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

European Heart Journal (2005) 26, 1700–1704

doi:10.1093/eurheartj/ehi282
Review

Dendritic cells in atherosclerosis: current status of the


problem and clinical relevance
Yuri V. Bobryshev*
Surgical Professorial Unit, Level 5, DeLacy Building, St Vincent’s Hospital Sydney, University of New South Wales,
Darlinghurst, New South Wales 2010, Australia
Received 8 December 2004; revised 3 March 2005; accepted 24 March 2005; online publish-ahead-of-print 26 April 2005

KEYWORDS Dendritic cells (DCs) are the most potent antigen-presenting cells. DCs were identified in arteries in
Dendritic cells; 1995 and, since then, further knowledge has been gained indicating the importance of DCs in athero-
Arteries; sclerosis. Vascular DCs have been shown to become activated from a very early stage of atherosclerosis.

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on April 22, 2013


Atherosclerosis; Some DCs cluster with T cells directly within atherosclerotic lesions, while others migrate to lymphoid
Plaque destabilization organs to activate T cells. Dyslipidaemia systemically alters DC function and recent findings suggest that
DCs play a role in plaque destabilization. This review summarizes the current status of the problem.

Introduction cell interactions, the presence of co-stimulatory molecules


on DCs is required for T cell activation and the differen-
Initially described in the skin by Langerhans in 1868, dendri- tiation into effector cells. In the absence of sufficient co-
tic cells (DCs) were identified as antigen-presenting cells in stimulation, T cells contacting DCs exhibit anergy or
1973 by the pioneering work of Steinman and Cohn.1 In all undergo apoptosis.4–9 In DC/T cell contacts, the secretion
tissues and all pathological conditions, DCs are minor cell or lack of secretion of cytokines, in particular interleikin-
populations but they are the most potent antigen-presenting 12 (IL-12), by DCs is instrumental in the differentiation of
cells.2 T cells into type 1 (Th1) or type 2 (Th2) effector T cells,
In arteries, DCs were originally identified in 1995.3 Since respectively.4–9
then, knowledge has been gained indicating the importance The growing interest in DC physiology reflects the import-
of DCs in atherogenesis. This review summarizes the current ance of DCs in the initiation and progression of various dis-
status of the problem. eases, including tumour, viral and bacterial infections, and
various autoimmune diseases.2,4–9 The ability of DCs to
elicit immune responses and the availability of DC culture
DCs and immunity
systems has led to their use in cancer immunotherapy
In recent years, the biology and pathophysiology of DCs have and therapeutic intervention in autoimmunity and
been highlighted in a number of reviews and mono- transplantation.2,4,9,10
graphs.2,4–9 DCs are members of both innate and adaptive
immune systems.4–8 As members of the innate immune
system, DCs can respond to danger signals by immediately Origin, migratory routes, and histological
generating protective cytokines.4–9 As the key element of nomenclature of DCs
the adaptive immune system, DCs respond to danger DCs originate from a common CD34þ progenitor in the bone
signals by the acquisition of the capacity to direct the devel- marrow.4–9 Their development involves three stages for
opment of primary immune responses appropriate to the which the terms, ‘precursors’, ‘immature’, and ‘mature’
type of danger.4–9 DCs have a potent antigen-presenting are used.2,4–9 DC precursors circulate via the bloodstream
capacity for the stimulation of naive, memory, and effector to reach their target tissues where they take up residence
T cells.4–9 DCs are capable of activating not only convention- at sites of potential antigen entry.2,4–6 In this stage, DCs
al T cells but also natural killer T (NKT) cells.9 During the can be identified in essentially all tissues but are mostly con-
development of an adaptive immune response, T cells form centrated along epithelial and body cavity surfaces.2,4–6 In
direct contacts with DCs and respond to peptide antigen dis- these locations, DCs continuously and efficiently sample
played on major histocompatibility complex (MHC) class II the antigenic content of their microenvironment by
and class I molecules present on DC surfaces.4–9 In DC/T phagocytosis, by high-volume fluid phase macropinocytosis,
by receptor-mediated endocytosis, or by direct contact
* Corresponding author. Tel: þ61 2 8382 2395; fax: þ61 2 9314 7654. with necrotic, apoptotic, or virally infected cells.2,4–6
E-mail address: y.bobryshev@unsw.edu.au Internalized antigens are degraded into short peptides that

