Apart from their canonical role in thrombosis and hemostasis, platelets amplify acute and chronic inflammation including atherosclerosis, and regulate the development and remodeling of blood and lymph vessels in health and disease. In this issue of Blood, Lievens and colleagues show evidence that activated platelets expressing CD40L promote atherogenesis and inflammation at least in part by mitigating the natural response to recruit immunomodulatory Tregs.1 

Mounting evidence indicates a pivotal role for regulatory T cells (Tregs; generally identified by positive expression of CD4, CD25, and FoxP3) in balancing inflammation and autoimmune disease via actively suppressing pathogenic immune activation (Th1, Th2, Th17, CTL) and maintaining immune tolerance.2  Although present approximately 1% to 2% in whole blood, they are actively recruited to inflammatory sites via various chemokines including CCL2 and CCL5.2  In addition, in atherosclerosis (ie, a chronic inflammatory disease of the vessel wall caused by progressive accumulation of lipids), various groups have shown an inverse relationship between the presence of Tregs versus atherosclerotic plaque growth, and even the induction of a “stable plaque” phenotype characterized by reduced inflammatory cell infiltration and increased plaque fibrosis.3,4  The latter is clinically important, as such stable plaques would prevent plaque rupture thereby reducing the associated life-threatening risk of thrombo-occlusive/ embolic stroke and myocardial infarction.5 

CD40 ligand (CD40L; CD154) is a transmembrane glycoprotein and member of the tumor necrosis factor (TNF) family. While existing in a membrane-bound and soluble form, CD40L can bind upon trimerization to its classical receptor CD40 leading—via TNF receptor–associated factor (TRAF) adaptor proteins—to intracellular signaling and transcriptional activation, but CD40L also signals by binding noncanonical receptors including integrins (reviewed in Seijkens et al6 ). Primarily known for its role as a costimulator of T-cell activation, it became clear that CD40L is also functionally expressed on several other hematopoietic and nonhematopoietic cell types including platelets and endothelial cells.6  In atherosclerosis, various groups showed that inhibition or genetic deletion of CD40L in mice reduced the growth of newly formed as well as established plaques, and promoted the acquisition of a stable plaque phenotype.7,8  Given the broad expression of CD40L on several cell types relevant in atherosclerosis, it seems important to identify the proatherogenic role of CD40L in a cell-type–specific manner.

Here, Lievens and colleagues demonstrate, in 3 laborious mouse models of atherosclerosis (early/advanced/established lesions), that repeated transfer of thrombin-activated platelets doubles plaque size in all phases, and this phenomenon is strictly dependent on CD40L expression on platelets (evidenced by using CD40L−/− platelets) in combination with the presence of natural Tregs (evidenced by using anti-CD25–depleting antibodies).1  Thus, the absence of platelet CD40L increases the number of circulating Tregs, thereby blocking plaque growth induced by activated platelets (see figure). Surprisingly, the authors found that activated CD40L+ platelets increased—not reduced—the plasma levels and endothelial deposition of CCL2, while CCL5 expression was not regulated by platelet CD40L.1  Therefore, the inverse relationship between platelet CD40L versus Tregs does not appear to be explained by unfavorable chemokine gradients. In addition, Treg function seemed unaltered as corroborated by in vitro assays and normal IL-10 plasma levels.1 

Potential effects of activated CD40L+ platelets in promoting atherosclerotic plaque growth. The endothelial cell surface can dynamically shift between pro- versus anti-inflammatory states. Under the influence of plaque-specific chemokine gradients (eg, CCL2), activated CD40L+ platelets tip the balance toward persistent proinflammation via a “pro-inflammatory cell coating” at the luminal side of the intimal endothelial cells, by increasing (1) endothelial translocation of circulating myeloid cells without augmenting their activation; (2) formation of circulating aggregates (platelet-leukocyte aggregates [PLAs]) between platelets and myeloid cells (left side of panel); and (3) their own endothelial translocation (right side of panel). Concomitantly, activated CD40L+ platelets reduce the circulating numbers of lymphoid immunosuppressive cells (right side of panel) via humoral, subcellular, and/or heterotypic cell-cell interactions. Unidentified potential underlying mechanisms might include CD40 activation and/or TRAF2/3/5 signaling, resulting in impaired Treg formation via suppression of FoxP3/CD25 signaling and accelerated Treg apoptosis and/or clearance via CD40L-induced overstimulation. In addition, activated CD40L+ platelets could cross-activate the endothelium (center of the panel) and affect plaque infiltration of inflammatory cells and smooth muscle cells (SMCs), but these effects might be less pronounced. This proinflammatory balance, caused by activated CD40L+ platelets, might progressively promote further plaque growth by enhanced neovascularization and capillary rarefaction, likely via CD40 signaling. After plaque rupture, activated CD40L+ platelets stimulate collagen/VWF-dependent thrombus formation and stability, likely via integrin-dependent and CD40-independent pathways. (Professional illustration by Kenneth X. Probst.)

