In this issue of Blood, Kaul et al1 report that stimulation by platelet-activating factor (PAF) of artificially perfused rat mesocecum ex vivo promotes the adhesion of human sickle red blood cells (SS RBC) to postcapillary endothelium. Notably, this adhesive interaction was blocked by two different monoclonal antibodies (MoAbs) to endothelial αVβ3 integrin receptor, with resulting improvement in microvascular hemodynamics. This study is an elegant and important contribution to our understanding of SS RBC interactions with the vessel wall and, hence, the mechanisms of vasoocclusion. Vasoocclusion of small and sometimes large vessels is the hallmark of sickle cell disease, accounting for much of its morbidity and mortality. The pathophysiology of the vasoocclusive episodes is complex, involving not only the polymerization of the mutant hemoglobin, but also interactions between SS RBC, endothelium, platelets, leukocytes, and plasma constituents. Intracapillary sickling and vasoocclusion occur when transit time through capillaries is longer than the lag time for deoxygenation-induced polymerization of sickle hemoglobin. Thus, processes that delay passage of SS RBC through the microvasculature may participate in the initiation and propagation of vasoocclusion. In particular, an increase in SS RBC adhesion to postcapillary endothelium could initiate vasoocclusion by impairing flow, thereby delaying transit time of less deformable SS RBC and propagating intracapillary sickling.2 Factors such as inflammatory mediators that activate endothelial cells3 and enhance endothelial adhesivity for SS RBC might, therefore, promote vasoocclusion. Conversely, anti-adhesive or anti-inflammatory therapies might attenuate vasoocclusion.

Seminal studies by Hebbel et al4 and Hoover et al5 two decades ago first demonstrated that SS RBC showed increased adherence to endothelial cells in vitro. Moreover, Hebbel et al6 showed that vasoocclusive severity correlated with adhesivity of SS RBC in vitro. Subsequently, these and many other investigators have defined adhesion pathways involved in SS RBC adhesion to cultured endothelium under static and flow conditions (Table; also reviewed in references 2 and 7). Adhesion receptors on SS RBC include α4β1 integrin8,9 and CD368,10 whose expression is increased on sickle versus normal reticulocytes8-10 and is down-regulated by treatment with hydroxyurea.11 Aggregated, membranous band 312 and sulfated glycolipids exposed on the surface of damaged RBC also have been implicated. On the endothelial side, cytokine-induced VCAM-1,9,13,14 a ligand for α4β1, and αvβ3 integrin,10 which binds von Willebrand factor (vWf) and thrombospondin (TSP), have been demonstrated to mediate SS RBC adhesion. GpIb and CD36 are also potential endothelial adhesion receptors. The adhesive protein TSP,15,16 released by platelets, and vWf,17released by endothelium, promote SS RBC adhesion to cultured endothelium, serving as bridging molecules between endothelial and SS RBC receptors.

Potential sickle red blood cell–vessel wall adhesion pathways

Endothelial Cell Adhesive Bridging Protein SS RBC
αVβ3 ←——— TSP ———→  sulfated glycolipid 
αVβ3 ←——— vWf ———→  sulfated glycolipid 
αVβ3 ←——— TSP ———→  CD36  
VCAM-1 ←————————→  α4β1 
CD36  ←——— TSP ———→  CD36  
GPIb ←——— vWf ———→  sulfated glycolipid  
←————————→  band 3 
Subendothelial Matrix   SS RBC  
vWf ←————————→  sulfated glycolipid  
TSP ←————————→  sulfated glycolipid  
LN ←————————→  sulfated glycolipid  
FN ←————————→  α4β1 
LN  ←————————→  B-CAM/LU 
Endothelial Cell Adhesive Bridging Protein SS RBC
αVβ3 ←——— TSP ———→  sulfated glycolipid 
αVβ3 ←——— vWf ———→  sulfated glycolipid 
αVβ3 ←——— TSP ———→  CD36  
VCAM-1 ←————————→  α4β1 
CD36  ←——— TSP ———→  CD36  
GPIb ←——— vWf ———→  sulfated glycolipid  
←————————→  band 3 
Subendothelial Matrix   SS RBC  
vWf ←————————→  sulfated glycolipid  
TSP ←————————→  sulfated glycolipid  
LN ←————————→  sulfated glycolipid  
FN ←————————→  α4β1 
LN  ←————————→  B-CAM/LU 

SS RBC, sickle red blood cell; TSP, thrombospondin; vWf, von Willebrand factor; VCAM-1, vascular cell adhesion molecule-1; GPIb, glycoprotein Ib; FN, fibronectin; LN, laminin; B-CAM/LU, basal cell adhesion molecule/lutheran protein.

