Next Article in Journal
New Insights into Potential Anti-Aging and Fatigue Effects of a Dietary Supplement from the Resveratrol Beverage in Aged SAMP8 Mice
Previous Article in Journal
Sesame Meal: The Ideal Alternative to Soybean Meal for Fattening Beef Cattle—Reducing Nitrogen Excretion and Bolstering Antioxidant Defenses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis

Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(11), 1338; https://doi.org/10.3390/antiox14111338
Submission received: 28 September 2025 / Revised: 2 November 2025 / Accepted: 3 November 2025 / Published: 6 November 2025
(This article belongs to the Topic Oxidative Stress and Inflammation, 3rd Edition)

Abstract

The vascular endothelium serves as a critical barrier preventing the transmigration of monocytes, circulating lipoproteins, and other molecules into the subendothelial space, and plays a vital role in regulating vascular tone. A dysfunctional and inflamed endothelial layer in response to disturbed blood flow or other proatherogenic risk factors is the initiating event in the pathogenesis of atherosclerosis, suggesting the importance of an intact and properly functioning endothelium in preventing the onset and progression of this disease. Accumulated evidence demonstrates the significant role of matricellular proteins, which are non-structural and secretory extracellular matrix (ECM) proteins, in the development of atherosclerosis. These proteins exert multifaceted effects on endothelial cells (ECs) ranging from reactive oxygen species (ROS) production, endoplasmic reticulum stress, and expression of adhesion molecules to autophagy and compromised barrier function via stimulating various molecular mechanisms. Given the critical roles of these processes in EC function and atherosclerosis, a better understanding of signaling pathways governed by matricellular proteins in ECs is required to develop therapeutic strategies for suppressing or preventing atherosclerosis and related cardiovascular diseases (CVDs). This review comprehensively summarizes the existing literature on the diverse roles of matricellular proteins in regulating EC inflammation and function, and highlights their potential as viable therapeutic targets for maintaining vascular health and inhibiting the progression of atherosclerosis.

1. Introduction

Atherosclerosis is a chronic inflammatory vascular disease characterized by the buildup of cholesterol-rich plaques that progressively narrow arterial blood vessels and reduce blood flow to vital organs like the heart and brain. This pathology is a key underlying cause of various CVDs, including myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral vascular disease, primarily due to arterial occlusion by plaques or embolism from ruptured plaques [1]. In 2022, the age-adjusted mortality rate attributable to CVDs in the United States was 224.3 per 100,000 [2].
A better understanding of the pathogenesis of atherosclerosis is important for the development of effective therapies to prevent and treat this complex vascular disease. Each stage of atherogenesis is marked by distinct histological features and cellular processes. Early stages, beginning in childhood, involve the accumulation of low-density lipoprotein (LDL) cholesterol in the arterial intima, leading to endothelial inflammation and the recruitment of immune cells [3]. As the disease progresses, monocyte-derived macrophages and vascular smooth muscle cells (VSMCs) contribute to plaque formation through lipid uptake, foam cell formation, ECM accumulation, and cell death. The stability of the plaque’s fibrous cap decreases over time, increasing the risk of rupture and subsequent thrombosis, which can result in severe cardiovascular events such as myocardial infarction [4,5,6]. Understanding the underlying molecular mechanisms is essential for developing targeted strategies to intervene at various stages of atherosclerosis, thereby improving outcomes for patients at risk of cardiovascular complications. ECs play a crucial role in the development of atherosclerosis by responding to various atherogenic stimuli present in the bloodstream, including increased lipid and glucose levels, elevated blood pressure, and inflammatory molecules/cells. These stimuli influence EC phenotypes, which are implicated in various stages of atherosclerotic lesion development. Several genetic studies have demonstrated the involvement of EC dysfunction in the pathogenesis of atherosclerosis [7,8,9,10]. However, our knowledge of the exact mechanisms responsible for EC dysfunction regulating atherogenesis is far from being completely understood and requires future investigations. Earlier studies have suggested the important roles of matricellular proteins in the development of atherosclerosis [11,12]. This review summarizes various types of matricellular proteins and their contributions to endothelial dysfunction and the development of atherosclerosis.

2. Role of Vascular Endothelium in Atherogenesis

Atherosclerosis, a chronic inflammatory disease affecting large- and medium-sized arteries, involves intricate interactions among different cell types, including vascular ECs, VSMCs, macrophages, T cells, and B cells. Additionally, it is accompanied by increased levels of lipoproteins, ECM constituents, and various pro-inflammatory cytokines and chemokines [13]. Both genetic and environmental factors regulate the initiation and progression of atherosclerosis. These factors include dietary patterns, adiposity, tobacco use, diabetes, hypertension, and lipidemia, which collectively regulate the expression and function of different proteins involved in lipid metabolism, inflammatory processes, oxidative stress, and immune responses. One significant determinant increasing the risk of atherosclerosis is the levels of small, dense LDL particles. These particles possess heightened atherogenic properties compared to larger, more buoyant LDL counterparts, due to increased arterial infiltration and entrapment, elevated susceptibility to oxidative and glycation modifications, aggregation tendency, reduced affinity for LDL receptors, and higher capacity to induce pro-inflammatory and prothrombotic events [14].
Vascular ECs are situated at the critical interface between semi-solid tissues and circulating blood, and play pivotal roles both in physiological and pathophysiological processes, particularly in the development of atherosclerosis. These roles include—(a) regulating the entry and retention of lipoproteins and leukocytes into the subendothelial space of the arterial wall, which are the main components of plaque [15], (b) producing and secreting various factors such as nitric oxide, cytokines, chemokines, and adhesion molecules, which regulate vascular tone, inflammation, oxidative stress, and coagulation [16,17,18,19,20,21], (c) changing their proliferation rate, turnover, metabolism, permeability, and plasticity, which can alter the structure and function of the vessel wall [22], (d) interacting with other cell types, such as VSMCs, macrophages, monocytes, T cells, and B cells, to modulate the immune response and the stability of the plaque [13,23], (e) contributing to the resolution of inflammation, thereby preventing or limiting the complications related to plaque formation.
Vascular EC inflammation refers to a condition where the endothelium layer of blood vessels becomes damaged or dysfunctional, potentially leading to several vascular diseases such as atherosclerosis, hypertension, and coronary artery disease (CAD). The risk factors contributing to endothelial dysfunction include diabetes, high blood pressure, smoking, obesity, and aging. These factors can reduce the production or availability of nitric oxide (NO), a crucial molecule for maintaining vascular homeostasis, and elevate the levels of various pro-inflammatory cytokines, and expression of various immune cell adhesion molecules or lipoprotein receptors on the surface of ECs [24,25,26,27]. Here, we provide a brief overview of various types of EC inflammation and the signaling pathways. These aspects have been comprehensively reviewed in previous publications [7,10,26].

2.1. Endothelial Activation

This type of EC inflammation occurs in response to various pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and IL-6, or endotoxins like lipopolysaccharide (LPS). Endothelial activation leads to an upregulation in the expression of adhesion molecules, including E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1), and increased secretion of pro-inflammatory chemokines/cytokines, which compromise endothelial barrier function, resulting in increased vascular permeability (Figure 1). This facilitates the recruitment, adhesion, and transmigration of leukocytes across the endothelial layer [28]. Furthermore, endothelial activation induces the expression of pro-coagulant factors, such as tissue factor and von Willebrand factor, while concurrently downregulating anti-coagulant factors, such as thrombomodulin and prostacyclin. This imbalance increases the risk of thrombus formation [29]. Endothelial activation plays an important role in the pathogenesis of various inflammatory diseases, such as atherosclerosis, sepsis, and rheumatoid arthritis [30,31]. The major signaling pathways involved in endothelial activation include nuclear factor-kappa B (NF-κB), MAPK, and PI3K/Akt pathways [32,33,34] (Figure 1 and Table 1).
Table 1. Major markers of EC inflammation.
Table 1. Major markers of EC inflammation.
Name of the MarkerFunctionRefs.
C-reactive protein (CRP)CRP is produced by the liver in response to inflammation. CRP binds to damaged ECs and exacerbates inflammation and oxidative stress. In addition, it reduces NO production, increases endothelin-1 synthesis, and suppresses endothelium-dependent arterial relaxation.[35,36]
IL-6IL-6 is a pro-inflammatory cytokine secreted by ECs and other cells during inflammation. It plays an important role in upregulating the production of CRP and other inflammatory mediators. It also promotes the expression of chemokines and adhesion molecules on ECs, which aids in leukocyte recruitment.[35,37]
VCAM-1VCAM-1, an adhesion molecule, binds to integrins found on leukocytes, facilitating their attachment and transmigration across the endothelium. VCAM-1 expression on ECs increases in response to various inflammatory stimuli.[38,39]
E-selectinIt interacts with sialylated glycoproteins on leukocytes, enabling leukocyte rolling and initial attachment to the endothelium.[35,40]
ICAM-1Similar to VCAM-1, it helps in leukocyte attachment and transmigration across the endothelium.[35,41]
Endothelin-1It is a potent vasoconstrictor and pro-inflammatory agent synthesized by ECs. It can stimulate the expression of adhesion molecules and chemokines on ECs. Additionally, it activates the NF-κB pathway, a key regulator of inflammation.[35,42]
MCP-1/CCL2ECs and VSMCs secrete MCP-1, which plays a crucial role in attracting monocytes and macrophages to the subendothelial cell layer. This recruitment process facilitates the accumulation of lipids within these immune cells, ultimately contributing to the formation of atherosclerotic lesions.[35,43,44]
TNF-αTNF-α, originally identified for its anti-tumor properties, is a key pro-inflammatory cytokine associated with various CVDs. TNF-α contributes to endothelial dysfunction via promoting oxidative stress and reducing NO production, thus impairing endothelium-dependent vasodilation across different vascular beds.[35,45]
Interleukin-1 β (IL-1β)IL-1β promotes the development of early atherosclerotic lesions by enhancing the adherence of monocytes to ECs through the elevation of adhesion molecules.[35,46]
Transforming growth factor β (TGF-β)TGF-β induces the expression of several pro-inflammatory chemokines, cytokines, their receptors, and adhesion molecules on ECs. Moreover, it stimulates the expression of matrix metalloproteinases and fibronectin (FN), which are closely associated with inflammation.[47]
Interleukin-18 (IL-18)IL-18 belongs to the IL-1 cytokine family and was originally found in macrophages and Kupffer cells. IL-18 triggers IFN-γ production by T cells.[35]
CD40/CD40LCD40L, a TNF family member, and its receptor CD40 are co-expressed in activated T lymphocytes, ECs, SMCs, and macrophages in atherosclerotic lesions.[35]
Interleukin-8 (IL-8)IL-8 is a pro-inflammatory cytokine that plays a significant role in EC inflammation, particularly in the context of CVDs. IL-8 functions through its receptors, CXCR1 and CXCR2, present on circulating immune cells, particularly neutrophils, leading to their chemotaxis and adhesion to the EC surface. IL-8 also plays a role in promoting ICAM-1 and VCAM-1 expression on ECs.[48,49]
CXCL12CXCL12, also known as stromal cell-derived factor 1 (SDF-1), is a chemokine that plays a crucial role in vascular development, tissue repair, and inflammation. In ECs, CXCL12 is involved in various processes: EC migration, inflammation, and angiogenesis.[50]
Figure 1. Key signaling pathways governed by ROS and/or matricellular proteins in EC inflammation: (A) MAPK/p38 pathway: The activation of the Mitogen-Activated Protein Kinase (MAPK) pathway, comprising ERK, JNK, and p38 subfamilies, stimulates the expression of various molecules, including IL6, IL1β, TNFα, p53, ELK1, ATF2, MYC, VCAM1, ICAM1, and E-selectin, in ECs [51,52]. ERK promotes cell survival, proliferation, and migration JNK induces cell death and inflammation, and p38 contributes to inflammation and apoptosis [53]. Crosstalk between these subfamilies regulates cellular responses, affecting processes critical to atherosclerosis, such as proliferation, differentiation, migration, and apoptosis. (B) NF-κB pathway and NLRP3 inflammasome: The NF-κB pathway regulates the expression of inflammatory genes in ECs. ROS-induced activation of IκB kinase (IKK) leads to the degradation of the inhibitory protein IκB, allowing nuclear translocation of NF-κB (p50/p65 heterodimer). Once in the nucleus, p50/p65 heterodimer activates the transcription of pro-inflammatory genes IL-6, IL-12, and TNFα, and adhesion molecules (VCAM1, ICAM1, and E-selectin) [54,55]. NLRP3 inflammasome, which senses ROS and danger signals, contributes to inflammatory responses in ECs. ROS or other stimuli activate NLRP3, forming a complex with the adaptor protein ASC and pro-caspase-1. Caspase-1 activation cleaves pro-IL1β and pro-IL18 into their active forms, inducing EC pyroptosis (a form of inflammatory cell death). This process amplifies inflammation within atherosclerotic arteries, potentially influencing plaque stability and progression [56,57,58]. (C) Nrf2 pathway: Counteracting the pro-inflammatory milieu, Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) pathway functions as a major regulator of antioxidant gene expression. ROS-triggered modification of Kelch-like ECH-associated protein 1 (Keap1) leads to the release and stabilization of Nrf2 [59]. Nuclear translocation of Nrf2 activates antioxidant response element (ARE)-mediated transcription of antioxidant genes such as heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO1), and superoxide dismutase (SOD) [60]. This antioxidative arm plays a crucial role in protecting ECs against oxidative stress, a hallmark of atherosclerosis [61]. Beyond these pathways, molecules such as Vascular Endothelial Growth Factors (VEGF) [62], Transforming Growth Factor-beta (TGFβ) [63], IL6 [37], and Platelet-Derived Growth Factor (PDGF) activate various signaling cascades and regulate angiogenesis, vascular remodeling, and inflammation, further shaping the atherosclerotic landscape [64].
Figure 1. Key signaling pathways governed by ROS and/or matricellular proteins in EC inflammation: (A) MAPK/p38 pathway: The activation of the Mitogen-Activated Protein Kinase (MAPK) pathway, comprising ERK, JNK, and p38 subfamilies, stimulates the expression of various molecules, including IL6, IL1β, TNFα, p53, ELK1, ATF2, MYC, VCAM1, ICAM1, and E-selectin, in ECs [51,52]. ERK promotes cell survival, proliferation, and migration JNK induces cell death and inflammation, and p38 contributes to inflammation and apoptosis [53]. Crosstalk between these subfamilies regulates cellular responses, affecting processes critical to atherosclerosis, such as proliferation, differentiation, migration, and apoptosis. (B) NF-κB pathway and NLRP3 inflammasome: The NF-κB pathway regulates the expression of inflammatory genes in ECs. ROS-induced activation of IκB kinase (IKK) leads to the degradation of the inhibitory protein IκB, allowing nuclear translocation of NF-κB (p50/p65 heterodimer). Once in the nucleus, p50/p65 heterodimer activates the transcription of pro-inflammatory genes IL-6, IL-12, and TNFα, and adhesion molecules (VCAM1, ICAM1, and E-selectin) [54,55]. NLRP3 inflammasome, which senses ROS and danger signals, contributes to inflammatory responses in ECs. ROS or other stimuli activate NLRP3, forming a complex with the adaptor protein ASC and pro-caspase-1. Caspase-1 activation cleaves pro-IL1β and pro-IL18 into their active forms, inducing EC pyroptosis (a form of inflammatory cell death). This process amplifies inflammation within atherosclerotic arteries, potentially influencing plaque stability and progression [56,57,58]. (C) Nrf2 pathway: Counteracting the pro-inflammatory milieu, Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) pathway functions as a major regulator of antioxidant gene expression. ROS-triggered modification of Kelch-like ECH-associated protein 1 (Keap1) leads to the release and stabilization of Nrf2 [59]. Nuclear translocation of Nrf2 activates antioxidant response element (ARE)-mediated transcription of antioxidant genes such as heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO1), and superoxide dismutase (SOD) [60]. This antioxidative arm plays a crucial role in protecting ECs against oxidative stress, a hallmark of atherosclerosis [61]. Beyond these pathways, molecules such as Vascular Endothelial Growth Factors (VEGF) [62], Transforming Growth Factor-beta (TGFβ) [63], IL6 [37], and Platelet-Derived Growth Factor (PDGF) activate various signaling cascades and regulate angiogenesis, vascular remodeling, and inflammation, further shaping the atherosclerotic landscape [64].
Antioxidants 14 01338 g001

2.2. Endothelial Dysfunction

Endothelial dysfunction arises due to impairment in the vasoprotective function of the endothelium, including vasodilation, anti-inflammation, and anti-oxidation [65]. It is primarily caused by the reduced production and/or bioavailability of NO [7], a key EC-derived relaxing factor that modulates vascular tone, inhibits platelet aggregation, and suppresses leukocyte adhesion. Endothelial dysfunction is closely linked to the development and progression of CVDs—hypertension, CAD, and stroke. It represents the earliest stage of atherosclerosis, characterized by the impaired endothelium’s physiological vasoprotective function [26]. Fatty streak formation is the stage where subendothelial accumulation of lipid-laden macrophages/VSMCs known as foam cells [38] occurs, along with the recruitment of T cells and secretion of pro-inflammatory cytokines and chemokines, such as IL-1, IL-6, TNF-α, and monocyte chemoattractant protein-1 (MCP-1) [39] (Figure 1 and Table 1).
It is important to note that endothelial activation and dysfunction are distinct yet interconnected processes, both involving changes in EC behavior. Endothelial activation is typically an acute and reversible response to injury or inflammation, characterized by increased expression of adhesion molecules, cytokine release, and enhanced leukocyte recruitment. In contrast, endothelial dysfunction represents a chronic pathological state, marked by impaired vasodilation, oxidative stress, inflammation, and loss of barrier integrity [26]. Prolonged endothelial activation can lead to dysfunction, as the persistent inflammation and oxidative stress damage the endothelium and impair its physiological functions [66]. In pathologies like atherosclerosis and hypertension, the transition from endothelial activation to endothelial dysfunction is a critical event. Thus, endothelial activation is often viewed as an early and potentially reversible phase, whereas endothelial dysfunction reflects a more advanced and irreversible stage of endothelial damage.

2.3. Endothelial Senescence

Senescence is a state of permanent cell cycle arrest, where cells remain metabolically active but lose their ability to proliferate and function properly. It is characterized by an increased expression of senescence-associated markers, such as p16, p21, and p53, along with decreased expression of telomerase, which maintains the length and integrity of telomeres [67]. Further, endothelial senescence induces the secretion of pro-inflammatory and pro-fibrotic factors, contributing to chronic low-grade inflammation and vascular remodeling [68]. Accelerated by processes such as oxidative stress, DNA damage, cellular inflammation, and shear stress [67], this condition is implicated in the pathogenesis of age-related vascular diseases, such as atherosclerosis, aneurysm, and vascular calcification [67,68]. EC apoptosis and necrosis reduce endothelial integrity and promote exposure of the subendothelial matrix, which promotes plaque growth and instability. Similarly, oxidative stress, driven by the accumulation and generation of ROS, is responsible for cellular senescence, ultimately contributing to age-related endothelial dysfunction. While moderate levels of ROS play crucial roles in cellular functions, an excess of ROS induces detrimental effects, including DNA damage, which activates the p53-dependent pathway, leading to cell cycle arrest via p21 [69,70,71].

2.4. LDL Transcytosis Across the Arterial Endothelium

The LDL transport from plasma to the arterial sub-endothelial space is considered a rate-limiting step in atherogenesis and plays a critical role in determining the spatial distribution of atherosclerotic lesions. While the arterial endothelium is permeable to water and small molecules (<6 nm diameter), the transport of macromolecules, including LDL particles (22 to 28 nm), across the EC layer is tightly regulated. Due to the restrictive nature of intercellular junctions, including tight junctions and gap junctions, paracellular transport of LDL is nil to minimal [72]. Instead, LDL primarily crosses the endothelium via transcytosis, a transcellular process [73,74,75]. Emerging evidence supports the role of endothelial transcytosis in LDL transport via caveolae, flask-shaped invaginations of the plasma membrane [76]. Key proteins localized within caveolae, such as activin-like kinase 1 (ALK1) and scavenger receptor class B type 1 (SR-B1), have been shown to directly bind LDL and mediate its transcytosis. The molecular mechanisms governing this process have been recently reviewed in detail [76,77].

2.5. Endothelial-to-Mesenchymal Transition

Endothelial-to-mesenchymal transition (EndMT), a dynamic cellular phenomenon, is recognized to play important roles in several CVDs, including atherosclerosis, valvular disease, and pulmonary arterial hypertension [78,79]. During EndMT, ECs lose their intercellular junctions, degrade the basement membrane, and undergo phenotypic transformation to acquire mesenchymal characteristics [80]. This transition is driven by the induction of key EndMT-related transcription factors (Snai1, Snai2, Twist-1, Zeb1, and Zeb2), leading to the progressive loss of typical EC markers and the upregulation of mesenchymal markers vimentin, fibronectin, SM22α, etc. [79]. These phenotypic changes contribute to vascular inflammation, plaque development, and destabilization [81,82,83,84,85]. EndMT also plays a critical role in endothelial activation and dysfunction, promoting maladaptive tissue responses across various disease states [86]. Radiation therapy has been reported to induce EndMT in HAoECs, marked by increased expression of mesenchymal markers and decreased levels of endothelial markers (CD31, VE-cadherin). These changes were also observed in irradiated Apoe/ mice with elevated oxLDL, linking oxLDL-driven EndMT to radiation-induced atherosclerosis [87]. The involvement of EndMT in atherosclerosis has been comprehensively reviewed previously [80,88].

3. Mechanisms of EC Inflammation

3.1. Oxidative Stress in EC Inflammation

The imbalance between the ROS production and their scavenging is called oxidative stress [89], which promotes EC inflammation and dysfunction. This section briefly covers different types of ROS, reactive nitrogen species (RNS), their sources, and roles in endothelial health.

3.1.1. Reactive Oxygen Species and Their Role in EC Inflammation

ROS comprise a group of molecules derived from molecular oxygen, which are highly reactive and capable of damaging cellular components and triggering inflammatory responses (Table 2). Vascular ECs produce ROS in response to various stimuli, such as shear stress, hyperglycemia, angiotensin II (Ang II), and cytokines. Multiple sources contribute to ROS production in ECs, including NADPH oxidases (NOXs), mitochondria, cytochrome P450, and xanthine oxidase.
Table 2. Common ROS and RNS molecules involved in EC inflammation [90].
Table 2. Common ROS and RNS molecules involved in EC inflammation [90].
MoleculeFunctionRefs.
Superoxide anion (O2•−)It is produced by the one-electron reduction of molecular oxygen (O2). Superoxide can activate various signaling pathways in ECs, such as MAPK, NF-κB, and nucleotide-binding domain, leucine-rich-containing family, pyrin domain–containing-3 (NLRP3), and induce the expression of pro-inflammatory cytokines, chemokines, and adhesion molecules.[90,91]
Hydrogen peroxide (H2O2)It is produced by the two-electron reduction of O2 or by the dismutation of superoxide. H2O2 can modulate the activity of various transcription factors in ECs, such as NF-κB, Nrf2, and AP-1, and regulate the expression of genes involved in inflammation, antioxidant defense, and cell death.[90,92,93]
Hydroxyl radical (OH)It is the most reactive and damaging ROS molecule, which is produced by the one-electron reduction of H2O2 or Fenton reaction. Hydroxyl radical can cause oxidative damage to various biomolecules in ECs, such as lipids, proteins, and DNA, and trigger inflammation, apoptosis, and senescence.[90,92]
NO radical (NO)NO is a free radical, which readily reacts with various molecules, particularly iron centers and oxygen. It plays critical roles in vasodilation, neuronal signaling, and microbial defense.[94]
Peroxynitrite (ONOO)It is reactive nitrogen species, produced by the reaction of superoxide with NO. Peroxynitrite can impair the function of various enzymes and proteins in ECs, such as nitric oxide synthase (NOS), cyclooxygenase (COX), and SOD, and induce inflammation, nitrosative stress, and endothelial dysfunction.[90,95]
Among these, flavocytochrome enzymes NOXs are the major ROS sources in ECs [96,97]. These enzymes catalyze the transfer of electrons from cytosolic NADPH to molecular oxygen, producing superoxide ion (O2•−) and hydrogen peroxide (H2O2). NOX-derived ROS serve several physiological roles, including host defense, biosynthesis, cellular signal transduction, and vascular function [98,99]. Mitochondria also produce ROS, but they are not the main source in ECs, as mitochondrial oxidative phosphorylation contributes minimally to ATP production in these cells [100,101]. Additional ROS sources in ECs include lysosomes, peroxisomes, and endoplasmic reticulum (ER) [102,103,104,105]. Various ROS types, their cellular sources, and functions in vascular health have been comprehensively reviewed previously [106,107]. In humans, 7 NOX isoforms—NOX1, NOX2, NOX3, NOX4, NOX5, Duox1, and Doux2 have been identified, while mice and rats lack NOX5 [108]. These NOX isoforms are localized in various subcellular locations, such as the plasma membrane, perinuclear membrane, and ER [97,98,99,109]. Four members of NOX family, including NOX1, NOX2, NOX4, and NOX5, are important sources of ROS in the vasculature [110]. NOX1, NOX2, and NOX5 produce O2•− in response to stimuli like shear stress, growth factors, and inflammatory cytokines, while NOX4 generates H2O2. ROS derived from NOX1, NOX2, and NOX5 have been shown to induce EC apoptosis, inflammation, and endothelial dysfunction [100,111]. Additionally, NOX1- and NOX4-derived ROS contribute to cell cycle arrest in the S phase and induce EC senescence, which are the features of EC activation [106].
NOX2 produces O2•− both intracellularly and extracellularly. Intracellular O2•− is neutralized by cytoplasmic superoxide dismutase (SOD1), while extracellular O2•− is scavenged by SOD3, converting it to H2O2. Superoxide radicals produced by NOX2 in the endothelium also react with NO, reducing its bioavailability and impairing endothelium-dependent vasorelaxation [112]. In isolated aortic tissue, the extent of endothelium-dependent relaxation is inversely correlated with NOX2 expression [113]. There is a strong interplay between NOX-derived ROS and NO signaling. For instance, ECs exposed to high glucose levels display increased expression and activity of NOX2, which is negatively correlated with endothelial nitric oxide synthase (eNOS) expression and NO production [114]. Conversely, treatment with NO donors downregulates NOX2 expression and O2•− generation [115]. Further, oscillatory shear stress (OSS) has been demonstrated to induce NOX2 expression, promote O2•− generation, and stimulate monocyte adhesion [116]. Moreover, NOX2-produced ROS in human microvascular ECs (MVECs) promote cell cycle arrest and apoptosis by increasing expression of cyclin-dependent kinase inhibitor, p21Cip1 and cell cycle regulatory protein, p53 [117].
NOX4 is the most abundantly expressed NOX isoform in the cardiovascular system [118]. Its expression in ECs is regulated by mechanical stimuli—downregulated by laminar shear stress and upregulated by OSS [119,120]. NOX4 plays a key role in EC proliferation and cell cycle progression [121]. Specifically, NOX4 knockdown reduces epidermal growth factor-induced proliferation, whereas its overexpression promotes EC growth [121,122]. The role of NOX4 in apoptosis appears context-dependent. In MVECs NOX4 overexpression protects against serum starvation-induced apoptosis, while it mediates apoptosis in response to TNF-α stimulation [121,123,124]. Similarly, in various cell types and tissues, NOX4-induced senescence contributes to the development of CVDs [125,126]. Notably, NOX4 expression increases with age, particularly in the aorta, renal cortex, and medulla [113,127,128]. The functional versatility of NOX4 is highlighted by its capacity to respond to diverse stimuli and its involvement in multiple cellular processes, including differentiation [129,130], migration [131,132], proliferation [133], apoptosis [124,134], senescence [126], and inflammation [135,136]. However, further research is required to completely determine if NOX4 has similar functions in the cardiovascular system.
ROS play a pivotal role in the pathogenesis of atherosclerosis via regulating various signaling pathways in ECs. Figure 1 summarizes ROS-stimulated signaling pathways, including MAPK, NF-κB, Nrf2, and NLRP3 cascades. Several in vivo studies have reported that different NOX isoforms promote atherosclerosis [122,137,138]. NOX1 expression is increased in diabetic atherosclerotic mice [139], and its deletion in diabetic mice suppresses lesion formation, ROS generation, leukocyte adherence, and macrophage infiltration [140,141]. In contrast, EC-specific NOX2 overexpression does not affect atherosclerosis development in the aortic root or descending aorta [142]. Interestingly, NOX4 deficiency in Apoe/ mice leads to reduced H2O2 generation and increased atherosclerotic lesion formation [143]. Moreover, patients with symptomatic carotid artery stenosis exhibit reduced NOX4 mRNA and H2O2 levels, suggesting a protective role of NOX4 in plaque stabilization [144]. These results indicate that NOX4 may exert atheroprotective effects, potentially through H2O2-mediated upregulation of eNOS. The absence of NOX5 in rodents has limited investigations into its role in the development of atherosclerosis. However, studies using EC-specific NOX5 knock-in mice indicate that its expression does not promote plaque formation [145]. This information suggests the complex and isoform-specific roles of NOX-derived ROS in endothelial biology and atherosclerosis.

