L.; Kasim, M.; Dreyfuss, G. SMN deficiency causes tissue-specific perturbations in the repertoire of snRNAs and widespread defects in splicing. Cell 2008, 133, 585–600. [Google Scholar] [CrossRef] [PubMed]
- Lefebvre, S.; Burglen, L.; Reboullet, S.; Clermont, O.; Burlet, P.; Viollet, L.; Benichou, B.; Cruaud, C.; Millasseau, P.; Zeviani, M.; et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995, 80, 155–165. [Google Scholar] [CrossRef] [PubMed]
- Lorson, C.L.; Hahnen, E.; Androphy, E.J.; Wirth, B. A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc. Natl. Acad. Sci. USA 1999, 96, 6307–6311. [Google Scholar] [CrossRef]
- Feldkotter, M.; Schwarzer, V.; Wirth, R.; Wienker, T.F.; Wirth, B. Quantitative analyses of SMN1 and SMN2 based on real-time lightCycler PCR: Fast and highly reliable carrier testing and prediction of severity of spinal muscular atrophy. Am. J. Hum. Genet. 2002, 70, 358–368. [Google Scholar] [CrossRef]
- Koch-Henriksen, N.; Magyari, M. Apparent changes in the epidemiology and severity of multiple sclerosis. Nat. Rev. Neurol. 2021, 17, 676–688. [Google Scholar] [CrossRef] [PubMed]
- Marcus, R. What Is Multiple Sclerosis? JAMA 2022, 328, 2078. [Google Scholar] [CrossRef]
- Correale, J.; Gaitan, M.I.; Ysrraelit, M.C.; Fiol, M.P. Progressive multiple sclerosis: From pathogenic mechanisms to treatment. Brain 2017, 140, 527–546. [Google Scholar] [CrossRef]
- Kuhlmann, T.; Moccia, M.; Coetzee, T.; Cohen, J.A.; Correale, J.; Graves, J.; Marrie, R.A.; Montalban, X.; Yong, V.W.; Thompson, A.J.; et al. Multiple sclerosis progression: Time for a new mechanism-driven framework. Lancet Neurol. 2023, 22, 78–88. [Google Scholar] [CrossRef]
- Pasinelli, P.; Brown, R.H. Molecular biology of amyotrophic lateral sclerosis: Insights from genetics. Nat. Rev. Neurosci. 2006, 7, 710–723. [Google Scholar] [CrossRef] [PubMed]
- Chaytow, H.; Faller, K.M.E.; Huang, Y.T.; Gillingwater, T.H. Spinal muscular atrophy: From approved therapies to future therapeutic targets for personalized medicine. Cell Rep. Med. 2021, 2, 100346. [Google Scholar] [CrossRef]
- Guadagno, J.; Xu, X.; Karajgikar, M.; Brown, A.; Cregan, S.P. Microglia-derived TNFalpha induces apoptosis in neural precursor cells via transcriptional activation of the Bcl-2 family member Puma. Cell Death Dis. 2013, 4, e538. [Google Scholar] [CrossRef] [PubMed]
- Kennedy, P.G.E.; George, W.; Yu, X. The Possible Role of Neural Cell Apoptosis in Multiple Sclerosis. Int. J. Mol. Sci. 2022, 23, 7584. [Google Scholar] [CrossRef] [PubMed]
- Pehar, M.; Vargas, M.R.; Cassina, P.; Barbeito, A.G.; Beckman, J.S.; Barbeito, L. Complexity of astrocyte-motor neuron interactions in amyotrophic lateral sclerosis. Neurodegener. Dis. 2005, 2, 139–146. [Google Scholar] [CrossRef]
- Sreedharan, J.; Blair, I.P.; Tripathi, V.B.; Hu, X.; Vance, C.; Rogelj, B.; Ackerley, S.; Durnall, J.C.; Williams, K.L.; Buratti, E.; et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 2008, 319, 1668–1672. [Google Scholar] [CrossRef]
- Genabai, N.K.; Ahmad, S.; Zhang, Z.; Jiang, X.; Gabaldon, C.A.; Gangwani, L. Genetic inhibition of JNK3 ameliorates spinal muscular atrophy. Hum. Mol. Genet. 2015, 24, 6986–7004. [Google Scholar] [CrossRef]
- Simon, C.M.; Van Alstyne, M.; Lotti, F.; Bianchetti, E.; Tisdale, S.; Watterson, D.M.; Mentis, G.Z.; Pellizzoni, L. Stasimon Contributes to the Loss of Sensory Synapses and Motor Neuron Death in a Mouse Model of Spinal Muscular Atrophy. Cell Rep. 2019, 29, 3885–3901.e3885. [Google Scholar] [CrossRef]
- Ito, Y.; Ofengeim, D.; Najafov, A.; Das, S.; Saberi, S.; Li, Y.; Hitomi, J.; Zhu, H.; Chen, H.; Mayo, L.; et al. RIPK1 mediates axonal degeneration by promoting inflammation and necroptosis in ALS. Science 2016, 353, 603–608. [Google Scholar] [CrossRef]
- Zhang, Y.; Su, S.S.; Zhao, S.; Yang, Z.; Zhong, C.Q.; Chen, X.; Cai, Q.; Yang, Z.H.; Huang, D.; Wu, R.; et al. RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat. Commun. 2017, 8, 14329. [Google Scholar] [CrossRef]
- Ofengeim, D.; Ito, Y.; Najafov, A.; Zhang, Y.; Shan, B.; DeWitt, J.P.; Ye, J.; Zhang, X.; Chang, A.; Vakifahmetoglu-Norberg, H.; et al. Activation of necroptosis in multiple sclerosis. Cell Rep. 2015, 10, 1836–1849. [Google Scholar] [CrossRef] [PubMed]
- Chehade, L.; Deguise, M.O.; De Repentigny, Y.; Yaworski, R.; Beauvais, A.; Gagnon, S.; Hensel, N.; Kothary, R. Suppression of the necroptotic cell death pathways improves survival in Smn (2B/-) mice. Front. Cell Neurosci. 2022, 16, 972029. [Google Scholar] [CrossRef] [PubMed]
