Next Article in Journal
Chronic Inflammation and Altered Immune Responses in LongCOVID Associate with Neurological Manifestations and Accelerated Aging
Previous Article in Journal
Therapeutic Potential of an Anti-CD44v6 Monoclonal Antibody in Xenograft Models of Colorectal and Gastric Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Regulated Cell Death Pathways in COPD: Mechanisms and Therapeutic Strategies

Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Cells 2025, 14(23), 1874; https://doi.org/10.3390/cells14231874
Submission received: 2 October 2025 / Revised: 8 November 2025 / Accepted: 25 November 2025 / Published: 26 November 2025

Abstract

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease defined by persistent airflow limitation, chronic inflammation, and ongoing airway remodeling, and poses a substantial global health challenge. Despite its clinical significance, the underlying cellular mechanisms remain poorly defined. Regulated cell death (RCD) incorporates various kinds of cell death that are typically regulated by specific molecular pathways. Numerous new kinds of RCD have been identified outside of the traditional apoptotic pathway, like necroptosis, pyroptosis, ferroptosis, autophagy, cuproptosis, and parthanatos. To date, there is growing evidence indicating that these pathways participate in the regulation of COPD development. However, their specific roles and therapeutic relevance remain poorly understood. In this review, we discuss a summary of the molecular mechanisms by which RCD pathways influence the onset and advancement of COPD. Additionally, the therapeutic benefits of agents that target these cell death pathways in COPD treatment were described. By integrating current insights, this review aims to broaden our knowledge of the pathophysiology of COPD and offer novel approaches to treatment.

1. Introduction

Chronic obstructive pulmonary disease (COPD) is a prevalent and widespread respiratory condition marked by a high mortality rate, ranking as the third primary cause of death globally [1]. Parameters like smoking and exposure to toxic airborne particles, can trigger the onset of COPD [2,3]. Exposure to these risk factors induces oxidative stress [4], inflammation [5], and airway remodeling, leading to increased mucus secretion, airway obstruction, and airflow limitation [6]. These changes also disrupt alveolar structures, resulting in emphysema development and, eventually, COPD. Additionally, a crucial aspect of this process is the cell death [7,8].
Cell death can be categorized into accidental cell death (ACD) and regulated cell death (RCD), all of which have been reported to contribute to COPD development [9] (Figure 1). Apoptosis, the classical type of RCD, significantly drives COPD progression. More recently, several non-apoptotic RCD pathways have been identified [10]. Necroptosis [11] occurs through death receptor signaling and leads to inflammatory cell death. Pyroptosis [12] is a form of inflammatory cell death triggered by inflammasome activation and gasdermin-mediated membrane pore formation. Ferroptosis [13] is an iron-dependent cell death driven by lipid peroxidation. Autophagy [14] functions as a cellular recycling mechanism, which can either promote cell survival or trigger cell death under stress. Cuproptosis [15] results from toxic copper ion accumulation, leading to cell death. Parthanatos [16] is a cell death pathway mediated by PARP1 and associated with extensive DNA damage. In COPD, the imbalance between cell death and tissue repair drives structural lung damage and emphysema, while dysregulated RCD further amplifies tissue-destructive inflammation, thereby exacerbating airway inflammation and remodeling. Given its inherent controllability, RCD represents a promising target for therapeutic intervention.
We have provided a systematic review of the cellular mechanisms and their function in COPD occurrence and progression. Furthermore, we focus on the therapeutic effects and mechanisms of pharmacological agents that target these RCD pathways, highlighting their potential applications in COPD management. The collective goal of these discoveries is to improve our knowledge of the pathophysiology of COPD and provide novel avenues for improved therapeutic development.

2. Apoptosis

2.1. Overview of Apoptosis

A distinct form of programmed cell death, or apoptosis, as coined by Kerr, was first introduced in 1972 [17]. The hallmark morphological changes in apoptosis include cell rounding, nuclear condensation, plasma membrane blebbing, and the formation of apoptotic bodies [18]. Furthermore, it can be categorized into intrinsic apoptosis and extrinsic apoptosis [19] (Figure 2). Intrinsic apoptosis is initiated with mitochondrial outer membrane permeabilization (MOMP), which releases cytochrome c (Cyto-C) into the cytoplasm. In the cytoplasm, Cyto-C interacts with Apaf-1 to form the apoptosome, which subsequently activates caspase-9. This initiator caspase then triggers the caspases-3/7, leading to cellular disintegration [20]. Notably, this classical pathway is tightly regulated by intrinsic inhibitory factors. For example, anti-apoptotic Bcl-2 family proteins (Bcl-2, Bcl-xL, and Mcl-1) are capable of preventing MOMP by directly binding and inhibiting Bax/Bak [21], while mitochondrial fusion protein Mfn2 has been found to inhibit Bax activation through direct interaction [22]. Extrinsic apoptosis is triggered by the binding of a specific ligand to its corresponding receptor (Fas and TNF receptor superfamily member 1A (TNFRSF1A)) on the cell surface. Following ligand binding, the death-inducing signal complex (DISC) is assembled through the clustering or trimerizing of the death receptors, which recruit Fas-associated death domain (FADD) together with procaspase-8/10. To increase the apoptotic signal, the activated caspase-8/10 either directly or indirectly cleaves Bid to produce tBid, which in turn activates Bax/Bak and promotes MOMP [23]. Moreover, caspase-9 or death-associated protein kinase 1 (DAPK1) can be activated by receptor-dependent pathways involving Unc-5 Netrin Receptor B (UNC5B) and DCC Netrin 1 Receptor (DCC) after dephosphorylation induced by ligand withdrawal [18,24]. In all cases, the caspase cascade is crucial to the successful completion of apoptosis [25].

2.2. Apoptosis in COPD

The percentage of apoptotic cells in the lung parenchymal cells of patients with COPD is approximately 1–5%, which is more than twice the rate observed in healthy lungs [26]. Chen et al. [27] demonstrated that apoptosis is markedly increased in both bronchial and alveolar epithelial cells in emphysema mouse models generated by exposure to cigarette smoke (CS) or intraperitoneal administration of cigarette smoke extract (CSE). In addition, widespread apoptosis of pulmonary vascular endothelial cells has been noted in COPD, with evidence suggesting that this process not only represents a key pathological feature but may also actively drive disease pathogenesis [28,29]. Importantly, the ability of alveolar macrophages to phagocytize apoptotic epithelial cells is compromised in COPD, due to both intrinsic defects in macrophage function and excessive apoptotic burden. The resulting persistence of uncleared apoptotic cells results in secondary necrosis, thereby exacerbating inflammation and accelerating COPD progression [26,30]. The regulatory network governing apoptosis in COPD is complex and involves multiple intracellular signaling pathways [31]. Sonic hedgehog (Shh) signaling pathway significantly participates in the maintenance of pulmonary cellular homeostasis and suppressing apoptosis, and its dysfunction has been closely linked to COPD development [32]. Li et al. [33] reported that CSE stimulation downregulates Shh pathway-associated protein expression in MLE cells. Treatment with recombinant mouse Shh protein partially reversed apoptosis, whereas inhibition of Shh signaling with cyclosporine A (an inhibitor of the Shh pathway) enhanced apoptosis in MLE-12 cells. Furthermore, apoptosis in COPD undergoes precise regulation via multiple epigenetic mechanisms. Fu et al. [34] demonstrated that miR-613 exacerbates CSE-induced bronchial epithelial cell apoptosis and epithelial–mesenchymal transition (EMT) through the targeting of ARHGAP1, thereby contributing to inflammatory responses and airway remodeling in COPD. Similarly, Li et al. [35] showed that CSE reduces PRMT6 expression and H3R2me2a levels in pulmonary epithelial cells while upregulating Bax expression and inflammatory cytokine production. Inflammation, another central feature of COPD pathology, is also closely linked to apoptosis [10,36]. Patients with COPD present with higher levels of inflammatory mediators such as TNF-α and interferon-γ [37]. TNF-α is primarily produced by alveolar macrophages and mediates apoptosis by activating the extrinsic apoptotic pathway [38]. Interferon-γ, mainly secreted by Th1 CD4+ and CD8+ T lymphocytes, induces apoptosis in type II alveolar epithelial cells [39]. These inflammatory mediators further recruit neutrophils, lymphocytes, and macrophages to the lung parenchyma and airways. The accumulation of macrophages and neutrophils leads to protease imbalance and oxidative stress, whereas adaptive immune cells undergo proliferative expansion and promote apoptosis of alveolar epithelial cells [40].
Accumulating evidence indicates that CS can induce endoplasmic reticulum stress, resulting in the activation of the unfolded protein response (UPR), disrupts protein homeostasis, and ultimately triggers apoptosis [41]. Feng et al. [42] found that CSE promotes apoptosis in Raw264.7 macrophages by upregulating ER stress-related proteins and activating the Ca2+/P38/STAT1 signaling pathways, suggesting that CS-induced macrophage dysfunction may increase susceptibility to pulmonary infections in COPD. Furthermore, Chen et al. [43] demonstrated in human airway smooth muscle cells (HASMCs) that CSE upregulates ORMDL3, leading to ATF6 activation, disruption of endoplasmic reticulum calcium homeostasis, and enhanced secretion of inflammatory cytokines, thereby facilitating airway remodeling, persistent inflammation, and airflow limitation in COPD.

3. Necroptosis

3.1. Overview of Necroptosis

Research on necroptosis commenced in 2005 [44]. Unlike apoptosis, necroptosis is caspase-independent and does not involve apoptotic body formation [45]. Instead, it is defined by the rupture of the plasma membrane, secretion of intracellular contents, and leakage of damage-associated molecular patterns (DAMPs), such as HMGB1, S100 proteins, and ATP, thereby triggering a potent inflammatory response [46]. Necroptosis is typically initiated through ligand-receptor signaling, whereby external or internal stimuli activate death receptors like TNFR1. Upon binding, TNF receptor-associated death domain protein (TRADD), TRAF2, RIPK1, and cIAPs are induced by TNFR1, thereby initiating NF-κB-mediated pro-survival signaling. Deubiquitination of RIPK1 promotes its release from complex I and subsequent formation of complex IIa with FADD and caspase-8, thereby initiating apoptosis. However, the inhibition of caspase-8 activity activates RIPK3, triggering the intracellular kinase cascade activation further [47]. Phosphorylated RIPK1 cooperates with RIPK3 and MLKL to form the necrosome, within which RIPK3 and MLKL are sequentially phosphorylated. Activated MLKL then oligomerises and inserts into the plasma membrane, generating disruptive pores that compromise membrane integrity and execute necroptosis [48,49] (Figure 3).

3.2. Necroptosis in COPD

Necroptosis is strongly upregulated in the lungs of patients with COPD and contributes to airway inflammation, airway remodeling, and emphysema formation [50]. Relevant studies have shown that MLKL expression in alveolar epithelial cells and macrophages of patients with severe disease is elevated, accompanied by increased levels of phosphorylated RIPK3(p-RIPK3) and phosphorylated MLKL(p-MLKL) in lung tissue [50]. Conversely, the absence of RIPK3 or MLKL reduces CS-induced macrophage death, thereby alleviating airway inflammation, remodeling, and the pathological progression of emphysema [51]. Furthermore, RIPK1 deficiency diminishes smoking-induced pulmonary cell apoptosis and necrotic cell death, thereby alleviating airway inflammation, remodeling, and emphysema [52] while also preventing cigarette-induced impairment of mucociliary clearance [53]. The specific mechanisms by which necroptosis contributes to COPD pathogenesis may include aberrant upregulation of pro-inflammatory mediators, inefficient removal of dead cells, and promoting oxidative stress. For example, Zeng et al. [54] demonstrated that miR-21 knockout reduces necrotic apoptosis-associated proteins (TNFR1 and p-MLKL) in bronchial epithelial cells, concurrently lowering inflammatory cytokine levels and thereby ameliorating COPD symptoms. According to Wang and colleagues [55], GRP78 facilitates CSE-induced inflammation of airway epithelial cells and excessive mucus production by upregulating necrotic apoptosis and subsequently activating NF-κB and AP-1 pathways. In addition, Chen and associates [56] showed that impaired macrophage clearance of necrotic cells and DAMPs reduces phagocytic efficiency, thereby amplifying pro-inflammatory factor expression and ultimately triggering pathological inflammatory responses. Another study further reported that CSE induces necroptosis in bronchial epithelial cells, triggering the secretion of mitochondrial DNA (mtDNA) into the cytoplasm and extracellular space, which in turn activates inflammatory signaling [57].

4. Ferroptosis

4.1. Overview of Ferroptosis

Ferroptosis is a controlled type of RCD that is initiated by oxidative disturbances within the intracellular microenvironment and defined by its reliance on iron and lipid peroxidation. Morphologically, it is characterized by mitochondrial shrinkage, increased membrane density, decreased cristae density, and eventual rupture of the mitochondrial membrane [58]. Its mechanisms are complex and involve interactions among iron metabolism, glutathione metabolism, and lipid peroxidation (Figure 4). In iron metabolism, Ferric iron (Fe3+) is taken up by cells via transferrin receptors (TFRs), where it is converted to ferrous iron (Fe2+) within endosomes, and subsequently transported into the labile intracellular iron pool. Under pathological conditions such as infection or inflammation, disruption in iron homeostasis may result in an accumulation of iron within cells. Excess iron accelerates lipid peroxidation of polyunsaturated fatty acids (PUFAs), ultimately driving ferroptosis. The cystine/glutamate antiporter system Xc is responsible for maintaining redox balance. This system consists of the light chain subunit xCT (SLC7A11) and the heavy chain subunit CD98hc (SLC3A2), which facilitates the import of cystine in exchange for glutamate [59]. Intracellular cystine is enzymatically converted into glutathione (GSH), which, as an electron donor, reduces phospholipid hydroperoxides (PL-OOH) to their corresponding alcohols through the action of glutathione peroxidase 4 (GPX4), thereby mitigating lipid toxicity and inhibiting ferroptosis [60]. Ferroptosis is a process that takes place via an iron-catalyzed lipid peroxidation mechanism. This process can be triggered by both non-enzymatic (Fenton reaction) and enzymatic mechanisms (lipoxygenases, ALOXs). The Fenton reaction, mediated by Fe2+, produces reactive oxygen species (ROS) such as hydroxyl radicals. These ROS engage in lipid peroxidation reactions with phospholipid-containing PUFAs to generate lipid peroxidation products (PL-PUFA-OOH) such as malondialdehyde (MDA) and 4-hydroxynonenal (HNE), which disrupt cell membrane fluidity and permeability, leading to cell death. In parallel, ALOXs, a family of non-heme iron-containing enzymes, catalyze the PUFA oxygenation to produce PL-PUFA-OOH, further amplifying lipid peroxidation and promoting ferroptosis.

4.2. Ferroptosis in COPD

In 2012, B. Stockwell introduced the concept of ferroptosis [61] and was implicated in the regulation of COPD formation in 2019 [62]. Growing evidence reveals that patients with COPD display noticeably raised levels of ACSL4, LPCAT3, and lipid peroxidation products compared to healthy controls [63]. CS increases the expression of the key ferroptosis protein NCOA4 in COPD, promoting epithelial ferroptosis, iron release, and iron accumulation. Excess intracellular iron enhances ROS production through the Fenton reaction, further depleting GPX4, exacerbating lipid peroxidation, and inducing cell death, airway inflammation, airway remodeling, and emphysema development [64]. Ferroptosis is therefore characterized by iron overload, excessive lipid peroxidation, and disruption of the system Xc/GSH/GPX4 antioxidant axis [65], collectively driving cellular damage and contributing to COPD pathogenesis.
As previously mentioned, ACSL4 is pivotal in lipid peroxidation and acts as a crucial regulator of ferroptosis. Wang et al. [66] reported that both Lv-shACSL4-treated COPD mouse models and siACSL4-transfected cells effectively suppressed ACSL4 expression, decreased lipid peroxidation, and downregulated ferroptosis markers in vivo and in vitro. This suggests that ACSL4 is a key factor in ferroptosis in COPD. Liu and colleagues [67] found that the deubiquitinating enzyme USP8 was downregulated in COPD models. Its overexpression activated the OTUB1/SLC7A11 pathway, enhancing cystine uptake and GSH synthesis to maintain GPx4 activity. This, in turn, suppressed ACSL4 expression, alleviating lung inflammation and airway remodeling. Epigenetic regulation influences the pathogenesis of COPD by affecting ferroptosis, offering new therapeutic directions. TET2, a DNA demethylase, catalyzes cytosine hydroxymethylation and chromatin remodeling to regulate gene expression, playing a crucial epigenetic role in myeloid hematopoiesis, inflammatory responses, and various pulmonary diseases. Zeng [68] et al. found that TET2 downregulation induces hypermethylation of the GPx4 promoter, impairing antioxidant defenses and exacerbating ferroptosis. Notably, co-treatment with the demethylating agent 5-AZA and the antioxidant NAC synergistically reduced ferroptosis and lung injury, suggesting that combining methylation inhibitors with antioxidants could provide a potentially new therapeutic approach for COPD. Xia et al. [64] identified the m6A-modified circSAV1/YTHDF1/IREB2 axis as the key driver of CS-induced ferroptosis in lung epithelial cells. This pathway promotes IREB2 mRNA translation, disrupts iron homeostasis, and exacerbates lipid peroxidation. Targeting circSAV1 or inhibiting key molecules in this axis significantly mitigated alveolar enlargement and airway remodeling in COPD mouse models.

