Open Access

Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review)

  • Authors:
    • Ruiying Wang
    • Min Wang
    • Jingxue Ye
    • Guibo Sun
    • Xiaobo Sun
  • View Affiliations

  • Published online on: November 24, 2020     https://doi.org/10.3892/ijmm.2020.4798
  • Pages: 65-76
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Atherosclerosis (AS) is a chronic disease with a complex pathology that may lead to several cardiovascular and cerebrovascular diseases; however, further research is necessary to fully elucidate its pathogenesis. The main risk factors for AS include lipid metabolism disorders, endothelial cell injury, inflammation and immune dysfunction, among which vascular endothelial cell damage is considered as the main trigger for AS occurrence and development. Endothelial cell damage leads to enhanced intimal permeability and leukocyte adhesion, promoting thrombus formation and accelerating disease progression. The function of endothelial cells is affected by glycolysis regulation, since 80% of ATP in these cells is produced via this pathway. Genes associated with AS and endothelial cell glycolysis, including AKT1, interleukin‑6, vascular endothelial growth factor A, TP53, signal transducer and activator of transcription 3, SRC and mitogen‑activated protein kinase 1, were screened. Through integrated analysis, these genes were found to play a key role in AS by regulating multiple signaling pathways associated with cell signal transduction, energy metabolism, immune function and thrombosis. In conclusion, endothelial cell injury in AS may be alleviated by glycolysis and is a potential clinical treatment strategy for AS.

1. Introduction

Atherosclerosis (AS) is a chronic disease with complex etiology, which involves early local injury of the arterial intima, followed by lipid deposition, proliferation of the intimal fibrous tissue, local thickening of the intima and, ultimately, plaque formation (1). Vascular plaque-induced stenosis by AS may lead to insufficient arterial blood supply and cardiovascular diseases (2). The most serious complications caused by plaque rupture are myocardial infarction, cerebral ischemia, and ischemia of the surrounding tissue (3). The pathogenesis of AS has not been fully elucidated to date. Previous studies have demonstrated that AS is associated with lipid metabolism disorders, endothelial cell damage, inflammation and immune dysfunction, involving macrophages, endothelial cells, vascular smooth muscle cells and platelets (4,5). In recent years, AS animal models mainly include mice, rabbits, miniature pigs, non-human primates and transgenic animals (6). AS is a vicious circle combining multiple factors and long-term effects. So, elucidating the underlying mechanism is crucial for the treatment and prevention of the disease.

Despite not having been fully elucidated, it is believed that lipid metabolism disorders, endothelial cell injury, inflammation and immune dysfunction are the most important factors implicated in the pathogenesis of AS (5,7). Functional damage of endothelial cells is the initiating step in the early stage of AS (8). Endothelial cell damage in AS plaques leads to further plaque instability, rupture (9) and secondary thrombosis, thus accelerating disease progression and affirming the important role of endothelial cell integrity (10). Glycolysis, the most important energy source for endothelial cells, is used to quickly produce energy, enabling cells to respond to environmental changes (11). Intermediate metabolic products produced during glycolysis affect cell survival (12); therefore, glycolytic rates in endothelial cells play a key role in maintaining their homeostasis and reducing the risk for AS.

The aim of the present study was to review the pathogenesis of AS, the role of endothelial cell damage and glycolysis, and the role of associated target genes and the involved signalling pathways, in order to indicate new approaches to the research on AS pathogenesis and intervention methods, and aid in the development of novel treatments for AS.

2. Factors implicated in AS

Pathogenesis

The pathogenesis of AS is extremely complicated (Fig. 1). AS is currently considered to be the result of the interaction among various mechanisms, including lipid metabolism disorder, inflammatory cell infiltration, oxidative stress, immune dysfunction and vascular endothelial cell damage, the latter of which ultimately leads to plaque rupture and thrombosis, leading to serious cardiovascular and cerebrovascular diseases (1,4,13).

Role of oxidative stress in AS

Oxidative stress is the initiating factor of AS inflammatory response, with reactive oxygen species (ROS) and oxidized low-density lipoprotein (Ox-LDL; formed by oxidative modification of LDL) being the main factors responsible for endothelial cell damage and for inducing the expression of pro-inflammatory factors in endothelial cells (14,15). When endothelial cells are continuously exposed to external as well as endogenous oxidants, oxidative stress is likely to induce production of various biologically active substances, which may cause endothelial cell functional damage and apoptosis (16,17). This process leads to the synthesis and release of inflammatory factors, further aggravating vascular inflammation (18). In addition, oxidative stress regulates the expression of vascular wall genes by acting on transcription factors of the vascular wall cells. For example, intracellular ROS directly oxidatively modify the transcription factor itself, thereby participating in the occurrence and development of AS (14).

Role of lipid metabolism disorder in AS

Excessive blood lipid levels are the main cause of AS. In the hyperlipidemic state, the elevated plasma LDL cholesterol (LDL-C) is deposited on the vascular intima and enters macrophages via membrane receptors (19). In addition, LDL also undergoes oxidative modification to form Ox-LDL, leading to changes in endothelial cell function, and causing increased permeability and lipid deposition in the inner membrane (20). Ox-LDL exhibits strong affinity for scavenger receptors found on mononuclear macrophages, leading to its quick internalisation (21). However, Ox-LDL is toxic for macrophages, causing them to become activated, rapidly proliferate, aggregate and degenerate (22). Finally, the macrophages undergo apoptosis and become foam cells, which then aggregate to form AS lipid plaques. Moreover, Ox-LDL binds to vascular endothelial cells through lectin-like oxidized LDL receptor-1 to disrupt intra-cellular signaling and cause endothelial cell dysfunction (23). Ox-LDL can also promote the continuous proliferation of vascular smooth muscle cells and their outward migration to form plaques on the inner wall of blood vessels.

Role of endothelial cell injury in AS

Disruption of endothelial cell morphology and function leads to vascular barrier function impairment, as well as to changes in the intimal integrity and permeability (24). The apoptosis and shedding of endothelial cells promote the adhesion and aggregation of platelets from the blood (25). Dysfunctional endothelial cells, macrophages and platelets secrete a variety of growth factors and vasoactive substances, stimulating the continuous proliferation of smooth muscle cells in the media, and enter the intima, while also causing contractions of the vascular wall (26). As a result, the fatty plaques increase in size while the lumen becomes progressively narrowed, promoting the formation of AS lesions (8).

Role of inflammation in AS

AS involves not only lipid deposition in blood vessel walls, but also chronic inflammation (27). Oxidative stress persists throughout AS. AS has been proven to be a chronic inflammatory disease initiated in the arterial wall, mainly driven by modification of endogenous structures and dysfunction of the vascular endothelium (28). Certain lipids act as signaling molecules, and bind to cell receptors to activate the expression of specific genes and produce a number of pro-inflammatory cytokines (19). This leads to an increase in the numbers of inflammatory cells, including macrophages, increased phagocytosis of Ox-LDL and production of pro-inflammatory factors, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1 and IL-8 (29). These factors further aggravate the pathology of AS.

Role of immune dysfunction in AS

AS is also an autoimmune disease, stimulated by accumulated lipoproteins, as well as specific T lymphocytes and their antibodies, in the blood vessel wall (30). It was previously demonstrated that T lymphocytes infiltrate the aorta, where they accumulate and express restricted T-cell receptors, thereby promoting AS through immune regulation (13). In addition, the cells of innate immune response, such as monocytes and neutrophils, play an important role in the occurrence and development of AS (31). Upon immune function impairment, these cells disrupt cytokine production (27). Thus, the expression of anti-inflammatory factors decreases and the expression of pro-inflammatory factors increases, ultimately promoting AS development (29).

Role of microRNAs (miRNA) of the circulatory system in AS

miRNAs are found in non-coding regions of the genome and play important roles in gene transcription, post-transcriptional processing, cell proliferation, cell differentiation, cell apoptosis, ontogeny, heredity and epigenetics (21,23). It has been recently discovered that miRNAs are involved in inflammatory cell regulation and, thus, all aspects of AS pathology, including vascular endothelial cell and monocyte development, differentiation and function (32). Studies have shown that miR-143 can affect the formation of AS plaques by inhibiting endothelial cell glycolysis, while miR-33 is closely associated with macrophage metabolism (33,34).

