Next Article in Journal
CsHY5 Regulates Light-Induced Anthocyanin Accumulation in Camellia sinensis
Previous Article in Journal
Characterization and Immune Functions of LcβLectin from Large Yellow Croaker (Larimichthys crocea): A Potential Antiviral Defense Molecule
Previous Article in Special Issue
Autophagy in High-Fat Diet and Streptozotocin-Induced Metabolic Cardiomyopathy: Mechanisms and Therapeutic Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis

1
Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China
2
Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China
3
Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(7), 3252; https://doi.org/10.3390/ijms26073252
Submission received: 27 January 2025 / Revised: 4 March 2025 / Accepted: 17 March 2025 / Published: 1 April 2025
(This article belongs to the Special Issue Cellular and Molecular Mechanisms of Myocardial Diseases)

Abstract

:
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.

1. Introduction

Atherosclerosis is a chronic inflammatory disease of the arterial wall, primarily driven by the accumulation of lipids and immune cells, such as macrophages, which contribute to plaque formation and progression. It remains the leading cause of cardiovascular disease (CVD), resulting in high morbidity and mortality worldwide. While traditionally associated with lipid metabolism disorders, recent advances in understanding atherosclerosis (AS) have highlighted the critical role of cellular processes such as autophagy and inflammation in disease pathogenesis [1,2]. Autophagy, a vital cellular process responsible for maintaining homeostasis, enables the degradation and recycling of damaged proteins and organelles. In macrophages, autophagy plays an essential role in regulating immune responses, lipid metabolism, and the inflammatory processes that are central to atherosclerotic plaque formation [3,4].
Macrophages in atherosclerotic lesions exhibit complex phenotypic changes, including polarization into pro-inflammatory M1 macrophages that exacerbate plaque instability and anti-inflammatory M2 macrophages that promote tissue repair [5,6]. These dynamic macrophage responses are further modulated by cellular pathways like autophagy, which not only maintains macrophage function but also influences the progression of atherosclerosis through its effects on lipid metabolism, cytokine production, and cellular senescence [7,8,9]. Autophagy defects in macrophages have been associated with increased oxidative stress, inflammatory cytokine release, and plaque rupture, thereby accelerating atherosclerosis progression [2,10].
Emerging evidence suggests that autophagy also modulates the fate of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) within the atherosclerotic microenvironment. For instance, lipophagy, a form of selective autophagy, regulates lipid accumulation in macrophages, facilitating cholesterol efflux and preventing foam cell formation [11,12,13]. However, atherosclerotic conditions impair autophagic activity, which leads to macrophage dysfunction, increased inflammation, and the destabilization of plaques [14,15,16]. This dysfunction is believed to be a critical factor in the progression of atherosclerosis and its complications [17], including myocardial infarction and stroke.
In addition to autophagy, clonal hematopoiesis (CH), a process whereby somatic mutations in hematopoietic stem and progenitor cells lead to the expansion of specific clones, has recently emerged as an important factor contributing to the development and progression of atherosclerosis. Mutations in genes such as ten-eleven translocation-2 (TET2), Janus kinase 2 (JAK2), and DNA methyltransferase 3 alpha (DNMT3A), which are frequently observed in patients with CH, have been shown to promote inflammatory responses and alter macrophage function in the context of atherosclerosis [18,19,20]. These genetic mutations lead to changes in immune cell activity and exacerbate the inflammatory processes underlying plaque formation, highlighting a potential intersection between genetic risk factors and inflammation in atherosclerosis [10].
Given the complex interplay between autophagy, inflammation, and CH in atherosclerosis (Figure 1), targeting these pathways holds promise for developing novel therapeutic strategies aimed at reducing plaque formation, preventing plaque rupture, and improving clinical outcomes in CVD [21,22]. Building upon recent advances in the field, we hypothesize that autophagy, cellular programmed cell death, and clonal hematopoiesis form a critical intrinsic axis, modulating the basic functions of macrophages, which in turn plays a multifaceted and complex role in the development and progression of atherosclerosis. Specifically, this axis is hypothesized to regulate macrophage homeostasis, inflammatory responses, and lipid metabolism, all of which are key drivers in atherosclerotic plaque formation and plaque instability. Moreover, autophagic processes in circulating and CH developed/maturated macrophages are essential not only for cellular maintenance and inflammation resolution but also for preventing exaggerated immune responses and cellular senescence. When this axis is dysregulated, as seen in the context of autophagy defects or somatic mutations associated with clonal hematopoiesis, macrophages are prone to heightened inflammatory states and abnormal lipid accumulation, leading to accelerated atherosclerosis progression. These insights suggest that a more integrated understanding of the molecular underpinnings linking autophagy and clonal hematopoiesis could reveal new therapeutic targets for managing atherosclerotic cardiovascular disease. Recent studies highlight the pharmacological modulation of autophagy as a promising strategy to restore macrophage function and stabilize atherosclerotic plaques. For instance, metformin activates autophagy and suppresses NLRP3 inflammasome activity, reducing plaque progression [23]. Chlorogenic acid enhances macrophage polarization and inflammation resolution by inducing autophagy, while rapamycin improves plaque stability by activating autophagy in macrophages [24,25,26]. Curcumin, a natural compound, also promotes autophagy in macrophages, alleviating atherosclerosis in animal models [27]. These findings, plus those discussed in this review, suggest that targeting autophagy could offer potential therapeutic benefits in preventing or treating atherosclerosis by improving macrophage function and plaque stability.

2. Chaperone-Mediated Autophagy (CMA) Initiates in Inflammation and Atherosclerosis

Chaperone-mediated autophagy (CMA) has emerged as a critical process connecting cellular stress responses to inflammation in the context of atherosclerosis. As a specialized form of autophagy, CMA selectively degrades damaged or misfolded proteins containing the KFERQ-like motif (Lys-Phe-Glu-Arg-Gln), which is often present in pro-inflammatory and stress-related proteins [28,29]. In macrophages, CMA plays a pivotal role in modulating the inflammatory environment within atherosclerotic plaques, influencing both plaque stability and disease progression [30]. Research indicates that impaired CMA activity in macrophages can exacerbate inflammatory responses by increasing the secretion of pro-inflammatory cytokines and the activation of inflammasomes, thereby accelerating atherosclerotic development [29]. Furthermore, CMA regulates lipid metabolism, cellular senescence, and oxidative stress, which are processes that are integral to the pathophysiology of atherosclerosis [31]. Expanding on recent findings, in conjunction with others from diverse cellular processes including other forms of autophagy, programmed cell death, and clonal hematopoiesis, CMA establishes an intricate regulatory network that governs macrophage function in atherosclerosis, in which macrophages can be resident from circulating blood and developed/matured CH. This network plays a critical role in macrophage homeostasis, inflammatory regulation, and lipid metabolism, all of which are essential for the development and progression of atherosclerosis. Dysregulation of this axis—whether through impaired CMA, autophagic dysfunction, or mutations related to clonal hematopoiesis—can amplify inflammation and lipid accumulation, accelerating disease progression. These insights highlight the potential for CMA as a therapeutic target for modulating inflammation and plaque stability, offering novel opportunities for therapeutic intervention aimed at controlling inflammation and stabilizing plaques.

2.1. Chaperone-Mediated Autophagy as a Specialized Form of Autophagy

Autophagy is a crucial catabolic process in cells, responsible for the degradation and recycling of cellular components such as proteins, lipids, nucleic acids, and organelles. This process not only maintains intracellular homeostasis but also supports cellular renewal by generating new components and energy [28]. Autophagy is broadly categorized into three forms: macroautophagy, microautophagy, and CMA. Unlike macroautophagy and microautophagy, CMA is a highly selective degradation pathway that specifically targets proteins containing the KFERQ motif [32].
In CMA, substrate proteins carrying the KFERQ motif are recognized by heat-shock cognate protein 70 (HSC70), which binds to the substrate and facilitates its translocation to lysosomes via lysosome-associated membrane protein type 2A (LAMP-2A). The protein is then unfolded, and the translocation complex is formed. HSC70 mediates the translocation of the substrate into the lysosome, where lysosomal proteases degrade it. Once degradation is complete, LAMP-2A dissociates from the complex, concluding the CMA process [33,34].
In the context of atherosclerosis, CMA may offer protective effects through several mechanisms. By degrading damaged proteins and organelles, CMA reduces oxidative stress and modulates inflammatory responses. For example, CMA helps prevent plaque progression by degrading oxidized low-density lipoprotein (ox-LDL) in foam cells [35]. Additionally, CMA has been shown to regulate the inflammatory response by inhibiting the secretion of pro-inflammatory cytokines, such as interleukin-1β (IL-1β), and controlling the activation of the NLRP3 inflammasome [29,36].

2.2. The Protective Role of Chaperone-Mediated Autophagy in Atherosclerosis

Chaperone-mediated autophagy (CMA) plays a critical protective role in atherosclerosis by modulating cellular homeostasis, particularly within macrophages, which are key players in the disease’s pathogenesis. A 2021 study using ApoE−/− mice and and macrophage-specific conditional LAMP-2A knockout mice revealed that CMA activity is significantly impaired during atherosclerosis progression. The study demonstrated reduced levels of LAMP-2A, a key marker of CMA, which correlated with increased secretion of pro-inflammatory cytokines IL-1β and IL-18. These cytokines, which are dependent on the NLRP3 inflammasome, were found to be elevated due to the impaired CMA-mediated degradation of NLRP3. Notably, the NLRP3 protein contains five KFERQ-like motifs that allow it to interact with HSC70 and LAMP-2A, which are essential components of CMA. This interaction is observed not only in macrophages exposed to lipopolysaccharide (LPS) and ATP but also in human coronary atherosclerotic plaques [29]. Thus, CMA-lysosomal degradation of NLRP3 plays a crucial role in reducing NLRP3 protein levels, which, in turn, helps to limit inflammation and atherosclerosis progression.
While CMA’s protective role has been studied in various diseases [30,37,38], its specific impact on atherosclerosis has only recently come under investigation. This research highlights the potential for pharmacologically activating CMA as a strategy to enhance NLRP3 inflammasome degradation, thus reducing inflammation and slowing atherosclerosis. This approach offers a promising avenue for the development of therapeutic interventions aimed at alleviating atherosclerotic disease [29].
Further corroborating CMA’s protective role in atherosclerosis, a 2022 study demonstrated that CMA inhibition exacerbates cardiovascular disease (CVD) in transgenic mouse models [39,40]. These studies, using KFERQ-PS-Dendra2 mice—expressing a fluorescent reporter for CMA—found that as atherosclerosis progressed, the fluorescence associated with CMA activity in plaques became nearly undetectable. In the systemic CMA-blocked LAMP-2A-deficient (L2AKO) mice, researchers showed that CMA deficiency worsens the metabolic and coagulation abnormalities induced by a high-cholesterol diet (Western-type diet, WD), making the mice more autophagy susceptible to atherosclerosis [35,41]. These findings underline that reduced CMA activity in atherosclerotic conditions accelerates disease progression, while promoting CMA could potentially mitigate its severity, presenting a valuable intervention strategy.
Macrophages, known for their pivotal role in atherosclerotic plaque dynamics, express high levels of key CMA effectors, including LAMP-2A and HSC70 [35]. When CMA is impaired, macrophages adopt a more pro-inflammatory phenotype, enhancing their contribution to plaque formation and inflammation. Moreover, CMA inhibition increases the vulnerability of vascular smooth muscle cells (VSMCs) to lipotoxicity, promoting their dedifferentiation and contributing to the autophagic pathological transformation of plaques [42,43]. These observations suggest that CMA is integral to maintaining macrophage homeostasis, modulating inflammatory responses, and protecting against lipid-induced damage in VSMCs. By regulating both macrophage polarization and VSMC function, CMA exerts a protective role in atherosclerosis progression.
Collectively, these studies highlight the importance of CMA in controlling lipid metabolism, inflammation, and cellular responses in the vascular wall. Activating CMA could offer a promising therapeutic strategy to modulate macrophage inflammatory status, prevent foam cell formation, and reduce plaque instability, thus halting the progression of atherosclerosis. This understanding opens the door to potential therapies aimed at enhancing CMA activity to curb cardiovascular disease.

3. Deficient Chaperone-Mediated Autophagy Promotes Lipid Accumulation in Macrophages

Macrophages play a pivotal role in the pathogenesis of atherosclerosis, primarily through their ability to internalize lipids and transform into foam cells, which contribute to plaque formation and instability. Lipid accumulation in macrophages is influenced by various processes, including lipid uptake, synthesis, and degradation [44,45]. Autophagy, particularly Chaperone-Mediated Autophagy (CMA), is critical for regulating lipid homeostasis by modulating lipid droplet turnover and degrading lipid-associated proteins. Dysfunction in CMA can lead to impaired lipid metabolism, promoting excessive lipid accumulation and accelerating atherosclerosis progression [46]. This disruption in lipid handling has been linked to altered macrophage function and inflammatory responses, further complicating atherosclerotic disease progression [47,48,49]. In this section, we examine the role of lipids in macrophage function and their contribution to atherosclerotic plaque development, with a particular focus on the impact of CMA dysfunction on lipid metabolism and foam cell formation (Table 1).

3.1. Macrophages and Their Fundemental Functions

Macrophages are highly versatile immune cells that perform a wide range of functions, critical to both homeostasis and disease pathology. They are able to adopt distinct phenotypic and functional states depending on the signals present in their local microenvironment. As central players in the immune response, macrophages maintain tissue homeostasis, regulate inflammation, and contribute to tissue repair by engulfing pathogens, cellular debris, and apoptotic cells, as well as secreting a diverse array of cytokines to modulate immune reactions [97].
Morphologically, macrophages are large, irregularly shaped cells with a highly adaptable, amoeboid form. Their cytoplasm is rich in organelles, including lysosomes, mitochondria, and rough endoplasmic reticulum, which are essential for their phagocytic activity and various other cellular processes. These cells are identified by surface markers such as CD14, CD40, CD11b, CD64, F4/80, EMR1, lysozyme M, MAC-1/MAC-3, and CD68, which serve to define their unique role in immune surveillance and response [98,99].
Macrophages exhibit remarkable plasticity, being classified into two primary polarization states: M1 and M2. M1 macrophages are typically induced by pro-inflammatory stimuli such as lipopolysaccharides (LPS) and interferon-gamma (IFN-γ). These cells are characterized by the production of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β), as well as reactive oxygen species (ROS), all of which contribute to inflammation and tissue damage [100]. On the other hand, M2 macrophages are driven by anti-inflammatory signals like interleukin-4 (IL-4) and IL-13, and they are primarily involved in tissue repair, wound healing, and the resolution of inflammation. M2 macrophages release anti-inflammatory cytokines such as IL-10 and facilitate the clearance of apoptotic cells [100].
Furthermore, macrophages exhibit functional diversity based on their anatomical location and activation status. For example, alveolar macrophages in the lungs play a crucial role in clearing inhaled pathogens and particulate matter, thereby contributing to pulmonary defense [101,102]. Kupffer cells in the liver are responsible for detoxifying metabolic byproducts and removing toxins from the bloodstream [103]. Microglia in the central nervous system monitor brain homeostasis, clear dead cells, and respond to injury. Inflammatory macrophages, recruited to sites of infection or injury, are classified into M1 and M2 subtypes based on their functional profiles and cytokine production [104,105].
Vascular macrophages represent a specialized subset of macrophages that reside in or are closely associated with blood vessels, where they play key roles in maintaining vascular integrity and homeostasis. These cells originate from multiple sources, including yolk sac progenitors, bone marrow-derived monocytes, and tissue-resident progenitors [106]. The developmental origins of vascular macrophages are complex, with embryonic yolk sac-derived CX3CR1+ endothelial microparticles (EMPs) and fetal liver monocytes contributing to the establishment of the tissue-resident macrophage population in the arterial wall during early development [107]. Vascular macrophages can be further classified into distinct subtypes, including M1, M2, Mox, M4, and Mhem macrophages, each with unique gene expression profiles and functional roles. For example, Mox macrophages are involved in heme detoxification and mitigating oxidative stress, while M4 macrophages produce chemokines and proteases that recruit additional immune cells and degrade extracellular matrix components [108,109].
In response to vascular injury, inflammation, or pathological conditions such as atherosclerosis or vascular calcification, circulating monocytes infiltrate the vessel wall and differentiate into macrophages. These macrophages play critical roles in vascular pathology, including foam cell formation in atherosclerotic plaques [110], promoting vascular calcification through the secretion of osteogenic factors [111], and facilitating tissue repair through the clearance of apoptotic cells and resolution of inflammation [112]. The plasticity of macrophages allows them to adapt to a variety of microenvironments, making them essential regulators of vascular health and disease.

3.2. Lipids Are Essential for Multiple Functions of Macrophages

Lipids play a crucial role in the diverse functions of macrophages, including efferocytosis (the process of clearing apoptotic cells), energy homeostasis, and the regulation of aging [113]. However, the role of lipids in macrophages is context-dependent, with specific lipid species serving different functions based on their cellular localization and the macrophage’s functional state. For example, macrophages can become adipogenic under certain conditions, contributing to diseases such as atherosclerosis [45,47,114]. Recent investigations have also highlighted the importance of lipid accumulation in tumor-associated macrophages (TAMs), lipid-associated macrophages (LAMs) in atherosclerosis, and the selective regulation of lipid metabolism in macrophages [48,115].
In the context of atherosclerosis, macrophage dysfunction is a central event in plaque formation. Macrophages are the predominant immune cell type within atherosclerotic plaques, where they are responsible for lipid uptake, foam cell formation, and the propagation of local inflammation. Macrophages internalize modified low-density lipoproteins (LDLs) and, through the accumulation of lipid droplets (LDs), transform into foam cells [116,117]. The mechanisms underlying lipid accumulation in macrophages involve a combination of altered lipophagy, dysfunction of chaperone-mediated autophagy (CMA), changes in lipid-metabolizing enzymes, and disruptions in lipid uptake and efflux pathways [118].
The autophagy-lysosome-mediated degradation system, including CMA, plays a key role in regulating lipid handling in macrophages. When CMA is disrupted, macrophages fail to properly degrade lipids, resulting in excessive lipid accumulation and foam cell formation, which exacerbates plaque development [119,120]. Recent studies have highlighted the importance of CMA dysfunction in atherosclerosis, with reduced expression of LAMP-2A, a critical component of CMA, linked to lipid accumulation and alterations in lipid-regulating enzymes [49,51].