3.1.2. Sources of NO and Endothelial Cell Biology

NO is synthesized endogenously by three isoforms of NOS, which catalyze the NADPH-dependent oxidation of L-arginine to L-citrulline and NO. These enzymes are expressed in various vascular and immune cell types [146]. The NOS family includes two constitutive isoforms—neuronal NOS (nNOS or NOS1) and eNOS (NOS3), as well as an inducible isoform (iNOS or NOS2). NO is a freely diffusible radical that readily crosses plasma membranes. It plays essential roles in neurotransmission, immunological defense, and maintenance of vascular tone [97]. The nNOS is a calcium-dependent enzyme predominantly found in the nervous system, gastrointestinal tract, and skeletal muscle. In contrast, iNOS is primarily expressed in activated immune cells, including macrophages and neutrophils, which generate a large amount of NO during inflammatory responses [97,147,148]. NO produced by eNOS promotes vasodilation by relaxing VSMCs, thereby reducing blood pressure. NOS enzyme activity requires cofactor tetrahydrobiopterin (BH4), and loss or oxidation of BH4 to dihydrobiopterin (BH2) leads to NOS uncoupling, resulting in the production of O2•− instead of NO. Uncoupling of eNOS reduces NO bioavailability and promotes EC dysfunction. NO derived from eNOS is atheroprotective as it inhibits LDL oxidation, leukocyte adhesion, VSMC proliferation, and platelet aggregation [147,149]. In contrast, excessive NO generated by iNOS contributes to oxidative stress and atherogenesis through the formation of peroxynitrite and further depletion of BH4, which exacerbates eNOS uncoupling [150]. Interestingly, nNOS is also found in blood vessels, and may facilitate vasodilation, and exert anti-atherogenic effects [151].

3.1.3. Effects of Various Circulating Risk Factors of Atherosclerosis on ROS Generation

Circulating risk factors for atherosclerosis, including oxidized LDL (oxLDL), high blood glucose levels, elevated free fatty acids (FFAs), Ang II, and pro-inflammatory cytokines, significantly stimulate ROS production by ECs, leading to endothelial dysfunction, and atherogenesis. These risk factors activate redox-sensitive pathways and impair NO bioavailability, which is a hallmark of vascular homeostasis disruption. OxLDL binds to different scavenger receptors (SR-B1, LOX-1, and CD36) present on the cell surface and augments intracellular ROS production specifically via NOX2 and NOX4 activation [152]. Concurrently, oXLDL suppresses eNOS expression and activation, consequently reducing NO release [153]. The increased ROS generation further causes oxidative modification of lipids and proteins, upregulation of adhesion molecules (VCAM-1, ICAM-1), and recruitment of monocytes, all of which contribute to EC inflammation and dysfunction [154]. High blood glucose levels (hyperglycemia) observed in diabetic individuals, promote ROS generation by multiple signaling pathways, which stimulate NOX enzymes and mitochondrial respiratory chain complexes, resulting in elevated ROS production and reduced NO bioavailability [155]. Moreover, elevated levels of circulating free fatty acids and Ang II also induce ROS production in ECs, leading to mitochondrial dysfunction and cellular inflammation [156,157]. Similar to the other circulating risk factors of atherosclerosis, various pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β upregulate NOX expression and induce ROS production. Further, these cytokines impair endothelial integrity by disrupting endothelial tight junctions, promote leukocyte adhesion, and decrease antioxidant capacity, thus facilitating atherosclerotic plaque development [158]. In summary, circulating atherogenic stimuli synergistically induce ROS generation in ECs, primarily via NOX activation and mitochondrial dysfunction.

3.2. Disturbed Flow in EC Inflammation and Atherosclerosis

The EC layer of blood vessels is exposed to varying magnitudes and patterns of shear stress exerted by blood flow, depending on blood viscosity and the vascular geometry. Shear stress alters EC structure and function, including cell orientation, cytoskeletal reorganization, permeability to macromolecules, and leukocyte attachment [159]. Unidirectional laminar blood flow, typically found in the straight region of arteries, is atheroprotective and shown to stimulate the secretion of potent vasodilators—NO and prostacyclin, while downregulating inflammatory cellular responses. In contrast, OSS caused by disturbed blood flow at branching points and curvatures triggers endothelial inflammation and contributes to plaque formation [160,161]. Therefore, atherosclerotic lesions are mainly detected in regions exposed to disturbed flow, including the inner curvature of the aortic arch, carotid bifurcations, branch points of the coronary, and infrarenal arteries, etc. Due to exposure to disturbed flow, ECs in these arterial regions exhibit reduced production of anti-inflammatory and vasodilatory NO, along with increased expression of adhesion molecules (ICAM-1, VCAM-1, and E-selectin), facilitating leukocyte attachment and transmigration across the intima [160,162,163].
Shear stress activates various EC mechanosensors, such as integrins, tyrosine kinase receptors (e.g., VEGFR-2), and G-protein coupled receptors. Integrins mediate the effects of shear stress on EC cytoskeleton by activating MAPKs and focal adhesion-associated kinases [164,165]. VEGFR-2 activation by shear stress promotes downstream AKT phosphorylation via various proteins, including VE-cadherin, β-catenin, and PI3K [166,167]. These signaling pathways induce EC inflammation, proliferation, and apoptosis. A key mechanism by which OSS promotes EC inflammation is the impaired expression of eNOS, the principal enzyme responsible for NO production in ECs. Another important regulator of vascular health is Krüppel-like factor 2 (KLF2), a transcription factor that is highly expressed in ECs and induces eNOS expression. Laminar blood flow, but not atheroprone-OSS, upregulates KLF2 mRNA levels, leading to increased eNOS expression and NO production [168]. Other transcription factors, including PKA [169], Nrf2 [170], SIRT1 [171], and HDACs (e.g., HDAC5) [172], also contribute to laminar shear stress-mediated activation of eNOS. Furthermore, in vivo studies have shown that disturbed flow promotes EC proliferation in the early stages of atherogenesis. Interestingly, prolonged laminar flow in vitro reduces EC proliferation and leads to cell cycle arrest in the G0/G1 phase, whereas OSS accelerates EC turnover and the G0/G1 to S transition [173,174]. Thus, the vascular EC layer, through its response to hemodynamic forces, serves as a critical regulator of vascular physiology and pathobiology in both health and disease. The role of disturbed flow in regulating vascular dysfunction has been nicely reviewed previously [160,175].

3.3. ER Stress in EC Inflammation and Atherosclerosis

The ER plays important roles in protein synthesis, folding, trafficking, lipid synthesis, and calcium homeostasis. However, the ER can become dysfunctional due to the accumulation of unfolded or misfolded proteins in its lumen [176,177]. During ER stress, unfolded protein response (UPR) is induced as a compensatory mechanism to restore ER homeostasis by enhancing protein folding, degrading misfolded proteins, and reducing protein synthesis [178]. However, under the setting of persistent ER stress, the UPR stimulates apoptosis, inflammation, and oxidative stress, which can further damage cells, and has been shown to promote atherosclerosis [178,179]. ER stress activates various signaling pathways in ECs, such as MAPK, NF-κB, Nrf2, and NLRP3, which modulate the expression of genes involved in inflammation, antioxidant defense, and cell death [180].
Exposure of ECs to various sources of ER stress, such as elevated cholesterol levels, ROS, calcium imbalance, and shear stress, can impair normal cellular functions, including vasodilation, anticoagulation, anti-inflammation, and anti-oxidation [181,182]. For instance, ECs exposed to oxidized phospholipids undergo ER stress response and express higher levels of adhesion molecule, VCAM-1 [183]. Within human atherosclerotic lesions, endothelial regions rich in oxidized phospholipids exhibit increased ER stress markers, which promote the initiation and progression of plaque formation [184]. Throughout the course of atherogenesis, modified LDL, characterized by modifications, including oxidation, glycosylation, or phospholipolysis, perturbs calcium homeostasis within the ER, triggering UPR in ECs and promoting oxidative stress [185]. Earlier investigations have shown that phospholipolyzed LDL elicits inflammatory responses in ECs via ER stress pathways [181,186,187]. Furthermore, studies have demonstrated that oxLDL induces inflammatory pathways in ECs, which are responsible for EC damage through the activation of inflammasome-mediated apoptosis signal-regulating kinase 1 (ASK1) and NLRP3 [186,188]. In summary, modified LDL exerts significant regulatory effects on ER stress-mediated endothelial dysfunction, inflammation, and apoptosis within atherosclerotic vessels [179,187,188].
Under ER stress, an imbalance of the Bcl-2 family, mediated by CHOP, activates proapoptotic proteins on the mitochondrial membrane, leading to the release of cytochrome c and subsequent mitochondrial-dependent apoptosis [189]. This process, combined with disrupted calcium homeostasis, results in diminished mitochondrial function and elevated levels of NADPH and ROS in ECs within the pathological milieu of atherosclerosis [190,191]. Moreover, ER stress induces cytosolic calcium overload, triggering the activation of procaspase-12 into caspase-12 on the ER membrane of ECs. Consequently, caspase-3 and apoptosis are activated in these cells, alongside calpain-mediated activation of caspase-9 [179]. In short, dysfunctional ER responses, triggered by factors like modified LDL and oxidative stress, lead to inflammation, apoptosis, and oxidative damage to ECs and exacerbate atherosclerotic plaque formation and progression.

3.4. Autophagy in EC Inflammation and Atherosclerosis

Autophagy is a catabolic cellular process that degrades and recycles damaged or unwanted cellular components, such as proteins, organelles, and lipids [192,193]. Autophagy serves as both a metabolic process for degrading organelles and intracellular substances and as a stress response to conditions like starvation and oxidative stress. It plays a crucial role in cell proliferation, differentiation, and aging. The regulation of autophagy involves several signaling pathways, among which the mammalian target of rapamycin complex 1/unc-51-like autophagy activating kinase 1 pathway and the silent mating type information regulation 2 homolog 1/Forkhead box protein O1 (Sirt1/FoxO1) pathway are relevant to ECs [194,195,196]. Sirt1 expression in vascular ECs promotes angiogenesis and exerts anti-inflammatory and anti-atherosclerotic effects by regulating transcription factors such as FoxO1 and NF-κB, as well as eNOS activation [197,198,199]. FoxO1 is also intricately linked to autophagy regulation, as it modulates the expression of key autophagy-related proteins like microtubule-associated protein 1 light chain 3, autophagy-related protein 5 (Atg5), and Beclin-1 [196,200,201].
Autophagy has both anti-atherogenic and pro-atherogenic effects, depending on the type, intensity, and duration of the stimulus, and the stage of atherosclerosis [202]. Numerous stimuli can activate autophagy within atherosclerotic plaques, including ROS, LDL, TNF-α, and other inflammatory factors [203,204]. Autophagy can reduce atherogenesis and help maintain a stable plaque phenotype. Inhibition of EC’s autophagy induces the expression and secretion of various factors VCAM-1, ICAM-1, E-selectin, MCP-1, and IL-8, which facilitate the recruitment and activation of leukocytes and promote foam cell formation, thereby increasing the risk of arterial thrombosis [202]. Additionally, Perrotta et al. demonstrated that pharmacological inhibition of glycolytic flux with a small molecule inhibitor 3-[3-pyridinyl]-1-[4-pyridinyl]-2-propen-1-one (3PO) induces autophagy and impairs NF-κB signaling, resulting in the inhibition of TNF-α-mediated upregulation of VCAM-1 and ICAM-1 [205]. In instances of oxidative stress, damaged mitochondrial DNA (mtDNA) evades autophagic clearance and triggers a potent inflammatory response in the arterial wall [206]. Enhanced autophagy significantly reduces chronic vascular inflammation and suppresses atherosclerosis. Conversely, inhibition or impairment of autophagy exacerbates the inflammatory response [202,207]. These findings suggest that autophagy protects against endothelial inflammation. Transient knockdown of the essential autophagy gene ATG7 results in increased intracellular levels of both intermediate-density lipoprotein and oxLDL, suggesting that in ECs, autophagy serves as an important mechanism for regulating excess exogenous lipids [208,209]. All these studies demonstrated that endothelial autophagy has a potential role in EC inflammation-dependent atherosclerosis. Therefore, a better understanding of the molecular mechanisms regulating autophagy in ECs may be essential in developing novel and effective strategies to prevent EC dysfunction/inflammation.

4. Matricellular Proteins in EC Inflammation and Atherosclerosis

Matricellular proteins are a group of non-structural proteins that modulate various cellular functions, such as adhesion, migration, proliferation, differentiation, and survival, by interacting with different molecules, including cell surface receptors, proteases, hormones, and ECM components. They are often highly expressed during embryonic development, repair, and remodeling, and regulate inflammation and immune responses. Remarkably, these proteins exhibit both pro-inflammatory and anti-inflammatory effects, depending on the context and receptors they interact [11,210]. Therefore, targeting these proteins or their receptors may offer new therapeutic strategies for modulating inflammation and associated pathologies. These proteins are characterized by distinct domains that possess enzymatic activity capable of modifying ECM components or regulating the activities of various growth factors. By serving as molecular bridges, these proteins facilitate communication and crosstalk among growth factors, proteases, cytokines, and other macromolecules within the cellular microenvironment [211]. The matricellular protein family includes a diverse range of molecules, including thrombospondins (TSPs), CCN proteins (CYR61, CTGF, NOV), SPARC (secreted protein acidic and rich in cysteine), osteopontin, tenascins, Rspondins and others [11]. Each member of this protein family exhibits unique structural features and functional properties, contributing to the dynamic regulation of cellular and tissue homeostasis. Furthermore, their dysregulated expression and activity have been implicated in various pathological conditions, including cancer, fibrosis, CVDs, and inflammatory disorders [212,213]. Here, we summarize the role of various matricellular proteins in EC inflammation and atherosclerosis (Table 3).
Table 3. Role of matricellular proteins in regulating vascular phenotype.
Table 3. Role of matricellular proteins in regulating vascular phenotype.
ProteinPrimary ReceptorsSignaling Pathway and PhenotypeCell/Animal/Human Model TypeRoleRefs.
TSP1CD47
CD36
Decreases cAMP/cGMP levels by inhibiting EC NO productionBovine aortic ECs
Human umbilical vein ECs
Thbs1−/− and Cd47−/− mice arteries
EC dysfunction and pro-hypertensive[214]
Thbs1 deletion prevents leptin-induced atherosclerosis

Deletion blocks leptin-induced vascular inflammation

Deletion inhibits SMC dedifferentiation
Apoe−/− and Apoe−/−/Thbs1−/− micePro-atherogenic



Pro-inflammatory


Pro-atherogenic
[215]
In the early stage, a deficiency of Thbs1 reduces plaque area

In the advanced stage, Thbs1 loss promotes plaque necrosis
Apoe−/− and Apoe−/−/Thbs1−/− micePro-atherogenic


Anti-plaque vulnerability
[216]
Thbs1 deletion in mice promotes maladaptive remodeling in response to pressure overload via inhibiting Thbs1/integrin β1/YAP signaling

Thbs1 deletion inhibits neointima formation upon carotid artery ligation
Thbs1−/− mice





Promotes intimal hyperplasia
[217]
COMPα5β1
α7β1
Lack of Comp induces aging-related vascular dysfunction, stiffness, and senescenceComp−/− micePromotes vascular function[218]
Comp deletion augments atherosclerosisApoe−/− and Comp−/−/Apoe−/− miceAnti-atherogenic[219]
RSPO2LGR4Suppresses lymphangiogenesis via PI3K-AKT-eNOS signaling and inhibits Wnt-β-catenin pathway in lymphatic ECsHuman dermal lymphatic ECs Anti-lymphangiogenic



Pro-atherogenic
[220]
Perivascular application of LGR4-ECD promotes arterial lymphangiogenesis and reduces atherosclerosisApoe−/− mice
RSPO1LGR4-5Wnt/β-catenin/VEGFaa-induced abnormal angiogenesisZebrafishPro-angiogenic[221]
RSPO3LGR4-5Non-canonical WNT/Ca2+/NFAT signaling and vascular defects EC-specific Rspo3-deficient micePro-angiogenic[222]
Tenascin-CIntegrinsTN-C polymorphisms correlate with atherosclerosis/CADHuman aorta samples and CATHGEN cardiovascular studyThree SNPs correlate with atherosclerosis[223]
Osteopontin
(OPN/SSP1)
Integrins
CD44
Deletion reduces atherosclerosis
Deletion stimulates vascular calcification
Deficiency reduces atherogenesis
Apo−/−/Spp1−/− mice


Apoe−/−/Ldlr−/− /Spp1−/− triple knockout mice
Pro-atherogenic



Pro-atherogenic
[224]
Expression levels associate with plaque severityHuman aorta samplesPro-atherogenic[225]
CCN1IntegrinsUpregulated levels in atherosclerotic aortas of Apoe−/− mice
Promotes atherosclerosis
Apoe−/− micePro-atherogenic[226]
Elevated Ccn1 expression in atherosclerotic arteriesApoe−/− mice and human ECsMediates TNFα-induced EC apoptosis[227]
Promotes neovascularizationC57BL/6 wild-type mice and human venous ECsPro-angiogenic[228]

4.1. Thrombospondins

There are five different types of thrombospondins, named TSP1 to TSP5, based on their molecular structures, functions, and oligomerization status, which are encoded by five distinct genes, namely THBS1 to 5 [229]. Among them, TSP1 and TSP2 are extensively studied and play significant roles in various biological processes. They contribute to inflammatory responses, regulate angiogenesis during tumor growth, and promote EC apoptosis. TSP1 and TSP2 via binding to various receptors found on ECs, including CD36 [230], CD47 [231], LRP-1 [232], and integrins (α9β1, α6β1, αvβ3, and αIIββ3) [233,234,235,236] regulate several signaling pathways critical for cellular function [11,237]. TSP2 has been demonstrated to inhibit the expression of vascular endothelial growth factor A (VEGF-A), a potent angiogenic factor. Via blocking binding of VEGF to its receptors on ECs, TSP2 prevents VEGF-VEGFR-mediated angiogenic signaling [238]. In addition, TSP2 exerts inhibitory effects on the activities of MMP-2 and MMP-9, enzymes involved in angiogenesis and inflammation [239]. Further, TSP2 activates the tissue inhibitor of metalloproteinases 3 (TIMP-3), an inhibitor of MMPs and other proteases. While TSP3, TSP4, and TSP5 are less characterized and involved in tissue development, repair, and remodeling. Notably, TSP4 via CD44 activation modulates the ECM composition and the mechanical properties of atherosclerotic plaques by interacting with collagen and elastin [240], and affects plaque vulnerability and rupture. Additionally, Frolova et al., reported that TSP4 triggers inflammatory signaling in ECs and monocytes/macrophages, promoting the accumulation of macrophages in atherosclerotic plaques [241]. TSP5, also known as cartilage oligomeric matrix protein (COMP), is mainly expressed in cartilage and modulates the differentiation and survival of chondrocytes [242]. TSP5 functions by interacting with integrins (α7β1, αvβ3, α5β1, α5β3) as well as CD47 [11,243,244].

4.1.1. Thrombospondin1 (TSP1)

TSP1 plays an important role in various biological processes, including angiogenesis [245], lymphangiogenesis [246], inflammation [247], and atherosclerosis development [215,216,246]. TSP1 has been shown to bind and activate latent TGF-β, thereby modulating TGF-β-stimulated signaling in various diseases, including diabetes, liver fibrosis, pulmonary arterial hypertension, arterial stiffening, cardiomyopathy, and tumors [248]. Antibody-mediated blockade of TSP1-induced signaling has been shown to promote re-endothelialization and suppress neointima formation in the carotid arteries of rats after balloon injury, indicating the detrimental role of TSP1-stimulated signaling in neointima formation [249]. Moreover, global Thbs1 deletion in mice on Apoe/ background led to reduced plaque area compared with control Apoe−/− mice following leptin stimulation [215]. In contrast, global Apoe−/−/Thbs1−/− double knockout mice exhibited augmented plaque maturation; however, this effect was observed only during the advanced stage of atherosclerosis [216]. In early atherogenesis, Thbs1 loss reduced lesion formation; however, in the later stages, its deficiency was associated with increased inflammation, impaired phagocytosis, and elevated ECM remodeling [216]. Despite these findings, the impact of EC-specific Thbs1 deletion or overexpression on atherosclerotic lesion formation remains unexplored.
The effects of TSP1 on EC biology have been extensively studied in vitro. Activation of CD36 by TSP1 initiates a cascade involving Fyn, JNK, and p38/MAPK, ultimately triggering apoptosis via the activation of caspase-8 and caspase-9-dependent pathways. Additionally, TSP1 via CD36 activation triggers caspase-dependent cell death mechanisms and ultimately induces EC apoptosis [250,251,252]. Consistently, CD36-silencing in human umbilical vein ECs (HUVECs) counteracts the inhibitory effects of TSP1 on migration and tube formation, indicating the role of TSP1-CD36 signaling axis in angiogenesis [253]. Further, in vitro studies demonstrated that TSP1 and stimulation of CD36 receptor with a CD36 agonist antibody inhibit NO-stimulated EC responses, including chemotaxis, adhesion, and proliferation [254], suggesting an antagonism between the TSP1-CD36 axis and proangiogenic signaling downstream of NO. Besides, CD36 knockdown in MVECs or genetic Cd36 deletion in mice revealed that TSR (CD36-binding domain of TSP1)-induced SHP-1/VEGFR2 complex formation is mediated by CD36 both in vitro and in vivo, and silencing of SHP-1 in MVECs abrogates TSR-mediated inhibition of VEGFR2 phosphorylation [255]. On the other hand, TSP1-mediated suppression of NO signaling remains unaltered in CD36-null ECs; however, it is reduced in CD47-deficient ECs, demonstrating the essential role of CD47 in this process [256]. Further, TSP1 by binding to CD47 in ECs inhibits eNOS, leading to reduced NO production and bioavailability [257]. Consistently, TSP1 treatment has been demonstrated to promote VCAM-1 and ICAM-1 expression and increase monocyte adhesion to human ECs [258]. These observations point out the significant role of TSP1-induced signaling in EC inflammation. Recently, Peng et al. reported elevated levels of TSP1 in the peripheral blood of pediatric patients with pulmonary hypertension, and suppression of Sugen/hypoxia-induced pulmonary hypertension and EndMT with pharmacological inhibition of TSP1 [259]. Additionally, silencing of THBS1 in human pulmonary ECs inhibited hypoxia-stimulated downregulation of EC markers, further supporting the role of TSP1 in hypoxia-induced EndMT [259]. Moreover, increased TSP1 expression has been associated with EndMT in a mouse model of complete left carotid artery ligation, indicating the potential role of TSP1 in EndMT-induced atherosclerotic lesion formation [260].
In addition, TSP1-CD47 signaling regulates thrombosis/hemostasis [261], immune responses, and mitochondrial function in various cell types [262,263]. Moreover, TSP1-CD47 signaling axis is involved in the regulation of senescence, self-renewal, and inflammation. Experimental data from CD47-deficient ECs and muscle explants from global Cd47 knockout mice revealed that TSP1 requires CD47 to inhibit NO signaling [256]. CD47 deficiency in ECs downregulates inflammatory cytokines and chemokines, including TSP1 [264,265]. Additionally, a recent investigation by Singh et al. reported that the deletion of Cd47 specifically in ECs attenuates atherosclerotic lesion formation in mice [266]. Further, the authors observed increased internalization of apoptotic cells (efferocytosis) by Cd47-deficient ECs compared with control cells possibly via upregulating expression of efferocytosis receptor FasL, CX3CL1, Gpr132, MerTK, Scarb1, Sirpα, TSP1, and HMGB1 [266]. In addition to this, the expression of CD47 on non-tumor cells, like ECs, is crucial in regulating tumor angiogenesis. Particularly, the absence of CD47 expression on ECs markedly boosts angiogenesis, thereby effectively mitigating hypoxia-induced tumor necrosis and expediting tumor progression [265]. This suggests that targeting CD47 and/or TSP1 can be a promising therapeutic strategy to delay EC senescence and mitigate inflammation.
TSP1 induces ROS generation in ECs, leading to oxidative stress and ultimately inducing cell senescence. TSP1-mediated Nox1 activation in human pulmonary artery ECs increases ROS production and elevates levels of transcription factor p53, promoting EC senescence [267]. Importantly, blocking of CD47 receptor with an antibody prevents TSP1-induced ROS generation [267], suggesting TSP1 promotes senescence, inflammation, and apoptosis in ECs by increasing ROS generation, inhibiting NO signaling, and/or decreasing antioxidant levels [245,268,269]. Various in vivo studies have explored the role of TSP1 in EC ROS generation and its implications in metabolic and CVDs. Mice deficient in TSP1 exhibit reduced ROS production, decreased p21 expression, attenuated p53 activity, and diminished aging-induced senescence in lung tissue compared with wild-type mice [267] (Figure 2A). Furthermore, TSP1-induced CD47 activation stimulates ROS generation in human pulmonary artery ECs, and this signaling pathway emerged as a pivotal player in sickle cell-associated vasculopathy, contributing to the development of pulmonary hypertension [270,271,272]. Recent research by our group demonstrated that TSP1 via CD47 activation in lymphatic ECs (LECs) suppresses lymphangiogenesis by inhibiting AKT-eNOS signaling and inducing ROS production [246]. Further, deletion of CD47 specifically in LECs augmented lymphangiogenesis and attenuated atherosclerosis in hypercholesterolemic mice [246] (Figure 2A). In another type of vascular cell, VSMCs, TSP1 stimulates Nox1-mediated superoxide production as determined using cytochrome c reduction and electron paramagnetic resonance assays [273]. Further, the inhibition of CD47-mediated signaling employing blocking antibody and gene silencing abrogates TSP1-induced ROS production in vitro and ex vivo. This increased ROS production, in turn, disrupts redox signaling within the vasculature, consequently impairing VSMC-dependent vasorelaxation, a crucial mechanism for maintaining vascular tone and blood pressure (BP) regulation [273]. Such vascular dysfunction contributes to pathological conditions like hypertension and atherosclerosis, highlighting the critical role of the TSP1-CD47 axis in vascular homeostasis and disease progression. Additionally, in macrophages, TSP1-stimulated Nox1 activation triggers the dephosphorylation of the actin-binding protein cofilin [274]. This process subsequently induces cytoskeletal rearrangement, which is associated with increased LDL uptake by macrophages via stimulating macropinocytosis, thus promoting atherosclerosis progression [274,275]. Altogether, these findings suggest that inhibition of TSP1-mediated signaling may be beneficial for EC function, vascular tone, and homeostasis.

4.1.2. Thrombospondin 5/Cartilage Oligomeric Matrix Protein (COMP)

TSP5/COMP functions by binding to α5β1 and is mainly expressed in cartilage, bone, and tendon, where it regulates the development and maintenance of the ECM [244]. Expression of COMP has also been detected in both inflammatory and fibrous atherosclerotic plaques in mice [219]. In a recent study, Hultman et al. associated arterial COMP expression with symptomatic carotid atherosclerosis [276]. Additionally, COMP levels were positively correlated with plaque lipid content and CD68-positive areas but inversely correlated with lesional collagen content, suggesting a link between COMP expression and plaque vulnerability. Another study reported higher circulating levels of COMP in patients with CAD and found a positive correlation between COMP levels and coronary artery calcium scores, advocating its potential as a biomarker for coronary artery calcification [277]. Interestingly, atheroprone Apoe/ mice lacking Comp exhibit increased plaque formation and vascular calcification [278]. Further, the authors of this study demonstrated that Comp deletion promotes atherosclerotic calcification by shifting macrophage phenotype toward pro-atherogenic and osteogenic types. Supporting this, Bond et al. reported larger plaques in brachiocephalic arteries of Comp-deficient Apoe/ mice [219]. Moreover, COMP suppresses disturbed flow-induced EC activation by interacting with integrin α5, thereby maintaining endothelial homeostasis. Increased EC activation was observed in Comp−/− mice, particularly in the aortic arch under normal conditions and following partial carotid artery ligation [279]. Notably, treatment with a COMP-derived peptidomimetic (CCPep24) that mimics COMP’s interaction with integrin α5 reduced endothelial activation and atherogenesis in vivo [279], indicating the protective role of COMP in EC inflammation and atherosclerosis.
Various studies have investigated TSP5 expression and function in vascular cells and angiogenesis [280,281]. TSP5 has been shown to have pro-angiogenic activity, potentially via inducing the expression of a proangiogenic gene HEY1, cytokine LTB, and antiapoptotic BCL2, while downregulating a proapoptotic gene, BIK [281] (Figure 2B). TSP5 enhances endothelium-dependent relaxation via regulating Piezo1/CaKMII/eNOS signaling pathway, thereby playing a protective role in BP regulation [282]. Moreover, overexpression of COMP in VSMCs reduces systolic and mean arterial BP during AngII-induced hypertension and promotes CaKMII/eNOS activation in arteries, suggesting the protective role of VSMC-derived COMP in BP regulation via NO release in ECs [282]. Similarly, COMP maintains endothelial homeostasis by attenuating disturbed flow-induced endothelial activation through its interaction with integrin α5 [279]. In both normal and pathological conditions, such as partially ligated carotid arteries in mouse models, global Comp-deficient mice exhibited increased aortic EC activation compared with control mice. Additionally, peptidomimetics derived from the C-terminal of COMP (CCPep24), which interacts with integrin α5, protect against EC activation and atherogenesis in vivo [279].
COMP also plays a pivotal role in the maintenance of VSMC contractile phenotype and confers protection against injury-induced neointima formation in Sprague-Dawley rats by interacting with α7β1 integrin [243]. Riessen et al. reported COMP expression in human VSMCs, human normal arteries, and those affected by atherosclerosis and restenosis [280]. In vitro experiments demonstrated that VSMCs strongly adhere to COMP-coated surfaces, facilitating their migration [280]. These findings suggest a potential role for COMP in vasculogenesis and vascular diseases such as atherosclerosis and restenosis by regulating VSMC migration and adhesion. Several studies have reported elevated COMP expression in fibrotic conditions through activation of skin fibroblasts and lung epithelial cells by TGF-β. TGF-β induces the conversion of fibroblasts and lung epithelial cells into myofibroblasts, leading to increased ECM synthesis and fibrosis. Upregulation of COMP expression enhances TGF-β signaling, establishing a positive feedback loop in idiopathic pulmonary fibrosis and fibrotic skin [283,284,285,286]. On the other hand, evidence from multiple mouse models indicates endothelial dysfunction and EndMT in vascular calcification [84,287,288]. Therefore, it is tempting to speculate a possible association of arterial COMP expression with TGF-β-dependent signaling in EC inflammation responsible for EndMT, atherosclerosis, and vascular calcification.

