ArticlePDF AvailableLiterature Review

The vascular biology of macrophage migration inhibitory factor (MIF). Expression and effects in inflammation, atherogenesis and angiogenesis

Authors:

Abstract and Figures

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with chemokine-like functions. MIF is a critical mediator of the host immune and inflammatory response. Dysregulated MIF expression has been demonstrated to contribute to various acute and chronic inflammatory conditions as well as cancer development. More recently, MIF has been identified as an important pro-atherogenic factor. Its blockade could even aid plaque regression in advanced atherosclerosis. Promotion of atherogenic leukocyte recruitment processes has been recognised as a major underlying mechanism of MIF in vascular pathology. However, MIF's role in vascular biology is not limited to immune cell recruitment as recent evidence also points to a role for this mediator in neo-angiogenesis / vasculogenesis by endothelial cell activation and endothelial progenitor cell recruitment. On the basis of introducing MIF's chemokine-like functions, the current article focusses on MIF's role in vascular biology and pathology.
Content may be subject to copyright.
© Schattauer 2013 Thrombosis and Haemostasis 109.3/2013
1Theme Issue Article
The vascular biology of macrophage migration inhibitory factor (MIF)
Expression and effects in inflammation, atherogenesis and angiogenesis
Yaw Asare1; Martin Schmitt2,3; Jürgen Bernhagen1
1Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany; 2Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen
University, Aachen, Germany; 3Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, The Netherlands
Summary
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine
with chemokine-like functions. MIF is a critical mediator of the host
immune and inflammatory response. Dysregulated MIF expression has
been demonstrated to contribute to various acute and chronic inflam-
matory conditions as well as cancer development. More recently, MIF
has been identified as an important pro-atherogenic factor. Its block-
ade could even aid plaque regression in advanced atherosclerosis.
Promotion of atherogenic leukocyte recruitment processes has been
recognised as a major underlying mechanism of MIF in vascular pa-
thology. However, MIF’s role in vascular biology is not limited to im-
mune cell recruitment as recent evidence also points to a role for this
mediator in neo-angiogenesis / vasculogenesis by endothelial cell acti-
vation and endothelial progenitor cell recruitment. On the basis of in-
troducing MIF’s chemokine-like functions, the current article focusses
on MIF’s role in vascular biology and pathology.
Keywords
Chemokine, monocyte/macrophage, cardiovascular disease, athero-
sclerosis, (neo-)angiogenesis/vasculogenesis
Correspondence to:
Univ.-Prof. Dr. rer. nat. Jürgen Bernhagen
Institute of Biochemistry and Molecular Cell Biology
RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
Tel.: +49 241 80 88 840/31/41, Fax: +49 241 80 82 427
E-mail: jbernhagen@ukaachen.de
Received: November 18, 2012
Accepted after minor revision: December 3, 2012
Prepublished online: January 17, 2013
doi:10.1160/TH12-11-0831
Thromb Haemost 2013; 109: ■■■
Introduction:
chemokines and vascular biology
The processes governing vascular homeostasis, vascular repair
after acute injury and vascular remodelling during chronic disease
are controlled and driven by a plethora of factors. Among them
chemokines play a pivotal role at all levels of regulation.
Chemokines are small chemoattractant cytokines which have a
molecular weight of 8-12 kDa. Chemokines exert a broad variety
of functions in physiology and pathophysiology. In the context of
the current review article, we will focus on their role in leukocyte
chemotaxis, extravasation, as well as augmentation and/or attenu-
ation of angiogenesis (1-3). Based on the arrangement of four con-
served cysteine residues, chemokines are divided into four families
(CC, CXC, CX3C, and XC) (4). Numerous examples of key roles of
chemokines in vascular function, atherogenesis and vascular re-
pair exist. A joint role for several of these molecular players can
easily be rationalised, if one considers that the different steps of
leukocyte recruitment process, i.e. rolling, firm adhesion, and
transmigration are controlled by functionally specialised chemo-
kines, which act in a sequential and cooperative manner. However,
in the context of this review, we can only refer to some excellent
previous review articles covering the aspects of a balance between
chemokine redundancy and cooperativity in both the mainten-
ance of vascular homeostasis and vascular pathology (3, 5-8).
Chemokine structure and function may be modulated by di-,
tetrameric or perhaps even higher-order interactions. These inter-
actions are usually homomeric (for a comprehensive review please
see [9]), but chemokine function can also be modulated by hetero-
merisation, e.g. between CC and CXC chemokines. This may serve
to promote chemokine/receptor interactions. An example for the
latter is the heterodimerisation between CXCL4/PF4 and
CCL5/RANTES, which takes place in α-granules of human pla-
telets deposited at the glycosaminoglycan surface of endothelial
cells in vivo. In vitro and in vivo studies revealed a pathophysiologi-
cal function of such chemokine heterodimers (10, 11). In fact, von
Hundelshausen et al. demonstrated that heterodimerisation of
CCL5 and CXCL4 enhances CCL5-mediated monocyte recruit-
ment, while Koenen et al. additionally identified peptidic in-
hibitors specifically interrupting the CCL5/CXCL4 interface and
inhibiting vascular lesion formation in atherosclerotic Apoe–/–
mice (10, 11).
A subfamily of the CXC chemokine family is known as the
ELR+ chemokines and is defined by a glutamic acid-leucine-argi-
nine (ELR)-motif near the CXC sequence. In contrast to non-ELR
chemokines (ELR– CXC chemokines), chemokines from the ELR+
subfamily are potent inducers of physiological and pathological
angiogenesis. They play important roles in diseases like cancer, fi-
broproliferative disorders and chronic inflammation like athero-
sclerosis (12-14). A representative example for an ELR+ chemo-
kine is CXCL8/interleukin (IL-8). Thus, besides being a potent
neutrophil chemoattractant, CXCL8 also plays an important role
in neovascularisation in human non-small cell lung cancer (15),
human gastrointestinal cancers (16) and human ovarian and pros-
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
Thrombosis and Haemostasis 109.3/2013 © Schattauer 2013
2Asare et al. Vascular biology of MIF
tate cancer (17), where the CXCL8-CXCR2 axis regulates tumour
angiogenesis accompanied by correlative reduction or enhance-
ment of tumour growth. Further evidence for CXCL8-mediated
angiogenesis in diseased tissue was found in human coronary ar-
tery plaques, where CXCL8 has been shown to be overexpressed
(18). This might point to CXCL8-mediated growth of intra-plaque
vessels and plaque destabilisation (19).
As an example for a prominent CC chemokine, the
CCL2/CCR2 ligand receptor axis is important in monocyte che-
moattraction and transendothelial migration into areas of vascular
inflammation. CCL2 expression has been shown in atherosclerotic
lesions, likely exacerbating lesion progression through extensive
monocyte attraction and also through triggering firm monocyte
adhesion to the inflamed endothelium (20, 21). A deletion or at-
tenuation of CCL2 expression in atherosclerotic mouse models re-
sulted in decreased lesion formation (22, 23).
In addition to the four canonical chemokine classes, a group of
molecules sharing functional similarities with chemokines has
emerged as a fifth subclass. This class has been referred to as ‘che-
mokine-like function’ (CLF) chemokines, non-canonical chemo-
kines, or micro-chemokines. By definition, the CLF family of che-
mokines encompasses certain inflammatory and immune proteins
that exhibit chemokine-like functions such as chemotactic proper-
ties or leukocyte arrest-promoting effects but which neither for-
mally carry the typical N-terminal cysteine motif of the classical
chemokines nor the chemokine fold. Most members of this func-
tional family have been found to act as non-canonical ligands for
classical chemokine receptors. CLF chemokines thus further ex-
pand the degree of redundancy and promiscuity in chemokine/
chemokine receptor interactions, with consequences for angiogen-
esis regulation, as discussed below.
Examples are a cleavage fragment of tyrosyl tRNA synthetase
(TyrRS) which has been shown to act as a non-canonical CXCR1
ligand via the presence of an ELR-like motif (24). The N-terminal
‘mini-TyrRS’ domain has proangiogenic properties and induces
neutrophil chemotaxis through interaction with CXCR1 but not
CXCR2 (24, 25). Interestingly, the ELR motif is only exposed and
available for interaction with CXCR1 in the cleaved fragment (26).
Similarly, autoantigenic aminoacyl-RS, released under apoptotic
conditions, have leukocyte recruitment properties by triggering
CC receptors. Both histidyl-RS and its N-terminal fragment are
chemoattractants for several CCR5-expressing immune cells. As-
paraginyl-RS interacts with CCR3 (27). For these aminoacyl-RS,
the presence of specific surface charge distributions has been sug-
gested to mediate chemokine receptor usage. Furthermore, the
human antimicrobial peptides β-defensin-1 and -2 were identified
as non-cognate ligands for CCR6, mediating chemotaxis (28).
Again, although the sequence similarity between the β-defensins
and CCL20 is limited, it appears that a cluster of cationic amino
acids and shared electrostatic charge patterns account for the over-
lap in chemotactic activities. Promiscuity and mimicry mechan-
isms not only can be found endogenously in the host, but there are
several examples of parasite or viral chemokine mimicry, en-
compassing both mimicry of classical host chemokine structures
and CLF-type mimicry mechanisms (29, 30). Most prominently,
HIV-1 capsid protein gp120 interacts with host CXCR4 (and
CCR5) to direct leukocyte infection (29, 31). The nuclear protein
high-mobility-group binding protein 1 (HMGB1) has been shown
to exert numerous extracellular inflammatory functions. HMGB1
signals through several receptors (32, 33) and most recently, het-
eromeric complex formation between HMGB1 and CXCL12 was
identified to mediate HMGB1 chemokine activities through
CXCR4 (34).
