• Open Access
    Review

    Decoding vascular aging: implications for atherosclerosis progression and clinical intervention

    Silumbwe Ceaser Wankumbu *
    Xiao-Man Ji
    Ming Xu *

    Explor Drug Sci. 2024;2:449–472 DOI: https://doi.org/10.37349/eds.2024.00056

    Received: March 15, 2024 Accepted: May 31, 2024 Published: August 02, 2024

    Academic Editor: Santiago J. Ballaz, Yachay Tech University, Ecuador

    Abstract

    Age-related pathologies, particularly cardiovascular disorders, pose a significant global health concern. The World Health Organization (WHO) predicts an increase in advanced mortality by 2030 unless critical interventions are implemented. Atherosclerosis remains the major cause of various cardiovascular diseases. Hence, this review focused on the interaction between known mechanisms of vascular aging, disease manifestation, and progression during atherosclerosis. In the review, we highlighted five altered vascular mechanisms in cardiovascular models: genomic instability, neurohormonal deregulation, epigenetics, protein regulation, and the gut microbiome. The articles were selected from various indexed scientific databases. It is important to note that the mechanisms are equally interrelated with other aging pathways, such as inflammation and senescence. In conclusion, atherosclerosis is multifaceted and cholesterol-lowering therapy has been widely used. However, more than one specific action line is required to eradicate or slow down its manifestation. Equally, establishing a balance between aging stressors resulting in vascular injuries and stress buffering mechanisms during aging is critical to the treatment of atherosclerosis. The promising therapeutic targets reviewed include the angiotensin (1–7)/MAS axis, the gut microbiome, histone deacetylases, DNA repair systems, noncoding RNAs, β3/dopamine adrenoceptors, senescence and inflammation checkpoints.

    Keywords

    Atherosclerosis, inflammation, vascular mechanisms, cardiovascular diseases, therapeutic targets

    Introduction

    Cardiovascular diseases (CVDs) pose a significant global health problem, accounting for 29.6% of deaths in 2010, reaching 17.9 million in 2019, with the World Health Organization predicting an increase by 2030. Atherosclerosis remains the major cause of various CVDs [1, 2]. Despite links to fatty degeneration and vessel stiffening, with the low-density lipoprotein (LDL)/high-density lipoprotein (HDL) ratio being significant risk factors, atherosclerosis is a chronic inflammatory condition that primarily causes CVDs, as demonstrated by studies such as canakinumab anti-inflammatory thrombosis outcome study (CANTOS), colchicine cardiovascular outcome and rescue trials [25]. Morphological, cellular, genetic, and consequently, functional alterations occur during atherosclerosis and aging [6]. The arterial walls harden, accompanied by a reduced elasticity [7]. Atherosclerosis involves the formation of plaques comprising necrotic cores, calcified regions, reorganized lipids, inflamed vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and inflammatory factors [8]. Aged vessels show similar modifications as observed in atherosclerosis [9]. However, even in the absence of atherosclerosis, vessels can undergo vascular remodeling, leading to decreased elasticity [10]. These mechanisms that lead to the manifestation of the disease are currently englobed as hallmarks of aging. Various authors have defined aging mechanisms and their significant role in scientific research, particularly in disease and drug development [1113]. In this review, we focus on five vascular alterations (genomic instability, neurohormonal regulation, epigenetics, protein regulation, and gut microbiome) leading to atherosclerosis during aging. In addition, we also discuss how these mechanisms also induce other aging phenomena, such as inflammation and senescence, because these mechanisms are significantly interrelated [13, 14]. Furthermore, we highlight current and potential therapeutic interventions.

    Pathogenesis of atherosclerosis

    Preliminary atherogenesis involves the trapping of lipoproteins at the injury site, allowing LDL cholesterol to enter ECs through endocytosis, increasing the concentration of LDL in the intima. Passive movement of oxidized LDLs (ox-LDLs) in ECs is facilitated by compromised endothelial barriers, activin-like kinase receptor 1 (ALK1), and scavenger receptors (SRs) [15]. The next process is endothelial activation, which occurs due to glycocalyx dysfunction and reduced cell contact, leading to weak junctions, promoting the absorption, uptake, and oxidation of triglyceride-rich lipoproteins in subendothelial cells [16]. CD36 and lectin-like oxidized LDL receptor 1 (LOX-1) regulate the effects of ox-LDLs on ECs. CD36 is the main binding receptor. ox-LDL through CD36 inhibits macrophage migration, potentially causing atherosclerotic lesions. The C-reactive protein-CD36 interaction reinforces macrophage uptake of ox-LDL [17, 18]. ECs are activated by ox-LDL in subendothelial spaces, causing monocyte adhesion and rolling through the expression of chemoattractant proteins [vascular adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), P-selectin, monocyte chemoattractant protein 1 (MCP-1), and CC-chemokine receptor 2 (CCR2) and CCR5] [19, 20]. The CCR pairs involved in macrophage transmigration also include; CCR2-CCL2, CX3C CCR1, and CCR5-CCL5. In addition, T cell helper 1 (Th1) cells express the T-box transcription factor TBX21 (T-bet) and CCRs such as the CXCR3, CCR5, and IFNγ. In atherosclerotic ApoE-/- mice, Th1 cells express the atherosclerotic plaque-homing receptor CCR5 in lymph nodes [21]. Blocking of these axes in ApoE-/- mice reduces atherosclerosis progression by 90% [22]. CD4+ T cells in the atherosclerotic plaque express other pro-inflammatory cytokines associated with Th1 in addition to IFNγ, such as IL-2, IL-3, tumor necrosis factor (TNF), and lymphotoxin, which can all activate macrophages and aggravate inflammatory responses [21, 23, 24]. On the other hand, Th2 cells activate the signal transducer and transcription 6 (STAT6) through IL-4, resulting in up-regulation of GATA-binding protein 3 (GATA3), the key regulator of Th2 cell development. Mouse atherosclerotic plaques include cytokines associated with Th2 cells such as IL-4, IL-5, IL-10 (secreted by regulatory T cells), and IL-13. However, whether Th2 cells possess a pro-atherogenic or atheroprotective effect is still a subject of debate [25]. Interestingly, individuals with elevated Th2 cells in the peripheral blood mononuclear cells (PBMC) fraction have less preclinical atherosclerosis risk, as indicated by a lower thickness of the common carotid intimal media, compared to people with reduced levels of Th2 cells. Furthermore, the release of IL-4 from human activated mononuclear leukocytes in vitro is inversely associated with clinical atherosclerosis in ApoE-/- and LDLR-/- mice [23, 24]. In addition to counteracting the effects of Th1 and Th2 cells, IL-4 also affects other cells, which complicates the understanding of its unique role in atherosclerosis. Mouse models have demonstrated that IL-4 production results in increased expression of CD36, SR-A1 on macrophages, VCAM1, matrix metalloproteinase 1, and MCP-1. These findings suggest a potential link between IL-4 production and the development of atherosclerosis [24]. It is also estimated that during aging, T cell subsets become more proinflammatory [12, 21]. To progress, through macrophage colony stimulating factor (M-CSF), monocytes in the tunica intima differentiate into inflammatory macrophages (M1) and anti-inflammatory macrophages (M2) [26]. Mice lacking M-CSF show resistance to atherosclerosis [27]. Atherosclerotic lipoproteins (ox-LDL) are then taken in by monocyte-differentiated macrophages through the SR-A1 and CD36 [28, 29]. SR-B1 targets macrophages to minimize cholesterol trafficking and foam cell formation in atherosclerosis [30]. Foam cell production increases the expression of chemoattractant factors, allowing VSMCs to penetrate the intima. Soluble N-cadherin inhibits VSMC proliferation and macrophage foam cell apoptosis in vitro [31, 32]. Intima monocyte-differentiated macrophages via IL-1 promote the proliferation of derived growth factors. This stimulates VSMC proliferation and formation of extracellular matrix components that form a fibrous cap. The fibrous cap, necrotic core thickening, and arterial remodeling index can cause plaque rupture and thrombosis [33]. Please see Figure 1 and Figure 2 below.

    Anatomy of the normal artery [tunica externa is the outer layer; tunica media is the middle layer comprising mostly vascular smooth muscle cells (VSMCs); tunica intima is the inner most layer encompassing endothelial cells (ECs)]. Created with BioRender.com

    Pathophysiology of atherosclerosis 1: entrance of oxidized low-density lipoprotein (ox-LDL); 2: entrance of monocytes in response to ox-LDL secreted by ox-LDL; 3: differentiation of monocytes into M1 macrophages via macrophage colony stimulating factor (M-CSF); 4: foam cell formation; 5: vascular smooth muscle cells (VSMCs) and fibrous cap formation; 6: necrosis and liberation of pro-inflammatory factors to enhance the process of atherosclerosis]. ICAM1: intercellular adhesion molecule 1; MCP-1: monocyte chemoattractant protein 1; VCAM1: vascular adhesion molecule 1. Created with BioRender.com