Drug intervention for AS

The aim of currently available anti-AS treatments, which mainly include statins, antithrombotic drugs and surgical intervention, is to reduce serum LDL levels (35). Statins are methylglutaryl-CoA reductase inhibitors that have been found to be effective at halting disease progression and reducing the incidence of cardiovascular and cerebrovascular complications. By inhibiting methylglutaryl-CoA reductase, statins lower total cholesterol and LDL-C, increase HDL-C, activate nitric oxide (NO) synthase, increase endothelial NO levels, and prevent NO decrease caused by Ox-LDL (36). Probucol, a strong synthetic antioxidant, is a symmetrical di-tert-butylphenol structure that is easily oxidized, thus reducing free oxygen radicals and diminishing their oxidizing capacity (37).

The irreversible antioxidant effect of probucol is attributed to its strong affinity for LDL-C, thus inhibiting LDL-C and reducing the formation of Ox-LDL. Anti-platelet aggregation drugs may also exert an anti-inflammatory effect on the injured vascular intima (38).

The combination of statins and antioxidants may prevent thrombosis and AS plaque formation (39). In addition, niacin, a broad-spectrum lipid-modulating drug, acts by inhibiting the expression of serum adhesion molecules and inflammatory cytokines (40). The anti-inflammatory effect of niacin is achieved by downregulating the NF-κB signaling pathway.

In addition to Western medicine, Traditional Chinese Medicine (TCM) has also achieved marked benefits in the treatment of AS. In TCM, AS is commonly categorised as 'blood stasis' and 'phlegm turbidity' (41). Modern TCM theories mostly attribute the pathogenesis of AS to phlegm, toxins and stasis. TCM arrests AS progression by activating blood and dredging collaterals, inhibiting inflammation and plaque formation, and stabilizing plaques (42,43). Commonly used TCM agents include Chaihu Shugan San, Yiqi Yangyin Recipe, Danhuang Tongmai Capsule and Simiao Yongan Decoction (44-46). From a macro perspective, TCM is guided by Chinese medicine principles, and the treatment is performed holistically. In recent years, TCM has been found to add unique advantages to the treatment of complex diseases, such as AS.

3. Vascular endothelial cells

Physiological functions of vascular endothelial cells

The inner vascular endothelium in the heart, blood and lymphatic vessels is composed of squamous epithelial cells, and is important for maintaining the complete structure and function of the blood vessel wall (47). Endothelial cells serve not onlyas a mechanical barrier, but also as receptors and endocrine organs, as they are capable of synthesizing and releasing a variety of endothelial-derived vasoactive factors (9). When damaged, vascular endothelial cells secrete a variety of molecules to promote leukocyte adhesion. They also secrete endothelium-dependent factors, such as endothelin, angiotensin, NO and prostacyclin, regulating vasoconstriction and vasodilation (48). Endothelium-dependent dilation factors may also inhibit platelet activation and aggregation. Plasma plasminogen activator synthesized by vascular endothelial cells has high affinity for fibrin and can activate plasminogen to dissolve thrombi (24). Under normal conditions, these cells reduce inflammation by regulating the release of various cytokines and inflammatory factors, such as intercellular adhesion molecule-1, vascular cell adhesion molecule-1 and monocyte chemokines (49). However, the injured endothelium promotes a series of inflammatory reactions, causing endothelial cell dysfunction and favouring AS development.

Mechanism underlying the development of AS caused by vascular endothelial cell injury

Vascular endothelial cell function is closely associated with AS, which is a chronic inflammatory response of the arterial wall to endothelial cell injury (8). Vascular endothelial cell injury and functional damage serve as the initiating factor. The dysfunction and morphological damage of endothelial cells may manifest as a variety of endothelial abnormalities, causing leukocyte adhesion, vasoconstriction, platelet activation, oxidative stress and inflammation, followed by thrombus formation which, in turn, promotes the formation of AS plaques (24,25,50,51). Endothelial progenitor cells participate in reendothelialization and angiogenesis, and may play a pivotal role in the occur-rence and development of AS (52).

Increased intimal permeability

Following endothelial cell injury, lipids accumulate in the blood vessels, resulting in increased permeability of the vascular intima. This causes monocytes and phagocytes to be released into the bloodstream, increasing the adhesion to endothelial cells (50). LDL is then oxidized to the cytotoxic Ox-LDL, which is damaging to endothelial and smooth muscle cells. Macrophage Ox-LDL uptake occurs through scavenger receptors, accelerating their transformation into foam cells that eventually form the AS plaques (47).

Increased leukocyte adhesion

Leukocytes, particularly monocytes, adhere to the endothelium through intercellular adhesion molecules, leukocyte adhesion molecules, TNF-α and vascular cell adhesion molecules. Subsequently, guided by chemokines, they migrate to the intima and differentiate into macrophages (53). Macrophages secrete a variety of inflammatory factors, such as TNF-α, IL-1 and IL-6, to promote the formation of AS plaques. In addition, damage to endothelial cells leads to increased expression of adhesion molecules, while also increasing the susceptibility of endothelial cells to inflammatory stimuli. This again increases the expression of adhesion molecules, promoting the adhesion of monocytes to endothelial cells and triggering AS development (34).

Promoting thrombosis

Stimulation or damage of endothelial cells causes a decrease in their antithrombotic properties, which promotes thrombus formation (25). Among other factors, increased secretion of the von Willebrand factor and thromboxane mediate platelet aggregation and adhesion to the endothelium to promote thrombosis, while plasminogen activator inhibitors inhibit thrombolysis (5). Thus, the micro-thrombi formed on the endothelial inner membrane are difficult to dissolve, contributing to the formation of plaques (8).

Regulation of endothelial progenitor cells

Endothelial progenitor cells, a type of precursor cell with a high proliferation potential, are a heterogeneous cell population with multiple origins and different phenotypes, capable of producing endothelial cells (54). Endothelial progenitor cells can selectively be recruited to damaged or ischemic areas by stress and inflammatory stimuli to form new blood vessels through differentiation and proliferation, without relying on the original vasculature (55). Inflammation is the pathophysiological basis of various cardiovascular diseases. Pro-inflammatory cytokines stimulate the expression of adhesion molecules on the surface of endothelial cells and promote the onset of AS (56). Inflammation can stimulate the release of endothelial progenitor cells from the bone marrow into peripheral blood, thus promoting tissue repair (57). Therefore, the occurrence of AS is associated with endothelial progenitor cells, which are involved in repair and intimal hyperplasia following endothelial cell injury, which also promote the formation and stability of AS plaques (52).

4. Glycolysis

It was previously demonstrated that 85% of ATP in endothelial cells is produced by glycolysis, of which ~60% is used for homeostatic maintenance and 40% for proliferation (11). The high glycolytic properties of endothelial cells result from their low content of mitochondria, which comprise only 5% of the cell. Although endothelial cells reside in a high-oxygen environment, they consume little oxygen and can, therefore, deliver oxygen to nearby tissues (58). Endothelial cells use glycolysis to quickly produce energy, which helps them adapt to changes in their environment. More metabolic intermediate products are produced through glycolysis, affecting cell survival (59). Compared with other cells, resting endothelial cells have a high-efficiency glycolysis rate. When cells undergo migration or proliferation, their glycolysis rates double (60). Glycolysis is required to meet the energy demands of blood vessel sprouting, since the apical cells at the front must continuously migrate forward to form filamentous or laminar pseudopodia, while stem cells at the back increase their proliferation rate and form a lumen (61). Endothelial cell dysfunction caused by Ox-LDL and other factors is the main cause of AS. It was previously demonstrated that excessive activation of glycolysis is a key factor leading to endothelial cell dysfunction and proliferation (12). Maintaining the metabolic balance of endothelial cells by inhibiting glycolysis to reduce dysfunction and inflammation may represent a novel treatment strategy for AS (11). The rate-limiting enzymes of glycolysis may be a bridge to elucidating the association between endothelial cell injury and AS.