SS RBC interactions with the vessel wall also may involve interactions with subendothelial matrix components such as laminin (LN),18,19 TSP,18 vWf, or fibronectin20 (Table). Matrix components may be exposed by vascular injury or by endothelial retraction induced by stimuli such as thrombin.21 A sulfated glycolipid isolated from SS RBC was shown to bind to LN and TSP,18 and sulfated glycolipids also have been reported to bind to vWf.22 B-CAM/LU (basal cell adhesion molecule/lutheran protein) was recently shown to be a major LN receptor on SS RBC.19 

The majority of studies of SS RBC interactions with endothelium or subendothelial matrix have used in vitro approaches. Studies with cultured cells or purified matrix components, under static or flow conditions, have been invaluable in identifying potential adhesive interactions. Ultimately, however, it is necessary to validate concepts and pathways in animal models. Fabry et al23 used an intact rat model with gamma camera imaging and visualization of the microvasculature by silicone injection. They demonstrated that desmopressin, possibly by releasing vWf, increased the retention of arterially injected deformable SS discocytes without producing overt obstruction, and suggested that narrowing of postcapillary venules by the adhesion of deformable SS RBC facilitated trapping of less deformable SS RBC, triggering vasoocclusion.

The century-old technique of intravital microscopy, coupled with modern video image analysis, allows precise quantification of blood cell interactions with the vessel wall. This approach has proven to be a powerful tool in defining leukocyte-endothelial interactions, leading to the formulation of the multistep model of leukocyte emigration with selectin-mediated tethering/rolling and integrin-dependent sticking/transmigration.24 Kaul and colleagues have been leaders in applying intravital microscopy to elucidate SS RBC interactions with the vessel wall, using the rat mesocecum ex vivo25 and the sickle transgenic mouse in vivo.26 Their studies demonstrated that deformable SS RBC are more likely to adhere than dense SS RBC25 and that adhesion was limited to postcapillary venules.25,26 They also showed that desmopressin-stimulated human SS RBC adhesion to postcapillary venules in the ex vivo rat mesocolon was inhibited by anti-vWf—but not anti-TSP—antibody, providing the first trial of adhesion blockade with specific reagents in an animal model of sickle disease.27 

Previous studies in vitro10,28 suggested that αVβ3 integrin receptor, which is expressed on the luminal surface of endothelial cells,29 may be involved in SS RBC adhesion to endothelium. Kumar et al28 found that a conformationally constrained RGD-containing peptide that inhibits αVβ3 significantly reduced plasma-induced SS RBC adhesion to cultured endothelial cells under flow. Sugihara et al10 reported that the anti-αVβ3 MoAb LM609 and anti-αIIbβ3 MoAb 7E3, which cross-reacts with αVβ3 but not the non–cross-reactive anti-αIIbβ3 MoAb 10E5, reduced plasma-dependent SS RBC static adhesion to cultured human endothelial cells. In the current study, Kaul and colleagues used the same reagents in the ex vivo rat model to address the role of αVβ3 integrin. Platelet-activating factor, a mediator that is increased in plasma of sickle cell patients, was used to stimulate SS RBC adhesion to postcapillary venules, possibly by releasing vWf or by provoking endothelial cell retraction. As shown in the dramatic videomicroscopy (vide infra), treatment with the αVβ3-blocking MoAb 7E3 largely abolished PAF-stimulated SS RBC adhesion to the vessel wall. MoAb 7E3 and the αVβ3-specific MoAb LM609, but not the αIIbβ3-specific MoAb 10E5, also improved hemodynamics in the PAF-treated vessels.

This study provides compelling evidence for a role of endothelial αVβ3 integrin in SS RBC adhesion to PAF-stimulated postcapillary venules in the rat. There are, of course, a number of obvious cautions in extrapolating these exciting observations in the animal model to sickle cell vasoocclusive crises. First, this is an ex vivo model with artificial perfusion of isolated cells in a plasma-free medium, certainly not reflecting the complex hemodynamics and rheology that occur in the microcirculation during inflammation and hypoxia. Second, although the in vitro studies with large vessel human endothelial cells are consistent with the ex vivo results in the rat regarding the role of αVβ3 integrin, the interaction of human SS RBC with human microvascular endothelium in vivo may involve adhesion pathways different from those observed in the rat microvasculature. Third, the model examines only a single inflammatory stimulus, PAF, in the absence of other blood cells, whereas vasoocclusion occurs in a complex milieu of platelets, leukocytes, plasma, and multiple inflammatory mediators that may markedly alter the adhesion pathways used.