    Vascular mechanisms and atherosclerosis

    Genomic instability and atherosclerosis

    Data from experimental aged animal models have portrayed the associations between DNA damage and atherosclerosis [34]. End-replication and oxidative DNA damage mechanisms alter DNA stability; therefore, failures in DNA damage response (DDR) sequences cause genome instability by activating DDR [35, 36]. The result of constant DDR is the activation of cyclin-dependent inhibitor pathways, including p53/p21Cip and p16INK4A/Rb [37]. With the successful repair of damaged DNA regions, standard cellular mechanisms are resumed [38]. However, unresolved DNA damage induces nuclear events that activate ataxia-telangiectasia mutated (ATM) and post-translational modifications of the nuclear factor kappa light chain enhancer of activated B cells (NF-κB) essential modulator (NEMO), thus activating NF-κB, senescence (characterized by the expression of pro-inflammatory and immune-modulatory chemokines and cytokines, growth factors and matrix metalloproteinases) and, consequently, inflammatory circuits in atherosclerosis [39, 40]. This is evident in aged animal models that have been studied [41, 42]. Another evidence of an association between atherosclerosis, inflammation, and genetic instability involves the immune checkpoints CD80/CD86-CD28 and CTLA4. Breaks in DNA cause the expression of class I MHC and costimulatory molecules (CD80/CD86-CD28) in fibroblasts, together with adhesive molecules such as VCAM1 and ICAM1 [43]. Evidently, genetic deletion of CD80/86 vitiated the development of atherosclerosis in LDLR-/- mice by lowering the inflammatory feedback driven by T cells [44]. However, in the aortic regions, which comprise more advanced plaques, αCTLA4 antibody (negative regulator CD80/CD86-CD28) increased plaque T cell composition, indicating the importance of this pathway in interventional therapy for accelerated atherosclerosis [44, 45]. Patients with Hutchinson-Gilford progeria syndrome (HGPS) and Werner syndrome (WS) also show DNA detrition, cellular senescence, deregulated mTOR pathways (nutritional sensing pathway), and upregulated inflammatory responses and consequently succumb to complications from atherosclerosis [19, 46]. For instance, administering mTOR inhibitors in HGPS patients delays senescence [47]. Microsatellite instability (MSI) and loss of heterozygosity (LOH) are also commonly observed in atherosclerosis. For example, LOH at the hMSH2, hPMS1, and hMLH1 loci occurs in atherosclerosis. MSI and LOH have also been found on chromosomes 1, 2, 8, 9, and 17 in cerebral atherosclerotic plaques [48]. DNA repair has been shown to be less accurate in atherosclerotic tissues due to changes found in mismatch repair (MMR) genes [48]. The aortic samples from people with atherosclerosis had more damage to mitochondrial DNA (mtDNA) than the samples of healthy donors of identical age. These samples also had metabolic enzymes involved in fatty acid oxidation [49]. Manganese superoxide dismutase 2 (MnSOD2) deficiency and p66sch overexpression increase reactive oxygen species (ROS)-induced damage in mtDNA, accelerating atherosclerosis in ApoE-/- mice [50, 51]. In the same way, ApoE-/- mice models that were haploinsufficient for ATM/angiotensin (Ang) II receptor (ATR) repair mechanisms show worsened atherosclerotic lesions together with inflammatory markers in the mitochondria [52, 53]. The telomerase enzyme also ensures stable telomere lengths (TLs) and stability through the telomerase RNA component (TERC), telomerase reverse transcriptase (TERT), and dyskerin [54]. Their activities are reduced with age progression [54]. Despite contradictory results, associations have been observed between shorter TLs and CVDs [55]. For example, data from the West of Scotland Coronary Prevention Study Group (WOSCOPS) trial and other studies indicate a shorter TL as a predictor of cardiovascular risk factors such as atherosclerosis. Fibroblasts derived from patients with HGPS show premature senescence and telomere shortening [56]. VSMC, EC, and fibrous cap leukocytes also show shorter telomeres than usual during aging and plaque progression [57, 58]. The decreased expression of SRY-box transcription factor 4 (SOX4) genes in peripheral blood cells during aging also causes a shortening of TL, which contributes to senescence and atherosclerosis [59]. This is achieved by activating the tumor suppressor p53, reduction in levels of the coactivator 1- and the transcriptional regulator peroxisome proliferator-activated receptor γ (PPARγ) coactivator 1-α (PGC1-α) and PGC1-β leading to mitochondrial dysfunction, deficient apoptosis, or EC senescence [60]. In short, DNA damage involves genomic alterations, adducts, and shortening of the telomere. In the same line, blood cells undergo somatic genetic mutations as we age, leading to expanded leukocyte clones and clonal hematopoiesis of indeterminate potential (CHIP) (another aging mechanism) [61]. Individuals with CHIP have increased cardiovascular related mortality. Data shows that mutations in DNA methyltransferase 3A (DNMT3A), tet methylcytosine dioxygenase 2 (TET2), and ASXL1 facilitate the clonal expansion of leukocytes. Furthermore, Macrophages from TET2-knockout hyperlipidemic mice result in abnormal activation of the NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) and are absent in melanoma 2 (AIM2) inflammasome and contribute to enhanced atherosclerosis [60, 62, 63].

    Neurohormonal regulation and atherosclerosis

    Renin-angiotensin system (RAS)

    The renin-Ang-aldosterone system is crucial in the pathobiology of vascular disease, with Ang II acting through receptors ATR1 and ATR2. ATR1 promotes vasoconstriction, endothelial and cardiac dysfunction, aldosterone release, ROS, inflammation, and prothrombin activity [28]. During atherogenesis, Ang II stimulates Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase, causing the generation of superoxides in arterial wall cells. Superoxide is converted to hydrogen peroxide (H2O2), which promotes degradation of the IκB kinase complex, nuclear translocation of NF-κB, and transcriptional induction of pro-inflammatory genes such as VCAM1, MCP-1, or IL-6 [29, 64]. NF-κB inhibition reduces the expression of endothelial lipase, an enzyme that plays a role in ApoE and leukocyte and EC uptake during atherosclerosis when Ang II is present [65]. Furthermore, Ang II can potentially induce the polarization of RAW264.7 macrophages to the M1 type through the connexin 43/NF-κB pathway which may play a role in the development of atherosclerosis [66]. ATR2 and Ang-1–7/renin-Ang system (RAS) axis ensure cardioprotection by increasing resistance to cardiac remodeling mechanisms by modulating the calcineurin nuclear factor of activated T cells (NFAT) pathway (calcineurin/NFAT) [67]. In a cecal ligation and puncture (CLP)-induced inflammatory model, Ang converting enzyme 2 (ACE2)-Ang-(1–7)-Mas axis modulated M1/M2 macrophage polarization to reduce inflammation via toll-like receptor 4 (TLR4)-Mediated NF-κB and mitogen-activated protein kinase (MAPK) pathways [68]. Ang II, renin, and ACE activities physiologically decline with age, but local tissues show a marked increase in expression [69]. Ang II, ACE, and ATR1 levels have been found to be higher in the hearts of older mice and the thoracic aorta of C57/BL6 mice [70]. Ang II also induces premature senescence of VSMC and accelerates atheroma progression through the p53/p21 pathway. Inhibition of this pathway decreases pro-inflammatory signals and arrests premature senescence, preventing age-induced atherosclerosis in mouse models [65]. Collectively, Ang II facilitates aging by increasing free radical production, mitochondrial dysfunction, and telomere attrition equally driving atherosclerosis [29].

    Transforming growth factor-β (TGF-β) and insulin-like growth factor-1 (IGF-1)

    Transforming growth factor-β (TGF-β) exhibits anti-aging properties. Some recent studies have described its involvement in cellular senescence and atherosclerosis via various mechanisms [71]. Older mice portray increased TGF-β expression [72]. At higher vascular levels, TGF-β with Ang II in the presence of T cells increases the collagen/elastin ratio in favor of collagen, drives inflammation, senescence, and consequently, EC dysfunction, a common characteristic of atherosclerosis [7275]. TGF-β has both anti- and pro-inflammatory properties. It lowers the expression of pro-inflammatory cytokines MCP-1 and inducible nitric oxide synthase (iNOS), while simultaneously increasing the expression of IL-10 via the SMAD/p21/MAPK/Rho/ROCK pathways, as reviewed by Chen et al. [76]. On the other hand, defective TGF-β expression of TGF-β could be a driver of cardiac aging and atherosclerosis at higher concentrations (observed in aged mice) by stimulating the expression of chemoattractants such as IL-1 and IL-6, negatively regulating noncoding atheroprotective mRNAs, collagen synthesis, and upregulating some adhesion molecules and pathways dependent on plasminogen activator inhibitor (PAI) [72, 76].

    Insulin-like growth factor-1 (IGF-1) also has cytoprotective properties through antioxidant and anti-inflammatory effects on the vascular systems, contributing to atherosclerotic stability and ensuring longevity at lower levels [77]. Generally, these observations are made in the early stages of an organism’s lifespan [47, 78]. Its involvement in cardiovascular pathologies and cell senescence remains vague [79]. Some models show that the over secretion of IGF-1 and growth hormone (GH) is associated with atherosclerosis, longevity, and inflammation [80]. Interestingly, GH receptor (GHR) knockout mice with decreased serum IGF-1 levels lived 38–55% longer than wild-type mice [81]. Increased non-conjugated IGF-1 in the circulation stimulates atherosclerosis and senescence [82]. To support these results, the models of Harrington et al. [83] showed a connection between the genetic deletion of pregnancy-associated plasma protein A (PAPP-A), a metalloproteinase in the IGF system that increases the bioactivity of IGF, and resistance to the development of atherosclerotic lesions in high-fat mice lacking ApoE, which was accompanied by an extended life span. Mechanistically, PAPP-A impairs liver X receptor expression through the IGF-1/PI3K/Akt signaling channel, hence reducing ATP binding cassette A1 (ABCA1), ABCG1, and SR-B1 expression and cholesterol efflux in macrophage-derived foam cells thereby promoting plaque formation [84, 85]. However, Mediterranean diet supplementation promotes IGF-1 inhibition downregulates mTORC1 which activates the fork head box O 3A (FOXO3A), resulting in anti-inflammatory and gene homeostasis effects, thereby promoting longevity [47]. Equally, FOXO3 protects vascular ECs and VSMCs against oxidative stress injury by up-regulating the expression of antioxidant enzymes such as catalase and MnSOD, enzymes implicated in atherosclerosis [86]. Therefore, in both cases, the results remain contradictory because vascular concentrations leading to the occurrence of lesions have not been universally established.

    Autonomic nervous system and atherosclerosis

    Catecholamine levels increase with age, leading to increased tissue spillover and reduced plasma clearance [87]. Studies in adults with prevalent CVDs show increased white blood cell count, reduced baroreflex sensitivity, and advanced carotid atherosclerosis compared to those without a history of CVD [20]. Age-related increases in catecholamine can lead to β-adrenergic desensitization, as seen in the infusion of isoproterenol and dobutamine models [88]. In atherosclerosis, excessive sympathetic activation leads to cardiac hypertrophy, necrosis, and apoptosis and promotes inflammatory signaling [88]. Non selective inhibition of β1 and β2 reduces M1 content in atherosclerotic lesions [89]. Opposingly, β3-adrenoceptor and dopamine D1-like activation improve atherosclerotic plaques in aged ApoE-/- mice by attenuating lipid profiles (free cholesterol, cholesteryl ester, and non-HDL triglycerides) and suppressing macrophage proliferation induced by ox-LDL through the PI3/AKT and MAPK/ERK signaling pathways [90, 91]. The implication of α adrenergic receptors in atherosclerosis is not well elucidated. However, impeding α1 receptors improves lipid profiles in favor of atherosclerosis prevention while the latter is observed in α2 activation [81, 90, 92]. Furthermore, catecholamines autoxidize into highly reactive pro-oxidant species, including amino chromes, o-semiquinones, OH-, and O2-, promoting lipid peroxidation and cellular necrosis. The activation of catecholamine monoamine oxidase (MAO) in the plasma membrane also generates H2O2 and aldehydes, which opens the mitochondrial permeability transition pore (MPTP) to promote mtDNA damage induced by oxidative stress, cardiomyocyte death, and heart failure [87]. Both enzymes (MAO-A/B) are expressed on the mitochondrial surface. MAO-A inhibits mitophagy and induces senescence. However, MAO-B impairment retards atherosclerosis and the progression of nonalcoholic fatty liver disease [93]. To further study outcomes of sympathetic and atherosclerosis association, Li et al. [94] designed a model in which they observed the effects of renal denervation (RDN) and atherosclerosis progression in ApoE-/- mice. RDN reduces atherosclerosis, EC mitochondrial oxidative stress and inflammation by impairing MAO-A activities [94]. Acetylcholine is a protective neurotransmitter in atherosclerosis due to its role in impeding NF-κB, IL-6, TNF-α, and C-reactive protein expression [95], modulating senescence, proliferation of VSMC and antioxidant pathways via the α7 nicotinic acetylcholine receptor (α7nAchR) [96]. Therefore, its decrease in aged models leads to a change in the immune reflex axis towards NF-κB, hence driving senescence and the inflammatory pathways that contribute to atherosclerosis [97].