In this review, we will focus on macrophage migration in-
hibitory factor (MIF), one of the first cytokines to be discovered
(35) and another recent addition to the CLF family of chemokines
(30, 36). In the next chapters, we will briefly outline both the
physiology and the pathophysiologic roles of MIF in inflammatory
and vascular disease, including underlying mechanisms such as its
non-cognate interactions with the chemokine receptors CXCR2
and CXCR4. With regard to MIF’s mechanisms of action, we will
solely focus on aspects relating to vascular biology and patho-
physiology.
MIF: structure, mechanism of action, and role
in inflammatory disease
MIF is an evolutionarily-conserved protein that is abundantly ex-
pressed in humans and non-primate mammals. In addition to its
functions as cytokine/chemokine and angiogenic factor (see
below), it has been suggested that MIF also has anti-oxidative in-
tracellular effects. The MIF structure is unique among cytokines.
MIF consists of 114 amino acids and has a molecular weight of
12.5 kDa. The three-dimensional structure of human MIF shows
that MIF crystallises as a trimer of three identical subunits, but
studies at more physiological concentrations imply that the
monomer may have crucial functions in vivo as well. Despite its
wide tissue distribution, the secretion of MIF is tightly regulated,
with relevant triggers such as hypoxia/ischaemia or oxidised low-
density lipoprotein (oxLDL) of particular importance for this re-
view article. Moreover, MIF expression is strongly up-regulated in
several disease conditions most importantly in vascular pathology
and tumourigenesis (37-39).
CD74, the membrane-expressed form of invariant chain (Ii)
and an MHC class II chaperone, was identified as the first MIF
plasma membrane receptor (40). CD74 expression is typically re-
stricted to class II-positive cells, but under inflammatory condi-
tions as well as in several tumour cell types, CD74 can be up-regu-
lated, even in the absence of class II expression. MIF binds to
CD74 by high affinity interaction in the nanomolar range, but sig-
nalling additionally requires the recruitment of signalling-compet-
ent co-receptors such as CD44 or CXCR2 and CXCR4 (see below).
CD74 alone mediates MIF binding, but MIF-induced MAPK sig-
nalling requires the coexpression of CD44. MIF signalling through
CD74/CD44 also involves the serine phosphorylation of the cyto-
plasmic tails of CD74 and CD44 and the recruitment of a Src-type
tyrosine kinase (41). An architectural similarity between the MIF
monomer and the CXCL8 dimer instigated biochemical investi-
gations to probe potential interactions between MIF and CXCR2.
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
© Schattauer 2013 Thrombosis and Haemostasis 109.3/2013
3Asare et al. Vascular biology of MIF
Receptor binding studies then revealed that MIF engages in a non-
cognate, high affinity interaction with CXCR2 (42). CXCR2 had
been known to be promiscuous previously, as it also is the receptor
for six other ELR+ CXC chemokines including CXCL8. This as
well as numerous functional data then suggested that MIF belongs
to the family of CLF chemokines. The signal transduction path-
ways triggered by MIF/CXCR2 have not been studied systemati-
cally, but initial evidence shows that MIF binding to CXCR2 leads
to Gi coupling and can elicit calcium transients. A limited screen of
other chemokine receptors as well as an observed chemotactic ef-
fects of MIF on T cells, which do not express CXCR2 and only low
levels of CD74, unraveled yet another MIF/chemokine receptor in-
teraction, i.e. that with CXCR4. MIF/CXCR4 interaction is less af-
fine than that between MIF and CXCR2 but still in the nanomolar
range. Further research into the receptors of MIF revealed that
CD74 forms heteromeric complexes with either CXCR2 or CXCR4
(42, 43).
During inflammation (44), endothelial cells do not only get ac-
tivated but also adjust their phenotypes to the inflammatory re-
sponse (45). Activation of endothelial cells (ECs), however, con-
tributes to both acute and chronic inflammatory diseases such as
sepsis, inflammatory bowel disease, rheumatoid arthritis, inflam-
matory lung disease, and atherosclerosis (46). There is ample evi-
dence now that MIF is a key mediator of all of these disease condi-
tions (37, 38, 47-49). Thus, MIF has been proven to play a pivotal
role in the pathogenesis of both acute and chronic inflammatory
diseases. Two prominent examples are sepsis and rheumatoid ar-
thritis.
First evidence implicating MIF in systemic infection and sepsis
dates back two decades ago when pituitary-derived MIF was found
to contribute to serum levels of MIF in the post-acute phase of en-
dotoxaemia. Employing a mouse model of endotoxic shock, MIF
was found to promote lethal endotoxaemia in mice (50). Indeed,
several and diverse models of septic shock have demonstrated the
crucial role of MIF in mediating sepsis (51-53).
Rheumatoid arthritis, a systemic chronic inflammatory dis-
order of the joints, is characterised by key pathological events in-
cluding diapedesis of leukocytes and the active participation of cy-
tokines such as tumour necrosis factor (TNF). MIF has been ex-
tensively described to play a role in rheumatoid arthritis by e.g. in-
ducing the secretion of CCL2 and to promote TNF production to
amplify leukocyte recruitment at yet another level (38, 54). Of
note, polymorphisms in the MIF gene functionally enhancing the
transcriptional activity of MIF have been linked to increased dis-
ease severity of rheumatoid arthritis and other inflammatory con-
ditions (38, 55).
MIF in the vasculature
Vascular endothelial cells exhibit a profound heterogeneity and
organ specificity in terms of their phenotype and protein ex-
pression patterns. Depending on the vessel or tissue they inhabit,
ECs are either strictly continuous with tight junctions, e.g. to
maintain the blood-brain barrier or discontinuous in the case of
the liver to allow for maximal fluid exchange. On the other hand,
ECs lining the glomerulus are strongly fenestrated to allow for op-
timal filtration results. In large arteries, an additional requirement
to resist high pressure and shear flow is found. ECs express various
molecules that have been described to be pivotal in the pathogen-
esis of numerous vascular diseases such as atherosclerosis and an-
giogenesis. Among these molecules are various adhesion mol-
ecules, such as vascular cell adhesion molecules (VCAMs), inter-
cellular adhesion molecules (ICAMs), selectins, and junctional ad-
hesion molecules (JAMs).
The expression levels of MIF in human ECs of both microvas-
cular and large artery origin have been shown to be upregulated
upon treatment with oxLDL (56) or thrombin (57) in a time- and
dose-dependent manner, suggesting a role for MIF in the vascula-
ture. MIF released upon oxLDL stimulation contributes to athero-
genic leukocyte recruitment (56, 58) (
Figure 1). Moreover, ex-
posing human ECs to hypoxia led to a release of substantial
amounts of MIF that was found to participate in the recruitment
and migration of endothelial progenitor cells (59). The expression
of MIF in the vasculature extends beyond the endothelial layer as
vascular smooth muscle cells (VSMCs) have also been shown to
express low levels of MIF (56); moreover, MIF expression was also
observed to be upregulated by oxLDL in VSMCs (60). Importantly,
VSMCs do not only express MIF but also migrate towards exogen-
ous MIF after 6 hours (h) of incubation (61) (
Figure 1).
MIF signalling in the vasculature has also been pursued. Subse-
quent to showing MIF-induced expression of ICAM-1 in ECs (62),
Cheng et al. reported the expression of VCAM-1, E-selectin,
ICAM-1 and CCL2 to be MIF-dependent. Indeed, TNF-induced
leukocyte rolling and adhesion to MIF-depleted human umbilical
vein endothelial cells (HUVECs) were impaired, an observation
that was attributed to a reduction of p38 MAPK activation, result-
ing in reduced expression of chemokines and adhesion molecules
(63). In addition, MIF was found to upregulate mononuclear cell
adhesion molecules such as VCAM-1 and ICAM-1 in a Src-, nu-
clear factor-κB (NF-κB), and phosphatidylinositol-3-kinase
(PI3K)-dependent manner (64). The above findings are in line
with a previous report where oxLDL- and MIF-induced monocyte
arrest on HAoEC was abrogated by anti-MIF neutralising antibody
(58). Moreover, MIF blockade leads to reduced VSMC prolifer-
ation and neointimal thickening (65), while at later stages during
the atherogenic process MIF contributes to plaque instability (58).
Thus, it is not surprising that there is extensive literature on the
functional role of vascular MIF in the pathogenesis of vascular dis-
ease like atherosclerosis or neointimal growth after vascular injury.
MIF in atherosclerosis
Inflammatory processes are key contributors to the pathogenesis
of atherosclerosis (66). The proinflammatory chemokine-like cy-
tokine MIF has been broadly implicated in atherogenesis (39)
(
Figure 1). Expression of MIF in developing atherosclerotic
plaques in humans was minimal in ECs and smooth muscle cells
(SMCs) in non-lesion-associated areas but was upregulated in
ECs, SMCs, macrophages and T cells upon atheroprogression sug-
gesting a role for MIF in plaque instability (56, 67). Interestingly,
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
Thrombosis and Haemostasis 109.3/2013 © Schattauer 2013
4Asare et al. Vascular biology of MIF
an interaction between MIF and CSN5/JAB1, a negative regulator
of NF-κB which is a key transcription factor involved in the in-
flammatory and immune processes associated with atherosclerosis
(68, 69), was revealed in human atherosclerotic lesions (56). As a
negative regulator of CSN5, MIF might synergise with JAB1 in
regulating NF-κB-driven inflammatory and immune signalling in
atherosclerosis (70).