& The European Society of Cardiology 2005. All rights reserved. For Permissions, please e-mail: journals.permissions@oupjournals.org
DCs in atherosclerosis 1701

are loaded onto nascent class I and class II MHC or CD1 mol- Langerhans cells which express E-cadherin and contain
ecules for subsequent display on the cell surface.2,4–6 Birbeck granules with Lag-antigen/Langerin. This pathway
Because, in this stage, DCs are not yet able to stimulate T is regulated by granulocyte macrophage colony stimulating
cells, these DCs are termed ‘immature’ or ‘processing’ factor (GM-CSF) and tumour necrosis factor a (TNF a). The
DCs.2,4–6 Processing DCs exit the non-lymphoid tissues and second is a monocyte-related pathway.2,4–6 This pathway
migrate via the afferent lymph into lymphoid tissues, such does not yield typical Langerhans cells in the presence of
as the spleen and lymph nodes, where DCs then complete GM-CSF and TNF a, but leads to interstitial DCs displaying
their maturation.2,4–6 Maturation of DCs involves the high levels of CD14 and CD68 during their development.
down-regulation of endocytotic activity and the up-regu- The third is a lymphoid pathway which provides DCs for
lation of the adhesion molecules (CD11a, CD50, CD54, and the thymus medulla and the T cell areas of lymphoid
CD58), co-stimulatory molecules (CD40, CD80/B7.1, CD86/ tissues. This subset of DCs lacks the myeloid marker
B7.2) and antigen-presenting molecules, including the CD11b. ‘Myeloid’ DCs are required for T cell activation
class I and class II of MHC proteins and CD1 molecules.2,4–6 whereas ‘lymphoid’ DCs induce T cell tolerance.2,4 The
Co-expression of the adhesion, co-stimulatory, and development of different DC subsets is influenced by
antigen-presenting molecules enables DCs to form contacts varying combinations of cytokine growth factors, including
with and activate T cells.2,4–6 It has been recognized also TNF family members (TNF-a, TRANCE/RANK), GM-CSF, IL-4,
that the maturation of some DCs can occur in the peripheral stem cell factor, transforming growth factor-b, and flt-3
non-lymphoid tissues4,6 and that DCs can present antigenic ligand.2,4–6
peptides to circulating T cells.4 The trafficking of DCs into
tissue sites is regulated by chemokines.2,4–6 At different

