IMR Press / FBL / Volume 29 / Issue 1 / DOI: 10.31083/j.fbl2901026
Open Access Review
The Regulatory Role and Mechanism of Energy Metabolism in Vascular Diseases
Show Less
1 Key Research Laboratory of Germplasm Resources and Standardized Planting of Genuine Regional Medicinal Materials Produced in Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
2 Laboratory of Vascular Biology and translational medicine, School of Medicine, Hunan University of Chinese Medicine, 410208 Changsha, Hunan, China
3 Department of Science and Education, The first Hospital of Hunan University of Chinese Medicine, 410007 Changsha, Hunan, China
*Correspondence: qinhuituo@hnucm.edu.cn (Qinhui Tuo)
Front. Biosci. (Landmark Ed) 2024, 29(1), 26; https://doi.org/10.31083/j.fbl2901026
Submitted: 26 August 2023 | Revised: 18 October 2023 | Accepted: 20 November 2023 | Published: 18 January 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Vascular diseases are amongst the most serious diseases affecting human life and health globally. Energy metabolism plays a crucial role in multiple vascular diseases, and the imbalance of energy metabolism in cells from the blood vessel wall can cause various vascular diseases. Energy metabolism studies have often focused on atherosclerosis (AS) and pulmonary hypertension (PH). However, the roles of energy metabolism in the development of other vascular diseases is becoming increasingly appreciated as both dynamic and essential. This review summarizes the role of energy metabolism in various vascular diseases, including AS, hemangioma, aortic dissection, PH, vascular aging, and arterial embolism. It also discusses how energy metabolism participates in the pathophysiological processes of vascular diseases and potential drugs that may interfere with energy metabolism. This review presents suggestions for the clinical prevention and treatment of vascular diseases from the perspective of energy metabolism.

Keywords
glycolysis
mitochondria
atherosclerosis
hemangioma
aortic dissection
pulmonary hypertension
1. Introduction

Various pathologies associated with the vasculature, including hypertension, atherosclerosis (AS), aortic aneurysm, aortic dissection, pulmonary hypertension (PH), and stenosis, are considered to be some of the most serious diseases affecting human life and health worldwide [1]. Furthermore, energy metabolism is known to play a crucial role in the etiology of a number of vascular diseases. An imbalance of energy metabolism has been closely associated with vascular diseases, such as AS [2], abdominal aortic aneurysm [3], PH [4], aortic dissection [5], and vascular aging [6]. Pathological changes in specific vascular diseases usually cause changes in the energy metabolism of cells in the vascular wall, including vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs), and macrophages [7, 8, 9].

Energy metabolism is critical for maintaining normal cell function and life. In eukaryotes, nutrients are converted by glycolysis to pyruvate, which then enters the mitochondria, where it is further decarboxylated to acetyl coenzyme A (acetyl-CoA). Under aerobic conditions, a acetyl-CoA enters the tricarboxylic acid (TCA) cycle where it is reduced through the oxidative respiratory chain pathway to produce nicotinamide adenine dinucleotide (NADH), with the generation of H2O, CO2 as end-products. The energy released in this process leads to mitochondrial oxidative phosphorylation (OXPHOS) of adenosine diphosphate (ADP) to generate adenosine triphosphate (ATP), which is used to maintain the normal physiological functions of cells. However, under hypoxic conditions or when mitochondrial function is damaged, pyruvate can instead be converted to lactate for use in anaerobic glycolysis [10, 11, 12] (Fig. 1). OXPHOS and glycolysis maintain the energy metabolism homeostasis of cells. Imbalance of energy metabolism homeostasis is linked to various pathological conditions; for instance, obesity brain homeostasis, cardiovascular disease, and cancer [13, 14, 15].

Fig. 1.

Mitochondrial oxidative phosphorylation and glycolytic pathways. Abbreviations: ATP, adenosine triphosphate; NAD+, nicotinamide adenine dinucleotide; NADH, nicotinamide-adenine dinucleotide-reduced; TCA, tricarboxylic acid; ETC, electron transport chain. By Figdraw.

Mitochondrial respiration and glycolysis are the major energy metabolism pathways in cells. However, they also regulate various cellular processes, including differentiation, proliferation, apoptosis, immune response, cell death, and the transmission of secondary signals to the nucleus, all of which play important roles in a variety of vascular diseases [16, 17, 18]. It is therefore imperative to study the role of energy metabolism in vascular diseases. This review will be elaborated the regulatory role and mechanism of energy metabolism in vascular diseases in two part. The first part of this review will summarize mechanisms responsible for the regulatory of energy metabolism in vascular diseases, such as AS, hemangioma, aortic dissection, PH, vascular aging, and arterial embolism. The second part will discuss drugs and clinical treatment strategies, which might help to improve energy metabolism function in vascular diseases.

2. Atherosclerosis and Energy Metabolism

AS is a chronic inflammatory vascular disease driven by abnormal lipids and characterized by progressive thickening and the gradual formation of plaques on the blood vessel wall [19]. Cell of the arterial wall are very vulnerable to mitochondrial dysfunction due to their barrier and metabolic functions. Mitochondrial dysfunction in AS causes alteration in cellular metabolism and respiration, leading to the production of excessive amounts of reactive oxygen species (ROS) and resulting in oxidative stress [20]. Mitochondrial DNA (mtDNA) is more susceptible to oxidative stress, making it vulnerable to oxidation. Induction of mtDNA damage is a critical mechanism in the development of AS [21, 22]. Damaged mtDNA is usually accompanied by a change in the number of abnormal mitochondria due to alterations in mitochondrial fusion and fission, thereby leading to abnormal energy metabolism in AS [23]. Mutations in mtDNA also lead to an increased number of in pro-inflammatory macrophages. These phagocytose large amounts of lipid to form foam cells, resulting in cell death and necrotic plaque cores. The destruction of mitochondrial membranes, inhibition of the respiratory chain, and the reduction in mitochondrial enzyme activity, can also affect energy metabolism in AS [24]. Yu E, et al. [25] confirmed that mtDNA damage is seen early in AS in Apolipoprotein E knockout (ApoE-/-) mice. To then determine whether mtDNA damage promotes AS, They studied ApoE-/- mice with defficient polymerase-γ proofreading activity (polG-/-/ApoE-/-). The results suggested that mtDNA damage promotes AS and affects plaque composition independently of ROS in polG-/-/ApoE-/- mice through effects on VSMCs, monocytes, and hyperlipidemia [25].

Besides mitochondrial metabolism, the glycolytic process is the chief source of rapid energy production in response to environmental changes, with the rate of glycolysis playing a critical role in maintaining cellular homeostasis. Intermediates produced during glycolysis can affect cell viability and thus increase the risk of AS [26, 27]. Moreover, abnormalities in mitochondrial and glycolytic energy metabolism can affect the function of VECs, the proliferation of VSMCs, and the polarization of macrophages, which ultimately promote the progression of AS lesions (Fig. 2).

Fig. 2.

Changes of energy metabolism in AS. (A) Energy metabolism imbalance and AS. (B) Signaling regulation of glycolysis and mitochondrial dysfunction in endothelial cells. (C) Signaling regulation of the glycolysis pathway in vascular smooth muscle cells. (D) Comparison of energy metabolism between M1 and M2 macrophages. Abbreviations: AS, atherosclerosis; ECs, endothelial cells; NOX2, NADPH oxidase 2; ROS, reactive oxygen species; GLUT1, Glucose transporter protein 1; M1, M1 macrophage; M2, M2 macrophage; GSH, glutathione; GSSG, glutathione disulfide; OCR, oxygen consumption rate. By Figdraw.

2.1 Energy Metabolism of Vascular Endothelial Cells and Atherosclerosis

Endothelial dysfunction is a key early event in the pathogenesis of AS. Unlike other cell types, the energy required for the physiological function of VECs primarily derives from glycolysis [28]. Even when oxygen is sufficient, VECs preferentially catabolize glucose through aerobic glycolysis rather than OXPHOS [29, 30]. Therefore, aerobic glycolysis provides various substrates for cell proliferation and DNA synthesis.

VECs that are exposed to disturbed flow patterns in areas of arterial bifurcation or curvature are more vulnerable to injury and loss of integrity, and show increased glycolysis both in vitro and in vivo [31]. Multiple mechanisms are involved in this process, protein kinase AMP-activated catalytic subunit alpha 1 (PRKAA1)/AMP-activated protein kinase α1 (AMPKα1), is a major regulator of endothelial glycolytic metabolism, and is upregulated under pathological shear stress. Increased levels of PRKAA1/AMPKα1 signaling promote the expression of hypoxia-inducible factor-1α (HIF-1α). This, in turn, drives the transcription of glycolytic enzymes, thereby increasing glycolysis in VECs. PRKAA1/AMPKα1-mediated glycolysis is critical for increased VEC proliferation, thus helping to preserve the integrity of the VEC barrier in regions that are vulnerable to AS and protecting against the development and progression of AS in a mouse model [27, 29]. Similarly, impaired endothelial glycolysis is associated with various vascular pathologies, including impaired angiogenesis and the development of AS. Interferon-γ inhibits basal glycolysis through global transcriptional suppression of glycolytic enzymes in quiescent primary human coronary artery ECs leading to basal reduced intracellular guanosine 3, 5-cyclic monophosphate (cGMP), impaired endothelial migration, and a shift towards a pro-inflammatory state [32].