3.3. CMA Plays an Important Role in Lipid Accumulation in Macrophages

Recent studies have established CMA as a crucial regulator of lipid catabolism, primarily by degrading lipid droplet-associated proteins and enzymes involved in lipid metabolism [121,122]. In the absence of CMA, lipid accumulation is observed in various cell types, including hepatocytes in vitro and in mouse liver [121,122]. In macrophages, lipid accumulation is a central event in the progression of atherosclerosis.
Using macrophage-specific L2AKO mice and primary peritoneal macrophages from these mice, a 2020 study confirmed that CMA deficiency leads to lipid accumulation in macrophages. This is likely due to the disruption of lipid metabolism-related enzymes. In advanced atherosclerotic lesions, the co-localization of LC3 with lipid droplets (LDs) was reduced, indicating impaired lipid autophagy [51]. Additionally, CMA dysfunction may contribute to lipid accumulation through altered expression of lipid-binding and transport proteins such as scavenger receptor type A (SR-A) and SR-B (CD36), as well as increased expression of long-chain-fatty-acid-CoA ligase 1 (ACSL1), a key enzyme involved in lipid synthesis [51,123]. Concurrently, enzymes responsible for lipid breakdown, such as lysosomal acid lipase (LAL), are upregulated, suggesting that CMA deficiency results in enhanced lipid synthesis and impaired lipid catabolism, further driving lipid accumulation in macrophages [124,125,126]. This disruption of lipophagy in advanced atherosclerotic lesions accelerates lipid buildup and the progression of atherosclerosis [51].
Studies propose that future research should focus on investigating CMA’s role in atherosclerosis using genetic interventions, such as blocking LAMP-2A [29], and the development of pharmacological agents that target LAMP-2A to promote lipid metabolism and alleviate atherosclerosis progression.

3.4. PYCARD Modulates CMA-Mediated Lipid Regulation in Macrophages

Recent research has explored the role of PYCARD in regulating chaperone-mediated autophagy (CMA) and its impact on lipid accumulation in macrophages [127,128]. Using pycard knockout (pycard−/−) mice and 3T3-L1 cells, a study demonstrated that PYCARD promotes microRNA maturation by inhibiting CMA-mediated degradation of the argonaute RISC catalytic subunit 2 (AGO2) [129]. This process plays a key role in regulating neointimal formation following vascular injury, independent of inflammasome activity. The study showed that PYCARD deficiency inhibits miRNA maturation and prevents neointimal formation by promoting CMA-dependent degradation of AGO2, suggesting a novel mechanism through which PYCARD modulates lipid regulation [127,130]. These findings point to the possibility of using PYCARD as a therapeutic target to control lipid accumulation and reduce the progression of atherosclerosis and other vascular diseases.

3.5. p62-ATG5-Mediated Autophagy and CMA in Macrophages

Autophagy, a fundamental cellular process for degrading dysfunctional proteins and organelles, plays a critical role in maintaining cellular homeostasis and preventing the accumulation of toxic materials [131,132]. This process is particularly important in macrophages, which are central players in the pathogenesis of atherosclerosis [7,133]. The autophagy pathway includes both bulk autophagy and chaperone-mediated autophagy (CMA), with CMA specifically targeting damaged proteins for lysosomal degradation [134,135]. Disruption of these pathways in macrophages, particularly through deficiencies in key autophagy-related proteins, can exacerbate lipid accumulation and atherosclerotic plaque formation [120,136].
Studies using p62−/− mice and macrophage-specific ATG5-deficient (mΦATG5−/−) mice have revealed the importance of p62 in regulating both bulk autophagy and CMA in the context of atherosclerosis. p62 is an autophagic adaptor protein that sequesters polyubiquitinated proteins, directing them to the lysosome for degradation [137]. In macrophages exposed to lipid-rich environments, such as those present in atherosclerosis, p62 accumulates and co-localizes with polyubiquitinated proteins, forming inclusion bodies. These inclusion bodies are typically composed of insoluble protein aggregates and serve a protective role by preventing the cytotoxic effects of protein aggregation [138,139,140].
In macrophages deficient in ATG5, a key protein required for autophagosome formation, the accumulation of p62 and polyubiquitinated proteins is further enhanced. These macrophages exhibit larger cytoplasmic inclusion bodies, which are characterized by an increase in protein aggregation [141]. Notably, in atherosclerotic lesions, the presence of p62-rich inclusion bodies was observed in both mouse aortic plaques and human endarterectomy samples, suggesting that this phenomenon is a hallmark of atherosclerotic disease [138].
The protective role of p62 is critical for the clearance of damaged proteins and the prevention of macrophage dysfunction [142]. In the absence of p62, protein aggregation becomes more pronounced, leading to increased protein insolubility, macrophage apoptosis, and activation of the inflammasome, which in turn promotes the production of pro-inflammatory cytokines such as IL-1β [143]. This inflammatory cascade contributes to the enlargement and increased complexity of atherosclerotic plaques [138].
Given the central role of p62 in managing protein homeostasis through autophagy and its protective effects in macrophages, enhancing p62 function or its mediated clearance pathway presents a promising therapeutic strategy [96]. By boosting the autophagic degradation of toxic protein aggregates and reducing inflammation, such strategies could help mitigate lipid accumulation, plaque instability, and the progression of atherosclerosis.

4. Autophagy-Mediated Macrophage Pyroptosis in Atherosclerosis

Atherosclerosis is a complex, multifactorial disease involving various forms of programmed cell death, each playing a distinct role in plaque development and instability. Among these, macrophage pyroptosis has emerged as a key contributor to inflammation and plaque destabilization [144]. Autophagy, a cellular process that helps maintain homeostasis, has been shown to interact with pyroptosis, influencing macrophage survival and inflammatory responses in the context of atherosclerosis [145]. Studies have demonstrated that dysregulated autophagy can exacerbate inflammatory processes in atherosclerotic plaques, while controlled autophagy may protect against excessive pyroptotic cell death [146,147]. This section explores the mechanisms through which macrophage pyroptosis contributes to atherosclerosis progression, particularly in relation to autophagy and other forms of programmed cell death.

4.1. Macrophage Pyroptosis

Atherosclerosis is a chronic inflammatory disease characterized by the gradual accumulation of plaque within the walls of medium and large arteries. The development and rupture of plaques in atherosclerosis are associated with damage to vascular cells, including ECs, VSMCs, and macrophages. Autophagy is a subcellular process that influences the pathogenesis of atherosclerosis by regulating the inflammatory response and cell death pathways [148]. Pyroptosis is a newly recognized form of caspase-dependent regulated cell death, characterized by cell swelling, pore formation, and membrane rupture, leading to the massive release of cytoplasmic contents [149]. This mode of cell death is strongly associated with inflammation as it involves the release of pro-inflammatory cytokines, such as IL-1β and IL-18 [150]. In contrast, ferroptosis is a regulated form of non-apoptotic cell death involving iron-dependent lipid peroxidation [151,152]. Programmed cell death encompasses several forms [144,153], including apoptosis, autophagy, pyroptosis, and ferroptosis. In atherosclerosis, these forms of cell death can impact plaque development and stability.
There is increasing evidence that pyroptosis and ferroptosis interact with autophagy and contribute to the development of various diseases, such as cancer, degenerative encephalopathies, and CVD [77,154]. Autophagy can protect macrophages from oxidative stress and inflammatory damage, thereby maintaining cell survival. However, in certain instances, excessive or dysregulated autophagy can lead to cell death, a phenomenon known as “autophagy-dependent cell death” [155]. Although the role of autophagy in atherosclerosis has been extensively studied, the interactions between autophagy, pyroptosis, and ferroptosis, as well as their specific mechanisms in atherosclerosis, require further exploration.

4.2. Pathological Studies and Mechanisms of Macrophage Pyroptosis

Pathological studies have identified a significant presence of dead macrophages within vulnerable atherosclerotic plaques, underscoring the association between macrophage death and plaque instability [156]. Macrophage pyroptosis, a pro-inflammatory form of programmed cell death, is induced by activation of the NLRP3 inflammasome and cleavage of caspase-1, both of which are central in triggering pyroptosis [74,157,158]. The activation of caspase-1 leads to the formation of the N-terminal domain of gasdermin D (GSDMD), which then migrates to the plasma membrane to form pores and initiate pyroptosis [159,160,161]. Notably, caspase-1 deficiency has been shown to slow the progression of atherosclerotic plaques in ApoE−/− mice, underscoring its role in disease advancement [162,163]. ox-LDL has been implicated in aberrant activation of NLRP3, which, in turn, activates GSDMD and exacerbates atherosclerosis in both mouse models and humans [164].
Studies on natural antioxidants have highlighted potential interventions for pyroptosis-related pathways. In 2022, a study using ApoE−/− mice and human myelomonocytic THP-1 cells found that quercetin effectively activates nuclear factor erythroid 2-related factor 2 (NRF2) by competitively binding to the Arg483 site of kelch like ECH associated protein 1(KEAP1), thereby inhibiting macrophage pyroptosis and reducing oxidative stress levels [165]. These effects were observed at both the cellular and organismal levels, where quercetin administration in high-fat diet-fed ApoE−/− mice reduced atherosclerosis progression. On the cellular level, quercetin inhibited ox-LDL-induced pyroptosis in THP-1 macrophages by suppressing NLRP3 inflammasome activation and lowering reactive oxygen species (ROS) levels. Further insights were provided by studies using KEAP1 mutants in THP-1 cells, which revealed that quercetin’s anti-pyroptotic effects were specifically associated with the Arg483 residue of KEAP1, not Arg415 [165]. This study suggests that quercetin’s modulation of the KEAP1/NRF2 interaction could be a promising target for reducing pyroptosis in atherosclerosis.

4.3. Mechanistic Insights and Therapeutic Targets of Autophagy-Mediated Macrophage Pyroptosis

Recent studies provide compelling evidence that autophagy plays a pivotal role in regulating gasdermin E (GSDME)-mediated pyroptosis, a form of programmed cell death that contributes significantly to inflammation and plaque instability in atherosclerosis [70]. GSDME-mediated pyroptosis has been shown to be induced by a variety of pro-inflammatory stimuli, including oxidized low-density lipoprotein (ox-LDL) in macrophages, which is a key contributor to atherosclerosis [71]. In 2023, research using GSDME knockout ApoE-deficient mice revealed that the deletion of GSDME resulted in reduced atherosclerotic lesion size and lower inflammatory cytokines such as IL-1β, TNF, MCP-1, and IL-6 [70,71]. Single-cell transcriptomic analysis confirmed that macrophages are the primary cells expressing GSDME in atherosclerotic plaques. This study also demonstrated that ox-LDL triggers the expression of GSDME in macrophages, which activates pyroptosis and inflammatory signaling. Notably, the STAT3 transcription factor was found to regulate GSDME expression, highlighting a transcriptional mechanism through which GSDME-mediated pyroptosis is upregulated in atherosclerotic lesions [71]. These findings suggest that targeting GSDME-mediated pyroptosis could serve as a novel therapeutic strategy for reducing atherosclerotic plaque burden and inflammation.
Moreover, autophagic regulation of macrophage pyroptosis is also observed in the context of hyperhomocysteinemia (HHcy), a common metabolic disorder associated with atherosclerosis. In a 2023 study, caspase-1-deficient (Casp1−/−) mice and THP-1-derived macrophages demonstrated that homocysteine promotes macrophage pyroptosis through endoplasmic reticulum (ER) stress and calcium dysregulation, both of which are influenced by autophagic processes. This study showed that hyperhomocysteinemia exacerbates atherosclerosis by increasing the size of atherosclerotic plaques and enhancing inflammatory cytokine production, including IL-1β [166,167]. Interestingly, autophagic flux and ER-mitochondrial coupling were found to play critical roles in this process. When autophagy is disrupted, the accumulated ER stress and calcium imbalances contribute to the activation of the NLRP3 inflammasome, which triggers macrophage pyroptosis [166,168]. Thus, targeting autophagy to modulate ER stress and calcium homeostasis may represent a promising approach for preventing HHcy-induced atherosclerosis progression.
Autophagy-mediated macrophage pyroptosis further exacerbates plaque instability and necrotic core formation in advanced atherosclerotic lesions. Dead macrophages release cellular contents, cytokines, and proteases, which promote inflammation and destabilize plaques, increasing the risk of acute cardiovascular events [76,79]. Pyroptosis, specifically through the GSDMD pathway, plays a crucial role in this process. Initially associated with monocytes, pyroptosis, particularly through GSDMD, is now recognized as a key driver of atherosclerotic plaque instability. Ox-LDL, a critical lipid component in atherosclerosis, induces ROS production, which oxidizes GSDMD and activates the NLRP3 inflammasome [169]. Once activated, GSDMD forms pores in the macrophage membrane, initiating pyroptosis and further amplifying inflammation [170]. This suggests that targeting the GSDMD/NLRP3 pathway could be an effective strategy for preventing macrophage-mediated plaque destabilization.
Moreover, autophagy-mediated macrophage pyroptosis is a significant source of cell death in atherosclerosis. Studies show that inhibition of pyroptosis can reduce atherosclerotic progression [165]. Autophagic processes are essential for maintaining macrophage homeostasis and preventing excessive cell death, thereby limiting the release of inflammatory mediators that promote plaque instability. Inhibition of the NLRP3 inflammasome and pyroptosis has been shown to mitigate the harmful effects of oxidative stress in atherosclerosis, further underscoring the therapeutic potential of autophagic modulation in treating cardiovascular disease [166,171]. As autophagy-mediated regulation of macrophage pyroptosis is a critical determinant of atherosclerosis progression, with significant implications for plaque instability and inflammation. Targeting the pathways that modulate autophagic flux and inflammasome activation may offer new therapeutic strategies to mitigate the detrimental effects of macrophage pyroptosis in cardiovascular disease.

4.4. Potential and Risk of Pharmacological Manipulation of Autophagic Pathways

As autophagy is an essential cellular degradation process through which damaged organelles, proteins, and other intracellular debris are sequestered in autophagosomes and degraded by lysosomal enzymes, thereby maintaining cellular homeostasis, pharmacological modulation of the autophagy pathway has garnered attention for its potential therapeutic applications across various diseases in recent years. However, this approach also carries inherent risks that require careful consideration.
In the context of atherosclerosis, autophagy plays a crucial role in both the progression and resolution of the disease. Pharmacologically activating autophagy in macrophages, such as through the use of rapamycin, has been shown to promote the polarization of an anti-inflammatory phenotype, reduce vascular inflammation, and improve vascular remodeling [172]. Furthermore, autophagy plays a key role in lipid metabolism, influencing the formation and resolution of atherosclerotic plaques. For example, autophagic activation can reduce plaque progression by facilitating the removal of lipid droplets within macrophages. In patients with non-alcoholic fatty liver disease (NAFLD), autophagy activation has been demonstrated to alleviate hepatic lipid accumulation. Resveratrol, a small molecule drug, can activate the AMPK pathway and inhibit mTORC1 activity, thereby promoting autophagic flux and regulating autophagy-related gene expression via the PI3K/AKT signaling pathway [172].
Beyond lipid metabolism, autophagy is integral to cellular homeostasis, particularly in neurons. For instance, autophagic activation reduces the accumulation of α-synuclein in the brains of Parkinson’s disease models [173,174]. Additionally, the autophagy-inducing drug trehalose has demonstrated protective effects in animal models of neurodegenerative diseases, highlighting its potential for clinical application [175].
Autophagy also has a dual role in cancer therapy. On one hand, autophagy can promote cancer cell survival and resistance to chemotherapy. On the other hand, autophagy modulation, such as through rapamycin, may enhance the efficacy of cancer therapies. However, excessive autophagy activation can lead to cellular dysfunction, including disrupted energy metabolism and the induction of apoptotic pathways [176]. Dysregulated autophagy may also contribute to lipotoxicity, thereby increasing oxidative stress within cells. In certain contexts, excessive autophagy can exacerbate disease progression, such as in tumors where autophagy supports growth and drug resistance.
While the pharmacological manipulation of autophagy pathways holds considerable therapeutic promise, it is not without its risks. Overactivation of autophagy can impair cellular function, leading to metabolic disturbances and heightened oxidative stress. Conversely, inhibition of autophagy may result in an exacerbated inflammatory response, potentially aggravating disease progression. For instance, chronic administration of rapamycin, although effective in inducing autophagy, has been associated with immunosuppression and metabolic disorders, underscoring the need for caution in its long-term use [177,178].
In summary, while pharmacological regulation of the autophagy pathway holds significant promise for the treatment of various diseases (Table 2), it is essential to evaluate the risks and fine-tune therapeutic strategies to ensure efficacy and minimize adverse effects. Future research should focus on elucidating the precise mechanisms governing autophagy and developing more selective and safer pharmacological agents that can harness the full therapeutic potential of autophagy without undesirable side effects.