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on April 22, 2013


DCs in the non-diseased arterial wall
stages of their differentiation, DCs produce distinct compo-
sitions of cytokines which affect neighbouring cells.2,4–6 Small numbers of DCs, predominantly localized along the
The histological nomenclature of DCs is rather complex. In endothelium in the subendothelial layer, are present in the
peripheral blood, circulating DCs are known as blood DCs.4 In intima of apparently normal, non-diseased arteries.3,11
peripheral blood, circulating DCs represent the migratory Electron microscopy revealed that the normal arterial
form of DC precursors. Immature DCs are known as intima harbours a distinct subtype of the DC family display-
Langerhans cells in the skin and other epithelial surfaces ing unique ultrastructural features.3,11,12 Vascular DCs,
and as interstitial DCs in interstitial spaces of organs such through their processes, form focal networks in some
as the heart, kidney, and lung.2,4–6 The migratory form of intimal areas.13 Wick et al. 14,15 have suggested that in the
processing DCs in the afferent lymph is known as veiled normal intima, DCs are a key element of the so-called
cells. Interdigitating cells in the lymphoid organs represent vascular-associated lymphoid tissue (VALT). VALT is
mature DCs.2,4–6 analogous to the mucosa-associated lymphoid tissue of the
The morphological appearance of DCs varies, but some respiratory and gastrointestinal tracts.14 VALT consists of
members of the DC family possess distinctive cytoplasmic aggregates of vascular DCs, T cells, and resident macro-
structures.4 Langerhans cells contain Birbeck granules, phages distributed throughout the subendothelial layer of
while the tubulovesicular system is well developed in inter- the arterial intima along the luminal endothelial mono-
digitating cells.4 Accumulating evidence suggests that both layer.15 It has been suggested that VALT screens ‘vascular
Birbeck granules and the tubulovesicular system are tissue’ for potentially harmful antigens.14,15 Besides their
involved in antigen processing and antigen presentation.4 intimal location, small numbers of resident vascular DCs
DCs are characterized by the presence of numerous thin are present also in the adventitia, usually in close proximity
cytoplasmic processes (dendrites) as well as large cyto- to capillaries forming the vasa vasorum.16
plasmic veils that are continuously extended and retracted.4
The surface molecular content of DCs varies depending on
Vascular DCs in pre-atherosclerotic stage
the tissue location and on the stage of cell maturation,
and DCs display either the molecules essential for antigen The numbers of vascular DCs in the intima of athero-
capture and antigen processing or the molecules essential sclerosis-prone and atherosclerosis-resistant areas of the
for their specialized interactions with lymphocytes.2,4–6 non-diseased aorta have been assessed and it has been
The expression of the CD83 molecule, a member of the found that, in atherosclerosis-prone areas, there were
immunoglobulin superfamily with unknown functions, as more DCs than in atherosclerosis-resistant areas.17 In ather-
well as the expression of antigen-presenting CD1 molecules osclerosis-prone areas, DCs were found to form cell clus-
including CD1a, CD1b, CD1c, and CD1d are essentially ters.17 The accumulation of DCs has also been identified in
restricted to DCs.4 Fascin (55 kDa actin-bundling protein) the intima of the carotid arteries of children, 8 weeks to
is a reliable cytoplasmic marker for identifying different 10 years of age, at sites subjected to major haemodynamic
DC subtypes, while Lag-antigen and Langerin are uniquely stress and predisposed to the development of atherosclero-
present in Birbeck granules and Birbeck granule-associated sis.18 In other pathological conditions, the accumulation of
structures in Langerhans cells.4 S100 (S100A1 and S100B) clustering DCs is thought to be an indicator of autoimmune
proteins, the function of which in DCs are poorly under- processes.2,4 According to the autoimmune hypothesis of
stood, are expressed intensely by both mature and imma- atherosclerosis proposed by Wick et al.,14 VALT activation
ture DCs. The cell adhesion molecule E-cadherin is by auto-antigens is responsible for initiating immune
expressed by only some subsets of DCs.4 responses in the arterial wall which lead to atherosclerotic
Three pathways by which the CD34þ DC precursor can alteration. The observations of DCs accumulating and clus-
develop into immature DCs have been recognized.2,4–6 The tering in atherosclerosis-prone areas of the normal aorta
first pathway leads to the differentiation of typical and carotid arteries3,15,17,18 support the concept that
1702 Y.V. Bobryshev

immune mechanisms are involved in the formation of ather- found to cluster with T cells.26 DCs clustering with T cells
osclerotic lesions from the very early stages of the display the intercellular cell adhesion molecule-1 and vascu-
disease.19–21 lar cell adhesion molecule-1,26 interactions of which with
leukocyte function-associated antigen-1 and very late acti-
vation antigen-4, respectively, are essential for T cell acti-
DCs in atherosclerotic lesions
vation.31 In DC–T cell interactions, DCs display CD4032 and
In atherosclerotic arteries, the number of DCs express high levels of HLA–DR.26 Vascular DCs also express
increase.11,22–24 Vascular DCs residing in the intima DC-SIGN33 and some DCs display CD83, a marker of DC acti-
become activated in the earlier stages of atherosclero- vation.22 In atherosclerotic lesions, DCs express not only
sis.17,22,25 Advanced atherosclerotic plaques become high levels of class II MHC molecules but also display on
enriched by DCs invading the atherosclerotic plaques from their surfaces CD1 molecules,3,25,26,34 which have been
the adventitia, along with neovascularization associated recognized as antigen-presenting molecules in the same
with vasa vasorum.26 In addition to these originally resident sense as the classical MHC class I and II molecules.35,36 It
vascular DCs, atherosclerotic plaques may be invaded by has been found also that CD1d, an antigen-presenting mol-
blood DCs via inflamed neovessels.22,26 Monocytes that infil- ecule responsible for the presentation of lipid antigens,35,36
trate the intima from the very early stages of atherosclero- is expressed in atherosclerotic lesions25,34,37 and that its
sis27 may contribute to an increased DC population, as well. expression is restricted to DCs.25 The effects of the athero-
Depending on the micro-environmental conditions such as sclerotic environment on DCs are still poorly studied but it
the content and composition of cytokines, monocytes has been shown that in atherosclerotic lesions, DCs are