    Epigenetics and atherosclerosis

    Epigenetics and aging mechanisms are linked to age-related diseases [98]. Individuals with advanced epigenetic caducity have an increased cardiovascular age and a higher risk of subclinical atherosclerosis progression [99]. Hypomethylation and DNA hypermethylation have been observed in the atheroma of ApoE-/- mice and monocyte/macrophage cell lines before and after the onset of atherosclerosis [100103]. Patients with atherogenesis and familial hypercholesterolemia (FH) often exhibit hypermethylation of promoters and protective genes such as ABCA1 and Krüppel-like factor 4 (KLF4) [104]. Atherogenic enzymes and receptors, including SOD, endothelial nitric oxide synthase (eNOS), and estrogen receptors, are hypomethylated [105, 106]. This overshadows the protective mechanisms. DNMT1 disrupts PPARγ signaling, leading to atherosclerosis. TET2 counteracts this, restoring autophagic activities in EC and VSMC [107]. The somatic mutation TET2, which leads to accelerated atherosclerosis, is associated with NLRP3-mediated IL-1β production in clonal hematopoiesis [108].

    Histone deacetylases (HDACs) affect the progression of atherosclerosis in various ways, from lipid metabolism, EC integrity, inflammatory pathways, and plaque disruption to modulation of oxidative pathways. The role of HDACs in atherosclerosis often leads to conflicting results. HDAC3 plays a role in EC integrity, but mice lacking it exhibit more stable plaques, decreased NADPH oxidase 4 (Nox4), reduced lipid content, and an anti-inflammatory phenotype during atherosclerosis. HDAC3 also inhibits PPAR and enhances foam cell formation [109]. HDAC9 interacts with the IκK/NF-κB p65 complex, leading to inflammatory phenotypes in macrophages and ECs linked to atherosclerosis. HDAC9-/- mice exhibit increased histone H3K9 acetylation at the ABCA1 and ABCG1 promoters, which affects macrophage cholesterol efflux and highlights its significant role in atherosclerosis [110]. HDAC6 causes EC injury and autophagy [111]. HDAC1/HDAC2 disrupts prolyl-4-hydroxylase alpha1 (P4Hα1) transcription, an enzyme in collagen formation, enhancing plaque destabilization. Furthermore, HDAC2 exhibits sexual dimorphic activity when inhibiting the expression of the antioxidant glutaredoxin-1 [111, 112]. HDAC7 suppresses VSMC proliferation and neointima formation by preventing β-catenin nuclear translocation [113, 114]. Recently, data suggest a more protective role for HDAC11 primarily through the expression of IL-10, IL-17, IL-10, IL-12, and TNF [115]. Members of the sirtuin (Sirt) family (Sirt1, Sirt3, and Sirt6) are studied for their antiaging and vasoprotective activities in atherosclerosis. Sirt1 modulates FOXO signaling, upregulates 5’-AMP-activated protein kinase (AMPK) (in low energy conditions), and restores normal autophagic activities (mTOR inhibition) [47], while Sirt6 targets inflammatory factors and loss of telomeres. SIRT3 protects against oxidative stress by upregulating PPAR‐α and eNOS and downregulating iNOS [116118]. Sirt activities decline with age.

    Non coding RNAs (ncRNAs) play a crucial role in atherogenesis, with their expression linked to EC dysfunction, inflammation, lipogenesis, and macrophage deregulation in atherosclerosis, with knockdown mice models demonstrating alleviation. Long ncRNAs (LncRNAs) and circular RNAs (circRNAs) modulate atherosclerotic progression via microRNAs (miRNAs) at the transcriptional and post-transcriptional levels. In cardiovascular development, three novel LncRNAs (TERMINATOR, ALIEN, and PUNISHER) have attracted great attention [119]. LncRNA H19 promotes the area of atherosclerotic plaque area in ApoE-/- by inhibiting cholesterol efflux mechanisms and upregulating NF-κB-mediated inflammatory responses in vascular ECs by miR-130b [120]. The lncRNA RP5–833A20.1 THP-1 was found to increase oxidized LDL uptake, lower cholesterol efflux, and increase the production of inflammatory mediators like TNF-α, IL-1β, and IL-6 in macrophages [121]. However, the expression of LncRNA-DYNLRB2-2 lowered the amount of cholesterol in macrophages by increasing the expression of G-protein-coupled receptor 119 (GPR119) and the cholesterol transporter ABCA1 [122]. LncRNA SMILR (smooth muscle-induced LncRNA) enhances replication and aggravates atherosclerosis by targeting the miR-10b-3p/KLF5 axis [123, 124]. LncRNA-p21 improves atherosclerosis by regulating the miR-221/SIRT1/Pcsk9 axis [125]. LncRNA-MALAT1 sponges miR-330-5p to activate the NF-κB signal pathway in THP-1 macrophage-derived foam cells in atherosclerosis pathogenesis [126]. Similarly, circRNAs are differentially expressed in atherosclerotic regions. Li et al. [127] found that 52 circRNAs were up-regulated more than twice and 47 circRNAs were down-regulated more than 1.5 times in atherosclerotic aortic vessels and of the circRNA examined, Mmu_circRNA_36781 (circRNA ABCA1) and 37699 (circRNA KHDRBS1) were significantly up-regulated in atherosclerotic aortic vessels and H2O2-treated mouse aortic ECs. They potentially regulated atherosclerosis and vascular endothelial injury by targeting the miR-30d-3p-TP53RK and miR-140-3p-MKK6 axes [127, 128].

    Protein homeostasis and atherosclerosis

    Protein homeostasis declines with age, leading to abnormalities in the neurological and cardiovascular systems [129]. Numerous studies show that one of the main factors influencing atherogenesis is cell death [130, 131]. The signaling channels involved include the ubiquitin- proteasome, chaperons, and lysosome-dependent autophagy, with evidence found in human and animal cardiovascular models leading to atherosclerosis [132]. Madrigal-Matute et al. [129] found a decrease in activities and elevated inflammatory responses after chaperone-mediated autophagy (CMA) blockage in a fluorescent reporter mouse model, consistent with the results observed by Qiao et al. [133] (2021). Subclinical models demonstrate the essential involvement of the ubiquitin proteasome system (UPS) in NF-κB activation, where impaired UPS is associated with uncontrolled NF-κB pathways that drive inflammation in inflammatory-related disorders [131, 134]. Exploring the connection between atherosclerosis and UPS has revealed both pro-atherogenic and potentially preventive impacts. Increased inhibition of the proteasome accelerates the progression of atherosclerosis, while reduced inhibition enhances cholesterol efflux through ABCA1 and ABCG1 [135]. UPS is essential for the intracellular breakdown of proteins in all cells. Under stress, these proteasome activities are replaced by immunoproteasomes (IP), which mediate immune responses in various inflammatory diseases. Nevertheless, in LDLR-/-LMP7-/- and LDLR-/- mice fed on a Western-type diet for 6 weeks or 24 weeks to induce early and advanced stage atherosclerosis, respectively, depleted IP subunit β5i/LMP7 did not alter protein homeostasis and macrophage polarization, thus not aggravating atherogenesis in LDLR-/- mice [96]. The degradation of perilipin (protects lipid droplets from lipase activities) through the UPS pathway leads to the progression of atherosclerosis, while inhibition of adiponectin receptor 1 (ADR1) protects atherosclerotic models by altering the generation of foam cells [136]. Ubiquitin-specific peptidase 9 X-linked (USP9X), a deubiquitinating enzyme, inhibits macrophage lipid uptake by downregulating SR-A1, preventing foam cell formation [137]. Furthermore, ubiquitin C-terminal hydrolase L1 (UCHL1) and USP14 promote CD36 autophagic degradation, but their mechanical effects have opposite results: Inhibition of USP14 leads to the suppression of foam cells while deubiquitinating UCHL1 promotes the formation and stabilization of foam cells [138, 139]. Heat shock protein 60 (HSP 60) is the most recognized HSP 60 with known direct atherogenic potential. However, HSP 70, HSP 90, and HSP 47 have shown the potential to aggravate atherosclerosis in ApoE-/- mice and THP-1 derived macrophage models as reviewed by various authors [139144]. In the same line, HSP 27 demonstrates atheroprotective activities in vitro [142].

    Gut microbiome and atherosclerosis

    The gut microbiome and the immune system are interdependent, and disruptions can increase vulnerability to disease and influence drug candidates’ pharmacokinetic or pharmacodynamic results. Studies show that as we age, the diversity of the gut microbiome decreases due to a loss in organisms’ physiological and symbiotic capacities, leading to reduced longevity and age-related anomalies [145]. Reprogramming of the gut microbiome induced by trimethylamine N-oxide (TMAO) is a typical example that serves as a prognostic index for CVDs and senescence [146, 147]. In aged animal models, TMAO is elevated and accelerates cell senescence by causing mitochondrial damage, superoxide formation (ROS), and promoting the generation of pro-inflammatory factors such as caspase-1 and IL-1β through the ROS-TXNIP-NLRP3 signaling pathway, thus aggravating atherosclerosis [148, 149]. It can also stop the SIRT3-SOD2-mitochondrial ROS signaling pathway, which damages ECs and leads to the development of atherosclerosis [148]. Zhu et al. [150] found that an enriched titanium dioxide diet (E71)/TMAO in ApoE-/- mice caused the appearance of lesions, mediated by E71, which transformed choline into pro-atherogenic TMAO. Consistently, Traughber et al. [151] reversed cadmium and high-fat-induced atherosclerosis by inhibiting TMAO production, NLRP3 machinery, and macrophage polarization in the gut microbiota of mice. TMAO also reduces bile acid formation, increases foam cell formation, and promotes oxidative stress-induced mitochondrial damage, leading to pro-inflammatory factors, senescence, and atherosclerosis during aging [148]. The alteration of the intestinal microbiome reduces butyrate-producing bacteria, increasing intestinal permeability and lipopolysaccharide levels, which activate inflammatory pathways, primarily in gram-negative species [152]. The intestinal microbiomes also produce hydrogen sulfide (H2S), which supports vascular tone, VSMC, cholesteryl ester (CE) activities, and energy production under normal conditions, but under strenuous conditions, H2S becomes pro-atherogenic [153156]. Specifically, H2S is produced by three key enzymes; cystathionine β-lyase (CBE), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (MST)/cysteine aminotransferase (CAT) [157]. CSE is predominantly in the cardiovascular system [147]. These enzymes can be suppressed under conditions of high glucose, oxidative stress, and high fat diet (HFD) thus leading to a reduction in H2S production [156, 158]. For example, inhibition of MST by 2-[(4-hydroxy-6-methylpyrimidin-2-yl)sulfanyl]-1-(naphthalen-1-yl)ethan-1-one (HMPSNE) resulted in adipocyte differentiation and lipid accumulation with increased inflammation of adipose tissue [159]. CSE exhibits its antiatherosclerotic effect through 3 mechanisms, namely reduction of the chemoattractant factor, ICAM1, and CX3C CCR 1 (CX3CR1), inhibition of macrophage lipid uptake, and induction of VSMCs apoptosis through the MAPK pathway [157]. Although it has not been clinically tested, H2S has excellent potential as a treatment for this pathophysiological condition. Figure 3 below summarizes vascular mechanisms associated with aging and atherosclerosis materialization.