Atherosclerosis may be preceeded by vascular injury and MIF
has been shown to regulate the biological response to the injured
tissue. In a carotid artery injury model of atherosclerosis-suscep-
tible mice, MIF was shown to potentiate neointimal thickening by
promoting the accumulation of inflammatory cells in the neointi-
ma and the proliferation of medial and intimal cells (65). Indeed,
neutralising MIF resulted in a reduction of neointimal macro-
phage content and an increase in SMCs and collagen type I content
in the neointimal lesions in a model of vascular injury-induced ac-
celerated lesion formation. This reduction in neointimal macro-
phage content was attributed to impaired monocyte recruitment as
exogenous MIF increased the number of monocytes adhering to
HAoECs, an effect that was dampened by neutralising MIF mAbs
(58). The notion that MIF is critical in the development of athero-
sclerosis was corroborated by Mif gene-inactivation in Ldlr–/–
mice. These mice showed impaired atherogenic diet-induced
lesion initiation and progression when compared with corre-
sponding wild-type mice. The ability of MIF to induce prolifer-
ation of SMCs which has previously been observed mainly by
using mAb was confirmed by this genetic study (71). In a sponta-
neous atherogenic Apoe–/– mouse model, MIF was found to be
elevated at the aortic wall and blocking aortic MIF with anti-MIF
neutralising antibody led to a reduction in intimal macrophage
content and inflammatory mediators (72). Notably, treatment with
a blocking antibody that targets MIF even resulted in athero-
sclerotic plaque regression and a more stable plaque phenotype in
diet-induced atherosclerotic Apoe–/– mice. This observation was
reasserted by the finding that MIF mediates atherogenic monocyte
and T cell recruitment in vivo by engaging its receptors CXCR2
and CXCR4, respectively (
Figure 1). Dual action of MIF
through CXCR2 and CXCR4 also may explain why anti-MIF
Figure 1: Expression and functional role of vascular MIF in angiogen-
esis and atherosclerosis. Left (angiogenesis): After tissue injury, MIF along
with other chemokines such as CXCL12, CXCL1, and VEGF is released to acti-
vate and recruit EPCs and also monocytes to the site of injury where they are
embedded into tube structures. Importantly, MIF drives tube formation and
the formation of new vessels that follows. EPCs carry angiogenic factors
(‘cargo’) themselves. Right (atherosclerosis): MIF expressed by ECs and mac-
rophages in the atherosclerotic plaque is upregulated upon stimulation with
inflammatory and atherogenic mediators such as oxLDL and thrombin. MIF
induces the expression of chemokines (CCL2) and adhesion molecules
(VCAM-1, ICAM-1) which regulates the recruitment and adhesion of mono-
cytes to the surface of the endothelium. Alternatively, MIF may employ its
chemokine receptors CXCR2 and CXCR4 (expressed on recruited monocytes
and T cells, respectively; receptors not shown) in exerting its chemokine-like
functions. After transmigration into the subendothelial space, monocytes
subsequently differentiate into macrophages. MIF and other pro-atherogenic
factors (latter not shown) stimulate these macrophages to secrete TNF-
α
,
IL-1
β
, iNOS and NO, enhancing the inflammatory milieu in the lesion. Addi-
tionally, MIF potentiates foam cell formation by accelerating macrophage
oxLDL uptake, thereby enhancing lesion formation. VSMCs express and also
migrate towards MIF and this may contribute to plaque stability, although
long term exposure to MIF has also been shown to inhibit PDGF-BB-induced
VSMCs migration and MMP upregulation which points in the direction of
plaque destabilisation by MIF (for references see text).
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
© Schattauer 2013 Thrombosis and Haemostasis 109.3/2013
5Asare et al. Vascular biology of MIF
blockade was more efficacious in the clinically highly relevant re-
gression model than treatment with anti-CXCL1 or anti-CXCL12
(42). Indeed, a MIF N-loop-derived peptide that disrupts the inter-
action between MIF and CXCR2 blocked MIF-induced leukocyte
adhesion in carotid arteries in vivo (73).
Taken together, data from several groups using either neutralis-
ing antibody or genetic deletion underpins the pathogenic role of
MIF in promoting atherogenic changes in the arterial vessel wall.
Importantly, the proatherogenic role of MIF was confirmed in epi-
demiologic studies where single nucleotide polymorphisms
(SNPs) in the human MIF gene were identified as a risk factor for
coronary heart disease, showing an association of a haplotype con-
taining the rs755622C allele, which has been reported before to in-
crease the susceptibility for various other proinflammatory condi-
tions (74), and showing that the GG genotype of the MIF SNP
rs1007888 was associated with myocardial infarction (MI) in
Czech female patients (75). Moreover, Makino et al. showed that
high plasma levels of MIF are associated with adverse long-term
outcome in patients with stable coronary artery disease and im-
paired glucose tolerance or type 2 diabetes mellitus (76). Similarly,
Müller et al. found that MIF expression is enhanced in acute cor-
onary syndromes (ACS), that it is associated with various markers
of the inflammatory response, that it correlates with the extent of
cardiac necrosis marker release after percutaneous intervention
and that it is increased in ACS patients with respective lesions (77).
Role of MIF in (neo-)angiogenesis /
vasculogenesis
MIF’s role in atherogenesis has been extensively studied (see
above); however, its roles in the processes controlling physiologic
and pathophysiologic angiogenesis are less well understood.
Angiogenesis is the growth of blood vessels from a pre-existing
vasculature. It occurs throughout life and is an important compo-
nent of different physiological and pathophysiological conditions
such as wound healing and pregnancy (78). Regulation of angio-
genesis is achieved by balancing angiogenic and angiostatic
triggers. If not properly controlled, angiogenesis can promote to
tumour growth, rheumatic arthritis, and retinopathies (78). The
therapeutic value of angiogenesis has become of great interest. In-
hibiting or decreasing angiogenesis possesses therapeutic potential
in treating cancer and rheumatic arthritis, while stimulation of an-
giogenesis can be helpful in ischaemic heart disease, peripheral ar-
terial disease, and wound healing responses by increasing reperfu-
sion of the tissue. Moreover, vasculogenesis, a process formerly
considered to be restricted to the de novo formation of vascular
structures from mesenchymal angioblasts in early embryonic vas-
cular development (79, 80), has now also been discussed to occur
postnatally, where it is triggered by endothelial progenitor cells
(EPCs1) (81-84), opening-up promising novel therapeutic avenues
as well (85, 86).
Oxygen plays a crucial role in controlling angiogenesis. Hy-
poxic conditions stimulate vessel growth by activation of ECs (i.e.
proliferation and migration). The cellular response to hypoxia is
mediated via up-regulation of hypoxia-inducible transcription fac-
tors (HIFs), most prominently HIF-1α. HIFs upregulate the tran-
scription of numerous genes, thereby affecting endothelial cell
growth, SMC recruitment, and leukocyte attraction. HIF-1α is the
best characterised inducer of the expression of vascular endothelial
growth factor (VEGF), the major endothelial growth factor in an-
giogenesis and a key trigger of angiogenesis. Hypoxia-triggered
VEGF production and the subsequent increase in oxygen supply
following newly formed vessel growth reciprocally regulate each
other to restore homeostasis following limited blood supply
(86-88).
MIF was first implicated in angiogenesis some 14 years ago,
when Chesney et al. found that MIF blockade by a neutralising
antibody reduces tumour vascularisation and tumour growth in a
murine model of B-cell lymphoma (89). Although several distinct
mechanisms have been suggested to underlie MIF’s potent pro-tu-
mourigenic capacity, a role for MIF in tumour angiogenesis was
confirmed in a model of colon adenocarcinoma formation, in
which blockade of MIF led to reduced microvessel formation (90).
Moreover, MIF expression is seen in non–small-cell lung cancer, a
tumour entity in which MIF now has been firmly established as a
crucial player (91-95). MIF expression occurs in association with
angiogenic CXC chemokines and increased vessel density (70). As
discussed above, MIF is a non-cognate ligand for CXCR2, the cog-
nate receptor for angiogenic CXCL8, and MIF also promiscuously
engages CXCR4, the cognate receptor for CXCL12. Although
CXCL12 is an ELR– CXC chemokine, both CXCR2 and CXCR4
have been found involved in numerous pro-angiogenic effects in
various models of postnatal angiogenesis, including post-is-
chaemic adaption (71–73), underscoring the notion that the che-
mokine receptors of MIF could be critical in mediating MIF-
driven pro-angiogenic responses, although direct evidence from
knockout mouse models is yet missing for this assumption (
Fig-
ure 1).
As for angiogenic factors such as VEGF, MIF expression was
also identified to be regulated by HIF-1α activity, as demonstrated
in lung tissue, ECs, hepatic stellate cells, and VSMCs (59, 96-98). A
number of studies have since confirmed MIF’s pro-angiogenic
properties and explored the underlying molecular and cellular
mechanisms. MIF mediates EC migration and tube formation in
matrigel assays and induces angiogenesis in matrigel plugs and the
cornea angiogenesis assay (
Figure 1). These effects rely on mi-
togen-activated protein kinase (MAPK) and PI3K signalling, ac-
tivities known to be triggered by MIF (42, 99-101). Accordingly,
1 Endothelial progenitor cells (EPCs) were initially considered to represent a single
entity of progenitor cells capable of supporting post-natal de novo blood vessel
formation and have been assigned a crucial role in neo-angiogenesis. However,
since their discovery in 1997 their phenotype has been refined and become more
complex. In fact, EPCs exhibit different characteristics: (i) the so-called early out-
growth EPCs (EOCs) are derived from circulating CD34-positive mononuclear
cells and additionally express CD45 and CD14; they exert enhanced adhesion
proprieties but fail to proliferate in vitro; (ii) the so-called late outgrowth EPCs
(LOCs), lack hematopoietic markers but have the ability to proliferate. Both sub-
types respond to angiogenic stimuli, express CD31 and secrete angiogenic factors
such as VEGF and angiogenic cytokines/chemokines by themselves (79-82).