5. Macrophage Autophagy and Clonal Hematopoiesis in Atherosclerosis

In adult mammals, circulating macrophages, derived from the monocyte-macrophage lineage, are generated from four primary sources: (1) the yolk sac at embryonic day 7.0 (E7.0), (2) the embryonic liver at E9.5, (3) the dorsal aorta, and (4) the bone marrow. These sources give rise to pre-macrophages/macrophages, monocytes, perivascular stromal cells (PDGFRA+), and hematopoietic stem cells (HSCs) through distinct waves of hematopoiesis [191,192]. Upon migration, circulating macrophages can localize to various tissues, including the heart, where atrioventricular (AV) node-resident macrophages directly influence cardiac function, as characterized by electrocardiogram (EKG) abnormalities [193,194,195]. Genetic defects in macrophage function can lead to severe cardiac dysfunction, particularly by disrupting the integrity of the AV node and its associated structures, such as the plane of insulation (POI) adjacent to the central fibrous body (CFB) [194,195]. It is plausible that circulating macrophages with impaired autophagic function localize to the aortic wall, where they play a critical role in the pathogenesis of atherosclerosis.
Clonal hematopoiesis (CH), driven by somatic mutations in hematopoietic stem cells (HSCs), is increasingly recognized as a significant risk factor for atherosclerosis. Mutations in genes such as TET2, JAK2, and DNMT3A promote the expansion of mutant clones in the blood, leading to inflammation, macrophage dysfunction, and plaque instability [196,197]. These mutations are particularly associated with clonal hematopoiesis of indeterminate potential (CHIP), a condition that accelerates atherosclerosis through the activation of inflammasomes and increased oxidative stress within macrophages [198]. While some studies suggest that CHIP mutations exacerbate atherosclerosis, others argue that CHIP may be a consequence of the chronic inflammatory state associated with atherosclerosis [199,200]. This section explores the genetic and inflammatory mechanisms through which CHIP mutations influence macrophage behavior and contribute to atherosclerotic plaque development.

5.1. Clonal Hematopoiesis as a Risk Factor for Atherosclerosis

Clonal hematopoiesis of indeterminate potential (CHIP) is a newly recognized, age-related risk factor for atherosclerosis, driven by somatic mutations in specific leukemia-related driver genes along with other age-related diseases including cardiac diseases [201]. These mutations lead to the expansion of mutant cell clones in peripheral blood. CH occurs when HSCs acquire genetic mutations and differentiate through polylineage hematopoiesis to produce terminally differentiated blood cells carrying clonal markers [202,203]. Common mutations associated with CHIP typically involve one of four genes—TET2, additional sex combs like-1 (ASXL1), DNMT3A or JAK2—which promote the clonal expansion of hematopoietic cells [89,187]. Although CH itself is not a malignancy, its presence significantly increases the risk of hematologic cancers. However, this alone does not explain the elevated mortality seen in individuals with CHIP [204]. The relationship between CHIP and atherosclerotic CVD has been explored in various studies. Some research suggests that certain CHIP mutations may directly contribute to atherosclerosis by exacerbating the inflammatory response, while other studies propose that CHIP may be a consequence of atherosclerosis, with the disease accelerating the expansion of mutant clones [18,96,196,205]. Furthermore, mutations commonly observed in CHIP, such as DNMT3A, TET2, ASXL1 and JAK2 [19,206] plus described above, have been linked to arterial disease, such as tubular artery disease.

5.2. Mechanisms Linking Clonal Hematopoiesis to Atherosclerosis

Recent studies have elucidated key mechanisms through which clonal hematopoiesis (CH) mutations contribute to atherosclerosis. A study by Fidler et al. demonstrated that mutations in JAK2, specifically the JAK2V617F mutation, promoted macrophage proliferation and necrotic core formation in atherosclerotic plaques in mutant mice. This process was associated with the activation of the AIM2 inflammasome, which led to an increase in IL-1β production and enhanced oxidative stress [187]. The study also highlighted the critical role of macrophage metabolic reprogramming in CH, with JAK2 mutations driving the accumulation of reactive oxygen species (ROS) and oxidized DNA [187]. Furthermore, the administration of the IL-1 receptor antagonist anakinra was shown to reduce plaque instability by normalizing macrophage proliferation, reducing necrotic core size, and stabilizing the fibrous cap [207,208,209]. These findings underscore the important connection between JAK2 mutations, inflammasome activation, oxidative damage, and atherosclerosis, suggesting potential therapeutic avenues for managing CHIP-related cardiovascular risk.
In another pivotal study, Bick et al. employed whole-genome sequencing in a cohort of nearly 100,000 individuals to investigate the genetic mechanisms of CHIP. The researchers identified several CHIP-associated mutations in genes such as DNMT3A, TET2, ASXL1, JAK2, and PPM1D, which were found to be significantly associated with lipid metabolism, immune function, and inflammatory responses [202]. Notably, a variant of TET2 was identified as being more prevalent in individuals of African ancestry, suggesting that genetic diversity may influence the impact of CHIP on atherosclerosis risk [210,211]. These findings provide important insights into the genetic basis of CHIP and open potential avenues for future diagnostics and therapeutic interventions for CHIP-related cardiovascular disease.
Additionally, research by Fuster et al. investigated the role of TET2 mutations in both CHIP and atherosclerosis. The loss of TET2 in hematopoietic stem cells (HSCs) was shown to disrupt DNA methylation patterns, thereby influencing hematopoietic cell clonality [200] and promoting inflammatory processes that accelerate atherosclerosis. TET2 deficiency led to altered immune responses and increased plaque formation in mouse models, emphasizing the significant role of epigenetic changes in CHIP and its potential contribution to the pathogenesis of atherosclerosis [212,213]. These findings provide critical insights into how genetic mutations in hematopoietic cells influence atherosclerosis development, reinforcing the importance of clonal hematopoiesis as a significant risk factor for cardiovascular disease.

5.3. Confirmed Function of Circulating Macrophages in the Cardiovascular System

The heart is composed of a diverse array of cell types, predominantly cardiomyocytes, endothelial cells, fibroblasts, pericytes, smooth muscle cells, and various leukocytes, including macrophages. Traditionally, the immune system has not been closely associated with the heart, but recent studies have shown that immune cells, particularly macrophages, play a vital role in maintaining heart health [191,193,194,214,215].
Using gene network analysis and single-cell regulatory network inference (SCENIC), Sano et al. provided mechanistic evidence that mosaic loss of the Y-chromosome (mLOY) in leukocytes can impact cardiac function [191]. Hulsmans et al. further demonstrated that hematopoietic macrophages can localize to the heart, particularly in the atrioventricular (AV) node, where they bind to conductive cardiomyocytes. These resident macrophages are essential for regulating the heart’s electrophysiological activity through the gap-junction protein connexin 43 (Cx43) [195].
These findings confirm that hematopoietic macrophages contribute to arrhythmias commonly observed in patients with heart failure. Specifically, macrophages localized to the AV node disrupt electroconductive gap-junctions, leading to arrhythmias and cardiac fibrosis, as demonstrated in their mouse models [191,195] (Figure 2). In the context of ischemia/reperfusion injury, bone marrow-derived monocytes increase in response to myocardial stress, localizing to macrophage niches within the myocardium, where they aid in tissue repair. This suggests that cardiac resident macrophages could also be pivotal in the development and progression of atherosclerosis.

5.4. TET2, JAK2, and DNMT3A-Associated Autophagy Could Lead Clonal Hematopoiesis to Atherosclerosis

The relationship between clonal hematopoiesis (CH) and atherosclerosis is increasingly recognized as a critical factor in cardiovascular disease pathogenesis [216,217]. Mutations in key genes such as TET2, JAK2, and DNMT3A not only promote clonal expansion of hematopoietic stem cells (HSCs) but also alter the function of macrophages, a major cell type in atherosclerotic lesions [200,218]. These mutations impact autophagic pathways, which are essential for macrophage homeostasis, inflammation resolution, and lipid metabolism [90,219]. The role of autophagy in macrophages is particularly pertinent, as its dysfunction contributes to the inflammatory milieu within atherosclerotic plaques [127,219]. Among the various genetic alterations associated with CH, JAK2 mutations have emerged as a key modulator of both autophagy and inflammation, influencing macrophage behavior and plaque development [218]. This section explores the mechanisms by which JAK2 mutations disrupt autophagic regulation in macrophages and how this dysregulation accelerates atherosclerosis, setting the stage for understanding potential therapeutic interventions targeting JAK2 and autophagic pathways in atherosclerosis [71,166].

5.4.1. JAK2 and Autophagic Regulation in Atherosclerosis

Clonal hematopoiesis (CH), driven by mutations in genes such as JAK2, has been implicated in the pathogenesis of atherosclerosis, particularly by modulating macrophage function and autophagy. JAK2, a central regulator of immune and inflammatory responses, contributes to autophagic regulation in macrophages. Interleukin-6 (IL-6), a key pro-inflammatory cytokine elevated in atherosclerosis, activates JAK2 signaling and induces the phosphorylation of Beclin 1 (BECN1), a critical protein involved in the initiation of autophagy [218]. This phosphorylation enhances BECN1’s role in autophagy, helping macrophages survive under stress conditions and contributing to inflammation within atherosclerotic plaques. JAK2 activation in macrophages thus not only promotes autophagy but also influences the inflammatory environment, exacerbating macrophage dysfunction and disease progression [218]. This suggests that JAK2 could be a potential therapeutic target to modulate macrophage autophagy and inflammation in atherosclerosis.

5.4.2. TET2 Mutations and Impaired Autophagy in Atherosclerosis

TET2, a DNA demethylase, plays a critical role in the regulation of macrophage autophagy. Mutations in TET2, commonly found in CH, disrupt autophagic flux, especially in the hypoxic microenvironment of atherosclerotic lesions. Hypoxia-induced alterations in TET2 activity lead to changes in DNA methylation patterns, affecting the expression of genes crucial for autophagy. Loss of TET2 activity results in enhanced macrophage inflammation and reprogramming, which accelerates atherosclerotic lesion formation [90,218,220]. These findings highlight the importance of TET2 in maintaining macrophage function and autophagic homeostasis within the atherosclerotic context, suggesting that TET2 mutations may contribute to atherosclerosis by disrupting macrophage autophagic processes.

5.4.3. DNMT3A and Epigenetic Regulation of Autophagy in Atherosclerosis

Mutations in DNMT3A, a DNA methyltransferase, further complicate the epigenetic regulation of autophagy in macrophages. González-Rodríguez et al. demonstrated that DNMT3A-mediated DNA methylation at the MAP1LC3 locus—critical for initiating autophagy—leads to reduced autophagic flux [219]. In DNMT3A-mutated macrophages, this impairment exacerbates lipid retention and inflammatory responses, accelerating the progression of atherosclerosis. These findings underscore the role of DNMT3A in regulating autophagic processes that modulate macrophage function and inflammation in atherosclerotic disease. Therefore, DNMT3A mutations, similar to TET2 and JAK2, link epigenetic regulation of autophagy to macrophage-driven atherosclerosis [218,219].
In conclusion, mutations in TET2, JAK2, and DNMT3A not only drive clonal hematopoiesis but also modulate autophagic pathways that exacerbate the inflammatory microenvironment within atherosclerotic plaques. These genetic alterations contribute to macrophage dysfunction, lipid retention, and plaque instability, highlighting CH as a significant risk factor for cardiovascular disease. The dual role of CH in both the hematopoietic and vascular systems underscores its potential as a therapeutic target in atherosclerosis. Targeting the autophagic regulation modulated by TET2, JAK2, and DNMT3A mutations may offer new avenues for managing atherosclerotic cardiovascular disease.

5.5. The Role of the Monocyte-Macrophage System in Cardiovascular Health

In the adult mammalian heart, there are three distinct populations of macrophages, each originating from different waves of hematopoiesis. The first wave originates from embryonic yolk sac macrophages around embryonic day 7.0. The second wave arises from fetal liver monocyte progenitors at embryonic day 9.5. The third wave consists of the traditionally recognized hematopoietic monocyte-macrophage system [221,222,223,224,225] (Figure 2).
Recent studies have highlighted the role of macrophages derived from embryonic progenitors in both myocardial and vascular repair. These macrophages, localized in the myocardium and within the walls of large and medium-sized arterioles, are involved in cardiac function recovery under pathological conditions [226,227,228]. Autophagic dysfunction in these macrophages can significantly affect their role in cardiovascular diseases, including atherosclerosis.
A recent study utilizing long-term in vivo tracing of yolk sac-derived hematopoietic stem cells (HSCs) demonstrated that these cells migrate to the aorta-gonad-mesonephros (AGM) region, umbilical vessels, and other extraembryonic tissues during development. Notably, these progenitors retain their hematopoietic potential even in adult mice [229]. Samokhvalov et al. showed that yolk sac-derived cells expressing Runx1 at embryonic day 7.5 give rise to fetal lymphoid progenitors and adult HSCs [229].
In a similar vein, Yokomizo et al. used genetic tracing to demonstrate that fetal yolk sac-derived HSCs, expressing the liver-specific transcription factor hepatic leukemia factor (HLF), play a critical role in the post-gestational development of intra-arterial hematopoietic clusters, rather than contributing significantly to fetal hematopoiesis [230]. These findings have been validated in zebrafish models [228]. Further evidence of the importance of embryonic HSCs in adult hematopoiesis comes from studies by Wattrus et al., which showed that embryonic macrophages are essential for establishing proper adult hematopoiesis through quality control mechanisms [231].
In aging humans, mLOY not only affects cardiac function but also has been linked to carcinogenesis, as the progressive accumulation of LOY in cells correlates with aging [232,233]. Thompson et al. proposed that macrophage LOY serves as a biomarker of general genetic instability, consistent with mLOY across various cell types throughout the body [195,233]. These mutations, particularly in leukocytes including macrophages, may have functional consequences, contributing to age-related diseases such as cancer and cardiovascular disorders. Moreover, mutations in genes involved in autophagy pathways may strongly influence the development of atherosclerosis [201,234,235,236], highlighting the broader implications of autophagic dysregulation in cardiovascular diseases.

6. Conclusions and Perspective

Atherosclerosis is a multifaceted, chronic inflammatory disease in which various proteolytic pathways, such as macroautophagy, the ubiquitin–proteasome system (UPS), and chaperone-mediated autophagy (CMA), play pivotal roles. Among these, macroautophagy has been well-characterized in atherosclerosis, while the contribution of CMA remains less understood. CMA is a selective protein degradation process that targets over 45% of cytoplasmic proteins containing KFERQ-like motifs, and it is crucial for maintaining cellular proteostasis and lysosomal function. Dysregulation of CMA can influence macroautophagy and exacerbate atherosclerosis progression. Notably, CMA facilitates the degradation of pro-atherogenic proteins, such as NLRP3 inflammasomes, making it a promising target for therapeutic intervention in atherosclerosis. Despite this potential, further research is required to fully understand how CMA affects NLRP3 inflammasome activity and its broader role in atherosclerotic pathogenesis.
CMA dysfunction, particularly in macrophages and their associated clonal hematopoiesis, results in lipid accumulation, oxidative stress, and inflammatory cytokine release—key contributors to atherosclerosis. While the link between CMA deficiency and disrupted lipid metabolism in either circulating and CH developed/maturated macrophages is established, the precise mechanisms by which CMA regulates lipid accumulation and atherogenesis remain unclear. CMA also plays a vital role in lipid homeostasis by modulating lipid-regulating enzymes, thereby preventing foam cell formation and plaque instability. Emerging evidence suggests that both macroautophagy and CMA contribute to macrophage function and plaque stability, pointing to the potential for therapeutic strategies that target both pathways to mitigate atherosclerosis progression. These findings underscore the necessity of further research into the intricate regulation of lipid metabolism by CMA.
Another critical aspect of atherosclerosis is macrophage pyroptosis, a pro-inflammatory form of cell death that destabilizes plaques. Pyroptosis, mediated by proteins such as GSDMD, exacerbates atherosclerosis by impairing cholesterol transport, with GSDMD knockout models showing reduced lesion areas [237,238]. Additionally, GSDME, another regulator of inflammatory cell death, can switch apoptosis to pyroptosis depending on caspase-3 activity. GSDME’s role in regulating cell death pathways presents a promising therapeutic target for atherosclerosis. Building on recent advances, we hypothesize that CMA, in conjunction with other forms of autophagy, programmed cell death, and clonal hematopoiesis, forms a crucial regulatory axis that governs all-source macrophage function in atherosclerosis. This network plays a pivotal role in regulating macrophage homeostasis, inflammation, and lipid metabolism, all of which are central to the development and progression of atherosclerosis. Disruption of this axis—whether through CMA dysfunction or clonal hematopoiesis—can lead to excessive inflammation, impaired lipid handling, and plaque destabilization. This hypothesis suggests that targeting the key components of this axis may offer a powerful approach for mitigating atherosclerosis and preventing its complications. Understanding the intricate relationships between autophagy, cell death, and clonal hematopoiesis will be essential in designing novel therapeutic interventions for atherosclerotic cardiovascular disease.
In conclusion, the intricate interplay between CMA, lipid metabolism, and pyroptosis represents an untapped opportunity for innovative therapeutic interventions. Targeting these pathways, either individually or synergistically, offers a powerful strategy to slow or even reverse the progression of atherosclerosis, with the potential to significantly impact the management of atherosclerotic cardiovascular disease.

Author Contributions

Conceptualization, project administration, supervision, resources and funding acquisition, H.C., X.Z., S.-P.G. and X.X.; writing—original draft preparation, S.L., X.Z., S.N., Q.D. and X.X.; writing, review and editing, X.Z., P.L., Q.D., Y.F., Y.Z., X.X. and H.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (# 82200513 to XZ); the Sanqin Talent Innovation Team Program (#202405 to HLC), the Key Program of Shaanxi Provincial Science and Technology Department (#2022ZDLSF05-15 to HLC; 2022ZDLSF04-01 to SPG); the Shaanxi Association for Science and Technology Youth Talent Support Program (#20220317 to XZ); the Young Talent Fund of Xi’an Association for Science and Technology (#959202313083 to YZ); the Key Program of Shaanxi Provincial Education Department (#20JS138 to XZ); the College Student Innovation Training Program of Xi’an Medical University (#121524083/S202411840083 to S-KN); the Scientific and technological capabilities enhancement project from Xi’an Medical University (#205152353/208240383 to XZ); the Key Program of Xianyang Science and Technology Department (#L2024-ZDYF-ZDYF-SF-0054 to HLC), the Talent Program of Xi’an Medical University (#RCYJ202401 to HLC).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Publicly available datasets were analyzed in this study as described in Section.

Acknowledgments

We acknowledge the National Natural Science Foundation of China, the Shaanxi Association for Science and Technology Youth Talent Support Program, the Young Talent Fund of Xi’an Association for Science and Technology, the Key Program of Shaanxi Provincial Education Department, the College Student Innovation Training Program and the Scientific and technological capabilities enhancement project from Xi’an Medical University.