    Vascular mechanisms (genomic instability, microbiome dysbiosis, neurohormonal deregulation, epigenetics, and proteostasis/protein deregulation) and atherosclerosis. These vascular mechanisms lead to increased senescent activities accompanied by elevated senescence associated secretory patterns which further aggravate inflammatory signals implicated in atherosclerosis. AMPK: 5’-AMP-activated protein kinase; ANS: autonomous nervous system; CircRNA: circular RNA; C-RP: C-reactive protein; H2S: hydrogen sulfide; ICAM1: intercellular adhesion molecule 1; IGF-1: insulin-like growth factor-1; LncRNA: long noncoding RNA; LPS: lipopolysaccharide; miRNA: micro RNA; mtDNA: mitochondrial DNA; NF-κB: nuclear factor kappa light chain enhancer of activated B cells; NLRP3: NACHT, LRR, and PYD domain-containing protein 3; ROS: reactive oxygen species; SASP: senescence-associated secretory phenotype; Sirt1: sirtuin 1; SR-A1: scavenger receptor A1; TGF-β: transforming growth factor-β; TMAO: trimethylamine N-oxide; TNF-α: tumor necrosis factor-α; UPS: ubiquitin-proteasome system; VCAM1: vascular adhesion molecule 1; VSMC: vascular smooth muscle cell. Created with BioRender.com

    Conclusions

    Statin therapy remains the gold standard in atherosclerosis due to its pleiotropic nature [160]. Given the complexity of atherosclerosis, it is essential to find more adjuvant compounds (Figure 4).

    Various pharmacological interests in the treatment of atherosclerosis. PCSK9: proprotein convertase subtilisin/kexin type 9; PPARα: peroxisome proliferator-activated receptor α; SCFAs: short-chain fatty acids; SGLT2: sodium glucose transporter 2; TET2: tet methylcytosine dioxygenase 2; TMAO: trimethylamine N-oxide

    Potentially beneficial HDAC inhibitors in the treatment of atherosclerosis mainly target class I and II HDACs. These include TMP195, RGFP966, MC1568, and sodium valproate (antiepileptic). Other nonspecific HDAC inhibitors are trichostatin and vorinostat. HDAC inhibitors that have entered clinical trial phases primarily target neurological and oncological disorders [161, 162]. Please see Table 1.

    Histone deacetylases (HDACs) inhibitors

    Drug Model and mechanismsStructureAuthorReference
    TMP195HDAC9 inhibition (hyperlipidemic mice)Asare et al. (2020)[163]
    RGFP966HDAC3 inhibition (ApoE-/- mice)Chen et al. (2021)[164]
    MC1568HDAC9 inhibition (ex vivo genetic knockout HDAC9 endothelial cells)Lecce et al.[165]
    Sodium valproateHDAC9 inhibition (human patients)Brookes et al. (2018) [166]
    Display full size

    Reconstitution of TERC or TERT expression in TERC- or TERT-deficient mice with critically short telomeres resulted in telomere elongation, less DNA damage, decreases in aging biomarkers, and delay in age-related pathologies. For example, in preclinical models, gene therapy involving the administration of TERT adeno-associated virus (AAV), lentivirus, anellovirus, and lipid nanoparticles to young and old mice resulted in elongated telomeres that can prolong life expectancy and delay age-associated pathologies [61, 167]. Partially, the delivery of modified mRNA encoding TERT to human fibroblasts and myoblasts increased telomere proliferation in a dose dependent manner by 1012 fold. However, after 4 days of treatment, all treated cell populations eventually stopped increasing in number and expressed senescence markers to the same extent as untreated cells. This method is rapid, shows minimal risks of mutagenesis, and could have wide utility in regenerative medicine, drug development, and disease comprehension [60]. Furthermore, studies demonstrate that the use of key regulatory enzymes 8-oxoguanine DNA glycosylase (OGG1) can recruit TET1 into damaged DNA regions, thus preventing the activation of the inflammasome [168]. TA-65 telomerase activator (Astragalus, an antiaging agent), resveratrol, pterostilbene, PPAR agonists (e.g., pioglitazone), and statins, alleviate inflammation, senescence, increase leukocyte TL, promote stabilization of the Nijmegen breakage syndrome-1 protein, upregulate telomere repeat binding factor 2 (TRF2) and improve endothelial function with dietary supplementation, caloric restriction, and inclusion of polyunsaturated fatty acid [61, 169171]. Clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR/CAS9) technology has shown significant success in telomere-targeted therapy. However, little effort has been directed towards cardiovascular treatment [172]. A recent study by Xu et al. [173] (2024) used a CRISPR/CAS9 method to target the proprotein convertase subtilisin/kexin type 9 (Psck9) gene and successfully prevented the occurrence of atherosclerosis in hyperlipidemic mice. However, telomerase-targeting therapies in CVD face carcinogenic concerns.

    Butyrate, a short-chain fatty acid (SCFA), has anti-inflammatory effects in animal models by creating barriers between lipopolysaccharides and the intestinal microbiome [174]. Research shows that reduction in choline substrate intake, along with acetylsalicylic acid and metformin therapy, can effectively lower TMAO in humans and ApoE-/- mice [175].

    It should be noted that cellular senescence is an aging hallmark characterized by the presence of senescence associated secretory phenotypes (SASPs) [1113, 176]. There is evidence atherosclerotic plaques encompass SASP components such as matrix metalloproteinases and multiple inflammatory factors [176]. SASPs drive atherosclerosis by upregulating inflammatory cascades involving the NF-κB, IL-1α, TGF-β, IL-8, and IL-6 [50]. Hence, senescence collectively alters DNA repair mechanisms and impairs antioxidant defenses, causing vascular inflammation and atherosclerosis in the presence of defective apoptotic pathways [176]. Clearing senescent cells in atherosclerosis is a therapeutic achievement. However, approved senolytic drugs, such as navitoclax (ABT-263) have been shown to make plaque less stable and increase mortality in atherosclerotic models [177]. Resveratrol indirectly prevents and attenuates cellular senescence by positively modulating energy and nutritional sensing pathways including AMPK and FOXO3A via Sirt1 [47]. Other drug candidates with antisenescent and anti-aging effects useful in atherosclerosis management have been summarized in the review by Sun et al. [176] (2022).

    Targeting non-coding RNAs produces both intended and unintended effects. Mechanisms of drug delivery to atherosclerotic regions remain challenging in atherosclerosis. Inclisiran (Leqvio®; Novartis) is a first-in-class, cholesterol-lowering small interfering RNA (siRNA) approved by the FDA/EMA to target PCSK9 and has a favorable administration regimen compared to normal PSCK9 antibodies [178, 179]. It is used in patients who cannot achieve LDL cholesterol goals with the maximum tolerated doses of statins [180].

    The multifaceted role of Ag II makes targeting RAAS equally crucial in the treatment of atherosclerosis. ACE inhibitors attenuate pro-inflammatory and oxidative pathways [70]. Ang (1–7)/RAS/ACE2 is atheroprotective, but its half-life and instability hinder drug delivery methods. Hydroxypropyl β-cyclodextrin-incorporated (HPβCD/Ang-1–7) formulations (HPβCD/Ang-1–7) address these problems by incorporating a thioether bridge, stabilizing the peptide against hydrolysis and increasing its half-life by eliminating the affinity of the AT1 receptor [181, 182] (Figure 5).

    Cyclic Ang-(1–7) with thioether bridge between amino acid numbers 4 and 7

    The development and progression of atherosclerosis are also related to various modalities of cell death (autophagy, apoptosis, necroptosis, pyroptosis, and ferroptosis) [183, 184]. Sodium glucose transporter 2 (SGLT2) and factor Xa inhibitors are additional main drugs that show beneficial effects when targeting these cell death pathways in atherosclerosis and require more extensive research [185, 186].

    Recent investigations also evoke inflammation-induced intraplaque angiogenesis as a potential therapeutic target, as reviewed by Perrotta et al. [187]. Documented angiopoietin antibodies decreased intimal fatty streak formation in some mouse models of hypercholesterolemia-induced atherosclerosis, but did not affect preexisting plaques [188190].

    Although not fully covered, the review also highlights the importance of phytoextracts (e.g., Astragalus polysaccharides) in the management of atherosclerosis. Other natural pharmacological interventions that stimulate autophagy and Sirt activities (e.g., flavonoids, trehalose, or spermidine treatment) have been reported to reverse aspects of arterial aging and atherosclerosis [11, 191, 192]. Above all, contextualizing atherosclerosis to one particular pathway is impossible; therefore, more extensive research is paramount.

    Abbreviations

    ABCA1:

    ATP binding cassette A1

    ACE2:

    angiotensin converting enzyme 2

    Ang:

    angiotensin

    ATR:

    angiotensin II receptor

    CCR2:

    CC-chemokine receptor 2

    circRNAs:

    circular RNAs

    CSE:

    cystathionine γ-lyase

    CVDs:

    cardiovascular diseases

    DDR:

    DNA damage response

    ECs:

    endothelial cells

    FOXO3A:

    fork head box O 3A

    H2O2:

    hydrogen peroxide

    H2S:

    hydrogen sulfide

    HDACs:

    histone deacetylases

    HGPS:

    Hutchinson-Gilford progeria syndrome

    HSP 60:

    heat shock protein 60

    ICAM1:

    intercellular adhesion molecule 1

    IGF-1:

    insulin-like growth factor-1

    LDL:

    low-density lipoprotein

    LncRNA:

    long noncoding RNA

    LOH:

    loss of heterozygosity

    MAO:

    monoamine oxidase

    MAPK:

    mitogen-activated protein kinase

    MCP-1:

    monocyte chemoattractant protein 1

    mtDNA:

    mitochondrial DNA

    NF-κB:

    nuclear factor kappa light chain enhancer of activated B cells

    NLRP3:

    NACHT, LRR, and PYD domain-containing protein 3

    ox-LDLs:

    oxidized low-density lipoproteins

    PPARγ:

    peroxisome proliferator-activated receptor γ

    RAS:

    renin-angiotensin system

    ROS:

    reactive oxygen species

    SASPs:

    senescence associated secretory phenotypes

    Sirt:

    sirtuin

    SRs:

    scavenger receptors

    TERC:

    telomerase RNA component

    TERT:

    telomerase reverse transcriptase

    TET2:

    tet methylcytosine dioxygenase 2

    TGF-β:

    transforming growth factor-β

    Th1:

    T cell helper 1

    TLs:

    telomere lengths

    TMAO:

    trimethylamine N-oxide

    TNF:

    tumor necrosis factor

    UPS:

    ubiquitin proteasome system

    VCAM1:

    vascular adhesion molecule 1

    VSMCs:

    vascular smooth muscle cells

    Declarations

    Acknowledgments

    We thank all members of the Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, and the National Natural Sciences Foundation of China for their support.

    Author contributions

    SCW: Writing—original draft. XMJ: Conceptualization, Investigation, Writing—review & editing. MX: Conceptualization, Investigation, Writing—review & editing, Funding acquisition.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    This work is supported by the National Natural Science Foundation of China [82373870]. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2024.