- Voet, S.; Srinivasan, S.; Lamkanfi, M.; van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 2019, 11, e10248. [Google Scholar] [CrossRef]
- McKenzie, B.A.; Mamik, M.K.; Saito, L.B.; Boghozian, R.; Monaco, M.C.; Major, E.O.; Lu, J.Q.; Branton, W.G.; Power, C. Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc. Natl. Acad. Sci. USA 2018, 115, E6065–E6074. [Google Scholar] [CrossRef]
- Jeong, S.Y.; Rathore, K.I.; Schulz, K.; Ponka, P.; Arosio, P.; David, S. Dysregulation of iron homeostasis in the CNS contributes to disease progression in a mouse model of amyotrophic lateral sclerosis. J. Neurosci. 2009, 29, 610–619. [Google Scholar] [CrossRef] [PubMed]
- McAlary, L.; Shephard, V.K.; Wright, G.S.A.; Yerbury, J.J. A copper chaperone-mimetic polytherapy for SOD1-associated amyotrophic lateral sclerosis. J. Biol. Chem. 2022, 298, 101612. [Google Scholar] [CrossRef]
- Van San, E.; Debruyne, A.C.; Veeckmans, G.; Tyurina, Y.Y.; Tyurin, V.A.; Zheng, H.; Choi, S.M.; Augustyns, K.; van Loo, G.; Michalke, B.; et al. Ferroptosis contributes to multiple sclerosis and its pharmacological targeting suppresses experimental disease progression. Cell Death Differ. 2023, 30, 2092–2103. [Google Scholar] [CrossRef]
- Colombo, E.; Triolo, D.; Bassani, C.; Bedogni, F.; Di Dario, M.; Dina, G.; Fredrickx, E.; Fermo, I.; Martinelli, V.; Newcombe, J.; et al. Dysregulated copper transport in multiple sclerosis may cause demyelination via astrocytes. Proc. Natl. Acad. Sci. USA 2021, 118, e2025804118. [Google Scholar] [CrossRef]
- Magrane, J.; Hervias, I.; Henning, M.S.; Damiano, M.; Kawamata, H.; Manfredi, G. Mutant SOD1 in neuronal mitochondria causes toxicity and mitochondrial dynamics abnormalities. Hum. Mol. Genet. 2009, 18, 4552–4564. [Google Scholar] [CrossRef]
- Forte, M.; Gold, B.G.; Marracci, G.; Chaudhary, P.; Basso, E.; Johnsen, D.; Yu, X.; Fowlkes, J.; Rahder, M.; Stem, K.; et al. Cyclophilin D inactivation protects axons in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Proc. Natl. Acad. Sci. USA 2007, 104, 7558–7563. [Google Scholar] [CrossRef]
- Mamontova, E.M.; Clement, M.J.; Sukhanova, M.V.; Joshi, V.; Bouhss, A.; Rengifo-Gonzalez, J.C.; Desforges, B.; Hamon, L.; Lavrik, O.I.; Pastre, D. FUS RRM regulates poly(ADP-ribose) levels after transcriptional arrest and PARP-1 activation on DNA damage. Cell Rep. 2023, 42, 113199. [Google Scholar] [CrossRef] [PubMed]
- He, L.; Liang, J.; Chen, C.; Chen, J.; Shen, Y.; Sun, S.; Li, L. C9orf72 functions in the nucleus to regulate DNA damage repair. Cell Death Differ. 2023, 30, 716–730. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Pleasure, D.; Deng, W.; Guo, F. Therapeutic Potentials of Poly (ADP-Ribose) Polymerase 1 (PARP1) Inhibition in Multiple Sclerosis and Animal Models: Concept Revisiting. Adv Sci (Weinh) 2022, 9, e2102853. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, S. Autophagy in spinal cord motor neurons in sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2011, 70, 349–359. [Google Scholar] [CrossRef]
- Rashid, S.; Dimitriadi, M. Autophagy in spinal muscular atrophy: From pathogenic mechanisms to therapeutic approaches. Front. Cell Neurosci. 2023, 17, 1307636. [Google Scholar] [CrossRef]
- Al-Kuraishy, H.M.; Jabir, M.S.; Al-Gareeb, A.I.; Saad, H.M.; Batiha, G.E.; Klionsky, D.J. The beneficial role of autophagy in multiple sclerosis: Yes or No? Autophagy 2024, 20, 259–274. [Google Scholar] [CrossRef]
- Naegele, M.; Tillack, K.; Reinhardt, S.; Schippling, S.; Martin, R.; Sospedra, M. Neutrophils in multiple sclerosis are characterized by a primed phenotype. J. Neuroimmunol. 2012, 242, 60–71. [Google Scholar] [CrossRef]
- Trias, E.; King, P.H.; Si, Y.; Kwon, Y.; Varela, V.; Ibarburu, S.; Kovacs, M.; Moura, I.C.; Beckman, J.S.; Hermine, O.; et al. Mast cells and neutrophils mediate peripheral motor pathway degeneration in ALS. JCI Insight 2018, 3, 123249. [Google Scholar] [CrossRef]
- Murdock, B.J.; Goutman, S.A.; Boss, J.; Kim, S.; Feldman, E.L. Amyotrophic Lateral Sclerosis Survival Associates with Neutrophils in a Sex-specific Manner. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e953. [Google Scholar] [CrossRef]
- Blennow, K.; Hardy, J.; Zetterberg, H. The neuropathology and neurobiology of traumatic brain injury. Neuron 2012, 76, 886–899. [Google Scholar] [CrossRef]
- Capizzi, A.; Woo, J.; Verduzco-Gutierrez, M. Traumatic Brain Injury: An Overview of Epidemiology, Pathophysiology, and Medical Management. Med. Clin. North. Am. 2020, 104, 213–238. [Google Scholar] [CrossRef] [PubMed]
