Next Article in Journal
Transcriptomic and Biochemical Analysis of the Antimicrobial Mechanism of Lipopeptide Iturin W against Staphylococcus aureus
Previous Article in Journal
Photo-Crosslinked Pro-Angiogenic Hydrogel Dressing for Wound Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases

1
College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
2
Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(18), 9947; https://doi.org/10.3390/ijms25189947
Submission received: 27 July 2024 / Revised: 7 September 2024 / Accepted: 12 September 2024 / Published: 15 September 2024
(This article belongs to the Special Issue Cell Apoptosis, 3rd Edition)

Abstract

:
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases.

1. Introduction

The Nomenclature of Cell Death Committee has established guidelines dividing cell death into two distinct categories: accidental cell death (ACD) and programmed cell death (PCD) [1]. ACD is an uncontrolled cellular process that occurs in response to accidental injury stimuli, such as necrosis [2]. PCD is essential for maintaining physiological homeostasis in mammals by clearing damaged cells, facilitating tissue renewal, and supporting organismal development, all of which are strictly regulated by intracellular signaling cascades [3]. The hallmark ultrastructural features of cells undergoing PCD, including cytoplasmic shrinkage, nuclear condensation, and chromatin fragmentation, were first observed in 1972, leading to the definition of this form of PCD as apoptosis. These features are evident in various tissues under both physiological and certain pathological conditions [4]. In the past five decades, novel forms of PCD and their corresponding signaling cascades have been identified, including necroptosis, pyroptosis, ferroptosis, cuproptosis, mitochondrial permeability transition (MPT)-driven necrosis, autophagy-dependent cell death (ADCD), lysosome-dependent cell death (LDCD), parthanatos, alkaliptosis, oxeiptosis, NET-release-induced necrotic cell death (NETosis), entotic cell death (ENTosis), and disulfidptosis [5,6,7].
The etiology of neurodegenerative diseases (NDDs) is multifactorial and is associated with abnormalities in various intracellular processes, such as autophagy, mitochondrial biogenesis, homeostasis of the endoplasmic reticulum (ER), and epigenetic modifications [8]. The most well-known NDDs include Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. As the global population continues to grow and age, NDDs have become one of the foremost medical and social concerns worldwide. According to clinical data, the number of people suffering from Parkinson’s disease (PD) tripled between 1996 and 2016 [9]. The prevalence of AD in China among those aged over 60 was 1.37%, and more than 50 million people are affected by AD worldwide [10]. The incidence of HD, ALS, SMA, MS, TBI, and stroke has also been increasing annually worldwide [11,12,13,14,15]. Notably, stroke remains the second-leading cause of death worldwide, with the estimated global cost of stroke being approximately 0.66% of global GDP [16].
The central nervous system (CNS) comprises the brain and spinal cord. Under normal conditions, PCD signaling cascades are tightly regulated at temporal and spatial levels to establish neural architecture in the CNS [17]. During normal neural embryonic and postnatal development, apoptosis controls the survival of embryonic stem cells that have the appropriate size and shape and have made proper connections with their axons and neurites [18]. In addition, regulators of apoptosis play a crucial role in cell survival during developmental neurogenesis, such as the anti-apoptotic Bcl-2 family members myeloid cell leukemia-1 (MCL-1) and Bcl-2-related gene long isoform (Bcl-XL) [19,20,21]. However, aberrant neuronal cell death is a hallmark of the pathology associated with NDDs, and different PCD pathways interact in the progression of these diseases [22]. In recent years, numerous clinical trials have focused on PCD pathways to develop therapeutic strategies for the treatment of NDDs, achieving inspiring progress.
In this review, we provide a comprehensive overview of the signaling pathways involved in various PCD subroutines. Subsequently, we elucidate the similarities and differences among these pathways. We further discuss the role of different PCD pathways in the pathogenesis and progression of NDDs. Finally, we discuss existing and potential therapeutic strategies focusing on the central regulators of various PCD pathways for the treatment of NDDs.

2. Forms of Programmed Cell Death

2.1. Apoptosis

There are two distinct pathways that trigger apoptosis: the intrinsic and extrinsic pathways. The intrinsic pathway, also called mitochondrial or Bcl-2-regulated apoptosis, is characterized by non-receptor-mediated initiation and mitochondrial dependence in response to intracellular stress, such as DNA damage, ER stress, hypoxia, extremely high concentrations of cytosolic calcium, microtubular alteration, and growth factor deprivation [23]. After the generation of intracellular stimuli, the expression of BH3 (bcl-2 homolog3r)-only proteins, including BIM, PUMA, BID, BMF, BAD, HRK, BIK, and NOXA, is upregulated. These BH3-only proteins then bind to anti-apoptotic proteins, such as Bcl-2, Bcl-XL, and Mcl-1, to liberate and activate pro-apoptotic proteins, such as BAX, BAK, and BOK [24,25,26]. Subsequently, pro-apoptotic proteins undergo oligomerization, causing the dissipation of mitochondrial membrane potential. This leads to the disruption of mitochondrial outer membrane permeability (MOMP) and the formation of the mitochondrial permeability transition pore (MPT), allowing apoptogenic factors such as cytochrome-c and small mitochondria-derived activator of caspase (Smac) to be released into the cytosol [27,28,29]. Of note, cytochrome-c binds to the apoptosis protease activating factor-1 (Apaf-1) and induces the formation of the apoptosome, which recruits procaspase-9 for its cleavage and activation. Next, activated caspase-9 cleaves and activates its downstream effectors, including caspase-3, caspase-6, and caspase-7 [30]. The presence of Smac in the cytosol prevents the activation of inhibitor of apoptosis proteins (IAPs) through direct binding, thereby allowing for the initiation of caspase-dependent pathways [31]. These apoptogenic factors aim to induce the activation of caspase-dependent cascades, resulting in the cleavage of hundreds of proteins and ultimately apoptosis. Similarly, ER stress triggered by an imbalance in calcium homeostasis can induce the expression of caspase-12, which is localized in the ER membrane, and then recruits caspase-7 to the ER membrane to initiate apoptosis [32,33,34]. In addition, apoptosis-inducing factors (AIFs) can induce apoptosis independently of caspase signals [35]. After the cleavage of calcium-dependent proteases, especially calpain, AIFs are translocated from the inner membrane of mitochondria to the nucleus by nuclear localization signals (NLSs), leading to genome instability and chromatin fragmentation [36].
The extrinsic pathway, also known as death receptor apoptosis, is induced by the interaction between extracellular ligands and death receptors anchored in the cell membrane [37]. The extracellular ligands consist mainly of the tumor necrosis factor (TNF) superfamily, Fas ligand (FasL), and TNF-related apoptosis-inducing ligand (TRAIL), which bind to death receptors, such as TNF receptor (TNFR)-1, TNFR-2, Fas, TRAILR1, and TRAILR2 [38]. Upon the binding of ligands and death receptors, the death receptors undergo oligomerization and conformational changes to expose their death domain (DD) for the recruitment of TNF receptor-associated death domain (TRADD) and Fas-associated death domain (FADD) adaptor proteins, leading to the formation of an intracellular death-inducing signaling complex (DISC) for Fas-FasL and TRAIL-TRAILR, as well as complex II for TNF-TNFR [39,40]. Next, DISC and complex II mediate the cleavage and activation of procaspase-8, initiating the cleavage of caspase-3 and caspase-7 to induce apoptotic signaling pathways and the proteolytic degradation of a variety of intracellular proteins [37,39,41]. In some specific situations, apoptosis cannot be activated by the extrinsic pathways. Therefore, it is necessary for cleaved caspase-8 to interact with BH3-interacting domain death agonist (BID) and then cleave BID to form activated tBID. tBID subsequently directly activates pro-apoptotic multi-domain proteins to induce MOMP and mitochondrial apoptosis [17,37,40]. The mechanisms of apoptotic pathways are depicted in Figure 1.

2.2. Necroptosis

Necroptosis can be regarded as a regulated form of necrosis, first introduced in 2005 [42]. Canonical necroptosis is an alternative to apoptosis because its activation relies on the engagement of apoptotic extracellular ligands and their corresponding death receptors when the activation of caspase-8 is inhibited by pharmacological agents or viral inhibitors [43,44]. After the binding of ligands and receptors, such as TNF-TNFR, FAS-FASL, and TRAIL-TRAILR, DD recruits receptor-interacting serine/threonine protein kinase 1 (RIPK1) through homotypic binding, and then RIPK1 undergoes autophosphorylation [45]. Phosphorylated RIPK1 binds and phosphorylates RIPK3 through shared RIP (receptor-interacting protein) homology interaction motifs (RHIMs) [46,47,48]. Subsequently, mixed-lineage kinase like (MLKL) is phosphorylated by the RIPK1-RIPK3 complex and then undergoes oligomerization to form a high-molecular-weight complex, termed the necrosome, in the cytosol [49,50]. Next, the necrosome translocates to the plasma membrane, causing rupture of the plasma membrane, cell swelling, the release of cytokines and chemokines, as well as potassium efflux, leading to inflammation and immune responses [51]. Similarly, tumor-cell-derived amyloid precursor protein (APP) can activate the RIPK1-RIPK3-MLKL axis-induced necroptosis through binding to death receptor 6 (DR6) in endothelial cells, which enhances the extravasation of circulating tumor cells (CTCs) [52,53]. In addition, viral RNA and DNA or RNA leaked from damaged mitochondria can also induce necroptosis by activating RHIM-containing Z-dsDNA/dsRNA-binding protein (ZBP1), which subsequently results in the activation of the RIPK3-MLKL axis [54,55]. Meanwhile, necroptosis can also be triggered by the recognition of toll-like receptor 3 (TLR3) and TLR4 to double-stranded RNA (dsRNA) from viruses and lipopolysaccharide (LPS) from bacteria, respectively [56,57]. Upon binding, TLR3 and TLR4 are capable of activating TIR-domain-containing adapter-inducing interferon-β (TRIF) containing RHIM, ensuing the activation of the RIPK3-MLKL axis [43]. Additionally, proinflammatory factors such as interferons (IFNs) can trigger RIPK1-RIPK3-MLKL axis-mediated necroptosis or ZBP1-RIPK3-MLKL axis-mediated necroptosis in the absence of RIPK1 via sensing IFN receptors (IFNRs), suggesting that necroptosis is essential for the induction of inflammation [58]. Figure 1 depicts the activation of various necroptotic pathways.

2.3. Pyroptosis

Pyroptosis, a proinflammatory form of programmed cell death first discovered in 1989, is triggered by a variety of inflammasomes and is executed by the caspase and gasdermin (GSDM) families [59]. Pyroptotic signaling cascades include the canonical, non-canonical, caspase-3-induced, caspase-8-induced, and granzyme (GZM)-mediated pathways [60]. The purpose of the canonical pathway is to respond to pathogen invasion and facilitate the development of adaptive immune responses [60]. Upon the activation of pattern recognition receptors (PRRs) by pathogenic exposures, such as bacteria and viruses, PRRs recruit pro-caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC) to assemble the inflammasome. After that, pro-caspase-1 undergoes self-cleavage to form activated caspase-1, leading to the cleavage of GSDMD and the release of its cytotoxic N-terminal P30 fragment containing the pore formation domain (PFD) as well as the synthesis of IL-18 and IL-1β [61]. Finally, the N-terminus inserts into the plasma membrane and oligomerizes to form pores with inner diameters of approximately 12–14 nm in the plasma membrane, leading to the expulsion of proinflammatory factors, chromatin degradation, and cell swelling [62]. The non-canonical pathway is triggered by the infection of Gram-negative bacteria [63]. In mice, caspase-11, or in humans, caspase-4 and caspase-5, can be activated by bacterial LPS through CARD, after which they proteolytically hydrolyze GSDMD to release the N-terminus containing PFD. The N-terminus finally undergoes oligomerization to translocate to the plasma membrane and cause plasma membrane perforation [64]. Significantly, activated caspase-11, caspase-4, and caspase-5 can also be packaged into the NLRP3 inflammasome to induce pyroptosis through cleaving GSDMD and causing the efflux of potassium ions (K+) [65,66,67]. In addition, activated caspase-11 can also cause pyroptosis through activating the Pannexin-1-ATP-P2X7 channel and the efflux of potassium ions (K+) [68]. Caspase-4, caspase-5, and caspase-11 participate in the maturation and secretion of IL-18 and IL-1β rather than their synthesis [69]. The protein phosphatase PtpB from Mycobacterium tuberculosis can dephosphorylate phosphatidylinositol-4-monophosphate and phosphatidylinositol-(4,5)-bisphosphate to disrupt the localization of the N-terminus of GSDMD in the cell membrane, suggesting a novel mechanism for the regulation of pyroptosis [70].
The caspase-3-induced pyroptotic pathway has crosstalk with mitochondrial apoptosis. The leakage of cytochrome c triggered by cytotoxic chemotherapy is involved in the formation of the apoptosome, thereby cleaving pro-caspase-3 to cleave GSDME for the release of its N-terminus containing PFD. Subsequently, the N-terminus of GSDME undergoes oligomerization and re-localizes to the cell membrane, thus converting intrinsic apoptosis to pyroptosis or secondary necrosis [71]. Simultaneously, the caspase-8-induced pyroptotic pathway can be executed by the extrinsic apoptotic pathway. After the formation of complex II, caspase-8 is activated to cleave GSDMC, releasing its N-terminus containing PFD. The resulting GSDMC-N fragment translocates to the plasma membrane, converting extrinsic apoptosis to pyroptosis [72]. In addition, PD-L1 can also convert apoptosis to pyroptosis by cleaving GSDMC in cancer cells under hypoxic conditions [73]. During the infection of yersiniosis, the Yersinia effector protein YopJ is able to facilitate the activation of caspase-8 and inhibit the activation of TGF-β-activated kinase 1 (TAK1), leading to the cleavage of GSDMD and the release of its N-terminus containing PFD, causing pore formation in the cell membrane and pyroptosis [74,75]. Additionally, the cysteine protease SpeB of Streptococcus can directly drive the proteolytic cleavage of GSDMA to release the N-terminus containing PFD, resulting in pore formation in the plasma membrane and pyroptosis [76]. Moreover, granzyme A from cytotoxic lymphocytes and granzyme B from natural killer (NK) cells can cleave GSDMB and GSDME, respectively, contributing to the release of their N-terminus and the activation of pyroptosis [77,78]. Meanwhile, GSDMB can be divided into six isoforms due to alternative splicing, and only GSDMB isoform 3 and isoform 4 are capable of recognizing granzyme A to induce pyroptosis because isoform 3 and isoform 4 have a belt motif, raising questions regarding the discovery of GSDMB alternative splicing mechanisms among various diseases [79]. Figure 1 offers a comprehensive overview of the process of pyroptosis.

