scholarly journals Tissue factor promotes activation of coagulation and inflammation in a mouse model of sickle cell disease

Blood ◽  
2012 ◽  
Vol 120 (3) ◽  
pp. 636-646 ◽  
Author(s):  
Pichika Chantrathammachart ◽  
Nigel Mackman ◽  
Erica Sparkenbaugh ◽  
Jian-Guo Wang ◽  
Leslie V. Parise ◽  
...  

Abstract Sickle cell disease (SCD) is associated with a complex vascular pathophysiology that includes activation of coagulation and inflammation. However, the crosstalk between these 2 systems in SCD has not been investigated. Here, we examined the role of tissue factor (TF) in the activation of coagulation and inflammation in 2 different mouse models of SCD (BERK and Townes). Leukocytes isolated from BERK mice expressed TF protein and had increased TF activity compared with control mice. We found that an inhibitory anti-TF antibody abrogated the activation of coagulation but had no effect on hemolysis or anemia. Importantly, inhibition of TF also attenuated inflammation and endothelial cell injury as demonstrated by reduced plasma levels of IL-6, serum amyloid P, and soluble vascular cell adhesion molecule-1. In addition, we found decreased levels of the chemokines MCP-1 and KC, as well as myeloperoxidase in the lungs of sickle cell mice treated with the anti-TF antibody. Finally, we found that endothelial cell-specific deletion of TF had no effect on coagulation but selectively attenuated plasma levels of IL-6. Our data indicate that different cellular sources of TF contribute to activation of coagulation, vascular inflammation, and endothelial cell injury. Furthermore, it appears that TF contributes to these processes without affecting intravascular hemolysis.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 375-375
Author(s):  
Pichika Chantrathammachart ◽  
Erica M Sparkenbaugh ◽  
Nigel Mackman ◽  
Nigel S Key ◽  
Rafal Pawlinski

Abstract Abstract 375 Sickle cell disease (SCD) is a hematologic disorder caused by a single nucleotide mutation of the beta-globin gene. It is associated with increased tissue factor (TF) expression, activation of coagulation and chronic vascular inflammation. Using two mouse models of SCD (BERK and Townes mice), we have recently demonstrated that inhibition of TF with a rat anti-mouse TF (1H1) antibody not only abolishes activation of coagulation (measured by plasma levels of thrombin anti-thrombin (TAT) complexes) but also reduces inflammation and endothelial cell (EC) injury, indicated by attenuation of plasma levels of IL-6 and sVCAM-1, respectively. Furthermore, we showed that EC-specific deletion of TF gene significantly reduced plasma levels of IL-6 but had no effect on activation of coagulation (TAT) or EC injury (sVCAM-1). These data suggest that EC-TF is primarily involved in signaling rather than activation of coagulation. Since TF:factor VIIa complex-dependent activation of protease activated receptor-2 (PAR-2) has been shown to promote inflammation, we have now investigated the role of PAR-2 expressed by non-hematopoietic cells in the pathology of SCD. PAR-2+/+ and PAR-2−/− mice were lethally irradiated and transplanted with bone marrow from BERK SS (sickle cell mice) or BERK AA (non-sickle control) mice(n=6–10). Four months after bone marrow transplantation, mice were sacrificed and the reconstitution of bone marrow was confirmed by electrophoretic analysis of the different forms of hemoglobin. PAR-2+/+ mice transplanted with bone marrow from BERK SS mice had reduced number of red blood cells and hematocrit compared to PAR-2+/+ mice transplanted with bone marrow from BERK AA mice. PAR-2 deficiency in all non-hematopoietic cells had no effect on these hematologic parameters. Furthermore, PAR-2+/+ mice transplanted with bone marrow from BERK SS mice had increased number of monocytes (3.1 fold, p<0.0001) and neutrophils (2.5 fold, p<0.05) in the blood. Interestingly, sickle cell mice lacking PAR-2 in non-hematopoietic cells had significantly reduced neutrophil counts compared to the sickle cell mice with normal levels of PAR-2 (1.9+/−0.2 vs. 5.4+/−1.4 X103/ul; p<0.05), whereas monocytes counts were not affected. Compared to non-sickle controls, sickle cell mice had increased plasma levels of TAT (1.9 fold, p<0.01), IL-6 (6.8 fold, P<0.0001), serum amyloid protein SAP (6.5 fold, p<0.01; mouse homolog of human C reactive protein) and sVCAM-1 (1.4 fold, p<0.01). Moreover, increased levels of myeloperoxidase (MPO) were observed in the livers of sickle cell mice (3 fold, p<0.0001). Importantly, sickle cell mice lacking PAR-2 expression in all non-hematopoietic cells demonstrated significant reduction of plasma levels of IL-6 (9.4+/−0.9 vs. 18.9+/−4.5 pg/ml; p<0.05) and SAP (60.5+/−12.9 vs. 182.8+/−62.5ug/ml; p<0.05) compared to sickle cell mice with normal levels of PAR2 expression. In addition, deletion of PAR-2 also significantly reduced MPO levels in the liver of sickle cell mice (53.7+/−3.5 vs. 117.4+/−16.9 ng/mg protein; p<0.0001). In contrast, PAR-2 deficiency in non-hematopoietic cells had no effect on activation of coagulation (TAT) or EC injury (sVCAM-1) in sickle mice. Our data demonstrate that vascular inflammation observed in a mouse model of sickle cell disease is mediated, in part, by PAR-2 expressed by non-hematopoietic cells. Activation of EC (sVCAM-1) was not affected by PAR-2 deficiency. Ongoing studies are investigating the possible contribution of the TF-thrombin-PAR1 pathway to the EC activation in SCD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 181-181 ◽  
Author(s):  
Hao Xu ◽  
Nancy J. Wandersee ◽  
YiHe H. Guo ◽  
Deron W. Jones ◽  
Sandra L. Holzhauer ◽  
...  