Although aerobic glycolysis is beneficial for VEC proliferation; pathological angiogenesis also increases [33]. Aerobic glycolysis can bypass OXPHOS, so that the rate of glucose hydrolysis is faster. This can produce large amounts of energy very rapidly for VECs, thus accelerating their migration and proliferation, and promoting the formation of AS plaques [34]. The new vessels developed as a consequence of aerobic glycolysis are immature and have structural and functional abnormalities, thereby leading to plaque hemorrhage which is a major risk factor for the development of AS [34, 35]. Thus, aerobic glycolysis in VECs can have both beneficial and harmful roles in the development of AS.

Mitochondrial respiration is not the main energy metabolism pathway in VECs, but the disruption of mitochondrial redox homeostasis in VECs can cause chronic inflammation, which then contributes substantially to AS development [36]. Recently, the disruption of mitochondrial redox homeostasis in VECs was shown to be a key factor in triggering inflammation. Reducing or neutralizing mitochondrial ROS levels may therefore be an effective strategy to mitigate inflammation, promote endothelial repair, and inhibit AS development [37]. The primary enzymatic source of ROS during cardiovascular pathogenesis is the nicotinamide adenine dinucleotide phosphate oxidase (NOX) family of proteins. These are mainly expressed in VECs and provoke oxidative inactivation of nitric oxide (NO) and the consequent uncoupling of endothelial nitric-oxide synthase (eNOS), leading to sustained oxidative stress [38] (Fig. 2B). NOX has become a target for drug therapy in AS, Tanshinone IIA, for example, can attenuates oxidative stress by decreasing NOX2 and NOX4 levels and downregulating the nuclear factor-κB (NF-κB) signaling pathway, which may help to ameliorate AS [39].

2.2 Energy Metabolism of Vascular Smooth Muscle Cells and Atherosclerosis

Abnormal proliferation of VSMCs promotes plaque formation in the early stage of AS lesions, but the apoptosis and senescence of VSMCs and VSMC-derived macrophage-like cells may promote inflammation and plaque rupture in the late stages of AS. Therefore, VSMCs are beneficial during the advanced stage of plaques, for example, by preventing the fibrous cap from rupturing [40, 41]. A major event leading to AS development is the change in VSMC phenotype from a physiological contractile state to an adverse proliferative or synthetic state, accompanied by enhanced aerobic glycolysis [42].

Aerobic glycolysis is the main energy metabolism pathway in VSMCs. Even though approximately 90% of glucose is used in aerobic glycolysis, this pathway only produces about 30% of the ATP is produced [42]. Currently there are two hypotheses to explain this: The first is that the ATP produced by glycolysis and glucose oxidation provides energy for different physiological processes. For example, ATP produced by glycolysis is used to supply energy for sodium and potassium transport, whereas ATP produced by OXPHOS supplies energy for other physiological processes [43]. The second is that the large amount of lactate produced by glycolysis can enhance the reserve capacity of mitochondria. The lactate can then be converted to pyruvate by lactate dehydrogenase after entering the mitochondria and being completely oxidized to produce CO2 and H2O. In this process, all of the energy stored in lactate is completely released and participates in various biochemical activities in cells and organisms. Under ATP-deficient conditions, glycolysis can promote the proliferation of VSMCs by producing lactate [44, 45, 46] (Fig. 2C).

The uptake of carbohydrates in VSMCs is determined by glucose transporter 1 (GLUT1), which can further affect energy metabolism and especially the glycolytic pathway [47, 48]. The GLUT1-mediated glycolytic pathway in VSMCs has been closely associated with AS [49]. In one study, GLUT1 overexpression led to a 44% increase in intracellular glucose concentration in rat aortic smooth muscle cell lines (A7r5) and in human VSMC lines. An increase in the glucose flux through glycolysis and the TCA cycle further promotes the proliferation of VSMCs [42] (Fig. 2C). Overexpression of GLUT1 can also inhibit VSMC apoptosis by inhibiting caspase-8 activation in mouse primary VSMCs, thereby promoting cell proliferation [50]. Metabolic reprogramming may be involved in VSMC phenotypic transition. Prohibitin 2 is a pleiotropic scaffold protein involved in the regulation of energy metabolism and proliferation, which maintains VSMC contractile phenotype by interacting with heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) to counteract hnRNPA1-mediated pyruvate kinase isoform M2 (PKM2) alternative splicing and glucose metabolic reprogramming [45].

Furthermore, VSMCs were recently found to rely on a combination of mitochondrial and glycolytic metabolism, with the energy switch occurring at different stages in the AS disease process. Mitochondrial dysfunction and DNA damage are defining traits of aged VSMCs in AS [51]. Klotho is an anti-aging protein that could play a significant role in improving the VSMC phenotype. Targeting Klotho during early stages of AS could induce energy switching and improve VSMC survival in the plaque [51]. A follow-up study demonstrated that phosphatase and tensin homolog deleted on chromosome ten (PTEN) induced putative kinase 1 (Pink1) kinase can regulate mitochondrial homeostasis and the recycling of dysfunctional organelles that are critical for maintaining energetic homeostasis [52]. Energy switching between oxidative and glycolytic metabolism were found to be regulated by Pink1.

2.3 Energy Metabolism of Macrophages and Atherosclerosis

Macrophages exist in two distinct subsets: the classically activated M1 macrophages, and the alternatively activated M2 macrophages [53]. Glycolysis is the main energy pathway in M1 macrophages, and the TCA cycle and OXPHOS are impaired in these cells. In contrast, M2 macrophages are more dependent on mitochondrial OXPHOS [54]. GLUT1 is a rate-limiting glucose transporter in pro-inflammatory polarized macrophages. Overexpression of GLUT1 in macrophage/monocyte-like leukemia cells (RAW264.7) resulted in a significant enhancement of glucose uptake and metabolism, and increased production of pentose phosphate pathway intermediates [55]. With increased the enhancement of glycolytic metabolism, the proportion of M1 macrophages also increases, followed by reduced lipid oxidative metabolism and lipid accumulation [56]. The polarization of M1 macrophages induces AS by increasing ROS levels, which then cause OXPHOS dysfunction. This in turn increases the rate of glycolysis and stimulates pro-inflammatory reactions and the synthesis of inflammatory factors [57]. As a consequence, glycolysis is increased in M1 macrophages together with the levels of NADH:NAD+ and glutathione (GSH):glutathione disulfide (GSSG), while the O2 consumption rate (OCR) and O2 consumption both decrease. In contrast, glycolysis is decreased in M2 macrophages, together with NADH:NAD+ and GSH:GSSG levels, whereas the OCR and O2 consumption levels increase (Fig. 2D) [58, 59].

Recent studies highlight the importance of mitochondria in the metabolic and signaling pathways necessary to maintain macrophage effector functions [60]. Lipoprotein, oxidized phospholipid, and oxidized low-density lipoprotein (ox-LDL) present within AS plaques can cause oxidative stress-induced damage to macrophage mitochondria [61]. Mild oxidative stress damages the mitochondrial respiratory chain, stimulates mtDNA replication and transcription, and increases the mtDNA copy number. However, under severe oxidative stress the damaged mitochondria gradually lose the ability to self-regulate, which then leads to decreased rates of replication and transcription and a decrease in mtDNA copy number. This ultimately affects mitochondrial function and leads to abnormal energy metabolism [62]. Concurrently, oxidative stress damage to mitochondria in macrophages causes dysfunction of the mitochondrial electron transport chain (ETC), leading to stalled electron transport and increased ROS production [63]. Macrophage polarization is activated by increased ROS through a variety of signaling pathways. For example, activation of the mitogen-activated protein kinase (MAPK)-dependent nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathways by ROS-induced cytotoxicity ultimately increases the transcription of pro-inflammatory cytokines and other inflammatory mediators that promote polarization of M1 macrophages in RAW264.7 cells [64]. Consequently, ROS is generated in AS plaques as a result of damaged ETC function and further induces the polarization of M1 macrophages. Once polarized, the energy metabolism of macrophages relies mainly on glycolysis, meaning that fat metabolism is reduced and leading to further lipid accumulation and the deterioration of AS plaques [65, 66].

In addition to oxidative stress damage to mitochondria in macrophages, HIF-1α is involved in ox-LDL-induced effects in macrophages, including the inflammatory response, angiogenesis, and metabolic reprogramming. HIF-1α signaling promotes the accumulation of triglycerides and cholesterol in macrophages. Hypoxia and HIF-1α increase the uptake of ox-LDLs, promote cholesterol and triglyceride synthesis, and decrease cholesterol efflux. The effect of HIF-1α on cholesterol homeostasis within macrophages, and activation of the inflammatory response by oxysterols via HIF-1α, could have deleterious consequences in AS [67]. HIF-1α can also activate the glycolytic pathway in macrophages. Meanwhile, mitochondrial function in these cells is decreased, thereby changing the mode of energy metabolism, increasing the level of inflammatory factors, accelerating macrophage polarization to the M1 type, and hence causing the progression of AS [57, 68].