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
Thrombosis and Haemostasis 109.3/2013 © Schattauer 2013
6Asare et al. Vascular biology of MIF
MIF could be detected in the tumour-associated neovasculature
and neointima following vascular injury in pro-atherogenic mouse
models (58, 89, 99).
Tissue repair after MI heavily relies on neoangiogenesis of the
infarcted area. Chemokines and their receptors play important
roles during these ‘repairprocesses. Both exacerbating and pro-
healing responses occur. For example, the genetic absence of CC
chemokine receptor Ccr1 in a corresponding mouse model re-
duces functional impairment and structural remodelling after MI
due to an abrogated early inflammatory recruitment of neutro-
phils and improved tissue healing including vessel regeneration
(102). Interestingly, the CXCL12/MIF receptor CXCR4 was re-
cently found to have a dual role in neo-angiogenesis after MI.
CXCR4 was found to play a crucial role in endogenous remodell-
ing processes after MI, contributing to inflammatory/progenitor
cell recruitment and neovascularisation, whereas its deficiency li-
mits infarct size and causes adaptation to hypoxic stress (103).
MIF has now been amply implicated as a protective factor in
MI-ischaemia/reperfusion (I/R) injury. Although the protective
mechanisms involved have been suggested to span from AMP ki-
nase activation to promotion of anti-oxidative pathways
(104-107), it is likely that MIF-driven angiogenic/vasculogenic
processes are equally important. Indeed, MIF secreted from ECs
by hypoxic stimulation has been identified to promote EPC che-
motaxis in a CXCR4- (59) and CXCR2- (D. Simons and J. Bern-
hagen, unpublished observations) dependent manner (
Figure
1). This finding was intriguing, because previously CXCL12 was
considered the main protagonist in driving EPC recruitment fol-
lowing hypoxic/ischaemic triggers (108, 109). However, these
seemingly divergent findings may be readily reconciled if one con-
siders the kinetics of chemokine production. Hypoxia-stimulated
HUVECs and human aortic endothelial cells (HAoECs) did not
secrete detectable CXCL12 levels within an early time window of 2
h, when MIF was predominantly secreted, peaking at 60 minutes
(59). This nicely coincides with the findings by Ceradini et al.
reporting that CXCL12 production from hypoxically challenged
HUVECs and ischaemic tissue in vivo occurred in a time interval
of 6-24 h and correlated subsequent EPC trafficking in vivo (109).
Interestingly, a second MIF secretion peak was observed 8 h after
the hypoxic trigger (59), indicating that within this intermediate
time window MIF and CXCL12 may jointly act to drive EPC re-
cruitment and neovascularisation (
Figure 1).
EPC recruitment and neovascularisation also represent impor-
tant mechanisms driving the healing process of acute or chronic
skin wounds. Accordingly, MIF was defined as a potential inducer
of EPC mobilisation after flap operations. In flap patients, the
number of circulating EPCs and serum levels of MIF but not
CXCL12 serum levels was increased, especially in the patient
group of free microvascular flaps. Also serum MIF and EPC levels
correlated and MIF blockade, and to a lesser extent CXCL12 in-
hibition, partially reverted the chemotactic effect of patient serum
on isolated human EPCs (110). The study also indicated that MIF-
mediated EPC mobilisation is dependent on the degree of ischae-
mia (110).
As mentioned above, the neovascularisation potential of EPCs
is in part due to the fact that these cells are carriers of numerous
potent angiogenic/vasculogenic factors (‘angiogenic cargo’).
Prominent cargo factor are VEGF and thymosin-β4, but interest-
ingly also MIF and other chemokines (111, 112). The differential
usage of these factors was recently further refined by in vivo im-
plantation experiments, indicating that the MIF/chemokine recep-
tor axis has an important role in differentiation towards an en-
dothelial and SMC phenotype (113). As murine embryonic EPCs
were previously found to induce blood vessel growth and cardio-
protection under conditions of severe acute and chronic ischaemia
in a mouse I/R model and a rat hind-limb ischaemia model, the
above study confirms the therapeutic neovascularisation potential
of EPCs in combination with selected sets of angiogenic chemo-
kines/factors, including MIF (112, 113).
Acknowledgements
We thank Elisa Liehn, Heidi Noels, David Simons, Nancy Tuch-
scheerer and other doctoral researchers of the EuCAR program as
well as numerous (inter)national collaborators and friends for
helpful discussions over the past years. Our studies on the role of
MIF and other chemokines in neovascularisation and athero-
sclerosis were supported by an IZKF Aachen grant of the Faculty
of Medicine, RWTH Aachen University to J. B. and by Deutsche
Forschungsgemeinschaft (DFG) grants BE1977/4-2/FOR 809 to J.
B. and DFG-GRK1508/1 to J. B., Y. A., and M. S.
Conflicts of interest
None declared.
References
1. Charo IF, Ransohoff RM. The many roles of chemokines and chemokine recep-
tors in inflammation. N Engl J Med 2006; 354: 610-621.
2. Laudanna C, Alon R. Right on the spot. Chemokine triggering of integrin-me-
diated arrest of rolling leukocytes. Thromb Haemost 2006; 95: 5-11.
3. Weber C, Schober A, Zernecke A. Chemokines: key regulators of mononuclear
cell recruitment in atherosclerotic vascular disease. Arterioscler Thromb Vasc
Biol 2004; 24: 1997-2008.
4. Murphy PM, Baggiolini M, Charo IF, et al. International union of pharmacol-
ogy. XXII. Nomenclature for chemokine receptors. Pharmacol Rev 2000; 52:
145-176.
5. Koenen RR, Weber C. Chemokines: established and novel targets in athero-
sclerosis. EMBO Mol Med 2011; 3: 713-725.
6. Weber C. Chemokines take centre stage in vascular biology. Thromb Haemost
2007; 97: 685-687.
7. Humbert M, Morrell NW, Archer SL, et al. Cellular and molecular pathobiology
of pulmonary arterial hypertension. J Am Coll Cardiol 2004; 43: 13S-24S.
8. Murdoch C, Finn A. Chemokine receptors and their role in vascular biology. J
Vasc Res 2000; 37: 1-7.
9. Thelen M. Dancing to the tune of chemokines. Nat Immunol 2001; 2: 129-134.
10. von Hundelshausen P, Koenen RR, Sack M, et al. Heterophilic interactions of
platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood
2005; 105: 924-930.
11. Koenen RR, von Hundelshausen P, Nesmelova IV, et al. Disrupting functional
interactions between platelet chemokines inhibits atherosclerosis in hyperlipi-
demic mice. Nat Med 2009; 15: 97-103.
12. Moore BB, Arenberg DA, Addison CL, et al. CXC chemokines mechanism of ac-
tion in regulating tumour angiogenesis. Angiogenesis 1998; 2: 123-134.
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
© Schattauer 2013 Thrombosis and Haemostasis 109.3/2013
7Asare et al. Vascular biology of MIF
13. Strieter RM, Polverini PJ, Kunkel SL, et al. The functional role of the ELR motif
in CXC chemokine-mediated angiogenesis. J Biol Chem 1995; 270:
27348-27357.
14. Belperio JA, Keane MP, Arenberg DA, et al. CXC chemokines in angiogenesis. J
Leukoc Biol 2000; 68: 1-8.
15. Arenberg DA, Kunkel SL, Polverini PJ, et al. Inhibition of interleukin-8 reduces
tumourigenesis of human non-small cell lung cancer in SCID mice. J Clin Invest
1996; 97: 2792-2802.
16. Takamori H, Oades ZG, Hoch OC, et al. Autocrine growth effect of IL-8 and
GROalpha on a human pancreatic cancer cell line, Capan-1. Pancreas 2000; 21:
52-56.
17. Yoneda J, Kuniyasu H, Crispens MA, et al. Expression of angiogenesis-related
genes and progression of human ovarian carcinomas in nude mice. J Natl
Cancer Inst 1998; 90: 447-454.
18. Simonini A, Moscucci M, Muller DW, et al. IL-8 is an angiogenic factor in
human coronary atherectomy tissue. Circulation 2000; 101: 1519-1526.
19. Peeters W, Hellings WE, de Kleijn DP, et al. Carotid atherosclerotic plaques sta-
bilize after stroke: insights into the natural process of atherosclerotic plaque sta-
bilisation. Arterioscler Thromb Vasc Biol 2009; 29: 128-133.
20. Gerszten RE, Garcia-Zepeda EA, Lim YC, et al. MCP-1 and IL-8 trigger firm ad-
hesion of monocytes to vascular endothelium under flow conditions. Nature
1999; 398: 718-723.
21. Weber KS, von Hundelshausen P, Clark-Lewis I, et al. Differential immobili-
sation and hierarchical involvement of chemokines in monocyte arrest and
transmigration on inflamed endothelium in shear flow. Eur J Immunol 1999; 29:
700-712.
22. Gu L, Okada Y, Clinton SK, et al. Absence of monocyte chemoattractant pro-
tein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient
mice. Mol Cell 1998; 2: 275-281.
23. Boring L, Gosling J, Cleary M, et al. Decreased lesion formation in CCR2-/-
mice reveals a role for chemokines in the initiation of atherosclerosis. Nature
1998; 394: 894-897.
24. Wakasugi K, Schimmel P. Two distinct cytokines released from a human ami-
noacyl-tRNA synthetase. Science 1999; 284: 147-151.
