Next Article in Journal
Gene Expression Profile of Uterine Leiomyoma from Women Exposed to Different Air Pollution Levels in Metropolitan Cities of Sao Paulo, Brazil
Next Article in Special Issue
Galectin-3, Inflammation, and the Risk of Atrial High-Rate Episodes in Patients with Dual Chamber Pacemakers
Previous Article in Journal
Characterization of the WRKY Gene Family Related to Anthocyanin Biosynthesis and the Regulation Mechanism under Drought Stress and Methyl Jasmonate Treatment in Lycoris radiata
Previous Article in Special Issue
Transplacental Therapeutic Drug Monitoring in Pregnant Women with Fetal Tachyarrhythmia Using HPLC-MS/MS
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis

1
Center for Translational Medicine, College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
2
Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2429; https://doi.org/10.3390/ijms24032429
Submission received: 8 December 2022 / Revised: 12 January 2023 / Accepted: 16 January 2023 / Published: 26 January 2023
(This article belongs to the Special Issue Molecular Pharmacology of Cardiovascular Disease)

Abstract

:
Traditional herbal medicines based on natural products play a pivotal role in preventing and managing atherosclerotic diseases, which are among the leading causes of death globally. Monoterpenes are a large class of naturally occurring compounds commonly found in many aromatic and medicinal plants. Emerging evidence has shown that monoterpenes have many biological properties, including cardioprotective effects. Remarkably, an increasing number of studies have demonstrated the therapeutic potential of natural monoterpenes to protect against the pathogenesis of atherosclerosis. These findings shed light on developing novel effective antiatherogenic drugs from these compounds. Herein, we provide an overview of natural monoterpenes’ effects on atherogenesis and the underlying mechanisms. Monoterpenes have pleiotropic and multitargeted pharmacological properties by interacting with various cell types and intracellular molecular pathways involved in atherogenesis. These properties confer remarkable advantages in managing atherosclerosis, which has been recognized as a multifaceted vascular disease. We also discuss limitations in the potential clinical application of monoterpenes as therapeutic agents against atherosclerosis. We propose perspectives to give new insights into future preclinical research and clinical practice regarding natural monoterpenes.

1. Introduction

Atherosclerosis is the major underlying pathological basis for coronary artery disease, cerebrovascular disease, and peripheral arterial disease, which causes significant morbidity and mortality worldwide [1,2,3,4]. A spectrum of mechanisms is involved in the initiation and progression of atherosclerosis, including endothelial dysfunction, abnormal lipid metabolism, oxidative stress, and inflammation [3,5,6]. Additionally, a wide array of risk factors such as dyslipidemia, obesity, diabetes, hypertension, aging, and smoking are recognized to be associated with atherosclerosis [7], and the interplay between them makes atherosclerosis a complex condition. Over the past decades, despite significant advances in prevention therapies using contemporary intervention and pharmacologic agents against atherosclerosis, the burden of ischemic cardiovascular conditions remains substantial [8,9]. Therefore, to better cope with the life-threatening atherosclerotic cardiovascular disease (ASCVD), it is urgent to seek new effective therapeutic agents targeting atherosclerosis without marked side effects.
Natural products derived from plants remain attractive sources of molecular entities for developing novel medicinal and therapeutic agents [10]. Due to fewer adverse effects and lower cost of natural compounds compared with chemotherapeutic agents, therapies based on natural products have long been widely used in traditional medicines to treat ASCVD in China and many other Asian countries [11,12]. Terpenes represent a large class of plant-derived secondary metabolites with five-carbon isoprene (C5H8) units as their primary structural component [13]. According to the number of isoprene units within their chemical structure, terpenes can be classified into hemiterpene (C5), monoterpene (C10), sesquiterpene (C15), diterpene (C20), sesterpene (C25), triterpene (C30), and polyterpene (>C30) [13]. The monoterpenes, a major chemical group of terpenes with two isoprene units in their structure, are commonly found in many bioactive essential oils and medicinal plants [14]. It has been well documented that monoterpenes have many biological properties, including anti-bacterial, anti-fungal, anti-oxidative, anti-inflammatory, and anti-tumor activities [14]. Moreover, they are widely utilized in the pharmaceutical preparations, food, and cosmetic industries [15]. Notably, a growing body of evidence has shown that monoterpenes are promising in their potential roles in protecting against cardiovascular disease. For example, geniposide, a well-known iridoid glycoside, has been reported to have remarkable therapeutic potential in managing cardiac fibrosis, cardiac hypertrophy, myocardial ischemia/reperfusion injury, obesity-related cardiac injury, atherosclerosis, ischemic stroke, and hypertension. This makes it an attractive candidate for cardiovascular medicine [16]. For a detailed overview of the protective roles of monoterpenoids in the cardiovascular system, we refer to an excellent recent review [17]. In light of the therapeutic potential of monoterpenes in cardiovascular disease, they are involved in an increasing number of patents registered within the cardiovascular field, highlighting the significant role of these natural compounds in developing new drugs intended to prevent and manage cardiovascular disease [18].
As mentioned above, atherosclerosis is the major contributor to CVD. Targeting atherosclerosis is of fundamental importance for counteracting ASCVD. In recent years, investigations on medicinal natural products have led to increased attention to the potential of naturally occurring monoterpenes and their derivatives in treating atherosclerosis. Various cellular and molecular targets of monoterpenes have also been identified. These findings shed light on the mechanisms by which monoterpenes protect against atherosclerosis in the clinic. However, there needs to be detailed reviews on the research progress of monoterpenes in the context of atherosclerosis. In this review, we overview the pharmacological effects of natural monoterpenes on atherosclerosis, focusing on the underlying cellular and molecular mechanisms. A total of 48 natural monoterpenes were included in this review, and their chemical structures are shown in Figure 1. We attempt to provide insights into the development of monoterpene-based pharmacotherapies to prevent and treat atherosclerosis and its related clinical complications.

2. Natural Monoterpenes Modulate Serum Lipid Profile

Hyperlipidemia refers to the dysregulated lipid metabolism manifesting high levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C) and a decreased level of high-density lipoprotein cholesterol (HDL-C) in the circulation. Hyperlipidemia is considered a prominent risk factor for the pathophysiology of atherosclerosis [19]. In the early stage of atherosclerotic lesions, LDL particles accumulate and undergo modification in the intima of the arterial wall, leading to subsequent monocyte recruitment and cholesterol-laden foam cell formation [3]. Multiple lines of evidence from experimental and clinical studies have established that excessive serum LDL-C is not merely associated with high risk but also a direct underlying mechanism of atherosclerosis [20]. Thus, LDL-C is currently an essential target for the intervention of ASCVD. The introduction of statin drugs, which effectively reduce LDL-C, has been the cornerstone for managing hyperlipidemia and ASCVD risk [9]. Nonetheless, safety issues related to statin therapy remain a chief concern because of the associated adverse effects, such as myopathy and hepatotoxicity [21], illustrating the need for new therapeutic strategies.
3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, the rate-limiting enzyme in the biosynthesis of cholesterol, has been a promising target for developing hypolipidemic drugs such as the statins [3]. The expression of HMG-CoA reductase is critically regulated by sterol regulatory element binding protein-2 (SREBP-2), a crucial transcription factor controlling cellular cholesterol homeostasis [22]. HMG-CoA reductase is also post-transcriptionally regulated by the ubiquitin-proteasome system, which depends on the insulin-induced gene 1 (Insig) protein and the ubiquitin ligase gp78 [23]. A growing body of evidence has demonstrated that many natural monoterpenes improve hypercholesterolemia by targeting HMG-CoA reductase. Linalool is a naturally occurring monoterpene present in essential oils of various aromatic medicinal plants. Oral administration of linalool significantly alleviated high-fat diet (HFD)-induced hyperlipidemia in mice by diminishing plasma TC, TG, and LDL-C with a concomitant reduction of HMG-CoA reductase expression [24]. Furthermore, mechanistic studies have found that linalool could reduce the expression of SREBP-2 and enhance Insig expression and ubiquitination of HMG-CoA reductase, thus attenuating SREBP-2-mediated HMG-CoA reductase transcription and accelerating ubiquitin-dependent proteolysis of HMG-CoA reductase [24]. Moreover, as a critical energy sensor of cell metabolism, the AMP-activated protein kinase (AMPK) has been implicated in suppressing HMG-CoA reductase. It thus has therapeutic importance for treating hypercholesterolemia [25]. Administration of monoterpenes, such as amarogentin, oleuropein, and aucubin, significantly reduces serum TC and LDL-C by activating AMPK, suggesting that the modulation of AMPK/HMG-CoA reductase signaling may contribute to the hypocholesterolemic property of these monoterpenes [26,27,28].
Reverse cholesterol transport (RCT) refers to the delivery of accumulated cholesterol from the blood and the peripheral tissue into the liver for excretion. Manipulation of this process is thus expected to achieve the hypocholesterolemic effect [29]. It has been known that low-density lipoprotein receptor (LDLR), scavenger receptor class B type 1 (SR-B1), and ATP-binding cassette G1 (ABCG1) are involved in RCT. Thymoquinone, the major bioactive component in Nigella sativa volatile oil, could serve as a cholesterol-lowering agent by increasing the uptake of serum LDL-C via elevation of hepatic LDLR expression and by inhibiting HMG-CoA reductase-mediated cholesterol synthesis [30]. Geniposide, a well-known monoterpenoid derived from the fruit of Gardenia jasminoides, was found to attenuate cholesterol accumulation in the plasma and the liver, at least partly through facilitating RCT via upregulation of LDLR, SR-B1, and ABCG1 in the liver [31]. Another critical aspect of cholesterol metabolism is converting cholesterol into bile acids and their subsequent excretion. These processes are critically orchestrated by the farnesoid X receptor (FXR). Specifically, when abundant bile acids are produced, FXR-mediated negative feedback regulation suppresses hepatic bile acid synthesis while accelerating ileal bile acid reabsorption [32,33]. Therefore, by inducing bile acid synthesis and inhibiting bile acid reabsorption through FXR suppression, more cholesterol can be converted into bile acids with a concomitant enhancement of bile acids excretion, eventually leading to decreased cholesterol and thus improving atherosclerosis. Notably, a recent study showed that FXR suppression is an important mechanism underlying the protective effects of geniposide on cholesterol homeostasis and atherosclerosis, in addition to the regulation of RCT, as mentioned above [31]. The administration of geniposide significantly modulated the FXR-small heterodimer partner (SHP)-hepatocyte nuclear factor 4 (HNF-4α)/liver receptor homolog-1 (LRH-1) axis in the liver and the FXR/ileal bile acid-binding protein (I-BABP) axis in the ileum. These effects were associated with the induction of bile acid synthesis and excretion processes in animals fed with or without HFD, suggesting that geniposide exerts a hypocholesterolemic effect by regulating FXR-mediated liver-gut crosstalk of bile acids [31]. Similarly, elevation of bile acid excretion and improved serum lipid profile were observed in a rat model of hypercholesterolemia after oral administration of swertiamarin, which is the main constituent of plants such as Enicostemma littorale, making swertiamarin a potent lipid-lowering and atheroprotective agent [34].
Other monoterpenes [35,36,37,38,39,40,41,42,43,44,45,46,47] that modify serum lipid profiles are listed in Table 1. However, the detailed mechanism of their hypocholesterolemic property remains elusive for many.

3. Natural Monoterpenes Protect against Atherosclerosis by Targeting Endothelial Cells

The endothelial cells are integral to the cardiovascular system and function as gatekeepers of vascular health and homeostasis. The dysfunction of vascular endothelial cells has been recognized as a critical component in the pathophysiology of atherosclerosis [48]. Mediated by endothelial dysfunction, circulating lipoprotein particles enter the artery wall, facilitating the recruitment of monocytes/macrophages and the formation of atherogenic foam cells, ultimately triggering a series of complex pathogenic processes to promote plaque formation [48].

3.1. Attenuation of Endothelial Pro-Inflammatory Activation

Triggered by various cardiovascular risk factors, endothelial cells undergo morphological and functional modifications, termed endothelial activation, manifesting increased expression of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), E-Selectin, and chemokines/pro-inflammatory cytokines such as interleukin-8 (IL-8), IL-6, tumor necrosis factor-α (TNF-α), and IL-1β. The results of endothelial activation are increased leukocyte adhesion and infiltration into the vascular wall leading to the propagation and development of vascular inflammation [48].
Nuclear factor κB (NF-κB) is a master transcription factor responsible for inflammatory responses [49]. Previous studies have demonstrated that NF-κB inactivation is an essential mechanism of the anti-endothelial activation effect of natural monoterpenes (Table 2). Peroxisome proliferator-activated receptor Gama (PPARγ) can attenuate inflammatory responses in the cardiovascular system, including endothelial cells. Growing evidence suggests that PPARγ is an upstream regulator of NF-κB in the anti-inflammatory process [50,51]. Eucalyptol is a monoterpene found naturally in many aromatic plants with anti-inflammatory effects. Pre-treatment with eucalyptol has been reported to suppress the expression of VCAM-1, E-selectin, IL-8, and IL-6 in lipopolysaccharide (LPS)-induced human umbilical vein endothelial cells (HUVECs), and this was achieved by blockade of NF-κB signaling. Importantly, by using PPARγ inhibitor or PPARγ gene silencing, LPS-induced activation of NF-κB and expression of inflammatory mediators in HUVECs could be reversed, suggesting that modulation of the PPARγ/NF-κB axis contributes to eucalyptol-mediated suppression of endothelial pro-inflammatory activation [52]. Similarly, several studies have shown that monoterpenes, including citral, citronellol, and genipin, can inhibit adhesion molecule expression in HUVECs and neutrophil/monocyte–endothelial cell adhesion by regulating the PPARγ-dependent NF-κB signaling pathway [53,54,55]. High mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP), secreted from endothelial cells and leukocytes that mediate inflammation, and correlates with the severity of atherosclerosis [56]. Paeoniflorin and cornuside, two natural monoterpenoids used in traditional oriental herbal medicine, were found to suppress the expression and release of HMGB1 in LPS- or lysophosphatidylcholine (LPC)-stimulated HUVECs, paralleling with reduced expression of endothelial cell-derived adhesion molecules and inflammatory factors [57,58]. The underlying mechanism may involve the induction of sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide–dependent protein deacetylase, which plays an important role in deacetylation of HMGB1 and thus inhibits HMGB1 release and subsequent NF-κB activation in HUVECs [57]. Additionally, reactive oxygen species (ROS) have been reported to induce the activation of NF-κB to promote the adhesiveness of endothelial cells [59]. Using a high glucose-induced HUVECs model, Wang et al. showed that geniposide exhibited a beneficial role in normalizing endothelial pro-inflammatory activation by inhibiting ROS overproduction, NF-κB activation, and monocyte–endothelial cell adhesion [60]. Recently, molecular docking analysis combined with in vitro cell culture-based approaches showed that amarogentin isolated from Gentianaceace plants could directly interact with AMPK to block NF-κB-mediated endothelial inflammation, indicating the pivotal role of AMPK/NF-κB in the protective effect of amarogentin on endothelial activation [26]. Notably, many monoterpenes with NF-κB inhibitory effects, such as cornuside, paeoniflorin, and geniposide, can attenuate endothelial activation through mitogen-activated protein kinase (MAPK) signaling (p38 MAPK, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK)), another major signaling pathway driving inflammatory response [57,61,62].
Endoplasmic reticulum (ER) stress, autophagy, and inflammation are tightly integrated biological processes in atherosclerosis’s pathogenesis [79]. ER homeostasis is crucial in determining cell survival or death based on the cellular stress factors present. When stimulated by pathological insults, intracellular unfolded protein responses can be induced to protect ER homeostasis. However, excessive and prolonged stimuli disturbing ER homeostasis can cause persistent ER stress responses, triggering inflammatory cascades and cell death events [79]. Thus, ER stress is acknowledged as a danger signal for inflammation. Paeoniflorin extracted from the traditional Chinese herb Paeonia lactiflora was reported to be able to modulate ER stress during endothelial pro-inflammatory activation [65]. Pre-treatment with paeoniflorin significantly reduced the expression of ER stress markers glucose regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), and spliced X-box binding protein-1 (XBP-1), as well as improved ultrastructural abnormalities of the ER. These events ultimately contributed to the attenuation of NF-κB-dependent production of inflammatory mediators in LPS-induced HUVECs [65]. Similarly, catalpol, another natural monoterpene, could also alleviate endothelial inflammation, at least in part, by inhibiting ER stress [66]. Autophagy is an evolutionarily conserved lysosomal catabolic process engaged in degrading dysfunctional or surplus protein aggregates and organelles to maintain cellular homeostasis. Nevertheless, aberrant autophagy causes detrimental effects on cellular homeostasis and thus leads to the induction of inflammation and facilitates the pathophysiology of atherosclerosis [79]. In addition to regulating ER stress, paeoniflorin was found to promote autophagy and attenuate endothelial pro-inflammatory activation through a SIRT1-dependent mechanism [64].
Other monoterpenes [63,67,68,69,70,71,72,73,74,75,76,77,78] that attenuate endothelial pro-inflammatory activation are shown in Table 2.

3.2. Inhibition of Endothelial Oxidative Stress

Oxidative stress refers to an imbalance favoring the production of ROS over intrinsic antioxidant mechanisms, which leads to extensive cellular and molecular damage. Endothelial cells are susceptible to risk factors of oxidative stress, such as oxidized lipids, homocysteine, angiotensin II, hyperglycemia, and inflammatory mediators. These risk factors result in the overproduction of ROS followed by endothelial cell damage, dysfunction, pro-inflammatory activation, and apoptosis, eventually promoting the development of atherosclerosis [80]. Accordingly, targeting endothelial oxidative stress by attenuating ROS overproduction and/or improving intracellular antioxidant activity would be beneficial for protecting against endothelial oxidative injury and atherosclerosis.
Studies have demonstrated that many monoterpenes can alleviate endothelial oxidative stress by either inhibiting pro-oxidant enzymes or by enhancing antioxidant enzymes to maintain cellular redox balance. For example, geraniol, paeoniflorin and harpagoside could suppress endothelial ROS production through the downregulation of NADPH oxidases (NOXs) and cyclooxygenase (COX), which are the predominant sources of ROS in the vasculature [81,82,83]. Other monoterpenes, such as thymoquinone, perillaldehyde, citronellal, geniposide, and monotropein, mitigate endothelial oxidative stress by strengthening the activities of antioxidant enzymes, including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) [67,77,84,85,86]. Many monoterpenes exert anti-oxidative stress effects via attenuating pro-oxidant pathways and inducing antioxidant mechanisms (Table 3).
The nuclear factor E2-related factor 2 (Nrf2) is a central transcription factor responsible for intracellular redox homeostasis [80]. Under basal conditions, Nrf2 binds to Kelch ECH associating protein 1 (Keap1) in the cytoplasm, promoting Nrf2 ubiquitination and degradation. Upon oxidative stress stimulation, Nrf2 dissociates from Keap1 and translocates into the nucleus, thereby inducing the expressions of phase II detoxifying enzymes and antioxidant enzymes for cellular defense [80]. A recent study demonstrated that paeoniflorin, a traditional Chinese herbal substance belonging to monoterpenoid, suppressed mitochondrial ROS production and restored mitochondrial functional damage in tert-butyl hydroperoxide (TBHP)-induced HUVECs [90]. Furthermore, a mechanistic study indicated that paeoniflorin directly interacted with cytoplasm Nrf2, resulting in the nuclear translocation of Nrf2 and activation of Nrf2-mediated antioxidant signaling, thus relieving TBHP-induced endothelial oxidative stress [90]. Another traditional Chinese herbal component with atheroprotective effects, geniposide, was found to attenuate H2O2-induced endothelial oxidative stress by activating the Nrf2 antioxidant pathway. Different from the paeoniflorin mentioned above, geniposide stimulated Nrf2 nuclear translocation by modulating AMPK/mechanistic target of rapamycin (mTOR) signaling [68]. Similarly, monoterpenes, such as eucalyptol, geraniol, and catalpol, have also been shown to have anti-oxidative stress effects via Nrf2 activation [42,87,92]. miR-21 is an endogenous miRNA implicated in various pathophysiological processes, including cardiovascular disorders [99]. Accumulating evidence suggests that the miR-21/phosphatase and tensin homolog (PTEN) pathway regulates cell survival and death in the cardiovascular system, including endothelial cells, which makes miR-21/PTEN a therapeutic target for cardiovascular disease [100,101]. Zhou et al. reported that pre-treatment with geniposide decreased ox-LDL-induced oxidative stress in HUVECs by reducing NOX2 expression and by upregulating antioxidant enzyme activities [67]. Using gain- and loss-of-function approaches, the investigators further revealed that geniposide could modulate the miR-21/PTEN pathway to restore the balance between intracellular oxidant and antioxidant states, thus preventing ox-LDL-induced endothelial oxidative injury [67]. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a well-known receptor for ox-LDL that plays a vital role in atherosclerosis. Homocysteine, a risk factor of atherosclerosis, has been reported to induce the expression of LOX-1 in endothelial cells and to promote ROS generation and oxidative injury [102,103]. Picroside II, a monoterpenoid isolated from traditional Chinese medicine Picrorhiza scrophulariiflora, was reported to counteract homocysteine-induced LOX-1 expression in a SIRT1-dependent manner and thus ameliorated HUVEC oxidative stress [76]. In addition, catalpol and monotropein, two naturally occurring monoterpenoids, were found to attenuate endothelial oxidative stress, at least in part, by inhibiting NF-κB activation, suggesting inflammation as a target of these compounds to regulate oxidative stress [66,77].
Other monoterpenes [89,91,95,97,98] that inhibit endothelial oxidative stress are listed in Table 3.