Despite these caveats, the current study by Kaul et al suggests that blockade of endothelial αVβ3 integrin might be beneficial in treatment of vasoocclusion in sickle cell disease. Integrin receptors have emerged as important therapeutic targets. Anti-adhesion therapies directed to platelet αIIbβ3 integrin receptor have been proven to be of considerable benefit in acute coronary syndromes.30These drugs include abciximab, a derivative of MoAb 7E3 that cross-reacts with αVβ3 integrin.31 Antagonists of leukocyte α4β1 and β2 integrin receptors have demonstrated efficacy in diverse animal models32 and are now in clinical trial in several acute inflammatory disorders. Small molecule and antibody-based specific inhibitors of αVβ3 integrin are being developed as anti-angiogenic agents.33 If αVβ3 antagonists continue to prove safe in other indications, what additional studies would be required before testing them for the prevention or acute treatment of vasoocclusive episodes in sickle cell disease? Certainly, confirmatory results in other animal models of sickle cell disease would be encouraging. However, given the strong rationale, the supporting ex vivo and in vitro evidence, and the limitations of any of the animal models, it would seem reasonable even now to consider a clinical trial.

Reprints:John M. Harlan, Division of Hematology, Box 359756, Harborview Medical Center, 325 Ninth Avenue, Seattle, WA 98104.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Kaul
DK
Tsai
HM
Liu
XD
Nakada
MT
Nagel
RL
Coller
BS
Monoclonal antibodies to αVβ3 (7E3 and LM609) inhibit sickle red blood cell–endothelium interactions induced by platelet-activating factor.
Blood.
94
1999
1
2
Hebbel
RP
Adhesive interactions of sickle erythrocytes with endothelium.
J Clin Invest.
100
1997
S83
3
Solovey
A
Lin
Y
Browne
P
Choong
S
Wayner
E
Hebbel
RP
Circulating activated endothelial cells in sickle cell anemia.
N Engl J Med.
337
1997
1584
4
Hebbel
RP
Yamada
O
Moldow
CF
Jacob
HS
White
JG
Eaton
JW
Abnormal adherence of sickle erythrocytes to cultured vascular endothelium: possible mechanism for microvascular occlusion in sickle cell disease.
J Clin Invest.
65
1980
154
5
Hoover
R
Rubin
R
Wise
G
Warren
R
Adhesion of normal and sickle erythrocytes to endothelial monolayer cultures.
Blood.
54
1979
872
6
Hebbel
RP
Boogaerts
MA
Eaton
JW
Steinberg
MH
Erythrocyte adherence to endothelium in sickle-cell anemia. A possible determinant of disease severity.
N Engl J Med.
302
1980
992
7
Bunn
HF
Pathogenesis and treatment of sickle cell disease.
N Engl J Med.
337
1997
762
8
Joneckis
CC
Ackley
RL
Orringer
EP
Wayner
EA
Parise
LV
Integrin alpha 4 beta 1 and glycoprotein IV (CD36) are expressed on circulating reticulocytes in sickle cell anemia.
Blood.
82
1993
3548
9
Swerlick
RA
Eckman
JR
Kumar
A
Jeitler
M
Wick
TM
Alpha 4 beta 1-integrin expression on sickle reticulocytes: vascular cell adhesion molecule-1-dependent binding to endothelium.
Blood.
82
1993
1891
10
Sugihara
K
Sugihara
T
Mohandas
N
Hebbel
RP
Thrombospondin mediates adherence of CD36+ sickle reticulocytes to endothelial cells.
Blood.
80
1992
2634
11
Styles
LA
Lubin
B
Vichinsky
E
et al
Decrease of very late activation antigen-4 and CD36 on reticulocytes in sickle cell patients treated with hydroxyurea.
Blood.
89
1997
2554
12
Thevenin
BJM
Crandall
I
Ballas
SK
Sherman
IW
Shohet
SB
Band 3 peptides block the adherence of sickle cells to endothelial cells in vitro.
Blood.
90
1997
4172
13
Gee
BE
Platt
OS
Sickle reticulocytes adhere to VCAM-1.
Blood.