25. Wakasugi K, Slike BM, Hood J, et al. Induction of angiogenesis by a fragment of
human tyrosyl-tRNA synthetase. J Biol Chem 2002; 277: 20124-20126.
26. Yang XL, Skene RJ, McRee DE, et al. Crystal structure of a human aminoacyl-
tRNA synthetase cytokine. Proc Natl Acad Sci U S A 2002; 99: 15369-15374.
27. Howard OM, Dong HF, Yang D, et al. Histidyl-tRNA synthetase and asparagi-
nyl-tRNA synthetase, autoantigens in myositis, activate chemokine receptors on
T lymphocytes and immature dendritic cells. J Exp Med 2002; 196: 781-791.
28. Yang D, Chertov O, Bykovskaia SN, et al. Beta-defensins: linking innate and
adaptive immunity through dendritic and T cell CCR6. Science 1999; 286:
525-528.
29. Alcami A. Viral mimicry of cytokines, chemokines and their receptors. Nat Rev
Immunol 2003; 3: 36-50.
30. Noels H, Bernhagen J, Weber C. Macrophage migration inhibitory factor: a
noncanonical chemokine important in atherosclerosis. Trends Cardiovasc Med
2009; 19: 76-86.
31. Murphy PM. Viral exploitation and subversion of the immune system through
chemokine mimicry. Nat Immunol 2001; 2: 116-122.
32. Andersson U, Tracey KJ. HMGB1 is a therapeutic target for sterile inflammation
and infection. Annu Rev Immunol 2011; 29: 139-162.
33. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear
weapon in the immune arsenal. Nat Rev Immunol 2005; 5: 331-342.
34. Schiraldi M, Raucci A, Munoz LM, et al. HMGB1 promotes recruitment of in-
flammatory cells to damaged tissues by forming a complex with CXCL12 and
signalling via CXCR4. J Exp Med 2012; 209: 551-563.
35. David JR. Delayed hypersensitivity in vitro: its mediation by cell-free substances
formed by lymphoid cell-antigen interaction. Proc Natl Acad Sci USA 1966; 56:
72-77.
36. Degryse B, de Virgilio M. The nuclear protein HMGB1, a new kind of chemo-
kine? FEBS Lett 2003; 553: 11-17.
37. Calandra T, Roger T. Macrophage migration inhibitory factor: a regulator of in-
nate immunity. Nat Rev Immunol 2003; 3: 791-800.
38. Morand EF, Leech M, Bernhagen J. MIF: a new cytokine link between rheuma-
toid arthritis and atherosclerosis. Nat Rev Drug Discov 2006; 5: 399-410.
39. Noels H, Bernhagen J, Weber C. MIF in atherosclerosis. In: Bucala R, editor. The
MIF Handbook. World Scientific Publishing, Hongkong; 2012.
40. Leng L, Metz CN, Fang Y, et al. MIF signal transduction initiated by binding to
CD74. J Exp Med 2003; 197: 1467-1476.
41. Shi X, Leng L, Wang T, et al. CD44 is the signalling component of the macro-
phage migration inhibitory factor-CD74 receptor complex. Immunity 2006; 25:
595-606.
42. Bernhagen J, Krohn R, Lue H, et al. MIF is a noncognate ligand of CXC chemo-
kine receptors in inflammatory and atherogenic cell recruitment. Nat Med 2007;
13: 587-596.
43. Schwartz V, Lue H, Kraemer S, et al. A functional heteromeric MIF receptor
formed by CD74 and CXCR4. FEBS Lett 2009; 583: 2749-2757.
44. Liu Y, Shaw SK, Ma S, et al. Regulation of leukocyte transmigration: cell surface
interactions and signalling events. J Immunol 2004; 172: 7-13.
45. Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation.
Nat Rev Immunol 2007; 7: 803-815.
46. Danese S, Dejana E, Fiocchi C. Immune regulation by microvascular endothe-
lial cells: directing innate and adaptive immunity, coagulation, and inflam-
mation. J Immunol 2007; 178: 6017-6022.
47. Donnelly SC, Bucala R. Macrophage migration inhibitory factor: a regulator of
glucocorticoid activity with a critical role in inflammatory disease. Mol Med
Today 1997; 3: 502-507.
48. Lolis E, Bucala R. Therapeutic approaches to innate immunity: severe sepsis and
septic shock. Nat Rev Drug Discov 2003; 2: 635-645.
49. Mitchell RA, Bucala R. Tumour growth-promoting properties of macrophage
migration inhibitory factor. Semin Cancer Biol 2000; 10: 359-366.
50. Bernhagen J, Calandra T, Mitchell RA, et al. MIF is a pituitary-derived cytokine
that potentiates lethal endotoxaemia. Nature 1993; 365: 756-759.
51. Calandra T, Spiegel LA, Metz CN, et al. Macrophage migration inhibitory factor
is a critical mediator of the activation of immune cells by exotoxins of Gram-
positive bacteria. Proc Natl Acad Sci USA 1998; 95: 11383-11388.
52. Bozza M, Satoskar AR, Lin G, et al. Targeted disruption of migration inhibitory
factor gene reveals its critical role in sepsis. J Exp Med 1999; 189: 341-346.
53. Calandra T, Echtenacher B, Roy DL, et al. Protection from septic shock by neu-
tralisation of macrophage migration inhibitory factor. Nat Med 2000; 6:
164-170.
54. Veillat V, Carli C, Metz CN, et al. Macrophage migration inhibitory factor elicits
an angiogenic phenotype in human ectopic endometrial cells and triggers the
production of major angiogenic factors via CD44, CD74, and MAPK signalling
pathways. J Clin Endocrinol Metab 2010; 95: E403-412.
55. Baugh JA, Chitnis S, Donnelly SC, et al. A functional promoter polymorphism
in the macrophage migration inhibitory factor (MIF) gene associated with dis-
ease severity in rheumatoid arthritis. Genes Immun 2002; 3: 170-176.
56. Burger-Kentischer A, Goebel H, Seiler R, et al. Expression of macrophage mi-
gration inhibitory factor in different stages of human atherosclerosis. Circu-
lation 2002; 105: 1561-1566.
57. Shimizu T, Nishihira J, Watanabe H, et al. Macrophage migration inhibitory fac-
tor is induced by thrombin and factor Xa in endothelial cells. J Biol Chem 2004;
279: 13729-13737.
58. Schober A, Bernhagen J, Thiele M, et al. Stabilisation of atherosclerotic plaques
by blockade of macrophage migration inhibitory factor after vascular injury in
apolipoprotein E-deficient mice. Circulation 2004; 109: 380-385.
59. Simons D, Grieb G, Hristov M, et al. Hypoxia-induced endothelial secretion of
macrophage migration inhibitory factor and role in endothelial progenitor cell
recruitment. J Cell Mol Med 2011; 15: 668-678.
60. Chen L, Yang G, Zhang X, et al. Induction of MIF expression by oxidized LDL
via activation of NF-κB in vascular smooth muscle cells. Atherosclerosis 2009;
207: 428-433.
61. Schrans-Stassen BH, Lue H, Sonnemans DG, et al. Stimulation of vascular
smooth muscle cell migration by macrophage migration inhibitory factor. Anti-
oxid Redox Signal 2005; 7: 1211-1216.
62. Lin SG, Yu XY, Chen YX, et al. De novo expression of macrophage migration in-
hibitory factor in atherogenesis in rabbits. Circ Res 2000; 87: 1202-1208.
63. Cheng Q, McKeown SJ, Santos L, et al. Macrophage migration inhibitory factor
increases leukocyte-endothelial interactions in human endothelial cells via pro-
motion of expression of adhesion molecules. J Immunol 2010; 185: 1238-1247.
64. Amin MA, Haas CS, Zhu K, et al. Migration inhibitory factor up-regulates vas-
cular cell adhesion molecule-1 and intercellular adhesion molecule-1 via Src,
PI3 kinase, and NFkappaB. Blood 2006; 107: 2252-2261.
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
Thrombosis and Haemostasis 109.3/2013 © Schattauer 2013
8Asare et al. Vascular biology of MIF
65. Chen Z, Sakuma M, Zago AC, et al. Evidence for a role of macrophage mi-
gration inhibitory factor in vascular disease. Arterioscler Thromb Vasc Biol
2004; 24: 709-714.
66. Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868-874.
67. Schmeisser A, Marquetant R, Illmer T, et al. The expression of macrophage mi-
gration inhibitory factor 1alpha (MIF 1alpha) in human atherosclerotic plaques
is induced by different proatherogenic stimuli and associated with plaque insta-
bility. Atherosclerosis 2005; 178: 83-94.
68. de Winther MP, Kanters E, Kraal G, et al. Nuclear factor kappaB signalling in
atherogenesis. Arterioscler Thromb Vasc Biol 2005; 25: 904-914.
69. Schweitzer K, Bozko PM, Dubiel W, et al. CSN controls NF-kappaB by deubi-
quitinylation of IkappaBalpha. Embo J 2007; 26: 1532-1541.
70. Kleemann R, Hausser A, Geiger G, et al. Intracellular action of the cytokine MIF
to modulate AP-1 activity and the cell cycle through Jab1. Nature 2000; 408:
211-216.
71. Pan JH, Sukhova GK, Yang JT, et al. Macrophage migration inhibitory factor
deficiency impairs atherosclerosis in low-density lipoprotein receptor-deficient
mice. Circulation 2004; 109: 3149-3153.
72. Burger-Kentischer A, Gobel H, Kleemann R, et al. Reduction of the aortic in-
flammatory response in spontaneous atherosclerosis by blockade of macro-
phage migration inhibitory factor (MIF). Atherosclerosis 2006; 184: 28-38.