- Ohashi, S.N.; DeLong, J.H.; Kozberg, M.G.; Mazur-Hart, D.J.; van Veluw, S.J.; Alkayed, N.J.; Sansing, L.H. Role of Inflammatory Processes in Hemorrhagic Stroke. Stroke 2023, 54, 605–619. [Google Scholar] [CrossRef] [PubMed]
- Campbell, B.C.V.; De Silva, D.A.; Macleod, M.R.; Coutts, S.B.; Schwamm, L.H.; Davis, S.M.; Donnan, G.A. Ischaemic stroke. Nat. Rev. Dis. Primers 2019, 5, 70. [Google Scholar] [CrossRef] [PubMed]
- Barthels, D.; Das, H. Current advances in ischemic stroke research and therapies. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165260. [Google Scholar] [CrossRef]
- Saini, V.; Guada, L.; Yavagal, D.R. Global Epidemiology of Stroke and Access to Acute Ischemic Stroke Interventions. Neurology 2021, 97, S6–S16. [Google Scholar] [CrossRef]
- Macrez, R.; Ali, C.; Toutirais, O.; Le Mauff, B.; Defer, G.; Dirnagl, U.; Vivien, D. Stroke and the immune system: From pathophysiology to new therapeutic strategies. Lancet Neurol. 2011, 10, 471–480. [Google Scholar] [CrossRef]
- MacManus, J.P.; Buchan, A.M.; Hill, I.E.; Rasquinha, I.; Preston, E. Global ischemia can cause DNA fragmentation indicative of apoptosis in rat brain. Neurosci. Lett. 1993, 164, 89–92. [Google Scholar] [CrossRef]
- Radak, D.; Katsiki, N.; Resanovic, I.; Jovanovic, A.; Sudar-Milovanovic, E.; Zafirovic, S.; Mousad, S.A.; Isenovic, E.R. Apoptosis and Acute Brain Ischemia in Ischemic Stroke. Curr. Vasc. Pharmacol. 2017, 15, 115–122. [Google Scholar] [CrossRef]
- Bonora, M.; Patergnani, S.; Rimessi, A.; De Marchi, E.; Suski, J.M.; Bononi, A.; Giorgi, C.; Marchi, S.; Missiroli, S.; Poletti, F.; et al. ATP synthesis and storage. Purinergic Signal 2012, 8, 343–357. [Google Scholar] [CrossRef]
- Dirnagl, U.; Iadecola, C.; Moskowitz, M.A. Pathobiology of ischaemic stroke: An integrated view. Trends Neurosci. 1999, 22, 391–397. [Google Scholar] [CrossRef]
- Liu, S.P.; Liu, C.; Xu, B.; Zhou, H.; Zhao, H. Disulfidptosis and its Role in Peripheral Blood Immune Cells after a Stroke: A New Frontier in Stroke Pathogenesis. Curr. Neurovasc Res. 2024, 20, 608–622. [Google Scholar] [CrossRef] [PubMed]
- Qin, R.; Huang, L.; Xu, W.; Qin, Q.; Liang, X.; Lai, X.; Huang, X.; Xie, M.; Chen, L. Identification of disulfidptosis-related genes and analysis of immune infiltration characteristics in ischemic strokes. Math. Biosci. Eng. 2023, 20, 18939–18959. [Google Scholar] [CrossRef] [PubMed]
- Tuo, Q.Z.; Zhang, S.T.; Lei, P. Mechanisms of neuronal cell death in ischemic stroke and their therapeutic implications. Med. Res. Rev. 2022, 42, 259–305. [Google Scholar] [CrossRef]
- Wu, M.H.; Huang, C.C.; Chio, C.C.; Tsai, K.J.; Chang, C.P.; Lin, N.K.; Lin, M.T. Inhibition of Peripheral TNF-alpha and Downregulation of Microglial Activation by Alpha-Lipoic Acid and Etanercept Protect Rat Brain Against Ischemic Stroke. Mol. Neurobiol. 2016, 53, 4961–4971. [Google Scholar] [CrossRef]
- Cantarella, G.; Pignataro, G.; Di Benedetto, G.; Anzilotti, S.; Vinciguerra, A.; Cuomo, O.; Di Renzo, G.F.; Parenti, C.; Annunziato, L.; Bernardini, R. Ischemic tolerance modulates TRAIL expression and its receptors and generates a neuroprotected phenotype. Cell Death Dis. 2014, 5, e1331. [Google Scholar] [CrossRef] [PubMed]
- Mira, R.G.; Lira, M.; Cerpa, W. Traumatic Brain Injury: Mechanisms of Glial Response. Front. Physiol. 2021, 12, 740939. [Google Scholar] [CrossRef]
- Liu, M.; Xu, Z.; Wang, L.; Zhang, L.; Liu, Y.; Cao, J.; Fu, Q.; Liu, Y.; Li, H.; Lou, J.; et al. Cottonseed oil alleviates ischemic stroke injury by inhibiting the inflammatory activation of microglia and astrocyte. J. Neuroinflammation 2020, 17, 270. [Google Scholar] [CrossRef]
- Hu, X.; Chen, H.; Xu, H.; Wu, Y.; Wu, C.; Jia, C.; Li, Y.; Sheng, S.; Xu, C.; Xu, H.; et al. Role of Pyroptosis in Traumatic Brain and Spinal Cord Injuries. Int. J. Biol. Sci. 2020, 16, 2042–2050. [Google Scholar] [CrossRef]
- Li, Q.S.; Jia, Y.J. Ferroptosis: A critical player and potential therapeutic target in traumatic brain injury and spinal cord injury. Neural Regen. Res. 2023, 18, 506–512. [Google Scholar] [CrossRef]
- Isaev, N.K.; Stelmashook, E.V.; Genrikhs, E.E. Role of zinc and copper ions in the pathogenetic mechanisms of traumatic brain injury and Alzheimer’s disease. Rev. Neurosci. 2020, 31, 233–243. [Google Scholar] [CrossRef]