    References

    Cardiovascular diseases (cvds) [Internet]. WHO; c2024 [cited 2023 Jun 13]. Available from: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)
    Shah SR, Abbasi Z, Fatima M, Ochani RK, Shahnawaz W, Khan MA, et al. Canakinumab and cardiovascular outcomes: results of the CANTOS trial. J Community Hosp Intern Med Perspect. 2018;8:212. [DOI] [PubMed] [PMC]
    Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM, Hovingh GK, Ivkovic M, et al.; RESCUE Investigators. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2021;397:20609. [DOI] [PubMed]
    Tardif J, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N Engl J Med. 2019;381:2497505. [DOI] [PubMed]
    Ridker PM, Rane M. Interleukin-6 Signaling and Anti-Interleukin-6 Therapeutics in Cardiovascular Disease. Circ Res. 2021;128:172846. [DOI] [PubMed]
    Minamino T, Miyauchi H, Yoshida T, Tateno K, Kunieda T, Komuro I. Vascular cell senescence and vascular aging. J Mol Cell Cardiol. 2004;36:17583. [DOI] [PubMed]
    Wang JC, Bennett M. Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ Res. 2012;111:24559. [DOI] [PubMed]
    Golledge J. Update on the pathophysiology and medical treatment of peripheral artery disease. Nat Rev Cardiol. 2022;19:45674. [DOI] [PubMed]
    O’Rourke MF, Hashimoto J. Mechanical factors in arterial aging: a clinical perspective. J Am Coll Cardiol. 2007;50:113. [DOI] [PubMed]
    Zhang W, Hui R, Yang S. Telomeres, cardiovascular aging, and potential intervention for cellular senescence. Sci China Life Sci. 2014;57:85862. [DOI] [PubMed]
    Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of Vascular Aging. Circ Res. 2018;123:84967. [DOI] [PubMed] [PMC]
    Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol. 2023;20:75477. [DOI] [PubMed]
    López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell. 2023;186:24378. [DOI] [PubMed]
    Bu L, Yuan H, Xie L, Guo M, Liao D, Zheng X. New Dawn for Atherosclerosis: Vascular Endothelial Cell Senescence and Death. Int J Mol Sci. 2023;24:15160. [DOI] [PubMed] [PMC]
    Glass CK, Witztum JL. Atherosclerosis. the road ahead. Cell. 2001;104:50316. [DOI] [PubMed]
    Björkegren JLM, Lusis AJ. Atherosclerosis: Recent developments. Cell. 2022;185:163045. [DOI] [PubMed] [PMC]
    Park YM. CD36, a scavenger receptor implicated in atherosclerosis. Exp Mol Med. 2014;46:e99. [DOI] [PubMed] [PMC]
    Xu S, Ogura S, Chen J, Little PJ, Moss J, Liu P. LOX-1 in atherosclerosis: biological functions and pharmacological modifiers. Cell Mol Life Sci. 2013;70:285972. [DOI] [PubMed] [PMC]
    Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Vidak S, Nevado RM, et al. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol Med. 2019;11:e9736. [DOI] [PubMed] [PMC]
    Ulleryd MA, Prahl U, Börsbo J, Schmidt C, Nilsson S, Bergström G, et al. The association between autonomic dysfunction, inflammation and atherosclerosis in men under investigation for carotid plaques. PLoS One. 2017;12:e0174974. [DOI] [PubMed] [PMC]
    Saigusa R, Winkels H, Ley K. T cell subsets and functions in atherosclerosis. Nat Rev Cardiol. 2020;17:387401. [DOI] [PubMed] [PMC]
    Combadière C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, et al. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6Chi and Ly6Clo monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:164957. [DOI] [PubMed]
    Chen J, Xiang X, Nie L, Guo X, Zhang F, Wen C, et al. The emerging role of Th1 cells in atherosclerosis and its implications for therapy. Front Immunol. 2023;13:1079668. [DOI] [PubMed] [PMC]
    Tse K, Tse H, Sidney J, Sette A, Ley K. T cells in atherosclerosis. Int Immunol. 2013;25:61522. [DOI] [PubMed] [PMC]
    Hinkley H, Counts DA, VonCanon E, Lacy M. T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease. Cells. 2023;12:2152. [DOI] [PubMed] [PMC]
    Song X, Song Y, Ma Q, Fang K, Chang X. M1-Type Macrophages Secrete TNF-α to Stimulate Vascular Calcification by Upregulating CA1 and CA2 Expression in VSMCs. J Inflamm Res. 2023;16:301932. [DOI] [PubMed] [PMC]
    Tabas I, Bornfeldt KE. Intracellular and Intercellular Aspects of Macrophage Immunometabolism in Atherosclerosis. Circ Res. 2020;126:120927. [DOI] [PubMed] [PMC]
    Crowley SD, Rudemiller NP. Immunologic Effects of the Renin-Angiotensin System. J Am Soc Nephrol. 2017;28:135061. [DOI] [PubMed] [PMC]
    Schmidt-Ott KM, Kagiyama S, Phillips MI. The multiple actions of angiotensin II in atherosclerosis. Regul Pept. 2000;93:6577. [DOI] [PubMed]
    Marques PE, Nyegaard S, Collins RF, Troise F, Freeman SA, Trimble WS, et al. Multimerization and Retention of the Scavenger Receptor SR-B1 in the Plasma Membrane. Dev Cell. 2019;50:28395.e5. [DOI] [PubMed]
    Koutsouki E, Beeching CA, Slater SC, Blaschuk OW, Sala-Newby GB, George SJ. N-cadherin-dependent cell-cell contacts promote human saphenous vein smooth muscle cell survival. Arterioscler Thromb Vasc Biol. 2005;25:9828. [DOI] [PubMed]
    Lyon CA, Johnson JL, White S, Sala-Newby GB, George SJ. EC4, a truncation of soluble N-cadherin, reduces vascular smooth muscle cell apoptosis and markers of atherosclerotic plaque instability. Mol Ther Methods Clin Dev. 2014;1:14004. [DOI] [PubMed] [PMC]
    Patel P, Rai V, Agrawal DK. Role of oncostatin-M in ECM remodeling and plaque vulnerability. Mol Cell Biochem. 2023;478:245160. [DOI] [PubMed] [PMC]
    Yu EPK, Bennett MR. Mitochondrial DNA damage and atherosclerosis. Trends Endocrinol Metab. 2014;25:4817. [DOI] [PubMed]
    Ricottini E, Madonna R, Grieco D, Zoccoli A, Stampachiacchiere B, Patti G, et al. Effect of High-Dose Atorvastatin Reload on the Release of Endothelial Progenitor Cells in Patients on Long-Term Statin Treatment Who Underwent Percutaneous Coronary Intervention (from the ARMYDA-EPC Study). Am J Cardiol. 2016;117:16571. [DOI] [PubMed]
    Pan M, Li K, Lin S, Hung W. Connecting the Dots: From DNA Damage and Repair to Aging. Int J Mol Sci. 2016;17:685. [DOI] [PubMed] [PMC]
    Engeland K. Cell cycle regulation: p53-p21-RB signaling. Cell Death Differ. 2022;29:94660. [DOI] [PubMed] [PMC]
    Yu E, Calvert PA, Mercer JR, Harrison J, Baker L, Figg NL, et al. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation. 2013;128:70212. [DOI] [PubMed]
    Miyamoto S. Nuclear initiated NF-κB signaling: NEMO and ATM take center stage. Cell Res. 2011;21:11630. [DOI] [PubMed] [PMC]
    Casella G, Tsitsipatis D, Abdelmohsen K, Gorospe M. mRNA methylation in cell senescence. Wiley Interdiscip Rev RNA. 2019;10:e1547. [DOI] [PubMed] [PMC]
    Lee JW, Ong EBB. Genomic Instability and Cellular Senescence: Lessons From the Budding Yeast. Front Cell Dev Biol. 2021;8:619126. [DOI] [PubMed] [PMC]
    Vijg J, Suh Y. Genome instability and aging. Annu Rev Physiol. 2013;75:64568. [DOI] [PubMed]
    Tang MLF, Khan MKN, Croxford JL, Tan KW, Angeli V, Gasser S. The DNA damage response induces antigen presenting cell-like functions in fibroblasts. Eur J Immunol. 2014;44:110818. [DOI] [PubMed]
    Ewing MM, Karper JC, Abdul S, Jong RCMd, Peters HAB, Vries MRd, et al. T-cell co-stimulation by CD28-CD80/86 and its negative regulator CTLA-4 strongly influence accelerated atherosclerosis development. Int J Cardiol. 2013;168:196574. [DOI] [PubMed]
    Poels K, Leent MMTv, Reiche ME, Kusters PJH, Huveneers S, Winther MPJd, et al. Antibody-Mediated Inhibition of CTLA4 Aggravates Atherosclerotic Plaque Inflammation and Progression in Hyperlipidemic Mice. Cells. 2020;9:1987. [DOI] [PubMed] [PMC]
    Wu Z, Zhang W, Song M, Wang W, Wei G, Li W, et al. Differential stem cell aging kinetics in Hutchinson-Gilford progeria syndrome and Werner syndrome. Protein Cell. 2018;9:33350. [DOI] [PubMed] [PMC]
    Yang K, Hou R, Zhao J, Wang X, Wei J, Pan X, et al. Lifestyle effects on aging and CVD: A spotlight on the nutrient-sensing network. Ageing Res Rev. 2023;92:102121. [DOI] [PubMed]
    Cervelli T, Borghini A, Galli A, Andreassi MG. DNA damage and repair in atherosclerosis: current insights and future perspectives. Int J Mol Sci. 2012;13:1692944. [DOI] [PubMed] [PMC]
    Ballinger SW, Patterson C, Knight-Lozano CA, Burow DL, Conklin CA, Hu Z, et al. Mitochondrial integrity and function in atherogenesis. Circulation. 2002;106:5449. [DOI] [PubMed]
    Tyrrell DJ, Goldstein DR. Ageing and atherosclerosis: vascular intrinsic and extrinsic factors and potential role of IL-6. Nat Rev Cardiol. 2021;18:5868. [DOI] [PubMed] [PMC]
    Zhang Y, Weng J, Huan L, Sheng S, Xu F. Mitophagy in atherosclerosis: from mechanism to therapy. Front Immunol. 2023;14:1165507. [DOI] [PubMed] [PMC]
    Mercer JR, Cheng K, Figg N, Gorenne I, Mahmoudi M, Griffin J, et al. DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circ Res. 2010;107:102131. [DOI] [PubMed] [PMC]
    Peng W, Cai G, Xia Y, Chen J, Wu P, Wang Z, et al. Mitochondrial Dysfunction in Atherosclerosis. DNA Cell Biol. 2019;38:597606. [DOI] [PubMed]
    Hoffmann J, Richardson G, Haendeler J, Altschmied J, Andrés V, Spyridopoulos I. Telomerase as a Therapeutic Target in Cardiovascular Disease. Arterioscler Thromb Vasc Biol. 2021;41:104761. [DOI] [PubMed]