3.3. Modulation of Nitric Oxide (NO) Pathway

Endothelium-derived NO plays essential roles in normal endothelial functions. NO is a multifunctional signaling molecule controlling cardiovascular homeostasis, including regulating vasomotor tone, modulation of platelet activation and leukocyte adhesion, and manipulating local cell growth [104]. All established risk factors for atherosclerosis, such as hyperlipidemia, diabetes mellitus, hypertension, and smoking, are found to be associated with diminished NO production [105]. Emerging evidence has also indicated that NO bioavailability dysfunction is implicated in the initiation and development of atherosclerosis [105]. Therefore, improvement of endothelial NO production provides a potential strategy for preventing and managing atherosclerosis.
Abundant evidence has demonstrated the implication of modulating the NO pathway in the effect of natural monoterpenes on endothelial dysfunction and atherosclerosis. NO is synthesized by nitric oxide synthase (NOS) using L-arginine as the substrate [104]. NOS includes different isoforms that play contrasting roles in atherosclerosis, with endothelial NOS (eNOS) being atheroprotective and inducible NOS (iNOS) being pro-atherogenic [105]. Thus, modulating NO by adjusting the eNOS/iNOS ratio is crucial for protecting against endothelial dysfunction and atherosclerosis. Catalpol, a monoterpenoid extracted from the root of the traditional Chinese herb Rehmanniae radix, could maintain the balance of endothelial NO by inhibiting the NF-κB/iNOS pathway and by activating the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/eNOS pathway. As a result, catalpol improved cell viability, enhanced endothelial integrity, and reduced inflammatory response in advanced glycation end-product (AGE)-treated endothelial cells [94]. Furthermore, two other monoterpenoids, paeoniflorin and eucalyptol, can upregulate SIRT1 expression to regulate the eNOS/iNOS ratio and to protect against endothelial dysfunction [88,93]. Notably, the activation of eNOS by these monoterpenoids depends on either eNOS Ser1177 phosphorylation [94] or upregulation of eNOS expression [88,93]. Oxidative stress plays a crucial role in determining NO bioavailability [104]. During endothelial dysfunction, oxidative stress mediates ONOO formation and oxidizes eNOS cofactor tetrahydrobiopterin (BH4), causing BH4 deficiency and eNOS uncoupling. Ultimately, it leads to increased ROS generation and reduced NO bioavailability [104]. Perillaldehyde, a major component in essential oil isolated from Perilla frutescens that has been used in traditional Chinese medicine, was reported to prevent endothelial dysfunction and to attenuate the growth of atherosclerosis [85]. Mechanically, perillaldehyde alleviated ROS-mediated oxidative stress and rescued BH4 deficiency, thereby promoting eNOS recoupling and improving endothelial dysfunction [85]. Other monoterpenoids, such as citronellal and aucubin, similarly induced eNOS recoupling and ameliorated vascular endothelial injury through an anti-oxidative mechanism [91,98]. Monoterpenes that modulate the endothelial NO pathway are shown in Table 3.

3.4. Attenuation of Endothelial Apoptosis

Apoptosis is a well-known programmed cell death pathway involved in various physiological and pathological processes. Two molecular pathways are acknowledged to regulate apoptosis: mitochondrial-dependent intrinsic pathway and death receptor-mediated extrinsic pathway. The mitochondrial-dependent apoptosis pathway is induced by cellular damage or stress, which upregulates B cell leukemia/lymphoma 2 (BCL2)-associated agonist of cell death (Bad) and facilitates the insertion of BCL2-associated X protein (Bax) into the mitochondrial outer membrane. This further causes cytochrome c to be released into the cytoplasm, followed by interaction between cytochrome c and apoptotic peptidase activating factor 1 (Apaf-1) to induce apoptosis through caspase activation [106]. The extrinsic apoptotic pathway is initiated by binding extracellular death ligands (such as TNF-α) to their respective cell-surface death receptors. The activation of the death receptors results in the formation of the death-inducing signaling complex mediated by an adaptor protein, thereby triggering caspase activation and the apoptosis process [107]. While apoptosis serves as a fundamental mechanism for physiological homeostasis, uncontrolled apoptosis may lead to cellular dysfunction and death. It has been observed that increased endothelial apoptosis is closely associated with atherosclerosis, and pro-atherosclerotic factors such as ox-LDL, oxidative stress, and low shear stress have been shown to induce vascular endothelial cell apoptosis [106,108]. Therefore, inhibition of endothelial apoptosis may represent a promising strategy to cope with atherosclerosis.
Accumulating evidence demonstrates the reduction of endothelial apoptosis as one of the mechanisms by which natural monoterpenes protect against endothelial injury and atherosclerosis. The anti-apoptotic effect of monoterpenes involves the modulation of intrinsic and/or extrinsic apoptotic pathways in endothelial cells. For instance, the beneficial impact of geniposide against atherosclerosis and endothelial dysfunction was related to the attenuation of endothelial apoptosis due to upregulation of anti-apoptotic protein Bcl-2, downregulation of pro-apoptotic protein Bax and caspase-3, and maintaining mitochondrial membrane potential [67,68]. Similarly, many other monoterpenes have also been shown to ameliorate endothelial cell apoptosis by modulating the Bcl-2/Bax ratio, caspase-9, caspase-3, the release of cytochrome c, and mitochondrial function [64,66,77,78,83,90,97,109]. All this evidence suggests a mitochondrial-dependent intrinsic pathway as a target in the effect of these monoterpenes. Furthermore, the extrinsic apoptotic pathway is likely implicated in the anti-endothelial apoptotic effect of monoterpenes. Monotropein, an active monoterpenoid isolated from the roots of Morinda officinalis, was reported to suppress the phosphorylation of NF-κB and activating protein-1 (AP-1), which further inhibited the expression of pro-inflammatory cytokine TNF-α and reduced cell apoptosis in H2O2-induced HUVECs. These studies indicated that the protective effect of monotropein on endothelial cells might involve the regulation of the TNF-mediated mitochondrial-independent apoptotic pathway [77]. Similarly, picroside II, the main active constituent of Picrorhiza scrophulariiflora belonging to monoterpenoid, was found to decrease the caspase-3 activity and the cleaved caspase-3 protein level to inhibit apoptosis in homocysteine-treated HUVECs, which might also be related to the attenuation of TNF-α production [76]. In addition, another study showed that harpagoside, a monoterpenoid extracted from the traditional Chinese herb Scrophulariae Radix, prevented angiotensin II (Ang II)-induced endothelial apoptosis via inactivation of caspase-8/caspase-9/caspase-3, suggesting that harpagoside could exert an anti-apoptosis effect by targeting both intrinsic and extrinsic apoptotic pathways [83]. Monoterpenes that attenuate endothelial apoptosis are listed in Table 4. Notably, various signaling pathways such as PI3K/AKT, SIRT1, Nrf2, NF-κB, MAPK, miR-21/PTEN, and AMPK were involved in the anti-endothelial apoptosis effect of these compounds, most of which are also related with oxidative stress and inflammation (Table 4).

4. Natural Monoterpenes Potentially Protect against Atherosclerosis by Targeting Macrophages

Macrophages, the major immune cells in atherosclerotic plaque, are fundamental contributors to the pathophysiology of atherosclerosis. After lipoproteins enter the artery wall, they elicit complex pathological processes involving macrophages, such as monocyte recruitment, macrophage differentiation, engulfment of modified lipoproteins, foam cell formation, and initiation of inflammatory responses, which instigate the progression of atherosclerosis [110]. Therefore, macrophages have become an attractive target for developing therapeutic agents against atherosclerosis.

4.1. Reduction of Macrophage-Related Inflammation

Atherosclerosis is a chronic inflammatory disease of the arterial wall where non-resolving inflammation acts as a critical underlying driver in all stages of the disease from its onset to its progression, eventually leading to intimal plaque rupture and atherothrombosis giving rise to myocardial infarction or stroke [6,7]. Large amounts of pro-inflammatory cytokines, chemokines, ROS, and NO are produced by inflammatory macrophages that enter the vascular intima, which drives vascular inflammation in atherosclerosis. Thus, anti-inflammation is considered one of the strategies for dealing with atherosclerosis [111].
Studies have been conducted to evaluate the anti-inflammation property of natural monoterpenes in macrophages to determine their potential for atherosclerosis management. Both NF-κB and MAPK inflammatory signaling pathways function as vital nodal points in regulating atherosclerosis, which drive the expression of a large panel of pro-inflammatory genes [112]. As shown in Table 5, most monoterpenes are potent macrophage NF-κB and MAPK inhibitors with concomitant suppression of inflammatory mediators (e.g., TNF-α, IL-1β, IL-6, CC, and CXC chemokines), suggesting that NF-κB and MAPK inactivation is the major mechanism for their anti-inflammatory and potential anti-atherosclerotic effects. For example, geniposide was reported to significantly inhibit the expression of toll-like receptor (TLR)-4 and phosphorylation of NF-κB p65, p38 MAPK, JNK, and ERK in macrophages, paralleling with an attenuated systemic inflammation as shown by lower serum levels of TNF-α, IL-6 and IL-8 in HFD-fed apolipoprotein E (ApoE)−/− mice, which suppressed the development of atherosclerosis [68,113,114]. Transcription factors SIRT1 [115,116,117,118], PPARγ [119,120], and Nrf2 [121,122,123,124,125,126], together with intracellular signaling modulators dual specificity phosphatase 1 (MKP-1) [113] and suppressor of cytokine signaling 3 (SOCS3) [127], were proposed to mediate the inhibitory effect of monoterpenes on NF-κB and/or MAPK activation.
Inflammasome is an integral component of the innate immune system that responds to pathogens and cellular stress. One of the most widely studied inflammasomes induced upon macrophage activation consists of the sensor NLR family PYRIN domain containing-3 (NLRP3), the adaptor apoptosis-associated speck-like protein containing CARD (ASC), and the effector enzyme caspase-1 [170]. It has been established that activation of the NLRP3 inflammasome requires NF-κB-mediated priming and subsequent inflammasome assembly steps [170]. Upon stimulation, the interaction between NLRP3 and ASC together with the formation of ASC oligomer provides a platform for pro-caspase-1 autocleavage and activation [170]. Subsequently, it triggers the production and release of mature IL-1β and IL-18 [170]. Experimental evidence has shown that NLRP3 inflammasome activation is the major driver of atherosclerosis [171,172,173]. Thus, modulating NLRP3 inflammasome may be beneficial for managing atherosclerosis. Carvacrol, a monoterpene commonly found in the essential oils of the Laminaceae family, was proven to inhibit cytokine secretion and NLRP3 inflammasome activation [136]. Treatment with carvacrol-containing active fractions significantly blunted protein interaction between NLRP3 and ASC, suppressed ASC oligomerization, abolished ATP-induced K+ efflux, and inhibited the release of caspase-1, IL-1β, and IL-18 in LPS-primed macrophages [136]. Furthermore, this carvacrol-containing extract attenuated LPS-induced NF-κB nuclear translocation. Therefore, these observations indicated that carvacrol inhibits NLRP3 inflammasome activation by interfering in both the priming and inflammasome assembly steps in activated macrophages [136]. Mitochondrial dysfunction plays a critical role in the NLRP3 inflammasome assembly process, characterized by the generation of mitochondrial ROS, a decrease in mitochondrial membrane potential, and the release of mitochondrial DNA [170]. Loganin, a major bioactive monoterpenoid derived from the traditional Chinese herb Cornus officinalis, was found to prevent mitochondrial stress as shown by decreased mitochondrial ROS level, increased mitochondrial membrane potential, and reduced mitochondrial DNA such as D-loop [156]. These molecular events ultimately suppressed monosodium uric acid (MSU) crystal-induced NLRP3 inflammasome activation in macrophages [156]. Moreover, Nrf2-mediated cytoprotective signaling pathway regulating intracellular redox homeostasis was found to mediate the inhibition of NLRP3 inflammasome activation upon treatment of monoterpenoids, such as loganin and oleocanthal [122,123,156]. Among others, gentiopicroside, a monoterpenoid separated from a traditional medicine Gentiana macrophylla, was reported to inhibit NLRP3 inflammasome activation by suppressing both mRNA and protein expression of NLRP3 inflammasome key components NLRP3, ASC, and caspase-1 in LPS-MSU-induced RAW264.7 macrophages [160]. An inhibitory effect on NLRP3 inflammasome activation has also been reported for other monoterpenes [115,129,135,144,147,159,166].
Monoterpenes that decrease macrophage-related inflammation are shown in Table 5.

4.2. Inhibition of Foam Cell Formation

The formation of foam cells in the arterial intima is a hallmark of atherosclerosis. During atherogenesis, circulating monocytes infiltrate into the intima and differentiate into macrophages. Mediated by relevant cell surface receptors, macrophages engorge modified lipoproteins, causing accumulation of cholesteryl ester and free cholesterol in the cells, leading to foam cell formation [110,174]. The resulting cholesterol-laden foam cells can further trigger the release of DAMPs and inflammatory responses, thereby promoting the development of atherosclerosis [174]. Accordingly, the macrophage foam cell is recognized as a target for therapeutic intervention of atherosclerosis [175].
Foam cell formation is finely orchestrated by cholesterol uptake and efflux processes. Engulfment of modified lipoproteins by macrophages to induce foam cell formation depends on a family of pattern recognition receptors known as scavenger receptors, including CD36, SR-A, and LOX-1 [176]. Some monoterpenoids have been shown to target macrophage scavenger receptors to inhibit foam cell formation. Using ox-LDL-treated human macrophages, it was found that oleacein, a monoterpenoid found primarily in olive fruit and leaves, significantly attenuated ox-LDL-induced foam cell formation by decreasing the expression of CD36, SR-A, and LOX-1 [177]. Notably, macrophage apoptosis, a major contributor to the progression of atherosclerotic plaques, was inhibited by oleacein [177,178]. This observation might also be related to the reduced expression of CD36 and SR-A [177,178]. Another two medicinal monoterpenoids, geniposide and albiflorin, showed similar inhibitory effects on ox-LDL-induced foam cell formation by suppressing CD36 or LOX-1 expression and by inhibiting NF-κB- and MAPK-mediated inflammatory response [179,180].
Apart from cholesterol uptake, foam cell formation is also regulated by cholesterol efflux from macrophages. Diminished efflux of intracellular cholesterol to extracellular receptors facilitates the formation of foam cells. It has been known that macrophage cholesterol efflux is mediated by ATP-binding cassette transporters ABCA1, ABCG1, and SR-BI [176]. Targeting ABCA1, ABCG1, and SR-BI to promote cholesterol efflux represents a potential strategy to inhibit foam cell formation and atherosclerosis. Liver X receptors (LXRs), members of the nuclear receptor superfamily, play a pivotal role in RCT by which accumulated cholesterol in the peripheral tissue is transported to the liver for excretion [181]. As ligand-activated transcription factors, LXRs regulate cellular cholesterol homeostasis by promoting the expression of genes related to cholesterol efflux, such as ABCA1 and ABCG1 [181]. Previous studies reported that eucalyptol, a principal monoterpene in plant-derived essential oils, significantly stimulated the expression of LXRα and LXRβ followed by induction of their target genes ABCA1 and ABCG1 expression in macrophages, which in turn promoted cholesterol efflux and suppressed ox-LDL-induced foam cell formation [182,183]. Eucalyptol decreased the expression of LXRα and lipogenesis-related genes in hepatocytes, resulting in attenuated hepatic lipid accumulation [183]. Given that many synthetic LXR agonists cause hepatic lipogenesis and hypertriglyceridemia, eucalyptol can serve as a potent pharmaceutical agent against atherosclerosis by activating LXRs in macrophages without inducing the side effects of hepatic lipogenesis and steatosis [183]. Moreover, a recent study showed that geniposide attenuated foam cell formation and atherosclerosis by inhibiting lipid uptake and by promoting cholesterol efflux [184]. Mechanistically, it was found that this natural monoterpenoid could reduce the expression of SR-A. At the same time, it promotes the expression of ABCA1 and SR-BI in both lysophosphatidic acid (LPA)-induced macrophages and HFD-fed ApoE−/− mice, leading to attenuated foam cell formation and atherosclerosis [184]. Notably, modulation of SR-A, ABCA1, and SR-BI by geniposide was proposed to be mediated by p38 MAPK and AKT signaling pathways [184].
Monoterpenes that inhibit foam cell formation have been listed in Table 6.

4.3. Induction of Macrophage Autophagy

Autophagy is an intracellular lysosomal degradation process that plays a housekeeping role in clearing dysfunctional proteins, damaged organelles, and intracellular pathogens. Key factors involved in autophagy regulation have been identified, including mTOR, AMPK, and Beclin1 [195]. As the central players in the pathogenesis of atherosclerosis, macrophages have been demonstrated to undergo a progressive autophagy dysfunction in the developing atherosclerotic lesion, resulting in hyperactivation of inflammation, increase in cell death, and reductions in lipophagy and cholesterol efflux [196]. Growing evidence suggests that induction of autophagy in macrophages can exert an atheroprotective effect by eliminating long-lived/damaged intracellular material, attenuating excessive inflammation and cell death, promoting macrophage efferocytosis, and accelerating cholesterol efflux from foam cells, eventually inhibiting plaque development and instability [195]. Thus, activation of macrophage autophagy is expected to be beneficial for preventing atherosclerosis [195].
Geniposide, a bioactive monoterpenoid isolated from Gardenia jasminoides, has been reported to be atheroprotective, as shown by improved serum lipid profile, decreased aortic lipid deposition, reduced macrophage-mediated inflammation, attenuated apoptosis, and smaller size of atherosclerotic plaques [68,184]. A recent study provided evidence that geniposide could inhibit the progression of atherosclerosis by promoting macrophage autophagy [188]. Mechanistic studies further indicated that geniposide inhibited the expression of triggering receptor expressed on myeloid cells 2 (TREM2) and the phosphorylation of mTOR, which in turn enhanced macrophage autophagy [188]. These results suggest TREM2/mTOR signaling as an intracellular target in the protective effect of geniposide on macrophage autophagy and atherosclerosis [188]. Paeoniflorin, a monoterpenoid in the traditional Chinese herb Paeonia lactiflora, was previously reported to promote autophagy in ox-LDL-induced HUVECs, as demonstrated by increased microtubule-associated protein 1 light chain 3-II (LC3-II)/LC3-I ratio and decreased p62 protein level [64]. Consistently, an increased autophagic response was also observed in IL-4-induced pro-fibrotic macrophages in the presence of paeoniflorin-containing compound combination, which was mediated by attenuation of AKT/mTOR signaling [197]. However, whether paeoniflorin can regulate macrophage autophagy under atherogenic conditions and exert an atheroprotective effect remains elusive and is worth further investigation.
Prevention of defective macrophage autophagy in plaques is considered a novel strategy to protect against atherosclerosis [196]. Although emerging data reveal that natural products are promising in stimulating autophagy [195], our understanding of the role of monoterpenes in the regulation of macrophage autophagy in atherosclerosis is somewhat limited, and it thus requires further studies. Since autophagy is a double-edged sword in cardiovascular disease [79], more selective and precise manipulation of autophagy is also needed to achieve better therapeutic outcomes.