- Fan, X.; Chen, H.; Jiang, F.; Xu, C.; Wang, Y.; Wang, H.; Li, M.; Wei, W.; Song, J.; Zhong, D.; et al. Comprehensive analysis of cuproptosis-related genes in immune infiltration in ischemic stroke. Front. Neurol. 2022, 13, 1077178. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Xu, G.; Fang, J. Association Between Serum Copper and Stroke Risk Factors in Adults: Evidence from the National Health and Nutrition Examination Survey, 2011-2016. Biol. Trace. Elem. Res. 2022, 200, 1089–1094. [Google Scholar] [CrossRef] [PubMed]
- Alano, C.C.; Garnier, P.; Ying, W.; Higashi, Y.; Kauppinen, T.M.; Swanson, R.A. NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J. Neurosci. 2010, 30, 2967–2978. [Google Scholar] [CrossRef] [PubMed]
- Nie, Z.; Tan, L.; Niu, J.; Wang, B. The role of regulatory necrosis in traumatic brain injury. Front. Mol. Neurosci. 2022, 15, 1005422. [Google Scholar] [CrossRef] [PubMed]
- Schinzel, A.C.; Takeuchi, O.; Huang, Z.; Fisher, J.K.; Zhou, Z.; Rubens, J.; Hetz, C.; Danial, N.N.; Moskowitz, M.A.; Korsmeyer, S.J. Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. Proc. Natl. Acad. Sci. USA 2005, 102, 12005–12010. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.Y.; Shim, M.S.; Kim, K.Y.; Weinreb, R.N.; Wheeler, L.A.; Ju, W.K. Inhibition of cyclophilin D by cyclosporin A promotes retinal ganglion cell survival by preventing mitochondrial alteration in ischemic injury. Cell Death Dis. 2014, 5, e1105. [Google Scholar] [CrossRef]
- Tam, H.H.; Zhu, D.; Ho, S.S.K.; Vong, H.W.; Wong, V.K.W.; Mok, S.W.; Wong, I.N. Potential enhancement of post-stroke angiogenic response by targeting the oligomeric aggregation of p53 protein. Front. Cell Neurosci. 2023, 17, 1193362. [Google Scholar] [CrossRef]
- Zeng, Z.; Zhang, Y.; Jiang, W.; He, L.; Qu, H. Modulation of autophagy in traumatic brain injury. J. Cell Physiol. 2020, 235, 1973–1985. [Google Scholar] [CrossRef]
- Denorme, F.; Portier, I.; Rustad, J.L.; Cody, M.J.; de Araujo, C.V.; Hoki, C.; Alexander, M.D.; Grandhi, R.; Dyer, M.R.; Neal, M.D.; et al. Neutrophil extracellular traps regulate ischemic stroke brain injury. J. Clin. Investig. 2022, 132, e154225. [Google Scholar] [CrossRef]
- Kang, L.; Yu, H.; Yang, X.; Zhu, Y.; Bai, X.; Wang, R.; Cao, Y.; Xu, H.; Luo, H.; Lu, L.; et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat. Commun. 2020, 11, 2488. [Google Scholar] [CrossRef]
- Jin, J.; Wang, F.; Tian, J.; Zhao, X.; Dong, J.; Wang, N.; Liu, Z.; Zhao, H.; Li, W.; Mang, G.; et al. Neutrophil extracellular traps contribute to coagulopathy after traumatic brain injury. JCI Insight 2023, 8, 141110. [Google Scholar] [CrossRef] [PubMed]
- Shi, G.; Liu, L.; Cao, Y.; Ma, G.; Zhu, Y.; Xu, J.; Zhang, X.; Li, T.; Mi, L.; Jia, H.; et al. Inhibition of neutrophil extracellular trap formation ameliorates neuroinflammation and neuronal apoptosis via STING-dependent IRE1alpha/ASK1/JNK signaling pathway in mice with traumatic brain injury. J. Neuroinflammation 2023, 20, 222. [Google Scholar] [CrossRef] [PubMed]
- Troy, C.M.; Jean, Y.Y. Caspases: Therapeutic targets in neurologic disease. Neurotherapeutics 2015, 12, 42–48. [Google Scholar] [CrossRef]
- Shimohama, S.; Tanino, H.; Fujimoto, S. Changes in caspase expression in Alzheimer’s disease: Comparison with development and aging. Biochem. Biophys. Res. Commun. 1999, 256, 381–384. [Google Scholar] [CrossRef]
- D’Amelio, M.; Cavallucci, V.; Middei, S.; Marchetti, C.; Pacioni, S.; Ferri, A.; Diamantini, A.; De Zio, D.; Carrara, P.; Battistini, L.; et al. Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat. Neurosci. 2011, 14, 69–76. [Google Scholar] [CrossRef]
- D’Amelio, M.; Sheng, M.; Cecconi, F. Caspase-3 in the central nervous system: Beyond apoptosis. Trends Neurosci. 2012, 35, 700–709. [Google Scholar] [CrossRef]
- Rohn, T.T.; Head, E. Caspases as therapeutic targets in Alzheimer’s disease: Is it time to “cut” to the chase? Int. J. Clin. Exp. Pathol. 2009, 2, 108–118. [Google Scholar] [PubMed]
- Huang, I.H.; Chung, W.H.; Wu, P.C.; Chen, C.B. JAK-STAT signaling pathway in the pathogenesis of atopic dermatitis: An updated review. Front. Immunol. 2022, 13, 1068260. [Google Scholar] [CrossRef]
- Hermel, E.; Gafni, J.; Propp, S.S.; Leavitt, B.R.; Wellington, C.L.; Young, J.E.; Hackam, A.S.; Logvinova, A.V.; Peel, A.L.; Chen, S.F.; et al. Specific caspase interactions and amplification are involved in selective neuronal vulnerability in Huntington’s disease. Cell Death Differ. 2004, 11, 424–438. [Google Scholar] [CrossRef]
- Skotte, N.H.; Pouladi, M.A.; Ehrnhoefer, D.E.; Huynh, K.; Qiu, X.; Nielsen, S.M.B.; Nielsen, T.T.; Norremolle, A.; Hayden, M.R. Compromised IGF signaling causes caspase-6 activation in Huntington disease. Exp. Neurol. 2020, 332, 113396. [Google Scholar] [CrossRef]
