You are on page 1of 8

High Blood Pressure & Cardiovascular Prevention

https://doi.org/10.1007/s40292-019-00356-y

REVIEW ARTICLE

From Endothelium to Lipids, Through microRNAs and PCSK9:


A Fascinating Travel Across Atherosclerosis
D. D’Ardes1,2 · F. Santilli1 · M. T. Guagnano1 · M. Bucci1,2 · F. Cipollone1,2 

Received: 16 September 2019 / Accepted: 17 December 2019


© Italian Society of Hypertension 2020

Abstract
Lipids and endothelium are pivotal players on the scene of atherosclerosis and their interaction is crucial for the establish-
ment of the pathological processes. The endothelium is not only the border of the arterial wall: it plays a key role in regu-
lating circulating fatty acids and lipoproteins and vice versa it is regulated by these lipidic molecules thereby promoting
atherosclerosis. Inflammation is another important element in the relationship between lipids and endothelium. Recently,
proprotein convertase subtilisin/kexin type 9 (PCSK9) has been recognized as a fundamental regulator of LDL-C and anti-
PCSK9 monoclonal antibodies have been approved for therapeutic use in hypercholesterolemia, with the promise to subvert
the natural history of the disease. Moreover, growing experimental and clinical evidence is enlarging our understanding of
the mechanisms through which this protein may facilitate the genesis of atherosclerosis, independently of its impact on lipid
metabolism. In addition, environmental stimuli may affect the post-transcriptional regulation of genes through micro-RNAs,
which in turn play a key role in orchestrating the crosstalk between endothelium and cholesterol. Advances in experimental
research, with development of high throughput techniques, have led, over the last century, to a tremendous progress in the
understanding and fine tuning of the molecular mechanisms leading to atherosclerosis. Identification of pivotal keystone
molecules bridging lipid metabolism, endothelial dysfunction and atherogenesis will provide the mechanistic substrate to
test valuable targets for prediction, prevention and treatment of atherosclerosis-related disease.

Keywords  Atherosclerosis · Endothelium · Lipids · PCSK9 · miRNA

1 A Couple at the Beginning a duplex role, having both sensory and executive functions,
of Atherosclerosis: Endothelium and can also generate molecules thus influencing multiple
and Lipids processes such as vascular tone and remodeling, inflamma-
tion and thrombosis. A lot of different forces act on endothe-
Atherosclerosis is the result of complex pathogenic mecha- lial cells (EC), one being the fluid shear stress. In fact,
nisms initiated by the lipid-endothelium intertwining, even- where blood flow is uniform and laminar, cells are ellipsoid
tually leading to the development of atherosclerotic plaques. in shape and aligned in the direction of flow, showing ele-
The endothelium is characterized by intercellular tight junc- vated nitric oxide (NO) production, reduced oxidant stress,
tions and represents a selectively permeable barrier. It has enhanced barrier function and limited proinflammatory
activity, through Erk5-dependent expression of Klf2, which
binds NF-κB and competes for binding of transcriptional
D. D’Ardes and F. Santilli equally contributed to this review coactivators. Vice versa, where flow is altered, cells have
article. polygonal shapes without a particular orientation, showing
an increased permeability to macromolecules, for example
* F. Cipollone
francesco.cipollone@unich.it low-density lipoprotein cholesterol (LDL-C) [1], reduced
NO production and enhanced oxidant stress, endothelial
1
Department of Medicine and Aging, “G. d’Annunzio” turnover and proinflammatory activation. Moreover, expo-
University, Chieti, Italy sure to laminar flow in culture actively suppresses endothe-
2
Clinica Medica Division and European Center of Excellence lial activation by cytokines and oxidized LDL-C by hinder-
on Atherosclerosis, Hypertension and Dyslipidemia ing NF-кB-dependent proinflammatory gene expression and
“SS. Annunziata” Hospital, Chieti, Italy

Vol.:(0123456789)
D. D’Ardes et al.

promoting NF-кB-dependent anti-apoptotic gene expression process is supported by observations showing a progressive
[2]. decrease in platelet formation of NO moving from patients
The accumulation of LDL-C in the subendothelial matrix with hereditary deficiency in gp91phox (NOX-2) to obese
is indeed one of the most important initiating events of ath- patients [8].
erosclerosis. The higher the LDL-C levels, the greater the NOX2 represents the catalytic core of nicotinamide
accumulation: LDL diffuses passively through EC junctions adenine dinucleotide phosphate (NADPH) oxidase, that is
and its retention in the vessel wall involves interactions believed to modulate arterial tone. In fact, vascular tone is
between the LDL-C constituent apolipoprotein B (apoB) modulated by NADPH oxidase-generated oxidative stress,
and matrix proteoglycans [3]. affecting NO generation. Since platelets are a pivotal source
However, atherosclerosis is not just a LDL-C-centered of systemic NOX-2, it is conceivable that they are the initia-
process: other apoB-containing lipoproteins, namely tors of this detrimental cascade in settings of persistently
lipoprotein(a) and remnants, can also accumulate in enhanced platelet activation, such as obesity and metabolic
the intima and promote atherosclerosis. In particular, syndrome [9].
lipoprotein(a) has been shown to be particularly atherogenic
for its effects on fibrinolysis and smooth muscle cells (SMC)
growth [4].
A strong relationship exists between LDL-C and arterial 2 The Lipids–Endothelium Couple
wall: the LDL-C are somehow ‘modified’ in the vessel wall, and a Third Wheel: Inflammation
undergoing oxidation, lipolysis, proteolysis and aggregation,
thus contributing to inflammation as well as to foam-cell Inflammation is another important element in the relation-
formation. Lipid peroxidation is one of the most significant ship between lipids and endothelium. If macrophages play
modifications in terms of early lesion formation as a result a fundamental role since the initial phases of the athero-
of the exposure of vascular cells to oxidative stress stimuli, sclerotic process, thus participating to plaque progression, a
a combination of enzymatic, such as lipoxygenases, and staggering array of immune cells migrate into the plaque and
non-enzymatic, free-radical catalyzed reactions. The main release different kinds of cytokines contributing to a further
modification of LDL-C takes place inside the intima, where progression or to a regression of the plaque.
proteoglycans can sequester LDL-C [5]. The cytokines associated with inflammatory macrophage
If endothelium influences lipids, their migration and mod- and Th1 T cell responses, such as interferon-γ (IFN-γ), inter-
ification, vice versa a large number of studies have shown leukin (IL)-1α, IL-1β, and tumor necrosis factor-α (TNF-α)
an impairment of vasodilatation mediated by endothelium- are responsible for encouraging atherosclerosis and these
derived NO in response to elevated total cholesterol and pro-atherogenic cytokines enhance foam cell formation by
particularly LDL-C. LDL-C effects have been demonstrated enhancing modified LDL-C uptake and inhibiting proper
at numerous sites of the NO signaling pathway including reverse cholesterol transport in macrophages. Other kinds
G-protein-coupled receptors, NO synthase (NOS) and NO of cytokines, such as IL-13, IL-10, IL-19 and transform-
bioactivity, with evidence for enhanced superoxide forma- ing growth factor-β (TGF-β), have been shown to reduce
tion and the consequent production of the less potent dilator atherosclerosis [10].
peroxynitrite. The effects of lipids on endothelium-depend- TNF-α and IFN-γ also contribute to macrophage and vas-
ent vasodilation can be reversed not only by reducing the cular smooth muscle cells (VSMCs) apoptosis, promoting
level of lipids but also by provision of the NOS substrate, the thinning of the fibrous cap and the growth of the necrotic
l-arginine and antioxidants, although the mechanisms under- core, whose progression is linked to the formation of cho-
lying these effects are not fully elucidated [6]. lesterol crystals in the cytosols, leading to inflammasome
Lipids and endothelium do not represent the only pro- activation and subsequent IL-1α/β production [11].
tagonists on the scene: a lot of other actors can influence this In addition, macrophages can contribute to oxidative
relationship and modulate endothelial dysfunction. Platelets stress through production of hydrogen peroxide, superox-
are an example of potential “influencers”. ides, neopterin and myeloperoxidases [12].
Interestingly, both platelet activation and endothelial dys- These radicals can promote further oxidation of LDL-C
function are early events in the natural history of atheroscle- within the plaques, as well as the triggering of endoplasmic
rosis. A continuous dialogue exists between platelets and reticulum (ER) stress that can initiate apoptosis. In contrast
endothelial cells, dating back to the bone marrow, where to atherogenic inflammatory responses, the atheroprotec-
haematopoietic precursors, megakaryocytes, and sinusoidal tive cytokines IL-10, IL-19 and TGF-β reduce leukocyte
endothelium interact directly [7]. In addition, platelets are recruitment and inflammation and slow down atherosclerosis
a source of oxygen free radicals, and trigger a cascade of progression [13]. Thus, the plaque may be considered as a
events that ultimately leads to endothelial dysfunction: this dynamic environment in which plaque progression is the
From Endothelium to Lipids, Through microRNAs and PCSK9: A Fascinating Travel Across…

result of the balance between pro-inflammatory and athero- Because PCSK9 induces the degradation of LDL-R, its
protective cytokines. pharmacological inhibition would prolong the duration
In this regard, a pivotal role has been reported for soluble of the receptor action, leading to a drastic reduction of
CD40 ligand (sCD40L), with particular reference to diabetic LDL-C [23].
patients. Upregulation of sCD40L as a consequence of per- Moreover, growing experimental and clinical evidence
sistent hyperglycaemia in diabetic patients seems to result is widening our understanding of the mechanisms through
in EC activation and monocyte recruitment to the arterial which this protein may facilitate the genesis of athero-
wall, possibly contributing to accelerated atherosclerosis sclerosis, independently of its impact on lipid metabolism
development in diabetes. In fact, elevated sCD40L levels [24].
have been associated with enhanced in vitro expression of An increasingly emerging hypothesis suggests that the
adhesion molecules, impaired migration in ECs, enhanced impact of PCSK9 on lipids and on atherosclerosis involves
­O2-generation in monocytes and with higher HbA1c; vice mechanisms going beyond the regulation of the clearance of
versa, metabolic profile improvement has been associated the LDL-C: PCSK9, in fact, does not bind exclusively the
with a reduction of plasma sCD40L [14]. LDL-R but promotes the degradation of other receptors such
In addition to inflammation, endothelial cells at athero- as the receptor of very low density lipoproteins (VLDL),
susceptible sites show signs of enhanced cellular injury as a CD36, ApoER2 and receptor of type 1 LDL (LRP1) [25,
result of mechanical, oxidative and metabolic stress. Tran- 26], whose contribution to the homeostasis of cholesterol
scriptome analysis of endothelial cells at athero-susceptible is still unclear, but seems to be linked to triglyceride-rich
sites suggests that ER stress may exert a pivotal role in ath- lipoprotein (TRL) metabolism. TRLs represent important
erogenic endothelial activation [15]. elements in atherosclerosis development and the impact of
ER stress is significant when protein rates overcome the PCSK9 on these highly pro-atherogenic particles provides
ER’s ability to promote proper protein folding. The accumu- an additional mechanism by which PCSK9 inhibitors can
lation of misfolded peptides initiates the unfolded protein reduce the risk of atherosclerotic cardiovascular diseases.
response and this process results in an upregulation of ER PCSK9 has also been shown to provide other proathero-
chaperone proteins. ER stress plays a role in inflammation, sclerotic effects not exclusively linked to plasma concentra-
oxidative stress and apoptosis [16]. tions of apoB-containing lipoproteins. Indeed, PCSK9 seems
It has also been noted that disturbed flow promotes ER to be expressed in the atheromatous plaque and in particular
stress in endothelial cells and mimicking ER stress enhances at the level of VSMCs and in ECs, where it exerts parac-
atherosclerotic plaque formation in laminar flow regions rine effects on an array of foam cell receptors, including the
[17]. LDL-R and CD36. This represents a potential mechanism
In contrast, laminar flow is known to reduce endothe- to modulate atherogenesis independently of plasma LDL-C
lial cell metabolism through Kruppel like factor 2 (Klf2)- concentrations, although its impact on each of these foam
dependent repression of endothelial glycolytic enzymes cell receptors and its relevance to atherosclerosis remains
[18], eliciting in turn endothelial cell autophagy to protect unclear [27].
endothelial cells from oxidant-induced cell injury [19]. PCSK9 released by VSMCs may influence the expression
Moreover, oscillatory flow stimulates a c-Jun N-terminal of the LDL-R on the surface of macrophages [28]. PCSK9
kinase (JNK)-dependent increase in mitochondrial super- has been also shown to promote the inflammatory response
oxide production limiting autophagy, thus accumulating in macrophages [29].
oxidant injuries at sites of disturbed flow [20]. The expression of PCSK9 in VSMCs appears to be influ-
enced by shear stress. Moreover, in the absence of PCSK9,
a protective effect has been observed on the formation of the
3 PCSK9: An Actor Not to be Missed neointima, after a vascular insult [30].
An interesting relationship may also exist between
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a PCSK9 and endothelial function. Indeed, treatment with a
fundamental regulator of LDL-C. Consistently, anti-PCSK9 PCSK9 inhibitor may improve endothelial function in sub-
monoclonal antibodies have been recently approved for the jects with increased cardiovascular risk and this improve-
treatment of heterozygous familial hyper-cholesterolemia ment is proportional to LDL-C reduction [31].
and clinical atherosclerotic cardiovascular disease as an One study reported an impressive improvement in
addition to diet and maximally-tolerated statin therapy in carotid-femoral pulse wave velocity after short-term treat-
adults who require additional LDL-C lowering [21]. ment with a PCSK9 inhibitor. This result was achieved
PCSK9 interacts with the LDL-C receptor (LDL-R) despite an increase in LDL-C, suggesting that the PCSK9
inducing its degradation by conveying it in lysosomes and inhibitors may exert a beneficial impact on vasomotor func-
blocking its recycling on the cytoplasmic membrane [22]. tion independently of their lipid-lowering effect [32].
D. D’Ardes et al.

Unexpectedly, given the body of evidence on endothe- MiRNAs are short non-coding RNA molecules (about
lial function, studies performed until now have shown no 18–24 nucleotides), exerting their function via the seed
correlation between PCSK9 and hypertension. Indeed, region, a sequence of 6–8 nucleotides that binds to messen-
PCSK9−/− mice display normal sodium balance and blood ger ribonucleic acid (mRNA) [38] identified over 25 years
pressure regulation [33]. Moreover, PCSK9 level is unre- ago [39]. This interaction with mRNA often results in the
lated to blood pressure in Chinese hypertensive patients prevention of protein production by different mechanisms.
[34]. Larger studies are needed to better clarify the link Both hyperlipidemia and shear stress are two important
between hypertension and PCSK9. pathogenetic elements in the vessel wall in terms of athero-
Another important element within the relationship between sclerotic lesion formation. When stimulated by biochemical
inflammation, PCSK9 and atherosclerosis is represented by and biomechanical stimuli, endothelial cells undergo a series
the lectin-like oxidized low-density lipoprotein receptor-1 of molecular and cellular conformational changes promot-
(LOX-1) whose expression is increased by inflammation and ing atherogenesis. First of all, early induction of adhesion
that is involved in oxidized LDL-C uptake with a self-regula- molecule expression such as vascular cell adhesion molecule
tion mechanism with PCSK9, that induces LOX-1 transcrip- (VCAM)-1, intracellular adhesion molecule (ICAM)-1 and
tion which in turn stimulates PCSK9 [35]. Furthermore, in an E-selectin facilitates leukocyte recruitment to the vessel wall
inflammatory context, high levels of PCSK9 potently stimu- and may be among the earliest hallmarks associated with
late the expression of LOX-1 and oxidized LDL-C uptake nascent plaques [40].
in macrophages and, moreover, PCSK9 has been shown to A lot of miRNAs have been implicated in atherogenesis
promote atherosclerosis directly through the modulation of thanks to the possibility to target the 3′-UTRs of these mol-
proinflammatory monocytes in the arterial wall [36]. ecules such as miR-17-3p, targeting ICAM-1, and miR-31,
In conclusion, PCSK9 appears to have pleiotropic actions targeting E-selectin [41].
and, consistently, the effect of anti-PCSK9 monoclonal anti-
bodies may determine not only a strong reduction of LDL-C, 4.1 MiRNAs, Endothelium and Inflammation
but also other important effects in the atherosclerotic pro-
cess, that are still the object of ongoing investigations. MiR-92a is expressed in ECs and is dynamically regulated
by shear stress both in vitro and in vivo [42]. When compar-
ing in vivo athero-prone areas of the aortic arch with athero-
4 micro‑RNAs: The Directors Behind resistant regions, miR-92a expression has been found highly
the Scene of Atherosclerosis induced in the first ones [43].
In the context of the dynamic relation between endothe-
MicroRNAs (miRNAs) play a key role in the context of the lium and physical forces that act within the vessels, a large
crosstalk between endothelium and cholesterol [37] (Fig. 1). number of miRNAs, such as miR-126, miR-10a, miR-19a,

Fig. 1  Summary of the influence of PCSK9 and hypercholesterolemia on the relationship between endothelium and lipids. In this context, also
microRNAs, shear stress and inflammation play an important role
From Endothelium to Lipids, Through microRNAs and PCSK9: A Fascinating Travel Across…

miR-23b, miR-21, miR-663, miR-143/145, miR-101, miR- and activity of LDL-R, that usually promotes the clearance of
712, miR-205, miR-155, 146a and miR-181b seem to have a circulating LDL-C particles [53].
role as mechano-miRNAs. These mechano-miRNAs mainly MiRNAs have also been reported to act as critical regula-
target key signaling pathways involved in endothelial cells tors of HDL-C biogenesis and cholesterol efflux. In fact, a
cycle, inflammation, apoptosis and NO signaling [44]. great number of miRNAs (miR-33 [54], miR-758 [55], miR-26
Moreover, miR-126 is one of the most widely expressed [56], miR-106 [57], miR-144 [58]) have been shown to target
miRNAs in the endothelium and it has been shown to play a ABCA1 to reduce cholesterol efflux to apoA1 in vitro. MiR-33
crucial role in regulating inflammation and angiogenesis [45]. inhibition in mouse models of atherosclerosis plays a pivotal
In fact, miR-126 has been involved in endothelial dys- role in the modulation of circulating HDL-C and regression of
function itself, by reducing VCAM-1 expression and thereby atherosclerosis. Targeted deletion of miR-33 increased plasma
leucocyte homing in the arteries, thus modulating, through levels of HDL-C and reduced atherosclerotic plaque size in
different pathways, vascular inflammation [46]. Apoe−/− mice and furthermore treatment of Ldlr−/− mice
A similar role in the regulation of vascular inflamma- increased plasma levels of HDL-C associated with a regres-
tion is played by miR-155 and miR-221/222 through NOS sion of atherosclerosis [59].
regulation [47]. MiRNAs have also been implicated in macrophage choles-
Moreover, miR-155 increased untreated monocyte terol efflux in stable plaque. In fact, adenosine triphosphate
adhesion to aortic endothelial cells by promoting ICAM-1 (ATP)-binding cassette (ABC) transporters A1 and G1 are the
expression during homeostasis and lipopolysaccharides main transporters involved in this process and we showed that
(LPS)-induced inflammation [48]. miR-758 and miR-33b are potential modulators of ABCA1
On the contrary, a number of miRNAs with anti-inflam- expression in atherosclerotic plaques [60].
matory properties has been identified. For example, miR-10a
is expressed in endothelial cells where it seems to contrast 4.3 MiRNAs and Atherosclerotic Plaques
pro-inflammatory gene expression in athero-susceptible
regions in vivo, thus exerting a possible atheroprotective MiRNAs have been also implicated in plaque instability and
role [49]. related cardiovascular events, such as stroke and myocardial
infarction. We demonstrated that profound differences exist
4.2 MiRNAs, Liver and Cholesterol Metabolism in the expression of 5 miRNAs (miR-100, mi-127, miR-145,
miR-133a, and miR-133b) in symptomatic versus asympto-
In addition to modulate inflammation, miRNAs are matic plaques. These data were confirmed by in vitro experi-
also involved in cholesterol homeostasis by controlling ments on endothelial cells transfected with miR-145 and miR-
LDL-C and high density lipoprotein cholesterol (HDL-C) 133a that further remarked the importance of these miRNAs
abundance. in the modulation of stroke-related proteins [61].
Since the liver has a key role in the production and clear- Moreover, hypertension may upregulate miR-145 expres-
ance of lipoproteins, a great number of hepatic-enriched sion in human atherosclerotic plaques [62].
miRNAs have been identified that functionally regulate lipo- In light of all these data, it seems clear that miRNAs rep-
protein metabolism. First of all, miR-122 has been implicated resent crucial messengers of the fascinating dialogue between
in lipoprotein metabolism. Studies based on loss-of-function endothelium and lipids. This relation has attracted the attention
experiments in animals, identified miR-122 as a crucial regu- of researchers and it will continue to deepen our knowledge,
lator of cholesterol and lipoprotein homeostasis [50]. thanks to further studies, with important implications in terms
In addition, miR-223 and miR-27b have been identified as of diagnosis and therapies for our patients.
post-transcriptional regulatory hubs involved in the control of
cholesterol and lipoprotein metabolism genes [51, 52]. Con-
cerning miR-223, it represses genes involved in cholesterol 5 Conclusions and Perspectives
biosynthesis (HMGS1, SC4MOL) and HDL-C uptake (SRB1)
[51], whereas miR-27b represses a number of targets (PPARG​, Advances in experimental research, with development of
GPAM, ANGPTL3 and NDST1) involved in lipoprotein metab- high throughput techniques, have led, over the last century,
olism and remodeling [52]. to a tremendous progress in the understanding and fine tun-
MiRNA regulation of plasma levels of LDL-C through tar- ing of the molecular mechanisms leading to atherosclerosis.
geting of the LDL-R has also been reported. LDL-R expres- Identification of pivotal keystone molecules bridging lipid
sion in the liver promotes the clearance of circulating LDL-C metabolism, endothelial dysfunction and atherogenesis, such
particles and is a major determinant of plasma cholesterol as LOX-1, PCSK9 and others, are emerging as valuable tar-
levels. MiRNAs are also involved in the regulation of LDL-C get for prediction, prevention and treatment of atheroscle-
plasma levels: miR-148a is a negative regulator of expression rosis-related disease.
D. D’Ardes et al.

Intensive LDL-C lowering, achieved through the com- DO, Evans PC. Laminar shear stress acts as a switch to regulate
bined therapy with PCSK9 antibody and statin, has been divergent functions of NF-kappaB in endothelial cells. FASEB J.
2007;21:3553–61.
associated with coronary plaque regression in a patient with 3. Cipollone F, Rocca B, Patrono C. Cyclooxygenase-2 expression
double-heterozygous familial hypercholesterolemia with a and inhibition in atherothrombosis. Arterioscler Thromb Vasc
LDL-R mutation and a PCSK9 V4I mutation. This finding Biol. 2004;24:246–55. 
provides the mechanistic framework for studies assessing the 4. Grainger DJ, Kemp PR, Liu AC, Lawn RM, Metcalfe JC. Acti-
vation of transforming growth factor-β is inhibited in transgenic
efficacy of combined therapy including PCSK9 antibodies in apolipoprotein(a) mice. Nature. 1994;370:460–2.
reducing cardiovascular events in patients with established 5. Skalen K, Gustafsson M, Rydberg EK, Hulten LM, Wiklund O,
cardiovascular disease [63]. Innerarity TL, Boren J. Subendothelial retention of atherogenic
The existence of a functional miRNAs machinery in lipoproteins in early atherosclerosis. Nature. 2002;417:750–4.
6. Anderson TJ, Meredith IT, Charbonneau F, et al. Endothelium-
human cells have deepen our knowledge about the molecular dependent coronary vasomotion relates to the susceptibility to
phenotype in health and disease, and opens up the possibility oxidation in humans. Circulation. 1996;93:1647–50.
of modulating gene expression using therapeutic RNAs to 7. Davì G, Santilli F. Platelets, oxidative stress and preservation of
prevent atherogenesis [64]. the vascular endothelium: is it a matter of fat? Intern Emerg Med.
2012;7(3):199–201.
An additional layer of complexity has been provided, over 8. Violi F, Sanguigni V, Carnevale R, Plebani A, Rossi P, Finocchi
the last 10 years, by the publication of large-scale genome- A, Pignata C, De Mattia D, Martire B, Pietrogrande MC, Martino
wide association studies (GWAS), identifying dozens of can- S, Gambineri E, Soresina AR, Pignatelli P, Martino F, Basili S,
didate genetic loci associated with risk for coronary artery Loffredo L. Hereditary deficiency of gp91(phox) is associated
with enhanced arterial dilatation: results of a multicenter study.
disease (CAD). Indeed, genetics accounts for approximately Circulation. 2009;120:1616–22.
50% heritability of CAD. Since the role and function of 9. Davì G, Guagnano MT, Ciabattoni G, Basili S, Falco A,
identified loci or genes remains often elusive, this kind of Marinopiccoli M, Nutini M, Sensi S, Patrono C. Platelet activa-
studies has promoted a novel wave of functional studies to tion in obese women: role of inflammation and oxidant stress.
JAMA. 2002;288:2008–14.
translate GWAS findings into novel discoveries involving 10. Ramji DP, Davies TS. Cytokines in atherosclerosis: key players in
the pathogenesis of atherosclerosis and atherothrombosis. In all stages of disease and promising therapeutic targets. Cytokine
this regard, the novel GWAS-identified CAD risk gene CAD/ Growth Fact Rev. 2015;26(6):673–85.
KIAA1462 has been shown to promote endothelial dysfunc- 11. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauern-
feind FG, Abela GS, Franchi L, Nunez G, Schnurr M, Espevik T,
tion and atherosclerosis. Indeed, junctional protein JCAD Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung
triggers the expression of inflammatory genes in endothelial V, Latz E. NLRP3 inflammasomes are required for atherogenesis
cells, driving an inflammatory process via the recruitment of and activated by cholesterol crystals. Nature. 2010;464:1357–61.
monocytes and resulting in the formation of atherosclerotic 12. Gieseg SP, Amit Z, Yang YT, Shchepetkina A, Katouah H.
Oxidant production, oxLDL uptake, and CD36 levels in human
plaques in ApoE−/− mice [65]. This approach, starting from monocyte-derived macrophages are downregulated by the mac-
the genetic loci associated with the risk of disease and mov- rophage-generated antioxidant 7,8-dihydroneopterin. Antioxid
ing back to the pathophysiology, has also therapeutic impli- Redox Signal. 2010;13:1525–34.
cations. Indeed, targeting GWAS-identified genes relevant 13. Ellison S, Gabunia K, Kelemen SE, England RN, Scalia R, Rich-
ards JM, Orr AW, Traylor JG Jr, Rogers T, Cornwell W, Berglund
to cardiovascular disease, such as JCAD, may represent a LM, Goncalves I, Gomez MF, Autieri MV. Attenuation of experi-
novel strategy for designing innovative drugs to prevent or mental atherosclerosis by interleukin–19. Arterioscler Thromb
delay atherosclerosis and thrombosis. Vasc Biol. 2013;33:2316–24.
14. Cipollone F, Chiarelli F, Davì G, Ferri C, Desideri G, Fazia
M, Iezzi A, Santilli F, Pini B, Cuccurullo C, Tumini S, Del
Compliance With Ethical Standards  Ponte A, Santucci A, Cuccurullo F, Mezzetti A. Enhanced
soluble CD40 ligand contributes to endothelial cell dysfunc-
Conflict of Interest  On behalf of all authors, the corresponding author tion in vitro and monocyte activation in patients with diabetes
states that there is no conflict of interest. mellitus: effect of improved metabolic control. Diabetologia.
2005;48(6):1216–24.
Research Involving Human Participants and/or Animals  Not applicable. 15. Civelek M, Manduchi E, Riley RJ, Stoeckert CJ Jr, Davies PF.
Chronic endoplasmic reticulum stress activates unfolded protein
Informed Consent  Not applicable. response in arterial endothelium in regions of susceptibility to
atherosclerosis. Circ Res. 2009;105:453–61.
16. Scull CM, Tabas I. Mechanisms of ER stress-induced apop-
tosis in atherosclerosis. Arterioscler Thromb Vasc Biol.
2011;31:2792–7.
References 17. Zeng L, Zampetaki A, Margariti A, Pepe AE, Alam S, Mar-
tin D, Xiao Q, Wang W, Jin ZG, Cockerill G, Mori K, Li YS,
1. Gimbrone MA. Vascular endothelium, hemodynamic forces, and Hu Y, Chien S, Xu Q. Sustained activation of XBP1 splicing
atherogenesis. Am J Pathol. 1999;155:1–5. leads to endothelial apoptosis and atherosclerosis develop-
2. Partridge J, Carlsen H, Enesa K, Chaudhury H, Zakkar M, Luong ment in response to disturbed flow. Proc Natl Acad Sci USA.
L, Kinderlerer A, Johns M, Blomhoff R, Mason JC, Haskard 2009;106:8326–31.
From Endothelium to Lipids, Through microRNAs and PCSK9: A Fascinating Travel Across…

18. Doddaballapur A, Michalik KM, Manavski Y, Lucas T, Houtkoo- 36. Ding Z, Liu S, Wang X, Theus S, Deng X, Fan Y, Zhou S,
per RH, You X, Chen W, Zeiher AM, Potente M, Dimmeler S, Mehta JL. PCSK9 regulates expression of scavenger recep-
Boon RA. Laminar shear stress inhibits endothelial cell metabo- tors and ox-LDL uptake in macrophages. Cardiovasc Res.
lism via KLF2-mediated repression of PFKFB3. Arterioscler 2018;114(8):1145–53.
Thromb Vasc Biol. 2015;35:137–45. 37. Santovito D, Mezzetti A, Cipollone F. MicroRNAs and athero-
19. Yang Q, Li X, Li R, Peng J, Wang Z, Jiang Z, Tang X, Peng sclerosis: new actors for an old movie. Nutr Metab Cardiovasc
Z, Wang Y, Wei D. Low shear stress inhibited endothelial cell Dis. 2012;22(11):937–43.
Autophagy through TET2 downregulation. Ann Biomed Eng. 38. Condorelli G, Latronico MVG, Cavarretta E. microRNAs in car-
2016;44(7):2218–27. diovascular diseases: current knowledge and the road ahead. J Am
20. Takabe W, Jen N, Ai L, Hamilton R, Wang S, Holmes K, Dhar- Coll Cardiol. 2014;63:2177–87.
bandi F, Khalsa B, Bressler S, Barr ML, Li R, Hsiai TK. Oscil- 39. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and
latory shear stress induces mitochondrial superoxide production: function. Cell. 2004;116:281–97.
implication of NADPH oxidase and c-Jun NH2-terminal kinase 40. Libby P, Ridker PM, Hansson GK. Progress and chal-
signaling. Antioxid Redox Signal. 2011;15:1379–88. lenges in translating the biology of atherosclerosis. Nature.
21. Karatasakis A, Danek BA, Karacsonyi J, Rangan BV, et al. Effect 2011;473:317–25.
of PCSK9 inhibitors on clinical outcomes in patients with hyper- 41. Suarez Y, Wang C, Manes TD, Pober JS. Cutting edge: TNF-
cholesterolemia: a meta-analysis of 35 randomized controlled tri- induced microRNAs regulate TNF-induced expression of
als. J Am Heart Assoc. 2017;6(12):e006910. E-selectin and intercellular adhesion molecule-1 on human
22. Zhang DW, Lagace TA, Garuti R, Zhao Z, et al. Binding of pro- endothelial cells: feedback control of inflammation. J Immunol.
protein convertase subtilisin/kexin type 9 to epidermal growth 2010;184:21–5.
factor-like repeat A of low density lipoprotein receptor decreases 42. Wu W, Xiao H, Laguna-Fernandez A, Villarreal G Jr, Wang KC,
receptor recycling and increases degradation. J Biol Chem. Geary GG, Zhang Y, Wang WC, Huang HD, Zhou J, Li YS, Chien
2007;282:8602–18612. S, Garcia-Cardena G, Shyy JY. Flow-dependent regulation of
23. Costet P, Krempf M, Cariou B. PCSK9 and LDL cholesterol: kruppel-like factor 2 is mediated by MicroRNA-92a. Circulation.
unravelling the target to design the bullet. Trends Biochem Sci. 2011;124:633–41.
2008;33:426–34. 43. Potteaux S, Vion AC, Guerin CL, Boulkroun S, Rautou PE, Ram-
24. Hess CN, Low Wang CC, Hiatt WR. PCSK9 inhibitors: mecha- khelawon B, Esposito B, Dalloz M, Paul JL, Julia P, Maccario J,
nisms of action, metabolic effects, and clinical outcomes. Annu Boulanger CM, Mallat Z, Tedgui A. Inhibition of microRNA-92a
Rev Med. 2018;69:133–45. prevents endothelial dysfunction and atherosclerosis in mice. Circ
25. Demers A, Samami S, Lauzier B, Des Rosiers C, et al. PCSK9 Res. 2014;114:434–43.
induces CD36 degradation and affects long-chain fatty acid uptake 44. Kumar S, Kim CW, Simmons RD, Jo H. Role of flow-sensitive
and triglyceride metabolism in adipocytes and in mouse liver. microRNAs in endothelial dysfunction and atherosclerosis. Arte-
Arterioscler Thromb Vasc Biol. 2015;35:2517–25. rioscler Thromb Vasc Biol. 2014;34:2206–16.
26. Canuel M, Sun X, Asselin MC, Paramithiotis E, et al. Proprotein 45. Fang Y, Davies PF. Site-specific microRNA-92a regulation of
convertase subtilisin/kexin type 9 (PCSK9) can mediate degra- Kruppel-like factors 4 and 2 in atherosusceptible endothelium.
dation of the low density lipoprotein receptor-related protein 1 Arterioscler Thromb Vasc Biol. 2012;32(4):979–87.
(LRP-1). PLoS One. 2013;8:e64145. 46. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowen-
27. Shapiro MD, Fazio S. PCSK9 and atherosclerosis—lipids and stein CJ. MicroRNA-126 regulates endothelial expression of
beyond. J Atheroscler Thromb. 2017;24:462–72. vascular cell adhesion molecule 1. Proc Natl Acad Sci USA.
28. Ferri N, Tibolla G, Pirillo A, Cipollone F, et al. Proprotein con- 2008;105:1516–21.
vertase subtilisin kexin type 9 (PCSK9) secreted by cultured 47. Poliseno L, Tuccoli A, Mariani L, Evangelista M, Citti L, Woods
smooth muscle cells reduces macrophages LDLR levels. Athero- K, et  al. MicroRNAs modulate the angiogenic properties of
sclerosis. 2012;220:381–6. HUVECs. Blood. 2006;108:3068–71.
29. Ricci C, Ruscica M, Camera M, Rossetti L, et  al. PCSK9 48. Virtue A, Johnson C, Lopez-Pastraña J, Shao Y, Fu H, Li X, Li
induces a pro-inflammatory response in macrophages. Sci Rep. YF, Yin Y, Mai J, Rizzo V, Tordoff M, Bagi Z, Shan H, Jiang X,
2018;8(1):2267. Wang H, Yang XF. MicroRNA-155 deficiency leads to decreased
30. Ferri N, Marchiano S, Tibolla G, Baetta R, et al. PCSK9 knock- atherosclerosis, increased white adipose tissue obesity, and non-
out mice are protected from neointimal formation in response alcoholic fatty liver disease: a novel mouse model of obesity para-
to perivascular carotid collar placement. Atherosclerosis. dox. J Biol Chem. 2017;292(4):1267–87.
2016;253:214–24. 49. Fang Y, Shi C, Manduchi E, Civelek M, Davies PF. MicroRNA-
31. Maulucci G, Cipriani F, Russo D, Casavecchia G, et al. Improved 10a regulation of proinflammatory phenotype in athero-suscep-
endothelial function after short-term therapy with evolocumab. J tible endothelium in vivo and in vitro. Proc Natl Acad Sci USA.
Clin Lipidol. 2018;12:669–73. 2010;107(30):13450–5.
32. Cicero AFG, Toth PP, Fogacci F, Virdis A, et al. Improvement in 50. Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts
arterial stiffness after short-term treatment with PCSK9 inhibitors. L, Booten SL, Graham M, McKay R, Subramaniam A, Propp S,
Nutr Metab Cardiovasc Dis. 2019;29:527–9. Lollo BA, Freier S, Bennett CF, Bhanot S, Monia BP. miR-122
33. Berger JM, Vaillant N, Le May C, Calderon C, et al. PCSK9- regulation of lipid metabolism revealed by in vivo antisense tar-
deficiency does not alter blood pressure and sodium balance in geting. Cell Metab. 2006;3:87–98.
mouse models of hypertension. Atherosclerosis. 2015;239:252–9. 51. Vickers KC, Landstreet SR, Levin MG, Shoucri BM, Toth CL,
34. Yang SH, Du Y, Li S, Zhang Y. Plasma PCSK9 level is unre- Taylor RC, Palmisano BT, Tabet F, Cui HL, Rye KA, Sethupathy
lated to blood pressure and not associated independently with P, Remaley AT. MicroRNA-223 coordinates cholesterol homeo-
carotid intima-media thickness in hypertensives. Hypertens Res. stasis. Proc Natl Acad Sci USA. 2014;111:14518–23.
2016;39:598–605. 52. Vickers KC, Shoucri BM, Levin MG, Wu H, Pearson DS, Osei-
35. Zufeng D, Shijie L, Xianwei W, Xiaoyan D, et al. Cross-talk Hwedieh D, Collins FS, Remaley AT, Sethupathy P. MicroRNA-
between LOX-1 and PCSK9 in vascular tissues. Cardiovasc Res. 27b is a regulatory hub in lipid metabolism and is altered in dys-
2015;107(4):556–67. lipidemia. Hepatology. 2013;57:533–42.
D. D’Ardes et al.

53. Goedeke L, Rotllan N, Canfran-Duque A, Aranda JF, Ramirez atherosclerotic plaque in ApoE −/− mice. J Am Heart Assoc.
CM, Araldi E, Lin CS, Anderson NN, Wagschal A, de Cabo R, 2012;1:e003376.
Horton JD, Lasuncion MA, Naar AM, Suarez Y, Fernandez-Her- 60. Mandolini C, Santovito D, Marcantonio P, Buttitta F, Bucci M,
nando C. MicroRNA-148a regulates LDL receptor and ABCA1 Ucchino S, Mezzetti A, Cipollone F. Identification of microR-
expression to control circulating lipoprotein levels. Nat Med. NAs 758 and 33b as potential modulators of ABCA1 expression
2015;21:1280–9. in human atherosclerotic plaques. Nutr Metab Cardiovasc Dis.
54. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, 2015;25(2):202–9. https:​ //doi.org/10.1016/j.numecd​ .2014.09.005.
Gerszten RE, Naar AM. MicroRNA-33 and the SREBP host 61. Cipollone F, Felicioni L, Sarzani R, Ucchino S, Spigonardo F,
genes cooperate to control cholesterol homeostasis. Science. Mandolini C, Malatesta S, Bucci M, Mammarella C, Santovito
2010;328:1566–9. D, de Lutiis F, Marchetti A, Mezzetti A, Buttitta F. A unique
55. Ramirez CM, Davalos A, Goedeke L, Salerno AG, Warrier N, Cir- microRNA signature associated with plaque instability in humans.
era-Salinas D, Suarez Y, Fernandez-Hernando C. MicroRNA-758 Stroke. 2011;42(9):2556–63.
regulates cholesterol efflux through posttranscriptional repression 62. Santovito D, Mandolini C, Marcantonio P, De Nardis V, Bucci M,
of ATP-binding cassette transporter A1. Arterioscler Thromb Vasc Paganelli C, Magnacca F, Ucchino S, Mastroiacovo D, Desideri
Biol. 2011;31:2707–14. G, Mezzetti A, Cipollone F. Overexpression of microRNA-145 in
56. Sun D, Zhang J, Xie J, Wei W, Chen M, Zhao X. MiR-26 con- atherosclerotic plaques from hypertensive patients. Expert Opin
trols LXR-dependent cholesterol efflux by targeting ABCA1 and Ther Targets. 2013;17(3):217–23.
ARL7. FEBS Lett. 2012;586:1472–9. 63. Shirahama R, Ono T, Nagamatsu S, Sueta D, Takashio S, et al.
57. Kim J, Yoon H, Ramirez CM, Lee SM, Hoe HS, Fernandez- Coronary artery plaque regression by a PCSK9 antibody and rosu-
Hernando C, Kim J. MiR-106b impairs cholesterol efflux and vastatin in double-heterozygous familial hypercholesterolemia
increases Abeta levels by repressing ABCA1 expression. Exp with an LDL receptor mutation and a PCSk9 V4I mutation. Intern
Neurol. 2012;235:476–83. Med. 2018;57:3551–7.
58. Ramirez CM, Rotllan N, Vlassov AV, Davalos A, Li M, Goedeke 64. Kumar S, Williams D, Sur S, Wang JY, Jo H. Role of flow-sensi-
L, Aranda JF, Cirera-Salinas D, Araldi E, Salerno A, Wanschel tive microRNAs and long noncoding RNAs in vascular dysfunc-
AC, Zavadil J, Castrillo A, Jungsu K, Suarez Y, Fernandez-Her- tion and atherosclerosis. Vascul Pharmacol. 2019;114:76–92.
nando C. Control of cholesterol metabolism and plasma HDL 65. Xu S, Xu Y, Liu P, Zhang S, Liu H, Slavin S, Kumar S, Koroleva
levels by miRNA-144. Circ Res. 2013;112:1592–601. M, Luo J, Wu X, Rahman A, Pelisek J, Jo H, Si S, Miller CL,
59. Horie T, Baba O, Kuwabara Y, Chujo Y, Watanabe S, Kinosh- Jin ZG. The novel coronary artery disease risk gene JCAD/
ita M, Horiguchi M, Nakamura T, Chonabayashi K, Hishi- KIAA1462 promotes endothelial dysfunction and atherosclerosis.
zawa M, Hasegawa K, Kume N, Yokode M, Kita T, Kimura T, Eur Heart J. 2019;40:2398–408.
Ono K. MicroRNA-33 deficiency reduces the progression of

You might also like