2.4. Ferroptosis

The term ferroptosis was first coined in 2012. As the name indicates, ferroptosis refers to a form of iron-dependent programmed cell death caused by iron overload in cells [80]. Specifically, iron is an essential trace element that maintains intracellular homeostasis, being involved in the transportation of oxygen, ATP generation, and DNA biosynthesis [81,82]. Under normal conditions, the membrane-bound protein transferrin receptor 1 (TFR1), which contains two ferric iron molecules, recognizes extracellular transferrin (TF) carrying ferric iron and imports iron into cells by triggering clathrin-dependent endocytosis of the entire holo-complex. The ferric iron is then transported into endosomes for reduction to ferrous iron by the STEAP (six-transmembrane epithelial antigen of the prostate) family of metalloreductases [83]. Subsequently, ferrous iron is released from the endosome into the cytosol via natural resistance-associated macrophage protein 2 (NRAMP2), and TFR1 is re-localized to the cell surface to uptake additional TF [84]. Ferritin, an iron-storage protein, can dynamically modulate the oxidation of intracellular ferrous iron to its ferric state, which is used in intracellular enzymatic reactions or stored for later use. Meanwhile, iron-saturated ferritin is degraded by nuclear receptor coactivator 4 (NCOA4)-mediated autophagy, termed ferritinophagy, to release its iron content [83,85]. Correspondingly, ferroportin is the only known iron exporter in mammalian cells, preventing the accumulation of excessive iron in the cell [86,87,88].
However, dysregulation of iron metabolism can cause an overload of ferrous iron in cells, inducing excessive generation of reactive oxygen species (ROS) through the Fenton reaction or oxidation of iron-binding enzymes. This results in the generation of fatal lipid peroxidation products, such as PL-hydroperoxide (PLOOH), malonaldehyde (MDA), and 4-hydroxynonenal (4-HNE) [88,89]. Lipid peroxidation is the final executor that induces cell damage and ferroptosis. In addition, the imbalance between the formation of oxidants and antioxidants triggers an abnormal redox system, which also contributes to the accumulation of lipid peroxides and ferroptosis. To be more specific, cells have evolved multiple antioxidant signaling cascades to protect themselves from ferroptosis, such as the cystine–glutamate antiporter (system Xc) comprising two subunits, SLC3A2 (solute carrier family 3 member 2) and SLC7A11 (solute carrier family 7 member 11), the glutathione peroxidase 4 (GPX4) pathway, the ferroptosis suppressor protein 1-coenzyme Q10 (FSP1-CoQ10) pathway, the dihydroorotate dehydrogenase (DHODH)-CoQ10 pathway, and the GTP cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4) pathway [90,91,92,93]. The final products of the above pathways are glutathione (GSH), CoQ10H2, and BH4, which have ferroptotic protective effects because they are able to decrease the level of oxidative stress [94]. Conversely, dysregulation of the above pathways contributes to the initiation of ferroptosis. Recently, research identified that cyclic GMP-AMP synthase (cGAS) anchored to the outer mitochondrial membrane can associate with dynamin-related protein 1 (DRP1) to facilitate its oligomerization, leading to decreased levels of mitochondrial ROS and avoidance of ferroptosis [95]. Similarly, sex hormone receptors, such as the estrogen receptor (ER) and androgen receptor (AR), can upregulate the expression of phospholipid-modifying enzyme membrane-associated O-acetyl transferase genes (MBOAT1 and MBOAT2), which can reshape the cellular phospholipid profile to protect cells from ferroptosis [96]. Additionally, excessive activation of ferritinophagy and digestion of lipid droplets by autophagy, termed lipophagy, can contribute to lipid peroxidation and ferroptosis via intracellular iron overload and excessive production of free fatty acids, respectively [97,98,99]. In addition, novel clues indicate that selective autophagy, clockphagy, and chaperone-mediated autophagy can modulate the expression of modulators in the redox system to manipulate the process of ferroptosis [100,101]. Ferroptosis is regulated by various molecules. We summarize numerous crucial molecules involved in the modulation of cellular iron homeostasis and ferroptosis in Table 1 and describe the mechanisms of ferroptosis in Figure 2. In general, discovering novel effectors and mechanisms participating in the modulation of ferroptosis, or protective patterns of ferroptosis independent of the aforementioned mechanisms, can help us gain insight into the etiology of various diseases.

2.5. Cuproptosis

Cuproptosis was first identified in 2022 as a novel form of programmed cell death caused by abnormalities in systemic copper metabolism [154]. Specifically, copper ions are essential micronutrients present in all living mammals, especially in humans. They act as co-factors for enzymes, regulating the activity of various key metabolic enzymes and a broad range of physiological processes, such as mitochondrial oxidative phosphorylation (OXPHOS), tyrosine metabolism, neurotransmitter metabolism, redox reactions, extracellular matrix remodeling, and cell proliferation [7]. Meanwhile, the concentration of copper ions must be tightly regulated to ensure normal biochemical processes. Regarding systemic copper metabolism, the uptake of copper ions is mainly mediated by copper transporter 1 (CTR1, also called solute carrier family 31 member 1, SLC31A1) located on the apical side of the enterocytes in the small intestine. Subsequently, copper ions are translocated to the other side of enterocytes via copper chaperone antioxidant 1 (ATOX1) for release into the bloodstream through ATPase copper transporting alpha (ATP7A) and ATPase copper transporting beta (ATP7B) [155]. In the blood, copper ions prefer to bind with soluble chaperones, such as ceruloplasmin (CP), serum albumin (SA), transcuprein, histidines, and macroglobulins, rather than remaining free, and are subsequently transported to the liver by the portal system [7,155]. Upon absorption by the liver, copper ions bind with metallothionein 1/2 (MT1/2) and other thiol-rich proteins in a pH-dependent manner via their cysteine residues for storage or later use [156]. Simultaneously, ATPase copper transporting beta (ATP7B) in hepatocytes pumps copper ions back into the blood or bile, targeting specific tissues and organs or preventing the excessive accumulation of copper ions in the liver, respectively [157,158]. Upon reaching target tissues and organs, copper ions are internalized into the cytosol of target cells by SLC31A1 and then transported to the trans-Golgi network (TGN) or nucleus by chaperone antioxidant-1 (ATOX1) to facilitate the synthesis of cuproenzymes, including lysyl oxidase, tyrosinase, and ceruloplasmin, or to regulate gene expression related to cell proliferation [7]. In addition, copper ions can also be transported into mitochondria through the Cu chaperone for the superoxide dismutase (CCS)-superoxide dismutase 1 (SOD1) axis or cytochrome oxidase 17 (COX17) for detoxification of ROS and oxidative phosphorylation [7,157,159]. Additionally, intracellular ATP7A and ATP7B are responsible for exporting copper ions to prevent their accumulation in cells [7].
Nevertheless, abnormal systemic or intracellular copper metabolism, especially dysfunction of modulators, can cause the accumulation of extracellular divalent copper ions (Cu2+). Cu2+ can form a complex by binding with Elesclomol (ES) or Disulfiram (DSF) to internalize into the cytosol [154,160]. The cytosolic Cu2+ enters mitochondria and undergoes reduction from Cu2+ to the toxic monovalent copper ion (Cu+) via catalysis by ionophore ferredoxin 1 (FDX1) [154]. Significantly, Cu2+ is transported from the inner membrane of mitochondria into the mitochondrial matrix via solute carrier family 25 member 3 (SLC25A3), but the mechanism of how Cu2+ crosses from the outer membrane to the inner membrane of mitochondria remains unclear [161]. Cu+ can attach to fatty acylated proteins of the tricarboxylic acid (TCA) cycle, further inducing the aggregation of lipoylated proteins and depletion of iron-sulfur (Fe-S) proteins, resulting in protein toxicity, intracellular stress, mitochondrial shrinkage, membrane rupture, and, ultimately, cuproptosis [154,162]. Systemic and intracellular copper metabolisms, as well as cuproptosis, are described in Figure 2.

2.6. Other Forms of PCD

Mitochondrial permeability transition (MPT) was first described in 1976. MPT-driven necrosis is characterized as a unique form of programmed cell death (PCD), manifesting necrotic morphology, initiated by specific perturbations of the intracellular microenvironment such as severe oxidative stress and cytosolic calcium ion (Ca2+) overload [163]. MPT-driven necrosis relies on the activation of cyclophilin D (CYPD) and the formation of a supramolecular complex termed the permeability transition pore complex (PTPC) in the intermembrane space (IMS), leading to an abrupt loss of impermeability of the inner mitochondrial membrane (IMM) to small solutes and a rapid dissipation of mitochondrial potential [164,165]. The abnormality of mitochondria causes cell swelling, rupture of cell membranes, and eventual cell death [165]. However, the detailed mechanisms of MPT-driven necrosis need further investigation. Particularly, several factors, such as BAX, BAK, BID, dynamin 1-like (DRP1), and p53, can regulate MPT-driven necrosis by manipulating the activation of CYPD and PTPC formation, indicating crosstalk among intrinsic apoptosis, mitophagy, the cell cycle, and MPT-driven necrosis [1].
Autophagy is an evolutionarily conserved, intracellular, self-protective mechanism that maintains energy balance in response to nutrient stress in cells. The activation of autophagy can also degrade misfolded or aggregated proteins, damaged organelles, and invading pathogens, thereby maintaining intracellular homeostasis [166,167]. Generally, autophagy-dependent cell death (ADCD), first observed in 2006, is a distinct form of programmed cell death (PCD) that occurs due to the abnormal stimulation of autophagy in specific developmental or pathophysiological contexts and relies on the autophagic machinery or its components [168,169]. ADCD can be triggered by three distinct mechanisms: excessive ER-phagy, excessive mitophagy, and autosis [170]. The endoplasmic reticulum (ER) is the largest organelle in eukaryotic cells, responsible for the folding and trafficking of proteins that enter the secretory pathway by assembling a complex cell quality-control network [171]. Stress conditions, such as the unfolded protein response (UPR), lack of nutrients or oxygen, and pharmacologic stimuli, can induce ER-phagy by interacting with a series of ER-phagy receptors to remove damaged ER by delivering ER fragments to lysosomes [170,172]. However, the continuous degradation of ER fragments by ER-phagy can cause the excessive formation of autophagosomes, eventually leading to cell death [173,174]. Mitophagy is a conserved intracellular process that ensures mitochondrial quality and quantity control [175]. Mitophagy is initiated by specific mitochondrial outer membrane receptors interacting with proteins on the mitochondrial surface, leading to the formation of autophagosomes surrounding mitochondria to degrade damaged or depolarized mitochondria [176]. Nevertheless, excessive mitophagy results in an abnormal mitochondrial membrane potential and cell death [177]. Autosis was first observed by treating HeLa cells with the autophagy-specific activator BECN1-derived peptide (Tat-Beclin 1) [178]. Tat-Beclin 1 can directly cause the perturbation of sodium (Na+) and potassium (K+)-adenosine triphosphatase (ATPase) circulation, ultimately resulting in changes in autophagic flux and cell death [179]. Additionally, the process of autosis manifests in a time-dependent manner. During the early phase of autosis, the number of autophagosomes increases significantly while focal nuclear concavities occur. Subsequently, focal ballooning of the perinuclear space (PNS) and the disappearance of subcellular organelles appear in the later phase [180]. Additionally, autophagy is associated with numerous forms of PCD, such as apoptosis, necroptosis, ferroptosis, and cuproptosis [170,181].
Lysosomes are dynamic, single-membrane, and heterogeneous organelles that contain a wide variety of hydrolytic enzymes for the digestion of toxic intracellular components and damaged organelles, as well as for the termination of signal transduction [182]. Lysosome-dependent cell death (LDCD), first coined in 2000, is a form of programmed cell death (PCD) initiated by lysosomal membrane permeabilization (LMP), leading to the leakage of lysosomal contents into the cytosol, such as proteolytic enzymes of the cathepsin family and iron [5,183]. Subsequently, these leaked contents engage with apoptotic effectors, such as the p53 effector DNA damage-regulated autophagy modulator 1 (DRAM1), leading to mitochondrial outer membrane permeabilization (MOMP) and the activation of caspase-dependent signaling, eventually causing cell death [5].
Parthanatos, a poly(ADP-ribose) polymerase 1 (PARP1)-dependent form of programmed cell death (PCD), was first described in 2008 [184]. The accumulation of cytotoxic stimuli, such as oxidative stress, hypoxia, hypoglycemia, and inflammatory conditions, can cause PARP1 hyperactivation, resulting in the depletion of nicotinamide adenine dinucleotide (NAD+) and adenosine triphosphate (ATP), as well as the aggregation of polymers and poly(ADP-ribosyl)ated proteins at mitochondria, ultimately causing cell death due to mitochondrial membrane dissipation and mitochondrial outer membrane permeabilization (MOMP) [185].
Alkaliptosis, first unveiled in 2018, is caused by intracellular alkalinization, which involves the suppression of the NF-κB (nuclear factor κB)-carbonic anhydrase 9 (CA9) pathway and the ATP6V0D1-STAT3 pathway [186,187]. This dysregulation of intracellular pH results in cell death.
Oxeiptosis, first termed in 2018, is an oxygen radical-induced form of programmed cell death (PCD) initiated by the hyperactivation of the KEAP1-PGAM5-AIFM1 signaling cascade [188].
Disulfidptosis is a novel form of programmed cell death (PCD), defined in 2023, initiated by glucose starvation in cells with high SLC7A11 expression [189]. High uptake of cystine, coupled with a shortage of nicotinamide adenine dinucleotide phosphate (NADPH) supply, results in NADPH depletion, aberrant disulfide binding to actin cytoskeleton proteins, actin network collapse, and subsequent cell death [189].
NETs are extracellular net-like DNA–protein structures released by cells in response to various cellular stresses, including pathogen infections or injuries. They can also be formed by other leukocyte types, such as mast cells, eosinophils, and basophils, as well as epithelial cells and cancer cells [5]. The term NETosis was first coined in 2004 and was observed in neutrophils upon exposure to phorbol myristate acetate or interleukin 8 (IL-8), describing the process of NET generation [190]. At the molecular level, NETosis is a dynamic process involving multiple signaling pathways, such as NADPH oxidase-mediated ROS production, protein kinase C (PKC) isoform-initiated signaling cascades, autophagy, the release and translocation of granular enzymes, and the trafficking of N-GSDMD from the cytosol to the nucleus. This leads to various abnormal biological processes, including histone citrullination, chromatin decondensation, the destruction of the nuclear envelope, the release of chromatin fibers, and the formation of pores in the plasma membrane [191,192,193,194,195].
ENTosis, first introduced in 2007, is characterized by one cell inserting itself into a neighboring cell, a process termed the cell-in-cell (CIC) pattern, ultimately causing the death of the invading cell [196]. Glucose starvation, matrix deadhesion, and mitotic stress can induce ENTosis through cell adhesion and cytoskeletal rearrangement pathways [196,197,198,199,200]. Although the underlying mechanisms of ENTosis are not well understood, adhesion proteins such as cadherin 1 (E-cadherin), catenin alpha 1 (CTNNA1), and microtubules play a central role in the formation of adherent junctions between cells, leading to the generation of CIC structures and cell death [5]. The aforementioned forms of PCD are described in Figure 3 and Figure 4.

