315
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases

, , , , , ORCID Icon, , , & ORCID Icon show all
Pages 3593-3617 | Received 21 Apr 2023, Accepted 02 Aug 2023, Published online: 23 Aug 2023

Abstract

Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.

Introduction

Endothelial cells (ECs), residing as the innermost layer of blood vessel walls, serve as a critical interface between circulating blood components and the vessel wall itself.Citation1 These cells interact with substances in the flowing blood while participating in numerous physiological and pathological processes, including metabolism, antioxidant reduction state, inflammation, and immune response.Citation1,Citation2 As principal regulators of vascular homeostasis, ECs fulfill various functions, such as modulating vascular tension and managing hemostasis and thrombosis.Citation3 Positioned at the nexus of blood and tissue, ECs are particularly vulnerable to alterations in blood flow and its constituents.Citation3 Consequently, exposure to specific cytokines or pro-inflammatory stimuli may prompt a transition from an anti-thrombotic, anti-inflammatory, and vasodilatory state to one predisposed to coagulation, inflammation, and vasoconstriction.Citation4

Inflammation refers to a complex set of biological response processes triggered by various stimulation, including biological, chemical, or physical factors.Citation5,Citation6 As pivotal effector cells in initiating inflammation, ECs orchestrate the body’s response to systemic inflammation, modulate vascular function, and contribute to the pathogenesis of vascular diseases.Citation7 Although inflammation functions as a self-protective mechanism, dysregulated inflammatory responses can ultimately give rise to various inflammatory disorders, including obesity, hypertension, atherosclerosis (AS), autoimmune diseases, neurodegenerative conditions, diabetes mellitus (DM), sepsis, cardiovascular disease (CVD), and cancer.Citation6,Citation8–10 Hence, a deeper understanding of the interplay between ECs and inflammation is essential. In recent years, reports have highlighted the central position of ECs in inflammatory processes and the involvement of inflammation in the onset and progression of diverse diseases.Citation4,Citation11,Citation12 This review delineates the role of ECs in inflammatory processes, the signaling pathways associated with AS, DM, and related complications, respiratory diseases (RD), sepsis, and CVD, and corresponding therapeutic effects. The objective is to enhance researchers’ comprehension of vascular-related diseases and provide a foundation for future in-depth investigations and clinical interventions involving ECs and inflammation.

There are reports suggesting that with increasing age, the vascular endothelium may develop into a pro-inflammatory state, potentially leading to vascular endothelial dysfunction and CVD.Citation13 Some findings demonstrate that aging is associated with the development of a proinflammatory phenotype in the vascular endothelium of healthy adults, which may be caused in part by a reduction in IκB-mediated NF-κB activation. The elevated nuclear content of NF-κB in ECs of healthy older adults with impaired vascular endothelial function, compared to younger healthy subjects, offers compelling evidence suggesting that NF-κB might be involved in the molecular mechanisms contributing to age-related vascular inflammation, endothelial dysfunction, and CVD in humans.Citation13 Moreover, some findings support the hypothesis that serum 25-hydroxyvitamin D status is associated with vascular endothelial function in middle-aged and elderly patients without clinical disease. Scholars’ research findings indicate that reduced levels of 25-hydroxy vitamin D are linked to higher expression of NF-κB and IL-6, along with increased NF-κB-associated suppression of vascular endothelial function. Additionally, it could be linked to the decrease in the expression of vitamin D receptors and 1-hydroxylase in vascular ECs.Citation14 Furthermore, in a randomized crossover experimental design, scholars discovered that habitual aerobic exercise training may enhance vascular endothelial function in older adults through targeting the NF-κB signaling pathway, which mediates age-related endothelium-dependent dilation in humans. The study also suggests that improving endothelial function in sedentary older individuals can potentially reduce the risk of CVD.Citation15

Literature Inclusion and Exclusion Criteria

Employing the keywords “endothelial cells”, “endothelium”, and “inflammation”, we systematically searched for English-language literature published between 2000, 2012, 2016 and 2022 within the Web of Science and PubMed databases. The initial phase involved screening article titles and abstracts, which was followed by a comprehensive full-text assessment of the articles. Ultimately, 102 papers that fulfilled the inclusion and exclusion criteria were selected.

The inclusion criteria for articles comprised clinical studies and fundamental research focusing on endothelial inflammation, as well as animal models, cell models, therapeutics, inducers, and signaling pathways. The exclusion criteria encompassed non-English studies and those deemed unsuitable for endothelial inflammation research. The investigations retrieved from the literature spanned five disease areas: atherosclerosis (AS), cardiovascular disease (CVD), diabetes mellitus (DM), respiratory disease (RD), and sepsis.

Endothelial Cells and Inflammation

Under physiological conditions, ECs maintain vascular health by regulating blood flow and distributing nutrients, hormones, and other essential substances.Citation16–18 They possess the ability to modulate vascular tension, manage hemostasis and thrombosis, inhibit leukocyte adhesion, and control vascular inflammation through vasoconstriction or relaxation.Citation3,Citation16,Citation19–22 Inflammation constitutes a vital component of innate immunity, safeguarding the host from infection.Citation23

As the innermost layer of blood vessels, ECs not only furnish a dynamic interface between circulating blood components and adjacent tissues but also play a crucial role in preserving blood homeostasis and preventing tissue damage.Citation24 ECs serve as potential targets for lipids, bacterial endotoxins, inflammatory cytokines (tumor necrosis factor (TNF)-α, ILs, interferon-γ), and microbial agents, with alterations in their functions eliciting inflammatory responses in tissues and organs. Concurrently, vascular inflammation provokes abnormal activation of ECs, leading to dysfunction and structural abnormalities in blood vessels.Citation25

Since ECs continuously perceive the extracellular environment, inflammatory stimuli can compromise their barrier function, making them indicative of systemic inflammation.Citation7 ECs not only safeguard human health but also operate as inflammation mediators, influencing the progression and outcomes of vascular inflammatory diseases.Citation26 Dysregulated EC activation or dysfunction is considered the initial step in the pathogenesis of vascular inflammatory disorders.Citation27 Inflammation lies at the core of vascular-related diseases, and endothelial inflammation contributes to a wide variety of diseases, such as highly prevalent conditions such as AS, DM, end-stage renal disease, CVD, etc.Citation28–32 Therefore, EC inflammation warrants attention and thorough investigation. With a deeper understanding of the mechanism of vascular inflammation, we hope to find relevant disease markers or novel therapeutics to assist clinical diagnosis and improve treatment effectiveness.

The Role of Endothelial Cells Inflammation in Different Diseases

Atherosclerosis

Atherosclerosis (AS) arises from the excessive accumulation of lipids and other substances within the arterial intima.Citation33 This vascular disease, characterized by endothelial inflammation, serves as a major underlying cause of CVD.Citation34,Citation35 The pathological process of AS is typified by chronic inflammatory reactions, resulting from excessive inflammatory responses to various forms of damage.Citation31 ECs not only constitute the interface between blood and the arterial intima but also represent the site of AS initiation.Citation33 Experimental findings indicate that inflammation participates in AS development, with lipids and other traditional risk factors linked to AS via numerous pathways facilitated by inflammatory reactions.Citation36,Citation37

Under homeostatic conditions, thrombo regulatory proteins and heparan sulfate proteoglycans on the EC surface, as well as nitric oxide and prostacyclin produced by ECs, contribute to the anticoagulant and anti-thrombotic properties of the normal endothelium.Citation33,Citation38,Citation39 However, in pathological contexts, inflammatory stimuli activate ECs, leading to the up-regulation of cell adhesion molecules such as E-selectin, intercellular adhesion molecules (ICAM), and vascular cell adhesion molecules (VCAM). This process triggers leukocyte exudation following their rolling on the endothelial surface.Citation39–41 In summary, EC inflammation is implicated in the progression of AS, and the suppression of inflammation also constitutes an effective treatment for AS.Citation42,Citation43

The impacts of specific key molecules on inflammation and AS are detailed below. In mouse aortic endothelial cells (MAECs) and human umbilical vein endothelial cells (HUVECs) stimulated by interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α), it has been demonstrated that Krüppel-like factor 14 (KLF14), a transcription factor linked to coronary artery disease (CAD), mitigates inflammation by inhibiting the NF-κB signaling pathway.Citation44 Similarly, in Ox-LDL-mediated HUVECs, silencing circular RNA circ_0003645 can mitigate inflammation and apoptosis by suppressing the NF-κB signaling pathway.Citation45 Moreover, in TNF-α-induced HUVECs, the cordycepin derivative IMM-H007—an activator of AMP-Activated Protein Kinase (AMPK)—can inhibit the inflammatory response by modulating NF-κB and JNK/AP1 signaling pathways.Citation46 In Ox-LDL-induced HUVECs, Kruppel-like factor 2 (KLF2) knockdown also abrogates the activation of the AMPK/SIRT1 signaling pathway elicited by protein tyrosine phosphatase 1B (PTP1B) knockdown, which can reduce inflammatory directional injury and dysfunction, thus ameliorating AS.Citation47 In Ox-LDL-induced human aortic endothelial cells (HAECs) models, overexpressed microRNA-20a attenuates the inflammatory response by inhibiting TLR4 and TXNIP signaling pathways, emerging as a potential therapeutic target for anti-AS development.Citation48 Some researchers have discovered that in Ox-LDL-induced MAECs, overexpressed C1q/tumor necrosis factor-related protein-3 (CTRP3) inhibits the inflammatory response and endothelial dysfunction by activating the PI3K/AKT/eNOS signaling pathway, suggesting that this may be an effective anti-AS strategy.Citation49 Additionally, in TNF-α-induced HUVECs, apolipoprotein M and sphingosine-1-phosphate (ApoM-S1P) activate the PI3K/AKT signaling pathway by binding to S1PR2, thereby reducing EC injury, inflammatory response, and pyrosis.Citation50 In the Ox-LDL-induced HUVECs model, overexpressed Vestigial-like 4 (VGLL4) ameliorates apoptosis, oxidative stress, inflammation, and EC dysfunction by activating the Hippo-YAP/TEAD1 signaling pathway.Citation51 Similarly, in Ox-LDL-treated HUVECs, cytoplasmic polyadenylation element binding protein 1 (CPEB1) deletion may suppress oxidative stress, inflammatory response, and apoptosis by modulating the SIRT1/LOX-1 signaling pathway.Citation52 It has also been demonstrated that in Ox-LDL-induced HUVECs, the biomarker Galectin-3 promotes endothelial dysfunction through the LOX-1-mediated LOX-1/ROS/p38/NF-κB signaling pathway, exacerbating AS.Citation53 In the AS rat model, overexpression of MiR-181b could alleviate inflammation and protect vascular endothelial function by inhibiting the Notch1 signaling pathway.Citation54 Another researcher found that in a mouse model of coronary AS, upregulated microRNA-107 (MiR107) activated the Notch signaling pathway by suppressing KRT1, thereby inhibiting the inflammatory response and endoplasmic reticulum stress of vascular ECs.Citation55 In the high-fat diet (HFD)-fed rabbit AS model, myosin light chain kinase inhibitor 7 (ML7) improves vascular endothelial dysfunction and permeability through the mitogen-activated protein kinase (MAPK) signaling pathway.Citation56 In the aging model of HUVECs, overexpressed MicroRNA-216a, acting as an endogenous inhibitor of the Smad3/IκBα pathway, accelerates the aging and inflammatory response of ECs, emerging as a potential target for aging-related AS.Citation57 Studies have shown that in LPS-induced rats and ECs models, the JAK/STAT pathway could inhibit the increase of endothelial adenosine deaminase (eADA) activity, attenuate the activation and inflammation of ECs, and thereby improve AS.Citation58 Collectively, these findings suggest that overexpression of key molecules can reduce endothelial inflammation in IL-1β, TNF-α, Ox-LDL, or LPS-induced cell and animal models.