Under physiological conditions, cells metabolize glucose mainly through oxidative phosphorylation, while the glycolytic pathway is activated only under hypoxic conditions (62). The glycolytic pathway comprises a series of enzymatic reactions in which glucose from tissues is degraded (63). Glucose is first transported into the cell via glucose transporters (GLUTs), and is then phosphorylated by hexokinase (HK) into glucose-6-phosphate, which cannot penetrate the cell membrane (64). Glucose-6-phosphate is then converted into pyruvate by hexose phosphate isomerase and phosphofructokinase (PFK) (65). The pyruvate produced through glycolysis may directly enter the tricarboxylic acid cycle or be converted into lactic acid by lactate dehydrogenase (LDHA) (66). The lactic acid is then transported outside the cell via the monocarboxylic acid transporter ¼ (67). The ATP produced by this process provides energy for the cell.

Regulation of HK in endothelial glycolysis

HK is the first rate-limiting enzyme in the glycolytic pathway. This enzyme catalyses the conversion of glucose to glucose-6-phosphate, and produces ATP through oxidative phosphorylation or glycolysis (68). It was previously demonstrated that the AKT/mTOR signalling pathway is associated with HK production. High HK expression in endothelial cells facilitates efficient glycolysis and promotes rapid cell proliferation (69). In addition, HK can bind to voltage-dependent anion channels in the outer mitochondrial membrane to prevent binding of the pro-apoptotic protein Bax. This prevents the release of cytochrome c from the mitochondria, thereby exerting an anti-apoptotic effect (70). Therefore, HK not only promotes cell proliferation, but also inhibits cell apoptosis.

Regulation of PFK in endothelial glycolysis

PFK, the second glycolytic rate-limiting enzyme in the glycolytic pathway, catalyses the conversion of fructose 6-phosphate to fructose 1,6-diphosphate. The expression of PFK is regulated by a number of factors, such as c-Src activation and hypoxia-inducible factor (HIF)-1α, promoting PFK2 expression (71). TP53-induced glycolysis and apoptosis regulator (TIGAR) may reduce PFK expression and inhibit glycolysis. The kinase activity of PFKFB3, a subtype of PFK2, in vascular endothelial cells, is affected by the RAS and AMP-activated protein kinase (AMPK) signalling pathways (72). In addition, PFKFB3 can also promote endothelial cell inflammation through TNF-α, thereby promoting AS development (73).

Regulation of PK in endothelial glycolysis

PK, the third rate-limiting enzyme in the glycolytic pathway, specifically catalyses the conversion of phosphoenolpyruvate to pyruvate. This irreversible reaction is a crucial regulatory step in the glycolytic pathway. PKM2 is the only PK subtype that can switch between high-activity (tetramer) and low-activity (dimer) forms. Following phosphorylation, PKM2 is converted to a dimer, promoting the upstream glycolytic products of PK to enter the biosynthetic pathway (74). In addition, the expression of PKM2 is differentially regulated by the PI3K/AKT signal-ling pathway (75). PKM2-regulated glycolysis contributes to the proliferation and migration of vascular smooth muscle cells, and is positively correlated with AS development (12).

Regulation of LDHA in endothelial glycolysis

LDHA, the fourth rate-limiting enzyme in the glycolytic pathway, catalyses the production of lactic acid from pyruvate. This enzyme enables recycling of pyruvate and reduced nicotinamide purine dinucleotide, and plays a key role in promoting efficient cell glycolysis (76). SRC can also phosphorylate LDHA and promote conversion enzyme activity. HIF-1α is the upstream regulator of LDHA, and both HIF-1α and LDHA stimulate the inflammatory response (77).

5. Prediction and analysis of targets associated with endothelial cell glycolysis in AS

Target prediction

Although endothelial cell glycolysis plays an important role in AS, related research is scarce. Therefore, in the present study, the term 'NCBI-gene' was used to search for genes associated with endothelial cell glycolysis in AS, and 76 genes were identified (Table I). The Comparative Toxicogenomics Database (CTD) is a database used to describe the association between chemical substances, genes and human diseases. CTD was used to conduct Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis (Table II); the pathways included in the analysis were the immune system pathway, the signal transduction pathway, pathways in cancer, the PI3K/AKT, IL-4, IL-13, HIF-1, NGF and FoxO signalling pathways, the platelet activation, signalling and aggregation pathway, as well as the fluid shear stress and AS pathways.

Table I

Gene information on atherosclerosis and endothelial cell glycolysis.

Table I

Gene information on atherosclerosis and endothelial cell glycolysis.

No.Gene nameFull nameChromosomeLocationID
1VEGFAVascular endothelial growth factor A66p21.17422
2CXCL8C-X-C motif chemokine ligand 844q13.33576
3PPARGPeroxisome proliferator activated receptor gamma33p25.25468
4IL33Interleukin 3399p24.190865
5IL6Interleukin 677p15.33569
6HIF1AHypoxia-inducible factor 1 subunit alpha1414q23.23091
7TP53Tumor protein p531717p13.17157
8KDRKinase insert domain receptor44q123791
9TGFB1Transforming growth factor beta 11919q13.27040
10miR21MicroRNA 211717q23.1406991
11CDKN2ACyclin-dependent kinase inhibitor 2A99p21.31029
12IL1BInterleukin 1 beta22q14.13553
13AKT1AKT serine/threonine kinase 11414q32.33207
14BCL2BCL2 apoptosis regulator1818q21.33596
15SIRT1Sirtuin 11010q21.323411
16STAT3Signal transducer and activator of transcription 31717q21.26774
17TLR4Toll-like receptor 499q33.17099
18PTGS2 Prostaglandin-endoperoxide synthase 211q31.15743
19ADIPOQAdiponectin C1Q and collagen domain containing33q27.39370
20NFE2L2Nuclear factor erythroid 2 like 222q31.24780
21NOTCH1Notch receptor 199q34.34851
22MTORMechanistic target of rapamycin kinase11p36.222475
23CTNNB1Catenin beta 133p22.11499
24PTENPhosphatase and tensin homolog1010q23.315728
25LEPLeptin77q32.13952
26ESR1Estrogen receptor 166q25.1-q25.22099
27SOD1Superoxide dismutase 12121q22.116647
28MAPK1Mitogen-activated protein kinase 12222q11.225594
29RELARELA proto-oncogene NF-kB subunit1111q13.15970
30miR34AMicroRNA 34a11p36.22407040
31IFNGInterferon gamma1212q153458
32AGTR1Angiotensin II receptor type 133q24185
33ABCB1ATP-binding cassette subfamily B member 177q21.125243
34CD44CD44 molecule (Indian blood group)1111p13960
35STAT1Signal transducer and activator of transcription 122q32.26772
36HGFHepatocyte growth factor77q21.113082
37NAMPTNicotinamide phosphoribosyl transferase77q22.310135
38CYBBCytochrome b-245 beta chainXXp21.1-p11.41536
39KLKlotho1313q13.19365
40SRCSRC proto-oncogene non-receptor tyrosine kinase2020q11.236714
41PPARAPeroxisome proliferator activated receptor alpha2222q13.315465
42BSGBasigin (Ok blood group)1919p13.3682
43TXNThioredoxin99q31.37295
44NOS2Nitric oxide synthase 21717q11.24843
45FOXO3Forkhead box O366q212309
46PRKCEProtein kinase C epsilon22p215581
47NR4A1Nuclear receptor subfamily 4 group A member 11212q13.133164
48NOX4NADPH oxidase 41111q14.350507
49PRKAA1Protein kinase AMP-activated catalytic subunit alpha 155p13.15562
50miR210MicroRNA 2101111p15.5406992
51MALAT1 Metastasis-associated lung adenocarcinoma transcript 11111q13.1378938
52IGF1RInsulin-like growth factor 1 receptor1515q26.33480
53AHRAryl hydrocarbon receptor77p21.1196
54CD36CD36 molecule77q21.11948
55EZH2Enhancer of zeste 2 polycomb repressive complex 2 subunit77q36.12146
56SIRT6Sirtuin 61919p13.351548
57IKBKBInhibitor of nuclear factor kappa B kinase subunit beta88p11.213551
58miR122MicroRNA 1221818q21.31406906
59UCP2Uncoupling protein 21111q13.47351
60ENO1Enolase 111p36.232023
61PPARGC1APPARG coactivator 1 alpha44p15.210891
62SNAI1Snail family transcriptional repressor 12020q13.136615
63PPBPPro-platelet basic protein44q13.35473
64DICER1Dicer 1 ribonuclease III1414q32.1323405
65TXNIP Thioredoxin-interacting protein11q21.110628
66IL22Interleukin 221212q1550616
67TNFSF13BTNF superfamily member 13b1313q33.310673
68SREBF2Sterol regulatory element-binding transcription factor 22222q13.26721
69miR497MicroRNA 4971717p13.1574456
70miR206MicroRNA 20666p12.2406989
71TUG1Taurine upregulated 12222q12.255000
72miR135BMicroRNA 135b11q32.1442891
73miR142MicroRNA 1421717q22406934
74miR135A1MicroRNA 135a-133p21.2406925
75miR148BMicroRNA 148b1212q13.13442892
76miR30C1MicroRNA 30c-111p34.2407031

Table II

Potential pathways of atherosclerosis and endothelial cell glycolysis.