    Brouilette SW, Moore JS, McMahon AD, Thompson JR, Ford I, Shepherd J, et al.; West of Scotland Coronary Prevention Study Group. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study. Lancet. 2007;369:10714. [DOI] [PubMed]
    Yegorov YE, Poznyak AV, Nikiforov NG, Starodubova AV, Orekhov AN. Role of Telomeres Shortening in Atherogenesis: An Overview. Cells. 2021;10:395. [DOI] [PubMed] [PMC]
    Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, et al. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ Res. 2006;99:15664. [DOI] [PubMed]
    Bloom SI, Liu Y, Tucker JR, Islam MT, Machin DR, Abdeahad H, et al. Endothelial cell telomere dysfunction induces senescence and results in vascular and metabolic impairments. Aging Cell. 2023;22:e13875. [DOI] [PubMed] [PMC]
    Stevanovic M, Lazic A, Schwirtlich M, Ninkovic DS. The Role of SOX Transcription Factors in Ageing and Age-Related Diseases. Int J Mol Sci. 2023;24:851. [DOI] [PubMed] [PMC]
    Ramunas J, Yakubov E, Brady JJ, Corbel SY, Holbrook C, Brandt M, et al. Transient delivery of modified mRNA encoding TERT rapidly extends telomeres in human cells. FASEB J. 2015;29:19309. [DOI] [PubMed] [PMC]
    Yeh J, Lin M, Wang C. Telomeres as Therapeutic Targets in Heart Disease. JACC Basic Transl Sci. 2019;4:85565. [DOI] [PubMed] [PMC]
    Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med. 2017;377:11121. [DOI] [PubMed] [PMC]
    Fidler TP, Xue C, Yalcinkaya M, Hardaway B, Abramowicz S, Xiao T, et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature. 2021;592:296301. [DOI] [PubMed] [PMC]
    Dandona P, Dhindsa S, Ghanim H, Chaudhuri A. Angiotensin II and inflammation: the effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockade. J Hum Hypertens. 2007;21:207. [DOI] [PubMed]
    Ekholm M, Kahan T. The Impact of the Renin-Angiotensin-Aldosterone System on Inflammation, Coagulation, and Atherothrombotic Complications, and to Aggravated COVID-19. Front Pharmacol. 2021;12:640185. [DOI] [PubMed] [PMC]
    Wu L, Chen K, Xiao J, Xin J, Zhang L, Li X, et al. Angiotensin II induces RAW264.7 macrophage polarization to the M1‑type through the connexin 43/NF‑κB pathway. Mol Med Rep. 2020;21:210312. [DOI] [PubMed] [PMC]
    Wang Y, Tikellis C, Thomas MC, Golledge J. Angiotensin converting enzyme 2 and atherosclerosis. Atherosclerosis. 2013;226:38. [DOI] [PubMed]
    Pan H, Huang W, Wang Z, Ren F, Luo L, Zhou J, et al. The ACE2-Ang-(1‑7)-Mas Axis Modulates M1/M2 Macrophage Polarization to Relieve CLP-Induced Inflammation via TLR4-Mediated NF-кb and MAPK Pathways. J Inflamm Res. 2021;14:204560. [DOI] [PubMed] [PMC]
    Diz DI, Varagic J, Groban L. Aging and the brain renin-angiotensin system: relevance to age-related decline in cardiac function. Future Cardiol. 2008;4:23745. [DOI] [PubMed] [PMC]
    Yoon HE, Kim EN, Kim MY, Lim JH, Jang I, Ban TH, et al. Age-Associated Changes in the Vascular Renin-Angiotensin System in Mice. Oxid Med Cell Longev. 2016;2016:6731093. [DOI] [PubMed] [PMC]
    Grainger DJ. Transforming growth factor β and atherosclerosis: so far, so good for the protective cytokine hypothesis. Arterioscler Thromb Vasc Biol. 2004;24:399404. [DOI] [PubMed]
    Tominaga K, Suzuki HI. TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int J Mol Sci. 2019;20:5002. [DOI] [PubMed] [PMC]
    Fleenor BS, Marshall KD, Durrant JR, Lesniewski LA, Seals DR. Arterial stiffening with ageing is associated with transforming growth factor-β1-related changes in adventitial collagen: reversal by aerobic exercise. J Physiol. 2010;588:397182. [DOI] [PubMed] [PMC]
    Wang B, Omar A, Angelovska T, Drobic V, Rattan SG, Jones SC, et al. Regulation of collagen synthesis by inhibitory Smad7 in cardiac myofibroblasts. Am J Physiol Heart Circ Physiol. 2007;293:H128290. [DOI] [PubMed]
    Toma I, McCaffrey TA. Transforming growth factor-β and atherosclerosis: interwoven atherogenic and atheroprotective aspects. Cell Tissue Res. 2012;347:15575. [DOI] [PubMed] [PMC]
    Chen P, Qin L, Li G, Wang Z, Dahlman JE, Malagon-Lopez J, et al. Endothelial TGF-β signalling drives vascular inflammation and atherosclerosis. Nat Metab. 2019;1:91226. [DOI] [PubMed] [PMC]
    Macvanin M, Gluvic Z, Radovanovic J, Essack M, Gao X, Isenovic ER. New insights on the cardiovascular effects of IGF-1. Front Endocrinol (Lausanne). 2023;14:1142644. [DOI] [PubMed] [PMC]
    Abdellatif M, Trummer-Herbst V, Heberle AM, Humnig A, Pendl T, Durand S, et al. Fine-Tuning Cardiac Insulin-Like Growth Factor 1 Receptor Signaling to Promote Health and Longevity. Circulation. 2022;145:185366. [DOI] [PubMed] [PMC]
    Luo X, Jiang X, Li J, Bai Y, Li Z, Wei P, et al. Insulin-like growth factor-1 attenuates oxidative stress-induced hepatocyte premature senescence in liver fibrogenesis via regulating nuclear p53-progerin interaction. Cell Death Dis. 2019;10:451. [DOI] [PubMed] [PMC]
    Andreassen M, Raymond I, Kistorp C, Hildebrandt P, Faber J, Kristensen LØ. IGF1 as predictor of all cause mortality and cardiovascular disease in an elderly population. Eur J Endocrinol. 2009;160:2531. [DOI] [PubMed]
    Wang Y, Anesi J, Maier MC, Myers MA, Oqueli E, Sobey CG, et al. Sympathetic Nervous System and Atherosclerosis. Int J Mol Sci. 2023;24:13132. [DOI] [PubMed] [PMC]
    Kawachi S, Takeda N, Sasaki A, Kokubo Y, Takami K, Sarui H, et al. Circulating insulin-like growth factor-1 and insulin-like growth factor binding protein-3 are associated with early carotid atherosclerosis. Arterioscler Thromb Vasc Biol. 2005;25:61721. [DOI] [PubMed]
    Harrington SC, Simari RD, Conover CA. Genetic deletion of pregnancy-associated plasma protein-A is associated with resistance to atherosclerotic lesion development in apolipoprotein E-deficient mice challenged with a high-fat diet. Circ Res. 2007;100:1696702. [DOI] [PubMed]
    Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, et al. Role of PI3K in the Progression and Regression of Atherosclerosis. Front Pharmacol. 2021;12:632378. [DOI] [PubMed] [PMC]
    Abdellatif M, Eisenberg T, Heberle AM, Thedieck K, Kroemer G, Sedej S. Cardiac PI3K p110α attenuation delays aging and extends lifespan. Cell Stress. 2022;6:725. [DOI] [PubMed] [PMC]
    Zhao Y, Liu Y. Longevity Factor FOXO3: A Key Regulator in Aging-Related Vascular Diseases. Front Cardiovasc Med. 2021;8:778674. [DOI] [PubMed] [PMC]
    Barthelemy J, Pichot V, Hupin D, Berger M, Celle S, Mouhli L, et al. Targeting autonomic nervous system as a biomarker of well-ageing in the prevention of stroke. Front Aging Neurosci. 2022;14:969352. [DOI] [PubMed] [PMC]
    Seals DR, Esler MD. Human ageing and the sympathoadrenal system. J Physiol. 2000;528:40717. [DOI] [PubMed] [PMC]
    Wang Y, Nguyen DT, Anesi J, Alramahi A, Witting PK, Chai Z, et al. Moxonidine Increases Uptake of Oxidised Low-Density Lipoprotein in Cultured Vascular Smooth Muscle Cells and Inhibits Atherosclerosis in Apolipoprotein E-Deficient Mice. Int J Mol Sci. 2023;24:3857. [DOI] [PubMed] [PMC]
    Shi S, Li Y, Guo Y, Wang Z. Effect of beta-3 adrenoceptor stimulation on the levels of ApoA-I, PPARα, and PPARγ in apolipoprotein E-deficient mice. J Cardiovasc Pharmacol. 2014;64:40711. [DOI] [PubMed]
    Yao Y, Yang D, Han Y, Wang W, Wang N, Yang J, et al. Dopamine D1-Like Receptors Suppress the Proliferation of Macrophages Induced by Ox-LDL. Cell Physiol Biochem. 2016;38:41526. [DOI] [PubMed]
    Manzella N, Santin Y, Maggiorani D, Martini H, Douin-Echinard V, Passos JF, et al. Monoamine oxidase-A is a novel driver of stress-induced premature senescence through inhibition of parkin-mediated mitophagy. Aging Cell. 2018;17:e12811. [DOI] [PubMed] [PMC]
    Wang Z, Li Y, Guo Y. β3-Adrenoceptor activation attenuates atherosclerotic plaque formation in ApoE−/− mice through lowering blood lipids and glucose. Acta Pharmacol Sin. 2013;34:115663. [DOI] [PubMed] [PMC]
    Li Z, Li Q, Wang L, Li C, Xu M, Duan Y, et al. Targeting mitochondria-inflammation circle by renal denervation reduces atheroprone endothelial phenotypes and atherosclerosis. Redox Biol. 2021;47:102156. [DOI] [PubMed] [PMC]
    Santanam N, Thornhill BA, Lau JK, Crabtree CM, Cook CR, Brown KC, et al. Nicotinic acetylcholine receptor signaling in atherogenesis. Atherosclerosis. 2012;225:26473. [DOI] [PubMed]
    Hewing B, Ludwig A, Dan C, Pötzsch M, Hannemann C, Petry A, et al. Immunoproteasome subunit ß5i/LMP7-deficiency in atherosclerosis. Sci Rep. 2017;7:13342. [DOI] [PubMed] [PMC]
    Ask TF, Lugo RG, Sütterlin S. The Neuro-Immuno-Senescence Integrative Model (NISIM) on the Negative Association Between Parasympathetic Activity and Cellular Senescence. Front Neurosci. 2018;12:726. [DOI] [PubMed] [PMC]
    Wang K, Liu H, Hu Q, Wang L, Liu J, Zheng Z, et al. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct Target Ther. 2022;7:374. [DOI] [PubMed] [PMC]
    Sánchez-Cabo F, Fuster V, Silla-Castro JC, González G, Lorenzo-Vivas E, Alvarez R, et al. Subclinical atherosclerosis and accelerated epigenetic age mediated by inflammation: a multi-omics study. Eur Heart J. 2023;44:2698709. [DOI] [PubMed] [PMC]