73. Kraemer S, Lue H, Zernecke A, et al. MIF-chemokine receptor interactions in
atherogenesis are dependent on an N-loop-based 2-site binding mechanism.
FASEB J 2011; 25: 894-906.
74. Herder C, Illig T, Baumert J, et al. Macrophage migration inhibitory factor
(MIF) and risk for coronary heart disease: results from the MONICA/KORA
Augsburg case-cohort study, 1984-2002. Atherosclerosis 2008; 200: 380-388.
75. Tereshchenko IP, Petrkova J, Mrazek F, et al. The macrophage migration in-
hibitory factor (MIF) gene polymorphism in Czech and Russian patients with
myocardial infarction. Clin Chim Acta 2009; 402: 199-202.
76. Makino A, Nakamura T, Hirano M, et al. High plasma levels of macrophage mi-
gration inhibitory factor are associated with adverse long-term outcome in pa-
tients with stable coronary artery disease and impaired glucose tolerance or type
2 diabetes mellitus. Atherosclerosis 2010; 213: 573-578.
77. Muller II, Muller KA, Schonleber H, et al. Macrophage migration inhibitory fac-
tor is enhanced in acute coronary syndromes and is associated with the inflam-
matory response. PLoS One 2012; 7: e38376.
78. Conway EM, Collen D, Carmeliet P. Molecular mechanisms of blood vessel
growth. Cardiovasc Res 2001; 49: 507-521.
79. Buschmann I, Schaper W. Arteriogenesis versus angiogenesis: two mechanisms
of vessel growth. News Physiol Sci 1999; 14: 121-125.
80. Risau W, Flamme I. Vasculogenesis. Annu Rev Cell Dev Biol 1995; 11: 73-91.
81. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor en-
dothelial cells for angiogenesis. Science 1997; 275: 964-967.
82. Ingram DA, Caplice NM, Yoder MC. Unresolved questions, changing defini-
tions, and novel paradigms for defining endothelial progenitor cells. Blood
2005; 106: 1525-1531.
83. Ingram DA, Mead LE, Tanaka H, et al. Identification of a novel hierarchy of en-
dothelial progenitor cells using human peripheral and umbilical cord blood.
Blood 2004; 104: 2752-2760.
84. Rehman J, Li J, Orschell CM, et al. Peripheral blood "endothelial progenitor
cells" are derived from monocyte/macrophages and secrete angiogenic growth
factors. Circulation 2003; 107: 1164-1169.
85. Simons M, Ware JA. Therapeutic angiogenesis in cardiovascular disease. Nat
Rev Drug Discov 2003; 2: 863-871.
86. Tuchscheerer N. The ligands of CXCR4 in vascularisation [Dissertation thesis].
Aachen: RWTH Aachen University; 2012.
87. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angio-
genesis. Nature 2011; 473: 298-307.
88. Yancopoulos GD, Davis S, Gale NW, et al. Vascular-specific growth factors and
blood vessel formation. Nature 2000; 407: 242-248.
89. Chesney J, Metz C, Bacher M, et al. An essential role for macrophage migration
inhibitory factor (MIF) in angiogenesis and the growth of a murine lymphoma.
Mol Med 1999; 5: 181-191.
90. Wilson JM, Coletta PL, Cuthbert RJ, et al. Macrophage migration inhibitory fac-
tor promotes intestinal tumourigenesis. Gastroenterology 2005; 129: 1485-1503.
91. Winner M, Meier J, Zierow S, et al. A novel, macrophage migration inhibitory
factor suicide substrate inhibits motility and growth of lung cancer cells. Cancer
Res 2008; 68: 7253-7257.
92. Rendon BE, Roger T, Teneng I, et al. Regulation of human lung adenocarcinoma
cell migration and invasion by macrophage migration inhibitory factor. J Biol
Chem 2007; 282: 29910-29918.
93. Brock SE, Rendon BE, Yaddanapudi K, et al. Negative Regulation of AMP-acti-
vated Protein Kinase (AMPK) Activity by Macrophage Migration Inhibitory
Factor (MIF) Family Members in Non-small Cell Lung Carcinomas. J Biol
Chem 2012; 287: 37917-37925.
94. Rendon BE, Willer SS, Zundel W, et al. Mechanisms of macrophage migration
inhibitory factor (MIF)-dependent tumour microenvironmental adaptation.
Exp Mol Pathol 2009; 86: 180-185.
95. Coleman AM, Rendon BE, Zhao M, et al. Cooperative regulation of non-small
cell lung carcinoma angiogenic potential by macrophage migration inhibitory
factor and its homolog, D-dopachrome tautomerase. J Immunol 2008; 181:
2330-2337.
96. Baugh JA, Gantier M, Li L, et al. Dual regulation of macrophage migration in-
hibitory factor (MIF) expression in hypoxia by CREB and HIF-1. Biochem Bio-
phys Res Commun 2006; 347: 895-903.
97. Copple BL, Bai S, Burgoon LD, et al. Hypoxia-inducible factor-1alpha regulates
the expression of genes in hypoxic hepatic stellate cells important for collagen
deposition and angiogenesis. Liver Int 2011; 31: 230-244.
98. Fu H, Luo F, Yang L, et al. Hypoxia stimulates the expression of macrophage mi-
gration inhibitory factor in human vascular smooth muscle cells via HIF-1alpha
dependent pathway. BMC Cell Biol 2010; 11: 66.
99. Amin MA, Volpert OV, Woods JM, et al. Migration inhibitory factor mediates
angiogenesis via mitogen-activated protein kinase and phosphatidylinositol ki-
nase. Circ Res 2003; 93: 321-329.
100.Lue H, Kapurniotu A, Fingerle-Rowson G, et al. Rapid and transient activation
of the ERK MAPK signalling pathway by macrophage migration inhibitory fac-
tor (MIF) and dependence on JAB1/CSN5 and Src kinase activity. Cell Signal
2006; 18: 688-703.
101.Mitchell RA, Metz CN, Peng T, et al. Sustained mitogen-activated protein kinase
(MAPK) and cytoplasmic phospholipase A2 activation by macrophage mi-
gration inhibitory factor (MIF). Regulatory role in cell proliferation and gluco-
corticoid action. J Biol Chem 1999; 274: 18100-18106.
102.Liehn EA, Merx MW, Postea O, et al. Ccr1 deficiency reduces inflammatory re-
modelling and preserves left ventricular function after myocardial infarction. J
Cell Mol Med 2008; 12: 496-506.
103.Liehn EA, Tuchscheerer N, Kanzler I, et al. Double-edged role of the
CXCL12/CXCR4 axis in experimental myocardial infarction. J Am Coll Cardiol
2011; 58: 2415-2423.
104.Koga K, Kenessey A, Powell S, et al. Macrophage migration inhibitory factor
provides cardioprotection during ischaemia/reperfusion by reducing oxidative
stress. Antioxid Redox Signal 2010; 14: 1191-1202.
105.Miller EJ, Li J, Leng L, et al. Macrophage migration inhibitory factor stimulates
AMP-activated protein kinase in the ischaemic heart. Nature 2008; 451:
578-582.
106.Qi D, Hu X, Wu X, et al. Cardiac macrophage migration inhibitory factor in-
hibits JNK pathway activation and injury during ischaemia/reperfusion. J Clin
Invest 2009; 119: 3807-3816.
107.Luedike P, Hendgen-Cotta UB, Sobierajski J, et al. Cardioprotection through
S-nitros(yl)ation of macrophage migration inhibitory factor. Circulation 2012;
125: 1880-1889.
108.Ceradini DJ, Gurtner GC. Homing to hypoxia: HIF-1 as a mediator of progeni-
tor cell recruitment to injured tissue. Trends Cardiovasc Med 2005; 15: 57-63.
109.Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is
regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med
2004; 10: 858-864.
110.Grieb G, Piatkowski A, Simons D, et al. Macrophage migration inhibitory factor
is a potential inducer of endothelial progenitor cell mobilisation after flap oper-
ation. Surgery 2012; 151: 268-277.
111.Kupatt C, Bock-Marquette I, Boekstegers P. Embryonic endothelial progenitor
cell-mediated cardioprotection requires Thymosin beta4. Trends Cardiovasc
Med 2008; 18: 205-210.
112.Kupatt C, Horstkotte J, Vlastos GA, et al. Embryonic endothelial progenitor cells
expressing a broad range of proangiogenic and remodelling factors enhance vas-
cularisation and tissue recovery in acute and chronic ischaemia. Faseb J 2005;
19: 1576-1578.
113.Kanzler I, Tuchscheerer N, Steffens G, et al. Differential roles of angiogenic che-
mokines in endothelial progenitor cell-induced angiogenesis. Basic Res Cardiol
For personal or educational use only. No other uses without permission. All rights reserved.
Note: Uncorrected proof, prepublished online
Downloaded from www.thrombosis-online.com on 2013-01-17 | ID: 1000416899 | IP: 134.130.15.1
... MIF was an evolutionarily-conserved protein that abundantly expressed in humans and non-primate mammals which consisted of 114 amino acids and had a molecular weight of 12.5 kDa. In addition to its functions as cytokine/chemokine and angiogenic factor, the expression of MIF was strongly upregulated in several disease [52][53][54]. Previous studies revealed that MIF mediates endothelial cell migration and tube formation via Akt and ERK signal pathways, and could induce angiogenesis [55,56]. ...