4.2. Osteopontin (OPN)

Osteopontin is a multifunctional glycoprotein, which plays a crucial role in inflammatory processes. It is also called secreted phosphoprotein 1 (SPP1). It is expressed in various tissues in adults, including bone, kidney, and epithelial linings [289,290]. OPN aids in cell migration, adhesion, and survival of various cell types including ECs, SMCs, and inflammatory cells through interactions with integrins (α4β1, α5β1, α8β1, α9β1, αvβ1, αvβ3, αvβ5, and αvβ6) and CD44 [291,292,293,294,295]. OPN expression is normally low but is remarkably upregulated at sites of inflammation and tissue remodeling. OPN is mainly produced by injured and inflamed epithelial cells, ECs, SMCs, and certain tumor cells, as well as by inflammatory infiltrating cells comprising T cells and macrophages [296,297,298,299]. Ang II has been shown to induce OPN expression in cardiac MVECs in a time- and concentration-dependent manner, and this effect was inhibited by treatment with Ang II receptor type 1 antagonist, losartan. Furthermore, inhibition of NOXs with diphenylene iodonium prevented Ang II-induced upregulation of OPN, highlighting the involvement of redox-sensitive signaling in this process [300].
Golledge et al. reported upregulated OPN expression in the proximal segments of atherosclerotic internal carotid arteries isolated from symptomatic patients compared with asymptomatic individuals [301]. Increased OPN levels in symptomatic patients may promote infiltration of macrophages and secretion of proteolytic enzymes, thereby reducing plaque stability [302]. Matsui et al. demonstrated that the deficiency of OPN reduces the size of atherosclerotic lesions in female Apoe/OPN double-knockout mice after 36 weeks of a normal chow diet [224]. These findings align with previous findings indicating that OPN attracts macrophages and lymphocytes, therefore, promotes vascular inflammation and atherogenesis [303]. However, no differences in atherosclerosis were observed among male control and OPN-deficient mice. On the other hand, endothelial dysfunction is known to play an important role in the early stages of atherosclerosis. OPN has been associated with reduced eNOS activity within atherosclerotic lesions, exacerbating endothelial dysfunction in individuals with CAD [304,305]. OPN overexpression in mice stimulates medial thickening, neointima formation, and accelerates atherosclerosis progression [224,306]. Moreover, transgenic mice overexpressing OPN exhibit accelerated development of fatty streaks and mononuclear cell-rich lesions, resulting in larger atherosclerotic plaques compared with control mice after being fed an atherogenic diet [306,307], suggesting the pro-atherogenic role of OPN.
OPN, via binding to α9β1 integrin, has been shown to activate ERK and p38 signaling pathways, which stimulate COX-2 expression in macrophages and promote angiogenesis [308]. OPN plays a significant role in orchestrating the inflammatory response and modulating EC function. Dai et al. reported that OPN promotes EC proliferation, migration, and tube formation, and induces activation of PI3K/AKT- and ERK-mediated pathways [309]. Further, OPN treatment stimulated VEGF mRNA and protein expression in dose- and time-dependent manners responsible for the second phase of AKT and ERK signaling. Additional in vivo experiments showed inhibition of angiogenesis with anti-OPN antibody treatment in the corneal pocket and MCF-7 xenograft models [309]. Furthermore, in vitro studies have highlighted the significant role of OPN in the chemotaxis of inflammatory cells [296,310]. Additionally, OPN stimulates EC proliferation via inducing tumor cells to release VEGF. OPN present in tumors interacts with CD44 and integrin receptors, thereby facilitating PI3K/Akt/eNOS- signaling, which in turn promotes EC proliferation, migration, and tube formation in vitro [311,312,313]. These cellular changes promote angiogenesis within tumors and stimulate tumor growth. Lyle et al. revealed that H2O2 stimulates OPN translation via phosphorylating 4E-BP1 (Ser65) in VSMCs [314]. Further in vivo study by the same group demonstrated increased OPN and H2O2 levels in ligated femoral arteries compared with controls [315] and scavenging of H2O2 with polyethylene glycol-catalase infusion and SMC-specific catalase overexpression blunted ischemia-induced OPN, suggesting a crucial role of H2O2 in the induction of OPN expression during ischemia. Additionally, the reduction in OPN expression resulted in impaired neovascularization [315]. Altogether, OPN plays a multifaceted role in EC inflammation and atherosclerosis, and further research is required to fully understand the underlying molecular mechanisms.

4.3. Roof Plate-Specific Spondins (RSPOs)

Roof plate-specific spondins (RSPOs) are involved in many biological processes, such as tissue homeostasis, stem cell control, and embryonic development [11,316,317,318,319]. There are four types of RSPOs, namely RSPO1, RSPO2, RSPO3, and RSPO4, all of which are crucial in regulating Wnt/β-catenin signaling, essential for various developmental processes and tissue regeneration [11]. RSPO3 has been shown to regulate angiogenesis and vascular remodeling [222]. Different RSPOs function by interacting with various receptors and co-receptors, including leucine-rich repeat-containing G-protein coupled receptors (LGR4, 5 and 6), ZNRF3, and RNF43, and modulate Wnt/β-catenin signaling [318,320,321,322]. This Wnt signaling, in turn, regulates key cellular functions such as proliferation and differentiation, thereby influencing vascular development and angiogenesis [323]. Bretón-Romero et al. have shown that ECs isolated from diabetic patients exhibit higher levels of Wnt5a, have increased JNK activation, and impaired eNOS phosphorylation [324]. Inhibition of Wnt5a and JNK restores eNOS activation and EC function, suggesting the detrimental role of Wnt5a/JNK pathway in endothelial dysfunction during diabetic conditions [324]. Stimulation of human aortic ECs with Wnt5a induces COX-2 expression and elevates inflammatory cytokine levels, while Wnt3a has limited effects, pointing to the role of β-catenin-independent Wnt signaling in inflammatory endothelial activation [325]. Further experiments revealed that Wnt5a increases endothelial permeability and promotes EC invasion, reflecting an inflammatory state of the endothelium. Collectively, this information suggests that Wnt5a plays a significant role in regulating endothelial permeability [324,325,326,327,328].
RSPO1 is one of the extensively studied members of the RSPO family. It aids in embryonic development, particularly in limb formation, hair follicle development, and sex determination. In the zebrafish model, RSPO1-Wnt signaling promotes angiogenesis by stimulating the Vegfc-Vegfr3 axis during development [329]. In line with this, blocking Rspo1 function through genetic mutation or morpholino-mediated Rspo1 knockdown suppresses angiogenic proliferation and leads to defective trunk and cranial vessel angiogenesis. Moreover, deficiency of the putative Rspo1 receptor Kremen, which is expressed in the vasculature, also results in angiogenic defects in zebrafish [329,330]. Furthermore, Caruso et al. reported that administration of R-spondin 1 rescues Dickkopf-1-induced inhibition of testicular angiogenesis during embryonic development [331]. Additionally, Rspo1/Wnt/β-catenin signaling has been demonstrated to upregulate VEGFaa expression and promote N-methyl-N-nitrosourea-induced abnormal angiogenesis in zebrafish [221]. However, the specific role of RSPO1 in endothelial inflammation remains understudied, given the pleiotropic role of Wnt/β-catenin signaling, RSPO1 may regulate EC inflammation and modulate atherosclerosis development.
In a recent study from our laboratory, we investigated the role of RSPO2 in arterial lymphangiogenesis and atherosclerotic lesion formation [220]. We discovered that RSPO2 negatively regulates VEGFC-VEGFR3-induced lymphangiogenesis both in vitro and in vivo. RSPO2 induces its anti-lymphangiogenic effects via activation of LGR4 receptor, which inhibits PI3K-AKT-eNOS signaling and suppresses canonical Wnt-β-catenin cascade [220] (Figure 3A). Interestingly, we observed an inhibitory role of RSPO2 in LECs on β-catenin activation. Furthermore, investigations into the significance of RSPO2-regulated lymphangiogenesis in atherosclerosis revealed that blockade of LGR4-mediated signaling employing the perivascular application of RSPO2’s decoy receptor (LGR4 extracellular domain) promotes arterial lymphatic vessel density, increases arterial cholesterol drainage and reduces atherosclerosis (Figure 3A). These findings suggest that the inhibition of RSPO2-induced signaling may be a therapeutic target to promote lymphangiogenesis and suppress atherosclerosis [220]. In the same study, we reported that RSPO2 inhibits lymphangiogenesis via impaired NO biogenesis rather than increased ROS-mediated impairment of NO bioavailability [220]. While Rspo2 deletion in widespread cells utilizing the UBC-Cre-ERT2 mouse line increased neutrophil migration into the alveolar space and induced lung permeability in Rspo2 knockout mice compared with control mice [332], hinting at a beneficial role of Rspo2 in regulating blood vessel permeability or promoting neutrophil migration. Similarly, Carmon et al. observed the activation of Wnt signaling following RSPO2 exposure across various cell types, including MDCK, HEK293T, and HeLa cells. These findings suggest that RSPO2’s effects on the Wnt-β-catenin pathway are cell-specific [333]. In addition, NF-κB activation has been shown to regulate RSPO2 expression in processes like heterotopic ossification [334,335], and NF-κB signaling pathway plays a significant role in EC activation and atherogenesis [336]. However, it remains to be investigated whether RSPO2 mediates NF-κB-induced EC dysfunction and impaired endothelium permeability. Additionally, Aithabathula et al. recently identified RSPO2 as a key suppressor of hepatic steatosis and fibrosis [337], which are closely associated with the development of atherosclerosis. Therefore, it is tempting to speculate on the beneficial role of hepatocyte RSPO2 in atherosclerosis, in contrast to its detrimental role in vascular cells.
Skaria et al. demonstrated that human EC monolayers treated with RSPO3 have increased permeability compared with vehicle-treated cell layers [338]. This increased permeability is attributable to the induction of intercellular gaps, resulting from the disruption of β-catenin and VE-cadherin alignment at adherent junctions [338]. Conversely, a study by Scholz and colleagues demonstrated RSPO3 as an inducer of Wnt signaling in ECs [222]. They observed that inducible deletion of Rspo3 results in perturbed developmental and tumor vascular remodeling, leading to reduced microvessel density in deficient mice (Figure 3B). Besides, EC-specific Rspo3-deficient mice phenocopied vascular defects observed in non-canonical Wnt signaling factor Evi/Wls knockout mice [222]. These results demonstrate the crucial function of RSPO3 in preserving vascular integrity and suggest it as a potential therapeutic target for pathologies associated with aberrant angiogenesis and vessel regression. The downstream effects of RSPO-induced signaling, particularly in ECs, primarily occur through the activation of the Wnt/β-catenin signaling pathway. However, while these effects are well-documented, the precise mechanisms by which RSPOs activate the Wnt/β-catenin signaling pathway are still not fully understood.

4.4. Tenascins

The tenascin family comprises four members: tenascin-C (TN-C), tenascin-R (TN-R), tenascin-X (TN-X), and tenascin-W (TN-W). They play important roles in tissue formation, homeostasis, wound healing, and various pathological processes [339,340,341]. TN-R is predominantly expressed in the central nervous system (CNS), especially in areas of axonal growth and guidance during development. While its expression is low in adult CNS, it increases in response to injury or inflammation, suggesting a potential role of TN-R in neural repair [342,343,344,345,346]. TN-X, primarily found in connective tissues, maintains the structural integrity of the ECM and regulates collagen fibrillogenesis. Further, TN-X induces inflammation via activation of TGF-β signaling and interaction with cell surface integrins (α11β1), altering tissue dynamics and immune responses [339,347,348]. TN-W is expressed in various tissues, including the nervous system and skeletal muscle. In brain tumors, TN-W is specifically expressed in blood vessels. Moreover, TN-W has pro-angiogenic activity as demonstrated by in vitro studies using EC cultures [349].
TN-C expression is highly expressed in VSMCs during embryonic development and its levels are also elevated in adults during tissue injury, inflammation, or remodeling [350,351,352]. High expression of TN-C has been linked with several arterial pathologies including intimal hyperplasia, pulmonary artery hypertension, aortic aneurysm/dissection, and cerebral vasospasm [353]. TN-C regulates a wide range of cellular processes, including adhesion, migration, proliferation, and differentiation, which are mediated via its interactions with cell surface receptors and ECM components.
Serum levels of TN-C are significantly higher in patients with CAD compared to non-CAD patients, and an association between TN-C levels and the severity of atherosclerosis was observed, indicating its involvement in lesion formation and disease progression [354,355,356]. Elevated plasma TN-C levels are also associated with a higher incidence of cardiovascular events and increased overall and cardiovascular-related mortality in patients with chronic kidney disease, indicating its potential as a prognostic marker in high-risk populations. Exosomes isolated from CAD patients also exhibit higher levels of TN-C compared to those from non-CAD patients, suggesting a potential role of TN-C in disease pathogenesis and intercellular/interorgan communication [357]. In atheroprone Apoe/ mice, TN-C expression increases with the progression of atherosclerosis, suggesting its involvement in disease development [358,359]. Additionally, TN-C expression is upregulated in neointimal lesions following balloon catheter-induced vascular injury, highlighting its potential involvement in vascular remodeling after injury [360].
Interestingly, Rupp et al. demonstrated higher sprouting of aortic rings from TN-C-deficient mice in comparison to rings from wild-type mice [361]. TN-C exposure represses EC tube formation, migration, and invasion and induces cell apoptosis via inhibition of YAP signaling in vitro. Elevated TN-C levels are also associated with increased tumor cell survival, enhanced angiogenesis, and higher vessel leakiness, thereby promoting metastasis [362]. Furthermore, in vitro studies demonstrate that treatment with an inflammatory agent LPS, augments TN-C expression in THP-1 macrophages in a dose- and time-dependent manner, and TN-C mediates LPS-induced foam cell formation [363]. Consistently, TN-C is involved in oxLDL-stimulated foam cell formation in THP-1 macrophages, which is abrogated by CD36 gene silencing and blockade of TLR4, suggesting its role in macrophage lipid uptake and atherogenesis [364]. Similarly, perivascular macrophages activated by TN-C via TLR4 activation release NO and TNF, inducing niche component expression in ECs [365].
Fibronectin and TN-C, important components of the cell matrix, exhibit pro- and anti-adhesive properties, respectively. TN-C induces fibronectin expression in ECs, which counteracts the anti-adhesive effects of TN-C responsible for reduced EC motility. Consequently, this triggers the assembly of a dense, highly branched subendothelial matrix that promotes tubulogenic activity [366]. Contrary to this, increased lymphangiogenesis was observed following the downregulation of TNC expression. TN-C demonstrated a modest downregulation of genes associated with nuclear division, cell division, and cell migration in lymphatic ECs, suggesting its inhibitory effects on these cellular processes [367] (Figure 4B). Additionally, TN-C is an important inducer of neuroinflammatory cascades and the resultant pathology of stroke. It is upregulated in cerebral arteries and brain tissues including astrocytes, neurons, and brain capillary ECs following subarachnoid hemorrhage [368]. Overall, the above studies suggest that TN-C plays a significant role in regulating angiogenesis, EC function and development. However, the exact mechanisms of how TN-C interacts with ECs and its overall impact on endothelial function require further investigation.

4.5. CCN Proteins

CCN family members are involved in the regulation of cell adhesion, proliferation, migration, differentiation, survival, angiogenesis, and wound healing. This family includes six members named CCN1 to CCN6. CCN proteins play a crucial role as adaptor molecules in multiple cellular processes involved in vascular development and vascular diseases such as atherosclerosis and restenosis. One of their important functions is to bind growth factors like VEGF and TGF-β and facilitate their localization near the cell surface through interactions with cell surface binding partners like integrins or heparan sulfate proteoglycans. CCN2 interacts with various cell surface receptors, including heparan sulfate proteoglycans, tropomyosin-related kinase A (TrkA), and LRP-1 [369,370,371]. On the other hand, CCN3 exerts its effects through interactions with notch receptors and integrins, specifically α6β1 and α6β5 [372,373,374]. CCN5 is mainly expressed in ECs, fibroblasts, and VSMCs within differentiating scar tissue and known to inhibit vascular cell growth and migration, indicating a potential anti-inflammatory protective role [375,376,377,378]. The expression of CCN proteins in vascular endothelium is regulated by shear stress, a mechanical force generated by blood flow in blood vessels [371,379,380,381,382,383,384].
As we know, atherosclerosis is a focal arterial disease, mainly manifesting at the sites of disturbed blood flow, including the lesser curvature of the arch and bifurcations. Human ECs, when exposed to OSS, mimicking disturbed flow at atherosclerotic sites, have increased CCN1 expression [371]. Elevated CCN1 levels contribute to endothelial dysfunction and promote atherosclerosis. The interaction between CCN1 and integrin α6β1 activates NF-κB, leading to increased CCN1 production, thus establishing a positive feedback loop that exacerbates endothelial activation and atherogenesis [385]. Hsu et al. reported that disrupting CCN1-integrin α6β1 interaction by a mutation (defective in binding α6β1) in Ccn1 gene or using an antagonist T1 peptide (derived from an α6β1-binding sequence of CCN1) may serve as a promising therapeutic strategy to prevent or treat atherosclerosis [385]. Knock-in mice (Ccn1dm/dm/Apoe/) carrying a mutant Ccn1 allele unable to bind integrin α6β1 exhibit reduced plaque formation, suggesting the importance of CCN1 and integrin α6β1 interaction in atherosclerosis development [385]. Moreover, experimental strategies to manipulate CCN1-induced signaling using gain-/loss-of-function assays in cultured macrophages have revealed that CCN1 promotes lipid accumulation and foam cell formation [226]. In Apoe/ mice, CCN1 treatment exacerbates atherosclerosis and induces systemic inflammation [226]. Mechanistically, CCN1 treatment downregulates hepatic expression of key genes involved in cholesterol metabolism, including ABCG5, ABCG8, liver X receptor α, cholesterol 7a-hydrolase, and LDL receptor [226]. These findings indicate that CCN1 via increasing hepatic lipid accumulation and inhibiting macrophage cholesterol efflux, promotes atherosclerotic lesion formation. Similar to reduced atherosclerosis in Ccn1dm/dm mice, Ccn1dm/dm mice are resistant to isoproterenol-induced cardiac injury and autophagy. CCN1/α6β1 interaction in cardiomyocytes promotes autophagy via ROS production and activation of ERK and JNK [386], further implicating CCN1 in the pathogenesis of CVDs and highlighting its potential as a target for therapeutic intervention.
CCN proteins stimulate angiogenesis, not only by promoting the migration, adhesion, and survival of vascular ECs but also by facilitating cell communication networks, which centralize the coordination of various growth factors and proteins, and ultimately induce the formation of new blood vessels [387]. In addition, CCN1 via αvβ3 activates VEGFR2 and downstream MAPK/PI3K signaling pathways, YAP/TAZ, as well as Rho effector mDia1 to enhance tip cell activity and autoregulate its own expression [388] (Figure 4A). Inhibition of αvβ3-mediated signaling represses tip cell number and sprouting in retinas of EC-specific Ccn1 transgenic mice, and allograft tumors in Ccn1-overexpressing mice have hyperactive vascular sprouting [388].
CCN1 activates various signaling pathways, such as ILK/Akt, MEK/ERK, and Wnt/β-catenin axis in various cell types [389,390,391,392]. In ECs, CCN1-induced by OSS promotes superoxide production, NF-κB activation, and expression of inflammatory genes [371,385]. This suggests a pro-inflammatory role of CCN1 in ECs under disturbed flow conditions. In addition, a previous study has demonstrated that the knockdown of CCN1 in human retinal vascular ECs reduces NOX4 expression and inhibits ROS production [393]. Consistently, treatment with recombinant CCN1 stimulates generation of ROS in human/mouse neutrophils, further supporting its role in promoting oxidative stress [394]. In macrophages, CCN1 interacts with integrin αMβ2 and syndecan-4 to enhance cell adhesion and activate NF-κB-mediated pro-inflammatory responses as evidenced by elevated levels of TNF-α, IL-1α, IL-1β, IL-6, and IL-12b [395]. CCN1 is also reported to be expressed in VSMCs of atheromatous plaques, where its expression is regulated by Ang II [379]. Consistently, CCN1 knockdown significantly suppresses neointimal hyperplasia in rats after 14 and 28 days of vascular injury [396]. CCN1 activation has several downstream effects pivotal for inflammation and tissue repair processes. It modulates immune cells (lymphocytes and monocytes) migration in a biphasic manner. Initially, it enhances actin polymerization and transwell migration, but prolonged exposure to CCN1 inhibits immune cell migration by suppressing the activation status of PI3Kγ, p38, and Akt [397]. Furthermore, CCN1 promotes the expression of various pro-inflammatory cytokines and chemokines in macrophages [395]. Collectively, existing literature suggests that novel approaches targeting CCN1 expression or secretion may hold therapeutic potential for treating atherosclerosis by attenuating endothelial dysfunction, foam cell formation, and hepatic lipid accumulation.

4.6. Secreted Protein Acidic and Rich in Cysteine

Secreted Protein Acidic and Rich in Cysteine (SPARC), also known as osteonectin (ON) or BM-40, is a calcium-binding glycoprotein [398,399,400]. It interacts with various components of the ECM, including collagen and fibronectin, and is found in several tissues and organs, such as bone, cartilage, skin, and the ECM of tumors [401,402]. Recent studies have implicated SPARC in vascular pathologies. Hu et al. reported elevated expression of SPARC in VSMCs of atherosclerotic rats and demonstrated a reduction in its expression with aerobic exercise [403]. Similarly, Li et al. observed higher serum levels of SPARC in hypertensive rats, with a positive correlation between SPARC levels and elevated BP [46]. Mechanistically, SPARC has been shown to inhibit endothelium-dependent vessel relaxation, suggesting a potential involvement in vascular dysfunction. However, the precise role of SPARC in atherosclerosis development, including its cell-specific functions, is far from being completely understood.
SPARC is known to modulate various cellular processes in ECs, including adhesion, migration, proliferation, and differentiation, consequently affecting angiogenesis via regulating responses to different growth factors [404,405,406,407,408]. Growth factors like TGF-β, PDGF, and IGF-1 have been demonstrated to stimulate the synthesis of SPARC in chondrocytes [401]. Additional experiments have shown that these growth factors rescue from the inhibitory effects of IL-1 on SPARC synthesis. Similarly, Sage et al. reported increased SPARC levels in aortic ECs exposed to endotoxin and an association between SPARC levels and proliferative cells in vivo, suggesting its cell proliferative effects [408]. Conversely, SPARC has been found to inhibit DNA synthesis and suppress ERK1/2 activation in VEGF-treated ECs by directly binding to VEGF and preventing its interaction with its cell surface receptors [407]. Other studies have also revealed similar findings that SPARC suppresses EC cycle progression [409], inhibits binding of PDGF-AB and -BB to its receptors [410], and regulate EC morphology and barrier function [411]. Interestingly, the role of SPARC has also been investigated under oxidative stress conditions within the tumor microenvironment. Studies using HUVECs have shown that SPARC expression is downregulated in the presence of H2O2 compared to control conditions; however, SPARC levels can be restored upon ROS inhibition with diphenyleneiodonium [412].
SPARC expression in cultured human cerebral MVECs positively correlates with cell proliferation and reduces as these cells mature and form a confluent monolayer [413]. Besides, treatment with recombinant SPARC increases permeability and decreases transendothelial electrical resistance. These findings indicate that SPARC may significantly affect the function of cerebral microvessels during development and inflammation at the blood–brain barrier. In another study, it was reported that the deficiency of SPARC inhibits endoglin-mediated pericyte migration, alters endoglin binding in focal complexes, increases TGF-β activation via αV integrin pathway, and results in decreased pericyte-associated vessels in an orthotopic model of pancreatic cancer [414]. These results highlight the function of SPARC in promoting pericyte migration through TGF-β. Further, these studies provide valuable insights into the role of SPARC in EC inflammation and its potential effects on vascular diseases and repair mechanisms. As described above, SPARC exhibits an anti-angiogenic role by either impeding the interaction of growth factors with their cell surface receptors or modulating the expression of MMPs and TGF-β1 [415,416,417]. This anti-angiogenic function is abolished in SPARC knockout mice [404,407]. Conversely, certain cleavage products of SPARC or intact SPARC have been observed to promote angiogenesis [408,418].

5. Therapeutic Potential of Targeting Different Matricellular Proteins

Matricellular proteins are minimally expressed in healthy adult tissues but get upregulated in response to stress, injury, or disease. As described above, various matricellular proteins have been demonstrated to promote atherosclerosis via stimulating vascular inflammation and regulating the phenotype of different vascular and immune cells involved in its pathogenesis. Therefore, targeting the signaling mechanisms induced by these proteins and/or regulating their expression may offer significant therapeutic potential to reduce and even regress plaque progression.
TSP1 is a well-known antiangiogenic matricellular protein that can either promote or inhibit inflammation, depending on the cellular context. In atherosclerosis, its therapeutic relevance primarily lies in the inhibition of TSP1-induced CD47 activation. TSP1 via CD47 activation in LECs inhibits lymphangiogenesis [246]. Earlier studies have demonstrated that lymphatic vasculature present in the adventitial layer of the arterial wall represents the primary route of cholesterol removal from atherosclerotic vessels, and improved lymphatic function leads to atherosclerosis regression [419,420]. We have previously demonstrated that blockade of TSP1-induced CD47 activation in LECs promotes in vitro lymphangiogenesis and mice with LEC-restricted Cd47 deletion have enhanced arterial lymphatic vessel density and attenuated atherosclerosis [246]. This signaling axis also promotes EC senescence, contributing to vascular aging and dysfunction [245], thereby, inhibiting TSP1-CD47 interactions may help reverse age-related endothelial impairments and atherosclerosis. Additionally, TSP1 suppresses NO production, a key regulator of vasodilation and vascular homeostasis. Therapeutic strategies targeting TSP1 to restore NO bioavailability represent another promising approach to combat vascular inflammation and dysfunction [273]. This information supports the therapeutic potential of blocking the TSP1-CD47 axis for preventing vascular inflammation, compromised endothelial barrier integrity, and atherosclerosis. Consistently, Kojima et al. demonstrated that a CD47-blocking antibody originally developed as an anti-cancer drug improves efferocytic removal of apoptotic cells from the vascular wall, leading to attenuated vascular inflammation and atherosclerosis in mice [421]. Jarr et al. reported in a phase 1b-2 trial that a humanized anti-CD47 antibody magrolimab induces tumor reduction in patients with relapsed or refractory lymphoma or acute myeloid leukemia (AML) [422]. Additionally, the authors reported reduced vascular inflammation in those patients as determined by 18F-fluorodeoxyglucose uptake in carotid arteries. Several other CD47-blocking antibodies are currently in various stages of clinical trials targeting different types of cancer (Supplementary Table S1) [423,424]. In addition, macrophage-specific nanotherapy approaches have been explored to inhibit the CD47-SIRPα antiphagocytic signaling axis [425]. For example, a novel nanotherapy using single-walled carbon nanotubes (SWNTs) loaded with an SHP1 inhibitor (SWNT-SHP1i) has been developed for systemic blockade of CD47 signaling. This approach was tested in a porcine model (large animal model) of early atherosclerosis [426], demonstrating the potential for targeted therapeutic strategies aimed at enhancing efferocytosis in atherosclerotic cardiovascular disease.
Singla et al. demonstrated that blockade of RSPO2–LGR4 signaling via perivascular application of Rspo2’s decoy receptor attenuates atherosclerosis. A mouse LGR4/Gpr48 Fc chimeric protein, containing the N-terminal extracellular domain (ECD) of LGR4, was used for this purpose. The ECD of LGR4 is the recognized binding site for RSPO ligands, and LGR4-ECD has been shown to effectively inhibit LGR4 signaling both in vitro and in vivo [220]. The antibodies 130M23, 130M24, 130M25, 130M26, 130M27, and 130M28 are specific inhibitors of RSPO2 and have been shown to reduce or completely block RSPO2-induced β-catenin signaling (Patent No.: US 9,644,034). These antibodies may serve as blocking agents to disrupt RSPO2-LGR4 interactions. Additionally, inhibition of Wnt signaling using small molecule inhibitors, recombinant proteins, or neutralizing antibodies has been shown to ameliorate atherosclerosis in preclinical models. For example, the porcupine inhibitor GNF-6231 and XAV939, small molecules that inhibit poly ADP-ribose polymerase tankyrase 1 and tankyrase 2, have demonstrated efficacy in this context. Similarly, LGK974 and SFRP4 are reported to block Wnt/β-catenin signaling [427]. Therefore, inhibition of Rspo2-induced LGR4 activation and downstream Wnt signaling utilizing recombinant proteins, blocking antibodies, and small molecule inhibitors has the potential to reduce atherosclerosis; however, this possibility requires further investigation.
OPN contains an RGD sequence that binds to integrins and CD44 on monocytes, macrophages, and immune cells, promoting their recruitment to plaques. This interaction drives macrophage activation, foam cell formation, and VSMCs migration, contributing to inflammation and plaque progression [428]. Blocking OPN-integrin binding reduces its pro-inflammatory effects. While primarily pro-atherogenic, OPN also inhibits vascular calcification, which can paradoxically stabilize plaques. Thus, OPN-targeted therapies should be able to balance suppress the inflammation while preserving OPN’s anti-calcific role [429]. Targeted drug delivery using nanoparticles, for example, those carrying the PPARδ agonist GW1516, has shown promise in reducing VSMC migration and apoptosis [430].
TN-C levels are upregulated in atherosclerotic lesions. Several peptides and vector fragments, including G11, G11-iRGD, TN11, PL1, and PL3 have been developed that recognize TN-C domains and can be used to direct therapeutic or diagnostic payloads to plaque tissue. Furthermore, ATN-RNA and IMA950 were investigated in clinical trials as therapeutic drugs and vaccines by targeting TN-C, respectively [431]. In other cardiovascular-remodeling settings (post-myocardial infarction), modulating TN-C may also be relevant—either inhibiting its excessive up-regulation (which promotes adverse ventricular remodeling via macrophage polarization to a pro-inflammatory M1 phenotype through TLR4) or harnessing its transient, wound-healing role during acute repair.
In short, a defining feature of these matricellular proteins is their context-dependent dual role in tissue remodeling, inflammation, and repair. Their biological (protective or pathogenic) effects are highly regulated by the cellular microenvironment, stage of disease, and type/duration of injury. For example, certain matricellular proteins promote cell adhesion, migration, proliferation, and repair during acute tissue injury, thereby supporting wound healing. However, the same molecules may drive fibrosis, chronic inflammation, or atherogenesis when their levels are persistently elevated. For example, TSP1 inhibits angiogenesis by activating TGF-β and suppressing EC proliferation, yet in other settings, it promotes tumor progression by enhancing matrix remodeling, cell migration, and cell invasive behavior. Similarly, OPN contributes to wound healing by promoting cell survival and migration, but its long-term higher expression induces pathological fibrosis, chronic inflammation, and tumor metastasis. These functional differences may arise from the expression levels of various interacting partners (integrins, CD44, CD47, CD36, LRP receptors), post-translational modifications, and crosstalk with other matricellular proteins and signaling molecules. Therefore, a precise temporal and spatial understanding of their protein expression and downstream signaling pathways is required for elucidating their multifaceted roles. Such insights will be critical to design specific targeted therapies to promote their protective functions while mitigating their deleterious effects in individual pathologies.

