• Aucun résultat trouvé

An overview of cytokine interactions in atherosclerosis and implications for peripheral arterial disease

N/A
N/A
Protected

Academic year: 2021

Partager "An overview of cytokine interactions in atherosclerosis and implications for peripheral arterial disease"

Copied!
12
0
0

Texte intégral

(1)

HAL Id: hal-00571370

https://hal.archives-ouvertes.fr/hal-00571370

Submitted on 1 Mar 2011

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

and implications for peripheral arterial disease

H.R.S. Girn, N.M. Orsi, S. Homer-Vanniasinkam

To cite this version:

H.R.S. Girn, N.M. Orsi, S. Homer-Vanniasinkam. An overview of cytokine interactions in atheroscle-

rosis and implications for peripheral arterial disease. Vascular Medicine, SAGE Publications, 2007,

12 (4), pp.299-309. �10.1177/1358863X07083387�. �hal-00571370�

(2)

Introduction

Atherosclerosis remains the leading cause of morbid- ity and mortality in the developed world. Chronic inflammation is the hallmark of atherosclerosis. It affects every step of the atherosclerotic process, from emergence of the fatty streak

1

– the first identifiable lesion of atherosclerosis – to endothelial activation and subsequent vascular dysfunction and vessel occlu- sion. More specifically, its clinical manifestations in the distal aorta and arteries in the legs are referred to as peripheral arterial disease (PAD) and affects about 4.5% of the population above 40 years of age, increas- ing to 14.5% above 70 years of age.

2

The PARTNERS study, which was a multi-centre, cross-sectional study

conducted in 25 cities and 350 primary care practices throughout the United States, reported a PAD preva- lence rate of 29%.

3

Although PAD is not directly life threatening, affected patients have a higher risk of mor- bidity and mortality due to generalized atherosclerosis.

It remains largely under-diagnosed,

3

with an estimated six asymptomatic patients for every known sympto- matic patient.

4

The inflammatory phenomenon related to atherosclerosis has been extensively studied over the past two decades and a variety of inflammatory and immunological mediators have been identified which positively correlate with the underlying disease load.

They regulate every stage of the inflammatory cascade, from endothelial activation to the adhesion of inflam- matory cells and platelets, and subsequent remodelling of the internal vascular environment.

Cytokines are key regulatory glycoproteins allied to inflammatory/immunological processes which modu- late all aspects of vascular inflammation by altering the proliferation, differentiation and function of an extensive array of cell types. They are intimately asso- ciated with atherogenesis and modulate plaque mor- phology and stabilization. Atheromatous plaque consists of a lipid core covered by a fibrotic cap on the luminal surface. Integrity of the fibrous cap depends

© 2007 SAGE Publications 10.1177/1358863x07083387

An overview of cytokine interactions in atherosclerosis and implications for peripheral arterial disease

HRS Girn

a

, NM Orsi

b

and S Homer-Vanniasinkam

a

Abstract: Over the last three decades, a surge in research into the inflammatory pathophysiology of atherosclerosis has highlighted an array of cytokines and other inflammatory mediators associated with underlying inflammatory burden. The ability to identify and simultaneously measure multiple cytokines in peripheral blood high- lights their potential as biomarkers of atherosclerosis. This has prompted much research in vascular medicine to identify the ‘at-risk’ groups for atherostenotic or atheroaneurysmal disease. This review is compiled with similar intentions and aims to discern the relevant evidence for cytokine profiling in peripheral arterial disease (PAD), where such information is lacking, while providing a holistic overview of cytokine interactions in atherosclerosis. This is pertinent given that cytokine profiles from coronary artery disease and aortic aneurysm studies cannot be directly extrap- olated to PAD due to differences in inflammatory environments that exist in these conditions. Whilst plaque morphology and blood rheology play an important role in the cardiac manifestations of atherosclerosis, tissue thrombogenecity is very impor- tant in PAD. Further, cytokines act in concert rather than in isolation in a disease process, and no single cytokine in a cross-sectional model is able to serve as an absolute screening marker. Thus, it is essential to understand the regulation of cytokine production in atherosclerosis prior to evaluating the viability and merits of a multimarker approach for clinical risk stratification in PAD.

Key words: atherosclerosis; biomarkers; cytokines; interleukins; peripheral arterial disease

a

Leeds Vascular Institute, Leeds General Infirmary, Leeds, UK;

b

Pathology & Tumour Biology, Leeds Institute of Molecular Medicine, St James University Hospital, Leeds, UK

Address for correspondence: Hardev Ramandeep Singh Girn, Leeds Vascular Institute, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK. Tel: ⫹ 44 (0)113 3922628;

Fax: ⫹ 44 (0)113 3922624; E-mail: hrsgirn@aol.com

Figure 1 is available in color online at http://vmj.sagepub.com

(3)

on the balance between the smooth muscle cell (SMC) proliferation and migration, and matrix metallopro- teinase (MMP)-induced degradation of the extracellu- lar matrix (ECM). The local balance between the pro- and anti-inflammatory cytokines is instrumental in determining structure of the fibrous cap and SMC degradation, thereby influencing plaque stability.

Cytokines were initially discovered about 50 years ago through their mitogenic and chemotactic effects on lymphocyte and monocyte populations. Although initial research focused more on pro-inflammatory cytokines [T-helper cell (Th)

1

] in relation to disease progression, it is the imbalance between pro- and anti- inflammatory (Th

2

cytokines) that is emerging to be pivotal to this process. Th

2

cytokines, which were initially considered to be protective, have now been found to be strongly associated with human abdominal aortic aneurysm (AAA) development. By comparison, stenotic atherosclerotic lesions are associated with Th

1

responses.

5

Cytokines act by binding to specific receptors, an interaction which has clarified their involvement in atherosclerosis and highlighted potential new avenues of therapeutic intervention. There are over 200 cytokines cloned to date and various classification systems have been used for the purpose of their catego- rization, although no single system has proven suffi- ciently comprehensive to be considered as a standard format. The difficulty in categorizing cytokines has largely related to the complexity and overlap of their biological roles. Structurally, cytokines can be classi- fied into nine different families (Table 1): the inter- leukin (IL)-1, IL-2/IL-4, IL-6/IL-12, interferon (IFN), tumour necrosis factor (TNF), chemokines, IL-10, transforming growth factor ␤ (TGF- ␤ ) and IL-17 fam- ilies. This system will be used for discussing represen- tative cytokines from each family in the context of

atherosclerosis and PAD for the purpose of this review.

In this respect, IL-17 is potentially pro-atherogenic because of its role in augmenting the expression of pro-inflammatory cytokines such as IL-6 from macrophages. Although it has recently been found to be associated with unstable angina,

6

there is no direct evidence implicating it with atherosclerosis and its family will therefore not be discussed as a part of this review.

The interleukin (IL)-1 family

IL -1 and IL -1 receptor antagonist

The IL-1 cytokine family comprises four main mem- bers: IL-1 ␣ , IL-1 ␤ , IL-1 receptor antagonist (IL-1ra) and IL-18. Although IL-1 can upregulate host defences and act as an immunoadjuvant, the family is primarily considered to be pro-inflammatory.

7

IL-1 was one of the first cytokines to be implicated in vessel wall inflammation in atherosclerosis, and in facilitating early lesion formation. Its effect on leukocyte recruit- ment and transmigration allows it to maintain a pro- inflammatory milieu once the inflammation has been initiated. Increased IL-1 levels have also been associ- ated with an increased tendency for plaque rupture.