- Duarte, A.I.; Petit, G.H.; Ranganathan, S.; Li, J.Y.; Oliveira, C.R.; Brundin, P.; Bjorkqvist, M.; Rego, A.C. IGF-1 protects against diabetic features in an in vivo model of Huntington’s disease. Exp. Neurol. 2011, 231, 314–319. [Google Scholar] [CrossRef] [PubMed]
- Jean, Y.Y.; Ribe, E.M.; Pero, M.E.; Moskalenko, M.; Iqbal, Z.; Marks, L.J.; Greene, L.A.; Troy, C.M. Caspase-2 is essential for c-Jun transcriptional activation and Bim induction in neuron death. Biochem. J. 2013, 455, 15–25. [Google Scholar] [CrossRef] [PubMed]
- Pozueta, J.; Lefort, R.; Ribe, E.M.; Troy, C.M.; Arancio, O.; Shelanski, M. Caspase-2 is required for dendritic spine and behavioural alterations in J20 APP transgenic mice. Nat. Commun. 2013, 4, 1939. [Google Scholar] [CrossRef]
- Pockes, S.; Walters, M.A.; Ashe, K.H. Targeting caspase-2 interactions with tau in Alzheimer’s disease and related dementias. Transl. Res. 2023, 254, 34–40. [Google Scholar] [CrossRef] [PubMed]
- Eckelman, B.P.; Salvesen, G.S. The human anti-apoptotic proteins cIAP1 and cIAP2 bind but do not inhibit caspases. J. Biol. Chem. 2006, 281, 3254–3260. [Google Scholar] [CrossRef]
- Robertson, G.S.; Crocker, S.J.; Nicholson, D.W.; Schulz, J.B. Neuroprotection by the inhibition of apoptosis. Brain Pathol. 2000, 10, 283–292. [Google Scholar] [CrossRef]
- Ando, S.; Funato, M.; Ohuchi, K.; Inagaki, S.; Sato, A.; Seki, J.; Kawase, C.; Saito, T.; Nishio, H.; Nakamura, S.; et al. The Protective Effects of Levetiracetam on a Human iPSCs-Derived Spinal Muscular Atrophy Model. Neurochem. Res. 2019, 44, 1773–1779. [Google Scholar] [CrossRef]
- Siddiqui, W.A.; Ahad, A.; Ahsan, H. The mystery of BCL2 family: Bcl-2 proteins and apoptosis: An update. Arch. Toxicol. 2015, 89, 289–317. [Google Scholar] [CrossRef]
- Reyes, N.A.; Fisher, J.K.; Austgen, K.; VandenBerg, S.; Huang, E.J.; Oakes, S.A. Blocking the mitochondrial apoptotic pathway preserves motor neuron viability and function in a mouse model of amyotrophic lateral sclerosis. J. Clin. Investig. 2010, 120, 3673–3679. [Google Scholar] [CrossRef]
- Iaccarino, C.; Mura, M.E.; Esposito, S.; Carta, F.; Sanna, G.; Turrini, F.; Carri, M.T.; Crosio, C. Bcl2-A1 interacts with pro-caspase-3: Implications for amyotrophic lateral sclerosis. Neurobiol. Dis. 2011, 43, 642–650. [Google Scholar] [CrossRef]
- Sanchez, I.; Xu, C.J.; Juo, P.; Kakizaka, A.; Blenis, J.; Yuan, J. Caspase-8 is required for cell death induced by expanded polyglutamine repeats. Neuron 1999, 22, 623–633. [Google Scholar] [CrossRef] [PubMed]
- Gafni, J.; Ellerby, L.M. Calpain activation in Huntington’s disease. J. Neurosci. 2002, 22, 4842–4849. [Google Scholar] [CrossRef] [PubMed]
- Matheoud, D.; Cannon, T.; Voisin, A.; Penttinen, A.M.; Ramet, L.; Fahmy, A.M.; Ducrot, C.; Laplante, A.; Bourque, M.J.; Zhu, L.; et al. Intestinal infection triggers Parkinson’s disease-like symptoms in Pink1(−/−) mice. Nature 2019, 571, 565–569. [Google Scholar] [CrossRef]
- Klingelhoefer, L.; Reichmann, H. Pathogenesis of Parkinson disease--the gut-brain axis and environmental factors. Nat. Rev. Neurol. 2015, 11, 625–636. [Google Scholar] [CrossRef] [PubMed]
- Johnson, M.E.; Stecher, B.; Labrie, V.; Brundin, L.; Brundin, P. Triggers, Facilitators, and Aggravators: Redefining Parkinson’s Disease Pathogenesis. Trends Neurosci. 2019, 42, 4–13. [Google Scholar] [CrossRef]
- Sarkar, S. Microglial ion channels: Key players in non-cell autonomous neurodegeneration. Neurobiol. Dis. 2022, 174, 105861. [Google Scholar] [CrossRef]
- Zhang, C.; Zhao, M.; Wang, B.; Su, Z.; Guo, B.; Qin, L.; Zhang, W.; Zheng, R. The Nrf2-NLRP3-caspase-1 axis mediates the neuroprotective effects of Celastrol in Parkinson’s disease. Redox Biol. 2021, 47, 102134. [Google Scholar] [CrossRef]
- Han, X.; Sun, S.; Sun, Y.; Song, Q.; Zhu, J.; Song, N.; Chen, M.; Sun, T.; Xia, M.; Ding, J.; et al. Small molecule-driven NLRP3 inflammation inhibition via interplay between ubiquitination and autophagy: Implications for Parkinson disease. Autophagy 2019, 15, 1860–1881. [Google Scholar] [CrossRef]
- Chen, J.; Mao, K.; Yu, H.; Wen, Y.; She, H.; Zhang, H.; Liu, L.; Li, M.; Li, W.; Zou, F. p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson’s disease. J. Neuroinflammation 2021, 18, 295. [Google Scholar] [CrossRef]
- Panicker, N.; Kam, T.I.; Wang, H.; Neifert, S.; Chou, S.C.; Kumar, M.; Brahmachari, S.; Jhaldiyal, A.; Hinkle, J.T.; Akkentli, F.; et al. Neuronal NLRP3 is a parkin substrate that drives neurodegeneration in Parkinson’s disease. Neuron 2022, 110, 2422–2437.e2429. [Google Scholar] [CrossRef]
