Anda di halaman 1dari 7

Brief Reviews

Transforming Growth Factor ␤ and Atherosclerosis: So Far,


So Good for the Protective Cytokine Hypothesis
David J. Grainger

Abstract—The role of the anti-inflammatory cytokine transforming growth factor ␤ (TGF-␤) in atherosclerosis has been
the subject of considerable debate for a decade. In the early 1990s, we postulated that TGF-␤ played an important role
in maintaining normal vessel wall structure and that loss of this protective effect contributed to the development of
atherosclerosis. We termed this the protective cytokine hypothesis. This proposal was slow to gain broad acceptance,
however, because at that time there were little data available on the role of TGF-␤ during the development of
atherosclerosis but much information about its role during trauma-induced neointima formation. Because TGF-␤
apparently aggravates neointima formation, both by inhibiting endothelial regeneration and by promoting fibrosis, it was
difficult to accept that its presence might ameliorate the superficially similar atherogenesis process. But several recent
studies revealed beyond doubt the fact that TGF-␤ protects against lipid lesion formation, at least in mouse models of
atherosclerosis. Therefore, two important questions remain. First, is the role of TGF-␤ in vascular biology similar in
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

humans and in mice? Secondly, how important, compared with defects in thrombosis or lipoprotein metabolism, is the
protective role of TGF-␤ during atherogenesis? (Arterioscler Thromb Vasc Biol. 2004;24:399-404.)
Key Words: TGF-␤ 䡲 myocardial infarction 䡲 inflammation

he transforming growth factor ␤ (TGF-␤) superfamily


T consists of a large number of structurally related cyto-
kines that participate in a vast range of biological processes
under the majority of conditions, and particularly under any
conditions in which proliferation was stimulated by growth
factors, that TGF-␤ inhibits smooth muscle proliferation.8 –10
from axis specification and tissue differentiation in early The small stimulatory effects seen in the early studies most
development through to regulation of a range of immune likely owe their existence to the exceptional ability of TGF-␤
functions in the adult organism. Members of the TGF-␤ to promote extracellular matrix (ECM) formation. Cells
superfamily are typically produced as dimeric latent precur- placed into culture simply adhere to the culture plastic better
sors with no biological activity, which are subsequently if TGF-␤ is present to stimulate ECM production. In addition
activated in the extracellular environment to release the to its affect on VSMC proliferation, TGF-␤ also inhibited
receptor-binding ligands that share a common structural motif VSMC migration11 and promoted the expression of an array
known as the cysteine knot. Several members of the TGF-␤ of proteins that make-up the contractile apparatus of the
superfamily are likely to play a role in vascular biology. cell.8 –10 In short, TGF-␤ possessed a portfolio of coordinated
TGF-␤1 (the first member of the family to be discovered and effects in vitro that would contribute to the maintenance of
the best-studied member to date) is present at high levels in the normal blood vessel wall architecture if similar effects
the healthy blood vessel wall, whereas the closely related occurred in vivo.5
TGF-␤2 and TGF-␤3 isoforms are either absent (␤2) or Early in its existence, the protective cytokine idea was
present only at low levels (␤3). Of the remaining members of expanded beyond the smooth muscle component of the vessel
the superfamily, activin1 and several of the bone morphoge- wall. Gene knockout studies demonstrated that TGF-␤ ex-
netic proteins2,3 are known to be expressed in the blood vessel erted a dramatic anti-inflammatory signal in constitutive
wall and, although substantially less well-studied than TGF- fashion. Loss of TGF-␤1 resulted in perinatal death after
␤1, are thought to play broadly similar roles. unbridled leukocyte extravasation in almost every organ
The protective cytokine hypothesis has its origins in cell system.12,13 Because accumulation of leukocytes, particularly
culture studies performed to examine the effects of TGF-␤ on macrophages but also T cells, is a characteristic signature of
vascular smooth muscle cells (VSMCs).4,5 Although some of the change from normal vessel wall structure to early athero-
the earliest studies suggested that TGF-␤ could promote sclerotic lesion,14 we postulated that lowered TGF-␤ activity
smooth muscle proliferation,6,7 it soon became clear that in the vessel wall might lead to leukocyte accumulation, in

Received July 9, 2003; revision accepted November 26, 2003.