Table II

Potential pathways of atherosclerosis and endothelial cell glycolysis.

PathwayNo.Genes
Immune system36AKT1, BCL2, CD36, CD44, CTNNB1, CXCL8, CYBB, FOXO3, HGF, HIF1A, IFNG, IKBKB, IL1B, IL22, IL33, IL6, KL, MAPK1, MTOR, NOS2, NR4A1, PPBP, PRKCE, PTEN, PTGS2, RELA, SRC, STAT1, STAT3, TGFB1, TLR4, TNFSF13B, TP53, TXN, TXNIP, VEGFA
Signal transduction30AGTR1, AKT1, CTNNB1, CXCL8, CYBB, ESR1, FOXO3, HGF, HIF1A, IGF1R, IKBKB, IL6, KDR, KL, LEP, MAPK1, MTOR, NOTCH1, NR4A1, PPBP, PRKAA1, PRKCE, PTEN, RELA, SRC, STAT1, STAT3, TGFB1, TP53, VEGFA
Pathways in cancer23AGTR1, AKT1, BCL2, CDKN2A, CTNNB1, CXCL8, HGF, HIF1A, IGF1R, IKBKB, IL6, MAPK1, MTOR, NOS2, PPARG, PTEN, PTGS2, RELA, STAT1, STAT3, TGFB1, TP53, VEGFA
PI3K-AKT signalling pathway17AKT1, BCL2, FOXO3, HGF, IGF1R, IKBKB, IL6, KDR, MAPK1, MTOR, NR4A1, PRKAA1, PTEN, RELA, TLR4, TP53, VEGFA
Cellular responses to stress17CDKN2A, CXCL8, CYBB, EZH2, HIF1A, IL6, MAPK1, MTOR, NOX4, PRKAA1, RELA, SIRT1, SOD1, STAT3, TP53, TXN, VEGFA
Interleukin-4 and 13 signalling16AKT1, BCL2, CD36, CXCL8, FOXO3, HGF, HIF1A, IL1B, IL6, NOS2, PTGS2, STAT1, STAT3, TGFB1, TP53, VEGFA
HIF-1 signalling pathway15AKT1, BCL2, CYBB, ENO1, HIF1A, IFNG, IGF1R, IL6, MAPK1, MTOR, NOS2, RELA, STAT3, TLR4, VEGFA
Platelet activation, signaling and aggregation10AKT1, CD36, HGF, MAPK1, PPBP, PRKCE, SOD1, SRC, TGFB1, VEGFA
NGF signalling via TRKA from the plasma membrane12AKT1, FOXO3, HGF, KL, MAPK1, MTOR, NR4A1, PRKCE, PTEN, SRC, STAT3, TP53
Fluid shear stress and atherosclerosis15AKT1, BCL2, CTNNB1, CYBB, IFNG, IKBKB, IL1B, KDR, NFE2L2, PRKAA1, RELA, SRC, TP53, TXN, VEGFA
FoxO signalling pathway11AKT1, FOXO3, IGF1R, IKBKB, IL6, MAPK1, PRKAA1, PTEN, SIRT1, STAT3, TGFB1
PI3K/AKT activation10AKT1, FOXO3, HGF, KL, MAPK1, MTOR, NR4A1, PTEN, SRC, TP53
LAT2/NTAL/LAB on calcium mobilization10AKT1, FOXO3, HGF, KL, MAPK1, MTOR, NR4A1, PTEN, SRC, TP53

The screened genes play important regulatory roles in multiple signalling pathways, indicating that such interactions should also be present among their corresponding proteins. The STRING database, used to study the protein interaction networks, helps to mine core regulatory genes. Based on that, a protein-protein interaction network was built and Cytoscape 3.2.1 software (https://cytoscape.org/download_old_versions.html) was used to visualize it (Fig. 2). The node size in Fig. 1 reflects its importance in the network. CytoHubba is a network analysis plug-in specifically used to identify key nodes in the Cytoscape software, providing a variety of key node identification algorithms, which focus on different network topology characteristics. To make the identification of key targets as accurate as possible, the top 10 key targets were identified based on two algorithms, namely Matthews correlation coefficient (MCC) and Stress (Fig. 3). Next, the intersection of the two results was considered as the key targets for AS endothelial glycolysis, including AKT1, IL-6, vascular endothelial growth factor (VEGF)A, TP53, signal transducer and activator of transcription 3 (STAT3), SRC and mitogen-activated protein kinase (MAPK)1.

Target analysis

All 7 predicted targets are factors proven to promote or inhibit the process of AS. AKT1 and TP53 can directly regulate glycolysis, thereby affecting AS. In addition to the regulation of glycolysis, IL-6 can also regulate AS through inflammation; VEGFA, STAT3 and SRC are also closely associated with immune system dysfunction. The regulatory effect of MAPK1 on AS is mainly mediated through immune response, but the association between glycolysis and MAPK1 has not been explored. The available research on these 7 targets is currently not sufficient, and further experiments are required to verify the results. The purpose of the present review was also to provide a direction for future AS studies.

Evaluation of AKT1 involvement in endothelial cell glycolysis in AS

AKT, including AKT1, AKT2 and AKT3, play an important role in cell proliferation, survival, metabolism and migration (78). AKT can activate mTOR, another major regulatory gene of cell metabolism, thereby promoting the expression of HIF-1α. HIF-1α then regulates the expression of glycolysis-related proteins, such as GLUTs, PK, HK2 and LDHA (79). These pathways promote glucose uptake and lactic acid production. In addition, endothelial glycolysis can be regulated by the FKBP51/AKT1 and AKT1/GSK/3β signalling pathways (80,81). Therefore, as regards energy metabolism, AKT1 may be a key target in AS.

Evaluation of IL-6 involvement in endothelial cell glycolysis in AS

IL-6 is an important pro-inflammatory cytokine (82). Chronic inflammation is also considered as part of AS pathology, suggesting a possible role for IL-6 (83). It has been shown that IL-6 affects AS development through the acute phase response, and exerts an effect on insulin resistance (84), while it also promotes glycolysis through PFKFB3 (85). The IL-6/STAT3 pathway is also a pathway involved in glycolysis regulation (86).

Evaluation of VEGFA involvement in endothelial cell glycolysis in AS

VEGFA promotes angiogenesis and regulates endothelial cell proliferation, macrophage infiltration and foam cell formation through signal transduction (87); therefore, it plays a key regulatory role in the formation and stabilization of AS plaques (88). In addition, it was previously demonstrated that endothelial glycolysis may be promoted by enhancing VEGFA expression, making it an important contributor to AS development and endothelial glycolysis (89). Therefore, VEGFA is an important target in AS and endothelial glycolysis.