Abstract High Mobility Group Box 1 (HMGB1) is a nuclear protein that aids in regulating gene expression and the organization of DNA structure. However, upon cellular activation or injury, HMGB1 can be secreted from activated immune cells through non-classical ‘leaderless’ pathways or released from tissues through necrosis. Once released, HMGB1 acts as a Damaged-Associated Molecular Pattern (DAMP) that binds with other DAMPs and cytokines to activate Toll-Like Receptor 4 (TLR4), resulting in pro-inflammatory signaling and impaired endothelial cell function. It is well established that sickle cell disease (SCD) increases neutrophil count and activation. In addition, tissue injury and inflammation is further exacerbated by the ischemia/reperfusion that occurs during acute vaso-occlusion in SCD. Very little is known concerning HMGB1 release in SCD and its role in the pathology of SCD. We hypothesize that SCD increases HMGB1 release and that SCD-dependent increases in HMGB1 and oxidative stress act in concert to impair endothelial cell (EC) function and increase vascular congestion and tissue injury. To explore this hypothesis, we assessed plasma levels of HMGB1 in SCD as compared to normal controls. In humans, we found that individuals with SCD have ∼4-fold increased plasma HMGB1 levels compared to plasma levels in control individuals (p=0.02). Similarly, the Berkeley mouse model of SCD has ∼2-fold higher levels of plasma HMGB1 compared to control animals (p<0.01). Next, we measured plasma HMGB1 levels in SCD and control mice after exposure to hypoxia (3 hrs 10% FIO2) to induce acute sickling followed by reoxygenation (2 hrs room air, H/R) as an experimental model of acute vaso-occlusion. Importantly, hypoxia/reoxygenation (H/R) increased HMGB1 levels in SS mice more than 3-fold, while having little effect on the plasma levels of HMGB1 in control mice (p<0.03). This indicates that H/R induces immune cell activation and/or tissue injury, which increases the release of HMGB1 into the plasma. Since H/R induced such high plasma concentrations of HMGB1 in SCD mice, we examined if HMGB1 alone was sufficient to induce vascular congestion. Mice were injected with recombinant HMGB1 to deliver levels similar to that found in SCD mice post H/R; after 3 hrs mice were euthanized and lungs harvested. Histologic examination of lung sections showed that HMGB1 directly increased vascular congestion in SCD but not control mice. These data indicate that, even in the absence of acute sickling, SCD mice are more susceptible than control mice to HMGB1-induced lung vascular injury. Finally, to begin to understand how SCD induces chronic states of inflammation, we determined the effects of human and mouse plasma on TLR4 receptor activity. To assess TLR4 receptor activity we used TLR4 “reporter cells” that secrete alkaline phosphatase upon binding and activation of TLR4. We found that plasma from individuals with SCD induced ∼4-fold greater increases in TLR4 reporter activity compared to plasma from healthy race-matched individuals (p<0.025). Likewise, after H/R treatment, plasma from SCD mice induced ∼2-fold greater increase in TLR4 reporter activity compared to plasma from control mice (p<0.05). The “gold standard” for determining if HMGB1 plays a role in inflammation is the use of neutralizing antibodies. Interestingly, anti-HMGB1 antibody treatment of H/R-exposed SCD mice markedly diminished the plasma-induced TLR4 receptor activity to levels similar to that observed from control mice (p<0.05), suggesting that much of the increase in TLR4 receptor activity induced by plasma from SS-H/R mice is HMGB1-dependent. Taken together, these data indicate that SCD increases the release of HMGB1 and suggests that HMGB1 plays an important role in the mechanisms by which SCD impairs vascular function and increases vascular congestion. Disclosures: Wandersee: Bayer: Consultancy. Guo:Bayer: Consultancy. Hillery:Bayer: Consultancy; Biogen Idec: Consultancy.