From the Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK.
Sources of Support: D.J.G. is supported by the British Heart Foundation.
Correspondence to David J. Grainger, Department of Medicine, University of Cambridge, Box 157, Addenbrooke’s Hospital, Cambridge, CB2 2QQ,
UK. E-mail djg15@cam.ac.uk
© 2004 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org DOI: 10.1161/01.ATV.0000114567.76772.33

399
400 Arterioscler Thromb Vasc Biol. March 2004

parallel with increased VSMC proliferation, migration, and


dedifferentiation.5
Over a period of years, a range of other potentially
protective effects of TGF-␤ in cell culture were identified,
including suppression of proinflammatory adhesion molecule
expression by the vascular endothelium,15 and reduced foam
cell formation in cultured macrophages.16 Recently, Mallat
and Tedgui17 have provided an excellent overview of the
immunomodulatory role of TGF-␤ during the development of
atherosclerosis, in which they highlight the central role of
TGF-␤ in the regulation of T-cell biology and it implications
for the chronic inflammatory component of atherogenesis.
Recent evidence from Gojova et al18 underlines the role of
TGF-␤ as a deactivating cytokine for T cells, because
disrupting TGF-␤ signaling specifically in the T-cell popula- Figure 1. A model for the action of TGF-␤ on VSMCs during
atherogenesis. In the normal vessel wall (left cycle), latent
tion increased the number of T cells, as well as MHC class TGF-␤ is efficiently converted into active TGF-␤ (black line),
II-positive macrophages, in the lesions. These T-cell–rich which signals to cells rich in type II TGF-␤ receptor (RII). This
lesions had a characteristic phenotype of rupture-prone maintains normal vessel wall structure and a high level of RII on
the VSMCs. If TGF-␤ activity is transiently diminished (red line;
plaques in humans: lowered collagen content and decreased
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

the “first hit”), then RII levels decrease and the vessel wall shifts
representation of smooth muscle cells expressing smooth to the right cycle. While TGF-␤ activity remains adequate, sig-
muscle-specific actin isoforms. As Mallat and Tedgui con- naling to cells rich in type I TGF-␤ receptor (RI) stimulates ECM
clude,17 the effect of TGF-␤ on T-cell differentiation is expression and a stable plaque phenotype results. Physiologi-
cally, this process is irreversible because TGF-␤ activity no
undoubtedly an important component of TGF-␤ action during longer promotes RII expression. If TGF-␤ activity is again
atherogenesis. reduced (thin red line; the “second hit”), inflammation and loss
However, TGF-␤ signaling in the inflammatory cell pop- of ECM results converting the lesion to an unstable phenotype
prone to rupture.
ulation cannot be the whole story: Gojova et al18 report a
small but significant decrease in lesion size in the aortic sinus
when TGF-␤ signaling is abrogated specifically in the T-cell matrix-rich lesions), but to lose TGF-␤ activity repeatedly is
population, whereas generalized suppression of TGF-␤ levels potentially fatal (leading to formation of leukocyte-rich,
(using neutralizing antibodies,19 soluble receptor fragments,20 matrix-poor lesions that are at high risk for rupture).
or heterozygous deletion of the tgfb1 gene21) led to either no More recently, a plausible molecular basis for this double
change in lesion size or an increase in lesion size. This is role of TGF-␤ during atherogenesis has evolved: McCaffrey
likely to reflect the complexity of the role of TGF-␤ in the et al demonstrated that VSMCs (the cells that make all the
vessel wall, with important effects not only on the leukocyte ECM in the vessel wall) had different TGF-␤ receptor
populations but also on the endothelial and vascular smooth expression profiles in atherosclerotic lesions compared with
muscle cell compartments. This review therefore comple- normal vessel wall.23 In the diseased vessel, the cells domi-
ments the article by Mallat and Tedgui17 by focusing primar- nantly expressed the type I TGF-␤ receptor, whereas in
ily on the impact of TGF-␤ signaling on the vascular smooth normal vessel wall the type II receptors dominated. This
muscle cell population in the vessel wall. change had functional consequences, at least in vitro. The
This interplay between the leukocyte and smooth muscle type I dominated VSMCs responded to TGF-␤ very differ-
cell compartments is likely to be particularly important when ently from the type II dominated cells, by massively elevating
considering the impact of TGF-␤ signaling on ECM produc- ECM production.23 In contrast, the type II dominated cells
tion. Elaboration of ECM is one of the major functions of the barely increase ECM production at all in response to TGF-␤.
smooth muscle cell, and the balance between leukocytes and We confirmed and extended these studies, demonstrating that
smooth muscle cell populations in the developing atheroscle- when VSMCs from healthy artery are first dispersed into cell
rotic plaque is therefore a major determinant of plaque culture, they have high levels of the type II receptor and
stability.22 Consequently, TGF-␤ is likely to play a central respond to TGF-␤ by increasing contractile protein expres-
role in regulating this “stability balance” in at least two sion, but not ECM production. In the absence of added
distinct ways: (1) through its immunoregulatory role it TGF-␤, over a number of cell cycles in culture, the ratio of
suppresses leukocyte recruitment; and (2) it is one of the most type II to type I receptor decreases, and the cells switch their
powerful fibrogenic cytokines described to date. TGF-␤ responses over. Now TGF-␤ stimulates ECM production but
therefore not only protects against the initial formation of not contractile protein expression. Most interestingly, if the
fatty streak lesions by maintaining normal blood vessel wall freshly dispersed cells were maintained continually in
structure19,21 but also protects against the development of TGF-␤, they maintained high levels of type II receptor
unstable lesions by driving matrix production by the smooth indefinitely in culture, and with it maintained a differentiated
muscle cells of the fibrous cap.19,20 phenotype with relatively low ECM production.
Potentially, suppression of TGF-␤ signaling is progres- Taken together, these studies allow a plausible molecular
sively and doubly dangerous. To lose TGF-␤ activity tempo- model for the role of TGF-␤ in the vessel wall to be
rarily is merely careless (leading to formation of stable constructed (Figure 1). High levels of TGF-␤ maintain a
Grainger Protective Cytokine Hypothesis 401

