In this issue of Blood, Langhauser and colleagues report that kininogen knockout mice (KNG−/−) are protected from stroke induced by transient mechanical occlusion of the middle artery (MCAO).1  KNG−/− mice develop smaller infarcts, less neurologic impairment, and exhibit lower mortality than wild-type (WT) mice studied at 24 hours and beyond. Benefit was retained in elderly mice and was conferred through reduction in microvascular thrombosis, preservation of blood-brain-barrier function, and attenuation of inflammation. These are important findings because they provide insight into the pathophysiology of stroke and identify potential novel targets for intervention.

Ischemic stroke remains the one of the most common causes of mortality and protracted morbidity. It is estimated that approximately 750 000 new patients a year in the United States are affected. Notwithstanding extensive investigation, thrombolysis by tissue-type plasminogen activator (tPA) remains the sole US Food and Drug Administration–approved intervention. However, only 3% to 6% of patients with ischemic stroke receive tPA because of delays in medical care, lesion size, and evidence of hemorrhagic transformation that make the risk of intracerebral hemorrhage prohibitive. Moreover, even patients who respond to tPA may be left with significant neurologic defects and risk for recurrent stroke. Thus, there is a clear need for new, more effective, and safer treatments.

This study by Langhauser and colleagues builds on emerging literature, which indicates that components of the contact factor pathway of coagulation, including factor XII, prekallekrein and KNG, the proteolytic product bradykinin (BK), and the BK 2 receptor, are involved in the response to vascular injury, but have less prominent roles in hemostasis. It has been posited that activation of the contact factor pathway in vivo requires higher concentrations of thrombin, found only in pathologic conditions, than does activation of the intrinsic pathway.2  On the other hand, inflammation and tissue injury expose activators of the contact factor pathway found in the extracellular matrix and others released by cell activation or injury, including polyphosphates from platelets, neutrophil serine proteases and nucleosomes, and RNA from degraded cells,3-5  some seemingly unassociated with activation of factor XI.6  This suggests that specific inhibition of KNG-related activities might impede propagation of thrombosis7  without potentiating bleeding.

This study also highlights the concept that the pathophysiology of stroke and the sequelae of treatment in this mouse model, as in humans, extend beyond intravascular thrombosis. Neuroprotection after transient MCAO simulates human ischemic stroke treated successfully with tPA. The finding that BK restored deleterious outcomes in KNG−/− animals and the absence of significant differences in cortical blood flow immediately after reperfusion speaks to the potential contribution of nonhemostatic pathways, including alterations in vascular contractility.8,9  Within minutes of MCAO, cerebrovascular autoregulation, which matches blood flow to local metabolic needs, is lost, reducing blood flow to ischemic neurons and the penumbra, while diverting flow to surrounding healthy tissue. Local release of glutamate and plasminogen activators from ischemic tissue causes intense activation of N-methyl-d-aspartate and the low-density lipoprotein receptors, which exacerbates loss of adaptive cerebral vasoregulation and leads to neuronal apoptosis.10,11  Improved outcomes are seen in models of thrombotic and mechanical occlusion when the deleterious effects of plasminogen activators on vascular contractility are blocked without affecting plasminogen activation.10,11  Such approaches might help dissociate the deleterious effects of plasminogen activators from their salutary effects on fibrinolysis.

This study by Langauser and colleagues also points to the need to better delineate the contribution of inflammation to vascular damage and thrombosis. Generation of cytokines, up-regulation of leukocyte adhesion molecules, leukocyte infiltration and generation of metalloproteinases, reactive oxygen species, and lipid peroxidation, among diverse other factors, enhance ischemic and reperfusion injury and promote clot stability and progression. Attenuating the inflammatory response with activated protein C, cytokine antagonists and inhibitors of cell adhesion have been effective in this model, although the clinical benefits remain modest or uncertain, highlighting the complexity of human stroke.

To the extent that thrombosis is superimposed on vascular injury caused by ischemia, (peri)vascular inflammation, and changes in contractility, among many other factors, it may be possible to intervene proximal to the onset of coagulation, breaking the so far inexorable link between the benefits and risks of antithrombotic therapy. Timing here may be critical. Understanding the most proximal events and the sequence of injury will help to delineate both the targets and the windows of opportunity for preventing vascular and parenchymal damage. Thus, this important study by Langhauser and colleagues should prompt further investigation into the diverse activities of KNG and the other members of this pathway and its implications for novel approaches to mitigate thrombotic occlusion and reperfusion injury.

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

1
Langhauser
 
F
Gob
 
E
Kraft
 
P
, et al. 
Kininogen deficiency protects from ischemic neurodegeneration in mice by reducing thrombosis, blood-brain barrier damage, and inflammation.
Blood
2012
, vol. 
120
 
19
(pg. 
4082
-
4092
)
2
Mackman
 
N
Gruber
 
A
Platelet polyphosphate: an endogenous activator of coagulation factor XII.
J Thromb Haemost
2010
, vol. 
8
 
5
(pg. 
865
-
867
)
3
Müller
 
F
Mutch
 
NJ
Schenk
 
WA
, et al. 
Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo.
Cell
2009
, vol. 
139
 
6
(pg. 
1143
-
1156
)
4
Massberg
 
S
Grahl
 
L
von Bruehl
 
ML
, et al. 
Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases.
Nat Med
2010
, vol. 
16
 
8
(pg. 
887
-
896
)
5
Kannemeier
 
C
Shibamiya
 
A
Nakazawa
 
F
, et al. 
Extracellular RNA constitutes a natural procoagulant cofactor in blood coagulation.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
15
(pg. 
6388
-
6393
)
6
Maas
 
C
Govers-Riemslag
 
JW
Bouma
 
B
, et al. 
Misfolded proteins activate factor XII in humans, leading to kallikrein formation without initiating coagulation.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3208
-
3218
)
7
Revenko
 
AS
Gao
 
D
Crosby
 
JR
, et al. 
Selective depletion of plasma prekallikrein or coagulation factor XII inhibits thrombosis in mice without increased risk of bleeding.
Blood
2011
, vol. 
118
 
19
(pg. 
5302
-
5311
)
8
Iwaki
 
T
Cruz-Topete
 
D
Castellino
 
FJ
A complete factor XII deficiency does not affect coagulopathy, inflammatory responses, and lethality, but attenuates early hypotension in endotoxemic mice.
J Thromb Haemost
2008
, vol. 
6
 
11
(pg. 
1993
-
1995
)
9
Shariat-Madar
 
Z
Mahdi
 
F
Warnock
 
M
, et al. 
Bradykinin B2 receptor knockout mice are protected from thrombosis by increased nitric oxide and prostacyclin.
Blood
2006
, vol. 
108
 
1
(pg. 
192
-
199
)
10
Armstead
 
WM
Cines
 
DB
Bdeir
 
K
Kulikovskaya
 
I
Stein
 
SC
Higazi
 
AA
uPA impairs cerebrovasodilation after hypoxia/ischemia through LRP and ERK MAPK.
Brain Res
2008
, vol. 
1231
 (pg. 
121
-
131
)
11
Armstead
 
WM
Nassar
 
T
Akkawi
 
S
, et al. 
Neutralizing the neurotoxic effects of exogenous and endogenous tPA.
Nat Neurosci
2006
, vol. 
9
 
9
(pg. 
1150
-
1155
)

National Institutes of Health

Sign in via your Institution