1995 ◽  
Vol 73 (05) ◽  
pp. 763-767 ◽  
Author(s):  
Kazuomi Kario ◽  
Takefumi Matsuo ◽  
Miyako Matsuo ◽  
Masanobu Koide ◽  
Tsutomu Yamada ◽  
...  

SummaryWe investigated plasma activated factor VII (FVIIa) levels in uremic patients (nondialysis group: n = 38; dialysis group: n = 36) and healthy controls (n = 32). We also measured the plasma levels of thrombomodulin (an indicator of endothelial cell injury) and tissue factor. Plasma FVIIa showed a marked increase in the nondialysis group (mean [95% confidence interval]: 4.6 [4.1-5.1] ng/ml, p <0.0001) with the progressive impairment of renal function, as indicated by the serum creatinine level, when compared with the 32 controls (2.8 [2.5-3.1] ng/ml), and was further increased in the dialysis group (6.1 [5.5-6.8] ng/ml, p <0.001 vs. nondialysis group). Plasma levels of thrombomodulin and tissue factor were also higher in the nondialysis group than the control group, and were further increased in the dialysis group. Plasma tissue factor levels did not show any correlation with FVIIa or thrombomodulin in both the nondialysis and dialysis groups. Thus, circulating tissue factor appears to be released by a different mechanism from thrombomodulin and may not contribute to the direct activation of factor VII in uremic patients. On the other hand, the plasma level of thrombomodulin was positively correlated with that of FVIIa in the nondialysis group, and this correlation was independent of renal function. Thus, enhanced conversion of factor VII zymogen to FVIIa, probably related to endothelial cell injury, may be a risk factor for cardiovascular events in uremic patients.


2005 ◽  
Vol 16 (3) ◽  
pp. 209-214 ◽  
Author(s):  
Junette S Mohan ◽  
Gregory YH Lip ◽  
Josh Wright ◽  
David Bareford ◽  
Andrew D Blann

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 236-236 ◽  
Author(s):  
Cheryl A. Hillery ◽  
Thomas D. Foster ◽  
Sandra L. Holzhauer ◽  
J. Paul Scott ◽  
Julie A. Panepinto ◽  
...  