4.4. Enhancement of M2 Macrophage Polarization

As important players in the innate immune system, macrophages are characterized by remarkable phenotypic and functional plasticity. Based on the dynamic microenvironmental stimuli in the atherosclerotic plaque, macrophages can be polarized into subsets with distinct functional properties, which include two well-known macrophage phenotypes: classically activated (M1) macrophages and alternatively activated (M2) macrophages [198]. M1 macrophages produce a variety of inflammatory mediators, which exacerbate the development of atherosclerosis [199]. In contrast, M2 macrophages exert a protective effect on atherosclerosis by releasing anti-inflammatory factors and promoting tissue reparative processes to inhibit plaque growth and enhance plaque stability [198,199]. Thus, modification of plaque macrophage polarization by enhancing M2 macrophage phenotype is a potential therapeutic strategy to ameliorate atherosclerosis.
Several studies have shown natural monoterpenes’ capacity to promote M2 macrophage polarization. Signal transducer and activator of transcription 3 (STAT3) and STAT6 are recognized as key signaling molecules responsible for M2 macrophage polarization, which are mediated by IL-4/13 and IL-10 signaling, respectively [200]. Oleacein, a monoterpenoid in extra-virgin olive oil, was reported to upregulate the expression of CD163 (a biomarker of M2-type macrophages) and increase the phosphorylation of Janus kinase (JAK1)/JAK2 and STAT3 in ox-LDL-induced human macrophages, suggesting that oleacein promotes the polarization of anti-inflammatory M2-type macrophages via the JAK/STAT3 pathway [177,194]. An ex vivo study using human carotid plaques obtained from patients with transient ischemic attacks revealed that oleacein could suppress plaque instability by attenuating related biomarkers such as HMGB1 and matrix metalloproteinase-9 (MMP-9) [201]. Therefore, it is reasoned that oleacein-mediated M2 macrophage polarization contributed to the protective effect of oleacein on atherosclerotic plaque vulnerability. Compound edaravone injection is an approved drug for acute ischemic stroke that consists of edaravone and borneol (a monoterpene) [186]. Pharmacological studies revealed that although borneol alone showed no apparent effect on modulating macrophage phenotype, it could reinforce the efficacy of edaravone in promoting M2 macrophage polarization, as indicated by increased expression of CD206, arginase-1 (Arg-1) and IL-10 [186]. Mechanistically, this pharmacological action of borneol was related to the activation of the JAK/STAT3 signaling pathway [186]. However, other monoterpenoids, such as genipin and paeoniflorin, were shown to enhance M2 macrophage polarization by activating STAT6 [189,191]. Estrogen deficiency is a known risk factor for atherosclerosis in postmenopausal women [190]. Catalpol, a monoterpenoid in the traditional Chinese herb Rehmanniae radix, was demonstrated to attenuate atherosclerotic lesions in a mouse model of postmenopausal atherosclerosis by accelerating M2 macrophage polarization [190]. The underlying mechanism was related to the molecular interaction between catalpol and the estrogen receptor (ER) leading to the activation of ERα, suggesting that catalpol is a potent therapeutic agent against postmenopausal atherosclerosis [190]. Monoterpenes involved in promoting M2 macrophage polarization are shown in Table 6.
Notably, other macrophage populations, in addition to M1/M2 macrophages, have been indicated in atherosclerotic plaque [202,203]. For example, M2-type macrophages are reported to be divided into M2a, M2b, M2c, and M2d subsets with distinct functions in atherosclerosis [203]. However, there is still a significant knowledge gap regarding macrophage heterogeneity and their roles in atherosclerosis, as well as the effects of monoterpenes on these macrophage sub-populations, which warrant further investigations.

5. Natural Monoterpenes Potentially Protect against Atherosclerosis by Targeting Vascular Smooth Muscle Cells

Vascular smooth muscle cell (VSMC) is a constitutive part of the blood vessel and plays essential roles in regulating arterial physiology and pathology. Under physiological conditions, VSMCs are characterized by the expression of a range of genes required for their differentiated and contractile phenotype [204]. In response to vascular injury or atherosclerosis, however, VSMCs undergo phenotypic switching, which refers to the de-differentiation of the contractile phenotype to an alternative non-contractile VSMC phenotype [204]. These phenotypically modulated VSMCs typically exhibit reduced expression of contractile markers and increased capacity for cell proliferation, migration, and secretion, which are critically involved in the pathological process of atherogenesis [205,206]. Within the atherosclerotic plaque, the modulated VSMCs can adopt a variety of phenotypes with different effects on atherosclerosis, some of which have been considered to be detrimental such as macrophage-like and foam cell-like VSMCs [205,206]. Remarkably, many human genomic loci identified to be associated with the risk of coronary artery disease are related to VSMC phenotypic modulation, suggesting that targeting the phenotypic transition of VSMCs may help to attenuate plaque burden and promote plaque stability [206].
Enhanced proliferation and migration are hallmarks of VSMC phenotypic switching that underlies many vascular diseases. A number of natural monoterpenes have been proven to inhibit abnormal proliferation and migration of VSMCs, indicating their potential pharmacological application to treat vascular disorders. The MAPK family, including p38 MAPK, JNK, and ERK is a class of serine/threonine protein kinases essential for cell proliferation and migration [207]. As shown in Table 7, most monoterpenes could target MAPK signaling to suppress aberrant VSMC proliferation and migration. For instance, linalool, a monoterpene in many aromatic medicinal plants, inhibited Ang II-induced VSMC proliferation and migration by suppressing cholinergic receptor muscarinic 3 (CHRM3)-dependent activation of p38 MAPK, JNK, and ERK signaling [208]. However, many other monoterpenes selectively inhibit MAPKs in stimulated VSMCs. Carvacrol, a monoterpene found in plant essential oils and commonly used as a food additive and flavoring agent, was shown to inhibit p38 MAPK and ERK activation, thereby attenuating platelet-derived growth factor-BB (PDGF-BB)-induced VSMC proliferation and migration [209]. Furthermore, carvacrol was observed to decrease ROS generation [209] markedly. Since ROS has been reported to promote VSMC proliferation and migration through MAPK signaling [210], these observations indicated that the anti-proliferative and anti-migratory effects of carvacrol on PDGF-BB-induced VSMCs were achieved by the regulation of the ROS-mediated p38 MAPK/ERK pathway. Consistently, thymoquinone, a monoterpene derived from the medicinal plant Nigella sativa, could activate the AMPK/PPARγ/PGC-1α pathway to downregulate ROS production and further attenuate p38 MAPK-mediated VSMC proliferation and migration [211,212]. Similarly, the monoterpenoid paeoniflorin was also reported to suppress ox-LDL- and PDGF-BB-induced proliferation and migration of VSMCs through ROS-mediated p38 MAPK and ERK signaling pathways [192,213]. Notably, the reduction of ROS by paeoniflorin was related to the increased expression of heme oxygenase-1 (HO-1), which is an essential cytoprotective gene regulating intracellular redox homeostasis [192]. In addition to MAPKs, AKT signaling is a classic pathway involved in cell proliferation and migration. The monoterpenoid, genipin, was shown to target not only HO-1/ERK but also AKT signaling to inhibit TNF-α-induced VSMC proliferation and migration [214]. Other cell proliferation and migration-related signaling molecules, such as phospholipase C-γ1 (PLC-γ1) and STAT3, were also reported to be targeted by monoterpenoids hinokitiol [215] and plumericin [216]. Monoterpenes that regulate VSMC proliferation and migration are listed in Table 7.
Although the above-mentioned monoterpenes can inhibit abnormal VSMC proliferation and migration, whether these compounds can achieve an atheroprotective effect through modulating VSMC phenotypic switching is unclear. VSMC phenotypic switching is a rather complex process in atherosclerosis. Depending on various environmental cues, VSMCs exhibit different phenotypes that may have either beneficial or maladaptive effects on atherosclerosis. However, the exact role and the regulatory mechanism of each VSMC phenotype in atherogenesis remain largely elusive. Therefore, much work remains to be conducted regarding VSMC phenotypic switching in order to perform a better pharmacological intervention to stimulate atheroprotective VSMC phenotypes and to eliminate detrimental VSMC phenotypes.

6. Natural Monoterpenes Potentially Protect against Atherosclerosis by Targeting Dendritic Cells

Atherosclerosis is characterized by chronic arterial inflammation involving innate and adaptive immune responses. Dendritic cells, a class of hematopoietic cells, function as the necessary connection between innate and adaptive immunity. Throughout atherogenesis, an increased accumulation of vascular dendritic cells has been demonstrated in atherosclerotic lesions, indicating their involvement in the development of atherosclerosis [223]. It has been shown that dendritic cells are highly heterogenous with multifaced roles in the pathophysiology of atherosclerosis, which include lipid uptake, efferocytosis, antigen presentation, T cell activation, and the production of cytokines/chemokines [224,225]. Targeting dendritic cells based on their immunoregulation property has provided encouraging results in attenuating experimental atherosclerosis, indicating the promise of its clinical application for atherosclerosis treatment [223].
Oral administration of geniposide, a monoterpenoid isolated from the traditional Chinese herb Gardenia jasminoides, was shown to ameliorate the development of atherosclerosis in HFD-fed ApoE−/− mice [222]. It was also demonstrated that geniposide decreased dendritic cell maturation in the bone marrow and infiltrated the atherosclerotic lesions [222]. As we know, upon maturation, the expression of biomarkers of dendritic cells is increased, such as CD11c, CD83, CD80, and CD86, which regulates antigen presentation and induction of T cell-related adaptive immunity and thus facilitates the development of a pro-atherogenic phenotype [223,225]. Nevertheless, geniposide treatment resulted in decreased expression of CD11c, CD83, CD80, and CD86 in ApoE−/− mice, indicating that geniposide might inhibit dendritic cell maturation and suppress the development of pro-atherogenic effector T cells, thus alleviating atherogenesis [222]. Moreover, dendritic cell-derived IL-12 decreased after geniposide treatment [222], an essential pro-atherogenic cytokine that activates T cells and induces the generation of pro-inflammatory T-helper 1 (Th1) cells [225]. Taken together, the atheroprotective role of geniposide may be attributed to the inhibition of both dendritic cell maturation and pro-inflammatory cytokine production. Similarly, the monoterpenoid paeoniflorin was demonstrated to suppress dendritic cell maturation and subsequent Th17 cell differentiation, which was related to the attenuation of NF-κB and JNK signaling pathways [219]. Another study reported that paeoniflorin could target TGF-β signaling to induce a regulatory dendritic cell subtype characterized by high expression of CD11b/c and low expression of co-stimulatory molecules [220]. This regulatory dendritic cell showed a striking stimulative effect on anti-inflammatory regulatory T (Treg) cells that protect against atherosclerosis, indicating the potential of paeoniflorin as an immunosuppressive and atheroprotective agent [220]. Other monoterpenes, such as carvacrol [217], thymol [217], and thymoquinone [218], exhibit similar inhibitory effects on dendritic cell maturation and function (Table 7).

7. Conclusions and Future Perspectives

Monoterpenes, a large class of naturally occurring compounds in many aromatic and medicinal plants, have drawn increasing attention due to their therapeutic potential in cardiovascular disease management. Herein, we reviewed published studies concerning natural monoterpenes that have potential atheroprotective properties and highlighted their underlying mechanisms at the cellular and molecular levels. In summary, the cellular targets of atheroprotective monoterpenes are diverse. Monoterpenes regulate the biological functions of endothelial cells, monocytes/macrophages, VSMCs, and dendritic cells through various pharmacological mechanisms, including serum lipid-lowering properties, anti-inflammatory effects, anti-oxidative effects, anti-apoptosis effects, inhibition of foam cell formation, suppression of ER stress, activation of autophagy, and attenuation of VSMC proliferation and migration (Figure 2). Moreover, molecular targets mediating the atheroprotective effect of monoterpenes mentioned in this review include SIRT1, PPARγ, Nrf2, STAT, NF-κB, MAPK, PI3K/AKT, AMPK, and mTOR (Figure 3).
Despite significant research progress on the effects and mechanisms of natural monoterpenes on atherosclerosis, developing novel therapeutic agents from these compounds as effective therapies for atherosclerosis requires more detailed investigations. First, many monoterpenes demonstrate low bioavailability upon oral administration, which significantly dampens their pharmacological actions in vivo. Therefore, it is imperative to develop alternative strategies for drug delivery to improve their bioavailability and pharmacokinetic properties. Currently, nanotechnology and structural modification techniques have emerged as promising approaches to facilitate in vivo drug delivery. Remarkably, preclinical studies using nanostructured lipid carriers for eucalyptol, thymoquinone, and oleuropein, three monoterpenoids with poor bioavailability due to low aqueous solubility and stability, have shown encouraging results with significantly improved bioavailability and efficacy in vivo [92,226,227]. In addition, structural alterations of swertiamarin, a monoterpenoid with a low plasma half-life, resulted in two new derivatives of swertiamarin with enhanced stability in the circulation and thus may accelerate its bioavailability [228]. However, these approaches that aim to improve the bioavailability of monoterpenes should be further tested on human subjects with atherosclerosis to evaluate their antiatherogenic efficacy. Second, although a number of monoterpenes have been demonstrated to be atheroprotective, few studies mention adverse effects or toxicity. Therefore, comprehensive toxicity studies should be carried out to investigate the safety of these compounds for drug development. Concurrently, given that there are not many data available on the pharmacokinetics of monoterpenes, it is necessary to perform comprehensive absorption–distribution–metabolism–excretion investigations in vivo to assess the effectiveness and safety of candidate monoterpenes. Third, while a large proportion of pharmacological studies regarding antiatherogenic monoterpenes have been conducted at the levels of experimental animals and cultured cells, further clinical studies are required to examine the therapeutic effects of candidate monoterpenes on atherosclerosis. In this regard, randomized, double-blind, placebo-controlled clinical trials with large samples are warranted for rigorous evaluation of the potency of clinical translation.
Notably, as shown in this review, the potential antiatherogenic effects of most of the monoterpenes may be mediated by multiple mechanisms involving different cell types and molecular pathways, suggesting their pleiotropic or multitargeted pharmacological properties. Since atherosclerosis has been established as a multifaceted vascular disease, the pleiotropic pharmacological actions of monoterpenes may hold great promise for the therapeutic intervention of atherosclerosis by targeting multiple pathological processes in atherogenesis. However, many current data are based on in vitro studies. Thus, more in vivo investigations using animal models of atherosclerosis should be conducted to confirm these in vitro findings. Additionally, it is tempting to expect that the combination of different monoterpenoids or other bioactive herbal ingredients may further improve the efficacy in treating atherosclerosis as a result of their potential synergistic effects. For example, a patented drug combination used for the management of atherosclerosis (patent no. CN201810425740.8) consists of a monoterpenoid geniposide and the other natural compound, notoginsenoside R1 [68]. It has been reported that this drug combination exhibits better efficacy in atherosclerosis alleviation than either geniposide or notoginsenoside R1 alone [68]. Therefore, comprehensive and rigorous analysis of the effectiveness and mechanisms of each monoterpene alone and in combination is of enormous significance and should be strengthened in future studies.

Author Contributions

C.Z. and J.Y. (Jun Yu) conceptualized the manuscript. J.Y. (Jing Yang), C.Z. and J.Y. (Jun Yu) wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (82104180), Jiangxi Provincial Natural Science Foundation (20212BAB216012), Research Fund of Jiangxi University of Chinese Medicine (2018BSZR002), Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling-related Diseases (20202BCD42014), Innovation Funds of the Jiangxi University of Chinese Medicine for Postgraduate Students (JZYC21S62, JZYC22S12), and from a grant from the American Heart Association (18EIA33900065).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Top Discipline of Jiangxi Province & Discipline of Chinese and Western Integrative Medicine (Jiangxi University of Chinese Medicine) for infrastructural and/or scientific support.

Conflicts of Interest

The authors declare that there is no conflict of interest.