Article
Full-text available
Intracerebral hemorrhage (ICH) is a subtype of stroke with the highest fatality and disability rate. Up to now, commonly used first-line therapies have limited value in improving prognosis. Angiogenesis is essential to neurological recovery after ICH. Recent studies have shown that microRNA-451(miR-451) plays an important role in angiogenesis by regulating the function of vascular endothelial cells. We found miR-451 was significantly decreased in the peripheral blood of ICH patients in the acute stage. Based on the clinical findings, we conducted this study to investigate the potential regulatory effect of miR-451 on angiogenesis after ICH. The expression of miR-451 in ICH mouse model and in a hemin toxicity model of human brain microvascular endothelial cells (hBMECs) was decreased the same as in ICH patients. MiR-451 negatively regulated the proliferation, migration, and tube formation of hBMECs in vitro. MiR-451 negatively regulated the microvessel density in the perihematoma tissue and affected neural functional recovery of ICH mouse model. Knockdown of miR-451 could recovered tight junction and protect the integrity of blood-brain barrier after ICH. Based on bioinformatic programs, macrophage migration inhibitory factor (MIF) was predicted to be the target gene and identified to be regulated by miR-451 inhibiting the protein translation. And p-AKT and p-ERK were verified to be downstream of MIF in angiogenesis. These results all suggest that miR-451 will be a potential target for regulating angiogenesis in ICH.
... 41 In contrast, upregulated VCAM and MIF signaling networks within APVSLevel-High crosstalk further promoted atherogenic leukocyte recruitment and lesional inflammation, exacerbating plaque instability. 42,43 Notably, consistent with our observation of intensified SPP1 signaling network in APVSLevel-Low crosstalk, previous reports have suggested that the plaque macrophage-mediated paracrine signaling by SPP1 enhanced fibroblast collagen secretion, contributing to the formation of the desmoplastic region. 44,45 Moreover, a group of dysregulated LRs between macrophages and myofibroblasts was identified as therapeutic targets, including IL34/CTSK, IL6/SOCS3, AGT/SPP1, APOE/SPP1, CXCL2/CDKN1A, and ADM/CCL2, which were demonstrated to promote acute inflammation response and ECM degradation in the AC. ...
Article
Full-text available
Acute myocardial infarction dominates coronary artery disease mortality. Identifying bio-signatures for plaque destabilization and rupture is important for preventing the transition from coronary stability to instability and the occurrence of thrombosis events. This computational systems biology study enrolled 2235 samples from 22 independent bulks cohorts and 14 samples from two single-cell cohorts. A machine-learning integrative program containing nine learners was developed to generate a warning classifier linked to atherosclerotic plaque vulnerability signature (APVS). The classifier displays the reliable performance and robustness for distinguishing ST-elevation myocardial infarction from chronic coronary syndrome at presentation, and revealed higher accuracy to 33 pathogenic biomarkers. We also developed an APVS-based quantification system (APVSLevel) for comprehensively quantifying atherosclerotic plaque vulnerability, empowering early-warning capabilities and accurate assessment of atherosclerosis severity. It unraveled the multidimensional dysregulated mechanisms at high resolution. This study provides a potential tool for macro-level differential diagnosis and evaluation of subtle genetic pathological changes in atherosclerosis.
... Previous studies have found that MIF could affect the development of CSVD by affecting a variety of pathophysiological processes [35]. Arteriosclerosis is the main cause of chronic hypoxia hypoperfusion, while MIF is a key mediator of arteriosclerosis [36], participating in the entire process of arteriosclerosis by promoting leukocyte recruitment and damaging inflammation [37,38]. Previous studies have found that MIF is involved in the preclinical atherosclerosis process based on low-grade inflammation [39], and is associated with hypoendothelial function and increased vascular stiffness [40], while arteriosclerosis may be a common pathogenesis of CSVD [41]. ...
Article
Full-text available
(1) Objective: To investigate the relationship between serum macrophage migration inhibitory factor (MIF) level and white matter hyperintensity (WMH) and executive function (EF) in cerebral small vascular disease (CSVD), and assess the impact and predictive value of MIF level and Fazekas scores in CSVD-related cognitive impairment (CI) (CSVD-CI); (2) Methods: A total of 117 patients with WMH admitted to the First Affiliated Hospital of Xinxiang Medical College from January 2022 to August 2022 were enrolled. According to the Montreal cognitive assessment (MoCA) scale, subjects were divided into a normal cognitive group and an impaired group. All subjects required serum MIF level, 3.0 T MRI, and neuropsychological evaluation to investigate the risk factors for CDVD-CI, analyze the correlation between MIF level, WMH, and EF, and to analyze the diagnostic value of MIF and WMH degree in predicting CSVD-CI; (3) Results: 1. Fazekas score and MIF level were the risk factors of CSVD-CI. 2. The Fazekas score was negatively correlated with MoCA score, positively correlated with Stroop C-Time, Stroop C-Mistake, Stroop interference effects (SIE)-Time, SIE-Mistake, and color trails test (CTT) interference effects (CIE) (B-A). 3. The MIF level was positively correlated with Fazekas score, Stroop C-Time, SIE-Time, CTT B-Time, and CIE (B-A), and negatively correlated with MoCA score. 4. Fazekas score and MIF level were significant factors for diagnosing CSVD-CI; (4) Conclusion: The Fazekas score and MIF level may be the risk factors of CSVD-CI, and they are closely correlated to CI, especially the EF, and they have diagnostic value for CSVD-CI.
... Further studies have revealed its diverse functions in different pathological reactions, including inflammatory processes (Asare et al., 2013), leukocyte recruitment (Ives et al., 2021), bone metabolism (Movila et al., 2016), angiogenesis (Liao et al., 2010), cell proliferation (Utispan & Koontongkaew, 2021), apoptosis (W. Liu et al., 2017), and autophagy (R. Li et al., 2021). ...
Article
Objective: Periodontitis is an inflammatory disease that involves an imbalance in the oral microbiota, activation of inflammatory and immune responses, and alveolar bone destruction. Macrophage migration inhibitory factor (MIF) is a versatile cytokine involved in several pathological reactions, including inflammatory processes and bone destruction, both of which are characteristics of periodontitis. While the roles of MIF in cancer and other immune diseases have been extensively characterized, its role in periodontitis remains inconclusive. Results: In this review, we describe a comprehensive analysis of the potential roles of MIF in periodontitis from the perspective of immune response and bone regulation at the cellular and molecular levels. Moreover, we discuss its potential reliability as a novel diagnostic and therapeutic target for periodontitis. Conclusion: This review can aid dental researchers and clinicians in understanding the current state of MIF-related pathogenesis, diagnosis, and treatment of periodontitis.
... Several studies have suggested that inflammation may contribute to atherogenesis. In addition to systemic inflammation caused by uremia, repeated cannulations in AVF result in a sustained local inflammatory response characterized by T cell and macrophage infiltrations and increased proinflammatory cytokines [28][29][30]. The importance of inflammation in AVF failure is further demonstrated in a study that showed significant decreases in intimal hyperplasia in MCP-1 knockout mouse models [31]. ...
Article
Full-text available
Systemic inflammation has been proposed as a relevant factor of vascular remodeling and dysfunction. We aimed to identify circulating inflammatory biomarkers that could predict future arteriovenous fistula (AVF) dysfunction in patients undergoing hemodialysis. A total of 282 hemodialysis patients were enrolled in this prospective multicenter cohort study. Plasma cytokine levels were measured at the time of data collection. The primary outcome was the occurrence of AVF stenosis and/or thrombosis requiring percutaneous transluminal angioplasty or surgery within the first year of enrollment. AVF dysfunction occurred in 38 (13.5%) patients during the study period. Plasma interleukin-6 (IL-6) levels were significantly higher in patients with AVF dysfunction than those without. Diabetes mellitus, low systolic blood pressure, and statin use were also associated with AVF dysfunction. The cumulative event rate of AVF dysfunction was the highest in IL-6 tertile 3 (p = 0.05), and patients in tertile 3 were independently associated with an increased risk of AVF dysfunction after multivariable adjustments (adjusted hazard ratio = 3.06, p = 0.015). In conclusion, circulating IL-6 levels are positively associated with the occurrence of incident AVF dysfunction in hemodialysis patients. Our data suggest that IL-6 may help clinicians identify those at high risk of impending AVF failure.
... Inflammatory mediators including macrophages, infiltrating lymphocytes, macrophage migration inhibitory factor, chemokine (C-X-C motif) receptor (CXCR)-2, CXCR-4, interleukin (IL)-8, and monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor (TNF)-α, IL-1β and growth factors such as vascular endothelial growth factor (VEGF)-A play a critical role in AVF failure [10]. These mediators also play a crucial role in regulating neointimal thickening, the proliferation of medial and intimal cells, and plaque formation [10,12]. The venous NIH and failure of outward remodeling are attributed to inflammation, proliferation, migration, and phenotypic changes of vascular smooth muscle cells (VSMCs), and extracellular remodeling due to increased matrix metalloproteinases (MMPs) [13][14][15][16]. ...
Article
Full-text available
Arteriovenous fistula (AVF) is the preferred vascular access in hemodialysis patients; however, it is afflicted with a high failure rate. Chronic inflammation, excessive neointimal hyperplasia (NIH), vessel stenosis, early thrombosis, and failure of outward remodeling are the major causes of AVF maturation failure. Inflammatory mediator toll-like receptor (TLR)-4 plays a critical role in NIH, arterial thrombosis, and stenosis. We investigated the effect of TLR-4 inhibition on early thrombosis. Yucatan miniswine were used to create AVF involving femoral artery and femoral vein and treated with TLR-4 inhibitor TAK-242 with ethanol as the vehicle. The vessels were assessed after 12 weeks using histomorphometry, immunostaining, ultrasound, angiography, and optical coherence tomography. Inhibition of TLR-4 attenuated inflammation and early thrombosis in 50% of animals, and blood flow was present through AVF in 25% of animals. Thus, targeting TLR-4 to attenuate inflammation and early thrombosis might be a therapeutic approach to keep AVF patent and maintain blood flow through the outflow vein.