3. Energy Metabolism and Hemangioma

Hemangioma is the most common benign tumor of infancy and is caused by dysfunction during embryogenesis and vasculogenesis. Hemangiomas progress during fetal life and manifest fully at birth. They arise in the skin and soft tissues and can occur at birth or shortly after birth [69]. The primary characteristics of hemangioma are a high incidence rate and strong destructiveness. Hypoxia and abnormal glucose metabolism are considered to be key factors in the pathogenesis of this disease [70].

Under normoxic and hypoxic conditions, glucose consumption and ATP production occur at higher levels in hemangioma-derived endothelial cells compared to human umbilical vein endothelial cells [70]. In addition, several glycolysis-related genes are expressed at higher levels, including GLUT1, hexokinase 2 (HK2), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), PKM2, and lactate dehydrogenase A (LDHA). Inhibition of glycolytic-related genes in hemangioma-derived ECs can reduce the proliferation, migration, and angiogenesis of these cells. The above results suggest that increased glycolytic metabolism is an important cause of infantile hemangioma [70, 71]. Some genes related to glycolysis are also highly expressed in infantile hemangioma. For example, the expression of LncRNA MCM3AP-AS1 is significantly upregulated in infantile hemangiomas and also promotes their progression by increasing glycolysis via regulation of the miR-138-5p/HIF-1α axis [72].

Although tumor cells rely on glycolysis for energy supply, mitochondrial transformations in bioenergetics, metabolism, and fission-fusion dynamics are also likely to be important. This is especially true for rapidly growing tumor cells that rely on mitochondrial function for their sustenance [73]. However, some studies have identified tumorigenic cell populations that rely more on mitochondrial respiration than on glycolysis [74, 75, 76]. Mouse hemangioendothelioma ECs have been used to study the mechanism of hemangioendothelioma, which depend heavily on mitochondria to thrive. The targeting of mitochondria may therefore be an effective strategy for the management of hemangiomas [77].

4. Energy Metabolism and Aortic Dissection

Aortic dissection is a life-threatening condition in which blood in the aortic cavity enters the aortic medial membrane from a tear in the aortic intima, resulting in separation of the different layers of the aortic wall. Although the incidence of aortic dissection is low, it has a major clinical impact due to its high mortality and poor prognosis [78]. The potential mechanisms underlying energy metabolism-induced aortic dissection primarily involve two aspects: ROS and inflammation [79, 80].

Fan LM, et al. [81] investigated the special role of endothelial ROS in the development of aortic dissection by establishing a mouse model with endothelium-specific Nox2 overexpression. These workers then determined the susceptibility of the Nox2 over-expressing mice to aortic dissection after infusion with angiotensin II (Ang II). Compared to wild-type mice, a specific increase in endothelial ROS production in the Nox2 transgenic mice was sufficient to cause Ang II-mediated aortic dissection. Increased expression of Nox2 in VECs can lead to increased ROS production, upregulated expression of vascular cell adhesion molecule-1 (VCAM-1), enhanced matrix metalloproteinase activity, and accelerated CD45 inflammatory cell infiltration. Compared to controls, ECs from Nox2-overexpressing mice grown in conditioned medium induced greater phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) in VSMCs by secreting cyclophilin A (CypA). These findings suggest that VEC-derived ROS plays a critical role in determining the susceptibility of the aortic wall to dissection mediated by Ang II. The ROS-dependent secretion of CypA by VECs is an important signaling mechanism that regulates the susceptibility of aortic wall structural components to aortic dissection. These results suggest that endothelial ROS and CypA are potential therapeutic targets for the prevention of aortic dissection [81].

Aortic dissection is a severe inflammatory vascular disease associated with the metabolic reprogramming of macrophages. The effect of macrophage metabolic reprogramming has been studied in a mouse model by inducing aortic dissection with β-amiopropanitrile (BAPN, a lipoxygenase inhibitor) and then collecting tissue samples from the vessels. Changes in the macrophage levels of HIF-1α in the animal model and in the tissues of patients with aortic dissection were evaluated by immunohistochemistry, immunofluorescence, Western blotting, and quantitative polymerase chain reaction. Macrophage HIF-1α expression was upregulated in the aorta of patients with aortic dissection, as well as in the mouse model. Mechanistically, activated HIF-1α in macrophages triggers vascular inflammation, extracellular matrix degradation, and elastic plate disruption by increasing the secretion of disintegrin and metalloprotease. Meanwhile, BAPN was found to impair mitochondrial oxidative phosphorylation in macrophages by enhancing HIF-1α activation and glycolytic metabolism, and by promoting metabolic reprogramming. Therefore, HIF-1α is activated by the metabolic reprogramming of macrophages, which subsequently favors aortic dissection. Inhibition of macrophage HIF-1αmay therefore be a potential treatment strategy for aortic dissection [80].

5. Energy Metabolism and Pulmonary Hypertension

Pulmonary Hypertension (PH) is a pulmonary vascular disease with complex etiology. It is characterized by the remodeling and occlusion of small peripheral pulmonary vessels, and a progressive increase in pulmonary vascular resistance. This eventually leads to hypertrophy of the right ventricle and right heart failure [82]. The pathological characteristics of PH are excessive proliferation and apoptosis tolerance of VSMCs, adventitial fibroblasts and VECs, eventually leading to continuous pulmonary vascular wall thickening and pulmonary vascular remodeling [83]. Changes in metabolism and bioenergetics are increasingly recognized as universal hallmarks of PH in patients and in animal models of this disease [84].

Excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) is the main cause of hypoxic PH. Molecules that induce abnormal proliferation of VSMCs may trigger PH [85]. For example, the increase in the main end products of intracellular lipid peroxidation, 4-HNE, leads to abnormal proliferation of VSMCs, thus promoting the development of PH. Therefore, inhibiting the intracellular increase of 4-HNE can prevent the occurrence and progression of PH. Aldehyde dehydrogenase 2 (ALDH2) is a detoxifying enzyme for 4-HNE, which can regulate mitochondrial fission and VSMC proliferation through the 4-HNE/HIF-1α/Drp1 (dynaminrelated protein 1) signaling pathway. Ultimately, this mechanism helps prevent hypoxia-induced PH [86]. Other enzymes in the ALDH family, such as ALDH1A3, can also regulate glycolysis in SMCs, thereby regulating the proliferation of SMCs [87]. The latest research shows that elevated ALDH1A3 can orchestrate changes in metabolism needed both for cell energy and for targeted epigenetic and transcriptional regulation of genes required by dividing cells. To elucidate mechanisms accounting for ALDH1A3-mediated PH PASMC proliferation, Li D, et al. [88] compared the transcriptome of PAH PASMC under basal conditions and when ALDH1A3 was reduced. They found that ALDH1A3-dependent regulation of mRNA levels of genes related to cell cycle and metabolism. Dihydrolipoyl dehydrogenase (DLD), PKM2, and isocitrate dehydrogenase 1 (IDH1) contribute to the metabolic shift initiated by ALDH1A3 in PH PASMC and also impact nucleotide synthesis via PKM2 and cell cycle genes required for cell proliferation [88].

An emerging metabolic theory for PH suggests that cellular and mitochondrial metabolic dysfunction underly the pathology of this disease [89]. Zhang et al. [89] showed that high expression of polypyrimidine tract-binding protein 1 (PTBP1), controlled by decreased expression of miR-124, underlies the elevated PKM2/PKM1 ratio and subsequent metabolic reprograming of pulmonary adventitial fibroblasts isolated from calves and humans with severe PH. These findings are similar to those made in cancer cells and support the idea that pyruvate metabolism can be exquisitely controlled by miR-124 and by PTBP1 [89].

Metabolic changes together with endothelial dysfunction that favor increased glycolysis are important in the pathophysiology of PH [90]. Lung ECs express high levels of glucose metabolic enzymes, such as PFKFB3, and consequently produce large amounts of glucose metabolites. Low levels of HIF-1α and HIF-2α were noted in PFKFB3-knockdown pulmonary artery endothelial cells (PAECs), whereas high levels of HIF-1α and HIF-2α were observed in PFKFB3-overexpressing PAECs. Knockdown of HIF-2α, but not HIF-1α, reduced the elevated expression of growth factors and proinflammatory cytokines in PFKFB3-overexpressing PAECs. This indicates the critical involvement of HIF-2α in PFKFB3-mediated expression of growth factors and proinflammatory cytokines in PAECs [7]. Furthermore, the blocking of endothelial PFKFB3 inhibits PH development in rodent models, suggesting that targeting of glucose metabolic enzymes may be a promising strategy for the treatment of PH. Recent research has shown that intracellular chloride channels regulate endothelial metabolic reprogramming in PH. In particular, chloride intracellular channel 1 (CLIC1) and CLIC4 are highly expressed in the lung endothelium of patients with PH and preclinical models of PH. Moreover, their activation has been linked with better endothelial repair, apoptosis resistance, and angiogenesis [91], CLIC1/4 is localized in the inner and outer mitochondrial membranes of VECs. Common and unique CLIC1- and CLIC4-interacting partners create a link between CLIC signaling, mitochondrial dysfunction, and mitofusin-2 deficiency in PH endothelium [4].

6. Energy Metabolism and Vascular Aging

Vascular aging involves a deterioration in the normal function of vessels, resulting in increased arterial stiffness and pulse wave propagation rate and accumulation of oxidative damage. Vascular aging also greatly increases the incidence of AS and PH [92, 93]. Mitochondrial dysfunction plays a critical role in disease progression. For example, mitochondria-targeted esculetin alleviates AS in the setting of aging by delaying vascular senescence and pro-inflammatory processes, and by improving mitochondrial biogenesis and function [92].

Senescence appears to be closely linked with changes in cell metabolism. Replicative senescent VECs show upregulation of glycolytic and oxidative glucose metabolism compared to young VECs. This was demonstrated by metabolic profiling and glucose flux measurements, and by analyzing the expression of key metabolic enzymes. Senescent cells exhibit higher glycolytic activity and lactate production, enhanced expression of LDHA, and increased activity of the TCA cycle and mitochondrial respiration [6].

Age-related endothelial dysfunction is mediated primarily by the reduced bioavailability of NO. Impaired NO-mediated endothelial function is largely due to excessive production of ROS, which reacts with NO to reduce its bioavailability. The resulting arterial oxidative stress leads in turn to vascular aging and dysfunction [94]. The effects of oxidative stress on vascular aging also correlate with endothelium-dependent dilation (EDD), which is a marker of endothelial dysfunction. Impaired or reduced EDD can promote vascular aging and the stiffening of arteries by increasing the tension in arterial activity. Increased expression of NADPH oxidase and abnormal mitochondrial function lead to increased ROS production, decreased NO bioavailability, damaged EDD, and age-related endothelial dysfunction [95]. Furthermore, the expression of serine biosynthetic enzyme phosphoglycerate dehydrogenase (PHGDH) is significantly reduced due to decreased transcription of the activating transcription factor 4 (ATF4) during senescence, which leads to reduction of intracellular serine. Mechanistically, PHGDH interacts with PKM2, which prevents p300/CBP-associated factor (PCAF)-catalyzed PKM2 K305 acetylation and subsequent degradation by autophagy. In addition, PHGDH facilitates p300-catalyzed PKM2 K433 acetylation, which promotes PKM2 nuclear translocation and stimulates its activity to phosphorylate H3T11 and regulate the transcription of senescence-associated genes. Vascular endothelium-targeted expression of PHGDH and PKM2 ameliorates ageing in mice. This findings reveal that enhancing serine biosynthesis could become a therapy to promote healthy ageing [96].

7. Energy Metabolism and Arterial Embolism

Arterial embolism involves the narrowing or occlusion of the lumen due to AS, thus making it difficult to transport nutrients in the blood to the limbs and resulting in ischemia and serious tissue necrosis [97]. The occurrence of arterial embolism causes changes in energy metabolism. Insufficient blood supply and hypoxia not only lead to changes in energy metabolism, but also decrease mitochondrial activity and increase the carbon dioxide level, which may cause hypercapnia [98]. However, most of research on the relationship between energy metabolism and arterial embolism was carried out in the 1990s, with almost no recent research. The reason may be that the inducing factors for arterial embolism are related to hypertension, hyperlipidemia, AS, etc. Therefore, study of the inducing factors for arterial embolism and energy metabolism may be more important for the prevention of arterial embolism. Here, we review the research progress on arterial embolism and energy metabolism during the 1990s.

In one study, cerebral ischemia was induced in rats by administering microspheres into the right carotid canal, and the time course of changes in brain energy metabolism was examined on day 7 post-surgery. Nearly 50% of the operated rats showed the typical symptoms of stroke, i.e., cerebral ATP and creatine phosphate (PCr) were significantly decreased in the right hemisphere of microsphere-induced cerebral ischemia from day-1 to day-7, while tissue lactate levels were increased on day-1, -3, and -5 after embolism [99]. These results indicate that ischemia alters the energy metabolism of rat cerebra. Subsequently, Takeo et al. [98] studied the sustained damage to cerebral regions of rat brain induced by cerebral ischemia. These workers examined animal behavior and cerebral energy metabolism after microsphere embolization. On day-1 of post-microsphere embolization, the lactate content increased to its peak, and the ATP and PCr contents decreased to their lowest level in these cerebral regions in the right hemisphere. The glucose and glycogen content in these regions increased to their peak on day-3. Although these markers returned to control levels over time, metabolic changes in all of these areas persisted for up to 28 days post-surgery. The behavioral scoring as a result of the temporal changes after microsphere embolization showed stroke-like symptoms on day-1 after embolization, with the symptoms disappearing relatively quickly with the passage of time. These observations indicate that microsphere embolization in brain regions can cause extensive and sustained damage to energy metabolism [98].

Hepatic energy metabolism and oxidative attack were also studied following transcatheter arterial embolization (TAE) and chemoembolization (TAC) in the left and median lobes of the liver in a rat model with thioacetamide-induced liver cirrhosis. The energy charge of the embolized lobes of the lungs decreased from 0.86 to 0.78 and 0.74 one hour after the TAE and TAC processes, respectively, but recovered after 3 h. The ATP and total adenine nucleotide content of embolized and non-embolized lobes were also temporarily reduced. Total hepatic blood flow in the catheter-embolized and chemo-embolized lobes decreased by nearly 50% and returned to normal after 1 week. After chemoembolization for 3 h, the total liver glutathione content decreased from 7.02 µmol/g to about 4.5 µmol/g, while after chemoembolization for 6 h the malondialdehyde content increased from 196.94 nmol/g to more than 300 nmol/g. However, these parameters did not change in the catheter-embolized liver. The above results suggest that hepatic energy metabolism is temporarily altered by ischemia after treatment with TAE and TAC [100].

8. Discussion and Perspective

The regulation of energy metabolism in various tissues and cells helps the body adapt to environmental changes. These regulatory disturbances are closely related to the molecular pathology of many vascular diseases, including AS, PH and hemangioma [70, 84, 101]. Under specific pathological conditions, glycolysis and the mitochondrial energy pathway can affect VEC function, macrophage polarization, and VSMC proliferation [51, 102, 103]. Abnormal glycolysis metabolism in VECs, VSMCs, and macrophages is related to AS, infantile hemangioma, and PH [70, 84, 101]. In addition to glycolysis, mitochondrial metabolism is also crucial for vascular diseases such as AS, aortic dissection, PH, vascular aging, and arterial embolism [20, 90, 104]. In vitro and in vivo studies indicate that regulation of energy metabolism-related molecules and pathways is used to maintain cellular metabolic homeostasis and to prevent vascular diseases. The mechanisms for preventing vascular diseases include multiple genes and signaling pathways involved in energy metabolism, such as PRKAA1/AMPKα1 [29], GLUT1 [47], Prohibitin 2 [45], NOX [39], Klotho [51], Pink1 [105], ROS- and MAPKs-dependent Nrf2/HO-1 signaling pathways [64], HIF-1α [67], key glycolytic enzymes (HK2, PFKFB3, PKM2, and LDHA) [70], ALDH2 [86], ALDH1A3 [87], microRNA-124/PTBP1 [89], CLIC1/CLIC4 [4], and PHGDH [96].

Through literature analysis, GLUT1 is closely associated with the energy metabolism of various vascular diseases, which overexpressed in high glycolytic flux, resulting in the overproduction of pyruvate, alanine, and lactate [106]. Concurrently, VECs and VSMCs display characteristics of cell proliferation and migration, while the polarization of M1 macrophages also increases [48, 107, 108]. NOX2, ROS, Pink1 and NF-κB are important molecules closely related to mitochondrial metabolism. Excessive ROS can directly target mitochondria, decouple mitochondrial ETC, and reduce ATP production. ROS is therefore highly deleterious to mitochondria and can lead to several vascular diseases [109, 110]. HIF-1α is involved in various pathophysiological processes, such as oxygen supply and cell metabolism, and is also important in AS, infantile hemangioma, and aortic dissection [58, 72, 80]. Inhibition of HIF-1α helps to shift from glycolysis to normal aerobic oxidation. Therapeutic targeting of HIF-1α can also partially reverse the promoting effect of glycolysis on infantile hemangioma by upregulating the expression of LncRNA MCM3AP-AS1 [72].

Treatment strategies for vascular diseases that target energy metabolism have attracted much attention, especially the use of drugs. Pharmacotherapies that target endothelial dysfunction caused by energy metabolism disorders can potentially be used to treat AS and many other vascular diseases [111]. For example, the small molecule 3PO (3-[3-pyridyl]-1-[4-pyridyl]-2-propen-1-ketone) may exert beneficial effects against plaque formation by partially inhibiting the glycolysis-mediated impairment of new angiogenesis without affecting plaque composition, as well as by increasing the number of autophagosomes in VECs [112]. 3PO is known to be an inhibitor of PFKFB3. Heterozygous global deficiency of Pfkfb3 was shown to protect mice from developing hypoxia-induced PH, while administration of 3PO almost completely prevented PH in rats treated with Sugen 5416/hypoxia [7]. Tetrahydrobiopterin (BH4) has been shown to reduce endothelial NO synthase uncoupling, reverse oxidative stress-mediated inhibition of EDD, and improve EDD in the elderly, thereby alleviating vascular aging [95]. Furthermore, natural products may be a good primary source of drugs for the treatment of vascular diseases. Extracts of Rhodiola and Salidroside have been suggested to provide beneficial effects for metabolic disorders and AS. The postulated mechanisms for Rhodiola extracts and Salidroside in the treatment of AS involve energy metabolism and include oxidative stress, inflammation, mitochondria, and cell death [113]. Apocynin, extracted from the roots of Apocynum cannabinum and Picrorhiza kurroa, is an inhibitor of NADPH oxidase and could therefore restore EDD and NO levels in vitro and alleviate vascular aging [95, 114]. Oral apigenin supplementation can reverse two clinically important and age-related indicators of arterial dysfunction, namely vascular endothelial dysfunction and large elastic artery stiffening [115]. This novel work also showed that apigenin prevents foam cell formation in an established cell culture model of early AS. The above preclinical findings provide an experimental basis for future translational studies to assess the potential of natural products in treating arterial dysfunction and reducing vascular disease risk. In addition, the non-selective-adrenergic receptor antagonist propranolol is already in clinical use and is becoming the first line therapy as an approved treatment for infantile hemangioma. The latest evidence showed that propranolol-induced apoptosis in VECs may occur via mitochondrial stress [116]. Thus, it would be advantageous to discover the mechanisms related to energy metabolism for drugs that are already used in clinical practice.

In terms of clinical research, a mitochondrial-targeted antioxidant (MitoQ) has been shown to improve vascular mitochondrial function. Acute oral administration of the MitoQ (80 mg) can effectively improve endothelial function and superoxide dismutase in patients with peripheral artery disease (PAD). In addition, acute intake of MitoQ has also been found to be effective in improving maximum walking capacity and delaying claudication onset in PAD patients [117]. Pyruvate dehydrogenase kinase (PDK), an inhibitor of the mitochondrial enzyme pyruvate dehydrogenase in the pulmonary arteries of human PH compared to healthy lungs. In a 4-month, open-label study, DCA (PDK inhibitor dichloroacetate, 3 to 6.25 mg/kg b.i.d.) administered to patients with idiopathic PH already on approved idiopathic PH therapies led to reduction in mean pulmonary artery pressure and pulmonary vascular resistance and improvement in functional capacity [118].

Besides medication, there may be other potential therapeutic methods for the treatment of vascular diseases, such as cytotherapy. ALDHs are a superfamily that includes 19 subtypes in humans. ALDH2 is the mitochondrial isoform of ALDH and a key metabolizer of acetaldehyde. It is known to be a protective factor for vascular diseases such as AS, aortic dissection, aortic aneurysm, and PH by clearing toxic aldehydes generated from lipid peroxidation [86, 119, 120]. A Bayesian network meta-analysis of randomized controlled trials showed that intra-arterial injection of large amounts of ALDH-positive cells could improve the activity of daily living in ischemic stroke patients [121]. Therefore, we speculate that cytotherapy is a potential treatment for vascular disease.

9. Conclusions

Cellular energy metabolism is a crucial factor in vascular disease, with a number of studies having been conducted over the years to investigate its role. The vascular disease types related to energy metabolism include AS, hemangioma, aortic dissection, PH, vascular aging, and arterial embolism. Some drugs can directly or indirectly affect the expression of key rate-limiting enzymes in glycolysis, or regulate the mitochondrial energy metabolism pathway. In summary, this review should increase our understanding of energy metabolism in various vascular diseases. It may also help to guide future research efforts aimed at exploring the pathogenesis of vascular diseases and clinical treatment methods.

Abbreviations

Acetyl-CoA, acetyl coenzyme A; ADP, adenosine diphosphate; ALDH1A3, aldehyde dehydrogenase 1A3; ALDH2, aldehyde dehydrogenase 2; AMPKα1, AMP-activated protein kinase α1; Ang II, angiotensin II; ApoE-/-, Apolipoprotein E knockout; AS, atherosclerosis; ATF4, activating transcription factor 4; ATP, adenosine triphosphate; A7r5, aortic smooth muscle cell lines; BAPN, β-amiopropanitrile, a lipoxygenase inhibitor; BH4, Tetrahydrobiopterin; cGMP, guanosine 3, 5-cyclic monophosphate; CLIC, Chloride intracellular channel; Cr, creatine; CypA, cyclophilin A; DLD, Dihydrolipoyl dehydrogenase; Drp1, dynaminrelated protein 1; ECs, endothelial cells; EDD, endothelium-dependent dilation; eNOS, endothelial nitric-oxide synthase; EOMA, Mouse hemangioendothelioma endothelial cells; ERK1/2, extracellular-regulated kinase 1/2; ETC, electron transport chain; GLUT1, glucose transporter 1; GSH, glutathione; GSSG, glutathione disulfide; HIF-1α, hypoxia-inducible factor-1α; HIF-2α, hypoxia-inducible factor-2α; HK2, hexokinase 2; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; H3K27, lysine 27 on histone H3; IDH1, isocitrate dehydrogenase 1; LDHA, lactate dehydrogenase A; LDLR-/-, low-density lipoprotein receptor-deficient; MAPK, mitogen-activated protein kinase; mtDNA, mitochondrial DNA; NADH, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NFYA, nuclear transcription factor Y subunit alpha; NF-κB, nuclear factor-κB; NO, nitric oxide; NOX, Nicotinamide adenine dinucleotide phosphate oxidase; NOX2, NADPH oxidase 2; NOX4, NADPH oxidase 4; Nrf2/HO-1, nuclear factor erythroid 2-related factor 2/heme oxygenase-1; OCR, O2 consumption rate; ox-LDL, oxidized low-density lipoprotein; OXPHOS, oxidative phosphorylation; PAECs, pulmonary artery endothelial cells; PASMC, pulmonary-artery smooth muscle cells; PH, pulmonary hypertension; PCAF, p300/CBP-associated factor; PCr, creatine phosphate; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3; PINK1, PTEN induced putative kinase 1; PKM2, pyruvate kinase isoform M2; polG-/-, polymerase-γ proofreading activity; PRKAA1, protein kinase AMP-activated catalytic subunit alpha 1; PTBP1, polypyrimidine tract-binding protein 1; PTEN, phosphatase and tensin homolog deleted on chromosome ten; RAW264.7, macrophage/monocyte-like leukemia cells; ROS, reactive oxygen species; TAC, chemoembolization; TAE, transcatheter arterial embolization; TCA, tricarboxylic acid; VCAM-1, vascular cell adhesion molecule-1; VECs, vascular endothelial cells; VSMCs, vascular smooth muscle cells; 3PO, 3-[3-pyridyl]-1-[4-pyridyl] -2-propen-1-ketone; 4-HNE, 4-hydroxynonenal.

Author Contributions

QT designed the study. TS, WY, YW and DL performed the literature research. TS and WY wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Figures were created by Figdraw (https://www.figdraw.com/#/).

Funding

This study was supported by the Key Research and Development Projects of Hunan Provincial Science and Technology Department (No. 2022SK2011), the Science and Technology Innovation team of Hunan Province (No. 2021RC4064), the Key Projects of Hunan Provincial Department of Education (No. 20A379) and Graduate Innovation Project of Hunan University of Chinese Medicine (No. 2022CX15). 2023 Undergraduate Innovation and Entrepreneurship Training Program of Hunan University of Chinese Medicine (No. S202310541108).

Conflict of Interest

The authors declare no conflicts of interest for this work.

References
[1]
Stratton MS, Farina FM, Elia L. Epigenetics and vascular diseases. Journal of Molecular and Cellular Cardiology. 2019; 133: 148–163.
[2]
Zhou LJ, Lin WZ, Meng XQ, Zhu H, Liu T, Du LJ, et al. Periodontitis exacerbates atherosclerosis through Fusobacterium nucleatum-promoted hepatic glycolysis and lipogenesis. Cardiovascular Research. 2023; 119: 1706–1717.
[3]
Chao de la Barca JM, Richard A, Robert P, Eid M, Fouquet O, Tessier L, et al. Metabolomic Profiling of Angiotensin-II-Induced Abdominal Aortic Aneurysm in Ldlr-/- Mice Points to Alteration of Nitric Oxide, Lipid, and Energy Metabolisms. International Journal of Molecular Sciences. 2022; 23: 6387.
[4]
Alzaydi MM, Abdul-Salam VB, Whitwell HJ, Russomanno G, Glynos A, Capece D, et al. Intracellular Chloride Channels Regulate Endothelial Metabolic Reprogramming in Pulmonary Arterial Hypertension. American Journal of Respiratory Cell and Molecular Biology. 2023; 68: 103–115.
[5]
Xia SB, Tian ZB, Zhang W, Zhang H. NORAD Promotes the Viability, Migration, and Phenotypic Switch of Human Vascular Smooth Muscle Cells during Aortic Dissection via LIN28B-Mediated TGF-β Promotion and Subsequent Enhanced Glycolysis. BioMed Research International. 2022; 2022: 5333928.
[6]
Stabenow LK, Zibrova D, Ender C, Helbing DL, Spengler K, Marx C, et al. Oxidative Glucose Metabolism Promotes Senescence in Vascular Endothelial Cells. Cells. 2022; 11: 2213.
[7]
Cao Y, Zhang X, Wang L, Yang Q, Ma Q, Xu J, et al. PFKFB3-mediated endothelial glycolysis promotes pulmonary hypertension. Proceedings of the National Academy of Sciences of the United States of America. 2019; 116: 13394–13403.
[8]
Lu X, Zhang J, Liu H, Ma W, Yu L, Tan X, et al. Cannabidiol attenuates pulmonary arterial hypertension by improving vascular smooth muscle cells mitochondrial function. Theranostics. 2021; 11: 5267–5278.
[9]
Karshovska E, Wei Y, Subramanian P, Mohibullah R, Geißler C, Baatsch I, et al. HIF-1α (Hypoxia-Inducible Factor-1α) Promotes Macrophage Necroptosis by Regulating miR-210 and miR-383. Arteriosclerosis, Thrombosis, and Vascular Biology. 2020; 40: 583–596.
[10]
Zhu J, Thompson CB. Metabolic regulation of cell growth and proliferation. Nature Reviews. Molecular Cell Biology. 2019; 20: 436–450.
[11]
Glancy B, Balaban RS. Energy metabolism design of the striated muscle cell. Physiological Reviews. 2021; 101: 1561–1607.
[12]
Li T, Han J, Jia L, Hu X, Chen L, Wang Y. PKM2 coordinates glycolysis with mitochondrial fusion and oxidative phosphorylation. Protein & Cell. 2019; 10: 583–594.
[13]
Cho YK, Yoon YC, Im H, Son Y, Kim M, Saha A, et al. Adipocyte lysoplasmalogenase TMEM86A regulates plasmalogen homeostasis and protein kinase A-dependent energy metabolism. Nature Communications. 2022; 13: 4084.
[14]
Bielanin JP, Sun D. Significance of Microglial Energy Metabolism in Maintaining Brain Homeostasis. Translational Stroke Research. 2023; 14: 435–437.
[15]
Li Y, Chen H, Xie X, Yang B, Wang X, Zhang J, et al. PINK1-Mediated Mitophagy Promotes Oxidative Phosphorylation and Redox Homeostasis to Induce Drug-Tolerant Persister Cancer Cells. Cancer Research. 2023; 83: 398–413.
[16]
Faas M, Ipseiz N, Ackermann J, Culemann S, Grüneboom A, Schröder F, et al. IL-33-induced metabolic reprogramming controls the differentiation of alternatively activated macrophages and the resolution of inflammation. Immunity. 2021; 54: 2531–2546.e5.
[17]
Zhang B, Pan C, Feng C, Yan C, Yu Y, Chen Z, et al. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Report: Communications in Free Radical Research. 2022; 27: 45–52.
[18]
Ye L, Jiang Y, Zhang M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine & Growth Factor Reviews. 2022; 68: 81–92.
[19]
Sun T, Quan W, Peng S, Yang D, Liu J, He C, et al. Network Pharmacology-Based Strategy Combined with Molecular Docking and in vitro Validation Study to Explore the Underlying Mechanism of Huo Luo Xiao Ling Dan in Treating Atherosclerosis. Drug Design, Development and Therapy. 2022; 16: 1621–1645.
[20]
Salnikova D, Orekhova V, Grechko A, Starodubova A, Bezsonov E, Popkova T, et al. Mitochondrial Dysfunction in Vascular Wall Cells and Its Role in Atherosclerosis. International Journal of Molecular Sciences. 2021; 22: 8990.
[21]
Li J, Huynh L, Cornwell WD, Tang MS, Simborio H, Huang J, et al. Electronic Cigarettes Induce Mitochondrial DNA Damage and Trigger TLR9 (Toll-Like Receptor 9)-Mediated Atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2021; 41: 839–853.
[22]
Konkova MS, Kaliyanov AA, Sergeeva VA, Abramova MS, Kostyuk SV. Oxidized Cell-Free DNA Is a Factor of Stress Signaling in Radiation-Induced Bystander Effects in Different Types of Human Cells. International Journal of Genomics. 2019; 2019: 9467029.
[23]
Shemiakova T, Ivanova E, Grechko AV, Gerasimova EV, Sobenin IA, Orekhov AN. Mitochondrial Dysfunction and DNA Damage in the Context of Pathogenesis of Atherosclerosis. Biomedicines. 2020; 8: 166.
[24]
Shemiakova T, Ivanova E, Wu WK, Kirichenko TV, Starodubova AV, Orekhov AN. Atherosclerosis as Mitochondriopathy: Repositioning the Disease to Help Finding New Therapies. Frontiers in Cardiovascular Medicine. 2021; 8: 660473.
[25]
Yu E, Calvert PA, Mercer JR, Harrison J, Baker L, Figg NL, et al. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation. 2013; 128: 702–712.
[26]
Edgar L, Akbar N, Braithwaite AT, Krausgruber T, Gallart-Ayala H, Bailey J, et al. Hyperglycemia Induces Trained Immunity in Macrophages and Their Precursors and Promotes Atherosclerosis. Circulation. 2021; 144: 961–982.
[27]
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. 2021; 47: 65–76.
[28]
An T, Zhang X, Gao X, Zhang X, Shen T, Li H, et al. Phosphoenolpyruvate induces endothelial dysfunction and cell senescence through stimulation of metabolic reprogramming. Journal of Bioenergetics and Biomembranes. 2023; 55: 103–114.
[29]
Yang Q, Xu J, Ma Q, Liu Z, Sudhahar V, Cao Y, et al. PRKAA1/AMPKα1-driven glycolysis in endothelial cells exposed to disturbed flow protects against atherosclerosis. Nature Communications. 2018; 9: 4667.
[30]
Wu W, Wang X, Liao L, Chen J, Wang Y, Yao M, et al. The TRPM7 channel reprograms cellular glycolysis to drive tumorigenesis and angiogenesis. Cell Death & Disease. 2023; 14: 183.
[31]
Feng S, Bowden N, Fragiadaki M, Souilhol C, Hsiao S, Mahmoud M, et al. Mechanical Activation of Hypoxia-Inducible Factor 1α Drives Endothelial Dysfunction at Atheroprone Sites. Arteriosclerosis, Thrombosis, and Vascular Biology. 2017; 37: 2087–2101.
[32]
Lee LYH, Oldham WM, He H, Wang R, Mulhern R, Handy DE, et al. Interferon-γ Impairs Human Coronary Artery Endothelial Glucose Metabolism by Tryptophan Catabolism and Activates Fatty Acid Oxidation. Circulation. 2021; 144: 1612–1628.
[33]
Rohlenova K, Veys K, Miranda-Santos I, De Bock K, Carmeliet P. Endothelial Cell Metabolism in Health and Disease. Trends in Cell Biology. 2018; 28: 224–236.
[34]
Vallée A, Vallée JN, Lecarpentier Y. Metabolic reprogramming in atherosclerosis: Opposed interplay between the canonical WNT/β-catenin pathway and PPARγ. Journal of Molecular and Cellular Cardiology. 2019; 133: 36–46.
[35]
Wong BW, Marsch E, Treps L, Baes M, Carmeliet P. Endothelial cell metabolism in health and disease: impact of hypoxia. The EMBO Journal. 2017; 36: 2187–2203.
[36]
Li Z, Li Q, Wang L, Li C, Xu M, Duan Y, et al. Targeting mitochondria-inflammation circle by renal denervation reduces atheroprone endothelial phenotypes and atherosclerosis. Redox Biology. 2021; 47: 102156.
[37]
Kattoor AJ, Pothineni NVK, Palagiri D, Mehta JL. Oxidative Stress in Atherosclerosis. Current Atherosclerosis Reports. 2017; 19: 42.
[38]
Jiang J, Huang K, Xu S, Garcia JGN, Wang C, Cai H. Erratum to Targeting NOX4 alleviates sepsis-induced acute lung injury via attenuation of redox-sensitive activation of CaMKII/ERK1/2/MLCK and endothelial cell barrier dysfunction, Redox Biology 36 (2020) 101638. Redox Biology. 2021; 48: 102200.
[39]
Xuan Y, Yu C, Ni K, Congcong L, Lixin Q, Qingxian L. Protective effects of tanshinone IIA on Porphyromonas gingivalis-induced atherosclerosis via the downregulation of the NOX2/NOX4-ROS mediation of NF-κB signaling pathway. Microbes and Infection. 2023; 105177.
[40]
Han Z, Hu H, Yin M, Lin Y, Yan Y, Han P, et al. HOXA1 participates in VSMC-to-macrophage-like cell transformation via regulation of NF-κB p65 and KLF4: a potential mechanism of atherosclerosis pathogenesis. Molecular Medicine (Cambridge, Mass.). 2023; 29: 104.
[41]
Miano JM, Fisher EA, Majesky MW. Fate and State of Vascular Smooth Muscle Cells in Atherosclerosis. Circulation. 2021; 143: 2110–2116.
[42]
Shi J, Yang Y, Cheng A, Xu G, He F. Metabolism of vascular smooth muscle cells in vascular diseases. American Journal of Physiology. Heart and Circulatory Physiology. 2020; 319: H613–H631.
[43]
Dhar-Chowdhury P, Malester B, Rajacic P, Coetzee WA. The regulation of ion channels and transporters by glycolytically derived ATP. Cellular and Molecular Life Sciences: CMLS. 2007; 64: 3069–3083.
[44]
Rabinowitz JD, Enerbäck S. Lactate: the ugly duckling of energy metabolism. Nature Metabolism. 2020; 2: 566–571.
[45]
Jia Y, Mao C, Ma Z, Huang J, Li W, Ma X, et al. PHB2 Maintains the Contractile Phenotype of VSMCs by Counteracting PKM2 Splicing. Circulation Research. 2022; 131: 807–824.
[46]
Zhu Y, Zhang JL, Yan XJ, Ji Y, Wang FF. Exploring a new mechanism between lactate and VSMC calcification: PARP1/POLG/UCP2 signaling pathway and imbalance of mitochondrial homeostasis. Cell Death & Disease. 2023; 14: 598.
[47]
Adhikari N, Basi DL, Carlson M, Mariash A, Hong Z, Lehman U, et al. Increase in GLUT1 in smooth muscle alters vascular contractility and increases inflammation in response to vascular injury. Arteriosclerosis, Thrombosis, and Vascular Biology. 2011; 31: 86–94.
[48]
Wall VZ, Barnhart S, Kanter JE, Kramer F, Shimizu-Albergine M, Adhikari N, et al. Smooth muscle glucose metabolism promotes monocyte recruitment and atherosclerosis in a mouse model of metabolic syndrome. JCI Insight. 2018; 3: e96544.
[49]
Chiong M, Morales P, Torres G, Gutiérrez T, García L, Ibacache M, et al. Influence of glucose metabolism on vascular smooth muscle cell proliferation. VASA. Zeitschrift Fur Gefasskrankheiten. 2013; 42: 8–16.
[50]
Hall JL, Chatham JC, Eldar-Finkelman H, Gibbons GH. Upregulation of glucose metabolism during intimal lesion formation is coupled to the inhibition of vascular smooth muscle cell apoptosis. Role of GSK3beta. Diabetes. 2001; 50: 1171–1179.
[51]
Docherty CK, Strembitska A, Baker CP, Schmidt FF, Reay K, Mercer JR. Inducing Energetic Switching Using Klotho Improves Vascular Smooth Muscle Cell Phenotype. International Journal of Molecular Sciences. 2021; 23: 217.
[52]
Docherty CK, Bresciani J, Carswell A, Chanderseka A, Friel E, Stasi M, et al. An Inducible and Vascular Smooth Muscle Cell-Specific Pink1 Knockout Induces Mitochondrial Energetic Dysfunction during Atherogenesis. International Journal of Molecular Sciences. 2021; 22: 9993.
[53]
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and function in health and disease. Journal of Cellular Physiology. 2018; 233: 6425–6440.
[54]
Liu Y, Xu R, Gu H, Zhang E, Qu J, Cao W, et al. Metabolic reprogramming in macrophage responses. Biomarker Research. 2021; 9: 1.
[55]
Freemerman AJ, Johnson AR, Sacks GN, Milner JJ, Kirk EL, Troester MA, et al. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. The Journal of Biological Chemistry. 2014; 289: 7884–7896.
[56]
Batista-Gonzalez A, Vidal R, Criollo A, Carreño LJ. New Insights on the Role of Lipid Metabolism in the Metabolic Reprogramming of Macrophages. Frontiers in Immunology. 2020; 10: 2993.
[57]
Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The Metabolic Signature of Macrophage Responses. Frontiers in Immunology. 2019; 10: 1462.
[58]
Zhu L, Zhao Q, Yang T, Ding W, Zhao Y. Cellular metabolism and macrophage functional polarization. International Reviews of Immunology. 2015; 34: 82–100.
[59]
Hu X, Cai X, Ma R, Fu W, Zhang C, Du X. Iron-load exacerbates the severity of atherosclerosis via inducing inflammation and enhancing the glycolysis in macrophages. Journal of Cellular Physiology. 2019; 234: 18792–18800.
[60]
Dumont A, Lee M, Barouillet T, Murphy A, Yvan-Charvet L. Mitochondria orchestrate macrophage effector functions in atherosclerosis. Molecular Aspects of Medicine. 2021; 77: 100922.
[61]
Zhong S, Li L, Shen X, Li Q, Xu W, Wang X, et al. An update on lipid oxidation and inflammation in cardiovascular diseases. Free Radical Biology & Medicine. 2019; 144: 266–278.
[62]
Timblin GA, Tharp KM, Ford B, Winchester JM, Wang J, Zhu S, et al. Mitohormesis reprogrammes macrophage metabolism to enforce tolerance. Nature Metabolism. 2021; 3: 618–635.
[63]
Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circulation Research. 2017; 120: 713–735.
[64]
Ren J, Su D, Li L, Cai H, Zhang M, Zhai J, et al. Anti-inflammatory effects of Aureusidin in LPS-stimulated RAW264.7 macrophages via suppressing NF-κB and activating ROS- and MAPKs-dependent Nrf2/HO-1 signaling pathways. Toxicology and Applied Pharmacology. 2020; 387: 114846.
[65]
Kothari V, Bornfeldt KE. Liver Kinase B1 Links Macrophage Metabolism Sensing and Atherosclerosis. Circulation Research. 2017; 121: 1024–1026.
[66]
Patterson MT, Williams JW. Metabolic regulation of macrophage proliferation and function in atherosclerosis. Current Opinion in Lipidology. 2021; 32: 293–300.
[67]
Thomas C, Leleu D, Masson D. Cholesterol and HIF-1α: Dangerous Liaisons in Atherosclerosis. Frontiers in Immunology. 2022; 13: 868958.
[68]
Wang X, Du H, Li X. Artesunate attenuates atherosclerosis by inhibiting macrophage M1-like polarization and improving metabolism. International Immunopharmacology. 2022; 102: 108413.
[69]
Hasbani DJ, Hamie L. Infantile Hemangiomas. Dermatologic Clinics. 2022; 40: 383–392.
[70]
Chen J, Wu D, Dong Z, Chen A, Liu S. The expression and role of glycolysis-associated molecules in infantile hemangioma. Life Sciences. 2020; 259: 118215.
[71]
Xu ZP, Chang H, Ni YY, Li C, Chen L, Hou M, et al. Schistosoma japonicum infection causes a reprogramming of glycolipid metabolism in the liver. Parasites & Vectors. 2019; 12: 388.
[72]
Mei H, Xian H, Ke J. LncRNA-MCM3AP-AS1 Promotes the Progression of Infantile Hemangiomas by Increasing miR-138-5p/HIF-1α Axis-Regulated Glycolysis. Frontiers in Molecular Biosciences. 2021; 8: 753218.
[73]
Ghosh P, Vidal C, Dey S, Zhang L. Mitochondria Targeting as an Effective Strategy for Cancer Therapy. International Journal of Molecular Sciences. 2020; 21: 3363.
[74]
Wen YA, Xiong X, Scott T, Li AT, Wang C, Weiss HL, et al. The mitochondrial retrograde signaling regulates Wnt signaling to promote tumorigenesis in colon cancer. Cell Death and Differentiation. 2019; 26: 1955–1969.
[75]
Vasan K, Werner M, Chandel NS. Mitochondrial Metabolism as a Target for Cancer Therapy. Cell Metabolism. 2020; 32: 341–352.
[76]
Missiroli S, Perrone M, Genovese I, Pinton P, Giorgi C. Cancer metabolism and mitochondria: Finding novel mechanisms to fight tumours. EBioMedicine. 2020; 59: 102943.
[77]
Gordillo GM, Biswas A, Singh K, Sen A, Guda PR, Miller C, et al. Mitochondria as Target for Tumor Management of Hemangioendothelioma. Antioxidants & Redox Signaling. 2021; 34: 137–153.
[78]
Tchana-Sato V, Sakalihasan N, Defraigne JO. La dissection aortique [Aortic dissection]. Revue Medicale de Liege. 2018; 73: 290–295. (In French)
[79]
Le S, Zhang H, Huang X, Chen S, Wu J, Chen S, et al. PKM2 Activator TEPP-46 Attenuates Thoracic Aortic Aneurysm and Dissection by Inhibiting NLRP3 Inflammasome-Mediated IL-1β Secretion. Journal of Cardiovascular Pharmacology and Therapeutics. 2020; 25: 364–376.
[80]
Lian G, Li X, Zhang L, Zhang Y, Sun L, Zhang X, et al. Macrophage metabolic reprogramming aggravates aortic dissection through the HIF1α-ADAM17 pathway✩. EBioMedicine. 2019; 49: 291–304.
[81]
Fan LM, Douglas G, Bendall JK, McNeill E, Crabtree MJ, Hale AB, et al. Endothelial cell-specific reactive oxygen species production increases susceptibility to aortic dissection. Circulation. 2014; 129: 2661–2672.
[82]
Poch D, Mandel J. Pulmonary Hypertension. Annals of Internal Medicine. 2021; 174: ITC49–ITC64.
[83]
Hernandez-Saavedra D, Sanders L, Freeman S, Reisz JA, Lee MH, Mickael C, et al. Stable isotope metabolomics of pulmonary artery smooth muscle and endothelial cells in pulmonary hypertension and with TGF-beta treatment. Scientific Reports. 2020; 10: 413.
[84]
Xu W, Janocha AJ, Erzurum SC. Metabolism in Pulmonary Hypertension. Annual Review of Physiology. 2021; 83: 551–576.
[85]
Ma C, Wang X, He S, Zhang L, Bai J, Qu L, et al. Ubiquitinated AIF is a major mediator of hypoxia-induced mitochondrial dysfunction and pulmonary artery smooth muscle cell proliferation. Cell & Bioscience. 2022; 12: 9.
[86]
Zhao Y, Wang B, Zhang J, He D, Zhang Q, Pan C, et al. ALDH2 (Aldehyde Dehydrogenase 2) Protects Against Hypoxia-Induced Pulmonary Hypertension. Arteriosclerosis, Thrombosis, and Vascular Biology. 2019; 39: 2303–2319.
[87]
Xie X, Urabe G, Marcho L, Stratton M, Guo LW, Kent CK. ALDH1A3 Regulations of Matricellular Proteins Promote Vascular Smooth Muscle Cell Proliferation. iScience. 2019; 19: 872–882.
[88]
Li D, Shao NY, Moonen JR, Zhao Z, Shi M, Otsuki S, et al. ALDH1A3 Coordinates Metabolism With Gene Regulation in Pulmonary Arterial Hypertension. Circulation. 2021; 143: 2074–2090.
[89]
Zhang H, Wang D, Li M, Plecitá-Hlavatá L, D’Alessandro A, Tauber J, et al. Metabolic and Proliferative State of Vascular Adventitial Fibroblasts in Pulmonary Hypertension Is Regulated Through a MicroRNA-124/PTBP1 (Polypyrimidine Tract Binding Protein 1)/Pyruvate Kinase Muscle Axis. Circulation. 2017; 136: 2468–2485.
[90]
Smolders VFED, Rodríguez C, Blanco I, Szulcek R, Timens W, Piccari L, et al. Metabolic profile in endothelial cells of chronic thromboembolic pulmonary hypertension and pulmonary arterial hypertension. Scientific Reports. 2022; 12: 2283.
[91]
Abdul-Salam VB, Russomanno G, Chien-Nien C, Mahomed AS, Yates LA, Wilkins MR, et al. CLIC4/Arf6 Pathway. Circulation Research. 2019; 124: 52–65.
[92]
Karnewar S, Pulipaka S, Katta S, Panuganti D, Neeli PK, Thennati R, et al. Mitochondria-targeted esculetin mitigates atherosclerosis in the setting of aging via the modulation of SIRT1-mediated vascular cell senescence and mitochondrial function in Apoe-/- mice. Atherosclerosis. 2022; 356: 28–40.
[93]
Born E, Lipskaia L, Breau M, Houssaini A, Beaulieu D, Marcos E, et al. Eliminating Senescent Cells Can Promote Pulmonary Hypertension Development and Progression. Circulation. 2023; 147: 650–666.
[94]
Brunt VE, Gioscia-Ryan RA, Richey JJ, Zigler MC, Cuevas LM, Gonzalez A, et al. Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. The Journal of Physiology. 2019; 597: 2361–2378.
[95]
Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, et al. Voluntary wheel running restores endothelial function in conduit arteries of old mice: direct evidence for reduced oxidative stress, increased superoxide dismutase activity and down-regulation of NADPH oxidase. The Journal of Physiology. 2009; 587: 3271–3285.
[96]
Wu Y, Tang L, Huang H, Yu Q, Hu B, Wang G, et al. Phosphoglycerate dehydrogenase activates PKM2 to phosphorylate histone H3T11 and attenuate cellular senescence. Nature Communications. 2023; 14: 1323.
[97]
Gaspar MR. Arterial embolism and thrombosis. Major Problems in Clinical Surgery. 1981; 4: 158–175.
[98]
Takeo S, Taguchi T, Tanonaka K, Miyake K, Horiguchi T, Takagi N, et al. Sustained damage to energy metabolism of brain regions after microsphere embolism in rats. Stroke. 1992; 23: 62–68.
[99]
Takeo S, Miyake K, Tanonaka K, Tanonaka R, Taguchi T, Okano K, et al. Time course of changes in brain energy metabolism of the rat after microsphere-induced cerebral embolism. Japanese Journal of Pharmacology. 1991; 55: 197–209.
[100]
Hashimoto L, Ouchi K, Suzuki M, Matsuno S. Energy metabolism changes and oxidative attack after hepatic arterial embolization and chemoembolization in thioacetamide-induced cirrhotic livers. Research in Experimental Medicine. Zeitschrift Fur Die Gesamte Experimentelle Medizin Einschliesslich Experimenteller Chirurgie. 1991; 191: 91–98.
[101]
Li L, Wang M, Ma Q, Ye J, Sun G. Role of glycolysis in the development of atherosclerosis. American Journal of Physiology. Cell Physiology. 2022; 323: C617–C629.
[102]
Peng H, Wang X, Du J, Cui Q, Huang Y, Jin H. Metabolic Reprogramming of Vascular Endothelial Cells: Basic Research and Clinical Applications. Frontiers in Cell and Developmental Biology. 2021; 9: 626047.
[103]
Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O’Sullivan D, et al. Polyamines and eIF5A Hypusination Modulate Mitochondrial Respiration and Macrophage Activation. Cell Metabolism. 2019; 30: 352–363.e8.
[104]
Qin HL, Bao JH, Tang JJ, Xu DY, Shen L. Arterial remodeling: the role of mitochondrial metabolism in vascular smooth muscle cells. American Journal of Physiology. Cell Physiology. 2023; 324: C183–C192.
[105]
Donato AJ, Machin DR, Lesniewski LA. Mechanisms of Dysfunction in the Aging Vasculature and Role in Age-Related Disease. Circulation Research. 2018; 123: 825–848.
[106]
Sarrazy V, Viaud M, Westerterp M, Ivanov S, Giorgetti-Peraldi S, Guinamard R, et al. Disruption of Glut1 in Hematopoietic Stem Cells Prevents Myelopoiesis and Enhanced Glucose Flux in Atheromatous Plaques of ApoE(-/-) Mice. Circulation Research. 2016; 118: 1062–1077.
[107]
Wu WZ, Bai YP. Endothelial GLUTs and vascular biology. Biomedicine & Pharmacotherapy. 2023; 158: 114151.
[108]
Kang YJ. MicroRNA-22 Regulates the Pro-inflammatory Responses and M1 Polarization of Macrophages by Targeting GLUT1 and 4-1BBL. Journal of Immunology Research. 2023; 2023: 2457006.
[109]
van Hameren G, Campbell G, Deck M, Berthelot J, Gautier B, Quintana P, et al. In vivo real-time dynamics of ATP and ROS production in axonal mitochondria show decoupling in mouse models of peripheral neuropathies. Acta Neuropathologica Communications. 2019; 7: 86.
[110]
Nolfi-Donegan D, Braganza A, Shiva S. Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biology. 2020; 37: 101674.
[111]
Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, et al. Endothelial Dysfunction in Atherosclerotic Cardiovascular Diseases and Beyond: From Mechanism to Pharmacotherapies. Pharmacological Reviews. 2021; 73: 924–967.
[112]
Perrotta P, Van der Veken B, Van Der Veken P, Pintelon I, Roosens L, Adriaenssens E, et al. Partial Inhibition of Glycolysis Reduces Atherogenesis Independent of Intraplaque Neovascularization in Mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 2020; 40: 1168–1181.
[113]
Bai XL, Deng XL, Wu GJ, Li WJ, Jin S. Rhodiola and salidroside in the treatment of metabolic disorders. Mini Reviews in Medicinal Chemistry. 2019; 19: 1611–1626.
[114]
Savla SR, Laddha AP, Kulkarni YA. Pharmacology of apocynin: a natural acetophenone. Drug Metabolism Reviews. 2021; 53: 542–562.
[115]
Clayton ZS, Hutton DA, Brunt VE, VanDongen NS, Ziemba BP, Casso AG, et al. Apigenin restores endothelial function by ameliorating oxidative stress, reverses aortic stiffening, and mitigates vascular inflammation with aging. American Journal of Physiology. Heart and Circulatory Physiology. 2021; 321: H185–H196.
[116]
Gomez-Acevedo H, Dai Y, Strub G, Shawber C, Wu JK, Richter GT. Identification of putative biomarkers for Infantile Hemangiomas and Propranolol treatment via data integration. Scientific Reports. 2020; 10: 3261.
[117]
Park SY, Pekas EJ, Headid RJ, 3rd, Son WM, Wooden TK, Song J, et al. Acute mitochondrial antioxidant intake improves endothelial function, antioxidant enzyme activity, and exercise tolerance in patients with peripheral artery disease. American Journal of Physiology. Heart and Circulatory Physiology. 2020; 319: H456–H467.
[118]
Michelakis ED, Gurtu V, Webster L, Barnes G, Watson G, Howard L, et al. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Science Translational Medicine. 2017; 9: eaao4583.
[119]
Yang K, Ren J, Li X, Wang Z, Xue L, Cui S, et al. Prevention of aortic dissection and aneurysm via an ALDH2-mediated switch in vascular smooth muscle cell phenotype. European Heart Journal. 2020; 41: 2442–2453.
[120]
Zhang J, Zhao X, Guo Y, Liu Z, Wei S, Yuan Q, et al. Macrophage ALDH2 (Aldehyde Dehydrogenase 2) Stabilizing Rac2 Is Required for Efferocytosis Internalization and Reduction of Atherosclerosis Development. Arteriosclerosis, Thrombosis, and Vascular Biology. 2022; 42: 700–716.
[121]
Wang K, Rong L, Wei X, Zhang Q, Xiao L. The effectiveness of various cytotherapeutic strategies for the treatment of ischemic stroke: a Bayesian network meta-analysis of randomized controlled trials. Neurological Sciences: Official Journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology. 2020; 41: 1705–1717.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top