References

  1. Herrington, W.; Lacey, B.; Sherliker, P.; Armitage, J.; Lewington, S. Epidemiology of Atherosclerosis and the Potential to Reduce the Global Burden of Atherothrombotic Disease. Circ. Res. 2016, 118, 535–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Barquera, S.; Pedroza-Tobías, A.; Medina, C.; Hernández-Barrera, L.; Bibbins-Domingo, K.; Lozano, R.; Moran, A.E. Global Overview of the Epidemiology of Atherosclerotic Cardiovascular Disease. Arch. Med. Res. 2015, 46, 328–338. [Google Scholar] [CrossRef]
  3. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Prim. 2019, 5, 56. [Google Scholar] [CrossRef]
  4. Tervaert, J.W.C. Cardiovascular disease due to accelerated atherosclerosis in systemic vasculitides. Best Pract. Res. Clin. Rheumatol. 2013, 27, 33–44. [Google Scholar] [CrossRef]
  5. Schaftenaar, F.; Frodermann, V.; Kuiper, J.; Lutgens, E. Atherosclerosis: The interplay between lipids and immune cells. Curr. Opin. Lipidol. 2016, 27, 209–215. [Google Scholar] [CrossRef]
  6. Li, B.; Li, W.; Li, X.; Zhou, H. Inflammation: A Novel Therapeutic Target/Direction in Atherosclerosis. Curr. Pharm. Des. 2017, 23, 1216–1227. [Google Scholar] [CrossRef] [PubMed]
  7. Christ, A.; Bekkering, S.; Latz, E.; Riksen, N.P. Long-term activation of the innate immune system in atherosclerosis. Semin. Immunol. 2016, 28, 384–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Libby, P.; Bornfeldt, K.E.; Tall, A.R. Atherosclerosis: Successes, surprises, and future challenges. Circ. Res. 2016, 118, 531–534. [Google Scholar] [CrossRef] [Green Version]
  9. Shapiro, M.; Fazio, S. From lipids to inflammation: New approaches to reducing atherosclerotic risk. Circ. Res. 2016, 118, 732–749. [Google Scholar] [CrossRef] [PubMed]
  10. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug. Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  11. Liu, Q.; Li, J.; Hartstone-Rose, A.; Wang, J.; Li, J.; Janicki, J.S.; Fan, D. Chinese Herbal Compounds for the Prevention and Treatment of Atherosclerosis: Experimental Evidence and Mechanisms. Evid.-Based Complement. Altern. Med. 2015, 2015, 752610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ai, X.; Yu, P.; Peng, L.; Luo, L.; Liu, J.; Li, S.; Lai, X.; Luan, F.; Meng, X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front. Pharmacol. 2021, 12, 762654. [Google Scholar] [CrossRef] [PubMed]
  13. Silva, B.I.M.; Nascimento, E.A.; Silva, C.J.; Silva, T.G.; Aguiar, J.S. Anticancer activity of monoterpenes: A systematic review. Mol. Biol. Rep. 2021, 48, 5775–5785. [Google Scholar] [CrossRef] [PubMed]
  14. De Cássia da Silveira e Sá, R.; Andrade, L.N.; de Sousa, D.P. A Review on Anti-Inflammatory Activity of Monoterpenes. Molecules 2013, 18, 1227–1254. [Google Scholar] [CrossRef] [PubMed]
  15. Koziol, A.; Stryjewska, A.; Librowski, T.; Sałat, K.; Gawel, M.; Moniczewski, A.; Lochynski, S. An Overview of the Pharmacological Properties and Potential Applications of Natural Monoterpenes. Mini-Rev. Med. Chem. 2015, 14, 1156–1168. [Google Scholar] [CrossRef]
  16. Li, N.; Li, L.; Wu, H.; Zhou, H. Antioxidative property and molecular mechanisms underlying genipo-side-mediated therapeutic effects in diabetes mellitus and cardiovascular disease. Oxid. Med. Cell Longev. 2019, 2019, 7480512. [Google Scholar] [PubMed] [Green Version]
  17. Silva, E.A.; Santos, D.M.; de Carvalho, F.O.; Menezes, I.A.; Barreto, A.S.; Souza, D.S.; Quintans-Junior, L.J.; Santos, M.R. Monoterpenes and their derivatives as agents for cardiovascular disease management: A systematic review and me-ta-analysis. Phytomedicine 2021, 88, 153451. [Google Scholar] [CrossRef]
  18. Silva, E.A.P.; Carvalho, J.S.; Guimarães, A.G.; Barreto, R.D.S.; Santos, M.R.; Barreto, A.S.; Quintans-Júnior, L.J. The use of terpenes and derivatives as a new perspective for cardiovascular disease treatment: A patent review (2008–2018). Expert Opin. Ther. Pat. 2018, 29, 43–53. [Google Scholar] [CrossRef]
  19. Nelson, R.H. Hyperlipidemia as a Risk Factor for Cardiovascular Disease. Prim. Care Clin. Off. Pract. 2012, 40, 195–211. [Google Scholar] [CrossRef] [Green Version]
  20. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef]
  21. Wilkinson, M.J.; Laffin, L.J.; Davidson, M.H. Overcoming toxicity and side-effects of lipid-lowering therapies. Best Pract. Res. Clin. Endocrinol. Metab. 2014, 28, 439–452. [Google Scholar] [CrossRef] [PubMed]
  22. Brown, M.S.; Goldstein, J.L. Sterol regulatory element binding proteins (SREBPs): Controllers of lipid synthesis and cellular uptake. Nutr. Rev. 1998, 56, S1–S3. [Google Scholar] [CrossRef] [PubMed]
  23. Song, B.-L.; Javitt, N.B.; DeBose-Boyd, R.A. Insig-mediated degradation of HMG CoA reductase stimulated by lanosterol, an intermediate in the synthesis of cholesterol. Cell Metab. 2005, 1, 179–189. [Google Scholar] [CrossRef] [Green Version]
  24. Cho, S.Y.; Jun, H.J.; Lee, J.H.; Jia, Y.; Kim, K.H.; Lee, S.J. Linalool reduces the expression of 3-hydroxy-3-methylglutaryl CoA reductase via sterol regulatory element binding protein-2- and ubiquitin-dependent mecha-nisms. FEBS Lett. 2011, 585, 3289–3296. [Google Scholar] [CrossRef] [Green Version]
  25. Loh, K.; Tam, S.; Murray-Segal, L.; Huynh, K.; Meikle, P.J.; Scott, J.W.; van Denderen, B.; Chen, Z.; Steel, R.; LeBlond, N.D.; et al. Inhibition of Adenosine Mono-phosphate-Activated Protein Kinase-3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Signaling Leads to Hypercholes-terolemia and Promotes Hepatic Steatosis and Insulin Resistance. Hepatol. Commun. 2018, 3, 84–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Potunuru, U.R.; Priya, K.V.; Varsha, M.S.; Mehta, N.; Chandel, S.; Manoj, N.; Raman, T.; Ramar, M.; Gromiha, M.M.; Dixit, M. Amarogentin, a secoiridoid glycoside, activates AMP- activated protein kinase (AMPK) to exert beneficial vasculo-metabolic effects. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2019, 1863, 1270–1282. [Google Scholar] [CrossRef] [PubMed]
  27. Hadrich, F.; Mahmoudi, A.; Bouallagui, Z.; Feki, I.; Isoda, H.; Feve, B.; Sayadi, S. Evaluation of hypocho-lesterolemic effect of oleuropein in cholesterol-fed rats. Chem. Biol. Interact. 2016, 252, 54–60. [Google Scholar] [CrossRef] [Green Version]
  28. Shen, B.; Zhao, C.; Wang, Y.; Peng, Y.; Cheng, J.; Li, Z.; Wu, L.; Jin, M.; Feng, H. Aucubin inhibited lipid accumulation and oxi-dative stress via Nrf2/HO-1 and AMPK signalling pathways. J. Cell. Mol. Med. 2019, 23, 4063–4075. [Google Scholar] [CrossRef] [Green Version]
  29. Pownall, H.J.; Rosales, C.; Gillard, B.K.; Gotto, A.M. High-density lipoproteins, reverse cholesterol transport and atherogenesis. Nat. Rev. Cardiol. 2021, 18, 712–723. [Google Scholar] [CrossRef]
  30. Al Naqeb, G.; Ismail, M.; Yazan, L.S. Effects of thymoquinone rich fraction and thymoquinone on plasma lipoprotein levels and hepatic low density lipoprotein receptor and 3-hydroxy-3-methylglutaryl coenzyme A reductase genes expression. J. Funct. Foods 2009, 1, 298–303. [Google Scholar] [CrossRef]
  31. Liu, J.; Li, Y.; Sun, C.; Liu, S.; Yan, Y.; Pan, H.; Fan, M.; Xue, L.; Nie, C.; Zhang, H.; et al. Geniposide reduces cholesterol accumulation and increases its excretion by regulating the FXR-mediated liver-gut crosstalk of bile acids. Pharmacol. Res. 2020, 152, 104631. [Google Scholar] [CrossRef] [PubMed]
  32. Chiang, J.Y. Negative feedback regulation of bile acid metabolism: Impact on liver metabolism and diseases. Hepatology 2015, 62, 1315–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Matsubara, T.; Li, F.; Gonzalez, F.J. FXR signaling in the enterohepatic system. Mol. Cell. Endocrinol. 2013, 368, 17–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Vaidya, H.; Rajani, M.; Sudarsanam, V.; Padh, H.; Goyal, R. Swertiamarin: A lead from Enico-stemma littorale Blume. for anti-hyperlipidaemic effect. Eur. J. Pharmacol. 2009, 617, 108–112. [Google Scholar] [CrossRef]
  35. Mahdavifard, S.; Nakhjavani, M. Preventive Effect of Eucalyptol on the Formation of Aorta Lesions in the Diabet-ic-Atherosclerotic Rat. Int. J. Prev. Med. 2021, 12, 45. [Google Scholar] [PubMed]
  36. Jayachandran, M.; Chandrasekaran, B.; Namasivayam, N. Effect of geraniol, a plant derived monoterpene on lipids and lipid metabolizing enzymes in experimental hyperlipidemic hamsters. Mol. Cell. Biochem. 2014, 398, 39–53. [Google Scholar] [CrossRef] [PubMed]
  37. Yu, Y.-M.; Chao, T.-Y.; Chang, W.-C.; Chang, M.J.; Lee, M.-F. Thymol reduces oxidative stress, aortic intimal thickening, and inflammation-related gene expression in hyperlipidemic rabbits. J. Food Drug Anal. 2016, 24, 556–563. [Google Scholar] [CrossRef] [Green Version]
  38. Ahmad, S.; Beg, Z.H. Hypolipidemic and antioxidant activities of thymoquinone and limonene in atherogenic suspension fed rats. Food Chem. 2013, 138, 1116–1124. [Google Scholar] [CrossRef]
  39. Sani, M.A.; Yaghmaei, P.; Hajebrahimi, Z.; Roodbari, N.H. Therapeutic Effect of P-Cymene on Lipid Profile, Liver Enzyme, and Akt/Mtor Pathway in Streptozotocin-Induced Diabetes Mellitus in Wistar Rats. J. Obes. 2022, 2022, 1015669. [Google Scholar] [CrossRef]
  40. Samarghandian, S.; Borji, A.; Delkhosh, M.B.; Samini, F. Safranal Treatment Improves Hyperglycemia, Hyperlipidemia and Oxidative Stress in Streptozotocin-Induced Diabetic Rats. J. Pharm. Pharm. Sci. 2013, 16, 352–362. [Google Scholar] [CrossRef]
  41. Zhong, H.; Chen, K.; Feng, M.; Shao, W.; Wu, J.; Chen, K.; Liang, T.; Liu, C. Genipin alleviates high-fat diet-induced hyper-lipidemia and hepatic lipid accumulation in mice via miR-142a-5p/SREBP-1c axis. FEBS J. 2018, 285, 501–517. [Google Scholar] [CrossRef]
  42. Liu, J.-Y.; Zhang, D.-J. Amelioration by Catalpol of Atherosclerotic Lesions in Hypercholesterolemic Rabbits. Planta Med. 2015, 81, 175–184. [Google Scholar] [CrossRef] [Green Version]
  43. Zhang, L.; Yang, B.; Yu, B. Paeoniflorin Protects against Nonalcoholic Fatty Liver Disease Induced by a High-Fat Diet in Mice. Biol. Pharm. Bull. 2015, 38, 1005–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sozański, T.; Kucharska, A.Z.; Rapak, A.; Szumny, D.; Trocha, M.; Merwid-Ląd, A.; Dzimira, S.; Piasecki, T.; Piórecki, N.; Magdalan, J.; et al. Iri-doid-loganic acid versus anthocyanins from the Cornus mas fruits (cornelian cherry): Common and different effects on di-et-induced atherosclerosis, PPARs expression and inflammation. Atherosclerosis 2016, 254, 151–160. [Google Scholar] [CrossRef] [PubMed]
  45. Yamabe, N.; Noh, J.S.; Park, C.H.; Kang, K.S.; Shibahara, N.; Tanaka, T.; Yokozawa, T. Evaluation of loganin, iridoid glycoside from Corni Fructus, on hepatic and renal glucolipotoxicity and inflammation in type 2 diabetic db/db mice. Eur. J. Pharmacol. 2010, 648, 179–187. [Google Scholar] [CrossRef]
  46. Lombardo, G.E.; Lepore, S.M.; Morittu, V.M.; Arcidiacono, B.; Colica, C.; Procopio, A.; Maggisano, V.; Bulotta, S.; Costa, N.; Mignogna, C.; et al. Effects of Oleacein on High-Fat Diet-Dependent Steatosis, Weight Gain, and Insulin Resistance in Mice. Front. Endocrinol. 2018, 9, 116. [Google Scholar] [CrossRef] [Green Version]
  47. Jin, M.; Feng, H.; Wang, Y.; Yan, S.; Shen, B.; Li, Z.; Qin, H.; Wang, Q.; Li, J.; Liu, G. Gentiopicroside Ameliorates Oxidative Stress and Lipid Accumulation through Nuclear Factor Erythroid 2-Related Factor 2 Activation. Oxidative Med. Cell. Longev. 2020, 2020, 2940746. [Google Scholar] [CrossRef] [PubMed]
  48. Gimbrone, M.A., Jr.; García-Cardeña, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hayden, M.; Ghosh, S. NF-κB in immunobiology. Cell Res. 2011, 21, 223–244. [Google Scholar] [CrossRef] [Green Version]
  50. Lim, S.; Lee, K.-S.; Lee, J.E.; Park, H.S.; Kim, K.M.; Moon, J.H.; Choi, S.H.; Park, K.S.; Kim, Y.B.; Jang, H.C. Effect of a new PPAR-gamma agonist, lobeglitazone, on neointimal formation after balloon injury in rats and the development of atherosclerosis. Atherosclerosis 2015, 243, 107–119. [Google Scholar] [CrossRef]
  51. He, X.; Liu, W.; Shi, M.; Yang, Z.; Zhang, X.; Gong, P. Docosahexaenoic acid attenuates LPS-stimulated inflammatory response by regulating the PPARγ/NF-κB pathways in primary bovine mammary epithelial cells. Res. Vet. Sci. 2017, 112, 7–12. [Google Scholar] [CrossRef]
  52. Linghu, K.G.; Wu, G.P.; Fu, L.Y.; Yang, H.; Li, H.Z.; Chen, Y.; Yu, H.; Tao, L.; Shen, X.C. 1,8-Cineole Ameliorates LPS-Induced Vascular Endothelium Dysfunction in Mice via PPAR-γ Dependent Regulation of NF-κB. Front. Pharmacol. 2019, 10, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Song, Y.; Zhao, H.; Liu, J.; Fang, C.; Miao, R. Effects of Citral on Lipopolysaccharide-Induced Inflam-mation in Human Umbilical Vein Endothelial Cells. Inflammation 2016, 39, 663–671. [Google Scholar] [CrossRef] [PubMed]
  54. Katsukawa, M.; Nakata, R.; Koeji, S.; Hori, K.; Takahashi, S.; Inoue, H. Citronellol and geraniol, components of rose oil, activate peroxisome proliferator-activated receptor α and γ and suppress cyclooxygenase-2 expression. Biosci. Biotechnol. Biochem. 2011, 75, 1010–1012. [Google Scholar] [CrossRef] [PubMed]
  55. Hwa, J.S.; Mun, L.; Kim, H.J.; Seo, H.G.; Lee, J.H.; Kwak, J.H.; Lee, D.-U.; Chang, K.C. Genipin Selectively Inhibits TNF-α-activated VCAM-1 But Not ICAM-1 Expression by Upregulation of PPAR-γ in Human Endothelial Cells. Korean J. Physiol. Pharmacol. 2011, 15, 157–162. [Google Scholar] [CrossRef] [Green Version]
  56. Zhang, X.; Fernández-Hernando, C. Endothelial HMGB1 (High-Mobility Group Box 1) Regulation of LDL (Low-Density Lipoprotein) Transcytosis: A Novel Mechanism of Intracellular HMGB1 in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2020, 41, 217–219. [Google Scholar]
  57. Kim, N.; Kim, C.; Ryu, S.H.; Lee, W.; Bae, J.-S. Anti-Septic Functions of Cornuside against HMGB1-Mediated Severe Inflammatory Responses. Int. J. Mol. Sci. 2022, 23, 2065. [Google Scholar] [CrossRef]
  58. Li, J.Z.; Wu, J.H.; Yu, S.Y.; Shao, Q.R.; Dong, X.M. Inhibitory effects of paeoniflorin on lysophosphati-dylcholine-induced inflammatory factor production in human umbilical vein endothelial cells. Int. J. Mol. Med. 2013, 31, 493–497. [Google Scholar] [CrossRef] [Green Version]
  59. Nishikawa, T.; Edelstein, D.; Brownlee, M. The missing link: A single unifying mechanism for diabetic complications. Kidney Int. 2000, 58, S26–S30. [Google Scholar] [CrossRef] [Green Version]
  60. Wang, G.F.; Wu, S.Y.; Xu, W.; Jin, H.; Zhu, Z.G.; Li, Z.H.; Tian, Y.X.; Zhang, J.J.; Rao, J.J.; Wu, S.G. Geniposide inhibits high glu-cose-induced cell adhesion through the NF-kappaB signaling pathway in human umbilical vein endothelial cells. Acta Pharmacol. Sin. 2010, 31, 953–962. [Google Scholar] [CrossRef] [Green Version]
  61. Bi, Y.; Han, X.; Lai, Y.; Fu, Y.; Li, K.; Zhang, W.; Wang, Q.; Jiang, X.; Zhou, Y.; Liang, H.; et al. Systems pharmacological study based on UHPLC-Q-Orbitrap-HRMS, network pharmacology and experimental validation to explore the potential mechanisms of Danggui-Shaoyao-San against atherosclerosis. J. Ethnopharmacol. 2021, 278, 114278. [Google Scholar] [CrossRef]
  62. Liu, H.T.; He, J.L.; Li, W.M.; Yang, Z.; Wang, Y.X.; Yin, J.; Du, Y.G.; Yu, C. Geniposide inhibits interleukin-6 and interleukin-8 production in lipopolysaccharide-induced human umbilical vein endothelial cells by blocking p38 and ERK1/2 signaling pathways. Inflamm. Res. 2010, 59, 451–461. [Google Scholar] [PubMed]
  63. Kang, D.G.; Moon, M.K.; Lee, A.S.; Kwon, T.O.; Kim, J.S.; Lee, H.S. Cornuside suppresses cyto-kine-induced proinflammatory and adhesion molecules in the human umbilical vein endothelial cells. Biol. Pharm. Bull. 2007, 30, 1796–1799. [Google Scholar] [PubMed] [Green Version]
  64. Wang, Y.; Che, J.; Zhao, H.; Tang, J.; Shi, G. Paeoniflorin attenuates oxidized low-density lipoprotein-induced apoptosis and adhesion molecule expression by autophagy enhancement in human umbilical vein endothelial cells. J. Cell. Biochem. 2018, 120, 9291–9299. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, J.; Zhang, M.; Zhu, M.; Gu, J.; Song, J.; Cui, L.; Liu, D.; Ning, Q.; Jia, X.; Feng, L. Paeoniflorin prevents endoplasmic reticulum stress-associated inflammation in lipopolysaccharide -stimulated human umbilical vein endothelial cells via the IRE1α/NF-κB signaling pathway. Food Funct. 2018, 9, 2386–2397. [Google Scholar] [CrossRef]
  66. Hu, H.; Wang, C.; Jin, Y.; Meng, Q.; Liu, Q.; Liu, Z.; Liu, K.; Liu, X.; Sun, H. Catalpol Inhibits Homocysteine-induced Ox-idation and Inflammation via Inhibiting Nox4/NF-κB and GRP78/PERK Pathways in Human Aorta Endothelial Cells. Inflammation 2019, 42, 64–80. [Google Scholar] [CrossRef] [Green Version]
  67. Zhou, S.; Sun, Y.; Zhao, K.; Gao, Y.; Cui, J.; Qi, L.; Huang, L. miR-21/PTEN pathway mediates the car-dioprotection of geniposide against oxidized low-density lipoprotein-induced endothelial injury via suppressing oxidative stress and inflammatory response. Int. J. Mol. Med. 2020, 45, 1305–1316. [Google Scholar]
  68. Liu, X.; Xu, Y.; Cheng, S.; Zhou, X.; Zhou, F.; He, P.; Hu, F.; Zhang, L.; Chen, Y.; Jia, Y. Geniposide Combined With Noto-ginsenoside R1 Attenuates Inflammation and Apoptosis in Atherosclerosis via the AMPK/mTOR/Nrf2 Signaling Pathway. Front. Pharmacol. 2021, 12, 687394. [Google Scholar] [CrossRef]
  69. Yang, L.; Liu, J.; Li, Y.; Qi, G. Bornyl acetate suppresses ox-LDL-induced attachment of THP-1 monocytes to endothelial cells. Biomed. Pharmacother. 2018, 103, 234–239. [Google Scholar] [CrossRef]
  70. Zhao, W.; Deng, C.; Han, Q.; Xu, H.; Chen, Y. Carvacrol may alleviate vascular inflammation in diabetic db/db mice. Int. J. Mol. Med. 2020, 46, 977–988. [Google Scholar] [CrossRef]
  71. Shih, M.F.; Pan, K.H.; Liu, C.C.; Shen, C.R.; Cherng, J.Y. Treatment of β-thujaplicin counteracts di(2-ethylhexyl)phthalate (DEHP)-exposed vascular smooth muscle activation, inflammation and atherosclerosis progression. Regul. Toxicol. Pharmacol. 2018, 92, 333–337. [Google Scholar] [CrossRef]
  72. Amartey, J.; Gapper, S.; Hussein, N.; Morris, K.; Withycombe, C.E. Nigella sativa Extract and Thy-moquinone Regulate Inflammatory Cytokine and TET-2 Expression in Endothelial Cells. Artery Res. 2019, 25, 157–163. [Google Scholar] [CrossRef]