6. Conclusions and Future Perspectives

Atherosclerosis is the underlying cause of CVD-related mortality worldwide. Among the various cell types involved in the pathogenesis of atherosclerosis, EC dysfunction triggered by disturbed blood flow or other proatherogenic risk factors is the initiating event in the pathogenesis of atherosclerosis. Matricellular proteins are recognized to play important roles in EC inflammation and the progression of atherosclerosis. Elevated levels of these proteins in arteries correlate with atherosclerosis; however, their precise role, either deleterious or protective during atherogenesis, requires further investigations. In vascular ECs, these proteins promote inflammation by facilitating leukocyte adhesion and transmigration across the endothelium. Moreover, they contribute to the chronic inflammatory environment characteristic of atherosclerosis by inducing pro-inflammatory signaling in ECs and remodeling of ECM. Additionally, these proteins regulate plaque stability by affecting VSMC proliferation and migration, as well as collagen deposition in the fibrous cap. Given the complex pathogenesis of atherosclerosis, further research on how these proteins stimulate EC damage, activation, inflammation, and plaque formation, and identification of involved cell surface receptor(s) is warranted. Novel animal models, including EC-restricted knockout/overexpression mice with/without lineage-tracing capabilities, advanced imaging techniques, and transcriptomic sequencing, would be useful in uncovering the precise mechanisms stimulated by these proteins in ECs during disease progression. Additionally, the development of novel pharmacological small-molecule inhibitors or blocking antibodies is crucial to explore the translational potential of inhibiting identified pathways. While gene-deficient mouse models provide valuable mechanistic insights, they may not fully replicate the outcome of pharmacological inhibition, as small molecules and antibodies may have off-target and adverse effects. Currently, limited information is available on the roles of matricellular proteins in regulating ROS generation, redox-sensitive signaling, and autophagy during endothelial dysfunction; therefore, future studies are warranted on these aspects. Similarly, the effects of matricellular proteins on LDL uptake and clearance by ECs remain underexplored, despite their relevance in atherogenesis.
Matricellular proteins may also play an important role in regulating the liver-vascular axis and consequently governing atherogenesis. Supporting this concept, our lab has recently demonstrated that nidogen-2 (NID2) levels are upregulated in both human atherosclerotic arteries and murine steatotic livers [432]. Adeno-associated virus (AAV)-mediated overexpression of NID2 in a mouse model promotes both hepatic steatosis and atherosclerotic lesion formation. Notably, since intraperitoneal injection of AAV primarily induces gene expression in the liver, it is plausible that liver-derived NID2 contributes to the observed vascular phenotype. However, future studies with hepatocyte-specific NID2 deletion/overexpression are required to establish its role in liver–vascular axis. Further, Hepatocyte RSPO2 has been identified as a key suppressor of hepatosteatosis [337], a condition closely associated with atherosclerosis. Therefore, it is tempting to speculate on the beneficial role of hepatocyte RSPO2 in atherosclerosis in contrast to its detrimental role in vascular cells. However, the role of matricellular proteins secreted from the liver and binding to their cognate receptors present on vascular cells remains to be investigated.
To better understand the expression of matricellular proteins in ECs at the single-cell level and across different stages of atherogenesis (early and advanced), studies using global or cell-specific knockout mice, combined with transcriptomic profiling of ECs during inflammation and atherogenesis with single-cell and/or bulk RNA sequencing (RNA-seq) of aortic ECs, are warranted. Some limited RNA-seq studies have been reported, such as those investigating SMCs and atherosclerosis using a CD47 SMC-specific knockout mouse model [433]. However, RNA-seq studies focusing on matricellular proteins, EC inflammation, and atherogenesis remain underexplored.
Despite knowing that matricellular proteins contribute to atherogenesis, targeting these presents significant challenges. They often exhibit both pro- and anti-inflammatory effects depending on their expression level, tissue type, disease stage, and microenvironment. Their expression is transient, spatially restricted, and induced by stress or injury, complicating timing and delivery of therapy. These proteins bind multiple, often weakly interacting partners/receptors (e.g., integrins, CD44, CD47, CD36, growth factors), making it difficult to block specific interactions without off-target effects. Being embedded in the ECM also limits drug accessibility, and large molecules, such as blocking antibodies, may not penetrate enough dense stroma to inhibit the respective signaling. Finally, as key mediators of tissue repair and remodeling, their inhibition risks disrupting normal physiological processes. These drug development challenges may be overcome with different potential strategies: (i) Antibody-drug conjugates or targeted delivery systems to selectively deliver therapeutics to matricellular protein-expressing tissues. (ii) Peptide inhibitors or mimetics that block specific binding domains. (iii) RNA-based therapeutics (e.g., siRNA, antisense oligonucleotides-ASOs) with potential tissue specificity.
Addressing these knowledge gaps will be useful in advancing our understanding of matricellular protein biology and for developing effective therapeutic strategies for CVDs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox14111338/s1, Table S1. Overview of clinical trials on various CD47-blocking antibodies.

Author Contributions

R.V.A. wrote, edited, and proofread the manuscript, and created figures using BioRender. S.K. proofread the article. B.S. contributed to manuscript editing, proofreading, and figure preparation. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health (NIH) grants K99HL146954, R00HL146954, and R01HL172809 and the UTHSC-College of Pharmacy Startup and Seed Research Grant awarded to BS.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

The authors thank Umamaheswara Sastry Pujyam, B.S. Biology Program, Stanford University, Stanford, CA 94305, USA, for his help with proofreading of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ECMExtracellular Matrix
EC/ECsEndothelial Cells
CVDsCardiovascular Diseases
LDLLow-Density Lipoprotein
VSMCsVascular Smooth Muscle Cells
CADCoronary Artery Disease
NONitric Oxide
TNF-αTumor Necrosis Factor-alpha
IL-1Interleukin-1
ICAM-1Intercellular Adhesion Molecule-1
VCAM-1Vascular Cell Adhesion Molecule-1
NF-κBNuclear Factor-Kappa B
MAPKMitogen-Activated Protein Kinase
MCP-1Monocyte Chemoattractant Protein-1
EndMTEndothelial-to-Mesenchymal Transition
ROSReactive oxygen species
NOXsNADPH Oxidases
O2•−Superoxide Ion
SODSuperoxide Dismutase
TSPsThrombospondins
OPNOsteopontin
RSPOsRoof Plate-Specific Spondins
TN-CTenascin-C
SPARCSecreted Protein Acidic and Rich in Cysteine
HUVECsHuman Umbilical Vein ECs
AMLAcute Myeloid Leukemia