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on April 22, 2013


migrating through the luminal endothelial monolayer from activated and display an abundance of heat shock protein-
the blood may differentiate into macrophages or into 70.25 In contrast to other members of the DC family, the
DCs.28 DC adhesion and migration are modulated by pathophysiological significance of DCs as pro-
changes in endothelial function.29 In vitro experiments inflammatory and auto-immune cells in atherosclerosis
have revealed that DC adhesion and transmigration are is poorly understood.
markedly increased after exposing endothelial cells to Atherosclerosis displays features of immune activation
hypoxia, oxidized low density lipoproteins, and TNF-a.29 both locally and systemically.19–21 Angeli et al. 38 demon-
The inhibition of endothelial NO synthase increases DC strated that dyslipidemia associated with atherosclerotic
binding and transmigration, while the augmentation of disease systemically alters the DC function. In different
endothelial NO synthase activity has been found to pathophysiological conditions, inflammatory cytokines and
prevent DC adhesion.29 These findings suggest that the chemokines are known to be responsible for DC
adhesion and migration of DCs are increased by stimuli activation and migration,2,4–9 but the peculiarities of the
known to accelerate atherogenesis.29 effects of cytokines and chemokines on DCs in
Accumulating evidence supports a view that, in the arter- atherosclerosis have not yet been investigated. Nicotine,
ial wall, DCs are involved in antigen capture and antigen which is known to accelerate atherosclerosis development,
processing, as are DCs in other peripheral tissues.22,26,30 It activates DCs and augments their capacity to stimulate T
has been suggested that the migratory routes of vascular- cell proliferation and cytokine secretion.39 Modified lipopro-
associated DCs are similar to those known for Langerhans teins accumulating in atherosclerotic lesions likely trigger
cells in the skin and DCs in other peripheral tissues.22,26 DC activation, especially as in in vitro studies, oxidized
After engulfing antigen in the arterial wall, vascular DCs low density lipoproteins have been shown to promote
likely migrate as veiled cells via the afferent lymph into mature DC transition from monocytes40 and induce promi-
regional lymph nodes where they activate T cells. nent DC clustering.41 C1q, which is thought to be involved
Supporting this possibility are the immunohistochemical in capturing immune complexes in the lymphoid tissue, has
observations showing that the number of interdigitating been found to be expressed by vascular DCs.42
cells in para-aortic and jugulodigastric lymph nodes DCs populating atherosclerotic lesions might be involved
attached to atherosclerotic arterial wall segments exceed in the regulation of innate immunity in athero-
those in the lymph nodes attached to non-atherosclerotic sclerosis,43–45 especially as the presence of Chlamydia
arterial segments.22 Besides their migration to lymph pneumoniae has been identified in the cytoplasm of vascular
tissues, it has been suggested that some DCs emigrate to DCs.45 Spanbroek et al. 46 reported the expression of
the blood stream.30 5-lipoxygenase by DCs in atherosclerotic lesions.
An experimental in vivo study revealed trafficking of 5-lipoxygenase pathway is the major source of potent pro-
monocyte-derived cells from atherosclerotic plaques inflammatory leukotrienes issued from the metabolism of
during lesion regression but little emigration was detected arachidonic acid.47 These lipid mediators released by vascu-
from progressive plaques, suggesting that the progression lar DCs may act as physiological autocrine and paracrine sig-
of atherosclerotic plaques may result not only from robust nalling molecules and play a central role in regulating the
monocyte recruitment into arterial walls but also from interaction between innate and adaptive immunity.46 The
reduced emigration of these cells from lesions.30 This is in expression of C1q by vascular DCs42 may also relate to
agreement with a histopathological study of human arterial innate immunity in atherosclerosis.
tissue which indicated that only some DCs may migrate to
the lymph nodes while other DCs may activate T cells
DCs and plaque destabilization
directly within the intima.26 Mapping of the distribution of
DCs in human atherosclerotic lesions revealed that DCs are In atherosclerotic plaques, the number of DCs markedly
most frequent in areas enriched with T cells and, particu- increases with more than 90% of DCs accumulating in
larly so, within inflammatory infiltrates where DCs are plaque shoulders, which represent plaque rupture-prone
DCs in atherosclerosis 1703