The following sections discuss the fundamental research on small molecules and drugs in AS models. Studies have demonstrated that in HUVECs and ApoE−/− mice models, exosomes derived from mature dendritic cells can exacerbate AS by increasing endothelial inflammation through the membrane TNF-α-mediated NF-κB signaling pathway.Citation59 In IL-1β-induced HUVECs and LPS-induced acute inflammatory mice models, chrysin mitigates vascular EC inflammation by suppressing the NF-κB signaling pathway, potentially emerging as a promising drug candidate for the treatment of inflammatory vascular diseases, such as AS.Citation60

Likewise, in IL-1β-induced HUVECs and LPS-induced acute inflammatory mouse models, neferine attenuates inflammatory injury by inhibiting the NF-κB signaling pathway, making it a promising candidate for AS treatment.Citation61 In Ox-LDL-induced HUVECs, triptolide counteracts EC inflammation by impeding the activation of the oxidative stress-dependent NF-κB pathway, contributing to AS prevention.Citation62 In LPS-induced HUVECs, the anti-inflammatory effect of Lactococcus lactis-fermented spinach juice is mediated through the inhibition of the NF-κB signaling pathway, providing a potential treatment for AS.Citation63

Moreover, in TNF-α-induced HUVECs and LPS-induced C57BL/6 mice models, the peptide lycosin-I ameliorates the inflammatory response by modulating the IκB/NF-κB signaling pathway, potentially emerging as a new drug candidate for treating inflammatory diseases.Citation64 In LPS-induced HUVECs, hyperoside hinders EC inflammation and apoptosis by suppressing the activation of the TLR4/NF-κB signaling pathway, potentially reducing the risk of AS.Citation65 Additionally, in LPS-induced human coronary endothelial cells (HCAECs) models, ficus deltoidea (FD) obstructs EC activation, inflammation, monocyte adhesion, and oxidative stress via NF-κB and eNOS pathways, thereby exerting anti-AS effects.Citation66

Previous research indicates that in homocysteine-induced HAECs, catalpol inhibits reactive oxygen species (ROS) production, oxidative stress, endoplasmic reticulum stress, inflammation, and apoptosis by suppressing the Nox4/NF-κB and GRP78/PERK pathways, potentially providing a therapeutic approach for AS prevention and treatment.Citation67 Similarly, in Ox-LDL-induced human vascular smooth muscle cells (hVSMCs) and HUVECs, myristicin inhibits cell proliferation, apoptosis, and inflammatory cytokine expression by modulating the PI3K/AKT/NF-κB signaling pathway, consequently suppressing AS development.Citation68

Furthermore, in LPS-induced human microvascular endothelial cells-1 (HMEC-1), hypaphorine curbs inflammatory responses by regulating TLR4 and PPAR-γ, which rely on the PI3K/AKT/mTOR signaling pathway, potentially serving as a therapeutic agent for endothelial inflammatory diseases, such as AS.Citation69 In hyperhomocysteinemia-induced HUVECs and mice models, picroside II may decrease EC injury in AS by inhibiting oxidative stress, inflammatory responses, and apoptosis through regulation of the SIRT1/LOX1 signaling pathway.Citation70 In LPS or Ox-LDL-induced cell models, active polypeptides from Hirudo may prevent AS onset by modulating the LOX-1/LXR-α/ABCA1 signaling pathway, inhibiting THP-1 cell adhesion to HUVECs, reducing the inflammatory response, and suppressing ROS production and apoptosis in RAW264.7 cells.Citation34 In Ox-LDL-induced HUVECs, naringin mitigates apoptosis and inflammatory responses by inhibiting the Hippo-YAP signaling pathway, thereby decreasing the formation and progression of AS plaques.Citation71

Previous findings suggest that in rat models of cigarette smoke extract (CSE)-induced HAECs and carotid artery injury exposed to cigarette smoke, melatonin reduces ROS generation and cell pyroptosis via the Nrf2/ROS/NLRP3 signaling pathway, preventing smoking-induced vascular injury and AS.Citation72 Additionally, in Ox-LDL-induced HUVECs and ApoE−/− mice, dihydrohomoplantagin and homoplantaginin minimize EC injury, ROS overproduction, and apoptosis by activating the Nrf2 antioxidative signaling pathway, thereby controlling AS development.Citation73

In the in vitro experimental HUVECs model, the choline-derived metabolite trimethylamine N-oxide (TMAO) induces oxidative stress and activates the ROS-TXNIP-NLRP3 inflammasome signaling pathway, resulting in EC inflammation and endothelial dysfunction, thereby increasing AS risk.Citation74 Intriguingly, in Ox-LDL-induced HUVECs, rapamycin abates the inflammatory response by suppressing the mTORC2/PKC/c-Fos pathway, thus exerting an anti-AS function.Citation75 Researchers have discovered that in Ox-LDL-induced HUVECs, nintedanib (a multityrosine kinase receptor inhibitor) downregulates arginase II by inhibiting the p53/p21 signaling pathway, improving endothelial inflammation, oxidative stress, and cellular senescence, potentially serving as a therapeutic agent for AS.Citation76

In HUVECs and Tlr4mut mice models, disordered blood flow locally activates the TLR4 signaling pathway in ECs by upregulating fibronectin containing the extra domain A in the subendothelial extracellular matrix, leading to endothelial inflammation and AS onset.Citation77 Significantly increased expression of NDRG1 was found in cytokine-stimulated ECs as well as in human and mouse models of AS, and the findings suggest that NDRG1 is a key signal influencing endothelial inflammation and vascular remodeling, and that inhibiting NDRG1 may be a potential clinical therapeutic target for the treatment of inflammatory vascular diseases such as AS.Citation78 In an AS model of HFD-fed ApoE −/− mice, a significant increase in plaque formation was observed in the model group; in the LPS-treated HUVECs and RAW264.7 inflammation models, isorhynchophylline reduced LPS-induced inflammatory responses through inhibition of the NF-κB/NLRP3 pathway and promoted the cell migration ability.Citation79

From the aforementioned findings, it is evident that AS frequently develops in medium and large arteries composed of ECs, vascular smooth muscle cells (VSMCs), and other vascular cells.Citation80 These vascular ECs in the table may serve as a valuable model for investigating the molecular mechanisms of vascular diseases (). AS is a chronic inflammatory vascular disease driven by both traditional and non-traditional risk factors.Citation31,Citation81 Several inflammation-related signaling pathways are involved in the regulation of AS pathogenesis, including the NF-κB signaling pathway, Toll-like receptor signaling pathway, and PI3K/AKT signaling pathway. These pathways hold significant implications for the progression of AS. Targeting inflammation-related signaling pathways may present a novel and effective approach for treating AS.Citation31 Consequently, we have summarized the cell models, animal models, and associated signaling pathways related to AS and introduced corresponding research findings. These studies collectively highlight the impact of EC inflammation on the onset and progression of AS.

Table 1 Endothelial Inflammation in Atherosclerosis

Cardiovascular Diseases

Cardiovascular disease (CVD) encompasses a range of conditions affecting blood vessels and the heart, including coronary heart disease (CHD), stroke, and peripheral vascular disease.Citation82,Citation83 CVD is a leading cause of mortality in numerous countries and a common endpoint for various chronic diseases.Citation84 Multiple potential causes of CVD have been identified, with inflammation being one of them.Citation85 Clinical and epidemiological studies have established a close link between EC function and CVD risk, revealing that the onset, progression, and alleviation of CVD are intimately associated with the inflammatory response.Citation43,Citation86 Chronic inflammation plays a pivotal role in the pathogenesis of CVD during its pathological process.Citation11 Consequently, mitigating endothelial inflammation has emerged as a crucial strategy in treating CVD. A thorough investigation of the connection between inflammation and disease could offer novel insights and approaches for the prevention and treatment of such conditions. In this section, we will discuss the significance of inflammation in the initiation and progression of CVD, as well as basic research methodologies aimed at drug-targeted ECs to enhance vascular function.Citation85

According to the latest research, arterial dysfunction, such as impaired endothelial function like reduced endothelium-dependent dilation (EDD), and large artery stiffening, are key factors in the development of CVD and tend to worsen with age. These are mainly mediated by an overproduction of ROS and an increase in chronic, low-grade inflammation.Citation87

In a clinical study, regular aerobic exercise has demonstrated its ability to inhibit oxidative stress and lower inflammatory marker levels, which may effectively improve endothelial dysfunction and large elastic artery stiffening. As a result, this intervention serves as a promising measure for preventing and treating CVD and promoting cardiovascular health.Citation88 Interestingly, multiple pilot trials have indicated that Inspiratory Muscle Strength Training (IMST), a high-resistance inspiratory muscle training, can reduce chronic low-grade inflammation and improve cardiovascular function, enhancing compliance among middle-aged and older individuals. This approach addresses the limitations of only a small proportion of adults meeting the aerobic exercise guidelines.Citation89,Citation90 In addition, in vehicle-treated animals, observations suggest that oral administration of apigenin reverses vascular endothelial dysfunction and large elastic artery stiffening and prevents foam cell formation in an established cell culture model of early AS. These preclinical research findings offer valuable insights into the inhibition of age-related intrinsic mechanical wall stiffening in the aorta and vascular inflammation. They lay the groundwork for future translational studies assessing the potential of apigenin therapy in treating arterial dysfunction and reducing the risk of CVD.Citation91 It is noteworthy that in a randomized, double-blind, placebo-controlled, single-point parallel group clinical trial, age-related increases in large elastic artery stiffening and systolic blood pressure were found to be associated with oxidative stress, inflammation, and increased vascular smooth muscle tension, leading to the development of CVD. However, the researchers discovered that supplementing with nicotinamide riboside could alleviate the rise in systolic blood pressure and arterial stiffness in middle-aged and older individuals, thereby improving cardiovascular health.Citation92

Endothelium-dependent dilation is a prerequisite for CVD. Recently, in a double-blind placebo-controlled study, it was confirmed that inhibiting acute systemic inflammation can improve endothelium-dependent dilation in women with a history of preeclampsia during pregnancy. This finding, based on a cutaneous microcirculation model, suggests that the vascular dysfunction observed during preeclamptic pregnancies may increase the lifelong risk of CVD in these women.Citation93 Secondly, in an epidemiologic and observational study, it was shown that acute systemic inflammation impairs endothelium-dependent dilatation of human veins in humans, and in this study, it was found that there is a close relationship between infections or inflammation, which is usually considered to be associated with CVD.Citation94 Notably, in a small open-label study, experts found that short-term treatment with the NF-κB inhibitor salicylate improved nitric oxide (NO)-mediated endothelium-dependent dilatation of the microvasculature in young adults with major depression. Besides, there is substantial evidence that adult major depression is associated with a substantially increased risk of future CVD development. Thus, the development of new therapeutic interventions to prevent or slow the progression of CVD has strong relevance.Citation95 In addition, two key findings need to be noted in a study. First, in a rat model, inhalation of multi-walled carbon nanotubes induced lung inflammation. Second, the inhalation of multi-walled carbon nanotubes resulted in profound changes in endothelium-dependent dilation of coronary arteries. Taken together, these results are the first report of coronary microvascular dysfunction after multiwall carbon nanotubes.Citation96 Scientists have also determined in recent years that the effects of pulmonary exposure to particulate matter on endothelium-dependent dilation of systemic microvascular are dependent on pulmonary and/or microvascular inflammation, and that, these systemic inflammations associated with particulate matter exposure have been considered to be linked to impaired cardiovascular function in affected individuals.Citation97