8

The only member of this family with paradoxical properties is IL-1ra, a naturally occurring cytokine antagonist, which plays an anti-inflammatory role in regulating IL-1 function. In this respect, IL-1ra injec- tions in rheumatoid arthritis patients can reduce inflammatory joint destruction.

9

However, no similar therapeutic approaches have been determined for this cytokine in cardiovascular medicine. IL-1ra has been shown to be up-regulated in atherosclerosis and to cor- relate with the clinical stage of the disease in a small cohort of PAD patients in a number of studies.

10,11

Table 1 Cytokine classification and implication in atherosclerosis.

Cytokines Family Implication in atherosclerosis

IL -1 ␣ , IL -1 ␤ , IL -1ra, IL -18 IL -1 family Pro-atherogenic except IL -1ra

IL-2, IL-4, GM-CSF IL -2 family Pro-atherogenic. IL -4 levels elevated in aortic aneurysms but precise categorization of IL -4 and GM-CSF as atherogenic or protective is not possible

IL -6, IL -12 IL -6 family Pro-atherogenic

CXCL1-16, CXCL -28, Chemokines Pro-atherogenic

CX3CL1/fractalkine, IL -8, MCP-1

IL -10 IL -10 family Protective

IL -17 IL -17 family Pro-atherogenic

IFN- ␥ Interferon family Pro-atherogenic

TGF- ␤ TGF- ␤ Protective

TNF- ␣ , TNF- ␤ , LT- ␤ Tumour necrosis factor family Pro-atherogenic LT, lymphotoxins.

Other abbreviations can be found in the text.

(4)

IL-1␤␤

IL-1 ␤ plays a significant role in inflammation, as demonstrated by studies on IL-1 ␤ -deficient mice:

these animals failed to exhibit an acute phase response, remained apyrexic and exhibited no rise in circulating IL-6 levels when receiving an inflamma- tory stimulus.

12

IL-1 ␤ has been implicated in enhanc- ing expression of cell adhesion molecules on the endothelial surface and has consequently been deemed to be pro-atherogenic.

13

Taken together, these findings highlight the potential role of IL-1 ␤ in PAD through its eliciting of IL-6 production, an independent predic- tor of PAD progression in the Edinburgh artery study.

14

However, while IL-1 ␤ levels have been found to be increased in the pericardial fluid of patients with unstable angina,

15

plasma levels of IL-1 ␤ have not been found to be indicative of PAD.

10,11

IL-18

IL-18 is a more recently identified member of the IL-1 family, initially identified for its role in inducing IFN- ␥ production. IL-18 is an immunomodulator and does not fall neatly into the typical Th

1

:Th

2

dichotomy, inasmuch as it can induce either of these responses depending on the prevailing immunological environ- ment. Like many cytokines, it acts synergistically with other similar proteins and mediators in order to poten- tiate its effects. Experimental evidence from studies using apo E –/– mice confirms IL-18 to be athero- genic. While exogenous administration of IL-18 in these mice enhances atherosclerotic lesions,

16

there is conversely a reduction in atherosclerosis by inhibiting IL-18.

17,18

IL-18 has a bearing on the progression of human atherosclerotic plaques, particularly in relation to their stability.

19

Furthermore, IL-18 genetic poly- morphisms have been associated with clinical out- come in coronary artery disease.

20

Although IL-18 is considered to be a predictor of future cardiovascular risk,

21

there are currently no data to indicate that IL-18 levels are elevated in association with PAD.

The IL-2 family

IL-2

The IL-2 family comprises of four representative members: IL-2, IL-4, IL-5 and granulocyte-monocyte colony-stimulating factor (GM-CSF). The most note- worthy action of IL-2 relates to its mitogenic effects on T lymphocytes in response to antigenic stimulation, including the generation of both cytotoxic and sup- pressor T cells. However, despite extensive attempts to fully understand the actions and signalling pathways of IL-2, the nature of its pleiotropic effects on an array of cell types remains elusive. Although IL-2 is detected in atheromatous lesions, its precise role in vascular plaque formation remains poorly understood.

Indeed, although IL-2 levels are elevated in the blood

and in the aortic tissue of patients with aortic aneurysm disease, the rate of aneurysm expansion appears to depend on the overall Th

1

:Th

2

cytokine imbalance

22

in favour of Th

2

cytokines. Although studies to date have failed to address the role of IL-2 in PAD, this cytokine is nevertheless integral to vascu- lar inflammatory processes and promotes the mainte- nance and function of naturally occurring regulatory T cells, mostly at the transcriptional levels both in vivo and in vitro.

23

IL-4

Since IL-4 is produced by Th

2

cells, it has been proposed to play a protective role in atherosclerosis. This is sup- ported by phenomena such as IL-4-induced suppression of pro-atherogenic cytokines (e.g. IL-12) and inhibition of SMC proliferation and macrophage adhesiveness.

24

However, IL-4 has been found to cause an increased expression of P-selectin,

25

vascular cell adhesion mole- cule-1 (VCAM-1), and MMP-1 and 12, which are impli- cated in aortic aneurysm formation. IL-4 responses have also been found to predominate in aortic aneurysms both in animal models and human studies.

26

More recent studies also indicate that IL-4 causes differential IL-12 expression,

27

rather than a distinct inhibitory effect, as was previously noted. Although there is some evidence that IL-4 levels are elevated in acute coronary syndrome in comparison to stable disease,

28

no definitive involve- ment of IL-4 has been implicated in the severity of man- ifestation of PAD to date; small cross-sectional studies reflect non-significant variations in IL-4 levels in associ- ation with PAD (which are very low overall). This may also be explained by a shift in the Th

1

:Th

2

balance in favour of a Th

1

milieu in atherostenotic disease, as detailed above.

GM-CSF

GM-CSF stimulates the proliferation, differentiation

and function of multiple myeloid cells including neu-

trophils, monocytes/macrophages, eosinophils and

dendritic cells. Its main clinical use is in mobilizing

peripheral blood stem cells in myelosuppressive disor-

ders, drug-induced agranulocytosis and immuno-

deficiency syndromes. It has been implicated in

atherosclerosis because a decrease in plasma choles-

terol levels has been observed in patients receiving

GM-CSF for haematological disorders. These findings

are mirrored by GM-CSF treatment of Watanabe heri-

table hyperlipidaemic rabbits, which also exhibit a

significant decrease in total cholesterol levels for up to

2 weeks following cessation of recombinant GM-CSF

administration.

29

Related leporine studies

30

have also

indicated that GM-CSF can prevent the progression of

atherosclerotic plaques. After 7.5 months of GM-CSF

treatment, a 45% reduction in the atherosclerotic

lesion of the aortic arch was noted along with a reduc-

tion in intimal thickening compared to untreated

animals.

(5)

GM-CSF has traditionally been considered a pro- inflammatory cytokine based on in vivo and in vitro studies using lipopolysaccharide challenge.

31

However, this distinction becomes blurred in atherosclerosis,

32

where GM-CSF has been shown to confer some protec- tive effects.

32

GM-CSF is also a potential marker of chronic and severe inflammation (as opposed to acute responses) as its levels become more resistant to sup- pression by inhibitors such as glucocorticoids in this situation.

33

Recently, there has been renewed interest in the role of GM-CSF in atherosclerosis because of its angio- genic properties. Following pioneering studies in rodents, subcutaneous GM-CSF administration has been shown to promote collateral flow in the coronary circulation, with a decrease in ECG signs of myocar- dial ischaemia.

34

Unfortunately, these results have not been reproduced in PAD patients

35

(START trial), despite beneficial effects being seen in porcine models of PAD.

36

Although there are currently no data sup- porting the role of GM-CSF as a cytokine signature of PAD, our preliminary studies in this area have never- theless indicated that there is a trend for GM-CSF levels to be marginally higher in claudicants (present authors, unpublished).

