Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

BASIC SCIENCE REVIEW

Current concepts in the pathogenesis of


abdominal aortic aneurysm
Gorav Ailawadi, MD, Jonathan L. Eliason, MD, and Gilbert R. Upchurch Jr, MD, Ann Arbor, Mich

Abdominal aortic aneurysms (AAAs) are a significant PROTEOLYTIC DEGRADATION OF AORTIC


medical problem with a high mortality. The primary diag- WALL CONNECTIVE TISSUE
nostic code for AAA accounts for approximately 150,000 Aneurysm formation involves a complex process of
inpatient hospital admissions per year. The most recent destruction of the aortic media and supporting lamina
published data from the National Vital Statistics Report on through degradation of elastin and collagen. Experimental
Deaths from the year 2000 show that AAAs and aortic data and studies of human AAAs suggest that matrix met-
dissection composed the tenth leading cause of death in alloproteinases (MMPs) and other proteases, derived from
white men 65 to 74 years old and accounted for nearly macrophages and aortic smooth muscle cells, are secreted
16,000 deaths overall. The year 2000 National Hospital into the extracellular matrix and are integral to aneurysm
Discharge Summary reports more than 30,000 open oper- formation (Table I).3,5 MMPs are also prominent in a
ations for repair of AAAs in the United States. Understand- number of pathologic conditions, eg, tumor metastasis and
ing the cause of AAAs therefore becomes an important various rheumatologic and orthopedic disorders. Fluctua-
undertaking. tion in MMP expression and activity occurs during normal
The pathogenesis of AAAs is complex and multifacto- physiologic aortic wall remodeling. However, in AAAs,
rial. Histologically, AAAs are characterized by destruction MMP activation favors collagen and elastin degradation.1
of elastin and collagen in the media and adventitia, smooth Interstitial collagen dissolution accompanies increased ex-
muscle cell loss with thinning of the medial wall, infiltration pression of collagenases MMP-1 and MMP-13 in AAAs in
humans. Elastases MMP-2 (gelatinase A), MMP-9 (gelati-
of lymphocytes and macrophages, and neovascularization.7
nase B), and MMP-12 (macrophage elastase) are also in-
Inflammation is a common underlying feature of both
creased in aneurysmal aortic tissue. MMP-12 is highly
aneurysm disease and atherosclerosis. However, atheroscle-
expressed along the proximal leading edge of AAAs in
rosis is primarily found within the intima and media,
humans, and may be important in aneurysm initiation.
whereas aneurysm disease typically affects the media and High concentrations of the constitutive enzyme MMP-2
adventitia. A National Heart, Lung and Blood Institute are found in small aneurysmal aortas, suggesting a role for
Request for Applications (HL-99-007) entitled “Patho- MMP-2 during early aneurysm formation. The inducible
genesis of Abdominal Aortic Aneurysms” identified four enzyme MMP-9 is elevated in aortic tissue and in serum
mechanisms relevant to AAA formation: proteolytic degra- from patients with AAA compared with patients with aor-
dation of aortic wall connective tissue, inflammation and toiliac occlusive disease.8 A critical role for MMP-9 is
immune responses, biomechanical wall stress, and molecu- supported by the observation that MMP-9 knockout mice
lar genetics.19 do not form experimental aneurysms. Wild-type bone mar-
row transplantation, however, restores the aneurysm phe-
notype. During AAA formation, the balance of vessel wall
remodeling between MMPs and their inhibitors, tissue
From the Jobst Vascular Research Laboratories, Section of Vascular Surgery,
Department of Surgery, University of Michigan Medical School. inhibitors of metalloproteinases, favors elastin and collagen
Supported by an American College of Surgeons Resident Research Scholar- degradation. The mechanisms for initiating and propagat-
ship (G.A.) and a National Institutes of Health Mentored Clinical Scien- ing these proteolytic enzymes in the aorta remain to be
tists Development Award (K08 HL67885-01) Lifeline Foundation Re- identified.
search Award (G.R.U.).
Competition of interest: none.
INFLAMMATION AND IMMUNE RESPONSES
Reprint requests: Gilbert R. Upchurch, Jr, MD, University of Michigan
Medical Center, 2210 Taubman Health Care Center, 1500 E Medical A prominent histologic feature of AAAs is extensive
Center Dr, Ann Arbor, MI 48109-0329 (e-mail: riversu@umich.edu). transmural infiltration by macrophages and lymphocytes. It
J Vasc Surg 2003;38:584-8.
Copyright © 2003 by The Society for Vascular Surgery.
is hypothesized that these cells subsequently release a cas-
0741-5214/2003/$30.00 ⫹ 0 cade of cytokines that result in activation of many proteases
doi:10.1016/S0741-5214(03)00324-0 (Table II). The trigger for influx and migration of leuko-
584
JOURNAL OF VASCULAR SURGERY
Volume 38, Number 3 Ailawadi, Eliason, and Upchurch 585

Table I. Enzymes documented to be altered in AAA in humans

Enzyme Other names Primary substrate Evidence/origin

Metalloproteinases
MMP-1 Collagenase-1, interstitial Collagen (type I, III) Epithelial, inflammatory, mesenchymal
collagenase
MMP-2 72 kD gelatinase A Collagen (type IV), elastin SMCs, fibroblasts
MMP-3 Stromelysin-1 Collagen
MMP-9 92 kD gelatinase B Elastin, collagen Macrophages, SMCs
MMP-12 Macrophage elastase Elastin Macrophages
MMP-13 Collagenase-3 Collagen (type IV) SMCs
MT-MMP-1 (MMP-14) Activates pro-MMP-2 Membrane-bound protein
TIMP-1 Inhibitor of MMPs AAA wall
TIMP-2 Inhibitor of MMP-2 Expression mildly increased
Cysteine proteases
Cathepsin S Elastin Macrophages, SMCs
Cathepsin K Elastin Macrophages, SMCs
Cathepsin D Not elastin or collagen Increased in AAA wall
Cathepsin L Not elastin or Collagen Increased in AAA wall
Cathepsin H Increased by gene array
Serine proteases
Plasmin Macrophages
Tissue-plasminogen Activates MMPs Protein, expression increased; macrophages
activator
Urokinase-plasminogen Activates MMPs Protein, expression increased; macrophages
activator
AAA, Abdominal aortic aneurysm; MMP, matrix metalloproteinase; SMC, smooth muscle cell; TIMP, tissue inhabitor of metalloproteinase.

creased in the aneurysmal aortic wall. Importantly, these


Table II. Cytokines increased in aortic wall or circulating inflammatory cytokines induce expression and activation of
blood levels in patients with AAA MMPs and tissue inhibitors of MMPs.
An infectious cause has also been suggested in several
Cytokine Effect
reports; as many as 55% of aneurysms demonstrate Chla-
Proinflammatory mydiae pneumoniae by immunohistochemistry. Tambiah
IL-1 ␤ SMC MMP production resulting in and Powell demonstrated experimental aneurysm forma-
increased matrix turnover; circulating tion after periaortic application with live and formalin-
levels higher than control levels
IL-6 Induces MMP expression; increased matrix inactivated C pneumoniae, but not with heat-inactivated C
turnover; circulating levels may correlate pneumoniae antigen.
with aneurysm size The role of reactive oxygen species and antioxidants in
IL-8 Induces MMP expression; increased matrix aneurysm disease has also been an area of interest. Super-
turnover
TNF- ␣ Induces MMP expression; increased matrix oxide (O2–) levels in aneurysmal tissue in human beings is
turnover 2.5-fold higher than adjacent, nonaneurysmal aortic tissue
IFN ␥ Elevated in women with AAAs and with and 10-fold higher than control aorta.10 A greater than
short-term expansion of AAA 50-fold increase in inducible nitric oxide synthase gene
Anti-inflammatory
IL-10 Inhibits inflammatory cytokine production; expression was seen 2 days after infusion of rodent aortas
decreases MMP expression; increases with porcine pancreatic elastase. In contrast, 10 days after
TIMP elastase infusion, expression of superoxide dismutase, a
AAA, Abdominal aortic aneurysm; IL, interleukin; SMC, smooth muscle potent antioxidant, was downregulated more than 20-fold
cell; MMP, matrix metalloproteinase TIMP, tissue inhibitor of metallopro- compared with saline solution–infused control aorta.9
teinase. Thus, an imbalance in gene expression promoting oxidant
stress has been demonstrated in AAAs in humans and in
cytes is unknown, but exposed elastin degradation products experimental AAAs. This pro-oxidant environment has im-
in the aortic wall may serve as a chemotactic agent for portant implications for matrix degradation; in in vitro
infiltrating macrophages. The concept that AAA formation studies reactive oxygen species activated MMPs.12,14
is an autoimmune response is supported by the extensive Reactive oxygen species also influence apoptosis, which
lymphocytic and monocytic infiltrate, primarily in the ad- may be important during AAA formation.6 AAAs histolog-
ventitia, and deposition of immunoglobulin G in the aortic ically demonstrate decreased vascular smooth muscle cell
wall. Xia et al described a new 40 kDa aortic aneurysm– density, contributing to loss of aortic wall structural integ-
associated protein that may be chemotactic.21 Macroph- rity. In situ end-labeling of DNA fragments (TUNEL) has
age-generated and lymphocyte-generated cytokines are in- shown that higher levels of DNA fragmentation, a marker
JOURNAL OF VASCULAR SURGERY
586 Ailawadi, Eliason, and Upchurch September 2003

Table III. Altered human expression in AAA by gene array*

Gene function Upregulated in AAA Downregulated in AAA

Extracellular matrix degradation Gelatinase B (matrix metalloproteinase 9)


Inflammation Myeloid cell nuclear differentiation antigen
Interleukin-8
CXC chemokine receptor 4
Tumor necrosis factor-␤R
Cellular adhesion ICAM-1 (CD54) Integrin ␣-5
Integrin ␣L (CD11a) Glycoprotein IIIA
Atherosclerosis Apolipoprotein E
Smooth muscle cell contraction Myosin light chain kinase
Intracellular protein degradation and turnover Cathepsin H
Growth factors Platelet-derived growth factor-A Chain
Oncogenes Fli-1
Cell signalling Transcriptional regulator ISGF3, ␥ subunit Ephrin A5
Rho/Rac GEF
*Genes included have at least a fourfold difference in expression when compared with nonaneurysmal aortic tissue.

of apoptosis, can be found in aneurysmal medial smooth groups have used sophisticated, computer-based three-
muscle cells. Collectively, these studies provide support for dimensional “finite element analysis” of the aorta. Studies
the hypothesis that oxidative stress is an important compo- by Vorp, Fillinger, and others have made significant contri-
nent in formation and enlargement of AAA through acti- butions in this field. In one study, analysis of computed
vation of MMPs and induction of apoptosis within the geometric factors suggested that aneurysm volume, rather
aortic wall. than simply AAA diameter, was the best indicator of peak
wall stress.13 From a biomechanical standpoint, the effect
BIOCHEMICAL WALL STRESS of intraluminal thrombus may serve to lower aortic wall
The preferential infrarenal site for AAA formation sug- stress.18 Furthermore, ruptured or symptomatic AAAs had
gests potential differences in aortic structure, biologic fea- significantly higher peak wall stress compared with diame-
tures, and stress along the length of the aorta. Structurally, ter-matched asymptomatic AAAs.4 This is a dynamic pro-
from proximal to distal along the aorta, the elastin-collagen cess with multiple variables, but recent advances in this area
ratio decreases; ie, the infrarenal aorta has low levels of may improve risk stratification and subsequent decision-
elastin compared with the aortic arch and descending tho- making as to which patients should be considered surgical
racic aorta. Further, a decrease in the elastin-collagen ratio candidates.
from proximal to distal aorta may be clinically relevant,
because diminished elastin is associated with aortic dilation, MOLECULAR GENETICS
and collagen degradation predisposes to aortic rupture. Certain phenotypes have been associated with AAAs.
Ailawadi et al documented that, in addition to diminished For example, the Hp-2-1 haptoglobin phenotype and de-
elastin, the abdominal aorta has increased MMP-9 expres- ficiencies in ␣1-antitrypsin are associated with aneurysm
sion and activity compared with aortic arch and descending formation. Patients with Rh-negative blood type have de-
thoracic aortic segments. creased frequency of AAA, whereas those with MN or
Flow studies have also suggested disordered flow and Kell-positive blood groups have increased frequency. Cur-
an increase in wall tension in the infrarenal aorta.11 Ex vivo rently no single genetic defect or polymorphism has been
models have documented activation of MMPs as a conse- identified as a common denominator for AAA. However,
quence of these mechanical alterations. Others have sug- familial clustering and a common HLA subtype suggest
gested that relative tissue hypoxia occurs in the infrarenal both a genetic and an immunologic role in the pathogen-
aorta as a discrete vasa vasorum ends near the renal artery esis of AAA. Patients with affected siblings are at substan-
branches. A combination of these factors likely contributes tially increased risk for AAA. Gene microarray has provided
to the preponderance for aneurysms to form in the infrare- information regarding altered gene expression in patients
nal aorta. with AAA (Table III).17 Specific polymorphisms have also
Once an AAA has developed, it is likely that increased been linked to AAA formation (Table IV). Genetic screen-
wall stress is important in accelerating dilation and increas- ing of patients predictably at high risk for AAA develop-
ing the risk for rupture. ␤-Blockers reduce wall stress, and it ment will likely become a reality in the future.
has been suggested that they protect against continued
aneurysm dilation and rupture in animal models. In human PROPOSED MECHANISM
trials, however, this observation has not been convincingly A combination of factors, eg, localized hemodynamic
reproduced. stress, fragmented medial proteins, and genetic predisposi-
In an attempt to better understand the relationship tion, through an unknown immunologic mechanism, likely
between wall stress and risk for aneurysm rupture, several attracts inflammatory cells into the aortic wall (Figure).
JOURNAL OF VASCULAR SURGERY
Volume 38, Number 3 Ailawadi, Eliason, and Upchurch 587

Table IV. Polymorphisms associated with AAA

Polymorphism Patients with AAAs Patients without AAAs

Heme oxygenase-1 gene promoter Homozygous or heterozygous for less than Homozygous or heterozygous for less than
25 GT repeats, 41% 25 GT repeats, 59%
Angiotensin converting enzyme gene Normotension Normotensive control patients
DD genotype frequency, 70% DD genotype frequency, 25%
II genotype frequency, 5% II genotype frequency, 32%
Hypertension
DD genotype frequency, 32%
II genotype frequency, 21%
Apolipoprotein E gene E3E3 genotype: expansion rate, 2.1 mm/y
E3E4 genotype: expansion rate, 1.3 mm/y
E2E3 genotype: expansion rate, 3.1 mm/y
E2E4 genotype: expansion rate, 4.2 mm/y
Plasminogen activator inhibitor-1 Familial AAA Healthy control subjects
gene promoter 4G4G geneotype frequency, 20% 4G4G geneotype frequency, 35%
5G5G geneotype frequency, 26% 5G5G geneotype frequency, 13%
Nonfamilial AAA
4G4G geneotype frequency, 38%
5G5G geneotype frequency, 14%
TIMP-1 and TIMP-2 genes Female TIMP-1 nt 434 Female TIMP-1 nt 434
C allele frequency, 62% C allele frequency, 27%
T allele frequency, 38% T allele frequency, 73%
Male TIMP-2 nt 573 Male TIMP-2 nt 573
G allele frequency, 91% G allele frequency, 79%
A allele frequency, 9% A allele frequency, 21%
Endothelial nitric oxide synthase gene Patients with AAA requiring operation Healthy control subjects
5 repeat intron 4 allele frequency, 79% 5 repeat intron 4 allele frequency, 90%
4 repeat intron 4 allele frequency, 21% 4 repeat intron 4 allele frequency, 10%
Patients with AAA under observation
5 repeat intron 4 allele frequency, 96%
4 repeat intron 4 allele frequency, 4%
HLA-DR B1 gene Patients with inflammatory AAAs Healthy control subjects
HLA-DR B1*15 allele frequency, 47% HLA-DR B1*15 allele frequency, 27%
HLA-DR B1*0404 allele frequency, 14% HLA-DR B1*0404 allele frequency, 3%
AAA, Abdominal aortic aneurysm; TIMP, tissue inhibitor of metallsproteinase; HLA, human leukocyte antigen.

Proposed mechanism of aortic aneurysm formation. IEL, Internal elastic lamina; EEL, external elastic lamina. (Courtesy
of B. S. Knipp, MS, University of Michigan Medical School, Ann Arbor, Mich.)
JOURNAL OF VASCULAR SURGERY
588 Ailawadi, Eliason, and Upchurch September 2003

Inflammatory cells then release chemokines, cytokines, and 7. Lopez-Candales A, Holmes DR, Liao S, Scott MJ, Wickline SA,
reactive oxygen species, resulting in further influx of leuko- Thompson RW. Decreased vascular smooth muscle cell density in
medial degeneration of human abdominal aortic aneurysms. Am J
cytes, with subsequent expression and activation of induc- Pathol 1997;150:993-1007.
ible and constitutive proteases, notably MMPs. These pro- 8. Mc Millan WD, Pearce WH. Increased plasma levels of metalloprotein-
teases result in medial degradation and aneurysmal dilation, ase-9 are associated with abdominal aortic aneurysms. J Vasc Surg
with ongoing remodeling. Increased wall stress enhances 1999;29:122-9.
9. Yajima N, Masuda M, Miyazaki M, Nakajima N, Chien S, Shyy JY.
proteolysis and progressive aneurysm dilation, with even-
Oxidative stress is involved in the development of experimental abdom-
tual aortic rupture if untreated. inal aortic aneurysms: a study of the transcription profile with compli-
mentary DNA microarray. J Vasc Surg 2002;36:379-85.
FUTURE THERAPIES 10. Miller FJ Jr, Sharp WJ, Fang X, Oberley LW, Oberlely TD, Weintraub
Initial clinical observations followed by intense basic NL. Oxidative stress in human abdominal aortic aneurysms: a potential
mediator of aneurysm remodeling. Arterioscler Thromb Vasc Biol
science and translational research on the underlying patho- 2002;22:560-5.
genesis of aortic aneurysms have underscored the complex- 11. Moore JE Jr, Ku DN, Zarins CK, Glagov S. Pulsatile flow visualization
ity of this inflammatory process. Progress is being made on in the abdominal aorta under differing conditions: implications for
many fronts. For example, a better understanding of which increased susceptibility to atherosclerosis. J Biomech Eng 1992;114:
AAAs are at risk for rupture from increased wall stress is 391-7.
12. Peppin GJ, Weiss SJ. Activation of the endogenous metalloproteinase,
being developed. In addition, ongoing studies examining gelatinase, by triggered human neutrophils. Proc Natl Acad Sci U S A
the genetic basis for aneurysm disease will shed further light 1986;83:4322-6.
on the interactions between the aortic wall and circulating 13. Raghavan ML, Vorp DA, Federle MP, Makaroun MS, Webster MW.
factors important in aneurysm development. Wall stress distribution on three-dimensionally reconstructed models of
human abdominal aortic aneurysm. J Vasc Surg 2000;31:760-9.
To date, surgical and endovascular therapy for end-
14. Rajagopalan S, Meng XP, Ramasamy S, Harrison DG, Galis ZS. Reac-
stage aortic aneurysm disease is all we have to offer the tive oxygen species produced by macrophage-derived foam cells regu-
patient with an AAA. Risk factor modification, eg, cessation late the activity of vascular matrix metalloproteinases in vitro. J Clin
of cigarette smoking and control of hypertension, may help Invest 1996;98:2572-9.
to slow AAA growth and reduce coronary deaths. How- 15. Tambiah J, Powell JT. Chlamydia pneumoniae antigens facilitate exper-
imental aortic dilatation: prevention with azithromycin. J Vasc Surg
ever, strategies aimed at determining who is at risk for 2002;36:1011-7.
development of an aortic aneurysm and at slowing the 16. Thompson RW, Geraghty PJ, Lee JK. Abdominal aortic aneurysms:
growth of small AAAs once present are needed. Clinical basic mechanisms and clinical implications. Curr Probl Surg 2002;39:
trials using antiproteinases and anti-inflammatory agents 110-230.
17. Tung WS, Lee JK, Thompson RW. Simultaneous analysis of 1176 gene
should be performed.
products in normal human aorta and abdominal aortic aneurysms using
a membrane-based complementary DNA expression array. J Vasc Surg
REFERENCES 2000;34:143-50.
1. The abdominal aortic aneurysm: genetics, pathophysiology, molecular 18. Wang DH, Makaroun MS, Webster MW, Vorp DA. Effect of intralu-
biology. Proceedings of a conference. New York City, March 7-9, 1996. minal thrombus on wall stress in patient-specific models of abdominal
Ann N Y Acad Sci 1996;800:1-300. aortic aneurysm. J Vasc Surg 2002;36:598-604.
2. Ailawadi G, Knipp BS, Lu G, Roelofs KJ, Ford JW, Hannawa KK et al. 19. Wassef M, Baxter BT, Chisholm RL, Dalman RL, Fillinger MF, Hei-
A nontrinsic regional basis for increased infrarenal aortic MMP-9 ex- necke J, et al. Pathogenesis of abdominal aortic aneurysms: a multidis-
pression and activity. J Vasc Surg 2003;37:1059-66. ciplinary research program supported by the National Heart, Lung and
3. Davies MJ. Aortic aneurysm formation: lessons from human studies and Blood Institute. J Vasc Surg 2001;34:730-8.
experimental models. Circulation 1998;98:193-5. 20. Wolinsky H, Glagov S. Comparison of abdominal and thoracic aortic
4. Fillinger MF, Raghavan ML, Marra SP, Cronenwett JL. In vivo analysis medial structure in mammals: deviation of man from the usual pattern.
of mechanical wall stress and abdominal aortic aneurysm rupture risk. J Circ Res 1969;25:677-86.
Vasc Surg 2002;36:589-97. 21. Xia S, Ozvath K, Hirose H, Tilson MD. Partial amino acid sequence of
5. Grange JJ, Davis V, Baxter BT. Pathogenesis of abdominal aortic a novel 40-kDa human aortic protein, with birtonectin-like, fibrinogen-
aneurysm: an update and look toward the future. Cardiovasc Surg like, and calcium binding domains: aortic aneurysm-associated pro-
1997;5:256-65. tein-40 (AAAP-40). Biochem Biophys Res Comm 1996;219:36-9.
6. Li PF, Dietz R, Von Harsdorf R. Reactive oxygen species induce
apoptosis of vascular smooth muscle cell. FEBS Lett 1997;404:249-52. Submitted Feb 5, 2003; accepted Feb 12, 2003.

You might also like