In acute arterial wall shear stress-induced saphenous vein ECs, the inflammatory response can be diminished by inhibiting the activation of the NF-κB signaling pathway.Citation98 Similarly, in TNF-α-induced HAECs, zafirlukast (a cysteinyl leukotriene receptor type 1 (CysLT1R) antagonist) reduces inflammatory injury and ROS by suppressing the NF-κB signaling pathway, potentially serving as a novel therapeutic agent for CVD.Citation99 Likewise, in angiotensin II–induced C57BL/6 mice and HUVECs, schizandrin B mitigates endothelial to mesenchymal transition, oxidative stress, and inflammation by inhibiting the NF-κB signaling pathway, thereby attenuating vascular remodeling and potentially reducing the progression of CAD.Citation100 In TNF-α-induced HCAECs, epigallocatechin gallate diminishes the inflammatory response by suppressing the NF-κB signaling pathway, potentially providing a treatment for CAD.Citation101 Interestingly, in a model of hypoxia-reoxygenation-induced human cardiac microvascular EC injury, overexpressed MicroRNA-106b inhibited B-cell linker (BLNK) by repressing the NF-κB signaling pathway, thereby reducing inflammatory injury in cardiac ECs.Citation102 Additionally, in a rat model of ischemia-reperfusion-induced SD, Vitamin D hinders the inflammatory response by inhibiting the RhoA/ROCK/NF-ĸB signaling pathway, thus decreasing myocardial ischemia-reperfusion injury.Citation103 Intriguingly, in HUVECs subjected to simulated microgravity, endoplasmic reticulum stress activates the iNOS/NO-NF-κB and NLRP3 inflammasome signaling pathways, promoting EC inflammation and apoptosis, ultimately leading to cardiovascular dysfunction.Citation104 In contrast, under endosulfan (a fat-soluble insecticide)-induced HUVECs, oxidative stress and endoplasmic reticulum stress are reduced by suppressing the IRE1α/NF-κB signaling pathway, thereby inhibiting EC inflammation and endothelial dysfunction, potentially decreasing the incidence of CVD.Citation105 Furthermore, in Ox-LDL-induced HUVECs and HASMCs, orexin A ameliorates endothelial inflammation by inhibiting THP-1 cell adhesion to ECs through the suppression of MAPK p38 and NF-κB signaling pathways.Citation106 Interestingly, in hexavalent chromium-induced THP-1 cells and HUVECs, taxifolin reduces oxidative stress and apoptosis by inhibiting NF-κB and p38 MAPK signaling pathways, thereby preventing CAD.Citation107 In palmitic acid (PA)-induced HUVECs, adiponectin decreases ROS, endothelial inflammation, and IR by modulating the ROS/IKKβ signaling pathway, providing new insights into the mechanism of cardiovascular protective action.Citation108 In a mouse model of heart failure with preserved ejection fraction (HFpEF), QiShenYiQi mitigates HFpEF by inhibiting microvascular endothelial inflammation and activating the NO-cGMP-PKG signaling pathway.Citation109 Additionally, in ApoE−/− mice fed HFD, the cordycepin derivative IMM-H007, an activator of AMP-Activated Protein Kinase (AMPK), suppresses vascular inflammation and improves endothelial dysfunction by regulating the AMPK-PI3K-AKT-eNOS signaling pathway, contributing to cardiovascular protection.Citation110 Moreover, in Ox-LDL-induced HUVECs, ginsenoside Rg1 attenuates apoptosis, senescence, and oxidative stress by regulating the AMPK/SIRT3/p53 signaling pathway, thus laying the groundwork for the treatment of CHD.Citation111 In leptin-induced HUVECs, 1.25-dihydroxycholecalciferol ([1,25 (OH)2D3]) reduces oxidative stress and inflammation by activating the Nrf2-antioxidant signaling pathway, thereby decreasing obesity and the risk of CVD and other health problems.Citation112 Some researchers have observed that in PA-induced SD rats, methotrexate can improve endothelial dysfunction by activating the AMPK/eNOS pathway to inhibit the inflammatory response in perivascular adipose tissue, thereby providing pharmacological evidence for the treatment of CVD.Citation113

The significance of vascular endothelial function in the initiation and progression of CVD cannot be understated.Citation114 Inflammation within the vascular endothelium serves as a primary contributor to CVD development.Citation115 To gain deeper insights into the amelioration of CVD through inflammation reduction, this section outlines various vascular ECs models and animal models employed to investigate the pathogenesis of CVD. We also present an overview of molecular mechanisms targeted for CVD treatment, encompassing the RhoA/ROCK/NF-ĸB signaling pathway, iNOS/NO-NF-κB/IκB signaling pathway, NLRP3 inflammasome signaling pathway, IRE1α/NF-κB signaling pathway, and the MAPK p38 signaling pathway ().

Table 2 Endothelial Inflammation in Cardiovascular Diseases

Diabetes Mellitus and Its Complications

Diabetes mellitus (DM) is a metabolic disorder characterized by persistent hyperglycemia resulting from inadequate insulin action, insufficient insulin secretion, or a combination of both.Citation116 Vascular complications associated with DM are the primary drivers of morbidity and mortality in affected individuals.Citation117 Hyperglycemia initially impairs blood vessels, with ECs considered the primary targets of hyperglycemic injury. Endothelial damage and inflammation play crucial roles in the pathogenesis of type 2 diabetes mellitus (T2DM) and its vascular complications.Citation32,Citation117,Citation118 Recent evidence also indicates that vascular EC dysfunction is present during the pre-DM stage, contributing significantly to the development and progression of both macrovascular and microangiopathic complications.Citation119 Chronic inflammation is a prevalent feature of T2DM.Citation120 Besides, the connection between inflammation and DM has sparked interest in targeting inflammation as a means to improve DM and its related complications.Citation121 This section provides an overview of recent treatments and preventive measures, which target different signaling pathways to help reduce inflammation and manage diabetes.

The following research highlights advancements in mitigating inflammatory responses in DM by modulating key molecular players. In high glucose (HG)-induced HUVECs, silencing long noncoding RNA MALAT1 has been shown to alleviate apoptosis and inflammatory responses by inhibiting the NF-κB signaling pathway.Citation122 Similarly, in HG-induced human retinal endothelial cells (HRCECs) and streptozotocin-induced diabetic retinopathy model mice, depletion of SOX4, a transcription factor expressed in the pancreas, inhibited endothelial inflammatory responses, migration, and angiogenesis via the NF-κB signaling pathway.Citation123 Studies have demonstrated that in HG-induced HUVECs, overexpression of cystic fibrosis transmembrane conductance regulator (CFTR) reduces ROS, oxidative stress, and inflammatory response by regulating NF-κB and MAPK signaling pathways, thereby mitigating diabetic CAD.Citation124 In HG-induced HUVECs, downregulation of hsa_circ_0068087 could inhibit endothelial dysfunction and inflammatory response by suppressing the TLR4/NF-κB/NLRP3 inflammasome signaling pathway.Citation125 In a human retinal endothelial cells model, soluble gp-130 fused chimera (sgp130-Fc) attenuated endothelial inflammation, apoptosis, and endothelial barrier disruption through inhibition of the IL-6 trans-signaling pathway, thereby reducing diabetic retinopathy.Citation126 In HG-induced HUVECs, overexpressed malignant fibrous histiocytoma amplified sequence 1 (MFHAS1) inhibits the inflammatory response by activating the AKT/HO-1 signaling pathway, thus controlling the development of diabetes.Citation127 In diabetic db/db mice and HUVECs models, overexpressed transcription factor EB (TFEB) attenuated vascular endothelial inflammation by inhibiting the IKK (IκB kinase)-p65 signaling pathway.Citation128 In HG-induced HUVECs, overexpression of microRNA-9-5p reduced apoptosis and inflammatory responses by inhibiting CXC chemokine receptor-4 (CXCR4) to suppress the mitogen-activated protein kinase (MAPK)/ERK and PI3K/AKT/mTOR signaling pathways.Citation129

The following studies showcase the research advancements involving small molecule compounds and drugs in diabetes models. In HG-induced HUVECs, ketamine has been shown to inhibit EC inflammation by attenuating ROS production, reducing phosphorylation of PKC βII Ser660, and deactivating PKC and NF-κB.Citation130 Interestingly, in db/db mice and HG and palmitate-induced MAECs, circulating metabolites of strawberries may improve vascular inflammation and endothelial dysfunction by inhibiting the NF-κB signaling pathway, thereby preventing diabetes-related vascular complications.Citation131 In advanced glycation end products (AGEs)-induced HUVECs, salidroside reduces EC inflammation and oxidative stress by modulating the AMPK/NF-κB/NLRP3 signaling pathway.Citation132 Among palmitate-induced HUVECs and SD rats, the ethyl acetate extract of C. chinense (CCE) reduces endothelial inflammation and IR by inhibiting TLR4-mediated NF-κB and MAPK signaling pathways, suggesting its potential as a therapeutic agent for treating IR and DM-related endothelial dysfunction.Citation133 In mouse models of diabetic nephropathy and AGEs-induced mouse glomerular endothelial cells (mGECs) injury, catalpol improved endothelial dysfunction and inflammatory response by inhibiting the RAGE/RhoA/ROCK signaling pathway, thereby ameliorating the pathological injury of diabetic kidneys.Citation134 In HG-induced HUVECs, sodium hydrosulfide (NaHS) reduces endothelial injury and inflammation by inhibiting the p38 MAPK signaling pathway, thus treating vascular complications of DM.Citation135 In palmitate and insulin-induced HUVECs and HFD fed mice models, spinosin improves IR and reduces ROS production and inflammation by modulating the PI3K/AKT/eNOS signaling pathway.Citation136 Albiflorin exerts a potential therapeutic effect on diabetes vascular complications by inhibiting HG-induced apoptosis and inflammatory response in HUVECs through the suppression of the PARP1/ NF-κB signaling pathway.Citation137

In light of these studies, our understanding of the inflammatory mechanisms associated with DM and its vascular complications has significantly advanced (). This section offers a comprehensive overview of the methodologies, experimental progress, and related pathways involved in DM treatment. Collectively, these studies underscore the crucial role of inflammation in ECs during the pathological process of DM and provide valuable insights for future research aimed at treating DM through anti-inflammatory approaches.