References

  1. Piepoli, M.F.; Hoes, A.W.; Agewall, S.; Albus, C.; Brotons, C.; Catapano, A.L.; Cooney, M.T.; Corra, U.; Cosyns, B.; Deaton, C.; et al. 2016 European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts)Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR). Eur. Heart J. 2016, 37, 2315–2381. [Google Scholar] [CrossRef] [PubMed]
  2. Martin, S.S.; Aday, A.W.; Allen, N.B.; Almarzooq, Z.I.; Anderson, C.A.M.; Arora, P.; Avery, C.L.; Baker-Smith, C.M.; Bansal, N.; Beaton, A.Z.; et al. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025, 151, e41–e660. [Google Scholar] [CrossRef] [PubMed]
  3. Stary, H.C. The sequence of cell and matrix changes in atherosclerotic lesions of coronary arteries in the first forty years of life. Eur. Heart J. 1990, 11 (Suppl. E), 3–19. [Google Scholar] [CrossRef]
  4. Wang, Y.; Nanda, V.; Direnzo, D.; Ye, J.; Xiao, S.; Kojima, Y.; Howe, K.L.; Jarr, K.U.; Flores, A.M.; Tsantilas, P.; et al. Clonally expanding smooth muscle cells promote atherosclerosis by escaping efferocytosis and activating the complement cascade. Proc. Natl. Acad. Sci. USA 2020, 117, 15818–15826. [Google Scholar] [CrossRef]
  5. Depuydt, M.A.C.; Prange, K.H.M.; Slenders, L.; Ord, T.; Elbersen, D.; Boltjes, A.; de Jager, S.C.A.; Asselbergs, F.W.; de Borst, G.J.; Aavik, E.; et al. Microanatomy of the Human Atherosclerotic Plaque by Single-Cell Transcriptomics. Circ. Res. 2020, 127, 1437–1455. [Google Scholar] [CrossRef]
  6. Virmani, R.; Kolodgie, F.D.; Burke, A.P.; Farb, A.; Schwartz, S.M. Lessons from sudden coronary death: A comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1262–1275. [Google Scholar] [CrossRef] [PubMed]
  7. Davignon, J.; Ganz, P. Role of endothelial dysfunction in atherosclerosis. Circulation 2004, 109, III27–III32. [Google Scholar] [CrossRef]
  8. Cahill, P.A.; Redmond, E.M. Vascular endothelium—Gatekeeper of vessel health. Atherosclerosis 2016, 248, 97–109. [Google Scholar] [CrossRef]
  9. Ricard, N.; Bailly, S.; Guignabert, C.; Simons, M. The quiescent endothelium: Signalling pathways regulating organ-specific endothelial normalcy. Nat. Rev. Cardiol. 2021, 18, 565–580. [Google Scholar] [CrossRef]
  10. Medina-Leyte, D.J.; Zepeda-Garcia, O.; Dominguez-Perez, M.; Gonzalez-Garrido, A.; Villarreal-Molina, T.; Jacobo-Albavera, L. Endothelial Dysfunction, Inflammation and Coronary Artery Disease: Potential Biomarkers and Promising Therapeutical Approaches. Int. J. Mol. Sci. 2021, 22, 3850. [Google Scholar] [CrossRef]
  11. Pervaiz, N.; Kathuria, I.; Aithabathula, R.V.; Singla, B. Matricellular proteins in atherosclerosis development. Matrix Biol. 2023, 120, 1–23. [Google Scholar] [CrossRef]
  12. Bornstein, P. Diversity of function is inherent in matricellular proteins: An appraisal of thrombospondin 1. J. Cell Biol. 1995, 130, 503–506. [Google Scholar] [CrossRef]
  13. Kong, P.; Cui, Z.Y.; Huang, X.F.; Zhang, D.D.; Guo, R.J.; Han, M. Inflammation and atherosclerosis: Signaling pathways and therapeutic intervention. Signal Transduct. Target. Ther. 2022, 7, 131. [Google Scholar] [CrossRef]
  14. Ivanova, E.A.; Myasoedova, V.A.; Melnichenko, A.A.; Grechko, A.V.; Orekhov, A.N. Small Dense Low-Density Lipoprotein as Biomarker for Atherosclerotic Diseases. Oxid. Med. Cell Longev. 2017, 2017, 1273042. [Google Scholar] [CrossRef]
  15. Mauersberger, C.; Hinterdobler, J.; Schunkert, H.; Kessler, T.; Sager, H.B. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front. Cardiovasc. Med. 2021, 8, 813984. [Google Scholar] [CrossRef]
  16. Rafieian-Kopaei, M.; Setorki, M.; Doudi, M.; Baradaran, A.; Nasri, H. Atherosclerosis: Process, indicators, risk factors and new hopes. Int. J. Prev. Med. 2014, 5, 927–946. [Google Scholar] [PubMed]
  17. Libby, P. Inflammation in atherosclerosis. Nature 2002, 420, 868–874. [Google Scholar] [CrossRef] [PubMed]
  18. 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]
  19. Forstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
  20. Loeffen, R.; Spronk, H.M.; ten Cate, H. The impact of blood coagulability on atherosclerosis and cardiovascular disease. J. Thromb. Haemost. 2012, 10, 1207–1216. [Google Scholar] [CrossRef]
  21. Girolami, A.; Sambado, L.; Lombardi, A.M. The impact of blood coagulability on atherosclerosis and cardiovascular disease: A rebuttal. J. Thromb. Haemost. 2013, 11, 213–214, discussion 215–216. [Google Scholar] [CrossRef] [PubMed]
  22. Botts, S.R.; Fish, J.E.; Howe, K.L. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front. Pharmacol. 2021, 12, 787541. [Google Scholar] [CrossRef]
  23. Zhu, Y.; Liao, H.; Wang, N.; Ma, K.S.; Verna, L.K.; Shyy, J.Y.; Chien, S.; Stemerman, M.B. LDL-activated p38 in endothelial cells is mediated by Ras. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1159–1164. [Google Scholar] [CrossRef]
  24. Pietersma, A.; Tilly, B.C.; Gaestel, M.; de Jong, N.; Lee, J.C.; Koster, J.F.; Sluiter, W. p38 mitogen activated protein kinase regulates endothelial VCAM-1 expression at the post-transcriptional level. Biochem. Biophys. Res. Commun. 1997, 230, 44–48. [Google Scholar] [CrossRef] [PubMed]
  25. Reustle, A.; Torzewski, M. Role of p38 MAPK in Atherosclerosis and Aortic Valve Sclerosis. Int. J. Mol. Sci. 2018, 19, 3761. [Google Scholar] [CrossRef]
  26. Gan, J.; Guo, L.; Zhang, X.; Yu, Q.; Yang, Q.; Zhang, Y.; Zeng, W.; Jiang, X.; Guo, M. Anti-inflammatory therapy of atherosclerosis: Focusing on IKKbeta. J. Inflamm. 2023, 20, 8. [Google Scholar] [CrossRef]
  27. Hajra, L.; Evans, A.I.; Chen, M.; Hyduk, S.J.; Collins, T.; Cybulsky, M.I. The NF-kappa B signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proc. Natl. Acad. Sci. USA 2000, 97, 9052–9057. [Google Scholar] [CrossRef]
  28. Chen, Y.; Ye, X.; Escames, G.; Lei, W.; Zhang, X.; Li, M.; Jing, T.; Yao, Y.; Qiu, Z.; Wang, Z.; et al. The NLRP3 inflammasome: Contributions to inflammation-related diseases. Cell Mol. Biol. Lett. 2023, 28, 51. [Google Scholar] [CrossRef]
  29. Blevins, H.M.; Xu, Y.; Biby, S.; Zhang, S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front. Aging Neurosci. 2022, 14, 879021. [Google Scholar] [CrossRef]
  30. Chen, M.Y.; Ye, X.J.; He, X.H.; Ouyang, D.Y. The Signaling Pathways Regulating NLRP3 Inflammasome Activation. Inflammation 2021, 44, 1229–1245. [Google Scholar] [CrossRef]
  31. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  32. Zgorzynska, E.; Dziedzic, B.; Walczewska, A. An Overview of the Nrf2/ARE Pathway and Its Role in Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 9592. [Google Scholar] [CrossRef]
  33. Mimura, J.; Itoh, K. Role of Nrf2 in the pathogenesis of atherosclerosis. Free Radic. Biol. Med. 2015, 88, 221–232. [Google Scholar] [CrossRef]
  34. Braile, M.; Marcella, S.; Cristinziano, L.; Galdiero, M.R.; Modestino, L.; Ferrara, A.L.; Varricchi, G.; Marone, G.; Loffredo, S. VEGF-A in Cardiomyocytes and Heart Diseases. Int. J. Mol. Sci. 2020, 21, 5294. [Google Scholar] [CrossRef] [PubMed]
  35. Yoshimura, A.; Wakabayashi, Y.; Mori, T. Cellular and molecular basis for the regulation of inflammation by TGF-beta. J. Biochem. 2010, 147, 781–792. [Google Scholar] [CrossRef] [PubMed]
  36. Kang, S.; Kishimoto, T. Interplay between interleukin-6 signaling and the vascular endothelium in cytokine storms. Exp. Mol. Med. 2021, 53, 1116–1123. [Google Scholar] [CrossRef]
  37. Immanuel, J.; Yun, S. Vascular Inflammatory Diseases and Endothelial Phenotypes. Cells 2023, 12, 1640. [Google Scholar] [CrossRef] [PubMed]
  38. Hu, D.; Yin, C.; Luo, S.; Habenicht, A.J.R.; Mohanta, S.K. Vascular Smooth Muscle Cells Contribute to Atherosclerosis Immunity. Front. Immunol. 2019, 10, 1101. [Google Scholar] [CrossRef]
  39. Kwaifa, I.K.; Bahari, H.; Yong, Y.K.; Noor, S.M. Endothelial Dysfunction in Obesity-Induced Inflammation: Molecular Mechanisms and Clinical Implications. Biomolecules 2020, 10, 291. [Google Scholar] [CrossRef]
  40. Dhananjayan, R.; Koundinya, K.S.; Malati, T.; Kutala, V.K. Endothelial Dysfunction in Type 2 Diabetes Mellitus. Indian. J. Clin. Biochem. 2016, 31, 372–379. [Google Scholar] [CrossRef]
  41. Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef]
  42. Sitia, S.; Tomasoni, L.; Atzeni, F.; Ambrosio, G.; Cordiano, C.; Catapano, A.; Tramontana, S.; Perticone, F.; Naccarato, P.; Camici, P.; et al. From endothelial dysfunction to atherosclerosis. Autoimmun. Rev. 2010, 9, 830–834. [Google Scholar] [CrossRef]
  43. Hunt, B.J.; Jurd, K.M. Endothelial cell activation. A central pathophysiological process. BMJ 1998, 316, 1328–1329. [Google Scholar] [CrossRef]
  44. Yau, J.W.; Teoh, H.; Verma, S. Endothelial cell control of thrombosis. BMC Cardiovasc. Disord. 2015, 15, 130. [Google Scholar] [CrossRef]
  45. Yang, X.; Chang, Y.; Wei, W. Endothelial Dysfunction and Inflammation: Immunity in Rheumatoid Arthritis. Mediat. Inflamm. 2016, 2016, 6813016. [Google Scholar] [CrossRef]
  46. Dolmatova, E.V.; Wang, K.; Mandavilli, R.; Griendling, K.K. The effects of sepsis on endothelium and clinical implications. Cardiovasc. Res. 2021, 117, 60–73. [Google Scholar] [CrossRef]
  47. Chen, J.; Somanath, P.R.; Razorenova, O.; Chen, W.S.; Hay, N.; Bornstein, P.; Byzova, T.V. Akt1 regulates pathological angiogenesis, vascular maturation and permeability in vivo. Nat. Med. 2005, 11, 1188–1196. [Google Scholar] [CrossRef]
  48. Lee, M.Y.; Luciano, A.K.; Ackah, E.; Rodriguez-Vita, J.; Bancroft, T.A.; Eichmann, A.; Simons, M.; Kyriakides, T.R.; Morales-Ruiz, M.; Sessa, W.C. Endothelial Akt1 mediates angiogenesis by phosphorylating multiple angiogenic substrates. Proc. Natl. Acad. Sci. USA 2014, 111, 12865–12870. [Google Scholar] [CrossRef]
  49. Ackah, E.; Yu, J.; Zoellner, S.; Iwakiri, Y.; Skurk, C.; Shibata, R.; Ouchi, N.; Easton, R.M.; Galasso, G.; Birnbaum, M.J.; et al. Akt1/protein kinase Balpha is critical for ischemic and VEGF-mediated angiogenesis. J. Clin. Investig. 2005, 115, 2119–2127. [Google Scholar] [CrossRef]
  50. Szmitko, P.E.; Wang, C.H.; Weisel, R.D.; de Almeida, J.R.; Anderson, T.J.; Verma, S. New markers of inflammation and endothelial cell activation: Part I. Circulation 2003, 108, 1917–1923. [Google Scholar] [CrossRef]
  51. Badimon, L.; Pena, E.; Arderiu, G.; Padro, T.; Slevin, M.; Vilahur, G.; Chiva-Blanch, G. C-Reactive Protein in Atherothrombosis and Angiogenesis. Front. Immunol. 2018, 9, 430. [Google Scholar] [CrossRef]
  52. Barthel, S.R.; Gavino, J.D.; Descheny, L.; Dimitroff, C.J. Targeting selectins and selectin ligands in inflammation and cancer. Expert Opin. Ther. Targets 2007, 11, 1473–1491. [Google Scholar] [CrossRef]
  53. Kaur, R.; Singh, V.; Kumari, P.; Singh, R.; Chopra, H.; Emran, T.B. Novel insights on the role of VCAM-1 and ICAM-1: Potential biomarkers for cardiovascular diseases. Ann. Med. Surg. 2022, 84, 104802. [Google Scholar] [CrossRef]
  54. Sutton, G.; Pugh, D.; Dhaun, N. Developments in the Role of Endothelin-1 in Atherosclerosis: A Potential Therapeutic Target? Am. J. Hypertens. 2019, 32, 813–815. [Google Scholar] [CrossRef]
  55. Harrington, J.R. The role of MCP-1 in atherosclerosis. Stem Cells 2000, 18, 65–66. [Google Scholar] [CrossRef]
  56. Lin, J.; Kakkar, V.; Lu, X. Impact of MCP-1 in atherosclerosis. Curr. Pharm. Des. 2014, 20, 4580–4588. [Google Scholar] [CrossRef]
  57. Picchi, A.; Gao, X.; Belmadani, S.; Potter, B.J.; Focardi, M.; Chilian, W.M.; Zhang, C. Tumor necrosis factor-alpha induces endothelial dysfunction in the prediabetic metabolic syndrome. Circ. Res. 2006, 99, 69–77. [Google Scholar] [CrossRef]
  58. Li, X.; Zhao, W.; Li, X.; Chen, X.; Li, Y.; He, J.; Qin, Y.; Li, L.; Zhang, H. The association of SPARC with hypertension and its function in endothelial-dependent relaxation. Atherosclerosis 2024, 388, 117390. [Google Scholar] [CrossRef]
  59. Chen, P.Y.; Qin, L.; Li, G.; Wang, Z.; Dahlman, J.E.; Malagon-Lopez, J.; Gujja, S.; Cilfone, N.A.; Kauffman, K.J.; Sun, L.; et al. Endothelial TGF-beta signalling drives vascular inflammation and atherosclerosis. Nat. Metab. 2019, 1, 912–926. [Google Scholar] [CrossRef]
  60. Li, A.; Dubey, S.; Varney, M.L.; Dave, B.J.; Singh, R.K. IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis. J. Immunol. 2003, 170, 3369–3376. [Google Scholar] [CrossRef]
  61. Yu, H.; Huang, X.; Ma, Y.; Gao, M.; Wang, O.; Gao, T.; Shen, Y.; Liu, X. Interleukin-8 regulates endothelial permeability by down-regulation of tight junction but not dependent on integrins induced focal adhesions. Int. J. Biol. Sci. 2013, 9, 966–979. [Google Scholar] [CrossRef] [PubMed]
  62. Doring, Y.; van der Vorst, E.P.C.; Duchene, J.; Jansen, Y.; Gencer, S.; Bidzhekov, K.; Atzler, D.; Santovito, D.; Rader, D.J.; Saleheen, D.; et al. CXCL12 Derived From Endothelial Cells Promotes Atherosclerosis to Drive Coronary Artery Disease. Circulation 2019, 139, 1338–1340. [Google Scholar] [CrossRef]
  63. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, C.; Tousoulis, D. Inflammatory Mechanisms Contributing to Endothelial Dysfunction. Biomedicines 2021, 9, 781. [Google Scholar] [CrossRef]
  64. 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]
  65. Alfaddagh, A.; Martin, S.S.; Leucker, T.M.; Michos, E.D.; Blaha, M.J.; Lowenstein, C.J.; Jones, S.R.; Toth, P.P. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am. J. Prev. Cardiol. 2020, 4, 100130. [Google Scholar] [CrossRef]
  66. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  67. Bloom, S.I.; Islam, M.T.; Lesniewski, L.A.; Donato, A.J. Mechanisms and consequences of endothelial cell senescence. Nat. Rev. Cardiol. 2023, 20, 38–51. [Google Scholar] [CrossRef]
  68. Han, Y.; Kim, S.Y. Endothelial senescence in vascular diseases: Current understanding and future opportunities in senotherapeutics. Exp. Mol. Med. 2023, 55, 1–12. [Google Scholar] [CrossRef]
  69. Yang, Z.; Li, H.; Luo, P.; Yan, D.; Yang, N.; Zhang, Y.; Huang, Y.; Liu, Y.; Zhang, L.; Yan, J.; et al. UNC5B Promotes Vascular Endothelial Cell Senescence via the ROS-Mediated P53 Pathway. Oxid. Med. Cell Longev. 2021, 2021, 5546711. [Google Scholar] [CrossRef]
  70. Davalli, P.; Mitic, T.; Caporali, A.; Lauriola, A.; D’Arca, D. ROS, Cell Senescence, and Novel Molecular Mechanisms in Aging and Age-Related Diseases. Oxid. Med. Cell Longev. 2016, 2016, 3565127. [Google Scholar] [CrossRef]
  71. Donato, A.J.; Machin, D.R.; Lesniewski, L.A. Mechanisms of Dysfunction in the Aging Vasculature and Role in Age-Related Disease. Circ. Res. 2018, 123, 825–848. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, X.; Sessa, W.C.; Fernandez-Hernando, C. Endothelial Transcytosis of Lipoproteins in Atherosclerosis. Front. Cardiovasc. Med. 2018, 5, 130. [Google Scholar] [CrossRef]
  73. Ramirez, C.M.; Zhang, X.; Bandyopadhyay, C.; Rotllan, N.; Sugiyama, M.G.; Aryal, B.; Liu, X.; He, S.; Kraehling, J.R.; Ulrich, V.; et al. Caveolin-1 Regulates Atherogenesis by Attenuating Low-Density Lipoprotein Transcytosis and Vascular Inflammation Independently of Endothelial Nitric Oxide Synthase Activation. Circulation 2019, 140, 225–239. [Google Scholar] [CrossRef]
  74. Huang, L.; Chambliss, K.L.; Gao, X.; Yuhanna, I.S.; Behling-Kelly, E.; Bergaya, S.; Ahmed, M.; Michaely, P.; Luby-Phelps, K.; Darehshouri, A.; et al. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. Nature 2019, 569, 565–569. [Google Scholar] [CrossRef]
  75. Kraehling, J.R.; Chidlow, J.H.; Rajagopal, C.; Sugiyama, M.G.; Fowler, J.W.; Lee, M.Y.; Zhang, X.; Ramirez, C.M.; Park, E.J.; Tao, B.; et al. Genome-wide RNAi screen reveals ALK1 mediates LDL uptake and transcytosis in endothelial cells. Nat. Commun. 2016, 7, 13516. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, X.; Fernandez-Hernando, C. Transport of LDLs into the arterial wall: Impact in atherosclerosis. Curr. Opin. Lipidol. 2020, 31, 279–285. [Google Scholar] [CrossRef]
  77. Bolanle, I.O.; de Liedekerke Beaufort, G.C.; Weinberg, P.D. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arterioscler. Thromb. Vasc. Biol. 2025, 45, 468–480. [Google Scholar] [CrossRef] [PubMed]
  78. Tamargo, I.A.; Baek, K.I.; Kim, Y.; Park, C.; Jo, H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat. Rev. Cardiol. 2023, 20, 738–753. [Google Scholar] [CrossRef]
  79. Gorelova, A.; Berman, M.; Al Ghouleh, I. Endothelial-to-Mesenchymal Transition in Pulmonary Arterial Hypertension. Antioxid. Redox Signal. 2021, 34, 891–914. [Google Scholar] [CrossRef]
  80. Zhang, Z.; Fang, Z.; Ge, J.; Li, H. Endothelial-to-mesenchymal transition in cardiovascular diseases. Trends Mol. Med. 2025, 25, S1471–S1486. [Google Scholar] [CrossRef]
  81. Chen, P.Y.; Schwartz, M.A.; Simons, M. Endothelial-to-Mesenchymal Transition, Vascular Inflammation, and Atherosclerosis. Front. Cardiovasc. Med. 2020, 7, 53. [Google Scholar] [CrossRef]
  82. Xiong, J.; Kawagishi, H.; Yan, Y.; Liu, J.; Wells, Q.S.; Edmunds, L.R.; Fergusson, M.M.; Yu, Z.X.; Rovira, I.I.; Brittain, E.L.; et al. A Metabolic Basis for Endothelial-to-Mesenchymal Transition. Mol. Cell 2018, 69, 689–698.e7. [Google Scholar] [CrossRef] [PubMed]
  83. Alvandi, Z.; Bischoff, J. Endothelial-Mesenchymal Transition in Cardiovascular Disease. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2357–2369. [Google Scholar] [CrossRef]
  84. Evrard, S.M.; Lecce, L.; Michelis, K.C.; Nomura-Kitabayashi, A.; Pandey, G.; Purushothaman, K.R.; d’Escamard, V.; Li, J.R.; Hadri, L.; Fujitani, K.; et al. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat. Commun. 2016, 7, 11853. [Google Scholar] [CrossRef] [PubMed]
  85. Cho, J.G.; Lee, A.; Chang, W.; Lee, M.S.; Kim, J. Endothelial to Mesenchymal Transition Represents a Key Link in the Interaction between Inflammation and Endothelial Dysfunction. Front. Immunol. 2018, 9, 294. [Google Scholar] [CrossRef] [PubMed]
  86. Hall, I.F.; Kishta, F.; Xu, Y.; Baker, A.H.; Kovacic, J.C. Endothelial to mesenchymal transition: At the axis of cardiovascular health and disease. Cardiovasc. Res. 2024, 120, 223–236. [Google Scholar] [CrossRef]
  87. Kim, M.; Choi, S.H.; Jin, Y.B.; Lee, H.J.; Ji, Y.H.; Kim, J.; Lee, Y.S.; Lee, Y.J. The effect of oxidized low-density lipoprotein (ox-LDL) on radiation-induced endothelial-to-mesenchymal transition. Int. J. Radiat. Biol. 2013, 89, 356–363. [Google Scholar] [CrossRef]
  88. Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [CrossRef]
  89. Shi, X.; Li, P.; Liu, H.; Prokosch, V. Oxidative Stress, Vascular Endothelium, and the Pathology of Neurodegeneration in Retina. Antioxidants 2022, 11, 543. [Google Scholar] [CrossRef]
  90. Singla, B.; Aithabathula, R.V.; Kiran, S.; Kapil, S.; Kumar, S.; Singh, U.P. Reactive Oxygen Species in Regulating Lymphangiogenesis and Lymphatic Function. Cells 2022, 11, 1750. [Google Scholar] [CrossRef]
  91. Meyer, J.W.; Holland, J.A.; Ziegler, L.M.; Chang, M.M.; Beebe, G.; Schmitt, M.E. Identification of a functional leukocyte-type NADPH oxidase in human endothelial cells: A potential atherogenic source of reactive oxygen species. Endothelium 1999, 7, 11–22. [Google Scholar] [CrossRef] [PubMed]
  92. Yan, S.; Zhang, X.; Zheng, H.; Hu, D.; Zhang, Y.; Guan, Q.; Liu, L.; Ding, Q.; Li, Y. Clematichinenoside inhibits VCAM-1 and ICAM-1 expression in TNF-alpha-treated endothelial cells via NADPH oxidase-dependent IkappaB kinase/NF-kappaB pathway. Free Radic. Biol. Med. 2015, 78, 190–201. [Google Scholar] [CrossRef]
  93. Kim, H.J.; Park, K.G.; Yoo, E.K.; Kim, Y.H.; Kim, Y.N.; Kim, H.S.; Kim, H.T.; Park, J.Y.; Lee, K.U.; Jang, W.G.; et al. Effects of PGC-1alpha on TNF-alpha-induced MCP-1 and VCAM-1 expression and NF-kappaB activation in human aortic smooth muscle and endothelial cells. Antioxid. Redox Signal. 2007, 9, 301–307. [Google Scholar] [CrossRef]
  94. Bloodsworth, A.; O’Donnell, V.B.; Freeman, B.A. Nitric oxide regulation of free radical- and enzyme-mediated lipid and lipoprotein oxidation. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1707–1715. [Google Scholar] [CrossRef] [PubMed]
  95. Beckman, J.S.; Koppenol, W.H. Nitric oxide, superoxide, and peroxynitrite: The good, the bad, and ugly. Am. J. Physiol. 1996, 271, C1424–C1437. [Google Scholar] [CrossRef] [PubMed]
  96. Brandes, R.P.; Weissmann, N.; Schroder, K. Nox family NADPH oxidases: Molecular mechanisms of activation. Free Radic. Biol. Med. 2014, 76, 208–226. [Google Scholar] [CrossRef]
  97. Ushio-Fukai, M.; Ash, D.; Nagarkoti, S.; Belin de Chantemele, E.J.; Fulton, D.J.R.; Fukai, T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid. Redox Signal. 2021, 34, 1319–1354. [Google Scholar] [CrossRef] [PubMed]
  98. Van Buul, J.D.; Fernandez-Borja, M.; Anthony, E.C.; Hordijk, P.L. Expression and localization of NOX2 and NOX4 in primary human endothelial cells. Antioxid. Redox Signal. 2005, 7, 308–317. [Google Scholar] [CrossRef]
  99. Bayraktutan, U.; Blayney, L.; Shah, A.M. Molecular characterization and localization of the NAD(P)H oxidase components gp91-phox and p22-phox in endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1903–1911. [Google Scholar] [CrossRef] [PubMed]
  100. Aldosari, S.; Awad, M.; Harrington, E.O.; Sellke, F.W.; Abid, M.R. Subcellular Reactive Oxygen Species (ROS) in Cardiovascular Pathophysiology. Antioxidants 2018, 7, 14. [Google Scholar] [CrossRef]
  101. Shafique, E.; Torina, A.; Reichert, K.; Colantuono, B.; Nur, N.; Zeeshan, K.; Ravichandran, V.; Liu, Y.; Feng, J.; Zeeshan, K.; et al. Mitochondrial redox plays a critical role in the paradoxical effects of NAPDH oxidase-derived ROS on coronary endothelium. Cardiovasc. Res. 2017, 113, 234–246. [Google Scholar] [CrossRef]
  102. Craige, S.M.; Chen, K.; Pei, Y.; Li, C.; Huang, X.; Chen, C.; Shibata, R.; Sato, K.; Walsh, K.; Keaney, J.F., Jr. NADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation. Circulation 2011, 124, 731–740. [Google Scholar] [CrossRef]
  103. Schroder, K.; Zhang, M.; Benkhoff, S.; Mieth, A.; Pliquett, R.; Kosowski, J.; Kruse, C.; Luedike, P.; Michaelis, U.R.; Weissmann, N.; et al. Nox4 is a protective reactive oxygen species generating vascular NADPH oxidase. Circ. Res. 2012, 110, 1217–1225. [Google Scholar] [CrossRef]
  104. Shafique, E.; Choy, W.C.; Liu, Y.; Feng, J.; Cordeiro, B.; Lyra, A.; Arafah, M.; Yassin-Kassab, A.; Zanetti, A.V.; Clements, R.T.; et al. Oxidative stress improves coronary endothelial function through activation of the pro-survival kinase AMPK. Aging 2013, 5, 515–530. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, Y.M.; Kim, S.J.; Tatsunami, R.; Yamamura, H.; Fukai, T.; Ushio-Fukai, M. ROS-induced ROS release orchestrated by Nox4, Nox2, and mitochondria in VEGF signaling and angiogenesis. Am. J. Physiol. Cell Physiol. 2017, 312, C749–C764. [Google Scholar] [CrossRef]
  106. Schilder, Y.D.; Heiss, E.H.; Schachner, D.; Ziegler, J.; Reznicek, G.; Sorescu, D.; Dirsch, V.M. NADPH oxidases 1 and 4 mediate cellular senescence induced by resveratrol in human endothelial cells. Free Radic. Biol. Med. 2009, 46, 1598–1606. [Google Scholar] [CrossRef]
  107. Konior, A.; Schramm, A.; Czesnikiewicz-Guzik, M.; Guzik, T.J. NADPH oxidases in vascular pathology. Antioxid. Redox Signal. 2014, 20, 2794–2814. [Google Scholar] [CrossRef]
  108. Nazari, B.; Jaquet, V.; Krause, K.H. NOX family NADPH oxidases in mammals: Evolutionary conservation and isoform-defining sequences. Redox Biol. 2023, 66, 102851. [Google Scholar] [CrossRef] [PubMed]
  109. Szekeres, F.L.M.; Walum, E.; Wikstrom, P.; Arner, A. A small molecule inhibitor of Nox2 and Nox4 improves contractile function after ischemia-reperfusion in the mouse heart. Sci. Rep. 2021, 11, 11970. [Google Scholar] [CrossRef]
  110. Lassegue, B.; Griendling, K.K. NADPH oxidases: Functions and pathologies in the vasculature. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 653–661. [Google Scholar] [CrossRef] [PubMed]
  111. Alhayaza, R.; Haque, E.; Karbasiafshar, C.; Sellke, F.W.; Abid, M.R. The Relationship Between Reactive Oxygen Species and Endothelial Cell Metabolism. Front. Chem. 2020, 8, 592688. [Google Scholar] [CrossRef] [PubMed]
  112. Takenouchi, Y.; Kobayashi, T.; Matsumoto, T.; Kamata, K. Gender differences in age-related endothelial function in the murine aorta. Atherosclerosis 2009, 206, 397–404. [Google Scholar] [CrossRef] [PubMed]
  113. Lassegue, B.; San Martin, A.; Griendling, K.K. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ. Res. 2012, 110, 1364–1390. [Google Scholar] [CrossRef]
  114. Taye, A.; Saad, A.H.; Kumar, A.H.; Morawietz, H. Effect of apocynin on NADPH oxidase-mediated oxidative stress-LOX-1-eNOS pathway in human endothelial cells exposed to high glucose. Eur. J. Pharmacol. 2010, 627, 42–48. [Google Scholar] [CrossRef] [PubMed]
  115. Duerrschmidt, N.; Stielow, C.; Muller, G.; Pagano, P.J.; Morawietz, H. NO-mediated regulation of NAD(P)H oxidase by laminar shear stress in human endothelial cells. J. Physiol. 2006, 576, 557–567. [Google Scholar] [CrossRef]
  116. Hwang, J.; Saha, A.; Boo, Y.C.; Sorescu, G.P.; McNally, J.S.; Holland, S.M.; Dikalov, S.; Giddens, D.P.; Griendling, K.K.; Harrison, D.G.; et al. Oscillatory shear stress stimulates endothelial production of O2 from p47phox-dependent NAD(P)H oxidases, leading to monocyte adhesion. J. Biol. Chem. 2003, 278, 47291–47298. [Google Scholar] [CrossRef]
  117. Li, J.M.; Fan, L.M.; George, V.T.; Brooks, G. Nox2 regulates endothelial cell cycle arrest and apoptosis via p21cip1 and p53. Free Radic. Biol. Med. 2007, 43, 976–986. [Google Scholar] [CrossRef]
  118. Gray, S.P.; Shah, A.M.; Smyrnias, I. NADPH oxidase 4 and its role in the cardiovascular system. Vasc. Biol. 2019, 1, H59–H66. [Google Scholar] [CrossRef]
  119. Goettsch, C.; Goettsch, W.; Arsov, A.; Hofbauer, L.C.; Bornstein, S.R.; Morawietz, H. Long-term cyclic strain downregulates endothelial Nox4. Antioxid. Redox Signal. 2009, 11, 2385–2397. [Google Scholar] [CrossRef]
  120. Hwang, J.; Ing, M.H.; Salazar, A.; Lassegue, B.; Griendling, K.; Navab, M.; Sevanian, A.; Hsiai, T.K. Pulsatile versus oscillatory shear stress regulates NADPH oxidase subunit expression: Implication for native LDL oxidation. Circ. Res. 2003, 93, 1225–1232. [Google Scholar] [CrossRef]
  121. Datla, S.R.; Peshavariya, H.; Dusting, G.J.; Mahadev, K.; Goldstein, B.J.; Jiang, F. Important role of Nox4 type NADPH oxidase in angiogenic responses in human microvascular endothelial cells in vitro. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2319–2324. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, K.; Kirber, M.T.; Xiao, H.; Yang, Y.; Keaney, J.F., Jr. Regulation of ROS signal transduction by NADPH oxidase 4 localization. J. Cell Biol. 2008, 181, 1129–1139. [Google Scholar] [CrossRef]
  123. Tang, X.; Wang, J.; Abboud, H.E.; Chen, Y.; Wang, J.J.; Zhang, S.X. Sustained Upregulation of Endothelial Nox4 Mediates Retinal Vascular Pathology in Type 1 Diabetes. Diabetes 2023, 72, 112–125. [Google Scholar] [CrossRef]
  124. Basuroy, S.; Bhattacharya, S.; Leffler, C.W.; Parfenova, H. Nox4 NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-alpha in cerebral vascular endothelial cells. Am. J. Physiol. Cell Physiol. 2009, 296, C422–C432. [Google Scholar] [CrossRef] [PubMed]
  125. Salazar, G. NADPH Oxidases and Mitochondria in Vascular Senescence. Int. J. Mol. Sci. 2018, 19, 1327. [Google Scholar] [CrossRef] [PubMed]
  126. Przybylska, D.; Janiszewska, D.; Gozdzik, A.; Bielak-Zmijewska, A.; Sunderland, P.; Sikora, E.; Mosieniak, G. NOX4 downregulation leads to senescence of human vascular smooth muscle cells. Oncotarget 2016, 7, 66429–66443. [Google Scholar] [CrossRef]