Potential effects of activated CD40L+ platelets in promoting atherosclerotic plaque growth. The endothelial cell surface can dynamically shift between pro- versus anti-inflammatory states. Under the influence of plaque-specific chemokine gradients (eg, CCL2), activated CD40L+ platelets tip the balance toward persistent proinflammation via a “pro-inflammatory cell coating” at the luminal side of the intimal endothelial cells, by increasing (1) endothelial translocation of circulating myeloid cells without augmenting their activation; (2) formation of circulating aggregates (platelet-leukocyte aggregates [PLAs]) between platelets and myeloid cells (left side of panel); and (3) their own endothelial translocation (right side of panel). Concomitantly, activated CD40L+ platelets reduce the circulating numbers of lymphoid immunosuppressive cells (right side of panel) via humoral, subcellular, and/or heterotypic cell-cell interactions. Unidentified potential underlying mechanisms might include CD40 activation and/or TRAF2/3/5 signaling, resulting in impaired Treg formation via suppression of FoxP3/CD25 signaling and accelerated Treg apoptosis and/or clearance via CD40L-induced overstimulation. In addition, activated CD40L+ platelets could cross-activate the endothelium (center of the panel) and affect plaque infiltration of inflammatory cells and smooth muscle cells (SMCs), but these effects might be less pronounced. This proinflammatory balance, caused by activated CD40L+ platelets, might progressively promote further plaque growth by enhanced neovascularization and capillary rarefaction, likely via CD40 signaling. After plaque rupture, activated CD40L+ platelets stimulate collagen/VWF-dependent thrombus formation and stability, likely via integrin-dependent and CD40-independent pathways. (Professional illustration by Kenneth X. Probst.)

Close modal

Interestingly, Lutgens et al recently reported that the absence of leukocytic TRAF2/3/5 signaling downstream of CD40 also increased Treg counts; yet, atherogenesis was unaffected (but no activated platelets were administered).9  These findings together suggest that interactions between activated CD40L+ platelets and CD40+ Tregs might result in TRAF2/3/5 signaling and suppress Treg recruitment. Hence, apart from distinct tissue-derived chemokine gradients,2  Treg trafficking into inflamed tissues might be regulated by activated platelets. It would perhaps be interesting to study whether (1) this interaction occurs after solubilization via rapid proteolytic shedding from activated platelets (eg, by ADAM10) or via CD40L+ microparticles10 ; (2) membrane-bound CD40L initiates heterotypic cell-cell interactions with CD40+ Tregs; and (3) CD40L-CD40 interaction results in reduced Treg formation via suppressing FoxP3 and/or CD25 signaling, or increased Treg apoptosis and/or clearance via overstimulation (see figure).

Consistent with previous observations,11  the authors show that, overall, activated CD40L+ platelets only modestly affect plaque composition, with small and inconsistent differences in macrophage infiltration and fibrosis.1  Hence, platelet CD40L does not seem to primarily promote plaque instability, as suggested by CD40L inhibition/deletion studies.7,8  Notably, activated CD40L+ platelets also did not alter the cytokine release and phagocytic activity of macrophages,1  and a recent report indicated that platelet CD40L was not required to cross-activate (noninflammatory) endothelium in vivo.12  Instead, the current report shows that platelet CD40L promoted the endothelial translocation of themselves, and of leukocytes in vivo—consistent with the increased CCL2 expression and endothelial deposition (see above).1  In addition, activated CD40L+ platelets formed more (proinflammatory) aggregates with each other and with (CD40+) leukocytes.1  Together, these results further underscore the premise that a complex union (a sort of thrombotic “ménage-à-trois”) among platelets, endothelial cells, and leukocytes strongly catalyzes acute and chronic inflammation.13  In fact, when considering that activated CD40L+ platelets strongly promote intraluminal interactions among platelets, endothelial cells, and leukocytes—including suppressing the circulating number of Tregs, whereas they, at best, modestly promote the intraplaque infiltration of inflammatory (and vascular smooth muscle) cells—one could speculate the following (see figure): analogous to the tightly regulated pro- versus antithrombotic balance, the endothelial cell surface can dynamically shift between pro- versus anti-inflammatory states. On atherosclerotic plaques, activated CD40L+ platelets might form some kind of “proinflammatory cell coating” at the luminal side of the intimal endothelial cells which consists of activated leukocytes, platelets, and platelet-leukocyte aggregates. This plaque-specific cell coating tips the balance (via multiple cellular and—as yet incompletely defined—humoral interactions) in a positive amplification loop toward persistent proinflammation, while it concomitantly shields the growing plaque from potentially plaque-growth–inhibiting effects such as the presence of Tregs (ie, reducing their circulating number, via unidentified mechanisms). In a second phase, this proinflammatory balance might further promote plaque growth by increased neovascularization10  and capillary rarefaction (as evidenced in the current study by the aberrant erythrocyte/iron deposits1 ).