regions.26,48 Yilmaz et al. 48 have found that up to 70% of DCs with aGalCer need to be ligated prior to returning them to
in the shoulders of vulnerable carotid plaques express the bloodstream.
markers of DC activation such as CD83 and DC-LAMP. The
clustering of T cells with activated DCs cells has been
observed in plaque rupture-prone regions.26,48 In these
Concluding remarks
regions, DCs form contacts not only with conventional T DCs are present in their immature forms in non-diseased
cells but also with NKT cells.49 It has been suggested that arteries and become activated during atherogenesis. Some
the formation of clusters of DCs with T cells in rupture- DCs cluster with T cells directly within atherosclerotic
prone regions is associated with plaque destabilization.48,49 lesions, while others migrate to lymphoid organs to activate
A comparison of DC numbers in atherosclerotic plaques T cells. The identification of activated DCs in atherosclerosis-
obtained from patients with and without acute ischaemic like lesions in animal models57–59 may facilitate the
symptoms has revealed that DC numbers are markedly elev- investigation of the functions of vascular DCs and their use
ated in patients with acute ischaemic symptoms.48 Yilmaz for atherosclerosis immunotherapy.
et al. 48 have also reported that in statin-treated patients,
atherosclerotic plaques contain significantly lower numbers
of DCs than plaques in patients without statin treatment. Acknowledgement
In vitro experiments have revealed that statins inhibit the I would like to thank St Vincent’s Clinic Foundation, Sydney, for sup-
maturation and antigen-presenting function of DCs, thus porting this work.
possibly contributing to the beneficial effects of statins in
atherosclerosis.50