85
1995
268
14
Setty
BN
Stuart
MJ
Vascular cell adhesion molecule-1 is involved in mediating hypoxia-induced sickle red blood cell adherence to endothelium: potential role in sickle cell disease.
Blood.
88
1996
2311
15
Brittain
HA
Eckman
JR
Swerlick
RA
Howard
RJ
Wick
TM
Thrombospondin from activated platelets promotes sickle erythrocyte adherence to human microvascular endothelium under physiologic flow: a potential role for platelet activation in sickle cell vaso-occlusion.
Blood.
81
1993
2137
16
Kaul
DK
Chen
D
Zhan
J
Adhesion of sickle cells to vascular endothelium is critically dependent on changes in density and shape of the cells.
Blood.
83
1994
3006
17
Wick
TM
Moake
JL
Udden
MM
Eskin
SG
Sears
DA
McIntire
LV
Unusually large von Willebrand factor multimers increase adhesion of sickle erythrocytes to human endothelial cells under controlled flow.
J Clin Invest.
80
1987
905
18
Hillery
CA
Du
MC
Montgomery
RR
Scott
JP
Increased adhesion of erythrocytes to components of the extracellular matrix: isolation and characterization of a red blood cell lipid that binds thrombospondin and laminin.
Blood.
87
1996
4879
19
Udani
M
Zen
Q
Cottman
M
et al
Basal cell adhesion molecule/lutheran protein. The receptor critical for sickle cell adhesion to laminin.
J Clin Invest.
101
1998
2550
20
Kasschau
MR
Barabino
GA
Bridges
KR
Golan
DE
Adhesion of sickle neutrophils and erythrocytes to fibronectin.
Blood.
87
1996
771
21
Manodori
AB
Matsui
NM
Chen
JY
Embury
SH
Enhanced adherence of sickle erythrocytes to thrombin-treated endothelial cells involves interendothelial cell gap formation.
Blood.
92
1998
3445
22
Roberts
DD
Williams
SB
Gralnick
HR
Ginsburg
V
Von Willebrand factor binds specifically to sulfated glycolipids.
J Biol Chem.
261
1986
3306
23
Fabry
ME
Fine
E
Rajanayagam
V
et al
Demonstration of endothelial adhesion of sickle cells in vivo: a distinct role for deformable sickle cell discocytes.
Blood.
79
1992
1602
24
Carlos
TM
Harlan
JM
Leukocyte-endothelial adhesion molecules.
Blood.
84
1994
2068
25
Kaul
DK
Fabry
ME
Nagel
RL
Microvascular sites and characteristics of sickle cell adhesion to vascular endothelium in shear flow conditions: pathophysiological implications.
Proc Natl Acad Sci U S A.
86
1989
3356
26
Kaul
DK
Fabry
ME
Costantini
F
Rubin
EM
Nagel
RL
In vivo demonstration of red cell–endothelial interaction, sickling and altered microvascular response to oxygen in the sickle transgenic mouse.
J Clin Invest.
96
1995
2845
27
Kaul
DK
Nagel
RL
Chen
D
Tsai
HM
Sickle erythrocyte–endothelial interactions in microcirculation: the role of von Willebrand factor and implications for vasoocclusion.
Blood.
81
1993
2429
28
Kumar
A
Eckman
JR
Wick
TM
Inhibition of plasma-mediated adherence of sickle erythrocytes to microvascular endothelium by conformationally constrained RGD-containing peptides.
Am J Hematol.
53
1996
92
29
Conforti
G
Dominguez-Jimenez
C
Zanetti
A
et al
Human endothelial cells express integrin receptors on the luminal aspect of their membrane.
Blood.
80
1992
437
30
Coller
BS
Platelet GPIIb/IIIa antagonists: the first anti-integrin receptor therapeutics.
J Clin Invest.
100
1997
S57
31
Coller
BS
Potential non-glycoprotein IIb/IIIa effects of abciximab.
Am Heart J.
38
1999
S1
32
Cornejo
CJ
Winn
RK
Harlan
JM
Anti-adhesion therapy.
Adv Pharmacol.
39
1997
99
33
Eliceiri
BP
Cheresh
DA
The role of alphaV integrins during angiogenesis: insights into potential mechanisms of action and clinical development.
J Clin Invest.
103
1999
1227
Sign in via your Institution