feedback loop in which type II receptor remains high and the TGF-␤1 must be reduced to promote susceptibility to
vessel wall retains its differentiated phenotype with lots of atherogenesis.
contractile proteins and relatively little ECM. If some exter- More recently, several other groups have used different
nal factor intervenes to reduce TGF-␤ activity sufficiently in methodological approaches to draw similar conclusions. Both
a particular region, the VSMCs respond not only by dedif- Mallat et al19 and Lutgens et al20 used in vivo neutralization
ferentiating, proliferating, and migrating into the vessel in- approaches to show that depletion of TGF-␤1 in the blood
tima but also by reducing type II receptor expression. This is vessel wall of adult apoE-deficient mice was sufficient to
the “first hit” of reduced TGF-␤ activity that promotes early exacerbate lipid lesion development. The amount of lipid
fatty streak lesion formation. deposited was increased in both studies, as was the number of
However, the changes in TGF-␤ receptor expression pat- macrophages and other inflammatory cells accumulating in
terns has the effect of locking in the vessel wall changes that the developing lesion. Lutgens et al20 went on to demonstrate
have occurred, because even if TGF-␤ activity returns to that neutralizing TGF-␤ decreased ECM deposition, confirm-
normal, those dedifferentiated intimal VSMCs respond by ing the role of this cytokine in regulating the balance between
producing ECM rather than by returning to the contractile an unstable, proinflammatory lesion phenotype and a stable,
state. The result would be a stable matrix-rich atherosclerotic matrix-rich phenotype.
plaque. However, if TGF-␤ activity remained suppressed or Taken together, these studies provide strong evidence that
was lowered still further, then the “second hit” of reduced TGF-␤ protects against unstable plaque development in at
TGF-␤ activity occurs. Enhanced recruitment of leukocytes least two ways. TGF-␤ acts constitutively to suppress the
changes in vessel wall architecture that are associated with
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

to the plaque coupled with reduced ECM production by the


remaining VSMCs results in formation of a rupture-prone lipid lesion development in mice19,21 (the “first hit” of
plaque typical of those that go on to have clinical sequelae, reduced TGF-␤). Decreased TGF-␤ activity later during the
including myocardial infarction and death. atherogenic process results in a switch to an unstable inflam-
Such a model provides one possible explanation for a range matory lesion phenotype19,20 (the “second hit” of reduced
TGF-␤).
of features of the atherogenic process, but is it correct? And
what might trigger localized suppression of TGF-␤ activity?
What Factors Might Trigger Suppression of
Direct Evidence Supports the Protective TGF-␤ Activity in Humans?
One of the best understood local triggers for suppression of
Cytokine Hypothesis in Mice TGF-␤ activity in the blood vessel wall is the variant
Genetic manipulation has become the standard method for
lipoprotein particle Lp(a). Lp(a) consists of an additional
determining the importance of gene products in disease
protein component, apo(a), covalently bonded to the apoB of
processes. The gene of interest is knocked out, and the impact
an LDL particle.27 The distinguishing apo(a) protein has
on disease progression monitored. Unfortunately, the tgfb1
sequence homology to plasminogen but has been inactivated
null mouse dies soon after birth from severe multifocal
in the catalytic triad, rendering it incapable of yielding an
inflammation,12,13 preventing us from studying vessel wall
enzymatically active form equivalent to plasmin. Rifkin et al
architecture in the absence of TGF-␤1. Two approaches have first demonstrated that Lp(a) could inhibit TGF-␤ activation
been taken to get around this problem: tgfb1 null mice can be in cell culture,11 an observation that has been repeated many
bred on a SCID background, allowing them to survive into times.28,29 We extended these studies and demonstrated that
adulthood;24 alternatively, mice heterozygous for the tgfb1 transgenic expression of apo(a) in mice, which have no
deletion are studied.21,25,26 These tgfb1⫹/⫺ mice have lower equivalent endogenous gene, results in impaired TGF-␤
levels of TGF-␤1 in a number of tissues,25 including the activation in the blood vessel wall.30 In a further study, we
blood vessel wall,21,26 and provide a direct method to study demonstrated that apo(a) accumulates preferentially at sites
the impact of lowered TGF-␤1 levels on vessel wall where TGF-␤ activation is already suppressed, leading to a
architecture. positive feedback loop that explains the highly focal suppres-
Adult tgfb1⫹/⫺ mice have reduced levels of smooth muscle sion of TGF-␤ activity in apo(a) and Lp(a) transgenic mice.31
differentiation markers, including smooth muscle specific-␣- Feedback loops such as these may contribute to the focal
actin and smooth muscle-specific myosin heavy chain in the nature of atherosclerotic lesions, which affect some regions of
aortic wall,21,26 consistent with the expected effect of reduced the vasculature but not others within the same individual. To
levels of TGF-␤1. However, neither tgfb1⫺/⫺ mice on a SCID date, however, there are no data to determine whether the
background24 nor tgfb1⫹/⫺ mice21 showed signs of endothelial presence of Lp(a) lowers TGF-␤ activation in humans.32
activation (marked by VCAM-1 and ICAM-1 expression). A range of other factors have also been postulated to
Interestingly, when the mice were fed a high-fat diet, endo- regulate TGF-␤ activation (Figure 2), including proteases that
thelial activation and lipid deposition was observed in the are thought to cleave the LAP thereby promoting release of
tgfb1⫹/⫺ mice but not in their wild-type littermate controls.21 mature TGF-␤, binding proteins that alter the conformation of
These observations demonstrate that reduced TGF-␤1 pro- the complex promoting its dissociation, and physicochemical
duction renders the vessel wall susceptible to endothelial factors (such as acidity and radiation) that also favor disso-
activation and development of early stage lipid lesions when ciation of protein complexes. Recently, Rifkin et al33 have
subjected to a proatherogenic challenge. This study, however, proposed a unifying scheme, suggesting that the LAP dimer
provides no information as to when or where the level of acts as a “sensor” component of the latent TGF-␤ complex,
402 Arterioscler Thromb Vasc Biol. March 2004

