Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Thrombosis and Haemostasis, Georg Thieme Verlag KG, Vol. 107, No. 05 ( 2012), p. 962-971
    Abstract: Although intravital microscopy models of thrombosis in mice have contributed to dissect the mechanisms of thrombus formation and stability, they have not been well adapted to study long-term evolution of occlusive thrombi. Here, we assessed the suitability of the dorsal skinfold chamber (DSC) for the study of thrombolysis and testing of thrombolytic agents by intravital microscopy. We show that induction of FeCl3-induced occlusive thrombosis is achievable in microvessels of DSCs, and that thrombi formed in DSCs can be visualised by intravital microscopy using brightfield transmitted light, or fluorescent staining of thrombus components such as fibrinogen, platelets, leukocytes, and von Willebrand factor. Direct application of control saline or recombinant tissue-plasminogen activator (rtPA) to FeCl3-produced thrombi in DSCs did not affect thrombus size or induce recanalisation. However, in the presence of hirudin, rtPA treatment caused a rapid dose-dependent lysis of occlusive thrombi, resulting in recanalisation within 1 hour after treatment. Skin haemorrhage originating from vessels located inside and outside the FeCl3-injured area was also observed in DSCs of rtPA-treated mice. We further show that rtPA-induced thrombolysis was enhanced in plasminogen activator inhibitor-1-deficient (PAI-1−/−) mice, and dropped considerably as the time between occlusion and treatment application increased. Together, our results show that by allowing visualization and measurement of thrombus lysis and potential bleeding complications of thrombolytic treatments, the DSC provides a model for studying endogenous fibrinolysis and for first-line screening of thrombolytic agents. Furthermore, using this system, we found that thrombin and clot aging impair the thrombolytic action of rtPA towards FeCl3-produced thrombi.
    Type of Medium: Online Resource
    ISSN: 0340-6245 , 2567-689X
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2012
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 618-618
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 618-618
    Abstract: Abstract 618 Disorders of hemostasis are frequent in patients suffering from classic myeloproliferative neoplasm (MPN) i.e. essential thrombocytemia (ET) or polycythemia vera (PV). They include high incidence of thrombosis, which represents a leading cause of morbidity and mortality, and a lower incidence of hemorrhagic events. The pathogenesis of hemostatic disorders is complex. The JAK2V617F mutation is found in 95% of PV and in 50% of ET. Since receptors coupled to JAK2V617F are hyper-responsive to growth factors, it is proposed that JAK2V617F primes platelets and, as a consequence, directly contributes to the thrombotic risk. However, the relation between the mutation and thrombosis has yet not clearly been established due to variable JAK2V617F allele burden and antithrombotic prophylaxis. Novel MPN animal models expressing the mutation have been developed with reported hemostasis disorders from unknown origin. Therefore, we took the opportunity to use these JAK2V617F knock-in (KI) mice to investigate the effects of JAK2V617F on platelet functions. We used irradiated WT recipient mice grafted with bone marrow (BM) cells from JAK2+/V617F KI donor mice. JAK2+/V617F KI mice were conditional floxed JAK2+/fl mice crossed with transgenic mice expressing Cre under the control of the vav promoter (vavCre/JAK2+/V617F) or Cre-tamoxifen-inducible transgenic mice expressing CreERt under the HSC-SCL promoter (sclCreERt/JAK2+/V617F). All recipient mice developed 4 weeks after transplantation (vavCre/JAK2+/V617F) and tamoxifen-induction (sclCreERt/JAK2+/V617F) a MPN mimicking PV and characterized by elevated hematocrit, platelet and leucocyte levels. Mice were studied between 4 and 9 weeks after the graft or 7, 14, 32 and 60 days after tamoxifen-induction (sclCreERt/JAK2+/V617F). Platelet functions were analyzed in vitro by flow cytometry and light transmission aggregometry, ex vivo by measuring thrombus formation in whole blood and flow conditions on collagen and vWF, and in vivo in a FeCl3-induced model of thrombosis. The analysis of platelet glycoprotein expression was focused on two ITAM receptors, GPVI and