Conflicts of Interest

The authors declare no conflicts of interest. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

ACSL1: long-chain-fatty-acid-CoA ligase 1; AGO2: argonaute RISC catalytic subunit 2; AIM2: Absent In Melanoma 2; ASXL1: addi-tional sex combs like-1; ATG5: Autophagy Related 5; AV node/AVN: atrioventricular node; BECN1: Beclin 1; CD36: cluster of differentiation 36; CFB: central fibrous body; CH: clonal hematopoiesis; CHIP: Clonal Hematopoiesis of Indeterminate Potential; CMA: Chaperone-mediated autophagy; CVD: cardiovascular disease; DNMT3A: DNA methyltransferase 3 alpha; ECs: endothelial cells; EKG: electrocardiogram; ER: endoplasmic reticulum; GSDMD: gasdermin D; GSDME: gasdermin E; Hcy: homocysteine; HHcy: hyperhomocysteinemia; HSC70: heat-shock cognate protein 70; HSCs: hematopoietic stem cells; HSPA8: heat shock protein family A member 8; IL-18: interleukin-18; IL-1β: interleukin-1β; IL-6: Interleukin 6; JAK2: Janus kinase 2; KEAP1: kelch like ECH associated protein 1; KFERQ: Lys-Phe-Glu-Arg-Gln; KPNA4: Karyopherin Subunit Alpha 4; L2AKO: LAMP-2A–deficient mice; LAL: lysosomal acid lipase; LAMP-2: lysosomal associated membrane protein 2; LAMs: lipid-associated macrophages; LDLs: low-density lipoproteins; LDs: lipid droplets; MAM: mitochondria-associated ER membrane; MAP1LC3: Microtubule-associated pro-tein 1 light chain 3; MCP-1: Monocyte chemoattractant protein-1; Mir17: MicroRNA 17; NLRP3: NOD-, LRR- and pyrin domain-containing protein 3; NRF2: Nuclear factor erythroid 2-related factor 2; ox-LDL: oxidized low-density lipoprotein; POI: plane of insulation; PPM1D: Protein Phosphatase Mg2+/Mn2+ Dependent 1D; PRMT 8: protein arginine N-methyltransferase 8; PYCARD: PYD And CARD Domain Containing; ROS: reactive oxygen species; SR-A: scavenger receptor type A; SR-B: scavenger receptor class B; STAT3: Signal transducer and activator of transcription 3; TAMs: Tumor-associated macrophages; TERT: Telomerase Reverse Transcriptase; TET2: ten-eleven translocation-2; TNF: Tumor Necrosis Factor; TRIM59: Tripartite Motif Containing 59; UPS: ubiquitin-proteasome system; VSMCs: vascular smooth muscle cells; WD: Western-type diet.

