Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

HHS Public Access

Author manuscript
Circ Res. Author manuscript; available in PMC 2020 February 15.
Author Manuscript

Published in final edited form as:


Circ Res. 2019 February 15; 124(4): 607–618. doi:10.1161/CIRCRESAHA.118.313187.

Cellular Mechanisms of Aortic Aneurysm Formation


Raymundo Alain Quintana, MD1 and W. Robert Taylor, MD, PhD1,2,3
1Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta
GA
2Division of Cardiology, Atlanta VA Medical Center, Decatur GA
3Wallace
H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and
Author Manuscript

Emory University School of Medicine, Atlanta GA

Abstract
Aortic aneurysms are a common vascular disease in Western populations that can involve virtually
any portion of the aorta. Abdominal aortic aneurysms are much more common than thoracic aortic
aneurysms and combined they account for over 25,000 deaths in the United States annually. While
thoracic and abdominal aortic aneurysms share some common characteristics including the gross
anatomic appearance, alterations in extracellular matrix and loss of smooth muscle cells, they are
distinct diseases. In recent years, advances in genetic analysis, robust molecular tools, and
increased availability of animal models have greatly enhanced our knowledge of the
pathophysiology of aortic aneurysms. This review examines the various proposed cellular
mechanisms responsible for aortic aneurysm formation and identifies opportunities for future
Author Manuscript

studies.

Keywords
Aneurysm; Pathophysiology; Vascular Biology

Subject Terms:
Aneurysm; Cell Biology/Structural Biology; Mechanisms

Introduction
Author Manuscript

The cellular mechanisms responsible for aortic aneurysm formation constitute a complex,
orchestrated series of events that result in dramatic pathological changes in the anatomy and
function of the arterial wall. Of fundamental importance is the understanding that there are
clear and distinct differences between the mechanisms underlying thoracic and abdominal
aortic aneurysms. Thus, while the physical appearance of thoracic and abdominal aortic
aneurysms have striking similarities, the pathophysiological change of these two diseases are

Correspondence should be addressed to: W. Robert Taylor, MD, PhD, Division of Cardiology, Emory University School of Medicine,
101 Woodruff Circle, Suite 319 WMB, Atlanta GA, 30322, Phone 404-727-3754, FAX 404-727- 3858, w.robert.taylor@emory.edu.
The authors have no conflicts of interest to disclose.
Quintana and Taylor Page 2

quite distinct. This review is organized by first focusing on the mechanisms of abdominal
Author Manuscript

aortic aneurysm (AAA) formation followed by similar considerations for thoracic aortic
aneurysm (TAA) formation. Aneurysms involving other arterial beds including cerebral and
peripheral arterial aneurysms are outside of the scope of the current review. The cellular
mechanisms discussed do not include recent advances in the studies of non-coding RNAs or
the genetic basis of aortic aneurysms which are specifically discussed in different sections of
this compendium.

Abdominal Aortic Aneurysms


Abdominal aortic aneurysms (AAA) are a major cause of morbidity and mortality and it is
estimated that the incidence of AAA in men increases by 6% per decade after age 65. A
predictive modeling study based on known risk factors suggested that there may be over 1
million people in the US today with AAA.1
Author Manuscript

The epidemiology of AAA formation appears to be distinct from that of atherosclerotic


disease. One of the earlier definitive studies of risks factors for AAA was the Aneurysm
Detection and Management (ADAM) Veterans Affairs Cooperative Study Group.2 This
retrospective analysis found that a history of cigarette smoking was by far the strongest risk
factor for AAA carrying a relative risk of 5.9 when compared to non-smokers. The second
most potent risk factor was an age-independent family history of AAA with a relative risk of
1.9. Hypertension, hypercholesterolemia and pre-existing coronary artery disease carried a
relative risk of less than 1.5 suggesting that the disease mechanisms of AAA may diverge
from those of atherosclerosis.

Contributions of Different Cell Types—Apoptosis of smooth muscle cells and


degeneration of the aortic media have long been identified as hallmark of AAA pathology.3
Author Manuscript

Inflammation, production of reactive oxygen species, and ER stress have all been associated
with smooth muscle cell apoptosis in AAA.4 This loss of structural integrity is key to aortic
dilation and rupture. Of interest is that the vast majority of AAA occur below the level of the
renal arteries which may reflect the differing embryologic origins of vascular smooth muscle
cells in the distal abdominal aorta where the mesoderm gives rise to aortic vascular smooth
muscle cells whereas the thoracic aorta smooth muscle cells arise from the neural crest.5

While it is obvious that changes in vascular smooth muscle cells of the media are pivotal to
the development of AAA, many other cell types are involved in addition to smooth muscle
cells including endothelial cells,6 neutrophils,7, 8 monocyte/macrophages,9, 10 lymphocytes,
10, 11 adipocytes,12, 13 mast cells,10, 14, 15 and platelets.16 The functional contributions of

these cell types is sometimes obvious as in the case of vascular smooth muscle cells. In other
Author Manuscript

cases, studies have utilized depletion strategies in order to define their relative contributions.

The precise role of the endothelium has not yet been fully explored but, it is clear that these
cells do play a critical role in that eNOS uncoupling17 and endothelial biomechanical signal
transduction18, 19 have roles in AAA formation. In the case of platelets, while von
Willebrand factor (VWF) may not be essential for AAA formation,20 the presence of
thrombus in AAA portends a worse outcome21, 22 and suggests that there are yet to be
determined functional contributions of platelets to AAA development and rupture.

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 3

Matrix Metalloproteinases—In both AAA and TAA, the amount of elastin, collagen and
Author Manuscript

glycosaminoglycans is reduced compared to normal aortas. An imbalance between the


amount of active matrix metalloproteinases (MMPs) and their inhibitors is responsible for
most of these changes.23, 24 Thus far, 23 different MMPs have been described in humans and
are divided into archetypal, matrilysins, gelatinases and furin-activated MMPs.23 In the
normal aorta, endothelial cells, smooth muscle cells, and adventitial fibroblasts are
responsible for MMP production. In the setting of AAA, inflammatory cells serve as
additional sources of MMPs.23, 24

Increased MMP-1 (collagenase-1) expression has been described in human AAA.25, 26


Along with increased MMP-1 expression, there is a concurrent decrease in the levels of
MMP-1 inhibitors.27 However, clinical studies have not been able to correlate genetic
polymorphisms of the MMP-1 promoter region with a clinical outcome.28 MMP-1 is
produced constitutively by fibroblasts and smooth muscle cells (SMCs) in the aortic wall
Author Manuscript

and MMP-3 or plasmin are required for its activation. Inflammatory cells represent a minor
source of MMP-1.29 (18)F-FDG uptake detected by PET in the abdominal aorta is seen in
patients with symptomatic AAAs and those at high risk of rupture. Histologically, these sites
of uptake correlate with an increase in adventitial inflammatory infiltrates, a reduction in
smooth muscle cells and increased in MMP-1 and MMP-13 expression.30

MMP-13 (collagenase-3) expression is increased in human AAAs, especially in


symptomatic AAAs and those at high-risk for rupture.25, 31, 32 MMP-13 is produced
primarily by SMCs in the aortic wall. A polymorphism (−77A/G) is an independent risk
factors for AAA formation, suggesting MMP-13 plays a relevant role in AAAs.27 In mice,
nitric oxide (NO) was found to increase the production of CD147 leading to greater levels of
MMP-13 and worsening of elastase-induced AAAs. Silencing of CD147 by RNA
Author Manuscript

interference or pharmacological inhibition of iNOS lead to the inhibition of MMP-13


expression and a subsequent decrease in aortic dilation.33

MMP-3 (stromelysin-1) is also expressed at high levels in the wall of AAAs.34 This enzyme,
produced by fibroblasts and epithelial cells, appears to be produced by macrophages in the
setting of AAA.23 A MMP-3 gene promoter region polymorphism termed 5A/6A (5
adenines vs. 6 adenines at −1612) augments transcriptional activity and serves as an
independent risk factor for AAA formation.27

MMP-12 (Metalloelastase) is produced and secreted primarily by macrophages. 35 The role


of MMP-12 in the pathogenesis of AAAs is not entirely clear. Experiments in MMP-9
knockout (KO), MMP-12 KO and MMP-9/MMP-12 KO mice using the elastase model of
AAA formation revealed that MMP-12 KO mice developed AAAs (similar to wild-type
Author Manuscript

controls). However, MMP-9 KO and MMP-9/MMP-12 were resistant to AAA development.


36 Recently, experiments using the CaCl model of AAA formation in mice with a genetic
2
inactivation of PI3-kinase delta revealed a significant upregulation of MMP-12 expression
and enhanced AAA formation.37 MMP-12 co-localizes with CD68-positive macrophages in
the aneurysmal wall. In vitro, genetic inactivation of PI3-kinase delta or treatment with a
PI3-kinase delta inhibitor led to enhanced macrophage migration and increased MMP-12

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 4

expressio.. 37 While MMP-12 may not be directly involved in the pathogenesis of AAAs, it
Author Manuscript

apparently facilitates other MMPs in extracellular matrix degradation.

MMP-2 (gelatinase A) degrades e;aston and is expressed constitutively in smooth muscle


cells, fibroblasts and in macrophages.24, 38 Ang II and CaCl2 augment the activity of
MMP-2 in the abdominal and thoracic aorta.38-40 In vivo studies revealed MMP-2 systemic
KO had no impact on Ang II-induced AAAs but was protective of CaCl2-induced AAAs.
25,27

MMP-9 (Gelatinase B) is constitutively produced by fibroblasts and smooth muscle cells


and by infiltrating adventitial macrophages during AAA formation.41 MMP-9 mRNA and
protein expression in the aortic wall and MMP-9 plasma levels are significantly greater in
patients with AAAs, and higher levels are seen in patients with a luminal thrombus.40, 42, 43
MMP-9 KO mice proved to be resistant to elastase-induced AAAs.44 Infusion of wild-type
Author Manuscript

macrophages into MMP-9 KO mice led to reconstitution of AAA, suggesting macrophage-


derived MMP-9 is crucial for AAA formation.40 Recently, a genomic-wide association study
of patients with AAA identified an interaction between the MMP-9 gene expression and the
expression of genes involved in inflammation/immune function) and cholesterol metabolism.
45

MMP-14 (Membrane type 1-MMP) is produced by infiltrating macrophages and smooth


muscle cells in the aortic wall. Higher MMP-14 mRNA and protein expression levels have
been found in human AAAs.46 In murine CaCl2-induced AAAs, macrophage-derived
MMP-14 plays a crucial role in the direct degradation of the extracellular matrix in the
tunica media and adventitia leading to AAA formation. Macrophage-derived MMP-14
directly regulates macrophage elastolytic activity even in the setting of MMP-2 deletion.47
Author Manuscript

Given the pathological process of AAA formation, it is not surprising that an array of MMPs
have been implicated involving multiple cell types. It is difficult to assign central causality to
a specific MMP and cell type. Indeed, as indicated above, it appears that several MMPs are
necessary, but not sufficient to support AAA development as it is most likely that the
different cell types act with temporal and spatial specificity during AAA initiation and
growth.

