Assessment of two contact activation reagents for the diagnosis of congenital factor XI deficiency

2018 ◽  
Vol 163 ◽  
pp. 64-70 ◽  
Author(s):  
Salam Salloum-Asfar ◽  
María E. de la Morena-Barrio ◽  
Julio Esteban ◽  
Antonia Miñano ◽  
Cristina Aroca ◽  
...  
2019 ◽  
Vol 153 (10) ◽  
pp. 373-379
Author(s):  
Carlos Bravo-Perez ◽  
Teresa Ródenas ◽  
Julio Esteban ◽  
Maria Eugenia de la Morena-Barrio ◽  
Salam Salloum-Asfar ◽  
...  

Blood ◽  
1997 ◽  
Vol 90 (3) ◽  
pp. 1055-1064 ◽  
Author(s):  
David Gailani ◽  
Mao-Fu Sun ◽  
Yuehui Sun

Factor XI is a plasma glycoprotein that is required for contact activation initiated fibrin formation in vitro and for normal hemostasis in vivo. In preparation for developing a mouse model of factor XI deficiency to facilitate investigations into this protease's contributions to coagulation, we cloned the complementary DNA for murine factor XI, expressed the protein in a mammalian expression system, and compared its properties with human recombinant factor XI. The 2.8-kb murine cDNA codes for a protein of 624 amino acids with 78% homology to human factor XI. Both recombinant murine and human factor XI are 160 kD homodimers comprised of two 80 kD polypeptides connected by disulfide bonds. Murine factor XI shortens the clotting time of human factor XI deficient plasma in an activated partial thromboplastin time assay, with a specific activity 50% to 70% that of the human protein. In a purified system, murine factor XI is activated by human factor XIIa and thrombin in the presence of dextran sulfate. Murine factor XI differs from human factor XI in that it undergoes autoactivation slowly in the presence of dextran sulfate. This is due primarily to murine factor XIa preferentially cleaving a site on zymogen factor XI within the light chain, rather than the activation site between Arg371 and Val372. Northern blots of polyadenylated messenger RNA show that murine factor XI message is expressed, as expected, primarily in the liver. In contrast, messenger RNA for human factor XI was identified in liver, pancreas, and kidney. The studies show that murine and human factor XI have similar structural and enzymatic properties. However, there may be variations in tissue specific expression and subtle differences in enzyme activity across species.


2018 ◽  
pp. 291-306
Author(s):  
Tahere Tabatabaei ◽  
Akbar Dorgalaleh

2019 ◽  
Vol 153 (10) ◽  
pp. 373-379
Author(s):  
Carlos Bravo-Perez ◽  
Teresa Ródenas ◽  
Julio Esteban ◽  
Maria Eugenia de la Morena-Barrio ◽  
Salam Salloum-Asfar ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1796-1796
Author(s):  
Vickie McDonald ◽  
Savidge F. Geoffrey ◽  
Savita Rangarajan ◽  
Mike Mitchell