Evaluation of TP53 involvement in endothelial cell glycolysis in AS

TP53, also referred to as 'the guardian of the genome', maintains gene stability. As it is mutated in >50% of tumour cells, TP53 is used for cancer prediction and risk assessment (90). In addition, TP53 regulates energy metabolism through the AKT/mTOR and NF-κB signalling pathways, and it inhibits glycolysis by downregulating the expression of rate-limiting enzymes and activating TP53-induced glycolysis and TIGAR (91,92). It was previously demonstrated that endothelial autophagy is inhibited in advanced AS by regulating mTOR and TIGAR (93). However, there is insufficient research on AS endothelial glycolysis, and the TP53 regulatory effects in AS require further investigation.

Evaluation of STAT3 involvement in endothelial cell glycolysis in AS

STAT3, a transcription factor, regulates cell growth, apoptosis and inflammation, and plays an important role in cancer, AS, as well as cardiovascular and cerebrovas-cular diseases (94). STAT3 is also strongly associated with endothelial cell dysfunction, macrophage polarization and inflammatory responses, thereby promoting AS development (95). In addition, it can enhance cell metabolism through HK2 upregulation. Previous studies have demonstrated that the IL-6/STAT3 and JAK2/STAT3 pathways can promote glycolysis, confirming the crucial role of STAT3 in AS and glycolysis (96,97). Therefore, STAT3 was proven to be an important factor in AS and glycolysis.

Evaluation of SRC involvement in endothelial cell glycolysis in AS

SRC is an oncogene that regulates intracellular signal transduction, cell proliferation and cell migration (98). SRC phosphorylation and activation also participates in intracellular metabolic processes (99). It was previously demonstrated that the SRC/AKT/LKB1/AMPKα signalling pathway can shift the intracellular metabolic pathway from glycolysis to aerobic metabolism (100). In addition, SRC-related pathways in endothelial cells promote AS by affecting leukocyte adhesion and monocyte transport (101). Thus, SRC is another important target in AS and endothelial cell glycolysis.

Evaluation of MAPK1 involvement in endothelial cell glycolysis in AS

MAPK is an important intracellular enzyme and the upstream signalling transduction molecule of several pathways (102). It was previously demonstrated that the P38/MAPK signalling pathway can aggravate AS, while MAPK1 affects cell adhesion and the immune response to promote AS development (103). However, further research is required to confirm the regulatory role of MAPK1 in AS and endothelial cell glycolysis.

6. Discussion

Although some progress has been made in the treatment of AS and its complications, with a consequent improvement in patient survival (1), long-term illness puts pressure on the heart and is associated with a major socioeconomic burden (3). AS is currently considered as a chronic inflammatory disease, and the underlying mechanisms mainly involve oxidative stress, lipid metabolism disorders, endothelial cell damage, inflammation and immune dysfunction (5,27). Eventually, plaque rupture and thrombosis causes acute life-threatening cardiovascular and cerebrovascular diseases (1). Vascular endothelium inflammation and cell dysfunction are considered as the initiating and central events in AS (29). Since glycolysis is the main metabolic pathway in endothelial cells, an in-depth study of the association between endothelial cell glycolysis and AS may further elucidate AS pathophysiology and provide clues for its prevention (11).

Through mining and analysis of endothelial cell glycolysis and AS-related genes, 8 key targets were identified, namely AKT1, IL-6, VEGFA, TP53, STAT3, SRC and MAPK1. These targets were found to directly or indirectly affect the expression of key rate-limiting enzymes in endothelial cell glycolysis. In addition, the pathways obtained by the enrichment of these genes included the immune system pathway, the signal transduction pathway, pathways in cancer, the PI3K/AKT, IL-4, IL-13, HIF-1, NGF and FoxO signalling pathways, the platelet activation, signalling and aggregation pathway, as well as the fluid shear stress and AS pathways. The aforementioned results are based on existing research and correlation prediction (104). At present, research on AS mainly uses genetically modified animal models, such as ApoE−/− and db/db mice, and the findings cannot yet be translated into clinical application. Therefore, these targets must be further verified before they reach the clinical stage. The aim of the present review was to contribute to the study of AS mechanisms and provide novel suggestions, since future AS therapies should be explored from a new perspective. Advanced science and technology, including high-content technology, single cell transcriptomics, lipometa-bolic technology and epigenetic modification technology, will hopefully accelerate the search for novel AS treatments.

Funding

This review was supported by the National Natural Science Foundation of China (grant no. 81973514), the Central Public-Interest Scientific Institution Basal Research Fund (grant no. 2018PT35030) and the Drug Innovation Major Project (grant no. 2018ZX09711001-009).

Availability of data and materials

Not applicable.

Authors' contributions

GS and XS contributed to the conception and design of the study. RW performed the literature search and wrote the manuscript. MW prepared the figures. JY revised the manuscript. All the authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Kobiyama K and Ley K: Atherosclerosis. Circ Res. 123:1118–1120. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Spinelli FR, Barone F, Cacciapaglia F, Pecani A and Piga M: Atherosclerosis and autoimmunity. Mediators Inflamm. 2018:67304212018. View Article : Google Scholar : PubMed/NCBI

3 

Lu H and Daugherty A: Atherosclerosis. Arterioscler Thromb Vasc Biol. 35:485–491. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgözoğlu L and Lewis EF: Atherosclerosis. Nat Rev Dis Primers. 5:562019. View Article : Google Scholar : PubMed/NCBI

5 

Bories GFP and Leitinger N: Macrophage metabolism in athero-sclerosis. FEBS Lett. 591:3042–3060. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Zhao Y, Qu H, Wang Y, Xiao W, Zhang Y and Shi D: Small rodent models of atherosclerosis. Biomed Pharmacother. 129:1104262020. View Article : Google Scholar : PubMed/NCBI

7 

Goonewardena SN, Prevette LE and Desai AA: Metabolomics and atherosclerosis. Curr Atheroscler Rep. 12:267–272. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Sitia S, Tomasoni L, Atzeni F, Ambrosio G, Cordiano C, Catapano A, Tramontana S, Perticone F, Naccarato P, Camici P, et al: From endothelial dysfunction to atherosclerosis. Autoimmun Rev. 9:830–834. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Chen PY, Schwartz MA and Simons M: Endothelial-to-mesenchymal transition, vascular inflammation, and atherosclerosis. Front Cardiovasc Med. 7:532020. View Article : Google Scholar : PubMed/NCBI

10 

Gao F, Chen J and Zhu H: A potential strategy for treating athero-sclerosis: Improving endothelial function via AMP-activated protein kinase. Sci China Life Sci. 61:1024–1029. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Pircher A, Treps L, Bodrug N and Carmeliet P: Endothelial cell metabolism: A novel player in atherosclerosis? Basic principles and therapeutic opportunities. Atherosclerosis. 253:247–257. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Zhao X, Tan F, Cao X, Cao Z, Li B, Shen Z and Tian Y: PKM2-dependent glycolysis promotes the proliferation and migration of vascular smooth muscle cells during atherosclerosis. Acta Biochim Biophys Sin (Shanghai). 52:9–17. 2020. View Article : Google Scholar

13 

Ilhan F and Kalkanli ST: Atherosclerosis and the role of immune cells. World J Clin Cases. 3:345–352. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Xiao W, Jia Z, Zhang Q, Wei C, Wang H and Wu Y: Inflammation and oxidative stress, rather than hypoxia, are predominant factors promoting angiogenesis in the initial phases of atherosclerosis. Mol Med Rep. 12:3315–3322. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Dinh QN, Chrissobolis S, Diep H, Chan CT, Ferens D, Drummond GR and Sobey CG: Advanced atherosclerosis is associated with inflammation, vascular dysfunction and oxida-tive stress, but not hypertension. Pharmacol Res. 116:70–76. 2017. View Article : Google Scholar

16 

Yang K, Zhang H, Luo Y, Zhang J, Wang M, Liao P, Cao L, Guo P, Sun G and Sun X: Gypenoside XVII prevents atherosclerosis by attenuating endothelial apoptosis and oxidative stress: Insight into the ERα-Mediated PI3K/Akt Pathway. Int J Mol Sci. 18:772017. View Article : Google Scholar

17 

Feletou M, Cohen RA, Vanhoutte PM and Verbeuren TJ: TP receptors and oxidative stress hand in hand from endothelial dysfunction to atherosclerosis. Adv Pharmacol. 60:85–106. 2010.PubMed/NCBI