  73. Carluccio, M.A.; Siculella, L.; Ancora, M.A.; Massaro, M.; Scoditti, E.; Storelli, C.; Visioli, F.; Distante, A.; De Caterina, R. Olive oil and red wine antioxidant polyphenols inhibit endothelial activation: Antiatherogenic properties of Mediterranean diet phytochemicals. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 622–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sindona, G.; Caruso, A.; Cozza, A.; Fiorentini, S.; Lorusso, B.; Marini, E.; Nardi, M.; Procopio, A.; Zicari, S. Anti-inflammatory effect of 3,4-DHPEA-EDA [2-(3,4 -hydroxyphenyl) ethyl (3S, 4E)-4-formyl-3-(2-oxoethyl)hex-4-enoate] on primary human vascular endothelial cells. Curr. Med. Chem. 2012, 19, 4006–4013. [Google Scholar] [CrossRef] [PubMed]
  75. Fakhrudin, N.; Waltenberger, B.; Cabaravdic, M.; Atanasov, A.G.; Malainer, C.; Schachner, D.; Heiss, E.H.; Liu, R.; Noha, S.M.; Grzywacz, A.M.; et al. Identification of plumericin as a potent new inhibitor of the NF-κB pathway with anti-inflammatory activity in vitro and in vivo. Br. J. Pharmacol. 2014, 171, 1676–1686. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, Y.; Hong, Y.; Zhang, C.; Shen, Y.; Pan, Y.S.; Chen, R.Z.; Zhang, Q.; Chen, Y.H. Picroside II attenuates hyper-homocysteinemia-induced endothelial injury by reducing inflammation, oxidative stress and cell apoptosis. J. Cell. Mol. Med. 2019, 23, 464–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Jiang, F.; Xu, X.R.; Li, W.M.; Xia, K.; Wang, L.F.; Yang, X.C. Monotropein alleviates H2O2-induced inflammation, oxidative stress and apoptosis via NF-κB/AP-1 signaling. Mol. Med. Rep. 2020, 22, 4828–4836. [Google Scholar] [CrossRef] [PubMed]
  78. Liu, Y.; Sun, Y.; Bai, X.; Li, L.; Zhu, G. Albiflorin Alleviates Ox-LDL-Induced Human Umbilical Vein En-dothelial Cell Injury through IRAK1/TAK1 Pathway. Biomed. Res. Int. 2022, 2022, 6584645. [Google Scholar]
  79. Zhang, C.; Syed, T.W.; Liu, R.; Yu, J. Role of Endoplasmic Reticulum Stress, Autophagy, and Inflammation in Cardiovascular Disease. Front. Cardiovasc. Med. 2017, 4, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef]
  81. Wang, X.; Zhao, S.; Su, M.; Sun, L.; Zhang, S.; Wang, D.; Liu, Z.; Yuan, Y.; Liu, Y.; Li, Y. Geraniol improves endothelial function by inhibiting NOX-2 derived oxidative stress in high fat diet fed mice. Biochem. Biophys. Res. Commun. 2016, 474, 182–187. [Google Scholar] [CrossRef] [PubMed]
  82. Song, S.; Xiao, X.; Guo, D.; Mo, L.; Bu, C.; Ye, W.; Den, Q.; Liu, S.; Yang, X. Protective effects of Paeoniflorin against AOPP-induced oxidative injury in HUVECs by blocking the ROS-HIF-1α/VEGF pathway. Phytomedicine 2017, 34, 115–126. [Google Scholar] [CrossRef]
  83. Lu, Y.W.; Hao, R.J.; Wei, Y.Y.; Yu, G.R. The protective effect of harpagoside on angiotensin II (Ang II )-induced blood–brain barrier leakage in vitro. Phytother. Res. 2021, 35, 6241–6254. [Google Scholar] [CrossRef]
  84. El-Agamy, D.S.; A Nader, M. Attenuation of oxidative stress-induced vascular endothelial dysfunction by thymoquinone. Exp. Biol. Med. 2012, 237, 1032–1038. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, L.; Liu, H. Perillaldehyde prevents the formations of atherosclerotic plaques through recoupling endothelial nitric oxide synthase. J. Cell. Biochem. 2018, 119, 10204–10215. [Google Scholar] [CrossRef]
  86. Lu, J.X.; Guo, C.; Ou, W.S.; Jing, Y.; Niu, H.F.; Song, P.; Li, Q.Z.; Liu, Z.; Xu, J.; Li, P.; et al. Citronellal prevents endothelial dysfunction and ath-erosclerosis in rats. J. Cell. Biochem. 2019, 120, 3790–3800. [Google Scholar] [CrossRef]
  87. Jayachandran, M.; Chandrasekaran, B.; Namasivayam, N. Geraniol attenuates oxidative stress by Nrf2 activation in diet-induced experimental atherosclerosis. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 335–346. [Google Scholar] [CrossRef]
  88. Wu, J.; Zhang, D.; Hu, L.; Zheng, X.; Chen, C. Paeoniflorin alleviates NG-nitro-L-arginine methyl ester (L-NAME)-induced gestational hypertension and upregulates silent information regulator 2 related enzyme 1 (SIRT1) to reduce H2O2-induced endothelial cell damage. Bioengineered 2022, 13, 2248–2258. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, T.; Guo, Z.P.; Jiao, X.Y.; Zhang, Y.H.; Li, J.Y.; Liu, H.J. Protective effects of peoniflorin against hy-drogen peroxide-induced oxidative stress in human umbilical vein endothelial cells. Can. J. Physiol. Pharmacol. 2011, 89, 445–453. [Google Scholar] [CrossRef] [PubMed]
  90. Jiang, J.; Dong, C.; Zhai, L.; Lou, J.; Jin, J.; Cheng, S.; Chen, Z.; Guo, X.; Lin, D.; Ding, J.; et al. Paeoniflorin Suppresses TBHP-Induced Oxidative Stress and Apoptosis in Human Umbilical Vein Endothelial Cells via the Nrf2/HO-1 Signaling Pathway and Im-proves Skin Flap Survival. Front. Pharmacol. 2021, 12, 735530. [Google Scholar] [CrossRef]
  91. Qiu, Y.; Chao, C.Y.; Jiang, L.; Zhang, J.; Niu, Q.Q.; Guo, Y.Q.; Song, Y.T.; Li, P.; Zhu, M.L.; Yin, Y.L. Citronellal alleviate macro- and micro-vascular damage in high fat diet/streptozotocin-Induced diabetic rats via a S1P/S1P1 dependent signaling pathway. Eur. J. Pharmacol. 2022, 920, 174796. [Google Scholar] [CrossRef] [PubMed]
  92. Peng, J.; Jiang, Z.; Wu, G.; Cai, Z.; Du, Q.; Tao, L.; Zhang, Y.; Chen, Y.; Shen, X. Improving protection effects of eucalyptol via carboxymethyl chitosan-coated lipid nanoparticles on hyperglycaemia-induced vascular endothelial injury in rats. J. Drug Target. 2020, 29, 520–530. [Google Scholar] [CrossRef] [PubMed]
  93. Linghu, K.; Lin, D.; Yang, H.; Xu, Y.; Zhang, Y.; Tao, L.; Chen, Y.; Shen, X. Ameliorating effects of 1,8-cineole on LPS-induced human umbilical vein endothelial cell injury by suppressing NF-κB signaling in vitro. Eur. J. Pharmacol. 2016, 789, 195–201. [Google Scholar] [CrossRef]
  94. Sun, W.; Gao, Y.; Ding, Y.; Cao, Y.; Chen, J.; Lv, G.; Lu, J.; Yu, B.; Peng, M.; Xu, H.; et al. Catalpol ameliorates advanced glycation end product-induced dysfunction of glomerular endothelial cells via regulating nitric oxide synthesis by inducible nitric oxide synthase and endothelial nitric oxide synthase. IUBMB Life 2019, 71, 1268–1283. [Google Scholar] [CrossRef]
  95. Safaeian, L.; Sajjadi, S.E.; Montazeri, H.; Ohadi, F.; Javanmard, S. Citral Protects Human Endothelial Cells Against Hydrogen Peroxide-induced Oxidative Stress. Turk. J. Pharm. Sci. 2020, 17, 549–554. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, G.F.; Wu, S.Y.; Rao, J.J.; Lü, L.; Xu, W.; Pang, J.X.; Liu, Z.Q.; Wu, S.G.; Zhang, J.J. Genipin inhibits endothelial exocytosis via nitric oxide in cultured human umbilical vein endothelial cells. Acta Pharmacol. Sin. 2009, 30, 589–596. [Google Scholar]
  97. Rahiman, N.; Akaberi, M.; Sahebkar, A.; Emami, S.A.; Tayarani-Najaran, Z. Protective effects of saffron and its active components against oxidative stress and apoptosis in endothelial cells. Microvasc. Res. 2018, 118, 82–89. [Google Scholar] [CrossRef]
  98. Lee, G.-H.; Lee, H.-Y.; Choi, M.-K.; Choi, A.-H.; Shin, T.-S.; Chae, H.-J. Eucommia ulmoides leaf (EUL) extract enhances NO production in ox-LDL-treated human endothelial cells. Biomed. Pharmacother. 2018, 97, 1164–1172. [Google Scholar] [CrossRef]
  99. Jazbutyte, V.; Thum, T. MicroRNA-21: From Cancer to Cardiovascular Disease. Curr. Drug Targets 2010, 11, 926–935. [Google Scholar] [CrossRef]
  100. Yang, Q.; Yang, K.; Li, A. microRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/Akt-dependent mechanism. Mol. Med. Rep. 2014, 9, 2213–2220. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, J.Y.; Ma, J.; Yu, P.; Tang, G.J.; Li, C.J.; Yu, D.M.; Zhang, Q.M. Reduced beta 2 glycoprotein I prevents high glucose-induced cell death in HUVECs through miR-21/PTEN. Am. J. Transl. Res. 2017, 9, 3935–3949. [Google Scholar] [PubMed]
  102. Hung, C.H.; Chan, S.H.; Chu, P.M.; Tsai, K.L. Homocysteine facilitates LOX-1 activation and endothelial death through the PKCβ and SIRT1/HSF1 mechanism: Relevance to human hyperhomocysteinaemia. Clin. Sci. 2015, 129, 477–487. [Google Scholar]
  103. Chen, X.-P.; Xun, K.-L.; Wu, Q.; Zhang, T.-T.; Shi, J.-S.; Du, G.-H. Oxidized low density lipoprotein receptor-1 mediates oxidized low density lipoprotein-induced apoptosis in human umbilical vein endothelial cells: Role of reactive oxygen species. Vasc. Pharmacol. 2007, 47, 1–9. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, J.Y.; Ye, Z.X.; Wang, X.F.; Chang, J.; Yang, M.W.; Zhong, H.H.; Hong, F.F.; Yang, S.L. Nitric oxide bioavailability dys-function involves in atherosclerosis. Biomed. Pharmacother. 2018, 97, 423–428. [Google Scholar] [CrossRef] [PubMed]
  105. Förstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Athero-sclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar]
  106. Duan, H.; Zhang, Q.; Liu, J.; Li, R.; Wang, D.; Peng, W.; Wu, C. Suppression of apoptosis in vascular endothelial cell, the promising way for natural medicines to treat atherosclerosis. Pharmacol. Res. 2021, 168, 105599. [Google Scholar] [CrossRef]
  107. Datta, A.; Sarmah, D.; Mounica, L.; Kaur, H.; Kesharwani, R.; Verma, G.; Veeresh, P.; Kotian, V.; Kalia, K.; Borah, A.; et al. Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl. Stroke Res. 2020, 11, 1185–1202. [Google Scholar] [CrossRef]
  108. Paone, S.; Baxter, A.A.; Hulett, M.D.; Poon, I.K.H. Endothelial cell apoptosis and the role of endothelial cell-derived extracellular vesicles in the progression of atherosclerosis. Cell. Mol. Life Sci. 2019, 76, 1093–1106. [Google Scholar] [CrossRef]
  109. Hu, L.; Sun, Y.; Hu, J. Catalpol inhibits apoptosis in hydrogen peroxide-induced endothelium by activating the PI3K/Akt signaling pathway and modulating expression of Bcl-2 and Bax. Eur. J. Pharmacol. 2010, 628, 155–163. [Google Scholar] [CrossRef] [PubMed]
  110. Jian, X.; Liu, Y.; Zhao, Z.; Zhao, L.; Wang, D.; Liu, Q. The role of traditional Chinese medicine in the treatment of atherosclerosis through the regulation of macrophage activity. Biomed. Pharmacother. 2019, 118, 109375. [Google Scholar] [CrossRef]
  111. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Anti-inflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  112. Bäck, M.; Hansson, G.K. Anti-inflammatory therapies for atherosclerosis. Nat. Rev. Cardiol. 2015, 12, 199–211. [Google Scholar] [CrossRef] [PubMed]
  113. Cheng, S.; Zhou, F.; Xu, Y.; Liu, X.; Zhang, Y.; Gu, M.; Su, Z.; Zhao, D.; Zhang, L.; Jia, Y. Geniposide regulates the miR-101/MKP-1/p38 pathway and alleviates atherosclerosis inflammatory injury in ApoE-/- mice. Immunobiology 2019, 224, 296–306. [Google Scholar] [CrossRef] [PubMed]
  114. Fu, Y.; Liu, B.; Liu, J.; Liu, Z.; Liang, D.; Li, F.; Li, D.; Cao, Y.; Zhang, X.; Zhang, N.; et al. Geniposide, from Gardenia jasminoides Ellis, inhibits the inflammatory response in the primary mouse macrophages and mouse models. Int. Immunopharmacol. 2012, 14, 792–798. [Google Scholar] [CrossRef] [PubMed]
  115. Pu, Z.; Liu, Y.; Li, C.; Xu, M.; Xie, H.; Zhao, J. Using Network Pharmacology for Systematic Under-standing of Geniposide in Ameliorating Inflammatory Responses in Colitis Through Suppression of NLRP3 Inflammasome in Macrophage by AMPK/Sirt1 Dependent Signaling. Am. J. Chin. Med. 2020, 48, 1693–1713. [Google Scholar] [CrossRef] [PubMed]
  116. Sousa, C.; Neves, B.; Leitão, A.; Mendes, A. Elucidation of the Mechanism Underlying the Anti-Inflammatory Properties of (S)-(+)-Carvone Identifies a Novel Class of Sirtuin-1 Activators in a Murine Macrophage Cell Line. Biomedicines 2021, 9, 777. [Google Scholar] [CrossRef]
  117. Liu, S.; Shen, H.; Li, J.; Gong, Y.; Bao, H.; Zhang, J.; Hu, L.; Wang, Z.; Gong, J. Loganin inhibits macrophage M1 polarization and modulates sirt1/NF-κB signaling pathway to attenuate ulcerative colitis. Bioengineered 2020, 11, 628–639. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, R.; Dong, Z.; Lan, X.; Liao, Z.; Chen, M. Sweroside Alleviated LPS-Induced Inflammation via SIRT1 Mediating NF-κB and FOXO1 Signaling Pathways in RAW264.7 Cells. Molecules 2019, 24, 872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Shen, Y.; Sun, Z.; Guo, X. Citral inhibits lipopolysaccharide-induced acute lung injury by activating PPAR-γ. Eur. J. Pharmacol. 2015, 747, 45–51. [Google Scholar] [CrossRef]
  120. Hotta, M.; Nakata, R.; Katsukawa, M.; Hori, K.; Takahashi, S.; Inoue, H. Carvacrol, a component of thyme oil, activates PPARalpha and gamma and suppresses COX-2 expression. J. Lipid Res. 2010, 51, 132–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Jeon, W.-K.; Hong, H.-Y.; Kim, B.-C. Genipin up-regulates heme oxygenase-1 via PI3-kinase-JNK1/2-Nrf2 signaling pathway to enhance the anti-inflammatory capacity in RAW264.7 macrophages. Arch. Biochem. Biophys. 2011, 512, 119–125. [Google Scholar] [CrossRef]
  122. Park, C.; Lee, H.; Kwon, C.-Y.; Kim, G.-Y.; Jeong, J.-W.; Kim, S.O.; Choi, S.H.; Jeong, S.-J.; Noh, J.S.; Choi, Y.H. Loganin Inhibits Lipopol-ysaccharide-Induced Inflammation and Oxidative Response through the Activation of the Nrf2/HO-1 Signaling Pathway in RAW264.7 Macrophages. Biol. Pharm. Bull. 2021, 44, 875–883. [Google Scholar] [CrossRef]
  123. Montoya, T.; Castejón, M.L.; Sánchez-Hidalgo, M.; González-Benjumea, A.; Fernández-Bolaños, J.G.; De-La-Lastra, C.A. Oleocanthal Modulates LPS-Induced Murine Peritoneal Macrophages Activation via Regulation of Inflammasome, Nrf-2/HO-1, and MAPKs Signaling Pathways. J. Agric. Food Chem. 2019, 67, 5552–5559. [Google Scholar] [CrossRef] [PubMed]
  124. Qiu, Y.-L.; Cheng, X.-N.; Bai, F.; Fang, L.-Y.; Hu, H.-Z.; Sun, D.-Q. Aucubin protects against lipopoly-saccharide-induced acute pulmonary injury through regulating Nrf2 and AMPK pathways. Biomed. Pharmacother. 2018, 106, 192–199. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, Y.-E.; Xu, S.-J.; Lu, Y.-Y.; Chen, S.-X.; Du, X.-H.; Hou, S.-Z.; Huang, H.Y.; Liang, J. Asperuloside suppressing oxi-dative stress and inflammation in DSS-induced chronic colitis and RAW 264.7 macrophages via Nrf2/HO-1 and NF-κB pathways. Chem. Biol. Interact. 2021, 344, 109512. [Google Scholar] [CrossRef] [PubMed]
  126. Park, C.; Cha, H.-J.; Lee, H.; Kim, G.-Y.; Choi, Y.H. The regulation of the TLR4/NF-κB and Nrf2/HO-1 signaling pathways is involved in the inhibition of lipopolysaccharide-induced inflammation and oxidative reactions by morroniside in RAW 264.7 macrophages. Arch. Biochem. Biophys. 2021, 706, 108926. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, Z.; Zhang, J.; Jiang, P.; Yin, Z.; Liu, Y.; Liu, Y.; Wang, X.; Hu, L.; Xu, Y.; Liu, W. Paeoniflorin inhibits the macrophage-related rosacea-like inflammatory reaction through the suppressor of cytokine signaling 3-apoptosis signal-regulating kinase 1-p38 pathway. Medicine 2021, 100, e23986. [Google Scholar] [CrossRef]
  128. Chen, N.; Sun, G.; Yuan, X.; Hou, J.; Wu, Q.; Soromou, L.W.; Feng, H. Inhibition of lung inflammatory re-sponses by bornyl acetate is correlated with regulation of myeloperoxidase activity. J. Surg. Res. 2014, 186, 436–445. [Google Scholar] [CrossRef]
  129. Lee, E.H.; Shin, J.H.; Kim, S.S.; Lee, H.; Yang, S.; Seo, S.R. Laurus nobilis leaf extract controls inflammation by suppressing NLRP3 inflammasome activation. J. Cell. Physiol. 2018, 234, 6854–6864. [Google Scholar] [CrossRef]
  130. Trinh, H.-T.; Lee, I.-A.; Hyun, Y.-J.; Kim, D.-H. Artemisia princeps Pamp. Essential oil and its constituents eucalyptol and α-terpineol ameliorate bacterial vaginosis and vulvovaginal candidiasis in mice by inhibiting bacterial growth and NF-κB activation. Planta Med. 2011, 77, 1996–2002. [Google Scholar] [CrossRef] [PubMed]
  131. Yadav, N.; Chandra, H. Suppression of inflammatory and infection responses in lung macrophages by eucalyptus oil and its constituent 1,8-cineole: Role of pattern recognition receptors TREM-1 and NLRP3, the MAP kinase regulator MKP-1, and NFκB. PLoS ONE 2017, 12, e0188232. [Google Scholar] [CrossRef] [Green Version]
  132. Kim, D.S.; Lee, H.J.; Jeon, Y.D.; Han, Y.H.; Kee, J.Y.; Kim, H.J.; Shin, H.J.; Kang, J.; Lee, B.S.; Kim, S.H.; et al. Alpha-Pinene Exhibits An-ti-Inflammatory Activity Through the Suppression of MAPKs and the NF-κB Pathway in Mouse Peritoneal Macrophages. Am. J. Chin. Med. 2015, 43, 731–742. [Google Scholar] [CrossRef] [PubMed]
  133. Su, Y.-W.; Chao, S.-H.; Lee, M.-H.; Ou, T.-Y.; Tsai, Y.-C. Inhibitory Effects of Citronellol and Geraniol on Nitric Oxide and Prostaglandin E2Production in Macrophages. Planta Med. 2010, 76, 1666–1671. [Google Scholar] [CrossRef] [PubMed]
  134. Huo, M.; Cui, X.; Xue, J.; Chi, G.; Gao, R.; Deng, X.; Guan, S.; Wei, J.; Soromou, L.W.; Feng, H.; et al. Anti-inflammatory effects of linalool in RAW 264.7 macrophages and lipopolysaccharide-induced lung injury model. J. Surg. Res. 2013, 180, e47–e54. [Google Scholar] [CrossRef] [PubMed]
  135. Ka, S.-M.; Lin, J.-C.; Lin, T.-J.; Liu, F.-C.; Chao, L.K.; Ho, C.-L.; Yeh, L.-T.; Sytwu, H.-K.; Hua, K.-F.; Chen, A. Citral alleviates an accelerated and severe lupus nephritis model by inhibiting the activation signal of NLRP3 inflammasome and enhancing Nrf2 activation. Thromb. Haemost. 2015, 17, 331. [Google Scholar] [CrossRef] [Green Version]
  136. Leu, W.-J.; Chen, J.-C.; Guh, J.-H. Extract From Plectranthus amboinicus Inhibit Maturation and Release of Interleukin 1β Through Inhibition of NF-κB Nuclear Translocation and NLRP3 Inflammasome Activation. Front. Pharmacol. 2019, 10, 573. [Google Scholar] [CrossRef]
  137. Gholijani, N.; Gharagozloo, M.; Farjadian, S.; Amirghofran, Z. Modulatory effects of thymol and carvacrol on inflammatory transcription factors in lipopolysaccharide-treated macrophages. J. Immunotoxicol. 2015, 13, 157–164. [Google Scholar] [CrossRef]
  138. Chen, J.; Li, D.-L.; Xie, L.-N.; Ma, Y.-R.; Wu, P.-P.; Li, C.; Liu, W.-F.; Zhang, K.; Zhou, R.-P.; Xu, X.-T.; et al. Synergistic anti-inflammatory effects of silibinin and thymol combination on LPS-induced RAW264.7 cells by inhibition of NF-κB and MAPK activation. Phytomedicine 2020, 78, 153309. [Google Scholar] [CrossRef] [PubMed]
  139. Uemura, T.; Yashiro, T.; Oda, R.; Shioya, N.; Nakajima, T.; Hachisu, M.; Kobayashi, S.; Nishiyama, C.; Arimura, G.I. Intestinal An-ti-Inflammatory Activity of Perillaldehyde. J. Agric. Food. Chem. 2018, 66, 3443–3448. [Google Scholar] [CrossRef]