signaling receptors,36 which may exacerbate the reduction in


TGF-␤ activity seen after dietary fat loading.
TGF-␤ activity may also be suppressed at regions of low
shear stress (for example, at vessel branch points37). These
preliminary observations in human studies have been repli-
cated in studies of the rodent vasculature.26 Again, several
molecular mechanisms may contribute, including a direct
shear response element in the tgfb1 gene promoter38 and a
flow-dependency in the expression pattern of the inhibitory
Smad proteins, Smad6 and Smad7.39
As in mice, the definitive experiment to implicate TGF-␤
in the atherogenesis process in humans would be to identify
an association between tgfb1 genotype and disease. Although
the experiments are more technically demanding, there have
been a number of genetic studies that have attempted to
determine whether polymorphisms in the tgfb1 gene locus are
associated with either myocardial infarction (a measure of
plaque stability as well as plaque load) or angiographically
defined luminal stenosis (a measure of plaque load). The first
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

Figure 2. Factors affecting the activation of TGF-␤1. Members


of the TGF-␤ superfamily are produced as pre-pro-proteins that study, by Cambien et al40 in the ECTIM cohort, found an
dimerize and lose the signal sequence during secretion. The association between polymorphisms in the TGF-␤1 signal
resulting pro-protein dimer must then undergo a 2-stage activa- peptide region of the gene and myocardial infarction but no
tion process to yield the active TGF-␤ ligand. In the first step, a association with angiographically defined arterial stenosis. A
member of the pro-protein convertase family, such as furin,
cleaves the polypeptide backbone at a tetrabasic site between follow-up study confirmed the lack of association between
the pro-peptide (termed the latency associated peptide or LAP) several tgfb1 polymorphisms and angiographically defined
and the mature TGF-␤ dimer. The LAP dimer and mature TGF-␤ disease.41 However, a recent study in Japanese men42 found a
dimer remain noncovalently associated, and this structure
(termed latent TGF-␤) has no known biological activity. In a sec-
larger association between tgfb1 polymorphisms and myocar-
ond step, the LAP becomes dissociated from the mature TGF-␤ dial infarction than that originally reported by Cambien.
dimer, which is thereby released to interact with the signaling Taken together, these studies provide tentative support for the
receptors. The precise mechanism of this second step is still protective cytokine hypothesis in humans: polymorphisms
poorly understood, but it is known to be promoted by a wide
range of factors (including proteases, physiochemical mediators, associated with lower TGF-␤1 secretion are over-represented
and the binding of other proteins). Several inhibitors of this acti- among men with myocardial infarction in several popula-
vation step have also been described. It is likely that the LAP is tions.43 The lack of association with angiographically defined
acting as a “sensor” component of the complex, integrating the
levels of a range of environmental factors to determine how
vessel stenosis41 may suggest that in humans, the role of
much of the “effector” mature TGF-␤ should be released. A third TGF-␤ is more associated with the switch between stable
component of the complex (namely various TGF-␤ binding pro- matrix-rich lesions and unstable inflammatory lesions (the
teins) that acts as a “localizer” to regulate the tissue distribution “second hit” of reduced TGF-␤) than in determining the total
of the complex is not shown for clarity. MMP indicates matrix
metalloproteinases; M-6-P receptor, cation-independent atherosclerotic plaque load (the “first hit”).17,19,20
mannose-6-phosphate receptor. Unfortunately, a more clear-cut resolution of this question
is unlikely to come from genetic studies of the tgfb1 gene.
integrating the activity of multiple environmental factors to Because TGF-␤1 is involved in such a wide range of
tightly regulate the release of the “effector” TGF-␤ complex. physiological and pathological functions, polymorphisms or
mutations that caused significant changes to the temporal,
Thus, factors such as PAI-1 that are known to be elevated
spatial, or quantitative production of TGF-␤ would likely
during atherogenesis,34 or urokinase and plasmin activity
have effects that manifested themselves much earlier in life
(which are known to be suppressed34), are likely to contribute
than atherosclerosis. Unlike in mice, in which definitive
to local suppression of TGF-␤ activity at the site of vascular
experiments were possible, it is likely that a fuller under-
lipid lesion formation. standing of the role of TGF-␤ in human cardiovascular
Various mechanisms also link defects in lipid metabolism disease will have to come from the weight of evidence
to suppression of TGF-␤ activity. After a standardized yielded by an array of experimental approaches.
high-fat meal, TGF-␤ activation is transiently suppressed.
Several mechanisms may contribute to this. PAI-1 production How Important Is TGF-␤ Signaling
is stimulated by dietary fat load, to an extent that depends on in Atherogenesis?
the 4G/5G genotype in the PAI-1 promoter.35 Suppression of Recent studies from a range of groups have confirmed that in
TGF-␤ activation was similarly associated with the PAI-1 rodents, suppression of TGF-␤ activity results in increased
promoter genotype, suggesting that PAI-1 may contribute to lipid lesion deposition during early lesion development and a
the reduction in TGF-␤ activity. Additionally, the hydropho- switch from a stable, matrix-rich plaque phenotype to an
bic TGF-␤ protein can be sequestered into triglyceride-rich unstable, proinflammatory plaque at risk for rupture.19 –21 The
lipoprotein particles, reducing its ability to interact with its available evidence suggest that similar pathways are likely to
Grainger Protective Cytokine Hypothesis 403

genetic factors associated with development of atherosclero-


sis. It is possible, therefore, that it occupies an important
nexus in the web of interacting pathways controlling blood
vessel wall structure. The challenge now lies in quantifying
the contribution of aberrant TGF-␤ activity to the develop-
ment of clinically significant atherosclerosis in the human
population. Unfortunately, there are no simple experiments
that will conclusively determine the relative importance of
suppressed TGF-␤ activity as a risk factor for myocardial
infarction. Instead, a gradual understanding is likely to evolve
as genetic and epidemiological evidence accumulates over a
period of years.
This task has more than academic importance. Identifica-
tion of the most prevalent pathways contributing to athero-
Figure 3. A 3-pathway model for atherogenesis. Epidemiology genesis is a key step to improving current clinical manage-
and reverse genetic approaches have unequivocally demon- ment of coronary heart disease. The widespread clinical
strated that there are at least 3 major pathways (represented as
colored circles) contributing to the risk of myocardial infarction. benefit of treatment with lipid-lowering therapies and anti-
For some individuals, a major defect in a single pathway is thrombotic agents is now well-established, yet tens of thou-
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

dominantly responsible. For individual 1, a major defect in sands of myocardial infarction events still occur each year in
lipoprotein metabolism is the major contributor to their risk. For
individual 2, a major defect in fibrinolysis might be responsible.
subjects receiving the current best therapy. By identifying
We propose, however, that for most individuals with coronary further pathways, independent of lipid metabolism and
artery disease (represented by individual 3), it is a combination thrombosis, which contribute to the disease process, we can
of mild defects in all 3 pathways that results in high risk for develop additional therapeutic options that may be effective
myocardial infarction. Effective preventative therapy will depend
on targeting each of the pathways contributing to the risk for a in at least a proportion of the population. To this end, we and
given individual. others have proposed the use of triphenylethylene-derived
drugs, such as tamoxifen44 or raloxifene,45 which stimulate
be operating in humans. Thus, defects in TGF-␤ metabolism the production of TGF-␤46,47 as therapies for coronary artery
join an ever-growing list of pathways that have been plausi- disease. The recent articles19 –21 describing direct evidence for
bly linked to the development of atherosclerosis resulting in the protective cytokine hypothesis in rodents provide yet
myocardial infarction, including defects in lipoprotein metab- further impetus toward clinical trials of these TGF-␤–stimu-
olism and imbalance between thrombotic and fibrinolytic lating drugs in humans.
cascades.
It is no longer sufficient merely to ask whether defects in Acknowledgments
particular pathways might (under certain circumstances) con- I am grateful to Profs Jim Metcalfe and Peter Weissberg (University
tribute to the an increased risk for myocardial infarction. of Cambridge, UK) for their support during the formulation of the
Instead, we need to ask how much of the variation in protective cytokine hypothesis, and to Dr David Mosedale for
susceptibility to myocardial infarction across the human helpful discussions over many years. D.J.G. is a British Heart
Foundation Senior Research Fellow.
population can be attributed to each of these pathways. It is
likely that for different individuals, different pathways may
dominate (Figure 3). For some individuals, such as those with References
1. Engelse MA, Neele JM, van Achterberg TA, van Aken BE, van Schaik
familial hypercholesterolemia caused by genetic defects in
RH, Pannekoek H, de Vries CJ. Human activin-A is expressed in the
the LDL receptor protein and some individuals with less atherosclerotic lesion and promotes the contractile phenotype of smooth
severe lipoprotein metabolism defects, the lipoprotein traf- muscle cells. Circ Res. 1999;85:931–939.
ficking and metabolism defect may be the dominant cause of 2. Dhore CR, Cleutjens JP, Lutgens E, Cleutjens KB, Geusens PP, Kitslaar
PJ, Tordoir JH, Spronk HM, Vermeer C, Daemen MJ. Differential
the increased risk of myocardial infarction by increasing the expression of bone matrix regulatory proteins in human atherosclerotic
atherosclerotic burden. For others, increased susceptibility to plaques. Arterioscler Thromb Vasc Biol. 2001;21:1998 –2003.
intravascular coagulation may be the dominant cause of their 3. Willette RN, Gu JL, Lysko PG, Anderson KM, Minehart H, Yue T.
increased risk. Yet for the majority, it seems plausible that the BMP-2 gene expression and effects on human vascular smooth muscle
cells. J Vasc Res. 1999;36:120 –125.
development of atherosclerosis and, ultimately, its progres- 4. Metcalfe JC, Grainger DJ. Transforming growth factor-beta and the
sion to yield myocardial infarction result from the combined protection from cardiovascular injury hypothesis. Biochem Soc Trans.
effects of milder defects in a number of contributing path- 1995;23:403– 406.
ways. Increased inflammatory responses, whether systemic or 5. Grainger DJ, Metcalfe JC. A pivotal role for TGF-beta in atherogenesis?
Biol Rev Camb Philos Soc. 1995;70:571–596.
local to the vessel wall, seem likely to contribute to the total 6. Stouffer GA, Owens GK. Angiotensin II-induced mitogenesis of sponta-
atherosclerotic burden and to the tendency to the develop- neously hypertensive rat-derived cultured smooth muscle cells is
ment of unstable, leukocyte-rich plaques that predispose to dependent on autocrine production of transforming growth factor-beta.
Circ Res. 1992;70:820 – 828.
myocardial infarction.
7. Battegay EJ, Raines EW, Seifert RA, Bowen-Pope DF, Ross R. TGF-beta
Our studies to date suggest that reduced TGF-␤ activity is induces bimodal proliferation of connective tissue cells via complex
a common consequence of a range of environmental and control of an autocrine PDGF loop. Cell. 1990;63:515–524.
404 Arterioscler Thromb Vasc Biol. March 2004

8. Bjorkerud S. Effects of transforming growth factor-beta 1 on human 28. Grainger DJ, Kirschenlohr HL, Metcalfe JC, Weissberg PL, Wade DP,
arterial smooth muscle cells in vitro. Arterioscler Thromb. 1991;11: Lawn RM. Proliferation of human smooth muscle cells promoted by
892–902. lipoprotein(a). Science. 1993;260:1655–1658.
9. Grainger DJ, Kemp PR, Witchell CM, Weissberg PL, Metcalfe JC. 29. Takami S, Yamashita S, Kihara S, Ishigami M, Takemura K, Kume N,
Transforming growth factor beta decreases the rate of proliferation of rat Kita T, Matsuzawa Y. Lipoprotein(a) enhances the expression of inter-
vascular smooth muscle cells by extending the G2 phase of the cell cycle cellular adhesion molecule-1 in cultured human umbilical vein endothe-
and delays the rise in cyclic AMP before entry into M phase. Biochem J. lial cells. Circulation. 1998;97:721–728.
1994;299(Pt 1):227–35. 30. Grainger DJ, Kemp PR, Liu AC, Lawn RM, Metcalfe JC. Activation of
10. Owens GK, Geisterfer AA, Yang YW, Komoriya A. Transforming transforming growth factor-beta is inhibited in transgenic apoli-
growth factor-beta-induced growth inhibition and cellular hypertrophy in poprotein(a) mice. Nature. 1994;370:460 – 462.
cultured vascular smooth muscle cells. J Cell Biol. 1988;107:771–780. 31. Lawn RM, Pearle AD, Kunz LL, Rubin EM, Reckless J, Metcalfe JC,
11. Kojima S, Harpel PC, Rifkin DB. Lipoprotein (a) inhibits the generation Grainger DJ. Feedback mechanism of focal vascular lesion formation in
of transforming growth factor beta: an endogenous inhibitor of smooth transgenic apolipoprotein(a) mice. J Biol Chem. 1996;271:31367–31371.
muscle cell migration. J Cell Biol. 1991;113:1439 –1445. 32. Grainger DJ, Metcalfe JC. Transforming growth factor-beta: the key to
12. Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, Flanders KC, understanding lipoprotein(a)? Curr Opin Lipidol. 1995;6:81– 85.
Roberts AB, Sporn MB, Ward JM, Karlsson S. Transforming growth 33. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta
factor beta 1 null mutation in mice causes excessive inflammatory activation. J Cell Sci. 2003;116:217–224.
response and early death. Proc Natl Acad Sci U S A. 1993;90:770 –774. 34. Martin-Paredero V, Vadillo J, Diaz J, Espinosa A, Berga C, Segura J,
13. Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, Allen Sanchez J, Barbod A, Villaverde C, Richard CM. Fibrinogen and fibri-
R, Sidman C, Proetzel G, Calvin D, et al. Targeted disruption of the nolysis in blood and in the arterial wall: its role in advanced athero-
mouse transforming growth factor-␤ 1 gene results in multifocal inflam- sclerotic disease. Cardiovasc Surg. 1998;6:457– 462.
matory disease. Nature. 1992;359:693– 699. 35. Byrne CD, Wareham NJ, Martensz ND, Humphries SE, Metcalfe JC,
14. Ross R. Atherosclerosis–an inflammatory disease. N Engl J Med. 1999; Grainger DJ. Increased PAI activity and PAI-1 antigen occurring with an
oral fat load: associations with PAI-1 genotype and plasma active
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

340:115–126.
15. Gamble JR, Khew-Goodall Y, Vadas MA. Transforming growth TGF-beta levels. Atherosclerosis. 1998;140:45–53.
factor-beta inhibits E-selectin expression on human endothelial cells. 36. Grainger DJ, Byrne CD, Witchell CM, Metcalfe JC. Transforming growth
J Immunol. 1993;150:4494 – 4503. factor beta is sequestered into an inactive pool by lipoproteins. J Lipid
16. Argmann CA, Van Den Diepstraten CH, Sawyez CG, Edwards JY, Res. 1997;38:2344 –2352.
37. Borkowski P, Robinson MJ, Kusiak JW, Borkowski A, Brathwaite C,
Hegele RA, Wolfe BM, Huff MW. Transforming growth factor-beta1
Mergner WJ. Studies on TGF-beta 1 gene expression in the intima of the
inhibits macrophage cholesteryl ester accumulation induced by native and
human aorta in regions with high and low probability of developing
oxidized VLDL remnants. Arterioscler Thromb Vasc Biol. 2001;21:
atherosclerotic lesions. Mod Pathol. 1995;8:478 – 482.
2011–2018.
38. Ohno M, Cooke JP, Dzau VJ, Gibbons GH. Fluid shear stress induces
17. Mallat Z, Tedgui A. The role of transforming growth factor beta in
endothelial transforming growth factor beta-1 transcription and produc-
atherosclerosis: novel insights and future perspectives. Curr Opin Lipidol.
tion. Modulation by potassium channel blockade. J Clin Invest. 1995;95:
2002;13:523–529.
1363–1369.
18. Gojova A, Brun V, Esposito B, Cottrez F, Gourdy P, Ardouin P, Tedgui
39. Topper JN, Cai J, Qiu Y, Anderson KR, Xu YY, Deeds JD, Feeley R,
A, Mallat Z, Groux H. Specific abrogation of transforming growth factor-
Gimeno CJ, Woolf EA, Tayber O, Mays GG, Sampson BA, Schoen FJ,
{beta} signaling in T cells alters atherosclerotic lesion size and compo-
Gimbrone MA, Jr., Falb D. Vascular MADs: two novel MAD-related
sition in mice. Blood. 2003. genes selectively inducible by flow in human vascular endothelium. Proc
19. Mallat Z, Gojova A, Marchiol-Fournigault C, Esposito B, Kamate C, Natl Acad Sci U S A. 1997;94:9314 –9319.
Merval R, Fradelizi D, Tedgui A. Inhibition of transforming growth 40. Cambien F, Ricard S, Troesch A, Mallet C, Generenaz L, Evans A,
factor-beta signaling accelerates atherosclerosis and induces an unstable Arveiler D, Luc G, Ruidavets JB, Poirier O. Polymorphisms of the
plaque phenotype in mice. Circ Res. 2001;89:930 –934. transforming growth factor-beta 1 gene in relation to myocardial
20. Lutgens E, Gijbels M, Smook M, Heeringa P, Gotwals P, Koteliansky infarction and blood pressure. The Etude Cas-Temoin de l’Infarctus du
VE, Daemen MJ. Transforming growth factor-beta mediates balance Myocarde (ECTIM) Study. Hypertension. 1996;28:881– 887.
between inflammation and fibrosis during plaque progression. Arte- 41. Syrris P, Carter ND, Metcalfe JC, Kemp PR, Grainger DJ, Kaski JC,
rioscler Thromb Vasc Biol. 2002;22:975–982. Crossman DC, Francis SE, Gunn J, Jeffery S, Heathcote K. Transforming
21. Grainger DJ, Mosedale DE, Metcalfe JC, Bottinger EP. Dietary fat and growth factor-␤1 gene polymorphisms and coronary artery disease. Clin
reduced levels of TGF␤1 act synergistically to promote activation of the Sci (Lond). 1998;95:659 – 667.
vascular endothelium and formation of lipid lesions. J Cell Sci. 2000; 42. Yokota M, Ichihara S, Lin TL, Nakashima N, Yamada Y. Association of
113:2355–2361. a T29 –⬎C polymorphism of the transforming growth factor-␤1 gene
22. Robbie L, Libby P. Inflammation and atherothrombosis. Ann N Y Acad with genetic susceptibility to myocardial infarction in Japanese. Circu-
Sci. 2001;947:167–180. lation. 2000;101:2783–2787.
23. McCaffrey TA, Consigli S, Du B, Falcone DJ, Sanborn TA, Spokojny 43. Andreotti F, Porto I, Crea F, Maseri A. Inflammatory gene poly-
AM, Bush HL, Jr. Decreased type II/type I TGF-beta receptor ratio in morphisms and ischaemic heart disease: review of population association
cells derived from human atherosclerotic lesions. Conversion from an studies. Heart. 2002;87:107–112.
antiproliferative to profibrotic response to TGF-␤1. J Clin Invest. 1995; 44. Grainger DJ, Metcalfe JC. Tamoxifen: teaching an old drug new tricks?
96:2667–2675. Nat Med. 1996;2:381–385.
24. Diebold RJ, Eis MJ, Yin M, Ormsby I, Boivin GP, Darrow BJ, Saffitz JE, 45. Saitta A, Morabito N, Frisina N, Cucinotte D, Corrado F, D’Anna R,
Doetschman T. Early-onset multifocal inflammation in the transforming Altavilla D, Squadrito G, Minutoli L, Arcoraci V, Cancellieri F, Squadrito
growth factor beta 1-null mouse is lymphocyte mediated. Proc Natl Acad F. Cardiovascular effects of raloxifene hydrochloride. Cardiovasc Drug
Sci U S A. 1995;92:12215–12219. Rev. 2001;19:57–74.
25. Tang B, Bottinger EP, Jakowlew SB, Bagnall KM, Mariano J, Anver MR, 46. Yang NN, Bryant HU, Hardikar S, Sato M, Galvin RJ, Glasebrook AL,
Letterio JJ, Wakefield LM. Transforming growth factor-␤1 is a new form Termine JD. Estrogen and raloxifene stimulate transforming growth
of tumor suppressor with true haploid insufficiency. Nat Med. 1998;4: factor-beta 3 gene expression in rat bone: a potential mechanism for
802– 807. estrogen- or raloxifene-mediated bone maintenance. Endocrinology.
26. Grainger DJ, Metcalfe JC, Grace AA, Mosedale DE. Transforming 1996;137:2075–2084.
growth factor-beta dynamically regulates vascular smooth muscle differ- 47. Grainger DJ, Weissberg PL, Metcalfe JC. Tamoxifen decreases the rate of
entiation in vivo. J Cell Sci. 1998;111:2977–2988. proliferation of rat vascular smooth-muscle cells in culture by inducing
27. Scanu AM, Lawn RM, Berg K. Lipoprotein(a) and atherosclerosis. Ann production of transforming growth factor beta. Biochem J.
Intern Med. 1991;115:209 –218. 1993;294:109 –112.
Transforming Growth Factor β and Atherosclerosis: So Far, So Good for the Protective
Downloaded from http://atvb.ahajournals.org/ by guest on April 30, 2018

Cytokine Hypothesis
David J. Grainger

Arterioscler Thromb Vasc Biol. 2004;24:399-404; originally published online December 29,
2003;
doi: 10.1161/01.ATV.0000114567.76772.33
Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272
Greenville Avenue, Dallas, TX 75231
Copyright © 2003 American Heart Association, Inc. All rights reserved.
Print ISSN: 1079-5642. Online ISSN: 1524-4636

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://atvb.ahajournals.org/content/24/3/399

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the
Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for
which permission is being requested is located, click Request Permissions in the middle column of the Web
page under Services. Further information about this process is available in the Permissions and Rights
Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online
at:
http://atvb.ahajournals.org//subscriptions/

Anda mungkin juga menyukai