18 

Armstrong AW, Voyles SV, Armstrong EJ, Fuller EN and Rutledge JC: Angiogenesis and oxidative stress: Common mechanisms linking psoriasis with atherosclerosis. J Dermatol Sci. 63:1–9. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Gupta M, Blumenthal C, Chatterjee S, Bandyopadhyay D, Jain V, Lavie CJ, Virani SS, Ray KK, Aronow WS and Ghosh RK: Novel emerging therapies in atherosclerosis targeting lipid metabolism. Expert Opin Investig Drugs. 29:611–622. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Lee J, Jung S, Kim N, Shin MJ, Ryu DH and Hwang GS: Myocardial metabolic alterations in mice with diet-induced atherosclerosis: Linking sulfur amino acid and lipid metabolism. Sci Rep. 7:135972017. View Article : Google Scholar : PubMed/NCBI

21 

Novák J, Olejníčková V, Tkáčová N and Santulli G: Mechanistic role of MicroRNAs in coupling lipid metabolism and atherosclerosis. Adv Exp Med Biol. 887:79–100. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Shao D, Lian Z, Di Y, Zhang L, Rajoka MSR, Zhang Y, Kong J, Jiang C and Shi J: Dietary compounds have potential in controlling atherosclerosis by modulating macrophage cholesterol metabolism and inflammation via miRNA. NPJ Sci Food. 2:132018. View Article : Google Scholar

23 

Giral H, Kratzer A and Landmesser U: MicroRNAs in lipid metabolism and atherosclerosis. Best Pract Res Clin Endocrinol Metab. 30:665–676. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Gimbrone MA Jr and García-Cardeña G: Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 118:620–636. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Mudau M, Genis A, Lochner A and Strijdom H: Endothelial dysfunction: The early predictor of atherosclerosis. Cardiovasc J Afr. 23:222–231. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Brix B, Mesters JR, Pellerin L and Johren O: Endothelial cell-derived nitric oxide enhances aerobic glycolysis in astrocytes via HIF-1α-mediated target gene activation. J Neurosci. 32:9727–9735. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Legein B, Temmerman L, Biessen EA and Lutgens E: Inflammation and immune system interactions in atherosclerosis. Cell Mol Life Sci. 70:3847–3869. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Libby P and Hansson GK: Taming immune and inflammatory responses to treat atherosclerosis. J Am Coll Cardiol. 71:173–176. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Wolf D and Ley K: Immunity and Inflammation in atherosclerosis. Circ Res. 124:315–327. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Hansson GK and Hermansson A: The immune system in athero-sclerosis. Nat Immunol. 12:204–212. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Iwata H and Nagai R: Novel immune signals and atherosclerosis. Curr Atheroscler Rep. 14:484–490. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Kuosmanen SM, Kansanen E, Kaikkonen MU, Sihvola V, Pulkkinen K, Jyrkkänen HK, Tuoresmäki P, Hartikainen J, Hippeläinen M, Kokki H, et al: NRF2 regulates endothelial glycolysis and proliferation with miR-93 and mediates the effects of oxidized phospholipids on endothelial activation. Nucleic Acids Res. 46:1124–1138. 2018. View Article : Google Scholar :

33 

Ouimet M, Ediriweera HN, Gundra UM, Sheedy FJ, Ramkhelawon B, Hutchison SB, Rinehold K, van Solingen C, Fullerton MD, Cecchini K, et al: MicroRNA-33-dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis. J Clin Invest. 125:4334–4348. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Kumar S, Kim CW, Simmons RD and Jo H: Role of flow-sensitive microRNAs in endothelial dysfunction and atherosclerosis: Mechanosensitive athero-miRs. Arterioscler Thromb Vasc Biol. 34:2206–2216. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Barton M: Mechanisms and therapy of atherosclerosis and its clinical complications. Curr Opin Pharmacol. 13:149–153. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Gonzalez L and Trigatti BL: Macrophage apoptosis and necrotic core development in atherosclerosis: A rapidly advancing field with clinical relevance to imaging and therapy. Can J Cardiol. 33:303–312. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Chen C, Wang Y, Cao Y, Wang Q, Anwaier G, Zhang Q and Qi R: Mechanisms underlying the inhibitory effects of probucol on elastase-induced abdominal aortic aneurysm in mice. Br J Pharmacol. 177:204–216. 2020. View Article : Google Scholar

38 

Guo X, Wang L, Xia X, Wang P and Li X: Effects of atorvastatin and/or probucol on recovery of atherosclerosis in high-fat-diet-fed apolipoprotein E-deficient mice. Biomed Pharmacother. 109:1445–1453. 2019. View Article : Google Scholar

39 

Profumo E, Buttari B, D'Arcangelo D, Tinaburri L, Dettori MA, Fabbri D, Delogu G and Riganò R: The nutraceutical dehydroz-ingerone and its dimer counteract inflammation- and oxidative stress-induced dysfunction of in vitro cultured human endothelial cells: A novel perspective for the prevention and therapy of atherosclerosis. Oxid Med Cell Longev. 2016:12464852016. View Article : Google Scholar

40 

Jiang Y, Jin M, Chen J, Yan J, Liu P, Yao M, Cai W and Pi R: Discovery of a novel niacin-lipoic acid dimer N2L attenuating atherosclerosis and dyslipidemia with non-flushing effects. Eur J Pharmacol. 868:1728712020. View Article : Google Scholar

41 

Ren Y, Qiao W, Fu D, Han Z, Liu W, Ye W and Liu Z: Traditional Chinese medicine protects against cytokine production as the potential immunosuppressive agents in atherosclerosis. J Immunol Res. 2017:74243072017. View Article : Google Scholar : PubMed/NCBI

42 

Li TT, Wang ZB, Li Y, Cao F, Yang BY and Kuang HX: The mechanisms of traditional Chinese medicine underlying the prevention and treatment of atherosclerosis. Chin J Nat Med. 17:401–412. 2019.PubMed/NCBI

43 

Tian F, Gu L, Si A, Yao Q, Zhang X, Zhao J and Hu D: Metabolomic study on the faecal extracts of atherosclerosis mice and its application in a traditional Chinese Medicine. J Chromatogr B Analyt Technol Biomed Life Sci. 1007:140–148. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Fan Q, Liu Y, Rao J, Zhang Z, Xiao W, Zhu T, Chai X, Ye K, Ning N, Yin Z, et al: Anti-atherosclerosis effect of angong niuhuang pill via regulating Th17/Treg immune balance and inhibiting chronic inflammatory on ApoE−/− mice model of early and mid-term atherosclerosis. Front Pharmacol. 10:15842020. View Article : Google Scholar

45 

Zhu ZB, Song K, Huang WJ, Li H, Yang H, Bai YQ, Guo KT, Yang RB, Lou WJ, Xia CH, et al: Si-Miao-Yong-An (SMYA) decoction may protect the renal function through regulating the autophagy-mediated degradation of ubiquitinated protein in an atherosclerosis model. Front Pharmacol. 11:8372020. View Article : Google Scholar : PubMed/NCBI

46 

Li L, Yu AL, Wang ZL, Chen K, Zheng W, Zhou JJ, Xie Q, Yan HB, Ren P and Huang X: Chaihu-Shugan-San and absorbed meranzin hydrate induce anti-atherosclerosis and behavioral improvements in high-fat diet ApoE−/− mice via anti-inflammatory and BDNF-TrkB pathway. Biomed Pharmacother. 115:1088932019. View Article : Google Scholar

47 

Haskard DO, Boyle JJ, Evans PC, Mason JC and Randi AM: Cytoprotective signaling and gene expression in endothelial cells and macrophages-lessons for atherosclerosis. Microcirculation. 20:203–216. 2013. View Article : Google Scholar

48 

Dong Y, Fernandes C, Liu Y, Wu Y, Wu H, Brophy ML, Deng L, Song K, Wen A, Wong S, et al: Role of endoplasmic reticulum stress signalling in diabetic endothelial dysfunction and athero-sclerosis. Diab Vasc Dis Res. 14:14–23. 2017. View Article : Google Scholar

49 

Chrysohoou C, Kollia N and Tousoulis D: The link between depression and atherosclerosis through the pathways of inflammation and endothelium dysfunction. Maturitas. 109:1–5. 2018. View Article : Google Scholar

50 

Jensen HA and Mehta JL: Endothelial cell dysfunction as a novel therapeutic target in atherosclerosis. Expert Rev Cardiovasc Ther. 14:1021–1033. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Theodorou K and Boon RA: Endothelial cell metabolism in atherosclerosis. Front Cell Dev Biol. 6:822018. View Article : Google Scholar : PubMed/NCBI

52 

Verma I, Syngle A, Krishan P and Garg N: Endothelial progenitor cells as a marker of endothelial dysfunction and atherosclerosis in Ankylosing Spondylitis: A cross-sectional study. Int J Angiol. 26:36–42. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Lee DY and Chiu JJ: Atherosclerosis and flow: Roles of epigenetic modulation in vascular endothelium. J Biomed Sci. 26:562019. View Article : Google Scholar : PubMed/NCBI

54 

Georgescu A, Alexandru N, Andrei E, Dragan E, Cochior D and Dias S: Effects of transplanted circulating endothelial progenitor cells and platelet microparticles in atherosclerosis development. Biol Cell. 108:219–243. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Xiang W, Hu ZL, He XJ and Dang XQ: Intravenous transfusion of endothelial progenitor cells that overexpress vitamin D receptor inhibits atherosclerosis in apoE-deficient mice. Biomed Pharmacother. 84:1233–1242. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Kong M, Zhao Y, Chen A and Lin A: The importance of physiologic ischemia training in preventing the development of atherosclerosis: The role of endothelial progenitor cells in athero-sclerotic rabbits. Coron Artery Dis. 30:377–383. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Pákozdi A, Besenyei T, Paragh G, Koch AE and Szekanecz Z: Endothelial progenitor cells in arthritis-associated vasculogenesis and atherosclerosis. Joint Bone Spine. 76:581–583. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Hu X, Cai X, Ma R, Fu W, Zhang C and Du X: Iron-load exacerbates the severity of atherosclerosis via inducing inflammation and enhancing the glycolysis in macrophages. J Cell Physiol. 234:18792–18800. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Flynn MC, Kraakman MJ, Tikellis C, Lee MKS, Hanssen NMJ, Kammoun HL, Pickering RJ, Dragoljevic D, Al-Sharea A, Barrett TJ, et al: Transient intermittent hyperglycemia accelerates atherosclerosis by promoting myelopoiesis. Circ Res. 127:877–892. 2020. View Article : Google Scholar : PubMed/NCBI

60 

Matsuura Y, Yamashita A, Zhao Y, Iwakiri T, Yamasaki K, Sugita C, Koshimoto C, Kitamura K, Kawai K, Tamaki N, et al: Altered glucose metabolism and hypoxic response in alloxan-induced diabetic atherosclerosis in rabbits. PLoS One. 12:e01759762017. View Article : Google Scholar : PubMed/NCBI

61 

Vojinovic D, van der Lee SJ, van Duijn CM, Vernooij MW, Kavousi M, Amin N, Demirkan A, Ikram MA, van der Lugt A and Bos D: Metabolic profiling of intra- and extracranial carotid artery atherosclerosis. Atherosclerosis. 272:60–65. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Akins NS, Nielson TC and Le HV: Inhibition of glycolysis and glutaminolysis: An emerging drug discovery approach to combat cancer. Curr Top Med Chem. 18:494–504. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Deng F, Zhou R, Lin C, Yang S, Wang H, Li W, Zheng K, Lin W, Li X, Yao X, et al: Tumor-secreted dickkopf2 accelerates aerobic glycolysis and promotes angiogenesis in colorectal cancer. Theranostics. 9:1001–1014. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Godfrey R and Quinlivan R: Skeletal muscle disorders of glycogenolysis and glycolysis. Nat Rev Neurol. 12:393–402. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Schoors S, De Bock K, Cantelmo AR, Georgiadou M, Ghesquière B, Cauwenberghs S, Kuchnio A, Wong BW, Quaegebeur A, Goveia J, et al: Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis. Cell Metab. 19:37–48. 2014. View Article : Google Scholar

66 

Fernie AR, Carrari F and Sweetlove LJ: Respiratory metabolism: Glycolysis, the TCA cycle and mitochondrial electron transport. Curr Opin Plant Biol. 7:254–261. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Tang BL: Glucose, glycolysis, and neurodegenerative diseases. J Cell Physiol. 235:7653–7662. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Paik JY, Lee KH, Ko BH, Choe YS, Choi Y and Kim BT: Nitric oxide stimulates 18F-FDG uptake in human endothelial cells through increased hexokinase activity and GLUT1 expression. J Nucl Med. 46:365–370. 2005.PubMed/NCBI

69 

Kim JW, Gao P, Liu YC, Semenza GL and Dang CV: Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol. 27:7381–7393. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Song J, Li Y, Song J, Hou F, Liu B and Li A: Mangiferin protects mitochondrial function by preserving mitochondrial hexokinase-II in vessel endothelial cells. Biochim Biophys Acta Mol Basis Dis. 1863:1829–1839. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Wang Y, Han X, Fu M, Wang J, Song Y, Liu Y, Zhang J, Zhou J and Ge J: Qiliqiangxin attenuates hypoxia-induced injury in primary rat cardiac microvascular endothelial cells via promoting HIF-1α-dependent glycolysis. J Cell Mol Med. 22:2791–2803. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Wik JA, Lundbäck P, la Cour Poulsen L, Haraldsen G, Skålhegg BS and Hol J: 3PO inhibits inflammatory NFκB and stress-activated kinase signaling in primary human endothelial cells independently of its target PFKFB3. PLoS One. 15:e02293952020. View Article : Google Scholar

73 

Zhang R, Li R, Liu Y, Li L and Tang Y: The glycolytic enzyme PFKFB3 controls TNF-α-induced endothelial proinflammatory responses. Inflammation. 42:146–155. 2019. View Article : Google Scholar

74 

Lu S, Deng J, Liu H, Liu B, Yang J, Miao Y, Li J, Wang N, Jiang C, Xu Q, et al: PKM2-dependent metabolic reprogramming in CD4+ T cells is crucial for hyperhomocysteinemia-accelerated atherosclerosis. J Mol Med (Berl). 96:585–600. 2018. View Article : Google Scholar

75 

Zhang X, Chen B, Wu J, Sha J, Yang B, Zhu J, Sun J, Hartung J and Bao E: Aspirin enhances the protection of Hsp90 from heat-stressed injury in cardiac microvascular endothelial cells through PI3K-Akt and PKM2 pathways. Cells. 9:2432020. View Article : Google Scholar :

76 

Serganova I, Cohen IJ, Vemuri K, Shindo M, Maeda M, Mane M, Moroz E, Khanin R, Satagopan J, Koutcher JA and Blasberg R: LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response. PLoS One. 13:e02039652018. View Article : Google Scholar : PubMed/NCBI

77 

Chen SF, Pan MX, Tang JC, Cheng J, Zhao D, Zhang Y, Liao HB, Liu R, Zhuang Y, Zhang ZF, et al: Arginine is neuroprotective through suppressing HIF-1α/LDHA-mediated inflammatory response after cerebral ischemia/reperfusion injury. Mol Brain. 13:632020. View Article : Google Scholar

78 

Fernández-Hernando C, József L, Jenkins D, Di Lorenzo A and Sessa WC: Absence of Akt1 reduces vascular smooth muscle cell migration and survival and induces features of plaque vulnerability and cardiac dysfunction during atherosclerosis. Arterioscler Thromb Vasc Biol. 29:2033–2040. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Pan C, Liu Q and Wu X: HIF1α/miR-520a-3p/AKT1/mTOR feedback promotes the proliferation and glycolysis of gastric cancer cells. Cancer Manag Res. 11:10145–10156. 2019. View Article : Google Scholar :

80 

Zhong ZW, Zhou WC, Sun XF, Wu QC, Chen WK and Miao CH: Dezocine regulates the malignant potential and aerobic glycolysis of liver cancer targeting Akt1/GSK-3β pathway. Ann Transl Med. 8:4802020. View Article : Google Scholar

81 

Zhao X, Wu X, Wang H, Yu H and Wang J: USP53 promotes apoptosis and inhibits glycolysis in lung adenocarcinoma through FKBP51-AKT1 signaling. Mol Carcinog. 59:1000–1011. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Song L and Schindler C: IL-6 and the acute phase response in murine atherosclerosis. Atherosclerosis. 177:43–51. 2004. View Article : Google Scholar : PubMed/NCBI

83 

Zhang P, Chen X, Zhang Y, Su H, Zhang Y, Zhou X, Sun M, Li L and Xu Z: Tet3 enhances IL-6 expression through up-regulation of 5-hmC in IL-6 promoter in chronic hypoxia induced athero-sclerosis in offspring rats. Life Sci. 232:1166012019. View Article : Google Scholar

84 

Bozic M, Alvarez A, de Pablo C, Sanchez-Niño MD, Ortiz A, Dolcet X, Encinas M, Fernandez E and Valdivielso JM: Impaired vitamin D signaling in endothelial cell leads to an enhanced leukocyte-endothelium interplay: Implications for atheroscle-rosis development. PLoS One. 10:e01368632015. View Article : Google Scholar

85 

Han J, Meng Q, Xi Q, Zhang Y, Zhuang Q, Han Y, Jiang Y, Ding Q and Wu G: Interleukin-6 stimulates aerobic glycolysis by regulating PFKFB3 at early stage of colorectal cancer. Int J Oncol. 48:215–224. 2016. View Article : Google Scholar

86 

Li H, Liang Q and Wang L: Icaritin inhibits glioblastoma cell viability and glycolysis by blocking the IL-6/Stat3 pathway. J Cell Biochem. Nov 2–2018.Epub ahead of print. View Article : Google Scholar

87 

Zhao N and Zhang J: Role of alternative splicing of VEGF-A in the development of atherosclerosis. Aging (Albany NY). 10:2695–2708. 2018. View Article : Google Scholar

88 

Wang X, Hu Z, Wang Z, Cui Y and Cui X: Angiopoietin-like protein 2 is an important facilitator of tumor proliferation, metastasis, angiogenesis and glycolysis in osteosarcoma. Am J Transl Res. 11:6341–6355. 2019.PubMed/NCBI

89 

Peek CB, Levine DC, Cedernaes J, Taguchi A, Kobayashi Y, Tsai SJ, Bonar NA, McNulty MR, Ramsey KM and Bass J: Circadian clock interaction with HIF1α mediates oxygenic metabolism and anaerobic glycolysis in skeletal muscle. Cell Metab. 25:86–92. 2017. View Article : Google Scholar

90 

Zhao M, Fan J, Liu Y, Yu Y, Xu J, Wen Q, Zhang J, Fu S, Wang B, Xiang L, et al: Oncogenic role of the TP53-induced glycolysis and apoptosis regulator in nasopharyngeal carcinoma through NF-κB pathway modulation. Int J Oncol. 48:756–764. 2016. View Article : Google Scholar

91 

Ko YH, Domingo-Vidal M, Roche M, Lin Z, Whitaker-Menezes D, Seifert E, Capparelli C, Tuluc M, Birbe RC, Tassone P, et al: TP53-inducible glycolysis and apoptosis regulator (TIGAR) metabolically reprograms carcinoma and stromal cells in breast cancer. J Biol Chem. 291:26291–26303. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Xiong Y, Yepuri G, Forbiteh M, Yu Y, Montani JP, Yang Z and Ming XF: ARG2 impairs endothelial autophagy through regulation of MTOR and PRKAA/AMPK signaling in advanced atherosclerosis. Autophagy. 10:2223–2238. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Li X, Wu L, Zopp M, Kopelov S and Du W: p53-TP53-Induced glycolysis regulator mediated glycolytic suppression attenuates DNA damage and genomic instability in fanconi anemia hematopoietic stem cells. Stem Cells. 37:937–947. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Chen Q, Lv J, Yang W, Xu B, Wang Z, Yu Z, Wu J, Yang Y and Han Y: Targeted inhibition of STAT3 as a potential treatment strategy for atherosclerosis. Theranostics. 9:6424–6442. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Zhou X, Li D, Yan W and Li W: Pravastatin prevents aortic atherosclerosis via modulation of signal transduction and activation of transcription 3 (STAT3) to attenuate interleukin-6 (IL-6) action in ApoE knockout mice. Int J Mol Sci. 9:2253–2264. 2008. View Article : Google Scholar

96 

Li Y, Wang Y, Liu Z, Guo X, Miao Z and Ma S: Atractylenolide I induces apoptosis and suppresses glycolysis by blocking the JAK2/STAT3 signaling pathway in colorectal cancer cells. Front Pharmacol. 11:2732020. View Article : Google Scholar : PubMed/NCBI

97 

Zheng M, Cao MX, Yu XH, Li L, Wang K, Wang SS, Wang HF, Tang YJ, Tang YL and Liang XH: STAT3 promotes invasion and aerobic glycolysis of human oral squamous cell carcinoma via inhibiting FoxO1. Front Oncol. 9:11752019. View Article : Google Scholar : PubMed/NCBI

98 

Seki N, Hashimoto N, Taira M, Yagi S, Yoshida Y, Ishikawa K, Suzuki Y, Sano H, Horiuchi S, Yoshida S, et al: Regulation of Src homology 2-containing protein tyrosine phosphatase by advanced glycation end products: The role on atherosclerosis in diabetes. Metabolism. 56:1591–1598. 2007. View Article : Google Scholar : PubMed/NCBI

99 

Park JS, Lee S, Jeong AL, Han S, Ka HI, Lim JS, Lee MS, Yoon DY, Lee JH and Yang Y: Hypoxia-induced IL-32β increases glycolysis in breast cancer cells. Cancer Lett. 356:800–808. 2015. View Article : Google Scholar

100 

Nam K, Oh S and Shin I: Ablation of CD44 induces glycolysis-to-oxidative phosphorylation transition via modulation of the c-Src-Akt-LKB1-AMPKα pathway. Biochem J. 473:3013–3030. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Byun S, Jung H, Chen J, Kim YC, Kim DH, Kong B, Guo G, Kemper B and Kemper JK: Phosphorylation of hepatic farnesoid X receptor by FGF19 signaling-activated Src maintains cholesterol levels and protects from atherosclerosis. J Biol Chem. 294:8732–8744. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Lovren F, Pan Y, Shukla PC, Quan A, Teoh H, Szmitko PE, Peterson MD, Gupta M, Al-Omran M and Verma S: Visfatin activates eNOS via Akt and MAP kinases and improves endothelial cell function and angiogenesis in vitro and in vivo: Translational implications for atherosclerosis. Am J Physiol Endocrinol Metab. 296:1440–1449. 2009. View Article : Google Scholar

103 

Gupta A, Mohanty P and Bhatnagar S: Integrative analysis of ocular complications in atherosclerosis unveils pathway convergence and crosstalk. J Recept Signal Transduct Res. 35:149–164. 2015. View Article : Google Scholar

104 

Perrotta P, Emini Veseli B, Van der Veken B, Roth L, Martinet W and De Meyer GRY: Pharmacological strategies to inhibit intra-plaque angiogenesis in atherosclerosis. Vascul Pharmacol. 112:72–78. 2019. View Article : Google Scholar

Related Articles

Journal Cover

January-2021
Volume 47 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang R, Wang M, Ye J, Sun G and Sun X: Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review). Int J Mol Med 47: 65-76, 2021
APA
Wang, R., Wang, M., Ye, J., Sun, G., & Sun, X. (2021). Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review). International Journal of Molecular Medicine, 47, 65-76. https://doi.org/10.3892/ijmm.2020.4798
MLA
Wang, R., Wang, M., Ye, J., Sun, G., Sun, X."Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review)". International Journal of Molecular Medicine 47.1 (2021): 65-76.
Chicago
Wang, R., Wang, M., Ye, J., Sun, G., Sun, X."Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review)". International Journal of Molecular Medicine 47, no. 1 (2021): 65-76. https://doi.org/10.3892/ijmm.2020.4798