The Renin-Angiotensin System—The demonstration of induction of AAA by


angiotensin II (Ang II) infusion in apoE48 and LDL49 receptor knockout mice is certainly
the most direct data in terms of documenting a causal relationship between the renin-
angiotensin system and AAA formation. Earlier reports in mice overexpressing
angiotensinogen and renin on a high salt diet first suggested this relationship as these
Author Manuscript

animals developed AAA.50 These data are consistent with additional studies showing that
angiotensin converting enzyme inhibition limited AAA formation in the elastase model.51
Thus, it appears that angiotensin II may induce AAA but, it may not be sufficient as in most
cases, additional factors (e.g., elevated cholesterol) appear to be required to induce AAA.
The source of Ang II generation is complex and several studies raise the possibility that, in
addition to angiotensin converting enzyme, chymase expressed in mast cells may contribute

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 5

to local generation of angiotensin II.15, 52 The absolute magnitude of the contribution is less
clear due to the additional role of chymase in MMP activation and apoptosis.53, 54
Author Manuscript

Angiotensin II effects on the cellular components of the aorta have been extensively
studied55–59 and include many of the cellular mechanisms described below including
production of reactive oxygen species, induction and activation of MMP’s, and infiltration of
inflammatory cells. Rather disappointingly, clinical trials have failed to demonstrate a
benefit of angiotensin converting enzyme inhibition on AAA growth rate60 and in at least
one study, may have had an adverse effect.61 The reason for this discrepancy is unclear and
may reflect deficiencies in the available animal models, effects on disease initiation vs.
progression, or the endpoint of aortic diameter.

Of note, recent data also suggest a role for the mineralocorticoid receptor in AAA formation
as aldosterone administration in non-hyperlipdemic mice was shown to induce AAA, an
Author Manuscript

effect that was age dependent with a more pronounced phenotype in older animals.62

Inflammation: Cells and Pathways—A hallmark of AAA formation is an intense


inflammatory response involving essentially all of the classic cellular constituents of
inflammation as well as local inflammatory responses in the arterial wall. Neutrophil
infiltration occurs very early on in AAAbut is transient. Observations in the elastase model
showed that treatment with neutrophil neutralizing antibodies slowed AAA expansion
suggesting a functionally relevant role for neutrophils 63, 7. Similarly, resolvins D1 and D2
have been shown to inhibit AAA formation.64, 65 This effect was initially attributed to
neutrophil-derived MMP’s. However, more recent data suggests that neutrophil extracellular
traps may be an important component of the continued inflammation in AAA.65, 66 Finally,
neutrophils are a source of reactive oxygen species as described below that can be generated
Author Manuscript

by several enzymatic sources including the NADPH oxidase and myeloperoxidase.67

Macrophage infiltration of AAA is one of the most consistent pathological findings and
certainly not surprising given the biological function of these cells and their production of
MMPs, cytokines, and chemokines as well as their ability to remove cellular debris.29, 68, 69
Interestingly, owing to their ability to express different phenotypes encompassing both
inflammatory and reparative functions, macrophages participate in both the pathogenesis of
AAA as well as the repair response.69 This can occur through classical cytokine pathway or
through more novel mechanisms. Macrophages can directly influence smooth muscle cell
function via macrophage-derived netrin-1 (a protein classified as a neuronal guidance cue
but known to promote macrophage retention in tissues) as evidenced by the finding that
deletion of netrin-1 results in a reduction in AAA formation via a MMP3-depedent
mechanism.70
Author Manuscript

Both T cells and B lymphocytes are relatively abundant in aneurysmal tissues and both cell
types have been implicated in the disease process. B cell depletion is protective against AAA
formation in both the angiotensin II and elastase models.71 The mechanism appears to
involve enhancement of a unique population of indole 2,3-dioxygenase–expressing
plasmacytoid dendritic cells and regulatory T cells with a subsequent decrease in
inflammatory gene exression.71 However, there exist additional layers of complexity in that

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 6

different B cell subtypes (B1 and B2) may play different and opposing roles in AAA.72 B2
Author Manuscript

cells appear to be the dominant subtype in AAA which is consistent with their known role in
atherosclerosis.71

T cells in general are important in the development of AAA as it has been shown that T cell
depletion attenuates AAA formation.73 Regulatory T cells play a role in protection against
AAA formation74 likely due to the fact that they secrete IL-10, an anti-inflammatory
cytokine75 as well as TGF-β, which can have a stabilizing role for AAA.76 While the precise
role of CD8+ T cells in AAA remains unclear,77 CD4+ Th1 and Th2 cells do appear to
contribute to the pathogenesis of AAA. In the setting of AAA, there are conflicting data
showing that Th1-derrived interferon gamma has a role in inducing AAA78 as well as
protecting against AAA formation.79 There are also conflicting studies as regards the ratio
of Th1 to Th2 cells.80 These differences potentially reflect different states of disease
progression or perhaps differences in animal models.
Author Manuscript

Cytokine production by both immune cells and cells native to the vessel wall ultimately
drive the inflammatory responses leading to AAA formation. As reviewed in detail
elsewhere, numerous studies have identified the involvement of multiple cytokines in AAA
formation.81 These encompass both inflammatory and anti-inflammatory cytokines. While
extremely complex and incompletely understood, suffice it to say that cytokine production
by both inflammatory cells and the resident cells of the vascular wall contribute to a
pathological feedback loop further enhancing inflammation.

Reactive Oxygen Species—Reactive Oxygen Species (ROS) play a central role in the
development of AAA. Some of the earlier observations in human tissues showed that
superoxide levels were elevated in the smooth muscle and inflammatory cells of AAA
Author Manuscript

specimens.82 Many of the known pathological effects of excess ROS including activation of
MMPs, induction of pro-inflammatory genes, and apoptosis, are key pathological features of
AAA. Animal studies have shown that administration of vitamin E as an antioxidant led to
decreases in AAA size and rupture.83 Work from our own laboratory has shown that smooth
muscle-specific overexpression of catalase prevented early mechanical changes in the aortic
wall after Ang II infusion84 as well as inhibition of AAA formation in the CaCl2 model.85

NADPH Oxidase86–88 is a major source of ROS in AAA as evidenced by increased


expression of the NADPH oxidase in segments of human aorta from patients with AAA.82
Specific deletion of the p47 subunit of NADPH oxidase resulted in attenuation of AAA in
two different models.17, 89 The NADPH oxidase system is complex and is composed of a
family of NOX homologs identified by their catalytic subunits (NOX1, NOX2, NOX3,
NOX4, NOX5, DUOX1, DUOX2). The different homologs exhibit varied tissue expression
Author Manuscript

and produce superoxide anion and/or hydrogen peroxide. There exists some controversy as
regards the precise role of the various NOX homologs in AAA. Siu et al17 reported that
deficiency of NOX1, NOX 2 or NOX 4 as well as deletion of the non-catalytic p47 subunit
resulted in complete inhibition of AAA. However, Kigawa et al90 showed that NOX2
deficiency resulted in increased diameter and extent of AAA that was associated with overall
decreased ROS production and polarization of macrophages towards aninflammatory
phenotype. The reason for this discrepancy may be due to the models used, potential

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 7

compensatory upregulation of other NOX homologs, or cell-specific effects. The above


Author Manuscript

studies allow one to conclude that, even though the involvement of the specific NOX
homologs is unclear, there is an abundance of evidence supporting the role of NADPH
oxidase in AAA formation.

Uncoupled endothelial nitric oxide synthase (eNOS) has been proposed to be a contributor
to ROS production in a variety of vascular diseases processes in general91 and specifically in
AAA.17, 92–94 AAA occur after angiotensin II infusion in hph-1 mice95 which have impaired
activity of GTP cyclohydrolase leading to a deficiency in tetrahydrobiopterin which in turn
causes uncoupling of eNOS leading to production of superoxide. Importantly, folic acid
administration was shown to prevent AAA formation. Ang II infusion in apoE mice leads to
decreased tissue and circulating levels of tetrahydrobiopterin,96 an effect likely due to the
degradation of tetrahydrobiopterin by oxidative stress. These data suggest that ROS derived
from uncoupled eNOS also contribute to AAA formation and that eNOS uncoupling occurs
Author Manuscript

as a result of superoxide production by NADPH oxidases. In essence, uncoupled eNOS can


act to amplify the effects of NADPH oxidases.

Myeloperoxidase may also contribute to ROS generation in AAAas neutrophil infiltration of


the abdominal aorta occurs early on in experimental models. Neutrophil depletion inhibits
AAA formation,7 an effect which likely has multiple mechanistic impacts given the
functional contributions of neutrophils. However, neutrophils are the primary source of
myeloperoxidase, accounting for up to 5% of the total protein content of neutrophils.97
Genetic deletion of myeloperoxidase or inhibition of myeloperoxidase by administering
taurine inhibits AAA formation, confirming the importance of this enzyme in AAA
formation.67 Hypochlorous acid (a product of myeloperoxidase) is highly reactive with
many biomolecules including lipids. Subsequent lipid oxidation may be one the key steps in
Author Manuscript

oxidative damage in the setting of AAA as upregulation of serum amyloid A, a major target
of lipid oxidation, occurs in AAA and genetic depletion of serum amyloid A also results in
inhibition of AAA formation.98

Other sources of reactive oxygen species including xanthine oxidase, iNOS,


cyclooxygenase, and mitochondrial metabolism may also play a role in the pathogenesis of
AAA however, studies from human AAA samples suggest that the contributions of xanthine
oxidase and mitochondrial respiration may be less important.99 Deletion of mitochondrial
uncoupling protein-2 (UCP-2) resulted in increased incidence of AAA in the Ang II model
of AAA though whether this a direct effect on production of ROS by mitochondria or a
secondary effect through regulation of other pro- and antioxidant enzyme systems is unclear.
100
Author Manuscript

Dysregulation of antioxidant systems may also determine local and systemic levels of ROS.
Catalase expression has been shown to be decreased in aortic tissue in animal models of
AAA84 as well as in circulating polymorphonuclear neutrophils and plasma obtained from
patients with AAA.101 Studies of the potential involvement of superoxide dismutase (SOD)
have shown both increased and decreased activity in AAA samples from patients102,103
Specific analysis of SOD isoform expression showed increases only in MnSOD

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 8

expression104 with another study reporting a decrease in ecSOD.105 Glutathione


peroxidase103 and paraoxinase-1106 also appear to be decreased in human AAA specimens.
Author Manuscript

The precise mechanisms through which ROS elicit cellular responses has been reviewed
previously107–111 and remains an active area of study that is well beyond the scope of this
review. It is clear that dysregulation of ROS leads to widespread changes in the arterial wall
that are central in the process of AAA development. The result being that increased levels of
reactive oxygen species lead to increased expression of pro-inflammatory gene products,
smooth muscle apoptosis, increased expression and activation of MMPs, and other key
events in the pathogenesis of AAA.