2.7. Characteristics of Different PCD Subroutines

Various PCD subroutines cause the disintegration of cells through distinct signaling cascades, resulting in differing morphological changes and immunological consequences. PCD can be classified into immunogenic cell death (ICD), also known as lytic forms of cell death, and tolerogenic cell death (TCD), also known as non-lytic forms of cell death [201,202]. ICD induces the activation of the immune system, whereas TCD does not provoke any inflammatory or immune reactions. ICD elicits acute or chronic inflammatory responses by releasing DAMPs from dead or dying cells into extracellular spaces. These DAMPs are subsequently recognized by pattern recognition receptors (PRRs) or other receptor systems expressed by neighboring macrophages and other bystander cells, triggering the release of proinflammatory cytokines [203]. Additionally, DAMPs play a fundamental role in regulating the balance between ICD and TCD [5]. ICD not only promotes tissue regeneration and organ development but also contributes to the progression of inflammation in numerous human diseases, especially neurodegenerative disorders. Therefore, agents that inhibit PCD may be critical components of future clinical therapeutic strategies. We summarize the immune and morphological hallmark features and major inhibitors of various PCD modalities in Table 2.

3. PCD in NDDs

3.1. PCD in AD

Alzheimer’s disease (AD), first described by Alois Alzheimer in 1906, is the primary cause of dementia. It has now become one of the most expensive, lethal, and burdensome diseases, with huge implications for individuals and society [207]. The onset of AD is relatively insidious, characterized by substantial progressive cognitive impairment and memory loss associated with age, impacting daily life functionality [208]. Amyloid precursor protein (APP) is widely present in the endoplasmic reticulum (ER) of neurons and glial cells, mediating neurotransmitter release, cell-to-cell adhesion, and neuronal signaling [209]. The cleavage of APP by α-secretase and γ-secretase produces non-toxic, soluble, and neuroprotective APPα peptides, whereas APP cleaved by β-secretase and γ-secretase produces neurotoxic amyloid β (Aβ) oligomers [210]. The aggregation of Aβ oligomers between nerve cells causes the formation of Aβ plaques, leading to neuronal cell death, particularly in the hippocampus [17]. Meanwhile, type 2 microtubule-associated protein (Tau) is expressed in neurons, astrocytes, and oligodendrocytes and is responsible for stabilizing microtubule structures by directly binding to them [211]. However, hyperphosphorylation of Tau due to abnormal post-translational modifications leads to the dissociation of microtubules and the aggregation of neurotoxic Tau proteins [212]. Aβ deposition and Tau aggregation facilitate the generation of neurofibrillary tangles (NFTs) in the cortex, leading to the progression of AD [213]. Generally, the formation of Aβ plaques, Tau aggregation, and NFT formation in neuronal cells are primary features of AD.
The effectors and signaling cascades of programmed cell death (PCD) play an essential role in the onset and progression of Alzheimer’s disease (AD). Specifically, the formation of the neurotoxic Aβ peptide can also be mediated by caspase-3 instead of β-secretase and γ-secretase, and members of the caspase superfamily can be activated by Aβ [214]. During NFT formation, anti-apoptotic factor expression is restrained, while proapoptotic protein levels are elevated via p53-dependent transcriptional upregulation [214,215]. Extracellular Aβ deposition can be recognized by apoptotic death receptors, leading to the activation of extrinsic apoptotic pathways [216]. Intracellular Aβ can insert into the mitochondrial outer membrane, leading to the formation of the mitochondrial permeability transition (MPT) and subsequent leakage of cytochrome c, causing mitochondrial apoptosis in neurons [192]. Aβ in the endoplasmic reticulum (ER) can also cause ER stress and initiate caspase-12/caspase-7-induced apoptosis in neurons [217]. Additionally, MAPK, JNK, BDNF-TrkB-CREB, JAK-STAT, PI3K-Akt-mTOR, and GSK-3β pathways are involved in the formation and aggregation of Aβ, as well as the hyperphosphorylation and aggregation of Tau through interactions with apoptotic signaling pathways [214]. Concurrently, the necroptotic RIPK1-RIPK3 complex facilitates the formation of Aβ structures, aiding in the translocation of Aβ to the cell surface and its aggregation [218]. Aβ plaques can stimulate microglia to secrete inflammatory factors, including TNF-α, thereby inducing the pyroptosis of neurons [219]. Hyperphosphorylated Tau can simultaneously activate necroptosis and the NF-κB pathway, contributing to the formation of NFTs and cytokine storm in microglia [220]. Similarly, Aβ and hyperphosphorylated Tau mediate the activation of the NLPR3-caspase 1-GSDMD axis and the release of caspase-1, IL-1β, and IL-8, leading to the activation of pyroptosis and formation of NFTs, ultimately causing chronic inflammation in microglia [221,222,223]. Notably, the effectors of ferroptotic antioxidant signaling cascades exhibit decreased expression in the neurons of AD patients, suggesting that ferroptosis may be a significant mechanism in AD [224]. Iron overload in the brain can exacerbate the production and accumulation of Aβ by enhancing the activity of β-secretase [225]. Moreover, Aβ can bind to ferrous iron and subsequently initiate lipid peroxidation and ferroptosis via the Fenton reaction [225]. Additionally, ferric iron can directly bind to Tau, causing its hyperphosphorylation and accumulation, along with increased expression of HO1 [226,227]. Comparatively, abnormal accumulation or deficiency of copper ions due to dysregulation of copper metabolism can also be observed in specific areas of the brain in AD patients [224]. A molar ratio of Cu2+ to Aβ oligomers of 0.25:1 can facilitate Aβ plaque formation [228] and can also be associated with the Tau R1 peptide to regulate Tau aggregation, while the Tau R2 peptide can reduce Cu2+ to Cu+ to induce cuproptosis [7]. Cu2+ can also trigger the NF-κB signaling pathway, increasing the release of inflammatory factors in microglia and impairing the brain’s ability to remove Aβ peptides by reducing the expression of lipoprotein receptor-related protein 1 (LRP1) [229,230]. Aβ oligomers in the mitochondria can activate CYPD by directly binding, causing mitochondrial perturbation and potentially activating MPT-driven necrosis in the neurons of the temporal cortex and hippocampus [231,232]. The accumulation of autophagosomes and lysosomes, along with higher expression of autophagy-related genes associated with increased levels of Aβ, has been observed in hippocampal CA1 pyramidal neurons and other neurons, eventually leading to cell death [233,234]. Aβ peptides in the hippocampus and microglia can also induce nitric oxide (NO) production, triggering DNA damage and PARP1 activation, potentially resulting in Parthanatos, neuroinflammation, and alterations in hippocampal synaptic integrity [235,236]. During the progression of AD, circulating neutrophils in the peripheral blood can be recruited to the vessel walls of the CNS via the LFA-1 β2 integrin–ICAM-1 complex released from cerebral endothelial cells. Subsequently, β2 integrins are activated, causing neutrophil arrest and the formation of NETs, resulting in damage to the blood–brain barrier, which is positively associated with Aβ depositions [237]. Figure 5 illustrates various PCD modalities in AD.

3.2. PCD in PD

Parkinson’s disease (PD), first described by James Parkinson in 1817, is a complex, progressive, and multisystem neurodegenerative disease with a range of causes and clinical presentations, elicited by the combined effects of environmental and genetic factors [238]. Clinical syndromes of PD include pathological motor features, such as a slowly progressive asymmetric resting tremor, cogwheel rigidity, and bradykinesia, as well as non-motor features, including anosmia, constipation, depression, sleep behavior disorder, autonomic dysfunction, pain, cognitive decline, and psychiatric symptoms [239]. α-synuclein (αSyn), consisting of amphipathic alpha-helical repeats, is an abundant neuronal protein enriched at synapses and mediating neurotransmission [240]. Pathologically, αSyn can undergo conformational changes capable of aggregation due to mutations in its encoding gene Alpha-synuclein (SNCA). Aggregated αSyn proteins then act as a major component of Lewy bodies and Lewy neurites, which are typical hallmarks of PD [241]. Significantly, Lewy bodies contain hundreds of other proteins and dysmorphic organelles, including lysosomes and mitochondria, packaged by abundant lipid membranes [242]. Additionally, the loss of dopamine neurotransmission due to the death of dopaminergic neurons projecting from the substantia nigra pars compacta to the caudate-putamen in the striatum is another leading cause of PD [241]. Furthermore, mutations in genes responsible for maintaining mitochondrial quality in neuronal cells, such as parkin RBR E3 ubiquitin protein ligase (PRKN), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1), and Parkinsonism-associated deglycase (PARK7), are associated with inherited PD [243].
In Parkinson’s disease (PD), intrinsic apoptosis is the predominant driver of dopaminergic neuronal death. Numerous pieces of evidence suggest that mutated PRKN, LRRK2, PINK1, and PARK7 localize in the outer mitochondrial membrane of Lewy body-positive neurons and can cause the disruption of mitochondrial outer membrane permeability (MOMP) and the formation of mitochondrial permeability transition (MPT), ensuing leakage of cytochrome c and activation of intrinsic apoptosis [243,244]. Additionally, observations of brain tissue from PD patients illustrate the abnormally increased expression of caspase 3 and BAX, along with reductions in Bcl-2 superfamily protein levels, which are associated with the upregulation of p53 [245,246,247]. The aggregation of αSyn in dopaminergic neurons can disrupt mitochondrial homeostasis, making dopaminergic neurons prone to apoptosis [248]. Meanwhile, microglial neuroinflammation in PD is induced by the formation of αSyn-derived NLRP3 inflammasomes and pyroptosis in dopaminergic neurons [249]. Mutated LRRK2 can also induce the activation of gasdermin D (GSDMD) to facilitate the release of reactive oxygen species (ROS) and necroptosis [250]. In the postmortem substantia nigra of individuals with PD, the expression of receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like protein (MLKL) is elevated, and mutated LRRK2 proteins are positively associated with the activation of MLKL and necroptosis [251,252]. Notably, the induction of ferroptosis is highly synchronized with the progression of PD. Ferric iron and αSyn coexist in the Lewy bodies of PD patients, and ferric iron is essential for the aggregation of αSyn [253,254]. Iron accumulation in the brain can activate microglia to release proinflammatory cytokines and cause oxidative stress, leading to the ferroptosis of dopaminergic neurons [255]. Comparatively, the N-terminal of αSyn contains a copper-binding site, and abnormally increased or decreased copper concentrations are associated with the progression of PD [7,256]. Moreover, an independent study illustrated that the ablation of cyclophilin D (CYPD) in PD-linked αSyn mutant transgenic mice delayed disease onset and extended lifespan, suggesting that CYPD may induce MPT-driven necrosis to regulate PD development [257]. The activation of autophagy and dysregulation of lysosomes in dopaminergic neurons can also be observed in PD patients [258,259]. The activation of poly (ADP-ribose) polymerase 1 (PARP1) can increase the neurotoxicity of αSyn by changing its conformation, inducing parthanatos [260]. The different PCD modalities in PD are comprehensively described in Figure 5.

3.3. PCD in HD

Huntington’s disease (HD), first described by George Huntington in 1872, is an autosomal dominantly inherited neurodegenerative disorder characterized by progressive motor, behavioral, and cognitive decline with high mortality [261]. The huntingtin (HTT) protein is present in spindle poles and microtubules, regulating cell division, ciliogenesis, endocytosis, transcription, vesicular transport, and autophagy [262]. The pathology of HD is monogenic and characterized by the production of mutant HTT proteins with an abnormally long polyglutamine repeat due to CAG trinucleotide repeat expansion in the HTT gene on chromosome 4, which eventually results in the aggregation of mutant huntingtin (mHTT) proteins in neurons and glial cells, particularly GABAergic and motor neurons [262]. As HD progresses, significant neuronal death can be observed in cortical, thalamic, and hypothalamic areas and even the entire brain, along with atrophy of the basal ganglia [263].
mHTT proteins can significantly facilitate the expression of pro-apoptotic factors, such as BIM and BAX, and apoptosis, while loss of BIM can decrease the amounts of mHTT proteins and neuronal cell death, indicating that pro-apoptotic factors potentially participate in the generation of mHTT proteins [264,265,266]. Additionally, cleaved caspase-3 can cleave mHTT proteins to produce more neurotoxic fragments that translocate into the nucleus and subsequently interact with different transcription factors, such as p53, eventually causing mitochondrial disruption, while wild-type (WT) HTT protein can prevent caspase-3 activation [267,268,269]. Meanwhile, mHTT proteins can cause mitochondrial dysfunction and the subsequent release of cytochrome c [270,271]. Moreover, a study confirms that mHTT proteins have the potential to activate receptor-interacting protein kinase 1 (RIPK1) and necroptosis in specific neurons, leading to neuroinflammation and the progression of HD [272]. Interestingly, the levels of caspase-1 and NLRP3 are intensely elevated in striatal spiny projection neurons and in parvalbumin interneurons, deteriorating the symptoms of HD [273]. Increased levels of toxic iron in the brain contribute to the HD process due to increased levels of reactive oxygen species (ROS) and depletion of glutathione peroxidase 4 (GPX4) in spinal motor neurons, showing the significant role of ferroptosis in HD progression [274,275]. Similarly, the abnormal elevation of copper concentrations in the brain contributes to the onset and progression of HD, and the wild-type (WT) HTT protein has two potential copper-binding residues [276]. Additionally, a similar scenario can be seen in HD, where the accumulation of mHTT is associated with attenuated autophagy [277]. Various PCD modalities in AD, PD, and HD are described in Figure 5. As shown in Figure 5, various PCD modalities are described in the context of HD.