- Van Opdenbosch, N.; Lamkanfi, M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019, 50, 1352–1364. [Google Scholar] [CrossRef]
- McKenzie, B.A.; Fernandes, J.P.; Doan, M.A.L.; Schmitt, L.M.; Branton, W.G.; Power, C. Activation of the executioner caspases-3 and -7 promotes microglial pyroptosis in models of multiple sclerosis. J. Neuroinflammation 2020, 17, 253. [Google Scholar] [CrossRef] [PubMed]
- Wu, A.G.; Zhou, X.G.; Qiao, G.; Yu, L.; Tang, Y.; Yan, L.; Qiu, W.Q.; Pan, R.; Yu, C.L.; Law, B.Y.; et al. Targeting microglial autophagic degradation in NLRP3 inflammasome-mediated neurodegenerative diseases. Ageing Res. Rev. 2021, 65, 101202. [Google Scholar] [CrossRef] [PubMed]
- Neel, D.V.; Basu, H.; Gunner, G.; Bergstresser, M.D.; Giadone, R.M.; Chung, H.; Miao, R.; Chou, V.; Brody, E.; Jiang, X.; et al. Gasdermin-E mediates mitochondrial damage in axons and neurodegeneration. Neuron 2023, 111, 1222–1240.e1229. [Google Scholar] [CrossRef] [PubMed]
- He, L.; Chen, Y.; Lin, S.; Shen, R.; Pan, H.; Zhou, Y.; Wang, Y.; Chen, S.; Ding, J. Regulation of Hsa-miR-4639-5p expression and its potential role in the pathogenesis of Parkinson’s disease. Aging Cell 2023, 22, e13840. [Google Scholar] [CrossRef] [PubMed]
- Soldati, C.; Bithell, A.; Conforti, P.; Cattaneo, E.; Buckley, N.J. Rescue of gene expression by modified REST decoy oligonucleotides in a cellular model of Huntington’s disease. J. Neurochem. 2011, 116, 415–425. [Google Scholar] [CrossRef]
- Hwang, J.Y.; Zukin, R.S. REST, a master transcriptional regulator in neurodegenerative disease. Curr. Opin. Neurobiol. 2018, 48, 193–200. [Google Scholar] [CrossRef]
- Wang, J.F.; Li, Y.; Song, J.N.; Pang, H.G. Role of hydrogen sulfide in secondary neuronal injury. Neurochem. Int. 2014, 64, 37–47. [Google Scholar] [CrossRef]
- Pandya, J.D.; Musyaju, S.; Modi, H.R.; Cao, Y.; Flerlage, W.J.; Huynh, L.; Kociuba, B.; Visavadiya, N.P.; Kobeissy, F.; Wang, K.; et al. Comprehensive evaluation of mitochondrial redox profile, calcium dynamics, membrane integrity and apoptosis markers in a preclinical model of severe penetrating traumatic brain injury. Free Radic. Biol. Med. 2023, 198, 44–58. [Google Scholar] [CrossRef]
- Callens, M.; Kraskovskaya, N.; Derevtsova, K.; Annaert, W.; Bultynck, G.; Bezprozvanny, I.; Vervliet, T. The role of Bcl-2 proteins in modulating neuronal Ca(2+) signaling in health and in Alzheimer’s disease. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118997. [Google Scholar] [CrossRef]
- Zundorf, G.; Reiser, G. Calcium dysregulation and homeostasis of neural calcium in the molecular mechanisms of neurodegenerative diseases provide multiple targets for neuroprotection. Antioxid. Redox Signal 2011, 14, 1275–1288. [Google Scholar] [CrossRef] [PubMed]
- Bao, W.D.; Pang, P.; Zhou, X.T.; Hu, F.; Xiong, W.; Chen, K.; Wang, J.; Wang, F.; Xie, D.; Hu, Y.Z.; et al. Loss of ferroportin induces memory impairment by promoting ferroptosis in Alzheimer’s disease. Cell Death Differ. 2021, 28, 1548–1562. [Google Scholar] [CrossRef]
- Origa, R.; Danjou, F.; Cossa, S.; Matta, G.; Bina, P.; Dessi, C.; Defraia, E.; Foschini, M.L.; Leoni, G.; Morittu, M.; et al. Impact of heart magnetic resonance imaging on chelation choices, compliance with treatment and risk of heart disease in patients with thalassaemia major. Br. J. Haematol. 2013, 163, 400–403. [Google Scholar] [CrossRef] [PubMed]
- Crapper McLachlan, D.R.; Dalton, A.J.; Kruck, T.P.; Bell, M.Y.; Smith, W.L.; Kalow, W.; Andrews, D.F. Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet 1991, 337, 1304–1308. [Google Scholar] [CrossRef]
- Guo, C.; Wang, T.; Zheng, W.; Shan, Z.Y.; Teng, W.P.; Wang, Z.Y. Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer’s disease. Neurobiol. Aging 2013, 34, 562–575. [Google Scholar] [CrossRef]
- Bush, A.I. The metallobiology of Alzheimer’s disease. Trends Neurosci. 2003, 26, 207–214. [Google Scholar] [CrossRef]
- Cherny, R.A.; Atwood, C.S.; Xilinas, M.E.; Gray, D.N.; Jones, W.D.; McLean, C.A.; Barnham, K.J.; Volitakis, I.; Fraser, F.W.; Kim, Y.; et al. Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 2001, 30, 665–676. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Wang, C.Y.; Shan, Z.Y.; Teng, W.P.; Wang, Z.Y. Clioquinol reduces zinc accumulation in neuritic plaques and inhibits the amyloidogenic pathway in AbetaPP/PS1 transgenic mouse brain. J. Alzheimers Dis. 2012, 29, 549–559. [Google Scholar] [CrossRef]