Table 3 Endothelial Inflammation in Diabetes Mellitus

Sepsis

Sepsis is a life-threatening systemic inflammatory response syndrome resulting from infection.Citation138 It is characterized by organ dysfunction stemming from the host’s dysregulated response to infection.Citation139 However, the host’s inflammatory response also contributes to the pathological and physiological changes observed in sepsis.Citation140,Citation141 At least 19 million people globally are affected by this condition annually.Citation142,Citation143 Research into the pathogenesis of sepsis has revealed that both inflammation and anti-inflammatory responses are triggered during the early stages of infection. Inflammatory mediators can also induce infection, leading to abnormal EC activation, endothelial barrier injury, and an imbalance between pro-inflammatory and anti-inflammatory responses. This imbalance results in an inflammatory cell storm that causes extensive cellular damage.Citation143,Citation144 Persistent and repeated inflammatory damage has been implicated in EC injury during sepsis.Citation145 ECs typically serve as crucial physical barriers that maintain vascular intima integrity. However, in sepsis, the release of substantial amounts of endotoxins and inflammatory cytokines directly contributes to vascular EC injury.Citation146

Numerous sepsis models are induced by LPS. For instance, in LPS-induced HUVECs, upregulation of hnRNPA2/B1 has been shown to inhibit EC injury by suppressing NF-κB and VE-cadherin/β-catenin signaling pathways.Citation147 Likewise, in LPS-induced endotoxemia animal models and mouse lung microvascular endothelial cells, endothelial calpain knockout mitigates acute kidney injury and apoptosis in mice by inhibiting the p38-iNOS signaling pathway.Citation148 Moreover, in LPS-induced endotoxemia mouse models, HPMECs, and HUVECs, yes-associated protein (YAP, a transcriptional coactivator) attenuates the inflammatory response by blocking tumor necrosis factor receptor-associated factor 6 (TRAF6)-mediated NF-кB activation.Citation149 Furthermore, in LPS-induced lung epithelial cells, HUVECs, and sepsis mice, downregulation of programmed death ligand 1 (PD-L1) reduces the inflammatory response and apoptosis by inhibiting the HIF-1α signaling pathway.Citation150 Intriguingly, researchers discovered in an LPS-induced sepsis mouse model that rigosertib improved the inflammatory response by inhibiting the MEK1-ERK signaling pathway.Citation151 In LPS-treated HUVECs and a mouse cecum ligation (CLP)-induced sepsis model, findings suggest that the upregulation of PCSK9 activates the TLR4/MyD88/NF-кB and NLRP3 pathways, inducing inflammation and resulting in endothelial dysfunction. Therefore, inhibiting PCSK9 may represent a novel strategy to improve vascular endothelial function in sepsis.Citation152

This section presents an overview of ECs’ role in sepsis development (). ECs undergo morphological and functional changes during infection or tissue damage, a process known as EC activation.Citation3 Activated ECs produce various pro-inflammatory cytokines. The studies mentioned above suggest that blocking inflammatory signaling pathways can diminish the inflammatory response in sepsis. Experiments have also demonstrated that LPS-induced endothelial inflammation is a well-established cell model of sepsis. Targeting hnRNPA2/B1, endothelial calpain, yes-associated protein (YAP), programmed cell death receptor (PD)-L1, PCSK9 and rigosertib may be effective in inhibiting the inflammatory response and, consequently, reducing sepsis. This section holds significant value for gaining a deeper understanding of vascular EC inflammation in sepsis treatment.

Table 4 Endothelial Inflammation in Sepsis

Respiratory Diseases

Respiratory diseases (RD), particularly non-communicable chronic inflammatory diseases of the airways, are among the leading causes of mortality worldwide.Citation153 Persistent inflammation in the respiratory tract underlies all respiratory diseases and is the primary characteristic of all chronic respiratory diseases.Citation154,Citation155 This section reviews RDs associated with endothelial inflammation, including asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), and acute lung injury (ALI). Asthma is a chronic RD characterized by airway inflammation with clinical manifestations such as dyspnea, wheezing, chest tightness, and cough.Citation156–158 COPD, characterized by irreversible airflow restriction, is an inflammatory disease affecting both the airway and lung tissue, particularly associated with an abnormal inflammatory response to cigarette smoke.Citation159 ALI and ARDS represent two different stages of a disease caused by direct lung injury and indirect systemic inflammatory reactions.Citation160

Previous research results have shown that in ovalbumin induced asthma mouse models and in vitro cell models, the deletion of ECs Sox17 (endothelium-specific transcription factor) could inhibit the adhesion of IL-33 stimulated THP-1 cells to HUVECs and HPMECs by inhibiting ERK and STAT3 signaling pathways, as well as allergic airway inflammation in mice.Citation161 Furthermore, in cigarette smoke extract-induced HUVECs, the antioxidant mitoquinone (MitoQ) reduces ROS, autophagy, endothelial barrier damage, and inflammation by inhibiting NF-κB and NLRP3 inflammasome signaling pathways, thereby mitigating COPD.Citation162 Studies have demonstrated that in LPS-induced HPMECs, inhibition of microRNA-92a targets integrin α5 (ITGA5) through the PI3K/AKT signaling pathway, reducing endothelial barrier dysfunction and thus improving ALI/ARDS.Citation163 Similarly, in LPS-induced ARDS mouse models and EA.hy 926 HUVECs, ghrelin inhibits EC damage and apoptosis by regulating the PI3K/AKT signaling pathway.Citation164 In LPS-induced HPMECs, ripaudil, a novel ROCK2 inhibitor, suppresses apoptosis and inflammatory response by regulating the ROCK2/eNOS signaling pathway, making it a potential drug for the clinical treatment of ALI.Citation165 Likewise, in LPS-induced acute lung injury mouse models and alveolar epithelial cells, simvastatin inhibits apoptosis by upregulating the Survivin/NF-κB/p65 signaling pathway.Citation166 In LPS-induced rat pulmonary microvascular endothelial cells (PMVECs) and a rat model of ARDS, overexpression of Sema3A can inhibit the ERK/JNK signaling pathway, thereby improving ECs apoptosis and angiogenesis in the ARDS model, ultimately reducing lung injury and inflammation in rats.Citation167

In conclusion, this section highlights the unique role of EC inflammation in the pathogenesis of RD. Additionally, we summarize the key signaling pathways that initiate different mechanisms and propagate the inflammatory response, including NF-κB and NLRP3 inflammasome signaling pathways, ERK and STAT3 signaling pathways, PI3K/AKT signaling pathways, Survivin/NF-κB/p65 signaling pathways, ERK/JNK signaling pathway and ROCK2/eNOS signaling pathways. Research advances in RD and endothelial inflammation may serve as a reference for the development of new drugs to prevent or treat these RDs ().

Table 5 Endothelial Inflammation in Respiratory Diseases

Clinical Researches

A recent clinical study revealed that an aqueous extract of Terminalia chebula considerably diminished endothelial dysfunction and oxidative stress, thereby reducing CVD risk in patients with type 2 diabetes mellitus (T2DM).Citation168 In a randomized, double-blind, placebo-controlled clinical investigation, a standardized aqueous extract of Phyllanthus emblica fruits significantly improved endothelial dysfunction, oxidative stress, inflammation, and lipid profiles in individuals with metabolic syndrome.Citation169 According to a randomized, double-blind clinical trial, moderate supplementation with docosahexaenoic acid (DHA)-rich fish oil significantly enhanced PPARγ activity in patients with T2DM, potentially mitigating cardiovascular complications in DM patients.Citation170 Likewise, a double-blind randomized controlled trial demonstrated that omega-3 fatty acids could improve vascular inflammation and decrease AS.Citation171 Intriguingly, a pilot study found that oral supplementation with L. plantarum 299v (Lp299v) improved vascular endothelial function and reduced systemic inflammation in men with CAD.Citation172 Small clinical studies have discovered that the anti-inflammatory properties of Tongmai Yangxin pill (TMYX) could enhance patients’ serum biochemical markers, subsequently reducing the risk of coronary heart disease (CHD).Citation173 Following a randomized, double-blind, parallel, placebo-controlled trial involving 100 hemodialysis patients, the combined administration of pomegranate peel extract (PPE) and vitamin E (Vit E) was found to mitigate endothelial inflammation and bolstering vascular endothelial function, thus preventing CVD development.Citation174 In a single-blind, two-group, prospective randomized controlled trial for cardiac rehabilitation with 120 eligible participants (70 men and 50 women) suffering from chronic heart failure, group-based high-intensity aerobic interval training substantially improved the inflammatory status.Citation175 In a clinical trial including 46 patients with stable CAD and chronic obstructive pulmonary disease (COPD), ticagrelor alleviated symptoms by reducing systemic inflammation and oxidative stress.Citation176 Lastly, a double-blind, placebo-controlled, randomized clinical trial showed that nano-curcumin (NC) supplementation for patients with severe sepsis diminished the inflammatory response and protected endothelial function.Citation177

EC injury is a crucial factor contributing to the inflammatory response, making the protection of vascular endothelial function essential for preventing and treating inflammatory vascular diseases. Consequently, the investigation of medications to maintain vascular health has become pivotal in managing vascular inflammatory diseases ().Citation178 Among the drugs currently in clinical use, statin lipid-lowering medications, herbal formulations, traditional Chinese medicines, fruit and plant extracts, and antiplatelet agents have demonstrated effectiveness in improving vascular inflammation.Citation26 These medications exert protective effects on vascular endothelial function through indirect mechanisms.Citation179 Furthermore, adopting a healthy diet and engaging in regular physical exercise can also contribute to preserving endothelial function.

Table 6 Clinical Study of Endothelial Inflammation-Related Diseases

Discussion and Summary

Preserving the integrity of ECs is vital for maintaining human health and preventing diseases. ECs serve as the natural lining of blood vessels, regulating vascular and organ integrity, and playing a critical role in the inflammatory response.Citation180 Under physiological conditions, ECs prevent the infiltration of inflammatory cells into tissues by regulating vascular tension and controlling hemorrhage and thrombosis.Citation3,Citation16,Citation22 Endothelial inflammation is a key initiating event under pathological conditions and an early indicator of disease.Citation4 Comprehending the various functions of ECs and elucidating inflammatory regulatory mechanisms can provide insight into disease progression and enhance treatment outcomes.

In summary, we have confirmed that ECs are key targets and crucial components of the inflammatory process.Citation6 Studies suggest that these cells, situated in areas susceptible to vascular endothelial lesions, may be an ideal model for examining the molecular mechanisms of vascular-related diseases ( and ). This review also summarizes the inducers of inflammation and the therapeutic agents that inhibit inflammation in these cell types. Concurrently, recent studies have shed light on the key signaling pathways that regulate pro-inflammatory and anti-inflammatory responses in ECs ().

Table 7 Directions for Mechanistic Studies in Different Cell Models

Figure 1 Relationship between endothelial cells inflammation models and related diseases.

Figure 1 Relationship between endothelial cells inflammation models and related diseases.

Figure 2 Signaling pathways involved in the regulation of endothelial cells inflammation.

Figure 2 Signaling pathways involved in the regulation of endothelial cells inflammation.

Despite the comprehensive analysis of the relationship between endothelial inflammation and vascular-related diseases, this study presents certain limitations. Relatively small sample sizes in clinical trials may cause fluctuations in research findings. In clinical practice, numerous anti-inflammatory treatments are available, such as the routine use of antibiotics. However, these therapeutic strategies can only inhibit or eliminate pathogenic microorganisms without reversing the changes in EC function. To thoroughly alleviate inflammation’s secondary effects on blood vessels and organs, we propose an approach that combines anti-inflammatory therapy with the restoration of EC function, offering new insights for treating vascular inflammation.

In conclusion, this article not only underscores the significance of endothelial inflammation in vascular-related diseases but also highlights how understanding the molecular interactions and pathways regulating the inflammatory response can improve therapeutic strategies and promote drug development ( and ). Nevertheless, individuals should be aware that adopting a healthy and reasonable lifestyle (eg, quitting smoking, losing weight, increasing physical activity) can significantly reduce the occurrence of various risk factors. It is hoped that this review will inspire new perspectives and lay a theoretical foundation for clinical research.

Table 8 Research Gaps and Future Directions in Endothelial Cells Inflammation in Vascular-Related Diseases

Figure 3 Inflammation- Vascular- Disease- Pathological States.

Figure 3 Inflammation- Vascular- Disease- Pathological States.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare no conflicts of interest in this work.

Acknowledgments

Thanks to the support of Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine.

Additional information

Funding

This study was supported by the Natural Science Foundation of Jilin Provincial Department of Science and Technology (No. 20190201146JC), Jilin Province Science and Technology Project of Traditional Chinese Medicine (No. 2021059 and No. 2021014), the “Ju jing Cup” academic research innovation project of Changchun University of Traditional Chinese Medicine (No. YK202102) and the Science and Technology Development Plan Project of Jilin Province, China (Grant No: YDZJ202301ZYTS153).