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on April 22, 2013


Recently, Ranjit et al. 51 have reported that, in patients References
with unstable angina pectoris, DCs were functionally 1. Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral
altered. In contrast to healthy donors, DC co-stimulatory lymphoid organs of mice. I. Morphology, quantification, tissue distri-
molecule CD86 was upregulated in patients with unstable bution. J Exp Med 1973;137:1142–1162.
2. Banchereau J, Steinman RM. Dendritic cells and control of immunity.
angina.51 The abilities of DCs to stimulate T cells and Nature 1998;392:245–252.
produce cytokines also differed in the cells obtained from 3. Bobryshev YV, Lord RSA. Ultrastructural recognition of cells with
patients with unstable angina and from healthy donors.51 dendritic cell morphology in human aortic intima. Contacting inter-
actions of vascular dendritic cells in athero-resistant and athero-prone
areas of the normal aorta. Arch Histol Cytol 1995;58:307–322.
4. Lotze MT, Thomson AW, eds. Dendritic cells: Biology and Clinical
DCs and therapeutic intervention Applications. 2nd ed. San Diego, CA: Academic Press; 2001.
5. Lipscomb MF, Masten BJ. Dendritic cells: immune regulators in health and
in atherosclerosis disease. Physiol Rev 2002;82:97–130.
DCs may be used for the suppression of damaging immune 6. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells.
Annu Rev Immunol 2003;21:685–711.
responses in atherosclerosis.52 Approaches in atherosclerosis 7. Heath WR, Belz GT, Behrens GM, Smith CM, Forehan SP, Parish IA, Davey
immunotherapy might be similar to those employed nowa- GM, Wilson NS, Carbone FR, Villadangos JA. Cross-presentation, dendritic
days for cancer immunotherapy.4,53,54 One of these is cell subsets, and the generation of immunity to cellular antigens. Immunol
based on a technique in which DCs, isolated from the periph- Rev 2004;199:9–26.
8. de Jong EC, Smits HH, Kapsenberg ML. Dendritic cell-mediated T cell
eral blood, are activated (‘pulsed’) with the appropriate
polarization. Springer Semin Immunopathol 2005;26:289–307.
antigen ex vivo and then the ‘pulsed’ cells are returned to 9. Morel PA, Feili-Hariri M, Coates PT, Thomson AW. Dendritic cells, T cell
the bloodstream.4,53 tolerance and therapy of adverse immune reactions. Clin Exp Immunol
In atherosclerosis, the nature of the antigen(s) respon- 2003;133:1–10.
sible for the activation of T cells is still not well under- 10. O’Neill DW, Adams S, Bhardwaj N. Manipulating dendritic cell biology for
the active immunotherapy of cancer. Blood 2004;10:2235–2246.
stood.19,20 For atherosclerosis immunotherapy, DCs 11. Bobryshev YV. Dendritic cells in atherosclerosis. In: Lotze MT, Thomson
obtained from the peripheral blood can be pulsed ex vivo AW, eds. Dendritic cells: Biology and Clinical Applications. 2nd ed. San
by cultivating them in a medium containing a total extract Diego, CA: Academic Press; 2001. p547–557.
or suspension of atherosclerotic plaque material. Such culti- 12. Bobryshev YV, Ikezawa T, Watanabe T. Formation of Birbeck granule-like
structures in vascular dendritic cells in human atherosclerotic aorta. Lag-
vation would imitate the activation of DCs occurring in
antibody to epidermal Langerhans cells recognizes cells in the aortic
plaques in vivo. In contrast to cancer immunotherapy, wall. Atherosclerosis 1997;133:193–202.
immune responses in atherosclerosis need to be suppressed. 13. Millonig G, Niederegger H, Rabl W, Hochleitner BW, Hoefer D, Romani N,
As the presentation of peptide–MHC complexes to T cells by Wick G. Network of vascular-associated dendritic cells in intima of
DCs in the absence of co-stimulatory signals leads to anergy healthy young individuals. Arterioscler Thromb Vasc Biol 2001;21:
503–508.
and apoptosis of T cells, co-stimulatory molecules on pulsed 14. Wick G, Romen M, Amberger A, Metzler B, Mayr M, Falkensammer G, Xu
DCs should be blocked ex vivo prior the cells being returned Q. Atherosclerosis, autoimmunity, and vascular-associated lymphoid
to the bloodstream. Recent achievements in developing the tissue. FASEB J 1997;11:1199–1207.
techniques for the suppression (ligation) of co-stimulatory 15. Waltner-Romen M, Falkensammer G, Rabl W, Wick G. A previously unrec-
ognized site of local accumulation of mononuclear cells. The vascular-
molecules and for the loading of DCs with antigens have
associated lymphoid tissue. J Histochem Cytochem 1998;46:1347–1350.
made it possible to generate DCs with desirable properties.4 16. Bobryshev YV, Lord RS. Vascular-associated lymphoid tissue (VALT) invol-
Recent studies indicate that NKT cells aggravate athero- vement in aortic aneurysm. Atherosclerosis 2001;154:15–21.
sclerosis.19 DCs may represent a useful tool for the regu- 17. Lord RS, Bobryshev YV. Clustering of dendritic cells in athero-prone areas
lation of NKT cell function in atherosclerosis. In one cancer of the aorta. Atherosclerosis 1999;146:197–198.
18. Millonig G, Malcom GT, Wick G. Early inflammatory-immunological lesions
immunotherapy technique, DCs pulsed with aGalCer ex in juvenile atherosclerosis from the Pathobiological Determinants of
vivo are returned to the bloodstream.55,56 For atherosclero- Atherosclerosis in Youth (PDAY)-study. Atherosclerosis 2002;160:
sis immunotherapy, co-stimulatory molecules on DCs pulsed 441–448.
1704 Y.V. Bobryshev