IL-7

IL-7 was first discovered almost two decades ago for its role in promoting the growth of B-cell precursors in bone marrow culture systems.

37

This cytokine is dis- cussed under the IL-2 family because its receptor shares the common ␥ chain with other IL-2 family members. IL-7 is now an established regulator of T-cell homeostasis in both thymic and extra-thymic tissues,

38

and has more recently been implicated as a pro- inflammatory factor in atherosclerosis, with higher levels detected in patients with unstable angina, com- pared to stable angina and controls.

39

This study also revealed that aspirin causes a significant decline in both spontaneous and stimulated IL-7 release from platelets in vitro. However, only one study has investi- gated IL-7 levels in relation to PAD (as a subgroup of an atherosclerotic cohort), and revealed that IL-7 was undetectable in these patients.

40

The IL -6/IL -12 family

IL-6

This pro-inflammatory cytokine is probably the most consistent biomarker of PAD identified thus far (Table 2).

The Edinburgh artery,

14

InCHIANTI

11

and MESA

45

studies have all individually established its role as an independent predictor of PAD in community screening, irrespective of ethnicity. Furthermore, IL-6 has been found to be associated with PAD severity

41

and its genetic polymorphisms have been implicated in increas- ing susceptibility of type 2 diabetics to develop PAD.

44

IL-6 enhances cell adhesion molecule (CAM) expres- sion by endothelial and smooth muscle cells, and enhances the production of acute phase reactants such as C-reactive protein (CRP) and TNF- ␣ by the hepato- cytes. As such, plasma IL-6 concentrations in healthy populations correlate with metabolic and inflamma- tory factors such as TNF- ␣ and CRP, which are con- sidered relevant to atherosclerotic progression,

53

and in the case of the latter, associated with increased risk of death from cardiovascular pathology. More specifi- cally, IL-6 has a positive feedback interaction with intimal cells and macrophages to further stimulate its own production, along with that of IL-8 and monocyte chemoattractant protein (MCP)-1. IL-6 also increases leucocyte recruitment into the atherosclerotic plaque by stimulating chemokine release from the endothe- lium and by enhancing expression of intercellular adhesion molecule-1 (ICAM-1) by the smooth muscle cells.

54

It may also stimulate smooth muscle cells to evolve into foam cells,

55

thereby adversely effecting plaque morphology.

IL-12

IL-12 (p70) is a heterodimeric protein, composed of a heavy chain (p40) and a covalently associated light chain (p35). The expression of each of these subunits is independently regulated, but biologically active IL-12 can only be produced by cells with the capacity to pro- duce both light and heavy chains. The primary func- tion of IL-12, as proven in both animal models and humans, is the induction and sustenance of Th

1

responses to infection/antigen exposure by facilitating the secretion of cytokines such as IL-2, IFN- ␥ , IL-18 and an array of chemokines. While IL-12 has been implicated in the progression of atherosclerosis, func- tional blockade of endogenous IL-12 production can reduce atherogenesis in LDL receptor-deficient mice and improve plaque stability.

56

Immunohistochemical studies have also revealed an increased presence of IL-12 (p70) in human atherosclerotic plaques com- pared to normal arteries,

57

thereby supporting its role in promoting plaque inflammation in human coronary arteries by enhancing local T-cell recruitment.

58

While raised IL-12 levels in the coronary arteries in vivo are a feature of unstable angina,

59,60

no evidence is avail- able regarding IL-12 profiles in PAD to date.

IL-12 acts synergistically with IL-18 to elicit Th

1

responses, as well as facilitating the release of IFN- ␥

by Th

1

cells. IFN- ␥ and IL-12 act in combination to pro-

mote Th

1

function too, with IFN- ␥ further up-regulating

IL-12 production. Paradoxically, IL-12 also regulates

Th

2

responses by inducing the production of IL-10 and

IL-13 while inhibiting IL-4 and IL-5 production.

27

This is part of a protective negative feedback mecha-

nism, which comes into play in chronic inflammatory

states in particular. IL-10 causes feedback inhibition

of IL-12

57

while IL-4 has a similar effect on IFN- ␥ .

The interaction between IL-4 and IL-12 is more

(6)

complex: although initial reports indicated that IL-4 inhibited IL-12, recent evidence suggests that IL-4 has more subtle effects by inducing differential IL-12 expression rather than overall inhibition.

61

Thus, although IL-12 enhances IFN- ␥ production, it also concomitantly enhances production of anti-inflamma- tory cytokines that limit IFN- ␥ production, highlight- ing the complex network system in which cytokines operate. Understanding these interactions is pivotal to clarifying the inflammatory pathophysiology allied to atherosclerotic progression and revealing the prognos- tic potential of cytokines in PAD (Figure 1).

The interferon (IFN) family IFN-␥␥

IFN- ␥ is a pleiotropic pro-inflammatory cytokine pri- marily secreted by T lymphocytes and natural killer

cells following activation by immune and inflamma- tory stimuli. It induces expression of ICAM-1, VCAM-1 and selectins by endothelial and smooth muscle cells, and promotes leucocyte adhesion at the site of the vascular lesion.

62

As outlined, it acts in con- cert with IL-12 and IL-18 to promote atherosclero- sis.

63

More specifically, a reduction in atherosclerosis was also noted in cholesterol-diet fed mice in the absence of IFN- ␥ .

64

By contrast, IFN- ␥ has also been associated with a reduction in atherosclerotic lesion size in apo E –/– mice.

65

IFN- ␥ is intimately related to plaque morphology, with enhanced expression reported in unstable plaques.

66

This relationship with atherosclerotic lesions has been consistently reproduced in clinical samples as well as rodent atherosclerosis models. There is also an associated rise in pro-inflammatory cytokines such as MCP-1 and macrophage inflammatory protein (MIP)-1 ␣ and ␤ within the plaque. IFN- ␥ potentially destabilizes

Table 2 Baseline cytokine trends in PAD patients in vivo.

Year Study PAD group size Conclusions

2006 Nylænde et al

41

127 MCP-1, CD40L and IL-6, significantly associated with extent of angiographically proven PAD and maximal treadmill walking

2006 DePalma et al

42

100 IL-6, TNF- ␣ R1 and CRP were significantly elevated in PAD;

(47) no statins no significant differences with statin dose except for (53) on statins IL-6 (higher in PAD patients on statins)

2006 Armitage et al

43

60 Significantly elevated IL-6 in PAD; endothelial reactive antibodies promote PAD by elevating IL-6

2006 Libra et al

44

290 IL-6, CRP, fibrinogen and VEGF higher in diabetic PAD;

(146) diabetic IL-6-174G/G phenotype promotes PAD in type 2 (144) non-diabetic diabetics through increased IL-6 release

2006 MESA study

45

275 African-American ethnicity associated with 50% increase Allison et al (6653) total cohort in odds of PAD; increase in IL-6 associated with

largest risk for PAD

2005 Edinburgh Artery 273 CRP, IL-6 and ICAM-1 as molecular markers of peripheral Study

14

Tzoulaki et al (46) symptomatic atherosclerosis and its progression; IL-6 strongest

(227) asymptomatic predictive power

2005 InCHIANTI study

11

107 PAD population features increased IL-6, IL-1ra, fibrinogen

McDermott et al and CRP; trends for lower TGF- ␤ in PAD

2005 ARIC study

46

209 Significant association of MCP-1 with PAD and incident

Hoogeveen et al coronary heart disease

2005 Danielsson et al

47

100 CLI was significantly associated with elevated white cell count, hsCRP, IL-6 and sTNF- ␣ R1 and 2; no effect of IL-6 promoter polymorphism

2005 Ziegler et al

48

66 OPG values correlate with PAD severity

2003 DePalma et al

49

91 IL-10 significantly lower in PAD, while IL-6, CRP and TNF- ␣ significantly elevated

2003 Silvestro et al

50

38 IL-6, CRP, sICAM-1 and sVCAM-1 associated with poor prognosis in PAD

2000 Testa et al

51

8 IL-1 ␤ and IL-6 gene expression upregulated in skeletal muscle of CLI patients

1999 Fiotti et al

10

16 Significant increase in TNF- ␣ R1 and 2, IL-6 and IL-1ra in PAD

1999 Kirk et al

52

26 Significantly elevated IL-8 at rest and post-exercise in PAD VEGF, vascular endothelial growth factor; CLI, critical limb ischaemia; hsCRP, high-sensitivity C-reactive protein;

sTNF- ␣ R1 and 2, soluble TNF- ␣ receptors 1 and 2; OPG, osteoprotegerin; sVCAM, soluble vascular cell adhesion molecule.