References

  • Xu S, Jin T, Weng J. Endothelial cells as a key cell type for innate immunity: a focused review on RIG-I signaling pathway. Front Immunol. 2022;13:951614. doi:10.3389/fimmu.2022.951614
  • Xu S, Ilyas I, Little PJ, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924–967. doi:10.1124/pharmrev.120.000096
  • Theofilis P, Sagris M, Oikonomou E, et al. Inflammatory mechanisms contributing to endothelial dysfunction. Biomedicines. 2021;9(7):781. doi:10.3390/biomedicines9070781
  • Michiels C. Endothelial cell functions. Cell Physiol. 2003;196(3):430–443. doi:10.1002/jcp.10333
  • Coggins M, Rosenzweig A. The fire within: cardiac inflammatory signaling in health and disease. Circ Res. 2012;110(1):116–125. doi:10.1161/CIRCRESAHA.111.243196
  • Xiao L, Liu Y, Wang N. New paradigms in inflammatory signaling in vascular endothelial cells. Am J Physiol Heart Circ Physiol. 2014;306(3):H317–H325. doi:10.1152/ajpheart.00182.2013
  • Hellenthal KEM, Brabenec L, Wagner NM. Regulation and dysregulation of endothelial permeability during systemic inflammation. Cells. 2022;11(12):1935. doi:10.3390/cells11121935
  • Tu Z, Zhong Y, Hu H, et al. Design of therapeutic biomaterials to control inflammation. Nat Rev Mater. 2022;7(7):557–574. doi:10.1038/s41578-022-00426-z
  • Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med. 2019;25(12):1822–1832. doi:10.1038/s41591-019-0675-0
  • Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov. 2016;15(2):125–142. doi:10.1038/nrd.2015.2
  • Pop RM, Popolo A, Trifa AP, Stanciu LA. Phytochemicals in cardiovascular and respiratory diseases: evidence in oxidative stress and inflammation. Oxid Med Cell Longev. 2018;2018:1603872. doi:10.1155/2018/1603872
  • Navarro-Gonzalez JF, Mora-Fernandez C, Muros de Fuentes M, Garcia-Perez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7(6):327–340. doi:10.1038/nrneph.2011.51
  • Donato A, Black A, Jablonski K, Gano L, Seals D. Aging is associated with greater nuclear NF kappa B, reduced I kappa B alpha, and increased expression of proinflammatory cytokines in vascular endothelial cells of healthy humans. Aging Cell. 2008;7(6):805–812. doi:10.1111/j.1474-9726.2008.00438.x
  • Jablonski K, Chonchol M, Pierce G, Walker A, Seals D. 25-Hydroxyvitamin D deficiency is associated with inflammation-linked vascular endothelial dysfunction in middle-aged and older adults. Hypertension. 2011;57(1):63–69. doi:10.1161/HYPERTENSIONAHA.110.160929
  • Walker A, Kaplon R, Pierce G, Nowlan M, Seals D. Prevention of age-related endothelial dysfunction by habitual aerobic exercise in healthy humans: possible role of nuclear factor κB. Clin Sci. 2014;127(11):645–654. doi:10.1042/CS20140030
  • Alexander Y, Osto E, Schmidt-Trucksass A, et al. Endothelial function in cardiovascular medicine: a consensus paper of the European society of cardiology working groups on atherosclerosis and vascular biology, aorta and peripheral vascular diseases, coronary pathophysiology and microcirculation, and thrombosis. Cardiovasc Res. 2021;117(1):29–42. doi:10.1093/cvr/cvaa085
  • Kalucka J, Bierhansl L, Conchinha NV, et al. Quiescent endothelial cells upregulate fatty acid beta-oxidation for vasculoprotection via redox homeostasis. Cell Metab. 2018;28(6):881–894 e813. doi:10.1016/j.cmet.2018.07.016
  • Shah AV, Birdsey GM, Peghaire C, et al. The endothelial transcription factor ERG mediates Angiopoietin-1-dependent control of Notch signalling and vascular stability. Nat Commun. 2017;8:16002. doi:10.1038/ncomms16002
  • Sena CM, Carrilho F, Seiça RM. Endothelial dysfunction in type 2 diabetes: targeting inflammation. Endoth Dysf. 2018;24:23110.
  • Cook-Mills JM, Deem TL. Active participation of endothelial cells in inflammation. J Leukoc Biol. 2005;77(4):487–495. doi:10.1189/jlb.0904554
  • Landmesser U, Hornig B, Drexler H. Endothelial function: a critical determinant in atherosclerosis? Circulation. 2004;109(21 Suppl 1):II27–33. doi:10.1161/01.CIR.0000129501.88485.1f
  • Tousoulis D, Antoniades C, Stefanadis C. Evaluating endothelial function in humans: a guide to invasive and non-invasive techniques. Heart. 2005;91(4):553–558. doi:10.1136/hrt.2003.032847
  • Valenzuela CA, Baker EJ, Miles EA, Calder PC. Eighteen‑carbon trans fatty acids and inflammation in the context of atherosclerosis. Prog Lipid Res. 2019;76:101009. doi:10.1016/j.plipres.2019.101009
  • Dehghani T, Panitch A. Endothelial cells, neutrophils and platelets: getting to the bottom of an inflammatory triangle. Open Biol. 2020;10(10):200161. doi:10.1098/rsob.200161
  • Okamoto T, Park EJ, Kawamoto E, et al. Endothelial connexin-integrin crosstalk in vascular inflammation. Biochim Biophys Acta Mol Basis Dis. 2021;1867(9):166168. doi:10.1016/j.bbadis.2021.166168
  • Brocq ML, Leslie SJ, Milliken P, Megson IL. Endothelial dysfunction: from molecular mechanisms to measurement, clinical implications, and therapeutic opportunities. Antioxid Redox Signal. 2008;10(9):1631–1674. doi:10.1089/ars.2007.2013
  • Aird WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ Res. 2007;100(2):174–190. doi:10.1161/01.RES.0000255690.03436.ae
  • Tracy R. Emerging relationships of inflammation, cardiovascular disease and chronic diseases of aging. Int J Obes Relat Metab Disord. 2003;27(3):S29–S34. doi:10.1038/sj.ijo.0802497
  • Ricci N, Cunha A. Physical Exercise for Frailty and Cardiovascular Diseases. Adv Exp Med Biol. 2020;1216:115–129.
  • Eloueyk A, Osta B, Alameldinne R, Awad D. Uremic serum induces inflammation in cultured human endothelial cells and triggers vascular repair mechanisms. Inflammation. 2019;42(6):2003–2010. doi:10.1007/s10753-019-01061-7
  • Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther. 2022;7(1):131. doi:10.1038/s41392-022-00955-7
  • Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107. doi:10.1038/nri2925
  • Libby P, Buring JE, Badimon L, et al. Atherosclerosis. Nat Rev Dis Primers. 2019;5(1):56. doi:10.1038/s41572-019-0106-z
  • Lu J, Chen X, Xu X, et al. Active polypeptides from Hirudo inhibit endothelial cell inflammation and macrophage foam cell formation by regulating the LOX-1/LXR-α/ABCA1 pathway. Biom Pharmacoth. 2019;115:108840. doi:10.1016/j.biopha.2019.108840
  • Ross R. Atherosclerosis—an inflammatory disease. New England J Med. 1999;340(2):115–126. doi:10.1056/NEJM199901143400207
  • Libby P. The changing landscape of atherosclerosis. Nature. 2021;592(7855):524–533. doi:10.1038/s41586-021-03392-8
  • Libby P, Hansson GK. From focal lipid storage to systemic inflammation. J Am Coll Cardiol. 2019;74(12):1594–1607. doi:10.1016/j.jacc.2019.07.061
  • Gimbrone MA, García-Cardeña G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 2016;118(4):620–636. doi:10.1161/CIRCRESAHA.115.306301
  • Mussbacher M, Schossleitner K, Kral-Pointner JB, Salzmann M, Schrammel A, Schmid JA. More than just a monolayer: the multifaceted role of endothelial cells in the pathophysiology of atherosclerosis. Curr Atheroscler Rep. 2022;24(6):483–492. doi:10.1007/s11883-022-01023-9
  • Xu K, Saaoud F, Yu S, et al. Monocyte adhesion assays for detecting endothelial cell activation in vascular inflammation and atherosclerosis. Atherosclerosis. 2022;2022:169–182.
  • Cybulsky MI, Gimbrone MA. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science. 1991;251(4995):788–791. doi:10.1126/science.1990440
  • Lusis A. Atherosclerosis. Nature. 2000;407(6801):233–241. doi:10.1038/35025203
  • Rader DJ, Puré E. Lipoproteins, macrophage function, and atherosclerosis: beyond the foam cell? Cell Metab. 2005;1(4):223–230. doi:10.1016/j.cmet.2005.03.005
  • Hu W, Lu H, Zhang J, et al. Kruppel-like factor 14, a coronary artery disease associated transcription factor, inhibits endothelial inflammation via NF-kappaB signaling pathway. Atherosclerosis. 2018;278:39–48.
  • Qin M, Wang W, Zhou H, Wang X, Wang F, Wang H. Circular RNA circ_0003645 silencing alleviates inflammation and apoptosis via the NF-κB pathway in endothelial cells induced by oxLDL. Gene. 2020;755:144900. doi:10.1016/j.gene.2020.144900
  • Yu J, Ming H, Li HY, et al. IMM-H007, a novel small molecule inhibitor for atherosclerosis, represses endothelium inflammation by regulating the activity of NF-κB and JNK/AP1 signaling. Toxicol Appl Pharmacol. 2019;381:114732. doi:10.1016/j.taap.2019.114732
  • Zhang Y, Guan Q, Wang Z. PTP1B inhibition ameliorates inflammatory injury and dysfunction in ox-LDL-induced HUVECs by activating the AMPK/SIRT1 signaling pathway via negative regulation of KLF2. Exp Ther Med. 2022;24(1):467. doi:10.3892/etm.2022.11394
  • Chen M, Li W, Zhang Y, Yang J. MicroRNA-20a protects human aortic endothelial cells from Ox-LDL-induced inflammation through targeting TLR4 and TXNIP signaling. Biom Pharmacoth. 2018;103:191–197. doi:10.1016/j.biopha.2018.03.129
  • Chen L, Qin L, Liu X, Meng X. CTRP3 alleviates Ox-LDL–induced inflammatory response and endothelial dysfunction in mouse aortic endothelial cells by activating the PI3K/Akt/eNOS pathway. Inflammation. 2019;42(4):1350–1359. doi:10.1007/s10753-019-00996-1
  • Liu Y, Tie L. Apolipoprotein M and sphingosine-1-phosphate complex alleviates TNF-α-induced endothelial cell injury and inflammation through PI3K/AKT signaling pathway. BMC Cardiovasc Disord. 2019;19(1):1–9. doi:10.1186/s12872-019-1263-4
  • Xu K, Zhao H, Qiu X, Liu X, Zhao F, Zhao Y. VGLL4 Protects against oxidized-LDL-Induced Endothelial cell dysfunction and inflammation by activating Hippo-YAP/TEAD1 signaling pathway. Mediators Inflamm. 2020;2020:1–9. doi:10.1155/2020/8292173
  • Xu K, Xiwen L, Ren G, Yin D, Guo S, Zhao Y. Depletion of CPEB1 protects against oxidized LDL-induced endothelial apoptosis and inflammation though SIRT1/LOX-1 signalling pathway. Life Sci. 2019;239:116874. doi:10.1016/j.lfs.2019.116874