Abstract Traditional treatment modalities for FXI deficiency (UK prevalence 400 cases) include antifibrinolytics, desmopressin, fresh frozen plasma (FFP) and FXI concentrates but there has been reluctance to use FXI concentrates because of reported incidence rates of thrombosis up to 10%. Concerns over the safety and efficacy of FFP, with additional viral inactivation steps possibly leading to reduced FXI recoveries, have led us to increase our use of FXI concentrates. We aimed to assess the indications, dosage, recovery, efficacy and safety of Hemoleven, a plasma derived, purified and virally inactivated FXI concentrate, which also contains heparin and antithrombin, in patients with congenital factor XI deficiency. A retrospective study was performed using hospital notes and laboratory records of all patients who had received Hemoleven over a 2-year period. Eleven patients (6 male, 5 female) had been treated with a median age of 38 years (range 7–74) and mean baseline FXI:C levels of 25.4U/dl (3–50). All patients received Hemoleven as prophylaxis for surgery or dental work and had all previously had excess bleeding when surgically challenged. One patient died of a condition unrelated to FXI treatment. Pre- and post-FXI:C levels were available for a total of 60 treatment episodes of which 25 were 1000-unit doses and 35 were 2000-unit doses. The mean increase in FXI:C per 1000-unit dose was 25.4 U/dl (12.4–43.9) while the mean increase in FXI:C per 2000-unit dose was 50.5 U/dl (11.8–106.5). This is consistent with the manufacturer’s data. Ten minute post infusion FXI:C levels were above the normal range (73–133 U/kg) in 8% of patients given 1000 units and 11% of patients given 2000 units but below the normal range in 24% of patients who received 1000 units and 20% of patients who received 2000 units. 90% of treatment episodes led to FXI:C levels above the usual treatment target of 65 U/dl. Genetic analysis of 9/11 patients showed that 2 were homozygous (one type II and one type III), 6 were heterozygous for other recognised mutations and one had no mutation identified but apparent absence of RNA from one allele demonstrated in a relative by qRT-PCR. No excess bleeding or inhibitor development was recorded even in one patient who had had a poor haemostatic response with FFP. There were no episodes of arterial or venous thrombotic complications within this group and no clinical or laboratory evidence of DIC following treatment. In summary, treatment with factor XI concentrates gave consistent increments in FXI:C at the doses given and achieved good haemostasis with no episodes of thrombosis in this study, even in patients over the age of 60y. While the risk of prion transmission is still unknown, use of FXI concentrates is not associated with the risks of fluid overload and TRALI that are seen with FFP. We acknowledge that the study includes small numbers of patients however the cohort of patients with a bleeding diathesis in this condition is small. We conclude that Hemoleven appears to be an effective and reliable treatment for patients with FXI:C deficiency but should be given in the context of FXI:C level monitoring in order to detect those patients who may develop high levels and possible thrombosis.


1993 ◽  
Vol 84 (1) ◽  
pp. 172-174 ◽  
Author(s):  
Steven S. Ginsberg ◽  
Lionel P. Clyne ◽  
Peter McPhedran ◽  
Thomas P. Duffy ◽  
T. Hanson†

1982 ◽  
Vol 69 (6) ◽  
pp. 1270-1276 ◽  
Author(s):  
David M. Stern ◽  
Hymie L. Nossel ◽  
John Owen

Blood ◽  
1981 ◽  
Vol 57 (3) ◽  
pp. 437-443
Author(s):  
S Schiffman ◽  
R Margalit ◽  
M Rosove ◽  
D Feinstein

Recently we have described a normal plasma activity that modulates contact activation by inhibiting adsorption of factor XI to activating surfaces. Here we report the first identified case in which a patient has abnormal clotting tests due to an excess of a similar activity. The patient's plasma had a prolonged partial thromboplastin time and low apparent factor XI assay. His plasma prolonged the partial thromboplastin time of normal plasma and partially neutralized normal factor XI activity in vivo and in vitro. Analysis in dilute plasma revealed normal amounts of factor XI activity and antigen. Factor XI adsorption from plasma to activating surfaces was tested by adding a small amount of 125I-labeled purified factor XI to plasma, exposing the mixture to a glass tube or kaolin, and determining the amount of factor XI adsorbed to the surface. Whereas normal plasma and plasmas deficient in factor XII, factor XI, or Fletcher factor yielded about 4% adsorption to glass, factor XI adsorption from patient's plasma was less than 1%, indicating the presence of an adsorption inhibitor. This inhibitor did not affect factor XI activation or the activity of preformed factor XIa. It was not adsorbed by AI(OH)3 and was present in serum and the macroglobulin peak on gel filtration of the plasma through Sephadex G-200. The patient's history does not allow a definitive conclusion as to whether this inhibitor was associated with abnormal bleeding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 201-201
Author(s):  
Charles E. Bane ◽  
Kelli L. Boyd ◽  
Erik I. Tucker ◽  
Stephen T. Smiley ◽  
Andras Gruber ◽  
...  