References

  1. Shao, B.Z.; Han, B.Z.; Zeng, Y.X.; Su, D.F.; Liu, C. The roles of macrophage autophagy in atherosclerosis. Acta Pharmacol. 2016, 37, 150–156. [Google Scholar] [CrossRef] [PubMed]
  2. Pan, W.; Zhang, J.; Zhang, L.; Zhang, Y.; Song, Y.; Han, L.; Tan, M.; Yin, Y.; Yang, T.; Jiang, T.; et al. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif. 2024, 57, e13525. [Google Scholar] [CrossRef]
  3. Poznyak, A.V.; Nikiforov, N.G.; Wu, W.K.; Kirichenko, T.V.; Orekhov, A.N. Autophagy and Mitophagy as Essential Components of Atherosclerosis. Cells 2021, 10, 443. [Google Scholar] [CrossRef]
  4. Chen, S.; Saeed, A.F.U.H.; Liu, Q.; Jiang, Q.; Xu, H.; Xiao, G.G.; Rao, L.; Duo, Y. Macrophages in immunoregulation and therapeutics. Signal. Transduct. Target. Ther. 2023, 8, 207. [Google Scholar] [CrossRef]
  5. Hou, P.; Fang, J.; Liu, Z.; Shi, Y.; Agostini, M.; Bernassola, F.; Bove, P.; Candi, E.; Rovella, V.; Sica, G.; et al. Macrophage polarization and metabolism in atherosclerosis. Cell. Death Dis. 2023, 14, 691. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, B.; Hang, S.; Xu, S.; Gao, Y.; Yu, W.; Zang, G.; Zhang, L.; Wang, Z. Macrophage polarisation and inflammatory mechanisms in atherosclerosis: Implications for prevention and treatment. Heliyon 2024, 10, e32073. [Google Scholar] [CrossRef]
  7. Wu, M.Y.; Lu, J.H. Autophagy and Macrophage Functions: Inflammatory Response and Phagocytosis. Cells 2019, 9, 70. [Google Scholar] [CrossRef] [PubMed]
  8. Moore, K.J.; Sheedy, F.J.; Fisher, E.A. Macrophages in atherosclerosis: A dynamic balance. Nat. Rev. Immunol. 2013, 13, 709–721. [Google Scholar] [CrossRef]
  9. Peled, M.; Fisher, E.A. Dynamic Aspects of Macrophage Polarization during Atherosclerosis Progression and Regression. Front. Immunol. 2014, 5, 579. [Google Scholar] [CrossRef]
  10. Ajoolabady, A.; Pratico, D.; Lin, L.; Mantzoros, C.S.; Bahijri, S.; Tuomilehto, J.; Ren, J. Inflammation in atherosclerosis: Pathophysiology and mechanisms. Cell Death Dis. 2024, 15, 817. [Google Scholar] [CrossRef]
  11. Nussenzweig, S.C.; Verma, S.; Finkel, T. The role of autophagy in vascular biology. Circ. Res. 2015, 116, 480–488. [Google Scholar] [CrossRef]
  12. Zhang, S.; Peng, X.; Yang, S.; Li, X.; Huang, M.; Wei, S.; Liu, J.; He, G.; Zheng, H.; Yang, L.; et al. The regulation, function, and role of lipophagy, a form of selective autophagy, in metabolic disorders. Cell Death Dis. 2022, 13, 132. [Google Scholar] [CrossRef] [PubMed]
  13. Narabayashi, K.; Ito, Y.; Eid, N.; Maemura, K.; Inoue, T.; Takeuchi, T.; Otsuki, Y.; Higuchi, K. Indomethacin suppresses LAMP-2 expression and induces lipophagy and lipoapoptosis in rat enterocytes via the ER stress pathway. J. Gastroenterol. 2015, 50, 541–554. [Google Scholar] [CrossRef]
  14. Wang, Z.; Gao, Z.; Zheng, Y.; Kou, J.; Song, D.; Yu, X.; Dong, B.; Chen, T.; Yang, Y.; Gao, X.; et al. Melatonin inhibits atherosclerosis progression via galectin-3 downregulation to enhance autophagy and inhibit inflammation. J. Pineal. Res. 2023, 74, e12855. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, J.; Li, C.; Lu, M.; Jiang, G.; Chen, S.; Li, H.; Lu, K. Pharmacological induction of autophagy reduces inflammation in macrophages by degrading immunoproteasome subunits. PLoS Biol. 2024, 22, e3002537. [Google Scholar] [CrossRef] [PubMed]
  16. Silvestre-Roig, C.; de Winther, M.P.; Weber, C.; Daemen, M.J.; Lutgens, E.; Soehnlein, O. Atherosclerotic plaque destabilization: Mechanisms, models, and therapeutic strategies. Circ. Res. 2014, 114, 214–226. [Google Scholar] [CrossRef]
  17. Xu, J.; Kitada, M.; Ogura, Y.; Koya, D. Relationship Between Autophagy and Metabolic Syndrome Characteristics in the Pathogenesis of Atherosclerosis. Front. Cell Dev. Biol. 2021, 9, 641852. [Google Scholar] [CrossRef]
  18. Zuriaga, M.A.; Fuster, J.J. Clonal hematopoiesis and atherosclerotic cardiovascular disease: A primer. Clin. Investig. Arterioscler. 2023, 35, 35–41. [Google Scholar] [CrossRef]
  19. Cobo, I.; Tanaka, T.; Glass, C.K.; Yeang, C. Clonal hematopoiesis driven by DNMT3A and TET2 mutations: Role in monocyte and macrophage biology and atherosclerotic cardiovascular disease. Curr. Opin. Hematol. 2022, 29, 1–7. [Google Scholar] [CrossRef]
  20. Liu, W.; Pircher, J.; Schuermans, A.; Ul Ain, Q.; Zhang, Z.; Honigberg, M.C.; Yalcinkaya, M.; Nakao, T.; Pournamadri, A.; Xiao, T.; et al. Jak2 V617F clonal hematopoiesis promotes arterial thrombosis via platelet activation and cross talk. Blood 2024, 143, 1539–1550. [Google Scholar] [CrossRef]
  21. Guo, H.; Wei, D.; Liu, R.; Zhang, C.; Jiang, S.; Wang, W.; Hu, H.; Shen, L.; Liang, X. A novel therapeutic strategy for atherosclerosis: Autophagy-dependent cholesterol efflux. J. Physiol. Biochem. 2022, 78, 557–572. [Google Scholar] [CrossRef]
  22. Amorós-Pérez, M.; Fuster, J.J. Clonal hematopoiesis driven by somatic mutations: A new player in atherosclerotic cardiovascular disease. Atherosclerosis 2020, 297, 120–126. [Google Scholar] [CrossRef]
  23. Tang, G.; Duan, F.; Li, W.; Wang, Y.; Zeng, C.; Hu, J.; Li, H.; Zhang, X.; Chen, Y.; Tan, H. Metformin inhibited Nod-like receptor protein 3 inflammasomes activation and suppressed diabetes-accelerated atherosclerosis in apoE−/− mice. Biomed Pharmacother. 2019, 119, 109410. [Google Scholar] [CrossRef]
  24. Gu, T.; Zhang, Z.; Liu, J.; Chen, L.; Tian, Y.; Xu, W.; Zeng, T.; Wu, W.; Lu, L. Chlorogenic Acid Alleviates LPS-Induced Inflammation and Oxidative Stress by Modulating CD36/AMPK/PGC-1α in RAW264.7 Macrophages. Int. J. Mol. Sci. 2023, 24, 13516. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, Y.; Qin, J.; Zhao, Q.; Yang, J.; Wei, X.; Huang, Y.; Xie, M.; Zhang, C.; Li, Y. Plaque-Targeted Rapamycin Spherical Nucleic Acids for Synergistic Atherosclerosis Treatment. Adv. Sci. 2022, 9, e2105875. [Google Scholar] [CrossRef]
  26. Grootaert, M.O.J.; Moulis, M.; Roth, L.; Martinet, W.; Vindis, C.; Bennett, M.R.; De Meyer, G.R.Y. Vascular smooth muscle cell death, autophagy and senescence in atherosclerosis. Cardiovasc. Res. 2018, 114, 622–634. [Google Scholar] [CrossRef] [PubMed]
  27. Li, X.; Zhu, R.; Jiang, H.; Yin, Q.; Gu, J.; Chen, J.; Ji, X.; Wu, X.; Fu, H.; Wang, H.; et al. Autophagy enhanced by curcumin ameliorates inflammation in atherogenesis via the TFEB-P300-BRD4 axis. Acta Pharm. Sin. B 2022, 12, 2280–2299. [Google Scholar] [CrossRef] [PubMed]
  28. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef]
  29. Qiao, L.; Ma, J.; Zhang, Z.; Sui, W.; Zhai, C.; Xu, D.; Wang, Z.; Lu, H.; Zhang, M.; Zhang, C.; et al. Deficient Chaperone-Mediated Autophagy Promotes Inflammation and Atherosclerosis. Circ. Res. 2021, 129, 1141–1157. [Google Scholar] [CrossRef]
  30. Yang, Q.; Wang, R.; Zhu, L. Chaperone-Mediated Autophagy. Adv. Exp. Med. Biol. 2019, 1206, 435–452. [Google Scholar] [CrossRef]
  31. Madrigal-Matute, J.; Cuervo, A.M.; Sluimer, J.C. Chaperone-mediated autophagy protects against atherosclerosis. Autophagy 2022, 18, 2505–2507. [Google Scholar] [CrossRef] [PubMed]
  32. Valdor, R.; Martinez-Vicente, M. The Role of Chaperone-Mediated Autophagy in Tissue Homeostasis and Disease Pathogenesis. Biomedicines 2024, 12, 257. [Google Scholar] [CrossRef] [PubMed]
  33. Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
  34. Qiao, L.; Hu, J.; Qiu, X.; Wang, C.; Peng, J.; Zhang, C.; Zhang, M.; Lu, H.; Chen, W. LAMP2A, LAMP2B and LAMP2C: Similar structures, divergent roles. Autophagy 2023, 19, 2837–2852. [Google Scholar] [CrossRef] [PubMed]
  35. Madrigal-Matute, J.; de Bruijn, J.; van Kuijk, K.; Riascos-Bernal, D.F.; Diaz, A.; Tasset, I.; Martín-Segura, A.; Gijbels, M.J.J.; Sander, B.; Kaushik, S.; et al. Protective role of chaperone-mediated autophagy against atherosclerosis. Proc. Natl. Acad. Sci. USA 2022, 119, e2121133119. [Google Scholar] [CrossRef]
  36. Grebe, A.; Hoss, F.; Latz, E. NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ. Res. 2018, 122, 1722–1740. [Google Scholar] [CrossRef]
  37. Endicott, S.J.; Boynton, D.N., Jr.; Beckmann, L.J.; Miller, R.A. Long-lived mice with reduced growth hormone signaling have a constitutive upregulation of hepatic chaperone-mediated autophagy. Autophagy 2021, 17, 612–625. [Google Scholar] [CrossRef]
  38. Arias, E.; Cuervo, A.M. Pros and Cons of Chaperone-Mediated Autophagy in Cancer Biology. Trends. Endocrinol. Metab. 2020, 31, 53–66. [Google Scholar] [CrossRef]
  39. Madrigal-Matute, J.; Scorrano, L.; Sadoshima, J. Leducq Network: Modulating Autophagy to Treat Cardiovascular Disease. Circ. Res. 2018, 123, 323–325. [Google Scholar] [CrossRef]
  40. Dong, S.; Aguirre-Hernandez, C.; Scrivo, A.; Eliscovich, C.; Arias, E.; Bravo-Cordero, J.J.; Cuervo, A.M. Monitoring spatiotemporal changes in chaperone-mediated autophagy in vivo. Nat. Commun. 2020, 11, 645. [Google Scholar] [CrossRef]
  41. Schneider, J.L.; Villarroya, J.; Diaz-Carretero, A.; Patel, B.; Urbanska, A.M.; Thi, M.M.; Villarroya, F.; Santambrogio, L.; Cuervo, A.M. Loss of hepatic chaperone-mediated autophagy accelerates proteostasis failure in aging. Aging Cell. 2015, 14, 249–264. [Google Scholar] [CrossRef] [PubMed]
  42. Moore, K.J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef]
  43. Wen, J.H.; Li, D.Y.; Liang, S.; Yang, C.; Tang, J.X.; Liu, H.F. Macrophage autophagy in macrophage polarization, chronic inflammation and organ fibrosis. Front. Immunol. 2022, 13, 946832. [Google Scholar] [CrossRef]
  44. Li, X.; Zhu, X.; Wei, Y. Autophagy in Atherosclerotic Plaque Cells: Targeting NLRP3 Inflammasome for Self-Rescue. Biomolecules 2022, 13, 15. [Google Scholar] [CrossRef] [PubMed]
  45. Florance, I.; Ramasubbu, S. Current Understanding on the Role of Lipids in Macrophages and Associated Diseases. Int. J. Mol. Sci. 2022, 24, 589. [Google Scholar] [CrossRef]
  46. Rodriguez-Navarro, J.A.; Kaushik, S.; Koga, H.; Dall’Armi, C.; Shui, G.; Wenk, M.R.; Di Paolo, G.; Cuervo, A.M. Inhibitory effect of dietary lipids on chaperone-mediated autophagy. Proc. Natl. Acad. Sci. USA 2012, 109, E705–E714. [Google Scholar] [CrossRef]
  47. Ye, G.; Gao, H.; Wang, Z.; Lin, Y.; Liao, X.; Zhang, H.; Chi, Y.; Zhu, H.; Dong, S. PPARα and PPARγ activation attenuates total free fatty acid and triglyceride accumulation in macrophages via the inhibition of Fatp1 expression. Cell Death Dis. 2019, 10, 39. [Google Scholar] [CrossRef]
  48. Vogel, A.; Brunner, J.S.; Hajto, A.; Sharif, O.; Schabbauer, G. Lipid scavenging macrophages and inflammation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2022, 1867, 159066. [Google Scholar] [CrossRef]
  49. Dib, L.; Koneva, L.A.; Edsfeldt, A.; Zurke, Y.X.; Sun, J.; Nitulescu, M.; Attar, M.; Lutgens, E.; Schmidt, S.; Lindholm, M.W.; et al. Lipid-associated macrophages transition to an inflammatory state in human atherosclerosis increasing the risk of cerebrovascular complications. Nat. Cardiovasc. Res. 2023, 2, 656–672. [Google Scholar] [CrossRef]
  50. Zhai, M.; Gong, S.; Luan, P.; Shi, Y.; Kou, W.; Zeng, Y.; Shi, J.; Yu, G.; Hou, J.; Yu, Q.; et al. Extracellular Traps from Activated Vascular Smooth Muscle Cells Drive the Progression of Atherosclerosis. Nat. Commun. 2022, 13, 7500. [Google Scholar] [CrossRef]
  51. Qiao, L.; Wang, H.F.; Xiang, L.; Ma, J.; Zhu, Q.; Xu, D.; Zheng, H.; Peng, J.Q.; Zhang, S.; Lu, H.X.; et al. Deficient Chaperone-Mediated Autophagy Promotes Lipid Accumulation in Macrophage. J. Cardiovasc. Transl. Res. 2021, 14, 661–669. [Google Scholar] [CrossRef]
  52. Sebastian, W.A.; Inoue, M.; Shimizu, N.; Sato, R.; Oguri, S.; Itonaga, T.; Kishimoto, S.; Shiraishi, H.; Hanada, T.; Ihara, K. Cardiac Manifestations of Human ACTA2 Variants Recapitulated in a Zebrafish Model. J. Hum. Genet. 2024, 69, 133–138. [Google Scholar] [CrossRef]
  53. Jackson, A.O.; Regine, M.A.; Subrata, C.; Long, S. Molecular Mechanisms and Genetic Regulation in Atherosclerosis. Int. J. Cardiol. Heart Vasc. 2018, 21, 36–44. [Google Scholar] [CrossRef]
  54. Madan, N.; Ghazi, A.R.; Kong, X.; Chen, E.S.; Shaw, C.A.; Edelstein, L.C. Functionalization of CD36 Cardiovascular Disease and Expression Associated Variants by Interdisciplinary High Throughput Analysis. PLoS Genet. 2019, 15, e1008287. [Google Scholar] [CrossRef]
  55. Wu, H.; Che, X.; Zheng, Q.; Wu, A.; Pan, K.; Shao, A.; Wu, Q.; Zhang, J.; Hong, Y. Caspases: A Molecular Switch Node in the Crosstalk between Autophagy and Apoptosis. Int. J. Biol. Sci. 2014, 10, 1072–1083. [Google Scholar] [CrossRef]
  56. Zhang, J.; Huang, J.; Gu, Y.; Xue, M.; Qian, F.; Wang, B.; Yang, W.; Yu, H.; Wang, Q.; Guo, X.; et al. Inflammation-Induced Inhibition of Chaperone-Mediated Autophagy Maintains the Immunosuppressive Function of Murine Mesenchymal Stromal Cells. Cell Mol. Immunol. 2021, 18, 1476–1488. [Google Scholar] [CrossRef] [PubMed]
  57. Tzeng, H.T.; Yang, J.L.; Tseng, Y.J.; Lee, C.H.; Chen, W.J.; Chyuan, I.T. Plasminogen Activator Inhibitor-1 Secretion by Autophagy Contributes to Melanoma Resistance to Chemotherapy through Tumor Microenvironment Modulation. Cancers 2021, 13, 1253. [Google Scholar] [CrossRef]
  58. Orenstein, S.J.; Cuervo, A.M. Chaperone-Mediated Autophagy: Molecular Mechanisms and Physiological Relevance. Semin. Cell Dev. Biol. 2010, 21, 719–726. [Google Scholar] [CrossRef] [PubMed]
  59. Sukhorukov, V.N.; Khotina, V.A.; Chegodaev, Y.S.; Ivanova, E.; Sobenin, I.A.; Orekhov, A.N. Lipid Metabolism in Macrophages: Focus on Atherosclerosis. Biomedicines 2020, 8, 262. [Google Scholar] [CrossRef]
  60. Hu, H.J.; Wang, X.H.; Zhang, T.Q.; Liu, Y.; Chen, Z.R.; Zhang, Z.Z.; Huang, H.; Tang, H.F.; Jiang, Z.S. PLK1 Promotes Cholesterol Efflux and Alleviates Atherosclerosis by Up-Regulating ABCA1 and ABCG1 Expression via the AMPK/PPARγ/LXRα Pathway. Biochim. Biophys. Acta Mol. Cell Biol. 2022, 1867, 159221. [Google Scholar] [CrossRef]
  61. Thevkar-Nagesh, P.; Habault, J.; Voisin, M.; Ruff, S.E.; Ha, S.; Ruoff, R.; Chen, X.; Rawal, S.; Zahr, T.; Szabo, G.; et al. Transcriptional Regulation of Acsl1 by CHREBP and NF-κB in Macrophages during Hyperglycemia and Inflammation. PLoS ONE 2022, 17, e0272986. [Google Scholar] [CrossRef]
  62. Zhou, Q.; Wang, Y.; Cheng, Y.; Zhou, J.; Liu, W.; Ma, X.; Tang, S.; Tang, S.; Tang, C. Ilexgenin A Inhibits Lipid Accumulation in Macrophages and Reduces the Progression of Atherosclerosis through PTPN2/ERK1/2/ABCA1 Signalling Pathway. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2024, 1869, 159533. [Google Scholar] [CrossRef]
  63. Cui, N.; Li, H.; Dun, Y.; Ripley-Gonzalez, J.W.; You, B.; Li, D.; Liu, Y.; Qiu, L.; Li, C.; Liu, S. Exercise Inhibits JNK Pathway Activation and Lipotoxicity via Macrophage Migration Inhibitory Factor in Nonalcoholic Fatty Liver Disease. Front. Endocrinol. 2022, 13, 961231. [Google Scholar] [CrossRef]
  64. Schlager, S.; Vujic, N.; Korbelius, M.; Duta-Mare, M.; Dorow, J.; Leopold, C.; Rainer, S.; Wegscheider, M.; Reicher, H.; Ceglarek, U.; et al. Lysosomal Lipid Hydrolysis Provides Substrates for Lipid Mediator Synthesis in Murine Macrophages. Oncotarget 2017, 8, 40037–40051. [Google Scholar] [CrossRef] [PubMed]
  65. Cho, K.Y.; Miyoshi, H.; Nakamura, A.; Greenberg, A.S.; Atsumi, T. Lipid Droplet Protein PLIN1 Regulates Inflammatory Polarity in Human Macrophages and Is Involved in Atherosclerotic Plaque Development by Promoting Stable Lipid Storage. J. Atheroscler. Thromb. 2023, 30, 170–181. [Google Scholar] [CrossRef] [PubMed]
  66. Tao, H.; Yancey, P.G.; Blakemore, J.L.; Zhang, Y.; Ding, L.; Jerome, W.G.; Brown, J.D.; Vickers, K.C.; Linton, M.F. Macrophage SR-BI Modulates Autophagy via VPS34 Complex and PPARα Transcription of Tfeb in Atherosclerosis. J. Clin. Investig. 2021, 131, e94229. [Google Scholar] [CrossRef]
  67. Li, M.; Wang, Z.; Wang, P.; Li, H.; Yang, L. TFEB: An Emerging Regulator in Lipid Homeostasis for Atherosclerosis. Front. Physiol. 2021, 12, 639920. [Google Scholar] [CrossRef]
  68. Pan, J.; Han, L.; Guo, J.; Wang, X.; Liu, D.; Tian, J.; Zhang, M.; An, F. AIM2 Accelerates the Atherosclerotic Plaque Progressions in ApoE-/- Mice. Biochem. Biophys. Res. Commun. 2018, 498, 487–494. [Google Scholar] [CrossRef]
  69. Wang, A.; Yue, K.; Zhong, W.; Zhang, G.; Zhang, X.; Wang, L. Targeted Delivery of Rapamycin and Inhibition of Platelet Adhesion with Multifunctional Peptide Nanoparticles for Atherosclerosis Treatment. J. Control Release 2024, 376, 753–765. [Google Scholar] [CrossRef]
  70. Hu, Y.; Liu, Y.; Zong, L.; Zhang, W.; Liu, R.; Xing, Q.; Liu, Z.; Yan, Q.; Li, W.; Lei, H.; et al. The Multifaceted Roles of GSDME-Mediated Pyroptosis in Cancer: Therapeutic Strategies and Persisting Obstacles. Cell Death Dis. 2023, 4, 836. [Google Scholar] [CrossRef]
  71. 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]
  72. Seimon, T.; Tabas, I. Mechanisms and Consequences of Macrophage Apoptosis in Atherosclerosis. J. Lipid Res. 2009, 50, S382–S387. [Google Scholar] [CrossRef]