  • Ou HC, Chou WC, Hung CH, et al. Galectin‐3 aggravates ox‐LDL‐induced endothelial dysfunction through LOX‐1 mediated signaling pathway. Environ Toxicol. 2019;34(7):825–835. doi:10.1002/tox.22750
  • Sun P, Li L, Liu Y, et al. MiR-181b regulates atherosclerotic inflammation and vascular endothelial function through Notch1 signaling pathway. Eur Rev Med Pharmacol Sci. 2019;23(7):3051–3057. doi:10.26355/eurrev_201904_17587
  • Gao ZF, Ji XL, Gu J, Wang XY, Ding L, Zhang H. microRNA‐107 protects against inflammation and endoplasmic reticulum stress of vascular endothelial cells via KRT1‐dependent Notch signaling pathway in a mouse model of coronary atherosclerosis. J Cell Physiol. 2019;234(7):12029–12041. doi:10.1002/jcp.27864
  • Ding J, Li Z, Li L, et al. Myosin light chain kinase inhibitor ML7 improves vascular endothelial dysfunction and permeability via the mitogen-activated protein kinase pathway in a rabbit model of atherosclerosis. Biom Pharmacoth. 2020;128:110258. doi:10.1016/j.biopha.2020.110258
  • Yang S, Mi X, Chen Y, et al. MicroRNA‐216a induces endothelial senescence and inflammation via Smad3/IκBα pathway. J Cell Mol Med. 2018;22(5):2739–2749. doi:10.1111/jcmm.13567
  • Kutryb-Zajac B, Mierzejewska P, Sucajtys-Szulc E, et al. Inhibition of LPS-stimulated ecto-adenosine deaminase attenuates endothelial cell activation. J Mol Cell Cardiol. 2019;128:62–76. doi:10.1016/j.yjmcc.2019.01.004
  • Gao W, Liu H, Yuan J, et al. Exosomes derived from mature dendritic cells increase endothelial inflammation and atherosclerosis via membrane TNF‐α mediated NF‐κB pathway. J Cell Mol Med. 2016;20(12):2318–2327. doi:10.1111/jcmm.12923
  • Zhao S, Liang M, Wang Y, et al. Chrysin suppresses vascular endothelial inflammation via inhibiting the NF-κB signaling pathway. J Cardiovasc Pharmacol Ther. 2019;24(3):278–287. doi:10.1177/1074248418810809
  • Zhong Y, He S, Huang K, Liang M. Neferine suppresses vascular endothelial inflammation by inhibiting the NF-κB signaling pathway. Arch Biochem Biophys. 2020;696:108595. doi:10.1016/j.abb.2020.108595
  • Zhang S, Xie S, Gao Y, Wang Y. Triptolide alleviates oxidized LDL-induced endothelial inflammation by attenuating the oxidative stress-mediated nuclear factor-kappa B pathway. Curr Ther Res Clin Exp. 2022;97:100683. doi:10.1016/j.curtheres.2022.100683
  • Lee SH, Han AR, Kim BM, Jeong Sung M, Hong SM. Lactococcus lactis-fermented spinach juice suppresses LPS-induced expression of adhesion molecules and inflammatory cytokines through the NF-kappaB pathway in HUVECs. Exp Ther Med. 2022;23(6):390. doi:10.3892/etm.2022.11317
  • Li X, Tang Y, Ma B, et al. The peptide lycosin-I attenuates TNF-α-induced inflammation in human umbilical vein endothelial cells via IκB/NF-κB signaling pathway. Inflammat Res. 2018;67(5):455–466. doi:10.1007/s00011-018-1138-7
  • Zhou YQ, Zhao YT, Zhao XY, et al. Hyperoside suppresses lipopolysaccharide-induced inflammation and apoptosis in human umbilical vein endothelial cells. Curr Med Sci. 2018;38(2):222–228. doi:10.1007/s11596-018-1869-2
  • Mohd Ariff A, Abu Bakar NA, Omar E, et al. Ficus deltoidea suppresses endothelial activation, inflammation, monocytes adhesion and oxidative stress via NF-κB and eNOS pathways in stimulated human coronary artery endothelial cells. BMC Complement Med Therap. 2020;20(1):1–13. doi:10.1186/s12906-020-2844-6
  • Hu H, Wang C, Jin Y, et al. Catalpol inhibits homocysteine-induced oxidation and inflammation via inhibiting Nox4/NF-kappaB and GRP78/PERK pathways in human aorta endothelial cells. Inflammation. 2019;42(1):64–80. doi:10.1007/s10753-018-0873-9
  • Luo L, Liang H, Liu L. Myristicin regulates proliferation and apoptosis in oxidized low-density lipoprotein-stimulated human vascular smooth muscle cells and human umbilical vein endothelial cells by regulating the PI3K/Akt/NF-kappaB signalling pathway. Pharm Biol. 2022;60(1):56–64. doi:10.1080/13880209.2021.2010775
  • Sun H, Zhu X, Cai W, Qiu L. Hypaphorine attenuates lipopolysaccharide-induced endothelial inflammation via regulation of TLR4 and PPAR-γ dependent on PI3K/Akt/mTOR signal pathway. Int J Mol Sci. 2017;18(4):844. doi:10.3390/ijms18040844
  • Wang Y, Hong Y, Zhang C, et al. Picroside II attenuates hyperhomocysteinemia‐induced endothelial injury by reducing inflammation, oxidative stress and cell apoptosis. J Cell Mol Med. 2019;23(1):464–475. doi:10.1111/jcmm.13949
  • Zhao H, Liu M, Liu H, Suo R, Lu C. Naringin protects endothelial cells from apoptosis and inflammation by regulating the Hippo-YAP Pathway. Biosci Rep. 2020;40(3). doi:10.1042/BSR20193431
  • Zhao Z, Wang X, Zhang R, et al. Melatonin attenuates smoking-induced atherosclerosis by activating the Nrf2 pathway via NLRP3 inflammasomes in endothelial cells. Aging. 2021;13(8):11363. doi:10.18632/aging.202829
  • Meng N, Chen K, Wang Y, et al. Dihydrohomoplantagin and homoplantaginin, major flavonoid glycosides from salvia plebeia R. Br. Inhibit oxLDL-induced endothelial cell injury and restrict atherosclerosis via activating Nrf2 anti-oxidation signal pathway. Molecules. 2022;27(6):1990. doi:10.3390/molecules27061990
  • Sun X, Jiao X, Ma Y, et al. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem Biophys Res Commun. 2016;481(1–2):63–70. doi:10.1016/j.bbrc.2016.11.017
  • Sun J, Yin X, Liu H, et al. Rapamycin inhibits ox-LDL-induced inflammation in human endothelial cells in vitro by inhibiting the mTORC2/PKC/c-Fos pathway. Acta Pharmacol Sin. 2018;39(3):336–344. doi:10.1038/aps.2017.102
  • Li L, Chen Y, Shi C. Nintedanib ameliorates oxidized low-density lipoprotein -induced inflammation and cellular senescence in vascular endothelial cells. Bioengineered. 2022;13(3):6196–6207. doi:10.1080/21655979.2022.2036913
  • Qu D, Wang L, Huo M, et al. Focal TLR4 activation mediates disturbed flow-induced endothelial inflammation. Cardiovasc Res. 2020;116(1):226–236. doi:10.1093/cvr/cvz046
  • Zhang G, Qin Q, Zhang C, et al. NDRG1 signaling is essential for endothelial inflammation and vascular remodeling. Circ Res. 2023;132(3):306–319. doi:10.1161/CIRCRESAHA.122.321837
  • Wang L, Gu Z, Li J, et al. Isorhynchophylline inhibits inflammatory responses in endothelial cells and macrophages through the NF-κB/NLRP3 signaling pathway. BMC Complement Med Ther. 2023;23(1):80. doi:10.1186/s12906-023-03902-3
  • Niu N, Xu S, Xu Y, Little PJ, Jin Z-G. Targeting mechanosensitive transcription factors in atherosclerosis. Trends Pharmacol Sci. 2019;40(4):253–266. doi:10.1016/j.tips.2019.02.004
  • Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol. 2022;22(4):251–265. doi:10.1038/s41577-021-00584-1
  • Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Endothelial cells: from dysfunction mechanism to pharmacological effect in cardiovascular disease. Cardiovasc Toxicol. 2019;19(1):13–22. doi:10.1007/s12012-018-9493-8
  • Naseem KM. The role of nitric oxide in cardiovascular diseases. Mol Aspects Med. 2005;26(1–2):33–65. doi:10.1016/j.mam.2004.09.003
  • Dhaun N, Webb DJ. Endothelins in cardiovascular biology and therapeutics. Nat Rev Cardiol. 2019;16(8):491–502. doi:10.1038/s41569-019-0176-3
  • Carnevale D. Neuroimmune axis of cardiovascular control: mechanisms and therapeutic implications. Nat Rev Cardiol. 2022;19(6):379–394. doi:10.1038/s41569-022-00678-w
  • Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17(11):1410–1422. doi:10.1038/nm.2538
  • Murray K, Mahoney S, Venkatasubramanian R, Seals D, Clayton Z. Aging, aerobic exercise, and cardiovascular health: barriers, alternative strategies and future directions. Exp Gerontol. 2023;173:112105. doi:10.1016/j.exger.2023.112105
  • Kozakova M, Palombo C. Vascular ageing and aerobic exercise. Int J Environ Res Public Health. 2021;18(20):10666. doi:10.3390/ijerph182010666
  • Craighead D, Heinbockel T, Freeberg K, et al. Time-efficient inspiratory muscle strength training lowers blood pressure and improves endothelial function, NO bioavailability, and oxidative stress in midlife/older adults with above-normal blood pressure. J Am Heart Assoc. 2021;10(13):e020980. doi:10.1161/JAHA.121.020980
  • Craighead D, Freeberg K, Maurer G, Myers V, Seals D. Translational potential of high-resistance inspiratory muscle strength training. Exerc Sport Sci Rev. 2022;50(3):107–117. doi:10.1249/JES.0000000000000293
  • Clayton Z, Hutton D, Brunt V, et al. Apigenin restores endothelial function by ameliorating oxidative stress, reverses aortic stiffening, and mitigates vascular inflammation with aging. Am J Physiol Heart Circ Physiol. 2021;321(1):H185–H196. doi:10.1152/ajpheart.00118.2021
  • Freeberg K, Craighead D, Martens C, You Z, Chonchol M, Seals D. Nicotinamide riboside supplementation for treating elevated systolic blood pressure and arterial stiffness in midlife and older adults. Front Cardiovasc Med. 2022;9:881703. doi:10.3389/fcvm.2022.881703
  • Stanhewicz A, Dillon G, Serviente C, Alexander L. Acute systemic inhibition of inflammation augments endothelium-dependent dilation in women with a history of preeclamptic pregnancy. Pregnancy Hypertens. 2022;27:81–86. doi:10.1016/j.preghy.2021.12.010
  • Hingorani A, Cross J, Kharbanda R, et al. Acute systemic inflammation impairs endothelium-dependent dilatation in humans. Circulation. 2000;102(9):994–999. doi:10.1161/01.CIR.102.9.994
  • Greaney J, Saunders E, Alexander L. Short-term salicylate treatment improves microvascular endothelium-dependent dilation in young adults with major depressive disorder. Am J Physiol Heart Circ Physiol. 2022;322(5):H880–H889. doi:10.1152/ajpheart.00643.2021