Abstract There is considerable evidence for increased levels of procoagulant proteins, thrombin generation and tissue factor activity in patients with sickle cell disease (SCD). Both thrombin and tissue factor have potent proinflammatory effects. We found increased fibrin deposition in organs that develop SCD vaso-occlusive pathologies in the Berkeley murine model of SCD. In addition, we demonstrated that transplantation of sickle hematopoietic stem cells (HSC) into mice deficient in fibrinogen (SCD-Fib−/− mice) resulted in a decreased number and size of liver infarcts. These data suggest that fibrin clot formation is critical for the final irreversible stoppage of blood flow that can progress to focal tissue infarction. However, the SCD-Fib−/− mice still develop other typical organ pathologies associated with SCD, including focal sites of prominent vascular congestion that are likely due to persistent endothelial injury and inflammation. Because patients with afibrinogenemia have high levels of circulating thrombin, the persistent organ pathology in the SCD-Fib−/− mice may also be linked to the increased exposure to thrombin’s potent proinflammatory effects. In agreement, we now have evidence for increased circulating thrombin-antithrombin (TAT) complexes in SCD-Fib−/− mice compared to SCD-wt transplant control mice (54 ± 25 vs 9.9 ± 3.5 ng/mL, p=0.04) demonstrating increased thrombin generation in the SCD-Fib−/− mice. In addition, soluble VCAM-1, a marker of endothelia injury, was increased in SCD-Fib−/−mice compared to SCD-wt mice (843 ± 90 vs 628 ± 110). In order to determine whether the tissue factor pathway contributes significantly to proinflammatory pathologies observed in SCD, we transplanted fetal liver HSC from mice that predominately express human sickle hemoglobin (mα−/−, mβ−/−, Tg[HbS]+: “SCD mice” or hemizygous mα−/−, mβ+/−, Tg[HbS]x2: “hSCD2 mice,” Berkeley model) into mice deficient in tissue factor (mTF−/−, hTF+: “TFLow mice” that express ~1%TF levels) or control heterozygote mice (mTF+/−, hTF+) following lethal irradiation. Four months following transplant, we obtained blood samples for engraftment and hematologic studies (&gt;90% engraftment required for inclusion in this study), followed by perfusion of anesthetized animals. Perfused organs were divided with one part fixed for histologic study. Frozen nonfixed sections of the organs were homogenized and organ Hb content determined per gram organ weight. We found that tissue factor deficiency (SCD-TFLow and hSCD2-TFLow mice) resulted in a dramatic reduction of vascular congestion in liver histologic sections that was confirmed by quantitation of organ-specific trapped Hb: 1.2 ± 0.3 vs 6.3 ± 1.7 mg Hb/gm liver in hSCD2-TFLow vs hSCD2-TF+/− control mice (n = 8–9 per group, p&lt;0.001) with similar results in a smaller number of SCD-TFLow mice. These data suggest that the tissue factor pathway contributes importantly to vascular inflammation and cellular stasis. Thus, while the coagulation pathway appears to be a critical component of HbS-induced tissue infarction, endothelial injury and inflammation also significantly contribute to HbS-induced organ pathology. These data also suggest that targeted anticoagulation therapies may ameliorate the focal organ infarction and/or vascular inflammation that are prevalent in SCD.


Blood ◽  
2014 ◽  
Vol 123 (11) ◽  
pp. 1630-1631 ◽  
Author(s):  
Wolfram Ruf

In this issue of Blood, Sparkenbaugh et al identify coagulation factor Xa (FXa), the target for new protease-selective oral anticoagulants, as a crucial mediator for both coagulation abnormalities and chronic vascular inflammation that characterize sickle cell disease.1


Sign in / Sign up

Export Citation Format

Share Document