3.4. PCD in ALS, SMA and MS

Amyotrophic lateral sclerosis (ALS), first described by Jean Martin Charcot in 1869, is a fatal neuromuscular disease characterized by progressive muscle weakness and atrophy due to the loss of both upper motor neurons (UMNs) and lower motor neurons (LMNs). This leads to patients experiencing dysphagia, dysarthria, and limb weakness, eventually dying from respiratory complications [278]. ALS can be classified into two types: familial ALS, which constitutes 10 to 15% of cases and is inherited, and sporadic ALS, which constitutes the remaining (approximately 85%) of cases [12]. Pathologically, ALS-associated genetic signatures vary in frequency, but the most common mutations occur in four genes: chromosome 9 open reading frame 72 (C9ORF72), TAR DNA-binding protein (TARDBP), superoxide dismutase 1 (SOD1), and fused in sarcoma/translocated in liposarcoma (FUS). These mutations can impair various intracellular functions and form protein aggregations, which accelerate UMN and LMN loss and contribute to the onset and progression of most ALS cases [279]. Spinal muscular atrophy (SMA), first identified by William R. Kennedy in 1966, is an autosomal recessive, progressive, and lethal neuromuscular disorder characterized by the degeneration of alpha motor neurons in the spinal cord [280].
Spinal muscular atrophy (SMA) can be clinically classified into four grades of severity (SMA I, SMA II, SMA III, and SMA IV) according to age of onset and motor function achieved [281]. Clinical hallmarks of SMA include muscular dystrophy, fasciculations, altered reflexes, joint contractures, dysphagia, dysarthria, and respiratory complications [282]. Survival motor neuron (SMN) protein is involved in the biogenesis of small nuclear RNA (snRNA) and ribonucleoproteins (snRNPs), which act as major components of the pre-mRNA splicing machinery [283]. More than 95% of SMA cases exhibit a homozygous deletion or point mutation in exon 7 of the SMN1 gene, leading to the loss of SMN production [284]. Additionally, SMN2, a paralogous copy of SMN1, has a single-nucleotide difference in exon 7 compared with SMN1, resulting in the alternative splicing of exon 7 in transcripts and the expression of only 5–10% of full-length functional SMN protein [285]. The copy number of SMN2 is inversely proportional to the age of onset and severity of SMA [286].
Multiple sclerosis (MS) is an immunological disease and a neurodegenerative condition that causes chronic inflammation and acute inflammatory lesions in the central nervous system (CNS), eventually resulting in tissue damage and disability [287]. MS often occurs in young populations, and the clinical manifestations of MS patients are highly variable, including optic neuritis, weakness or changes in sensation in the body, dizziness, memory or cognitive impairment, dysregulation of bladder control, and depression or anxiety [288]. MS can be divided into three representative types according to the onset of recurring clinical symptoms followed by total or partial recovery: relapsing–remitting MS (RRMS), primary progressive MS (PPMS), and secondary progressive MS (SPMS). These types are influenced by factors, such as low uptake of vitamin D, low levels of sunlight exposure, Epstein-Barr virus (EBV) infection, and genetic predisposition [288]. The two pathologic hallmarks of MS are axonal degeneration and neuronal cell death, which are induced by oxidative stress and mitochondrial dysfunction in active MS lesions [289,290].
Research has discovered that the expression of pro-apoptotic factors and the caspase superfamily is significantly elevated in the motor neurons of ALS and SMA patients, along with abnormally low levels of anti-apoptotic factors [291,292,293]. Meanwhile, apoptosis is widely induced in astrocytes, microglia, oligodendrocytes, and neurons in MS lesions, contributing to the progression of MS [294]. Specifically, mutant SOD1 can bind to the Bcl-2 anti-apoptotic factor, suppressing its activity [295]. Mutations in a highly conserved region of TARDBP can cause the formation of mutant forms of TAR DNA-binding protein 43 (TDP-43), which can induce neural apoptosis [296]. Loss of SMN contributes to the activation of p53 and JNK signaling pathways, subsequently inducing apoptosis [297,298]. Additionally, the expression of RIPK1 and RIPK3, as well as the formation of necrosomes, is enhanced in pathological tissues of SOD1 (G93A) ALS transgenic mice [299,300]. A similar scenario is observed in the neuronal cells of cortical lesions in the human MS brain [301]. Furthermore, an independent study identified that the knockout (KO) of RIPK3 can significantly increase the survival and motor function of SMN deletion mice [302].
The elevated formation of inflammasomes, expression of IL-1β and IL-18, and excessive cleavage of GSDMD have been reported in amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) cases, indicating a crucial role for pyroptosis in the neuroinflammation and development of ALS [67,303,304]. Studies have also suggested that dysregulation of iron and copper homeostasis, as well as excessive reactive oxygen species (ROS) production, can be observed in the brains of ALS and MS patients, which may induce ferroptosis and cuproptosis, leading to neuronal damage [17,305,306,307,308]. Furthermore, several studies have revealed that cyclophilin D (CYPD) and mitochondrial permeability transition (MPT)-driven necrosis may contribute to the pathogenesis of ALS and MS. For example, mutant SOD1 can interact with CYPD to cause CYPD hyperactivation, thereby inducing the formation of mitochondrial permeability transition pore (mPTP) [309]. The activation of CYPD and formation of mPTP can also be observed in the axonal damage occurring during MS, which weakens the resistance to reactive oxygen and nitrogen species, thereby mediating axonal damage [310]. In addition, FUS can be recruited by PARP-1, activated by DNA damage, to stimulate the synthesis of long poly (ADP-ribose) (PAR) chains, indicating a role for parthanatos in ALS [311]. C9ORF72 deficiency or mutations in the brains of ALS patients may exacerbate the accumulation of DNA damage and PARP1 overactivation, leading to the activation of parthanatos [312]. Excessive PARP1 hyperactivation and parthanatos can also be detected in oligodendrocytes, astrocytes, and microglia or macrophages in the active areas of brain lesions in MS patients [313]. The abnormal accumulation of autophagosomes in the neuronal cells of ALS, SMA, and MS patients has been reported. The hallmark of autophagy-dependent cell death (ADCD) is the excessive formation of autophagosomes, revealing that ADCD is another form of neuronal death in the context of ALS [314,315,316]. The activation of neutrophils and the subsequent formation of neutrophil extracellular traps (NETosis) are elevated during the occurrence and progression of ALS and MS [317,318,319]. We describe various PCD modalities in ALS, SMA, and MS in Figure 6.

3.5. PCD in TBI and Stroke

Traumatic brain injury (TBI) refers to a physical injury caused by an external mechanical force, which induces transitory or permanent damage to brain tissues [320]. The pathophysiology of TBI involves both primary and secondary injury mechanisms. Primary injury occurs at the moment of impact, causing immediate damage to brain tissue or brain structures. Secondary injury mechanisms involve a neurodegenerative process manifesting from hours to days following the initial trauma, inducing chronic inflammation and neuron loss [320]. The secondary mechanism exacerbates the initial damage caused by the primary injury, and clinical presentations of secondary TBI include a combination of cognitive, emotional, and behavioral changes [321]. Stroke is a chronic neurodegenerative disease caused by insufficient blood supply to the brain. It can be divided into two major types: ischemic stroke, caused by occlusion of carotid and vertebral arteries, and hemorrhagic stroke, caused by subarachnoid or intraparenchymal hemorrhage [322,323]. Notably, ischemic strokes account for the majority of all stroke cases [324]. Common clinical symptoms of stroke include sudden weakness or numbness on one side of the body, aphasia, dysphasia, dizziness, and severe headache [325]. The primary pathophysiology of stroke involves the damage and death of brain cells due to the interruption of blood flow to the brain, which restrains the oxygen and nutrients reaching brain cells [326].
Generally, programmed cell death (PCD) plays an important role in the later stages of traumatic brain injury (TBI) and stroke progression, impacting the recovery of brain tissue and neurological function. Specifically, the formation of apoptotic bodies can be observed in post-ischemic stroke neurons, and apoptosis in neurons of the ischemic penumbra may be recoverable [327,328]. Additionally, neurons alter the glucose metabolism pathway from aerobic oxidation to anaerobic oxidation to deal with glucose starvation during stroke, leading to a lower production of ATP [329]. This change can impair Na+/Ca2+ influx and K+ efflux, causing the accumulation of intracellular Ca2+ and calpain activation, which leads to the cleavage of the anti-apoptotic protein Bcl-2 and intrinsic apoptosis [330]. Importantly, glucose starvation initiates disulfidoptosis, and research has discovered that disulfidoptosis-related genes (DRGs) are significantly associated with stroke in immune cells from peripheral blood samples of stroke patients [331,332]. Moreover, DNA damage-associated activation of the p53 signaling pathway and large amounts of ROS generation also contribute to the activation of intrinsic apoptosis during stroke progression [333]. Additionally, in the early stages of ischemic stroke, the activation of immune cells, such as microglia, can release TNFα and FasL, engaging death receptors to initiate extrinsic apoptosis [334,335]. Similarly, Ca2+ overload, DNA damage, excessive ROS generation, and activation of immune cells can also be observed in the CNS of TBI patients, activating intrinsic and extrinsic apoptosis of neuronal cells [336]. However, necroptosis may occur even if the apoptotic signal is suppressed during stroke and TBI progression. Research has identified that the secretion of TNF-α, TRAIL, and FasL by microglial cells can be recognized by death receptors on neurons, triggering RIPK1-RIPK3-MLKL signaling cascade activation and necrosome formation [45]. Meanwhile, microglial cells also secrete proinflammatory cytokines, such as IL-1β, causing caspase-1-mediated pyroptosis in neuronal cells under TBI and stroke conditions [337,338]. Additionally, iron accumulation and lipid peroxidation can be observed in multiple areas of the brains of TBI and stroke patients due to the loss of antioxidant signaling cascades [333,339]. Similarly, the disruption of copper homeostasis caused by insufficient ATP generation can also be observed in the CNS of TBI and stroke patients, and cuproptosis-related genes regulate immune infiltration in ischemic stroke [340,341,342]. Moreover, hyperactivation of PARP1 and depletion of NAD+ and ATP can be observed in stroke and TBI mouse models, triggering parthanatos in neurons [343,344]. Meanwhile, ATP depletion in neurons of TBI and stroke mouse models can also induce the activation of CYPD-dependent MPT-driven necrosis, while hypoxia-induced activation of p53 can interact with CYPD to exert an anti-angiogenic effect in the brain after ischemic stroke [345,346,347]. Additionally, excessive activation of the hypoxia-inducible factor 1α (HIF-1α) signaling pathway and ER stress mTOR signaling pathways can be observed in microglia and neurons in the brains of stroke patients, subsequently activating ADCD [323]. Similarly, excessive mitophagy and ER-phagy, as well as autosis, can also be observed in immune cells and neurons in the brains of TBI patients [348]. LDCD of endothelial cells can also be detected in stroke patients, potentially causing damage to the blood–brain barrier (BBB) [15]. Additionally, the release of high-mobility group box 1 (HMGB1) by platelets can facilitate NETosis in the acute phase of stroke, exacerbating disease progression [349]. During stroke development, NETosis generated by neutrophils exacerbates neuroinflammation and impairs revascularization and vascular remodeling after stroke due to the upregulation of peptidylarginine deiminase 4 (PAD4) [350]. NETosis also contributes to coagulopathy and neuroinflammation after TBI through the release of HMGB1 by platelets and the formation of neutrophil–platelet aggregates [351,352]. Various PCD modalities in stroke and TBI are illustrated in Figure 7.