5. Autophagy

5.1. Overview of Autophagy

Christian de Duve termed his observation of the lysosome-dependent degradation in cells, autophagy, in 1963 [69]. Autophagy is a highly conserved, pivotal biological process in eukaryotic cells that involves the breakdown of and recycling of damaged organelles, misfolded proteins, or invading pathogens. This process occurs within a double-membraned autophagosome, which fuses with lysosomes for degradation [70]. Autophagy-dependent cell death (ACD) represents a type of RCD governed by autophagic molecular processes. The Nomenclature Committee on Cell Death (NCCD) states that ACD is fundamentally different from classical types of cell death, like apoptosis and necrosis, and requires activation of the autophagy pathway or its key molecular components [19,71]. Based on substrate transport mechanisms, autophagy is primarily categorized into three major types: macroautophagy, microautophagy, and chaperone-mediated autophagy. Of these, macroautophagy represents the predominant form, commonly referred to simply as autophagy [72]. This process is strictly governed by a set of autophagy-related (ATG) proteins and occurs through five stages: induction, nucleation, elongation, maturation, and degradation (Figure 5). Under stress conditions, such as smoking, activation of AMPK or inhibition of mTORC1 leads to dissociation of mTORC1 from UNC-51-like kinase 1 (ULK1), resulting in ULK1 dephosphorylation and activation [73]. The activated ULK1 complex (ULK1-ATG13-FIP200-ATG101) subsequently recruits and activates the downstream Class-III PI3K complex (Beclin1-ATG14-VPS15-VPS34), which generates phosphatidylinositol 3-phosphate (PI3P) to initiate autophagosome nucleation [74]. Subsequently, microtubule-associated protein 1 light chain 3 (LC3) is cleaved by the ATG4 protease into LC3-I, which is then conjugated to phosphatidylethanolamine (PE) via the ATG12-ATG5-ATG16L1 ubiquitin-like conjugation system, forming membrane-bound LC3-II. LC3-II facilitates the extension and invagination of the phagophore membrane, ultimately enveloping substrates for degradation to form mature autophagosomes [75]. Autophagosomes are then fused with lysosomes to generate autolysosomes, which are responsible for the lysis of autophagosome contents using acidic hydrolases. The resulting metabolites, including amino acids and fatty acids, are recycled into cellular metabolic pathways, supplying both energy and biosynthetic precursors [76].

5.2. Autophagy in COPD

Accumulating evidence indicates that autophagy is dysregulated in COPD patients, exhibiting extremely dynamic and context-dependent bidirectional regulation. Compared to age-matched non-smokers, peripheral lung tissue from patients with severe COPD exhibits increased levels of p62, LC3, and protein aggregates, suggesting impaired autophagy in COPD [77]. Consistently, p62 accumulation in peripheral lung tissue positively correlates with pulmonary function severity and is closely associated with upregulation of LC3 and BICD1 expression [78]. In addition, alveolar macrophages from COPD patients and smokers display increased autophagosome formation, further supporting the notion that defective autophagic flux contributes to COPD pathogenesis [79]. Galectin-8 functions as a danger receptor, recognizing destroyed intracellular host vesicles that are damaged and recruits the autophagy adaptor protein NDP52 to facilitate forming and breaking down specific autophagosomes. Yuta Kono and associates [80] found lung homogenates from individuals with COPD demonstrated elevated galectin-8 and NDP52 levels, with significantly increased serum galectin-8 levels in those experiencing frequent acute exacerbations. At the cellular level, it has been demonstrated that CSE disrupts autophagic flux in U937 macrophage-like cells, resulting in the intracellular concentration of NDP52 and galactosidase-8. Conversely, chronic smoke exposure may also induce excessive autophagy activation, resulting in alveolar structural disruption, ciliary shortening, and impaired mucus clearance, thereby exacerbating airway remodeling and emphysema formation [81]. Bulk and single-cell RNA sequencing datasets from COPD and non-COPD tissues were examined by Liao et al. [82], who found significant upregulation of autophagy-related genes in COPD. Moreover, autophagy in macrophages and monocytes displayed extensive interactions with other cell types, highlighting its potential role as a key regulatory process in intercellular communication. Chen et al. [83] further demonstrated that mTOR deficiency enhances inflammatory responses, whereas autophagy inhibition via Atg5 deletion attenuates inflammation, suggesting that autophagy activation contributes to particulate aerosol-induced pulmonary inflammation.
Autophagy can be classified as either selective or non-selective forms based on the specificity of the degraded material [84]. Mitophagy, a selective type, specifically removes dysfunctional mitochondria to prevent ROS accumulation and apoptosis [85]. Abnormal activation of mitophagy is considered a key mechanism by which CS promotes COPD development. Yang et al. [86] demonstrated that ARSK alleviates COPD by inhibiting autophagy and restricting excessive mitophagy through suppression of Parkin Ser65 phosphorylation. Correspondingly, Wei and colleagues [87] revealed in a CS-induced mouse model that STAT3 activation enhances PINK1/Parkin pathway activity by upregulating TGF-β1, with excessive mitochondrial autophagy further inducing epithelial–mesenchymal transition (EMT) and small airway wall thickening. Beyond CS, environmental pollutants PM2.5 and microplastics also modulate mitochondrial autophagy through distinct mechanisms. Wang et al. [88] revealed that PM2.5 uncouples Klotho-mediated inhibition of IGF-1R, leading to Parkin-dependent mitophagy defects. Impaired clearance of damaged mitochondria subsequently triggers oxidative stress. Conversely, Wei et al. [89] demonstrated that polystyrene microplastics (PS-MPs) activate autophagy-dependent ferroptosis (ADF) via mitochondrial ROS accumulation, where NCOA4-mediated ferritinophagy releases free iron, exacerbating lipid peroxidation. These outcomes emphasize the complex and context-dependent function of mitochondrial autophagy in COPD. Insufficient mitophagy promotes oxidative stress, whereas excessive activation disrupts mitochondrial homeostasis. Despite operating through distinct mechanisms, both processes converge to drive airway remodeling and COPD progression [86].

6. Pyroptosis

6.1. Overview of Pyroptosis

The term “pyroptosis” was first introduced in 2005 [63]. Pyroptosis represents a type of RCD modulated by gasdermin (GSDM) proteins (Figure 6). It is structurally defined by chromatin condensation, cytoplasmic vesicle formation, and the rupturing of the plasma membrane. External factors like smoking and PM2.5 can trigger the release of pathogen-associated molecular patterns (PAMPs) or DAMPs. These molecules are recognized by pattern recognition receptors (PRRs), which trigger and induce the formation of inflammasomes [90,91,92,93,94]. Two major classes of PRRs involved in pyroptosis in COPD are nucleotide-binding domain leucine-rich repeat-containing proteins (NLRs) and AIM2-like receptors [95,96]. The formed inflammasome cleaves pro-caspase-1 into an active form, which then processes GSDM into the N-terminal fragment (GSDM-N) and matures pro-IL-1β and pro-IL-18. GSDM-N produces pores in the plasma membrane, enabling IL-1β/IL-18 release and amplifying inflammation [97,98,99]. In addition to this canonical pathway, pyroptosis can also occur through inflammasome-independent mechanisms. Intracellular lipopolysaccharide (LPS) activates caspase-4/5/11 directly, which then induces GSDMD cleavage and the release of GSDMD-N-terminal fragments [4,5].

6.2. Pyroptosis in COPD

Numerous chronic respiratory conditions, including COPD, have been linked to pyroptosis [100,101]. By 2013, studies demonstrated that IL-18 expression induces pulmonary inflammation, emphysematous changes, excessive mucus secretion, and fibrotic alterations in the blood vessels and airways of mice [102]. Nevertheless, the exact function of pyroptosis in COPD is still not fully determined. Faner and associates observed markedly elevated NLRP3, IL-1β, and IL-18 expressions in lung tissue from patients with stable COPD [103]. Subsequent studies further revealed that multiple pyroptosis-associated molecules, including NLRP3, caspase-1, and IL-18, are further upregulated during acute exacerbations of COPD [96,97]. Nicotine, a major constituent of CS, has been identified as a major driver of pyroptotic pathways [104]. Wang et al. [90] reported that nicotine exposure upregulates pyroptosis-associated molecules in 16HBE cells, potentially contributing to COPD progression. Similarly, Zhang et al. [105] demonstrated that CSE induces pyroptosis in 16HBE cells via the classical ROS/NLRP3/caspase-1 pathway, leading to GSDMD cleavage and release of IL-1β and IL-18. Rao et al. [106] further found that TRPV4 upregulation promotes pyroptosis by activating caspase-1 and increasing GSDM-N protein expression. Mechanistically, TRPV4 modulates the Ca2+/NLRP3/caspase-1 axis, while its inhibition attenuates NLRP3 inflammasome activation, lactate dehydrogenase release, and inflammatory cytokine production. While ferroptosis is defined by lipid peroxidation, lipid peroxidation can also initiate pyroptosis. Hou et al. [107] demonstrated that inhibiting xCT ubiquitination alleviates lipid peroxidation-mediated pyroptosis in COPD patients through downregulating the xCT/GSH/GPx4 axis. In vitro, Mo et al. [108] established a COPD cell model by treating the human lung fibroblast line MRC-5 with lipopolysaccharide and demonstrated that lncRNA GAS5 overexpression facilitates pyroptosis in pulmonary fibroblasts through activation of the miR-223-3p/NLRP3 axis. However, other studies yielded conflicting findings. One study reported that CSE suppresses NLRP3 inflammasome expression and its downstream cytokines IL-1β and IL-18 at the transcriptional level, while simultaneously activating caspase-1 via the noncanonical TLR4/TRIF/caspase-8 pathway [109]. This unique caspase-1 activation reduced glycolytic flux in macrophages, suggesting distinct and context-dependent cellular responses to CSE that require further investigation.

7. Other Forms of RCD

7.1. Cuproptosis

Cuproptosis (copper-induced cell death) denotes a recently identified form of programmed cell death, first described in 2022 by Tsvetkov and colleagues [110]. Its core mechanism involves the excessive binding of intracellular copper ions (Cu2+) to lipoylated proteins in the mitochondrial tricarboxylic acid (TCA) cycle, forming protein aggregation and destabilizing iron-sulfur (Fe-S) cluster proteins. This cascade induces proteotoxic stress, impairs mitochondrial respiration, and ultimately triggers cell death [111]. In contrast to other types of cell death, like ferroptosis and apoptosis, cuproptosis depends on copper–protein interactions and TCA cycle dysfunction, and cannot be blocked by conventional cell death inhibitors [112]. Evidence from COPD patients and cigarette smoke exposure models indicates significant dysregulation of cuproptosis-associated genes, including GLS, DLD, CDKN2A, and DLST. Among these, GLS expression is reduced and correlates with lung function decline, whereas DLD and CDKN2A are upregulated in response to smoking and related factors [113,114]. These genes not only serve as diagnostic biomarkers for COPD but are also implicated in copper homeostasis and cuproptosis-associated pathways, thereby offering potential avenues for targeted therapy [115]. Nevertheless, the exact mechanisms by which cuproptosis participates in the development of COPD pathogenesis remain unresolved, underscoring the need for further mechanistic studies and the development of targeted intervention strategies.

7.2. Parthanatos

Parthanatos is classified as a type of RCD that is reliant on Poly(ADP-ribose) (PAR) polymerase-1 (PARP-1). PARP1 is a chromatin-associated nuclear protein that significantly participates in DNA repair during strand breaks [116,117,118]. Parthanatos can be induced by oxidative stress, such as that caused by smoking [119]. ROS can induce DNA damage, leading to the overactivation of PARP1 and the accumulation of PAR. This cascade promotes the nuclear translocation of mitochondrial proteins, including apoptosis-inducing factor (AIF) and Endonuclease G (EndoG). Once in the nucleus, AIF and EndoG cause chromatin dissolution and cell death [120]. Studies have shown that the EndoG inhibitor BMN673 can reduce levels of AIF and EndoG in epithelial cells [121]. However, BMN673’s potential role in modulating parthanatos and attenuating cell death in COPD remains a subject for further investigation [121].

8. Therapeutic Potential

8.1. Targeting Apoptosis

Research has demonstrated that multiple compounds confer anti-apoptotic effects by selectively modulating key pathways involved in apoptosis (Table 1). As previously noted, Bax and Bcl-2 are critical proteins of the Bcl-2 protein family, functioning as pro-apoptotic and anti-apoptotic factors, respectively, and jointly mediating the process of programmed cell death through the mitochondrial apoptosis pathway. Asiaticoside activates the PGC-1α/Nrf2 pathway, resulting in downregulation of Bax and upregulation of Bcl-2. This action not only mitigates inflammation but also suppresses EMT, thereby reducing small airway wall thickening in CS-induced lung tissue [122]. Another study showed that Korean Red Ginseng can also counteract the toxicity of cigarette smoke condensate (CSC), significantly reducing apoptosis levels in NCI-H292 cells and mouse lung tissue by inhibiting the overactivation of the VEGF/PI3K/AKT pathway, which alleviates alveolar damage and emphysema [123]. Caspase-3, the central executioner of apoptosis, integrates signals from multiple pathways and induces DNA fragmentation, leading to cell death. Formononetin (FMN) [124] and N-acetyl-L-cysteine (NAC) [125] were reported to inhibit the AhR/CYP1A1/AKT/mTOR pathway, while NAC also replenishes GSH and suppresses the p53 pathway. Both compounds significantly reduce Caspase-3 activation, alleviating alveolar damage and demonstrating therapeutic potential for restoring lung function.
Mounting evidence further highlights oxidative stress as a key regulator of apoptosis, making it an attractive therapeutic target [129]. Anthrahydroquinone-2,6-disulfonate (AH2QDS) is a labdane diterpene lactone compound derived from Andrographis paniculata that exhibits anti-inflammatory, antioxidant, and antiviral activities. Li and colleagues [126] demonstrated that AH2QDS activates the Keap1/Nrf2 pathway, upregulating downstream antioxidant proteins HO-1 and NQO1, which enhances cellular antioxidant capacity, reduces inflammation, and decreases apoptosis in rat lung tissue induced by LPS and CS, ultimately contributing to the restoration of lung function. Progesterone (P4), a key steroid hormone, was documented to also protect breast cancer cells and uterine leiomyoma tissue from oxidative stress-induced apoptosis [130]. Li et al. [128] further reported that P4 activates the c-MYC/SIRT1/PGC-1α pathway to maintain the function of the mitochondria and reduce H2O2-induced apoptosis in BEAS-2B cells and airway smooth muscle cells. This suggests that P4 may have therapeutic potential beyond reproductive health, particularly in ameliorating the symptoms and advancement of COPD. In addition, Gaylussacin, a potential therapeutic agent for COPD, markedly suppresses MMP-12 expression and activity in macrophages by activating the SIRT1 pathway, thereby mitigating pulmonary inflammation, oxidative stress, and apoptosis in models induced by lead/cadmium (Pb/Cd) or porcine pancreatic elastase (PPE). These effects lead to improved lung function and slowed disease progression. Additionally, its favorable oral bioavailability and safety profile further support its therapeutic promise [127].

8.2. Targeting Necroptosis

Intervening in necroptosis-related key kinases has shown therapeutic potential in COPD (Table 2). GSK’547 is a RIPK1 inhibitor that significantly reduces elastase-induced emphysema and lung function decline, diminishes CS-induced peri-bronchial inflammatory cell infiltration, airway collagen deposition, and emphysema-like changes [52]. Necrostatin-1 (NEC-1), another RIPK1 inhibitor, has been reported to suppress cigarette smoke-induced inflammatory cytokine secretion from bone marrow–derived macrophages (BMDMs) [51]. However, Chen et al. [56] suggested that NEC-1 does not alleviate cigarette-induced emphysema and airway inflammation, and hypothesized that in COPD, there may exist mechanisms of RIPK3 activation that are independent of RIPK1. In contrast, RIPK3 inhibitor GSK’872 significantly reduces smoking-induced lung macrophage and epithelial cell death [51,56] and markedly improves COPD-related pathological changes [56]. The discrepancy in findings regarding NEC-1’s effects on CSE-induced cell death may result from differing assessment methods. To further clarify RIPK1’s role, gene knockout models are needed. Notably, Hannelore et al. [52] further employed single-cell sequencing and demonstrated elevated RIPK1 expression in multiple lung cell types in both COPD patients and experimental mouse models. Genetic or pharmacological inhibition of RIPK1 using GSK’ 547 markedly lowered airway inflammation following acute and subacute exposure to CS, reduced apoptosis and necrosis induced by chronic exposure, and mitigated airway remodeling and emphysema. Collectively, these findings reveal that RIPK1 is upregulated in COPD and represents a potential therapeutic target, though its interaction with RIPK3 remains to be fully defined.
Additionally, traditional Chinese medicine extracts targeting necroptosis have also shown promising effects in COPD treatment. Theaflavin 3,3′ digallate (TF-3), an extract from black tea, significantly improves CS-induced lung inflammation and emphysema. TF-3 inhibits necroptosis by reducing RIPK3 and MLKL phosphorylation [131]. Thymoquinone (Tq), an active ingredient extracted from black cumin, inhibits RIPK1 and RIPK3 expression, reduces MLKL phosphorylation, and attenuates CSE-induced epithelial cell death. Its protective effects on epithelial cells warrant further investigation in preclinical studies [132].

8.3. Targeting Ferroptosis

Various chemical agents targeting ferroptosis reduce cell death and ameliorate COPD pathology [133,134,135] (Table 3). Ferrostatin-1 (Fer-1) is a ferroptosis inhibitor. Deferoxamine (DFO) chelates iron, reducing intracellular iron levels. Both Fer-1 and DFO markedly reduce CS-induced epithelial cell death, which in turn decreases inflammation and potentially helps mitigate inflammatory cell infiltration and lung injury in COPD [135,136,137,138]. In addition, Fer-1 inhibits macrophage M2 polarization induced by epithelial cells [139]. Plant extracts can also improve COPD pathology by regulating ferroptosis. Ginsenoside Rg1 [140], Ginkgo biloba extract (GBE) [141], and Sea buckthorn extract (SBE) [142] can effectively reduce inflammatory cell infiltration and promote alveolar repair by triggering the Nrf2/SLC7A11/GPX4 pathway and inhibiting the occurrence of ferroptosis. Acacetin [143], Curcumin (CUR) [136], scutellarein (STR) [36], and dihydroquercetin (DHQ) [144] are natural compounds that significantly inhibit CS-induced ferroptosis, thereby alleviating airway inflammation and ameliorating emphysema. CUR and DHQ further enhance cellular antioxidant capacity by promoting System Xc and glutathione (GSH) expression [36,136]. In addition to promoting GSH and NRF2, DHQ also inhibits ALOX15, reducing lipid peroxidation [144]. Other antioxidants, including hydrogen sulfide (H2S) and sodium pyruvate (NaPyr), exert similar protective effects by upregulating NRF2, GSH, and GPX4, suppressing CS-induced epithelial ferroptosis and inflammation, and alleviating PM2.5-triggered airway injury [145]. Moreover, Lipoxin A4 (LXA4), an endogenous lipid mediator produced through lipoxygenase-dependent metabolism of arachidonic acid, exhibits both anti-inflammatory and antioxidant effects [146]. Li et al. [147] demonstrated that LXA4 activates the ALX/FPR2 receptor and inhibits p38/MAPK phosphorylation, suppressing ferroptosis and inflammatory responses while improving lung function and tissue integrity. However, clinical translation of LXA4 is hindered by its rapid inactivation and short half-life. Consequently, developing stabilized LXA4 analogs represents a potentially viable treatment approach for COPD.

8.4. Targeting Autophagy

Various agents can precisely modulate the balance between LC3B II and p62 expression, restoring autophagic homeostasis and alleviating lung injury (Table 4). The traditional Chinese medicine formula Bufei Yishen Formula (BYF) and its active components effectively upregulate LC3B II and downregulate p62, activating the autophagy pathway to promote the clearance of damaged organelles and suppress cellular senescence. It also mitigates inflammation and oxidative stress, preserves epithelial barrier function, and improves lung performance [148,149]. Similarly, natural compounds such as dihydromyricetin (DHM) [150] and quercetin [151], as well as the sildenafil analog vardenafil [152], can restore autophagy function, clear CS-induced lung tissue damage components, and improve lung function. DHM additionally reduces mucus hypersecretion and repairs ciliary dysfunction by rebalancing autophagy [150]. Conversely, when excessive autophagy contributes to cell injury, diindolylmethane (DIM) has demonstrated regulatory effects. Jung et al. [153] showed that in HEL299 human lung fibroblasts exposed to cadmium, a recognized risk factor for COPD, DIM suppresses excessive autophagy and attenuates autophagy-related cell death, revealing its potential as a viable treatment candidate for cadmium-associated diseases, including COPD.
Pharmacological regulation of mitophagy has also been recognized as a therapeutic avenue for COPD, either by repressing its overactivation or restoring its impaired activity. Puerarin, an isoflavone derived from kudzu root, exhibits antioxidant and anti-apoptotic properties. By activating the PI3K/AKT/mTOR pathway, it inhibits CSE-induced excessive mitophagy, as demonstrated by Wang et al. [154], thereby rescuing mitochondrial function and protecting bronchial epithelial cells from CSE-induced injury, providing a novel potential strategy for COPD therapy. In contrast, hydrogen sulfide (H2S) has been reported to enhance impaired Parkin-mediated mitophagy in a PM2.5 model by upregulating Klotho expression and inhibiting IGF-1R, clearing abnormal mitochondria, and alleviating cellular senescence and inflammation [88]. These effects significantly mitigate emphysema and airway inflammation, inhibit COPD progression, and underscore the potential clinical applications of H2S-based therapies. Collectively, these findings emphasize the importance of maintaining mitophagy at an appropriate level to preserve mitochondrial homeostasis and slow disease progression.

8.5. Targeting Pyroptosis

The strategy of suppressing pyroptosis by inhibiting NLRP3 inflammasome activation, blocking inflammatory caspase maturation, and preventing GSDMD cleavage offers a promising therapeutic approach for COPD (Table 5). A variety of natural plant extracts, including Resveratrol [155], DHM [156], Schisandrin A (SchA) [157], Tanshinone(TS) [158], grape seed proanthocyanidin extract(GSPE) [159], Daphnetin(Daph) [160], osthole [161], the flower buds of Tussilago farfara L (FTF) [162], (−)-Epicatechin(EC) [163], and isoforskolin (ISOF) [164], have demonstrated therapeutic efficacy in COPD treatment. These extracts inhibit lung cell pyroptosis by suppressing the NLRP3 inflammasome, alleviating airway inflammation and emphysema, and improving lung function. For instance, DHM can directly bind to and stabilize SLC7A11, enhancing glutathione synthesis and subsequently inhibiting lipid peroxidation and pyroptosis [156]. SchA also activates the NRF2/HO-1 signaling pathway, an important target in COPD treatment [157], whereas TS inhibits both NF-κB activation and the secretion of inflammatory cytokines IL-6 and IL-8 [158]. Daph additionally reduces airway inflammation and mucus hypersecretion resulting from PM2.5 exposure or cigarette smoke [160]. Consistent with these findings, traditional Chinese medicine exhibits complex pharmacological properties, particularly in regulating pyroptosis. Both Astragaloside IV [165] and Tianlong Kechuanling (TL) [166] were reported to control the NLRP3-GSDMD signaling pathway, thus participating in the regulation of pyroptosis. Notably, TL inhibits pyroptosis in pulmonary vascular endothelial cells, alleviates inflammatory responses and vascular remodeling, and consequently reduces pulmonary arterial hypertension while improving pulmonary function. It shows great clinical promise for COPD complicated by pulmonary arterial hypertension.
In addition, other chemical agents targeting pyroptosis have also shown potential in COPD therapy [168]. Hydrogen sulfide, a chemical agent, alleviates smoking-induced lung inflammation [169,171]. Magnesium isoglycyrrhizinate (MgIG), a derivative of glycyrrhizic acid, mitigates smoking and LPS-induced lung function decline, suppressing airway inflammation, remodeling, and emphysema [172]. A range of specific inhibitors has currently been developed primarily for use in experimental research settings, such as the NLRP3 inhibitor MCC950 [170] and the caspase-1 inhibitor VX-765 [105], both of which markedly attenuate lung inflammation. As previously noted, GSDMD is the central executor of pyroptosis, mediating lytic and inflammatory cell death. Gao et al. [167] demonstrated that disulfiram (DSF) directly targets GSDMD and, through covalent binding, prohibits the pore-forming activity of its N-terminal fragment (GSDMD-NT), thus preserving airway epithelial barrier integrity and attenuating ozone-induced COPD pathology and lung function impairment. Given its established safety profile as an approved drug, DSF holds significant translational potential as a therapeutic agent for COPD. However, its efficacy and mechanism of action in CS-induced models should be well-established in future research to support clinical application. Notably, although AIM2-mediated pyroptosis also participates in the development of COPD [96,173], little is currently known about pharmacological agents capable of regulating the AIM2 inflammasome.

9. The Future of Translational Research in RCD for COPD

Beyond their mechanistic roles, RCD pathways offer significant translational potential. Insights from therapeutic targeting of RCD in oncology provide a conceptual framework for developing novel COPD treatments, particularly through the identification of prognostic biomarkers and the design of pharmacological modulators. In colorectal and hepatocellular carcinoma [174,175], RCD-based prognostic models not only facilitate patient stratification but also uncover differential sensitivities to chemotherapy and immunotherapy. Comparable strategies are now being applied to respiratory diseases. For instance, a seven-gene “cell death score” stratifies risk and predicts therapy response in idiopathic pulmonary fibrosis [176], while a pyroptosis-related gene set was shown to predict 28-day survival and correlates with immune cell infiltration in sepsis-induced acute respiratory distress syndrome (ARDS) [177]. In COPD, serum ferroptosis-related biomarkers such as sTfR1 and GPX4 have been identified as independent predictors of exercise capacity and exacerbation risk [178]. These findings highlight the potential of molecular signatures related to regulated cell death to serve as tools for risk stratification, disease monitoring, and personalized therapy in COPD.
Encouragingly, both preclinical and early clinical studies have begun to validate several regulators of RCD as potential therapeutic targets. NLRP3 inhibitors, such as NT-0796 [179] and DFV890 [180], have demonstrated favorable safety and pharmacodynamic profiles, including central nervous system penetration and dose-dependent suppression of IL-1β in early human trials. In parallel, activators of the Nrf2 pathway such as dimethyl fumarate (DMF) and its metabolite monomethyl fumarate (MMF) exhibit combined antioxidant and anti-inflammatory properties [181]. In experimental models of COPD, DMF attenuates oxidative stress, reduces macrophage infiltration, and partially reverses emphysematous changes. Given its established clinical use in multiple sclerosis [182] and psoriasis [183], DMF represents a particularly promising candidate for translational application in COPD. Furthermore, RIPK1 inhibitors, including eclitasertib [184], SIR2446M [185], and GFH312 [186] have shown potent target engagement and favorable tolerability in early-phase trials, supporting the feasibility of pharmacologically modulating necroptosis and inflammation through selective kinase inhibition.

10. Conclusions

Disruption of apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and parthanatos is well established as a driver of COPD progression. In contrast, autophagy exhibits a context-dependent dual role: protective in early stages by clearing damaged organelles and alleviating stress, yet potentially detrimental in advanced disease when dysregulated or impaired [187]. This Janus-faced nature of autophagy highlights the importance of considering timing, extent, and microenvironmental context when designing therapeutic strategies that target RCD in COPD.
While distinct molecular mechanisms independently regulate various kinds of RCD, they also have potential interconnections. Multiple RCD pathways can be initiated by common external stimuli, such as ROS, as seen in ferroptosis and parthanatos [18]. Oxeiptosis is another ROS-initiated RCD, though its role in COPD remains unclear. DAMPs and PAMPs that activate pyroptosis can be released by cells undergoing necroptosis [121]. Necroptosis itself can be triggered by apoptosis inhibitors. Additionally, key components regulating different RCDs may overlap. For instance, RIPK1, a central mediator of necroptosis, and AIF, a key player in parthanatos, can also mediate apoptosis [52,188]. Notably, pyroptosis and ferroptosis are closely linked. Inactivation of GPX4 during ferroptosis causes lipid peroxidation that activates caspase 11 and gasdermin D cleavage, thereby inducing pyroptosis. Conversely, activation of the NLRP3 inflammasome during pyroptosis can promote iron accumulation and lipid peroxidation, potentially leading to ferroptosis [189]. Thus, the independent yet intertwined relationships between various RCDs present both opportunities and challenges for targeting RCDs in COPD treatment.
The temporal relationships among different RCD pathways in COPD remain unclear, representing a major conceptual gap. Most studies offer static observations within isolated disease stages, making it difficult to define their sequence or dominance. However, it can be speculated that apoptosis and autophagy likely predominate early in response to cigarette smoke, supporting repair and clearance, while necroptosis, pyroptosis, and ferroptosis become more prominent as oxidative stress and inflammation progress. These pathways may act concurrently rather than sequentially. Time-resolved preclinical studies are needed to clarify the dynamics of these RCD pathways. Insights from such studies may help define diagnostic and therapeutic windows, thereby facilitating the development of stage-specific biomarkers and targeted treatment strategies.
This review summarizes the roles of diverse RCD pathways in the pathogenesis of COPD and highlights the possible therapeutic modulation of these processes. The development of novel COPD treatments may be facilitated by the modification of key components within RCD pathways. However, whether other emerging RCD pathways, like oxeiptosis and alkaliptosis, also play roles in COPD pathogenesis remains an area for future research.

Author Contributions

Q.H. and J.X. developed the research and revised the manuscript. H.F. drafted the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was endorsed by the National Natural Science Foundation of China (No. 82170049, 81973986), the Clinical Collaboration Project of Traditional Chinese and Western Medicine in the Major Difficult Diseases in Hubei Province (Respiratory system Diseases), the Leading talents of public health in Hubei Province (2022SCZ047), the Project of Key R&D Program in Hubei Province (2023BCB127) and the Major Project of National Science and Technology (2023ZD0506300).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

Regulated cell death (RCD), chronic obstructive pulmonary disease (COPD), accidental cell death (ACD), mitochondrial outer membrane permeabilisation (MOMP), cytochrome c (Cyto-C), TNF receptor superfamily member 1A(TNFRSF1A), death-inducing signal complex (DISC), Unc-5 Netrin Receptor B (UNC5B), DCC Netrin 1 Receptor (DCC), death-associated protein kinase 1 (DAPK1), cigarette smoke extract (CSE), epithelial–mesenchymal transition (EMT), unfolded protein response (UPR), human airway smooth muscle cells (HASMCs), damage-associated molecular patterns (DAMPs), TNF receptor-associated death domain protein (TRADD), phosphorylated RIPK3 (p-RIPK3), phosphorylated MLKL (p-MLKL), mitochondrial DNA (mtDNA), Ferric iron (Fe3+), transferrin receptors (TFRs), ferrous iron (Fe2+), glutathione (GSH), phospholipid hydroperoxides (PL-OOH), glutathione peroxidase 4 (GPX4), oxygen species (ROS), polyunsaturated fatty acids (PUFAs), Autophagic cell death (ACD), Nomenclature Committee on Cell Death (NCCD), autophagy-related (ATG), UNC-51-like kinase 1 (ULK1), phosphatidylinositol 3-phosphate (PI3P), light chain 3 (LC3), phosphatidylethanolamine (PE), polystyrene microplastics (PS-MPs), autophagy-dependent ferroptosis (ADF), gasdermin (GSDM), pathogen-associated molecular patterns (PAMPs), leucine-rich repeat-containing proteins (NLRs), N-terminal fragment (GSDM-N), lipopolysaccharide (LPS), apoptosis-inducing factor (AIF), Endonuclease G (EndoG), cigarette smoke condensate(CSC), porcine pancreatic elastase (PPE), bone marrow-derived macrophages (BMDMs), acute respiratory distress syndrome (ARDS), dimethyl fumarate (DMF), metabolite monomethyl fumarate (MMF).

References

  1. de Oca, M.M.; Perez-Padilla, R.; Celli, B.; Aaron, S.D.; Wehrmeister, F.C.; Amaral, A.F.S.; Mannino, D.; Zheng, J.; Salvi, S.; Obaseki, D.; et al. The global burden of COPD: Epidemiology and effect of prevention strategies. Lancet Respir. Med. 2025, 13, 709–724. [Google Scholar] [CrossRef]
  2. Agustí, A.; Melén, E.; DeMeo, D.L.; Breyer-Kohansal, R.; Faner, R. Pathogenesis of chronic obstructive pulmonary disease: Understanding the contributions of gene-environment interactions across the lifespan. Lancet Respir. Med. 2022, 10, 512–524. [Google Scholar] [CrossRef] [PubMed]
  3. Stolz, D.; Mkorombindo, T.; Schumann, D.M.; Agusti, A.; Ash, S.Y.; Bafadhel, M.; Bai, C.; Chalmers, J.D.; Criner, G.J.; Dharmage, S.C.; et al. Towards the elimination of chronic obstructive pulmonary disease: A Lancet Commission. Lancet 2022, 400, 921–972. [Google Scholar] [CrossRef] [PubMed]
  4. Barnes, P.J. Oxidative stress-based therapeutics in COPD. Redox Biol. 2020, 33, 101544. [Google Scholar] [CrossRef] [PubMed]
  5. Brightling, C.; Greening, N. Airway inflammation in COPD: Progress to precision medicine. Eur. Respir. J. 2019, 54, 1900651. [Google Scholar] [CrossRef]
  6. Rao, W.; Wang, S.; Duleba, M.; Niroula, S.; Goller, K.; Xie, J.; Mahalingam, R.; Neupane, R.; Liew, A.-A.; Vincent, M.; et al. Regenerative Metaplastic Clones in COPD Lung Drive Inflammation and Fibrosis. Cell 2020, 181, 848–864.E18. [Google Scholar] [CrossRef]
  7. Sauler, M.; Bazan, I.S.; Lee, P.J. Cell Death in the Lung: The Apoptosis-Necroptosis Axis. Annu. Rev. Physiol. 2019, 81, 375–402. [Google Scholar] [CrossRef]
  8. Zhou, Y.; Xiang, Y.; Liu, S.; Li, C.; Dong, J.; Kong, X.; Ji, X.; Cheng, X.; Zhang, L. RIPK3 signaling and its role in regulated cell death and diseases. Cell Death Discov. 2024, 10, 200. [Google Scholar] [CrossRef]
  9. Tummers, B.; Green, D.R. The evolution of regulated cell death pathways in animals and their evasion by pathogens. Physiol. Rev. 2022, 102, 411–454. [Google Scholar] [CrossRef]
  10. Hadian, K.; Stockwell, B.R. The therapeutic potential of targeting regulated non-apoptotic cell death. Nat. Rev. Drug Discov. 2023, 22, 723–742. [Google Scholar] [CrossRef]
  11. Minagawa, S.; Yoshida, M.; Araya, J.; Hara, H.; Imai, H.; Kuwano, K. Regulated Necrosis in Pulmonary Disease. A Focus on Necroptosis and Ferroptosis. Am. J. Respir. Cell Mol. Biol. 2020, 62, 554–562. [Google Scholar] [CrossRef]
  12. Feng, Y.; Li, M.; Yangzhong, X.; Zhang, X.; Zu, A.; Hou, Y.; Li, L.; Sun, S. Pyroptosis in inflammation-related respiratory disease. J. Physiol. Biochem. 2022, 78, 721–737. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, S.; Jia, J.; Zheng, J.; Zhou, Y.; Jia, D.; Wang, J. Recent Progress of Ferroptosis in Lung Diseases. Front. Cell Dev. Biol. 2021, 9, 789517. [Google Scholar] [CrossRef] [PubMed]
  14. Ornatowski, W.; Lu, Q.; Yegambaram, M.; Garcia, A.E.; Zemskov, E.A.; Maltepe, E.; Fineman, J.R.; Wang, T.; Black, S.M. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol. 2020, 36, 101679. [Google Scholar] [CrossRef] [PubMed]
  15. Shen, Z.; Liu, Z.; Cai, S.; Fu, H.; Gan, Y.; Li, X.; Wang, X.; Liu, C.; Ma, W.; Chen, J.; et al. Copper homeostasis and cuproptosis in myocardial infarction: Molecular mechanisms, treatment strategies and potential therapeutic targets. Front. Pharmacol. 2025, 16, 1525585. [Google Scholar] [CrossRef]
  16. Liu, C.; Lai, F.; Zhang, T.; Mao, K.; Wan, H.; He, Y. Roles and therapeutic potential of PARP-1 in neurodegenerative diseases. Biochem. Pharmacol. 2025, 242, 117373. [Google Scholar] [CrossRef]
  17. 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]
  18. 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]
  19. 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]
  20. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
  21. Cao, Y.; Yang, S.; Baima, Q.; Zhen, Y.; Hang, X.; Meng, X. Dual role mechanisms of regulated cell death in apical periodontitis: From pathogenic destruction to therapeutic potential. Cell Death Discov. 2025, 11, 386. [Google Scholar] [CrossRef] [PubMed]
  22. Vogler, M.; Braun, Y.; Smith, V.M.; Westhoff, M.-A.; Pereira, R.S.; Pieper, N.M.; Anders, M.; Callens, M.; Vervliet, T.; Abbas, M.; et al. The BCL2 family: From apoptosis mechanisms to new advances in targeted therapy. Signal Transduct. Target. Ther. 2025, 10, 91. [Google Scholar] [CrossRef]
  23. Yang, C.-Y.; Lien, C.-I.; Tseng, Y.-C.; Tu, Y.-F.; Kulczyk, A.W.; Lu, Y.-C.; Wang, Y.-T.; Su, T.-W.; Hsu, L.-C.; Lo, Y.-C.; et al. Deciphering DED assembly mechanisms in FADD-procaspase-8-cFLIP complexes regulating apoptosis. Nat. Commun. 2024, 15, 3791. [Google Scholar] [CrossRef] [PubMed]
  24. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  25. Kesavardhana, S.; Malireddi, R.K.S.; Kanneganti, T.-D. Caspases in Cell Death, Inflammation, and Pyroptosis. Annu. Rev. Immunol. 2020, 38, 567–595. [Google Scholar] [CrossRef]
  26. Hodge, S.; Hodge, G.; Holmes, M.; Reynolds, P.N. Increased airway epithelial and T-cell apoptosis in COPD remains despite smoking cessation. Eur. Respir. J. 2005, 25, 447–454. [Google Scholar] [CrossRef]
  27. Chen, L.; Luo, L.; Kang, N.; He, X.; Li, T.; Chen, Y. The Protective Effect of HBO1 on Cigarette Smoke Extract-Induced Apoptosis in Airway Epithelial Cells. Int. J. Chron. Obstruct. Pulmon. Dis. 2020, 15, 15–24. [Google Scholar] [CrossRef]
  28. Song, Q.; Chen, P.; Liu, X.-M. The role of cigarette smoke-induced pulmonary vascular endothelial cell apoptosis in COPD. Respir. Res. 2021, 22, 39. [Google Scholar] [CrossRef]
  29. Tran, T.T.V.; Jeong, Y.; Kim, S.; Yeom, J.E.; Lee, J.; Lee, W.; Bae, G.; Kang, J. PRMT1 Ablation in Endothelial Cells Causes Endothelial Dysfunction and Aggravates COPD Attributable to Dysregulated NF-κB Signaling. Adv. Sci. 2025, 12, 2411514. [Google Scholar] [CrossRef]
  30. Kasahara, Y.; Tuder, R.M.; Cool, C.D.; Lynch, D.A.; Flores, S.C.; Voelkel, N.F. Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphysema. Am. J. Respir. Crit. Care Med. 2001, 163, 737–744. [Google Scholar] [CrossRef]
  31. Wang, C.; Zhou, J.; Wang, J.; Li, S.; Fukunaga, A.; Yodoi, J.; Tian, H. Progress in the mechanism and targeted drug therapy for COPD. Signal Transduct. Target. Ther. 2020, 5, 248. [Google Scholar] [CrossRef] [PubMed]
  32. Lu, F.Y.; Chen, R.; Zhou, M.; Guo, Y. Hedgehog signaling modulates cigarette-induced COPD development. Exp. Ther. Med. 2021, 22, 729. [Google Scholar] [CrossRef] [PubMed]
  33. Li, J.; Zong, D.; Chen, Y.; Chen, P. Anti-apoptotic effect of the Shh signaling pathway in cigarette smoke extract induced MLE 12 apoptosis. Tob. Induc. Dis. 2019, 17, 49. [Google Scholar] [CrossRef] [PubMed]
  34. Fu, H.; Liu, K.; Zheng, Y.; Zhao, J.; Xie, T.; Ding, Y. Upregulation of ARHGAP18 by miR-613 Inhibits Cigarette Smoke Extract-Induced Apoptosis and Epithelial-Mesenchymal Transition in Bronchial Epithelial Cells. Int. J. Chron. Obstruct. Pulmon. Dis. 2025, 20, 2525–2537. [Google Scholar] [CrossRef]
  35. Li, T.; He, X.; Luo, L.; Zeng, H.; Ren, S.; Chen, Y. F-Box Protein FBXW17-Mediated Proteasomal Degradation of Protein Methyltransferase PRMT6 Exaggerates CSE-Induced Lung Epithelial Inflammation and Apoptosis. Front. Cell Dev. Biol. 2021, 9, 599020. [Google Scholar] [CrossRef]
  36. Liu, L.; Zhang, Y.; Wang, L.; Liu, Y.; Chen, H.; Hu, Q.; Xie, C.; Meng, X.; Shen, X. Scutellarein alleviates chronic obstructive pulmonary disease through inhibition of ferroptosis by chelating iron and interacting with arachidonate 15-lipoxygenase. Phytother. Res. 2023, 37, 4587–4606. [Google Scholar] [CrossRef]
  37. Xu, J.; Zeng, Q.; Li, S.; Su, Q.; Fan, H. Inflammation mechanism and research progress of COPD. Front. Immunol. 2024, 15, 1404615. [Google Scholar] [CrossRef]
  38. Byrne, A.J.; Mathie, S.A.; Gregory, L.G.; Lloyd, C.M. Pulmonary macrophages: Key players in the innate defence of the airways. Thorax 2015, 70, 1189–1196. [Google Scholar] [CrossRef]
  39. Wang, Z.; Zheng, T.; Zhu, Z.; Homer, R.J.; Riese, R.J.; Chapman, H.A.; Shapiro, S.D.; Elias, J.A. Interferon gamma induction of pulmonary emphysema in the adult murine lung. J. Exp. Med. 2000, 192, 1587–1600. [Google Scholar] [CrossRef]
  40. Kheradmand, F.; Zhang, Y.; Corry, D.B. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol. Rev. 2023, 103, 1059–1093. [Google Scholar] [CrossRef]
  41. Yu, Y.; Yang, A.; Yu, G.; Wang, H. Endoplasmic Reticulum Stress in Chronic Obstructive Pulmonary Disease: Mechanisms and Future Perspectives. Biomolecules 2022, 12, 1637. [Google Scholar] [CrossRef]
  42. Feng, H.; Li, M.; Altawil, A.; Yin, Y.; Zheng, R.; Kang, J. Cigarette smoke extracts induce apoptosis in Raw264.7 cells via endoplasmic reticulum stress and the intracellular Ca2+/P38/STAT1 pathway. Toxicol. Vitr. 2021, 77, 105249. [Google Scholar] [CrossRef]
  43. Chen, R.; Michaeloudes, C.; Liang, Y.; Bhavsar, P.K.; Chung, K.F.; Ip, M.S.M.; Mak, J.C.W. ORMDL3 regulates cigarette smoke–induced endoplasmic reticulum stress in airway smooth muscle cells. J. Allergy Clin. Immunol. 2022, 149, 1445–1457.e5. [Google Scholar] [CrossRef] [PubMed]
  44. Degterev, A.; Huang, Z.; Boyce, M.; Li, Y.; Jagtap, P.; Mizushima, N.; Cuny, G.D.; Mitchison, T.J.; Moskowitz, M.A.; Yuan, J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005, 1, 112–119. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, Z.; Zhang, F.; Xie, W.; Niu, Y.; Wang, H.; Li, G.; Zhao, L.; Wang, X.; Xie, W. Induced Necroptosis and Its Role in Cancer Immunotherapy. Int. J. Mol. Sci. 2024, 25, 10760. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, M.; Chen, W.; He, L.; Liu, D.; Zhao, L.; Wang, X. A Glimpse of necroptosis and diseases. Biomed. Pharmacother. 2022, 156, 113925. [Google Scholar] [CrossRef]
  47. Seo, J.; Nam, Y.W.; Kim, S.; Oh, D.-B.; Song, J. Necroptosis molecular mechanisms: Recent findings regarding novel necroptosis regulators. Exp. Mol. Med. 2021, 53, 1007–1017. [Google Scholar] [CrossRef]
  48. Zhu, T.; Wu, B.-W. Recognition of necroptosis: From molecular mechanisms to detection methods. Biomed. Pharmacother. 2024, 178, 117196. [Google Scholar] [CrossRef]
  49. Chen, Y.; Gu, Y.; Xiong, X.; Zheng, Y.; Liu, X.; Wang, W.; Meng, G. Roles of the adaptor protein tumor necrosis factor receptor type 1-associated death domain protein (TRADD) in human diseases. Biomed. Pharmacother. 2022, 153, 113467. [Google Scholar] [CrossRef]
  50. Lu, Z.; Van Eeckhoutte, H.P.; Liu, G.; Nair, P.M.; Jones, B.; Gillis, C.M.; Nalkurthi, B.C.; Verhamme, F.; Buyle-Huybrecht, T.; Vandenabeele, P.; et al. Necroptosis Signaling Promotes Inflammation, Airway Remodeling, and Emphysema in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2021, 204, 667–681. [Google Scholar] [CrossRef]
  51. Wang, Y.; Wang, X.-K.; Wu, P.-P.; Wang, Y.; Ren, L.-Y.; Xu, A.-H. Necroptosis Mediates Cigarette Smoke-Induced Inflammatory Responses in Macrophages. Int. J. Chron. Obstruct. Pulmon. Dis. 2020, 15, 1093–1101. [Google Scholar] [CrossRef]
  52. Van Eeckhoutte, H.P.; Donovan, C.; Kim, R.Y.; Conlon, T.M.; Ansari, M.; Khan, H.; Jayaraman, R.; Hansbro, N.G.; Dondelinger, Y.; Delanghe, T.; et al. RIPK1 kinase-dependent inflammation and cell death contribute to the pathogenesis of COPD. Eur. Respir. J. 2023, 61, 2201506. [Google Scholar] [CrossRef]
  53. Mizumura, K.; Cloonan, S.M.; Nakahira, K.; Bhashyam, A.R.; Cervo, M.; Kitada, T.; Glass, K.; Owen, C.A.; Mahmood, A.; Washko, G.R.; et al. Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J. Clin. Investig. 2014, 124, 3987–4003. [Google Scholar] [CrossRef]
  54. Zeng, Z.; Liu, X.; Xiang, F.; He, X.; Li, J.; Liu, H.; Xie, L. MicroRNA-21 plays a role in exacerbating chronic obstructive pulmonary disease by regulating necroptosis and apoptosis in bronchial epithelial cells. Tob. Induc. Dis. 2025, 23, 10.18332. [Google Scholar] [CrossRef]
  55. Wang, Y.; Zhou, J.-S.; Xu, X.-C.; Li, Z.-Y.; Chen, H.-P.; Ying, S.-M.; Li, W.; Shen, H.-H.; Chen, Z.-H. Endoplasmic reticulum chaperone GRP78 mediates cigarette smoke-induced necroptosis and injury in bronchial epithelium. Int. J. Chron. Obstruct. Pulmon. Dis. 2018, 13, 571–581. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, D.; Gregory, A.D.; Li, X.; Wei, J.; Burton, C.L.; Gibson, G.; Scott, S.J.; St Croix, C.M.; Zhang, Y.; Shapiro, S.D. RIP3-dependent necroptosis contributes to the pathogenesis of chronic obstructive pulmonary disease. JCI Insight 2021, 6, e144689. [Google Scholar] [CrossRef]
  57. Mizumura, K.; Ozoe, R.; Nemoto, Y.; Furusho, N.; Kurosawa, Y.; Kozu, Y.; Oki, T.; Maruoka, S.; Gon, Y. Cigarette Smoke Extract-Induced Necroptosis Causes Mitochondrial DNA Release and Inflammation of Bronchial Epithelial Cells. Int. J. Chron. Obstruct. Pulmon. Dis. 2025, 20, 2685–2695. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, X.; Comish, P.B.; Tang, D.; Kang, R. Characteristics and Biomarkers of Ferroptosis. Front. Cell Dev. Biol. 2021, 9, 637162. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, J.; Kang, R.; Tang, D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022, 289, 7038–7050. [Google Scholar] [CrossRef]
  60. Jin, X.; Tang, J.; Qiu, X.; Nie, X.; Ou, S.; Wu, G.; Zhang, R.; Zhu, J. Ferroptosis: Emerging mechanisms, biological function, and therapeutic potential in cancer and inflammation. Cell Death Discov. 2024, 10, 45. [Google Scholar] [CrossRef]
  61. 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]
  62. Yoshida, M.; Minagawa, S.; Araya, J.; Sakamoto, T.; Hara, H.; Tsubouchi, K.; Hosaka, Y.; Ichikawa, A.; Saito, N.; Kadota, T.; et al. Involvement of cigarette smoke-induced epithelial cell ferroptosis in COPD pathogenesis. Nat. Commun. 2019, 10, 3145. [Google Scholar] [CrossRef] [PubMed]
  63. Fink, S.L.; Cookson, B.T. Apoptosis, pyroptosis, and necrosis: Mechanistic description of dead and dying eukaryotic cells. Infect. Immun. 2005, 73, 1907–1916. [Google Scholar] [CrossRef] [PubMed]
  64. Xia, H.; Wu, Y.; Zhao, J.; Cheng, C.; Lin, J.; Yang, Y.; Lu, L.; Xiang, Q.; Bian, T.; Liu, Q. N6-Methyladenosine-modified circSAV1 triggers ferroptosis in COPD through recruiting YTHDF1 to facilitate the translation of IREB2. Cell Death Differ. 2023, 30, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
  65. Stockwell, B.R. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell 2022, 185, 2401–2421. [Google Scholar] [CrossRef]
  66. Wang, Y.; Xia, S. Relationship Between ACSL4-Mediated Ferroptosis and Chronic Obstructive Pulmonary Disease. Int. J. Chron. Obstruct. Pulmon. Dis. 2023, 18, 99–111. [Google Scholar] [CrossRef]
  67. Liu, L.; Zhang, Y.; Xu, D.; Zhu, D.; Zhou, Y.; Chen, Z.; Huang, X. Overexpression of USP8 inhibits inflammation and ferroptosis in chronic obstructive pulmonary disease by regulating the OTUB1/SLC7A11 signaling pathway. Allergol. Immunopathol. 2024, 52, 60–67. [Google Scholar] [CrossRef]
  68. Zeng, Z.; Li, T.; Liu, X.; Ma, Y.; Luo, L.; Wang, Z.; Zhao, Z.; Li, H.; He, X.; Zeng, H.; et al. DNA dioxygenases TET2 deficiency promotes cigarette smoke induced chronic obstructive pulmonary disease by inducing ferroptosis of lung epithelial cell. Redox Biol. 2023, 67, 102916. [Google Scholar] [CrossRef]
  69. Galluzzi, L.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cecconi, F.; Choi, A.M.; Chu, C.T.; Codogno, P.; Colombo, M.I.; et al. Molecular definitions of autophagy and related processes. EMBO J. 2017, 36, 1811–1836. [Google Scholar] [CrossRef]
  70. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef]
  71. Galluzzi, L.; Vitale, I.; Abrams, J.M.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; Dawson, T.M.; Dawson, V.L.; El-Deiry, W.S.; Fulda, S.; et al. Molecular definitions of cell death subroutines: Recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012, 19, 107–120. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, S.; Yao, S.; Yang, H.; Liu, S.; Wang, Y. Autophagy: Regulator of cell death. Cell Death Dis. 2023, 14, 648. [Google Scholar] [CrossRef]
  73. Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and autophagy-related pathways in cancer. Nat. Rev. Mol. Cell Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, Q.; Liang, S.; Shi, G.-J.; Meng, G.-L.; Yang, S.-J. Distinct Types of Regulated Cell Death in Melanoma. Cells 2025, 14, 823. [Google Scholar] [CrossRef] [PubMed]
  75. Xia, S.; Gu, X.; Wang, G.; Zhong, Y.; Ma, F.; Liu, Q.; Xie, J. Regulated Cell Death of Alveolar Macrophages in Acute Lung Inflammation: Current Knowledge and Perspectives. J. Inflamm. Res. 2024, 17, 11419–11436. [Google Scholar] [CrossRef]
  76. Huang, X.; Yan, H.; Xu, Z.; Yang, B.; Luo, P.; He, Q. The inducible role of autophagy in cell death: Emerging evidence and future perspectives. Cell Commun. Signal. 2025, 23, 151. [Google Scholar] [CrossRef]
  77. Barnes, P.J.; Baker, J.; Donnelly, L.E. Autophagy in asthma and chronic obstructive pulmonary disease. Clin. Sci. 2022, 136, 733–746. [Google Scholar] [CrossRef]
  78. Mercado, N.; Colley, T.; Baker, J.R.; Vuppussetty, C.; Kono, Y.; Clarke, C.; Tooze, S.; Johansen, T.; Barnes, P.J. Bicaudal D1 impairs autophagosome maturation in chronic obstructive pulmonary disease. FASEB Bioadv. 2019, 1, 688–705. [Google Scholar] [CrossRef]
  79. Fujii, S.; Hara, H.; Araya, J.; Takasaka, N.; Kojima, J.; Ito, S.; Minagawa, S.; Yumino, Y.; Ishikawa, T.; Numata, T.; et al. Insufficient autophagy promotes bronchial epithelial cell senescence in chronic obstructive pulmonary disease. Oncoimmunology 2012, 1, 630–641. [Google Scholar] [CrossRef]
  80. Kono, Y.; Colley, T.; To, M.; Papaioannou, A.I.; Mercado, N.; Baker, J.R.; To, Y.; Abe, S.; Haruki, K.; Ito, K.; et al. Cigarette smoke-induced impairment of autophagy in macrophages increases galectin-8 and inflammation. Sci. Rep. 2021, 11, 335. [Google Scholar] [CrossRef]
  81. Liu, Y.; Levine, B. Autosis and autophagic cell death: The dark side of autophagy. Cell Death Differ. 2015, 22, 367–376. [Google Scholar] [CrossRef]
  82. Liao, S.-X.; Zhang, L.-Y.; Shi, L.-M.; Hu, H.-Y.; Gu, Y.-H.; Wang, T.-H.; Ouyang, Y.; Sun, P.-P. Integrating bulk and single-cell RNA sequencing data: Unveiling RNA methylation and autophagy-related signatures in chronic obstructive pulmonary disease patients. Sci. Rep. 2025, 15, 4005. [Google Scholar] [CrossRef] [PubMed]
  83. Wu, Y.-F.; Li, Z.-Y.; Dong, L.-L.; Li, W.-J.; Wu, Y.-P.; Wang, J.; Chen, H.-P.; Liu, H.-W.; Li, M.; Jin, C.-L.; et al. Inactivation of MTOR promotes autophagy-mediated epithelial injury in particulate matter-induced airway inflammation. Autophagy 2020, 16, 435–450. [Google Scholar] [CrossRef] [PubMed]
  84. Vargas, J.N.S.; Hamasaki, M.; Kawabata, T.; Youle, R.J.; Yoshimori, T. The mechanisms and roles of selective autophagy in mammals. Nat. Rev. Mol. Cell Biol. 2023, 24, 167–185. [Google Scholar] [CrossRef] [PubMed]
  85. Li, A.; Gao, M.; Liu, B.; Qin, Y.; Chen, L.; Liu, H.; Wu, H.; Gong, G. Mitochondrial autophagy: Molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022, 13, 444. [Google Scholar] [CrossRef]
  86. Yang, R.; Zhan, Y.; Deng, Z.; Zhang, J.; Chen, S.; Zhang, Y.; Fu, H.; Meng, X.; Wu, J.; Gu, Y.; et al. Arylsulfatase K attenuates airway epithelial cell senescence in COPD by regulating parkin-mediated mitophagy. Redox Biol. 2025, 86, 103793. [Google Scholar] [CrossRef]
  87. Wei, Y.; Li, Q.; He, K.; Liao, G.; Cheng, L.; Li, M.; He, Z. Mechanism of cigarette smoke in promoting small airway remodeling in mice via STAT 3/PINK 1-Parkin/EMT. Free Radic. Biol. Med. 2024, 224, 447–456. [Google Scholar] [CrossRef]
  88. Wang, Y.; Han, G.; Yang, J.; Xue, L.; Chen, Y. Hydrogen sulfide attenuates PM2.5-induced COPD by inhibiting cellular senescence via the Klotho/Parkin-mediated mitophagy signaling pathway. Ecotoxicol. Environ. Saf. 2025, 293, 117987. [Google Scholar] [CrossRef]
  89. Wei, Y.Y.; Chen, T.T.; Zhang, D.W.; Zhang, Y.; Li, F.; Ding, Y.C.; Wang, M.Y.; Zhang, L.; Chen, K.G.; Fei, G.H. Microplastics exacerbate ferroptosis via mitochondrial reactive oxygen species-mediated autophagy in chronic obstructive pulmonary disease. Autophagy 2025, 21, 1717–1743. [Google Scholar] [CrossRef]
  90. Mo, R.; Zhang, J.; Chen, Y.; Ding, Y. Nicotine promotes chronic obstructive pulmonary disease via inducing pyroptosis activation in bronchial epithelial cells. Mol. Med. Rep. 2022, 25, 92. [Google Scholar] [CrossRef]
  91. Fu, X.; Hong, W.; Li, S.; Chen, Z.; Zhou, W.; Dai, J.; Deng, X.; Zhou, H.; Li, B.; Ran, P. Wood smoke particulate matter (WSPM2.5) induces pyroptosis through both Caspase-1/IL-1β/IL-18 and ATP/P2Y-dependent mechanisms in human bronchial epithelial cells. Chemosphere 2022, 307, 135726. [Google Scholar] [CrossRef]
  92. Buscetta, M.; Cristaldi, M.; Cimino, M.; La Mensa, A.; Dino, P.; Bucchieri, F.; Rappa, F.; Amato, S.; Aronica, T.S.; Pace, E.; et al. Cigarette smoke promotes inflammasome-independent activation of caspase-1 and -4 leading to gasdermin D cleavage in human macrophages. FASEB J. 2022, 36, e22525. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, H.; Xu, F.; Lu, H.; Chen, J.; Huang, X.; Chen, Y.; Lin, L. S1PR2 is Important for Cigarette Smoke-induced Pyroptosis in Human Bronchial Epithelial Cells. Arch. Med. Res. 2023, 54, 277–286. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Q.; Liu, M.; Liu, Y.; Zhang, Z.; Bai, Z. Cigarette Smoke Extract and Lipopolysaccharide Induce Pyroptosis in Pulmonary Microvascular Endothelial Cells of Rats. Bull. Exp. Biol. Med. 2023, 174, 728–733. [Google Scholar] [CrossRef] [PubMed]
  95. Tran, H.B.; Hamon, R.; Jersmann, H.; Ween, M.P.; Asare, P.; Haberberger, R.; Pant, H.; Hodge, S.J. AIM2 nuclear exit and inflammasome activation in chronic obstructive pulmonary disease and response to cigarette smoke. J. Inflamm. 2021, 18, 19. [Google Scholar] [CrossRef]
  96. Ruwanpura, S.M.; McLeod, L.; Dousha, L.F.; Seow, H.J.; West, A.C.; West, A.J.; Weng, T.; Alanazi, M.; MacDonald, M.; King, P.T.; et al. Cross-talk between IL-6 trans-signaling and AIM2 inflammasome/IL-1β axes bridge innate immunity and epithelial apoptosis to promote emphysema. Proc. Natl. Acad. Sci. USA 2022, 119, e2201494119. [Google Scholar] [CrossRef]
  97. Ju, J.; Liu, Y.; Liang, H.; Yang, B. The role of pyroptosis in endothelial dysfunction induced by diseases. Front. Immunol. 2022, 13, 1093985. [Google Scholar] [CrossRef]
  98. Wu, Y.; Di, X.; Zhao, M.; Li, H.; Bai, L.; Wang, K. The role of the NLRP3 inflammasome in chronic inflammation in asthma and chronic obstructive pulmonary disease. Immun. Inflamm. Dis. 2022, 10, e750. [Google Scholar] [CrossRef]
  99. Fu, Y.-S.; Kang, N.; Yu, Y.; Mi, Y.; Guo, J.; Wu, J.; Weng, C.-F. Polyphenols, flavonoids and inflammasomes: The role of cigarette smoke in COPD. Eur. Respir. Rev. 2022, 31, 220028. [Google Scholar] [CrossRef]
  100. Nachmias, N.; Langier, S.; Brzezinski, R.Y.; Siterman, M.; Stark, M.; Etkin, S.; Avriel, A.; Schwarz, Y.; Shenhar-Tsarfaty, S.; Bar-Shai, A. NLRP3 inflammasome activity is upregulated in an in-vitro model of COPD exacerbation. PLoS ONE 2019, 14, e0214622. [Google Scholar] [CrossRef]
  101. Sun, J.; Li, Y. Pyroptosis and respiratory diseases: A review of current knowledge. Front. Immunol. 2022, 13, 920464. [Google Scholar] [CrossRef]
  102. Sawada, M.; Kawayama, T.; Imaoka, H.; Sakazaki, Y.; Oda, H.; Takenaka, S.; Kaku, Y.; Azuma, K.; Tajiri, M.; Edakuni, N.; et al. IL-18 Induces Airway Hyperresponsiveness and Pulmonary Inflammation via CD4+ T Cell and IL-13. PLoS ONE 2013, 8, e54623. [Google Scholar] [CrossRef] [PubMed]
  103. Faner, R.; Sobradillo, P.; Noguera, A.; Gomez, C.; Cruz, T.; López-Giraldo, A.; Ballester, E.; Soler, N.; Arostegui, J.I.; Pelegrín, P.; et al. The inflammasome pathway in stable COPD and acute exacerbations. ERJ Open Res. 2016, 2, 00002-2016. [Google Scholar] [CrossRef] [PubMed]
  104. Gellner, C.A.; Belluzzi, J.D.; Leslie, F.M. Self-administration of nicotine and cigarette smoke extract in adolescent and adult rats. Neuropharmacology 2016, 109, 247–253. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, M.-Y.; Jiang, Y.-X.; Yang, Y.-C.; Liu, J.-Y.; Huo, C.; Ji, X.-L.; Qu, Y.-Q. Cigarette smoke extract induces pyroptosis in human bronchial epithelial cells through the ROS/NLRP3/caspase-1 pathway. Life Sci. 2021, 269, 119090. [Google Scholar] [CrossRef]
  106. Rao, Y.; Gai, X.; Xiong, J.; Le, Y.; Sun, Y. Transient Receptor Potential Cation Channel Subfamily V Member 4 Mediates Pyroptosis in Chronic Obstructive Pulmonary Disease. Front. Physiol. 2021, 12, 783891. [Google Scholar] [CrossRef]
  107. Hou, T.; Zhu, L.; Zhang, Y.; Tang, Y.; Gao, Y.; Hua, S.; Ci, X.; Peng, L. Lipid peroxidation triggered by the degradation of xCT contributes to gasdermin D-mediated pyroptosis in COPD. Redox Biol. 2024, 77, 103388. [Google Scholar] [CrossRef]
  108. Mo, R.; Li, J.; Chen, Y.; Ding, Y. lncRNA GAS5 promotes pyroptosis in COPD by functioning as a ceRNA to regulate the miR-223-3p/NLRP3 axis. Mol. Med. Rep. 2022, 26, 219. [Google Scholar] [CrossRef]
  109. Buscetta, M.; Di Vincenzo, S.; Miele, M.; Badami, E.; Pace, E.; Cipollina, C. Cigarette smoke inhibits the NLRP3 inflammasome and leads to caspase-1 activation via the TLR4-TRIF-caspase-8 axis in human macrophages. FASEB J. 2020, 34, 1819–1832. [Google Scholar] [CrossRef]
  110. Chen, L.; Min, J.; Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 2022, 7, 378. [Google Scholar] [CrossRef]
  111. Tian, Z.; Jiang, S.; Zhou, J.; Zhang, W. Copper homeostasis and cuproptosis in mitochondria. Life Sci. 2023, 334, 122223. [Google Scholar] [CrossRef]
  112. Boaru, D.L.; Leon-Oliva, D.D.; Castro-Martinez, P.D.; Garcia-Montero, C.; Fraile-Martinez, O.; García-González, B.; Pérez-González, I.; Michael Alhaddadin, M.N.; Barrena-Blázquez, S.; Lopez-Gonzalez, L.; et al. Cuproptosis: Current insights into its multifaceted role in disease, cancer, and translational/therapeutic opportunities. Biomed. Pharmacother. 2025, 190, 118422. [Google Scholar] [CrossRef] [PubMed]
  113. Han, L.; Zhu, W.; Qi, H.; He, L.; Wang, Q.; Shen, J.; Song, Y.; Shen, Y.; Zhu, Q.; Zhou, J. The cuproptosis-related gene glutaminase promotes alveolar macrophage copper ion accumulation in chronic obstructive pulmonary disease. Int. Immunopharmacol. 2024, 129, 111585. [Google Scholar] [CrossRef] [PubMed]
  114. Shen, Q.; Huang, J.-B.; Zhu, M.; Ji, D.-J.; Huang, S.-J.; Li, J. Identification of Cuproptosis-Related Genes and Their Potential Role in COPD Pathogenesis: A Bioinformatics Analysis. Int. J. Chron. Obstruct. Pulmon. Dis. 2025, 20, 1083–1096. [Google Scholar] [CrossRef] [PubMed]
  115. Song, W.; Yue, Y.; Zhang, Q.; Wang, X. Copper homeostasis dysregulation in respiratory diseases: A review of current knowledge. Front. Physiol. 2024, 15, 1243629. [Google Scholar] [CrossRef]
  116. Wang, Y.; Luo, W.; Wang, Y. PARP-1 and its associated nucleases in DNA damage response. DNA Repair 2019, 81, 102651. [Google Scholar] [CrossRef]
  117. Mashimo, M.; Bu, X.; Aoyama, K.; Kato, J.; Ishiwata-Endo, H.; Stevens, L.A.; Kasamatsu, A.; Wolfe, L.A.; Toro, C.; Adams, D.; et al. PARP1 inhibition alleviates injury in ARH3-deficient mice and human cells. JCI Insight 2019, 4, e124519. [Google Scholar] [CrossRef]
  118. Pandey, N.; Black, B.E. Rapid Detection and Signaling of DNA Damage by PARP-1. Trends Biochem. Sci. 2021, 46, 744–757. [Google Scholar] [CrossRef]
  119. Künzi, L.; Holt, G.E. Cigarette smoke activates the parthanatos pathway of cell death in human bronchial epithelial cells. Cell Death Discov. 2019, 5, 127. [Google Scholar] [CrossRef]
  120. Huang, P.; Chen, G.; Jin, W.; Mao, K.; Wan, H.; He, Y. Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int. J. Mol. Sci. 2022, 23, 7292. [Google Scholar] [CrossRef]
  121. Pouwels, S.D.; Zijlstra, G.J.; van der Toorn, M.; Hesse, L.; Gras, R.; Ten Hacken, N.H.T.; Krysko, D.V.; Vandenabeele, P.; de Vries, M.; van Oosterhout, A.J.M.; et al. Cigarette smoke-induced necroptosis and DAMP release trigger neutrophilic airway inflammation in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2016, 310, L377–L386. [Google Scholar] [CrossRef]
  122. Zhu, F.; Ji, Y.; You, Q.; Dong, Q.; Tang, Y.; Zhang, Y. Asiaticoside Mitigates Chronic Obstructive Pulmonary Disease by Modulating TRIM27 Stability and Activating PGC-1α/Nrf2 Signaling. Appl. Biochem. Biotechnol. 2025, 197, 6234–6254. [Google Scholar] [CrossRef]
  123. Kim, J.-W.; Kim, J.-H.; Kim, C.-Y.; Jeong, J.-S.; Ko, J.-W.; Kim, T.-W. Korean Red Ginseng suppresses emphysematous lesions induced by cigarette smoke condensate through inhibition of macrophage-driven apoptosis pathways. J. Ginseng Res. 2024, 48, 181–189. [Google Scholar] [CrossRef] [PubMed]
  124. Li, X.; Jiang, X.; Zeng, R.; Lai, X.; Wang, J.; Liu, H.; Wu, H.; He, J.; Liu, L.; Zhu, Z.; et al. Formononetin attenuates cigarette smoke-induced COPD in mice by suppressing inflammation, endoplasmic reticulum stress, and apoptosis in bronchial epithelial cells via AhR/CYP1A1 and AKT/mTOR signaling pathways. Phytother. Res. 2024, 38, 1278–1293. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, J.; Cheng, Y.; Cui, H.; Li, S.; Duan, L.; Jiao, Z. N-acetyl-L-cysteine protects rat lungs and RLE-6TN cells from cigarette smoke-induced oxidative stress. Mol. Med. Rep. 2025, 31, 97. [Google Scholar] [CrossRef] [PubMed]
  126. Li, Z.; Chen, J.; Zhang, J.; Li, N.; Yi, Y.; Lu, H.; Li, M.; Liu, R.; Chen, Y.; Liu, X. Anthrahydroquinone-2,6-Disulfonate Restores Lung Function in COPD Through Keap1/Nrf2 Pathway Activation. J. Inflamm. Res. 2025, 18, 9559–9579. [Google Scholar] [CrossRef]
  127. Min, H.-Y.; Sim, J.Y.; Ahn, J.H.; Kang, N.-W.; Boo, H.-J.; Kim, J.; Yu, N.-Y.; Bottacin, M.; Huh, J.; Park, C.-S.; et al. Gaylussacin, a stilbene glycoside, inhibits chronic obstructive pulmonary disease in mice. Redox Biol. 2025, 85, 103744. [Google Scholar] [CrossRef]
  128. Xie, B.; Chen, Q.; Dai, Z.; Jiang, C.; Chen, X. Progesterone (P4) ameliorates cigarette smoke-induced chronic obstructive pulmonary disease (COPD). Mol. Med. 2024, 30, 123. [Google Scholar] [CrossRef]
  129. Lee, J.; Jang, J.; Park, S.-M.; Yang, S.-R. An Update on the Role of Nrf2 in Respiratory Disease: Molecular Mechanisms and Therapeutic Approaches. Int. J. Mol. Sci. 2021, 22, 8406. [Google Scholar] [CrossRef]
  130. Feng, Q.; Crochet, J.R.; Dai, Q.; Leppert, P.C.; Price, T.M. Expression of a Mitochondrial Progesterone Receptor (PR-M) in Leiomyomata and Association with Increased Mitochondrial Membrane Potential. J. Clin. Endocrinol. Metab. 2014, 99, E390–E399. [Google Scholar] [CrossRef]
  131. Luan, G.; Zhu, Z.; Wu, K.; Yin, S. Theaflavin-3,3’-digallate attenuates cigarette smoke extract-induced pulmonary emphysema in mice by suppressing necroptosis. Exp. Ther. Med. 2022, 23, 11. [Google Scholar] [CrossRef]
  132. Dera, A.A.; Al Fayi, M.; Otifi, H.; Alshyarba, M.; Alfhili, M.; Rajagopalan, P. Thymoquinone (Tq) protects necroptosis induced by autophagy/mitophagy-dependent oxidative stress in human bronchial epithelial cells exposed to cigarette smoke extract (CSE). J. Food Biochem. 2020, 44, e13366. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, M.-J.; Xu, Z.-P.; Guan, Y.-Q.; Wang, Y.-Y.; Wen, X.-S.; Li, G.-H.; Wang, X.-N.; Shen, T. Ethyl acetate fraction of Thesium chinense Turcz. alleviates chronic obstructive pulmonary disease through inhibition of ferroptosis mediated by activating Nrf2/SLC7A11/GPX4 axis. J. Ethnopharmacol. 2025, 337, 118776. [Google Scholar] [CrossRef] [PubMed]
  134. Wu, Y.; Li, B.; Xuan, Y.; Jiang, Y.; Chen, J.; Liao, H.; Feng, J.; Zhang, J. Fluorofenidone alleviates cigarette smoke exposure-induced chronic lung injury by targeting ferroptosis. Sci. Rep. 2024, 14, 32149. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, Y.; Liao, S.; Pan, Z.; Jiang, S.; Fan, J.; Yu, S.; Xue, L.; Yang, J.; Ma, S.; Liu, T.; et al. Hydrogen sulfide alleviates particulate matter-induced emphysema and airway inflammation by suppressing ferroptosis. Free Radic. Biol. Med. 2022, 186, 1–16. [Google Scholar] [CrossRef]
  136. Tang, X.; Li, Z.; Yu, Z.; Li, J.; Zhang, J.; Wan, N.; Zhang, J.; Cao, J. Effect of curcumin on lung epithelial injury and ferroptosis induced by cigarette smoke. Hum. Exp. Toxicol. 2021, 40, S753–S762. [Google Scholar] [CrossRef]
  137. Lian, N.; Zhang, Q.; Chen, J.; Chen, M.; Huang, J.; Lin, Q. The Role of Ferroptosis in Bronchoalveolar Epithelial Cell Injury Induced by Cigarette Smoke Extract. Front. Physiol. 2021, 12, 751206. [Google Scholar] [CrossRef]
  138. Sul, O.J.; Park, S.H.; Choi, H.W.; Kim, D.J.; Ra, S.W. NADPH oxidase-dependent heme oxygenase-1 expression mediates cigarette smoke-induced ferroptosis via intracellular Fe(II) accumulation. Free Radic. Biol. Med. 2025, 237, 131–146. [Google Scholar] [CrossRef]
  139. Liu, J.; Zhang, Z.; Yang, Y.; Di, T.; Wu, Y.; Bian, T. NCOA4-Mediated Ferroptosis in Bronchial Epithelial Cells Promotes Macrophage M2 Polarization in COPD Emphysema. Int. J. Chron. Obstruct. Pulmon. Dis. 2022, 17, 667–681. [Google Scholar] [CrossRef]
  140. Tan, W.; Liang, Z.; Tan, X.; Tan, G. Ginsenoside Rg1 improves cigarette smoke-induced ferroptosis in COPD by regulating PERK/ATF4 axis to inhibit endoplasmic reticulum stress. Biochem. Biophys. Res. Commun. 2024, 739, 150946. [Google Scholar] [CrossRef]
  141. Xu, A.; Xu, Y.; Chen, H.; Xiang, L.; Zhao, X. Ginkgo biloba extract alleviates ferroptosis in lung epithelial cells induced by cigarette smoke extract through miR-3,619-5p/GPX4 axis. Toxicol. Res. 2025, 14, tfae225. [Google Scholar] [CrossRef]
  142. Liu, L.; Wen, T.; Xiao, Y.; Chen, H.; Yang, S.; Shen, X. Sea buckthorn extract mitigates chronic obstructive pulmonary disease by suppression of ferroptosis via scavenging ROS and blocking p53/MAPK pathways. J. Ethnopharmacol. 2025, 336, 118726. [Google Scholar] [CrossRef]
  143. Chen, Y.; Wu, Y.; Dong, J.; Zhang, C.; Tang, J. Acacetin Attenuates Cigarette Smoke Extract-Induced Human Bronchial Epithelial Cell Injury by Activating NRF2/SLC7A11/GPX4 Signaling to Inhibit Ferroptosis. Cell Biochem. Biophys. 2025, 83, 2499–2510. [Google Scholar] [CrossRef]
  144. Liu, X.; Ma, Y.; Luo, L.; Zong, D.; Li, H.; Zeng, Z.; Cui, Y.; Meng, W.; Chen, Y. Dihydroquercetin suppresses cigarette smoke induced ferroptosis in the pathogenesis of chronic obstructive pulmonary disease by activating Nrf2-mediated pathway. Phytomedicine 2022, 96, 153894. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, Z.; Xu, Z.; Chang, J.; He, L.; Zhang, Z.; Song, X.; Hou, X.; Fan, F.; Jiang, Z. Sodium pyruvate exerts protective effects against cigarette smoke extract-induced ferroptosis in alveolar and bronchial epithelial cells through the GPX4/Nrf2 axis. J. Inflamm. 2023, 20, 28. [Google Scholar] [CrossRef] [PubMed]
  146. Zhou, Y.; You, C.-G. Lipoxin alleviates oxidative stress: A state-of-the-art review. Inflamm. Res. 2022, 71, 1169–1179. [Google Scholar] [CrossRef]
  147. Li, X.; Xu, H.; Liu, K.; Shi, M.; Zeng, X.; Liu, X. LXA4 alleviates inflammation and ferroptosis in cigarette smoke induced chronic obstructive pulmonary disease via the ALX/FPR2 receptor. Int. Immunopharmacol. 2025, 151, 114322. [Google Scholar] [CrossRef] [PubMed]
  148. Cheng, M.; Yang, M.; Tian, Y.; Liu, X.; Li, J.; Zhao, P. Bufei Yishen formula alleviates airway epithelial cell senescence in COPD by activating AMPK-Sirt1-FoxO3a pathway and promoting autophagy. Sci. Rep. 2025, 15, 16584. [Google Scholar] [CrossRef]
  149. Jia, L.; Liu, X.; Liu, X.; Guan, Q.; Tian, Y.; Li, J.; Zhao, P. Bufei Yishen formula protects the airway epithelial barrier and ameliorates COPD by enhancing autophagy through the Sirt1/AMPK/Foxo3 signaling pathway. Chin. Med. 2024, 19, 32. [Google Scholar] [CrossRef]
  150. Liu, Y.; Di, N.; Li, C.; Cui, Y.; He, J.; Wei, L. Dihydromyricetin restores mucus hypersecretion in Air–Liquid interface cultures in COPD by targeting the SRC-MAPK signaling pathway. Eur. J. Pharmacol. 2025, 1000, 177703. [Google Scholar] [CrossRef]
  151. Zhou, H.; Lai, Y.; Zhu, Y.; Shao, F.; Ma, G.; Yang, N.; Ma, X.; Sun, Y.; Shi, Q. Quercetin improves airway remodeling in COPD rats by suppressing phenotypic switch of ASMCs via inhibiting the Wnt5a/β-catenin pathway. Phytomedicine 2025, 139, 156491. [Google Scholar] [CrossRef] [PubMed]
  152. Li, W.; Yan, J.; Xu, J.; Zhu, L.; Zhai, C.; Wang, Y.; Wang, Y.; Feng, Y.; Cao, H. Vardenafil alleviates cigarette smoke-induced chronic obstructive pulmonary disease by activating autophagy via the AMPK/mTOR signalling pathway: An in vitro and in vivo study. Cell. Dev. Biol. Anim. 2023, 59, 717–728. [Google Scholar] [CrossRef] [PubMed]
  153. Jung, Y.-S.; Lee, H.J.; Hyun, M.; Kim, H.-J.; Kim, J.-H.; Hwang, K.-H.; Kim, W.-S.; Choi, J.; Heo, J.D. Diindolylmethane Inhibits Cadmium-Induced Autophagic Cell Death via Regulation of Oxidative Stress in HEL299 Human Lung Fibroblasts. Molecules 2022, 27, 5215. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, L.; Jiang, W.; Wang, J.; Xie, Y.; Wang, W. Puerarin inhibits FUNDC1-mediated mitochondrial autophagy and CSE-induced apoptosis of human bronchial epithelial cells by activating the PI3K/AKT/mTOR signaling pathway. Aging 2022, 14, 1253–1264. [Google Scholar] [CrossRef]
  155. Zhang, M.; Hu, C.; Yang, G.; Hu, Y.; Qu, Y. Cytoprotective role of resveratrol in cigarette smoke-induced pyroptosis through Nrf2 pathway activation. Cell Stress Chaperones 2025, 30, 100107. [Google Scholar] [CrossRef]
  156. Hou, T.; Tang, Y.; Wang, L.; Peng, L.; Ci, X. Dihydromyricetin alleviates lipid peroxidation-induced Pyroptosis by inhibiting xCT ubiquitination and degradation in experimental COPD model. Phytomedicine 2025, 144, 156929. [Google Scholar] [CrossRef]
  157. Zeng, J.; Liao, S.; Liang, Z.; Li, C.; Luo, Y.; Wang, K.; Zhang, D.; Lan, L.; Hu, S.; Li, W.; et al. Schisandrin A regulates the Nrf2 signaling pathway and inhibits NLRP3 inflammasome activation to interfere with pyroptosis in a mouse model of COPD. Eur. J. Med. Res. 2023, 28, 217. [Google Scholar] [CrossRef]
  158. Yang, N.; Zhang, L.; Tian, D.; Wang, P.; Men, K.; Ge, Y.; Zhang, C. Tanshinone increases Hemopexin expression in lung cells and macrophages to protect against cigarette smoke-induced COPD and enhance antiviral responses. Cell Cycle 2023, 22, 645–665. [Google Scholar] [CrossRef]
  159. Sul, O.J.; Choi, H.W.; Oh, J.; Ra, S.W. GSPE attenuates CSE-induced lung inflammation and emphysema by regulating autophagy via the reactive oxygen species/TFEB signaling pathway. Food Chem. Toxicol. 2023, 177, 113795. [Google Scholar] [CrossRef]
  160. Fan, X.; Gao, Y.; Hua, C.; Peng, L.; Ci, X. Daphnetin ameliorates PM2.5-induced airway inflammation by inhibiting NLRP3 inflammasome-mediated pyroptosis in CS-exposed mice. Biomed. Pharmacother. 2023, 165, 115047. [Google Scholar] [CrossRef]
  161. Chen, P.; Li, Q.; Su, X.; Zhang, Z.-Q.; Li, G.-P. Osthole, an ingredient from Cnidium monnieri, reduces the pyroptosis and apoptosis in bronchial epithelial cells. J. Asian Nat. Prod. Res. 2023, 25, 999–1011. [Google Scholar] [CrossRef]
  162. Xu, L.-T.; Wang, T.; Fang, K.-L.; Zhao, Y.; Wang, X.-N.; Ren, D.-M.; Shen, T. The ethanol extract of flower buds of Tussilago farfara L. attenuates cigarette smoke-induced lung inflammation through regulating NLRP3 inflammasome, Nrf2, and NF-κB. J. Ethnopharmacol. 2022, 283, 114694. [Google Scholar] [CrossRef]
  163. Tian, X.; Xue, Y.; Xie, G.; Zhou, Y.; Xiao, H.; Ding, F.; Zhang, M. (−)-Epicatechin ameliorates cigarette smoke-induced lung inflammation via inhibiting ROS/NLRP3 inflammasome pathway in rats with COPD. Toxicol. Appl. Pharmacol. 2021, 429, 115674. [Google Scholar] [CrossRef] [PubMed]
  164. Xiao, C.; Cheng, S.; Li, R.; Wang, Y.; Zeng, D.; Jiang, H.; Liang, Y.; Huang, R.; Pan, H.; Wu, X.; et al. Isoforskolin Alleviates AECOPD by Improving Pulmonary Function and Attenuating Inflammation Which Involves Downregulation of Th17/IL-17A and NF-κB/NLRP3. Front. Pharmacol. 2021, 12, 721273. [Google Scholar] [CrossRef] [PubMed]
  165. Wu, K.; Luan, G.; Hu, J.; Zhu, Z.; Kong, Z.; Yin, S. Astragaloside IV Reduces Lung Epithelial Cell Pyroptosis via TXNIP-NLRP3-GSDMD pathway. Cell Biochem. Biophys. 2024, 82, 3695–3702. [Google Scholar] [CrossRef] [PubMed]
  166. Li, H.; Li, Z.; Gong, J.; Yan, Y.; Cui, X.; Yu, Q.; Shi, Y.; Tian, X.; Jiang, Q.; Tan, Z.; et al. Tianlong kechuanling ameliorated COPD-PH through DNMT3A-GSDMD axis-dependent regulation of endothelial cell pyroptosis. J. Ethnopharmacol. 2025, 353, 120332. [Google Scholar] [CrossRef]
  167. Gao, J.; Han, L.; Zhang, Y.; Zhang, X.; Fei, X.; Zhang, M. Disulfiram alleviates epithelial barrier disruption in ozone-induced chronic obstructive pulmonary disease mouse models via inhibiting Gasdermin D-mediated pyroptosis. Int. Immunopharmacol. 2025, 159, 114887. [Google Scholar] [CrossRef]
  168. Xu, L.; Wang, Y.; Chen, Q.; Zhu, X.; Hong, J. Propofol modulates Nrf2/NLRP3 signaling to ameliorate cigarette smoke-induced damage in human bronchial epithelial cells. Tissue Cell 2024, 88, 102341. [Google Scholar] [CrossRef]
  169. Wang, L.; Meng, J.; Wang, C.; Wang, Y.; Yang, C.; Li, Y. Hydrogen sulfide attenuates cigarette smoke-induced pyroptosis through the TLR4/NF-κB signaling pathway. Int. J. Mol. Med. 2022, 49, 56. [Google Scholar] [CrossRef]
  170. Donovan, C.; Kim, R.Y.; Galvao, I.; Jarnicki, A.G.; Brown, A.C.; Jones-Freeman, B.; Gomez, H.M.; Wadhwa, R.; Hortle, E.; Jayaraman, R.; et al. Aim2 suppresses cigarette smoke-induced neutrophil recruitment, neutrophil caspase-1 activation and anti-Ly6G-mediated neutrophil depletion. Immunol. Cell Biol. 2022, 100, 235–249. [Google Scholar] [CrossRef]
  171. Jia, G.; Yu, S.; Sun, W.; Yang, J.; Wang, Y.; Qi, Y.; Chen, Y. Hydrogen Sulfide Attenuates Particulate Matter-Induced Emphysema and Airway Inflammation Through Nrf2-Dependent Manner. Front. Pharmacol. 2020, 11, 29. [Google Scholar] [CrossRef]
  172. Yang, Y.; Huang, L.; Tian, C.; Qian, B. Magnesium isoglycyrrhizinate inhibits airway inflammation in rats with chronic obstructive pulmonary disease. BMC Pulm. Med. 2021, 21, 371. [Google Scholar] [CrossRef] [PubMed]
  173. Colarusso, C.; Terlizzi, M.; Molino, A.; Imitazione, P.; Somma, P.; Rega, R.; Saccomanno, A.; Aquino, R.P.; Pinto, A.; Sorrentino, R. AIM2 Inflammasome Activation Leads to IL-1α and TGF-β Release from Exacerbated Chronic Obstructive Pulmonary Disease-Derived Peripheral Blood Mononuclear Cells. Front. Pharmacol. 2019, 10, 257. [Google Scholar] [CrossRef]
  174. Chen, Y.; Zhang, Y.; Lu, J.; Liu, Z.; Zhao, S.; Zhang, M.; Lu, M.; Xu, W.; Sun, F.; Wu, Q.; et al. Characteristics of Prognostic Programmed Cell Death–Related Long Noncoding RNAs Associated with Immune Infiltration and Therapeutic Responses to Colon Cancer. Front. Immunol. 2022, 13, 828243. [Google Scholar] [CrossRef]
  175. Yang, H.; Liu, Q.; Cao, S.; Fang, H.; Li, J.; Shi, X.; Pang, C.; Lu, D.; Zhao, X.; Li, J.; et al. Programmed cell death-related genes define distinct molecular subtypes and risk profiles in hepatocellular carcinoma. Sci. Rep. 2025, 15, 37117. [Google Scholar] [CrossRef] [PubMed]
  176. Sun, J.; Jiang, R.; Li, M.; Zhai, Q.; Dong, N.; Dong, H.; Zhang, G.; Zhang, Y. Development of a prognostic prediction signature for idiopathic pulmonary fibrosis by integrating multiple programmed cell death-related genes and machine learning algorithms. J. Thorac. Dis. 2025, 17, 7056–7073. [Google Scholar] [CrossRef] [PubMed]
  177. Zhang, P.; Xu, C.-L.; Xu, L.; Wu, J.J.; Wang, L. Pyroptosis-Related Genes as Prognostic Biomarkers and Immune Infiltration Features in Sepsis-Induced ARDS: A Single-Cell and Bulk RNA-Sequencing Analysis. J. Inflamm. Res. 2025, 18, 14261–14282. [Google Scholar] [CrossRef]
  178. Ghadban, C.; García-Unzueta, M.; Agüero, J.; Martín-Audera, P.; Lavín, B.A.; Guerra, A.R.; Berja, A.; Aranda, N.; Guzun, A.; Insua, A.I.; et al. Associations between serum levels of ferroptosis-related molecules and outcomes in stable COPD: An exploratory prospective observational study. Intern. Emerg. Med. 2025, 20, 1761–1773. [Google Scholar] [CrossRef]
  179. Clarke, N.; Thornton, P.; Reader, V.; Lindsay, N.; Digby, Z.; Mullen, B.; Gorman, M.; Jacobson, E.; Langdon, G.; Johnstone, H.; et al. Anti-Neuroinflammatory and Anti-Inflammatory Effects of the NLRP3 Inhibitor NT-0796 in Subjects with Parkinson’s Disease. Mov. Disord. 2025, 40, 2199–2208. [Google Scholar] [CrossRef]
  180. Gatlik, E.; Mehes, B.; Voltz, E.; Sommer, U.; Tritto, E.; Lestini, G.; Liu, X.; Pal, P.; Velinova, M.; Denney, W.S.; et al. First-in-human safety, tolerability, and pharmacokinetic results of DFV890, an oral low-molecular-weight NLRP3 inhibitor. Clin. Transl. Sci. 2024, 17, e13789. [Google Scholar] [CrossRef]
  181. Oliveira, A.P.d.; Figueiredo-Junior, A.T.; Mineiro, P.C.d.O.; Mota, E.C.; Amorim, C.S.d.; Valenca, H.d.M.; Gomes, A.C.C.d.A.; Serra, S.S.d.S.; Silva, P.L.; Takiya, C.M.; et al. Modulation of Pulmonary Inflammation and the Redox Pathway In Vitro and In Vivo by Fumaric Ester. Antioxidants 2025, 14, 1141. [Google Scholar] [CrossRef] [PubMed]
  182. Bomprezzi, R. Dimethyl fumarate in the treatment of relapsing-remitting multiple sclerosis: An overview. Ther. Adv. Neurol. Disord. 2015, 8, 20–30. [Google Scholar] [CrossRef] [PubMed]
  183. Megna, M.; Lauletta, G.; Tommasino, N.; Salsano, A.; Battista, T.; Ruggiero, A.; Martora, F.; Potestio, L. Management of Psoriasis Patients with Serious Infectious Diseases. Adv. Ther. 2024, 41, 2099–2111. [Google Scholar] [CrossRef] [PubMed]
  184. Farenc, C.; Clot, P.-F.; Badalamenti, S.; Kruger, A.J.; Pomponio, R.J.; Krahnke, T.; Staudinger, H.; Lin, Y. A Randomized Phase I Trial Evaluating Safety and Pharmacokinetics of Single and Multiple Ascending Doses of Eclitasertib, a RIPK1 Inhibitor, in Healthy Participants. Adv. Ther. 2025, 42, 3993–4012. [Google Scholar] [CrossRef]
  185. Sun, A.L.A.; Gillies, J.D.; Shen, Y.; Deng, H.; Xue, F.; Ma, Y.; Song, L. A phase I randomized study to evaluate safety, pharmacokinetics, and pharmacodynamics of SIR2446M, a selective RIPK1 inhibitor, in healthy participants. Clin. Transl. Sci. 2024, 17, e13857. [Google Scholar] [CrossRef]
  186. Lickliter, J.; Wang, S.; Zhang, W.; Zhu, H.; Wang, J.; Zhao, C.; Shen, H.; Wang, Y. A phase I randomized, double-blinded, placebo-controlled study assessing the safety and pharmacokinetics of RIPK1 inhibitor GFH312 in healthy subjects. Clin. Transl. Sci. 2023, 16, 1691–1703. [Google Scholar] [CrossRef]
  187. Chen, R.; Hu, X.; Huang, Y.; Jiang, Y.; Chen, G.; Shan, Q.; Xu, X.; Zheng, S. Regulated Cell Death in Lenvatinib Resistance of Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Strategies. Int. J. Biol. Sci. 2025, 21, 2012–2026. [Google Scholar] [CrossRef]
  188. Zhang, X.; Wang, Y.; Chen, M.; Zeng, M. Hexavalent chromium-induced apoptosis in Hep3B cells is accompanied by calcium overload, mitochondrial damage, and AIF translocation. Ecotoxicol. Environ. Saf. 2021, 208, 111391. [Google Scholar] [CrossRef]
  189. Khawas, S.; Sharma, N. Cell death crosstalk in respiratory diseases: Unveiling the relationship between pyroptosis and ferroptosis in asthma and COPD. Mol. Cell. Biochem. 2025, 480, 1305–1326. [Google Scholar] [CrossRef]
Figure 1. Emerging regulated cell death in COPD: apoptosis, necroptosis, ferroptosis, autophagy, pyrptosis, cuproptosis, parthanatos.
Figure 1. Emerging regulated cell death in COPD: apoptosis, necroptosis, ferroptosis, autophagy, pyrptosis, cuproptosis, parthanatos.
Cells 14 01874 g001
Figure 2. Potential molecular mechanisms of apoptosis in COPD.
Figure 2. Potential molecular mechanisms of apoptosis in COPD.
Cells 14 01874 g002
Figure 3. Potential molecular mechanisms of necroptosis in COPD.
Figure 3. Potential molecular mechanisms of necroptosis in COPD.
Cells 14 01874 g003
Figure 4. Potential molecular mechanisms of ferroptosis in COPD.
Figure 4. Potential molecular mechanisms of ferroptosis in COPD.
Cells 14 01874 g004
Figure 5. Potential molecular mechanisms of autophagy in COPD.
Figure 5. Potential molecular mechanisms of autophagy in COPD.
Cells 14 01874 g005
Figure 6. Potential molecular mechanisms of pyrptosis in COPD.
Figure 6. Potential molecular mechanisms of pyrptosis in COPD.
Cells 14 01874 g006
Table 1. Agents targeting COPD via regulating apoptosis pathway.
Table 1. Agents targeting COPD via regulating apoptosis pathway.
CompoundRCD TypesTargetCOPD ModelFunctionsRef.
Anthrahydroquinone-2,6-disulfonate (AH2QDS)ApoptosisNrf2, HO-1, NQO1↑SD ratsAttenuates CS-induced lung inflammation[126]
Asiaticoside (AS)ApoptosisPGC-1α, Nrf2↑BEAS-2B cellsReduces CSE-induced TNF-α, IL-6 and EMT[122]
Formononetin (FMN)ApoptosisAhR, CYP1A1, AKT, mTOR↓BEAS-2B cellsReduces CSE-induced TNF-α, IL-1β[124]
GaylussacinApoptosisSIRT1↑, MMP-12↓MiceAttenuates Pb/Cd-induced emphysema and lung inflammation[127]
ApoptosisSIRT1↑, MMP-12↓MH-S cellsReduces Pb/Cd-induced TNF-α, IL-1β[127]
Korean Red Ginseng (KRG)ApoptosisBax, Caspase 3↓; Bcl-2↑ NCI-H292 cellsReduces CSC-induced TNF-α, IL-1β[123]
ApoptosisBax, Caspase 3↓; Bcl-2↑MiceAttenuates CS-induced emphysema and lung inflammation[123]
N-acetyl-L-cysteine (NAC)Apoptosisp53, Caspase 3↓; Bcl-2, HO-1, GSH↑ RLE-6TN cells-[125]
Apoptosisp53, Caspase 3↓; Bcl-2, HO-1, GSH↑ Wistar ratsAttenuates CS-induced emphysema and lung inflammation[125]
Progesterone (P4)ApoptosisSIRT1, PGC-1α, Nrf1↑ MiceAttenuates CS-induced emphysema and lung inflammation[128]
BEAS-2B: human bronchial epithelial cell; MH-S: murine alveolar macrophage cell; NCI-H292: national cancer institute-H292 cell; RLE-6TN: rat lung epithelial-6 tumor necrosis factor-responsive cell; CS: cigarette smoke; CSE: cigarette smoke extract; IL-6: interleukin 6; IL-1β: interleukin 1β; TNF: tumor necrosis factor. “↓”: down-regulation; “↑”: up-regulation. “-“: none.
Table 2. Agents targeting COPD via regulating necroptosis pathway.
Table 2. Agents targeting COPD via regulating necroptosis pathway.
CompoundRCD TypesTargetCOPD ModelFunctionsRef.
GSK’547NecroptosisRIPK1 kinase inhibitorMice Attenuates CS-induced parenchymal inflammation, airway remodeling and emphysema[52]
Theaflavin-3,3′-digallate
(TF-3)
Necroptosisp-RIPK3, p-MLKL↓MiceAttenuates CS-induced emphysema and lung inflammation[131]
GSK’872NecroptosisRIPK3 kinase inhibitorMiceAttenuates CS-induced emphysema and lung inflammation[56]
NecroptosisRIPK3 kinase inhibitorMLE-12 cellsReduces CSE-induced TNF-α, IL-6 and cell death[56]
NecroptosisRIPK3 kinase inhibitorBMDMsReduces CSE-induced CXCL1, CXCL2, IL-6 and cell death[51]
Necrostatin-1 (NEC-1)NecroptosisRIPK1 kinase inhibitorBMDMsReduces CSE-induced CXCL1, CXCL2, IL-6 and cell death[51]
NecroptosisRIPK1 kinase inhibitorMiceSuppresses the CS-induced neutrophilic airway inflammation[121]
Thymoquinone (Tq)Necroptosisp-MLKL, RIP-1, RIP-3↓BEAS-2B cellsReduces cell death[132]
BEAS-2B: human bronchial epithelial cell; CS: cigarette smoke; CSE: cigarette smoke extract; IL-6: interleukin 6; CXCL-1: C-X-C motif chemokine ligand 1; CXCL-2: C-X-C motif chemokine ligand 2; TNF: tumor necrosis factor. “↓”: down-regulation.
Table 3. Agents targeting COPD via regulating ferroptosis pathway.
Table 3. Agents targeting COPD via regulating ferroptosis pathway.
CompoundRCD TypesTargetCOPD ModelFunctionsRef.
Ginkgo biloba extract (GBE)FerroptosisGSH, GPX4, FTH1↑; ACSL4↓MiceAlleviates PM2.5-induced emphysema and airway inflammation[141]
AcacetinFerroptosisGSH, GPX4, SLC7A11↑; Fe2+16HBE cellsDecreases cell death[143]
Ginsenoside Rg1FerroptosisGSH, GPX4↑; Fe2+BEAS-2B cellsReduces CSE-induced IL-6, TNF-α and IL-1β[140]
Ethyl acetate fraction of Thesium chinense Turcz (TCEA)FerroptosisGPX4, SLC7A11↑; ACSL4, ALOX15↓MiceAttenuates CS-induced emphysema and lung inflammation[133]
Fluorofenidone (AKF)FerroptosisGSH, GPX4, SLC7A11↑; MDA, Fe2+BEAS-2B cellsReduces CSE-induced IL-6, TNF-α and IL-1β[134]
FerroptosisGSH, GPX4, SLC7A11↑; MDA, Fe2+MiceAttenuates CS-induced emphysema, lung fibrosis and lung inflammation[134]
Sea buckthorn extract (SBE)FerroptosisSLC7A11, GSH, GPX4↑; MDA, ACSL4↓MiceAlleviates CS-induced emphysema and airway inflammation[142]
Sodium pyruvate (NaPyr)FerroptosisGSH, GPX4, NRF2↑; COX2↓A549 and
BEAS-2B cells
Reduces CSE-induced TNF-α and IL-8[145]
Ferrostatin-1 (Fer-1)FerroptosisNRF2, GPX4↑; COX2↓A549 and
BEAS-2B cells
Reduces CSE-induced TNF-α and IL-8[145]
FerroptosisFe2+, ALOX15↓MiceAttenuates LPS/CS-induced airway inflammation and MUC5AC[36]
FerroptosisGPX4↑; COX2↓BEAS-2B cellsalleviates PM2.5-induced IL-6, IL-8, TNF-α[135]
FerroptosisGPX4, SLC7A11↑BEAS-2B cellsReduces CSE-induced TNF-α, IL-6 and cell death[136]
FerroptosisGPX4, SLC7A11↑Bronchoalveolar epithelial cells (BAECs)Reduces CSE-induced TNF-α, IL-6 and cell death[137]
FerroptosisGPX4↑16HBE cellsDecreases cell death[137]
FerroptosisGPX4↑BEAS-2B cellsDecreases cell death[138]
Scutellarein (STR)FerroptosisGPX4, NRF2↑; ALOX15↓MiceAttenuates LPS/CS-induced airway inflammation and MUC5AC[36]
Sodium hydrosulfideFerroptosisNRF2, GPX4, NCOA4, PPAR-γ↑; COX2↓MiceAlleviates PM2.5-induced emphysema and airway inflammation[135]
FerroptosisNRF2, GPX4, NCOA4, PPAR-γ↑; COX2↓BEAS-2B cellsAlleviates PM2.5-induced IL-6, IL-8, TNF-α[135]
Deferoxamine (DFO)FerroptosisGPX4↑, COX2↓BEAS-2B cellsAlleviates PM2.5-induced IL-6, IL-8, TNF-α[135]
FerroptosisGPX4, SLC7A11↑BEAS-2B cellsReduces CSE-induced TNF-α, IL-6 and cell death[136]
FerroptosisGPX4↑16HBE cellsDecreases cell death[62]
Dihydroquercetin (DHQ)FerroptosisGPX4, SLC7A11↑MiceAlleviates CS-induced emphysema[148,149]
Curcumin (CUR)FerroptosisGPX4, SLC7A11↑BEAS-2B cellsReduces CSE-induced TNF-α, IL-6 and cell death[136]
16HBE: human airway epithelial cell; A549: alveolar epithelial cell; BEAS-2B: human bronchial epithelial cell; CS: cigarette smoke; CSE: cigarette smoke extract; LPS: lipopolysaccharide; PM: particulate matter; IL-6: interleukin 6; IL-8: interleukin 8; IL-1β: interleukin 1β; TNF: tumor necrosis factor. “↓”: down-regulation; “↑”: up-regulation.
Table 4. Agents targeting COPD via regulating autophagy pathway.
Table 4. Agents targeting COPD via regulating autophagy pathway.
CompoundRCD TypesTargetCOPD ModelFunctionsRef.
Bufei Yishen Formula (BYF)AutophagyLC3BII↑, p62↓16HBE cellsReduces CSE-induced IL-1β, IL-6, TNF-α, MMP-2, MMP-9[148]
AutophagyLC3BII↑BEAS-2B cellsReduces CSE-induced IL-1β, IL-6, TNF-α; maintains epithelial barrier integrity[149]
Dihydromyricetin (DHM)AutophagyBeclin1, LC3BII↑, p62↓Airway organoidsReduces mucus hypersecretion and repairs ciliary function[150]
Diindolylmethane (DIM)AutophagyLC3BII↓, p62↑HEL299 cellsReduces CdCl2-induced oxidative stress[153]
QuercetinAutophagyp62↑; ROS, LC3BII, MDA↓SD ratsAlleviates CS-induced collagen deposition and airway inflammation; improves lung function[151]
VardenafilAutophagyBeclin1, LC3BII↑, p62↓MiceAlleviates CS-induced emphysema and airway inflammation[152]
Hydrogen sulfide (H2S)MitophagyPINK1, Parkin↑, ROS, MDA↓MiceAlleviates CS-induced emphysema and airway inflammation; improves lung function[88]
PuerarinMitophagyPINK1, Parkin, DRP1, FUNDC1↓16HBE cellsReduces CSE-induced cell death and restores mitochondrial function [154]
16HBE: human airway epithelial cell; BEAS-2B: human bronchial epithelial cell; HEL299: human embryonic lung fibroblast 299 Cell; CS: cigarette smoke; CSE: cigarette smoke extract; MMP-2: matrix metallopeptidase 2; MMP-9: matrix metallopeptidase 9; IL-6: interleukin 6; IL-1β: interleukin 1β. “↓”: down-regulation; “↑”: up-regulation.
Table 5. Agents targeting COPD via regulating pyroptosis pathway.
Table 5. Agents targeting COPD via regulating pyroptosis pathway.
CompoundRCD TypesTargetCOPD ModelFunctionsRef.
Tianlong kechuanling (TL)PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓MiceAlleviates LPS + CS + Hx-induced lung function decline, pulmonary hypertension and airway inflammation[166]
ResveratrolPyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓; NRF2/HO-1↑BEAS-2B, 16HBE and A549 cellsReduces CSE-induced IL-18, IL-1β and cell death[155]
DihydromyricetinPyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓; SLC7A11, GPX4↑MiceAlleviates LPS + CS-induced lung function decline and airway inflammation[156]
Disulfiram (DSF)PyroptosisGSDMD-N↓MiceAlleviates O3-induced emphysema and airway inflammation; maintains epithelial barrier integrity[167]
Astragaloside IVPyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓BEAS-2B cellsReduces cell death[165]
PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓MiceAlleviates LPS + CS-induced emphysema and airway inflammation[165]
PropofolPyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓; NRF2↑16HBE cellsAmeliorates CSE-induced IL-6, TNF-α, IL-1β and reduces cell death[168]
Schisandrin A (SchA)PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓; NRF2/HO-1↑Mice Alleviates CS-induced emphysema and airway inflammation; improves lung function[157]
Tanshinone (TS)Pyroptosis-MiceAlleviates LPS + CS + H1N1-induced lung function decline and airway inflammation[158]
PyroptosisNLRP3 inflammasome activation↓; NF-κB signaling activation↓BEAS-2B and Raw264.7 cellsAlleviates LPS + CSE-induced IL-6, IL-8, TNF-α and IL-1β[158]
grape seed proanthocyanidin extract (GSPE)Pyroptosis-MiceAmeliorates lung inflammation and emphysema induced by intraperitoneal injection of CSE.[159]
PyroptosisNLRP3 inflammasome activation↓RAW 264.7 cells-[159]
Daphnetin (Daph)PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓MiceAlleviates PM2.5/PM2.5 + CS-induced airway inflammation and hypersecretion[160]
PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓BEAS-2BAlleviates PM2.5/PM2.5 + CSE-induced IL-1β and cell death[160]
MCC950 (Also known as cRId3)PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓MiceAlleviates PM2.5/PM2.5 + CS-induced airway inflammation[160]
PyroptosisThe NLRP3 inhibitor16HBE cellsReduces CSE-induced IL-1β and TLR4[169]
PyroptosisThe NLRP3 inhibitorMiceAlleviates CS-induced airway inflammation[170]
OstholePyroptosisNLRP3 inflammasome activation↓16HBE cellsReduces CSE-induced IL-6, TNF-α and IL-1β[161]
The flower buds of Tussilago farfara L. (FTF)PyroptosisNLRP3 inflammasome activation↓MiceAlleviates CS-induced airway inflammation [162]
Hydrogen sulfidePyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓SD ratsAttenuates CS-induced lung inflammation[169]
PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓16HBE cellsReduces CSE-induced IL-1β[169]
PyroptosisNLRP3 inflammasome activation↓MiceAttenuates CS-induced airway inflammation and emphysema[171]
PyroptosisNLRP3 inflammasome activation↓A549 cellsAlleviates PM2.5-induced IL-1β and cell death[171]
VX-765PyroptosisSpecific Caspase-1 inhibitor16HBE cellsAlleviates PM2.5-induced IL-1β, IL-6, IL-8, CXCL-1 and CXCL-2[91]
PyroptosisSpecific Caspase-1 inhibitor16HBE cellsAlleviates CSE-induced IL-1β, IL-18 and cell death[105]
Magnesium isoglycyrrhizinate (MgIG)PyroptosisNLRP3 inflammasome activation↓Wistar rats Alleviates LPS + CS-induced lung function decline, airway inflammation, airway remodeling and emphysema[172]
Isoforskolin (ISOF)PyroptosisNLRP3 inflammasome activation↓MiceAlleviates CS + H1N1-induced lung function decline and airway inflammation[164]
(−)-Epicatechin (EC)PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓Rats Alleviates CS-induced lung inflammation and emphysema[163]
PyroptosisNLRP3 inflammasome activation↓; GSDMD-N↓BEAS-2B cellsReduces CSE-induced IL-18, IL-1β and cell death[163]
16HBE: human airway epithelial cell; A549: alveolar epithelial cell; BEAS-2B: human bronchial epithelial cell; RAW 264.7: mouse mononuclear macrophage; CS: cigarette smoke; CSE: cigarette smoke extract; LPS: lipopolysaccharide; PM: particulate matter; IL-6: interleukin 6; IL-8: interleukin 8; IL-1β: interleukin 1β; IL-18: interleukin 18; CXCL-1: C-X-C motif chemokine ligand 1; CXCL-2: C-X-C motif chemokine ligand 2; TNF: tumor necrosis factor. “↓”: down-regulation; “↑”: up-regulation. “-“: none.
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

Fu, H.; Huang, Q.; Xie, J. Targeting Regulated Cell Death Pathways in COPD: Mechanisms and Therapeutic Strategies. Cells 2025, 14, 1874. https://doi.org/10.3390/cells14231874

AMA Style

Fu H, Huang Q, Xie J. Targeting Regulated Cell Death Pathways in COPD: Mechanisms and Therapeutic Strategies. Cells. 2025; 14(23):1874. https://doi.org/10.3390/cells14231874

Chicago/Turabian Style

Fu, Hao, Qian Huang, and Jungang Xie. 2025. "Targeting Regulated Cell Death Pathways in COPD: Mechanisms and Therapeutic Strategies" Cells 14, no. 23: 1874. https://doi.org/10.3390/cells14231874

APA Style

Fu, H., Huang, Q., & Xie, J. (2025). Targeting Regulated Cell Death Pathways in COPD: Mechanisms and Therapeutic Strategies. Cells, 14(23), 1874. https://doi.org/10.3390/cells14231874

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