- Ritchie, C.W.; Bush, A.I.; Mackinnon, A.; Macfarlane, S.; Mastwyk, M.; MacGregor, L.; Kiers, L.; Cherny, R.; Li, Q.X.; Tammer, A.; et al. Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: A pilot phase 2 clinical trial. Arch. Neurol. 2003, 60, 1685–1691. [Google Scholar] [CrossRef]
- Chen, L.L.; Fan, Y.G.; Zhao, L.X.; Zhang, Q.; Wang, Z.Y. The metal ion hypothesis of Alzheimer’s disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem. 2023, 131, 106301. [Google Scholar] [CrossRef]
- Zhong, X.; Liu, M.; Yao, W.; Du, K.; He, M.; Jin, X.; Jiao, L.; Ma, G.; Wei, B.; Wei, M. Epigallocatechin-3-Gallate Attenuates Microglial Inflammation and Neurotoxicity by Suppressing the Activation of Canonical and Noncanonical Inflammasome via TLR4/NF-kappaB Pathway. Mol. Nutr. Food Res. 2019, 63, e1801230. [Google Scholar] [CrossRef] [PubMed]
- Rong, S.; Wan, D.; Fan, Y.; Liu, S.; Sun, K.; Huo, J.; Zhang, P.; Li, X.; Xie, X.; Wang, F.; et al. Amentoflavone Affects Epileptogenesis and Exerts Neuroprotective Effects by Inhibiting NLRP3 Inflammasome. Front. Pharmacol. 2019, 10, 856. [Google Scholar] [CrossRef]
- Ghavami, S.; Shojaei, S.; Yeganeh, B.; Ande, S.R.; Jangamreddy, J.R.; Mehrpour, M.; Christoffersson, J.; Chaabane, W.; Moghadam, A.R.; Kashani, H.H.; et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog. Neurobiol. 2014, 112, 24–49. [Google Scholar] [CrossRef]
- Querfurth, H.; Lee, H.K. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol. Neurodegener. 2021, 16, 44. [Google Scholar] [CrossRef] [PubMed]
- Granatiero, V.; Sayles, N.M.; Savino, A.M.; Konrad, C.; Kharas, M.G.; Kawamata, H.; Manfredi, G. Modulation of the IGF1R-MTOR pathway attenuates motor neuron toxicity of human ALS SOD1(G93A) astrocytes. Autophagy 2021, 17, 4029–4042. [Google Scholar] [CrossRef]
- Hoeffer, C.A.; Klann, E. mTOR signaling: At the crossroads of plasticity, memory and disease. Trends Neurosci. 2010, 33, 67–75. [Google Scholar] [CrossRef] [PubMed]
- Stoica, L.; Zhu, P.J.; Huang, W.; Zhou, H.; Kozma, S.C.; Costa-Mattioli, M. Selective pharmacogenetic inhibition of mammalian target of Rapamycin complex I (mTORC1) blocks long-term synaptic plasticity and memory storage. Proc. Natl. Acad. Sci. USA 2011, 108, 3791–3796. [Google Scholar] [CrossRef]
- Zheng, S.; Clabough, E.B.; Sarkar, S.; Futter, M.; Rubinsztein, D.C.; Zeitlin, S.O. Deletion of the huntingtin polyglutamine stretch enhances neuronal autophagy and longevity in mice. PLoS Genet. 2010, 6, e1000838. [Google Scholar] [CrossRef]
- Salminen, A.; Kaarniranta, K.; Kauppinen, A.; Ojala, J.; Haapasalo, A.; Soininen, H.; Hiltunen, M. Impaired autophagy and APP processing in Alzheimer’s disease: The potential role of Beclin 1 interactome. Prog. Neurobiol. 2013, 106–107, 33–54. [Google Scholar] [CrossRef]
- Spencer, B.; Potkar, R.; Trejo, M.; Rockenstein, E.; Patrick, C.; Gindi, R.; Adame, A.; Wyss-Coray, T.; Masliah, E. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson’s and Lewy body diseases. J. Neurosci. 2009, 29, 13578–13588. [Google Scholar] [CrossRef]
- Rodriguez-Muela, N.; Parkhitko, A.; Grass, T.; Gibbs, R.M.; Norabuena, E.M.; Perrimon, N.; Singh, R.; Rubin, L.L. Blocking p62-dependent SMN degradation ameliorates spinal muscular atrophy disease phenotypes. J. Clin. Investig. 2018, 128, 3008–3023. [Google Scholar] [CrossRef] [PubMed]
Modulators | Functions | Refs. |
---|---|---|
hepcidin | degrading the ferroportin via ubiquitination | [102] |
IRP1/2 | promoting the expression of TFR1 | [103] |
PCBP1 | delivering ferrous iron to ferritin | [104] |
SLC25A37/8 | promoting the absorption of iron | [105] |
HO1 | catalyzing the synthesis of ferrous iron | [106] |
SLC40A1 | assisting the export of iron | [107] |
SLC39A14 | assisting the import of iron | [108] |
SLC25A28 | regulating the generation of ROS | [109] |
SFXN1 | regulating the generation of ROS | [110] |
PROM2 | regulating the storage of iron in ferritin | [111] |
PHKG2 | modulating the oxidative reactions | [112] |
HMOX1 | participating in the synthesis of ferrous iron | [113] |
SLC11A2 | assisting the absorption of iron | [87] |
CP | converting ferrous iron to ferric iron | [114] |
CISD1/2 | participating in the absorption of iron | [115,116] |
DMT1 | controlling the absorption of iron | [117] |
FBXL5 | degrading the IRP1 via ubiquitination | [118] |
HSF1 | regulating the iron metabolism-related genes | [119] |
HSPA5 | binding to GPX4 to prevent its degradation | [120] |
NRF2 | inducing the expression of antioxidant genes | [121] |
CISD1 | alleviating the accumulation of lipid | [122] |