3.6. Therapeutic Strategies Targeting PCD Signaling Pathways in NDDs

Following this review, it is evident that multiple programmed cell death (PCD) pathways collectively play a role in neurodegenerative diseases (NDDs). The regulation of PCD occupies a significant position in the complex pathogenesis of NDDs. As research continues to uncover the mechanisms underlying disease progression, our understanding of targeting PCD to modulate NDDs has deepened. Specifically, numerous key factors involved in PCD pathways have been identified as the most direct and promising targets for therapeutic development. These include genes and proteins, such as the caspase family, apoptosis-related factors, necrosome-related factors, and inflammasome-related factors, each of which directly influences PCD processes in NDDs. Furthermore, the development of PCD pathways in NDDs is often accompanied by various associated biological phenomena. Strategies targeting processes such as oxidative stress, neuroinflammation, and metabolic imbalance also hold great promise and therapeutic value for disease treatment. This review discusses the targets and strategies for treating NDDs by focusing on PCD pathways.
As major participants in cell death pathways, the caspase family is extensively involved in both apoptotic and non-apoptotic cell death processes. Inhibiting the caspase family to interrupt PCD processes mediated by them has been demonstrated in numerous studies to be beneficial for improving and treating symptoms of NDDs. Caspases with short prodomains (Caspase-3, -6, and -7) are known as effectors of apoptosis, while those with long prodomains can be further classified into initiators of apoptosis (Caspase-2, -8, -9, and -10) and inflammatory caspases that cleave cytokines (Caspase-1, -4, -5, -11, and -12) [353]. Caspase-2 may function as both an initiator and an effector of apoptosis [353]. The expression level of caspase-3 is significantly higher in Alzheimer’s disease (AD) patients compared to age-matched controls. Inhibiting caspase-3 activity can alleviate Alzheimer-like phenotypes in transgenic mice, and pharmacological experiments in AD models have demonstrated clinical improvements that mitigate symptoms [354,355,356,357]. Furthermore, inhibiting the JAK-STAT-caspase-3 axis to prevent neurodegenerative diseases has also been reported [214,358]. Caspase-6 is another factor considered a potential diagnostic marker and therapeutic target for Huntington’s disease (HD) patients. Enhancing the insulin-like growth factor 1 (IGF-1) signaling pathway reduces HTT toxicity changes associated with increased caspase-6 activation, and IGF-1 treatment has demonstrated therapeutic benefits in HD mouse models [356,359,360,361]. Additionally, the co-expression of caspase-2 and Bcl-2-interacting mediator of cell death (Bim) has been observed in neurons of AD brains [362]. Studies have shown that caspase-2 deficiency can ameliorate spine density reduction in the J20 mouse model of AD, preventing behavioral changes in these mice [363]. In Alzheimer’s disease and related dementias (ADRD), Tau is cleaved by caspase-2, producing Δtau314, which promotes Tau mislocalization and accumulation (Casp2/tau/Δtau314). Inhibiting caspase-2 as a drug target can ameliorate synaptic dysfunction in ADRD [364]. The inhibitor of the apoptosis protein (IAP) family, as endogenous inhibitors of the caspase family, suppresses cell death by directly acting on caspases or serving as targets for protein degradation [353,365]. In spinal muscular atrophy (SMA), neuron-specific IAP family members NAIP and XIAP effectively block the enzymatic activity of group II caspases (3 and 7) and reduce the expression levels of cleaved-caspase-3, thereby protecting spinal motor neurons (MNs) and preventing severe SMA [366,367]. In a mouse stroke model, early ischemic activation of the apoptotic pathway in the striatum is associated with caspase-9 activation. Treatment with the endogenous caspase-9 inhibitor XIAP-BIR3 has been shown to protect neuroanatomy and function in the disease model [353]. As critical anti-apoptotic proteins, Bcl-2 family proteins exhibit suppressed expression across various NDDs, profoundly influencing the progression of these conditions. The practical role and physiological significance of targeting Bcl-2 family proteins to regulate the onset and development of NDDs have been extensively documented in numerous studies [368].
Strategies employing Bcl-2 family-mediated apoptosis to treat amyotrophic lateral sclerosis (ALS) have been validated in ALS mouse models; the knockout of two Bcl-2 proteins, Bax and Bak, counteracts the toxic effects of mutant superoxide dismutase 1 (SOD1) by inhibiting the activation of pro-caspase-3, thereby preventing neuronal damage [369,370]. Additionally, in HD, huntingtin (HTT) enhances the activity of caspase-8 and calpain, leading to the cleavage of full-length Bid. In the superior cervical ganglion (SCG), mutant HTT induces Bax-independent cell death [264,371,372].
Inflammation and oxidative stress are pervasive in NDDs and PCD, serving both as triggers and concomitant biological phenomena. These processes play a central role in the pathogenesis of NDDs as critical pathophysiological features [373,374,375,376]. The caspase family, which initiates apoptosis, is not only involved in regulating cell death but also plays a crucial role in modulating neuroinflammation in PCD. Numerous studies have demonstrated that inhibiting caspase-1 alleviates PD symptoms by suppressing neuroinflammation [377,378,379,380]. Research indicates that the NLRP3/caspase-1 axis and the gasdermin (GSDM) family represent substantial interactions between neuroinflammation and the initiation of apoptosis [381]. Inhibiting the NLRP3/caspase-1 axis reduces amyloid-beta deposition in the AD APP/PS1 model, and disease models in Nlrp3−/− or Casp1−/− mice also exhibit symptom relief [381]. Additionally, the therapeutic potential of inhibiting p38 through the suppression of the NLRP3 inflammasome pathway has been shown. p38 inhibitors such as SB203580 and NLRP3 inhibitors like MCC950 not only prevent neurodegeneration in vivo but also alleviate motor deficits in the α-Syn-A53T transgenic mouse model of PD [379]. Beyond these two inhibitors, Celastrol and the small-molecule kaempferol (Ka) also mitigate PD symptoms via the same pathway, showing similar trends in multiple sclerosis (MS) studies [377,378,382]. The overactivation of the NLRP3 inflammasome impairs microglial autophagy, exacerbating neurodegenerative disease mechanisms, thus supporting the application of microglial autophagy inducers and NLRP3 inhibitors [383]. In amyotrophic lateral sclerosis, knockdown of GSDME reduces neuroinflammation and rescues the loss of motor neurons derived from patient-induced pluripotent stem cells (iPSCs). Deleting GSDME in SOD1G93A ALS mice also shows effective therapeutic outcomes [377,384].
Furthermore, the regulation of oxidative stress in cell death pathways has been characterized in NDDs. The post-transcriptional regulator hsa-miR-4639-5p of PARK7 reduces PARK7 protein levels when upregulated, exacerbating oxidative stress and leading to neuronal death [385]. Silencing transcription factor RE1-silencing transcription factor (REST) and REST-dependent epigenetic remodeling have been reported to inhibit genes associated with oxidative stress and β-amyloid toxicity, thereby preventing neuronal death, which also plays a crucial role in other NDDs [386,387]. During acute neuronal insult events, hydrogen sulfide (H2S) acts as an antioxidant, anti-inflammatory, and anti-apoptotic mediator, protecting neurons from secondary neuronal damage [388]. In traumatic brain-penetrating injury (PTBI), a significant increase in reactive oxygen species (ROS) and reactive nitrogen species (RNS) production and elevated oxidative stress markers are observed, ultimately leading to cell death. Adjusting abnormal oxidative stress levels offers important insights for disease treatment and target development [389].
Microglial cells, which serve as critical sites for ion metabolism and neuroinflammation, contain various ion channels, including potassium (K+), calcium (Ca2+), chloride (Cl), sodium (Na+), and proton (H+) channels. These ion channels are responsible for the dynamic characteristics of brain immune cells and play essential roles in regulating microglial proliferation, chemotaxis, phagocytosis, antigen recognition and presentation, apoptosis, and inflammatory cell signaling [376]. In neurodegenerative diseases, oxidative stress, energy metabolism disorders, and disease-related protein alterations lead to Ca2+-dependent synaptic dysfunction, impaired plasticity, and neuronal death. Dysregulation of the Bcl-2-Ca2+ signaling axis has been associated with the progression of AD. Targeting Ca2+ checkpoints, such as G protein-coupled receptors, ion channels, Ca2+-binding proteins, transcription networks, and ion exchangers, to maintain Ca2+ homeostasis may represent novel therapeutic targets [389,390,391]. In addition, the dysregulation of metal metabolism, including iron (Fe) and copper (Cu), also contributes to the regulation of NDDs and the induction of cell death.
Therefore, we discussed the use of metal chelators related to ferroptosis and cuproptosis [224,225,226,227,228]. Studies have shown that Liproxstatin-1, a specific inhibitor of ferroptosis, can prevent amyloid-beta (Aβ)-induced neuronal death and memory loss [392]. Deferoxamine (DFO), an iron chelator, has demonstrated beneficial effects in clinical trials for AD patients and improves cognitive deficits induced by iron overload in APP/PS1 transgenic mice by inhibiting the processing of amyloid precursor protein (APP) [393,394,395]. Specifically, metal–protein attenuating compounds (MPACs) inhibit Aβ production and toxicity formation by chelating Cu or zinc (Zn) ions, showing efficacy in multiple clinical trials by slowing disease progression and improving cognitive function [396,397,398,399]. Additionally, Fe chelators, such as epigallocatechin-3-gallate (EGCG), and Cu chelators, such as amentoflavone, play significant roles in regulating neuroinflammation and the progression of NDDs [400,401,402].
The pathways of PCD in NDDs represent a complex process regulated by multiple factors. Beyond targeting key elements responsible for executing functions as therapeutic targets, it is crucial to focus on the balance and coordination of multiple pathways. The normal function and balance of autophagy and apoptosis are vital for neuronal homeostasis, and their dysfunction leads to the onset of neurodegenerative diseases. Regulation of some autophagy and apoptosis modulators must be carried out cautiously to avoid excessive autophagy, which can lead to cell death, or excessive inhibition of apoptosis, which can result in the accumulation of toxic substances [403]. In AD, the c-Jun N-terminal kinase (JNK) pathway is upregulated, leading to a reduction in anti-apoptotic protein expression and triggering Janus kinase-signal transducer and activator of transcription (JAK-STAT)/caspase-3 axis-mediated apoptosis. Meanwhile, the phosphoinositide 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) pathway regulates the imbalance between autophagy and apoptosis, and this balancing act also impacts PD, HD, frontotemporal dementia (FTD), and ALS [214,404,405].
Consequently, mTOR inhibitors, such as rapamycin, have shown broad prospects in targeting PCD for the treatment of NDDs [406,407]. Moreover, the upregulation of autophagy can extend the lifespan of HD mice by clearing aggregates containing HTT [408]. Autophagy defects and autophagosome accumulation are attributed to Beclin-1. Beclin-1 serves as a molecular platform for initiating autophagosome formation, and its interaction with anti-apoptotic protein Bcl-2 or inflammasomes leads to autophagy dysfunction and promotes the onset of PD [409,410]. Studies have revealed that the autophagy adaptor protein p62 (sequestosome 1, SQSTM1) mediates the degradation of survival motor neuron (SMN) through interaction, resulting in reduced autophagosome clearance and overactivation of mTOR complex 1 (mTORC1) signaling in spinal muscular atrophy (SMA) neurons [411]. Lowering p62 levels significantly enhances the therapeutic effects for SMA [411].

4. Conclusions

Since the discovery of neurodegenerative diseases (NDDs), significant progress has been made in component identification, understanding pathogenesis, development, treatment, and regulation. Current drug development and treatment strategies are based on the exploration and understanding of these mechanisms. As the role of programmed cell death (PCD) in the mechanistic network of NDDs has become clearer, we have gained a deeper understanding of these diseases, creating more possibilities for mechanistic research and clinical treatment. However, breakthroughs based on mechanistic research have been limited, and the translation to clinical progress has encountered bottlenecks. Firstly, in terms of mechanistic exploration, the challenges lie in the insufficient depth of overall research, limited individual research directions and progress, and the inability to integrate multi-spatial, multi-omics, and multi-pathway approaches to establish a comprehensive interpretation. We still need more experimental methods to elucidate the specific roles and dynamic changes of key factors such as the caspase family and B-cell lymphoma 2 (Bcl-2) family in the PCD pathways of diseases and to develop targeted drugs and strategies. Secondly, in terms of clinical development, there is a lack of sufficiently realistic experimental models. Disease models, constrained by research progress, can only replicate relatively singular phenotypes, and there is also a lack of validation of the reliability of existing research findings. Additionally, drug clinicalization faces challenges such as a shortage of development ideas, severe homogenization within the same category, and limitations on drug delivery pathways imposed by the blood–brain barrier. Given these issues, it is crucial to conduct comprehensive assessments of feasibility and biosafety during research to ensure that the final clinical strategies and drugs meet contemporary pharmacokinetic and biosafety standards.
In summary, our review first elucidates the current research on cell death mechanisms by detailing apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other forms of PCD. Furthermore, we discuss the roles and processes of various PCD pathways in regulating networks within NDDs, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Finally, we briefly introduce disease treatment strategies and approaches targeting key factors in these pathways, including inflammation, oxidative stress, and metabolic balance, thereby establishing a common link between these complex biological processes. Through these three aspects, we preliminarily construct the relationship network between PCD and NDDs, which deepens our understanding of the corresponding mechanisms and identifies potential therapeutic targets for development. Our discussion leads to the following conclusions: (1) The regulation of NDDs by PCD involves the collaborative interaction of multiple factors and pathways. (2) Based on the common understanding of multiple systems, we can overcome the limitations of focusing on single research factors and drug choices, allowing for new experimental attempts. (3) The practical significance of this review lies in providing a reference for the development of therapeutic targets and strategies through elucidation from different perspectives.
As a recognized challenge in human disease, the treatment of NDDs has consistently faced significant obstacles. Based on the current research progress, we are still unable to achieve a complete cure for these diseases, and our ability to prevent their onset remains limited. At best, we can only slow disease progression or alleviate symptoms. Nevertheless, we anticipate that in the near future, the development of drugs targeting PCD for the treatment of NDDs and their clinical application will help improve the condition of patients with neurodegenerative diseases. Despite numerous challenges, we have taken a significant step forward in humanity’s battle against these diseases. We hope to make the greatest possible contribution to curing these diseases within the limits of our current capabilities.

Author Contributions

D.G. and Z.L. contributed equally to this article. They drafted the original manuscript and prepared the figures. J.Z. conducted the literature search and contributed to the writing of the paper. C.K. and D.L. proposed the research design, conducted literature searches, and contributed to the writing of the paper. Additionally, C.K. and D.L. provided professional advice and revisions. All authors have read and agreed to the published version of the manuscript.