Abstract Sepsis is a systemic host inflammatory response to entry and dissemination of an infectious agent. Interconnected pathways including inflammation, coagulation, and complement that serve protective functions when activated locally, contribute to morbidity and mortality when activated systemically during sepsis. Factor XIa (fXIa), the active form of the zymogen factor XI (fXI), contributes to thrombin generation in response to tissue injury. Prior work using a model of polymicrobial sepsis induced by cecal ligation and puncture (CLP) in mice suggests that fXI contributes to the pathology of sepsis by promoting disseminated intravascular coagulation (DIC) and inflammation (Tucker et al. J Infect Dis 2008;198:271 and Blood 2012;119:4762). We investigated the role of fXI in the CLP model, concentrating on the first 24 hours post-injury, using a high-level ligation that affects ≥75% of the length of the cecum. This level of injury causes near-100% mortality in wild type (WT) C57Bl/6 mice (median survival 42 hrs). Survival 7 days post-CLP was significantly greater for fXI-deficient (fXI-/-, 30% p=0.002) and partially deficient (fXI+/-, 17% p=0.003) mice compared to WT littermates (3%). For animals dying after CLP, median time to death was similar in the three groups. Four hours post-CLP, there were peaks in TNFα and IL-10 plasma levels in WT mice that were significantly smaller in fXI-/- mice (TNFα 76 ± 44 and 24 ± 22 pg/mL [p=0.009]; IL-10 10,000 ± 11,000 and 1,000 ± 900 pg/mL [p=0.0003], respectively), with levels also higher in WT mice 24 hrs post-CLP. There was significant white blood cell accumulation in the peritoneum 24 hrs post-CLP in WT and fXI-/- mice, with the lymphocyte fraction significantly elevated only in WT mice (p=0.008). The major rodent acute phase protein serum amyloid P was 5-fold higher in WT mice than in sham-treated mice, while levels in fXI-/-mice were indistinguishable from shams, 24 hrs post-CLP. In contrast to the indicators of inflammation, evidence for DIC was not strong for either genotype 24 hours post-CLP. Platelet counts were reduced similarly in both groups from a baseline mean of ∼600,000/μL, but mean counts remained above 300,000/μL. In both groups, plasma thrombin-antithrombin complex levels did not increase above baseline over the first 24 hrs post-CLP, and fibrinogen levels, which often decrease during DIC, increased modestly. No evidence of gross hemorrhage was noted at necropsy 4, 8, or 24 hrs post-CLP, and there was no histologic evidence of microvascular thrombosis (considered a “gold standard” for diagnosing DIC in laboratory animals) in kidney, liver or brain. The contact system zymogens factor XII (fXII) and prekallikrein (PK) are activated on bacterial surfaces during infection, resulting in cleavage of high molecular weight kininogen (HK) to produce antimicrobial peptides and the pro-inflammatory peptide bradykinin. We measured plasma fXII, PK, and fXI using western blots and densitometry. FXI decreased by ∼30% on average 24 hrs post-CLP in WT mice, consistent with protease activation. PK levels decreased by ∼50% in WT mice 24 hrs post-CLP (p 0.007), but were unchanged in fXI-/- mice. FXII levels decreased slightly in WT mice post-CLP, but increased in fXI-/-mice 4 hrs after CLP, returning to baseline 8 hrs post-CLP. FXI, PK and fXII levels were unchanged in sham-treated animals. The data suggest that fXI contributes to contact activation after CLP, and its absence leads to reduced PK and fXII activation. Previously, we reported that fXI deficiency reduced mortality after CLP (Tucker et al. J Infect Dis 2008;198:271) and infection with the gram-positive organism Listeria monocytogenes (Luo et al., Infect Immun. 2012;80:91), and proposed that this was in part related to down-regulation of coagulation, mitigating consumptive coagulopathy. The data presented here suggest that the high-level injury employed in the CLP-model produces an intense early systemic inflammatory response that leads to early death, before changes characteristic of DIC become obvious. The survival benefit conferred by fXI deficiency in this model then, may be primarily due to early modulation of inflammation, perhaps by altering contact activation, rather than to a prominent antithrombotic effect. Given the relatively mild bleeding diathesis associated with fXI deficiency, pharmacologic inhibition of fXIa may be a safe option for reducing the inflammatory response in septic patients. Disclosures: Tucker: Aoronora, Inc.: Employment. Gruber:Aronora, Inc.: Employment, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees. Gailani:Aronora, Inc.: Membership on an entity’s Board of Directors or advisory committees; Merck: Consultancy; Bayer Pharma: Consultancy; Novartis: Consultancy.


2019 ◽  
Vol 30 (8) ◽  
pp. 409-412
Author(s):  
Akbar Dorgalaleh ◽  
Masoume Gholaminezhad ◽  
Yavar Shiravand ◽  
Majid Naderi ◽  
Majid Safa

Sign in / Sign up

Export Citation Format

Share Document