Finally, this report1  does not provide evidence that blocking CD40L+-activated platelets might prevent atherosclerosis, but it does reinforce earlier clinical findings that antiplatelet drugs including aspirin (typically prescribed to cardiovascular patients) might, apart from preventing thrombotic complications after plaque rupture, also suppress plaque growth.14  Although the authors as well as previous studies already highlighted that blocking platelet CD40L might reduce thrombus formation and stability,1,15  the current report identifies—for the first time—that blocking platelet CD40L might preserve the natural Treg response, as a potential novel mechanism to explain the putative plaque-growth–suppressing effects of antiplatelet drugs.

Conflict-of-interest disclosure: The authors declare no competing financial interests. ■

R.M. is a postdoctoral fellow of IWT.

1
Lievens
 
D
Zernecke
 
A
Seijkens
 
T
, et al. 
Platelet CD40L mediates thrombotic and inflammatory processes in atherosclerosis.
Blood
2010
, vol. 
116
 
20
(pg. 
4317
-
4327
)
2
Wei
 
S
Kryczek
 
I
Zou
 
W
Regulatory T-cell compartmentalization and trafficking.
Blood
2006
, vol. 
108
 
2
(pg. 
426
-
431
)
3
Ait-Oufella
 
H
Salomon
 
BL
Potteaux
 
S
, et al. 
Natural regulatory T cells control the development of atherosclerosis in mice.
Nat Med
2006
, vol. 
12
 
2
(pg. 
178
-
180
)
4
Mor
 
A
Planer
 
D
Luboshits
 
G
, et al. 
Role of naturally occurring CD4+ CD25+ regulatory T cells in experimental atherosclerosis.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
4
(pg. 
893
-
900
)
5
Moura
 
R
Tjwa
 
M
Vandervoort
 
P
Van Kerckhoven
 
S
Holvoet
 
P
Hoylaerts
 
MF
Thrombospondin-1 deficiency accelerates atherosclerotic plaque maturation in ApoE−/− mice.
Circ Res
2008
, vol. 
103
 
10
(pg. 
1181
-
1189
)
6
Seijkens
 
T
Engel
 
D
Tjwa
 
M
Lutgens
 
E
The role of CD154 in haematopoietic development.
Thromb Haemost
2010
, vol. 
104
 
4
7
Lutgens
 
E
Gorelik
 
L
Daemen
 
MJ
, et al. 
Requirement for CD154 in the progression of atherosclerosis.
Nat Med
1999
, vol. 
5
 
11
(pg. 
1313
-
1316
)
8
Mach
 
F
Schonbeck
 
U
Sukhova
 
GK
Atkinson
 
E
Libby
 
P
Reduction of atherosclerosis in mice by inhibition of CD40 signalling.
Nature
1998
, vol. 
394
 
6689
(pg. 
200
-
203
)
9
Lutgens
 
E
Lievens
 
D
Beckers
 
L
, et al. 
Deficient CD40-TRAF6 signaling in leukocytes prevents atherosclerosis by skewing the immune response toward an antiinflammatory profile.
J Exp Med
, vol. 
207
 
2
(pg. 
391
-
404
)
10
Leroyer
 
AS
Rautou
 
PE
Silvestre
 
JS
, et al. 
CD40 ligand+ microparticles from human atherosclerotic plaques stimulate endothelial proliferation and angiogenesis a potential mechanism for intraplaque neovascularization.
J Am Coll Cardiol
2008
, vol. 
52
 
16
(pg. 
1302
-
1311
)
11
Huo
 
Y
Schober
 
A
Forlow
 
SB
, et al. 
Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E.
Nat Med
2003
, vol. 
9
 
1
(pg. 
61
-
67
)
12
Dole
 
VS
Bergmeier
 
W
Mitchell
 
HA
Eichenberger
 
SC
Wagner
 
DD
Activated platelets induce Weibel-Palade-body secretion and leukocyte rolling in vivo: role of P-selectin.
Blood
2005
, vol. 
106
 
7
(pg. 
2334
-
2339
)
13
Tjwa
 
M
Bellido-Martin
 
L
Lin
 
Y
, et al. 
Gas6 promotes inflammation by enhancing interactions between endothelial cells, platelets, and leukocytes.
Blood
2008
, vol. 
111
 
8
(pg. 
4096
-
4105
)
14
Ranke
 
C
Hecker
 
H
Creutzig
 
A
Alexander
 
K
Dose-dependent effect of aspirin on carotid atherosclerosis.
Circulation
1993
, vol. 
87
 
6
(pg. 
1873
-
1879
)
15
Andre
 
P
Prasad
 
KS
Denis
 
CV
, et al. 
CD40L stabilizes arterial thrombi by a beta3 integrin–dependent mechanism.
Nat Med
2002
, vol. 
8
 
3
(pg. 
247
-
252
)
Sign in via your Institution