Other abbreviations can be found in the text.

(7)

the fibrous cap by inhibiting SMC proliferation as well as IL-1 ␤ and TGF- ␤ -mediated collagen synthesis.

However, there is an ambivalent element to its other- wise pro-atherogenic role through its attenuation of constitutive and inducible MMP expression within the plaque.

67

In addition, it regulates foam cell apoptosis within the lipid core of the plaque.

68

It is, therefore, difficult to determine whether IFN- ␥ forms unstable plaque from the outset or if there is a shift in the plaque modulation mechanism of IFN- ␥ with advanced atherosclerosis, causing plaque instability at a later stage. Although IFN- ␥ has not been investi- gated in the context of PAD, its profiles should be studied in relation to IL-12, IL-4 and IL-10 levels given what has been discussed above (Figure 1B).

The tumour necrosis factor (TNF) family TNF-␣␣

TNF- ␣ is a pro-inflammatory prototype member of this family. Initially identified as an anti-tumour agent, it is now recognized as a pleiotropic cytokine with multiple, dose-dependent actions on both local and

systemic inflammation. TNF- ␣ is involved in athero- sclerotic progression from the initial stages of intimal thickening to the subsequent vessel occlusion.

69

It stimulates selectin and adhesion molecule expression by endothelial cells, SMCs and macrophages, promot- ing interaction between endothelium, leucocytes and platelets, thus creating a pro-inflammatory and pro- thrombotic local environment. These changes magnify the impact of the local vascular injury and enhance plaque formation. TNF- ␣ has also been implicated in modulating plaque morphology and is associated with plaque rupture. It stimulates production of MMP-1- 9,11 and 13 production in the endothelium, SMCs and macrophages.

67,70

Locally, within the atheroma, it increases expression of tissue factor, a potent throm- bogenic protein, by macrophage-derived foam cells and the endothelial cells.

71

There is ample evidence implicating TNF- ␣ in PAD progression (Table 2).

10,42,47,49

Osteoprotegerin, another member of the TNF family, has also been associated with PAD severity.

48

Along with CRP and IL-6, TNF- ␣ production has also been found to have a significant correlation in atherosclerotic parents and their offspring (STANISLAS cohort),

53

implicating it as an atherosclerotic risk indicator in healthy families.

The impact of TNF- ␣ on vascular injury in both acute and chronic inflammatory conditions has made it an important therapeutic target. Unfortunately, results allied to TNF neutralization therapies have been inconsistent thus far. Trials using TNF- ␣ antagonist, such as infliximab (ATTACH trial)

72

and etanercept (RENNAISANCE and RECOVER trials),

73

failed to show any improvement in cardiac failure, while the ATTACH trial was associated with an increased all- cause mortality. Although there have been no studies focusing on anti-atherogenic therapeutic interventions in PAD, a recent study by DePalma and co-workers,

42

indicated that there were no marked differences in TNF- ␣ levels in PAD patients receiving/not receiving statin therapy.

The chemokine family

Chemokines are a large superfamily of small proteins that direct leucocytes to sites of inflammation and can be broadly classified into three distinct groups on the basis of structure and function: CC (e.g. monocyte chemoattractant protein-1), CXC (e.g. IL-8) and CX3C (e.g. fractalkine) chemokine families. MCP-1 and IL-8 are both pro-inflammatory cytokines and contribute to atherogenesis by mediating monocyte/

neutrophil–endothelial interactions.

MCP-1

MCP-1 is the most widely studied chemokine in PAD and it is consistently expressed in both animal and human atherosclerotic plaques. This atherogenic

Figure 1 (A) Conventional Th

1

:Th

2

paradigm. Shift of balance in the favour of Th

1

responses promotes ather- osclerotic plaque progression in the vessel wall. (B) Schematic representation of the negative and positive feedback interactions between Th

1

and Th

2

cytokines.

IL-10 is the dominant anti-inflammatory cytokine that hin-

ders expression of IL-12, IFN- ␥ and IL -18. IL -12 enhances

production of IFN- ␥ and IL -18. It inhibits production of

TGF- ␤ and thereby contributes to plaque instability and

fibrin cap degradation. Whilst promoting pro-atherogenic

mechanisms, IL -12 also regulates atherogenic responses

by enhancing production of IL -10.

(8)

cytokine plays a crucial role in monocyte recruitment into the vascular lesion, which is one of the earliest steps in atherogenesis. While sub-endothelial migra- tion of monocytes is facilitated by engagement of this cytokine with its receptor (CCR2), this interaction is only necessary to allow entry of monocytes into the intima and does not play a significant role in the sub- sequent macrophage action.

74

There is a plethora of data from mouse studies clearly demonstrating the role of MCP-1 and its receptor in modulation of the atherosclerotic process.

75

MCP-1 has also been found to enhance SMC proliferation by stimulating binding activity of activator protein-1 and promotes expression of integrins and tissue factor on SMCs, generating a pro-inflammatory and a pro-thrombotic environment.

Recently, the ARIC study

46

found MCP-1 to be an independent risk factor for PAD and reported signifi- cantly higher levels of MCP-1 in the PAD population.

Incidental coronary heart disease was also found to increase significantly in association with MCP-1 levels. Another study correlated MCP-1 levels with the severity of PAD, as assessed by angiographic scores and maximal treadmill walking distance.

41

Consequently, MCP-1 has been suggested as a novel potential target for atherosclerotic therapy. Indeed, statins have been found to have an independent effect on reducing MCP-1 levels.

76

In this respect, adminis- tration of 20 mg simvastatin/day can reduce MCP-1 levels by 7% from baseline in hypercholesterolaemic patients.

77

IL-8 (CXCL8)

First described in 1987, IL-8 is probably one of the best characterized neutrophil chemoattractants consistently found in macrophage-rich atherosclerotic plaques.

78

It enhances all aspects of endothelial–neutrophil interac- tion and migration, facilitating neutrophil recruitment at the site of the vascular inflammation. IL-8 also trig- gers firm adhesion of rolling monocytes to vascular endothelium and infiltration of the vessel wall, and is thus implicated in early atherosclerotic progression.

79

Mice deficient in IL-8 receptor show decreased macrophage clustering in the plaque and smaller ather- osclerotic lesions.

80

A Japanese study involving tissue sampling from human abdominal aortic aneurysms and carotid artery stenoses showed significantly increased expression of IL-8 and its receptor (CXCR2), implicat- ing this chemokine in both aneurysmal and athero- occlusive disease processes.

81

The role of IL-8 in destabilizing the atherosclerotic plaque has been a subject of close scrutiny because of its inhibitory effect on tissue inhibitor of metalloproteinase (TIMP)- 1 expression in macrophages, which leads to a conse- quent increase in MMP activity.

82

Furthermore, the potentially detrimental effects of IL-8 may be com- pounded by its effects on SMC proliferation. While IL-8 has been associated with unstable angina and acute myocardial infarction,

83

there are some studies

addressing its role in PAD. Kirk et al.

52

indicated that significantly higher levels of IL-8 were detectable in PAD patients compared with controls, both at rest and post-exercise. However, this observation was not reproduced in the study of Danielsson et al.

47

which compared the cytokine profiles of PAD patients with/without diabetes and controls.

The IL-10 family

IL-10

IL-10 was initially described as a cytokine synthesis inhibitory factor because of its ability to directly inhibit Th

1

and Th

2

cytokine production. This role makes it one of the key regulators of both local and systemic inflammatory/immune responses, and it has been shown to confer a protective effect in atheroscle- rosis. This is illustrated by the study of Mallat and co-workers, who demonstrated that IL-10-deficient mice fed an atherogenic diet had a threefold increase in lipid accumulation compared with their wild-type counterparts.

84

The plaques in the study group were also more vulnerable and had increased thrombogenic tendencies. Furthermore, Namiki et al showed that intramuscular IL-10 gene transfer significantly reduced the atherosclerotic plaque area in such mice, with an associated decline in IL-12 and IFN- ␥ mRNA expression in the spleens of the study group animals.

85

IL-10 is produced by a variety of inflammatory cells and is found in advanced atherosclerotic plaques where it has been associated with decreased apoptosis in the lipid central core, thereby reducing the likelihood of rupture.

86

Systemic and intralesional delivery of IL-10 has been shown to diminish atherosclerosis in a variety of experimental models.

87

One of the reasons for this effect could be a reduction in inflammatory cell recruit- ment within the plaque: IL-10 has been shown to inhibit monocyte adherence to human aortic endothe- lial cells in vitro.

88

The ability of IL-10 to suppress certain CD40/CD40L ligand-mediated monocyte responses may account for some of its anti-atherogenic effects,

89

while others may be mediated indirectly through a suppression of pro-inflammatory cytokine (e.g. MCP-1, IL-6, IL-12 and IFN- ␥ ) secretion.

Data regarding the role of IL-10 in PAD in humans

are surprisingly scant. It is difficult to determine

whether higher or lower IL-10 levels are to be

expected as these will vary according to the timing of

onset of the disease. On one hand, lower IL-10 levels

in PAD would favour disease progression. On the

other, higher levels might be expected as a modulatory

phenomenon to the associated increase in local

inflammatory burden. This complex relationship has

been illustrated in coronary artery disease patients,

where lower IL-10 levels coupled with higher pro-

inflammatory cytokine levels were observed in

(9)

patients with unstable angina who subsequently devel- oped a cardiovascular event.

90,91

Conversely, higher IL-10 levels have been associated with a favourable prognosis in patients with acute coronary syndromes (CAPTURE trial).

92

As such, this suggests that IL-10 may have poor predictive merit for clinical outcome in atherosclerosis. In this respect, the ratio between IL-10 and various pro-inflammatory cytokines may prove more useful as they account for some of the dynamics of the inflammatory processes involved. While IL-10 levels have also been consistently found to be elevated in asso- ciation with abdominal aortic aneurysms,

93

DePalma et al

49

found IL-10 to be significantly lower in PAD.

These findings were resonated by the large InCHIANTI study, which pointed to a marked trend for circulatory IL-10 concentrations to be lower in patients with PAD.

11

The transforming growth factor (TGF) family

TGF- ␤␤

The TGF family of growth, differentiation and mor- phogenic glycosylated preproteins comprises of over 50 members. TGF- ␤ is both the prototype member of this superfamily as well as one of the most studied cytokines in vascular biology due to its role in athero- sclerosis and neo-intimal hyperplasia. Following the work done by Metcalfe and Grainger in the mid- 1990s, TGF- ␤ is considered to be beneficial in athero- sclerosis, leading to the so-called ‘protective cytokine’

hypothesis.

94

Although this hypothesis was initially postulated based on the role of TGF- ␤ in inhibiting vascular smooth muscle cell (VSMC) proliferation and migration, its other immunomodulatory effect on T-cell regulation both within and outside the plaque,

95

and inhibition of pro-inflammatory cytokine (IL-6, granulocyte colony stimulating factor and MCP-1) gene expression

96

gave further momentum to this concept. Whilst inhibition of VSMC proliferation is consistent across studies, the effects of TGF- ␤ on macrophages and cytokine inhibition is variable and governed by the local inflammatory environment.

97,98

As for many other cytokines, most of the initial evidence regarding the anti-atherogenic properties of TGF- ␤ was drawn from studies using knockout mice.

However, TGF- ␤ -deficient mice died shortly after birth due to systemic inflammation associated with leucocytic infiltration in virtually every organ sys- tem,

99

highlighting the anti-inflammatory effects of TGF- ␤ by down-regulation of CAM expression and the consequent reduction in leucocyte infiltration.

Studies using heterozygous mouse models proved equally informative, as these animals presented with reduced rather than absent TGF- ␤ profiles. Exposing these mice to a high-fat diet enhanced endothelial activation and arterial lipid deposition compared with controls.

100

In line with these observations, Mallat

et al noted that inhibition of TGF-␤ signalling is also associated with accelerated atherosclerotic lesion development in apoE-deficient mice and favours the development of those with an increased inflammatory component and decreased collagen content.

101

A related study further showed that TGF-␤ has a defini- tive role in influencing plaque morphology, with more vulnerable plaques formed in the case of TGF-␤ defi- ciency due to decreased fibrosis, increased plaque inflammatory cell counts, greater areas of rupture and increased intraplaque haemorrhage.

102

The anti-atherosclerotic role of TGF-␤ is supported by the negative correlation between its plasma levels and coronary artery disease,

103

with similar trends also noted in association with PAD in the InCHIANTI study.

11

These findings suggest that together with IL-10, TGF-␤ may have anti-atherogenic properties which may prove useful in a therapeutic setting.

Conclusion

There are multiple regulatory levels of vascular inflammation with direct or indirect involvement of cytokines at every step. Traditional risk factors such as smoking, hypertension, diabetes, obesity and high cholesterol levels – though useful in population studies – fail to explain the variable nature of disease progression between individuals. Although cytokines are clearly implicated in the pathophysiology of ather- osclerosis, there is currently scant information of their role in PAD in particular. However, early evidence in human studies indicates that markers such as IL-6 are closely associated with disease manifestation. Given the intimate interrelationships between cytokines in physiological immune/inflammatory networks, this argues in favour of the case that other cytokines whose role has not yet been implicated in cardiovascular pathophysiology are very likely to also play a role (and likely above and beyond the conventional Th

1

:Th

2

dichotomy). This postulate is further sup- ported by the animal studies reviewed herein.

The findings summarized in this review substantiate

the role of cytokines in atherosclerosis and highlight

the differences in cytokine responses in different sub-

catergorizations of atherostenotic or atheroaneurysmal

disease processes. As proteins involved in the patho-

physiological process per se, cytokines have the

potential to provide useful information on disease

progression on an individual basis and serve as valu-

able adjuncts to existing clinical screening and inves-

tigative tools in forming a composite picture. Also, the

potential role of cytokines in gauging the response to

the medical treatment (e.g. statins, aspirin) of athero-

sclerosis needs to be investigated through larger longi-

tudinal clinical trials. Only then would we be in a

position to assess the merit of cytokines as clinical

risk stratification tools in cardiovascular medicine.

(10)

References

1 Stary HC, Chandler AB, Glagov S et al. A definition of initial, fatty streak, and intermediate lesions of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1994; 89: 2462–78.

2 Selvin E, Erlinger TP. Prevalence of and risk factors for peri- pheral arterial disease in the United States: results from the National Health and Nutrition Examination Survey, 1999–2000. Circulation 2004; 110: 738–43.

3 Hirsch AT, Criqui MH, Treat-Jacobson D et al. Peripheral arte- rial disease detection, awareness, and treatment in primary care. JAMA 2001; 286: 1317–24.

4 Dormandy JA, Rutherford RB. Management of peripheral arte- rial disease (PAD). TASC working group. TransAtlantic Intersociety consensus (TASC). J Vasc Surg 2000; 31: 1–296.

5 Schonbeck U, Sukhova GK, Gerdes N, Libby P. T(H)2 pre- dominant immune responses prevail in human abdominal aortic aneurysm. Am J Pathol 2002; 161: 499–506.

6 Hashmi S, Zeng QT. Role of interleukin-17 and interleukin-17- induced cytokines interleukin-6 and interleukin-8 in unstable coronary artery disease. Coron Artery Dis 2006; 17: 699–706.

7 Dinarello CA. Interleukin-1. Cytokine Growth Factor Rev 1997; 8: 253–65.

8 Libby P, Sukhova G, Lee RT, Galis ZS. Cytokines regulate vas- cular functions related to stability of the atherosclerotic plaque. J Cardiovasc Pharmacol 1995; 25(suppl 2): S9–12.

9 Bresnihan B, Alvaro-Gracia JM, Cobby M et al. Treatment of rheumatoid arthritis with recombinant human interleukin-1 receptor antagonist. Arthritis Rheum 1998; 41: 2196–204.

10 Fiotti N, Giansante C, Ponte E et al. Atherosclerosis and inflammation. Patterns of cytokine regulation in patients with peripheral arterial disease. Atherosclerosis 1999; 145: 51–60.

11 McDermott MM, Guralnik JM, Corsi A et al. Patterns of inflammation associated with peripheral arterial disease: the InCHIANTI study. Am Heart J 2005; 150: 276–81.

12 Zheng H, Fletcher D, Kozak W et al. Resistance to fever induc- tion and impaired acute-phase response in interleukin-1 beta- deficient mice. Immunity 1995; 3: 9–19.

13 Dinarello CA. IL-18: A TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol 1999; 103(1 Pt 1): 11–24.

14 Tzoulaki I, Murray GD, Lee AJ, Rumley A, Lowe GD, Fowkes FG. C-reactive protein, interleukin-6, and soluble adhesion molecules as predictors of progressive peripheral atherosclero- sis in the general population: Edinburgh Artery Study.

Circulation 2005; 112: 976–83.

15 Oyama J, Shimokawa H, Morita S, Yasui H, Takeshita A.

Elevated interleukin-1beta in pericardial fluid of patients with ischemic heart disease. Coron Artery Dis 2001; 12: 567–71.

16 Whitman SC, Ravisankar P, Daugherty A. Interleukin-18 enhances atherosclerosis in apolipoprotein E(–/–) mice through release of interferon-gamma. Circ Res 2002; 90: E34–38.

17 Mallat Z, Corbaz A, Scoazec A et al. Interleukin-18/inter- leukin-18 binding protein signaling modulates atherosclerotic lesion development and stability. Circ Res 2001; 89: E41–45.

18 Elhage R, Jawien J, Rudling M et al. Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice.

Cardiovasc Res 2003; 59: 234–40.

19 Mallat Z, Corbaz A, Scoazec A et al. Expression of interleukin- 18 in human atherosclerotic plaques and relation to plaque instability. Circulation 2001; 104: 1598–603.

20 Tiret L, Godefroy T, Lubos E et al. Genetic analysis of the interleukin-18 system highlights the role of the interleukin-18 gene in cardiovascular disease. Circulation 2005; 112: 643–50.

21 Gilardini L, McTernan PG, Girola A et al. Adiponectin is a candidate marker of metabolic syndrome in obese children and adolescents. Atherosclerosis 2006; 189: 401–407.

22 Lindholt JS, Shi GP. Chronic inflammation, immune response, and infection in abdominal aortic aneurysms. Eur J Vasc Endovasc Surg 2006; 31: 453–63.

23 Wan YY, Flavell RA. The roles for cytokines in the generation and maintenance of regulatory T cells. Immunol Rev 2006;

212: 114–30.

24 Vadiveloo PK, Stanton HR, Cochran FW, Hamilton JA.

Interleukin-4 inhibits human smooth muscle cell proliferation.

Artery 1994; 21: 161–81.

25 Khew-Goodall Y, Wadham C, Stein BN, Gamble JR, Vadas MA. Stat6 activation is essential for interleukin-4 induction of P-selectin transcription in human umbilical vein endothelial cells. Arterioscler Thromb Vasc Biol 1999; 19: 1421–29.

26 Shimizu K, Libby P, Mitchell RN. Local cytokine environ- ments drive aneurysm formation in allografted aortas. Trends Cardiovasc Med 2005; 15: 142–48.

27 Morita Y, Gupta R, Seidl KM, McDonagh KT, Fox DA.

Cytokine production by dendritic cells genetically engineered to express IL-4: induction of Th2 responses and differential regulation of IL-12 and IL-23 synthesis. J Gene Med 2005;

7: 869–77.

28 Szodoray P, Timar O, Veres K et al. TH1/TH2 imbalance, measured by circulating and intracytoplasmic inflammatory cytokines – immunological alterations in acute coronary syndrome and stable coronary artery disease. Scand J Immunol 2006; 64: 336–44.

29 Ishibashi T, Yokoyama K, Shindo J et al. Potent cholesterol- lowering effect by human granulocyte-macrophage colony- stimulating factor in rabbits. Possible implications of enhancement of macrophage functions and an increase in mRNA for VLDL receptor. Arterioscler Thromb 1994; 14: 1534–41.

30 Shindo J, Ishibashi T, Yokoyama K et al. Granulocyte- macrophage colony-stimulating factor prevents the progression of atherosclerosis via changes in the cellular and extracellular composition of atherosclerotic lesions in Watanabe heritable hyperlipidemic rabbits. Circulation 1999; 99: 2150–56.

31 Hamilton JA, Stanley ER, Burgess AW, Shadduck RK.

Stimulation of macrophage plasminogen activator activity by colony-stimulating factors. J Cell Physiol 1980; 103: 435–45.

32 Hamilton JA, Anderson GP. GM-CSF biology. Growth Factors 2004; 22: 225–31.

33 Cosio BG, Mann B, Ito K, Jazrawi E, Barnes PJ, Chung KF, Adcock IM. Histone acetylase and deacetylase activity in alve- olar macrophages and blood monocytes in asthma. Am J Respir Crit Care Med 2004; 170: 141–47.

34 Seiler C, Pohl T, Wustmann K et al. Promotion of collateral growth by granulocyte-macrophage colony-stimulating factor in patients with coronary artery disease: a randomized, double- blind, placebo-controlled study. Circulation 2001; 104:

2012–17.

35 Van Royen N, Schirmer SH, Atasever B et al. START Trial: a pilot study on STimulation of ARTeriogenesis using subcuta- neous application of granulocyte-macrophage colony-stimulat- ing factor as a new treatment for peripheral vascular disease.

Circulation 2005; 112: 1040–46.

36 Grundmann S, Hoefer I, Ulusans S et al. Granulocyte-

macrophage colony-stimulating factor stimulates arteriogenesis

(11)

in a pig model of peripheral artery disease using clinically applicable infusion pumps. J Vasc Surg 2006; 43: 1263–69.

37 Namen AE, Lupton S, Hjerrild K et al. Stimulation of B-cell progenitors by cloned murine interleukin-7. Nature 1988; 333:

571–73.

38 Jiang Q, Li WQ, Aiello FB et al. Cell biology of IL-7, a key lymphotrophin. Cytokine Growth Factor Rev 2005; 16:

513–33.

39 Damas JK, Waehre T, Yndestad A et al. Interleukin-7-mediated inflammation in unstable angina: possible role of chemokines and platelets. Circulation 2003; 107: 2670–76.

40 Prunet C, Montange T, Vejux A et al. Multiplexed flow cyto- metric analyses of pro- and anti-inflammatory cytokines in the culture media of oxysterol-treated human monocytic cells and in the sera of atherosclerotic patients. Cytometry A 2006; 69:

359–73.

41 Nylaende M, Kroese A, Stranden E et al. Markers of vascular inflammation are associated with the extent of atherosclerosis assessed as angiographic score and treadmill walking distances in patients with peripheral arterial occlusive disease. Vasc Med 2006; 11: 21–28.

42 DePalma RG, Hayes VW, May PE et al. Statins and biomarkers in claudicants with peripheral arterial disease: cross-sectional study. Vascular 2006; 14: 193–200.

43 Armitage JD, Lindsey NJ, Homer-Vanniasinkam S. The role of endothelial cell reactive antibodies in peripheral arterial disease.

Eur J Vasc Endovasc Surg 2006; 31: 170–75.

44 Libra M, Signorelli SS, Bevelacqua Y et al. Analysis of G(-174)C IL-6 polymorphism and plasma concentrations of inflammatory markers in patients with type 2 diabetes and peripheral arterial disease. J Clin Pathol 2006; 59: 211–15.

45 Allison MA, Criqui MH, McClelland RL et al. The effect of novel cardiovascular risk factors on the ethnic-specific odds for peripheral arterial disease in the Multi-Ethnic Study of Atherosclerosis (MESA). J Am Coll Cardiol 2006; 48: 1190–97.

46 Hoogeveen RC, Morrison A, Boerwinkle E et al. Plasma MCP-1 level and risk for peripheral arterial disease and incident coronary heart disease: Atherosclerosis Risk in Communities study. Atherosclerosis 2005; 183: 301–307.

47 Danielsson P, Truedsson L, Eriksson KF, Norgren L.

Inflammatory markers and IL-6 polymorphism in peripheral arterial disease with and without diabetes mellitus. Vasc Med 2005; 10: 191–98.

48 Ziegler S, Kudlacek S, Luger A, Minar E. Osteoprotegerin plasma concentrations correlate with severity of peripheral artery disease. Atherosclerosis 2005; 182: 175–80.

49 DePalma RG, Hayes VW, Cafferata HT et al. Cytokine signatures in atherosclerotic claudicants. J Surg Res 2003; 111: 215–21.

50 Silvestro A, Scopacasa F, Ruocco A et al. Inflammatory status and endothelial function in asymptomatic and symptomatic peripheral arterial disease. Vasc Med 2003; 8: 225–32.

51 Testa M, De Ruvo E, Russo A et al. Induction of interleukin- 1beta and interleukin-6 gene expression in hypoperfused skeletal muscle of patients with peripheral arterial disease. Ital Heart J 2000; 1: 64–67.

52 Kirk G, Hickman P, McLaren M, Stonebridge PA, Belch JJ.

Interleukin-8 (IL-8) may contribute to the activation of neu- trophils in patients with peripheral arterial occlusive disease (PAOD). Eur J Vasc Endovasc Surg 1999; 18: 434–38.

53 Haddy N, Sass C, Droesch S et al. IL-6, TNF-alpha and ather- osclerosis risk indicators in a healthy family population: the STANISLAS cohort. Atherosclerosis 2003; 170: 277–83.

54 Von der Thusen JH, Kuiper J, van Berkel TJ, Biessen EA.

Interleukins in atherosclerosis: molecular pathways and thera- peutic potential. Pharmacol Rev 2003; 55: 133–66.

55 Klouche M, Rose-John S, Schmiedt W, Bhakdi S.

Enzymatically degraded, nonoxidized LDL induces human vascular smooth muscle cell activation, foam cell transforma- tion, and proliferation. Circulation 2000; 101: 1799–805.

56 Hauer AD, Uyttenhove C, de Vos P et al. Blockade of inter- leukin-12 function by protein vaccination attenuates athero- sclerosis. Circulation 2005; 112: 1054–62.

57 Uyemura K, Demer LL, Castle SC et al. Cross-regulatory roles of interleukin (IL)-12 and IL-10 in atherosclerosis. J Clin Invest 1996; 97: 2130–38.

58 Zhang X, Niessner A, Nakajima T et al. Interleukin 12 induces T-cell recruitment into the atherosclerotic plaque. Circ Res 2006; 98: 524–31.

59 Baidya SG, Zeng QT, Wang X, Guo HP. T helper cell related interleukins and the angiographic morphology in unstable angina. Cytokine 2005; 30: 303–10.

60 Steppich BA, Moog P, Matissek C et al. Cytokine profiles and T cell function in acute coronary syndromes. Atherosclerosis 2007; 190: 443–51.

61 Hochrein H, O’Keeffe M, Luft T et al. Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 produc- tion by mouse and human dendritic cells. J Exp Med 2000;

192: 823–33.

62 Chung HK, Lee IK, Kang H et al. Statin inhibits interferon- gamma-induced expression of intercellular adhesion molecule-1 (ICAM-1) in vascular endothelial and smooth muscle cells.

Exp Mol Med 2002; 34: 451–61.

63 Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev 2006; 86: 515–81.

64 Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH. Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol 2003; 23: 454–60.

65 Xiong W, Zhao Y, Prall A, Greiner TC, 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.

66 Leon ML, Zuckerman SH. Gamma interferon: a central medi- ator in atherosclerosis. Inflamm Res. 2005; 54: 395–411.

67 Young JL, Libby P, Schonbeck U. Cytokines in the pathogen- esis of atherosclerosis. Thromb Haemost 2002; 88: 554–67.

68 Harvey EJ, Ramji DP. Interferon-gamma and atherosclerosis:

pro- or anti-atherogenic? Cardiovasc Res 2005; 67: 11–20.

69 Barath P, Fishbein MC, Cao J, Berenson J, Helfant RH, Forrester JS. Detection and localization of tumor necrosis fac- tor in human atheroma. Am J Cardiol 1990; 65: 297–302.

70 Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Libby P. Enhanced expression of vascular matrix metallopro- teinases induced in vitro by cytokines and in regions of human atherosclerotic lesions. Ann N Y Acad Sci 1995; 748: 501–507.

71 Taubman MB, Fallon JT, Schecter AD et al. Tissue factor in the pathogenesis of atherosclerosis. Thromb Haemost. 1997; 78:

200–204.

72 Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT.

Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure:

results of the anti-TNF Therapy Against Congestive Heart

Failure (ATTACH) trial. Circulation 2003; 107: 3133–40.

(12)

73 Muller-Ehmsen J, Schwinger RH. TNF and congestive heart failure: therapeutic possibilities. Expert opinion on therapeutic targets. 2004; 8: 203–209.

74 Boisvert WA. Modulation of atherogenesis by chemokines.

Trends Cardiovasc Med 2004; 14: 161–65.

75 Shin WS, Szuba A, Rockson SG. The role of chemokines in human cardiovascular pathology: enhanced biological insights. Atherosclerosis 2002; 160: 91–102.

76 Ballantyne CM, Nambi V. Markers of inflammation and their clinical significance. Atheroscler Suppl 2005; 6: 21–29.

77 Leu HB, Wu CC, Wu TC, Lin SJ, Chen JW. Fluvastatin reduces oxidative stress, decreases serum monocyte chemotac- tic protein-1 level and improves endothelial function in patients with hypercholesterolemia. J Formos Med Assoc 2004; 103: 914–20.

78 Charo IF, Taubman MB. Chemokines in the pathogenesis of vascular disease. Circ Res 2004; 95: 858–66.

79 Ito T, Ikeda U. Inflammatory cytokines and cardiovascular dis- ease. Curr Drug Targets 2003; 2: 257–65.

80 Boisvert WA, Santiago R, Curtiss LK, Terkeltaub RA. A leuko- cyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest 1998; 101: 353–63.

81 Yamagishi M, Higashikata T, Ishibashi-Ueda H et al. Sustained upregulation of inflammatory chemokine and its receptor in aneurysmal and occlusive atherosclerotic disease: results from tissue analysis with cDNA macroarray and real-time reverse transcriptional polymerase chain reaction methods. Circ J 2005; 69: 1490–95.

82 Moreau M, Brocheriou I, Petit L, Ninio E, Chapman MJ, Rouis M. Interleukin-8 mediates downregulation of tissue inhibitor of metalloproteinase-1 expression in cholesterol- loaded human macrophages: relevance to stability of athero- sclerotic plaque. Circulation 1999; 99: 420–26.

83 Martins TB, Anderson JL, Muhlestein JB et al. Risk factor analysis of plasma cytokines in patients with coronary artery disease by a multiplexed fluorescent immunoassay. Am J Clin Pathol 2006; 125: 906–13.

84 Mallat Z, Besnard S, Duriez M et al. Protective role of inter- leukin-10 in atherosclerosis. Circ Res 1999; 85: e17–24.

85 Namiki M, Kawashima S, Yamashita T et al. Intramuscular gene transfer of interleukin-10 cDNA reduces atherosclerosis in apolipoprotein E-knockout mice. Atherosclerosis 2004; 172:

21–29.

86 Mallat Z, Heymes C, Ohan J, Faggin E, Leseche G, Tedgui A.

Expression of interleukin-10 in advanced human atheroscle- rotic plaques: relation to inducible nitric oxide synthase expression and cell death. Arterioscler Thromb Vasc Biol 1999;

19: 611–16.

87 Terkeltaub RA. IL-10: An ‘immunologic scalpel’ for athero- sclerosis? Arterioscler Thromb Vasc Biol 1999; 19: 2823–25.

88 Pinderski Oslund LJ, Hedrick CC, Olvera T et al. Interleukin-10 blocks atherosclerotic events in vitro and in vivo. Arterioscler Thromb Vasc Biol 1999; 19: 2847–53.

89 Poe JC, Wagner DH Jr, Miller RW, Stout RD, Suttles J. IL-4 and IL-10 modulation of CD40-mediated signaling of mono- cyte IL-1beta synthesis and rescue from apoptosis. J Immunol 1997; 159: 846–52.

90 Anguera I, Miranda-Guardiola F, Bosch X et al. Elevation of serum levels of the anti-inflammatory cytokine interleukin-10 and decreased risk of coronary events in patients with unsta- ble angina. Am Heart J 2002; 144: 811–17.

91 Smith DA, Irving SD, Sheldon J, Cole D, Kaski JC. Serum levels of the antiinflammatory cytokine interleukin-10 are decreased in patients with unstable angina. Circulation 2001;

104: 746–49.

92 Heeschen C, Dimmeler S, Hamm CW et al. Serum level of the antiinflammatory cytokine interleukin-10 is an important prognostic determinant in patients with acute coronary syn- dromes. Circulation 2003; 107: 2109–14.

93 Davis VA, Persidskaia RN, Baca-Regen LM, Fiotti N, Halloran BG, Baxter BT. Cytokine pattern in aneurysmal and occlusive disease of the aorta. J Surg Res 2001; 101: 152–56.

94 Metcalfe JC, Grainger DJ. Transforming growth factor-beta and the protection from cardiovascular injury hypothesis.

Biochem Soc Trans 1995; 23: 403–406.

95 Grainger DJ. Transforming growth factor beta and atheroscle- rosis: so far, so good for the protective cytokine hypothesis.

Arterioscler Thromb Vasc Biol 2004; 24: 399–404.

96 Yamagami S, Yokoo S, Mimura T, Amano S. Effects of TGF- beta2 on immune response-related gene expression profiles in the human corneal endothelium. Invest Ophthalmol Vis Sci 2004; 45: 515–21.

97 Ashcroft GS. Bidirectional regulation of macrophage func- tion by TGF-beta. Microbes Infect 1999; 1: 1275–82.

98 Turner M, Chantry D, Feldmann M. Transforming growth factor beta induces the production of interleukin 6 by human peripheral blood mononuclear cells. Cytokine 1990; 2: 211–16.

99 Kulkarni AB, Huh CG, Becker D et al. Transforming growth factor beta 1 null mutation in mice causes excessive inflam- matory response and early death. Proc Natl Acad Sci U S A 1993; 90: 770–74.

100 Grainger DJ, Mosedale DE, Metcalfe JC, Bottinger EP.

Dietary fat and reduced levels of TGFbeta1 act synergistically to promote activation of the vascular endothelium and forma- tion of lipid lesions. J Cell Sci 2000; 113(Pt 13): 2355–61.

101 Mallat Z, Gojova A, Marchiol-Fournigault C et al. Inhibition of transforming growth factor-beta signaling accelerates athero- sclerosis and induces an unstable plaque phenotype in mice.

Circ Res 2001; 89: 930–34.

102 Lutgens E, Gijbels M, Smook M et al. Transforming growth factor-beta mediates balance between inflammation and fibrosis during plaque progression. Arterioscler Thromb Vasc Biol 2002; 22: 975–82.

103 Erren M, Reinecke H, Junker R et al. Systemic inflammatory parameters in patients with atherosclerosis of the coronary and peripheral arteries. Arterioscler Thromb Vasc Biol 1999;

19: 2355–63.

Références

Documents relatifs

À partir de ces variables, l’élaboration d’un questionnaire a ciblé les éléments suivants : démographie (une question), pratiques existantes en termes d’évaluation

The oxidative stress linked to inflammation plays an important role in (i) endothelial dysfunction, with reduced nitric oxide (NO) bioavailability, (ii)

One of the interventional studies in our narrative review [ 72 ] demonstrates that a physical exercise program can induce an increase in cardiorespiratory fitness that correlates

Electrocardiographic and haemodynamic alterations caused by three different test solutions of local anaesthetics to detect accidental intravascular injection in children..

Rapamycin given in vivo to rats therefore inhibited the mTOR signaling pathway in a manner very similar to that seen with direct cell exposure, inhibiting both mTORC1

Expression of insulin receptor (IR) A and B isoforms, IGF-IR, and IR/IGF-IR hybrid receptors in vascular smooth muscle cells and their role in cell migration in

The apoptosis observed in ApoE -/- :PAI-1 -/- plaques appears to be dependent on plasminogen activation, as suggested by the six-fold higher plasmin activity detected

To elucidate these mechanisms, and to better understand plaque calcification heterogeneity among vascular beds, we used our ECLAGEN biocollection of human atherosclerotic lesions