Risk factors and mechanisms—The strongest risk factors for AAA are male sex,
family history, and cigarette smoking. How these risk factors impact the known cellular
mechanisms of AAA is key to our understanding of the disease. The overall rate at which
Author Manuscript

AAA affects men is higher than women at ratio of 5:1.112 In both the elastase113 and the
angiotensin II models of AAA114, male mice exhibit a higher incidence and larger AAAs
when compared to females. A very striking gender-based difference in the disease is its
progression over time. A population based study examining the incidence of AAA in men
and women as a function of age showed that in the 40–60 year age group (pre- and early
post-menopausal women), the rate of AAA was eleven times higher in men than women. In
the 60–90 year group this difference falls to three times, and by age 90 men and women have
AAA at an equivalent rate.115

There is not a single, causal mechanism responsible for the sex-dependent differences in the
incidence of AAA. As recently reviewed116 there are many clinical and pre-clinical studies
examining the role of sex hormones on AAA demonstrating complex and sometimes
Author Manuscript

paradoxical effects. While endogenous estradiol is likely protective, exogenous estrogen


replacement has shown conflicting results in human studies with both increases117 and
decreases118 in AAA events in women receiving hormone replacement therapy. Conversely,
androgen signaling increases production of reactive oxygen species, expression of
angiotensin II receptors,119 inflammation in the arterial wall116 and counteracts the effects
of exogenous estrogen.120 Very interestingly, specific loci on the Y chromosome have been
linked to activation of the renin angiotensin system and subsequent AAA formation.114, 121
These data suggest that androgen-mediated events are the primary drivers of sex differences
in AAA. However, in human studies, low testosterone levels were shown to be
independently associated with the presence of AAA.122 Whether this discrepancy is due to
differences in disease state or is related to lack of translation of animal models to the human
disease state is unclear.
Author Manuscript

Work from our laboratory has suggested that local difference in vascular hemodynamics may
also contribute to the sex differences in AAA in that local shear stress patterns differ
between males and females as a result of differences in abdominal blood flow patterns to the
reproductive organs.19 In addition, mechanical properties of the aorta exhibit sex-dependent
differences that are also age-dependent.123–126 This may be due to altered extracellular
matrix content126 and cross linking of the extracellular matrix which ultimately reflect
androgen-mediated physiology as described above. The downstream effectors of differential

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 9

androgen and estrogen signaling involve essentially all of the same cellular mechanisms of
AAA as described above with reported differences in production of ROS,127 inflammatory
Author Manuscript

mediators, extracellular matrix composition,128 and inflammatory cells.

The mechanisms of other risk factors is less clear. Genetic factors are discussed in a separate
section of this compendium. Cigarette smoking is a complex stimulus and there are likely
multiple components in cigarette smoke that promote vascular disease. It has been shown
that cigarette smoke extract induces expression of MMP-2 as well as MMP-9 and that
nicotine, through increased expression of MMP-2 induces AAA.129, 130 The mechanisms of
other, protective risk factors including diabetes and race remain unknown.

Thoracic Aortic Aneurysms


The prevalence of TAA is less than that of abdominal aortic aneurysms but they still occur in
up to 6–10 people per 100,000. Approximately 20% of TAA can be attributed to a specific
Author Manuscript

genetic variant. The differences between TAA and AAA reflects several fundamental
differences in their pathophysiology as exemplified by the fact that multiple genetic variants
have been identified for TAA which do not confer increased risk for AAA. While there is
clear evidence for increased risk for AAA among family members of individuals with AAA
and clusters of AAA within families, there is surprisingly little overlap between AAA and
TAA.

TAAs can be broadly categorized as syndromic, familial non-syndromic, and sporadic with
the latter category being quite diverse. This heterogeneity of TAA complicates mechanistic
studies using human samples and animal models as TAA is a multi-factorial disease with
multiple unique mechanisms resulting in similar clinical presentations. The classic
syndromic TAAs include Marfan, Loeys Dietz, Ehlers-Danlos, etc., while the familial non-
Author Manuscript

syndromic TAAs encompass a wide variety of genetic variants (see the section of this
compendium on the genetics of aneurysms). Overlapping this construct is TAA associated
with a bicuspid aortic valve as a bicuspid valve occurs at higher rates in some specific
syndromes (e.g., Marfan Syndrome) and also in the general population where a single
genetic variant is not present. 131

Much of the fundamental science of TAA stems from genetic studies and observations made
in mouse models that focused on alterations in extracellular matrix components, smooth
muscle function, and related proteins. The volume of data related to mechanisms of TAA is
less than that for AAA, likely reflecting differences in the ease and applicability of small
animal models for AAA vs. TAA. However, significant insights into the mechanisms of this
disease have been obtained over the past several decades and we now more fully appreciate
Author Manuscript

that TAAs represent a spectrum of disease pathologies that are the result of complex changes
in the cellular and extracellular environment and not a simple degenerative process.

A central role of the Extracellular Matrix—As is the case for AAA, TAA is
characterized in part by abnormalities in the extracellular matrix that compromise the
structural integrity of the aorta. Studies have implicated several genetic variants in proteins
that directly impact the mechanical characteristics of the aorta leading to TAA. Among these
is lysl oxidase, which is responsible for cross linking collagen and elastin. Genetic data are

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 10

supported by experimental studies in which administration of beta-aminopropionitrile (a lysl


Author Manuscript

oxidase inhibitor) to mice in the setting of angiotensin infusion leads to aneurysmal dilation
of the aorta at multiple locations.132 Interestingly, beta-aminopropionitrile is naturally
occurring in sweet peas133 and either feeding experimental animals sweet peas133 or farm
animals eating sweet peas results in aortic aneurysms among other manifestations related to
loss of collagen and elastin structure. It is important to note that the effects of beta-
aminopropionitrile are not specific to the thoracic aorta suggesting that other factors are
likely involved. In addition, it is unknown if genetic variants in other members of the lysl
oxidase family contribute to TAA formation.

Some forms of Ehlers-Danlos Syndrome are associated with TAA and aortic dissection.
Ehlers-Danlos Syndrome encompasses a group of diseases due to mutations in collagen
genes. The vascular phenotype variant of Ehlers-Danlos (type IV) is due to one of several
mutations in the type III pro-collagen gene and is associated with TAA and other vascular
Author Manuscript

abnormalities.134 The formation of aneurysm and dissection in affected individuals is


presumably due to the compromise of the mechanical integrity of the wall.

One of the initial, major contributions to our understanding of TAA came from observations
related to Marfan Syndrome with the finding there was a mutation in the fibrillin 1 gene.135
Subsequently, additional mutations in fibrillin 1 have been identified. Fibrillin 1 is an
extracellular matrix glycoprotein that serves as the major structural component of
microfibrils in the aortic wall and while the structural contributions of this protein to the
arterial wall may be important, Neptune and colleagues proposed that the mutations in
fibrillin 1 actually functioned through dysregulation of TGF-β signalling.136 The model that
emerged was that fibrillin-1 binds to the large latent TGF binding protein that sequesters
TGF-β in a complex and prevents it from becoming active.137–141 Subsequent work has also
Author Manuscript

implicated variants in the large latent TGF binding protein.142 In Marfan syndrome, it was
proposed that the mutations in fibrillin 1 led to increased availability of TGF-β and
subsequent activation of TGF-β mediated signaling pathways.141 With this model,
transcriptional regulation occurs through pathways that are both Smad-dependent and -
independent.143 Animal studies performed with either mutated or knocked out fibrillin-1
mimicked Marfan Syndrome. While there are reports of increased TGF-β in aortic tissue of
Marfan patients, other reports demonstrated increased total TGF-β but no change in active
TGF-β despite increased expression of total TGF-β and activation of Smad2.144
Furthermore, while Smad2 is a downstream effector of TGF-β, other signaling pathways
also converge on Smad2. Recent publications using a Marfan mouse model with mutated
fibrillin-1 demonstrated that genetic disruption of TGF-β receptor increased aortic dilatation
suggesting a protective effect of TGF-β signaling as opposed to a causal role.145, 146 Thus,
Author Manuscript

the precise role of TGF-β signaling in Marfan Syndrome remains unclear and is a source of
significant controversy.143, 145, 147 Fibrillin-1 is clearly important and may have alternative
mechanisms of action related to its structural contributions, integrin binding capacity,
involvement in sequestering other growth factors, or other, as yet undetermined functions.

Additional variants in the structural components of the arterial wall have been linked to TAA
are reviewed in detail in the section of this compendium devoted to the genetic basis of
aneurysmal diseases. There is clearly a significant contribution of variants in the structural

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 11

proteins of the aortic wall to the pathogenesis of TAA in both syndromic and sporadic
Author Manuscript

disease.

Contributions from Smooth Muscle Cells—Smooth muscle cells are the major
cellular constituent of the aorta and their loss through apoptosis or necroptosis is a major
defining feature of both AAA and TAA. Genetic studies have shown that several mutations
in the contractile proteins of smooth muscle cells predispose individuals to TAA. This
suggests that smooth muscle contractile function plays an important role in TAA though the
mechanism is unclear. Possibilities include both a structural, load bearing function or a
signal mechanical signal transduction function as proposed in the section of this
compendium devoted to the genetic basis of aortic aneurysms.

Matrix Metalloproteinases—As is the case with AAA, matrix metalloproteinases play a


pivotal role in the remodeling of TAA but they are far less studied. MMPs are potentially
Author Manuscript

involved in both the maladaptive degradation of the extracellular matrix as well as the
adaptive remodeling of extracellular matrix to stabilize an enlarging aorta. The largest body
of data related to TAA involve MMP-2 where the results are somewhat paradoxical. In
contrast to AAA, MMP-2 deletion increased Ang II-induced TAA formation but was also
protective of CaCl2-induced TAAs.36 A potential explanation for these findings is that there
is a higher content of elastin in the thoracic aorta, making it more susceptible to extracellular
matrix synthesis alterations whereas the abdominal aorta more susceptible to excess
extracellular matrix degradation. Systemic deletion of the MMP-2 gene in a mouse model of
Marfan Syndrome delayed TAA rupture, inhibited the activation of TGF-β and
phosphorylation of ERK1/2 and SMAD2, preserved aortic lamellar integrity, and prolonged
mice survival. MMP-9 protein and activity levels in the aortic wall are increased in the
setting of TAA.148, 149 An ex vivo study revealed that after Ang II administration, there is
Author Manuscript

SMAD2 activation, increased MMP-9 expression via ERK, and subsequent extracellular
matrix degradation.148 However, in TAAs associated with bicuspid aortic valves, MMP-9
expression was not augmented.149 In addition, MMP-3 may play a role in the development
of human ascending TAAs (4.0- to 5.9-cm size range), especially in older patients.150

Inflammation—Inflammation is a cardinal feature of AAA but the data supporting a role


for inflammation in TAA are less extensive. There are intriguing data suggesting that there is
some commonality between AAA and TAA and that the structural changes described above
may lead to increased production of reactive oxygen species and inflammatory responses.
Both T cells and macrophages are present in the media of TAA151 a finding that is supported
by genomic analysis of human TAA samples which revealed upregulation of multiple
inflammatory pathways.152 T cells may be a source of FAS ligand that results in smooth
Author Manuscript

muscle apoptosis, a cardinal feature of TAA. Specific mediators of inflammation in TAA are
not well defined. One study reported elevated IL-6 levels in TAA relative to both normal
tissue and tissue from AAA.153 Furthermore, inhibition of IL-6 signaling inhibited aneurysm
growth in the murine elastase model of TAA.153 Similar data from the same group were
obtained with regard to IL-1β.154 Thus, while there is evidence of inflammation in TAA and
plausible mechanisms for the induction of inflammation in TAA, the data set currently
available is somewhat limited.