19. Hansson GK. Immune mechanisms in atherosclerosis. Arterioscler Thromb immunoregulatory component of the atherogenic micro-environment?
Vasc Biol 2001;21:1876–1890. Cardiovasc Res 2002;55:806–819.
20. Hansson GK, Libby P, Schonbeck U, Yan ZQ. Innate and adaptive immunity 42. Cao W, Bobryshev YV, Lord RS, Oakley RE, Lee SH, Lu J. Dendritic cells in
in the pathogenesis of atherosclerosis. Circ Res 2002;91:281–291. the arterial wall express C1q: potential significance in atherogenesis.
21. Wick G, Knoflach M, Xu Q. Autoimmune and inflammatory mechanisms in Cardiovasc Res 2003;60:175–186.
atherosclerosis. Annu Rev Immunol 2004;22:361–364. 43. Kis Z, Pallinger E, Endresz V, Burian K, Jelinek I, Gonczol E, Valyi-Nagy
22. Bobryshev YV. Dendritic cells and their involvement in atherosclerosis. I. The interactions between human dendritic cells and microbes; possible
Curr Opin Lipidol 2000;11:511–517. clinical applications of dendritic cells. Inflamm Res 2004;53:413–423.
23. Bobryshev YV, Lord RS. S-100 positive cells in human arterial intima and in 44. Netea MG, van der Graaf C, Van der Meer JW, Kullberg BJ. Toll-like recep-
atherosclerotic lesions. Cardiovasc Res 1995;29:689–696. tors and the host defense against microbial pathogens: bringing speci-
24. Bobryshev YV, Watanabe T. Subset of vascular dendritic cells transforming ficity to the innate-immune system. J Leukoc Biol 2004;75:749–555.
into foam cells in human atherosclerotic lesions. Cardiovasc Pathol 45. Bobryshev YV, Cao W, Phoon MC, Tran D, Chow VT, Lord RS, Lu J. Detec-
1997;6:321–331. tion of Chlamydophila pneumoniae in dendritic cells in atherosclerotic
25. Bobryshev YV, Lord RS. Expression of heat shock protein-70 by dendritic lesions. Atherosclerosis 2004;173:185–195.
cells in the arterial intima and its potential significance in atherogenesis. 46. Spanbroek R, Grabner R, Lotzer K, Hildner M, Urbach A, Ruhling K, Moos
J Vasc Surg 2002;35:368–375. MP, Kaiser B, Cohnert TU, Wahlers T, Zieske A, Plenz G, Robenek H,
26. Bobryshev YV, Lord RSA. Mapping of vascular dendritic cells in athero- Salbach P, Kuhn H, Radmark O, Samuelsson B, Habenicht AJ. Expanding
sclerotic arteries suggests their involvement in local immune- expression of the 5-lipoxygenase pathway within the arterial wall
inflammatory reactions. Cardiovasc Res 1998;37:799–810. during human atherogenesis. Proc Natl Acad Sci USA 2003;100:
27. Ross R. Atherosclerosis - an inflammatory disease. N Engl J Med 1999; 1238–1243.
340:115–126. 47. Castellano G, Woltman AM, Schena FP, Roos A, Daha MR, van Kooten
28. Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. Differen- C. Dendritic cells and complement: at the cross road of innate and adap-
tiation of monocytes into dendritic cells in a model of transendothelial tive immunity. Mol Immunol 2004;41:133–140.
48. Yilmaz A, Lochno M, Traeg F, Cicha I, Reiss C, Stumpf C, Raaz D, Anger T,