Funding

Joint Funds for the innovation of science and Technology, Fujian province (grant number: 2023Y9415).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We thank Figdraw (https://www.figdraw.com/ (accessed on 26 July 2024)) for expert assistance in the pattern drawing.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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]
  2. 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]
  3. 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]
  4. 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]
  5. 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]
  6. Liu, X.; Zhuang, L.; Gan, B. Disulfidptosis: Disulfide stress-induced cell death. Trends Cell Biol. 2024, 34, 327–337. [Google Scholar] [CrossRef]
  7. Chen, L.; Min, J.; Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 2022, 7, 378. [Google Scholar] [CrossRef]
  8. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
  9. 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]
  10. 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]
  11. 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]
  12. 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]
  13. 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]
  14. Owens, B. Multiple sclerosis. Nature 2016, 540, S1. [Google Scholar] [CrossRef]
  15. 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]
  16. 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]
  17. 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]
  18. 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]
  19. 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]
  20. 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]
  21. 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]
  22. 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]
  23. 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]
  24. 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]
  25. 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]
  26. 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]
  27. 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]
  28. Danial, N.N.; Korsmeyer, S.J. Cell death: Critical control points. Cell 2004, 116, 205–219. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, X. The expanding role of mitochondria in apoptosis. Genes. Dev. 2001, 15, 2922–2933. [Google Scholar]
  30. 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]
  31. 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]
  32. 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]
  33. 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]
  34. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
  35. 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]
  36. 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]
  37. 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]
  38. 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]
  39. Fulda, S.; Debatin, K.M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef]
  40. 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]
  41. Green, D.R. Apoptotic pathways: Paper wraps stone blunts scissors. Cell 2000, 102, 1–4. [Google Scholar] [CrossRef] [PubMed]
  42. 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]
  43. Yan, J.; Wan, P.; Choksi, S.; Liu, Z.G. Necroptosis and tumor progression. Trends Cancer 2022, 8, 21–27. [Google Scholar] [CrossRef]
  44. Tummers, B.; Green, D.R. Caspase-8: Regulating life and death. Immunol. Rev. 2017, 277, 76–89. [Google Scholar] [CrossRef] [PubMed]
  45. Yuan, J.; Amin, P.; Ofengeim, D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat. Rev. Neurosci. 2019, 20, 19–33. [Google Scholar] [CrossRef]
  46. 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]
  47. 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]
  48. 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]
  49. 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]
  50. 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]
  51. 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]
  52. 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]
  53. 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]
  54. 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]
  55. 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]
  56. 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]
  57. Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef]
  58. 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]
  59. 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]
  60. 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]
  61. 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]
  62. 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]
  63. 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]
  64. 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]
  65. 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]
  66. 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]
  67. 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]
  68. 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]
  69. Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef]
  70. 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]
  71. 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]
  72. 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]
  73. 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]
  74. 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]
  75. 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]
  76. 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]
  77. 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]
  78. 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]
  79. 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]
  80. 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]
  81. 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]
  82. 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]
  83. 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]
  84. 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]
  85. 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]
  86. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef]
  87. 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]
  88. 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]
  89. 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]
  90. Zheng, J.; Conrad, M. The Metabolic Underpinnings of Ferroptosis. Cell Metab. 2020, 32, 920–937. [Google Scholar] [CrossRef]
  91. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef] [PubMed]
  92. 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]
  93. 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]
  94. 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]
  95. 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]
  96. 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]
  97. 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]
  98. 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]
  99. 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]
  100. 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]
  101. 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]
  102. 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]
  103. 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]
  104. 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]
  105. 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]
  106. 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]
  107. 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]
  108. 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]
  109. 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]
  110. 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]
  111. 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]
  112. 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]
  113. 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]
  114. 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]
  115. 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]
  116. 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]
  117. 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]
  118. 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]
  119. 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]
  120. 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]
  121. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. 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]
  124. 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]
  125. 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]
  126. 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]
  127. 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]
  128. 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]
  129. 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]
  130. 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]
  131. 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]
  132. 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]
  133. 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]
  134. 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]
  135. 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]
  136. 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]
  137. 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]
  138. Liu, J.; Kang, R.; Tang, D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022, 289, 7038–7050. [Google Scholar] [CrossRef]
  139. 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]
  140. 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]
  141. 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]
  142. 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]
  143. Hassannia, B.; Vandenabeele, P.; Vanden Berghe, T. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef]
  144. 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]
  145. 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]
  146. 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]
  147. 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]
  148. 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]
  149. 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]
  150. 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]
  151. 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]
  152. 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]
  153. 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]
  154. 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]
  155. Xie, J.; Yang, Y.; Gao, Y.; He, J. Cuproptosis: Mechanisms and links with cancers. Mol. Cancer 2023, 22, 46. [Google Scholar] [CrossRef]
  156. 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]
  157. Lutsenko, S. Copper trafficking to the secretory pathway. Metallomics 2016, 8, 840–852. [Google Scholar] [CrossRef] [PubMed]
  158. 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]
  159. Lutsenko, S.; Bhattacharjee, A.; Hubbard, A.L. Copper handling machinery of the brain. Metallomics 2010, 2, 596–608. [Google Scholar] [CrossRef]
  160. 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]
  161. 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]
  162. 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]
  163. 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]
  164. 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]
  165. 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]
  166. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [PubMed]
  167. 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]
  168. Denton, D.; Kumar, S. Autophagy-dependent cell death. Cell Death Differ. 2019, 26, 605–616. [Google Scholar] [CrossRef]
  169. 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]
  170. Liu, S.; Yao, S.; Yang, H.; Liu, S.; Wang, Y. Autophagy: Regulator of cell death. Cell Death Dis. 2023, 14, 648. [Google Scholar] [CrossRef]
  171. 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]
  172. Gubas, A.; Dikic, I. ER remodeling via ER-phagy. Mol. Cell 2022, 82, 1492–1500. [Google Scholar] [CrossRef] [PubMed]
  173. 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]
  174. 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]
  175. 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]
  176. 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]
  177. 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]
  178. 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]
  179. 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]
  180. 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]
  181. 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]
  182. Yang, C.; Wang, X. Lysosome biogenesis: Regulation and functions. J. Cell Biol. 2021, 220, e202102001. [Google Scholar] [CrossRef] [PubMed]
  183. Aits, S.; Jaattela, M. Lysosomal cell death at a glance. J. Cell Sci. 2013, 126, 1905–1912. [Google Scholar] [CrossRef]
  184. 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]
  185. 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]
  186. 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]
  187. 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]
  188. 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]
  189. 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]
  190. 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]
  191. 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]
  192. 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]
  193. 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]
  194. 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]
  195. 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]
  196. 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]
  197. 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]
  198. 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]
  199. 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]
  200. 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]
  201. 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]
  202. Green, D.R.; Ferguson, T.; Zitvogel, L.; Kroemer, G. Immunogenic and tolerogenic cell death. Nat. Rev. Immunol. 2009, 9, 353–363. [Google Scholar] [CrossRef]
  203. 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]
  204. 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]
  205. Mishima, E.; Conrad, M. Nutritional and Metabolic Control of Ferroptosis. Annu. Rev. Nutr. 2022, 42, 275–309. [Google Scholar] [CrossRef]
  206. 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]
  207. 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]
  208. 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]
  209. 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]
  210. Jucker, M.; Walker, L.C. Alzheimer’s disease: From immunotherapy to immunoprevention. Cell 2023, 186, 4260–4270. [Google Scholar] [CrossRef]
  211. 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]
  212. 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]
  213. 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]
  214. 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]
  215. 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]
  216. 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]
  217. 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]
  218. 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]
  219. 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]
  220. 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]
  221. 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]
  222. 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]
  223. 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]
  224. 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]
  225. 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]
  226. 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]
  227. 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]
  228. 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]
  229. 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]
  230. 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]
  231. 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]
  232. 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]
  233. 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]
  234. 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]
  235. 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]
  236. 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]
  237. Pietronigro, E.C.; Della Bianca, V.; Zenaro, E.; Constantin, G. NETosis in Alzheimer’s Disease. Front. Immunol. 2017, 8, 211. [Google Scholar] [CrossRef]
  238. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  239. 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]
  240. 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]
  241. 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]
  242. 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]
  243. Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef] [PubMed]
  244. 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]
  245. 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]
  246. 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]
  247. 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]
  248. 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]
  249. 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]
  250. 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]
  251. 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]
  252. 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]
  253. 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]
  254. 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]
  255. 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]
  256. 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]
  257. 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]
  258. 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]
  259. 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]
  260. 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]
  261. 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]
  262. 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]
  263. 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]
  264. 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]
  265. 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]
  266. 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]
  267. Erekat, N.S. Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin. Anat. 2022, 35, 65–78. [Google Scholar] [CrossRef]
  268. 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]
  269. Bossy-Wetzel, E.; Petrilli, A.; Knott, A.B. Mutant huntingtin and mitochondrial dysfunction. Trends Neurosci. 2008, 31, 609–616. [Google Scholar] [CrossRef]
  270. 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]
  271. 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]
  272. 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]
  273. 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]
  274. 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]
  275. 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]
  276. 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]
  277. 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]
  278. 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]
  279. 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]
  280. 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]
  281. 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]
  282. Wirth, B. Spinal Muscular Atrophy: In the Challenge Lies a Solution. Trends Neurosci. 2021, 44, 306–322. [Google Scholar] [CrossRef] [PubMed]
  283. 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]
  284. 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]
  285. 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]
  286. 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]
  287. 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]