  140. Byeon, S.E.; Lee, Y.G.; Kim, J.-C.; Han, J.G.; Lee, H.Y.; Cho, J.Y. Hinokitiol, a natural tropolone derivative, inhibits TNF-alpha production in LPS-activated macrophages via suppression of NF-kappaB. Planta Med. 2008, 74, 828–833. [Google Scholar] [CrossRef]
  141. Zhong, W.; Chi, G.; Jiang, L.; Soromou, L.W.; Chen, N.; Huo, M.; Guo, W.; Deng, X.; Feng, H. p-Cymene modulates in vitro and in vivo cytokine production by inhibiting MAPK and NF-κB activation. Inflammation 2013, 36, 529–537. [Google Scholar] [CrossRef] [PubMed]
  142. Hossen, M.J.; Yang, W.S.; Kim, D.; Aravinthan, A.; Kim, J.-H.; Cho, J.Y. Thymo-quinone: An IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities. Sci. Rep. 2017, 7, 42995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ramalho, T.R.; Filgueiras, L.R.; de Oliveira, M.T.P.; de Araujo Lima, A.L.; Bezerra-Santos, C.R.; Jancar, S.; Piuvezam, M.R. Gamma-Terpinene Modulation of LPS-Stimulated Macrophages is Dependent on the PGE2/IL-10 Axis. Planta Med. 2016, 82, 1341–1345. [Google Scholar] [CrossRef] [PubMed]
  144. Gupta, M.; Wani, A.; Ahsan, A.U.; Ali, M.; Chibber, P.; Singh, S.; Digra, S.K.; Datt, M.; Bharate, S.B.; Vishwakarma, R.A.; et al. Safranal inhibits NLRP3 inflammasome activation by preventing ASC oligomerization. Toxicol. Appl. Pharmacol. 2021, 423, 115582. [Google Scholar] [CrossRef]
  145. Lertnimitphun, P.; Jiang, Y.; Kim, N.; Fu, W.; Zheng, C.; Tan, H.; Zhou, H.; Zhang, X.; Pei, W.; Lu, Y.; et al. Safranal Alleviates Dextran Sulfate Sodium-Induced Colitis and Suppresses Macrophage-Mediated Inflammation. Front. Pharmacol. 2019, 10, 1281. [Google Scholar] [CrossRef]
  146. Koo, H.J.; Song, Y.S.; Kim, H.J.; Lee, Y.H.; Hong, S.M.; Kim, S.J.; Kim, B.C.; Jin, C.; Lim, C.J.; Park, E.H. Antiinflammatory effects of genipin, an active principle of gardenia. Eur. J. Pharmacol. 2004, 495, 201–208. [Google Scholar] [CrossRef]
  147. Zhu, T.; Zhang, L.; Ling, S.; Duan, J.; Qian, F.; Li, Y.; Xu, J.W. Scropolioside B inhibits IL-1β and cytokines expression through NF-κB and inflammasome NLRP3 pathways. Mediat. Inflamm. 2014, 2014, 819053. [Google Scholar] [CrossRef] [Green Version]
  148. Fu, K.; Piao, T.; Wang, M.; Zhang, J.; Jiang, J.; Wang, X.; Liu, H. Protective effect of catalpol on lipopoly-saccharide-induced acute lung injury in mice. Int. Immunopharmacol. 2014, 23, 400–406. [Google Scholar] [CrossRef]
  149. Saravanan, S.; Islam, V.H.; Babu, N.P.; Pandikumar, P.; Thirugnanasambantham, K.; Chellappandian, M.; Raj, C.S.D.; Paulraj, M.G.; Ignacimuthu, S. Swer-tiamarin attenuates inflammation mediators via modulating NF-κB/IκB and JAK2/STAT3 transcription factors in adjuvant induced arthritis. Eur. J. Pharm. Sci. 2014, 56, 70–86. [Google Scholar] [CrossRef]
  150. Zhang, M.; Ma, X.; Xu, H.; Wu, W.; He, X.; Wang, X.; Jiang, M.; Hou, Y.; Bai, G. A natural AKT inhibitor swertiamarin targets AKT-PH domain, inhibits downstream signaling, and alleviates inflammation. FEBS J. 2020, 287, 1816–1829. [Google Scholar] [CrossRef]
  151. Li, W.; Tao, W.; Chen, J.; Zhai, Y.; Yin, N.; Wang, Z. Paeoniflorin suppresses IL-33 production by macrophages. Immunopharmacol. Immunotoxicol. 2020, 42, 286–293. [Google Scholar] [CrossRef]
  152. Huang, D.; Li, Z.; Chen, Y.; Fan, Y.; Yu, T. Paeoniflorin reduces the inflammatory response of THP-1 cells by up-regulating microRNA-124: Paeoniflorin reduces the inflammatory response of THP-1 cells through microRNA-124. Genes Genomics 2021, 43, 623–631. [Google Scholar] [CrossRef] [PubMed]
  153. Zhang, T.; Zhu, Q.; Shao, Y.; Wang, K.; Wu, Y. Paeoniflorin prevents TLR2/4-mediated inflammation in type 2 diabetic nephropathy. Biosci. Trends 2017, 11, 308–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Bi, X.; Han, L.; Qu, T.; Mu, Y.; Guan, P.; Qu, X.; Wang, Z.; Huang, X. Anti-Inflammatory Effects, SAR, and Action Mechanism of Monoterpenoids from Radix Paeoniae Alba on LPS-Stimulated RAW 264.7 Cells. Molecules 2017, 22, 715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Choi, Y.H.; Jin, G.Y.; Li, G.Z.; Yan, G.H. Cornuside Suppresses Lipopolysaccharide-Induced Inflammatory Mediators by Inhibiting Nuclear Factor-Kappa B Activation in RAW 264.7 Macrophages. Biol. Pharm. Bull. 2011, 34, 959–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Choi, N.; Yang, G.; Jang, J.; Kang, H.; Cho, Y.-Y.; Lee, H.; Lee, J. Loganin Alleviates Gout Inflammation by Suppressing NLRP3 Inflammasome Activation and Mitochondrial Damage. Molecules 2021, 26, 1071. [Google Scholar] [CrossRef]
  157. Cui, Y.; Gao, H.; Han, S.; Yuan, R.; He, J.; Zhuo, Y.; Feng, Y.L.; Tang, M.; Feng, J.; Yang, S. Oleuropein Attenuates Lipopolysac-charide-Induced Acute Kidney Injury In Vitro and In Vivo by Regulating Toll-Like Receptor 4 Dimerization. Front. Pharmacol. 2021, 12, 617314. [Google Scholar] [CrossRef]
  158. Cirmi, S.; Maugeri, A.; Russo, C.; Musumeci, L.; Navarra, M.; Lombardo, G.E. Oleacein Attenuates Lipopolysaccharide-Induced Inflammation in THP-1-Derived Macrophages by the Inhibition of TLR4/MyD88/NF-κB Pathway. Int. J. Mol. Sci. 2022, 23, 1206. [Google Scholar] [CrossRef]
  159. Yao, H.; Yan, J.; Yin, L.; Chen, W. Picroside II alleviates DSS-induced ulcerative colitis by suppressing the production of NLRP3 inflammasomes through NF-κB signaling pathway. Immunopharmacol. Immunotoxicol. 2022, 44, 437–446. [Google Scholar] [CrossRef]
  160. He, M.; Hu, C.; Chen, M.; Gao, Q.; Li, L.; Tian, W. Effects of Gentiopicroside on activation of NLRP3 inflammasome in acute gouty arthritis mice induced by MSU. J. Nat. Med. 2021, 76, 178–187. [Google Scholar] [CrossRef]
  161. Wang, Q.; Zhou, X.; Yang, L.; Luo, M.; Han, L.; Lu, Y.; Shi, Q.; Wang, Y.; Liang, Q. Gentiopicroside (GENT) protects against sepsis induced by lipopolysaccharide (LPS) through the NF-κB signaling pathway. Ann. Transl. Med. 2019, 7, 731. [Google Scholar] [CrossRef]
  162. Huang, T.H.W.; Tran, V.H.; Duke, R.K.; Tan, S.; Chrubasik, S.; Roufogalis, B.D.; Duke, C.C. Harpagoside suppresses lipopolysaccharide-induced iNOS and COX-2 expression through inhibition of NF-kappa B activation. J. Ethnopharmacol. 2006, 104, 149–155. [Google Scholar] [CrossRef]
  163. An, S.J.; Pae, H.O.; Oh, G.S.; Choi, B.M.; Jeong, S.; Jang, S.I.; Oh, H.; Kwon, T.O.; Song, C.E.; Chung, H.T. Inhibition of TNF-alpha, IL-1beta, and IL-6 productions and NF-kappa B activation in lipopolysaccharide-activated RAW 264.7 macrophages by catalposide, an iridoid glycoside isolated from Catalpa ovata G. Don (Bignoniaceae). Int. Immunopharmacol. 2002, 2, 1173–1181. [Google Scholar] [CrossRef] [PubMed]
  164. Shin, J.-S.; Yun, K.-J.; Chung, K.-S.; Seo, K.-H.; Park, H.-J.; Cho, Y.-W.; Baek, N.-I.; Jang, D.; Lee, K.-T. Monotropein isolated from the roots of Morinda officinalis ameliorates proinflammatory mediators in RAW 264.7 macrophages and dextran sulfate sodium (DSS)-induced colitis via NF-κB inactivation. Food Chem. Toxicol. 2013, 53, 263–271. [Google Scholar] [CrossRef] [PubMed]
  165. He, J.; Lu, X.; Wei, T.; Dong, Y.; Cai, Z.; Tang, L.; Liu, M. Asperuloside and Asperulosidic Acid Exert an Anti-Inflammatory Effect via Suppression of the NF-κB and MAPK Signaling Pathways in LPS-Induced RAW 264.7 Macro-phages. Int. J. Mol. Sci. 2018, 19, 2027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Yang, G.; Jang, J.H.; Kim, S.W.; Han, S.-H.; Ma, K.-H.; Jang, J.-K.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Lee, J.Y. Sweroside Prevents Non-Alcoholic Steatohepatitis by Suppressing Activation of the NLRP3 Inflammasome. Int. J. Mol. Sci. 2020, 21, 2790. [Google Scholar] [CrossRef]
  167. Wang, Q.Q.; Han, S.; Li, X.X.; Yuan, R.; Zhuo, Y.; Chen, X.; Zhang, C.; Chen, Y.; Gao, H.; Zhao, L.C.; et al. Nuezhenide Exerts An-ti-Inflammatory Activity through the NF-κB Pathway. Curr. Mol. Pharmacol. 2021, 14, 101–111. [Google Scholar] [CrossRef]
  168. He, J.; Li, J.; Liu, H.; Yang, Z.; Zhou, F.; Wei, T.; Dong, Y.; Xue, H.; Tang, L.; Liu, M. Scandoside Exerts Anti-Inflammatory Effect Via Suppressing NF-κB and MAPK Signaling Pathways in LPS-Induced RAW 264.7 Macrophages. Int. J. Mol. Sci. 2018, 19, 457. [Google Scholar] [CrossRef] [Green Version]
  169. Lee, D.S.; Keo, S.; Ko, W.; Kim, K.S.; Ivanova, E.; Yim, J.H.; Kim, Y.C.; Oh, H. Secondary metabolites isolated from Castilleja rubra exert anti-inflammatory effects through NF-κB inactivation on lipopolysaccharide-induced RAW264.7 macrophages. Arch. Pharm. Res. 2014, 37, 947–954. [Google Scholar] [CrossRef]
  170. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activa-tion and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  171. Duewell, P.; Kono, H.; Rayner, K.; Sirois, C.; Vladimer, G.; Bauernfeind, F.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 in-flammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Stachon, P.; Heidenreich, A.; Merz, J.; Hilgendorf, I.; Wolf, D.; Willecke, F.; von Garlen, S.; Albrecht, P.; Härdtner, C.; Ehrat, N.; et al. P2X 7 Deficiency Blocks Lesional Inflammasome Activity and Ameliorates Atherosclerosis in Mice. Circulation 2017, 135, 2524–2533. [Google Scholar] [CrossRef] [PubMed]
  173. van der Heijden, T.; Kritikou, E.; Venema, W.; van Duijn, J.; van Santbrink, P.J.; Slütter, B.; Foks, A.C.; Bot, I.; Kuiper, J. NLRP3 Inflammasome Inhibition by MCC950 Reduces Atherosclerotic Lesion Development in Apolipoprotein E-Deficient Mice-Brief Report. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1457–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Williams, J.W.; Huang, L.-H.; Randolph, G.J. Cytokine Circuits in Cardiovascular Disease. Immunity 2019, 50, 941–954. [Google Scholar] [CrossRef]
  175. Maguire, E.M.; Pearce, S.W.; Xiao, Q. Foam cell formation: A new target for fighting atherosclerosis and cardiovascular disease. Vasc. Pharmacol. 2018, 112, 54–71. [Google Scholar] [CrossRef]
  176. Chistiakov, D.A.; Melnichenko, A.A.; Myasoedova, V.A.; Grechko, A.V.; Orekhov, A.N. Mechanisms of foam cell formation in atherosclerosis. J. Mol. Med. 2017, 95, 1153–1165. [Google Scholar] [CrossRef]
  177. Filipek, A.; Mikołajczyk, T.P.; Guzik, T.J.; Naruszewicz, M. Oleacein and Foam Cell Formation in Human Monocyte-Derived Macrophages: A Potential Strategy against Early and Advanced Atherosclerotic Lesions. Pharmaceuticals 2020, 13, 64. [Google Scholar] [CrossRef] [PubMed]
  178. Manning-Tobin, J.J.; Moore, K.J.; Seimon, T.A.; Bell, S.A.; Sharuk, M.; Alvarez-Leite, J.I.; De Winther, M.P.J.; Tabas, I.; Freeman, M.W. Loss of SR-A and CD36 Activity Reduces Atherosclerotic Lesion Complexity Without Abrogating Foam Cell Formation in Hyperlipidemic Mice. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 19–26. [Google Scholar] [CrossRef] [Green Version]
  179. Li, Z.-Q.; Huang, X.-Y.; Hu, C.-Y.; Zhu, Z.-S.; Chen, Y.; Gong, M. Geniposide protects against ox-LDL-induced foam cell formation through inhibition of MAPKs and NF-κB signaling pathways. Pharmazie 2019, 74, 601–605. [Google Scholar]
  180. Sun, J.; Li, X.; Jiao, K.; Zhai, Z.; Sun, D. Albiflorin inhibits the formation of THP-1-derived foam cells through the LOX-1/NF-κB pathway. Minerva Med. 2019, 110, 107–114. [Google Scholar] [CrossRef]
  181. Li, L.; Yin, H. Cholesterol Homeostasis and Liver X Receptor (LXR) in Atherosclerosis. Cardiovasc. Hematol. Disord. Targets 2018, 18, 27–33. [Google Scholar] [CrossRef]
  182. Cho, K.-H. 1,8-cineole protected human lipoproteins from modification by oxidation and glycation and exhibited serum lipid-lowering and anti-inflammatory activity in zebrafish. BMB Rep. 2012, 45, 565–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Jun, H.-J.; Hoang, M.-H.; Yeo, S.-K.; Jia, Y.; Lee, S.-J. Induction of ABCA1 and ABCG1 expression by the liver X receptor modulator cineole in macrophages. Bioorganic Med. Chem. Lett. 2013, 23, 579–583. [Google Scholar] [CrossRef]
  184. Shen, D.; Zhao, D.; Yang, X.; Zhang, J.; He, H.; Yu, C. Geniposide against atherosclerosis by inhibiting the formation of foam cell and lowering reverse lipid transport via p38/MAPK signaling pathways. Eur. J. Pharmacol. 2019, 864, 172728. [Google Scholar] [CrossRef]
  185. Wang, Y.; Li, Z.; Liu, B.; Wu, R.; Gong, H.; Su, Z.; Zhang, S. Isoborneol Attenuates Low-Density Lipoprotein Accumulation and Foam Cell Formation in Macrophages. Drug Des. Dev. Ther. 2020, 14, 167–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Zhang, X.; Xu, F.; Liu, L.; Feng, L.; Wu, X.; Shen, Y.; Sun, Y.; Wu, X.; Xu, Q. (+)-Borneol improves the efficacy of edaravone against DSS-induced colitis by promoting M2 macrophages polarization via JAK2-STAT3 signaling pathway. Int. Immunopharmacol. 2017, 53, 1–10. [Google Scholar] [CrossRef]
  187. Jin, Z.; Li, J.; Pi, J.; Chu, Q.; Wei, W.; Du, Z.; Qing, L.; Zhao, X.; Wu, W. Geniposide alleviates atherosclerosis by regulating macrophage polarization via the FOS/MAPK signaling pathway. Biomed. Pharmacother. 2020, 125, 110015. [Google Scholar] [CrossRef] [PubMed]
  188. Xu, Y.-L.; Liu, X.-Y.; Cheng, S.-B.; He, P.-K.; Hong, M.-K.; Chen, Y.-Y.; Zhou, F.-H.; Jia, Y.-H. Geniposide Enhances Macrophage Autophagy through Downregulation of TREM2 in Atherosclerosis. Am. J. Chin. Med. 2020, 48, 1821–1840. [Google Scholar] [CrossRef] [PubMed]
  189. Isali, I.; McClellan, P.; Shankar, E.; Gupta, S.; Jain, M.; Anderson, J.M.; Hijaz, A.; Akkus, O. Genipin guides and sustains the polarization of macrophages to the pro-regenerative M2 subtype via activation of the pSTAT6-PPAR-gamma pathway. Acta Biomater. 2021, 131, 198–210. [Google Scholar] [CrossRef] [PubMed]
  190. Chen, Q.; Qi, X.; Zhang, W.; Zhang, Y.; Bi, Y.; Meng, Q.; Bian, H.; Li, Y. Catalpol Inhibits Macrophage Polarization and Prevents Postmenopausal Atherosclerosis Through Regulating Estrogen Receptor Alpha. Front. Pharmacol. 2021, 12, 1073. [Google Scholar] [CrossRef] [PubMed]
  191. Zhai, T.; Sun, Y.; Li, H.; Zhang, J.; Huo, R.; Li, H.; Shen, B.; Li, N. Unique immunomodulatory effect of pae-oniflorin on type I and II macrophages activities. J. Pharmacol. Sci. 2016, 130, 143–150. [Google Scholar] [CrossRef] [Green Version]
  192. Li, W.; Zhi, W.; Liu, F.; Zhao, J.; Yao, Q.; Niu, X. Paeoniflorin inhibits VSMCs proliferation and migration by arresting cell cycle and activating HO-1 through MAPKs and NF-κB pathway. Int. Immunopharmacol. 2018, 54, 103–111. [Google Scholar] [CrossRef]
  193. Zhang, J.; Wang, C.; Wang, H.; Li, X.; Xu, J.; Yu, K. Loganin alleviates sepsis-induced acute lung injury by regulating macrophage polarization and inhibiting NLRP3 inflammasome activation. Int. Immunopharmacol. 2021, 95, 107529. [Google Scholar] [CrossRef] [PubMed]
  194. Filipek, A.; Czerwińska, M.E.; Kiss, A.K.; Wrzosek, M.; Naruszewicz, M. Oleacein enhances an-ti-inflammatory activity of human macrophages by increasing CD163 receptor expression. Phytomedicine 2015, 22, 1255–1261. [Google Scholar] [CrossRef] [PubMed]
  195. Shao, B.-Z.; Han, B.-Z.; Zeng, Y.-X.; Su, D.-F.; Liu, C. The roles of macrophage autophagy in atherosclerosis. Acta Pharmacol. Sin. 2016, 37, 150–156. [Google Scholar] [CrossRef] [Green Version]
  196. Sergin, I.; Evans, T.D.; Zhang, X.; Bhattacharya, S.; Stokes, C.J.; Song, E.; Ali, S.; Dehestani, B.; Holloway, K.B.; Micevych, P.S.; et al. Exploiting macro-phage autophagy-lysosomal biogenesis as a therapy for atherosclerosis. Nat. Commun. 2017, 8, 15750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Zhao, P.; Cai, Z.; Tian, Y.; Li, J.; Li, K.; Li, M.; Bai, Y.; Li, J. Effective-compound combination inhibits the M2-like polarization of macrophages and attenuates the development of pulmonary fibrosis by increasing autophagy through mTOR signaling. Int. Immunopharmacol. 2021, 101, 108360. [Google Scholar] [CrossRef]
  198. Moore, K.J.; Sheedy, F.J.; Fisher, E.A. Macrophages in atherosclerosis: A dynamic balance. Nat. Rev. Immunol. 2013, 13, 709–721. [Google Scholar] [CrossRef] [Green Version]
  199. Yang, S.; Yuan, H.-Q.; Hao, Y.-M.; Ren, Z.; Qu, S.-L.; Liu, L.-S.; Wei, D.-H.; Tang, Z.-H.; Zhang, J.-F.; Jiang, Z.-F. Macrophage polarization in athero-sclerosis. Clin. Chim. Acta 2020, 501, 142–146. [Google Scholar]
  200. Wang, N.; Liang, H.; Zen, K. Molecular mechanisms that influence the macrophage m1-m2 polarization balance. Front. Immunol. 2014, 5, 614. [Google Scholar]
  201. Filipek, A.; Czerwińska, M.; Kiss, A.; Polański, J.; Naruszewicz, M. Oleacein may inhibit destabi-lization of carotid plaques from hypertensive patients. Impact on high mobility group protein-1. Phytomedicine 2017, 32, 68–73. [Google Scholar] [CrossRef] [PubMed]
  202. Tabas, I.; Bornfeldt, K.E. Macrophage Phenotype and Function in Different Stages of Atherosclerosis. Circ. Res. 2016, 118, 653–667. [Google Scholar] [CrossRef] [Green Version]
  203. Xu, H.; Jiang, J.; Chen, W.; Li, W.; Chen, Z. Vascular Macrophages in Atherosclerosis. J. Immunol. Res. 2019, 2019, 4354786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Bennett, M.; Sinha, S.; Owens, G. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016, 118, 692–702. [Google Scholar] [CrossRef] [Green Version]
  205. Zhang, F.; Guo, X.; Xia, Y.; Mao, L. An update on the phenotypic switching of vascular smooth muscle cells in the pathogenesis of atherosclerosis. Cell. Mol. Life Sci. 2021, 79, 6. [Google Scholar] [CrossRef]
  206. Grootaert, M.O.J.; Bennett, M.R. Vascular smooth muscle cells in atherosclerosis: Time for a re-assessment. Cardiovasc. Res. 2021, 117, 2326–2339. [Google Scholar] [CrossRef]
  207. Muslin, A.J. MAPK signalling in cardiovascular health and disease: Molecular mechanisms and therapeutic targets. Clin. Sci. 2008, 115, 203–218. [Google Scholar] [CrossRef] [Green Version]
  208. Liang, Y.; Zhong, Y.; Li, X.; Xiao, Y.; Wu, Y.; Xie, P. Biological evaluation of linalool on the function of blood vessels. Mol. Med. Rep. 2021, 24, 874. [Google Scholar] [CrossRef]
  209. Lee, K.P.; Sudjarwo, G.W.; Jung, S.H.; Lee, D.; Lee, D.-Y.; Lee, G.B.; Baek, S.; Kim, D.-Y.; Lee, H.M.; Kim, B.; et al. Carvacrol inhibits atherosclerotic neointima formation by downregulating reactive oxygen species production in vascular smooth muscle cells. Atherosclerosis 2015, 240, 367–373. [Google Scholar] [CrossRef]