Article
Full-text available
Background The conditioned medium from human dermal fibroblasts (dermal fibroblast‐conditioned medium; DFCM) contains a diverse array of secretory proteins, including growth factors and wound repair‐promoting proteins. Angiogenesis, a crucial process that facilitates the infiltration of inflammatory cells during wound repair, is induced by a hypoxic environment and inflammatory cytokines. Methods In this study, we conducted a comprehensive bioinformatic analysis of 337 proteins identified through proteomics analysis of DFCM. We specifically focused on 64 DFCM proteins with potential involvement in angiogenesis. These proteins were further classified based on their characteristics, and we conducted a detailed analysis of their protein–protein interactions. Results Gene Ontology protein classification categorized these 64 DFCM proteins into various classes, including metabolite interconversion enzymes (N = 11), protein modifying enzymes (N = 10), protein‐binding activity modulators (N = 9), cell adhesion molecules (N = 6), extracellular matrix proteins (N = 6), transfer/carrier proteins (N = 3), calcium‐binding proteins (N = 2), chaperones (N = 2), cytoskeletal proteins (N = 2), RNA metabolism proteins (N = 1), intercellular signal molecules (N = 1), transporters (N = 1), scaffold/adaptor proteins (N = 1), and unclassified proteins (N = 9). Furthermore, our protein–protein interaction network analysis of DFCM proteins revealed two distinct networks: one with medium confidence level interaction scores, consisting of 60 proteins with significant connections, and another at a high confidence level, comprising 52 proteins with significant interactions. Conclusions Our bioinformatic analysis highlights the presence of a multitude of secretory proteins in DFCM that form significant protein–protein interaction networks crucial for regulating angiogenesis. These findings underscore the critical roles played by DFCM proteins in various stages of angiogenesis during the wound repair process.
Article
Full-text available
Atherosclerosis is a chronic inflammatory condition of our arteries and the main underlying pathology of myocardial infarction and stroke. The pathogenesis is age‐dependent, but the links between disease progression, age, and atherogenic cytokines and chemokines are incompletely understood. Here, we studied the chemokine‐like inflammatory cytokine macrophage migration inhibitory factor (MIF) in atherogenic Apoe−/− mice across different stages of aging and cholesterol‐rich high‐fat diet (HFD). MIF promotes atherosclerosis by mediating leukocyte recruitment, lesional inflammation, and suppressing atheroprotective B cells. However, links between MIF and advanced atherosclerosis across aging have not been systematically explored. We compared effects of global Mif‐gene deficiency in 30‐, 42‐, and 48‐week‐old Apoe−/− mice on HFD for 24, 36, or 42 weeks, respectively, and in 52‐week‐old mice on a 6‐week HFD. Mif‐deficient mice exhibited reduced atherosclerotic lesions in the 30/24‐ and 42/36‐week‐old groups, but atheroprotection, which in the applied Apoe−/− model was limited to lesions in the brachiocephalic artery and abdominal aorta, was not detected in the 48/42‐ and 52/6‐week‐old groups. This suggested that atheroprotection afforded by global Mif‐gene deletion differs across aging stages and atherogenic diet duration. To characterize this phenotype and study the underlying mechanisms, we determined immune cells in the periphery and vascular lesions, obtained a multiplex cytokine/chemokine profile, and compared the transcriptome between the age‐related phenotypes. We found that Mif deficiency promotes lesional macrophage and T‐cell counts in younger but not aged mice, with subgroup analysis pointing toward a role for Trem2⁺ macrophages. The transcriptomic analysis identified pronounced MIF‐ and aging‐dependent changes in pathways predominantly related to lipid synthesis and metabolism, lipid storage, and brown fat cell differentiation, as well as immunity, and atherosclerosis‐relevant enriched genes such as Plin1, Ldlr, Cpne7, or Il34, hinting toward effects on lesional lipids, foamy macrophages, and immune cells. Moreover, Mif‐deficient aged mice exhibited a distinct plasma cytokine/chemokine signature consistent with the notion that mediators known to drive inflamm'aging are either not downregulated or even upregulated in Mif‐deficient aged mice compared with the corresponding younger ones. Lastly, Mif deficiency favored formation of lymphocyte‐rich peri‐adventitial leukocyte clusters. While the causative contributions of these mechanistic pillars and their interplay will be subject to future scrutiny, our study suggests that atheroprotection due to global Mif‐gene deficiency in atherogenic Apoe−/− mice is reduced upon advanced aging and identifies previously unrecognized cellular and molecular targets that could explain this phenotype shift. These observations enhance our understanding of inflamm'aging and MIF pathways in atherosclerosis and may have implications for translational MIF‐directed strategies.
Article
Full-text available
Cerebral small vascular disease (CSVD) is a common type of cerebrovascular disease, and an important cause of vascular cognitive impairment (VCI) and stroke. The disease burden is expected to increase further as a result of population aging, an ongoing high prevalence of risk factors (e.g., hypertension), and inadequate management. Due to the poor understanding of pathophysiology in CSVD, there is no effective preventive or therapeutic approach for CSVD. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that is related to the occurrence and development of vascular dysfunction diseases. Therefore, MIF may contribute to the pathogenesis of CSVD and VCI. Here, reviewed MIF participation in chronic cerebral ischemia-hypoperfusion and neurodegeneration pathology, including new evidence for CSVD, and its potential role in protection against VCI.
Article
Full-text available
Background— Endothelial progenitor cells (EPCs) have been isolated from peripheral blood and can enhance angiogenesis after infusion into host animals. It is not known whether the proangiogenic effects are a result of such events as endothelial differentiation and subsequent proliferation of EPCs or secondary to secretion of angiogenic growth factors. Methods and Results— Human EPCs were isolated as previously described, and their phenotypes were confirmed by uptake of acetylated LDL and binding of ulex-lectin. EPC proliferation and surface marker expression were analyzed by flow cytometry, and conditioned medium was assayed for growth factors. The majority of EPCs expressed monocyte/macrophage markers such as CD14 (95.7±0.3%), Mac-1 (57.6±13.5%), and CD11c (90.8±4.9%). A much lower percentage of cells expressed the specific endothelial marker VE-cadherin (5.2±0.7%) or stem/progenitor-cell markers AC133 (0.16±0.05%) and c-kit (1.3±0.7%). Compared with circulating monocytes, cultured EPCs showed upregulation of monocyte activation and macrophage differentiation markers. EPCs did not demonstrate any significant proliferation but did secrete the angiogenic growth factors vascular endothelial growth factor, hepatocyte growth factor, granulocyte colony–stimulating factor, and granulocyte-macrophage colony–stimulating factor. Conclusions— Our findings suggest that acetylated LDL(+)ulex-lectin(+) cells, commonly referred to as EPCs, do not proliferate but release potent proangiogenic growth factors. The majority of acetylated LDL(+)ulex-lectin(+) cells are derived from monocyte/macrophages. The findings of low proliferation and endothelial differentiation suggest that their angiogenic effects are most likely mediated by growth factor secretion. These findings may allow for development of novel angiogenic therapies relying on secreted growth factors or on recruitment of endogenous monocytes/macrophages to sites of ischemia.
Article
Full-text available
The CXC chemokines have recently been identified as a family of molecules which can regulate angiogenesis. Members of this family which contain the amino acid motif Glu–Leu–Arg in their amino terminus (ELR^+) act as angiogenic factors, while ELR^-members act as angiostatic molecules. The balance of these angiogenic versus angiostatic factors is critical in regulating homeostasis. As we detail in this review, there is increasing evidence from a variety of tumor model systems to suggest that the angiogenic members of this family and their receptors may be playing an important role in the neovascular pathology of solid tumors. In contrast, the angiostatic effects of the ELR- family members may provide novel therapeutic strategies for treating many tumors.
Article
The basic molecular mechanisms governing how endothelial cells, periendothelial cells and matrix molecules interact with each other and with numerous growth factors and receptors, to form blood vessels have been presented. The many insights gained from this basic knowledge are being extended to further understand pathological angiogenesis associated with disorders such as arterial stenosis, myocardial ischemia, atherosclerosis, allograft transplant stenosis. wound healing and tissue repair. As a result, novel angiogenic and anti-angiogenic molecules are rapid-ly entering the clinic, with the promise of relief from a host of medical disorders.
Article
Atherosclerosis, formerly considered a bland lipid storage disease, actually involves an ongoing inflammatory response. Recent advances in basic science have established a fundamental role for inflammation in mediating all stages of this disease from initiation through progression and, ultimately, the thrombotic complications of atherosclerosis. These new findings provide important links between risk factors and the mechanisms of atherogenesis. Clinical studies have shown that this emerging biology of inflammation in atherosclerosis applies directly to human patients. Elevation in markers of inflammation predicts outcomes of patients with acute coronary syndromes, independently of myocardial damage. In addition, low-grade chronic inflammation, as indicated by levels of the inflammatory marker C-reactive protein, prospectively defines risk of atherosclerotic complications, thus adding to prognostic information provided by traditional risk factors. Moreover, certain treatments that reduce coronary risk also limit inflammation. In the case of lipid lowering with statins, this anti-inflammatory effect does not appear to correlate with reduction in low-density lipoprotein levels. These new insights into inflammation in atherosclerosis not only increase our understanding of this disease, but also have practical clinical applications in risk stratification and targeting of therapy for this scourge of growing worldwide importance.