  288. Marcus, R. What Is Multiple Sclerosis? JAMA 2022, 328, 2078. [Google Scholar] [CrossRef]
  289. 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]
  290. 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]
  291. 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]
  292. 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]
  293. 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]
  294. 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]
  295. 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]
  296. 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]
  297. 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]
  298. 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]
  299. 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]
  300. 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]
  301. 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]
  302. 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]
  303. Voet, S.; Srinivasan, S.; Lamkanfi, M.; van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 2019, 11, e10248. [Google Scholar] [CrossRef]
  304. 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]
  305. 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]
  306. 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]
  307. 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]
  308. 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]
  309. 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]
  310. 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]
  311. 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]
  312. 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]
  313. 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]
  314. Sasaki, S. Autophagy in spinal cord motor neurons in sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2011, 70, 349–359. [Google Scholar] [CrossRef]
  315. Rashid, S.; Dimitriadi, M. Autophagy in spinal muscular atrophy: From pathogenic mechanisms to therapeutic approaches. Front. Cell Neurosci. 2023, 17, 1307636. [Google Scholar] [CrossRef]
  316. 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]
  317. 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]
  318. 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]
  319. 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]
  320. Blennow, K.; Hardy, J.; Zetterberg, H. The neuropathology and neurobiology of traumatic brain injury. Neuron 2012, 76, 886–899. [Google Scholar] [CrossRef]
  321. 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]
  322. 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]
  323. 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]
  324. Barthels, D.; Das, H. Current advances in ischemic stroke research and therapies. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165260. [Google Scholar] [CrossRef]
  325. 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]
  326. 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]
  327. 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]
  328. 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]
  329. 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]
  330. Dirnagl, U.; Iadecola, C.; Moskowitz, M.A. Pathobiology of ischaemic stroke: An integrated view. Trends Neurosci. 1999, 22, 391–397. [Google Scholar] [CrossRef]
  331. 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]
  332. 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]
  333. 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]
  334. 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]
  335. 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]
  336. Mira, R.G.; Lira, M.; Cerpa, W. Traumatic Brain Injury: Mechanisms of Glial Response. Front. Physiol. 2021, 12, 740939. [Google Scholar] [CrossRef]
  337. 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]
  338. 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]
  339. 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]
  340. 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]
  341. 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]
  342. 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]
  343. 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]
  344. 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]
  345. 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]
  346. 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]
  347. 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]
  348. 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]
  349. 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]
  350. 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]
  351. 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]
  352. 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]
  353. Troy, C.M.; Jean, Y.Y. Caspases: Therapeutic targets in neurologic disease. Neurotherapeutics 2015, 12, 42–48. [Google Scholar] [CrossRef]
  354. 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]
  355. 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]
  356. 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]
  357. 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]
  358. 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]
  359. 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]
  360. 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]
  361. 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]
  362. 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]
  363. 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]
  364. 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]
  365. 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]
  366. 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]
  367. 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]
  368. 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]
  369. 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]
  370. 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]
  371. 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]
  372. Gafni, J.; Ellerby, L.M. Calpain activation in Huntington’s disease. J. Neurosci. 2002, 22, 4842–4849. [Google Scholar] [CrossRef] [PubMed]
  373. 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]
  374. 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]
  375. 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]
  376. Sarkar, S. Microglial ion channels: Key players in non-cell autonomous neurodegeneration. Neurobiol. Dis. 2022, 174, 105861. [Google Scholar] [CrossRef]
  377. 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]
  378. 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]
  379. 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]
  380. 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]
  381. Van Opdenbosch, N.; Lamkanfi, M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019, 50, 1352–1364. [Google Scholar] [CrossRef]
  382. 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]
  383. 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]
  384. 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]
  385. 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]
  386. 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]
  387. Hwang, J.Y.; Zukin, R.S. REST, a master transcriptional regulator in neurodegenerative disease. Curr. Opin. Neurobiol. 2018, 48, 193–200. [Google Scholar] [CrossRef]
  388. 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]
  389. 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]
  390. 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]
  391. 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]
  392. 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]
  393. 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]
  394. 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]
  395. 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]
  396. Bush, A.I. The metallobiology of Alzheimer’s disease. Trends Neurosci. 2003, 26, 207–214. [Google Scholar] [CrossRef]
  397. 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]
  398. 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]
  399. 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]
  400. 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]
  401. 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]
  402. 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]
  403. 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]
  404. Querfurth, H.; Lee, H.K. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol. Neurodegener. 2021, 16, 44. [Google Scholar] [CrossRef] [PubMed]
  405. 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]
  406. 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]
  407. 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]
  408. 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]
  409. 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]
  410. 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]
  411. 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]
Figure 1. The execution mechanisms of apoptosis, necroptosis, and pyroptosis are described in detail. (a) Both the intrinsic and extrinsic pathways of apoptosis are illustrated, highlighting the key signaling molecules and processes involved; (b) the assembly and function of various necroptosome structures are depicted, emphasizing the key proteins and their roles; (c) the pathways of pyroptosis execution are presented, detailing the stimuli and their effects on cellular components. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AIFs, apoptosis-inducing factors; Apaf-1, apoptosis protease activating factor-1; APP, amyloid precursor protein; ASC, apoptosis-associated speck-like protein containing a CARD; BH3-only proteins, Bcl-2 homology 3 domain only proteins; BID, BH3-interacting-domain death agonist; CARD, caspase recruitment domain; CTCs, circulating tumor cells; Cyt-c, cytochrome-c; DISC, death inducing signaling complex; DR6, death receptor 6; dsDNA, double-stranded DNA; ER, endoplasmic reticulum; FADD, Fas-associated death domain; GSDM, gasdermin; IAPs, inhibitors of apoptosis proteins; IFNARs, interferon alpha receptors; IFNs, interferons; IL-18, interleukin-18; IL-1β, interleukin-1β; K, potassium; LPS, lipopolysaccharide; MLKL, mixed-lineage kinase-like; NK, natural killer; NLR, nucleotide-binding oligomerization domain-like receptor; NLRP3, NLR family pyrin domain containing 3; NLS, nuclear localization signal; PD-1, programmed death 1; PD-L1, programmed cell death-ligand 1; PRRs, pattern recognition receptors; PtpB, protein tyrosine phosphatase B; RHIM, RIP (receptor-interacting protein) homology interaction motifs; RIPK1, serine/threonine protein kinase 1; RIPK3, serine/threonine protein kinase 3; Smac, small mitochondria-derived activator of caspase; SpeB, streptococcal pyrogenic exotoxin B; TAK1, TGF-β-activated kinase 1; T-BID, truncated BID; TLR3, toll-like receptor 3; TLR4, toll-like receptor 4; TRADD, TNF receptor-associated death domain; TRIF, TIR-domain-containing adapter-inducing interferon-β; YopJ, yersinia outer protein J; ZBP1, Z-DNA/RNA-binding protein; Z-dsDNA/RNA, Z-form double-stranded DNA/RNA.
Figure 1. The execution mechanisms of apoptosis, necroptosis, and pyroptosis are described in detail. (a) Both the intrinsic and extrinsic pathways of apoptosis are illustrated, highlighting the key signaling molecules and processes involved; (b) the assembly and function of various necroptosome structures are depicted, emphasizing the key proteins and their roles; (c) the pathways of pyroptosis execution are presented, detailing the stimuli and their effects on cellular components. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AIFs, apoptosis-inducing factors; Apaf-1, apoptosis protease activating factor-1; APP, amyloid precursor protein; ASC, apoptosis-associated speck-like protein containing a CARD; BH3-only proteins, Bcl-2 homology 3 domain only proteins; BID, BH3-interacting-domain death agonist; CARD, caspase recruitment domain; CTCs, circulating tumor cells; Cyt-c, cytochrome-c; DISC, death inducing signaling complex; DR6, death receptor 6; dsDNA, double-stranded DNA; ER, endoplasmic reticulum; FADD, Fas-associated death domain; GSDM, gasdermin; IAPs, inhibitors of apoptosis proteins; IFNARs, interferon alpha receptors; IFNs, interferons; IL-18, interleukin-18; IL-1β, interleukin-1β; K, potassium; LPS, lipopolysaccharide; MLKL, mixed-lineage kinase-like; NK, natural killer; NLR, nucleotide-binding oligomerization domain-like receptor; NLRP3, NLR family pyrin domain containing 3; NLS, nuclear localization signal; PD-1, programmed death 1; PD-L1, programmed cell death-ligand 1; PRRs, pattern recognition receptors; PtpB, protein tyrosine phosphatase B; RHIM, RIP (receptor-interacting protein) homology interaction motifs; RIPK1, serine/threonine protein kinase 1; RIPK3, serine/threonine protein kinase 3; Smac, small mitochondria-derived activator of caspase; SpeB, streptococcal pyrogenic exotoxin B; TAK1, TGF-β-activated kinase 1; T-BID, truncated BID; TLR3, toll-like receptor 3; TLR4, toll-like receptor 4; TRADD, TNF receptor-associated death domain; TRIF, TIR-domain-containing adapter-inducing interferon-β; YopJ, yersinia outer protein J; ZBP1, Z-DNA/RNA-binding protein; Z-dsDNA/RNA, Z-form double-stranded DNA/RNA.
Ijms 25 09947 g001
Figure 2. The pathways of cuproptosis and ferroptosis are illustrated. (a) Ferroptosis is a form of iron-dependent programmed cell death resulting from intracellular iron overload. The figure depicts the lipid peroxidation induced by dysregulated iron metabolism and the subsequent execution of ferroptosis; (b) abnormal copper metabolism and accumulation can lead to protein toxicity, mitochondrial damage, and cuproptosis. For details, refer to the corresponding section of this article. Abbreviations: Cu, copper; DSF, disulfiram; ES, elesclomol; FDX1, ferredoxin 1; Fe, iron; NCOA4, nuclear receptor coactivator 4; NRAMP2 (also known as SLC11A2), natural resistance-associated macrophage protein 2; ROS, reactive oxygen species; S, sulfur; SLC25A3, solute carrier family 25 member 3; STEAP, six-transmembrane epithelial antigen of prostate; TCA, tricarboxylic acid cycle; TF, transferrin; TFR1, transferrin receptor 1.
Figure 2. The pathways of cuproptosis and ferroptosis are illustrated. (a) Ferroptosis is a form of iron-dependent programmed cell death resulting from intracellular iron overload. The figure depicts the lipid peroxidation induced by dysregulated iron metabolism and the subsequent execution of ferroptosis; (b) abnormal copper metabolism and accumulation can lead to protein toxicity, mitochondrial damage, and cuproptosis. For details, refer to the corresponding section of this article. Abbreviations: Cu, copper; DSF, disulfiram; ES, elesclomol; FDX1, ferredoxin 1; Fe, iron; NCOA4, nuclear receptor coactivator 4; NRAMP2 (also known as SLC11A2), natural resistance-associated macrophage protein 2; ROS, reactive oxygen species; S, sulfur; SLC25A3, solute carrier family 25 member 3; STEAP, six-transmembrane epithelial antigen of prostate; TCA, tricarboxylic acid cycle; TF, transferrin; TFR1, transferrin receptor 1.
Ijms 25 09947 g002
Figure 3. An overview of the mechanisms of various other forms of PCD. (a) MPT-driven necrosis is mediated by the activation of CYPD and the formation of PTPC, leading to a loss of selective permeability of the inner mitochondrial membrane, resulting in cell swelling and membrane rupture; (b) oxeiptosis is a form of cell death induced by oxygen radicals and mediated by the hyperactivation of the KEAP1-PGAM5-AIFM1 signaling cascade; (c) LDCD is a form of cell death caused by changes in lysosomal membrane permeability, resulting in the leakage of lysosomal contents and subsequent alterations in mitochondrial outer membrane permeability; (d) parthanatos is a form of cell death induced by DNA damage, resulting in the overactivation of PARP1; (e) alkaliptosis is a form of cell death induced by intracellular alkalinization caused by JTC-801, an opioid receptor-like 1 (ORL1) receptor selective antagonist [5]; (f) in cells with high SLC7A11 expression, increased cystine uptake leads to NADPH depletion, abnormal disulfide bond formation, cytoskeletal collapse, and disulfidptosis; (g) aberrant autophagy leads to excessive ER-phagy, excessive mitophagy, and ADCD. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; AIFM1, apoptosis-inducing factor mitochondria-associated 1; ATP, adenosine triphosphate; ATP6V0D1, ATPase H+ transporting V0 subunit d1; ATPase, adenosine triphosphatase; CA9, carbonic anhydrase 9; CYPD, cyclophilin D; Cyt-c, cytochrome c; DRAM1, DNA damage-regulated autophagy modulator 1; DRP1, dynamin-related protein 1; ER, endoplasmic reticulum; Fe, iron; IMS, intermembrane space; K, potassium; KEAP1, kelch-like ECH-associated protein 1; LDCD, lysosome-dependent cell death; LMP, lysosomal membrane permeabilization; Mito, mitochondria; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; Na, sodium; NADH, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor κB; PARP1, poly(ADP-ribose) polymerase 1; PGAM, phosphoglycerate mutase; PGAM5, PGAM family member 5; pH, potential of hydrogen; reticulophagy, selective autophagy of the endoplasmic reticulum; PINK1, PTEN-induced kinase 1; PTPC, permeability transition pore complex; ROS, reactive oxygen species; S, sulfur; SLC7A11, solute carrier family 7 member 11; STAT3, signal transducer and activator of transcription 3; UPR, unfolded protein response.
Figure 3. An overview of the mechanisms of various other forms of PCD. (a) MPT-driven necrosis is mediated by the activation of CYPD and the formation of PTPC, leading to a loss of selective permeability of the inner mitochondrial membrane, resulting in cell swelling and membrane rupture; (b) oxeiptosis is a form of cell death induced by oxygen radicals and mediated by the hyperactivation of the KEAP1-PGAM5-AIFM1 signaling cascade; (c) LDCD is a form of cell death caused by changes in lysosomal membrane permeability, resulting in the leakage of lysosomal contents and subsequent alterations in mitochondrial outer membrane permeability; (d) parthanatos is a form of cell death induced by DNA damage, resulting in the overactivation of PARP1; (e) alkaliptosis is a form of cell death induced by intracellular alkalinization caused by JTC-801, an opioid receptor-like 1 (ORL1) receptor selective antagonist [5]; (f) in cells with high SLC7A11 expression, increased cystine uptake leads to NADPH depletion, abnormal disulfide bond formation, cytoskeletal collapse, and disulfidptosis; (g) aberrant autophagy leads to excessive ER-phagy, excessive mitophagy, and ADCD. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; AIFM1, apoptosis-inducing factor mitochondria-associated 1; ATP, adenosine triphosphate; ATP6V0D1, ATPase H+ transporting V0 subunit d1; ATPase, adenosine triphosphatase; CA9, carbonic anhydrase 9; CYPD, cyclophilin D; Cyt-c, cytochrome c; DRAM1, DNA damage-regulated autophagy modulator 1; DRP1, dynamin-related protein 1; ER, endoplasmic reticulum; Fe, iron; IMS, intermembrane space; K, potassium; KEAP1, kelch-like ECH-associated protein 1; LDCD, lysosome-dependent cell death; LMP, lysosomal membrane permeabilization; Mito, mitochondria; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; Na, sodium; NADH, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor κB; PARP1, poly(ADP-ribose) polymerase 1; PGAM, phosphoglycerate mutase; PGAM5, PGAM family member 5; pH, potential of hydrogen; reticulophagy, selective autophagy of the endoplasmic reticulum; PINK1, PTEN-induced kinase 1; PTPC, permeability transition pore complex; ROS, reactive oxygen species; S, sulfur; SLC7A11, solute carrier family 7 member 11; STAT3, signal transducer and activator of transcription 3; UPR, unfolded protein response.
Ijms 25 09947 g003
Figure 4. The pathways of NETosis and Entosis are depicted. (a) Cellular stress responses induce autophagy, granzyme release and translocation, chromatin decondensation, and cell membrane pore formation, leading to the release of web-like DNA–protein structures and resulting in NETosis; (b) cells undergo entosis, an intracellular cell death process, by inserting themselves into neighboring cells through adhesion proteins. For details, refer to the corresponding section of this article. Abbreviations: Ca, calcium; CTNNA1, catenin alpha 1; ENTosis, entotic cell death; ERK, extracellular signal-regulated kinase; GSDM, gasdermin; GSDMD-N,Gasdermin D N-terminal; LC3, microtubule-associated protein 1 light chain 3; LPS, lipopolysaccharide; MEK, MAP kinase kinase; MPO, myeloperoxidase; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; NADPH, nicotinamide adenine dinucleotide phosphate; NE, neutrophil elastase; NETosis, neutrophil extracellular trap cell death; NETs, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PKC, protein kinase C; RAF, RAF proto-oncogene serine/threonine-protein kinase; TLR, toll-like receptor.
Figure 4. The pathways of NETosis and Entosis are depicted. (a) Cellular stress responses induce autophagy, granzyme release and translocation, chromatin decondensation, and cell membrane pore formation, leading to the release of web-like DNA–protein structures and resulting in NETosis; (b) cells undergo entosis, an intracellular cell death process, by inserting themselves into neighboring cells through adhesion proteins. For details, refer to the corresponding section of this article. Abbreviations: Ca, calcium; CTNNA1, catenin alpha 1; ENTosis, entotic cell death; ERK, extracellular signal-regulated kinase; GSDM, gasdermin; GSDMD-N,Gasdermin D N-terminal; LC3, microtubule-associated protein 1 light chain 3; LPS, lipopolysaccharide; MEK, MAP kinase kinase; MPO, myeloperoxidase; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; NADPH, nicotinamide adenine dinucleotide phosphate; NE, neutrophil elastase; NETosis, neutrophil extracellular trap cell death; NETs, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PKC, protein kinase C; RAF, RAF proto-oncogene serine/threonine-protein kinase; TLR, toll-like receptor.
Ijms 25 09947 g004
Figure 5. The process of programmed cell death in the development of AD, PD and HD is significant. (a) Various forms of programmed cell death play roles in the pathogenesis and progression of AD, including their effects on Tau protein hyperphosphorylation, Aβ plaque formation, and neuronal cell death; (b) in PD, various forms of PCD induce dopaminergic neuronal loss and death by promoting αSyn aggregation, leading to mitochondrial dysfunction and neuroinflammation. (c) In HD, mHTT induces mitochondrial dysfunction and neuroinflammation by promoting the expression of pro-apoptotic factors and activating necroptosis and ferroptosis. Additionally, the aggregation of mHTT proteins is associated with impaired autophagy, further exacerbating neuronal damage. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AD, Alzheimer’s disease; Akt, protein kinase B; APP, amyloid precursor protein; Aβ, amyloid β; BAX, Bcl-2 associated x-protein; Bcl-2, B-cell lymphoma-2; BDNF, brain-derived neurotrophic factor; BIM, Bcl-2 interacting mediator of cell death; CGA, cytosine-guanine-adenine triplet; CREB, cAMP-response element binding protein; Cu, cuprum; CYPD, cyclophilin D; Cyt-c, cytochrome-c; ER, endoplasmic reticulum; Fe, ferrum; GPX4, glutathione peroxidase 4; GSDMD, gasdermin-D; GSK-3β, glycogen synthase kinase 3β; HD, Huntington’s disease; ICAM-1, intercellular adhesion molecule-1; IL-18, interleukin-18; IL-1β, interleukin-1β; JAK, janus kinase; JNK, c-Jun N-terminal kinase; LFA-1, lymphocyte function-associated antigen 1; LRP1, low-density lipoprotein receptor-related protein 1; LRRK2, leucine-rich repeat kinase 2; MAPK, mitogen-activated protein kinase; mHTT, mutant huntingtin; MLKL, mixed lineage kinase domain-like protein; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; mTOR, mammalian target of rapamycin; NET, neutrophil extracellular traps; NFTs, neurofibrillary tangles; NF-κB, nuclear factor κB; NLRP3, NLR family pyrin domain containing 3; NLR, nucleotide-binding oligomerization domain-like receptor; NO, nitric oxide; PARK7, parkinsonism associated deglycase; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; PD, Parkinson’s disease; PI3K, phosphoinositide 3-kinase; PINK1, PTEN induced kinase 1; polyQ, polyglutamine; PRKN, parkin RBR E3 ubiquitin protein ligase; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SNCA, alpha-synuclein; αSyn, α-synuclein; STAT, signal transducer and activator of transcription; Tau, microtubule-associated protein Tau; TNF-α, tumor necrosis factor-α; TrkB, tropomyosin receptor kinase B.
Figure 5. The process of programmed cell death in the development of AD, PD and HD is significant. (a) Various forms of programmed cell death play roles in the pathogenesis and progression of AD, including their effects on Tau protein hyperphosphorylation, Aβ plaque formation, and neuronal cell death; (b) in PD, various forms of PCD induce dopaminergic neuronal loss and death by promoting αSyn aggregation, leading to mitochondrial dysfunction and neuroinflammation. (c) In HD, mHTT induces mitochondrial dysfunction and neuroinflammation by promoting the expression of pro-apoptotic factors and activating necroptosis and ferroptosis. Additionally, the aggregation of mHTT proteins is associated with impaired autophagy, further exacerbating neuronal damage. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AD, Alzheimer’s disease; Akt, protein kinase B; APP, amyloid precursor protein; Aβ, amyloid β; BAX, Bcl-2 associated x-protein; Bcl-2, B-cell lymphoma-2; BDNF, brain-derived neurotrophic factor; BIM, Bcl-2 interacting mediator of cell death; CGA, cytosine-guanine-adenine triplet; CREB, cAMP-response element binding protein; Cu, cuprum; CYPD, cyclophilin D; Cyt-c, cytochrome-c; ER, endoplasmic reticulum; Fe, ferrum; GPX4, glutathione peroxidase 4; GSDMD, gasdermin-D; GSK-3β, glycogen synthase kinase 3β; HD, Huntington’s disease; ICAM-1, intercellular adhesion molecule-1; IL-18, interleukin-18; IL-1β, interleukin-1β; JAK, janus kinase; JNK, c-Jun N-terminal kinase; LFA-1, lymphocyte function-associated antigen 1; LRP1, low-density lipoprotein receptor-related protein 1; LRRK2, leucine-rich repeat kinase 2; MAPK, mitogen-activated protein kinase; mHTT, mutant huntingtin; MLKL, mixed lineage kinase domain-like protein; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; mTOR, mammalian target of rapamycin; NET, neutrophil extracellular traps; NFTs, neurofibrillary tangles; NF-κB, nuclear factor κB; NLRP3, NLR family pyrin domain containing 3; NLR, nucleotide-binding oligomerization domain-like receptor; NO, nitric oxide; PARK7, parkinsonism associated deglycase; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; PD, Parkinson’s disease; PI3K, phosphoinositide 3-kinase; PINK1, PTEN induced kinase 1; polyQ, polyglutamine; PRKN, parkin RBR E3 ubiquitin protein ligase; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SNCA, alpha-synuclein; αSyn, α-synuclein; STAT, signal transducer and activator of transcription; Tau, microtubule-associated protein Tau; TNF-α, tumor necrosis factor-α; TrkB, tropomyosin receptor kinase B.
Ijms 25 09947 g005
Figure 6. The role of PCD in the progression of ALS, SMA and MS is critical. The figure illustrates the involvement of PCD pathways in the pathogenesis of ALS, SMA, and MS, highlighting both the factors that contribute to disease progression and those that are beneficial for disease control. Additionally, it describes the common pathways through which PCD exerts its effects across these diseases. For more details, refer to the corresponding section of this article. Abbreviations: ADCD, autophagy-dependent cell death; ALS, amyotrophic lateral sclerosis; Bcl-2, B-cell lymphoma-2; C9ORF72, chromosome 9 open reading frame 72; Cu, cuprum; CYPD, cyclophilin D; Fe, ferrum; FUS, fused in sarcoma/translocated in liposarcoma; GSDMD, gasdermin-D; IL-18, interleukin-18; IL-1β, interleukin-1β; JNK, c-Jun N-terminal kinase; KO, knockout; MPT, mitochondrial permeability transition; mPTP, mitochondrial permeability transition pore; MS, multiple sclerosis; NET, neutrophil extracellular traps; NETosis, neutrophil extracellular trap cell death; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SMA, spinal muscular atrophy; SMN, survival motor neuron; SOD1, superoxide dismutase 1; TARDBP, TAR DNA-binding protein; TDP-43, TAR DNA-binding protein 43.
Figure 6. The role of PCD in the progression of ALS, SMA and MS is critical. The figure illustrates the involvement of PCD pathways in the pathogenesis of ALS, SMA, and MS, highlighting both the factors that contribute to disease progression and those that are beneficial for disease control. Additionally, it describes the common pathways through which PCD exerts its effects across these diseases. For more details, refer to the corresponding section of this article. Abbreviations: ADCD, autophagy-dependent cell death; ALS, amyotrophic lateral sclerosis; Bcl-2, B-cell lymphoma-2; C9ORF72, chromosome 9 open reading frame 72; Cu, cuprum; CYPD, cyclophilin D; Fe, ferrum; FUS, fused in sarcoma/translocated in liposarcoma; GSDMD, gasdermin-D; IL-18, interleukin-18; IL-1β, interleukin-1β; JNK, c-Jun N-terminal kinase; KO, knockout; MPT, mitochondrial permeability transition; mPTP, mitochondrial permeability transition pore; MS, multiple sclerosis; NET, neutrophil extracellular traps; NETosis, neutrophil extracellular trap cell death; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SMA, spinal muscular atrophy; SMN, survival motor neuron; SOD1, superoxide dismutase 1; TARDBP, TAR DNA-binding protein; TDP-43, TAR DNA-binding protein 43.
Ijms 25 09947 g006
Figure 7. The role of programmed cell death (PCD) in the progression of traumatic brain injury (TBI) and stroke. The figure illustrates various aspects that contribute to disease progression, revealing the role of PCD in these conditions and considerations beneficial for disease control and treatment. Additionally, it describes the pathways through which PCD exerts its effects both individually and collectively in TBI and stroke. For details, refer to the corresponding section of this article. The upward and downward arrows represent an increase and decrease in content or concentration, respectively. The circular arrow signifies that "apoptosis in neurons of the ischemic penumbra may be recoverable." The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; ATP, adenosine triphosphate; Bcl-2, B-cell lymphoma-2; Ca, calcium; CNS, central nervous system; Cu, cuprum; CYPD, cyclophilin D; DRGs, dorsal root ganglions; ER, endoplasmic reticulum; FasL, Fas ligand; Fe, ferrum; HIF-1α, hypoxia-inducible factor 1α; HMGB1, high-mobility group box 1; IL-1β, interleukin-1β; IP, ischemic penumbra; K, kalium; LDCD, lysosome-dependent cell death; Mito, mitochondria; MLKL, mixed lineage kinase domain-like protein; MPT, mitochondrial permeability transition; Na, natrium; NAD, nicotinamide adenine dinucleotide; NET, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; TBI, traumatic brain injury; TNF-α, tumor necrosis factor-α; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand.
Figure 7. The role of programmed cell death (PCD) in the progression of traumatic brain injury (TBI) and stroke. The figure illustrates various aspects that contribute to disease progression, revealing the role of PCD in these conditions and considerations beneficial for disease control and treatment. Additionally, it describes the pathways through which PCD exerts its effects both individually and collectively in TBI and stroke. For details, refer to the corresponding section of this article. The upward and downward arrows represent an increase and decrease in content or concentration, respectively. The circular arrow signifies that "apoptosis in neurons of the ischemic penumbra may be recoverable." The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; ATP, adenosine triphosphate; Bcl-2, B-cell lymphoma-2; Ca, calcium; CNS, central nervous system; Cu, cuprum; CYPD, cyclophilin D; DRGs, dorsal root ganglions; ER, endoplasmic reticulum; FasL, Fas ligand; Fe, ferrum; HIF-1α, hypoxia-inducible factor 1α; HMGB1, high-mobility group box 1; IL-1β, interleukin-1β; IP, ischemic penumbra; K, kalium; LDCD, lysosome-dependent cell death; Mito, mitochondria; MLKL, mixed lineage kinase domain-like protein; MPT, mitochondrial permeability transition; Na, natrium; NAD, nicotinamide adenine dinucleotide; NET, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; TBI, traumatic brain injury; TNF-α, tumor necrosis factor-α; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand.
Ijms 25 09947 g007
Table 1. Key regulators involved in the iron metabolism and ferroptosis.
Table 1. Key regulators involved in the iron metabolism and ferroptosis.
ModulatorsFunctionsRefs.
hepcidindegrading the ferroportin via ubiquitination[102]
IRP1/2promoting the expression of TFR1[103]
PCBP1delivering ferrous iron to ferritin[104]
SLC25A37/8promoting the absorption of iron[105]
HO1catalyzing the synthesis of ferrous iron[106]
SLC40A1assisting the export of iron[107]
SLC39A14assisting the import of iron[108]
SLC25A28regulating the generation of ROS[109]
SFXN1regulating the generation of ROS[110]
PROM2regulating the storage of iron in ferritin[111]
PHKG2modulating the oxidative reactions[112]
HMOX1participating in the synthesis of ferrous iron[113]
SLC11A2assisting the absorption of iron[87]
CPconverting ferrous iron to ferric iron[114]
CISD1/2participating in the absorption of iron[115,116]
DMT1controlling the absorption of iron[117]
FBXL5degrading the IRP1 via ubiquitination[118]
HSF1regulating the iron metabolism-related genes[119]
HSPA5binding to GPX4 to prevent its degradation[120]
NRF2inducing the expression of antioxidant genes[121]
CISD1alleviating the accumulation of lipid[122]
ALOXsfacilitating the lipoxygenase[123]
PEBP1enhancing the lipid death pathway via 15-LO[124]
NOXsfacilitating the generation of ROS[125]
DPP4/CD26causing the lipid peroxidation[126,127]
VDAC2/3activating ferroptotic agonist erastin[128]
MUC1activating GPX4[129]
GCLCaccelerating the synthesis of GSH[130]
GLS2increasing the ROS production via αKG[131]
CARSinhibiting the generation of GSH[132]
CHAC1promoting oxidative reactions[133]
LSHpromoting SLC7A11 transcription[134]
FADS2desaturating the free fatty acids[135]
ACSL3upregulating the lipid droplet biogenesis[136]
ACSL4shaping cellular composition[137]
LPCAT3upregulating polyunsaturated free fatty acid[138]
PHGDHupregulating the expression of PHGDH[139]
G6PDpreventing oxidative reactions via inhibiting POR[140]
ME1facilitating the generation of GSH[141]
PHKG2regulating lipoxygenase enzyme ALOX5[142]
HMGCRincreasing GPX4 and CoQ10 biosynthesis[143]
SQLEpreventing oxidative via squalene[144]
NRF2regulating the antioxidant-related genes expression[145]
P53modulating GPX4 pathway and ROS production[146]
HIF-1αimproving the expression of SLC7A11 via PMAN[147]
BACH1enhancing iron metabolisom-related gene expression[148]
STAT3enhancing the expression of GPX4 and SLC7A11[149]
ATF3restraining the activity of system Xc[150]
ATF4facilitating the expression of SLC7A11[151]
CHOPfacilitating the expression of CHAC1[152]
YAP/TAZinducing the expression of SLC7A11[153]
Abbreviation: IRP1/IRP2, iron regulatory protein 1/2; PCBP1, poly(rC)-binding protein 1; SLC25A37/SLC25A38, mitoferrin 1/2; HMOX1, heme oxygenase 1; SLC40A1, solute carrier family 40 member 1; SLC39A14, solute carrier family 39 member 14; SLC25A28, mitoferrin 2; SFXN1, sideroflexin 1; PROM2, prominin 2; PHKG2, phosphorylase b kinase γ-catalytic chain, liver/testis isoform; SLC11A2, natural resistance-associated macrophage protein 2; CP, ceruloplasmin; CISD1/CISD2, CDGSH iron–sulfur domain-containing protein 1/2; ACO1, cytoplasmic aconitate hydratase; DMT1, divalent metal transporter 1; FBXL5, F-box and leucine-rich repeat protein 5; HSF1, heat shock factor 1; HSPA5, heat shock 70-kDa protein 5; NRF2, nuclear factor erythroid 2-related factor 2; ALOXs, arachidonic acid lipoxygenases; PEBP1, phosphatidylethanolamine (PE)-binding protein 1; NOXs, NADPH oxidases; DPP4/CD26, dipeptidyl peptidase-4; VDAC2/3, voltage-dependent anion channel 2/3; MUC1, mucin 1; GCLC, glutamate-cysteine ligase catalytic subunit; GLS2, glutaminase 2; CARS, cysteinyl-tRNA synthetase; CHAC1, cation transport regulator-like protein 1; LSH, lymphoid-specific helicase; SCD, stearoyl-CoA desaturase; FADS2, fatty acid desaturase 2; ACSL3/4, acyl-CoA synthetase long-chain family member 3/4; LPCAT3, lysophosphatidylcholine acyltransferase 3; PHGDH, phosphoglycerate dehydrogenase; G6PD, glucose 6-phosphate dehydrogenase; ME1, malic enzyme 1; αKG, α-ketoglutarate; POR, cytochrome P450 oxidoreductase; ALOX5, arachidonate 5-lipoxygenase; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; SQLE, squalene epoxidase; HIF-1α, hypoxia-inducible factor 1α; BACH1, BTB domain and CNC homolog 1; STAT3, signal transducer and activator of transcription 3; ATF3/4, activating transcription factor 3/4; CHOP, C/EBP homologous protein; YAP/TAZ, Yes-associated protein/Transcriptional coactivator with PDZ-binding motif.
Table 2. Hallmark features of aforementioned PCD modalities.
Table 2. Hallmark features of aforementioned PCD modalities.
FormsImmune FeaturesMorphological FeaturesMajor
Inhibitors
Refs.
ApoptosisTCDApoptotic bodies formation;
Nuclear condensation;
Plasma membrane blebbing;
Cell shrinkage.
Z-VAD-FMK;
Q-VD-OPh;
Z-VAD (OH)-FMK.
[4]
[17]
NecroptosisICDCell swelling and oncosis
Rupture of plasma membrane;
Swelling of organelles
Chromatin condensation.
Nec-1;
GSK872;
NSA;
HS-1371.
[46]
[49]
PyroptosisICDLack of cell swelling;
Rupture of plasma membrane;
Cell bubbling;
Chromatin condensation.
Ac-YVAD-cmk;
VX765;
Ac-FLTD-CMK.
[67]
[204]
FerroptosisICDSmaller mitochondria;
Rupture of mitochondrial membrane;
Decreased mitochondrial cristae;
Normal nucleus.
Deferiprone;
Ferrostatin-1;
Alogliptin;
Selenium;
CoQ10;
Vildagliptin;
Vitamin E.
[80]
[86]
[205]
CuproptosisICDmitochondrial condensation;
Rupture of plasma membrane;
ER damage;
Chromatin condensation.
NSC689534
EMeramide
Penicillamine
AT-VI
[7]
[154]
MPT-driven necrosisICDPlasma membrane rupture;
Swelling of organelles;
Lack of inter-nucleosomal DNA fragmentation;
Depletion of ATP.
SfA[5]
ADCDICDAutophagic vacuolization.Chloroquline;
Spactin-1.
[168]
LDCDICDRupture of lysosome and plasma membrane.NAC;
CA-074Me
[1]
ParthanatosICDChromatin condensation;
Large DNA fragmentation;
Loss of cell swelling.
BYK204165;
AG-14361;
Iniparib.
[184]
[185]
OxeiptosisTCDApoptosis-like morphology.NAC.[188]
AlkaliptosisICDNecrosis-like morphology.NAC;
CAY10657;
SC514.
[5]
DisulfidptosisICDCell shrinkage;
Nuclear condensation;
Formation of aberrant disulfide bonds between actin cytoskeleton proteins;
Chromatin condensation.
GLUT inhibitor[6]
[189]
NETosisTCD or
ICD
Rupture of plasma membrane and nuclear membrane;
Release of chromatin fragments.
lactoferrin;
DNase;
Cl-amidine.
[190]
[206]
ENTosisTCD or ICDFormation of cell-in-cell structure.C3-toxin;
γ-27632
[196]
Abbreviations: MPT, mitochondrial permeability transition; ADCD, autophagy-dependent cell death; LDCD, lysosome-dependent cell death; NETosis, neutrophil extracellular trap cell death; ENTosis, entotic cell death; ICD, immunogenic cell death; TCD, tolerogenic cell death; ER, endoplasmic reticulum; AT-VI, ammonium tetrathiomolybdate (VI); Nec-1, necrostatin-1; GLUT inhibitor, glucose transporter 1 inhibitor; NSA, necrosulfonamide; SfA, sanglifehrin A.
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.

Share and Cite

MDPI and ACS Style

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

AMA Style

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 Style

Guo, 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

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop