Next Article in Journal
Definition and Discovery of Tandem SH3-Binding Motifs Interacting with Members of the p47phox-Related Protein Family
Previous Article in Journal
Characterization of Novel Variants in P2YRY12, GP6 and TBXAS1 in Patients with Lifelong History of Bleeding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comprehensive Review of Mechanisms and Translational Perspectives on Programmed Cell Death in Vascular Calcification

1
The Queen Mary School, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330031, China
2
School of Pharmacy, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330031, China
3
Jiangxi Province Key Laboratory of Bioengineering Drugs, The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330031, China
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(12), 1640; https://doi.org/10.3390/biom15121640
Submission received: 11 September 2025 / Revised: 7 November 2025 / Accepted: 19 November 2025 / Published: 21 November 2025
(This article belongs to the Section Molecular Medicine)

Abstract

Programmed cell death (PCD), a regulated cell death (RCD) subtype essential for physiological homeostasis, encompasses distinct mechanisms including apoptosis, necroptosis, autophagy, ferroptosis, cuproptosis, and pyroptosis. This evolutionarily conserved process critically regulates disease pathogenesis across degenerative disorders, malignancies, fungal infections, and vascular calcification (VC). VC manifests as pathological calcium deposition in cardiovascular tissues, impairing vascular elasticity and hemodynamics. These structural alterations elevate risks of atherosclerotic events, cerebrovascular accidents, and peripheral vascular dysfunction, while concomitantly inducing vital organ hypoperfusion and cardiac overload that predispose individuals to myocardial ischemia, left ventricular hypertrophy, and heart failure. Despite advances in delineating associated signaling networks, the molecular etiology of VC remains elusive, and effective therapeutic interventions are currently lacking. While systematically examining the pathophysiological contributions of both established and novel PCD mechanisms to VC progression, this review incorporates a discussion of cuproptosis as a novel form of PCD, which may serve as a target for atherosclerosis treatment. The inclusion of cuproptosis, alongside other PCD modalities, allows for a more integrated and updated perspective on the complex regulatory networks governing VC. Our objective is to synthesize the current understanding of how these diverse PCD pathways, both classical and emerging, collectively contribute to the disease pathogenesis and to explore the broader therapeutic potential of targeting PCD in VC.

Graphical Abstract

1. Introduction

Vascular calcification (VC), a hallmark of aging, commonly occurs in atherosclerosis [1] and diabetes mellitus [2], posing significant health risks. This pathological process involves excessive calcium deposition in the cardiovascular system, leading to reduced arterial elasticity, impaired blood circulation, and compromised organ perfusion [3]. Associated cardiovascular complications include myocardial hypertrophy and hypertension [4]. Clinically, VC severity serves as an independent predictor for assessing cardiovascular morbidity and mortality.
The scientific community recognizes VC as a complex pathophysiological process [5]. Unlike physiological bone mineralization during remodeling, VC progression is driven by the phenotypic transformation of vascular smooth muscle cells (VSMCs) into osteoblast-like cells, a process marked by the downregulation of contractile markers and the concomitant upregulation of osteogenic transcription factors [6]. This phenotypic switching is principally orchestrated by the activation of specific osteogenic signaling pathways and transcription factors. Key among these are the Runt-related transcription factor 2 (Runx2) and Msh homeobox 2 (Msx2), which serve as master regulators of bone-forming protein expression, including Alkaline Phosphatase (ALP), Osteopontin (OPN), and Osteocalcin [7,8,9]. Established inducers of this process include dysregulated mineral metabolism (particularly hyperphosphatemia) [10], Bone Morphogenetic Protein (BMP), and Wnt/β-catenin signaling pathways [11], which collaboratively promote the deposition of hydroxyapatite crystals within the vascular wall. While VSMCs constitute the primary cellular mediators, emerging evidence suggests synergistic contributions from endothelial cells through paracrine signaling and pericytes via their multipotent differentiation potential, amplifying the calcification cascade [12,13].
VC is classified into three types based on calcification sites: intimal, medial, and valvular. Intimal calcification, strongly linked to atherosclerotic plaques, involves chondrocyte-like cells regulated by macrophage/foam cell-derived cytokines that stabilize plaques. Inflammatory stimuli and hyperphosphatemia drive VSMCs to adopt osteoblast-like phenotypes, accelerating calcification [3]. Microcalcifications may evolve into calcified nodules or bone-like structures [14], whose hazards are closely related to the pathological process. In atherosclerotic plaques, microcalcifications disrupt the integrity of the fibrous cap through local stress concentration, significantly increasing the risk of plaque rupture and acute coronary events [15]. Meanwhile, these calcified particles can activate macrophages to release pro-inflammatory factors, exacerbating the vascular inflammatory microenvironment [16]. Medial calcification arises in elastin-rich vascular walls where VSMCs undergo osteogenic transformation via osteoprogenitor cells and transcription factors (Msx2, SRY-box transcription factor 9 [Sox9], Runx2). Bone morphogenetic proteins and inflammatory mediators activate Msx2/Wnt pathways, inducing bone-associated protein expression [17,18]. This is particularly prevalent in patients with diabetes and chronic kidney disease (CKD) [19]. Hyperphosphatemia contributes to VC by inducing osteoblastic differentiation of VSMCs, leading to abnormal hydroxyapatite deposition within the vascular elastic lamina, resulting in increased arterial stiffness and systolic hypertension [20]. In lower extremity arteries, medial calcification critically impairs blood flow perfusion, amplifying amputation risks in affected populations [21]. Healthy aortic valves consist of three thin leaflets containing valve endothelial cells and fibroblast-like interstitial cells [22]. Mechanical stress-induced endothelial damage promotes lipoprotein deposition, oxidized lipid accumulation, and immune infiltration, initiating valve calcification and aortic stenosis [23].
As a multisystem pathology, VC elevates disease incidence and mortality. Limited therapeutic options exist due to non-traditional risk factors (hyperphosphatemia, hypercalcemia, thyroid dysfunction), with current strategies focusing on early intervention: dietary phosphorus control, phosphate binders, vitamin D receptor activators, and calcium-sensing receptor agonists to normalize calcium/phosphorus levels and prevent secondary hyperparathyroidism [24,25]. Osteoprotegerin (OPG) inhibits VC by blocking osteoclast activity [26], suggesting the potential for bone homeostasis-targeting drugs (bisphosphonates, denosumab). Klotho protein demonstrates anti-calcification effects, though its therapeutic potential via endogenous/exogenous supplementation requires validation [27]. Most mechanisms remain incompletely understood, necessitating further research.
Cellular homeostasis relies on balanced regulated cell death (RCD) and accidental cell death. RCD, encompassing programmed cell death (PCD) subforms like apoptosis, necroptosis, and pyroptosis, maintains physiological integrity through controlled signaling cascades [28]. PCD critically influences development (cardiac morphogenesis [29] and immunity [30]) and disease progression (degenerative disorders, cancers [31], and VC [32]). In VC, dynamic interactions among cell types, molecular mediators, and PCD pathways regulate calcification dynamics. For instance, Wang et al. showed that peptide N-acetylgalactosaminyltransferase 3 reduces VSMC apoptosis, alleviating aortic calcification in mice [33]. González-Salvatierra et al. identified sclerostin’s vascular protection via apoptosis suppression [34]. These findings highlight PCD regulation as a therapeutic nexus. Deciphering these networks may unveil novel biomarkers and therapeutic targets.
Building upon this foundation, this review aims to systematically synthesize and delineate the specific mechanistic contributions of major PCD pathways, including apoptosis, necroptosis, ferroptosis, cuproptosis, and pyroptosis, to the pathogenesis of VC. We will critically evaluate how these distinct cell death modalities interact with and drive the osteogenic differentiation of VSMCs, focusing on key signaling cross-talks and the release of critical mediators. Furthermore, another objective of this review is to explore the emerging role of cuproptosis, a newly identified PCD, in VC pathophysiology, an area that has not been comprehensively covered in previous reviews. Finally, by integrating these insights, we seek to discuss the translational potential of targeting PCD pathways as novel therapeutic strategies for mitigating VC.

2. PCD and VC

2.1. Apoptosis and VC

2.1.1. Apoptosis

Apoptosis, an evolutionarily conserved PCD mechanism, eliminates damaged cells while paradoxically enabling proliferative signaling during tissue remodeling [35]. Key morphological features include cell shrinkage, chromatin margination, membrane blebbing, DNA fragmentation, and apoptotic body formation—membrane-bound vesicles phagocytosed by neighboring cells [36,37].
Apoptosis proceeds through two classical pathways: the mitochondrial (intrinsic) pathway initiated by cellular stress, and the cell surface death receptor (extrinsic) pathway [38]. In the intrinsic pathway, mitochondrial cytochrome c release combines with Apaf-1 and caspase-9 to form the apoptosome [39], recruiting caspase-3 to initiate a proteolytic cascade that amplifies death signals. Simultaneously, the second mitochondria-derived activator of caspase (Smac) neutralizes the inhibitor of apoptosis proteins (IAP) [40], apoptosis-inducing factor induces nuclear condensation [41], and endonuclease G cleaves DNA—collectively enhancing apoptotic progression. The Bcl-2 family regulates mitochondrial permeability through anti-apoptotic (Bcl-2, Bcl-xL, Mcl-1) and pro-apoptotic (Bax, Bak) proteins, together with BH3-only activators (Bid, Bim, Puma, etc.) [42]. Caspase-3 cleavage of Bcl-2 at Asp34 generates pro-apoptotic fragments that accelerate apoptosis [43]. The extrinsic pathway is activated through death receptor-ligand binding (e.g., FasL-Fas, TNF-α-TNFR1) [44], triggering receptor oligomerization and caspase-8/FAS-associated death domains recruitment to form the death-inducing signaling complex. Caspase-8 activates Bid, bridging extrinsic and intrinsic pathways to execute apoptosis [45].

2.1.2. The Role of Apoptosis in VC

Apoptosis, a major form of PCD, significantly contributes to VC pathogenesis. Key mediators, including endoplasmic reticulum (ER) stress, hyperphosphatemia, growth factor deficiency, and cellular hypoxia, can trigger apoptosis-driven VC.
As a membrane-bound organelle, the ER maintains proteostasis through synthesizing, processing, and transporting cellular components. Accumulation of misfolded/unfolded proteins disrupts proteostasis, initiating ER stress. Substantial evidence confirmed that ERS regulates VC through apoptotic mechanisms, primarily via four pathways: PERK, IRE1α, ATF6, and Ca2+, with C/EBP-homologous protein (CHOP) as the pivotal downstream effector [46]. Notably, Liu et al. revealed that C5a-C5aR1 activates the PERK-eIF2α-ATF4 pathway by inducing ERS in CKD models, promoting VSMC osteogenic transdifferentiation [47]. This finding aligns with Masuda et al.’s demonstration of TNF-α-mediated ERS activating the same pathway in CKD-associated VC [48]. These findings highlight the therapeutic potential of targeting this pro-apoptotic axis. Recent studies further identified parathyroid hormone and 25-hydroxycholesterol as ERS-mediated VC promoters [49,50], contrasting with fibroblast growth factor-21 (FGF-21)’s protective effects [51].
The FGF family regulates cell migration, proliferation, differentiation, and tissue homeostasis through FGF/FGFR signaling [52]. Mechanistically, Shi et al. demonstrated that FGF-21 suppresses ERS-induced apoptosis by downregulating GRP78, CHOP, and caspase-12 expression, evidenced by reduced TUNEL-positive cells in VC models [53]. This establishes apoptosis inhibition as a viable therapeutic strategy. Clinical observations confirmed that serum phosphorus levels critically regulate VC progression in CKD patients. Hyperphosphatemia induces VSMC calcification through multiple mechanisms: releasing calcifying vesicles, upregulating osteogenic markers (BMP2), and activating pro-apoptotic pathways [54,55]. The OPG/Klotho axis exerts regulatory control [56], while Gas6/Axl/Akt signaling emerges as central to phosphate-induced calcification. Son et al. identified statins attenuate VSMC apoptosis/calcification by enhancing Gas6/Axl expression and Akt/Bcl2/Bad phosphorylation, thereby suppressing caspase-3 activation [57]. Complementarily, Kim et al. demonstrated α-lipoic acid’s dual antioxidant and pro-survival effects through this pathway, suggesting multi-target intervention potential [58]. In addition to this, Liu et al. found that Bone marrow mesenchymal stem cell-derived exosomes inhibited hyperphosphate-induced apoptosis and VC by carrying miR-381-3p [59]. Cui et al. provided evidence that Mitoquinone attenuates VC by inhibiting VSMC oxidative stress and apoptosis through the Keap1/Nrf2 pathway [60]. As summarized in Figure 1, apoptosis intersects with VC through direct calcification and osteogenic differentiation [61,62,63,64]. These mechanistic insights provide validated therapeutic targets and multimodal intervention strategies for VC management.

2.2. Necroptosis and VC

2.2.1. Necroptosis

Necroptosis, a caspase-independent programmed cell death pathway, is initiated through apoptosis inhibition. Its molecular mechanism involves receptor-interacting protein kinase 3 (RIPK3) -mediated phosphorylation of mixed lineage kinase domain-like protein (MLKL) [65,66]. Phosphorylated MLKL forms plasma membrane pore complexes that release damage-associated molecular patterns (DAMP), induce cell swelling, and cause membrane rupture. Characteristic morphological features include organelle swelling, membrane blebbing, moderate chromatin condensation, and cytoplasmic/nuclear disintegration [67].

2.2.2. The Role of Necroptosis in VC

Emerging evidence suggests necroptosis influences VC through multiple pathways, though mechanistic studies remain limited. As a programmed necrosis pathway, necroptosis contributes to cardiovascular pathologies, including myocardial infarction and atherosclerosis [68]. DAMPs, such as calprotectin, amplify VC-associated inflammation by stimulating oxidative stress and cytokine production [69]. Amaya-Garrido et al. demonstrated serum calprotectin elevation correlates with VC severity, while pharmacological inhibitors (paquinimod, AGE/TLR4 blockers) attenuate calcification by modulating VSMC changes [70,71]. Recent studies implicated TLR4-mediated DAMP signaling in modulating IL-6, vascular endothelial growth factor, and collagen secretion, with TGF-β critically driving VSMC phenotypic switching during calcification [72]. Necroptotic cell lysis releases cellular contents that nucleate calcium phosphate crystals, a critical step in VC progression. These deposits form on calcifiable templates like collagen and cellular debris [73].
Phosphorylated RIPK3/MLKL complexes disrupt mitochondrial membranes, inducing hyperpolarization, fragmentation, and ROS overproduction—key drivers of VSMC osteogenic transformation. MLKL directly impairs mitochondrial function, while RIPK3 activates metabolic enzymes (pyruvate dehydrogenase, glutamine synthetase) to exacerbate oxidative stress [74]. Excessive mitochondrial fission promotes VSMC calcification, which Irisin inhibits via AMPK/Drp1 signaling, whereas lactate exacerbates it through NR4A1/DNA-PKcs/p53 activation [75,76]. While RIPK1 independently promotes VC via NF-κB-mediated inflammation, its mechanisms are distinct from necroptosis and warrant separate analysis.
As summarized in Figure 2, necroptosis intersects with VC through inflammatory amplification, mitochondrial dysfunction, and mineralization nucleation [77,78,79]. Elucidating these mechanisms could yield targeted therapies to mitigate cardiovascular calcification.

2.3. Autophagy and VC

2.3.1. Autophagy

Autophagy, a PCD mechanism, maintains cellular homeostasis by selectively degrading damaged organelles, harmful proteins, and intracellular pathogens through lysosomal recycling [80]. This process is evolutionarily conserved and controlled by autophagy-related genes (Atg) [81]. When mTOR activity is suppressed, the Unc-51-like kinase complex becomes activated and stimulates the class III PI3K complex (composed of Beclin1, Atg14L, hVps34, and p150). Once activated, this PI3K complex generates phosphatidylinositol-3-phosphate (PI3P) at pre-autophagosomal membranes, recruiting Atg proteins through hierarchical assembly mechanisms [82]. Among various forms of autophagy, mitophagy represents a selective process targeting mitochondria [83].

2.3.2. The Role of Autophagy in VC

Various studies have identified that the activity of cell autophagy fluctuates with the development of VC, indicating a mechanistic link between these processes. As Atg proteins critically regulate autophagy, multiple studies have explored their roles in VC pathogenesis and therapeutic potential. For example, metformin suppresses VSMC calcification by enhancing autophagic flux to degrade Runx2, whereas Atg3 silencing increases nuclear Runx2 accumulation, exacerbating VC [84]. Peng et al. further demonstrated that estrogen inhibits VSMC osteogenic differentiation through autophagy activation, evidenced by upregulated LC3I/II and Atg5 expression [85].
Different metal ions bridge autophagy and VC. Calcium signaling modulates autophagy through multiple pathways: IP3R/beclin1 interactions, CaMKKβ-AMPK-mTOR signaling, mitochondrial Ca2+ uptake, and lysosomal regulation. Notably, thonningianin A attenuates VC by activating L-type calcium channels to induce autophagy [86]. Paradoxically, another research showed that low potassium environment aggravates VC via elevated calcium level-induced autophagy [87], demonstrating dual functions of autophagy and intracellular calcium towards VC. Concurrently, magnesium also exerts anti-calcification effects via the Erk/autophagy pathway, protecting extracellular matrix integrity [88].
Non-coding RNAs are also an emphasis of the latest research. The lncRNA CAIF inhibits p53-mediated myocardin transcription while suppressing autophagy, accelerating VC by impairing SOX9 antagonism [89,90]. Conversely, small nucleolar RNA host gene 1 reduces VSMC calcification under high glucose by stabilizing Bhlhe40 mRNA to suppress Atg10 expression and excessive autophagy [91]. Furthermore, ectopic calcification has been reported to correlate with particular microRNAs regulating the epigenome. miRNAs like miR-30b, a regulator of SOX9, alleviate VC via mTOR pathway-mediated autophagy induction [92], whereas macrophage-derived miR-32 promotes VSMC osteogenesis by inhibiting autophagy, counteracted by Mef2d through cGMP/PKG signaling [93].
Most studies mentioned the positive facets of autophagy, whereas research also establishes the critical role of autophagy in promoting osteoblast differentiation and mineralization in several specific situations [64]. For example, under a low potassium environment, autophagy activation can exacerbate VC by enhancing VSMC osteogenesis [87], while cold exposure suppresses it [94]. However, specific mechanisms remain unclear.
In summary, autophagy serves dual roles in VC pathogenesis (inhibition via degradation of Runx2 calcium-sensing receptor [CaSR] and mtROS; aggravation via vascular osteogenic conversion and mineralization promotion) (Figure 3) [84,95,96,97]. While pro-autophagy agents hold therapeutic promises, certain pathways paradoxically aggravate calcification. Critical gaps remain in understanding autophagosome-lysosome fusion dynamics and degradation mechanisms. Elucidating these processes is essential for developing targeted VC therapies.

2.4. Ferroptosis and VC

2.4.1. Ferroptosis

Ferroptosis, typically involving glutathione peroxidase 4 (GPX4), glutathione (GSH), and System Xc complex, is a distinct cell death modality brought on by iron overload that leads to ROS accumulation [98]. A redox imbalance between oxidant generation and antioxidant defense primarily drives ferroptosis, mediated by dysregulated redox-active enzymes [99]. Morphologically, ferroptosis, differing from other PCD, manifests as mitochondrial shrinkage with condensed membranes and reduced/absent cristae [100].
Mechanistically, ferroptosis is primarily controlled by the System Xc–GSH–GPX4 axis, where System Xc (SLC7A11/SLC3A2) imports cystine for glutathione (GSH) synthesis via GCLC and GSS, and GPX4 subsequently detoxifies phospholipid hydroperoxides (PLOOH) into non-toxic lipid alcohols [101,102]. Parallel to the GPX4 axis, the NADPH/FSP1 system also suppresses ferroptosis by regenerating lipid antioxidants independently of glutathione [103]. The tumor suppressor p53 exerts dual regulatory roles in ferroptosis: promoting it via SAT1/GLS2 upregulation and SLC7A11 inhibition, while suppressing it through DPP4–NOX1 axis inhibition and CDKN1A/p21 induction [104].

2.4.2. The Role of Ferroptosis in VC

Ferroptosis is induced by iron overload, leading to ROS accumulation, indicating iron is an imperative bridge connecting ferroptosis and VC. Iron serves as an indispensable mental ion in our body, regulating several important molecules like ROS and calcium phosphate deposition, which are pathophysiologically linked to cardiovascular diseases, including atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, and VC [105,106,107]. Ferritin H, an iron storage protein with iron oxidase activity and antioxidant properties, whose mRNA expression level is directly proportional to the iron concentration in the aorta, demonstrates anti-calcification effects by suppressing phosphate-induced osteogenic differentiation in SMCs [108]. Clinical investigations further revealed that hemodialysis patients with reduced serum iron and transferrin saturation exhibit elevated coronary artery calcification scores, implicating iron deficiency in calcification progression [108]. Collectively, it can be predicted that iron is a crucial regulator of VC, and a suitable concentration of iron may attenuate VC. Notably, iron overload, a principal inducer of ferroptosis, stimulated aortic calcium deposition through osteoblast differentiation mediators (Runx2, BMP-2, Msx-2, RANKL) [109]. Furthermore, 2018 research in Nature Portfolio indicated interleukin-24 as a critical mediator of iron-induced VSMC calcification, demonstrating the calcification capacity of interleukin-24 even in iron-deficient environments [110]. Accordingly, aiming to unearth more effective treatments for VC, research finding the optimum iron concentration in the serum of patients should be encouraged.
ROS accumulation caused by the availability of redox-active iron and loss of lipid hydroperoxide repair capacity is the direct inducer of ferroptosis [98]. Experimental models created by palmitic acid, the most prevalent long-chain saturated fatty acid in plasma, created an in vitro oxidative stress-related calcification model and caused lipid overload, which was classified to facilitate ferroptosis in VSMCs via promoting the protein expression of the extracellular matrix protein periostin and its secretion into the extracellular environment [111]. Concomitantly, PA-mediated SIRT6 suppression in VSMCs further accelerates calcification [112], while 1-Palmitoyl-2-(5′-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) also exacerbates osteogenic differentiation of VSMCs via ferroptosis activation [113]. These discoveries further demonstrated the close relationship between ferroptosis and VC. Additionally, SLC7A11, a component of System Xc−, plays a crucial role in ROS regulation in the ferroptosis process [114]. Recent research has demonstrated that reducing levels of transcription factor, Fra-1 (also known as FOSL1), attenuated calcification of VSMCs, through the inhibition of ferroptosis via the p53/SLC7A11 signaling pathway [115], highlighting SLC7A11 as a valuable research target. Furthermore, POVPC-induced mitochondrial ROS promotes VSMC ferroptosis [115], while calcium-dependent Nox5-generated ROS facilitates VSMC phenotypic transition—a calcification prerequisite [116]. As mentioned, ROS-induced PLOOH was also an indispensable factor in ferroptosis [98]. Currently, voluminous chemical compounds have been discovered useful to treat VC based on the mechanisms of reducing the production and accumulation of ROS, such as Wogonin, 3-Arylcoumarin, osteoprotegerin, and Intermedin [117,118,119,120].
Notably, the latest research provided more evidence about the regulatory role of ferroptosis on VC. For instance, lipocalin-2 exacerbates ferroptosis in VSMCs via NCOA4/FTH1-mediated ferritinophagy, thereby promoting VC associated with CKD [121]. The hypothalamic peptide Nesfatin-1 alleviated calcific aortic valve disease by inhibiting cell ferroptosis mediated through the GSH/GPX4 and ZIP8/SOD2 signaling axes [122]. They both indicate the great potential for further research about ferroptosis and VC.
To summarize, ferroptosis promotes VC via mineral deposition coupling and an amplification loop (Figure 4) [123,124]. Considering that iron acts as an important antioxidant to maintain cell survival, and ferroptosis is typically triggered by ROS, we can draw a prediction that ferroptosis may be a prospective therapeutic target for VC treatment.

2.5. Cuproptosis and VC

2.5.1. Cuproptosis

Cuproptosis is a recently defined form of programmed cell death triggered by excessive copper ions (Cu2+) that directly bind to lipoylated proteins within the tricarboxylic acid (TCA) cycle, inducing their aberrant aggregation and destabilizing iron–sulfur (Fe–S) cluster proteins [125,126,127]. Key regulators include ferredoxin-1 (FDX1) and lipoyl synthase, which mediate the lipoylation of TCA enzymes such as dihydrolipoamide S-acetyltransferase [128,129]. Cellular copper homeostasis, maintained through importers (solute carrier family 31 member 1 [SLC31A1]) and exporters (ATPase copper transporting [ATP7] B), critically determines cuproptosis susceptibility [130]. Notably, GSH exerts protective effects by copper chelation, revealing that antioxidant capacity modulates this death pathway [131].

2.5.2. The Role of Cuproptosis in VC

Emerging evidence has revealed significant connections between copper regulation and VC. Clinical observations in 2015 revealed elevated serum copper levels in hemodialysis patients with severe corneal or AAC, though mechanistic insights remained elusive [132]. Subsequent work by Wang et al. (2018) identified abnormally high plasma copper concentrations in hemodialysis patients, suggesting copper imbalance exacerbates oxidative stress and inflammation to accelerate VC progression [133]. Paradoxically, Liu and Liang’s cross-sectional study (2019) associated high dietary copper intake with reduced AAC risk, highlighting the complex duality of copper’s role in VC pathogenesis [134].
The latest research conducted by Qi et al. indicates the fact that calcification in the aortas of VitD3-treated mice is aggravated by cuproptosis induced by elesclomol, a copper transporter. Furthermore, they proved that Elabela, a recently identified peptide, prevents cuproptosis by activating the PPAR-γ/FDX1 signaling pathway and promoting copper efflux through ATP7a-mediated mechanisms, thereby mitigating VC resulting from vitamin D3 excess [135]. A close association between cuproptosis and cardiovascular disease, such as atherosclerosis, has been identified [136], indicating that undiscovered links between cuproptosis and VC exist. Patients with atherosclerosis typically experience VC, which is more prevalent and severe in those with diabetes and CKD. Concurrently, calcification can worsen atherosclerosis by causing blood vessels to stiffen and blood pressure to rise [137]. Recently, a new observational study proved that copper plays a non-negligible role in atherosclerosis [138]. With ceRNA network analyses confirming their regulatory roles, three cuproptosis-related genes (SLC31A1, SLC31A2, SOD1) can serve as diagnostic biomarkers for atherosclerosis [139]. Additionally, using similar strategies, another research showed that cuproptosis-specific expressed genes, FDX1 and GLS, were also important atherosclerosis regulators [140]. These findings demonstrated that cuproptosis is a crucial regulator of atherosclerosis, and VC is a high possibility.
Mechanistically, the copper transporter CTR1 (encoded by SLC31A1, which can also be inhibited by Elabela [135]) governs intracellular copper balance and activates pro-calcification pathways, including MAPK/ERK1/2 and PI3K/Akt/mTOR signaling [141,142,143]. GSH, a molecule closely related to ferroptosis, protects cells from the cytotoxic effects of copper by forming a complex with excessive copper ions through its mercaptan group, while the deficiency of GSH could be a direct inducer of cuproptosis [144,145]. Several studies have identified that the depletion of GSH, which can be induced by the lower expression of GSH synthetase, significantly promoted the calcification of VSMCs under osteogenic conditions by the SLC7A11/GSH/GPX4 axis [114,146]. Notably, GSH also plays a crucial role in the inflammatory response, regulating inflammation by maintaining redox balance through NF-κB and AP-1 signaling regulation [147]. Copper overload also induces mitochondrial ROS generation, which promotes VSMC osteogenic differentiation through BMP-2, Wnt, and NF-κB pathways while impairing redox-sensitive inflammatory regulation [141,148]. They are all evidence supporting that cuproptosis may be a novel inducer of VC development.
Although direct cuproptosis-VC investigations are limited, current findings underscore the harm caused by cuproptosis to VC, potentially via osteogenic signaling driven by mitochondrial proteotoxic stress and direct activation of pro-calcific kinases (Figure 5) [135]. Future research should prioritize two targets: elucidating spatiotemporal expression patterns of cuproptosis-related genes in VC microenvironments and deciphering crosstalk between copper regulatory networks and calcification signaling cascades.

2.6. Pyroptosis and VC

2.6.1. Pyroptosis

Pyroptosis constitutes a programmed cell death modality characterized by distinct morphological changes, primarily triggered by extracellular/intracellular homeostasis disruption during innate immune responses [28]. This caspase-dependent process manifests through two principal pathways. In the classical Pathway, Caspase-1 activation via inflammasomes such as NOD-like receptor pyrin domain containing 3 (NLRP3) promotes recruitment of the adaptor ASC and procaspase-1, leading to caspase-1 activation. Activated caspase-1 then cleaves pro-IL-1β and pro-IL-18 into their mature forms and simultaneously cleaves gasdermin D (GSDMD), releasing its N-terminal fragment (GSDMD-NT) that forms membrane pores and drives pyroptotic lysis [149,150,151,152,153,154,155]. The non-classical pathway is mediated by caspase-4/5 (human) or caspase-11 (murine), which directly cleaves GSDMD to initiate pore formation. These events also secondarily activate NLRP3 inflammasomes, linking the two pathways [156,157].

2.6.2. The Role of Pyroptosis in VC

Emerging evidence establishes pyroptosis as a critical regulator of VC. Studies demonstrated that phosphate overload activated the NLRP3-caspase-1-pyroptosis axis in VSMCs, directly driving calcium deposition [158]. This pathway identifies NLRP3 and caspase-1 as potential therapeutic targets for VC intervention. The NLRP3 inflammasome, a crucial component of innate immunity, mediates VC progression through dual mechanisms: activating caspase-1 to initiate pyroptosis and IL-1β/IL-18 release during cellular stress [159] and promoting osteoclastic bone resorption that exacerbates vascular mineralization [160]. Pharmacological inhibition of NLRP3 may therefore attenuate both inflammatory cascades and bone-vascular axis dysregulation in VC.
Current research found that serum GSDMD levels were strikingly higher in end-stage kidney disease patients with moderate-to-severe VC, suggesting its utility as a clinical biomarker [161]. Mechanistically, caspase-1 cleaves GSDMD to generate GSDMD-NT that oligomerizes at cell membranes, forming lytic pores that execute pyroptosis and pro-calcific cytokine release [162]. Similarly, the gasdermin family member GSDME modulates cellular fate determination—while low expression permits apoptosis, sufficient GSDME converts cell death to pyroptosis through caspase-dependent cleavage, amplifying inflammatory responses in cardiovascular pathologies including atherosclerosis and VC [163]. Additionally, experimental models confirmed that VSMC-specific GSDME knockdown reduces macrophage infiltration, RAGE signaling, and calcification in murine.
VC [164]. Figure 6 illustrates this regulatory network, where activated pyroptosis drives VC through pore formation and pro-calcific factor release [165]. Their clinical correlations position these mediators as priority targets for translational VC research.

3. Clinical Perspectives

VC management employs severity-stratified therapeutic strategies. Intravascular ultrasound remains the gold standard for calcified lesion assessment [166], complemented by optical coherence tomography for detailed calcium characterization [167]. These imaging modalities enable precise calcium quantification and stent expansion prediction for pre-procedural planning. Advanced interventional techniques, including rotational atherectomy, orbital atherectomy, and intravascular lithotripsy, demonstrate efficacy in modifying calcific deposits and optimizing stent deployment for complex coronary lesions [168,169]. However, the potential drugs treating VC remain ambiguous. Emerging research identifies PCD pathways as novel therapeutic targets, with recent evidence revealing their regulatory potential in VC progression, suggesting future precision medicine approaches could combine mechanical modification with biological pathway modulation. Table 1 presents the most valuable emerging drugs targeting PCD pathways, which show the ability to attenuate VC [24,170,171,172,173,174,175,176,177,178,179,180,181,182].

4. Conclusions

PCD, encompassing apoptosis, necroptosis, autophagy, ferroptosis, cuproptosis, and pyroptosis, exerts a substantial influence on the pathogenesis of multiple diseases. VC constitutes a pivotal pathogenic mechanism underlying cardiovascular disorders. Although increasing evidence suggests that PCD critically regulates VC progression, a comprehensive review that systematically constructs the PCD-related regulatory network and identifies therapeutic targets remains essential for translational research.
Previous reviews have outlined the regulatory functions of PCD in VC pathogenesis and highlighted the therapeutic potential of targeting PCD in VC treatment [32,183]. Building upon these findings, the present review updates recently discovered molecular pathways and, for the first time, explores the potential involvement of cuproptosis—a newly characterized form of PCD—in VC regulation. Notably, inhibition of cuproptosis through Elabela-mediated activation of the PPAR-γ/FDX1 signaling axis has been shown to protect against arterial calcification [135]. However, further clarification of how cuproptosis regulates osteogenic conversion of VSMCs is warranted. This review emphasizes the potential of cuproptosis in VC through its mechanistic involvement, interaction with other cell death pathways, association with VC-related diseases, and its therapeutic implications, highlighting cuproptosis as a promising target for future research and intervention.
It is noteworthy that although all forms of PCD ultimately promote VC, they do so through distinct mechanisms. Apoptosis primarily contributes by generating apoptotic bodies that serve as physical “seeds” for calcium-phosphate crystal deposition [61], coupled with the direct upregulation of osteogenic genes by the ER stress downstream factor CHOP [62]. At the same time, lytic forms of death, such as necroptosis and pyroptosis, drive calcification by releasing DAMPs and pro-inflammatory cytokines (IL-1β, IL-18), thereby creating a potent inflammatory microenvironment [77,165]. Ferroptosis and cuproptosis rely on iron-dependent lipid peroxidation and copper-induced mitochondrial proteotoxic stress, respectively, with their core mechanisms centering on disordered metal ion metabolism and intense oxidative stress [123,135]. Autophagy plays the most unique role, exhibiting a dual regulatory function where the net effect depends on the integrity of the autophagic flux and the specific substrates degraded [84,95]. These fundamental differences underscore the necessity for highly specific therapeutic strategies tailored to VC subtypes dominated by different PCD pathways.
Moreover, emerging molecular targets and bioactive compounds that mitigate VC via PCD modulation were summarized in the current review, reinforcing the central role of PCD in VC regulation and offering valuable clues for translational studies. Nevertheless, critical knowledge gaps persist in delineating the mechanistic interplay between PCD and VC. Establishing definitive causal relationships remains difficult despite growing identification of upstream and downstream components. Additionally, most studies have been limited to in vitro or animal models. Thus, human-based investigations are urgently needed to validate the clinical relevance of PCD-mediated mechanisms in VC.
Addressing these challenges through advanced molecular and translational studies may pave the way for novel therapeutic strategies. A systematic and integrative understanding of PCD-driven VC mechanisms will ultimately facilitate the development of precise, mechanism-based interventions for cardiovascular calcification.

Author Contributions

Conceptualization, Y.C. (Yiyang Cao) and M.J.; investigation, Y.Y.; writing—original draft preparation, Y.C. (Yiyang Cao), Y.C. (Yulong Cao); writing—review and editing, M.J.; visualization, J.L.; supervision, M.J.; funding acquisition, M.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by National Natural Science Foundation of China grants (82160094, 82200509 to MXJ), Natural Science Foundation of Jiangxi Province (20252BAC240143 to MXJ), Jiangxi province Science and Technology Program of the Health Commission (202311127 to MXJ), and the Jiangxi Province Key Laboratory of bioengineering drugs (No.2024SSY07061).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The illustration material for this review was kindly provided by BioRender (accessed on 7 November 2025).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
VCVascular Calcification
VSMCVascular Smooth Muscle Cell
Runx2Runt-Related Transcription Factor 2
Msx2Msh Homeobox 2
ALPAlkaline Phosphatase
OPNOsteopontin
BMPBone Morphogenetic Protein
Sox9SRY-Box Transcription Factor 9
OPGOsteoprotegerin
RCDRegulated Cell Death
PCDProgrammed Cell Death
SmacThe Second Mitochondria-Derived Activator of Caspase
IAPInhibitor of Apoptosis Protein
EREndoplasmic Reticulum
CHOPC/EBP-Homologous Protein
FGF-21Fibroblast Growth Factor-21
RIPK3Receptor-Interacting Protein Kinase 3
MLKLMixed Lineage Kinase Domain-Like Protein
DAMPDamage-Associated Molecular Pattern
ROSReactive Oxygen Species
AtgAutophagy-Related Genes
PI3Pphosphatidylinositol-3-Phosphate
CaSRCalcium-Sensing Receptor
GPX4Glutathione Peroxidase 4
GSHGlutathione
PLOOHPhospholipid Hydroperoxide
RANKLReceptor Activator for Nuclear Factor-κ B Ligand
POVPC1-Palmitoyl-2-(5′-Oxo-Valeroyl)-Sn-Glycero-3-Phosphocholine
NCOA4Nuclear Receptor Coactivator 4
TCATricarboxylic Acid
FDX1Ferredoxin-1
SLC31A1Solute Carrier Family 31 Member 1
ATP7ATPase Copper Transporting
CTR1Copper Transporter 1
NLRP3NOD-like Receptor Pyrin Domain Containing 3
GSDMDGasdermin D
Gas6Growth Arrest-Specific Gene 6
RIP 1Receptor-Interacting Protein 1
FPSPolysaccharide from Fuzi
AMPKAdenosine 5′-Monophosphate-Activated Protein Kinase
mTORMammalian Target of Rapamycin
POSTNExtracellular Matrix Protein Periostin
PHGDHPhosphoglycerate Dehydrogenase
OTC2-Oxothiazolidine-4-Carboxylic Acid
p53Tumor Protein 53
PPARαPeroxisome Proliferator-Activated Receptor α

References

  1. Bessueille, L.; Magne, D. Inflammation: A culprit for vascular calcification in atherosclerosis and diabetes. Cell Mol. Life Sci. 2015, 72, 2475–2489. [Google Scholar] [CrossRef]
  2. Yahagi, K.; Kolodgie, F.D.; Lutter, C.; Mori, H.; Romero, M.E.; Finn, A.V.; Virmani, R. Pathology of Human Coronary and Carotid Artery Atherosclerosis and Vascular Calcification in Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 191–204. [Google Scholar] [CrossRef] [PubMed]
  3. Lanzer, P.; Boehm, M.; Sorribas, V.; Thiriet, M.; Janzen, J.; Zeller, T.; St Hilaire, C.; Shanahan, C. Medial vascular calcification revisited: Review and perspectives. Eur. Heart J. 2014, 35, 1515–1525. [Google Scholar] [CrossRef]
  4. Boutouyrie, P.; Chowienczyk, P.; Humphrey, J.D.; Mitchell, G.F. Arterial Stiffness and Cardiovascular Risk in Hypertension. Circ. Res. 2021, 128, 864–886. [Google Scholar] [CrossRef] [PubMed]
  5. Vidavsky, N.; Kunitake, J.; Estroff, L.A. Multiple Pathways for Pathological Calcification in the Human Body. Adv. Healthc. Mater. 2021, 10, e2001271. [Google Scholar] [CrossRef] [PubMed]
  6. Ballester-Servera, C.; Cañes, L.; Alonso, J.; Puertas-Umbert, L.; Vázquez-Sufuentes, P.; Taurón, M.; Roselló-Díez, E.; Marín, F.; Rodríguez, C.; Martínez-González, J. Upregulation of NOR-1 in calcified human vascular tissues: Impact on osteogenic differentiation and calcification. Transl. Res. 2024, 264, 1–14. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, L.; Dai, R.; Wu, H.; Cai, Z.; Xie, N.; Zhang, X.; Shen, Y.; Gong, Z.; Jia, Y.; Yu, F.; et al. Unspliced XBP1 Counteracts β-Catenin to Inhibit Vascular Calcification. Circ. Res. 2022, 130, 213–229. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, S.; Xie, Y.; Yan, F.; Zhang, Y.; Yang, Z.; Chen, Z.; Zhao, Y.; Huang, Z.; Cai, L.; Deng, Z. Negative pressure wound therapy improves bone regeneration by promoting osteogenic differentiation via the AMPK-ULK1-autophagy axis. Autophagy 2022, 18, 2229–2245. [Google Scholar] [CrossRef] [PubMed]
  9. Lee, A.E.; Choi, J.G.; Shi, S.H.; He, P.; Zhang, Q.Z.; Le, A.D. DPSC-Derived Extracellular Vesicles Promote Rat Jawbone Regeneration. J. Dent. Res. 2023, 102, 313–321. [Google Scholar] [CrossRef]
  10. Macrì, F.; Vigorito, I.; Castiglione, S.; Faggiano, S.; Casaburo, M.; Fanotti, N.; Piacentini, L.; Vigetti, D.; Vinci, M.C.; Raucci, A. High Phosphate-Induced JAK-STAT Signalling Sustains Vascular Smooth Muscle Cell Inflammation and Limits Calcification. Biomolecules 2023, 14, 29. [Google Scholar] [CrossRef] [PubMed]
  11. Zhang, Y.; Zhao, Y.; Xie, Z.; Li, M.; Liu, Y.; Tu, X. Activating Wnt/β-Catenin Signaling in Osteocytes Promotes Osteogenic Differentiation of BMSCs through BMP-7. Int. J. Mol. Sci. 2022, 23, 16045. [Google Scholar] [CrossRef] [PubMed]
  12. Lin, X.; Shan, S.K.; Xu, F.; Zhong, J.Y.; Wu, F.; Duan, J.Y.; Guo, B.; Li, F.X.; Wang, Y.; Zheng, M.H.; et al. The crosstalk between endothelial cells and vascular smooth muscle cells aggravates high phosphorus-induced arterial calcification. Cell Death Dis. 2022, 13, 650. [Google Scholar] [CrossRef] [PubMed]
  13. Dabravolski, S.A.; Markin, A.M.; Andreeva, E.R.; Eremin, I.I.; Orekhov, A.N.; Melnichenko, A.A. Emerging role of pericytes in therapy of cardiovascular diseases. Biomed. Pharmacother. 2022, 156, 113928. [Google Scholar] [CrossRef]
  14. Leopold, J.A. Vascular calcification: Mechanisms of vascular smooth muscle cell calcification. Trends Cardiovasc. Med. 2015, 25, 267–274. [Google Scholar] [CrossRef]
  15. Nakahara, T.; Dweck, M.R.; Narula, N.; Pisapia, D.; Narula, J.; Strauss, H.W. Coronary Artery Calcification: From Mechanism to Molecular Imaging. JACC Cardiovasc. Imaging 2017, 10, 582–593. [Google Scholar] [CrossRef]
  16. Kawakami, R.; Katsuki, S.; Travers, R.; Romero, D.C.; Becker-Greene, D.; Passos, L.S.A.; Higashi, H.; Blaser, M.C.; Sukhova, G.K.; Buttigieg, J.; et al. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1838–1853. [Google Scholar] [CrossRef]
  17. Boström, K.I.; Rajamannan, N.M.; Towler, D.A. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ. Res. 2011, 109, 564–577. [Google Scholar] [CrossRef]
  18. Lee, S.J.; Lee, I.K.; Jeon, J.H. Vascular Calcification-New Insights Into Its Mechanism. Int. J. Mol. Sci. 2020, 21, 2685. [Google Scholar] [CrossRef]
  19. Lanzer, P.; Hannan, F.M.; Lanzer, J.D.; Janzen, J.; Raggi, P.; Furniss, D.; Schuchardt, M.; Thakker, R.; Fok, P.W.; Saez-Rodriguez, J.; et al. Medial Arterial Calcification: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 78, 1145–1165. [Google Scholar] [CrossRef]
  20. Nikolajević, J.; Šabovič, M. Inflammatory, Metabolic, and Coagulation Effects on Medial Arterial Calcification in Patients with Peripheral Arterial Disease. Int. J. Mol. Sci. 2023, 24, 3132. [Google Scholar] [CrossRef] [PubMed]
  21. Lanzer, P.; Ferraresi, R. Medial Sclerosis-Epidemiology and Clinical Significance. Dtsch. Arztebl. Int. 2023, 120, 365–372. [Google Scholar] [CrossRef]
  22. Rajamannan, N.M.; Evans, F.J.; Aikawa, E.; Grande-Allen, K.J.; Demer, L.L.; Heistad, D.D.; Simmons, C.A.; Masters, K.S.; Mathieu, P.; O’Brien, K.D.; et al. Calcific aortic valve disease: Not simply a degenerative process: A review and agenda for research from the National Heart and Lung and Blood Institute Aortic Stenosis Working Group. Executive summary: Calcific aortic valve disease-2011 update. Circulation 2011, 124, 1783–1791. [Google Scholar] [CrossRef] [PubMed]
  23. Goody, P.R.; Hosen, M.R.; Christmann, D.; Niepmann, S.T.; Zietzer, A.; Adam, M.; Bönner, F.; Zimmer, S.; Nickenig, G.; Jansen, F. Aortic Valve Stenosis: From Basic Mechanisms to Novel Therapeutic Targets. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 885–900. [Google Scholar] [CrossRef]
  24. Khundmiri, S.J.; Murray, R.D.; Lederer, E. PTH and Vitamin D. Compr. Physiol. 2016, 6, 561–601. [Google Scholar] [CrossRef] [PubMed]
  25. Molostvov, G.; Hiemstra, T.F.; Fletcher, S.; Bland, R.; Zehnder, D. Arterial Expression of the Calcium-Sensing Receptor Is Maintained by Physiological Pulsation and Protects against Calcification. PLoS ONE 2015, 10, e0138833. [Google Scholar] [CrossRef]
  26. Ndip, A.; Wilkinson, F.L.; Jude, E.B.; Boulton, A.J.; Alexander, M.Y. RANKL-OPG and RAGE modulation in vascular calcification and diabetes: Novel targets for therapy. Diabetologia 2014, 57, 2251–2260. [Google Scholar] [CrossRef] [PubMed]
  27. Prud’homme, G.J.; Kurt, M.; Wang, Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. Front. Aging 2022, 3, 931331. [Google Scholar] [CrossRef]
  28. 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]
  29. Martínez-Lagunas, K.; Yamaguchi, Y.; Becker, C.; Geisen, C.; DeRuiter, M.C.; Miura, M.; Fleischmann, B.K.; Hesse, M. In vivo detection of programmed cell death during mouse heart development. Cell Death Differ. 2020, 27, 1398–1414. [Google Scholar] [CrossRef] [PubMed]
  30. Hänggi, K.; Ruffell, B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer 2023, 9, 381–396. [Google Scholar] [CrossRef]
  31. Hu, J.; Pan, D.; Li, G.; Chen, K.; Hu, X. Regulation of programmed cell death by Brd4. Cell Death Dis. 2022, 13, 1059. [Google Scholar] [CrossRef]
  32. Li, M.; Wang, Z.W.; Fang, L.J.; Cheng, S.Q.; Wang, X.; Liu, N.F. Programmed cell death in atherosclerosis and vascular calcification. Cell Death Dis. 2022, 13, 467. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Y.K.; Li, S.J.; Zhou, L.L.; Li, D.; Guo, L.W. GALNT3 protects against vascular calcification by reducing oxidative stress and apoptosis of smooth muscle cells. Eur. J. Pharmacol. 2023, 939, 175447. [Google Scholar] [CrossRef] [PubMed]
  34. González-Salvatierra, S.; García-Fontana, C.; Lacal, J.; Andújar-Vera, F.; Martínez-Heredia, L.; Sanabria-de la Torre, R.; Ferrer-Millán, M.; Moratalla-Aranda, E.; Muñoz-Torres, M.; García-Fontana, B. Cardioprotective function of sclerostin by reducing calcium deposition, proliferation, and apoptosis in human vascular smooth muscle cells. Cardiovasc. Diabetol. 2023, 22, 301. [Google Scholar] [CrossRef]
  35. 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] [PubMed]
  36. Green, D.R.; Llambi, F. Cell Death Signaling. Cold Spring Harb. Perspect. Biol. 2015, 7, a006080. [Google Scholar] [CrossRef]
  37. Yuan, J.; Ofengeim, D. A guide to cell death pathways. Nat. Rev. Mol. Cell Biol. 2024, 25, 379–395. [Google Scholar] [CrossRef] [PubMed]
  38. Boada-Romero, E.; Martinez, J.; Heckmann, B.L.; Green, D.R. The clearance of dead cells by efferocytosis. Nat. Rev. Mol. Cell Biol. 2020, 21, 398–414. [Google Scholar] [CrossRef]
  39. Acehan, D.; Jiang, X.; Morgan, D.G.; Heuser, J.E.; Wang, X.; Akey, C.W. Three-Dimensional Structure of the Apoptosome: Implications for Assembly, Procaspase-9 Binding, and Activation. Mol. Cell 2002, 9, 423–432. [Google Scholar] [CrossRef]
  40. Ehrmann, J.F.; Grabarczyk, D.B.; Heinke, M.; Deszcz, L.; Kurzbauer, R.; Hudecz, O.; Shulkina, A.; Gogova, R.; Meinhart, A.; Versteeg, G.A.; et al. Structural basis for regulation of apoptosis and autophagy by the BIRC6/SMAC complex. Science 2023, 379, 1117–1123. [Google Scholar] [CrossRef] [PubMed]
  41. Candé, C.; Vahsen, N.; Kouranti, I.; Schmitt, E.; Daugas, E.; Spahr, C.; Luban, J.; Kroemer, R.T.; Giordanetto, F.; Garrido, C.; et al. AIF and cyclophilin A cooperate in apoptosis-associated chromatinolysis. Oncogene 2004, 23, 1514–1521. [Google Scholar] [CrossRef]
  42. Shoshan-Barmatz, V.; De Pinto, V.; Zweckstetter, M.; Raviv, Z.; Keinan, N.; Arbel, N. VDAC, a multi-functional mitochondrial protein regulating cell life and death. Mol. Aspects Med. 2010, 31, 227–285. [Google Scholar] [CrossRef]
  43. Cheng, E.H.; Kirsch, D.G.; Clem, R.J.; Ravi, R.; Kastan, M.B.; Bedi, A.; Ueno, K.; Hardwick, J.M. Conversion of Bcl-2 to a Bax-like death effector by caspases. Science 1997, 278, 1966–1968. [Google Scholar] [CrossRef] [PubMed]
  44. Kashyap, D.; Garg, V.K.; Goel, N. Chapter Four—Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. In Advances in Protein Chemistry and Structural Biology; Donev, R., Ed.; Academic Press: Cambridge, MA, USA, 2021; Volume 125, pp. 73–120. [Google Scholar]
  45. Wajant, H. The Fas signaling pathway: More than a paradigm. Science 2002, 296, 1635–1636. [Google Scholar] [CrossRef]
  46. Zhang, J.; Guo, J.; Yang, N.; Huang, Y.; Hu, T.; Rao, C. Endoplasmic reticulum stress-mediated cell death in liver injury. Cell Death Dis. 2022, 13, 1051. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, A.; Chen, Z.; Li, X.; Xie, C.; Chen, Y.; Su, X.; Chen, Y.; Zhang, M.; Chen, J.; Yang, T.; et al. C5a-C5aR1 induces endoplasmic reticulum stress to accelerate vascular calcification via PERK-eIF2α-ATF4-CREB3L1 pathway. Cardiovasc. Res. 2023, 119, 2563–2578. [Google Scholar] [CrossRef]
  48. Masuda, M.; Miyazaki-Anzai, S.; Levi, M.; Ting, T.C.; Miyazaki, M. PERK-eIF2α-ATF4-CHOP signaling contributes to TNFα-induced vascular calcification. J. Am. Heart Assoc. 2013, 2, e000238. [Google Scholar] [CrossRef]
  49. Duang, S.; Zhang, M.; Liu, C.; Dong, Q. Parathyroid hormone-induced vascular smooth muscle cells calcification by endoplasmic reticulum stress. J. Physiol. Pharmacol. 2022, 73, 597–604. [Google Scholar] [CrossRef]
  50. Dong, Q.; Chen, Y.; Liu, W.; Liu, X.; Chen, A.; Yang, X.; Li, Y.; Wang, S.; Fu, M.; Ou, J.S.; et al. 25-Hydroxycholesterol promotes vascular calcification via activation of endoplasmic reticulum stress. Eur. J. Pharmacol. 2020, 880, 173165. [Google Scholar] [CrossRef]
  51. Shi, Y.; Zheng, Z.; Luo, J.; Li, Y.; He, S.; Shen, X.; Liu, J. Possible effects of fibroblast growth factor 21 on vascular calcification via suppressing activating transcription factor 4 mediated apoptosis and osteogenic transformation in rats. Cell Biochem. Funct. 2022, 40, 349–358. [Google Scholar] [CrossRef] [PubMed]
  52. Xie, Y.; Su, N.; Yang, J.; Tan, Q.; Huang, S.; Jin, M.; Ni, Z.; Zhang, B.; Zhang, D.; Luo, F.; et al. FGF/FGFR signaling in health and disease. Signal Transduct. Target. Ther. 2020, 5, 181. [Google Scholar] [CrossRef] [PubMed]
  53. Shi, Y.; Wang, S.; Peng, H.; Lv, Y.; Li, W.; Cheng, S.; Liu, J. Fibroblast Growth Factor 21 Attenuates Vascular Calcification by Alleviating Endoplasmic Reticulum Stress Mediated Apoptosis in Rats. Int. J. Biol. Sci. 2019, 15, 138–147. [Google Scholar] [CrossRef]
  54. Reynolds, J.L.; Joannides, A.J.; Skepper, J.N.; McNair, R.; Schurgers, L.J.; Proudfoot, D.; Jahnen-Dechent, W.; Weissberg, P.L.; Shanahan, C.M. Human vascular smooth muscle cells undergo vesicle-mediated calcification in response to changes in extracellular calcium and phosphate concentrations: A potential mechanism for accelerated vascular calcification in ESRD. J. Am. Soc. Nephrol. 2004, 15, 2857–2867. [Google Scholar] [CrossRef]
  55. Sage, A.P.; Lu, J.; Tintut, Y.; Demer, L.L. Hyperphosphatemia-induced nanocrystals upregulate the expression of bone morphogenetic protein-2 and osteopontin genes in mouse smooth muscle cells in vitro. Kidney Int. 2011, 79, 414–422. [Google Scholar] [CrossRef]
  56. Kaur, R.; Singh, R. Mechanistic insights into CKD-MBD-related vascular calcification and its clinical implications. Life Sci. 2022, 311, 121148. [Google Scholar] [CrossRef]
  57. Son, B.-K.; Kozaki, K.; Iijima, K.; Eto, M.; Nakano, T.; Akishita, M.; Ouchi, Y. Gas6/Axl-PI3K/Akt pathway plays a central role in the effect of statins on inorganic phosphate-induced calcification of vascular smooth muscle cells. Eur. J. Pharmacol. 2007, 556, 1–8. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, H.; Kim, H.J.; Lee, K.; Kim, J.M.; Kim, H.S.; Kim, J.R.; Ha, C.M.; Choi, Y.K.; Lee, S.J.; Kim, J.Y.; et al. α-Lipoic acid attenuates vascular calcification via reversal of mitochondrial function and restoration of Gas6/Axl/Akt survival pathway. J. Cell Mol. Med. 2012, 16, 273–286. [Google Scholar] [CrossRef]
  59. Liu, Y.; Guo, Y.; Bao, S.; Huang, H.; Liu, W.; Guo, W. Bone marrow mesenchymal stem cell-derived exosomal microRNA-381-3p alleviates vascular calcification in chronic kidney disease by targeting NFAT5. Cell Death Dis. 2022, 13, 278. [Google Scholar] [CrossRef] [PubMed]
  60. Cui, L.; Zhou, Q.; Zheng, X.; Sun, B.; Zhao, S. Mitoquinone attenuates vascular calcification by suppressing oxidative stress and reducing apoptosis of vascular smooth muscle cells via the Keap1/Nrf2 pathway. Free Radic. Biol. Med. 2020, 161, 23–31. [Google Scholar] [CrossRef] [PubMed]
  61. Neels, J.G.; Gollentz, C.; Chinetti, G. Macrophage death in atherosclerosis: Potential role in calcification. Front. Immunol. 2023, 14, 1215612. [Google Scholar] [CrossRef] [PubMed]
  62. Tóth, A.; Lente, G.; Csiki, D.M.; Balogh, E.; Szöőr, Á.; Nagy, B., Jr.; Jeney, V. Activation of PERK/eIF2α/ATF4/CHOP branch of endoplasmic reticulum stress response and cooperation between HIF-1α and ATF4 promotes Daprodustat-induced vascular calcification. Front. Pharmacol. 2024, 15, 1399248. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, Y.; You, Y.; Wang, X.; Jin, Y.; Zeng, Y.; Pan, Z.; Li, D.; Ling, W. β-Hydroxybutyrate Alleviates Atherosclerotic Calcification by Inhibiting Endoplasmic Reticulum Stress-Mediated Apoptosis via AMPK/Nrf2 Pathway. Nutrients 2024, 17, 111. [Google Scholar] [CrossRef] [PubMed]
  64. Zhou, X.; Xu, S.N.; Yuan, S.T.; Lei, X.; Sun, X.; Xing, L.; Li, H.J.; He, C.X.; Qin, W.; Zhao, D.; et al. Multiple functions of autophagy in vascular calcification. Cell Biosci. 2021, 11, 159. [Google Scholar] [CrossRef]
  65. Wang, H.; Sun, L.; Su, L.; Rizo, J.; Liu, L.; Wang, L.F.; Wang, F.S.; Wang, X. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol. Cell 2014, 54, 133–146. [Google Scholar] [CrossRef] [PubMed]
  66. Dondelinger, Y.; Declercq, W.; Montessuit, S.; Roelandt, R.; Goncalves, A.; Bruggeman, I.; Hulpiau, P.; Weber, K.; Sehon, C.A.; Marquis, R.W.; et al. MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates. Cell Rep. 2014, 7, 971–981. [Google Scholar] [CrossRef] [PubMed]
  67. Khoury, M.K.; Gupta, K.; Franco, S.R.; Liu, B. Necroptosis in the Pathophysiology of Disease. Am. J. Pathol. 2020, 190, 272–285. [Google Scholar] [CrossRef] [PubMed]
  68. Gupta, K.; Phan, N.; Wang, Q.; Liu, B. Necroptosis in cardiovascular disease—A new therapeutic target. J. Mol. Cell Cardiol. 2018, 118, 26–35. [Google Scholar] [CrossRef]
  69. Inciarte-Mundo, J.; Frade-Sosa, B.; Sanmartí, R. From bench to bedside: Calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front. Immunol. 2022, 13, 1001025. [Google Scholar] [CrossRef] [PubMed]
  70. Amaya-Garrido, A.; Brunet, M.; Buffin-Meyer, B.; Piedrafita, A.; Grzesiak, L.; Agbegbo, E.; Del Bello, A.; Ferrandiz, I.; Ardeleanu, S.; Bermudez-Lopez, M.; et al. Calprotectin is a contributor to and potential therapeutic target for vascular calcification in chronic kidney disease. Sci. Transl. Med. 2023, 15, eabn5939. [Google Scholar] [CrossRef]
  71. Son, M.; Oh, S.; Jang, J.T.; Park, C.H.; Son, K.H.; Byun, K. Attenuating Effects of Pyrogallol-Phloroglucinol-6,6-Bieckol on Vascular Smooth Muscle Cell Phenotype Changes to Osteoblastic Cells and Vascular Calcification Induced by High Fat Diet. Nutrients 2020, 12, 2777. [Google Scholar] [CrossRef]
  72. Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016, 118, 692–702. [Google Scholar] [CrossRef] [PubMed]
  73. Dorozhkin, S.V.; Epple, M. Biological and medical significance of calcium phosphates. Angew. Chem. Int. Ed. Engl. 2002, 41, 3130–3146. [Google Scholar] [CrossRef]
  74. Yang, Z.; Wang, Y.; Zhang, Y.; He, X.; Zhong, C.Q.; Ni, H.; Chen, X.; Liang, Y.; Wu, J.; Zhao, S.; et al. RIP3 targets pyruvate dehydrogenase complex to increase aerobic respiration in TNF-induced necroptosis. Nat. Cell Biol. 2018, 20, 186–197. [Google Scholar] [CrossRef]
  75. Phadwal, K.; Vrahnas, C.; Ganley, I.G.; MacRae, V.E. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front. Cell Dev. Biol. 2021, 9, 611922. [Google Scholar] [CrossRef] [PubMed]
  76. Zhu, Y.; Han, X.Q.; Sun, X.J.; Yang, R.; Ma, W.Q.; Liu, N.F. Lactate accelerates vascular calcification through NR4A1-regulated mitochondrial fission and BNIP3-related mitophagy. Apoptosis 2020, 25, 321–340. [Google Scholar] [CrossRef]
  77. More, S.A.; Ghosh, A.; Kulkarni, O.P.; Mulay, S.R. Role of Persistent Necroinflammation in Chronic Tissue Remodelling and Organ Fibrosis. Am. J. Physiol. Cell Physiol. 2025. [Google Scholar] [CrossRef]
  78. Byun, K.A.; Oh, S.; Yang, J.Y.; Lee, S.Y.; Son, K.H.; Byun, K. Ecklonia cava extracts decrease hypertension-related vascular calcification by modulating PGC-1α and SOD2. Biomed. Pharmacother. 2022, 153, 113283. [Google Scholar] [CrossRef] [PubMed]
  79. Ding, Z.; Wang, R.; Li, Y.; Wang, X. MLKL activates the cGAS-STING pathway by releasing mitochondrial DNA upon necroptosis induction. Mol. Cell 2025, 85, 2610–2625.e2615. [Google Scholar] [CrossRef]
  80. Levine, B.; Kroemer, G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 2019, 176, 11–42. [Google Scholar] [CrossRef] [PubMed]
  81. Yamamoto, H.; Zhang, S.; Mizushima, N. Autophagy genes in biology and disease. Nat. Rev. Genet. 2023, 24, 382–400. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, Z.; Klionsky, D.J. Mammalian autophagy: Core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 2010, 22, 124–131. [Google Scholar] [CrossRef] [PubMed]
  83. Okamoto, K. Organellophagy: Eliminating cellular building blocks via selective autophagy. J. Cell Biol. 2014, 205, 435–445. [Google Scholar] [CrossRef]
  84. Phadwal, K.; Koo, E.; Jones, R.A.; Forsythe, R.O.; Tang, K.; Tang, Q.; Corcoran, B.M.; Caporali, A.; MacRae, V.E. Metformin protects against vascular calcification through the selective degradation of Runx2 by the p62 autophagy receptor. J. Cell Physiol. 2022, 237, 4303–4316. [Google Scholar] [CrossRef]
  85. Peng, Y.Q.; Xiong, D.; Lin, X.; Cui, R.R.; Xu, F.; Zhong, J.Y.; Zhu, T.; Wu, F.; Mao, M.Z.; Liao, X.B.; et al. Oestrogen Inhibits Arterial Calcification by Promoting Autophagy. Sci. Rep. 2017, 7, 3549. [Google Scholar] [CrossRef] [PubMed]
  86. Shen, J.; Zhang, C.; Liu, Y.; Zhao, M.; Wang, Q.; Li, P.; Liu, R.; Wai Wong, V.K.; Zhang, C.; Sun, X. L-type calcium ion channel-mediated activation of autophagy in vascular smooth muscle cells via thonningianin A (TA) alleviates vascular calcification in type 2 diabetes mellitus. Eur. J. Pharmacol. 2023, 959, 176084. [Google Scholar] [CrossRef] [PubMed]
  87. Sun, Y.; Byon, C.H.; Yang, Y.; Bradley, W.E.; Dell’Italia, L.J.; Sanders, P.W.; Agarwal, A.; Wu, H.; Chen, Y. Dietary potassium regulates vascular calcification and arterial stiffness. JCI Insight 2017, 2, e94920. [Google Scholar] [CrossRef] [PubMed]
  88. Yue, J.; Jin, S.; Gu, S.; Sun, R.; Liang, Q. High concentration magnesium inhibits extracellular matrix calcification and protects articular cartilage via Erk/autophagy pathway. J. Cell Physiol. 2019, 234, 23190–23201. [Google Scholar] [CrossRef]
  89. Liu, C.Y.; Zhang, Y.H.; Li, R.B.; Zhou, L.Y.; An, T.; Zhang, R.C.; Zhai, M.; Huang, Y.; Yan, K.W.; Dong, Y.H.; et al. LncRNA CAIF inhibits autophagy and attenuates myocardial infarction by blocking p53-mediated myocardin transcription. Nat. Commun. 2018, 9, 29. [Google Scholar] [CrossRef]
  90. Xu, Z.; Ji, G.; Shen, J.; Wang, X.; Zhou, J.; Li, L. SOX9 and myocardin counteract each other in regulating vascular smooth muscle cell differentiation. Biochem. Biophys. Res. Commun. 2012, 422, 285–290. [Google Scholar] [CrossRef]
  91. Li, S.; Ni, Y.; Li, C.; Xiang, Q.; Zhao, Y.; Xu, H.; Huang, W.; Wang, Y.; Wang, Y.; Zhan, J.; et al. Long noncoding RNA SNHG1 alleviates high glucose-induced vascular smooth muscle cells calcification/senescence by post-transcriptionally regulating Bhlhe40 and autophagy via Atg10. J. Physiol. Biochem. 2023, 79, 83–105. [Google Scholar] [CrossRef]
  92. Xu, T.H.; Qiu, X.B.; Sheng, Z.T.; Han, Y.R.; Wang, J.; Tian, B.Y.; Yao, L. Restoration of microRNA-30b expression alleviates vascular calcification through the mTOR signaling pathway and autophagy. J. Cell Physiol. 2019, 234, 14306–14318. [Google Scholar] [CrossRef] [PubMed]
  93. Cao, J.; Chen, C.; Chen, Q.; Gao, Y.; Zhao, Z.; Yuan, Q.; Li, A.; Yang, S.; He, Y.; Zu, X.; et al. Extracellular vesicle miR-32 derived from macrophage promotes arterial calcification in mice with type 2 diabetes via inhibiting VSMC autophagy. J. Transl. Med. 2022, 20, 307. [Google Scholar] [CrossRef] [PubMed]
  94. Li, F.X.; Liu, J.J.; Xu, F.; Shan, S.K.; Zheng, M.H.; Lei, L.M.; Lin, X.; Guo, B.; Li, C.C.; Wu, F.; et al. Cold exposure protects against medial arterial calcification development via autophagy. J. Nanobiotechnol. 2023, 21, 226. [Google Scholar] [CrossRef]
  95. Liu, C.J.; Cheng, C.W.; Tsai, Y.S.; Huang, H.S. Crosstalk between Renal and Vascular Calcium Signaling: The Link between Nephrolithiasis and Vascular Calcification. Int. J. Mol. Sci. 2021, 22, 3590. [Google Scholar] [CrossRef]
  96. Hou, Y.C.; Zheng, C.M.; Chiu, H.W.; Liu, W.C.; Lu, K.C.; Lu, C.L. Role of Calcimimetics in Treating Bone and Mineral Disorders Related to Chronic Kidney Disease. Pharmaceuticals 2022, 15, 952. [Google Scholar] [CrossRef] [PubMed]
  97. Zhu, Y.; Zhang, J.L.; Yan, X.J.; Ji, Y.; Wang, F.F. Exploring a new mechanism between lactate and VSMC calcification: PARP1/POLG/UCP2 signaling pathway and imbalance of mitochondrial homeostasis. Cell Death Dis. 2023, 14, 598. [Google Scholar] [CrossRef]
  98. Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef]
  99. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  100. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef]
  101. Kumar, P.; Liu, C.; Suliburk, J.; Hsu, J.W.; Muthupillai, R.; Jahoor, F.; Minard, C.G.; Taffet, G.E.; Sekhar, R.V. Supplementing Glycine and N-Acetylcysteine (GlyNAC) in Older Adults Improves Glutathione Deficiency, Oxidative Stress, Mitochondrial Dysfunction, Inflammation, Physical Function, and Aging Hallmarks: A Randomized Clinical Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 75–89. [Google Scholar] [CrossRef]
  102. Mokhtari, S.; Mahdavi, A.H.; Jafarpour, F.; Andani, M.R.; Dattilo, M.; Nasr-Esfahani, M.H. Taurine, alpha lipoic acid and vitamin B6 ameliorate the reduced developmental competence of immature mouse oocytes exposed to methylglyoxal. Sci. Rep. 2024, 14, 17937. [Google Scholar] [CrossRef]
  103. Dixon, S.J.; Olzmann, J.A. The cell biology of ferroptosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 424–442. [Google Scholar] [CrossRef]
  104. Kang, R.; Kroemer, G.; Tang, D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic. Biol. Med. 2019, 133, 162–168. [Google Scholar] [CrossRef] [PubMed]
  105. Pan, W.; Jie, W.; Huang, H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm 2023, 4, e200. [Google Scholar] [CrossRef]
  106. Nakamura, T.; Naguro, I.; Ichijo, H. Iron homeostasis and iron-regulated ROS in cell death, senescence and human diseases. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 1398–1409. [Google Scholar] [CrossRef]
  107. Wen, Z.; Zhang, L.; Chen, C.; Liu, Y.; Wu, C.; Dai, C. A construction of novel iron-foam-based calcium phosphate/chitosan coating biodegradable scaffold material. Mater. Sci. Eng. C Mater. Biol. Appl. 2013, 33, 1022–1031. [Google Scholar] [CrossRef]
  108. Nakanishi, M.; Goto, A.; Iwasaki, T.; Nakanishi, T.; Kuma, A.; Nanami, M.; Kuragano, T. Effect of iron administration on the aortic iron content and vascular calcification in phosphorus-loaded chronic kidney disease rats. BMC Nephrol. 2023, 24, 373. [Google Scholar] [CrossRef] [PubMed]
  109. Song, Y.; Yang, N.; Si, H.; Liu, T.; Wang, H.; Geng, H.; Qin, Q.; Guo, Z. Iron overload impairs renal function and is associated with vascular calcification in rat aorta. Biometals 2022, 35, 1325–1339. [Google Scholar] [CrossRef] [PubMed]
  110. Kawada, S.; Nagasawa, Y.; Kawabe, M.; Ohyama, H.; Kida, A.; Kato-Kogoe, N.; Nanami, M.; Hasuike, Y.; Kuragano, T.; Kishimoto, H.; et al. Iron-induced calcification in human aortic vascular smooth muscle cells through interleukin-24 (IL-24), with/without TNF-alpha. Sci. Rep. 2018, 8, 658. [Google Scholar] [CrossRef] [PubMed]
  111. Ma, W.Q.; Sun, X.J.; Zhu, Y.; Liu, N.F. Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic. Biol. Med. 2021, 165, 229–242. [Google Scholar] [CrossRef]
  112. Tao, Y.; Wu, Y.; Jiang, C.; Wang, Q.; Geng, X.; Chen, L.; Zhou, S.; Wang, X.; Han, M.; Du, D.; et al. Saturated fatty acid promotes calcification via suppressing SIRT6 expression in vascular smooth muscle cells. J. Hypertens. 2023, 41, 393–401. [Google Scholar] [CrossRef] [PubMed]
  113. Lu, L.; Ye, Y.; Chen, Y.; Feng, L.; Huang, J.; Liang, Q.; Lan, Z.; Dong, Q.; Liu, X.; Li, Y.; et al. Oxidized phospholipid POVPC contributes to vascular calcification by triggering ferroptosis of vascular smooth muscle cells. FASEB J. 2024, 38, e23592. [Google Scholar] [CrossRef]
  114. Ye, Y.; Chen, A.; Li, L.; Liang, Q.; Wang, S.; Dong, Q.; Fu, M.; Lan, Z.; Li, Y.; Liu, X.; et al. Repression of the antiporter SLC7A11/glutathione/glutathione peroxidase 4 axis drives ferroptosis of vascular smooth muscle cells to facilitate vascular calcification. Kidney Int. 2022, 102, 1259–1275. [Google Scholar] [CrossRef]
  115. Shao, S.; Liu, Y.; Hong, W.; Mo, Y.; Shu, F.; Jiang, L.; Tan, N. Influence of FOSL1 Inhibition on Vascular Calcification and ROS Generation through Ferroptosis via P53-SLC7A11 Axis. Biomedicines 2023, 11, 635. [Google Scholar] [CrossRef]
  116. Furmanik, M.; Chatrou, M.; van Gorp, R.; Akbulut, A.; Willems, B.; Schmidt, H.; van Eys, G.; Bochaton-Piallat, M.L.; Proudfoot, D.; Biessen, E.; et al. Reactive Oxygen-Forming Nox5 Links Vascular Smooth Muscle Cell Phenotypic Switching and Extracellular Vesicle-Mediated Vascular Calcification. Circ. Res. 2020, 127, 911–927. [Google Scholar] [CrossRef]
  117. Lu, L.; Li, Y.; Dong, Q.; Fang, J.; Chen, A.; Lan, Z.; Ye, Y.; Yan, J.; Liang, Q. Wogonin inhibits oxidative stress and vascular calcification via modulation of heme oxygenase-1. Eur. J. Pharmacol. 2023, 958, 176070. [Google Scholar] [CrossRef] [PubMed]
  118. Li, Y.; Pan, Y.; Wang, L.; Wang, X.; Chu, H.; Li, Y.; Mu, Y.; Sun, J. 3-Arylcoumarin inhibits vascular calcification by inhibiting the generation of AGEs and anti-oxidative stress. J. Enzyme Inhib. Med. Chem. 2022, 37, 2147–2157. [Google Scholar] [CrossRef] [PubMed]
  119. Alves-Lopes, R.; Neves, K.B.; Strembitska, A.; Harvey, A.P.; Harvey, K.Y.; Yusuf, H.; Haniford, S.; Hepburn, R.T.; Dyet, J.; Beattie, W.; et al. Osteoprotegerin regulates vascular function through syndecan-1 and NADPH oxidase-derived reactive oxygen species. Clin. Sci. 2021, 135, 2429–2444. [Google Scholar] [CrossRef]
  120. Liu, S.M.; Zhang, Y.R.; Chen, Y.; Ji, D.R.; Zhao, J.; Fu, S.; Jia, M.Z.; Yu, Y.R.; Tang, C.S.; Huang, W.; et al. Intermedin Alleviates Vascular Calcification in CKD through Sirtuin 3-Mediated Inhibition of Mitochondrial Oxidative Stress. Pharmaceuticals 2022, 15, 1224. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, Y.; Zhang, Y.; Gao, M.; Chen, Z.; Lu, J.; Li, Y.; Di, Y.; Zhao, Y.; Liu, B.; Tang, R. Lipocalin-2 promotes CKD vascular calcification by aggravating VSMCs ferroptosis through NCOA4/FTH1-mediated ferritinophagy. Cell Death Dis. 2024, 15, 865. [Google Scholar] [CrossRef]
  122. Zhu, X.X.; Meng, X.Y.; Chen, G.; Su, J.B.; Fu, X.; Xu, A.J.; Liu, Y.; Hou, X.H.; Qiu, H.B.; Sun, Q.Y.; et al. Nesfatin-1 enhances vascular smooth muscle calcification through facilitating BMP-2 osteogenic signaling. Cell Commun. Signal 2024, 22, 488. [Google Scholar] [CrossRef] [PubMed]
  123. Zhong, P.; Li, L.; Feng, X.; Teng, C.; Cai, W.; Zheng, W.; Wei, J.; Li, X.; He, Y.; Chen, B.; et al. Neuronal ferroptosis and ferroptosis-mediated endoplasmic reticulum stress: Implications in cognitive dysfunction induced by chronic intermittent hypoxia in mice. Int. Immunopharmacol. 2024, 138, 112579. [Google Scholar] [CrossRef]
  124. Wu, H.; Liu, Q.; Shan, X.; Gao, W.; Chen, Q. ATM orchestrates ferritinophagy and ferroptosis by phosphorylating NCOA4. Autophagy 2023, 19, 2062–2077. [Google Scholar] [CrossRef]
  125. Chen, L.; Min, J.; Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 2022, 7, 378. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, D.; Tian, Z.; Zhang, P.; Zhen, L.; Meng, Q.; Sun, B.; Xu, X.; Jia, T.; Li, S. The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed. Pharmacother. 2023, 163, 114830. [Google Scholar] [CrossRef]
  127. Przybyla-Toscano, J.; Maclean, A.E.; Franceschetti, M.; Liebsch, D.; Vignols, F.; Keech, O.; Rouhier, N.; Balk, J. Protein lipoylation in mitochondria requires Fe-S cluster assembly factors NFU4 and NFU5. Plant Physiol. 2022, 188, 997–1013. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, W.; Lu, K.; Jiang, X.; Wei, Q.; Zhu, L.; Wang, X.; Jin, H.; Feng, L. Ferroptosis inducers enhanced cuproptosis induced by copper ionophores in primary liver cancer. J. Exp. Clin. Cancer Res. 2023, 42, 142. [Google Scholar] [CrossRef] [PubMed]
  129. Li, S.R.; Bu, L.L.; Cai, L. Cuproptosis: Lipoylated TCA cycle proteins-mediated novel cell death pathway. Signal Transduct. Target. Ther. 2022, 7, 158. [Google Scholar] [CrossRef] [PubMed]
  130. Tang, D.; Chen, X.; Kroemer, G. Cuproptosis: A copper-triggered modality of mitochondrial cell death. Cell Res. 2022, 32, 417–418. [Google Scholar] [CrossRef]
  131. Xiong, C.; Ling, H.; Hao, Q.; Zhou, X. Cuproptosis: p53-regulated metabolic cell death? Cell Death Differ. 2023, 30, 876–884. [Google Scholar] [CrossRef] [PubMed]
  132. Sun, W.; Sun, M.; Zhang, M.; Liu, Y.; Lin, X.; Zhao, S.; Ma, L. Correlation between conjunctival and corneal calcification and cardiovascular calcification in patients undergoing maintenance hemodialysis. Hemodial. Int. 2015, 19, 270–278. [Google Scholar] [CrossRef]
  133. Wang, C.L.; Lin, K.P.; Hsu, G.W.; Liu, K.L.; Guo, C.H. Altered Mineral Metabolism and Disequilibrium Between Calcification Promoters and Inhibitors in Chronic Hemodialysis Patients. Biol. Trace Elem. Res. 2020, 193, 14–22. [Google Scholar] [CrossRef]
  134. Liu, C.; Liang, D. High Copper Intake Is Associated with Decreased Likelihood of Abdominal Aortic Calcification in Middle-Aged and Older US Adults. Biol. Trace Elem. Res. 2024, 202, 1390–1400. [Google Scholar] [CrossRef]
  135. Qi, R.Q.; Chen, Y.F.; Cheng, J.; Song, J.W.; Chen, Y.H.; Wang, S.Y.; Liu, Y.; Yan, K.X.; Liu, X.Y.; Li, J.; et al. Elabela alleviates cuproptosis and vascular calcification in vitaminD3- overloaded mice via regulation of the PPAR-γ /FDX1 signaling. Mol. Med. 2024, 30, 223. [Google Scholar] [CrossRef] [PubMed]
  136. Chen, X.; Cai, Q.; Liang, R.; Zhang, D.; Liu, X.; Zhang, M.; Xiong, Y.; Xu, M.; Liu, Q.; Li, P.; et al. Copper homeostasis and copper-induced cell death in the pathogenesis of cardiovascular disease and therapeutic strategies. Cell Death Dis. 2023, 14, 105. [Google Scholar] [CrossRef]
  137. Reiss, A.B.; Miyawaki, N.; Moon, J.; Kasselman, L.J.; Voloshyna, I.; D’Avino, R., Jr.; De Leon, J. CKD, arterial calcification, atherosclerosis and bone health: Inter-relationships and controversies. Atherosclerosis 2018, 278, 49–59. [Google Scholar] [CrossRef] [PubMed]
  138. Zhou, D.; Mao, Q.; Sun, Y.; Cheng, H.; Zhao, J.; Liu, Q.; Deng, M.; Xu, S.; Zhao, X. Association of Blood Copper With the Subclinical Carotid Atherosclerosis: An Observational Study. J. Am. Heart Assoc. 2024, 13, e033474. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, Y.T.; Xu, X.H.; Lin, L.; Tian, S.; Wu, G.F. Identification of Three Cuproptosis-specific Expressed Genes as Diagnostic Biomarkers and Therapeutic Targets for Atherosclerosis. Int. J. Med. Sci. 2023, 20, 836–848. [Google Scholar] [CrossRef]
  140. Cui, Y.; Chen, Y.; Gan, N.; Li, M.; Liao, W.; Zhou, Y.; Xiang, Q.; Gong, X.; Guo, Q.; Hu, P.; et al. A novel cuproptosis-related diagnostic gene signature and differential expression validation in atherosclerosis. Mol. Biomed. 2023, 4, 21. [Google Scholar] [CrossRef] [PubMed]
  141. Tsvetkov, P.; Coy, S.; Petrova, B.; Dreishpoon, M.; Verma, A.; Abdusamad, M.; Rossen, J.; Joesch-Cohen, L.; Humeidi, R.; Spangler, R.D.; et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 2022, 375, 1254–1261. [Google Scholar] [CrossRef]
  142. Ba, L.; Gao, J.; Chen, Y.; Qi, H.; Dong, C.; Pan, H.; Zhang, Q.; Shi, P.; Song, C.; Guan, X.; et al. Allicin attenuates pathological cardiac hypertrophy by inhibiting autophagy via activation of PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways. Phytomedicine 2019, 58, 152765. [Google Scholar] [CrossRef]
  143. Guo, J.; Cheng, J.; Zheng, N.; Zhang, X.; Dai, X.; Zhang, L.; Hu, C.; Wu, X.; Jiang, Q.; Wu, D.; et al. Copper Promotes Tumorigenesis by Activating the PDK1-AKT Oncogenic Pathway in a Copper Transporter 1 Dependent Manner. Adv. Sci. 2021, 8, e2004303. [Google Scholar] [CrossRef] [PubMed]
  144. Zhao, L.; Yang, N.; Song, Y.; Si, H.; Qin, Q.; Guo, Z. Effect of iron overload on endothelial cell calcification and its mechanism. Ann. Transl. Med. 2021, 9, 1658. [Google Scholar] [CrossRef]
  145. Xue, Q.; Kang, R.; Klionsky, D.J.; Tang, D.; Liu, J.; Chen, X. Copper metabolism in cell death and autophagy. Autophagy 2023, 19, 2175–2195. [Google Scholar] [CrossRef] [PubMed]
  146. Patel, J.J.; Bourne, L.E.; Thakur, S.; Farrington, K.; Gorog, D.A.; Orriss, I.R.; Baydoun, A.R. 2-Oxothiazolidine-4-carboxylic acid inhibits vascular calcification via induction of glutathione synthesis. J. Cell Physiol. 2021, 236, 2696–2705. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, H.; Zhang, S.J.; Lyn, N.; Florentino, A.; Li, A.; Davies, K.J.A.; Forman, H.J. Down regulation of glutathione and glutamate cysteine ligase in the inflammatory response of macrophages. Free Radic. Biol. Med. 2020, 158, 53–59. [Google Scholar] [CrossRef] [PubMed]
  148. Tóth, A.; Balogh, E.; Jeney, V. Regulation of Vascular Calcification by Reactive Oxygen Species. Antioxidants 2020, 9, 963. [Google Scholar] [CrossRef]
  149. Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef] [PubMed]
  150. Zhong, Y.; Kinio, A.; Saleh, M. Functions of NOD-Like Receptors in Human Diseases. Front. Immunol. 2013, 4, 333. [Google Scholar] [CrossRef] [PubMed]
  151. Xue, Y.; Enosi Tuipulotu, D.; Tan, W.H.; Kay, C.; Man, S.M. Emerging Activators and Regulators of Inflammasomes and Pyroptosis. Trends Immunol. 2019, 40, 1035–1052. [Google Scholar] [CrossRef]
  152. Van Opdenbosch, N.; Lamkanfi, M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019, 50, 1352–1364. [Google Scholar] [CrossRef]
  153. Menu, P.; Vince, J.E. The NLRP3 inflammasome in health and disease: The good, the bad and the ugly. Clin. Exp. Immunol. 2011, 166, 1–15. [Google Scholar] [CrossRef]
  154. Nakashima, T.; Kobayashi, Y.; Yamasaki, S.; Kawakami, A.; Eguchi, K.; Sasaki, H.; Sakai, H. Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-kappaB ligand: Modulation of the expression by osteotropic factors and cytokines. Biochem. Biophys. Res. Commun. 2000, 275, 768–775. [Google Scholar] [CrossRef]
  155. Wang, K.; Sun, Q.; Zhong, X.; Zeng, M.; Zeng, H.; Shi, X.; Li, Z.; Wang, Y.; Zhao, Q.; Shao, F.; et al. Structural Mechanism for GSDMD Targeting by Autoprocessed Caspases in Pyroptosis. Cell 2020, 180, 941–955.e920. [Google Scholar] [CrossRef] [PubMed]
  156. Frank, D.; Vince, J.E. Pyroptosis versus necroptosis: Similarities, differences, and crosstalk. Cell Death Differ. 2019, 26, 99–114. [Google Scholar] [CrossRef] [PubMed]
  157. Zhaolin, Z.; Guohua, L.; Shiyuan, W.; Zuo, W. Role of pyroptosis in cardiovascular disease. Cell Prolif. 2019, 52, e12563. [Google Scholar] [CrossRef]
  158. Ho, L.C.; Chen, Y.H.; Wu, T.Y.; Kao, L.Z.; Hung, S.Y.; Liou, H.H.; Chen, P.C.; Tsai, P.J.; Lin, H.K.; Lee, Y.C.; et al. Phosphate burden induces vascular calcification through a NLRP3-caspase-1-mediated pyroptotic pathway. Life Sci. 2023, 332, 122123. [Google Scholar] [CrossRef] [PubMed]
  159. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed]
  160. Yu, C.; Zhang, C.; Kuang, Z.; Zheng, Q. The Role of NLRP3 Inflammasome Activities in Bone Diseases and Vascular Calcification. Inflammation 2021, 44, 434–449. [Google Scholar] [CrossRef] [PubMed]
  161. Zhang, X.; Yang, B. The serum levels of gasdermin D in uremic patients and its relationship with the prognosis: A prospective observational cohort study. Ren. Fail. 2024, 46, 2312534. [Google Scholar] [CrossRef]
  162. Li, Z.; Ji, S.; Jiang, M.L.; Xu, Y.; Zhang, C.J. The Regulation and Modification of GSDMD Signaling in Diseases. Front. Immunol. 2022, 13, 893912. [Google Scholar] [CrossRef]
  163. Wei, Y.; Lan, B.; Zheng, T.; Yang, L.; Zhang, X.; Cheng, L.; Tuerhongjiang, G.; Yuan, Z.; Wu, Y. GSDME-mediated pyroptosis promotes the progression and associated inflammation of atherosclerosis. Nat. Commun. 2023, 14, 929. [Google Scholar] [CrossRef] [PubMed]
  164. Yang, H.; Liu, Y.; Chen, G.; Zhou, B.; Xu, G.; Li, Q.; Zhu, L. Caspase-3/gasdermin-E axis facilitates the progression of coronary artery calcification by inducing the release of high mobility group box protein 1. Int. Immunopharmacol. 2024, 127, 111454. [Google Scholar] [CrossRef] [PubMed]
  165. Ruan, H.; Zhang, H.; Feng, J.; Luo, H.; Fu, F.; Yao, S.; Zhou, C.; Zhang, Z.; Bian, Y.; Jin, H.; et al. Inhibition of Caspase-1-mediated pyroptosis promotes osteogenic differentiation, offering a therapeutic target for osteoporosis. Int. Immunopharmacol. 2023, 124, 110901. [Google Scholar] [CrossRef] [PubMed]
  166. Truesdell, A.G.; Alasnag, M.A.; Kaul, P.; Rab, S.T.; Riley, R.F.; Young, M.N.; Batchelor, W.B.; Maehara, A.; Welt, F.G.; Kirtane, A.J. Intravascular Imaging During Percutaneous Coronary Intervention: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2023, 81, 590–605. [Google Scholar] [CrossRef]
  167. Mori, H.; Torii, S.; Kutyna, M.; Sakamoto, A.; Finn, A.V.; Virmani, R. Coronary Artery Calcification and its Progression: What Does it Really Mean? JACC Cardiovasc. Imaging 2018, 11, 127–142. [Google Scholar] [CrossRef] [PubMed]
  168. Okamoto, N.; Egami, Y.; Nohara, H.; Kawanami, S.; Sugae, H.; Kawamura, A.; Ukita, K.; Matsuhiro, Y.; Nakamura, H.; Yasumoto, K.; et al. Direct Comparison of Rotational vs Orbital Atherectomy for Calcified Lesions Guided by Optical Coherence Tomography. JACC Cardiovasc. Interv. 2023, 16, 2125–2136. [Google Scholar] [CrossRef] [PubMed]
  169. Kereiakes, D.J.; Virmani, R.; Hokama, J.Y.; Illindala, U.; Mena-Hurtado, C.; Holden, A.; Hill, J.M.; Lyden, S.P.; Ali, Z.A. Principles of Intravascular Lithotripsy for Calcific Plaque Modification. JACC Cardiovasc. Interv. 2021, 14, 1275–1292. [Google Scholar] [CrossRef]
  170. Kale, A.; Sankrityayan, H.; Anders, H.J.; Gaikwad, A.B. Klotho in kidney diseases: A crosstalk between the renin-angiotensin system and endoplasmic reticulum stress. Nephrol. Dial. Transplant. 2023, 38, 819–825. [Google Scholar] [CrossRef]
  171. Hu, H.J.; Zhou, S.H.; Liu, Q.M. The magic and mystery of statins in aging: The potent preventive and therapeutic agent. Int. J. Cardiol. 2015, 187, 58–59. [Google Scholar] [CrossRef] [PubMed]
  172. de Cavanagh, E.M.; Inserra, F.; Ferder, L. Angiotensin II blockade: How its molecular targets may signal to mitochondria and slow aging. Coincidences with calorie restriction and mTOR inhibition. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H15–H44. [Google Scholar] [CrossRef] [PubMed]
  173. Navarro-González, J.F.; Sánchez-Niño, M.D.; Donate-Correa, J.; Martín-Núñez, E.; Ferri, C.; Pérez-Delgado, N.; Górriz, J.L.; Martínez-Castelao, A.; Ortiz, A.; Mora-Fernández, C. Effects of Pentoxifylline on Soluble Klotho Concentrations and Renal Tubular Cell Expression in Diabetic Kidney Disease. Diabetes Care 2018, 41, 1817–1820. [Google Scholar] [CrossRef] [PubMed]
  174. Liang, Z.; Yang, Y.; Wu, X.; Lu, C.; Zhao, H.; Chen, K.; Zhao, A.; Li, X.; Xu, J. GAS6/Axl is associated with AMPK activation and attenuates H2O2-induced oxidative stress. Apoptosis 2023, 28, 485–497. [Google Scholar] [CrossRef] [PubMed]
  175. Lu, C.L.; Liao, M.T.; Hou, Y.C.; Fang, Y.W.; Zheng, C.M.; Liu, W.C.; Chao, C.T.; Lu, K.C.; Ng, Y.Y. Sirtuin-1 and Its Relevance in Vascular Calcification. Int. J. Mol. Sci. 2020, 21, 1593. [Google Scholar] [CrossRef]
  176. Van Campenhout, A.; Golledge, J. Osteoprotegerin, vascular calcification and atherosclerosis. Atherosclerosis 2009, 204, 321–329. [Google Scholar] [CrossRef] [PubMed]
  177. Fu, Y.X.; Gu, J.H.; Zhang, Y.R.; Tong, X.S.; Zhao, H.Y.; Yuan, Y.; Liu, X.Z.; Bian, J.C.; Liu, Z.P. Influence of osteoprotegerin on differentiation, activation, and apoptosis of Gaoyou duck embryo osteoclasts in vitro. Poult. Sci. 2013, 92, 1613–1620. [Google Scholar] [CrossRef]
  178. Jones, N.S.; Kshirsagar, S.; Mohanan, V.; Ramakrishnan, V.; Di Nucci, F.; Ma, L.; Mao, J.; Ding, H.; Klabunde, S.; Vucic, D.; et al. A phase I, randomized, ascending-dose study to assess safety, pharmacokinetics, and activity of GDC-8264, a RIP1 inhibitor, in healthy volunteers. Clin. Transl. Sci. 2023, 16, 1997–2009. [Google Scholar] [CrossRef] [PubMed]
  179. 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]
  180. 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]
  181. Weisel, K.; Scott, N.E.; Tompson, D.J.; Votta, B.J.; Madhavan, S.; Povey, K.; Wolstenholme, A.; Simeoni, M.; Rudo, T.; Richards-Peterson, L.; et al. Randomized clinical study of safety, pharmacokinetics, and pharmacodynamics of RIPK1 inhibitor GSK2982772 in healthy volunteers. Pharmacol. Res. Perspect. 2017, 5, e00365. [Google Scholar] [CrossRef]
  182. Yang, H.; Xu, G.; Li, Q.; Zhu, L. Ligustrazine alleviates the progression of coronary artery calcification by inhibiting caspase-3/GSDME mediated pyroptosis. Biosci. Trends 2024, 18, 482–491. [Google Scholar] [CrossRef] [PubMed]
  183. Zheng, J.; Lin, Z.; Zhong, X.; Liu, J. The relationship between programmed cell death and vascular calcification. Front. Cardiovasc. Med. 2025, 12, 1549857. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The regulatory network of apoptosis in vascular calcification (VC). Apoptosis is initiated through three major signaling cascades: extrinsic, intrinsic, and endoplasmic reticulum (ER) stress pathways. These pathways converge on effector caspases and the formation of apoptotic bodies. Apoptosis contributes to VC through two primary mechanisms: (i) Direct calcification: Apoptotic bodies serve as nucleation sites for calcium and phosphate crystal deposition. (ii) Osteogenic differentiation: Key apoptotic mediators, particularly C/EBP-homologous protein (CHOP) downstream of ER stress, directly transactivate osteogenic genes (Runx2, BMP2), promoting vascular smooth muscle cells (VSMCs) transdifferentiation into an osteoblast-like phenotype. Additionally, the Gas6/Axl pathway mediates apoptotic cells to become foci for calcium phosphate crystal deposition.
Figure 1. The regulatory network of apoptosis in vascular calcification (VC). Apoptosis is initiated through three major signaling cascades: extrinsic, intrinsic, and endoplasmic reticulum (ER) stress pathways. These pathways converge on effector caspases and the formation of apoptotic bodies. Apoptosis contributes to VC through two primary mechanisms: (i) Direct calcification: Apoptotic bodies serve as nucleation sites for calcium and phosphate crystal deposition. (ii) Osteogenic differentiation: Key apoptotic mediators, particularly C/EBP-homologous protein (CHOP) downstream of ER stress, directly transactivate osteogenic genes (Runx2, BMP2), promoting vascular smooth muscle cells (VSMCs) transdifferentiation into an osteoblast-like phenotype. Additionally, the Gas6/Axl pathway mediates apoptotic cells to become foci for calcium phosphate crystal deposition.
Biomolecules 15 01640 g001
Figure 2. The regulatory network of necroptosis in vascular calcification (VC). Necroptosis involves the sequential assembly of the core necrosome complex, which results in the membrane translocation and pore formation. Necroptosis drives VC progression through two primary mechanisms: (i) Mitochondrial dysfunction: Necrosome activation impairs mitochondria, leading to the generation of reactive oxygen species (ROS) and metabolic changes that drive vascular smooth muscle cells (VSMCs) osteogenic transformation; (ii) Inflammatory amplification: Membrane rupture causes the release of damage-associated molecular patterns (DAMPs), establishing a robust pro-inflammatory microenvironment rich in cytokines that promotes osteogenic transdifferentiation. Various alternative triggers and modulatory factors are also included.
Figure 2. The regulatory network of necroptosis in vascular calcification (VC). Necroptosis involves the sequential assembly of the core necrosome complex, which results in the membrane translocation and pore formation. Necroptosis drives VC progression through two primary mechanisms: (i) Mitochondrial dysfunction: Necrosome activation impairs mitochondria, leading to the generation of reactive oxygen species (ROS) and metabolic changes that drive vascular smooth muscle cells (VSMCs) osteogenic transformation; (ii) Inflammatory amplification: Membrane rupture causes the release of damage-associated molecular patterns (DAMPs), establishing a robust pro-inflammatory microenvironment rich in cytokines that promotes osteogenic transdifferentiation. Various alternative triggers and modulatory factors are also included.
Biomolecules 15 01640 g002
Figure 3. The regulatory network of autophagy in vascular calcification (VC). Autophagy is a sequential cellular process involving the formation, elongation, and closure of the autophagosome, culminating in lysosomal degradation. The autophagic process is tightly modulated by various upstream regulators, including intracellular ion concentrations, non-coding RNAs, hormonal factors, and diverse environmental cues. Autophagy directly regulates VC through the degradation of key pro-calcific factors. The autophagic degradation of Runx2 represents a primary protective mechanism by suppressing osteogenic gene expression. Conversely, excessive autophagy may promote calcification via degradation of the calcium-sensing receptor (CaSR), disrupting calcium homeostasis. Excessive autophagy also promotes VC via facilitating vascular osteogenic conversion and mineralization, but the underlying mechanisms remain unclear.
Figure 3. The regulatory network of autophagy in vascular calcification (VC). Autophagy is a sequential cellular process involving the formation, elongation, and closure of the autophagosome, culminating in lysosomal degradation. The autophagic process is tightly modulated by various upstream regulators, including intracellular ion concentrations, non-coding RNAs, hormonal factors, and diverse environmental cues. Autophagy directly regulates VC through the degradation of key pro-calcific factors. The autophagic degradation of Runx2 represents a primary protective mechanism by suppressing osteogenic gene expression. Conversely, excessive autophagy may promote calcification via degradation of the calcium-sensing receptor (CaSR), disrupting calcium homeostasis. Excessive autophagy also promotes VC via facilitating vascular osteogenic conversion and mineralization, but the underlying mechanisms remain unclear.
Biomolecules 15 01640 g003
Figure 4. The regulatory network of ferroptosis in vascular calcification (VC). Ferroptosis is an iron-dependent form of regulated cell death, marked by the accumulation of lipid peroxides and reactive oxygen species (ROS). Ferroptosis directly promotes VC through the following downstream mechanisms: (i) Mineral deposition coupling: Ferroptosis-generated ROS induces endoplasmic reticulum (ER) stress, triggering Ca2+ release that facilitates calcium-phosphate deposition and directly initiates mineralization; (ii) Amplification loop: Ferroptosis-associated oxidative stress triggers nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy, which degrades iron storage proteins and increases labile iron, thereby amplifying ferroptosis and its pro-calcific effects; (iii) 1-Palmitoyl-2-(5′-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) upregulates the expression of Runx2 and BMP2, inducing vascular smooth muscle cells (VSMCs) osteogenic transdifferentiation.
Figure 4. The regulatory network of ferroptosis in vascular calcification (VC). Ferroptosis is an iron-dependent form of regulated cell death, marked by the accumulation of lipid peroxides and reactive oxygen species (ROS). Ferroptosis directly promotes VC through the following downstream mechanisms: (i) Mineral deposition coupling: Ferroptosis-generated ROS induces endoplasmic reticulum (ER) stress, triggering Ca2+ release that facilitates calcium-phosphate deposition and directly initiates mineralization; (ii) Amplification loop: Ferroptosis-associated oxidative stress triggers nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy, which degrades iron storage proteins and increases labile iron, thereby amplifying ferroptosis and its pro-calcific effects; (iii) 1-Palmitoyl-2-(5′-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) upregulates the expression of Runx2 and BMP2, inducing vascular smooth muscle cells (VSMCs) osteogenic transdifferentiation.
Biomolecules 15 01640 g004
Figure 5. The regulatory network of cuproptosis in vascular calcification (VC). The cuproptosis and VC are linked by cuprooptosis-specific and related expressed genes, atherosclerosis, ferroptosis, and reactive oxygen species (ROS). Copper ions enter cells via copper transporter 1 (CTR1) and can activate multiple signaling pathways that lead to cuproptosis. Cuproptosis directly contributes to VC through two primary downstream pathways: (i) Mitochondrial proteotoxic stress driving osteogenic signaling: Copper-induced aggregation of lipoylated proteins and Fe-S cluster destabilization trigger mitochondrial proteotoxic stress. This results in a burst of mitochondrial ROS, which directly activates pro-osteogenic pathways (BMP-2); (ii) Direct activation of pro-calcific kinases: Intracellular copper ions directly activate key signaling cascades, including MAPK/ERK1/2 and PI3K/Akt/mTOR, which are known to phosphorylate and activate transcription factors driving the osteogenic transition of vascular smooth muscle cells (VSMCs).
Figure 5. The regulatory network of cuproptosis in vascular calcification (VC). The cuproptosis and VC are linked by cuprooptosis-specific and related expressed genes, atherosclerosis, ferroptosis, and reactive oxygen species (ROS). Copper ions enter cells via copper transporter 1 (CTR1) and can activate multiple signaling pathways that lead to cuproptosis. Cuproptosis directly contributes to VC through two primary downstream pathways: (i) Mitochondrial proteotoxic stress driving osteogenic signaling: Copper-induced aggregation of lipoylated proteins and Fe-S cluster destabilization trigger mitochondrial proteotoxic stress. This results in a burst of mitochondrial ROS, which directly activates pro-osteogenic pathways (BMP-2); (ii) Direct activation of pro-calcific kinases: Intracellular copper ions directly activate key signaling cascades, including MAPK/ERK1/2 and PI3K/Akt/mTOR, which are known to phosphorylate and activate transcription factors driving the osteogenic transition of vascular smooth muscle cells (VSMCs).
Biomolecules 15 01640 g005
Figure 6. The regulatory network of pyroptosis in vascular calcification (VC). Pyroptosis is an inflammatory lytic cell death initiated by multiple convergence points: the canonical pathway, the incomplete pyroptosis pathway, and a distinct non-canonical pathway. All three cascades culminate in the cleavage of a gasdermin family protein, generating pore-forming fragments that induce cell lysis and the release of copious inflammatory cytokines. Pyroptosis directly drives VC through the downstream pathway: pore formation and pro-calcific factor release. Activated inflammatory caspases cleave GSDMD/GSDME, generating N-terminal fragments that oligomerize to form plasma membrane pores. This leads to the release of mature IL-1β and IL-18, which act directly on vascular smooth muscle cells (VSMCs), activating signaling pathways that directly upregulate the expression of key osteogenic master genes such as Runx2, thereby initiating the calcification program.
Figure 6. The regulatory network of pyroptosis in vascular calcification (VC). Pyroptosis is an inflammatory lytic cell death initiated by multiple convergence points: the canonical pathway, the incomplete pyroptosis pathway, and a distinct non-canonical pathway. All three cascades culminate in the cleavage of a gasdermin family protein, generating pore-forming fragments that induce cell lysis and the release of copious inflammatory cytokines. Pyroptosis directly drives VC through the downstream pathway: pore formation and pro-calcific factor release. Activated inflammatory caspases cleave GSDMD/GSDME, generating N-terminal fragments that oligomerize to form plasma membrane pores. This leads to the release of mature IL-1β and IL-18, which act directly on vascular smooth muscle cells (VSMCs), activating signaling pathways that directly upregulate the expression of key osteogenic master genes such as Runx2, thereby initiating the calcification program.
Biomolecules 15 01640 g006
Table 1. Summarization of emerging drugs focusing on PCD and their corresponding targets.
Table 1. Summarization of emerging drugs focusing on PCD and their corresponding targets.
Targeted PCDNameMolecular Target
ApoptosisRAS inhibitorsα-Klotho protein
 Statinsα-Klotho protein
 mTOR inhibitorsα-Klotho protein
 Vitamin Dα-Klotho protein
 Pentophyllineα-Klotho protein
 Vitamin K2Gas6
 Sirt1 activatorsSirtuin1
 Vitamin DSirtuin1
 OsteoprotegerinRANKL
NecroptosisGDC-8264RIP 1
 SIR2446MRIPK 1
 GFH312RIPK 1
 DNL104RIPK 1
 GSK2982772RIPK 1
AutophagyIrisinNLRP3
 β-hydroxybutyric acidLC3B
 BavachinmTOR
 CalycosinmTOR
 FPSNot discovered yet
 Atorvastatinβ-catenin
 OestrogenErα
 GhrelinAMPK
 Exendin 4mTOR
 MelatoninOptic Atrophy 1
FerroptosisMetforminPOSTN
 DNA polymerase GammaP53
 PHGDHP53
 OTCGSH
 OleoylethanolamidePPARα
CuproptosisElabelaPPAR-γ
PyroptosisPuerarinNLRP3
 IL-38NLRP3
 EmpagliflozinNLRP3
 CanagliflozinNLRP3
 SinomenineNLRP3
 GenisteinNLRP3
 VX-765Caspase 1
 LigustrazineCaspase 3
Gas6 = Growth Arrest-Specific Gene 6; RANKL = Receptor Activator for Nuclear Factor-κ B Ligand; RIP 1 = Receptor-Interacting Protein 1; FPS = Polysaccharide from Fuzi; NLRP3 = NOD-like Receptor Pyrin Domain Containing 3; LC3B = Microtubule-Associated Protein 1A/1B-Light Chain 3B; AMPK = Adenosine 5′-Monophosphate-Activated Protein Kinase; mTOR = Mammalian Target of Rapamycin; POSTN = Extracellular Matrix Protein Periostin; PHGDH = Phosphoglycerate Dehydrogenase; OTC = 2-Oxothiazolidine-4-Carboxylic Acid; SLC7A11 = Solute Carrier Family 7 Member 11; p53 = Tumor Protein 53; GSH = glutathione; PPARα = Peroxisome Proliferator-Activated Receptor α; GSDME = Gasdermin E; IL-38 = Interleukin-38.
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

Cao, Y.; Cao, Y.; Liu, J.; Ye, Y.; Jiang, M. Comprehensive Review of Mechanisms and Translational Perspectives on Programmed Cell Death in Vascular Calcification. Biomolecules 2025, 15, 1640. https://doi.org/10.3390/biom15121640

AMA Style

Cao Y, Cao Y, Liu J, Ye Y, Jiang M. Comprehensive Review of Mechanisms and Translational Perspectives on Programmed Cell Death in Vascular Calcification. Biomolecules. 2025; 15(12):1640. https://doi.org/10.3390/biom15121640

Chicago/Turabian Style

Cao, Yiyang, Yulong Cao, Jiaheng Liu, Yifei Ye, and Meixiu Jiang. 2025. "Comprehensive Review of Mechanisms and Translational Perspectives on Programmed Cell Death in Vascular Calcification" Biomolecules 15, no. 12: 1640. https://doi.org/10.3390/biom15121640

APA Style

Cao, Y., Cao, Y., Liu, J., Ye, Y., & Jiang, M. (2025). Comprehensive Review of Mechanisms and Translational Perspectives on Programmed Cell Death in Vascular Calcification. Biomolecules, 15(12), 1640. https://doi.org/10.3390/biom15121640

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