  127. Munoz, M.; Lopez-Oliva, M.E.; Rodriguez, C.; Martinez, M.P.; Saenz-Medina, J.; Sanchez, A.; Climent, B.; Benedito, S.; Garcia-Sacristan, A.; Rivera, L.; et al. Differential contribution of Nox1, Nox2 and Nox4 to kidney vascular oxidative stress and endothelial dysfunction in obesity. Redox Biol. 2020, 28, 101330. [Google Scholar] [CrossRef]
  128. Canugovi, C.; Stevenson, M.D.; Vendrov, A.E.; Hayami, T.; Robidoux, J.; Xiao, H.; Zhang, Y.Y.; Eitzman, D.T.; Runge, M.S.; Madamanchi, N.R. Increased mitochondrial NADPH oxidase 4 (NOX4) expression in aging is a causative factor in aortic stiffening. Redox Biol. 2019, 26, 101288. [Google Scholar] [CrossRef]
  129. Cucoranu, I.; Clempus, R.; Dikalova, A.; Phelan, P.J.; Ariyan, S.; Dikalov, S.; Sorescu, D. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ. Res. 2005, 97, 900–907. [Google Scholar] [CrossRef]
  130. Hahner, F.; Moll, F.; Warwick, T.; Hebchen, D.M.; Buchmann, G.K.; Epah, J.; Abplanalp, W.; Schader, T.; Gunther, S.; Gilsbach, R.; et al. Nox4 promotes endothelial differentiation through chromatin remodeling. Redox Biol. 2022, 55, 102381. [Google Scholar] [CrossRef]
  131. Kim, J.; Yoo, J.Y.; Suh, J.M.; Park, S.; Kang, D.; Jo, H.; Bae, Y.S. The flagellin-TLR5-Nox4 axis promotes the migration of smooth muscle cells in atherosclerosis. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef]
  132. Miyano, K.; Okamoto, S.; Yamauchi, A.; Kawai, C.; Kajikawa, M.; Kiyohara, T.; Tamura, M.; Taura, M.; Kuribayashi, F. The NADPH oxidase NOX4 promotes the directed migration of endothelial cells by stabilizing vascular endothelial growth factor receptor 2 protein. J. Biol. Chem. 2020, 295, 11877–11890. [Google Scholar] [CrossRef]
  133. Petry, A.; Djordjevic, T.; Weitnauer, M.; Kietzmann, T.; Hess, J.; Gorlach, A. NOX2 and NOX4 mediate proliferative response in endothelial cells. Antioxid. Redox Signal. 2006, 8, 1473–1484. [Google Scholar] [CrossRef]
  134. Yan, F.; Wang, Y.; Wu, X.; Peshavariya, H.M.; Dusting, G.J.; Zhang, M.; Jiang, F. Nox4 and redox signaling mediate TGF-beta-induced endothelial cell apoptosis and phenotypic switch. Cell Death Dis. 2014, 5, e1010. [Google Scholar] [CrossRef]
  135. Hendricks, K.S.; To, E.E.; Luong, R.; Liong, F.; Erlich, J.R.; Shah, A.M.; Liong, S.; O’Leary, J.J.; Brooks, D.A.; Vlahos, R.; et al. Endothelial NOX4 Oxidase Negatively Regulates Inflammation and Improves Morbidity During Influenza A Virus Lung Infection in Mice. Front. Cell. Infect. Microbiol. 2022, 12, 883448. [Google Scholar] [CrossRef]
  136. Yuan, S.; Hahn, S.A.; Miller, M.P.; Sanker, S.; Calderon, M.J.; Sullivan, M.; Dosunmu-Ogunbi, A.M.; Fazzari, M.; Li, Y.; Reynolds, M.; et al. Cooperation between CYB5R3 and NOX4 via coenzyme Q mitigates endothelial inflammation. Redox Biol. 2021, 47, 102166. [Google Scholar] [CrossRef]
  137. Cathcart, M.K. Regulation of superoxide anion production by NADPH oxidase in monocytes/macrophages: Contributions to atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 23–28. [Google Scholar] [CrossRef]
  138. Schurmann, C.; Rezende, F.; Kruse, C.; Yasar, Y.; Lowe, O.; Fork, C.; van de Sluis, B.; Bremer, R.; Weissmann, N.; Shah, A.M.; et al. The NADPH oxidase Nox4 has anti-atherosclerotic functions. Eur. Heart J. 2015, 36, 3447–3456. [Google Scholar] [CrossRef]
  139. Gray, S.P.; Di Marco, E.; Kennedy, K.; Chew, P.; Okabe, J.; El-Osta, A.; Calkin, A.C.; Biessen, E.A.; Touyz, R.M.; Cooper, M.E.; et al. Reactive Oxygen Species Can Provide Atheroprotection via NOX4-Dependent Inhibition of Inflammation and Vascular Remodeling. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 295–307. [Google Scholar] [CrossRef]
  140. Sheehan, A.L.; Carrell, S.; Johnson, B.; Stanic, B.; Banfi, B.; Miller, F.J., Jr. Role for Nox1 NADPH oxidase in atherosclerosis. Atherosclerosis 2011, 216, 321–326. [Google Scholar] [CrossRef]
  141. Gray, S.P.; Di Marco, E.; Okabe, J.; Szyndralewiez, C.; Heitz, F.; Montezano, A.C.; de Haan, J.B.; Koulis, C.; El-Osta, A.; Andrews, K.L.; et al. NADPH oxidase 1 plays a key role in diabetes mellitus-accelerated atherosclerosis. Circulation 2013, 127, 1888–1902. [Google Scholar] [CrossRef]
  142. Douglas, G.; Bendall, J.K.; Crabtree, M.J.; Tatham, A.L.; Carter, E.E.; Hale, A.B.; Channon, K.M. Endothelial-specific Nox2 overexpression increases vascular superoxide and macrophage recruitment in ApoE-/- mice. Cardiovasc. Res. 2012, 94, 20–29. [Google Scholar] [CrossRef]
  143. Batty, M.; Bennett, M.R.; Yu, E. The Role of Oxidative Stress in Atherosclerosis. Cells 2022, 11, 3843. [Google Scholar] [CrossRef]
  144. Hofmann, A.; Frank, F.; Wolk, S.; Busch, A.; Klimova, A.; Sabarstinski, P.; Gerlach, M.; Egorov, D.; Kopaliani, I.; Weinert, S.; et al. NOX4 mRNA correlates with plaque stability in patients with carotid artery stenosis. Redox Biol. 2022, 57, 102473. [Google Scholar] [CrossRef] [PubMed]
  145. Ho, F.; Watson, A.M.D.; Elbatreek, M.H.; Kleikers, P.W.M.; Khan, W.; Sourris, K.C.; Dai, A.; Jha, J.; Schmidt, H.; Jandeleit-Dahm, K.A.M. Endothelial reactive oxygen-forming NADPH oxidase 5 is a possible player in diabetic aortic aneurysm but not atherosclerosis. Sci. Rep. 2022, 12, 11570. [Google Scholar] [CrossRef] [PubMed]
  146. Mayer, B.; Hemmens, B. Biosynthesis and action of nitric oxide in mammalian cells. Trends Biochem. Sci. 1997, 22, 477–481. [Google Scholar] [CrossRef] [PubMed]
  147. Forstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837. [Google Scholar] [CrossRef]
  148. Lundberg, J.O.; Weitzberg, E. Nitric oxide signaling in health and disease. Cell 2022, 185, 2853–2878. [Google Scholar] [CrossRef]
  149. Li, H.; Forstermann, U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr. Opin. Pharmacol. 2013, 13, 161–167. [Google Scholar] [CrossRef]
  150. Zamora, R.; Vodovotz, Y.; Billiar, T.R. Inducible nitric oxide synthase and inflammatory diseases. Mol. Med. 2000, 6, 347–373. [Google Scholar] [CrossRef]
  151. Li, H.; Horke, S.; Forstermann, U. Vascular oxidative stress, nitric oxide and atherosclerosis. Atherosclerosis 2014, 237, 208–219. [Google Scholar] [CrossRef] [PubMed]
  152. Cominacini, L.; Rigoni, A.; Pasini, A.F.; Garbin, U.; Davoli, A.; Campagnola, M.; Pastorino, A.M.; Lo Cascio, V.; Sawamura, T. The binding of oxidized low density lipoprotein (ox-LDL) to ox-LDL receptor-1 reduces the intracellular concentration of nitric oxide in endothelial cells through an increased production of superoxide. J. Biol. Chem. 2001, 276, 13750–13755. [Google Scholar] [CrossRef] [PubMed]
  153. Chavakis, E.; Dernbach, E.; Hermann, C.; Mondorf, U.F.; Zeiher, A.M.; Dimmeler, S. Oxidized LDL inhibits vascular endothelial growth factor-induced endothelial cell migration by an inhibitory effect on the Akt/endothelial nitric oxide synthase pathway. Circulation 2001, 103, 2102–2107. [Google Scholar] [CrossRef]
  154. Mehta, J.L.; Chen, J.; Hermonat, P.L.; Romeo, F.; Novelli, G. Lectin-like, oxidized low-density lipoprotein receptor-1 (LOX-1): A critical player in the development of atherosclerosis and related disorders. Cardiovasc. Res. 2006, 69, 36–45. [Google Scholar] [CrossRef] [PubMed]
  155. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [CrossRef]
  156. Dikalov, S.I.; Nazarewicz, R.R. Angiotensin II-induced production of mitochondrial reactive oxygen species: Potential mechanisms and relevance for cardiovascular disease. Antioxid. Redox Signal. 2013, 19, 1085–1094. [Google Scholar] [CrossRef] [PubMed]
  157. Khoi, C.S.; Lin, T.Y.; Chiang, C.K. Targeting Insulin Resistance, Reactive Oxygen Species, Inflammation, Programmed Cell Death, ER Stress, and Mitochondrial Dysfunction for the Therapeutic Prevention of Free Fatty Acid-Induced Vascular Endothelial Lipotoxicity. Antioxidants 2024, 13, 1486. [Google Scholar] [CrossRef]
  158. Schulz, E.; Anter, E.; Keaney, J.F., Jr. Oxidative stress, antioxidants, and endothelial function. Curr. Med. Chem. 2004, 11, 1093–1104. [Google Scholar] [CrossRef]
  159. Langille, B.L. Arterial remodeling: Relation to hemodynamics. Can. J. Physiol. Pharmacol. 1996, 74, 834–841. [Google Scholar] [CrossRef] [PubMed]
  160. Chiu, J.J.; Chien, S. Effects of disturbed flow on vascular endothelium: Pathophysiological basis and clinical perspectives. Physiol. Rev. 2011, 91, 327–387. [Google Scholar] [CrossRef] [PubMed]
  161. Lee, D.Y.; Chiu, J.J. Atherosclerosis and flow: Roles of epigenetic modulation in vascular endothelium. J. Biomed. Sci. 2019, 26, 56. [Google Scholar] [CrossRef]
  162. Zhou, J.; Li, Y.S.; Chien, S. Shear stress-initiated signaling and its regulation of endothelial function. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 2191–2198. [Google Scholar] [CrossRef]
  163. Davies, P.F. Flow-mediated endothelial mechanotransduction. Physiol. Rev. 1995, 75, 519–560. [Google Scholar] [CrossRef]
  164. Tzima, E.; Irani-Tehrani, M.; Kiosses, W.B.; Dejana, E.; Schultz, D.A.; Engelhardt, B.; Cao, G.; DeLisser, H.; Schwartz, M.A. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 2005, 437, 426–431. [Google Scholar] [CrossRef]
  165. Jalali, S.; del Pozo, M.A.; Chen, K.; Miao, H.; Li, Y.; Schwartz, M.A.; Shyy, J.Y.; Chien, S. Integrin-mediated mechanotransduction requires its dynamic interaction with specific extracellular matrix (ECM) ligands. Proc. Natl. Acad. Sci. USA 2001, 98, 1042–1046. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, Y.; Miao, H.; Li, S.; Chen, K.D.; Li, Y.S.; Yuan, S.; Shyy, J.Y.; Chien, S. Interplay between integrins and FLK-1 in shear stress-induced signaling. Am. J. Physiol. Cell Physiol. 2002, 283, C1540–C1547. [Google Scholar] [CrossRef] [PubMed]
  167. Dimmeler, S.; Fleming, I.; Fisslthaler, B.; Hermann, C.; Busse, R.; Zeiher, A.M. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 1999, 399, 601–605. [Google Scholar] [CrossRef]
  168. Wu, W.; Xiao, H.; Laguna-Fernandez, A.; Villarreal, G., Jr.; Wang, K.C.; Geary, G.G.; Zhang, Y.; Wang, W.C.; Huang, H.D.; Zhou, J.; et al. Flow-Dependent Regulation of Kruppel-Like Factor 2 Is Mediated by MicroRNA-92a. Circulation 2011, 124, 633–641. [Google Scholar] [CrossRef] [PubMed]
  169. Boo, Y.C.; Hwang, J.; Sykes, M.; Michell, B.J.; Kemp, B.E.; Lum, H.; Jo, H. Shear stress stimulates phosphorylation of eNOS at Ser(635) by a protein kinase A-dependent mechanism. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H1819–H1828. [Google Scholar] [CrossRef]
  170. Takabe, W.; Warabi, E.; Noguchi, N. Anti-atherogenic effect of laminar shear stress via Nrf2 activation. Antioxid. Redox Signal. 2011, 15, 1415–1426. [Google Scholar] [CrossRef]
  171. Chen, Z.; Peng, I.C.; Cui, X.; Li, Y.S.; Chien, S.; Shyy, J.Y. Shear stress, SIRT1, and vascular homeostasis. Proc. Natl. Acad. Sci. USA 2010, 107, 10268–10273. [Google Scholar] [CrossRef]
  172. Wang, W.; Ha, C.H.; Jhun, B.S.; Wong, C.; Jain, M.K.; Jin, Z.G. Fluid shear stress stimulates phosphorylation-dependent nuclear export of HDAC5 and mediates expression of KLF2 and eNOS. Blood 2010, 115, 2971–2979. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, K.C.; Garmire, L.X.; Young, A.; Nguyen, P.; Trinh, A.; Subramaniam, S.; Wang, N.; Shyy, J.Y.; Li, Y.S.; Chien, S. Role of microRNA-23b in flow-regulation of Rb phosphorylation and endothelial cell growth. Proc. Natl. Acad. Sci. USA 2010, 107, 3234–3239. [Google Scholar] [CrossRef]
  174. Lin, K.; Hsu, P.P.; Chen, B.P.; Yuan, S.; Usami, S.; Shyy, J.Y.; Li, Y.S.; Chien, S. Molecular mechanism of endothelial growth arrest by laminar shear stress. Proc. Natl. Acad. Sci. USA 2000, 97, 9385–9389. [Google Scholar] [CrossRef]
  175. Abe, J.; Berk, B.C. Novel mechanisms of endothelial mechanotransduction. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 2378–2386. [Google Scholar] [CrossRef]
  176. Lin, J.H.; Walter, P.; Yen, T.S. Endoplasmic reticulum stress in disease pathogenesis. Annu. Rev. Pathol. 2008, 3, 399–425. [Google Scholar] [CrossRef]
  177. Han, J.; Kaufman, R.J. The role of ER stress in lipid metabolism and lipotoxicity. J. Lipid Res. 2016, 57, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
  178. Hetz, C.; Zhang, K.; Kaufman, R.J. Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 2020, 21, 421–438. [Google Scholar] [CrossRef]
  179. Yang, S.; Wu, M.; Li, X.; Zhao, R.; Zhao, Y.; Liu, L.; Wang, S. Role of Endoplasmic Reticulum Stress in Atherosclerosis and Its Potential as a Therapeutic Target. Oxid. Med. Cell Longev. 2020, 2020, 9270107. [Google Scholar] [CrossRef]
  180. Darling, N.J.; Cook, S.J. The role of MAPK signalling pathways in the response to endoplasmic reticulum stress. Biochim. Biophys. Acta 2014, 1843, 2150–2163. [Google Scholar] [CrossRef] [PubMed]
  181. Lenna, S.; Han, R.; Trojanowska, M. Endoplasmic reticulum stress and endothelial dysfunction. IUBMB Life 2014, 66, 530–537. [Google Scholar] [CrossRef] [PubMed]
  182. Galan, M.; Kassan, M.; Kadowitz, P.J.; Trebak, M.; Belmadani, S.; Matrougui, K. Mechanism of endoplasmic reticulum stress-induced vascular endothelial dysfunction. Biochim. Biophys. Acta 2014, 1843, 1063–1075. [Google Scholar] [CrossRef]
  183. Sanson, M.; Auge, N.; Vindis, C.; Muller, C.; Bando, Y.; Thiers, J.C.; Marachet, M.A.; Zarkovic, K.; Sawa, Y.; Salvayre, R.; et al. Oxidized low-density lipoproteins trigger endoplasmic reticulum stress in vascular cells: Prevention by oxygen-regulated protein 150 expression. Circ. Res. 2009, 104, 328–336. [Google Scholar] [CrossRef]
  184. Civelek, M.; Manduchi, E.; Riley, R.J.; Stoeckert, C.J., Jr.; Davies, P.F. Chronic endoplasmic reticulum stress activates unfolded protein response in arterial endothelium in regions of susceptibility to atherosclerosis. Circ. Res. 2009, 105, 453–461. [Google Scholar] [CrossRef]
  185. Tabas, I. The role of endoplasmic reticulum stress in the progression of atherosclerosis. Circ. Res. 2010, 107, 839–850. [Google Scholar] [CrossRef]
  186. Jiang, C.; Xie, S.; Yang, G.; Wang, N. Spotlight on NLRP3 Inflammasome: Role in Pathogenesis and Therapies of Atherosclerosis. J. Inflamm. Res. 2021, 14, 7143–7172. [Google Scholar] [CrossRef] [PubMed]
  187. Gora, S.; Maouche, S.; Atout, R.; Wanherdrick, K.; Lambeau, G.; Cambien, F.; Ninio, E.; Karabina, S.A. Phospholipolyzed LDL induces an inflammatory response in endothelial cells through endoplasmic reticulum stress signaling. FASEB J. 2010, 24, 3284–3297. [Google Scholar] [CrossRef]
  188. Hang, L.; Peng, Y.; Xiang, R.; Li, X.; Li, Z. Ox-LDL Causes Endothelial Cell Injury Through ASK1/NLRP3-Mediated Inflammasome Activation via Endoplasmic Reticulum Stress. Drug Des. Devel Ther. 2020, 14, 731–744. [Google Scholar] [CrossRef]
  189. Wei, M.C.; Zong, W.X.; Cheng, E.H.; Lindsten, T.; Panoutsakopoulou, V.; Ross, A.J.; Roth, K.A.; MacGregor, G.R.; Thompson, C.B.; Korsmeyer, S.J. Proapoptotic BAX and BAK: A requisite gateway to mitochondrial dysfunction and death. Science 2001, 292, 727–730. [Google Scholar] [CrossRef]
  190. Li, G.; Scull, C.; Ozcan, L.; Tabas, I. NADPH oxidase links endoplasmic reticulum stress, oxidative stress, and PKR activation to induce apoptosis. J. Cell Biol. 2010, 191, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  191. Timmins, J.M.; Ozcan, L.; Seimon, T.A.; Li, G.; Malagelada, C.; Backs, J.; Backs, T.; Bassel-Duby, R.; Olson, E.N.; Anderson, M.E.; et al. Calcium/calmodulin-dependent protein kinase II links ER stress with Fas and mitochondrial apoptosis pathways. J. Clin. Investig. 2009, 119, 2925–2941. [Google Scholar] [CrossRef] [PubMed]
  192. Ryter, S.W.; Cloonan, S.M.; Choi, A.M. Autophagy: A critical regulator of cellular metabolism and homeostasis. Mol. Cells 2013, 36, 7–16. [Google Scholar] [CrossRef]
  193. Hua, Y.; Zhang, J.; Liu, Q.; Su, J.; Zhao, Y.; Zheng, G.; Yang, Z.; Zhuo, D.; Ma, C.; Fan, G. The Induction of Endothelial Autophagy and Its Role in the Development of Atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 831847. [Google Scholar] [CrossRef]
  194. Lyu, Z.S.; Cao, X.N.; Wen, Q.; Mo, X.D.; Zhao, H.Y.; Chen, Y.H.; Wang, Y.; Chang, Y.J.; Xu, L.P.; Zhang, X.H.; et al. Autophagy in endothelial cells regulates their haematopoiesis-supporting ability. EBioMedicine 2020, 53, 102677. [Google Scholar] [CrossRef]
  195. Schaaf, M.B.; Houbaert, D.; Mece, O.; Agostinis, P. Autophagy in endothelial cells and tumor angiogenesis. Cell Death Differ. 2019, 26, 665–679. [Google Scholar] [CrossRef]
  196. Wu, Q.; Hu, Y.; Jiang, M.; Wang, F.; Gong, G. Effect of Autophagy Regulated by Sirt1/FoxO1 Pathway on the Release of Factors Promoting Thrombosis from Vascular Endothelial Cells. Int. J. Mol. Sci. 2019, 20, 4132. [Google Scholar] [CrossRef]
  197. Potente, M.; Ghaeni, L.; Baldessari, D.; Mostoslavsky, R.; Rossig, L.; Dequiedt, F.; Haendeler, J.; Mione, M.; Dejana, E.; Alt, F.W.; et al. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes. Dev. 2007, 21, 2644–2658. [Google Scholar] [CrossRef]
  198. Dou, Y.Q.; Kong, P.; Li, C.L.; Sun, H.X.; Li, W.W.; Yu, Y.; Nie, L.; Zhao, L.L.; Miao, S.B.; Li, X.K.; et al. Smooth muscle SIRT1 reprograms endothelial cells to suppress angiogenesis after ischemia. Theranostics 2020, 10, 1197–1212. [Google Scholar] [CrossRef]
  199. Mattagajasingh, I.; Kim, C.S.; Naqvi, A.; Yamamori, T.; Hoffman, T.A.; Jung, S.B.; DeRicco, J.; Kasuno, K.; Irani, K. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 2007, 104, 14855–14860. [Google Scholar] [CrossRef] [PubMed]
  200. Liu, J.; Bi, X.; Chen, T.; Zhang, Q.; Wang, S.X.; Chiu, J.J.; Liu, G.S.; Zhang, Y.; Bu, P.; Jiang, F. Shear stress regulates endothelial cell autophagy via redox regulation and Sirt1 expression. Cell Death Dis. 2015, 6, e1827. [Google Scholar] [CrossRef] [PubMed]
  201. Hirota, Y.; Yamashita, S.; Kurihara, Y.; Jin, X.; Aihara, M.; Saigusa, T.; Kang, D.; Kanki, T. Mitophagy is primarily due to alternative autophagy and requires the MAPK1 and MAPK14 signaling pathways. Autophagy 2015, 11, 332–343. [Google Scholar] [CrossRef]
  202. 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. 2021, 9, 809955. [Google Scholar] [CrossRef]
  203. Shao, B.Z.; Han, B.Z.; Zeng, Y.X.; Su, D.F.; Liu, C. The roles of macrophage autophagy in atherosclerosis. Acta Pharmacol. Sin. 2016, 37, 150–156. [Google Scholar] [CrossRef]
  204. Zhu, L.; Wu, G.; Yang, X.; Jia, X.; Li, J.; Bai, X.; Li, W.; Zhao, Y.; Li, Y.; Cheng, W.; et al. Low density lipoprotein mimics insulin action on autophagy and glucose uptake in endothelial cells. Sci. Rep. 2019, 9, 3020. [Google Scholar] [CrossRef]
  205. Perrotta, P.; Van der Veken, B.; Van Der Veken, P.; Pintelon, I.; Roosens, L.; Adriaenssens, E.; Timmerman, V.; Guns, P.J.; De Meyer, G.R.Y.; Martinet, W. Partial Inhibition of Glycolysis Reduces Atherogenesis Independent of Intraplaque Neovascularization in Mice. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1168–1181. [Google Scholar] [CrossRef]
  206. Ding, Z.; Liu, S.; Wang, X.; Khaidakov, M.; Dai, Y.; Mehta, J.L. Oxidant stress in mitochondrial DNA damage, autophagy and inflammation in atherosclerosis. Sci. Rep. 2013, 3, 1077. [Google Scholar] [CrossRef]
  207. Cho, K.; Choi, S.H. ASK1 Mediates Apoptosis and Autophagy during oxLDL-CD36 Signaling in Senescent Endothelial Cells. Oxid. Med. Cell Longev. 2019, 2019, 2840437. [Google Scholar] [CrossRef] [PubMed]
  208. Torisu, K.; Singh, K.K.; Torisu, T.; Lovren, F.; Liu, J.; Pan, Y.; Quan, A.; Ramadan, A.; Al-Omran, M.; Pankova, N.; et al. Intact endothelial autophagy is required to maintain vascular lipid homeostasis. Aging Cell 2016, 15, 187–191. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, Y.L.; Cao, Y.J.; Zhang, X.; Liu, H.H.; Tong, T.; Xiao, G.D.; Yang, Y.P.; Liu, C.F. The autophagy-lysosome pathway: A novel mechanism involved in the processing of oxidized LDL in human vascular endothelial cells. Biochem. Biophys. Res. Commun. 2010, 394, 377–382. [Google Scholar] [CrossRef]
  210. Bornstein, P. Matricellular proteins: An overview. J. Cell Commun. Signal. 2009, 3, 163–165. [Google Scholar] [CrossRef] [PubMed]
  211. Frangogiannis, N.G. Matricellular proteins in cardiac adaptation and disease. Physiol. Rev. 2012, 92, 635–688. [Google Scholar] [CrossRef]
  212. Wong, G.S.; Rustgi, A.K. Matricellular proteins: Priming the tumour microenvironment for cancer development and metastasis. Br. J. Cancer 2013, 108, 755–761. [Google Scholar] [CrossRef]
  213. Vincent, K.M.; Postovit, L.M. Matricellular proteins in cancer: A focus on secreted Frizzled-related proteins. J. Cell Commun. Signal. 2018, 12, 103–112. [Google Scholar] [CrossRef]
  214. Bornstein, P. Thrombospondins as matricellular modulators of cell function. J. Clin. Investig. 2001, 107, 929–934. [Google Scholar] [CrossRef]
  215. Armstrong, L.C.; Bjorkblom, B.; Hankenson, K.D.; Siadak, A.W.; Stiles, C.E.; Bornstein, P. Thrombospondin 2 inhibits microvascular endothelial cell proliferation by a caspase-independent mechanism. Mol. Biol. Cell 2002, 13, 1893–1905. [Google Scholar] [CrossRef] [PubMed]
  216. Krady, M.M.; Zeng, J.; Yu, J.; MacLauchlan, S.; Skokos, E.A.; Tian, W.; Bornstein, P.; Sessa, W.C.; Kyriakides, T.R. Thrombospondin-2 modulates extracellular matrix remodeling during physiological angiogenesis. Am. J. Pathol. 2008, 173, 879–891. [Google Scholar] [CrossRef] [PubMed]
  217. Sadvakassova, G.; Dobocan, M.C.; Congote, L.F. Osteopontin and the C-terminal peptide of thrombospondin-4 compete for CD44 binding and have opposite effects on CD133+ cell colony formation. BMC Res. Notes 2009, 2, 215. [Google Scholar] [CrossRef] [PubMed]
  218. Frolova, E.G.; Pluskota, E.; Krukovets, I.; Burke, T.; Drumm, C.; Smith, J.D.; Blech, L.; Febbraio, M.; Bornstein, P.; Plow, E.F.; et al. Thrombospondin-4 regulates vascular inflammation and atherogenesis. Circ. Res. 2010, 107, 1313–1325. [Google Scholar] [CrossRef]
  219. Unger, S.; Hecht, J.T. Pseudoachondroplasia and multiple epiphyseal dysplasia: New etiologic developments. Am. J. Med. Genet. 2001, 106, 244–250. [Google Scholar] [CrossRef]
  220. Wang, L.; Zheng, J.; Du, Y.; Huang, Y.; Li, J.; Liu, B.; Liu, C.J.; Zhu, Y.; Gao, Y.; Xu, Q.; et al. Cartilage oligomeric matrix protein maintains the contractile phenotype of vascular smooth muscle cells by interacting with alpha(7)beta(1) integrin. Circ. Res. 2010, 106, 514–525. [Google Scholar] [CrossRef]
  221. Chen, F.H.; Thomas, A.O.; Hecht, J.T.; Goldring, M.B.; Lawler, J. Cartilage oligomeric matrix protein/thrombospondin 5 supports chondrocyte attachment through interaction with integrins. J. Biol. Chem. 2005, 280, 32655–32661. [Google Scholar] [CrossRef]
  222. Gao, Q.; Chen, K.; Gao, L.; Zheng, Y.; Yang, Y.G. Thrombospondin-1 signaling through CD47 inhibits cell cycle progression and induces senescence in endothelial cells. Cell Death Dis. 2016, 7, e2368. [Google Scholar] [CrossRef]
  223. Singla, B.; Aithbathula, R.V.; Pervaiz, N.; Kathuria, I.; Swanson, M.; Ekuban, F.A.; Ahn, W.; Park, F.; Gyamfi, M.; Cherian-Shaw, M.; et al. CD47 Activation by Thrombospondin-1 in Lymphatic Endothelial Cells Suppresses Lymphangiogenesis and Promotes Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2023, 43, 1234–1250. [Google Scholar] [CrossRef]
  224. Stein, E.V.; Miller, T.W.; Ivins-O’Keefe, K.; Kaur, S.; Roberts, D.D. Secreted Thrombospondin-1 Regulates Macrophage Interleukin-1beta Production and Activation through CD47. Sci. Rep. 2016, 6, 19684. [Google Scholar] [CrossRef]
  225. Ganguly, R.; Khanal, S.; Mathias, A.; Gupta, S.; Lallo, J.; Sahu, S.; Ohanyan, V.; Patel, A.; Storm, K.; Datta, S.; et al. TSP-1 (Thrombospondin-1) Deficiency Protects ApoE(-/-) Mice Against Leptin-Induced Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2021, 41, e112–e127. [Google Scholar] [CrossRef] [PubMed]
  226. Moura, R.; Tjwa, M.; Vandervoort, P.; Van Kerckhoven, S.; Holvoet, P.; Hoylaerts, M.F. Thrombospondin-1 deficiency accelerates atherosclerotic plaque maturation in ApoE-/- mice. Circ. Res. 2008, 103, 1181–1189. [Google Scholar] [CrossRef]
  227. Murphy-Ullrich, J.E.; Suto, M.J. Thrombospondin-1 regulation of latent TGF-beta activation: A therapeutic target for fibrotic disease. Matrix Biol. 2018, 68–69, 28–43. [Google Scholar] [CrossRef]
  228. Chen, D.; Asahara, T.; Krasinski, K.; Witzenbichler, B.; Yang, J.; Magner, M.; Kearney, M.; Frazier, W.A.; Isner, J.M.; Andres, V. Antibody blockade of thrombospondin accelerates reendothelialization and reduces neointima formation in balloon-injured rat carotid artery. Circulation 1999, 100, 849–854. [Google Scholar] [CrossRef] [PubMed]
  229. Isenberg, J.S.; Roberts, D.D. THBS1 (thrombospondin-1). Atlas Genet. Cytogenet. Oncol. Haematol. 2020, 24, 291–299. [Google Scholar] [CrossRef]
  230. Asch, A.S.; Silbiger, S.; Heimer, E.; Nachman, R.L. Thrombospondin sequence motif (CSVTCG) is responsible for CD36 binding. Biochem. Biophys. Res. Commun. 1992, 182, 1208–1217. [Google Scholar] [CrossRef] [PubMed]
  231. Gao, A.G.; Lindberg, F.P.; Finn, M.B.; Blystone, S.D.; Brown, E.J.; Frazier, W.A. Integrin-associated protein is a receptor for the C-terminal domain of thrombospondin. J. Biol. Chem. 1996, 271, 21–24. [Google Scholar] [CrossRef] [PubMed]
  232. Sun, X.; Mosher, D.F.; Rapraeger, A. Heparan sulfate-mediated binding of epithelial cell surface proteoglycan to thrombospondin. J. Biol. Chem. 1989, 264, 2885–2889. [Google Scholar] [CrossRef]
  233. Calzada, M.J.; Annis, D.S.; Zeng, B.; Marcinkiewicz, C.; Banas, B.; Lawler, J.; Mosher, D.F.; Roberts, D.D. Identification of novel beta1 integrin binding sites in the type 1 and type 2 repeats of thrombospondin-1. J. Biol. Chem. 2004, 279, 41734–41743. [Google Scholar] [CrossRef]
  234. Calzada, M.J.; Sipes, J.M.; Krutzsch, H.C.; Yurchenco, P.D.; Annis, D.S.; Mosher, D.F.; Roberts, D.D. Recognition of the N-terminal modules of thrombospondin-1 and thrombospondin-2 by alpha6beta1 integrin. J. Biol. Chem. 2003, 278, 40679–40687. [Google Scholar] [CrossRef]
  235. Lawler, J.; Weinstein, R.; Hynes, R.O. Cell attachment to thrombospondin: The role of ARG-GLY-ASP, calcium, and integrin receptors. J. Cell Biol. 1988, 107, 2351–2361. [Google Scholar] [CrossRef]
  236. Lawler, J.; Hynes, R.O. An integrin receptor on normal and thrombasthenic platelets that binds thrombospondin. Blood 1989, 74, 2022–2027. [Google Scholar] [CrossRef] [PubMed]
  237. Mirochnik, Y.; Kwiatek, A.; Volpert, O.V. Thrombospondin and apoptosis: Molecular mechanisms and use for design of complementation treatments. Curr. Drug Targets 2008, 9, 851–862. [Google Scholar] [CrossRef] [PubMed]
  238. Jimenez, B.; Volpert, O.V.; Crawford, S.E.; Febbraio, M.; Silverstein, R.L.; Bouck, N. Signals leading to apoptosis-dependent inhibition of neovascularization by thrombospondin-1. Nat. Med. 2000, 6, 41–48. [Google Scholar] [CrossRef]
  239. Nor, J.E.; Mitra, R.S.; Sutorik, M.M.; Mooney, D.J.; Castle, V.P.; Polverini, P.J. Thrombospondin-1 induces endothelial cell apoptosis and inhibits angiogenesis by activating the caspase death pathway. J. Vasc. Res. 2000, 37, 209–218. [Google Scholar] [CrossRef]
  240. Dawson, D.W.; Pearce, S.F.; Zhong, R.; Silverstein, R.L.; Frazier, W.A.; Bouck, N.P. CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on endothelial cells. J. Cell Biol. 1997, 138, 707–717. [Google Scholar] [CrossRef]