ALOXs | facilitating the lipoxygenase | [123] |
PEBP1 | enhancing the lipid death pathway via 15-LO | [124] |
NOXs | facilitating the generation of ROS | [125] |
DPP4/CD26 | causing the lipid peroxidation | [126,127] |
VDAC2/3 | activating ferroptotic agonist erastin | [128] |
MUC1 | activating GPX4 | [129] |
GCLC | accelerating the synthesis of GSH | [130] |
GLS2 | increasing the ROS production via αKG | [131] |
CARS | inhibiting the generation of GSH | [132] |
CHAC1 | promoting oxidative reactions | [133] |
LSH | promoting SLC7A11 transcription | [134] |
FADS2 | desaturating the free fatty acids | [135] |
ACSL3 | upregulating the lipid droplet biogenesis | [136] |
ACSL4 | shaping cellular composition | [137] |
LPCAT3 | upregulating polyunsaturated free fatty acid | [138] |
PHGDH | upregulating the expression of PHGDH | [139] |
G6PD | preventing oxidative reactions via inhibiting POR | [140] |
ME1 | facilitating the generation of GSH | [141] |
PHKG2 | regulating lipoxygenase enzyme ALOX5 | [142] |
HMGCR | increasing GPX4 and CoQ10 biosynthesis | [143] |
SQLE | preventing oxidative via squalene | [144] |
NRF2 | regulating the antioxidant-related genes expression | [145] |
P53 | modulating GPX4 pathway and ROS production | [146] |
HIF-1α | improving the expression of SLC7A11 via PMAN | [147] |
BACH1 | enhancing iron metabolisom-related gene expression | [148] |
STAT3 | enhancing the expression of GPX4 and SLC7A11 | [149] |
ATF3 | restraining the activity of system Xc− | [150] |
ATF4 | facilitating the expression of SLC7A11 | [151] |
CHOP | facilitating the expression of CHAC1 | [152] |
YAP/TAZ | inducing the expression of SLC7A11 | [153] |
Forms | Immune Features | Morphological Features | Major Inhibitors | Refs. |
---|---|---|---|---|
Apoptosis | TCD | Apoptotic bodies formation; Nuclear condensation; Plasma membrane blebbing; Cell shrinkage. | Z-VAD-FMK; Q-VD-OPh; Z-VAD (OH)-FMK. | [4] [17] |
Necroptosis | ICD | Cell swelling and oncosis Rupture of plasma membrane; Swelling of organelles Chromatin condensation. | Nec-1; GSK872; NSA; HS-1371. | [46] [49] |
Pyroptosis | ICD | Lack of cell swelling; Rupture of plasma membrane; Cell bubbling; Chromatin condensation. | Ac-YVAD-cmk; VX765; Ac-FLTD-CMK. | [67] [204] |
Ferroptosis | ICD | Smaller mitochondria; Rupture of mitochondrial membrane; Decreased mitochondrial cristae; Normal nucleus. | Deferiprone; Ferrostatin-1; Alogliptin; Selenium; CoQ10; Vildagliptin; Vitamin E. | [80] [86] [205] |
Cuproptosis | ICD | mitochondrial condensation; Rupture of plasma membrane; ER damage; Chromatin condensation. | NSC689534 EMeramide Penicillamine AT-VI | [7] [154] |
MPT-driven necrosis | ICD | Plasma membrane rupture; Swelling of organelles; Lack of inter-nucleosomal DNA fragmentation; Depletion of ATP. | SfA | [5] |
ADCD | ICD | Autophagic vacuolization. | Chloroquline; Spactin-1. | [168] |
LDCD | ICD | Rupture of lysosome and plasma membrane. | NAC; CA-074Me | [1] |
Parthanatos | ICD | Chromatin condensation; Large DNA fragmentation; Loss of cell swelling. | BYK204165; AG-14361; Iniparib. | [184] [185] |
Oxeiptosis | TCD | Apoptosis-like morphology. | NAC. | [188] |
Alkaliptosis | ICD | Necrosis-like morphology. | NAC; CAY10657; SC514. | [5] |
Disulfidptosis | ICD | Cell shrinkage; Nuclear condensation; Formation of aberrant disulfide bonds between actin cytoskeleton proteins; Chromatin condensation. | GLUT inhibitor | [6] [189] |
NETosis | TCD or ICD | Rupture of plasma membrane and nuclear membrane; Release of chromatin fragments. | lactoferrin; DNase; Cl-amidine. | [190] [206] |
ENTosis | TCD or ICD | Formation of cell-in-cell structure. | C3-toxin; γ-27632 | [196] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Guo, D.; Liu, Z.; Zhou, J.; Ke, C.; Li, D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. Int. J. Mol. Sci. 2024, 25, 9947. https://doi.org/10.3390/ijms25189947
Guo D, Liu Z, Zhou J, Ke C, Li D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2024; 25(18):9947. https://doi.org/10.3390/ijms25189947
Chicago/Turabian StyleGuo, Dong, Zhihao Liu, Jinglin Zhou, Chongrong Ke, and Daliang Li. 2024. "Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases" International Journal of Molecular Sciences 25, no. 18: 9947. https://doi.org/10.3390/ijms25189947
APA StyleGuo, D., Liu, Z., Zhou, J., Ke, C., & Li, D. (2024). Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. International Journal of Molecular Sciences, 25(18), 9947. https://doi.org/10.3390/ijms25189947