  • Stapleton P, Minarchick V, Cumpston A, et al. Impairment of coronary arteriolar endothelium-dependent dilation after multi-walled carbon nanotube inhalation: a time-course study. Int J Mol Sci. 2012;13(11):13781–13803. doi:10.3390/ijms131113781
  • Nurkiewicz T, Porter D, Barger M, Castranova V, Boegehold M. Particulate matter exposure impairs systemic microvascular endothelium-dependent dilation. Environ Health Perspect. 2004;112(13):1299–1306. doi:10.1289/ehp.7001
  • Ward AO, Angelini GD, Caputo M, et al. NF-κB inhibition prevents acute shear stress-induced inflammation in the saphenous vein graft endothelium. Sci Rep. 2020;10(1):1–10. doi:10.1038/s41598-020-71781-6
  • Zhou X, Cai J, Liu W, Wu X, Gao C. Cysteinyl leukotriene receptor type 1 (CysLT1R) antagonist zafirlukast protects against TNF-α-induced endothelial inflammation. Biom Pharmacoth. 2019;111:452–459. doi:10.1016/j.biopha.2018.12.064
  • You S, Qian J, Wu G, et al. Schizandrin B attenuates angiotensin II induced endothelial to mesenchymal transition in vascular endothelium by suppressing NF-κB activation. Phytomedicine. 2019;62:152955. doi:10.1016/j.phymed.2019.152955
  • Reddy AT, Lakshmi SP, Prasad EM, Varadacharyulu NC, Kodidhela LD. Epigallocatechin gallate suppresses inflammation in human coronary artery endothelial cells by inhibiting NF-κB. Life Sci. 2020;258:118136. doi:10.1016/j.lfs.2020.118136
  • An Z, Yang G, Nie W, Ren J, Wang D. MicroRNA-106b overexpression alleviates inflammation injury of cardiac endothelial cells by targeting BLNK via the NF-kappaB signaling pathway. J Cell Biochem. 2018;119(4):3451–3463. doi:10.1002/jcb.26517
  • Qian X, Zhu M, Qian W, Song J. Vitamin D attenuates myocardial ischemia–reperfusion injury by inhibiting inflammation via suppressing the RhoA/ROCK/NF‐ĸB pathway. Biotechnol Appl Biochem. 2019;66(5):850–857. doi:10.1002/bab.1797
  • Jiang M, Wang H, Liu Z, et al. Endoplasmic reticulum stress‐dependent activation of iNOS/NO‐NF‐κB signaling and NLRP3 inflammasome contributes to endothelial inflammation and apoptosis associated with microgravity. FASEB J. 2020;34(8):10835–10849. doi:10.1096/fj.202000734R
  • Sun S, Ji Z, Fu J, Wang X, Zhang L. Endosulfan induces endothelial inflammation and dysfunction via IRE1α/NF-κB signaling pathway. Environ Sci Pollut Res Int. 2020;27(21):26163–26171. doi:10.1007/s11356-020-09023-5
  • Zhang H, Liang B, Li T, Zhou Y, Shang D, Du Z. Orexin A suppresses oxidized LDL induced endothelial cell inflammation via MAPK p38 and NF‐κB signaling pathway. IUBMB Life. 2018;70(10):961–968. doi:10.1002/iub.1890
  • Cao X, Bi R, Hao J, et al. A study on the protective effects of taxifolin on human umbilical vein endothelial cells and THP-1 cells damaged by hexavalent chromium: a probable mechanism for preventing cardiovascular disease induced by heavy metals. Food Funct. 2020;11(5):3851–3859. doi:10.1039/D0FO00567C
  • Zhao W, Wu C, Li S, Chen X. Adiponectin protects palmitic acid induced endothelial inflammation and insulin resistance via regulating ROS/IKKβ pathways. Cytokine. 2016;88:167–176. doi:10.1016/j.cyto.2016.09.005
  • Huang Y, Zhang K, Liu M, et al. An herbal preparation ameliorates heart failure with preserved ejection fraction by alleviating microvascular endothelial inflammation and activating NO-cGMP-PKG pathway. Phytomedicine. 2021;91:153633. doi:10.1016/j.phymed.2021.153633
  • Wang M, Peng X, Lian Z, Zhu H. The cordycepin derivative IMM-H007 improves endothelial dysfunction by suppressing vascular inflammation and promoting AMPK-dependent eNOS activation in high-fat diet-fed ApoE knockout mice. Eur J Pharmacol. 2019;852:167–178. doi:10.1016/j.ejphar.2019.02.045
  • Lyu TJ, Zhang ZX, Chen J, Liu ZJ. Ginsenoside Rg1 ameliorates apoptosis, senescence and oxidative stress in ox-LDL-induced vascular endothelial cells via the AMPK/SIRT3/p53 signaling pathway. Exp Ther Med. 2022;24(3):545. doi:10.3892/etm.2022.11482
  • Teixeira TM, Da costa DC, Resende AC, Soulage CO, Bezerra FF, Daleprane JB. Activation of Nrf2-antioxidant signaling by 1, 25-dihydroxycholecalciferol prevents leptin-induced oxidative stress and inflammation in human endothelial cells. J Nutr. 2017;147(4):506–513. doi:10.3945/jn.116.239475
  • Ma Y, Li L, Shao Y, Bai X, Bai T, Huang X. Methotrexate improves perivascular adipose tissue/endothelial dysfunction via activation of AMPK/eNOS pathway. Mol Med Rep. 2017;15(4):2353–2359. doi:10.3892/mmr.2017.6225
  • Deanfield JE, Halcox JP, Rabelink T. Endothelial function and dysfunction: testing and clinical relevance. Circulation. 2007;115(10):1285–1295. doi:10.1161/CIRCULATIONAHA.106.652859
  • Yao Mattisson I, Christoffersen C. Apolipoprotein M and its impact on endothelial dysfunction and inflammation in the cardiovascular system. Atherosclerosis. 2021;334:76–84. doi:10.1016/j.atherosclerosis.2021.08.039
  • Seino Y, Nanjo K, Tajima N, et al. Report of the committee on the classification and diagnostic criteria of diabetes mellitus. J Diabetes Investig. 2010;2010:1.
  • Tang R, Li Q, Lv L, et al. Angiotensin II mediates the high-glucose-induced endothelial-to-mesenchymal transition in human aortic endothelial cells. Cardiovasc Diabetol. 2010;9:31. doi:10.1186/1475-2840-9-31
  • Lee HM, Kim JJ, Kim HJ, Shong M, Ku BJ, Jo EK. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes. 2013;62(1):194–204. doi:10.2337/db12-0420
  • Balletshofer BM, Rittig K, Enderle MD, et al. Endothelial dysfunction is detectable in young normotensive first-degree relatives of subjects with type 2 diabetes in association with insulin resistance. Circulation. 2000;101(15):1780–1784. doi:10.1161/01.CIR.101.15.1780
  • Mobasseri M, Ostadrahimi A, Tajaddini A, et al. Effects of saffron supplementation on glycemia and inflammation in patients with type 2 diabetes mellitus: a randomized double-blind, placebo-controlled clinical trial study. Diabetes Metab Syndr. 2020;14(4):527–534. doi:10.1016/j.dsx.2020.04.031
  • Lontchi-Yimagou E, Sobngwi E, Matsha TE, Kengne AP. Diabetes mellitus and inflammation. Curr Diab Rep. 2013;13(3):435–444.
  • Gong Y, Zhang Y, Su X, Gao H. Inhibition of long noncoding RNA MALAT1 suppresses high glucose-induced apoptosis and inflammation in human umbilical vein endothelial cells by suppressing the NF-κB signaling pathway. Biochemist Cell Bio. 2020;98(6):669–675. doi:10.1139/bcb-2019-0403
  • Wei H, Gu Q. SOX4 promotes high-glucose-induced inflammation and angiogenesis of retinal endothelial cells by activating NF-kappaB signaling pathway. Open Life Sci. 2022;17(1):393–400. doi:10.1515/biol-2022-0045
  • Fei Y, Sun L, Yuan C, Jiang M, Lou Q, Xu Y. CFTR ameliorates high glucose-induced oxidative stress and inflammation by mediating the NF-κB and MAPK signaling pathways in endothelial cells. Int J Mol Me. 2018;41(6):3501–3508.
  • Cheng J, Liu Q, Hu N, et al. Downregulation of hsa_circ_0068087 ameliorates TLR4/NF-κB/NLRP3 inflammasome-mediated inflammation and endothelial cell dysfunction in high glucose conditioned by sponging miR-197. Gene. 2019;709:1–7. doi:10.1016/j.gene.2019.05.012
  • Valle ML, Dworshak J, Sharma A, Ibrahim AS, Al-Shabrawey M, Sharma S. Inhibition of interleukin-6 trans-signaling prevents inflammation and endothelial barrier disruption in retinal endothelial cells. Exp Eye Res. 2019;178:27–36. doi:10.1016/j.exer.2018.09.009
  • Wang H, Sun P, Chen W, et al. High glucose stimulates expression of MFHAS1 to mitigate inflammation via Akt/HO-1 pathway in human umbilical vein endothelial cells. Inflammation. 2018;41(2):400–408. doi:10.1007/s10753-017-0696-0
  • Song W, Zhang C-L, Gou L, et al. Endothelial TFEB (transcription factor EB) restrains IKK (IκB kinase)-p65 pathway to attenuate vascular inflammation in diabetic db/db mice. Arterioscler Thromb Vasc Biol. 2019;39(4):719–730. doi:10.1161/ATVBAHA.119.312316
  • Yi J, Gao Z-F. MicroRNA-9-5p promotes angiogenesis but inhibits apoptosis and inflammation of high glucose-induced injury in human umbilical vascular endothelial cells by targeting CXCR4. Int J Biol Macromol. 2019;130:1–9. doi:10.1016/j.ijbiomac.2019.02.003
  • Wang T, Zhu H, Hou Y, Duan W, Meng F, Liu Y. Ketamine attenuates high-glucose-mediated endothelial inflammation in human umbilical vein endothelial cells. Can J Physiol Pharmacol. 2020;98(3):156–161. doi:10.1139/cjpp-2019-0185
  • Petersen C, Bharat D, Cutler BR, et al. Circulating metabolites of strawberry mediate reductions in vascular inflammation and endothelial dysfunction in db/db mice. Int J Cardiol. 2018;263:111–117. doi:10.1016/j.ijcard.2018.04.040
  • Hu R, Wang MQ, Ni SH, et al. Salidroside ameliorates endothelial inflammation and oxidative stress by regulating the AMPK/NF-kappaB/NLRP3 signaling pathway in AGEs-induced HUVECs. Eur J Pharmacol. 2020;867:172797. doi:10.1016/j.ejphar.2019.172797
  • Shi X, Wang S, Luan H, et al. Clinopodium chinense attenuates palmitic acid-induced vascular endothelial inflammation and insulin resistance through TLR4-mediated NF-κB and MAPK pathways. Am J Chin Med. 2019;47(01):97–117. doi:10.1142/S0192415X19500058
  • Shu A, Du Q, Chen J, et al. Catalpol ameliorates endothelial dysfunction and inflammation in diabetic nephropathy via suppression of RAGE/RhoA/ROCK signaling pathway. Chem Biol Interact. 2021;348:109625. doi:10.1016/j.cbi.2021.109625
  • Lin J, Li X, Lin Y, Huang Z, Wu W. Exogenous sodium hydrosulfide protects against high glucose‑induced injury and inflammation in human umbilical vein endothelial cells by inhibiting necroptosis via the p38 MAPK signaling pathway. Mol Med Rep. 2021;23(1):1–1.
  • Ge CY, Yang L, Zhang JL, Wei ZF, Feng F. Spinosin ameliorates insulin resistance by suppressing reactive oxygen species-associated inflammation. Iran J Basic Med Sci. 2022;25(7):850–858. doi:10.22038/IJBMS.2022.64154.14127
  • Yang R, Yang Y. Albiflorin attenuates high glucose-induced endothelial apoptosis via suppressing PARP1/NF-κB signaling pathway. Inflamm Res. 2023;72(1):159–169. doi:10.1007/s00011-022-01666-z
  • Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest. 1992;101(6):1644–1655. doi:10.1378/chest.101.6.1644
  • Kumar S, Tripathy S, Jyoti A, Singh SG. Recent advances in biosensors for diagnosis and detection of sepsis: a comprehensive review. Bio Bioelect. 2019;124:205–215. doi:10.1016/j.bios.2018.10.034
  • Salomao R, Brunialti MKC, Rapozo MM, Baggio-Zappia GL, Galanos C, Freudenberg M. Bacterial sensing, cell signaling, and modulation of the immune response during sepsis. Shock. 2012;38(3):227–242. doi:10.1097/SHK.0b013e318262c4b0
  • van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–420. doi:10.1038/nri.2017.36
  • Schrijver IT, Théroude C, Roger T. Myeloid-derived suppressor cells in sepsis. Front Immunol. 2019;10:327. doi:10.3389/fimmu.2019.00327
  • Salomão R, Ferreira B, Salomão M, Santos S, Azevedo L, Brunialti M. Sepsis: evolving concepts and challenges. Braz J Med Biol Res. 2019;52(4):e8595. doi:10.1590/1414-431x20198595
  • Geven C, Peters E, Schroedter M, et al. Effects of the humanized anti-adrenomedullin antibody adrecizumab (HAM8101) on vascular barrier function and survival in rodent models of systemic inflammation and sepsis. Shock. 2018;50(6):648–654. doi:10.1097/SHK.0000000000001102
  • Bone RC. The pathogenesis of sepsis. Ann Intern Med. 1991;115(6):457–469. doi:10.7326/0003-4819-115-6-457
  • Konradt C, Hunter CA. Pathogen interactions with endothelial cells and the induction of innate and adaptive immunity. Eur J Immunol. 2018;48(10):1607–1620. doi:10.1002/eji.201646789
  • Chen Y, Tang D, Zhu L, et al. hnRNPA2/B1 Ameliorates LPS-Induced Endothelial Injury through NF-κB Pathway and VE-Cadherin/β-catenin signaling modulation in vitro. Mediators Inflamm. 2020;2020. doi:10.1155/2020/6458791
  • Liu Z, Ji J, Zheng D, Su L, Peng T, Tang J. Protective role of endothelial calpain knockout in lipopolysaccharide-induced acute kidney injury via attenuation of the p38-iNOS pathway and NO/ROS production. Exp Mol Med. 2020;52(4):702–712. doi:10.1038/s12276-020-0426-9
  • Lv Y, Kim K, Sheng Y, et al. YAP controls endothelial activation and vascular inflammation through TRAF6. Circ Res. 2018;123(1):43–56. doi:10.1161/CIRCRESAHA.118.313143
  • Zhao S, Gao J, Li J, Wang S, Yuan C, Liu Q. PD-L1 regulates inflammation in LPS-Induced lung epithelial cells and vascular endothelial cells by interacting with the HIF-1α Signaling Pathway. Inflammation. 2021;44(5):1969–1981. doi:10.1007/s10753-021-01474-3
  • Wang Y, Du P, Jiang D. Rigosertib inhibits MEK1–ERK pathway and alleviates lipopolysaccharide‐induced sepsis. Immun Inflam Dis. 2021;9(3):991–999.
  • Huang L, Li Y, Cheng Z, Lv Z, Luo S, Xia Y. PCSK9 promotes endothelial dysfunction during sepsis via the TLR4/MyD88/NF-κB and NLRP3 pathways. Inflammation. 2023;46(1):115–128. doi:10.1007/s10753-022-01715-z
  • Mehta M, Dhanjal DS, Paudel KR, et al. Cellular signalling pathways mediating the pathogenesis of chronic inflammatory respiratory diseases: an update. Inflammopharmacology. 2020;28(4):795–817. doi:10.1007/s10787-020-00698-3
  • Arora VK, Chopra KK. Inflammation plays a central role in respiratory diseases, including tuberculosis. Indian J Tuberc. 2018;65(2):103–105. doi:10.1016/j.ijtb.2018.03.001
  • Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Autophagy. 2018;14(2):221–232. doi:10.1080/15548627.2017.1389823
  • Huang S, Zeng R, Wang J, et al. Follistatin-like 1 induces the activation of type 2 innate lymphoid cells to promote airway inflammation in asthma. Inflammation. 2022;45(2):904–918. doi:10.1007/s10753-021-01594-w
  • Fehrenbach H, Wagner C, Wegmann M. Airway remodeling in asthma: what really matters. Cell Tissue Res. 2017;367(3):551–569.
  • Wei L, Gou X, Su B, et al. Mahuang decoction attenuates airway inflammation and remodeling in asthma via suppression of the SP1/FGFR3/PI3K/AKT axis. Drug Des Devel Ther. 2022;16:2833–2850. doi:10.2147/DDDT.S351264
  • Nakanishi K, Takeda Y, Tetsumoto S, et al. Involvement of endothelial apoptosis underlying chronic obstructive pulmonary disease–like phenotype in adiponectin-null mice: implications for therapy. Am J Respir Crit Care Med. 2011;183(9):1164–1175. doi:10.1164/rccm.201007-1091OC
  • Fan EK, Fan J. Regulation of alveolar macrophage death in acute lung inflammation. Respir Res. 2018;19(1):1–13. doi:10.1186/s12931-018-0756-5
  • Ha EH, Choi J-P, Kwon H-S, et al. Endothelial Sox17 promotes allergic airway inflammation. J Allergy Clin Immunol. 2019;144(2):561–573. e566. doi:10.1016/j.jaci.2019.02.034
  • Chen S, Wang Y, Zhang H, et al. The antioxidant MitoQ protects against CSE-induced endothelial barrier injury and inflammation by inhibiting ROS and autophagy in human umbilical vein endothelial cells. Int J Biol Sci. 2019;15(7):1440. doi:10.7150/ijbs.30193
  • Xu F, Zhou F. Inhibition of microRNA-92a ameliorates lipopolysaccharide-induced endothelial barrier dysfunction by targeting ITGA5 through the PI3K/Akt signaling pathway in human pulmonary microvascular endothelial cells. Int Immunopharmacol. 2020;78:106060. doi:10.1016/j.intimp.2019.106060
  • Zhang L, Ge S, He W, Chen Q, Xu C, Zeng M. Ghrelin protects against lipopolysaccharide-induced acute respiratory distress syndrome through the PI3K/AKT pathway. J Biol Chem. 2021;297(3):101111. doi:10.1016/j.jbc.2021.101111
  • Yang J, Ruan F, Zheng Z. Ripasudil attenuates lipopolysaccharide (LPS)-mediated apoptosis and inflammation in pulmonary microvascular endothelial cells via ROCK2/eNOS signaling. Med Sci Monit. 2018;24:3212. doi:10.12659/MSM.910184
  • Nezic L, Amidzic L, Skrbic R, et al. Amelioration of endotoxin-induced acute lung injury and alveolar epithelial cells apoptosis by simvastatin is associated with up-regulation of survivin/NF-kB/p65 pathway. Int J Mol Sci. 2022;23(5). doi:10.3390/ijms23052596
  • Qiu Q, Yu X, Chen Q, He X. Sema3A inactivates the ERK/JNK signalling pathways to alleviate inflammation and oxidative stress in lipopolysaccharide-stimulated rat endothelial cells and lung tissues. Autoimmunity. 2023;56(1):2200908. doi:10.1080/08916934.2023.2200908
  • Pingali U, Sukumaran D, Nutalapati C. Effect of an aqueous extract of Terminalia chebula on endothelial dysfunction, systemic inflammation, and lipid profile in type 2 diabetes mellitus: a randomized double‐blind, placebo‐controlled clinical study. Phytoth Res. 2020;34(12):3226–3235. doi:10.1002/ptr.6771
  • Usharani P, Merugu PL, Nutalapati C, Cha -Y-Y, An H-J. Evaluation of the effects of a standardized aqueous extract of Phyllanthus emblica fruits on endothelial dysfunction, oxidative stress, systemic inflammation and lipid profile in subjects with metabolic syndrome: a randomised, double blind, placebo controlled clinical study. BMC Complement Altern Med. 2019;19(1):1–8. doi:10.1186/s12906-018-2420-5
  • Naeini Z, Toupchian O, Vatannejad A, et al. Effects of DHA-enriched fish oil on gene expression levels of p53 and NF-κB and PPAR-γ activity in PBMCs of patients with T2DM: a randomized, double-blind, clinical trial. Nutr Metab Cardiovasc Dis. 2020;30(3):441–447. doi:10.1016/j.numecd.2019.10.012
  • Pisaniello AD, Psaltis PJ, King PM, et al. Omega-3 fatty acids ameliorate vascular inflammation: a rationale for their atheroprotective effects. Atherosclerosis. 2021;324:27–37. doi:10.1016/j.atherosclerosis.2021.03.003
  • Malik M, Suboc TM, Tyagi S, et al. Lactobacillus plantarum 299v supplementation improves vascular endothelial function and reduces inflammatory biomarkers in men with stable coronary artery disease. Circ Res. 2018;123(9):1091–1102. doi:10.1161/CIRCRESAHA.118.313565
  • Fan Y, Liu J, Miao J, et al. Anti-inflammatory activity of the Tongmai Yangxin pill in the treatment of coronary heart disease is associated with estrogen receptor and NF-κB signaling pathway. J Ethnopharmacol. 2021;276:114106. doi:10.1016/j.jep.2021.114106
  • Jafari T, Fallah AA, Reyhanian A, Sarmast E. Effects of pomegranate peel extract and vitamin E on the inflammatory status and endothelial function in hemodialysis patients: a randomized controlled clinical trial. Food Funct. 2020;11(9):7987–7993. doi:10.1039/D0FO01012J
  • Papathanasiou JV, Petrov I, Tsekoura D, et al. Does group-based high-intensity aerobic interval training improve the inflammatory status in patients with chronic heart failure? Eur J Phys Rehabil Med. 2022;58(2):242–250. doi:10.23736/S1973-9087.21.06894-5
  • Aquila G, Vieceli Dalla Sega F, Marracino L, et al. Ticagrelor Increases SIRT1 and HES1 mRNA levels in peripheral blood cells from patients with stable coronary artery disease and chronic obstructive pulmonary disease. Int J Mol Sci. 2020;21(5):1576. doi:10.3390/ijms21051576
  • Karimi A, Naeini F, Niazkar HR, et al. Nano-curcumin supplementation in critically ill patients with sepsis: a randomized clinical trial investigating the inflammatory biomarkers, oxidative stress indices, endothelial function, clinical outcomes and nutritional status. Food Funct. 2022;13:6596–6612. doi:10.1039/D1FO03746C
  • De cheng R, Guan Hua D, Juntian Z. High throughput screening for intercellular adhesion molecule-1 inhibitor. Yao Xue Xue Bao. 2003;38:405–408.
  • Opar A. Where now for new drugs for atherosclerosis? Nat Rev Drug Discov. 2007;6(5):334–335. doi:10.1038/nrd2326
  • Dhananjayan R, Koundinya K, Malati T, Kutala VK. Endothelial dysfunction in type 2 diabetes mellitus. Indian J Clin Biochem. 2016;31(4):372–379. doi:10.1007/s12291-015-0516-y