  241. Isenberg, J.S.; Ridnour, L.A.; Perruccio, E.M.; Espey, M.G.; Wink, D.A.; Roberts, D.D. Thrombospondin-1 inhibits endothelial cell responses to nitric oxide in a cGMP-dependent manner. Proc. Natl. Acad. Sci. USA 2005, 102, 13141–13146. [Google Scholar] [CrossRef]
  242. Chu, L.Y.; Ramakrishnan, D.P.; Silverstein, R.L. Thrombospondin-1 modulates VEGF signaling via CD36 by recruiting SHP-1 to VEGFR2 complex in microvascular endothelial cells. Blood 2013, 122, 1822–1832. [Google Scholar] [CrossRef]
  243. Isenberg, J.S.; Ridnour, L.A.; Dimitry, J.; Frazier, W.A.; Wink, D.A.; Roberts, D.D. CD47 is necessary for inhibition of nitric oxide-stimulated vascular cell responses by thrombospondin-1. J. Biol. Chem. 2006, 281, 26069–26080. [Google Scholar] [CrossRef]
  244. Rogers, N.M.; Sharifi-Sanjani, M.; Csanyi, G.; Pagano, P.J.; Isenberg, J.S. Thrombospondin-1 and CD47 regulation of cardiac, pulmonary and vascular responses in health and disease. Matrix Biol. 2014, 37, 92–101. [Google Scholar] [CrossRef] [PubMed]
  245. Narizhneva, N.V.; Razorenova, O.V.; Podrez, E.A.; Chen, J.; Chandrasekharan, U.M.; DiCorleto, P.E.; Plow, E.F.; Topol, E.J.; Byzova, T.V. Thrombospondin-1 up-regulates expression of cell adhesion molecules and promotes monocyte binding to endothelium. FASEB J. 2005, 19, 1158–1160. [Google Scholar] [CrossRef] [PubMed]
  246. Peng, B.; Zhou, Y.; Fu, X.; Chen, L.; Pan, Z.; Yi, Q.; Zhao, T.; Fu, Z.; Wang, T. THBS1 mediates hypoxia driven EndMT in pulmonary hypertension. Pulm. Circ. 2024, 14, e70019. [Google Scholar] [CrossRef] [PubMed]
  247. Yamashiro, Y.; Thang, B.Q.; Shin, S.J.; Lino, C.A.; Nakamura, T.; Kim, J.; Sugiyama, K.; Tokunaga, C.; Sakamoto, H.; Osaka, M.; et al. Role of Thrombospondin-1 in Mechanotransduction and Development of Thoracic Aortic Aneurysm in Mouse and Humans. Circ. Res. 2018, 123, 660–672. [Google Scholar] [CrossRef]
  248. Aburima, A.; Berger, M.; Spurgeon, B.E.J.; Webb, B.A.; Wraith, K.S.; Febbraio, M.; Poole, A.W.; Naseem, K.M. Thrombospondin-1 promotes hemostasis through modulation of cAMP signaling in blood platelets. Blood 2021, 137, 678–689. [Google Scholar] [CrossRef]
  249. Stirling, E.R.; Terabe, M.; Wilson, A.S.; Kooshki, M.; Yamaleyeva, L.M.; Alexander-Miller, M.A.; Zhang, W.; Miller, L.D.; Triozzi, P.L.; Soto-Pantoja, D.R. Targeting the CD47/thrombospondin-1 signaling axis regulates immune cell bioenergetics in the tumor microenvironment to potentiate antitumor immune response. J. Immunother. Cancer 2022, 10, e004712. [Google Scholar] [CrossRef]
  250. Roberts, D.D.; Isenberg, J.S. CD47 and thrombospondin-1 regulation of mitochondria, metabolism, and diabetes. Am. J. Physiol. Cell Physiol. 2021, 321, C201–C213. [Google Scholar] [CrossRef]
  251. Zhao, W.; Shen, B.; Cheng, Q.; Zhou, Y.; Chen, K. Roles of TSP1-CD47 signaling pathway in senescence of endothelial cells: Cell cycle, inflammation and metabolism. Mol. Biol. Rep. 2023, 50, 4579–4585. [Google Scholar] [CrossRef]
  252. Gao, L.; Chen, K.; Gao, Q.; Wang, X.; Sun, J.; Yang, Y.G. CD47 deficiency in tumor stroma promotes tumor progression by enhancing angiogenesis. Oncotarget 2017, 8, 22406–22413. [Google Scholar] [CrossRef]
  253. Singh, B.; Cui, K.; Peng, Q.; Li, K.; Zhu, B.; Bhattacharjee, S.; Osorio, D.; Wang, B.; Dong, Y.; Wang, D. Novel Role Of Endothelial Cd47 In The Regulation Of Pathogenesis Of Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2023, 43, A338. [Google Scholar] [CrossRef]
  254. Meijles, D.N.; Sahoo, S.; Al Ghouleh, I.; Amaral, J.H.; Bienes-Martinez, R.; Knupp, H.E.; Attaran, S.; Sembrat, J.C.; Nouraie, S.M.; Rojas, M.M.; et al. The matricellular protein TSP1 promotes human and mouse endothelial cell senescence through CD47 and Nox1. Sci. Signal. 2017, 10, eaaj1784. [Google Scholar] [CrossRef] [PubMed]
  255. LeBlanc, A.J.; Kelm, N.Q. Thrombospondin-1, Free Radicals, and the Coronary Microcirculation: The Aging Conundrum. Antioxid. Redox Signal. 2017, 27, 785–801. [Google Scholar] [CrossRef] [PubMed]
  256. Ridnour, L.A.; Isenberg, J.S.; Espey, M.G.; Thomas, D.D.; Roberts, D.D.; Wink, D.A. Nitric oxide regulates angiogenesis through a functional switch involving thrombospondin-1. Proc. Natl. Acad. Sci. USA 2005, 102, 13147–13152. [Google Scholar] [CrossRef]
  257. Novelli, E.M.; Little-Ihrig, L.; Knupp, H.E.; Rogers, N.M.; Yao, M.; Baust, J.J.; Meijles, D.; St Croix, C.M.; Ross, M.A.; Pagano, P.J.; et al. Vascular TSP1-CD47 signaling promotes sickle cell-associated arterial vasculopathy and pulmonary hypertension in mice. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 316, L1150–L1164. [Google Scholar] [CrossRef]
  258. Kale, A.; Rogers, N.M.; Ghimire, K. Thrombospondin-1 CD47 Signalling: From Mechanisms to Medicine. Int. J. Mol. Sci. 2021, 22, 4062. [Google Scholar] [CrossRef]
  259. Rogers, N.M.; Sharifi-Sanjani, M.; Yao, M.; Ghimire, K.; Bienes-Martinez, R.; Mutchler, S.M.; Knupp, H.E.; Baust, J.; Novelli, E.M.; Ross, M.; et al. TSP1-CD47 signaling is upregulated in clinical pulmonary hypertension and contributes to pulmonary arterial vasculopathy and dysfunction. Cardiovasc. Res. 2017, 113, 15–29. [Google Scholar] [CrossRef]
  260. Csanyi, G.; Yao, M.; Rodriguez, A.I.; Al Ghouleh, I.; Sharifi-Sanjani, M.; Frazziano, G.; Huang, X.; Kelley, E.E.; Isenberg, J.S.; Pagano, P.J. Thrombospondin-1 regulates blood flow via CD47 receptor-mediated activation of NADPH oxidase 1. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 2966–2973. [Google Scholar] [CrossRef]
  261. Csanyi, G.; Feck, D.M.; Ghoshal, P.; Singla, B.; Lin, H.; Nagarajan, S.; Meijles, D.N.; Al Ghouleh, I.; Cantu-Medellin, N.; Kelley, E.E.; et al. CD47 and Nox1 Mediate Dynamic Fluid-Phase Macropinocytosis of Native LDL. Antioxid. Redox Signal. 2017, 26, 886–901. [Google Scholar] [CrossRef]
  262. Lin, H.P.; Singla, B.; Ahn, W.; Ghoshal, P.; Blahove, M.; Cherian-Shaw, M.; Chen, A.; Haller, A.; Hui, D.Y.; Dong, K.; et al. Receptor-independent fluid-phase macropinocytosis promotes arterial foam cell formation and atherosclerosis. Sci. Transl. Med. 2022, 14, eadd2376. [Google Scholar] [CrossRef] [PubMed]
  263. Bond, A.R.; Hultgardh-Nilsson, A.; Knutsson, A.; Jackson, C.L.; Rauch, U. Cartilage oligomeric matrix protein (COMP) in murine brachiocephalic and carotid atherosclerotic lesions. Atherosclerosis 2014, 236, 366–372. [Google Scholar] [CrossRef] [PubMed]
  264. Hultman, K.; Edsfeldt, A.; Bjorkbacka, H.; Duner, P.; Sundius, L.; Nitulescu, M.; Persson, A.; Boyle, J.J.; Nilsson, J.; Hultgardh-Nilsson, A.; et al. Cartilage Oligomeric Matrix Protein Associates With a Vulnerable Plaque Phenotype in Human Atherosclerotic Plaques. Stroke 2019, 50, 3289–3292. [Google Scholar] [CrossRef] [PubMed]
  265. Wang, F.F.; Ha, L.; Yu, H.Y.; Mi, L.; Han, J.L.; Gao, W. Altered serum level of cartilage oligomeric matrix protein and its association with coronary calcification in patients with coronary heart disease. J. Geriatr. Cardiol. 2017, 14, 87–92. [Google Scholar] [CrossRef]
  266. Fu, Y.; Gao, C.; Liang, Y.; Wang, M.; Huang, Y.; Ma, W.; Li, T.; Jia, Y.; Yu, F.; Zhu, W.; et al. Shift of Macrophage Phenotype Due to Cartilage Oligomeric Matrix Protein Deficiency Drives Atherosclerotic Calcification. Circ. Res. 2016, 119, 261–276. [Google Scholar] [CrossRef]
  267. Lv, H.; Wang, H.; Quan, M.; Zhang, C.; Fu, Y.; Zhang, L.; Lin, C.; Liu, X.; Yi, X.; Chen, J.; et al. Cartilage oligomeric matrix protein fine-tunes disturbed flow-induced endothelial activation and atherogenesis. Matrix Biol. 2021, 95, 32–51. [Google Scholar] [CrossRef]
  268. Riessen, R.; Fenchel, M.; Chen, H.; Axel, D.I.; Karsch, K.R.; Lawler, J. Cartilage oligomeric matrix protein (thrombospondin-5) is expressed by human vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 47–54. [Google Scholar] [CrossRef]
  269. Muqri, F.; Helkin, A.; Maier, K.G.; Gahtan, V. Thrombospondin-5 and fluvastatin promote angiogenesis and are protective against endothelial cell apoptosis. J. Cell. Biochem. 2020, 121, 4154–4165. [Google Scholar] [CrossRef]
  270. Wang, H.; Yuan, Z.; Wang, B.; Li, B.; Lv, H.; He, J.; Huang, Y.; Cui, Z.; Ma, Q.; Li, T.; et al. COMP (Cartilage Oligomeric Matrix Protein), a Novel PIEZO1 Regulator That Controls Blood Pressure. Hypertension 2022, 79, 549–561. [Google Scholar] [CrossRef]
  271. Agarwal, P.; Schulz, J.N.; Blumbach, K.; Andreasson, K.; Heinegard, D.; Paulsson, M.; Mauch, C.; Eming, S.A.; Eckes, B.; Krieg, T. Enhanced deposition of cartilage oligomeric matrix protein is a common feature in fibrotic skin pathologies. Matrix Biol. 2013, 32, 325–331. [Google Scholar] [CrossRef]
  272. Vuga, L.J.; Milosevic, J.; Pandit, K.; Ben-Yehudah, A.; Chu, Y.; Richards, T.; Sciurba, J.; Myerburg, M.; Zhang, Y.; Parwani, A.V.; et al. Cartilage oligomeric matrix protein in idiopathic pulmonary fibrosis. PLoS ONE 2013, 8, e83120. [Google Scholar] [CrossRef]
  273. Farina, G.; Lemaire, R.; Korn, J.H.; Widom, R.L. Cartilage oligomeric matrix protein is overexpressed by scleroderma dermal fibroblasts. Matrix Biol. 2006, 25, 213–222. [Google Scholar] [CrossRef]
  274. Farina, G.; Lemaire, R.; Pancari, P.; Bayle, J.; Widom, R.L.; Lafyatis, R. Cartilage oligomeric matrix protein expression in systemic sclerosis reveals heterogeneity of dermal fibroblast responses to transforming growth factor beta. Ann. Rheum. Dis. 2009, 68, 435–441. [Google Scholar] [CrossRef]
  275. Sanchez-Duffhues, G.; Garcia de Vinuesa, A.; Ten Dijke, P. Endothelial-to-mesenchymal transition in cardiovascular diseases: Developmental signaling pathways gone awry. Dev. Dyn. 2018, 247, 492–508. [Google Scholar] [CrossRef]
  276. Yao, J.; Guihard, P.J.; Blazquez-Medela, A.M.; Guo, Y.; Moon, J.H.; Jumabay, M.; Bostrom, K.I.; Yao, Y. Serine Protease Activation Essential for Endothelial-Mesenchymal Transition in Vascular Calcification. Circ. Res. 2015, 117, 758–769. [Google Scholar] [CrossRef]
  277. Bauer, E.M.; Qin, Y.; Miller, T.W.; Bandle, R.W.; Csanyi, G.; Pagano, P.J.; Bauer, P.M.; Schnermann, J.; Roberts, D.D.; Isenberg, J.S. Thrombospondin-1 supports blood pressure by limiting eNOS activation and endothelial-dependent vasorelaxation. Cardiovasc. Res. 2010, 88, 471–481. [Google Scholar] [CrossRef] [PubMed]
  278. Yamashiro, Y.; Thang, B.Q.; Ramirez, K.; Shin, S.J.; Kohata, T.; Ohata, S.; Nguyen, T.A.V.; Ohtsuki, S.; Nagayama, K.; Yanagisawa, H. Matrix mechanotransduction mediated by thrombospondin-1/integrin/YAP in the vascular remodeling. Proc. Natl. Acad. Sci. USA 2020, 117, 9896–9905. [Google Scholar] [CrossRef]
  279. Wang, M.; Fu, Y.; Gao, C.; Jia, Y.; Huang, Y.; Liu, L.; Wang, X.; Wang, W.; Kong, W. Cartilage oligomeric matrix protein prevents vascular aging and vascular smooth muscle cells senescence. Biochem. Biophys. Res. Commun. 2016, 478, 1006–1013. [Google Scholar] [CrossRef] [PubMed]
  280. Singla, B.; Lin, H.P.; Chen, A.; Ahn, W.; Ghoshal, P.; Cherian-Shaw, M.; White, J.; Stansfield, B.K.; Csanyi, G. Role of R-spondin 2 in arterial lymphangiogenesis and atherosclerosis. Cardiovasc. Res. 2021, 117, 1489–1509. [Google Scholar] [CrossRef] [PubMed]
  281. Fu, S.; Tan, R.; Feng, Y.; Yu, P.; Mo, Y.; Xiao, W.; Wang, S.; Zhang, J. N-methyl-N-nitrosourea induces zebrafish anomalous angiogenesis through Wnt/beta-catenin pathway. Ecotoxicol. Environ. Saf. 2022, 239, 113674. [Google Scholar] [CrossRef]
  282. Scholz, B.; Korn, C.; Wojtarowicz, J.; Mogler, C.; Augustin, I.; Boutros, M.; Niehrs, C.; Augustin, H.G. Endothelial RSPO3 Controls Vascular Stability and Pruning through Non-canonical WNT/Ca(2+)/NFAT Signaling. Dev. Cell 2016, 36, 79–93. [Google Scholar] [CrossRef] [PubMed]
  283. Minear, M.A.; Crosslin, D.R.; Sutton, B.S.; Connelly, J.J.; Nelson, S.C.; Gadson-Watson, S.; Wang, T.; Seo, D.; Vance, J.M.; Sketch, M.H., Jr.; et al. Polymorphic variants in tenascin-C (TNC) are associated with atherosclerosis and coronary artery disease. Hum. Genet. 2011, 129, 641–654. [Google Scholar] [CrossRef]
  284. Matsui, Y.; Rittling, S.R.; Okamoto, H.; Inobe, M.; Jia, N.; Shimizu, T.; Akino, M.; Sugawara, T.; Morimoto, J.; Kimura, C.; et al. Osteopontin deficiency attenuates atherosclerosis in female apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1029–1034. [Google Scholar] [CrossRef]
  285. Hirota, S.; Imakita, M.; Kohri, K.; Ito, A.; Morii, E.; Adachi, S.; Kim, H.M.; Kitamura, Y.; Yutani, C.; Nomura, S. Expression of osteopontin messenger RNA by macrophages in atherosclerotic plaques. A possible association with calcification. Am. J. Pathol. 1993, 143, 1003–1008. [Google Scholar] [PubMed]
  286. Zhao, J.F.; Chen, H.Y.; Wei, J.; Jim Leu, S.J.; Lee, T.S. CCN family member 1 deregulates cholesterol metabolism and aggravates atherosclerosis. Acta Physiol. 2019, 225, e13209. [Google Scholar] [CrossRef] [PubMed]
  287. Zhang, J.; Wu, G.; Dai, H. The matricellular protein CCN1 regulates TNF-alpha induced vascular endothelial cell apoptosis. Cell Biol. Int. 2016, 40, 1–6. [Google Scholar] [CrossRef]
  288. Grote, K.; Salguero, G.; Ballmaier, M.; Dangers, M.; Drexler, H.; Schieffer, B. The angiogenic factor CCN1 promotes adhesion and migration of circulating CD34+ progenitor cells: Potential role in angiogenesis and endothelial regeneration. Blood 2007, 110, 877–885. [Google Scholar] [CrossRef]
  289. Chen, J.; Singh, K.; Mukherjee, B.B.; Sodek, J. Developmental expression of osteopontin (OPN) mRNA in rat tissues: Evidence for a role for OPN in bone formation and resorption. Matrix 1993, 13, 113–123. [Google Scholar] [CrossRef]
  290. Brown, L.F.; Berse, B.; Van de Water, L.; Papadopoulos-Sergiou, A.; Perruzzi, C.A.; Manseau, E.J.; Dvorak, H.F.; Senger, D.R. Expression and distribution of osteopontin in human tissues: Widespread association with luminal epithelial surfaces. Mol. Biol. Cell 1992, 3, 1169–1180. [Google Scholar] [CrossRef]
  291. Xu, C.; Wu, Y.; Liu, N. Osteopontin in autoimmune disorders: Current knowledge and future perspective. Inflammopharmacology 2022, 30, 385–396. [Google Scholar] [CrossRef] [PubMed]
  292. Hu, D.D.; Lin, E.C.; Kovach, N.L.; Hoyer, J.R.; Smith, J.W. A biochemical characterization of the binding of osteopontin to integrins alpha v beta 1 and alpha v beta 5. J. Biol. Chem. 1995, 270, 26232–26238. [Google Scholar] [CrossRef]
  293. Yokosaki, Y.; Tanaka, K.; Higashikawa, F.; Yamashita, K.; Eboshida, A. Distinct structural requirements for binding of the integrins alphavbeta6, alphavbeta3, alphavbeta5, alpha5beta1 and alpha9beta1 to osteopontin. Matrix Biol. 2005, 24, 418–427. [Google Scholar] [CrossRef]
  294. Denda, S.; Reichardt, L.F.; Muller, U. Identification of osteopontin as a novel ligand for the integrin alpha8 beta1 and potential roles for this integrin-ligand interaction in kidney morphogenesis. Mol. Biol. Cell 1998, 9, 1425–1435. [Google Scholar] [CrossRef]
  295. Liaw, L.; Skinner, M.P.; Raines, E.W.; Ross, R.; Cheresh, D.A.; Schwartz, S.M.; Giachelli, C.M. The adhesive and migratory effects of osteopontin are mediated via distinct cell surface integrins. Role of alpha v beta 3 in smooth muscle cell migration to osteopontin in vitro. J. Clin. Investig. 1995, 95, 713–724. [Google Scholar] [CrossRef]
  296. O’Regan, A.; Berman, J.S. Osteopontin: A key cytokine in cell-mediated and granulomatous inflammation. Int. J. Exp. Pathol. 2000, 81, 373–390. [Google Scholar] [CrossRef] [PubMed]
  297. Liaw, L.; Lindner, V.; Schwartz, S.M.; Chambers, A.F.; Giachelli, C.M. Osteopontin and beta 3 integrin are coordinately expressed in regenerating endothelium in vivo and stimulate Arg-Gly-Asp-dependent endothelial migration in vitro. Circ. Res. 1995, 77, 665–672. [Google Scholar] [CrossRef]
  298. Qu, H.; Brown, L.F.; Senger, D.R.; Geng, L.L.; Dvorak, H.F.; Dvorak, A.M. Ultrastructural immunogold localization of osteopontin in human gallbladder epithelial cells. J. Histochem. Cytochem. 1994, 42, 351–361. [Google Scholar] [CrossRef]
  299. O’Brien, E.R.; Garvin, M.R.; Stewart, D.K.; Hinohara, T.; Simpson, J.B.; Schwartz, S.M.; Giachelli, C.M. Osteopontin is synthesized by macrophage, smooth muscle, and endothelial cells in primary and restenotic human coronary atherosclerotic plaques. Arterioscler. Thromb. 1994, 14, 1648–1656. [Google Scholar] [CrossRef] [PubMed]
  300. Xie, Z.; Pimental, D.R.; Lohan, S.; Vasertriger, A.; Pligavko, C.; Colucci, W.S.; Singh, K. Regulation of angiotensin II-stimulated osteopontin expression in cardiac microvascular endothelial cells: Role of p42/44 mitogen-activated protein kinase and reactive oxygen species. J. Cell. Physiol. 2001, 188, 132–138. [Google Scholar] [CrossRef] [PubMed]
  301. Golledge, J.; McCann, M.; Mangan, S.; Lam, A.; Karan, M. Osteoprotegerin and osteopontin are expressed at high concentrations within symptomatic carotid atherosclerosis. Stroke 2004, 35, 1636–1641. [Google Scholar] [CrossRef]
  302. Gravallese, E.M. Osteopontin: A bridge between bone and the immune system. J. Clin. Investig. 2003, 112, 147–149. [Google Scholar] [CrossRef] [PubMed]
  303. Ashkar, S.; Weber, G.F.; Panoutsakopoulou, V.; Sanchirico, M.E.; Jansson, M.; Zawaideh, S.; Rittling, S.R.; Denhardt, D.T.; Glimcher, M.J.; Cantor, H. Eta-1 (osteopontin): An early component of type-1 (cell-mediated) immunity. Science 2000, 287, 860–864. [Google Scholar] [CrossRef]
  304. Kadoglou, N.P.E.; Khattab, E.; Velidakis, N.; Gkougkoudi, E. The Role of Osteopontin in Atherosclerosis and Its Clinical Manifestations (Atherosclerotic Cardiovascular Diseases)-A Narrative Review. Biomedicines 2023, 11, 3178. [Google Scholar] [CrossRef]
  305. Moschetta, D.; Di Minno, M.N.D.; Porro, B.; Perrucci, G.L.; Valerio, V.; Alfieri, V.; Massaiu, I.; Orekhov, A.N.; Di Minno, A.; Songia, P.; et al. Relationship Between Plasma Osteopontin and Arginine Pathway Metabolites in Patients With Overt Coronary Artery Disease. Front. Physiol. 2020, 11, 982. [Google Scholar] [CrossRef]
  306. Isoda, K.; Kamezawa, Y.; Ayaori, M.; Kusuhara, M.; Tada, N.; Ohsuzu, F. Osteopontin transgenic mice fed a high-cholesterol diet develop early fatty-streak lesions. Circulation 2003, 107, 679–681. [Google Scholar] [CrossRef]
  307. Chiba, S.; Okamoto, H.; Kon, S.; Kimura, C.; Murakami, M.; Inobe, M.; Matsui, Y.; Sugawara, T.; Shimizu, T.; Uede, T.; et al. Development of atherosclerosis in osteopontin transgenic mice. Heart Vessels 2002, 16, 111–117. [Google Scholar] [CrossRef]
  308. Kale, S.; Raja, R.; Thorat, D.; Soundararajan, G.; Patil, T.V.; Kundu, G.C. Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via alpha9beta1 integrin. Oncogene 2014, 33, 2295–2306. [Google Scholar] [CrossRef] [PubMed]
  309. Dai, J.; Peng, L.; Fan, K.; Wang, H.; Wei, R.; Ji, G.; Cai, J.; Lu, B.; Li, B.; Zhang, D.; et al. Osteopontin induces angiogenesis through activation of PI3K/AKT and ERK1/2 in endothelial cells. Oncogene 2009, 28, 3412–3422. [Google Scholar] [CrossRef] [PubMed]
  310. Zhu, B.; Suzuki, K.; Goldberg, H.A.; Rittling, S.R.; Denhardt, D.T.; McCulloch, C.A.; Sodek, J. Osteopontin modulates CD44-dependent chemotaxis of peritoneal macrophages through G-protein-coupled receptors: Evidence of a role for an intracellular form of osteopontin. J. Cell. Physiol. 2004, 198, 155–167. [Google Scholar] [CrossRef]
  311. Wang, Y.; Yan, W.; Lu, X.; Qian, C.; Zhang, J.; Li, P.; Shi, L.; Zhao, P.; Fu, Z.; Pu, P.; et al. Overexpression of osteopontin induces angiogenesis of endothelial progenitor cells via the avbeta3/PI3K/AKT/eNOS/NO signaling pathway in glioma cells. Eur. J. Cell Biol. 2011, 90, 642–648. [Google Scholar] [CrossRef]
  312. Scatena, M.; Almeida, M.; Chaisson, M.L.; Fausto, N.; Nicosia, R.F.; Giachelli, C.M. NF-kappaB mediates alphavbeta3 integrin-induced endothelial cell survival. J. Cell Biol. 1998, 141, 1083–1093. [Google Scholar] [CrossRef]
  313. Raja, R.; Kale, S.; Thorat, D.; Soundararajan, G.; Lohite, K.; Mane, A.; Karnik, S.; Kundu, G.C. Hypoxia-driven osteopontin contributes to breast tumor growth through modulation of HIF1alpha-mediated VEGF-dependent angiogenesis. Oncogene 2014, 33, 2053–2064. [Google Scholar] [CrossRef] [PubMed]
  314. Lyle, A.N.; Remus, E.W.; Fan, A.E.; Lassegue, B.; Walter, G.A.; Kiyosue, A.; Griendling, K.K.; Taylor, W.R. Hydrogen peroxide regulates osteopontin expression through activation of transcriptional and translational pathways. J. Biol. Chem. 2014, 289, 275–285. [Google Scholar] [CrossRef]
  315. Lyle, A.N.; Joseph, G.; Fan, A.E.; Weiss, D.; Landazuri, N.; Taylor, W.R. Reactive oxygen species regulate osteopontin expression in a murine model of postischemic neovascularization. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1383–1391. [Google Scholar] [CrossRef]
  316. Okumura, N.; Nakamura, T.; Kay, E.P.; Nakahara, M.; Kinoshita, S.; Koizumi, N. R-spondin1 regulates cell proliferation of corneal endothelial cells via the Wnt3a/beta-catenin pathway. Invest. Ophthalmol. Vis. Sci. 2014, 55, 6861–6869. [Google Scholar] [CrossRef]
  317. Yoon, J.K.; Lee, J.S. Cellular signaling and biological functions of R-spondins. Cell Signal 2012, 24, 369–377. [Google Scholar] [CrossRef]
  318. Chen, P.H.; Chen, X.; Lin, Z.; Fang, D.; He, X. The structural basis of R-spondin recognition by LGR5 and RNF43. Genes. Dev. 2013, 27, 1345–1350. [Google Scholar] [CrossRef]
  319. Kim, K.A.; Wagle, M.; Tran, K.; Zhan, X.; Dixon, M.A.; Liu, S.; Gros, D.; Korver, W.; Yonkovich, S.; Tomasevic, N.; et al. R-Spondin family members regulate the Wnt pathway by a common mechanism. Mol. Biol. Cell 2008, 19, 2588–2596. [Google Scholar] [CrossRef] [PubMed]
  320. Jin, Y.R.; Yoon, J.K. The R-spondin family of proteins: Emerging regulators of WNT signaling. Int. J. Biochem. Cell Biol. 2012, 44, 2278–2287. [Google Scholar] [CrossRef] [PubMed]
  321. Nam, J.S.; Turcotte, T.J.; Smith, P.F.; Choi, S.; Yoon, J.K. Mouse cristin/R-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate beta-catenin-dependent gene expression. J. Biol. Chem. 2006, 281, 13247–13257. [Google Scholar] [CrossRef]
  322. de Lau, W.B.; Snel, B.; Clevers, H.C. The R-spondin protein family. Genome Biol. 2012, 13, 242. [Google Scholar] [CrossRef]
  323. Zerlin, M.; Julius, M.A.; Kitajewski, J. Wnt/Frizzled signaling in angiogenesis. Angiogenesis 2008, 11, 63–69. [Google Scholar] [CrossRef]
  324. Breton-Romero, R.; Feng, B.; Holbrook, M.; Farb, M.G.; Fetterman, J.L.; Linder, E.A.; Berk, B.D.; Masaki, N.; Weisbrod, R.M.; Inagaki, E.; et al. Endothelial Dysfunction in Human Diabetes Is Mediated by Wnt5a-JNK Signaling. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 561–569. [Google Scholar] [CrossRef]
  325. Kim, J.; Kim, J.; Kim, D.W.; Ha, Y.; Ihm, M.H.; Kim, H.; Song, K.; Lee, I. Wnt5a induces endothelial inflammation via beta-catenin-independent signaling. J. Immunol. 2010, 185, 1274–1282. [Google Scholar] [CrossRef] [PubMed]
  326. Skaria, T.; Burgener, J.; Bachli, E.; Schoedon, G. IL-4 Causes Hyperpermeability of Vascular Endothelial Cells through Wnt5A Signaling. PLoS ONE 2016, 11, e0156002. [Google Scholar] [CrossRef]
  327. Wright, M.; Aikawa, M.; Szeto, W.; Papkoff, J. Identification of a Wnt-responsive signal transduction pathway in primary endothelial cells. Biochem. Biophys. Res. Commun. 1999, 263, 384–388. [Google Scholar] [CrossRef]
  328. Masckauchan, T.N.; Shawber, C.J.; Funahashi, Y.; Li, C.M.; Kitajewski, J. Wnt/beta-catenin signaling induces proliferation, survival and interleukin-8 in human endothelial cells. Angiogenesis 2005, 8, 43–51. [Google Scholar] [CrossRef] [PubMed]
  329. Gore, A.V.; Swift, M.R.; Cha, Y.R.; Lo, B.; McKinney, M.C.; Li, W.; Castranova, D.; Davis, A.; Mukouyama, Y.S.; Weinstein, B.M. Rspo1/Wnt signaling promotes angiogenesis via Vegfc/Vegfr3. Development 2011, 138, 4875–4886. [Google Scholar] [CrossRef]
  330. Binnerts, M.E.; Kim, K.A.; Bright, J.M.; Patel, S.M.; Tran, K.; Zhou, M.; Leung, J.M.; Liu, Y.; Lomas, W.E., 3rd; Dixon, M.; et al. R-Spondin1 regulates Wnt signaling by inhibiting internalization of LRP6. Proc. Natl. Acad. Sci. USA 2007, 104, 14700–14705. [Google Scholar] [CrossRef]
  331. Caruso, M.; Ferranti, F.; Corano Scheri, K.; Dobrowolny, G.; Ciccarone, F.; Grammatico, P.; Catizone, A.; Ricci, G. R-spondin 1/dickkopf-1/beta-catenin machinery is involved in testicular embryonic angiogenesis. PLoS ONE 2015, 10, e0124213. [Google Scholar] [CrossRef]
  332. Jackson, S.R.; Costa, M.; Pastore, C.F.; Zhao, G.; Weiner, A.I.; Adams, S.; Palashikar, G.; Quansah, K.; Hankenson, K.; Herbert, D.R.; et al. R-spondin 2 mediates neutrophil egress into the alveolar space through increased lung permeability. BMC Res. Notes 2020, 13, 54. [Google Scholar] [CrossRef] [PubMed]
  333. Carmon, K.S.; Gong, X.; Yi, J.; Thomas, A.; Liu, Q. RSPO-LGR4 functions via IQGAP1 to potentiate Wnt signaling. Proc. Natl. Acad. Sci. USA 2014, 111, E1221–E1229. [Google Scholar] [CrossRef] [PubMed]
  334. Liu, F.; Zhao, Y.; Pei, Y.; Lian, F.; Lin, H. Role of the NF-kB signalling pathway in heterotopic ossification: Biological and therapeutic significance. Cell Commun. Signal 2024, 22, 159. [Google Scholar] [CrossRef]
  335. Tachibana, N.; Chijimatsu, R.; Okada, H.; Oichi, T.; Taniguchi, Y.; Maenohara, Y.; Miyahara, J.; Ishikura, H.; Iwanaga, Y.; Arino, Y.; et al. RSPO2 defines a distinct undifferentiated progenitor in the tendon/ligament and suppresses ectopic ossification. Sci. Adv. 2022, 8, eabn2138. [Google Scholar] [CrossRef]
  336. Kempe, S.; Kestler, H.; Lasar, A.; Wirth, T. NF-kappaB controls the global pro-inflammatory response in endothelial cells: Evidence for the regulation of a pro-atherogenic program. Nucleic Acids Res. 2005, 33, 5308–5319. [Google Scholar] [CrossRef] [PubMed]
  337. Aithabathula, R.V.; Kathuria, I.; Pervaiz, N.; Sharma, B.K.; Samake, T.I.; Ofosu-Boateng, M.; Gebreyesus, L.H.; Gyamfi, M.A.; Sprague, C.; Stayton, A.; et al. R-spondin 2 suppresses hepatic steatosis via activation of AMPK-ACC signaling. JHEP Rep. 2025, 101551, in press. [Google Scholar] [CrossRef]
  338. Skaria, T.; Bachli, E.; Schoedon, G. RSPO3 impairs barrier function of human vascular endothelial monolayers and synergizes with pro-inflammatory IL-1. Mol. Med. 2018, 24, 45. [Google Scholar] [CrossRef]
  339. Matsumoto, K.I.; Aoki, H. The Roles of Tenascins in Cardiovascular, Inflammatory, and Heritable Connective Tissue Diseases. Front. Immunol. 2020, 11, 609752. [Google Scholar] [CrossRef]
  340. Hashimoto, K.; Kajitani, N.; Miyamoto, Y.; Matsumoto, K.I. Wound healing-related properties detected in an experimental model with a collagen gel contraction assay are affected in the absence of tenascin-X. Exp. Cell Res. 2018, 363, 102–113. [Google Scholar] [CrossRef]
  341. Egging, D.; van Vlijmen-Willems, I.; van Tongeren, T.; Schalkwijk, J.; Peeters, A. Wound healing in tenascin-X deficient mice suggests that tenascin-X is involved in matrix maturation rather than matrix deposition. Connect. Tissue Res. 2007, 48, 93–98. [Google Scholar] [CrossRef]
  342. Rathjen, F.G.; Hodge, R. Early Days of Tenascin-R Research: Two Approaches Discovered and Shed Light on Tenascin-R. Front. Immunol. 2020, 11, 612482. [Google Scholar] [CrossRef]
  343. David, L.S.; Schachner, M.; Saghatelyan, A. The extracellular matrix glycoprotein tenascin-R affects adult but not developmental neurogenesis in the olfactory bulb. J. Neurosci. 2013, 33, 10324–10339. [Google Scholar] [CrossRef] [PubMed]
  344. Saghatelyan, A.; de Chevigny, A.; Schachner, M.; Lledo, P.M. Tenascin-R mediates activity-dependent recruitment of neuroblasts in the adult mouse forebrain. Nat. Neurosci. 2004, 7, 347–356. [Google Scholar] [CrossRef]