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on April 22, 2013


trafficking. Science 1998;282:480–483.
Amann K, Probst T, Ludwig J, Daniel WG, Garlichs CD. Emergence of den-
29. Weis M, Schlichting CL, Engleman EG, Cooke JP. Endothelial determinants
dritic cells in rupture-prone regions of vulnerable carotid plaques.
of dendritic cell adhesion and migration: new implications for vascular
Atherosclerosis 2004;176:101–110.
diseases. Arterioscler Thromb Vasc Biol 2002;22:1817–1823.
49. Bobryshev YV, Lord RSA. Co-accumulation of dendritic cells and natural
30. Llodra J, Angeli V, Liu J, Trogan E, Fisher EA, Randolph GJ. Emigration of
killer T cells within rupture-prone regions in human atherosclerotic
monocyte-derived cells from atherosclerotic lesions characterizes
plaques. J Histochem Cytochem 2005, in press.
regressive, but not progressive, plaques. Proc Natl Acad Sci USA
50. Yilmaz A, Reiss C, Tantawi O, Weng A, Stumpf C, Raaz D, Ludwig J, Berger
2004;101:11779–11784.
T, Steinkasserer A, Daniel WG, Garlichs CD. HMG-CoA reductase inhibitors
31. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte
suppress maturation of human dendritic cells: new implications for ather-
emigration: the multistep paradigm. Cell 1994;76:301–314.
osclerosis. Atherosclerosis 2004;172:85–93.
32. Ozmen J, Bobryshev YV, Lord RS. CD40 co-stimulatory molecule
51. Ranjit S, Dazhu L, Qiutang Z, Yibo F, Yushu L, Xiang W, Shen CL, Yuan
expression by dendritic cells in primary atherosclerotic lesions in
T. Differentiation of dendritic cells in monocyte cultures isolated from
carotid arteries and in stenotic saphenous vein coronary artery grafts.
patients with unstable angina. Int J Cardiol 2004;97:551–555.
Cardiovasc Surg 2001;9:329–333.
52. Bobryshev YV. Can dendritic cells be exploited for therapeutic interven-
33. Soilleux EJ, Morris LS, Trowsdale J, Coleman N, Boyle JJ. Human athero- tion in atherosclerosis? Atherosclerosis 2001;154:511–512.
sclerotic plaques express DC-SIGN, a novel protein found on dendritic 53. Timmerman JM, Levy R. Dendritic cell vaccines for cancer immunother-
cells and macrophages. J Pathol 2002;198:511–516. apy. Annu Rev Med 1999;50:507–529.
34. Bobryshev YV, Lord RS. CD1 expression and the nature of CD1- 54. Markiewicz MA, Kast WM. Progress in the development of immunotherapy
expressing cells in human atherosclerotic plaques. Am J Pathol 2000; of cancer using ex vivo-generated dendritic cells expressing multiple
156:1477–1478. tumor antigen epitopes. Cancer Invest 2004;22:417–434.
35. Jackman RM, Moody DB, Porcelli SA. Mechanisms of lipid antigen presen- 55. Nieda M, Okai M, Tazbirkova A, Lin H, Yamaura A, Ide K, Abraham R, Juji
tation by CD1. Crit Rev Immunol 1999;19:49–63. T, Macfarlane DJ, Nicol AJ. Therapeutic activation of Valpha24 þ
36. Brigl M, Brenner MB. CD1: antigen presentation and T cell function. Annu Vbeta11þ NKT cells in human subjects results in highly coordinated sec-
Rev Immunol 2004;22:817–890. ondary activation of acquired and innate immunity. Blood 2004;
37. Melian A, Geng YJ, Sukhova GK, Libby P, Porcelli SA. CD1 expression in 103:383–389.
human atherosclerosis. A potential mechanism for T cell activation by 56. Seino KI, Fujii SI, Harada M, Motohashi S, Nakayama T, Fujisawa T,
foam cells. Am J Pathol 1999;15:775–786. Taniguchi M. Valpha14 NKT cell-mediated anti-tumor responses and
38. Angeli V, Llodra J, Rong JX, Satoh K, Ishii S, Shimizu T, Fisher EA, their clinical application. Springer Semin Immunopathol 2005, (published
Randolph GJ. Dyslipidemia associated with atherosclerotic disease sys- online 14 January 2005).
temically alters dendritic cell mobilization. Immunity 2004;21:561–574. 57. Bobryshev YV, Taksir T, Lord RS, Freeman MW. Evidence that dendritic
39. Aicher A, Heeschen C, Mohaupt M, Cooke JP, Zeiher AM, Dimmeler S. Nic- cells infiltrate atherosclerotic lesions in apolipoprotein E-deficient
otine strongly activates dendritic cell-mediated adaptive immunity: mice. Histol Histopathol 2001;16:801–808.
potential role for progression of atherosclerotic lesions. Circulation 58. Afek A, Harats D, Roth A, Keren G, George J. Evidence for the involve-
2003;107:604–611. ment of T cell costimulation through the B-7/CD28 pathway in athero-
40. Perrin-Cocon L, Coutant F, Agaugue S, Deforges S, Andre P, Lotteau sclerotic plaques from apolipoprotein E knockout mice. Exp Mol Pathol
V. Oxidized low-density lipoprotein promotes mature dendritic cell trans- 2004;76:219–223.
ition from differentiating monocyte. J Immunol 2001;167:3785–3791. 59. Ozmen J, Bobryshev YV, Lord RS, Ashwell KW. Identification of dendritic
41. Alderman CJ, Bunyard PR, Chain BM, Foreman JC, Leake DS, Katz DR. cells in aortic atherosclerotic lesions in rats with diet-induced hypercho-
Effects of oxidised low density lipoprotein on dendritic cells: a possible lesterolaemia. Histol Histopathol 2002;17:223–237.

You might also like