  210. Clempus, R.E.; Griendling, K. Reactive oxygen species signaling in vascular smooth muscle cells. Cardiovasc. Res. 2006, 71, 216–225. [Google Scholar] [CrossRef] [PubMed]
  211. Pei, X.; Li, X.; Chen, H.; Han, Y.; Fan, Y. Thymoquinone Inhibits Angiotensin II-Induced Proliferation and Migration of Vascular Smooth Muscle Cells Through the AMPK/PPARγ/PGC-1α Pathway. DNA Cell Biol. 2016, 35, 426–433. [Google Scholar] [CrossRef] [PubMed]
  212. Zhu, N.; Xiang, Y.; Zhao, X.; Cai, C.; Chen, H.; Jiang, W.; Wang, Y.; Zeng, C. Thymoquinone suppresses plate-let-derived growth factor-BB-induced vascular smooth muscle cell proliferation, migration and neointimal formation. J. Cell. Mol. Med. 2019, 23, 8482–8492. [Google Scholar] [CrossRef] [PubMed]
  213. Fan, X.; Wu, J.; Yang, H.; Yan, L.; Wang, S. Paeoniflorin blocks the proliferation of vascular smooth muscle cells induced by platelet-derived growth factor-BB through ROS mediated ERK1/2 and p38 signaling pathways. Mol. Med. Rep. 2018, 17, 1676–1682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Jiang, F.; Jiang, R.; Zhu, X.; Zhang, X.; Zhan, Z. Genipin Inhibits TNF-α-Induced Vascular Smooth Muscle Cell Proliferation and Migration via Induction of HO-1. PLoS ONE 2013, 8, e74826. [Google Scholar] [CrossRef] [PubMed]
  215. Yang, P.S.; Wang, M.J.; Jayakumar, T.; Chou, D.S.; Ko, C.Y.; Hsu, M.J.; Hsieh, C.Y. Antiprolif-erative Activity of Hinokitiol, a Tropolone Derivative, Is Mediated via the Inductions of p-JNK and p-PLCγ1 Signaling in PDGF-BB-Stimulated Vascular Smooth Muscle Cells. Molecules 2015, 20, 8198–8212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Heiss, E.H.; Liu, R.; Waltenberger, B.; Khan, S.; Schachner, D.; Kollmann, P.; Zimmermann, K.; Cabaravdic, M.; Uhrin, P.; Stuppner, H.; et al. Plumericin inhibits proliferation of vascular smooth muscle cells by blocking STAT3 signaling via S-glutathionylation. Sci. Rep. 2016, 6, 20771. [Google Scholar] [CrossRef] [Green Version]
  217. Amirghofran, Z.; Ahmadi, H.; Karimi, M.H.; Kalantar, F.; Gholijani, N.; Malek-Hosseini, Z. In vitro inhibitory effects of thymol and carvacrol on dendritic cell activation and function. Pharm. Biol. 2016, 54, 1125–1132. [Google Scholar]
  218. Xuan, N.T.; Shumilina, E.; Qadri, S.M.; Götz, F.; Lang, F. Effect of thymoquinone on mouse den-dritic cells. Cell. Physiol. Biochem. 2010, 25, 307–314. [Google Scholar] [CrossRef]
  219. Zhang, H.; Qi, Y.; Yuan, Y.; Cai, L.; Xu, H.; Zhang, L.; Su, B.; Nie, H. Paeoniflorin Ameliorates Experimental Au-toimmune Encephalomyelitis via Inhibition of Dendritic Cell Function and Th17 Cell Differentiation. Sci. Rep. 2017, 7, 41887. [Google Scholar] [CrossRef]
  220. Chen, D.; Li, Y.; Wang, X.; Li, K.; Jing, Y.; He, J.; Qiang, Z.; Tong, J.; Sun, K.; Ding, W.; et al. Generation of regulatory dendritic cells after treatment with paeoniflorin. Immunol. Res. 2015, 64, 988–1000. [Google Scholar] [CrossRef]
  221. Abe, R.; Beckett, J.; Nixon, A.; Rochier, A.; Yamashita, N.; Sumpio, B. Olive Oil Polyphenol Oleuropein Inhibits Smooth Muscle Cell Proliferation. Eur. J. Vasc. Endovasc. Surg. 2011, 41, 814–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Liu, L.; Liao, P.; Wang, B.; Fang, X.; Li, W.; Guan, S. Oral administration of baicalin and geniposide induces regression of atherosclerosis via inhibiting dendritic cells in ApoE-knockout mice. Int. Immunopharmacol. 2014, 20, 197–204. [Google Scholar] [CrossRef] [PubMed]
  223. Subramanian, M.; Tabas, I. Dendritic cells in atherosclerosis. Semin. Immunopathol. 2014, 36, 93–102. [Google Scholar] [CrossRef]
  224. Gil-Pulido, J.; Zernecke, A. Antigen-presenting dendritic cells in atherosclerosis. Eur. J. Pharmacol. 2017, 816, 25–31. [Google Scholar] [CrossRef] [PubMed]
  225. Zhao, Y.; Zhang, J.; Zhang, W.; Xu, Y. A myriad of roles of dendritic cells in atherosclerosis. Clin. Exp. Immunol. 2021, 206, 12–27. [Google Scholar] [CrossRef]
  226. Elmowafy, M.; Samy, A.; Raslan, M.A.; Salama, A.; Said, R.A.; Abdelaziz, A.E.; Eleraky, W.; el Awdan, S.; Viitala, T. Enhancement of Bioavailability and Pharmacodynamic Effects of Thymoquinone Via Nanostructured Lipid Carrier (NLC) Formulation. AAPS PharmSciTech 2015, 17, 663–672. [Google Scholar] [CrossRef]
  227. Huguet-Casquero, A.; Xu, Y.; Gainza, E.; Pedraz, J.L.; Beloqui, A. Oral delivery of oleuropein-loaded lipid nanocarriers alleviates inflammation and oxidative stress in acute colitis. Int. J. Pharm. 2020, 586, 119515. [Google Scholar] [CrossRef]
  228. Vaidya, H.B.; Goyal, R.K.; Cheema, S.K. Acetylated and propionated derivatives of swertiamarin have an-ti-adipogenic effects. J. Pharmacol. Pharmacother. 2014, 5, 232–238. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of natural monoterpenes with potential antiatherogenic effects.
Figure 1. Chemical structures of natural monoterpenes with potential antiatherogenic effects.
Ijms 24 02429 g001
Figure 2. Pharmacological actions of natural monoterpenes on atherosclerosis. Atheroprotective monoterpenes can target vascular cells involved in atherosclerosis, including endothelial cells, monocytes/macrophages, VSMCs, and dendritic cells. Moreover, they modulate various atherogenesis-related pathological processes, such as hyperlipidemia, inflammation, oxidative stress, apoptosis, NO bioavailability, foam cell formation, autophagy, and abnormal cell proliferation and migration. This figure schematizes atherosclerotic plaque formation in the middle panel, with various target cell types (or pro-atherogenic factor) and relevant pharmacological actions of antiatherogenic monoterpenes shown around the outer margins.
Figure 2. Pharmacological actions of natural monoterpenes on atherosclerosis. Atheroprotective monoterpenes can target vascular cells involved in atherosclerosis, including endothelial cells, monocytes/macrophages, VSMCs, and dendritic cells. Moreover, they modulate various atherogenesis-related pathological processes, such as hyperlipidemia, inflammation, oxidative stress, apoptosis, NO bioavailability, foam cell formation, autophagy, and abnormal cell proliferation and migration. This figure schematizes atherosclerotic plaque formation in the middle panel, with various target cell types (or pro-atherogenic factor) and relevant pharmacological actions of antiatherogenic monoterpenes shown around the outer margins.
Ijms 24 02429 g002
Figure 3. The cellular and molecular mechanisms of natural monoterpenes in the development of atherosclerosis. Monoterpenes target various pro-atherogenic factors and intracellular molecular pathways in atherogenesis-related cell types, thus leading to the attenuation of atherosclerosis.
Figure 3. The cellular and molecular mechanisms of natural monoterpenes in the development of atherosclerosis. Monoterpenes target various pro-atherogenic factors and intracellular molecular pathways in atherogenesis-related cell types, thus leading to the attenuation of atherosclerosis.
Ijms 24 02429 g003
Table 1. Modulation of serum lipid profile.
Table 1. Modulation of serum lipid profile.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
LinaloolC57BL/6J mice fed with HFDLowers TC, TG, and LDL-C, and improves atherogenic indexSuppresses SREBP-2-mediated HMG-CoA reductase expression and induces ubiquitin-dependent proteolysis of the HMG-CoA reductase[24]
AmarogentinC57BL/6J mouse model of diabetesAttenuates neointimal thickening, and collagen and lipid deposition in the aorta, lowers TC, TG, LDL-C, and VLDL-C, and increases HDL-CInduces phosphorylation of AMPK[26]
OleuropeinWistar rats fed with a cholesterol rich dietDecreases TC and TG, and elevates HDL-CSuppresses lipid synthesis and promotes fatty acid oxidation mediated by AMPK[27]
AucubinC57BL/6J mice treated with tyloxapolDownregulates TC, TG, LDL-C and VLDL-C, and increases HDL-CActivates AMPK and Nrf2, and promotes the expression of PPARα and PPARγ[28]
ThymoquinoneSD rats fed with HFDDecreases TC and LDL-C levelsDownregulates gene expression of HMG-CoA reductase and upregulates the expression LDLR[30]
GeniposideC57BL/6J mice and ApoE−/− mice fed with HFDSuppresses atherosclerotic plaque progression, reduces serum TC and TG, and attenuates hepatic lipid depositionRegulates FXR-mediated liver-gut crosstalk of bile acids[31]
SwertiamarinSD rats fed with a cholesterol rich dietLowers TC, TG, LDL-C, VLDL-C, and atherogenic indexInhibits HMG-CoA reductase activity and enhances the fecal bile acid and total sterols excretion[34]
EucalyptolWistar rat model of diabetic-atherosclerosisPrevents the formation of the atheromatous lesions, reduces TC, TG, and LDL-C, and upregulates HDL-CNot shown[35]
GeraniolSyrian hamsters fed with an atherogenic dietDecreases TC, TG, free fatty acids, phospholipids, LDL-C, and VLDL-C, upregulates HDL-C, and lowers atherogenic indexInhibits HMG-CoA reductase and suppresses lipogenesis[36]
ThymolNew Zealand white rabbits fed with HFDDecreases TC, TG, and LDL-C, and elevates HDL-CNot shown[37]
LimoneneWistar rats fed with an atherogenic dietReduces TG, TC, VLDL-C, LDL-C and non-HDL-C levels, and elevates HDL-C/LDL-C, HDL-C/TC and HDL-C/TGReduces HMG-CoA reductase activity[38]
p-CymeneWistar rat model of diabetesImproves TC, TG, LDL-C, VLDL-C, and HDL-CSuppresses AKT/mTOR signaling[39]
SafranalWistar rat model of diabetesDecreases total lipids, TC, TG, and LDL-C, and elevates HDL-CNot shown[40]
GenipinC57BL/6J mice fed with HFD; mouse primary hepatocytes treated with free fatty acidsAntagonizes HFD-induced hyperlipidemia and hepatic lipid accumulationRegulates miR-142a-5p/SREBP-1c axis[41]
CatalpolNew Zealand white rabbits fed with HFD and EA. hy926 cells treated with ox-LDLAttenuates atherosclerotic lesions, decreases TC, TG, and LDL-C, and increases HDL-CPromotes Nrf2/HO-1-mediated anti-oxidative stress and inhibits NF-κB-mediated inflammation[42]
PaeoniflorinC57BL/6J mice fed with HFDExerts an antagonistic effect on hyperlipidemia and lipid ectopic depositionLowers the lipid synthesis pathway, promotes fatty acid oxidation and increases cholesterol output[43]
Loganic acidNew Zealand rabbits fed with a cholesterol rich dietDecreases intima thickness and intima/media ratio in the thoracic aorta, lowers TG and ox-LDL, and increases HDL-CPromotes the expression of PPARα and PPARγ[44]
LoganinC57BLKS/J type 2 diabetic db/db miceReduces TG and LDL-C/VLDL-C, and increases HDL-CSuppresses gene expressions related to lipid synthesis and adjusts the abnormal expression of PPARα and SREBPs in the nucleus[45]
OleaceinC57BL/6J mice fed with HFDReduces TC, TG, and LDL-CDownregulates the expression of FAS, SREBP-1, and phospho-ERK[46]
GentiopicrosideC57BL/6J mice treated with tyloxapol and HepG2 cells treated with free fatty acidReduces TC and TGRegulates Nrf2-mediated PPARα activation and SREBP-1c inactivation[47]
Table 2. Attenuation of endothelial pro-inflammatory activation.
Table 2. Attenuation of endothelial pro-inflammatory activation.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
EucalyptolHUVECs treated with LPSAttenuates adhesion molecules and pro-inflammatory cytokinesRegulates PPAR-γ dependent modulation of IκBα/NF- κB signaling[52]
CitralHUVECs treated with LPSSuppresses the adhesion of neutrophils to HUVECs, and decreases adhesion molecules and pro-inflammatory cytokinesRegulates PPAR-γ dependent modulation of IκBα/NF- κB signaling[53]
CitronellolBovine arterial endothelial cells treated with LPSSuppresses LPS-induced COX-2 expression and attenuates vascular endothelial inflammationActivates PPARγ signaling[54]
GenipinHUVECs treated with TNF-αAttenuates the adhesion of U937 monocytic cells to HUVECs and ameliorates adhesion moleculesInduces the expression of PPAR-γ[55]
CornusideHUVECs treated with TNF-αAttenuates pro-inflammatory mediator and adhesion moleculesSuppresses NF-κB signaling[63]
HUVECs treated with LPS or HMGB1Inhibits endothelial permeability, decreases pro-inflammatory mediators, and reduces adhesion eventsModulates SIRT1/HMGB1-mediated NF-κB, ERK and p38 MAPK signaling[57]
PaeoniflorinHUVECs treated with LPSSuppresses the expression of adhesion molecules and pro-inflammatory cytokinesDecreases the activation of IκBα/NF- κB, p38 MAPK and JNK pathway[61]
HUVECs treated with ox-LDLAttenuates adhesion molecule expressionEnhances autophagy via upregulation of SIRT1[64]
HUVECs treated with LPCSuppresses LPC-induced inflammatory factor productionInhibits the HMGB1-RAGE/TLR-2/TLR-4-NF-κB pathway[58]
HUVECs treated with LPSSuppresses pro-inflammatory cytokines and chemokineInhibits ER stress-dependent IRE1α /NF-κB signaling[65]
CatalpolHuman aortic endothelial cells treated with homocysteineInhibits the expression of adhesion molecules and chemokineSuppresses ER stress and NF-κB signaling[66]
GeniposideHUVECs treated with LPSInhibits LPS-induced expression of IL-6 and IL-8, and suppresses U937 monocyte adhesion to HUVECsAttenuates IκBα/NF-κB, p38 MAPK and ERK signaling[62]
HUVECs treated with a high level of glucoseSuppresses the adhesion of monocytes to HUVECs and reduces adhesion moleculesAttenuates ROS/NF-κB signaling[60]
HUVECs treated with ox-LDLDecreases the production of pro-inflammatory cytokinesEnhances the miR-21/PTEN pathway[67]
ApoE−/− mice fed with HFD and HUVECs treated with H2O2Suppresses atherosclerosis and inhibits endothelial inflammationModulates AMPK/mTOR/Nrf2 signaling pathway[68]
Bornyl acetateHUVECs treated with ox-LDLSuppresses the attachment of THP-1 monocytes to HUVECs, and ameliorates adhesion molecules and pro-inflammatory cytokinesMitigates the activation of the IκBα/NF-κB signaling pathway[69]
CarvacrolC57BLKS/J type 2 diabetic db/db mice and HUVECs treated with a high level of glucoseAlleviates the histological abnormalities of the abdominal aorta and reduces vascular inflammationReduces the activation of the TLR4/NF-κB signaling[70]
HinokitiolSEVC4-10 endothelial cells treated with culture medium of LPS-stimulated RAW 264.7 cellInhibits pro-inflammatory cytokine-induced adhesion moleculesNot shown[71]
ThymoquinoneHUVECs treated with LPSSuppresses pro-inflammatory cytokines and chemokineModulates TET2/NLRP3 inflammasome axis[72]
AmarogentinC57BL/6J diabetic mice, and EAhy926 cells or HUVECs treated with TNF-αExerts anti-atherosclerotic effects and inhibits endothelial inflammation and the adherence of THP-1 monocytes onto HUVECsRegulates AMPK/NF-κB pathway[26]
OleuropeinHUVECs treated with LPS, TNF-α or PMASuppresses the adhesion of monocytes to HUVECs and reduces the adhesion molecule VCAM-1Inhibits the binding of NF-κB, AP-1, and possibly GATA to the promoter of VCAM-1[73]
OleaceinHUVECs treated with LPS or TNF-αSuppresses the adhesion of monocytes to HUVECs and reduces adhesion molecules and pro-inflammatory chemokineInhibits NF-κB-mediated CCL2 expression[74]
PlumericinHUVECs treated with TNF-αAttenuates adhesion molecule expressionRegulates IKK/IκB/NF-κB pathway[75]
Picroside IIHUVECs treated with homocysteineReduces the production of inflammatory mediatorsModulates the SIRT1/LOX-1/NF-κB signaling pathway[76]
MonotropeinHUVECs treated with H2O2Alleviates the inflammatory response of HUVECsAttenuates NF-κB/AP-1 signaling[77]
AlbiflorinHUVECs treated with ox-LDLAlleviates the production of pro-inflammatory cytokinesBlocks IRAK1/TAK1 pathway[78]
Table 3. Inhibition of endothelial oxidative stress and modulation of nitric oxide pathway.
Table 3. Inhibition of endothelial oxidative stress and modulation of nitric oxide pathway.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
GeraniolC57BL/6J mice fed with HFD and HUVECs treated with palmitic acidProtects against HFD-induced endothelial dysfunctionDecreases the expression of NOX-2 to suppress ROS production[81]
Syrian hamsters fed with an atherogenic dietAttenuates endothelial dysfunction and prevents tissue oxidative injuryIncreases the expression of Nrf2, inhibits lipid peroxidation and reduces antioxidant enzymes (SOD, CAT, GPx, and GR)[87]
PaeoniflorinHUVECs treated with H2O2Attenuates H2O2-induced endothelial cell damageIncreases the expression of SIRT1 and modulate balance between eNOS/iNOS[88]
HUVECs treated with H2O2Suppresses H2O2-induced oxidative stressScavenges intracellular ROS, and rescues abnormalities of MDA, SOD and GSH-Px[89]
HUVECs treated with TBHPSuppresses TBHP-induced oxidative damageActivates Nrf2/HO-1 signaling to reduce ROS level and increase activities of CAT, GPx, and SOD[90]
HUVECs treated with AOPPsProtects against AOPP-induced oxidative damageSuppresses ROS generation through the inhibition of RAGE-NOX2/NOX4[82]
HarpagosidebEnd.3 endothelial cells treated with Ang IIInhibits Ang II-induced oxidative stressDecreases NOX2/NOX4/COX-2/ROS and lipid peroxidation level[83]
ThymoquinoneRabbit aortic rings treated with pyrogallolExerts antioxidant capacity, increases NO production, and improves pyrogallol-induced endothelial dysfunctionReduces lipid peroxidation and enhances activity or content of SOD and GSH[84]
PerillaldehydeRats and ApoE−/− mice with HFD or plus balloon injury and HUVECs treated with ox-LDLSuppresses oxidative stress to improve endothelial dysfunction with increased NO generation and inhibits atherosclerosisIncreases endogenous BH4 generation, rescues abnormalities of ROS, MDA and SOD, and elevates consequent eNOS recoupling[85]
CitronellalSD rats fed with HFD plus balloon injurySuppresses oxidative stress, increases NO production, and prevents endothelial dysfunction and the progression of atherosclerosisDownregulates the expression of NHE1 and rescues abnormalities of MDA and SOD activity[86]
SD rats fed with HFD plus streptozotocin (STZ) administration, and HUVECs treated with a high level of glucoseAlleviates oxidative stress, increases NO production, and improves high glucose-induced endothelial injuryInduction of S1P/S1P1 signaling, increases eNOS expression, recouples eNOS, and rescues abnormalities of NOx, ROS, MDA and SOD and other anti-oxidant enzymes[91]
GeniposideHUVECs treated with ox-LDLInhibits ox-LDL-induced oxidative stressModulates the miR-21/PTEN/NOX2 pathway, and rescues abnormalities of ROS, MDA, SOD, GSH-Px, and CAT[67]