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 12

Reactive Oxygen Species—The role of ROS in TAA has received less attention as
Author Manuscript

compared to studies of AAA. Aortic tissue from patients with Marfan syndrome have
increased levels of oxidative stress, decreased expression of antioxidant enzymes, and
increased expression of iNOS, xanthine oxidase, and NADPH oxidase subunits.155 Similarly
superoxide dismutase 3 expression in the ascending aorta is downregulated in patients with
bicuspid aortic valves. Smooth muscle cells from the aortas of patients with Marfan
Syndrome have increased expression of NOX4 and enhanced production of H2O2.156 More
mechanistic data have been obtained in models of Marfan and Loeys-Dietz syndrome where
altered mitochondrial respiration and increased production of ROS have been reported.157

Risk factors and mechanisms—The familial risk of TAA is discussed in a separate


section of this compendium. For sporadic TAA, hypertension, cigarette smoking, and
atherosclerotic disease are the strongest risk factors. The mechanisms behind these risk
factors is unclear is generally unclear except in the case of atherosclerosis which shares
Author Manuscript

many common disease mechanisms. Hypertension can impact the vessel wall through direct
mechanical effects on inflammation or through humoral stimuli. Female sex and diabetes are
also protective in TAA but the mechanisms are unknown.

Discussion
Perhaps the major conclusion to be derived from the evolving literature defining the cellular
and molecular mechanisms of thoracic and abdominal aortic aneurysms is that while
thoracic and abdominal aortic aneurysms have striking similarities at the gross anatomical
level, the underlying pathophysiology has quite distinct differences. AAA and TAA are best
considered as different disease processes with inflammation as hallmark of AAA formation
and distinct alterations in extracellular matrix formation perhaps being a common feature of
Author Manuscript

TAA. However, we must be cautious about such sweeping generalizations as they may be
driven by the experimental approaches used to study these diseases and in essence, be self-
fulfilling prophesies because of these approaches.

The vast majority of the studies of AAA have been fueled by the emergence of several
relatively simple models. While informative that angiotensin II can induce AAA in the
appropriate settings, the extensive use of this model raises the question; Does the fact that
angiotensin II is a potent pro-inflammatory stimulus inform us about the fundamental
pathophysiology of AAA or does it constrain us to an exclusive focus on inflammatory
mechanisms? Similarly, in the case of TAA, much of the mechanistic work has been driven
by genetic models that are often derived from human disease where unique genetic
mutations have been identified. While extraordinarily informative, this approach is also
Author Manuscript

somewhat limited in terms of the generalizability to the greater population of individuals


with sporadic TAA. Indeed, the relatively limited number of more “generic” animal models
of TAA (i.e., those that are not based on a specific genetic mutation) may be a limiting factor
in the study of TAA disease mechanisms. As discussed in other sections of this
compendium, clinical trials in humans using pharmacological approaches sometimes fail to
produce the predicted effects which is likely a consequence of the limitations of available
animal models.

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 13

It is also important to consider the potential differences between aneurysm initiation,


Author Manuscript

growth, and rupture. Much of the animal work has focused on the mechanisms responsible
for the initiation of aneurysms and while these studies have provided extremely valuable
insights into the potential pathogenesis of aortic aneurysms, they cannot help to define the
entire disease process and in some instances, they can diverge from the normal progression
of human disease. There is clear opportunity for additional studies to help us better
understand the mechanisms of aortic rupture and dissection as they relate to both AAA and
TAA.Human studies must also be placed into the correct context both in terms of disease
state and anatomic location. As the majority of human samples are obtained at the time of
surgery, they represent more advanced disease. For TAA, the heterogeneity of the disease is
also an important consideration as while syndromic, non-syndromic and sporadic TAA share
some common features, it would be naïve to think that they share a single, common cellular
disease mechanism.
Author Manuscript

The past two decades have seen an enormous growth in our understanding of the pathology
of both thoracic and abdominal aortic aneurysm formation. While the entirety of the field
cannot be summarized in a single review, we have attempted to define some of the key
themes that have emerged from this growing body of work. Much of the experimental work
to date has focused on cellular and molecular events that occur early in the disease process
leaving much to be learned about the more chronic events in aneurysm pathology. In
addition, we do not yet fully understand how various risk factors like cigarette smoking,
diabetes, and race impact these disease mechanisms. The mere fact that there are no specific,
mechanistically based curative therapies for patients with aortic aneurysms tells us that we
have much to learn about the cellular mechanisms of both thoracic and abdominal aortic
aneurysms.
Author Manuscript

Acknowledgments
This work was supported by a grant from the National Institutes of Health (PO1 HL095070).

Non-standard Abbreviations and Acronyms


AAA Abdominal Aortic Aneurysm

Ang II Angiotensin II

eNOS Endothelial Nitric Oxide Synthase

FDG Fluorodeoxyglucose

IL Interleukin
Author Manuscript

IFN Interferon

LDLR Low Density Lipoprotein Receptor

MMP Matrix Metalloproteinase

PI3-kinase Phosphatidylinositol-4,5-bisphosphate 3-kinase delta

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 14

PET Positron Emission Tomography


Author Manuscript

ROS Reactive Oxygen Species

SMC Smooth Muscle Cell

TAA Thoracic Abdominal Aortic Aneurysm

TGF-β Transforming Growth Factor Beta

Citations
1. Kent KC, Zwolak RM, Egorova NN, Riles TS, Manganaro A, Moskowitz AJ, Gelijns AC and Greco
G. Analysis of risk factors for abdominal aortic aneurysm in a cohort of more than 3 million
individuals. J Vasc Surg. 2010;52:539–48. [PubMed: 20630687]
2. Lederle FA, Johnson GR, Wilson SE, Chute EP, Littooy FN, Bandyk D, Krupski WC, Barone GW,
Author Manuscript

Acher CW and Ballard DJ. Prevalence and associations of abdominal aortic aneurysm detected
through screening. Aneurysm Detection and Management (ADAM) Veterans Affairs Cooperative
Study Group. Ann Intern Med. 1997;126:441–9. [PubMed: 9072929]
3. Lopez-Candales A, Holmes DR, Liao S, Scott MJ, Wickline SA and Thompson RW. Decreased
vascular smooth muscle cell density in medial degeneration of human abdominal aortic aneurysms.
Am J Pathol. 1997;150:993–1007. [PubMed: 9060837]
4. Qin Y, Wang Y, Liu O, Jia L, Fang W, Du J and Wei Y. Tauroursodeoxycholic Acid Attenuates
Angiotensin II Induced Abdominal Aortic Aneurysm Formation in Apolipoprotein E-deficient Mice
by Inhibiting Endoplasmic Reticulum Stress. Eur J Vasc Endovasc Surg. 2017;53:337–345.
[PubMed: 27889204]
5. Tromp G, Kuivaniemi H, Hinterseher I and Carey DJ. Novel genetic mechanisms for aortic
aneurysms. Curr Atheroscler Rep. 2010;12:259–66. [PubMed: 20446064]
6. Sun J, Deng H, Zhou Z, Xiong X and Gao L. Endothelium as a Potential Target for Treatment of
Abdominal Aortic Aneurysm. Oxid Med Cell Longev. 2018;2018:6306542. [PubMed: 29849906]
7. Eliason JL, Hannawa KK, Ailawadi G, Sinha I, Ford JW, Deogracias MP, Roelofs KJ, Woodrum DT,
Author Manuscript

Ennis TL, Henke PK, Stanley JC, Thompson RW and Upchurch GR, Jr. Neutrophil depletion
inhibits experimental abdominal aortic aneurysm formation. Circulation. 2005;112:232–40.
[PubMed: 16009808]
8. Hannawa KK, Eliason JL, Woodrum DT, Pearce CG, Roelofs KJ, Grigoryants V, Eagleton MJ,
Henke PK, Wakefield TW, Myers DD, Stanley JC and Upchurch GR, Jr. L-selectin-mediated
neutrophil recruitment in experimental rodent aneurysm formation. Circulation. 2005;112:241–7.
[PubMed: 15998669]
9. Rateri DL, Howatt DA, Moorleghen JJ, Charnigo R, Cassis LA and Daugherty A. Prolonged
infusion of angiotensin II in apoE(−/−) mice promotes macrophage recruitment with continued
expansion of abdominal aortic aneurysm. Am J Pathol. 2011;179:1542–8. [PubMed: 21763672]
10. Wang J, Lindholt JS, Sukhova GK, Shi MA, Xia M, Chen H, Xiang M, He A, Wang Y, Xiong N,
Libby P, Wang JA and Shi GP. IgE actions on CD4+ T cells, mast cells, and macrophages
participate in the pathogenesis of experimental abdominal aortic aneurysms. EMBO Mol Med.
2014;6:952–69. [PubMed: 24963147]
Author Manuscript

11. Zhang L and Wang Y. B lymphocytes in abdominal aortic aneurysms. Atherosclerosis.


2015;242:311–7. [PubMed: 26233918]
12. Kugo H, Tanaka H, Moriyama T and Zaima N. Pathological Implication of Adipocytes in AAA
Development and the Rupture. Ann Vasc Dis. 2018;11:159–168. [PubMed: 30116407]
13. Police SB, Thatcher SE, Charnigo R, Daugherty A and Cassis LA. Obesity promotes inflammation
in periaortic adipose tissue and angiotensin II-induced abdominal aortic aneurysm formation.
Arterioscler Thromb Vasc Biol. 2009;29:1458–64. [PubMed: 19608970]
14. Tsuruda T, Kato J, Hatakeyama K, Kojima K, Yano M, Yano Y, Nakamura K, Nakamura-
Uchiyama F, Matsushima Y, Imamura T, Onitsuka T, Asada Y, Nawa Y, Eto T and Kitamura K.

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 15

Adventitial mast cells contribute to pathogenesis in the progression of abdominal aortic aneurysm.
Circ Res. 2008;102:1368–77. [PubMed: 18451339]
Author Manuscript

15. Sun J, Zhang J, Lindholt JS, Sukhova GK, Liu J, He A, Abrink M, Pejler G, Stevens RL,
Thompson RW, Ennis TL, Gurish MF, Libby P and Shi GP. Critical role of mast cell chymase in
mouse abdominal aortic aneurysm formation. Circulation. 2009;120:973–82. [PubMed: 19720934]
16. Martinez-Pinna R, Madrigal-Matute J, Tarin C, Burillo E, Esteban-Salan M, Pastor-Vargas C,
Lindholt JS, Lopez JA, Calvo E, de Ceniga MV, Meilhac O, Egido J, Blanco-Colio LM, Michel JB
and Martin-Ventura JL. Proteomic analysis of intraluminal thrombus highlights complement
activation in human abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol. 2013;33:2013–
20. [PubMed: 23702661]
17. Siu KL, Miao XN and 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. [PubMed: 24558445]
18. Arzani A and Shadden SC. Characterizations and Correlations of Wall Shear Stress in Aneurysmal
Flow. J Biomech Eng. 2016;138.
19. Taylor WR, Iffrig E, Veneziani A, Oshinski JN and Smolensky A. Sex and Vascular Biomechanics:
Author Manuscript

A Hypothesis for the Mechanism Underlying Differences in the Prevalence of Abdominal Aortic
Aneurysms in Men and Women. Trans Am Clin Climatol Assoc. 2016;127:148–161. [PubMed:
28066050]
20. Portier I, Martinod K, Desender L, Vandeputte N, Deckmyn H, Vanhoorelbeke K and De Meyer
SF. von Willebrand factor deficiency does not influence angiotensin II-induced abdominal aortic
aneurysm formation in mice. Sci Rep. 2018;8:16645. [PubMed: 30413751]
21. Zambrano BA, Gharahi H, Lim C, Jaberi FA, Choi J, Lee W and Baek S. Association of
Intraluminal Thrombus, Hemodynamic Forces, and Abdominal Aortic Aneurysm Expansion Using
Longitudinal CT Images. Ann Biomed Eng. 2016;44:1502–14. [PubMed: 26429788]
22. Haller SJ, Crawford JD, Courchaine KM, Bohannan CJ, Landry GJ, Moneta GL, Azarbal AF and
Rugonyi S. Intraluminal thrombus is associated with early rupture of abdominal aortic aneurysm. J
Vasc Surg. 2018;67:1051–1058 e1. [PubMed: 29141786]
23. Fanjul-Fernandez M, Folgueras AR, Cabrera S and Lopez-Otin C. Matrix metalloproteinases:
evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta.
2010;1803:3–19. [PubMed: 19631700]
Author Manuscript

24. Kadoglou NP and Liapis CD. Matrix metalloproteinases: contribution to pathogenesis, diagnosis,
surveillance and treatment of abdominal aortic aneurysms. Curr Med Res Opin. 2004;20:419–32.
[PubMed: 15119978]
25. Knox JB, Sukhova GK, Whittemore AD and Libby P. Evidence for altered balance between matrix
metalloproteinases and their inhibitors in human aortic diseases. Circulation. 1997;95:205–12.
[PubMed: 8994438]
26. Verschuren L, Lindeman JH, van Bockel JH, Abdul-Hussien H, Kooistra T and Kleemann R. Up-
regulation and coexpression of MIF and matrix metalloproteinases in human abdominal aortic
aneurysms. Antioxid Redox Signal. 2005;7:1195–202. [PubMed: 16115023]
27. Saracini C, Bolli P, Sticchi E, Pratesi G, Pulli R, Sofi F, Pratesi C, Gensini GF, Abbate R and Giusti
B. Polymorphisms of genes involved in extracellular matrix remodeling and abdominal aortic
aneurysm. J Vasc Surg. 2012;55:171–179 e2. [PubMed: 22094117]
28. Saratzis A, Abbas AA, Kiskinis D, Melas N, Saratzis N and Kitas GD. Abdominal aortic
aneurysm: a review of the genetic basis. Angiology. 2011;62:18–32. [PubMed: 20566578]
Author Manuscript

29. Newman KM, Jean-Claude J, Li H, Scholes JV, Ogata Y, Nagase H and Tilson MD. Cellular
localization of matrix metalloproteinases in the abdominal aortic aneurysm wall. J Vasc Surg.
1994;20:814–20. [PubMed: 7526009]
30. Courtois A, Nusgens BV, Hustinx R, Namur G, Gomez P, Somja J, Defraigne JO, Delvenne P,
Michel JB, Colige AC and Sakalihasan N. 18F-FDG uptake assessed by PET/CT in abdominal
aortic aneurysms is associated with cellular and molecular alterations prefacing wall deterioration
and rupture. J Nucl Med. 2013;54:1740–7. [PubMed: 24009278]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 16

31. Mao D, Lee JK, VanVickle SJ and Thompson RW. Expression of collagenase-3 (MMP-13) in
human abdominal aortic aneurysms and vascular smooth muscle cells in culture. Biochem Biophys
Author Manuscript

Res Commun. 1999;261:904–10. [PubMed: 10441523]


32. Tromp G, Gatalica Z, Skunca M, Berguer R, Siegel T, Kline RA and Kuivaniemi H. Elevated
expression of matrix metalloproteinase-13 in abdominal aortic aneurysms. Ann Vasc Surg.
2004;18:414–20. [PubMed: 15156361]
33. Lizarbe TR, Tarin C, Gomez M, Lavin B, Aracil E, Orte LM and Zaragoza C. Nitric oxide induces
the progression of abdominal aortic aneurysms through the matrix metalloproteinase inducer
EMMPRIN. Am J Pathol. 2009;175:1421–30. [PubMed: 19779140]
34. Carrell TW, Burnand KG, Wells GM, Clements JM and Smith A. Stromelysin-1 (matrix
metalloproteinase-3) and tissue inhibitor of metalloproteinase-3 are overexpressed in the wall of
abdominal aortic aneurysms. Circulation. 2002;105:477–82. [PubMed: 11815431]
35. Johnson JL. Metalloproteinases in atherosclerosis. Eur J Pharmacol. 2017;816:93–106. [PubMed:
28893577]
36. Pyo R, Lee JK, Shipley JM, Curci JA, Mao D, Ziporin SJ, Ennis TL, Shapiro SD, Senior RM and
Thompson RW. Targeted gene disruption of matrix metalloproteinase-9 (gelatinase B) suppresses
Author Manuscript

development of experimental abdominal aortic aneurysms. J Clin Invest. 2000;105:1641–9.


[PubMed: 10841523]
37. Zheng L, Xing L, Zeng C, Wu T, Gui Y, Li W, Lan T, Yang Y, Gu Q, Qi C, Zhang Q, Tang F, He X
and Wang L. Inactivation of PI3Kdelta induces vascular injury and promotes aneurysm
development by upregulating the AP-1/MMP-12 pathway in macrophages. Arterioscler Thromb
Vasc Biol. 2015;35:368–77. [PubMed: 25503990]
38. Lu H, Rateri DL, Bruemmer D, Cassis LA and Daugherty A. Novel mechanisms of abdominal
aortic aneurysms. Curr Atheroscler Rep. 2012;14:402–12. [PubMed: 22833280]
39. Shen M, Lee J, Basu R, Sakamuri SS, Wang X, Fan D and Kassiri Z. Divergent roles of matrix
metalloproteinase 2 in pathogenesis of thoracic aortic aneurysm. Arterioscler Thromb Vasc Biol.
2015;35:888–98. [PubMed: 25657308]
40. Longo GM, Xiong W, Greiner TC, Zhao Y, Fiotti N and Baxter BT. Matrix metalloproteinases 2
and 9 work in concert to produce aortic aneurysms. J Clin Invest. 2002;110:625–32. [PubMed:
12208863]
41. Thompson RW, Holmes DR, Mertens RA, Liao S, Botney MD, Mecham RP, Welgus HG and Parks
Author Manuscript

WC. Production and localization of 92-kilodalton gelatinase in abdominal aortic aneurysms. An


elastolytic metalloproteinase expressed by aneurysm-infiltrating macrophages. J Clin Invest.
1995;96:318–26. [PubMed: 7615801]
42. Baxter BT. Could medical intervention work for aortic aneurysms? Am J Surg. 2004;188:628–32.
[PubMed: 15619475]
43. Fontaine V, Jacob MP, Houard X, Rossignol P, Plissonnier D, Angles-Cano E and Michel JB.
Involvement of the mural thrombus as a site of protease release and activation in human aortic
aneurysms. Am J Pathol. 2002;161:1701–10. [PubMed: 12414517]
44. Visse R and Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases:
structure, function, and biochemistry. Circ Res. 2003;92:827–39. [PubMed: 12730128]
45. Jones GT, Tromp G, Kuivaniemi H, Gretarsdottir S, Baas AF, Giusti B, Strauss E, Van’t Hof FN,
Webb TR, Erdman R, Ritchie MD, Elmore JR, Verma A, Pendergrass S, Kullo IJ, Ye Z, Peissig
PL, Gottesman O, Verma SS, Malinowski J, Rasmussen-Torvik LJ, Borthwick KM, Smelser DT,
Crosslin DR, de Andrade M, Ryer EJ, McCarty CA, Bottinger EP, Pacheco JA, Crawford DC,
Author Manuscript

Carrell DS, Gerhard GS, Franklin DP, Carey DJ, Phillips VL, Williams MJ, Wei W, Blair R, Hill
AA, Vasudevan TM, Lewis DR, Thomson IA, Krysa J, Hill GB, Roake J, Merriman TR, Oszkinis
G, Galora S, Saracini C, Abbate R, Pulli R, Pratesi C, Saratzis A, Verissimo AR, Bumpstead S,
Badger SA, Clough RE, Cockerill G, Hafez H, Scott DJ, Futers TS, Romaine SP, Bridge K, Griffin
KJ, Bailey MA, Smith A, Thompson MM, van Bockxmeer FM, Matthiasson SE, Thorleifsson G,
Thorsteinsdottir U, Blankensteijn JD, Teijink JA, Wijmenga C, de Graaf J, Kiemeney LA, Lindholt
JS, Hughes A, Bradley DT, Stirrups K, Golledge J, Norman PE, Powell JT, Humphries SE, Hamby
SE, Goodall AH, Nelson CP, Sakalihasan N, Courtois A, Ferrell RE, Eriksson P, Folkersen L,
Franco-Cereceda A, Eicher JD, Johnson AD, Betsholtz C, Ruusalepp A, Franzen O, Schadt EE,
Bjorkegren JL, Lipovich L, Drolet AM, Verhoeven EL, Zeebregts CJ, Geelkerken RH, van

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 17

Sambeek MR, van Sterkenburg SM, de Vries JP, Stefansson K, Thompson JR, de Bakker PI,
Deloukas P, Sayers RD, Harrison SC, van Rij AM, Samani NJ and Bown MJ. Meta-Analysis of
Author Manuscript

Genome-Wide Association Studies for Abdominal Aortic Aneurysm Identifies Four New Disease-
Specific Risk Loci. Circ Res. 2017;120:341–353. [PubMed: 27899403]
46. Nollendorfs A, Greiner TC, Nagase H and Baxter BT. The expression and localization of
membrane type-1 matrix metalloproteinase in human abdominal aortic aneurysms. J Vasc Surg.
2001;34:316–22. [PubMed: 11496285]
47. Xiong W, Knispel R, MacTaggart J, Greiner TC, Weiss SJ and Baxter BT. Membrane-type 1 matrix
metalloproteinase regulates macrophage-dependent elastolytic activity and aneurysm formation in
vivo. J Biol Chem. 2009;284:1765–71. [PubMed: 19010778]
48. Daugherty A, Manning MW and Cassis LA. Angiotensin II promotes atherosclerotic lesions and
aneurysms in apolipoprotein E-deficient mice. J Clin Invest. 2000;105:1605–12. [PubMed:
10841519]
49. Daugherty A and Cassis L. Chronic angiotensin II infusion promotes atherogenesis in low density
lipoprotein receptor −/− mice. Ann N Y Acad Sci. 1999;892:108–18. [PubMed: 10842656]
50. Nishijo N, Sugiyama F, Kimoto K, Taniguchi K, Murakami K, Suzuki S, Fukamizu A and Yagami
Author Manuscript

K. Salt-sensitive aortic aneurysm and rupture in hypertensive transgenic mice that overproduce
angiotensin II. Lab Invest. 1998;78:1059–66. [PubMed: 9759650]
51. Liao S, Miralles M, Kelley BJ, Curci JA, Borhani M and Thompson RW. Suppression of
experimental abdominal aortic aneurysms in the rat by treatment with angiotensin-converting
enzyme inhibitors. J Vasc Surg. 2001;33:1057–64. [PubMed: 11331849]
52. Sun J, Sukhova GK, Yang M, Wolters PJ, MacFarlane LA, Libby P, Sun C, Zhang Y, Liu J, Ennis
TL, Knispel R, Xiong W, Thompson RW, Baxter BT and Shi GP. Mast cells modulate the
pathogenesis of elastase-induced abdominal aortic aneurysms in mice. J Clin Invest.
2007;117:3359–68. [PubMed: 17932568]
53. Furubayashi K, Takai S, Jin D, Miyazaki M, Katsumata T, Inagaki S, Kimura M, Tanaka K,
Nishimoto M and Fukumoto H. Chymase activates promatrix metalloproteinase-9 in human
abdominal aortic aneurysm. Clin Chim Acta. 2008;388:214–6. [PubMed: 17964292]
54. Leskinen MJ, Heikkila HM, Speer MY, Hakala JK, Laine M, Kovanen PT and Lindstedt KA. Mast
cell chymase induces smooth muscle cell apoptosis by disrupting NF-kappaB-mediated survival
signaling. Exp Cell Res. 2006;312:1289–98. [PubMed: 16460729]
Author Manuscript

55. Griendling KK, Lassegue B and Alexander RW. Angiotensin receptors and their therapeutic
implications. Annu Rev Pharmacol Toxicol. 1996;36:281–306. [PubMed: 8725391]
56. Griendling KK, Ushio-Fukai M, Lassegue B and Alexander RW. Angiotensin II signaling in
vascular smooth muscle. New concepts. Hypertension. 1997;29:366–73. [PubMed: 9039129]
57. Garrido AM and Griendling KK. NADPH oxidases and angiotensin II receptor signaling. Mol Cell
Endocrinol. 2009;302:148–58. [PubMed: 19059306]
58. Montezano AC and Touyz RM. Oxidative stress, Noxs, and hypertension: experimental evidence
and clinical controversies. Ann Med. 2012;44 Suppl 1:S2–16. [PubMed: 22713144]
59. Savoia C, Burger D, Nishigaki N, Montezano A and Touyz RM. Angiotensin II and the vascular
phenotype in hypertension. Expert Rev Mol Med. 2011;13:e11. [PubMed: 21450123]
60. Bicknell CD, Kiru G, Falaschetti E, Powell JT, Poulter NR and Collaborators A. An evaluation of
the effect of an angiotensin-converting enzyme inhibitor on the growth rate of small abdominal
aortic aneurysms: a randomized placebo-controlled trial (AARDVARK). Eur Heart J.
2016;37:3213–3221. [PubMed: 27371719]
Author Manuscript

61. Sweeting MJ, Thompson SG, Brown LC, Greenhalgh RM and Powell JT. Use of angiotensin
converting enzyme inhibitors is associated with increased growth rate of abdominal aortic
aneurysms. J Vasc Surg. 2010;52:1–4. [PubMed: 20494541]
62. Liu S, Xie Z, Daugherty A, Cassis LA, Pearson KJ, Gong MC and Guo Z. Mineralocorticoid
receptor agonists induce mouse aortic aneurysm formation and rupture in the presence of high salt.
Arterioscler Thromb Vasc Biol. 2013;33:1568–79. [PubMed: 23661677]
63. Ricci MA, Strindberg G, Slaiby JM, Guibord R, Bergersen LJ, Nichols P, Hendley ED and Pilcher
DB. Anti-CD 18 monoclonal antibody slows experimental aortic aneurysm expansion. J Vasc
Surg. 1996;23:301–7. [PubMed: 8637107]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 18

64. Pope NH, Salmon M, Davis JP, Chatterjee A, Su G, Conte MS, Ailawadi G and Upchurch GR, Jr.
D-series resolvins inhibit murine abdominal aortic aneurysm formation and increase M2
Author Manuscript

macrophage polarization. FASEB J. 2016;30:4192–4201. [PubMed: 27619672]


65. Spinosa M, Su G, Salmon MD, Lu G, Cullen JM, Fashandi AZ, Hawkins RB, Montgomery W,
Meher AK, Conte MS, Sharma AK, Ailawadi G and Upchurch GR, Jr. Resolvin D1 decreases
abdominal aortic aneurysm formation by inhibiting NETosis in a mouse model. J Vasc Surg.
2018;68:93S–103S. [PubMed: 30470363]
66. Meher AK, Spinosa M, Davis JP, Pope N, Laubach VE, Su G, Serbulea V, Leitinger N, Ailawadi G
and Upchurch GR, Jr. Novel Role of IL (Interleukin)-1beta in Neutrophil Extracellular Trap
Formation and Abdominal Aortic Aneurysms. Arterioscler Thromb Vasc Biol. 2018;38:843–853.
[PubMed: 29472233]
67. Kim HW, Blomkalns AL, Ogbi M, Thomas M, Gavrila D, Neltner BS, Cassis LA, Thompson RW,
Weiss RM, Lindower PD, Blanco VM, McCormick ML, Daugherty A, Fu X, Hazen SL, Stansfield
BK, Huo Y, Fulton DJ, Chatterjee T and Weintraub NL. Role of myeloperoxidase in abdominal
aortic aneurysm formation: mitigation by taurine. Am J Physiol Heart Circ Physiol.
2017;313:H1168–H1179. [PubMed: 28971841]
Author Manuscript

68. Curci JA, Liao S, Huffman MD, Shapiro SD and Thompson RW. Expression and localization of
macrophage elastase (matrix metalloproteinase-12) in abdominal aortic aneurysms. J Clin Invest.
1998;102:1900–10. [PubMed: 9835614]
69. Raffort J, Lareyre F, Clement M, Hassen-Khodja R, Chinetti G and Mallat Z. Monocytes and
macrophages in abdominal aortic aneurysm. Nat Rev Cardiol. 2017;14:457–471. [PubMed:
28406184]
70. Hadi T, Boytard L, Silvestro M, Alebrahim D, Jacob S, Feinstein J, Barone K, Spiro W, Hutchison
S, Simon R, Rateri D, Pinet F, Fenyo D, Adelman M, Moore KJ, Eltzschig HK, Daugherty A and
Ramkhelawon B. Macrophage-derived netrin-1 promotes abdominal aortic aneurysm formation by
activating MMP3 in vascular smooth muscle cells. Nat Commun. 2018;9:5022. [PubMed:
30479344]
71. Schaheen B, Downs EA, Serbulea V, Almenara CC, Spinosa M, Su G, Zhao Y, Srikakulapu P,
Butts C, McNamara CA, Leitinger N, Upchurch GR, Jr., Meher AK and Ailawadi G. B-Cell
Depletion Promotes Aortic Infiltration of Immunosuppressive Cells and Is Protective of
Experimental Aortic Aneurysm. Arterioscler Thromb Vasc Biol. 2016;36:2191–2202. [PubMed:
Author Manuscript

27634836]
72. Ponnuswamy P, Joffre J, Herbin O, Esposito B, Laurans L, Binder CJ, Tedder TF, Zeboudj L,
Loyer X, Giraud A, Zhang Y, Tedgui A, Mallat Z and Ait-Oufella H. Angiotensin II synergizes
with BAFF to promote atheroprotective regulatory B cells. Sci Rep. 2017;7:4111. [PubMed:
28646220]
73. Xiong W, Zhao Y, Prall A, Greiner TC and Baxter BT. Key roles of CD4+ T cells and IFN-gamma
in the development of abdominal aortic aneurysms in a murine model. J Immunol. 2004;172:2607–
12. [PubMed: 14764734]
74. Ait-Oufella H, Wang Y, Herbin O, Bourcier S, Potteaux S, Joffre J, Loyer X, Ponnuswamy P,
Esposito B, Dalloz M, Laurans L, Tedgui A and Mallat Z. Natural regulatory T cells limit
angiotensin II-induced aneurysm formation and rupture in mice. Arterioscler Thromb Vasc Biol.
2013;33:2374–9. [PubMed: 23908246]
75. Kasashima S, Kawashima A, Zen Y, Ozaki S, Kasashima F, Endo M, Matsumoto Y and Kawakami
K. Upregulated interleukins (IL-6, IL-10, and IL-13) in immunoglobulin G4-related aortic
aneurysm patients. J Vasc Surg. 2018;67:1248–1262. [PubMed: 28434701]
Author Manuscript

76. Wang Y, Ait-Oufella H, Herbin O, Bonnin P, Ramkhelawon B, Taleb S, Huang J, Offenstadt G,


Combadiere C, Renia L, Johnson JL, Tharaux PL, Tedgui A and Mallat Z. TGF-beta activity
protects against inflammatory aortic aneurysm progression and complications in angiotensin II-
infused mice. J Clin Invest. 2010;120:422–32. [PubMed: 20101093]
77. Li H, Bai S, Ao Q, Wang X, Tian X, Li X, Tong H, Hou W and Fan J. Modulation of Immune-
Inflammatory Responses in Abdominal Aortic Aneurysm: Emerging Molecular Targets. J
Immunol Res. 2018;2018:7213760. [PubMed: 29967801]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 19

78. Galle C, Schandene L, Stordeur P, Peignois Y, Ferreira J, Wautrecht JC, Dereume JP and Goldman
M. Predominance of type 1 CD4+ T cells in human abdominal aortic aneurysm. Clin Exp
Author Manuscript

Immunol. 2005;142:519–27. [PubMed: 16297165]


79. King VL, Lin AY, Kristo F, Anderson TJ, Ahluwalia N, Hardy GJ, Owens AP, 3rd, Howatt DA,
Shen D, Tager AM, Luster AD, Daugherty A and Gerszten RE. Interferon-gamma and the
interferon-inducible chemokine CXCL10 protect against aneurysm formation and rupture.
Circulation. 2009;119:426–35. [PubMed: 19139386]
80. Schonbeck U, Sukhova GK, Gerdes N and Libby P. T(H)2 predominant immune responses prevail
in human abdominal aortic aneurysm. Am J Pathol. 2002;161:499–506. [PubMed: 12163375]
81. Peshkova IO, Schaefer G and Koltsova EK. Atherosclerosis and aortic aneurysm - is inflammation
a common denominator? FEBS J. 2016;283:1636–52. [PubMed: 26700480]
82. Miller FJ, Jr., Sharp WJ, Fang X, Oberley LW, Oberley TD and Weintraub NL. Oxidative stress in
human abdominal aortic aneurysms: a potential mediator of aneurysmal remodeling. Arterioscler
Thromb Vasc Biol. 2002;22:560–5. [PubMed: 11950691]
83. Gavrila D, Li WG, McCormick ML, Thomas M, Daugherty A, Cassis LA, Miller FJ, Jr., Oberley
LW, Dellsperger KC and Weintraub NL. Vitamin E inhibits abdominal aortic aneurysm formation
Author Manuscript

in angiotensin II-infused apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol.


2005;25:1671–7. [PubMed: 15933246]
84. Maiellaro-Rafferty K, Weiss D, Joseph G, Wan W, Gleason RL and Taylor WR. Catalase
overexpression in aortic smooth muscle prevents pathological mechanical changes underlying
abdominal aortic aneurysm formation. Am J Physiol Heart Circ Physiol. 2011;301:H355–62.
[PubMed: 21551275]
85. Parastatidis I, Weiss D, Joseph G and Taylor WR. Overexpression of catalase in vascular smooth
muscle cells prevents the formation of abdominal aortic aneurysms. Arterioscler Thromb Vasc
Biol. 2013;33:2389–96. [PubMed: 23950141]
86. Griendling KK, Minieri CA, Ollerenshaw JD and Alexander RW. Angiotensin II stimulates NADH
and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res. 1994;74:1141–8.
[PubMed: 8187280]
87. Lassegue B, San Martin A and Griendling KK. Biochemistry, physiology, and pathophysiology of
NADPH oxidases in the cardiovascular system. Circ Res. 2012;110:1364–90. [PubMed:
22581922]
Author Manuscript

88. Amanso AM and Griendling KK. Differential roles of NADPH oxidases in vascular physiology
and pathophysiology. Front Biosci (Schol Ed). 2012;4:1044–64. [PubMed: 22202108]
89. Thomas M, Gavrila D, McCormick ML, Miller FJ, Jr., Daugherty A, Cassis LA, Dellsperger KC
and Weintraub NL. Deletion of p47phox attenuates angiotensin II-induced abdominal aortic
aneurysm formation in apolipoprotein E-deficient mice. Circulation. 2006;114:404–413. [PubMed:
16864727]
90. Kigawa Y, Miyazaki T, Lei XF, Nakamachi T, Oguchi T, Kim-Kaneyama JR, Taniyama M,
Tsunawaki S, Shioda S and Miyazaki A. NADPH oxidase deficiency exacerbates angiotensin II-
induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol. 2014;34:2413–20.
[PubMed: 25189573]
91. Gielis JF, Lin JY, Wingler K, Van Schil PE, Schmidt HH and Moens AL. Pathogenetic role of
eNOS uncoupling in cardiopulmonary disorders. Free Radic Biol Med. 2011;50:765–76.
[PubMed: 21172428]
92. Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard MH and Huang
Author Manuscript

PL. Accelerated atherosclerosis, aortic aneurysm formation, and ischemic heart disease in
apolipoprotein E/endothelial nitric oxide synthase double-knockout mice. Circulation.
2001;104:448–54. [PubMed: 11468208]
93. Johanning JM, Franklin DP, Han DC, Carey DJ and Elmore JR. Inhibition of inducible nitric oxide
synthase limits nitric oxide production and experimental aneurysm expansion. J Vasc Surg.
2001;33:579–86. [PubMed: 11241130]
94. Fatini C, Sofi F, Sticchi E, Bolli P, Sestini I, Falciani M, Azas L and Pratesi G. eNOS G894T
polymorphism as a mild predisposing factor for abdominal aortic aneurysm. J Vasc Surg.
2005;42:415–9. [PubMed: 16171581]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 20

95. Gao L, Siu KL, Chalupsky K, Nguyen A, Chen P, Weintraub NL, Galis Z and Cai H. Role of
uncoupled endothelial nitric oxide synthase in abdominal aortic aneurysm formation: treatment
Author Manuscript

with folic acid. Hypertension. 2012;59:158–66. [PubMed: 22083158]


96. Siu KL and Cai H. Circulating tetrahydrobiopterin as a novel biomarker for abdominal aortic
aneurysm. Am J Physiol Heart Circ Physiol. 2014;307:H1559–64. [PubMed: 25260610]
97. Vlasova II. Peroxidase Activity of Human Hemoproteins: Keeping the Fire under Control.
Molecules. 2018;23.
98. Webb NR, De Beer MC, Wroblewski JM, Ji A, Bailey W, Shridas P, Charnigo RJ, Noffsinger VP,
Witta J, Howatt DA, Balakrishnan A, Rateri DL, Daugherty A and De Beer FC. Deficiency of
Endogenous Acute-Phase Serum Amyloid A Protects apoE−/− Mice From Angiotensin II-Induced
Abdominal Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol. 2015;35:1156–65.
[PubMed: 25745063]
99. Guzik B, Sagan A, Ludew D, Mrowiecki W, Chwala M, Bujak-Gizycka B, Filip G, Grudzien G,
Kapelak B, Zmudka K, Mrowiecki T, Sadowski J, Korbut R and Guzik TJ. Mechanisms of
oxidative stress in human aortic aneurysms--association with clinical risk factors for
atherosclerosis and disease severity. Int J Cardiol. 2013;168:2389–96. [PubMed: 23506637]
Author Manuscript

100. Yan P, Chen K, Wang Q, Yang D, Li Y. UCP-2 is involved in angiotensin-II-induced abdominal


aortic aneurysm in apolipoprotein E-knockout mice. PLoS One. 2017;12:e0179743. [PubMed:
28683125]
101. Ramos-Mozo P, Madrigal-Matute J, Martinez-Pinna R, Blanco-Colio LM, Lopez JA, Camafeita
E, Meilhac O, Michel JB, Aparicio C, Vega de Ceniga M, Egido J and Martin-Ventura JL.
Proteomic analysis of polymorphonuclear neutrophils identifies catalase as a novel biomarker of
abdominal aortic aneurysm: potential implication of oxidative stress in abdominal aortic
aneurysm progression. Arterioscler Thromb Vasc Biol. 2011;31:3011–9. [PubMed: 21940941]
102. Lucas ML, Carraro CC, Bello-Klein A, Kalil AN, Aerts NR, Carvalho FB, Fernandes MC and
Zettler CG. Oxidative Stress in Aortas of Patients with Advanced Occlusive and Aneurysmal
Diseases. Ann Vasc Surg. 2018;52:216–224. [PubMed: 29758327]
103. Dubick MA, Keen CL, DiSilvestro RA, Eskelson CD, Ireton J and Hunter GC. Antioxidant
enzyme activity in human abdominal aortic aneurysmal and occlusive disease. Proc Soc Exp Biol
Med. 1999;220:39–45. [PubMed: 9893167]
104. Sinha I, Pearce CG, Cho BS, Hannawa KK, Roelofs KJ, Stanley JC, Henke PK and Upchurch
Author Manuscript

GR, Jr. Differential regulation of the superoxide dismutase family in experimental aortic
aneurysms and rat aortic explants. J Surg Res. 2007;138:156–62. [PubMed: 17196988]
105. Yajima N, Masuda M, Miyazaki M, Nakajima N, Chien S and Shyy JY. Oxidative stress is
involved in the development of experimental abdominal aortic aneurysm: a study of the
transcription profile with complementary DNA microarray. J Vasc Surg. 2002;36:379–85.
[PubMed: 12170221]
106. Burillo E, Tarin C, Torres-Fonseca MM, Fernandez-Garcia CE, Martinez-Pinna R, Martinez-
Lopez D, Llamas-Granda P, Camafeita E, Lopez JA, Vega de Ceniga M, Aviram M, Egido J,
Blanco-Colio LM and Martin-Ventura JL. Paraoxonase-1 overexpression prevents experimental
abdominal aortic aneurysm progression. Clin Sci (Lond). 2016;130:1027–38. [PubMed:
26993251]
107. Forrester SJ, Kikuchi DS, Hernandes MS, Xu Q and Griendling KK. Reactive Oxygen Species in
Metabolic and Inflammatory Signaling. Circ Res. 2018;122:877–902. [PubMed: 29700084]
108. Brown DI and Griendling KK. Regulation of signal transduction by reactive oxygen species in the
Author Manuscript

cardiovascular system. Circ Res. 2015;116:531–49. [PubMed: 25634975]


109. Holmstrom KM and Finkel T. Cellular mechanisms and physiological consequences of redox-
dependent signalling. Nat Rev Mol Cell Biol. 2014;15:411–21. [PubMed: 24854789]
110. Finkel T Signal transduction by reactive oxygen species. J Cell Biol. 2011;194:7–15. [PubMed:
21746850]
111. Miki H and Funato Y. Regulation of intracellular signalling through cysteine oxidation by reactive
oxygen species. J Biochem. 2012;151:255–61. [PubMed: 22287686]
112. Villard C, Swedenborg J, Eriksson P and Hultgren R. Reproductive history in women with
abdominal aortic aneurysms. J Vasc Surg. 2011;54:341–5, 345 e1–2. [PubMed: 21620618]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 21

113. Ailawadi G, Eliason JL, Roelofs KJ, Sinha I, Hannawa KK, Kaldjian EP, Lu G, Henke PK,
Stanley JC, Weiss SJ, Thompson RW and Upchurch GR, Jr. Gender differences in experimental
Author Manuscript

aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2004;24:2116–22. [PubMed:


15331435]
114. Alsiraj Y, Thatcher SE, Charnigo R, Chen K, Blalock E, Daugherty A and Cassis LA. Female
Mice With an XY Sex Chromosome Complement Develop Severe Angiotensin II-Induced
Abdominal Aortic Aneurysms. Circulation. 2017;135:379–391. [PubMed: 27815372]
115. Bengtsson H, Sonesson B and Bergqvist D. Incidence and prevalence of abdominal aortic
aneurysms, estimated by necropsy studies and population screening by ultrasound. Ann N Y
Acad Sci. 1996;800:1–24.
116. Boese AC, Chang L, Yin KJ, Chen YE, Lee JP and Hamblin MH. Sex differences in abdominal
aortic aneurysms. Am J Physiol Heart Circ Physiol. 2018;314:H1137–H1152. [PubMed:
29350999]
117. Hsia J, Criqui MH, Herrington DM, Manson JE, Wu L, Heckbert SR, Allison M, McDermott
MM, Robinson J, Masaki K and Women’s Health Initiative Research G. Conjugated equine
estrogens and peripheral arterial disease risk: the Women’s Health Initiative. Am Heart J.
Author Manuscript

2006;152:170–6. [PubMed: 16824852]


118. Lederle FA, Larson JC, Margolis KL, Allison MA, Freiberg MS, Cochrane BB, Graettinger WF,
Curb JD and Women’s Health Initiative Cohort S. Abdominal aortic aneurysm events in the
women’s health initiative: cohort study. BMJ. 2008;337:a1724. [PubMed: 18854591]
119. Henriques T, Zhang X, Yiannikouris FB, Daugherty A and Cassis LA. Androgen increases AT1a
receptor expression in abdominal aortas to promote angiotensin II-induced AAAs in
apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2008;28:1251–6. [PubMed:
18451329]
120. Costa TJ, Ceravolo GS, dos Santos RA, de Oliveira MA, Araujo PX, Giaquinto LR, Tostes RC,
Akamine EH, Fortes ZB, Dantas AP and Carvalho MH. Association of testosterone with estrogen
abolishes the beneficial effects of estrogen treatment by increasing ROS generation in aorta
endothelial cells. Am J Physiol Heart Circ Physiol. 2015;308:H723–32. [PubMed: 25637546]
121. Milsted A, Underwood AC, Dunmire J, DelPuerto HL, Martins AS, Ely DL and Turner ME.
Regulation of multiple renin-angiotensin system genes by Sry. J Hypertens. 2010;28:59–64.
[PubMed: 19809364]
Author Manuscript

122. Yeap BB, Hyde Z, Norman PE, Chubb SA and Golledge J. Associations of total testosterone, sex
hormone-binding globulin, calculated free testosterone, and luteinizing hormone with prevalence
of abdominal aortic aneurysm in older men. J Clin Endocrinol Metab. 2010;95:1123–30.
[PubMed: 20061425]
123. Astrand H, Stalhand J, Karlsson J, Karlsson M, Sonesson B and Lanne T. In vivo estimation of
the contribution of elastin and collagen to the mechanical properties in the human abdominal
aorta: effect of age and sex. J Appl Physiol (1985). 2011;110:176–87. [PubMed: 21071586]
124. Sonesson B, Lanne T, Vernersson E and Hansen F. Sex difference in the mechanical properties of
the abdominal aorta in human beings. J Vasc Surg. 1994;20:959–69. [PubMed: 7990192]
125. Sonesson B, Hansen F, Stale H and Lanne T. Compliance and diameter in the human abdominal
aorta--the influence of age and sex. Eur J Vasc Surg. 1993;7:690–7. [PubMed: 8270073]
126. Tong J, Schriefl AJ, Cohnert T and Holzapfel GA. Gender differences in biomechanical
properties, thrombus age, mass fraction and clinical factors of abdominal aortic aneurysms. Eur J
Vasc Endovasc Surg. 2013;45:364–72. [PubMed: 23395130]
Author Manuscript

127. Cathapermal S, Lavigne MC, Leong-Son M, Alibadi T and Ramwell PW. Stereoisomer-specific
inhibition of superoxide anion-induced rat aortic smooth-muscle cell proliferation by 17beta-
estradiol is estrogen receptor dependent. J Cardiovasc Pharmacol. 1998;31:499–505. [PubMed:
9554796]
128. Natoli AK, Medley TL, Ahimastos AA, Drew BG, Thearle DJ, Dilley RJ and Kingwell BA. Sex
steroids modulate human aortic smooth muscle cell matrix protein deposition and matrix
metalloproteinase expression. Hypertension. 2005;46:1129–34. [PubMed: 16230520]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 22

129. Wang S, Zhang C, Zhang M, Liang B, Zhu H, Lee J, Viollet B, Xia L, Zhang Y and Zou MH.
Activation of AMP-activated protein kinase alpha2 by nicotine instigates formation of abdominal
Author Manuscript

aortic aneurysms in mice in vivo. Nat Med. 2012;18:902–10. [PubMed: 22561688]


130. Ghosh A, Pechota A, Coleman D, Upchurch GR, Jr. and Eliason JL. Cigarette smoke-induced
MMP2 and MMP9 secretion from aortic vascular smooth cells is mediated via the Jak/Stat
pathway. Hum Pathol. 2015;46:284–94. [PubMed: 25537973]
131. Fedak PW, Verma S, David TE, Leask RL, Weisel RD and Butany J. Clinical and
pathophysiological implications of a bicuspid aortic valve. Circulation. 2002;106:900–4.
[PubMed: 12186790]
132. Remus EW, O’Donnell RE, Jr., Rafferty K, Weiss D, Joseph G, Csiszar K, Fong SF and Taylor
WR. The role of lysyl oxidase family members in the stabilization of abdominal aortic
aneurysms. Am J Physiol Heart Circ Physiol. 2012;303:H1067–75. [PubMed: 22904155]
133. Bean WB and Ponseti IV. Dissecting aneurysm produced by diet. Circulation. 1955;12:185–92.
[PubMed: 13240798]
134. De Paepe A and Malfait F. Bleeding and bruising in patients with Ehlers-Danlos syndrome and
other collagen vascular disorders. Br J Haematol. 2004;127:491–500. [PubMed: 15566352]
Author Manuscript

135. Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM, Puffenberger EG, Hamosh
A, Nanthakumar EJ, Curristin SM and et al. Marfan syndrome caused by a recurrent de novo
missense mutation in the fibrillin gene. Nature. 1991;352:337–9. [PubMed: 1852208]
136. Neptune ER, Frischmeyer PA, Arking DE, Myers L, Bunton TE, Gayraud B, Ramirez F, Sakai LY
and Dietz HC. Dysregulation of TGF-beta activation contributes to pathogenesis in Marfan
syndrome. Nat Genet. 2003;33:407–11. [PubMed: 12598898]
137. Ng CM, Cheng A, Myers LA, Martinez-Murillo F, Jie C, Bedja D, Gabrielson KL, Hausladen JM,
Mecham RP, Judge DP and Dietz HC. TGF-beta-dependent pathogenesis of mitral valve prolapse
in a mouse model of Marfan syndrome. J Clin Invest. 2004;114:1586–92. [PubMed: 15546004]
138. Judge DP, Biery NJ, Keene DR, Geubtner J, Myers L, Huso DL, Sakai LY and Dietz HC.
Evidence for a critical contribution of haploinsufficiency in the complex pathogenesis of Marfan
syndrome. J Clin Invest. 2004;114:172–81. [PubMed: 15254584]
139. Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK, Myers L, Klein EC, Liu G,
Calvi C, Podowski M, Neptune ER, Halushka MK, Bedja D, Gabrielson K, Rifkin DB, Carta L,
Ramirez F, Huso DL and Dietz HC. Losartan, an AT1 antagonist, prevents aortic aneurysm in a
Author Manuscript

mouse model of Marfan syndrome. Science. 2006;312:117–21. [PubMed: 16601194]


140. Robinson PN, Arteaga-Solis E, Baldock C, Collod-Beroud G, Booms P, De Paepe A, Dietz HC,
Guo G, Handford PA, Judge DP, Kielty CM, Loeys B, Milewicz DM, Ney A, Ramirez F,
Reinhardt DP, Tiedemann K, Whiteman P and Godfrey M. The molecular genetics of Marfan
syndrome and related disorders. J Med Genet. 2006;43:769–87. [PubMed: 16571647]
141. Kaartinen V and Warburton D. Fibrillin controls TGF-beta activation. Nat Genet. 2003;33:331–2.
[PubMed: 12610545]
142. Guo DC, Regalado ES, Pinard A, Chen J, Lee K, Rigelsky C, Zilberberg L, Hostetler EM, Aldred
M, Wallace SE, Prakash SK, University of Washington Center for Mendelian G, Leal SM,
Bamshad MJ, Nickerson DA, Natowicz M, Rifkin DB and Milewicz DM. LTBP3 Pathogenic
Variants Predispose Individuals to Thoracic Aortic Aneurysms and Dissections. Am J Hum
Genet. 2018;102:706–712. [PubMed: 29625025]
143. Lin F and Yang X. TGF-beta signaling in aortic aneurysm: another round of controversy. J Genet
Genomics. 2010;37:583–91. [PubMed: 20933212]
Author Manuscript

144. Gomez D, Al Haj Zen A, Borges LF, Philippe M, Gutierrez PS, Jondeau G, Michel JB and
Vranckx R. Syndromic and non-syndromic aneurysms of the human ascending aorta share
activation of the Smad2 pathway. J Pathol. 2009;218:131–42. [PubMed: 19224541]
145. Wei H, Hu JH, Angelov SN, Fox K, Yan J, Enstrom R, Smith A and Dichek DA. Aortopathy in a
Mouse Model of Marfan Syndrome Is Not Mediated by Altered Transforming Growth Factor
beta Signaling. J Am Heart Assoc. 2017;6.
146. Li W, Li Q, Jiao Y, Qin L, Ali R, Zhou J, Ferruzzi J, Kim RW, Geirsson A, Dietz HC, Offermanns
S, Humphrey JD and Tellides G. Tgfbr2 disruption in postnatal smooth muscle impairs aortic
wall homeostasis. J Clin Invest. 2014;124:755–67. [PubMed: 24401272]

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 23

147. Daugherty A, Chen Z, Sawada H, Rateri DL and Sheppard MB. Transforming Growth Factor-beta
in Thoracic Aortic Aneurysms: Good, Bad, or Irrelevant? J Am Heart Assoc. 2017;6.
Author Manuscript

148. Nagasawa A, Yoshimura K, Suzuki R, Mikamo A, Yamashita O, Ikeda Y, Tsuchida M and


Hamano K. Important role of the angiotensin II pathway in producing matrix metalloproteinase-9
in human thoracic aortic aneurysms. J Surg Res. 2013;183:472–7. [PubMed: 23295196]
149. Rabkin SW. Differential expression of MMP-2, MMP-9 and TIMP proteins in thoracic aortic
aneurysm - comparison with and without bicuspid aortic valve: a meta-analysis. Vasa.
2014;43:433–42. [PubMed: 25339161]
150. Ikonomidis JS, Jones JA, Barbour JR, Stroud RE, Clark LL, Kaplan BS, Zeeshan A, Bavaria JE,
Gorman JH, 3rd, Spinale FG and Gorman RC. Expression of matrix metalloproteinases and
endogenous inhibitors within ascending aortic aneurysms of patients with bicuspid or tricuspid
aortic valves. J Thorac Cardiovasc Surg. 2007;133:1028–36. [PubMed: 17382648]
151. He R, Guo DC, Sun W, Papke CL, Duraisamy S, Estrera AL, Safi HJ, Ahn C, Buja LM, Arnett
FC, Zhang J, Geng YJ and Milewicz DM. Characterization of the inflammatory cells in
ascending thoracic aortic aneurysms in patients with Marfan syndrome, familial thoracic aortic
aneurysms, and sporadic aneurysms. J Thorac Cardiovasc Surg. 2008;136:922–9, 929 e1.
Author Manuscript

[PubMed: 18954631]
152. Sulkava M, Raitoharju E, Mennander A, Levula M, Seppala I, Lyytikainen LP, Jarvinen O, Illig T,
Klopp N, Mononen N, Laaksonen R, Kahonen M, Oksala N and Lehtimaki T. Differentially
expressed genes and canonical pathways in the ascending thoracic aortic aneurysm - The
Tampere Vascular Study. Sci Rep. 2017;7:12127. [PubMed: 28935963]
153. Pope NH, Salmon M, Johnston WF, Lu G, Lau CL, Upchurch GR, Jr. and Ailawadi G.
Interleukin-6 Receptor Inhibition Prevents Descending Thoracic Aortic Aneurysm Formation.
Ann Thorac Surg. 2015;100:1620–6. [PubMed: 26165482]
154. Johnston WF, Salmon M, Pope NH, Meher A, Su G, Stone ML, Lu G, Owens GK, Upchurch GR,
Jr. and Ailawadi G. Inhibition of interleukin-1beta decreases aneurysm formation and progression
in a novel model of thoracic aortic aneurysms. Circulation. 2014;130:S51–9. [PubMed:
25200056]
155. Yang HH, van Breemen C and Chung AW. Vasomotor dysfunction in the thoracic aorta of Marfan
syndrome is associated with accumulation of oxidative stress. Vascul Pharmacol. 2010;52:37–45.
[PubMed: 19879959]
Author Manuscript

156. Jimenez-Altayo F, Meirelles T, Crosas-Molist E, Sorolla MA, Del Blanco DG, Lopez-Luque J,
Mas-Stachurska A, Siegert AM, Bonorino F, Barbera L, Garcia C, Condom E, Sitges M,
Rodriguez-Pascual F, Laurindo F, Schroder K, Ros J, Fabregat I and Egea G. Redox stress in
Marfan syndrome: Dissecting the role of the NADPH oxidase NOX4 in aortic aneurysm. Free
Radic Biol Med. 2018;118:44–58. [PubMed: 29471108]
157. van der Pluijm I, Burger J, van Heijningen PM, A IJ, van Vliet N, Milanese C, Schoonderwoerd
K, Sluiter W, Ringuette LJ, Dekkers DHW, Que I, Kaijzel EL, Te Riet L, MacFarlane EG, Das D,
van der Linden R, Vermeij M, Demmers JA, Mastroberardino PG, Davis EC, Yanagisawa H,
Dietz HC, Kanaar R and Essers J. Decreased mitochondrial respiration in aneurysmal aortas of
Fibulin-4 mutant mice is linked to PGC1A regulation. Cardiovasc Res. 2018;114:1776–1793.
[PubMed: 29931197]
Author Manuscript

Circ Res. Author manuscript; available in PMC 2020 February 15.


Quintana and Taylor Page 24
Author Manuscript
Author Manuscript
Author Manuscript

Diagrammatic representation of the most critical components of the cellular mechanisms of


thoracic and abdominal aortic aneurysm formation
Author Manuscript

Circ Res. Author manuscript; available in PMC 2020 February 15.

You might also like