    Hiltunen MO, Turunen MP, Häkkinen TP, Rutanen J, Hedman M, Mäkinen K, et al. DNA hypomethylation and methyltransferase expression in atherosclerotic lesions. Vasc Med. 2002;7:511. [DOI] [PubMed]
    Jia L, Zhu L, Wang JZ, Wang XJ, Chen JZ, Song L, et al. Methylation of FOXP3 in regulatory T cells is related to the severity of coronary artery disease. Atherosclerosis. 2013;228:34652. [DOI] [PubMed]
    Valencia-Morales MdP, Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, et al. The DNA methylation drift of the atherosclerotic aorta increases with lesion progression. BMC Med Genomics. 2015;8:7. [DOI] [PubMed] [PMC]
    Jiang Y, Jiménez JM, Ou K, McCormick ME, Zhang L, Davies PF. Hemodynamic disturbed flow induces differential DNA methylation of endothelial Kruppel-Like Factor 4 promoter in vitro and in vivo. Circ Res. 2014;115:3243. [DOI] [PubMed] [PMC]
    Guay S, Brisson D, Munger J, Lamarche B, Gaudet D, Bouchard L. ABCA1 gene promoter DNA methylation is associated with HDL particle profile and coronary artery disease in familial hypercholesterolemia. Epigenetics. 2012;7:46472. [DOI] [PubMed]
    Zhang Y, Mei J, Li J, Zhang Y, Zhou Q, Xu F. DNA Methylation in Atherosclerosis: A New Perspective. Evid Based Complement Alternat Med. 2021;2021:6623657. [DOI] [PubMed] [PMC]
    Lacey M, Baribault C, Ehrlich KC, Ehrlich M. Atherosclerosis-associated differentially methylated regions can reflect the disease phenotype and are often at enhancers. Atherosclerosis. 2019;280:18391. [DOI] [PubMed] [PMC]
    Zhang Q, Zhao K, Shen Q, Han Y, Gu Y, Li X, et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature. 2015;525:38993. [DOI] [PubMed] [PMC]
    Liu Y, Peng W, Qu K, Lin X, Zeng Z, Chen J, et al. TET2: A Novel Epigenetic Regulator and Potential Intervention Target for Atherosclerosis. DNA Cell Biol. 2018;37:51723. [DOI] [PubMed] [PMC]
    Jiang L, Yu X, Chen J, Hu M, Zhang Y, Lin H, et al. Histone Deacetylase 3: A Potential Therapeutic Target for Atherosclerosis. Aging Dis. 2022;13:77386. [DOI] [PubMed] [PMC]
    Das S, Natarajan R. HDAC9: An Inflammatory Link in Atherosclerosis. Circ Res. 2020;127:8246. [DOI] [PubMed] [PMC]
    Zheng X, Zhou T, Wang X, Tong X, Ding J. Histone deacetylases and atherosclerosis. Atherosclerosis. 2015;240:35566. [DOI] [PubMed]
    Wang L, Ahn YJ, Asmis R. Inhibition of myeloid HDAC2 upregulates glutaredoxin 1 expression, improves protein thiol redox state and protects against high-calorie diet-induced monocyte dysfunction and atherosclerosis. Atherosclerosis. 2021;328:2332. [DOI] [PubMed]
    Zhou B, Margariti A, Zeng L, Habi O, Xiao Q, Martin D, et al. Splicing of histone deacetylase 7 modulates smooth muscle cell proliferation and neointima formation through nuclear β-catenin translocation. Arterioscler Thromb Vasc Biol. 2011;31:267684. [DOI] [PubMed]
    Wang F, Liu Z, Park S, Gwag T, Lu W, Ma M, et al. Myeloid β-Catenin Deficiency Exacerbates Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient Mice. Arterioscler Thromb Vasc Biol. 2018;38:146878. [DOI] [PubMed] [PMC]
    Chen H, Xie C, Chen Q, Zhuang S. HDAC11, an emerging therapeutic target for metabolic disorders. Front Endocrinol (Lausanne). 2022;13:989305. [DOI] [PubMed] [PMC]
    Sosnowska B, Mazidi M, Penson P, Gluba-Brzózka A, Rysz J, Banach M. The sirtuin family members SIRT1, SIRT3 and SIRT6: Their role in vascular biology and atherogenesis. Atherosclerosis. 2017;265:27582. [DOI] [PubMed]
    Gong H, Liu J, Xue Z, Wang W, Li C, Xu F, et al. SIRT3 attenuates coronary atherosclerosis in diabetic patients by regulating endothelial cell function. J Clin Lab Anal. 2022;36:e24586. [DOI] [PubMed] [PMC]
    Zierold S, Buschmann K, Gachkar S, Bochenek ML, Velmeden D, Hobohm L, et al. Brain-Derived Neurotrophic Factor Expression and Signaling in Different Perivascular Adipose Tissue Depots of Patients With Coronary Artery Disease. J Am Heart Assoc. 2021;10:e018322. [DOI] [PubMed] [PMC]
    Zhang Z, Salisbury D, Sallam T. Long Noncoding RNAs in Atherosclerosis: JACC Review Topic of the Week. J Am Coll Cardiol. 2018;72:238090. [DOI] [PubMed] [PMC]
    Rezaei M, Mokhtari MJ, Bayat M, Safari A, Dianatpuor M, Tabrizi R, et al. Long non-coding RNA H19 expression and functional polymorphism rs217727 are linked to increased ischemic stroke risk. BMC Neurol. 2021;21:54. [DOI] [PubMed] [PMC]
    Kang C, Hu Y, Nie Y, Zhao J, Li S, Chu S, et al. Long non-coding RNA RP5-833A20.1 inhibits proliferation, metastasis and cell cycle progression by suppressing the expression of NFIA in U251 cells. Mol Med Rep. 2016;14:528896. [DOI] [PubMed]
    Hu Y, Yang J, Ma X, Chen Z, Hu Y, Zhao J, et al. A lincRNA-DYNLRB2-2/GPR119/GLP-1R/ABCA1-dependent signal transduction pathway is essential for the regulation of cholesterol homeostasis. J Lipid Res. 2014;55:68197. [DOI] [PubMed] [PMC]
    Ballantyne MD, Pinel K, Dakin R, Vesey AT, Diver L, Mackenzie R, et al. Smooth Muscle Enriched Long Noncoding RNA (SMILR) Regulates Cell Proliferation. Circulation. 2016;133:205065. [DOI] [PubMed] [PMC]
    Li H, Pan Z, Chen Q, Yang Z, Zhang D. SMILR Aggravates the Progression of Atherosclerosis by Sponging miR-10b-3p to Regulate KLF5 Expression. Inflammation. 2020;43:162033. [DOI] [PubMed]
    Wang H, He F, Liang B, Jing Y, Zhang P, Liu W, et al. LincRNA-p21 alleviates atherosclerosis progression through regulating the miR-221/SIRT1/Pcsk9 axis. J Cell Mol Med. 2021;25:914153. [DOI] [PubMed] [PMC]
    Shi Z, Zheng Z, Lin X, Ma H. Long Noncoding RNA MALAT1 Regulates the Progression of Atherosclerosis by miR-330-5p/NF-κB Signal Pathway. J Cardiovasc Pharmacol. 2021;78:23546. [DOI] [PubMed]
    Li H, Liu X, Sun N, Wang T, Zhu J, Yang S, et al. Differentially Expressed Circular Non-coding RNAs in Atherosclerotic Aortic Vessels and Their Potential Functions in Endothelial Injury. Front Cardiovasc Med. 2021;8:657544. [DOI] [PubMed] [PMC]
    Tong K, Tan K, Lim Y, Tien X, Wong P. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem. 2022;477:270333. [DOI] [PubMed]
    Madrigal-Matute J, Cuervo AM, Sluimer JC. Chaperone-mediated autophagy protects against atherosclerosis. Autophagy. 2022;18:25057. [DOI] [PubMed] [PMC]
    Martinet W, Meyer GRYD. Autophagy in atherosclerosis: a cell survival and death phenomenon with therapeutic potential. Circ Res. 2009;104:30417. [DOI] [PubMed]
    Salminen A, Ojala J, Kaarniranta K, Kauppinen A. Mitochondrial dysfunction and oxidative stress activate inflammasomes: impact on the aging process and age-related diseases. Cell Mol Life Sci. 2012;69:29993013. [DOI] [PubMed] [PMC]
    Chattopadhyay A, Kwartler CS, Kaw K, Li Y, Kaw A, Chen J, et al. Cholesterol-Induced Phenotypic Modulation of Smooth Muscle Cells to Macrophage/Fibroblast-like Cells Is Driven by an Unfolded Protein Response. Arterioscler Thromb Vasc Biol. 2021;41:30216. [DOI] [PubMed] [PMC]
    Qiao L, Ma J, Zhang Z, Sui W, Zhai C, Xu D, et al. Deficient Chaperone-Mediated Autophagy Promotes Inflammation and Atherosclerosis. Circ Res. 2021;129:114157. [DOI] [PubMed] [PMC]
    Marfella R, D’Amico M, Filippo CD, Baldi A, Siniscalchi M, Sasso FC, et al. Increased activity of the ubiquitin-proteasome system in patients with symptomatic carotid disease is associated with enhanced inflammation and may destabilize the atherosclerotic plaque: effects of rosiglitazone treatment. J Am Coll Cardiol. 2006;47:244455. [DOI] [PubMed]
    Wang F, Lerman A, Herrmann J. Dysfunction of the ubiquitin-proteasome system in atherosclerotic cardiovascular disease. Am J Cardiovasc Dis. 2015;5:83100. [PubMed] [PMC]
    Paul A, Chang BH, Li L, Yechoor VK, Chan L. Deficiency of adipose differentiation-related protein impairs foam cell formation and protects against atherosclerosis. Circ Res. 2008;102:1492501. [DOI] [PubMed] [PMC]
    Wang B, Tang X, Yao L, Wang Y, Chen Z, Li M, et al. Disruption of USP9X in macrophages promotes foam cell formation and atherosclerosis. J Clin Invest. 2022;132:e154217. [DOI] [PubMed] [PMC]
    Zhang F, Xia X, Chai R, Xu R, Xu Q, Liu M, et al. Inhibition of USP14 suppresses the formation of foam cell by promoting CD36 degradation. J Cell Mol Med. 2020;24:3292302. [DOI] [PubMed] [PMC]
    Wick G, Jakic B, Buszko M, Wick MC, Grundtman C. The role of heat shock proteins in atherosclerosis. Nat Rev Cardiol. 2014;11:51629. [DOI] [PubMed]
    Kilic A, Mandal K. Heat shock proteins: pathogenic role in atherosclerosis and potential therapeutic implications. Autoimmune Dis. 2012;2012:502813. [DOI] [PubMed] [PMC]
    Xu Q, Metzler B, Jahangiri M, Mandal K. Molecular chaperones and heat shock proteins in atherosclerosis. Am J Physiol Heart Circ Physiol. 2012;302:H50614. [DOI] [PubMed] [PMC]
    Lu X, Kakkar V. The role of heat shock protein (HSP) in atherosclerosis: Pathophysiology and clinical opportunities. Curr Med Chem. 2010;17:95773. [DOI] [PubMed]
    Zhao Z, Zhang M, Chen L, Gong D, Xia X, Yu X, et al. Heat shock protein 70 accelerates atherosclerosis by downregulating the expression of ABCA1 and ABCG1 through the JNK/Elk-1 pathway. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863:80622. [DOI] [PubMed]
    Qi S, Yi G, Yu K, Feng C, Deng S. The Role of HSP90 Inhibitors in the Treatment of Cardiovascular Diseases. Cells. 2022;11:3444. [DOI] [PubMed] [PMC]
    Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:12141. [DOI] [PubMed] [PMC]
    Gatarek P, Kaluzna-Czaplinska J. Trimethylamine N-oxide (TMAO) in human health. EXCLI J. 2021;20:30119. [DOI] [PubMed] [PMC]
    Videja M, Sevostjanovs E, Upmale-Engela S, Liepinsh E, Konrade I, Dambrova M. Fasting-Mimicking Diet Reduces Trimethylamine N-Oxide Levels and Improves Serum Biochemical Parameters in Healthy Volunteers. Nutrients. 2022;14:1093. [DOI] [PubMed] [PMC]
    Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20:112667. [DOI] [PubMed] [PMC]
    Zhang L, Yu F, Xia J. Trimethylamine N-oxide: role in cell senescence and age-related diseases. Eur J Nutr. 2023;62:52541. [DOI] [PubMed]
    Zhu X, Zhao L, Wang Y, Hu X, Zhu Y, Yang X. Dietary titanium dioxide particles (E171) promote diet-induced atherosclerosis through reprogramming gut microbiota-mediated choline metabolism in APOE-/- mice. J Hazard Mater. 2022;436:129179. [DOI] [PubMed]
    Traughber CA, Iacano AJ, Neupane K, Khan MR, Opoku E, Nunn T, et al. Impavido attenuates inflammation, reduces atherosclerosis, and alters gut microbiota in hyperlipidemic mice. iScience. 2023;26:106453. [DOI] [PubMed] [PMC]
    Nicese MN, Bijkerk R, Zonneveld AJV, Berg BMVd, Rotmans JI. Sodium Butyrate as Key Regulator of Mitochondrial Function and Barrier Integrity of Human Glomerular Endothelial Cells. Int J Mol Sci. 2023;24:13090. [DOI] [PubMed] [PMC]
    Gui D, Luo W, Yan B, Ren Z, Tang Z, Liu L, et al. Effects of gut microbiota on atherosclerosis through hydrogen sulfide. Eur J Pharmacol. 2021;896:173916. [DOI] [PubMed]
    Zhang H, Bai Z, Zhu L, Liang Y, Fan X, Li J, et al. Hydrogen sulfide donors: Therapeutic potential in anti-atherosclerosis. Eur J Med Chem. 2020;205:112665. [DOI] [PubMed]
    Mani S, Untereiner A, Wu L, Wang R. Hydrogen sulfide and the pathogenesis of atherosclerosis. Antioxid Redox Signal. 2014;20:80517. [DOI] [PubMed]
    Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol. 2023;20:10925. [DOI] [PubMed] [PMC]
    Huang S, Li H, Ge J. A cardioprotective insight of the cystathionine γ-lyase/hydrogen sulfide pathway. Int J Cardiol Heart Vasc. 2015;7:517. [DOI] [PubMed] [PMC]
    Merz T, Wepler M, Nußbaum B, Vogt J, Calzia E, Wang R, et al. Cystathionine-γ-lyase expression is associated with mitochondrial respiration during sepsis-induced acute kidney injury in swine with atherosclerosis. Intensive Care Med Exp. 2018;6:43. [DOI] [PubMed] [PMC]
    Rao SP, Dobariya P, Bellamkonda H, More SS. Role of 3-Mercaptopyruvate Sulfurtransferase (3-MST) in Physiology and Disease. Antioxidants (Basel). 2023;12:603. [DOI] [PubMed] [PMC]
    Bittencourt MS, Cerci RJ. Statin effects on atherosclerotic plaques: regression or healing? BMC Med. 2015;13:260. [DOI] [PubMed] [PMC]
    Luan Y, Liu H, Luan Y, Yang Y, Yang J, Ren K. New Insight in HDACs: Potential Therapeutic Targets for the Treatment of Atherosclerosis. Front Pharmacol. 2022;13:863677. [DOI] [PubMed] [PMC]
    Neele AE, Willemsen L, Chen H, Dzobo KE, Winther MPJd. Targeting epigenetics as atherosclerosis treatment: an updated view. Curr Opin Lipidol. 2020;31:32430. [DOI] [PubMed] [PMC]
    Asare Y, Campbell-James TA, Bokov Y, Yu LL, Prestel M, Bounkari OE, et al. Histone Deacetylase 9 Activates IKK to Regulate Atherosclerotic Plaque Vulnerability. Circ Res. 2020;127:81123. [DOI] [PubMed]
    Chen L, Shang C, Wang B, Wang G, Jin Z, Yao F, et al. HDAC3 inhibitor suppresses endothelial-to-mesenchymal transition via modulating inflammatory response in atherosclerosis. Biochem Pharmacol. 2021;192:114716. [DOI] [PubMed]
    Lecce L, Xu Y, V’Gangula B, Chandel N, Pothula V, Caudrillier A, et al. Histone deacetylase 9 promotes endothelial-mesenchymal transition and an unfavorable atherosclerotic plaque phenotype. J Clin Invest. 2021;131:e131178. [DOI] [PubMed] [PMC]
    Brookes RL, Crichton S, Wolfe CDA, Yi Q, Li L, Hankey GJ, et al. Sodium Valproate, a Histone Deacetylase Inhibitor, Is Associated With Reduced Stroke Risk After Previous Ischemic Stroke or Transient Ischemic Attack. Stroke. 2018;49:5461. [DOI] [PubMed] [PMC]
    Fossel M, Bean J, Khera N, Kolonin MG. A Unified Model of Age-Related Cardiovascular Disease. Biology (Basel). 2022;11:1768. [DOI] [PubMed] [PMC]
    Shah A, Gray K, Figg N, Finigan A, Starks L, Bennett M. Defective Base Excision Repair of Oxidative DNA Damage in Vascular Smooth Muscle Cells Promotes Atherosclerosis. Circulation. 2018;138:144662. [DOI] [PubMed] [PMC]
    Fernandez ML, Thomas MS, Lemos BS, DiMarco DM, Missimer A, Melough M, et al. TA-65, A Telomerase Activator improves Cardiovascular Markers in Patients with Metabolic Syndrome. Curr Pharm Des. 2018;24:190511. [DOI] [PubMed]
    Li Y, Li S, Lin C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors. 2018;44:6982. [DOI] [PubMed]
    Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, et al. CCR5+T-bet+FoxP3+ Effector CD4 T Cells Drive Atherosclerosis. Circ Res. 2016;118:154052. [DOI] [PubMed] [PMC]
    Brane AC, Tollefsbol TO. Targeting Telomeres and Telomerase: Studies in Aging and Disease Utilizing CRISPR/Cas9 Technology. Cells. 2019;8:186. [DOI] [PubMed] [PMC]
    Xu C, Zhang X, Yang W, Gao S, Zhao N, Li P, et al. Effective prevention of atherosclerosis by non-viral delivery of CRISPR/Cas9. Nano Today. 2024;54:102097. [DOI]
    Jie Z, Xia H, Zhong S, Feng Q, Li S, Liang S, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 2017;8:845. [DOI] [PubMed] [PMC]
    Constantino-Jonapa LA, Espinoza-Palacios Y, Escalona-Montaño AR, Hernández-Ruiz P, Amezcua-Guerra LM, Amedei A, et al. Contribution of Trimethylamine N-Oxide (TMAO) to Chronic Inflammatory and Degenerative Diseases. Biomedicines. 2023;11:431. [DOI] [PubMed] [PMC]
    Sun Y, Wang X, Liu T, Zhu X, Pan X. The multifaceted role of the SASP in atherosclerosis: from mechanisms to therapeutic opportunities. Cell Biosci. 2022;12:74. [DOI] [PubMed] [PMC]
    Blagosklonny MV. Paradoxes of senolytics. Aging (Albany NY). 2018;10:428993. [DOI] [PubMed] [PMC]
    Albosta MS, Grant JK, Taub P, Blumenthal RS, Martin SS, Michos ED. Inclisiran: A New Strategy for LDL-C Lowering and Prevention of Atherosclerotic Cardiovascular Disease. Vasc Health Risk Manag. 2023;19:42131. [DOI] [PubMed] [PMC]
    Yurtseven E, Ural D, Baysal K, Tokgözoğlu L. An Update on the Role of PCSK9 in Atherosclerosis. J Atheroscler Thromb. 2020;27:90918. [DOI] [PubMed] [PMC]
    Fitzgerald K, White S, Borodovsky A, Bettencourt BR, Strahs A, Clausen V, et al. A Highly Durable RNAi Therapeutic Inhibitor of PCSK9. N Engl J Med. 2017;376:4151. [DOI] [PubMed] [PMC]
    Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, et al. Angiotensin-converting enzyme 2 and angiotensin 1–7: novel therapeutic targets. Nat Rev Cardiol. 2014;11:41326. [DOI] [PubMed] [PMC]
    Badae NM, Naggar ASE, Sayed SME. Is the cardioprotective effect of the ACE2 activator diminazene aceturate more potent than the ACE inhibitor enalapril on acute myocardial infarction in rats? Can J Physiol Pharmacol. 2019;97:63846. [DOI] [PubMed]
    Wahid A, Chen W, Wang X, Tang X. High-mobility group box 1 serves as an inflammation driver of cardiovascular disease. Biomed Pharmacother. 2021;139:111555. [DOI] [PubMed]
    Xu H, Xiang Q, Li S, Liu Y. High serum Bhlhe40 levels are associated with subclinical atherosclerosis in patients with type 2 diabetes mellitus: A cross-sectional study. Diab Vasc Dis Res. 2023;20:14791641231169246. [DOI] [PubMed] [PMC]
    Zhang Y, He Y, Liu S, Deng L, Zuo Y, Huang K, et al. SGLT2 Inhibitors in Aging-Related Cardiovascular Disease: A Review of Potential Mechanisms. Am J Cardiovasc Drugs. 2023;23:64162. [DOI] [PubMed]
    Posma JJN, Sluimer JC. Pleiotropic, Cellular Effects of Rivaroxaban on Autophagy Explain Atheroprotective Effects. JACC Basic Transl Sci. 2021;6:9813. [DOI] [PubMed] [PMC]
    Perrotta P, Emini Veseli B, Van der Veken B, Roth L, Martinet W, De Meyer GRY. Pharmacological strategies to inhibit intra-plaque angiogenesis in atherosclerosis. Vascul Pharmacol. 2019;112:728. [DOI] [PubMed]
    Theelen TL, Lappalainen JP, Sluimer JC, Gurzeler E, Cleutjens JP, Gijbels MJ, et al. Angiopoietin-2 blocking antibodies reduce early atherosclerotic plaque development in mice. Atherosclerosis. 2015;241:297304. [DOI] [PubMed] [PMC]
    Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov. 2017;16:63561. [DOI] [PubMed]
    Tikka A, Jauhiainen M. The role of ANGPTL3 in controlling lipoprotein metabolism. Endocrine. 2016;52:18793. [DOI] [PubMed] [PMC]
    Carnevale R, Nocella C, Schiavon S, Cammisotto V, Cotugno M, Forte M, et al. Beneficial effects of a combination of natural product activators of autophagy on endothelial cells and platelets. Br J Pharmacol. 2021;178:214659. [DOI] [PubMed]
    Akter R, Afrose A, Rahman MR, Chowdhury R, Nirzhor SSR, Khan RI, et al. A Comprehensive Analysis into the Therapeutic Application of Natural Products as SIRT6 Modulators in Alzheimer’s Disease, Aging, Cancer, Inflammation, and Diabetes. Int J Mol Sci. 2021;22:4180. [DOI] [PubMed] [PMC]