  73. Kurdi, A.; De Meyer, G.R.; Martinet, W. Potential Therapeutic Effects of mTOR Inhibition in Atherosclerosis. Br. J. Clin. Pharmacol. 2016, 82, 1267–1279. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, S.; Chen, H.; Ni, H.; Dai, Q. Targeting HDAC6 Attenuates Nicotine-Induced Macrophage Pyroptosis via NF-κB/NLRP3 Pathway. Atherosclerosis 2020, 317, 1–9. [Google Scholar] [CrossRef] [PubMed]
  75. Fan, X.; Li, Q.; Wang, Y.; Zhang, D.M.; Zhou, J.; Chen, Q.; Sheng, L.; Passerini, A.G.; Sun, C. Non-Canonical NF-κB Contributes to Endothelial Pyroptosis and Atherogenesis Dependent on IRF-1. Transl. Res. 2023, 255, 1–13. [Google Scholar] [CrossRef]
  76. Xu, X.D.; Chen, J.X.; Zhu, L.; Xu, S.T.; Jiang, J.; Ren, K. The Emerging Role of Pyroptosis-Related Inflammasome Pathway in Atherosclerosis. Mol. Med. 2022, 28, 160. [Google Scholar] [CrossRef]
  77. Qian, Z.; Zhao, Y.; Wan, C.; Deng, Y.; Zhuang, Y.; Xu, Y.; Zhu, Y.; Lu, S.; Bao, Z. Pyroptosis in the Initiation and Progression of Atherosclerosis. Front. Pharmacol. 2021, 12, 652963. [Google Scholar] [CrossRef]
  78. Chen, Q.; Lv, J.; Yang, W.; Xu, B.; Wang, Z.; Yu, Z.; Wu, J.; Yang, Y.; Han, Y. Targeted Inhibition of STAT3 as a Potential Treatment Strategy for Atherosclerosis. Theranostics 2019, 9, 6424–6442. [Google Scholar] [CrossRef]
  79. Fan, X.; Han, J.; Zhong, L.; Zheng, W.; Shao, R.; Zhang, Y.; Shi, S.; Lin, S.; Huang, Z.; Huang, W.; et al. Macrophage-Derived GSDMD Plays an Essential Role in Atherosclerosis and Cross Talk Between Macrophages via the Mitochondria-STING-IRF3/NF-κB Axis. Arterioscler. Thromb. Vasc. Biol. 2024, 44, 1365–1378. [Google Scholar] [CrossRef]
  80. Sun, W.; Lu, H.; Dong, S.; Li, R.; Chu, Y.; Wang, N.; Zhao, Y.; Zhang, Y.; Wang, L.; Sun, L.; et al. Beclin1 Controls Caspase-4 Inflammasome Activation and Pyroptosis in Mouse Myocardial Reperfusion-Induced Microvascular Injury. Cell Commun. Signal. 2021, 19, 107. [Google Scholar] [CrossRef]
  81. Yang, G.; Zhang, Q.; Tan, J.; Xiong, Y.; Liang, Y.; Yan, J.; Gu, F.; Xu, Y. HMGB1 Induces Macrophage Pyroptosis in Chronic Endometritis. Int. Immunopharmacol. 2023, 123, 110706. [Google Scholar] [CrossRef]
  82. Peng, X.; Chen, H.; Li, Y.; Huang, D.; Huang, B.; Sun, D. Effects of NIX-Mediated Mitophagy on ox-LDL-Induced Macrophage Pyroptosis in Atherosclerosis. Cell Biol. Int. 2020, 44, 1481–1490. [Google Scholar] [CrossRef]
  83. Cao, Y.; Chen, X.; Pan, F.; Wang, M.; Zhuang, H.; Chen, J.; Lu, L.; Wang, L.; Wang, T. Xinmaikang-Mediated Mitophagy Attenuates Atherosclerosis via the PINK1/Parkin Signaling Pathway. Phytomedicine 2023, 119, 154955. [Google Scholar] [CrossRef]
  84. Joo, L.; Bradley, C.C.; Lin, S.H.; Scheet, P.A.; Nead, K.T. Causes of Clonal Hematopoiesis: A Review. Curr. Oncol. Rep. 2023, 25, 211–220. [Google Scholar] [CrossRef] [PubMed]
  85. Köhnke, T.; Majeti, R. Clonal Hematopoiesis: From Mechanisms to Clinical Intervention. Cancer Discov. 2021, 11, 2987–2997. [Google Scholar] [CrossRef] [PubMed]
  86. Páramo Fernández, J.A. Atherosclerosis and Clonal Hematopoiesis: A New Risk Factor. Clin. Investig. Arterioscler. 2018, 30, 133–136. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, W.; Yalcinkaya, M.; Maestre, I.F.; Olszewska, M.; Ampomah, P.B.; Heimlich, J.B.; Wang, R.; Vela, P.S.; Xiao, T.; Bick, A.G.; et al. Blockade of IL-6 Signaling Alleviates Atherosclerosis in Tet2-Deficient Clonal Hematopoiesis. Nat. Cardiovasc. Res. 2023, 2, 572–586. [Google Scholar] [CrossRef]
  88. Fidler, T.P.; Dunbar, A.; Kim, E.; Hardaway, B.; Pauli, J.; Xue, C.; Abramowicz, S.; Xiao, T.; O’Connor, K.; Sachs, N.; et al. Suppression of IL-1β Promotes Beneficial Accumulation of Fibroblast-Like Cells in Atherosclerotic Plaques in Clonal Hematopoiesis. Nat. Cardiovasc. Res. 2024, 3, 60–75. [Google Scholar] [CrossRef]
  89. Asada, S.; Kitamura, T. Clonal Hematopoiesis and Associated Diseases: A Review of Recent Findings. Cancer Sci. 2021, 112, 3962–3971. [Google Scholar] [CrossRef]
  90. Yalcinkaya, M.; Liu, W.; Thomas, L.A.; Olszewska, M.; Xiao, T.; Abramowicz, S.; Papapetrou, E.P.; Westerterp, M.; Wang, N.; Tabas, I.; et al. BRCC3-Mediated NLRP3 Deubiquitylation Promotes Inflammasome Activation and Atherosclerosis in Tet2 Clonal Hematopoiesis. Circulation 2023, 148, 1764–1777. [Google Scholar] [CrossRef]
  91. Abplanalp, W.T.; Mas-Peiro, S.; Cremer, S.; John, D.; Dimmeler, S.; Zeiher, A.M. Association of Clonal Hematopoiesis of Indeterminate Potential with Inflammatory Gene Expression in Patients with Severe Degenerative Aortic Valve Stenosis or Chronic Postischemic Heart Failure. JAMA Cardiol. 2020, 5, 1170–1175. [Google Scholar] [CrossRef] [PubMed]
  92. Polizio, A.H.; Park, E.; Walsh, K. Clonal Hematopoiesis: Connecting Aging and Inflammation in Atherosclerosis. Curr. Atheroscler. Rep. 2023, 25, 105–111. [Google Scholar] [CrossRef]
  93. Kiefer, K.C.; Cremer, S.; Pardali, E.; Assmus, B.; Abou-El-Ardat, K.; Kirschbaum, K.; Dorsheimer, L.; Rasper, T.; Berkowitsch, A.; Serve, H.; et al. Full Spectrum of Clonal Haematopoiesis-Driver Mutations in Chronic Heart Failure and Their Associations with Mortality. ESC. Heart. Fail. 2021, 8, 1873–1884. [Google Scholar] [CrossRef]
  94. Zekavat, S.M.; Viana-Huete, V.; Matesanz, N.; Jorshery, S.D.; Zuriaga, M.A.; Uddin, M.M.; Trinder, M.; Paruchuri, K.; Zorita, V.; Ferrer-Pérez, A.; et al. TP53-Mediated Clonal Hematopoiesis Confers Increased Risk for Incident Atherosclerotic Disease. Nat. Cardiovasc. Res. 2023, 2, 144–158. [Google Scholar] [CrossRef] [PubMed]
  95. Dutta, A.; Yang, Y.; Le, B.T.; Zhang, Y.; Abdel-Wahab, O.; Zang, C.; Mohi, G. U2af1 Is Required for Survival and Function of Hematopoietic Stem/Progenitor Cells. Leukemia 2021, 35, 2382–2398. [Google Scholar] [CrossRef]
  96. Díez-Díez, M.; Ramos-Neble, B.L.; de la Barrera, J.; Silla-Castro, J.C.; Quintas, A.; Vázquez, E.; Rey-Martín, M.A.; Izzi, B.; Sánchez-García, L.; García-Lunar, I.; et al. Unidirectional Association of Clonal Hematopoiesis with Atherosclerosis Development. Nat. Med. 2024, 30, 2857–2866. [Google Scholar] [CrossRef] [PubMed]
  97. Wynn, T.A.; Vannella, K.M. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 2016, 44, 450–462. [Google Scholar] [CrossRef]
  98. Lendeckel, U.; Venz, S.; Wolke, C. Macrophages: Shapes and functions. ChemTexts 2022, 8, 12. [Google Scholar] [CrossRef]
  99. Wei, Q.; Deng, Y.; Yang, Q.; Zhan, A.; Wang, L. The markers to delineate different phenotypes of macrophages related to metabolic disorders. Front. Immunol. 2023, 14, 1084636. [Google Scholar] [CrossRef]
  100. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  101. Hussell, T.; Bell, T.J. Alveolar macrophages: Plasticity in a tissue-specific context. Nat. Rev. Immunol. 2014, 14, 81–93. [Google Scholar] [CrossRef] [PubMed]
  102. Misharin, A.V.; Morales-Nebreda, L.; Reyfman, P.A.; Cuda, C.M.; Walter, J.M.; McQuattie-Pimentel, A.C.; Chen, C.I.; Anekalla, K.R.; Joshi, N.; Williams, K.J.N.; et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 2017, 214, 2387–2404. [Google Scholar] [CrossRef]
  103. Dong, T.; Hu, G.; Fan, Z.; Wang, H.; Gao, Y.; Wang, S.; Xu, H.; Yaffe, M.B.; Vander Heiden, M.G.; Lv, G.; et al. Activation of GPR3-β-arrestin2-PKM2 pathway in Kupffer cells stimulates glycolysis and inhibits obesity and liver pathogenesis. Nat. Commun. 2024, 15, 807. [Google Scholar] [CrossRef]
  104. Salter, M.W.; Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 2017, 23, 1018–1027. [Google Scholar] [CrossRef] [PubMed]
  105. Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef]
  106. Lapenna, A.; De Palma, M.; Lewis, C.E. Perivascular macrophages in health and disease. Nat. Rev. Immunol. 2018, 18, 689–702. [Google Scholar] [CrossRef] [PubMed]
  107. Chen, R.; Zhang, H.; Tang, B.; Luo, Y.; Yang, Y.; Zhong, X.; Chen, S.; Xu, X.; Huang, S.; Liu, C. Macrophages in cardiovascular diseases: Molecular mechanisms and therapeutic targets. Signal Transduct. Target Ther. 2024, 9, 130. [Google Scholar] [CrossRef]
  108. Yu, B.; Jia, S.; Chen, Y.; Guan, R.; Chen, S.; Tang, W.; Bao, T.; Tian, Z. CXCL4 deficiency limits M4 macrophage infiltration and attenuates hyperoxia-induced lung injury. Mol. Med. 2024, 30, 253. [Google Scholar] [CrossRef]
  109. Naito, Y.; Takagi, T.; Higashimura, Y. Heme oxygenase-1 and anti-inflammatory M2 macrophages. Arch. Biochem. Biophys. 2014, 564, 83–88. [Google Scholar] [CrossRef]
  110. Lin, A.; Miano, J.M.; Fisher, E.A.; Misra, A. Chronic inflammation and vascular cell plasticity in atherosclerosis. Nat. Cardiovasc. Res. 2024, 3, 1408–1423. [Google Scholar] [CrossRef]
  111. Li, Y.; Sun, Z.; Zhang, L.; Yan, J.; Shao, C.; Jing, L.; Li, L.; Wang, Z. Role of Macrophages in the Progression and Regression of Vascular Calcification. Front. Pharmacol. 2020, 11, 661. [Google Scholar] [CrossRef] [PubMed]
  112. Hong, H.; Tian, X.Y. The Role of Macrophages in Vascular Repair and Regeneration after Ischemic Injury. Int. J. Mol. Sci. 2020, 21, 6328. [Google Scholar] [CrossRef]
  113. Ouimet, M.; Ediriweera, H.; Afonso, M.S.; Ramkhelawon, B.; Singaravelu, R.; Liao, X.; Bandler, R.C.; Rahman, K.; Fisher, E.A.; Rayner, K.J.; et al. microRNA-33 Regulates Macrophage Autophagy in Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1058–1067. [Google Scholar] [CrossRef]
  114. Wu, J.; Bai, X.; Yan, L.; Baimanov, D.; Cong, Y.; Quan, P.; Cai, R.; Guan, Y.; Bu, W.; Lin, B.; et al. Selective regulation of macrophage lipid metabolism via nanomaterials’ surface chemistry. Nat. Commun. 2024, 15, 8349. [Google Scholar] [CrossRef]
  115. Yan, J.; Horng, T. Lipid Metabolism in Regulation of Macrophage Functions. Trends. Cell. Biol. 2020, 30, 979–989. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, B.; Tang, X.; Yao, L.; Wang, Y.; Chen, Z.; Li, M.; Wu, N.; Wu, D.; Dai, X.; Jiang, H.; et al. Disruption of USP9X in macrophages promotes foam cell formation and atherosclerosis. J. Clin. Investig. 2022, 132, e154217. [Google Scholar] [CrossRef] [PubMed]
  117. Canfrán-Duque, A.; Rotllan, N.; Zhang, X.; Andrés-Blasco, I.; Thompson, B.M.; Sun, J.; Price, N.L.; Fernández-Fuertes, M.; Fowler, J.W.; Gómez-Coronado, D.; et al. Macrophage-Derived 25-Hydroxycholesterol Promotes Vascular Inflammation, Atherogenesis, and Lesion Remodeling. Circulation 2022, 147, 388–408. [Google Scholar] [CrossRef]
  118. Filali-Mouncef, Y.; Hunter, C.; Roccio, F.; Zagkou, S.; Dupont, N.; Primard, C.; Proikas-Cezanne, T.; Reggiori, F. The ménage à trois of autophagy, lipid droplets and liver disease. Autophagy 2022, 18, 50–72. [Google Scholar] [CrossRef]
  119. Emanuel, R.; Sergin, I.; Bhattacharya, S.; Turner, J.; Epelman, S.; Settembre, C.; Diwan, A.; Ballabio, A.; Razani, B. Induction of lysosomal biogenesis in atherosclerotic macrophages can rescue lipid-induced lysosomal dysfunction and downstream sequelae. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1942–1952. [Google Scholar] [CrossRef]
  120. Sergin, I.; Evans, T.D.; Zhang, X.; Bhattacharya, S.; Stokes, C.J.; Song, E.; Ali, S.; Dehestani, B.; Holloway, K.B.; Micevych, P.S.; et al. Exploiting macrophage autophagy-lysosomal biogenesis as a therapy for atherosclerosis. Nat. Commun. 2017, 8, 15750. [Google Scholar] [CrossRef]
  121. Kaushik, S.; Cuervo, A.M. Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Nat. Cell. Biol. 2015, 17, 759–770. [Google Scholar] [CrossRef]
  122. Schneider, J.L.; Suh, Y.; Cuervo, A.M. Deficient chaperone-mediated autophagy in liver leads to metabolic dysregulation. Cell. Metab. 2014, 20, 417–432. [Google Scholar] [CrossRef] [PubMed]
  123. Al-Rashed, F.; Haddad, D.; Al Madhoun, A.; Sindhu, S.; Jacob, T.; Kochumon, S.; Obeid, L.M.; Al-Mulla, F.; Hannun, Y.A.; Ahmad, R. ACSL1 is a key regulator of inflammatory and macrophage foaming induced by short-term palmitate exposure or acute high-fat feeding. iScience 2023, 26, 107145. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, H. Lysosomal acid lipase and lipid metabolism: New mechanisms, new questions, and new therapies. Curr. Opin. Lipidol. 2018, 29, 218–223. [Google Scholar] [CrossRef]
  125. Vassiliou, E.; Farias-Pereira, R. Impact of Lipid Metabolism on Macrophage Polarization: Implications for Inflammation and Tumor Immunity. Int. J. Mol. Sci. 2023, 24, 12032. [Google Scholar] [CrossRef] [PubMed]
  126. Reiner, Ž.; Guardamagna, O.; Nair, D.; Soran, H.; Hovingh, K.; Bertolini, S.; Jones, S.; Ćorić, M.; Calandra, S.; Hamilton, J.; et al. Lysosomal acid lipase deficiency--an under-recognized cause of dyslipidaemia and liver dysfunction. Atherosclerosis 2014, 235, 21–30. [Google Scholar] [CrossRef]
  127. Li, J.; Yao, H.; Zhao, F.; An, J.; Wang, Q.; Mu, J.; Liu, Z.; Zou, M.H.; Xie, Z. Pycard deficiency inhibits microRNA maturation and prevents neointima formation by promoting chaperone-mediated autophagic degradation of AGO2/argonaute 2 in adipose tissue. Autophagy 2024, 20, 629–644. [Google Scholar] [CrossRef]
  128. Shen, W.; Wu, T.; Liu, Q.; Ke, B. Analysis of regulatory patterns of NLRP3 corpuscles and related genes and the role of macrophage polarization in atherosclerosis based on online database. Mol. Genet. Genom. 2024, 300, 7. [Google Scholar] [CrossRef]
  129. Elbarbary, R.A.; Maquat, L.E. Evaluating the susceptibility of AGO2-loaded microRNAs to degradation by nucleases in vitro. Methods 2019, 152, 18–22. [Google Scholar] [CrossRef]
  130. Malekmohammad, K.; Bezsonov, E.E.; Rafieian-Kopaei, M. Role of Lipid Accumulation and Inflammation in Atherosclerosis: Focus on Molecular and Cellular Mechanisms. Front. Cardiovasc. Med. 2021, 8, 707529. [Google Scholar] [CrossRef]
  131. Wang, D.W.; Peng, Z.J.; Ren, G.F.; Wang, G.X. The different roles of selective autophagic protein degradation in mammalian cells. Oncotarget 2015, 6, 37098–37116. [Google Scholar] [CrossRef] [PubMed]
  132. Hassanpour, M.; Rahbarghazi, R.; Nouri, M.; Aghamohammadzadeh, N.; Safaei, N.; Ahmadi, M. Role of autophagy in atherosclerosis: Foe or friend? J. Inflamm. 2019, 16, 8. [Google Scholar] [CrossRef]
  133. Sergin, I.; Razani, B. Self-eating in the plaque: What macrophage autophagy reveals about atherosclerosis. Trends. Endocrinol. Metab. 2014, 25, 225–234. [Google Scholar] [CrossRef] [PubMed]
  134. Wen, X.; Yang, Y.; Klionsky, D.J. Moments in autophagy and disease: Past and Present. Mol. Aspects. Med. 2021, 82, 100966. [Google Scholar] [CrossRef]
  135. Yao, R.; Shen, J. Chaperone-mediated autophagy: Molecular mechanisms, biological functions, and diseases. MedComm 2020, 4, e347. [Google Scholar] [CrossRef]
  136. Ni, D.; Mo, Z.; Yi, G. Recent insights into atherosclerotic plaque cell autophagy. Exp. Biol. Med. 2021, 246, 2553–2558. [Google Scholar] [CrossRef]
  137. Liu, W.J.; Ye, L.; Huang, W.F.; Guo, L.J.; Xu, Z.G.; Wu, H.L.; Yang, C.; Liu, H.F. p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell. Mol. Biol. Lett. 2016, 21, 29. [Google Scholar] [CrossRef]
  138. Sergin, I.; Bhattacharya, S.; Emanuel, R.; Esen, E.; Stokes, C.J.; Evans, T.D.; Arif, B.; Curci, J.A.; Razani, B. Inclusion bodies enriched for p62 and polyubiquitinated proteins in macrophages protect against atherosclerosis. Sci. Signal. 2016, 9, ra2. [Google Scholar] [CrossRef]
  139. Kim, S.; Lee, W.; Cho, K. P62 Links the Autophagy Pathway and the Ubiquitin-Proteasome System in Endothelial Cells during Atherosclerosis. Int. J. Mol. Sci. 2021, 22, 7791. [Google Scholar] [CrossRef]
  140. Huang, X.; Yao, J.; Liu, L.; Chen, J.; Mei, L.; Huang, F.J.; Luo, D.; Wang, X.; Lin, C.; Chen, X.; et al. S-Acylation of p62 promotes p62 droplet recruitment into autophagosomes in mammalian autophagy. Mol. Cell. 2023, 83, 3485–3501. [Google Scholar] [CrossRef]
  141. Shi, Y.X.; Sun, Z.W.; Jia, D.L.; Wang, H.B. Autophagy deficiency promotes lung metastasis of prostate cancer via stabilization of TWIST1. Clin. Transl. Oncol. 2022, 24, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  142. Hennig, P.; Fenini, G.; Di Filippo, M.; Karakaya, T.; Beer, H.D. The Pathways Underlying the Multiple Roles of p62 in Inflammation and Cancer. Biomedicines 2021, 9, 707. [Google Scholar] [CrossRef]
  143. Wang, L.; Cano, M.; Handa, J.T. p62 provides dual cytoprotection against oxidative stress in the retinal pigment epithelium. Biochim. Biophys. Acta 2014, 1843, 1248–1258. [Google Scholar] [CrossRef] [PubMed]
  144. De Meyer, G.R.Y.; Zurek, M.; Puylaert, P.; Martinet, W. Programmed death of macrophages in atherosclerosis: Mechanisms and therapeutic targets. Nat. Rev. Cardiol. 2024, 21, 312–325. [Google Scholar] [CrossRef] [PubMed]
  145. Martinet, W.; Coornaert, I.; Puylaert, P.; De Meyer, G.R.Y. Macrophage Death as a Pharmacological Target in Atherosclerosis. Front. Pharmacol. 2019, 10, 306. [Google Scholar] [CrossRef]
  146. Yang, R.; Wang, D.; Ding, Y.; Liu, Q. Exploring biomarkers for autophagy-mediated macrophage pyroptosis in atherosclerosis. Cell. Biol. Int. 2023, 47, 1905–1925. [Google Scholar] [CrossRef]
  147. He, X.; Fan, X.; Bai, B.; Lu, N.; Zhang, S.; Zhang, L. Pyroptosis is a critical immune-inflammatory response involved in atherosclerosis. Pharmacol. Res. 2021, 165, 105447. [Google Scholar] [CrossRef]
  148. Lin, L.; Zhang, M.X.; Zhang, L.; Zhang, D.; Li, C.; Li, Y.L. Autophagy, Pyroptosis, and Ferroptosis: New Regulatory Mechanisms for Atherosclerosis. Front. Cell. Dev. Biol. 2022, 9, 809955. [Google Scholar] [CrossRef]
  149. Liu, Y.; Pan, R.; Ouyang, Y.; Gu, W.; Xiao, T.; Yang, H.; Tang, L.; Wang, H.; Xiang, B.; Chen, P. Pyroptosis in health and disease: Mechanisms, regulation and clinical perspective. Signal. Transduct. Target. Ther. 2024, 9, 245. [Google Scholar] [CrossRef]
  150. Zheng, Z.; Li, G. Mechanisms and Therapeutic Regulation of Pyroptosis in Inflammatory Diseases and Cancer. Int. J. Mol. Sci. 2020, 21, 1456. [Google Scholar] [CrossRef]
  151. 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]
  152. 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]
  153. Liu, C.; Jiang, Z.; Pan, Z.; Yang, L. The Function, Regulation and Mechanism of Programmed Cell Death of Macrophages in Atherosclerosis. Front Cell Dev. Biol. 2022, 9, 809516. [Google Scholar] [CrossRef] [PubMed]
  154. Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal. Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef] [PubMed]
  155. Marchio, P.; Guerra-Ojeda, S.; Vila, J.M.; Aldasoro, M.; Victor, V.M.; Mauricio, M.D. Targeting Early Atherosclerosis: A Focus on Oxidative Stress and Inflammation. Oxid. Med. Cell. Longev. 2019, 2019, 8563845. [Google Scholar] [CrossRef] [PubMed]
  156. Jinnouchi, H.; Sato, Y.; Sakamoto, A.; Cornelissen, A.; Mori, M.; Kawakami, R.; Gadhoke, N.V.; Kolodgie, F.D.; Virmani, R.; Finn, A.V. Calcium deposition within coronary atherosclerotic lesion: Implications for plaque stability. Atherosclerosis 2020, 306, 85–95. [Google Scholar] [CrossRef]
  157. Mao, C.; Li, D.; Zhou, E.; Zhang, J.; Wang, C.; Xue, C. Nicotine exacerbates atherosclerosis through a macrophage-mediated endothelial injury pathway. Aging 2021, 13, 7627–7643. [Google Scholar] [CrossRef]
  158. Lin, J.; Shou, X.; Mao, X.; Dong, J.; Mohabeer, N.; Kushwaha, K.K.; Wang, L.; Su, Y.; Fang, H.; Li, D. Oxidized low density lipoprotein induced caspase-1 mediated pyroptotic cell death in macrophages: Implication in lesion instability? PLoS ONE 2013, 8, e62148. [Google Scholar] [CrossRef]
  159. 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.e20. [Google Scholar] [CrossRef]
  160. Coll, R.C.; Robertson, A.A.; Chae, J.J.; Higgins, S.C.; Muñoz-Planillo, R.; Inserra, M.C.; Vetter, I.; Dungan, L.S.; Monks, B.G.; Stutz, A.; et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat. Med. 2015, 21, 248–255. [Google Scholar] [CrossRef]
  161. Sharma, A.; Choi, J.S.Y.; Stefanovic, N.; Al-Sharea, A.; Simpson, D.S.; Mukhamedova, N.; Jandeleit-Dahm, K.; Murphy, A.J.; Sviridov, D.; Vince, J.E.; et al. Specific NLRP3 Inhibition Protects Against Diabetes-Associated Atherosclerosis. Diabetes 2021, 70, 772–787. [Google Scholar] [CrossRef]
  162. Yao, F.; Jin, Z.; Zheng, Z.; Lv, X.; Ren, L.; Yang, J.; Chen, D.; Wang, B.; Yang, W.; Chen, L.; et al. HDAC11 promotes both NLRP3/Caspase-1/GSDMD and caspase-3/GSDME pathways causing pyroptosis via ERG in vascular endothelial cells. Cell Death Discov. 2022, 8, 112. [Google Scholar] [CrossRef]
  163. Jin, Y.; Liu, Y.; Xu, L.; Xu, J.; Xiong, Y.; Peng, Y.; Ding, K.; Zheng, S.; Yang, N.; Zhang, Z.; et al. Novel role for caspase 1 inhibitor VX765 in suppressing NLRP3 inflammasome assembly and atherosclerosis via promoting mitophagy and efferocytosis. Cell Death Dis. 2022, 13, 512. [Google Scholar] [CrossRef] [PubMed]
  164. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  165. Luo, X.; Weng, X.; Bao, X.; Bai, X.; Lv, Y.; Zhang, S.; Chen, Y.; Zhao, C.; Zeng, M.; Huang, J.; et al. A novel anti-atherosclerotic mechanism of quercetin: Competitive binding to KEAP1 via Arg483 to inhibit macrophage pyroptosis. Redox. Biol. 2022, 57, 102511. [Google Scholar] [CrossRef]
  166. Zhang, S.; Lv, Y.; Luo, X.; Weng, X.; Qi, J.; Bai, X.; Zhao, C.; Zeng, M.; Bao, X.; Dai, X.; et al. Homocysteine promotes atherosclerosis through macrophage pyroptosis via endoplasmic reticulum stress and calcium disorder. Mol. Med. 2023, 29, 73. [Google Scholar] [CrossRef]
  167. Wang, R.; Wang, Y.; Mu, N.; Lou, X.; Li, W.; Chen, Y.; Fan, D.; Tan, H. Activation of NLRP3 inflammasomes contributes to hyperhomocysteinemia-aggravated inflammation and atherosclerosis in apoE-deficient mice. Lab. Investig. 2017, 97, 922–934. [Google Scholar] [CrossRef]
  168. Rocha, M.; Apostolova, N.; Diaz-Rua, R.; Muntane, J.; Victor, V.M. Mitochondria and T2D: Role of Autophagy, ER Stress, and Inflammasome. Trends Endocrinol. Metab. 2020, 31, 725–741. [Google Scholar] [CrossRef]
  169. Wang, Y.; Shi, P.; Chen, Q.; Huang, Z.; Zou, D.; Zhang, J.; Gao, X.; Lin, Z. Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J. Mol. Cell Biol. 2019, 11, 1069–1082. [Google Scholar] [CrossRef]
  170. Sun, S.; Gong, D.; Liu, R.; Wang, R.; Chen, D.; Yuan, T.; Wang, S.; Xing, C.; Lv, Y.; Du, G.; et al. Puerarin Inhibits NLRP3-Caspase-1-GSDMD-Mediated Pyroptosis via P2X7 Receptor in Cardiomyocytes and Macrophages. Int. J. Mol. Sci. 2023, 24, 13169. [Google Scholar] [CrossRef]
  171. Wu, P.; Chen, J.; Chen, J.; Tao, J.; Wu, S.; Xu, G.; Wang, Z.; Wei, D.; Yin, W. Trimethylamine N-oxide promotes apoE-/- mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. J. Cell. Physiol. 2020, 235, 6582–6591. [Google Scholar] [CrossRef]
  172. Chen, W.; Xiao, W.; Liu, X.; Yuan, P.; Zhang, S.; Wang, Y.; Wu, W. Pharmacological manipulation of macrophage autophagy effectively rejuvenates the regenerative potential of biodegrading vascular graft in aging body. Bioact. Mater. 2021, 11, 283–299. [Google Scholar] [CrossRef]
  173. Utpal, B.K.; Mokhfi, F.Z.; Zehravi, M.; Sweilam, S.H.; Gupta, J.K.; Kareemulla, S.; Darwin, C.R.; Rao, A.A.; Kumar, V.V.; Krosuri, P.; et al. Resveratrol: A Natural Compound Targeting the PI3K/Akt/mTOR Pathway in Neurological Diseases. Mol. Neurobiol. 2024. [Google Scholar] [CrossRef]
  174. Nixon, R.A.; Rubinsztein, D.C. Mechanisms of autophagy-lysosome dysfunction in neurodegenerative diseases. Nat. Rev. Mol. Cell. Biol. 2024, 25, 926–946. [Google Scholar] [CrossRef] [PubMed]
  175. Sevriev, B.; Dimitrova, S.; Kehayova, G.; Dragomanova, S. Trehalose: Neuroprotective Effects and Mechanisms-An Updated Review. NeuroSci 2024, 5, 429–444. [Google Scholar] [CrossRef] [PubMed]
  176. Kocot, A.M.; Wróblewska, B. Nutritional strategies for autophagy activation and health consequences of autophagy impairment. Nutrition 2022, 103, 111686. [Google Scholar] [CrossRef]
  177. Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and autophagy-related pathways in cancer. Nat. Rev. Mol. Cell. Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef]
  178. Zhao, B.; Zhang, J.; Zhao, K.; Wang, B.; Liu, J.; Wang, C.; Zeng, L.; Zeng, X.; Luo, Y. Effect of rapamycin on hepatic metabolomics of non-alcoholic fatty liver rats based on non-targeted platform. J. Pharm. Biomed. Anal. 2025, 253, 116541. [Google Scholar] [CrossRef]
  179. Johnston, R.; Uthman, O.; Cummins, E. Canagliflozin, Dapagliflozin and Empagliflozin Monotherapy for Treating Type 2 Diabetes: Systematic Review and Economic Evaluation. Health Technol. Assess 2017, 21, 1–218. [Google Scholar] [CrossRef]
  180. Sun, R.Z.; Fan, Y.; Liang, X.; Gong, T.T.; Wang, Q.; Liu, H.; Shan, Z.Y.; Lei, L. Rapamycin and FTY720 Alleviate Atherosclerosis by Cross Talk of Macrophage Polarization and Autophagy. Biomed. Res. Int. 2018, 2018, 1010248. [Google Scholar] [CrossRef]
  181. Li, J.; Li, C.; Huang, Z.; Huang, C.; Liu, J.; Wu, T.; Xu, S.; Mai, P.; Geng, D.; Zhou, S.; et al. Empagliflozin Alleviates Atherosclerotic Calcification by Inhibiting Osteogenic Differentiation of Vascular Smooth Muscle Cells. Front. Pharmacol. 2023, 14, 1295463. [Google Scholar] [CrossRef]
  182. Suda, M.; Tchkonia, T.; Kirkland, J.L.; Minamino, T. Targeting Senescent Cells for the Treatment of Age-Associated Diseases. J. Biochem. 2024, 177, 177–187. [Google Scholar] [CrossRef]
  183. Gao, K.; Liu, Y.; Sun, C.; Wang, Y.; Bao, H.; Liu, G.; Ou, J.; Sun, P. TNF-α Induces Mitochondrial Dysfunction to Drive NLRP3/Caspase-1/GSDMD-Mediated Pyroptosis in MCF-7 Cells. Sci. Rep. 2024, 14, 25880. [Google Scholar] [CrossRef]
  184. Traughber, C.A.; Timinski, K.; Prince, A.; Bhandari, N.; Neupane, K.; Khan, M.R.; Opoku, E.; Opoku, E.; Brubaker, G.; Shin, J.; et al. Disulfiram Reduces Atherosclerosis and Enhances Efferocytosis, Autophagy, and Athereprotective Gut Microbiota in Hyperlipidemic Mice. J. Am. Heart Assoc. 2024, 13, e033881. [Google Scholar] [CrossRef] [PubMed]
  185. Xiang, L.; Wang, Y.; Liu, S.; Ying, L.; Zhang, K.; Liang, N.; Li, H.; Luo, G.; Xiao, L. Quercetin Attenuates KLF4-Mediated Phenotypic Switch of VSMCs to Macrophage-Like Cells in Atherosclerosis: A Critical Role for the JAK2/STAT3 Pathway. Int. J. Mol. Sci. 2024, 25, 7755. [Google Scholar] [CrossRef] [PubMed]
  186. Dotan, I.; Yang, J.; Ikeda, J. Macrophage Jak2 Deficiency Accelerates Atherosclerosis through Defects in Cholesterol Efflux. Commun. Biol. 2022, 5, 132. [Google Scholar] [CrossRef]
  187. Fidler, T.P.; Xue, C.; Yalcinkaya, M.; Hardaway, B.; Abramowicz, S.; Xiao, T.; Liu, W.; Thomas, D.G.; Hajebrahimi, M.A.; Pircher, J.; et al. The AIM2 Inflammasome Exacerbates Atherosclerosis in Clonal Haematopoiesis. Nature 2021, 592, 296–301. [Google Scholar] [CrossRef]
  188. Cheung, B.M.; Lauder, I.J.; Lau, C.P.; Kumana, C.R. Meta-Analysis of Large Randomized Controlled Trials to Evaluate the Impact of Statins on Cardiovascular Outcomes. Br. J. Clin. Pharmacol. 2004, 57, 640–651. [Google Scholar] [CrossRef]
  189. Yuan, J.; Liu, C.; Jiang, C.; Liu, N.; Yang, Z.; Xing, H. RSL3 Induces Ferroptosis by Activating the NF-κB Signalling Pathway to Enhance the Chemosensitivity of Triple-Negative Breast Cancer Cells to Paclitaxel. Sci. Rep. 2025, 15, 1654. [Google Scholar] [CrossRef]
  190. Ouyang, S.; You, J.; Zhi, C.; Li, P.; Lin, X.; Tan, X.; Ma, W.; Li, L.; Xie, W. Ferroptosis: The Potential Value Target in Atherosclerosis. Cell Death Dis. 2021, 12, 782. [Google Scholar] [CrossRef]
  191. Xu, X.; Zhou, R.; Duan, Q.; Miao, Y.; Zhang, T.; Wang, M.; Jones, O.D.; Xu, M.M. Circulating macrophages as the mechanistic link between mosaic loss of Y-chromosome and cardiac disease. Cell Biosci. 2023, 13, 135. [Google Scholar] [CrossRef]
  192. Sano, S.; Horitani, K.; Ogawa, H.; Halvardson, J.; Chavkin, N.W.; Wang, Y.; Sano, M.; Mattisson, J.; Hata, A.; Danielsson, M.; et al. Hematopoietic loss of Y chromosome leads to cardiac fibrosis and heart failure mortality. Science 2022, 377, 292–297. [Google Scholar] [CrossRef] [PubMed]
  193. Hulsmans, M.; Sam, F.; Nahrendorf, M. Monocyte and macrophage contributions to cardiac remodeling. J. Mol. Cell. Cardiol. 2016, 93, 149–155. [Google Scholar] [CrossRef]
  194. Zhou, X.; Li, Z.; Wang, Z.; Chen, E.; Wang, J.; Chen, F.; Jones, O.D.; Tan, T.; Chen, S.; Takeshima, H.; et al. Syncytium calcium signaling and macrophage function in the heart. Cell. Biosci. 2018, 8, 24. [Google Scholar] [CrossRef]
  195. Hulsmans, M.; Clauss, S.; Xiao, L.; Aguirre, A.D.; King, K.R.; Hanley, A.; Hucker, W.J.; Wülfers, E.M.; Seemann, G.; Courties, G.; et al. Macrophages facilitate electrical conduction in the heart. Cell 2017, 169, 510–522. [Google Scholar] [CrossRef] [PubMed]
  196. Oren, O.; Small, A.M.; Libby, P. Clonal hematopoiesis and atherosclerosis. J. Clin. Investig. 2024, 134, e180066. [Google Scholar] [CrossRef]
  197. Jaiswal, S.; Natarajan, P.; Silver, A.J.; Gibson, C.J.; Bick, A.G.; Shvartz, E.; McConkey, M.; Gupta, N.; Gabriel, S.; Ardissino, D.; et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 111–121. [Google Scholar] [CrossRef]
  198. Marnell, C.S.; Bick, A.; Natarajan, P. Clonal hematopoiesis of indeterminate potential (CHIP): Linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J. Mol. Cell. Cardiol. 2021, 161, 98–105. [Google Scholar] [CrossRef]
  199. Fuster, J.J.; MacLauchlan, S.; Zuriaga, M.A.; Polackal, M.N.; Ostriker, A.C.; Chakraborty, R.; Wu, C.L.; Sano, S.; Muralidharan, S.; Rius, C.; et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017, 355, 842–847. [Google Scholar] [CrossRef]
  200. Bick, A.G.; Weinstock, J.S.; Nandakumar, S.K.; Fulco, C.P.; Bao, E.L.; Zekavat, S.M.; Szeto, M.D.; Liao, X.; Leventhal, M.J.; Nasser, J.; et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 2021, 586, 763–768. [Google Scholar] [CrossRef]
  201. Li, Z.; Duan, Q.; Cui, Y.; Jones, O.D.; Shao, D.; Zhang, J.; Gao, Y.; Cao, X.; Wang, S.; Li, J.; et al. Cardiac-Specific Expression of Cre Recombinase Leads to Age-Related Cardiac Dysfunction Associated with Tumor-like Growth of Atrial Cardiomyocyte and Ventricular Fibrosis and Ferroptosis. Int. J. Mol. Sci. 2023, 24, 3094. [Google Scholar] [CrossRef] [PubMed]
  202. Li, Z.; Cui, Y.; Duan, Q.; Zhang, J.; Shao, D.; Cao, X.; Gao, Y.; Wang, S.; Li, J.; Jones, O.D.; et al. The Prognostic Significance of FKBP1A and Its Related Immune Infiltration in Liver Hepatocellular Carcinoma. Int. J. Mol. Sci. 2022, 23, 12797. [Google Scholar] [CrossRef] [PubMed]
  203. Uddin, M.M.; Saadatagah, S.; Niroula, A.; Yu, B.; Hornsby, W.E.; Ganesh, S.; Lannery, K.; Schuermans, A.; Honigberg, M.C.; Bick, A.G.; et al. Long-term longitudinal analysis of 4,187 participants reveals insights into determinants of clonal hematopoiesis. Nat. Commun. 2024, 15, 7858. [Google Scholar] [CrossRef]
  204. Walsh, K. The emergence of clonal hematopoiesis as a disease determinant. J. Clin. Investig. 2024, 134, e180063. [Google Scholar] [CrossRef] [PubMed]
  205. Izzi, B.; Fuster, J.J. Clonal Hematopoiesis and Cardiovascular Risk: Atherosclerosis, Thrombosis, and beyond. Hamostaseologie 2024, 44, 13–20. [Google Scholar] [CrossRef]
  206. Kinzhebay, A.; Salybekov, A.A. The Role of Somatic Mutations in Ischemic Stroke: CHIP’s Impact on Vascular Health. Neurol. Int. 2025, 17, 19. [Google Scholar] [CrossRef]
  207. Bird, L. Taking AIM2 at atherosclerotic plaques. Nat. Rev. Immunol. 2021, 21, 273. [Google Scholar] [CrossRef]
  208. Sano, S.; Walsh, K. Hematopoietic JAK2V617F-mediated clonal hematopoiesis: AIM2 understand mechanisms of atherogenesis. J. Cardiovasc. Aging 2021, 1, 5. [Google Scholar] [CrossRef]
  209. Du, L.; Wang, X.; Chen, S.; Guo, X. The AIM2 inflammasome: A novel biomarker and target in cardiovascular disease. Pharmacol. Res. 2022, 186, 106533. [Google Scholar] [CrossRef]
  210. Jaiswal, S.; Libby, P. Clonal haematopoiesis: Connecting ageing and inflammation in cardiovascular disease. Nat. Rev. Cardiol. 2020, 17, 137–144. [Google Scholar] [CrossRef]
  211. Sano, S.; Oshima, K.; Wang, Y.; MacLauchlan, S.; Katanasaka, Y.; Sano, M.; Zuriaga, M.A.; Yoshiyama, M.; Goukassian, D.; Cooper, M.A.; et al. Tet2-Mediated Clonal Hematopoiesis Accelerates Heart Failure Through a Mechanism Involving the IL-1β/NLRP3 Inflammasome. J. Am. Coll. Cardiol. 2017, 71, 875–886. [Google Scholar] [CrossRef]
  212. Tall, A.R.; Levine, R.L. Cardiovascular disease: Commonality with cancer. Nature 2017, 543, 45–47. [Google Scholar] [CrossRef]
  213. Zhu, Y.P.; Hedrick, C.C.; Gaddis, D.E. Hematopoietic stem cells gone rogue. Science 2017, 355, 798–799. [Google Scholar] [CrossRef] [PubMed]
  214. Lafuse, W.P.; Wozniak, D.J.; Rajaram, M.V.S. Role of Cardiac Macrophages on Cardiac Inflammation, Fibrosis and Tissue Repair. Cells 2020, 10, 51. [Google Scholar] [CrossRef]
  215. Frangogiannis, N.G. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol. Aspects. Med. 2019, 65, 70–99. [Google Scholar] [CrossRef]
  216. Zeiher, A.; Braun, T. Mosaic loss of Y chromosome during aging. Science 2022, 377, 266–267. [Google Scholar] [CrossRef] [PubMed]
  217. Polizio, A.H.; Marino, L.; Min, K.D.; Yura, Y.; Rolauer, L.; Cochran, J.D.; Evans, M.A.; Park, E.; Doviak, H.; Miura-Yura, E.; et al. Experimental TET2 Clonal Hematopoiesis Predisposes to Renal Hypertension Through an Inflammasome-Mediated Mechanism. Circ. Res. 2024, 135, 933–950. [Google Scholar] [CrossRef]
  218. Hu, F.; Song, D.; Yan, Y.; Huang, C.; Shen, C.; Lan, J.; Chen, Y.; Liu, A.; Wu, Q.; Sun, L.; et al. IL-6 regulates autophagy and chemotherapy resistance by promoting BECN1 phosphorylation. Nat. Commun. 2021, 12, 3651. [Google Scholar] [CrossRef] [PubMed]
  219. González-Rodríguez, P.; Cheray, M.; Füllgrabe, J.; Salli, M.; Engskog-Vlachos, P.; Keane, L.; Cunha, V.; Lupa, A.; Li, W.; Ma, Q.; et al. The DNA methyltransferase DNMT3A contributes to autophagy long-term memory. Autophagy 2021, 17, 1259–1277. [Google Scholar] [CrossRef]
  220. de la Calle-Fabregat, C.; Calafell-Segura, J.; Gardet, M.; Dunsmore, G.; Mulder, K.; Ciudad, L.; Silvin, A.; Moreno-Càceres, J.; Corbí, Á.L.; Muñoz-Pinedo, C.; et al. NF-κB and TET2 promote macrophage reprogramming in hypoxia that overrides the immunosuppressive effects of the tumor microenvironment. Sci. Adv. 2024, 10, 5226. [Google Scholar] [CrossRef]
  221. Epelman, S.; Lavine, K.J.; Beaudin, A.E.; Sojka, D.K.; Carrero, J.A.; Calderon, B.; Brija, T.; Gautier, E.L.; Ivanovm, S.; Satpathy, A.T.; et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 2014, 40, 91–104. [Google Scholar] [CrossRef] [PubMed]
  222. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and functions of tissue macrophages. Immunity 2014, 41, 21–35. [Google Scholar] [CrossRef]
  223. Svedberg, F.R.; Guilliams, M. Cellular origin of human cardiac macrophage populations. Nat. Med. 2018, 24, 1091–1092. [Google Scholar] [CrossRef] [PubMed]
  224. Bajpai, G.; Schneider, C.; Wong, N.; Bredemeyer, A.; Hulsmans, M.; Nahrendorf, M.; Epelman, S.; Kreisel, D.; Liu, Y.; Itoh, A.; et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat. Med. 2018, 24, 1234–1245. [Google Scholar] [CrossRef] [PubMed]
  225. Delgado, M.G.; Lennon-Duménil, A.M. How cell migration helps immune sentinels. Front. Cell. Dev. Biol. 2022, 10, 932472. [Google Scholar] [CrossRef]
  226. Peet, C.; Ivetic, A.; Bromage, D.I.; Shah, A.M. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc. Res. 2020, 116, 1101–1112. [Google Scholar] [CrossRef]
  227. Deniset, J.F.; Belke, D.; Lee, W.Y.; Jorch, S.K.; Deppermann, C.; Hassanabad, A.F.; Turnbull, J.D.; Teng, G.; Rozich, I.; Hudspeth, K.; et al. Gata6+ Pericardial Cavity Macrophages Relocate to the Injured Heart and Prevent Cardiac Fibrosis. Immunity 2019, 51, 131–140. [Google Scholar] [CrossRef]
  228. Chandrakanthan, V.; Rorimpandey, P.; Zanini, F.; Chacon, D.; Olivier, J.; Joshi, S.; Kang, Y.C.; Knezevic, K.; Huang, Y.; Qiao, Q.; et al. Mesoderm-derived PDGFRA+ cells regulate the emergence of hematopoietic stem cells in the dorsal aorta. Nat. Cell. Biol. 2022, 24, 1211–1225. [Google Scholar] [CrossRef]
  229. Samokhvalov, I.M.; Samokhvalova, N.I.; Nishikawa, S. Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature 2007, 446, 1056–1061. [Google Scholar] [CrossRef]
  230. Yokomizo, T.; Ideue, T.; Morino-Koga, S.; Tham, C.Y.; Sato, T.; Takeda, N.; Kubota, Y.; Kurokawa, M.; Komatsu, N.; Ogawa, M.; et al. Independent origins of fetal liver haematopoietic stem and progenitor cells. Nature 2022, 609, 779–784. [Google Scholar] [CrossRef]
  231. Wattrus, S.J.; Smith, M.L.; Rodrigues, C.P.; Hagedorn, E.J.; Kim, J.W.; Budnik, B.; Zon, L.I. Quality assurance of hematopoietic stem cells by macrophages determines stem cell clonality. Science 2022, 377, 1413–1419. [Google Scholar] [CrossRef]
  232. Forsberg, L.A.; Rasi, C.; Malmqvist, N.; Davies, H.; Pasupulati, S.; Pakalapati, G.; Sandgren, J.; Diaz de Ståhl, T.; Zaghlool, A.; Giedraitis, V.; et al. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat. Genet. 2014, 46, 624–628. [Google Scholar] [CrossRef]
  233. Duan, Q.C.; Gao, Y.; Cao, X.; Wang, S.; Xu, M.M.; Jones, O.D.; Xu, X.H. Mosaic loss of chromosome Y in peripheral blood cells is associated with age-related macular degeneration in men. Cell. Biosci. 2022, 12, 73. [Google Scholar] [CrossRef]
  234. Zhang, Q.; Zhao, L.; Yang, Y.; Li, S.; Liu, Y.; Chen, C. Mosaic loss of chromosome Y promotes leukemogenesis and clonal hematopoiesis. JCI Insight. 2022, 7, e153768. [Google Scholar] [CrossRef] [PubMed]
  235. Yamamoto, H.; Zhang, S.; Mizushima, N. Autophagy genes in biology and disease. Nat. Rev. Genet. 2023, 24, 382–400. [Google Scholar] [CrossRef]
  236. Levine, B.; Kroemer, G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 2019, 176, 11–42. [Google Scholar] [CrossRef] [PubMed]
  237. Opoku, E.; Traughber, C.A.; Zhang, D.; Iacano, A.J.; Khan, M.; Han, J.; Smith, J.D.; Gulshan, K. Gasdermin D Mediates Inflammation-Induced Defects in Reverse Cholesterol Transport and Promotes Atherosclerosis. Front. Cell. Dev. Biol. 2021, 9, 715211. [Google Scholar] [CrossRef]
  238. Wang, Y.; Liu, X.; Shi, H.; Yu, Y.; Li, M.; Chen, R. NLRP3 inflammasome, an immune-inflammatory target in pathogenesis and treatment of cardiovascular diseases. Clin. Transl. Med. 2020, 10, 91–106. [Google Scholar] [CrossRef]
Figure 1. Signaling molecular pathways are altered from circumstance in a normal physiological artery (top) to that in an atherosclerotic artery (bottom) through chaperone-mediated autophagy, macrophage pyroptosis, and clonal hematopoiesis (middle) to atherosclerosis (AS). (A) Chaperone-mediated autophagy (CMA) is a selective autophagic process. (B) Dysfunction of autophagy affects macrophage lipid accumulation. (C) Autophagy mediates pyroptosis in macrophages. (D) Mutations in clonal hematopoiesis (CH) are the AS risk factors (figure not to scale).
Figure 1. Signaling molecular pathways are altered from circumstance in a normal physiological artery (top) to that in an atherosclerotic artery (bottom) through chaperone-mediated autophagy, macrophage pyroptosis, and clonal hematopoiesis (middle) to atherosclerosis (AS). (A) Chaperone-mediated autophagy (CMA) is a selective autophagic process. (B) Dysfunction of autophagy affects macrophage lipid accumulation. (C) Autophagy mediates pyroptosis in macrophages. (D) Mutations in clonal hematopoiesis (CH) are the AS risk factors (figure not to scale).
Ijms 26 03252 g001
Figure 2. Cellular and developmental mechanisms of macrophages in clonal hematopoiesis associated with atherosclerosis. This figure illustrates the various stem cell lineages involved in the development of macrophages during clonal hematopoiesis (CH) and their contributions to cardiovascular pathophysiology. Macrophages originating from these lineages play a significant role in cardiac fibrosis, heart failure, and overall cardiovascular function. These macrophages arise from hematopoietic stem cells (HSCs), which begin their development from yolk sac cells during the early embryonic stage [139]. Hematopoiesis continues through the embryonic liver and dorsal aorta, ultimately transitioning to bone marrow for sustained blood cell production. Pathological autophagy in these macrophages contributes to cardiovascular diseases, including cardiac fibrosis and coronary ischemia. In contrast, physiological autophagy, promoted by a healthy lifestyle, supports cardiac health and enhances cognitive function, potentially mitigating the effects of aging (figure not to scale).
Figure 2. Cellular and developmental mechanisms of macrophages in clonal hematopoiesis associated with atherosclerosis. This figure illustrates the various stem cell lineages involved in the development of macrophages during clonal hematopoiesis (CH) and their contributions to cardiovascular pathophysiology. Macrophages originating from these lineages play a significant role in cardiac fibrosis, heart failure, and overall cardiovascular function. These macrophages arise from hematopoietic stem cells (HSCs), which begin their development from yolk sac cells during the early embryonic stage [139]. Hematopoiesis continues through the embryonic liver and dorsal aorta, ultimately transitioning to bone marrow for sustained blood cell production. Pathological autophagy in these macrophages contributes to cardiovascular diseases, including cardiac fibrosis and coronary ischemia. In contrast, physiological autophagy, promoted by a healthy lifestyle, supports cardiac health and enhances cognitive function, potentially mitigating the effects of aging (figure not to scale).
Ijms 26 03252 g002
Table 1. Autophagy involved atherosclerosis through inhibition and promotion cellular pathways.
Table 1. Autophagy involved atherosclerosis through inhibition and promotion cellular pathways.
Autophagy Related Cellular ProcessesInvolved Autophagy Genes for Functional Atherosclerosis
Signal Molecules Related to PromotionSignal Molecules Related to Inhibition
Genes InvolvedRef.Genes InvolvedRef.
Chaperone-Mediated AutophagyCD68, ERK, HSC70, LAMP-2A, LAL[33,34,35,50]ACSL1, ACTA2, COL1A1-3, COX2, CD36, caspase-1, HMGB1, iNOS, IL-1β, IL-18, NLRP3, P53, PAI-1, SR-A[29,51,52,53,54,55,56,57,58]
Lipid Accumulation in MacrophageAkt, MAPK, mTOR, NF-κB, SR-A[56,59]AMPK, ACSL1, ABCA1, FABP4, JNK, LAL, PPAR, PLIN/ADRP, SR-B, TFEB[47,60,61,62,63,64,65,66,67]
Macrophage PyroptosisAIM2, caspase-1, DAMPs, GSDMD, GSDME, IL-1β, IL-18, mTOR, NLRP3, NF-κB, PAMPs, ROS, STAT3, SESN2, STING, TNF-α[68,69,70,71,72,73,74,75,76,77,78,79]beclin-1, HMGB1, LC3, PINK1/Parkin[80,81,82,83]
Clonal HematopoiesisASXL1, CBL, DNMT3A, cAMP, IDH2, IL-1β, IL-18, IL-6, JAK2, MCP-1, NLRP3, PPM1D, SF3B1, SRSF2, TET2, TP53, TNF-α, U2AF1[84,85,86,87,88,89,90,91,92,93,94,95]IL-10, IL-13, TGF-β[96]
Abbreviation: ABCA1: ATP-binding cassette transporter A1; ACSL1: acyl-CoA synthetase long chain family member 1; ACTA2: Actin Alpha 2; AIM2: Absent in Melanoma 2; Akt: Protein Kinase B; AMPK: Adenosine 5’-monophosphate(AMP)-activated protein kinase; ASXL1: Additional Sex Combs Like Transcriptional Regulator 1; beclin-1: B-cell lymphoma 2-interacting myosin-like coiled-coil protein 1; cAMP: cyclic adenosine monophosphate; caspase-1: Cysteine Aspartate-Specific Protease 1; CBL: Casitas B-lineage Lymphoma; CD36: CD36 molecule (thrombospondin receptor); CD68: Cluster of Differentiation 68; COL1A1-3: Collagen Type I Alpha 1 Chain, Collagen Type I Alpha 2 Chain, Collagen Type I Alpha 3 Chain; COX2: Cyclooxygenase-2; DAMPs: Damage-associated molecular patterns; DNMT3A: DNA methyltransferase 3 alpha; ERK: Extracellular signal-regulated kinase; FABP4: fatty acid binding protein 4; GSDMD: gasdermin D; GSDME: gasdermin E; HMGB1: High mobility group box 1 protein; HSC70: Heat shock cognate protein 70; IDH2: isocitrate dehydrogenase (NADP+)2; IL-10: interleukin 10; IL-13: interleukin 13; IL-18: interleukin-18; IL-1β: Interleukin-1β; IL-6:interleukin 6; iNOS: Inducible Nitric Oxide Synthase; JAK2: Janus kinase 2; JNK: c-Jun N-terminal Kinase; LAL: Limulus Amebocyte Lysate; LAMP-2A: Lysosomal-associated membrane protein 2A; LC3: Microtubule-Associated Protein 1 Light Chain 3; MAPK: Mitogen-Activated Protein Kinase; MCP-1: Monocyte chemoattractant protein-1; mTOR: Mammalian Target of Rapamycin; NF-kB: Nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3: NOD-like receptor pyrin domain containing 3; P53: Tumor protein p53; PAI-1: Plasminogen Activator Inhibitor-1; PAMPs: Pathogen-Associated Molecular Patterns; Parkin: Parkin RBR E3 ubiquitin-protein ligase; PINK1: PTEN induced putative kinase 1; PLIN/ADRP: Perilipin/Adipose differentiation-related protein; PPAR: Peroxisome Proliferators-Activated Receptors; PPM1D: Protein Phosphatase, Mg2+/Mn2+Dependent 1D; ROS: Reactive Oxygen Species; SESN2: Sestrin2; SF3B1: Spliceosome factor 3b subunit 1; SR-A: Scavenger receptor class A; SR-B: Scavenger Receptor Class B; SRSF2: Serine/Arginine-Rich Splicing Factor 2; STAT3: Signal Transducer and Activator of Transcription 3; STING: Stimulator of interferon genes; TET2: Ten-eleven translocation 2; TFEB: Trancription Factor EB; TGF-β: Transforming Growth Factor Beta; TNF-α: Tumor Necrosis Factor-alpha; U2AF1: U2 Small Nuclear RNA Auxiliary Factor 1.
Table 2. Current Treatments and Preclinical/Clinical Data for Atherosclerosis.
Table 2. Current Treatments and Preclinical/Clinical Data for Atherosclerosis.
PathwayDrug Targeting MoleculeAgent ProductBiological Process/Mechanism of ActionPreclinical/Clinical DataClass/PhaseReferences
Autophagy associated drugs/molecules against atherosclerosisLipid Accumulation in MacrophageSGLT2Enpagliflozin, DagliliflozinActivation of autophagy through the AMPK signaling pathway → clears intracellular lipid accumulation and damaged organelles, reducing inflammatory responsesImproved glycemic control in T2DM and reduces the risk of cardiovascular adverse cardiovascular eventsClass II[76,179]
Adiponectin receptor agonistsAdipoRonActivate AMPK → enhance macrophage autophagy →promote cholesterol effluxCholesterol efflux increased by 40%, autophagy marker LC3-II/LC3-I ratio increased by 3 timesN/A[62]
mTORRapamycinInhibit mTOR→activation of autophagy→promote the burial effectRapamycin significantly reduced the atherosclerotic lesions in the aorta in a high-fat diet-induced atherosclerosis mouse modelPhase II[69,180]
SGLT2EmpagliflozinActivation AMPK→inhibition of the mTORC1 activity→deregulate the inhibitory effect of mTOR on autophagyReduced mortality in patients with type 2 diabetes mellitus complicated with atherosclerotic cardiovascular diseasePhase III[33,181]
TBK1/IKKεBX795The TBK1/IKKε signaling pathway activates autophagy → clears damaged organelles and lipid accumulation within cells → reduces the production of inflammatory factorsTNF- α and IL-6 secretion were reduced by 70%, and autophagy-related genes (Atg 5) were upregulatedN/A[182]
Macrophage PyroptosisColchicineColchicine, low doseInhibition of NLRP3 inflammasome activation; it may interfere with autophagosome-lysosome fusionReduce the risk of cardiovascular events and improve coronary plaque stabilityClass II[74,183]
NLRP3TranilastInhibition of the NLRP3 protein →reduce in IL-1 β release; Increase the LC3-II/LC3-I ratio and reduce p62 accumulationSignificantly reduced the atherosclerotic plaque formation in the aortaPhase II[78]
Caspase-1VX-765Blocking caspase-1 downstream of the inflammasome → inhibition of the GSDMD-mediated pyroptosisThe plaque pyroptotic cells were decreased by 55%, and the plaque stability was enhancedN/A[67]
GasderminDisulfiramBlocking of the gasdermin D pore tract → inhibition of pyroptosisInhibition of pyroptosis reduced the plaque necrotic corePhase I[184]
GSDMEGasdermin E inhibitorBlocking of the GSDME shearing →inhibit pyrodead pore formationPlaque area decreased by 30%, fibrous cap thickness increased by 50%, and 60% decreased macrophage pyroptosis markers (caspase-3 activity)N/A[80,185]
Clonal HematopoiesisJAK2FedratinibInhibition of the expansion of the JAK 2 mutant clones → reduction of the proinflammatory macrophagesReduce IL-6, and TNF- α secretionN/A[185,186,187]
TET2N/AReduce the methylation level of the Beclin1 promoter region → promote the expression of Beclin1Increase the expression of autophagy marker LC3-II, while reducing the accumulation of autophagy disorder marker p62N/A[81]
Drugs or molecules of other pathways against atherosclerosisStatinsAtorvastatin, RosuvastatinInhibits HMG-CoA reductase → lowers LDL-CLDL-C decreased to <100 mg/dLClass I[188]
PCSK9Alirocumab, ElosulfaseBlocking of the PCSK 9→increasing the LDL receptor expression →reduce LDL-CLower plasma LDL-C levels by 50–60%Class I[189]
IL-1βCanakinumabNeutralization of IL-1 β→inhibition of the NLRP3 inflammasomeSignificant reduction in the hs-CRP levelsClass II[29]
Antiplatelet drugsAspirin, ClopidogrelInhibition of platelet aggregationCould significantly reduce the risk of stent thrombosis and cardiovascular eventsClass I[87,183]
GPX4RSL3Triggering of macrophage iron death by induction of lipid peroxidation →reduce intraplaque necrotic coreThe plaque necrotic core was reduced by 40%N/A[67,189]
NLRP3/TRPV2TranilastInhibition of the NLRP3 inflammasome blocks the TRPV 2 channels → synergy anti-inflammatorySpot collagen content increased by 25% and macrophage apoptosis decreased by 40%N/A[78]
CCR2RS504393Blocking CCR2 → inhibition of monocyte recruitment to the plaquesThe plaque macrophage number decreased by 45% and the overall plaque load decreased by 28%N/A[190]
PLK1BI-2536Inhibition of PLK1 → blocking of macrophage proliferation → reduce cell accumulation within the plaqueThe proliferating macrophages (Ki67 +) were reduced by 60% and plaque stability was improvedN/A[60]
Anti-aging drugsDasatinib, QuercetinReduced inflammatory factor release in senescent macrophagesElimination of senescent clonal hematopoietic cells → lighten SASPN/A[31]
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

Li, S.; Zhou, X.; Duan, Q.; Niu, S.; Li, P.; Feng, Y.; Zhang, Y.; Xu, X.; Gong, S.-P.; Cao, H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int. J. Mol. Sci. 2025, 26, 3252. https://doi.org/10.3390/ijms26073252

AMA Style

Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong S-P, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. International Journal of Molecular Sciences. 2025; 26(7):3252. https://doi.org/10.3390/ijms26073252

Chicago/Turabian Style

Li, Shuanhu, Xin Zhou, Qinchun Duan, Shukun Niu, Pengquan Li, Yihan Feng, Ye Zhang, Xuehong Xu, Shou-Ping Gong, and Huiling Cao. 2025. "Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis" International Journal of Molecular Sciences 26, no. 7: 3252. https://doi.org/10.3390/ijms26073252

APA Style

Li, S., Zhou, X., Duan, Q., Niu, S., Li, P., Feng, Y., Zhang, Y., Xu, X., Gong, S.-P., & Cao, H. (2025). Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. International Journal of Molecular Sciences, 26(7), 3252. https://doi.org/10.3390/ijms26073252

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