  345. Xu, J.C.; Xiao, M.F.; Jakovcevski, I.; Sivukhina, E.; Hargus, G.; Cui, Y.F.; Irintchev, A.; Schachner, M.; Bernreuther, C. The extracellular matrix glycoprotein tenascin-R regulates neurogenesis during development and in the adult dentate gyrus of mice. J. Cell Sci. 2014, 127, 641–652. [Google Scholar] [CrossRef] [PubMed]
  346. Hargus, G.; Cui, Y.; Schmid, J.S.; Xu, J.; Glatzel, M.; Schachner, M.; Bernreuther, C. Tenascin-R promotes neuronal differentiation of embryonic stem cells and recruitment of host-derived neural precursor cells after excitotoxic lesion of the mouse striatum. Stem Cells 2008, 26, 1973–1984. [Google Scholar] [CrossRef] [PubMed]
  347. Chiovaro, F.; Chiquet-Ehrismann, R.; Chiquet, M. Transcriptional regulation of tenascin genes. Cell Adh Migr. 2015, 9, 34–47. [Google Scholar] [CrossRef]
  348. Alcaraz, L.B.; Exposito, J.Y.; Chuvin, N.; Pommier, R.M.; Cluzel, C.; Martel, S.; Sentis, S.; Bartholin, L.; Lethias, C.; Valcourt, U. Tenascin-X promotes epithelial-to-mesenchymal transition by activating latent TGF-beta. J. Cell Biol. 2014, 205, 409–428. [Google Scholar] [CrossRef]
  349. Martina, E.; Degen, M.; Ruegg, C.; Merlo, A.; Lino, M.M.; Chiquet-Ehrismann, R.; Brellier, F. Tenascin-W is a specific marker of glioma-associated blood vessels and stimulates angiogenesis in vitro. FASEB J. 2010, 24, 778–787. [Google Scholar] [CrossRef]
  350. Midwood, K.S.; Hussenet, T.; Langlois, B.; Orend, G. Advances in tenascin-C biology. Cell Mol. Life Sci. 2011, 68, 3175–3199. [Google Scholar] [CrossRef]
  351. Midwood, K.; Sacre, S.; Piccinini, A.M.; Inglis, J.; Trebaul, A.; Chan, E.; Drexler, S.; Sofat, N.; Kashiwagi, M.; Orend, G.; et al. Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat. Med. 2009, 15, 774–780. [Google Scholar] [CrossRef]
  352. Willems, I.E.; Arends, J.W.; Daemen, M.J. Tenascin and fibronectin expression in healing human myocardial scars. J. Pathol. 1996, 179, 321–325. [Google Scholar] [CrossRef]
  353. Imanaka-Yoshida, K.; Yoshida, T.; Miyagawa-Tomita, S. Tenascin-C in development and disease of blood vessels. Anat. Rec. 2014, 297, 1747–1757. [Google Scholar] [CrossRef]
  354. Mehri, H.; Aslanabadi, N.; Nourazarian, A.; Shademan, B.; Khaki-Khatibi, F. Evaluation of the serum levels of Mannose binding lectin-2, tenascin-C, and total antioxidant capacity in patients with coronary artery disease. J. Clin. Lab. Anal. 2021, 35, e23967. [Google Scholar] [CrossRef]
  355. Liabeuf, S.; Barreto, D.V.; Kretschmer, A.; Barreto, F.C.; Renard, C.; Andrejak, M.; Choukroun, G.; Massy, Z. High circulating levels of large splice variants of tenascin-C is associated with mortality and cardiovascular disease in chronic kidney disease patients. Atherosclerosis 2011, 215, 116–124. [Google Scholar] [CrossRef]
  356. Gao, W.; Li, J.; Ni, H.; Shi, H.; Qi, Z.; Zhu, S.; Hao, C.; Xie, Q.; Luo, X.; Xie, K. Tenascin C: A Potential Biomarker for Predicting the Severity of Coronary Atherosclerosis. J. Atheroscler. Thromb. 2019, 26, 31–38. [Google Scholar] [CrossRef]
  357. Gholipour, A.; Shakerian, F.; Zahedmehr, A.; Oveisee, M.; Maleki, M.; Mowla, S.J.; Malakootian, M. Tenascin-C as a noninvasive biomarker of coronary artery disease. Mol. Biol. Rep. 2022, 49, 9267–9273. [Google Scholar] [CrossRef] [PubMed]
  358. Li, Y.; Liu, J.; Huang, J.W.; Song, J.C.; Ma, Z.L.; Shi, H.B. In vivo MRI detection of atherosclerosis in ApoE-deficient mice by using tenascin-C-targeted USPIO. Acta Radiol. 2018, 59, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
  359. Wang, Z.; Wei, Q.; Han, L.; Cao, K.; Lan, T.; Xu, Z.; Wang, Y.; Gao, Y.; Xue, J.; Shan, F.; et al. Tenascin-c renders a proangiogenic phenotype in macrophage via annexin II. J. Cell. Mol. Med. 2018, 22, 429–438. [Google Scholar] [CrossRef]
  360. Hedin, U.; Holm, J.; Hansson, G.K. Induction of tenascin in rat arterial injury. Relationship to altered smooth muscle cell phenotype. Am. J. Pathol. 1991, 139, 649–656. [Google Scholar] [PubMed]
  361. Rupp, T.; Langlois, B.; Koczorowska, M.M.; Radwanska, A.; Sun, Z.; Hussenet, T.; Lefebvre, O.; Murdamoothoo, D.; Arnold, C.; Klein, A.; et al. Tenascin-C Orchestrates Glioblastoma Angiogenesis by Modulation of Pro- and Anti-angiogenic Signaling. Cell Rep. 2016, 17, 2607–2619. [Google Scholar] [CrossRef]
  362. Saupe, F.; Schwenzer, A.; Jia, Y.; Gasser, I.; Spenle, C.; Langlois, B.; Kammerer, M.; Lefebvre, O.; Hlushchuk, R.; Rupp, T.; et al. Tenascin-C downregulates wnt inhibitor dickkopf-1, promoting tumorigenesis in a neuroendocrine tumor model. Cell Rep. 2013, 5, 482–492. [Google Scholar] [CrossRef]
  363. Luo, H.; Wang, J.; Qiao, C.; Zhang, X.; Zhang, W.; Ma, N. ATF3 Inhibits Tenascin-C-induced Foam Cell Formation in LPS-Stimulated THP-1 Macrophages by Suppressing TLR-4. J. Atheroscler. Thromb. 2015, 22, 1214–1223. [Google Scholar] [CrossRef]
  364. Liu, R.; He, Y.; Li, B.; Liu, J.; Ren, Y.; Han, W.; Wang, X.; Zhang, L. Tenascin-C produced by oxidized LDL-stimulated macrophages increases foam cell formation through Toll-like receptor-4. Mol. Cells 2012, 34, 35–41. [Google Scholar] [CrossRef] [PubMed]
  365. Hongu, T.; Pein, M.; Insua-Rodriguez, J.; Gutjahr, E.; Mattavelli, G.; Meier, J.; Decker, K.; Descot, A.; Bozza, M.; Harbottle, R.; et al. Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs. Nat. Cancer 2022, 3, 486–504. [Google Scholar] [CrossRef]
  366. Radwanska, A.; Grall, D.; Schaub, S.; Divonne, S.B.F.; Ciais, D.; Rekima, S.; Rupp, T.; Sudaka, A.; Orend, G.; Van Obberghen-Schilling, E. Counterbalancing anti-adhesive effects of Tenascin-C through fibronectin expression in endothelial cells. Sci. Rep. 2017, 7, 12762. [Google Scholar] [CrossRef]
  367. Matsui, K.; Torii, S.; Hara, S.; Maruyama, K.; Arai, T.; Imanaka-Yoshida, K. Tenascin-C in Tissue Repair after Myocardial Infarction in Humans. Int. J. Mol. Sci. 2023, 24, 10184. [Google Scholar] [CrossRef] [PubMed]
  368. Okada, T.; Suzuki, H. The Role of Tenascin-C in Tissue Injury and Repair After Stroke. Front. Immunol. 2020, 11, 607587. [Google Scholar] [CrossRef]
  369. Babic, A.M.; Chen, C.C.; Lau, L.F. Fisp12/mouse connective tissue growth factor mediates endothelial cell adhesion and migration through integrin alphavbeta3, promotes endothelial cell survival, and induces angiogenesis in vivo. Mol. Cell. Biol. 1999, 19, 2958–2966. [Google Scholar] [CrossRef] [PubMed]
  370. Zaykov, V.; Chaqour, B. The CCN2/CTGF interactome: An approach to understanding the versatility of CCN2/CTGF molecular activities. J. Cell Commun. Signal 2021, 15, 567–580. [Google Scholar] [CrossRef]
  371. Wang, Y.K.; Weng, H.K.; Mo, F.E. The regulation and functions of the matricellular CCN proteins induced by shear stress. J. Cell Commun. Signal 2023, 17, 361–370. [Google Scholar] [CrossRef]
  372. Luan, Y.; Zhang, H.; Ma, K.; Liu, Y.; Lu, H.; Chen, X.; Liu, Y.; Zhang, Z. CCN3/NOV Regulates Proliferation and Neuronal Differentiation in Mouse Hippocampal Neural Stem Cells via the Activation of the Notch/PTEN/AKT Pathway. Int. J. Mol. Sci. 2023, 24, 10324. [Google Scholar] [CrossRef]
  373. Jun, J.I.; Lau, L.F. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 945–963. [Google Scholar] [CrossRef] [PubMed]
  374. Naughton, M.; Moffat, J.; Eleftheriadis, G.; de la Vega Gallardo, N.; Young, A.; Falconer, J.; Hawkins, K.; Pearson, B.; Perbal, B.; Hogan, A.; et al. CCN3 is dynamically regulated by treatment and disease state in multiple sclerosis. J. Neuroinflammation 2020, 17, 349. [Google Scholar] [CrossRef]
  375. Russo, J.W.; Castellot, J.J. CCN5: Biology and pathophysiology. J. Cell Commun. Signal 2010, 4, 119–130. [Google Scholar] [CrossRef] [PubMed]
  376. Karagiannis, E.D.; Popel, A.S. Peptides derived from type I thrombospondin repeat-containing proteins of the CCN family inhibit proliferation and migration of endothelial cells. Int. J. Biochem. Cell Biol. 2007, 39, 2314–2323. [Google Scholar] [CrossRef]
  377. Tanaka, I.; Morikawa, M.; Okuse, T.; Shirakawa, M.; Imai, K. Expression and regulation of WISP2 in rheumatoid arthritic synovium. Biochem. Biophys. Res. Commun. 2005, 334, 973–978. [Google Scholar] [CrossRef] [PubMed]
  378. Delmolino, L.M.; Stearns, N.A.; Castellot, J.J., Jr. COP-1, a member of the CCN family, is a heparin-induced growth arrest specific gene in vascular smooth muscle cells. J. Cell. Physiol. 2001, 188, 45–55. [Google Scholar] [CrossRef]
  379. Hilfiker, A.; Hilfiker-Kleiner, D.; Fuchs, M.; Kaminski, K.; Lichtenberg, A.; Rothkotter, H.J.; Schieffer, B.; Drexler, H. Expression of CYR61, an angiogenic immediate early gene, in arteriosclerosis and its regulation by angiotensin II. Circulation 2002, 106, 254–260. [Google Scholar] [CrossRef]
  380. Malik, A.R.; Liszewska, E.; Jaworski, J. Matricellular proteins of the Cyr61/CTGF/NOV (CCN) family and the nervous system. Front. Cell. Neurosci. 2015, 9, 237. [Google Scholar] [CrossRef]
  381. Zuo, G.W.; Kohls, C.D.; He, B.C.; Chen, L.; Zhang, W.; Shi, Q.; Zhang, B.Q.; Kang, Q.; Luo, J.; Luo, X.; et al. The CCN proteins: Important signaling mediators in stem cell differentiation and tumorigenesis. Histol. Histopathol. 2010, 25, 795–806. [Google Scholar] [CrossRef]
  382. Perbal, B.V.; Takigawa, M. CCN Proteins: A New Family of Cell Growth and Differentiation Regulators; London Imperial College Press: London, UK, 2005. [Google Scholar]
  383. Parisi, M.S.; Gazzerro, E.; Rydziel, S.; Canalis, E. Expression and regulation of CCN genes in murine osteoblasts. Bone 2006, 38, 671–677. [Google Scholar] [CrossRef]
  384. Lau, L.F.; Lam, S.C. The CCN family of angiogenic regulators: The integrin connection. Exp. Cell Res. 1999, 248, 44–57. [Google Scholar] [CrossRef] [PubMed]
  385. Hsu, P.L.; Chen, J.S.; Wang, C.Y.; Wu, H.L.; Mo, F.E. Shear-Induced CCN1 Promotes Atheroprone Endothelial Phenotypes and Atherosclerosis. Circulation 2019, 139, 2877–2891. [Google Scholar] [CrossRef]
  386. Su, B.C.; Hsu, P.L.; Mo, F.E. CCN1 triggers adaptive autophagy in cardiomyocytes to curb its apoptotic activities. J. Cell Commun. Signal 2020, 14, 93–100. [Google Scholar] [CrossRef] [PubMed]
  387. Kubota, S.; Takigawa, M. CCN family proteins and angiogenesis: From embryo to adulthood. Angiogenesis 2007, 10, 1–11. [Google Scholar] [CrossRef]
  388. Park, M.H.; Kim, A.K.; Manandhar, S.; Oh, S.Y.; Jang, G.H.; Kang, L.; Lee, D.W.; Hyeon, D.Y.; Lee, S.H.; Lee, H.E.; et al. CCN1 interlinks integrin and hippo pathway to autoregulate tip cell activity. Elife 2019, 8, e46012. [Google Scholar] [CrossRef]
  389. Niu, C.C.; Zhao, C.; Yang, Z.; Zhang, X.L.; Pan, J.; Zhao, C.; Si, W.K. Inhibiting CCN1 blocks AML cell growth by disrupting the MEK/ERK pathway. Cancer Cell Int. 2014, 14, 74. [Google Scholar] [CrossRef]
  390. Xie, D.; Yin, D.; Tong, X.; O’Kelly, J.; Mori, A.; Miller, C.; Black, K.; Gui, D.; Said, J.W.; Koeffler, H.P. Cyr61 is overexpressed in gliomas and involved in integrin-linked kinase-mediated Akt and beta-catenin-TCF/Lef signaling pathways. Cancer Res. 2004, 64, 1987–1996. [Google Scholar] [CrossRef] [PubMed]
  391. Yeger, H.; Perbal, B. The CCN axis in cancer development and progression. J. Cell Commun. Signal 2021, 15, 491–517. [Google Scholar] [CrossRef]
  392. Li, Z.Q.; Ding, W.; Sun, S.J.; Li, J.; Pan, J.; Zhao, C.; Wu, W.R.; Si, W.K. Cyr61/CCN1 is regulated by Wnt/beta-catenin signaling and plays an important role in the progression of hepatocellular carcinoma. PLoS ONE 2012, 7, e35754. [Google Scholar] [CrossRef]
  393. Li, H.; Li, T.; Wang, H.; He, X.; Li, Y.; Wen, S.; Peng, R.; Nie, Y.; Lu, Y.; Yang, H.; et al. Diabetes Promotes Retinal Vascular Endothelial Cell Injury by Inducing CCN1 Expression. Front. Cardiovasc. Med. 2021, 8, 689318. [Google Scholar] [CrossRef] [PubMed]
  394. Li, T.; Qian, Y.; Li, H.; Wang, T.; Jiang, Q.; Wang, Y.; Zhu, Y.; Li, S.; He, X.; Shi, G.; et al. Cellular communication network factor 1 promotes retinal leakage in diabetic retinopathy via inducing neutrophil stasis and neutrophil extracellular traps extrusion. Cell Commun. Signal 2024, 22, 275. [Google Scholar] [CrossRef] [PubMed]
  395. Bai, T.; Chen, C.C.; Lau, L.F. Matricellular protein CCN1 activates a proinflammatory genetic program in murine macrophages. J. Immunol. 2010, 184, 3223–3232. [Google Scholar] [CrossRef]
  396. Matsumae, H.; Yoshida, Y.; Ono, K.; Togi, K.; Inoue, K.; Furukawa, Y.; Nakashima, Y.; Kojima, Y.; Nobuyoshi, M.; Kita, T.; et al. CCN1 knockdown suppresses neointimal hyperplasia in a rat artery balloon injury model. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1077–1083. [Google Scholar] [CrossRef]
  397. Lobel, M.; Bauer, S.; Meisel, C.; Eisenreich, A.; Kudernatsch, R.; Tank, J.; Rauch, U.; Kuhl, U.; Schultheiss, H.P.; Volk, H.D.; et al. CCN1: A novel inflammation-regulated biphasic immune cell migration modulator. Cell Mol. Life Sci. 2012, 69, 3101–3113. [Google Scholar] [CrossRef] [PubMed]
  398. Jorgensen, L.H.; Petersson, S.J.; Sellathurai, J.; Andersen, D.C.; Thayssen, S.; Sant, D.J.; Jensen, C.H.; Schroder, H.D. Secreted protein acidic and rich in cysteine (SPARC) in human skeletal muscle. J. Histochem. Cytochem. 2009, 57, 29–39. [Google Scholar] [CrossRef]
  399. Parfenova, O.K.; Kukes, V.G.; Grishin, D.V. Follistatin-Like Proteins: Structure, Functions and Biomedical Importance. Biomedicines 2021, 9, 999. [Google Scholar] [CrossRef]
  400. Sage, H.; Vernon, R.B.; Funk, S.E.; Everitt, E.A.; Angello, J. SPARC, a secreted protein associated with cellular proliferation, inhibits cell spreading in vitro and exhibits Ca+2-dependent binding to the extracellular matrix. J. Cell Biol. 1989, 109, 341–356. [Google Scholar] [CrossRef]
  401. Chandrasekhar, S.; Harvey, A.K.; Johnson, M.G.; Becker, G.W. Osteonectin/SPARC is a product of articular chondrocytes/cartilage and is regulated by cytokines and growth factors. Biochim. Biophys. Acta 1994, 1221, 7–14. [Google Scholar] [CrossRef]
  402. Ham, S.M.; Song, M.J.; Yoon, H.S.; Lee, D.H.; Chung, J.H.; Lee, S.T. SPARC Is Highly Expressed in Young Skin and Promotes Extracellular Matrix Integrity in Fibroblasts via the TGF-beta Signaling Pathway. Int. J. Mol. Sci. 2023, 24, 12179. [Google Scholar] [CrossRef]
  403. Hu, S.; Zhang, X.; Ding, Y.; Liu, X.; Xia, R.; Wang, X. Inhibition of SPARC signal by aerobic exercise to ameliorate atherosclerosis. Int. Immunopharmacol. 2024, 132, 111856. [Google Scholar] [CrossRef]
  404. Bradshaw, A.D.; Sage, E.H. SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury. J. Clin. Investig. 2001, 107, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  405. Wong, S.L.; Sukkar, M.B. The SPARC protein: An overview of its role in lung cancer and pulmonary fibrosis and its potential role in chronic airways disease. Br. J. Pharmacol. 2017, 174, 3–14. [Google Scholar] [CrossRef] [PubMed]
  406. Kelly, K.A.; Allport, J.R.; Yu, A.M.; Sinh, S.; Sage, E.H.; Gerszten, R.E.; Weissleder, R. SPARC is a VCAM-1 counter-ligand that mediates leukocyte transmigration. J. Leukoc. Biol. 2007, 81, 748–756. [Google Scholar] [CrossRef]
  407. Kupprion, C.; Motamed, K.; Sage, E.H. SPARC (BM-40, osteonectin) inhibits the mitogenic effect of vascular endothelial growth factor on microvascular endothelial cells. J. Biol. Chem. 1998, 273, 29635–29640. [Google Scholar] [CrossRef] [PubMed]
  408. Sage, H.; Decker, J.; Funk, S.; Chow, M. SPARC: A Ca2+-binding extracellular protein associated with endothelial cell injury and proliferation. J. Mol. Cell. Cardiol. 1989, 21 (Suppl. 1), 13–22. [Google Scholar] [CrossRef]
  409. Funk, S.E.; Sage, E.H. The Ca2(+)-binding glycoprotein SPARC modulates cell cycle progression in bovine aortic endothelial cells. Proc. Natl. Acad. Sci. USA 1991, 88, 2648–2652. [Google Scholar] [CrossRef]
  410. Raines, E.W.; Lane, T.F.; Iruela-Arispe, M.L.; Ross, R.; Sage, E.H. The extracellular glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-AB and -BB and inhibits the binding of PDGF to its receptors. Proc. Natl. Acad. Sci. USA 1992, 89, 1281–1285. [Google Scholar] [CrossRef]
  411. Goldblum, S.E.; Ding, X.; Funk, S.E.; Sage, E.H. SPARC (secreted protein acidic and rich in cysteine) regulates endothelial cell shape and barrier function. Proc. Natl. Acad. Sci. USA 1994, 91, 3448–3452. [Google Scholar] [CrossRef]
  412. Zhao, Y.; Yu, J.; Huang, A.; Yang, Q.; Li, G.; Yang, Y.; Chen, Y. ROS impairs tumor vasculature normalization through an endocytosis effect of caveolae on extracellular SPARC. Cancer Cell Int. 2023, 23, 152. [Google Scholar] [CrossRef]
  413. Alkabie, S.; Basivireddy, J.; Zhou, L.; Roskams, J.; Rieckmann, P.; Quandt, J.A. SPARC expression by cerebral microvascular endothelial cells in vitro and its influence on blood-brain barrier properties. J. Neuroinflammation 2016, 13, 225. [Google Scholar] [CrossRef]
  414. Rivera, L.B.; Brekken, R.A. SPARC promotes pericyte recruitment via inhibition of endoglin-dependent TGF-beta1 activity. J. Cell Biol. 2011, 193, 1305–1319. [Google Scholar] [CrossRef]
  415. Arnold, S.; Mira, E.; Muneer, S.; Korpanty, G.; Beck, A.W.; Holloway, S.E.; Manes, S.; Brekken, R.A. Forced expression of MMP9 rescues the loss of angiogenesis and abrogates metastasis of pancreatic tumors triggered by the absence of host SPARC. Exp. Biol. Med. 2008, 233, 860–873. [Google Scholar] [CrossRef]
  416. Francki, A.; Bradshaw, A.D.; Bassuk, J.A.; Howe, C.C.; Couser, W.G.; Sage, E.H. SPARC regulates the expression of collagen type I and transforming growth factor-beta1 in mesangial cells. J. Biol. Chem. 1999, 274, 32145–32152. [Google Scholar] [CrossRef] [PubMed]
  417. Jendraschak, E.; Sage, E.H. Regulation of angiogenesis by SPARC and angiostatin: Implications for tumor cell biology. Semin. Cancer Biol. 1996, 7, 139–146. [Google Scholar] [CrossRef] [PubMed]
  418. Sage, E.H.; Reed, M.; Funk, S.E.; Truong, T.; Steadele, M.; Puolakkainen, P.; Maurice, D.H.; Bassuk, J.A. Cleavage of the matricellular protein SPARC by matrix metalloproteinase 3 produces polypeptides that influence angiogenesis. J. Biol. Chem. 2003, 278, 37849–37857. [Google Scholar] [CrossRef] [PubMed]
  419. Martel, C.; Li, W.; Fulp, B.; Platt, A.M.; Gautier, E.L.; Westerterp, M.; Bittman, R.; Tall, A.R.; Chen, S.H.; Thomas, M.J.; et al. Lymphatic vasculature mediates macrophage reverse cholesterol transport in mice. J. Clin. Investig. 2013, 123, 1571–1579. [Google Scholar] [CrossRef]
  420. Vuorio, T.; Nurmi, H.; Moulton, K.; Kurkipuro, J.; Robciuc, M.R.; Ohman, M.; Heinonen, S.E.; Samaranayake, H.; Heikura, T.; Alitalo, K.; et al. Lymphatic vessel insufficiency in hypercholesterolemic mice alters lipoprotein levels and promotes atherogenesis. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1162–1170. [Google Scholar] [CrossRef]
  421. Kojima, Y.; Volkmer, J.P.; McKenna, K.; Civelek, M.; Lusis, A.J.; Miller, C.L.; Direnzo, D.; Nanda, V.; Ye, J.; Connolly, A.J.; et al. CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis. Nature 2016, 536, 86–90. [Google Scholar] [CrossRef]
  422. Jarr, K.U.; Nakamoto, R.; Doan, B.H.; Kojima, Y.; Weissman, I.L.; Advani, R.H.; Iagaru, A.; Leeper, N.J. Effect of CD47 Blockade on Vascular Inflammation. N. Engl. J. Med. 2021, 384, 382–383. [Google Scholar] [CrossRef]
  423. Bouwstra, R.; van Meerten, T.; Bremer, E. CD47-SIRPalpha blocking-based immunotherapy: Current and prospective therapeutic strategies. Clin. Transl. Med. 2022, 12, e943. [Google Scholar] [CrossRef]
  424. Maute, R.; Xu, J.; Weissman, I.L. CD47-SIRPalpha-targeted therapeutics: Status and prospects. Immunooncol. Technol. 2022, 13, 100070. [Google Scholar] [CrossRef]
  425. Flores, A.M.; Hosseini-Nassab, N.; Jarr, K.U.; Ye, J.; Zhu, X.; Wirka, R.; Koh, A.L.; Tsantilas, P.; Wang, Y.; Nanda, V.; et al. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat. Nanotechnol. 2020, 15, 154–161. [Google Scholar] [CrossRef]
  426. Bamezai, S.; Zhang, Y.; Kumari, M.; Lotfi, M.; Alsaigh, T.; Luo, L.; Kumar, G.S.; Wang, F.; Ye, J.; Puri, M.; et al. Pro-efferocytic nanotherapies reduce vascular inflammation without inducing anemia in a large animal model of atherosclerosis. Nat. Commun. 2024, 15, 8034. [Google Scholar] [CrossRef]
  427. Afroz, R.; Goodwin, J.E. Wnt Signaling in Atherosclerosis: Mechanisms to Therapeutic Implications. Biomedicines 2024, 12, 276. [Google Scholar] [CrossRef]
  428. Lang, F.; Li, Y.; Yao, R.; Jiang, M. Osteopontin in Chronic Inflammatory Diseases: Mechanisms, Biomarker Potential, and Therapeutic Strategies. Biology 2025, 14, 428. [Google Scholar] [CrossRef]
  429. Ge, Q.; Ruan, C.C.; Ma, Y.; Tang, X.F.; Wu, Q.H.; Wang, J.G.; Zhu, D.L.; Gao, P.J. Osteopontin regulates macrophage activation and osteoclast formation in hypertensive patients with vascular calcification. Sci. Rep. 2017, 7, 40253. [Google Scholar] [CrossRef] [PubMed]
  430. Huang, X.; Zhang, Y.; Zhang, W.; Qin, C.; Zhu, Y.; Fang, Y.; Wang, Y.; Tang, C.; Cao, F. Osteopontin-Targeted and PPARdelta-Agonist-Loaded Nanoparticles Efficiently Reduce Atherosclerosis in Apolipoprotein E(-/-) Mice. ACS Omega 2022, 7, 28767–28778. [Google Scholar] [CrossRef] [PubMed]
  431. Chen, W.; Wang, Y.; Ren, C.; Yu, S.; Wang, C.; Xing, J.; Xu, J.; Yan, S.; Zhang, T.; Li, Q.; et al. The role of TNC in atherosclerosis and drug development opportunities. Int. J. Biol. Sci. 2024, 20, 127–136. [Google Scholar] [CrossRef] [PubMed]
  432. Kathuria, I.; Prasad, A.; Sharma, B.K.; Aithabathula, R.V.; Ofosu-Boateng, M.; Gyamfi, M.A.; Jiang, J.; Park, F.; Singh, U.P.; Singla, B. Nidogen 2 Overexpression Promotes Hepatosteatosis and Atherosclerosis. Int. J. Mol. Sci. 2024, 25, 12782. [Google Scholar] [CrossRef] [PubMed]
  433. Pervaiz, N.; Mehmood, R.; Aithabathula, R.V.; Kathuria, I.; Ahn, W.; Le, B.T.; Kim, K.S.; Singh, U.P.; Csanyi, G.; Singla, B. Smooth muscle cell-specific CD47 deletion suppresses atherosclerosis. Life Sci. 2025, 361, 123315. [Google Scholar] [CrossRef] [PubMed]
Figure 2. Role of TSP1 and TSP5 in endothelial dysfunction: Schematic representation of various receptors present on ECs that interact with matricellular proteins TSP1 and TSP5 and downstream signaling pathways. (A) TSP1 via binding to CD36 facilitates the formation of a CD36-Fyn complex. This complex amplifies Fyn-mediated signal transduction. Additionally, TSP1 induces the dissociation of VEGFR2 from CD47, leading to the association of VEGFR2 with CD36. (B) The illustration of interactions between TSP5 and integrins and downstream key signaling pathways. TSP5-integrin complex modulates the expression of cytokine LTB, BCL2, HEY1, and BIK, ultimately promoting angiogenic responses. ↑ (red): increased signaling/expression; ↓ (red): increased signaling/expression; ↔ (black): interaction; Ⱶ (red): inhibition.
Figure 2. Role of TSP1 and TSP5 in endothelial dysfunction: Schematic representation of various receptors present on ECs that interact with matricellular proteins TSP1 and TSP5 and downstream signaling pathways. (A) TSP1 via binding to CD36 facilitates the formation of a CD36-Fyn complex. This complex amplifies Fyn-mediated signal transduction. Additionally, TSP1 induces the dissociation of VEGFR2 from CD47, leading to the association of VEGFR2 with CD36. (B) The illustration of interactions between TSP5 and integrins and downstream key signaling pathways. TSP5-integrin complex modulates the expression of cytokine LTB, BCL2, HEY1, and BIK, ultimately promoting angiogenic responses. ↑ (red): increased signaling/expression; ↓ (red): increased signaling/expression; ↔ (black): interaction; Ⱶ (red): inhibition.
Antioxidants 14 01338 g002
Figure 3. Role of RSPOs in endothelial dysfunction: Schematic representation of various receptors present on ECs that interact with RSPO2 and RSPO3, and their signaling pathways. (A) RSPO2 binds to LGR4 receptors on LECs. This interaction inhibits VEGF-C-induced activation of AKT and eNOS, reducing NO production and impairing lymphatic vessel formation. Furthermore, RSPO2 inhibits the canonical Wnt-β-catenin pathway in LECs in a NO-dependent manner, decreasing lymphatic vessel-mediated LDL drainage from arterial walls and contributing to atherosclerosis development. (B) RSPO3 activates non-canonical Wnt signaling, including WNT/Ca2+/NFAT, which is crucial for vascular remodeling and inhibits angiogenesis. ↑ (red): increased signaling/expression; Ⱶ (red): inhibition.
Figure 3. Role of RSPOs in endothelial dysfunction: Schematic representation of various receptors present on ECs that interact with RSPO2 and RSPO3, and their signaling pathways. (A) RSPO2 binds to LGR4 receptors on LECs. This interaction inhibits VEGF-C-induced activation of AKT and eNOS, reducing NO production and impairing lymphatic vessel formation. Furthermore, RSPO2 inhibits the canonical Wnt-β-catenin pathway in LECs in a NO-dependent manner, decreasing lymphatic vessel-mediated LDL drainage from arterial walls and contributing to atherosclerosis development. (B) RSPO3 activates non-canonical Wnt signaling, including WNT/Ca2+/NFAT, which is crucial for vascular remodeling and inhibits angiogenesis. ↑ (red): increased signaling/expression; Ⱶ (red): inhibition.
Antioxidants 14 01338 g003
Figure 4. Role of CCN1- and TN-C-regulated signaling in angiogenesis: (A) The interaction between CCN1 and αvβ3 activates VEGFR2, leading to downstream MAPK/PI3K-YAP/TAZ signaling and regulation of actin polymerization via Rho effector mDia1. (B) TN-C interaction with various cell surface receptors and ECM components. Signaling via integrins, TLR4, and Wnt receptors to intracellular effectors such as MAPK leads to changes in gene transcription, resulting in altered expression of proteins involved in proliferation, adhesion, and cell survival/apoptotic responses. ↑ (red): increased signaling/expression; ↔ (black): interaction; Ⱶ (red): inhibition.
Figure 4. Role of CCN1- and TN-C-regulated signaling in angiogenesis: (A) The interaction between CCN1 and αvβ3 activates VEGFR2, leading to downstream MAPK/PI3K-YAP/TAZ signaling and regulation of actin polymerization via Rho effector mDia1. (B) TN-C interaction with various cell surface receptors and ECM components. Signaling via integrins, TLR4, and Wnt receptors to intracellular effectors such as MAPK leads to changes in gene transcription, resulting in altered expression of proteins involved in proliferation, adhesion, and cell survival/apoptotic responses. ↑ (red): increased signaling/expression; ↔ (black): interaction; Ⱶ (red): inhibition.
Antioxidants 14 01338 g004
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

Aithabathula, R.V.; Kumar, S.; Singla, B. Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis. Antioxidants 2025, 14, 1338. https://doi.org/10.3390/antiox14111338

AMA Style

Aithabathula RV, Kumar S, Singla B. Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis. Antioxidants. 2025; 14(11):1338. https://doi.org/10.3390/antiox14111338

Chicago/Turabian Style

Aithabathula, Ravi Varma, Santosh Kumar, and Bhupesh Singla. 2025. "Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis" Antioxidants 14, no. 11: 1338. https://doi.org/10.3390/antiox14111338

APA Style

Aithabathula, R. V., Kumar, S., & Singla, B. (2025). Role of Matricellular Proteins in Endothelial Cell Inflammation and Atherosclerosis. Antioxidants, 14(11), 1338. https://doi.org/10.3390/antiox14111338

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