ApoE−/− mice fed with HFD and HUVECs treated with H2O2Suppresses atherosclerosis and inhibits ox-LDL-induced oxidative stressModulates AMPK/mTOR/Nrf2 pathway and rescues abnormalities of NOX2, ROS, MDA, GSH, and SOD[68]
MonotropeinHUVECs treated with H2O2Ameliorates H2O2-mediated oxidative injuryAttenuates NF-κB/AP-1 signaling and rescues abnormalities of MDA, SOD, and GSH-Px[77]
EucalyptolHUVECs treated with a high level of glucoseProtects against high glucose-induced vascular endothelial injuryModulation of Keap1/Nrf2/HO-1 signaling to reduce ROS generation[92]
HUVECs treated with LPSSustains the balance of endothelial NO and ameliorates LPS-induced HUVEC injurySuppresses NF-κB signaling to reduce iNOS-derived NO, and recovers eNOS-derived NO to the normal level[93]
CatalpolNew Zealand white rabbits fed with HFD and EA.hy926 cells treated with ox-LDLExerts beneficial effects on atherosclerosis progression, oxidative stress and inflammationRescues abnormalities of MDA, SOD and GSH-Px, and induces the activation of Nrf2/HO-1 axis in HUVECs[42]
Mouse glomerular endothelial cells treated with AGEAmeliorates AGEs-induced endothelial dysfunctionInhibits the NF-κB/iNOS pathway and activates the PI3K/AKT/eNOS pathway[94]
Human aortic endothelial cells treated with homocysteineInhibits homocysteine-induced oxidative damageDecreases NOX4/ROS signaling and rescues abnormalities of MDA and GSH[66]
CitralHUVECs treated with H2O2Exerts antioxidant capacity and protects against oxidative damage induced by H2O2 in HUVECsReduces hydroperoxide levels and elevates total antioxidant activity[95]
GenipinHUVECs treated with thrombinInhibits thrombin-induced VWF release and P-selectin translocation in HUVECsActivates eNOS phosphorylation, promotes enzyme activation and increases NO production[96]
SafranalBovine aortic endothelial cells treated with H2O2Suppresses H2O2-induced oxidative stressDecreases ROS production[97]
AmarogentinC57BL/6J diabetic mice and EA.hy926 cells or HUVECs treated with TNF-αExerts anti-atherosclerotic effects and inhibits endothelial dysfunctionRegulates AMPK/eNOS pathway[26]
Picroside IIHUVECs treated with homocysteineAttenuates homocysteine-induced oxidative stressModulates the SIRT1/LOX-1 pathway, reduces ROS production, and rescues abnormalities of NOX, MDA, SOD, and CAT[76]
AucubinHUVECs treated with ox-LDLImproves vascular endothelial dysfunctionReduces ROS generation and protects against eNOS uncoupling[98]
Table 4. Attenuation of endothelial cell apoptosis.
Table 4. Attenuation of endothelial cell apoptosis.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
CatalpolHUVECs treated with H2O2Attenuates H2O2-induced apoptosis in HUVECsActivates PI3K/AKT pathway, decreases Bax and cleaved Caspase-3, and increases Bcl-2 and p-Bad[109]
Human aortic endothelial cells treated with homocysteineProtects against endothelial apoptosis induced by homocysteineInhibits ER stress-mediated apoptosis, enhances Bcl-2 and mitochondrial membrane potential, and reduces Bax, cleaved caspase-3, caspase-9 and cytochrome c release[66]
PaeoniflorinHUVECs treated with ox-LDLAttenuates ox-LDL-induced apoptosis in HUVECsEnhances autophagy via upregulation of SIRT1, decreases Bax, and increases Bcl-2[64]
HUVECs treated with TBHPSuppresses apoptosis in HUVECs mediated by TBHPActivates Nrf2/HO-1 signaling, decreases Bax, cleaved Caspase-3 and cytochrome c release, and increases Bcl-2[90]
HUVECs treated with H2O2Protects against HUVEC apoptosis induced by H2O2Suppresses ERK signaling and Caspase-3 activity[89]
GeniposideHUVECs treated with ox-LDLInhibits apoptosis in HUVECs subjected to ox-LDLModulates the miR-21/PTEN pathway, decreases Bax and Caspase-3 activity, and increases Bcl-2 and mitochondrial membrane potential[67]
ApoE−/− mice fed with HFD and HUVECs treated with H2O2Inhibits the growth of atherosclerosis and ameliorates H2O2-induced apoptosis in HUVECsModulates AMPK/mTOR/Nrf2 pathway, decreases Bax and Caspase-3, and increases Bcl-2[68]
Picroside IIHUVECs treated with homocysteineProtects against endothelial apoptosis induced by homocysteineModulates SIRT1/LOX-1 pathway, and reduces cleaved Caspase-3 as well as Caspase-3 activity[76]
MonotropeinHUVECs treated with H2O2Attenuates H2O2-induced apoptosis in HUVECsAttenuates NF-κB/AP-1 signaling, decreases Bax and cleaved Caspase-3, increases Bcl-2[77]
AlbiflorinHUVECs treated with ox-LDLAlleviates ox-LDL-induced apoptosis in HUVECsBlocks IRAK1/TAK1 pathway, and decreases Bax and Caspase-3[78]
HarpagosidebEnd.3 endothelial cells treated with Ang IISuppresses Ang II-induced apoptosis in HUVECsKeeps Bax/Bcl-2 balance, decreases cytochrome c release, and inactivates caspase-8, caspase-9, and caspase-3[83]
SafranalBovine aortic endothelial cells treated with H2O2Suppresses H2O2-induced endothelial apoptosisModulates MAPK signaling, decreases Caspase-3 and cytochrome c release, and increases Bcl-2 and survivin[97]
Table 5. Reduction of macrophage-related inflammation.
Table 5. Reduction of macrophage-related inflammation.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
Bornyl acetateRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, and IL-6Suppresses p38 MAPK/JNK/ERK signaling and IκBα/NF-κB signaling[128]
EucalyptolMurine peritoneal macrophages, BMDMs, or alveolar macrophages treated with LPS or LPS+ATPDecreases TNF-α, IL-1α, IL-1β, IL-6, COX-2, iNOS, and NO and increases IL-10Suppresses NF-κB, JNK, p38 MAPK, STAT3, and NLRP3 inflammasome activation[129,130,131]
α-PineneMurine peritoneal macrophages treated with LPSDecreases IL-6, TNF-α, COX-2, iNOS, PGE2, and NOSuppresses JNK, ERK, and IKK/NF-κB signaling[132]
GeraniolRAW 264.7 macrophages treated with LPSDecreases COX-2, iNOS, PGE2, and NOSuppresses IκBα/NF-κB signaling[133]
LinaloolRAW 264.7 macrophages treated with LPSDecreases TNF-α and IL-6Suppresses p38 MAPK/JNK/ERK and IκBα/NF-κB signaling[134]
CitralMurine alveolar macrophages or J774A.1 macrophages treated with LPS or LPS+ATPDecreases TNF-α, IL-1β, and IL-6Regulates PPAR-γ/NF-κB signaling and inhibits NLRP3 inflammasome activation[119,135]
CitronellolRAW 264.7 macrophages treated with LPSDecreases COX-2, iNOS, PGE2, and NOSuppresses IκBα/NF-κB signaling[133]
CarvacrolTHP-1 cells, J774A.1 cells, or macrophage-like U937 cells treated with LPS or LPS+ATPDecreases TNF-α, IL-1β, IL-18, and COX-2Suppresses NF-κB, JNK, ERK, STAT-3, AP-1, NFATs, and NLRP3 inflammasome and activates PPAR-γ[120,136,137]
ThymolRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, COX-2, and NOSuppresses NF-κB, MAPK, STAT-3, AP-1, and NFATs[137,138]
PerillaldehydeRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, and IL-6Suppresses JNK signaling[139]
HinokitiolRAW 264.7 macrophages treated with LPSDecreases TNF-αSuppresses the phosphorylation of PDK1, AKT/PKB, and ERK and consequently reduces NF-κB activation[140]
CarvoneRAW 264.7 macrophages treated with LPSAnti-inflammatory effectSuppresses JNK signaling and promotes SIRT1-mediated NF-κB-p65 deacetylation[116]
p-CymeneRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, and IL-6Suppresses p38 MAPK/JNK/ERK signaling and IκBα/NF-κB signaling[141]
ThymoquinoneRAW 264.7 macrophages treated with LPSDecreases iNOS, COX-2, TNF-α, IL-1β, and IL-6Suppresses IRAK1-linked AP-1/NF-κB pathways[142]
Gamma-TerpineneMurine peritoneal macrophages treated with LPSDecreases IL-1β and IL-6 and enhances IL-10Promotes the PGE2/IL-10 axis[143]
SafranalRAW 264.7 macrophages or J774A.1 cells treated with LPS or LPS+ATPDecreases TNF-α, IL-1β, IL-6, COX-2, iNOS, and NOSuppresses MAPK/AP-1 and IKK/NF-κB signaling and inhibits NLRP3 inflammasome activation[144,145]
GeniposideApoE−/− mice fed with HFD, RAW 264.7 macrophages or primary mouse macrophages treated with LPS or LPS+ATPAttenuates atherosclerosis and decreases TNF-α, IL-1β, and IL-6Modulates miR-101/ MKP-1/p38, TLR4-mediated NF-κB and MAPK signaling, and AMPK/SIRT1/NLRP3 inflammasome activation[113,114,115]
GenipinRAW 264.7 macrophages treated with LPSDecreases NO, iNOS and COX-2Suppresses IκBβ/NF-κB and promotes PI3K/JNK/Nrf2/HO-1 signaling[121,146]
CatalpolMurine alveolar macrophages or THP-1 cells treated with LPSDecreases TNF-α, IL-1β, IL-6, and IL-4 and increases IL-10Suppresses NLRP3 inflammasome activation and TLR4-mediated NF-κB and MAPK signaling[147,148]
SwertiamarinRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, IL-6, IL-8, iNOS, and COX-2 and increases IL-10 and IL-4Suppresses IκBα/NF-κB and JAK2/STAT3 signaling and targets the AKT-PH domain to reduce the phosphorylation of AKT[149,150]
PaeoniflorinRAW 264.7 macrophages or THP-1 cells treated with AGEs or LPSDecreases TNF-α, IL-1β, IL-6, IL-33, MCP-1, and iNOSSuppresses miR-124, TLR2/4, NF-κB and p38 MAPK with the regulation of Ca2+ mobilization and modulates the SOCS3-ASK1-p38 pathway[127,151,152,153]
MBPFRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, and NOSuppresses NF-κB, MAPK and PI3K/AKT[154]
CornusideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, IL-6, iNOS, COX-2, NO, and PGE2Suppresses IκBα/NF-κB and MAPK signaling[155]
LoganinMice fed with dextran sulfate sodium, RAW 264.7 macrophages, or BMDMs treated with LPS or LPS+MSUDecreases TNF-α, IL-1β, IL-6, MCP-1, CXCL10, iNOS, COX-2, NO, and PGE2Suppresses SIRT1/NF-κB and mitochondrial dysfunction-mediated NLRP3 inflammasome activation and induces Nrf2/HO-1 signaling[117,122,156]
OleuropeinJ774A.1 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, COX-2, and NOModulates CD14/TLR4-MyD88-NF-κB/MAPK pathways[157]
OleaceinTHP-1 cells treated with LPSDecreases TNF-α, IL-1β, IL-6, NO, and PGE2 and increases IL-10Suppresses TLR4/MyD88/NF-κB Pathway[158]
OleocanthalMouse peritoneal Macrophages treated with LPSDecreases TNF-α, IL-1β, IL-6, IL-17, IL-18, INF-γ, iNOS, COX-2, NO, and PGE2Activates Nrf2/HO-1 and inhibits MAPK and NLRP3 inflammasome activation[123]
Picroside IITHP-1 cells treated with LPS+ATPDecreases IL-1βSuppresses NF-κB-mediated NLRP3 inflammasome activation[159]
GentiopicrosideRAW 264.7 macrophages or primary mouse macrophages treated with LPS+INF-γ or LPS+MSUDecreases TNF-α, IL-1β, IL-6, IL-18, CCL-5, CXCL10, and iNOSSuppresses IKKα/β/NF-κB signaling and NLPR3 inflammasome activation[160,161]
AucubinRAW 264.7 macrophages and THP1 cells treated with LPSDecreases TNF-α, IL-1β, iNOS, and COX-2Induces the AMPK/Nrf2 pathway[124]
HarpagosideRAW 264.7 macrophages treated with LPSDecreases iNOS and COX-2Suppresses IκBα/NF-κB signaling[162]
Scropolioside BTHP-1 cells treated with LPSDecreases TNF-α, IL-1β, and IL-32Suppresses NLRP3 inflammasome activation[147]
CatalposideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, and IL-6Suppresses the binding of LPS to CD14 on the surface of cells thereby inhibiting NF-kB signaling[163]
MonotropeinRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, iNOS, COX-2, NO, and PGE2Suppresses IKKβ/NF-κB and MAPK signaling[164]
Asperulosidic AcidRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, COX-2, NO, and PGE2Suppresses JNK, ERK, and IκBα/NF-κB signaling[165]
AsperulosideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, COX-2, NO, and PGE2Induces Nrf2/HO-1 and suppresses MAPK and IκBα/NF-κB signaling[125,165]
SwerosideBMDMs or RAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, IL-6, COX-2, iNOS, PGE2, and NO and increases IL-10Increases SIRT1 signaling and suppresses NLRP3 inflammasome activation[118,166]
NuezhenideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, COX-2, and NOSuppresses IKKα/β/NF-κB signaling[167]
MorronisideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, iNOS, COX-2, NO, and PGE2Modulates TLR4/NF-κB and Nrf2/HO-1 signaling[126]
ScandosideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-6, iNOS, COX-2, NO, and PGE2Suppresses IκBα/NF-κB and MAPK signaling[168]
MussaenosideRAW 264.7 macrophages treated with LPSDecreases TNF-α, IL-1β, iNOS, COX-2, NO, and PGE2Suppresses NF-κB signaling[169]
Table 6. Modulation of foam cell formation, autophagy and M2 macrophage polarization.
Table 6. Modulation of foam cell formation, autophagy and M2 macrophage polarization.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
IsoborneolRAW 264.7 macrophages treated with ox-LDLReduces the absorption of ox-LDL and the accumulation of intracellular lipidsModulation of cell migration and polarity-related pathways may be involved[185]
RAW 264.7 macrophages treated with LPS+INF-γ or IL-4+IL-13Promotes M2 macrophage polarization as shown by elevated expression of CD206, Arg-1 and IL-10Activates the JAK2-STAT3 signaling pathway[186]
EucalyptolTHP-1 or RAW 264.7 macrophages treated with ox-LDLSuppresses foam cell formation and promotes cholesterol effluxUpregulates the expression of LXRs and their target genes ABCA1 and ABCG1[182,183]
GeniposideApoE−/− mice fed with HFD, RAW 264.7 cells treated with LPAInhibits atherosclerosis and attenuates foam cell formation by regulating both lipid uptake and effluxSuppresses p38 MAPK and AKT signaling pathways[184]
BMDMs treated with ox-LDLInhibits foam cell formation and inflammatory responseSuppresses CD36 expression and NF-κB and MAPK signaling pathways[179]
New Zealand rabbits fed with HFDInhibits atherosclerosis, suppresses M1 macrophage polarization, and promotes M2 polarizationSuppresses the FOS/ MAPK signaling pathway[187]
ApoE−/− mice fed with HFD, RAW 264.7 cells treated with ox-LDLInhibits the progression of atherosclerosis and reinforces macrophage autophagySuppresses the TREM2/mTOR axis[188]
GenipinBMDMs (M0, M1, M2-type)M2 polarization induction and maintenance, along with suppressed pro-inflammatory M1/iNOS responseActivates the pSTAT6/PPARγ pathway[189]
CatalpolPostmenopausal atherosclerosis mouse model, J774A-1 macrophages treated with LPS+INF-γPrevents postmenopausal atherosclerosis, suppresses M1 macrophage polarization and promotes M2 polarizationIncreases the expression of ERα[190]
PaeoniflorinMouse BMDMs treated with LPS or IL-4Suppresses M1 macrophage polarization and promotes M2 polarizationDecreases NF-κB and increases STAT6 signaling[191]
Mouse peritoneal macrophages treated with ox-LDLAttenuates ox-LDL-induced foam cell formationSuppression of NF-κB, ERK and p38 MAPK may be involved[192]
AlbiflorinTHP-1 cells treated with ox-LDLBlocks foam cell formationModulates the LOX-1/NF-κB signaling pathway[180]
LoganinRAW 264.7 macrophages or peritoneal macrophages treated with LPSSuppresses M1 macrophage polarization and promotes M2 polarizationSuppresses ERK and NF-κB signaling[193]
OleaceinHuman monocyte-derived macrophages treated with ox-LDLDecreases foam cell formation, reduces apoptosis, and shifts the polarization towards M2 macrophage phenotypeSuppresses the expression of CD36, SRA1 and LOX-1 and activates JAK/STAT3 pathway[177]
Human monocyte-derived macrophages treated with hemoglobin/haptoglobin complexesEnhances M2 macrophage phenotypeIncreases the expression of CD163 and IL-10 receptors as well as HO-1[194]
Table 7. Modulation of VSMC proliferation and migration and inhibition of dendritic cell maturation.
Table 7. Modulation of VSMC proliferation and migration and inhibition of dendritic cell maturation.
MonoterpeneExperimental ModelEfficacyTarget/PathwayRefs.
LinaloolRat aortic VSMCs (A7r5) treated with Ang IIInhibits Ang II-induced VSMC proliferation and migrationSuppresses CHRM3-mediated p38 MAPK/JNK/ERK signaling[208]
CarvacrolRat aortic VSMCs treated with PDGF-BBInhibits VSMC proliferation and migration and attenuates atherosclerotic neointima formationSuppresses ROS-mediated p38 MAPK/ERK signaling[209]
Mouse splenic dendritic cellsInhibits dendritic cell maturation and adaptive immunitySuppresses CD40[217]
HinokitiolRat aortic VSMCs treated with PDGF-BBInhibits the PDGF-BB-stimulated proliferation of VSMCsSuppresses JNK and PLC-γ1 and induces p27kip1 expression[215]
ThymoquinoneRat aortic VSMCs treated with Ang II or PDGF-BBInhibits VSMC proliferation, migration, and neointimal formation and promotes VSMC apoptosisPromotes AMPK/PPARγ/PGC-1α and inhibits p38 MAPK and MMP-2[211,212]
BMDCs treated with LPSInhibits maturation, cytokine release and survival of dendritic cellsSuppresses CD11c, CD86, MHCII, CD54, and CD40 and reduces PI3K/AKT and ERK signaling[218]
GenipinRat aortic VSMCs treated with TNF-αInhibits TNF-α-induced VSMC proliferation and migrationSuppresses ERK and AKT signaling via upregulating HO-1 expression[214]
PaeoniflorinRat aortic VSMCs treated with ox-LDL or PDGF-BBInhibits VSMCs proliferation, migration and inflammationActivates HO-1 and then inhibits ROS-mediated p38 MAPK, ERK and NF-κB pathways[192,213]
BMDCs or monocyte-derived dendritic cells treated with LPSInhibits dendritic cell function, impairs Th17 cell differentiation, and generates regulatory dendritic cellsReduces IKK/NF-κB and JNK-mediated IL-6 and costimulatory molecule expression and induces TGF-β/IDO signaling[219,220]
OleuropeinBovine vascular SMCsInhibits SMC proliferation with a cell cycle block between the G1 and the S phasesSuppresses ERK signaling[221]
PlumericinRat aortic VSMCs stimulated with serumArrested VSMCs in the G1/G0-phase of the cell cycleSuppresses STAT3 signaling via S-glutathionylation[216]
ThymolMouse splenic dendritic cellsInhibits dendritic cell maturation and adaptive immunitySuppresses CD86[217]
GeniposideApoE−/− mice fed with HFDAttenuates atherosclerosis and inhibits dendritic cell maturation in bone marrow and infiltration into lesionsSuppresses CD11c, CD80, CD86, and CD83 in bone marrow or in atherosclerotic lesions[222]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, J.; Zhong, C.; Yu, J. Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis. Int. J. Mol. Sci. 2023, 24, 2429. https://doi.org/10.3390/ijms24032429

AMA Style

Yang J, Zhong C, Yu J. Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis. International Journal of Molecular Sciences. 2023; 24(3):2429. https://doi.org/10.3390/ijms24032429

Chicago/Turabian Style

Yang, Jing, Chao Zhong, and Jun Yu. 2023. "Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis" International Journal of Molecular Sciences 24, no. 3: 2429. https://doi.org/10.3390/ijms24032429

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop