Next Article in Journal
Mass Spectrometry-Based Proteome Profiling of Extracellular Vesicles Derived from the Cerebrospinal Fluid of Adult Rhesus Monkeys Exposed to Cocaine throughout Gestation
Next Article in Special Issue
Fludrocortisone Induces Aortic Pathologies in Mice
Previous Article in Journal
Feature Reduction for Molecular Similarity Searching Based on Autoencoder Deep Learning
Previous Article in Special Issue
Imaging Techniques for Aortic Aneurysms and Dissections in Mice: Comparisons of Ex Vivo, In Situ, and Ultrasound Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm

Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(4), 509; https://doi.org/10.3390/biom12040509
Submission received: 30 January 2022 / Revised: 18 March 2022 / Accepted: 27 March 2022 / Published: 28 March 2022

Abstract

:
Abdominal aortic aneurysm (AAA), defined as a focal dilation of the abdominal aorta beyond 50% of its normal diameter, is a common and potentially life-threatening vascular disease. The molecular and cellular mechanisms underlying AAA pathogenesis remain unclear. Healthy endothelial cells (ECs) play a critical role in maintaining vascular homeostasis by regulating vascular tone and maintaining an anti-inflammatory, anti-thrombotic local environment. Increasing evidence indicates that endothelial dysfunction is an early pathologic event in AAA formation, contributing to both oxidative stress and inflammation in the degenerating arterial wall. Recent studies utilizing single-cell RNA sequencing revealed heterogeneous EC sub-populations, as determined by their transcriptional profiles, in aortic aneurysm tissue. This review summarizes recent findings, including clinical evidence of endothelial dysfunction in AAA, the impact of biomechanical stress on EC in AAA, the role of endothelial nitric oxide synthase (eNOS) uncoupling in AAA, and EC heterogeneity in AAA. These studies help to improve our understanding of AAA pathogenesis and ultimately may lead to the generation of EC-targeted therapeutics to treat or prevent this deadly disease.

1. Introduction

Abdominal aortic aneurysm (AAA) is defined as a greater than 1.5-fold increase in the diameter of the aorta beyond the normal range [1]. The dilated abdominal aorta is both structurally compromised and significantly weakened, and consequently often unable to tolerate the forces to which it is exposed from high pressure, pulsatile blood flow. When wall stress exceeds that which the aneurysmal aorta can tolerate, rupture occurs often with catastrophic consequences [2]. AAA rupture is associated with a high overall mortality rate; approximately half of people with ruptured aneurysms will not survive to present to the hospital, and 50% of those who undergo emergency surgery will not survive [3,4]. Risk factors for rupture include large aneurysm diameter and rapid rate of expansion [2]. The annual risk of rupture for patients with aneurysms <5 cm is low, but increases substantially for patients with aneurysms >5 cm (5.0–5.9 cm annual risk of rupture 1–11%, 6.0–6.9 cm annual risk of rupture 10–22%, >7.0 cm annual risk of rupture >30%) [2,5]. Aneurysm size of >5.5 cm in males, >5 cm in females, or rates of growth of >0.5 cm in six months or >1 cm in one year are all indications for repair [5]. Interestingly, AAA exhibits sexual dimorphism, with a 4:1 male to female predominance. While men are more likely to develop AAAs, women have an accelerated rate of aneurysm growth and a higher risk of rupture at smaller aneurysm diameters [6,7,8]. Besides clinical risk factors such as male sex, advanced age, and tobacco use, AAA development is also heavily influenced by genetics [9,10]. Genome-wide association studies (GWAS) have revealed 24 loci associated with increased risk of AAA formation, including genes such as LRP1, LDLR, PLTP, APOE, LPA, and PCSK9, indicating that both family history and individual phenotype influence the onset of AAA [11].
The pathophysiology of AAA formation, expansion, and rupture, including its sexual dimorphism, is not fully understood, although existing studies indicate that local inflammation, smooth muscle cell death, and extracellular matrix degradation play a substantial role [2,12]. A feature ubiquitous to nearly all AAAs is a layer of thick intraluminal thrombus (ILT) with associated destruction of the adjacent endothelial lining. While the relationship between ILT and AAA progression has historically garnered interest and been investigated, the consequences of endothelial cell (EC) destruction and endothelial phenotypic changes in the context of AAA have been inadequately investigated [13].
ECs play a major role in regulating vascular homeostasis and blood flow. ECs also modulate vascular tone, angiogenesis, wound healing, smooth muscle cell proliferation, fibrosis, and inflammation. Furthermore, ECs are essential in maintaining a non-thrombogenic blood-tissue interface with limited permeability [14]. Endothelial dysfunction has been shown to initiate various vascular diseases, including AAA [14]. ECs contribute to AAA expansion through increased oxidative stress, nitric oxide (NO) bioavailability, adhesion molecules expression, and inflammatory cell recruitment [15]. In this review, we summarize recent findings on endothelial dysfunction in AAA pathogenesis including clinical evidence of a dysfunctional endothelium in AAA patients, the relationship between biomechanical stress and ECs in AAA, endothelial nitric oxide synthase (eNOS) uncoupling in AAA, and a transcriptional profile of EC dysfunction in AAA revealed by single-cell RNA sequencing (scRNA-seq).

2. Endothelial Dysfunction and AAA Progression: Early Clinical Evidence and Common Risk Factors

Currently, AAAs are diagnosed, followed, and intervened upon largely based on abdominal imaging and size-based criteria. The risk of aneurysm rupture increases as aortic diameter increases, from a less than 5% annual risk of rupture for aneurysms 4–4.9 cm in diameter to a 30–50% annual risk of rupture for aneurysms >8 cm in diameter [5,16]. Intervention in the form of endovascular or open surgical aneurysm exclusion is typically offered when AAAs reach a diameter of 5.5 cm in men and 5.0 cm in women, or when growth exceeds 0.5 cm in 6 months or 1 cm in 12 months [5,17]. As our understanding of AAA pathophysiology improves, clinical tests beyond abdominal CT, MRI, and ultrasound and circulating biomarkers may begin to play a role in AAA diagnosis and interventional decision-making. Tests of endothelial function, which have been shown to be abnormal in patients with AAA, are one such promising area [18,19,20,21,22,23].
Flow mediated dilation (FMD) of the brachial artery is a non-invasive measure of endothelial function [22,24,25]. Briefly, FMD indirectly measures the abundance and bioavailability of NO, a soluble gas produced by the endothelium that acts as the main vasodilator within the vasculature [22]. NO is produced in the endothelium by the enzyme eNOS, which catalyzes the conversion of L-arginine into NO and L-citrulline [26]. Circulating NO is able to diffuse through the vascular wall to the medial layer, where it stimulates production of cyclic guanosine monophosphate (cGMP), setting off a cascade of intracellular events that results in smooth muscle relaxation [27,28]. In order to measure FMD in the clinical setting, arterial dilation is induced by inflating and deflating a blood pressure cuff on the extremity of interest, consequently increasing both flow and shear stress and therefore causing an endothelium-dependent release of vasodilating agents (ex: NO) [24]. Ultrasound-based comparisons of the target artery diameter at baseline and after cuff deflation are used to calculate FMD, with a lower FMD indicating endothelial dysfunction [25]. In a cross-sectional observational study of 30 all male patients with CT scan-confirmed AAAs, Medina et al. [18] found a negative correlation between FMD in the brachial artery and aneurysm diameter (R = –0.78, p < 0.001). In prospectively recruited cohorts of patients with confirmed or suspected AAA, both Lee et al. [19] and Sung et al. [23] similarly found an inverse correlation between maximum aneurysm diameter and FMD, although of a weaker magnitude than that reported by Medina et al. (Lee et al.: R = –0.28, p < 0.001; Sung et al.: R = –0.32, p = 0.005). Moreover, Lee et al. found that baseline FMD inversely correlated with aneurysm diameter progression over time and recovered after surgical (n = 22) or endovascular (n = 28) AAA repair [19]. Finally, in a study comparing similarly aged AAA patients (n = 22) and healthy adults (n = 22), Bailey et al. found lower FMD in AAA patients compared to the healthy population (1.10% lower FMD, 95% CI 0.72–0.81) [20].
While promising as a potential sensitive biomarker for AAA diagnosis and progression (Table 1), further investigation is required to better define the specificity of low FMD for AAA. Evidence exists that FMD may be impacted in the setting of peripheral arterial disease (PAD) [29], a pathology showing a moderate association with AAA [30]. Furthermore, Bailey et al. found several baseline differences between AAA patients with lower FMD and healthy peers with higher FMD, namely a higher incidence of hypertension, hyperlipidemia, and poorer peak cardiovascular fitness [20]. Future studies addressing these potential confounders would be of great interest.
While challenging to study, indirect epidemiological evidence also suggests an intimate link between smoking (the strongest modifiable risk factor for AAA formation), endothelial dysfunction, and AAA development. Tobacco use has been shown to be associated with a 5 to 7-fold increased risk of AAA formation [31,32], and smoking-induced endothelial dysfunction has been well documented over time. Celermajer et al. [33] demonstrated that adult smokers have significantly reduced FMD of the superficial femoral artery and brachial arteries, a finding also reported by Ozaki et al. in a study of young men reported nearly two decades later [34]. The proposed mechanisms by which smoking may drive endothelial dysfunction include generation of reactive oxygen species (ROS) and increased leukocyte adhesion [35,36], with both oxidative stress and vascular inflammation being key pathophysiological events in AAA formation [37,38].

3. Endothelial Dysfunction and AAA: Circulating Biomarkers

Beyond investigating FMD as a potential biomarker in AAA, Sung et al. also analyzed circulating endothelial progenitor cell (EPC) burden in AAA patients compared to healthy controls [23]. EPCs are bone marrow-derived cells that circulate in adults and may contribute to endothelial repair [39]. Sung et al. found not only that EPC numbers were reduced in patients with AAA compared to healthy controls, but also that EPC functions (ex: adhesion, proliferation, migration, tube formation) were attenuated in AAA patients [23]. Wu et al. similarly found that pre-intervention number of circulating EPCs were significantly lower in AAA patients compared to healthy controls, and that endovascular aneurysm repair increased EPC numbers as early as two weeks post-repair [40]. While overall a limited number of studies have been performed investigating circulating EPCs in patients with AAA, it is worth noting that one of the earliest studies in this field identified a different pattern with respect to EPCs in AAA patients: Dawson et al. found that AAA patients had significantly more circulating EPCs compared to healthy control patients [41]. These discrepancies may be due to changes in the surface markers used to accurately identify EPCs over time.
Finally, a limited number of studies have identified several circulating biomarkers for AAA that could be of an endothelial source. Both Ramos-Mozo et al. and Soto et al. found that circulating chemokine (C-C motif) ligand 20 (CCL20), a chemoattractant for lymphocytes and neutrophils that has been implicated in several autoimmune diseases, was elevated in plasma from AAA patients compared to healthy controls [42,43]. Gene expression analysis by Soto et al. revealed significantly higher CCL20 mRNA in human AAA samples compared to control aorta from healthy multi-organ donors. While subsequent immunohistochemical analysis of adjacent AAA tissue revealed that CCL20 localized to both the endothelial and medial layers, the definitive source of circulating CCL20 in AAA patients remains to be determined. Thrombomodulin, an endothelium-bound protein that plays a role in the activated-protein C anticoagulant pathway and has been found to associate with endothelial dysfunction [44], has also been implicated as a potential biomarker in AAA, although conflicting data exists. In a study of 58 male AAA patients and 60 male control patients, plasma thrombomodulin was significantly higher in AAA patients compared to controls [45]. In contrast, a study of 21 AAA patients and 42 healthy controls found no difference in circulating thrombomodulin between groups [46].
Until further research is performed, caution should be applied when considering the specificity of these candidate biomarkers for AAA. Studies have shown that CCL20 can be elevated in disease states commonly found in patients with AAA, such as ischemic heart disease and hyperlipidemia [47,48]. Thrombomodulin has also been implicated as a potential biomarker in ischemic heart disease [49] and PAD [44]. Circulating EPC number has also been found to have an inverse correlation with risk factors for PAD [50]. Further research into the sensitivity and specificity of AAA biomarkers of potentially endothelial origin will be merited as our understanding of endothelial dysfunction in AAA pathophysiology continues to grow.

4. Endothelial Dysfunction and Thrombosis in AAA

ILT, while variably sized and shaped, is found within the vast majority of AAAs abutting the intima [51,52,53,54,55]. The ILT observed in AAA is structurally complex and far from biologically inert. Numerous canaliculi of various sizes course from the luminal to abluminal surface, and cellular penetration is observed up to 1 cm in depth from the luminal surface. Erythrocytes, platelets, macrophages, and neutrophils can all be found within the cell-rich regions of the thrombus [53,54]. Conflicting theories have been proposed regarding the role of ILT in AAA pathogenesis. Some posit that ILT accelerates disease progression, while others argue that the thrombus lining decreases wall stress and lowers the risk of rupture. Overall, the current burden of evidence suggests that the prevailing effect of ILT is pathologic, rather than protective, in AAA progression [56].
A number of studies have shown that as thrombus volume and thickness increase, the rate of aneurysm growth and risk of rupture both increase [57,58,59]. The sequestered platelets, neutrophils, and macrophages within the body of the thrombus are thought to create a hostile local environment close to the vessel wall that is rich in inflammatory cytokines and proteolytic enzymes [54,56]. Vessel wall hypoxia created by a large luminal thrombus is thought to further contribute to wall weakening and aneurysmal degeneration [55]. ILT has been found to not only correlate with vessel diameter but also with matrix metalloproteinase (MMP) levels, elastin degradation, and smooth muscle cell apoptosis, which are all pathologic hallmarks of aneurysmal degeneration [54,60,61]. Circulating markers of hemostasis have also been found to correlate with AAA and ILT size [62]. Finally, in animal models of AAA, administration of anticoagulants (ex: enoxaparin, fondaparinux) has been shown to reduce intramural thrombus formation and decrease AAA diameter [63]. Of note, studies examining the effect of anticoagulants on AAA progression in humans are lacking. One randomized control trial examining the effect of low dose rivaroxaban plus aspirin in patients with PAD on cardiovascular death, myocardial infarction, or stroke showed reduced major adverse cardiovascular events in the rivaroxaban plus aspirin group compared to aspirin alone [64]. The incidence of AAA in this cohort of patients was not reported. Studies have found that anticoagulation with warfarin after endovascular aneurysm repair for AAA may predispose patients to ongoing type II endoleaks and aneurysm sac growth [65]. A hesitancy to offer anticoagulants to patients at risk for aortic rupture may explain the lack of studies investigating these therapies [66].
The arterial endothelium provides a surface for thrombosis formation and regulates blood fluidity and vascular homeostasis. Under conditions of laminar blood flow and high shear stress, healthy endothelium produces a variety of anticoagulant and anti-platelet substances (ex: tissue factor pathway inhibitor, thrombomodulin, NO). However, in the setting of turbulent flow, low shear stress, and endothelial injury (conditions present in the setting of AAA), the endothelial phenotype changes. Under these conditions, ECs shift the balance of their function from anti-thrombotic to pro-coagulant through the binding and activating of platelets and leukocytes, induction of tissue factor, uncoupling of eNOS, and release of von Willebrand factor (VWF), among other actions [67,68,69,70,71]. In a rat xenograft model of AAA, Franck et al. reported that endovascular infusion of ECs prevented AAA formation and stabilized formed AAAs [13]. EC-infusion treated aortas demonstrated reendothelialization, an absence of intraluminal thrombus, the formation of a thick neointima rich in α-smooth muscle actin-positive cells, and a reduction in MMP activity and macrophage infiltration. With respect to mechanism, the authors noted that transplanted rat aortic ECs exerted these effects by paracrine-driven upregulation of endothelial-stabilizing factors and recruitment of native vascular cells, as opposed to directly participating in vessel-wall restoration [13]. Overall, the direct relationship between the endothelium and ILT accumulation within AAAs remains inadequately defined and merits further investigation.

5. Biomechanical Stress on EC

Biomechanical stress plays an important role in the development of various vascular diseases, including atherosclerosis and AAA [72]. Pulsatile blood flow within a vessel exposes the vessel wall to several different biomechanical stresses (axial, longitudinal, shear stress). [73,74]. Shear stress is influenced by blood velocity, viscosity, and vessel diameter [72]. The tangential force that blood exerts along a vessel through flow and friction is referred to as the wall shear stress, a vector measurement influenced by the magnitude and direction of blood flow. [72,75,76,77]. Blood within a vessel maintains a laminar flow pattern and generates high wall shear stress until regions of curvature, branch points, or intraluminal disease are encountered. In these regions, the normally laminar flow pattern is lost and the vessel is exposed to an altered pattern of shear stress [76]. While aneurysms can form anywhere along the aorta, development commonly occurs where normal blood flow is disrupted due to vessel anatomy, such as the curved thoracic aortic arch or infrarenal abdominal aorta immediately proximal to the aortic bifurcation [77,78].
In the infrarenal abdominal aorta, the aortic bifurcation generates areas of low oscillatory flow and reduced wall shear stress, along with high shear stress gradients. Continued growth of an existing aneurysm can increase the regions of the wall experiencing low shear stress and local re-circulation of flow [78]. AAAs have been shown to occur when wall shear stress is low but a large wall shear stress gradient exists [79]. In a study conducted by Boyd et al., areas of rupture in AAA had low wall shear stress and thrombus formation, reinforcing that areas with low wall shear stress are prone to aneurysm development [80]. Given how sensitive ECs are to alterations in wall shear stress, it is important to consider how biomechanical stress influences EC function in the context of AAA.
The EC monolayer serves a crucial role in the regulation and transportation of molecules to other layers of the vessel. Aberrant conditions (disturbed flow or changes in wall shear stress) within the lumen disrupt EC function and can have pathological effects. ECs under disturbed flow conditions undergo a change in morphology, moving from a linear rectangular shape to a circular shape with weakened junctions between cells, increasing the permeability of the EC layer [76,78]. Wall shear stress can be sensed by ECs through receptors facing the vessel lumen, including ion channels, integrins, and G-coupled protein receptors [76,78]. Vascular endothelial growth factor (VEGF) and cell-adhesion molecules VCAM-1 and ICAM-1 have been found to be upregulated in the setting of disturbed flow and low wall shear stress, causing gaps in the EC monolayer and increasing binding of inflammatory cells to both endothelial and vascular smooth muscle cells through gaps in EC junctions [78,81]. Importantly, heightened vascular permeability and inflammation are hallmarks of AAA pathogenesis [82,83]. As a point of interest, several studies have noted sexual dimorphism in human umbilical vein endothelial cells (HUVECs) with respect to baseline expression of genes, including those regulated by shear stress, in addition to differences in the magnitude and direction of transcriptional response to shear stress [84,85,86]. Several studies have also noted sex-based differences in FMD, a measurement that captures vasodilation in response to changes in shear stress [87,88]. Whether or not these different responses to biomechanical stress contribute to differences in aneurysm incidence and rupture risk between men and women remains to be investigated.

6. eNOS Uncoupling in AAA

eNOS is a key enzyme involved in the production of NO. The normal function of eNOS requires dimerization of the enzyme and its co-factor tetrahydrobiopterin (BH4). In the absence of BH4, eNOS becomes "uncoupled" and generates superoxide (O2–) as opposed to NO, which consequently causes endothelial dysfunction. Bioavailable BH4 levels are determined by a number of factors, including the activity of GTP-cyclohydrolase I (GTPCHI, the rate limiting enzyme in BH4 synthesis), loss of BH4 secondary to oxidation to BH2, and regeneration of BH4 from BH2 by dihydrofolate reductase (DHFR) [89]. Across animal models of AAA, evidence is growing that pathologic eNOS uncoupling contributes meaningfully to aneurysm growth.
The Angiotensin II (Ang II) infusion model is one of the most widely used mouse models of AAA [90]. In this model, Ang II is subcutaneously delivered to apolipoprotein-E null (Apoe−/−) mice via osmotic pumps for an extended time, typically 28 days. Certain features of human AAA, including the presence of hyperlipidemia and hypertension, as well as higher incidence in males, are well captured by this model. Other aspects, however, are less well reflected. In the Ang II model, AAAs most commonly develop at the suprarenal rather than infrarenal abdominal aorta, and aortic dissection often precedes dilatation [91]. Using this model, Gao et al. found eNOS and DHFR expression was decreased by Ang II treatment in the intimal layer [92]. The eNOS uncoupling activity, assessed by L-NAME-sensitive superoxide production, was minimal at baseline but greatly exaggerated with Ang II infusion [93]. In cultured bovine aortic ECs, silencing DHFR via RNA interference reduced endothelial BH4 and NO bioavailability [94]. DHFR-deficient mice developed AAAs when infused with Ang II. DHFR-deficient aneurysm-prone mice were also found to have high eNOS uncoupling activity as measured by low BH4 and NO levels, adverse vascular remodeling, and heightened inflammation [95]. This phenotype was rescued by scavenging of mitochondrial ROS with Mito-Tempo [95]. In eNOS pre-uncoupled hyperphenylalaninemia (hph)-1 mice (deficient in GTPCHI), Gao et al. demonstrated that 14 days of Ang II infusion (0.7 mg/kg per day) resulted in 79% AAA incidence and 14% rupture, while none of the wild-type mice infused with Ang II died or developed AAA [96]. BH4 and DHFR were decreased in hph-1 mice, and further reduced by Ang II infusion. Tail vein administration of DHFR expression vector and lipid-based reagent attenuated eNOS uncoupling and prevented AAA formation [96]. Chuaiphichai et al. generated endothelial-specific GTPCH1-deficient mice (Gch1fl/fl Tie2cre). When challenged with Ang II (0.4 mg/kg per day), these mice showed a significant increase in AAA incidence as well as decreased circulating BH4 levels and increased vascular oxidative stress [97]. Administration of folic acid, which was reported to increase DHFR function and recouple eNOS, attenuated AAA development in Ang II-infused Apoe-deficient mice [93]. In contrast, pre-treating C57BL/6 mice with Nω-nitro-L-arginine methyl ester (a NO synthase inhibitor) followed by Ang II and β-aminopropionitrile (BAPN) infusion augmented aortic dissection and aneurysm rupture compared to Ang II and BAPN infusion alone [98]. These studies demonstrated eNOS uncoupling is likely to be involved in AAA progression.

7. EC Heterogeneity Revealed by Single-Cell RNA Sequencing

ScRNA-seq has emerged as a powerful and unbiased method in decoding cellular and molecular information in complex tissues by generating transcriptomic profiles of individual cells [99,100]. It has been employed by multiple groups to investigate molecular signatures of human and experimental aortic aneurysm tissue at single-cell resolution (Table 2) [101,102,103,104,105]. As a monolayer covering the aorta, ECs were shown to be a smaller cell population compared to other vascular cell types such as smooth muscle cells and fibroblasts. Of note, the relatively low cell number makes EC analysis challenging, given cell populations with higher numbers will generate the most reliable results with numerous observations to analyze.
Yang et al. conducted scRNA-seq on aortic tissue harvested from mice exposed to the CaCl2 AAA model [104]. Briefly, the infrarenal abdominal aortas of C57BL/6J mice were peri-vascularly treated with 0.5 M CaCl2 (AAA group) or NaCl (control group). Aortas were collected four days after AAA induction to capture acute transcriptional responses within the aortic wall. This dataset contained 3896 cells in total, including 2537 cells from the control group and 1359 cells from the AAA group. There were 77 ECs in the control group (3%) and 60 ECs (4.1%) in AAA. Unbiased clustering further identified two EC sub-populations based on the transcriptional features EC1 and EC2. EC1 was enriched in lipid metabolism genes such as Gpihbp1, Fabp4, Cd36, while EC2 highly expressed matrix remodeling genes such as Mgp, Bgn, Sulf1. Among total ECs, there were 61% EC1 and 39% EC2 in the control group, and 78% EC1 and 22% EC2 in the AAA group. Zhao et al. analyzed aneurysmal tissue from the peri-adventitial Elastase model [105]. In this model, infrarenal abdominal aortas from C57BL/6J mice were treated with 30 μL elastase or heat-inactivated elastase (control). Aortas were collected 7 or 14 days after elastase exposure or 14 days after heat-inactivated elastase exposure (control group). Consistent with the CaCl2 dataset, two EC sub-populations were also identified in this dataset, with similarly enriched genes. Additional analysis of scRNA-seq data from Ang II infusion-treated Apoe-deficient mice and human AAA samples confirmed the presence of these two EC functional states, suggesting the universal existence of these two EC sub-populations in both human AAA and mouse models of AAA. As spatial information is lost in scRNA-seq analysis, further spatial orientation-preserving validation in mouse and human AAA samples would be necessary and informative, along with a deeper analysis of how these two EC sub-populations contribute to AAA progression.

8. Conclusions

Aneurysmal degeneration of the abdominal aorta is a relatively common phenomenon that can be life threatening when an aneurysm becomes sufficiently large or grows at a rapid rate. Unfortunately, no current therapies exist outside of open surgery or endovascular intervention to halt the progression of aortic degeneration and reduce the risk of rupture. Better understanding of the cellular and molecular pathologic processes that drive AAA formation is the first step in being able to develop non-invasive therapies to treat, or even prevent, aneurysm formation. While the roles of local inflammation, matrix degradation, and smooth muscle cell death have been investigated in the context of AAA, limited attention has been given to the role that endothelial dysfunction may play in AAA development. This review provides an overview of studies showing evidence of endothelial dysfunction as a pathologic contributor to AAA formation across both animal models and in humans. Collectively, these studies show that the mechanisms by which dysfunctional ECs drive AAA formation are diverse, ranging from decreased NO production/eNOS uncoupling to a phenotypic switch that results in a pro-thrombotic, pro-inflammatory state with differing distributions of EC sub-populations in the setting of pathologic biomechanical stresses in aneurysm prone regions (Figure 1).
This review is not without limitations. The role of EC dysfunction as a contributor to AAA formation has been relatively under investigated, and as such a limited amount of data is available. As further studies focusing on EC dysfunction in the context of AAA become available, a systematic review, as opposed to narrative review, would benefit the field. Moreover, given that endothelial dysfunction is observed in cardiovascular disease beyond AAA, continuing to pursue experimental studies that directly manipulate endothelial genes/phenotypes in the context of AAA models is necessary. This will not only provide further insight into the mechanisms by which ECs contribute to AAA but will also bolster confidence that EC dysfunction is an active contributor to aneurysm pathophysiology. As our understanding of EC contributions to aneurysm formation evolves, our ability to explore endothelial-targeted or protective therapies in the context of aneurysm treatment will also be enhanced.

Author Contributions

Writing—original draft preparation, E.D., A.S., H.Y., M.H., C.S. and T.Z.; writing—review and editing, E.D. and T.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health (F32HL158171-01 to E. DeRoo) and the American Heart Association (20CDA35350009 to T. Zhou).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript, or in the decision to publish the results. Figure was generated with Biorender.

Abbreviations

AAAabdominal aortic aneurysm
GWASgenome-wide association studies
ILTintraluminal thrombus
ECendothelial cell
NOnitric oxide
PADperipheral arterial disease
eNOSendothelial nitric oxide synthase
scRNA-seqsingle-cell RNA sequencing
FMDflow mediated dilation
EPCendothelial progenitor cell
CCL20chemokine (C-C motif) ligand 20
BH4tetrahydrobiopterin
GTPCHIGTP-cyclohydrolase I
DHFRdihydrofolate reductase
Ang IIangiotensin II
BAPNβ-aminopropionitrile
cGMPcyclic guanosine monophosphate
MMPmatrix metalloproteinase
VWFvon Willebrand factor
HUVEChuman umbilical vein endothelial cell

References

  1. Evans, G.H.; Stansby, G.; Hamilton, G. Suggested standards for reporting on arterial aneurysms. J. Vasc. Surg. 1992, 15, 456. [Google Scholar] [CrossRef] [Green Version]
  2. Kuivaniemi, H.; Ryer, E.J.; Elmore, J.R.; Tromp, G. Understanding the pathogenesis of abdominal aortic aneurysms. Expert Rev. Cardiovasc. Ther. 2015, 13, 975–987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Nordon, I.M.; Hinchliffe, R.J.; Loftus, I.M.; Thompson, M.M. Pathophysiology and epidemiology of abdominal aortic aneurysms. Nat. Rev. Cardiol. 2011, 8, 92–102. [Google Scholar] [CrossRef] [PubMed]
  4. Harris, L.M.; Faggioli, G.L.; Fiedler, R.; Curl, G.R.; Ricotta, J.J. Ruptured abdominal aortic aneurysms: Factors affecting mortality rates. J. Vasc. Surg. 1991, 14, 812–820, discussion 819–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Chaikof, E.L.; Dalman, R.L.; Eskandari, M.K.; Jackson, B.M.; Lee, W.A.; Mansour, M.A.; Mastracci, T.M.; Mell, M.; Murad, M.H.; Nguyen, L.L.; et al. The Society for Vascular Surgery practice guidelines on the care of patients with an abdominal aortic aneurysm. J. Vasc. Surg. 2018, 67, 2–77.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lo, R.C.; Bensley, R.P.; Hamdan, A.D.; Wyers, M.; Adams, J.E.; Schermerhorn, M.L.; Vascular Study Group of New, E. Gender differences in abdominal aortic aneurysm presentation, repair, and mortality in the Vascular Study Group of New England. J. Vasc. Surg. 2013, 57, 1261–1268.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. McPhee, J.T.; Hill, J.S.; Eslami, M.H. The impact of gender on presentation, therapy, and mortality of abdominal aortic aneurysm in the United States, 2001–2004. J. Vasc. Surg. 2007, 45, 891–899. [Google Scholar] [CrossRef] [Green Version]
  8. Mofidi, R.; Goldie, V.J.; Kelman, J.; Dawson, A.R.; Murie, J.A.; Chalmers, R.T. Influence of sex on expansion rate of abdominal aortic aneurysms. Br. J. Surg. 2007, 94, 310–314. [Google Scholar] [CrossRef]
  9. Sidloff, D.; Stather, P.; Dattani, N.; Bown, M.; Thompson, J.; Sayers, R.; Choke, E. Aneurysm global epidemiology study: Public health measures can further reduce abdominal aortic aneurysm mortality. Circulation 2014, 129, 747–753. [Google Scholar] [CrossRef] [Green Version]
  10. Pinard, A.; Jones, G.T.; Milewicz, D.M. Genetics of Thoracic and Abdominal Aortic Diseases. Circ. Res. 2019, 124, 588–606. [Google Scholar] [CrossRef]
  11. Klarin, D.; Verma, S.S.; Judy, R.; Dikilitas, O.; Wolford, B.N.; Paranjpe, I.; Levin, M.G.; Pan, C.; Tcheandjieu, C.; Spin, J.M.; et al. Genetic Architecture of Abdominal Aortic Aneurysm in the Million Veteran Program. Circulation 2020, 142, 1633–1646. [Google Scholar] [CrossRef]
  12. Lu, H.; Rateri, D.L.; Bruemmer, D.; Cassis, L.A.; Daugherty, A. Novel mechanisms of abdominal aortic aneurysms. Curr. Atheroscler. Rep. 2012, 14, 402–412. [Google Scholar] [CrossRef] [Green Version]
  13. Franck, G.; Dai, J.; Fifre, A.; Ngo, S.; Justine, C.; Michineau, S.; Allaire, E.; Gervais, M. Reestablishment of the endothelial lining by endothelial cell therapy stabilizes experimental abdominal aortic aneurysms. Circulation 2013, 127, 1877–1887. [Google Scholar] [CrossRef] [Green Version]
  14. Widmer, R.J.; Lerman, A. Endothelial dysfunction and cardiovascular disease. Glob. Cardiol. Sci. Pract. 2014, 2014, 291–308. [Google Scholar] [CrossRef]
  15. Sun, J.; Deng, H.; Zhou, Z.; Xiong, X.; Gao, L. Endothelium as a Potential Target for Treatment of Abdominal Aortic Aneurysm. Oxid. Med. Cell. Longev. 2018, 2018, 6306542. [Google Scholar] [CrossRef] [Green Version]
  16. Aggarwal, S.; Qamar, A.; Sharma, V.; Sharma, A. Abdominal aortic aneurysm: A comprehensive review. Exp. Clin. Cardiol. 2011, 16, 11–15. [Google Scholar]
  17. Mortality results for randomised controlled trial of early elective surgery or ultrasonographic surveillance for small abdominal aortic aneurysms. The UK Small Aneurysm Trial Participants. Lancet 1998, 352, 1649–1655. [CrossRef]
  18. Medina, F.; de Haro, J.; Florez, A.; Acin, F. Relationship between endothelial dependent vasodilation and size of abdominal aortic aneurysms. Ann. Vasc. Surg. 2010, 24, 752–757. [Google Scholar] [CrossRef]
  19. Lee, R.; Bellamkonda, K.; Jones, A.; Killough, N.; Woodgate, F.; Williams, M.; Cassimjee, I.; Handa, A.; Oxford Abdominal Aortic Aneurysm, S. Flow Mediated Dilatation and Progression of Abdominal Aortic Aneurysms. Eur. J. Vasc. Endovasc. Surg. 2017, 53, 820–829. [Google Scholar] [CrossRef]
  20. Bailey, T.G.; Perissiou, M.; Windsor, M.T.; Schulze, K.; Nam, M.; Magee, R.; Leicht, A.S.; Green, D.J.; Greaves, K.; Golledge, J.; et al. Effects of acute exercise on endothelial function in patients with abdominal aortic aneurysm. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H19–H30. [Google Scholar] [CrossRef] [Green Version]
  21. Konno, N.; Harada, T.; Akamatsu, D.; Goto, H.; Miki, T.; Kamei, T.; Kohzuki, M. The area under curve for time-course analysis parameters is associated with abdominal aortic aneurysms and the severity of peripheral artery disease in men. Int. J. Cardiol. Hypertens. 2021, 8, 100080. [Google Scholar] [CrossRef]
  22. Bellamkonda, K.; Williams, M.; Handa, A.; Lee, R. Flow Mediated Dilatation as a Biomarker in Vascular Surgery Research. J. Atheroscler. Thromb. 2017, 24, 779–787. [Google Scholar] [CrossRef] [Green Version]
  23. Flam, B.R.; Eichler, D.C.; Solomonson, L.P. Endothelial nitric oxide production is tightly coupled to the citrulline-NO cycle. Nitric Oxide 2007, 17, 115–121. [Google Scholar] [CrossRef]
  24. Furchgott, R.F. The 1996 Albert Lasker Medical Research Awards. The discovery of endothelium-derived relaxing factor and its importance in the identification of nitric oxide. JAMA 1996, 276, 1186–1188. [Google Scholar] [CrossRef]
  25. Murad, F. Shattuck Lecture. Nitric oxide and cyclic GMP in cell signaling and drug development. N. Engl. J. Med. 2006, 355, 2003–2011. [Google Scholar] [CrossRef]
  26. Harris, R.A.; Nishiyama, S.K.; Wray, D.W.; Richardson, R.S. Ultrasound assessment of flow-mediated dilation. Hypertension 2010, 55, 1075–1085. [Google Scholar] [CrossRef]
  27. Raitakari, O.T.; Celermajer, D.S. Flow-mediated dilatation. Br. J. Clin. Pharmacol. 2000, 50, 397–404. [Google Scholar] [CrossRef] [Green Version]
  28. Sung, S.H.; Wu, T.C.; Chen, J.S.; Chen, Y.H.; Huang, P.H.; Lin, S.J.; Shih, C.C.; Chen, J.W. Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm. Int. J. Cardiol. 2013, 168, 1070–1077. [Google Scholar] [CrossRef]
  29. Maldonado, F.J.; Miralles Jde, H.; Aguilar, E.M.; Gonzalez, A.F.; Garcia, J.R.; Garcia, F.A. Relationship between noninvasively measured endothelial function and peripheral arterial disease. Angiology 2009, 60, 725–731. [Google Scholar] [CrossRef]
  30. Cornuz, J.; Sidoti Pinto, C.; Tevaearai, H.; Egger, M. Risk factors for asymptomatic abdominal aortic aneurysm: Systematic review and meta-analysis of population-based screening studies. Eur. J. Public Health 2004, 14, 343–349. [Google Scholar] [CrossRef] [Green Version]
  31. Aune, D.; Schlesinger, S.; Norat, T.; Riboli, E. Tobacco smoking and the risk of abdominal aortic aneurysm: A systematic review and meta-analysis of prospective studies. Sci. Rep. 2018, 8, 14786. [Google Scholar] [CrossRef] [PubMed]
  32. Wilmink, T.B.; Quick, C.R.; Day, N.E. The association between cigarette smoking and abdominal aortic aneurysms. J. Vasc. Surg. 1999, 30, 1099–1105. [Google Scholar] [CrossRef] [Green Version]
  33. Celermajer, D.S.; Sorensen, K.E.; Gooch, V.M.; Spiegelhalter, D.J.; Miller, O.I.; Sullivan, I.D.; Lloyd, J.K.; Deanfield, J.E. Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis. Lancet 1992, 340, 1111–1115. [Google Scholar] [CrossRef]
  34. Ozaki, K.; Hori, T.; Ishibashi, T.; Nishio, M.; Aizawa, Y. Effects of chronic cigarette smoking on endothelial function in young men. J. Cardiol. 2010, 56, 307–313. [Google Scholar] [CrossRef] [Green Version]
  35. Barua, R.S.; Ambrose, J.A.; Srivastava, S.; DeVoe, M.C.; Eales-Reynolds, L.J. Reactive oxygen species are involved in smoking-induced dysfunction of nitric oxide biosynthesis and upregulation of endothelial nitric oxide synthase: An in vitro demonstration in human coronary artery endothelial cells. Circulation 2003, 107, 2342–2347. [Google Scholar] [CrossRef] [Green Version]
  36. Cavusoglu, Y.; Timuralp, B.; Us, T.; Akgun, Y.; Kudaiberdieva, G.; Gorenek, B.; Unalir, A.; Goktekin, O.; Ata, N. Cigarette smoking increases plasma concentrations of vascular cell adhesion molecule-1 in patients with coronary artery disease. Angiology 2004, 55, 397–402. [Google Scholar] [CrossRef]
  37. Guzik, B.; Sagan, A.; Ludew, D.; Mrowiecki, W.; Chwala, M.; Bujak-Gizycka, B.; Filip, G.; Grudzien, G.; Kapelak, B.; Zmudka, K.; et al. Mechanisms of oxidative stress in human aortic aneurysms--association with clinical risk factors for atherosclerosis and disease severity. Int. J. Cardiol. 2013, 168, 2389–2396. [Google Scholar] [CrossRef] [Green Version]
  38. Miller, F.J., Jr.; Sharp, W.J.; Fang, X.; Oberley, L.W.; Oberley, T.D.; Weintraub, N.L. Oxidative stress in human abdominal aortic aneurysms: A potential mediator of aneurysmal remodeling. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 560–565. [Google Scholar] [CrossRef] [Green Version]
  39. Zhang, M.; Malik, A.B.; Rehman, J. Endothelial progenitor cells and vascular repair. Curr. Opin. Hematol. 2014, 21, 224–228. [Google Scholar] [CrossRef] [Green Version]
  40. Wu, W.; Zhang, J.; Shao, L.; Huang, H.; Meng, Q.; Shen, Z.; Teng, X. Evaluation of Circulating Endothelial Progenitor Cells in Abdominal Aortic Aneurysms after Endovascular Aneurysm Repair. Int. J. Stem Cells 2021, 1–8. [Google Scholar] [CrossRef]
  41. Dawson, J.; Tooze, J.; Cockerill, G.; Choke, E.; Loftus, I.; Thompson, M.M. Endothelial progenitor cells and abdominal aortic aneurysms. Ann. N. Y. Acad. Sci. 2006, 1085, 327–330. [Google Scholar] [CrossRef]
  42. Ramos-Mozo, P.; Rodriguez, C.; Pastor-Vargas, C.; Blanco-Colio, L.M.; Martinez-Gonzalez, J.; Meilhac, O.; Michel, J.B.; Vega de Ceniga, M.; Egido, J.; Martin-Ventura, J.L. Plasma profiling by a protein array approach identifies IGFBP-1 as a novel biomarker of abdominal aortic aneurysm. Atherosclerosis 2012, 221, 544–550. [Google Scholar] [CrossRef]
  43. Soto, B.; Gallastegi-Mozos, T.; Rodriguez, C.; Martinez-Gonzalez, J.; Escudero, J.R.; Vila, L.; Camacho, M. Circulating CCL20 as a New Biomarker of Abdominal Aortic Aneurysm. Sci. Rep. 2017, 7, 17331. [Google Scholar] [CrossRef] [Green Version]
  44. Constans, J.; Conri, C. Circulating markers of endothelial function in cardiovascular disease. Clin. Chim. Acta 2006, 368, 33–47. [Google Scholar] [CrossRef]
  45. Brunelli, T.; Prisco, D.; Fedi, S.; Rogolino, A.; Farsi, A.; Marcucci, R.; Giusti, B.; Pratesi, C.; Pulli, R.; Gensini, G.F.; et al. High prevalence of mild hyperhomocysteinemia in patients with abdominal aortic aneurysm. J. Vasc. Surg. 2000, 32, 531–536. [Google Scholar] [CrossRef] [Green Version]
  46. Blann, A.D.; Devine, C.; Amiral, J.; McCollum, C.N. Soluble adhesion molecules, endothelial markers and atherosclerosis risk factors in abdominal aortic aneurysm: A comparison with claudicants and healthy controls. Blood Coagul. Fibrinolysis 1998, 9, 479–484. [Google Scholar] [CrossRef]
  47. Safa, A.; Rashidinejad, H.R.; Khalili, M.; Dabiri, S.; Nemati, M.; Mohammadi, M.M.; Jafarzadeh, A. Higher circulating levels of chemokines CXCL10, CCL20 and CCL22 in patients with ischemic heart disease. Cytokine 2016, 83, 147–157. [Google Scholar] [CrossRef]
  48. Calvayrac, O.; Rodriguez-Calvo, R.; Alonso, J.; Orbe, J.; Martin-Ventura, J.L.; Guadall, A.; Gentile, M.; Juan-Babot, O.; Egido, J.; Beloqui, O.; et al. CCL20 is increased in hypercholesterolemic subjects and is upregulated by LDL in vascular smooth muscle cells: Role of NF-kappaB. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2733–2741. [Google Scholar] [CrossRef] [Green Version]
  49. Blann, A.D.; Amiral, J.; McCollum, C.N. Prognostic value of increased soluble thrombomodulin and increased soluble E-selectin in ischaemic heart disease. Eur. J. Haematol. 1997, 59, 115–120. [Google Scholar] [CrossRef]
  50. Vasa, M.; Fichtlscherer, S.; Aicher, A.; Adler, K.; Urbich, C.; Martin, H.; Zeiher, A.M.; Dimmeler, S. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ. Res. 2001, 89, E1–E7. [Google Scholar] [CrossRef] [Green Version]
  51. Harter, L.P.; Gross, B.H.; Callen, P.W.; Barth, R.A. Ultrasonic evaluation of abdominal aortic thrombus. J. Ultrasound Med. 1982, 1, 315–318. [Google Scholar] [CrossRef]
  52. Speelman, L.; Schurink, G.W.; Bosboom, E.M.; Buth, J.; Breeuwer, M.; van de Vosse, F.N.; Jacobs, M.H. The mechanical role of thrombus on the growth rate of an abdominal aortic aneurysm. J. Vasc. Surg. 2010, 51, 19–26. [Google Scholar] [CrossRef] [Green Version]
  53. Adolph, R.; Vorp, D.A.; Steed, D.L.; Webster, M.W.; Kameneva, M.V.; Watkins, S.C. Cellular content and permeability of intraluminal thrombus in abdominal aortic aneurysm. J. Vasc. Surg. 1997, 25, 916–926. [Google Scholar] [CrossRef] [Green Version]
  54. Piechota-Polanczyk, A.; Jozkowicz, A.; Nowak, W.; Eilenberg, W.; Neumayer, C.; Malinski, T.; Huk, I.; Brostjan, C. The Abdominal Aortic Aneurysm and Intraluminal Thrombus: Current Concepts of Development and Treatment. Front. Cardiovasc. Med. 2015, 2, 19. [Google Scholar] [CrossRef] [Green Version]
  55. Vorp, D.A.; Lee, P.C.; Wang, D.H.; Makaroun, M.S.; Nemoto, E.M.; Ogawa, S.; Webster, M.W. Association of intraluminal thrombus in abdominal aortic aneurysm with local hypoxia and wall weakening. J. Vasc. Surg. 2001, 34, 291–299. [Google Scholar] [CrossRef] [Green Version]
  56. Boyd, A.J. Intraluminal thrombus: Innocent bystander or factor in abdominal aortic aneurysm pathogenesis? JVS Vasc. Sci. 2021, 2, 159–169. [Google Scholar] [CrossRef]
  57. Wolf, Y.G.; Thomas, W.S.; Brennan, F.J.; Goff, W.G.; Sise, M.J.; Bernstein, E.F. Computed tomography scanning findings associated with rapid expansion of abdominal aortic aneurysms. J. Vasc. Surg. 1994, 20, 529–535, discussion 535–538. [Google Scholar] [CrossRef] [Green Version]
  58. Satta, J.; Laara, E.; Juvonen, T. Intraluminal thrombus predicts rupture of an abdominal aortic aneurysm. J. Vasc. Surg. 1996, 23, 737–739. [Google Scholar] [CrossRef]
  59. Hans, S.S.; Jareunpoon, O.; Balasubramaniam, M.; Zelenock, G.B. Size and location of thrombus in intact and ruptured abdominal aortic aneurysms. J. Vasc. Surg. 2005, 41, 584–588. [Google Scholar] [CrossRef] [Green Version]
  60. Kazi, M.; Thyberg, J.; Religa, P.; Roy, J.; Eriksson, P.; Hedin, U.; Swedenborg, J. Influence of intraluminal thrombus on structural and cellular composition of abdominal aortic aneurysm wall. J. Vasc. Surg. 2003, 38, 1283–1292. [Google Scholar] [CrossRef] [Green Version]
  61. Khan, J.A.; Abdul Rahman, M.N.; Mazari, F.A.; Shahin, Y.; Smith, G.; Madden, L.; Fagan, M.J.; Greenman, J.; McCollum, P.T.; Chetter, I.C. Intraluminal thrombus has a selective influence on matrix metalloproteinases and their inhibitors (tissue inhibitors of matrix metalloproteinases) in the wall of abdominal aortic aneurysms. Ann. Vasc. Surg. 2012, 26, 322–329. [Google Scholar] [CrossRef] [PubMed]
  62. Wallinder, J.; Bergqvist, D.; Henriksson, A.E. Haemostatic markers in patients with abdominal aortic aneurysm and the impact of aneurysm size. Thromb. Res. 2009, 124, 423–426. [Google Scholar] [CrossRef] [PubMed]
  63. Moran, C.S.; Seto, S.W.; Krishna, S.M.; Sharma, S.; Jose, R.J.; Biros, E.; Wang, Y.; Morton, S.K.; Golledge, J. Parenteral administration of factor Xa/IIa inhibitors limits experimental aortic aneurysm and atherosclerosis. Sci. Rep. 2017, 7, 43079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Anand, S.S.; Bosch, J.; Eikelboom, J.W.; Connolly, S.J.; Diaz, R.; Widimsky, P.; Aboyans, V.; Alings, M.; Kakkar, A.K.; Keltai, K.; et al. Rivaroxaban with or without aspirin in patients with stable peripheral or carotid artery disease: An international, randomised, double-blind, placebo-controlled trial. Lancet 2018, 391, 219–229. [Google Scholar] [CrossRef] [Green Version]
  65. Bobadilla, J.L.; Hoch, J.R.; Leverson, G.E.; Tefera, G. The effect of warfarin therapy on endoleak development after endovascular aneurysm repair (EVAR) of the abdominal aorta. J. Vasc. Surg. 2010, 52, 267–271. [Google Scholar] [CrossRef] [Green Version]
  66. Cameron, S.J.; Russell, H.M.; Owens, A.P., 3rd. Antithrombotic therapy in abdominal aortic aneurysm: Beneficial or detrimental? Blood 2018, 132, 2619–2628. [Google Scholar] [CrossRef]
  67. Yau, J.W.; Teoh, H.; Verma, S. Endothelial cell control of thrombosis. BMC Cardiovasc. Disord. 2015, 15, 130. [Google Scholar] [CrossRef] [Green Version]
  68. Van Hinsbergh, V.W. Endothelium—Role in regulation of coagulation and inflammation. Semin. Immunopathol. 2012, 34, 93–106. [Google Scholar] [CrossRef] [Green Version]
  69. Heiss, C.; Rodriguez-Mateos, A.; Kelm, M. Central role of eNOS in the maintenance of endothelial homeostasis. Antioxid. Redox Signal. 2015, 22, 1230–1242. [Google Scholar] [CrossRef] [Green Version]
  70. Forstermann, U.; Munzel, T. Endothelial nitric oxide synthase in vascular disease: From marvel to menace. Circulation 2006, 113, 1708–1714. [Google Scholar] [CrossRef] [Green Version]
  71. Freedman, J.E.; Loscalzo, J. Nitric oxide and its relationship to thrombotic disorders. J. Thromb. Haemost. 2003, 1, 1183–1188. [Google Scholar] [CrossRef]
  72. Hoefer, I.E.; den Adel, B.; Daemen, M.J. Biomechanical factors as triggers of vascular growth. Cardiovasc. Res. 2013, 99, 276–283. [Google Scholar] [CrossRef] [Green Version]
  73. Kwak, B.R.; Back, M.; Bochaton-Piallat, M.L.; Caligiuri, G.; Daemen, M.J.; Davies, P.F.; Hoefer, I.E.; Holvoet, P.; Jo, H.; Krams, R.; et al. Biomechanical factors in atherosclerosis: Mechanisms and clinical implications. Eur. Heart J. 2014, 35, 3013–3020. [Google Scholar] [CrossRef]
  74. Thondapu, V.; Bourantas, C.V.; Foin, N.; Jang, I.K.; Serruys, P.W.; Barlis, P. Biomechanical stress in coronary atherosclerosis: Emerging insights from computational modelling. Eur. Heart J. 2017, 38, 81–92. [Google Scholar] [CrossRef] [Green Version]
  75. Arzani, A.; Shadden, S.C. Characterizations and Correlations of Wall Shear Stress in Aneurysmal Flow. J. Biomech. Eng. 2016, 138, 014503. [Google Scholar] [CrossRef] [Green Version]
  76. Dhawan, S.S.; Avati Nanjundappa, R.P.; Branch, J.R.; Taylor, W.R.; Quyyumi, A.A.; Jo, H.; McDaniel, M.C.; Suo, J.; Giddens, D.; Samady, H. Shear stress and plaque development. Expert Rev. Cardiovasc. Ther. 2010, 8, 545–556. [Google Scholar] [CrossRef] [Green Version]
  77. Platt, M.O.; Shockey, W.A. Endothelial cells and cathepsins: Biochemical and biomechanical regulation. Biochimie 2016, 122, 314–323. [Google Scholar] [CrossRef] [Green Version]
  78. Sunderland, K.; Jiang, J.; Zhao, F. Disturbed flow’s impact on cellular changes indicative of vascular aneurysm initiation, expansion, and rupture: A pathological and methodological review. J. Cell. Physiol. 2022, 237, 278–300. [Google Scholar] [CrossRef]
  79. Tanweer, O.; Wilson, T.A.; Metaxa, E.; Riina, H.A.; Meng, H. A comparative review of the hemodynamics and pathogenesis of cerebral and abdominal aortic aneurysms: Lessons to learn from each other. J. Cerebrovasc. Endovasc. Neurosurg. 2014, 16, 335–349. [Google Scholar] [CrossRef] [Green Version]
  80. Boyd, A.J.; Kuhn, D.C.; Lozowy, R.J.; Kulbisky, G.P. Low wall shear stress predominates at sites of abdominal aortic aneurysm rupture. J. Vasc. Surg. 2016, 63, 1613–1619. [Google Scholar] [CrossRef] [Green Version]
  81. Chiu, J.J.; Chien, S. Effects of disturbed flow on vascular endothelium: Pathophysiological basis and clinical perspectives. Physiol. Rev. 2011, 91, 327–387. [Google Scholar] [CrossRef] [Green Version]
  82. Adams, L.C.; Brangsch, J.; Reimann, C.; Kaufmann, J.O.; Nowak, K.; Buchholz, R.; Karst, U.; Botnar, R.M.; Hamm, B.; Makowski, M.R. Noninvasive imaging of vascular permeability to predict the risk of rupture in abdominal aortic aneurysms using an albumin-binding probe. Sci. Rep. 2020, 10, 3231. [Google Scholar] [CrossRef]
  83. Yuan, Z.; Lu, Y.; Wei, J.; Wu, J.; Yang, J.; Cai, Z. Abdominal Aortic Aneurysm: Roles of Inflammatory Cells. Front. Immunol. 2020, 11, 609161. [Google Scholar] [CrossRef]
  84. Addis, R.; Campesi, I.; Fois, M.; Capobianco, G.; Dessole, S.; Fenu, G.; Montella, A.; Cattaneo, M.G.; Vicentini, L.M.; Franconi, F. Human umbilical endothelial cells (HUVECs) have a sex: Characterisation of the phenotype of male and female cells. Biol. Sex Differ. 2014, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  85. Lorenz, M.; Koschate, J.; Kaufmann, K.; Kreye, C.; Mertens, M.; Kuebler, W.M.; Baumann, G.; Gossing, G.; Marki, A.; Zakrzewicz, A.; et al. Does cellular sex matter? Dimorphic transcriptional differences between female and male endothelial cells. Atherosclerosis 2015, 240, 61–72. [Google Scholar] [CrossRef]
  86. Mudrovcic, N.; Arefin, S.; Van Craenenbroeck, A.H.; Kublickiene, K. Endothelial maintenance in health and disease: Importance of sex differences. Pharmacol. Res. 2017, 119, 48–60. [Google Scholar] [CrossRef]
  87. Skaug, E.A.; Aspenes, S.T.; Oldervoll, L.; Morkedal, B.; Vatten, L.; Wisloff, U.; Ellingsen, O. Age and gender differences of endothelial function in 4739 healthy adults: The HUNT3 Fitness Study. Eur. J. Prev. Cardiol. 2013, 20, 531–540. [Google Scholar] [CrossRef]
  88. Routledge, F.S.; Hinderliter, A.L.; Blumenthal, J.A.; Sherwood, A. Sex differences in the endothelial function of untreated hypertension. J. Clin. Hypertens. 2012, 14, 228–235. [Google Scholar] [CrossRef] [Green Version]
  89. Crabtree, M.J.; Hale, A.B.; Channon, K.M. Dihydrofolate reductase protects endothelial nitric oxide synthase from uncoupling in tetrahydrobiopterin deficiency. Free Radic. Biol. Med. 2011, 50, 1639–1646. [Google Scholar] [CrossRef] [Green Version]
  90. Daugherty, A.; Cassis, L.A. Mouse models of abdominal aortic aneurysms. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 429–434. [Google Scholar] [CrossRef] [Green Version]
  91. Saraff, K.; Babamusta, F.; Cassis, L.A.; Daugherty, A. Aortic dissection precedes formation of aneurysms and atherosclerosis in angiotensin II-infused, apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1621–1626. [Google Scholar] [CrossRef] [PubMed]
  92. Gao, L.; Chalupsky, K.; Stefani, E.; Cai, H. Mechanistic insights into folic acid-dependent vascular protection: Dihydrofolate reductase (DHFR)-mediated reduction in oxidant stress in endothelial cells and angiotensin II-infused mice: A novel HPLC-based fluorescent assay for DHFR activity. J. Mol. Cell. Cardiol. 2009, 47, 752–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Siu, K.L.; Miao, X.N.; Cai, H. Recoupling of eNOS with folic acid prevents abdominal aortic aneurysm formation in angiotensin II-infused apolipoprotein E null mice. PLoS ONE 2014, 9, e88899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Chalupsky, K.; Cai, H. Endothelial dihydrofolate reductase: Critical for nitric oxide bioavailability and role in angiotensin II uncoupling of endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 2005, 102, 9056–9061. [Google Scholar] [CrossRef] [Green Version]
  95. Li, Q.; Youn, J.Y.; Siu, K.L.; Murugesan, P.; Zhang, Y.; Cai, H. Knockout of dihydrofolate reductase in mice induces hypertension and abdominal aortic aneurysm via mitochondrial dysfunction. Redox Biol. 2019, 24, 101185. [Google Scholar] [CrossRef]
  96. Gao, L.; Siu, K.L.; Chalupsky, K.; Nguyen, A.; Chen, P.; Weintraub, N.L.; Galis, Z.; Cai, H. Role of uncoupled endothelial nitric oxide synthase in abdominal aortic aneurysm formation: Treatment with folic acid. Hypertension 2012, 59, 158–166. [Google Scholar] [CrossRef]
  97. Chuaiphichai, S.; Rashbrook, V.S.; Hale, A.B.; Trelfa, L.; Patel, J.; McNeill, E.; Lygate, C.A.; Channon, K.M.; Douglas, G. Endothelial Cell Tetrahydrobiopterin Modulates Sensitivity to Ang (Angiotensin) II-Induced Vascular Remodeling, Blood Pressure, and Abdominal Aortic Aneurysm. Hypertension 2018, 72, 128–138. [Google Scholar] [CrossRef]
  98. Izawa-Ishizawa, Y.; Imanishi, M.; Zamami, Y.; Toya, H.; Nagao, T.; Morishita, M.; Tsuneyama, K.; Horinouchi, Y.; Kihira, Y.; Takechi, K.; et al. Development of a novel aortic dissection mouse model and evaluation of drug efficacy using in-vivo assays and database analyses. J. Hypertens. 2019, 37, 73–83. [Google Scholar] [CrossRef]
  99. Li, Y.; LeMaire, S.A.; Shen, Y.H. Molecular and Cellular Dynamics of Aortic Aneurysms Revealed by Single-Cell Transcriptomics. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2671–2680. [Google Scholar] [CrossRef]
  100. Dawson, A.; Wang, Y.; Li, Y.; LeMaire, S.A.; Shen, Y.H. New Technologies With Increased Precision Improve Understanding of Endothelial Cell Heterogeneity in Cardiovascular Health and Disease. Front. Cell Dev. Biol. 2021, 9, 679995. [Google Scholar] [CrossRef]
  101. Hadi, T.; Boytard, L.; Silvestro, M.; Alebrahim, D.; Jacob, S.; Feinstein, J.; Barone, K.; Spiro, W.; Hutchison, S.; Simon, R.; et al. Macrophage-derived netrin-1 promotes abdominal aortic aneurysm formation by activating MMP3 in vascular smooth muscle cells. Nat. Commun. 2018, 9, 5022. [Google Scholar] [CrossRef]
  102. Boytard, L.; Hadi, T.; Silvestro, M.; Qu, H.; Kumpfbeck, A.; Sleiman, R.; Fils, K.H.; Alebrahim, D.; Boccalatte, F.; Kugler, M.; et al. Lung-derived HMGB1 is detrimental for vascular remodeling of metabolically imbalanced arterial macrophages. Nat. Commun. 2020, 11, 4311. [Google Scholar] [CrossRef]
  103. Davis, F.M.; Tsoi, L.C.; Melvin, W.J.; denDekker, A.; Wasikowski, R.; Joshi, A.D.; Wolf, S.; Obi, A.T.; Billi, A.C.; Xing, X.; et al. Inhibition of macrophage histone demethylase JMJD3 protects against abdominal aortic aneurysms. J. Exp. Med. 2021, 218, 1–19. [Google Scholar] [CrossRef]
  104. Yang, H.; Zhou, T.; Stranz, A.; DeRoo, E.; Liu, B. Single-Cell RNA Sequencing Reveals Heterogeneity of Vascular Cells in Early Stage Murine Abdominal Aortic Aneurysm-Brief Report. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 1158–1166. [Google Scholar] [CrossRef]
  105. Zhao, G.; Lu, H.; Chang, Z.; Zhao, Y.; Zhu, T.; Chang, L.; Guo, Y.; Garcia-Barrio, M.T.; Chen, Y.E.; Zhang, J. Single-cell RNA sequencing reveals the cellular heterogeneity of aneurysmal infrarenal abdominal aorta. Cardiovasc. Res. 2021, 117, 1402–1416. [Google Scholar] [CrossRef]
  106. Li, B.; Song, X.; Guo, W.; Hou, Y.; Hu, H.; Ge, W.; Fan, T.; Han, Z.; Li, Z.; Yang, P.; et al. Single-Cell Transcriptome Profiles Reveal Fibrocytes as Potential Targets of Cell Therapies for Abdominal Aortic Aneurysm. Front. Cardiovasc. Med. 2021, 8, 753711. [Google Scholar] [CrossRef]
Figure 1. Diagram depicting mechanisms by which endothelial dysfunction contributes to AAA growth, including eNOS uncoupling, a phenotypic switch to a pro-thrombotic, pro-adhesive, permeable state in the setting of altered biomechanical stress, and EC stress generated by intraluminal thrombus accumulation. EC populations 1 and 2 as determined by single-cell RNA sequencing are shown in distinct colors (pink, purple) in low (light) and high (dark) stress states. Potential biomarkers (circulating TM, CCL2, EPCs, and FMD analysis) are depicted. Tobacco use is highlighted as a clinical risk factor for endothelial dysfunction and AAA progression. Abbreviations: AAA (abdominal aortic aneurysm); ARG (arginine); CCL20 (C-C Motif Chemokine Ligand 20); CIT (citrulline); EC (endothelial cell); eNOS (endothelial nitric oxide synthase); EPC (endothelial progenitor cell); FMD (flow mediated dilation); MMP (matrix metalloproteinase); NO (nitric oxide); TM (thrombomodulin).
Figure 1. Diagram depicting mechanisms by which endothelial dysfunction contributes to AAA growth, including eNOS uncoupling, a phenotypic switch to a pro-thrombotic, pro-adhesive, permeable state in the setting of altered biomechanical stress, and EC stress generated by intraluminal thrombus accumulation. EC populations 1 and 2 as determined by single-cell RNA sequencing are shown in distinct colors (pink, purple) in low (light) and high (dark) stress states. Potential biomarkers (circulating TM, CCL2, EPCs, and FMD analysis) are depicted. Tobacco use is highlighted as a clinical risk factor for endothelial dysfunction and AAA progression. Abbreviations: AAA (abdominal aortic aneurysm); ARG (arginine); CCL20 (C-C Motif Chemokine Ligand 20); CIT (citrulline); EC (endothelial cell); eNOS (endothelial nitric oxide synthase); EPC (endothelial progenitor cell); FMD (flow mediated dilation); MMP (matrix metalloproteinase); NO (nitric oxide); TM (thrombomodulin).
Biomolecules 12 00509 g001
Table 1. Clinical studies of flow mediated dilation (FMD) in abdominal aortic aneurysm (AAA).
Table 1. Clinical studies of flow mediated dilation (FMD) in abdominal aortic aneurysm (AAA).
AuthorsStudy DesignParticipantsMethodsFindings
Medina et al., 2010 [18]Cross SectionalN = 30 (30 men)Correlate FMD with AAA diameter at a static point in time
  • Negative correlation between AAA diameter and FMD (R = −0.78 p < 0.001)
  • FMD significantly differed across AAA diameter quartiles (p < 0.001)
Sung et al., 2013 [23]Cross SectionalN = 78 (15 healthy controls [100% men], 27 small AAA [93% men], 36 large AAA [89% men])Evaluate FMD in patients with normal aortic diameter (M < 3.5 cm, W < 3 cm), small aneurysm (M 3.5–5.5 cm, W 3–5 cm), large aneurysm (M > 5.5 cm, W > 5 cm)
  • FMD was significantly lower in large (5.26 ± 3.11%) and small (6.31 ± 3.66%) AAA patients compared to controls (8.88 ± 4.83%, p = 0.008)
Lee et al., 2017 [19]Prospective CohortN = 162 (147 men, 15 women)Measure AAA diameter and FMD over time
  • Negative correlation between AAA diameter and FMD (R = −0.28, p < 0.001)
  • FMD inversely correlated with AAA diameter progression (R = −0.35, p = 0.001)
  • FMD deteriorates over AAA surveillance (median 2% at baseline to 1.2% at follow up; p = 0.004)
  • Surgical repair of AAA leads to improved FMD (1.1% pre-op to 3.8% post op, p < 0.001)
Bailey et al., 2018 [20]Prospective CohortN = 44 (22 AAA patients, 22 healthy adults, 100% men)Measure FMD in AAA patients and healthy controls at baseline and after exercise
  • Baseline brachial FMD was 1.10% lower (95% CI 0.72–0.81) in AAA patients compared to healthy controls
Flow mediated dilation (FMD); Abdominal aortic aneurysm (AAA); M (men); W (women).
Table 2. Single-cell RNA sequencing studies of abdominal aortic aneurysm (AAA).
Table 2. Single-cell RNA sequencing studies of abdominal aortic aneurysm (AAA).
AuthorsHuman AAA or Mouse AAA ModelControl GroupDuration of AAA Induction
Davis et al., 2021 [103]Tissue from AAA patients undergoing open aortic aneurysm repair Tissue from patients undergoing open aortobifemoral bypassNot applicable
Hadi et al., 2018 [101]1000 ng/kg/min Ang II infusion in Apoe−/− micePBS-infused Apoe−/− mice28 days
Boytard et al., 2020 [102]1000 ng/kg/min Ang II infusion in Apoe−/− micePBS-infused Apoe−/− mice28 days
Li et al., 2021 [106]1000 ng/kg/min Ang II infusion in Apoe−/− mice Saline-infused Apoe−/− mice28 days
Zhao et al., 2021 [105]Peri-adventitial incubation of elastase in C57BL/6J micePeri-adventitial incubation of heat-inactivated elastase in C57BL/6J mice7 and 14 days
Yang et al., 2021 [104]Peri-adventitial incubation of 0.5 M CaCl2 in C57BL/6J micePeri-adventitial incubation of 0.5 M NaCl in C57BL/6J mice4 days
AAA (abdominal aortic aneurysm); Ang II (Angiotensin II).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

DeRoo, E.; Stranz, A.; Yang, H.; Hsieh, M.; Se, C.; Zhou, T. Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm. Biomolecules 2022, 12, 509. https://doi.org/10.3390/biom12040509

AMA Style

DeRoo E, Stranz A, Yang H, Hsieh M, Se C, Zhou T. Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm. Biomolecules. 2022; 12(4):509. https://doi.org/10.3390/biom12040509

Chicago/Turabian Style

DeRoo, Elise, Amelia Stranz, Huan Yang, Marvin Hsieh, Caitlyn Se, and Ting Zhou. 2022. "Endothelial Dysfunction in the Pathogenesis of Abdominal Aortic Aneurysm" Biomolecules 12, no. 4: 509. https://doi.org/10.3390/biom12040509

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

Article Metrics

Back to TopTop