CLEC2. The principal observation is a deficiency in GPVI up to 50% in vavCre/JAK2+/V617F mice (p 〈 0.01) while expression of other platelet membrane glycoproteins (GPIb, GPV, GPIIbIIIa and CLEC2) was normal. GPVI deficiency was confirmed by immunoblot while the expression of the FcRg chain was normal. At arterial shear rates, platelet adhesion and aggregation on immobilized collagen and vWF were profoundly impaired with a platelet surface coverage decreased by up to 80% (p 〈 0.01). In vivo, an enlargement of mesenteric vessels was noticed in vavCre/JAK2+/V617F mice. When vessels of the same caliber as WT mice were submitted to FeCl3-induced thrombosis, platelet aggregates formed rapidly (5-8 min) but surprisingly occlusion was not often reached and the aggregates were highly unstable. In sclCreERt/JAK2+/V617F mice, GPVI deficiency was not observed at day 7 and 14 but appeared at day 60 after JAK2V617F-induction by tamoxifen. In vivo, time to vessel occlusion and thrombus stability were initially (day 7 and 14) similar in KI and WT mice. However, the rate to which platelet aggregates formed and their stability became impaired at 32 days after JAK2V617F-induction. In conclusion, these data demonstrate that JAK2V617F induces complex modifications of platelet functions including platelet GPVI deficiency, in vitro loss of collagen-induced aggregation and facilitation of in vivo thrombosis initiation associated with instability of the clot. Therefore, this study provides new insight into the pathophysiology of hemostatic disorders in MPN, thrombus instability being able to drive microvascular thrombosis disorders and pulmonary embolism. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3200-3200
    Abstract: Background: Myeloproliferative neoplasms (MPN) are associated with an increased risk of arterial and venous thrombosis with an annual incidence varying from 1.1% to 6.6% (Patrono, Blood 2013). Comparable to that recommended in non-MPN high-risk patients, low-dose aspirin (81-100 mg/day) is recommended in primary and secondary prophylaxis of thrombosis in polycythemia vera and essential thrombocytemia (Tefferi, Am J Hematol 2012). However, the daily dose of aspirin to optimize efficacy and safety for MPN patients has never been challenged. Aim of the study: Determine the optimal dose of aspirin to achieve biological efficacy in Philadelphia-negative MPN patients. Methods: Patients with Philadelphia-negative MPN who were treated with low-dose aspirin 75 or 100 mg/day were enrolled in this observational study. Patients without any cytostatic drug and patients treated with either pegylated interferon alpha or hydroxyurea were eligible. Major exclusion criteria included inability to adhere to aspirin therapy and chronic oral anticoagulation. Biological efficacy to aspirin was evaluated by platelet aggregation induced by arachidonic acid 1.33 mM on platelet rich plasma and tested at trough level and 24 hours after last aspirin intake. Resistance to aspirin was defined as a maximal platelet aggregation over 20%. According to the results of platelet aggregation, aspirin dose and dosing regimen were modified as follows: from 100 mg/day to 160 mg/day or 75 mg x2/day. Patients enrolled in this have a median follow up of at least 6 months after the analysis. Results: Between January 2012 and February 2014, 77 patients with Philadelphia-negative MPN were included. 53 were treated with aspirin 75 mg/day (69%) and 24 with aspirin 100 mg/day (31%). Out of the 53 patients treated with aspirin 75 mg/day, 12 patients (23%) were resistant to aspirin. Resistance to aspirin was not correlated to sex, age, presentation, JAK2 status, treatment, history of thrombotic or bleeding and hematologic values (see following table). Table 1:The MPN Grade 1 Fibrosis PhenotypePMF (N:33)PV/ET (N:58)Total (N:91)Median age63 yrs55 yrs58 yrsSex1:31:11:2JAK V617F +16/33: (49%)40/58: (69%)56/91: (62%)Median Hgb(g/dL)11.9 (range 7.9-16.4)12.5 (range 8.0-19.8)12.2 (range 7.9-19.8)Median WBC(X10 (9))26.9 (range 1.3-188)8.9 (range 3.5-51.3)10.8 (range 1.3-188 )Median Platelet(X10 (9))179 (range 18.0-1194)505 (range 67-2286)370 (range 18-2286)Leukoerythroblastic Blood Smear15/33 (45%)PV: 17 ET: 5 Total: 22/58 (38%)37/91 (41%)Splenomegaly(cm below costal margin)18/33: (55%) Median: 1025/58: (43%) Median: 643/91(48%) Median: 4Transfusion dependence6/33: (18%)2/58: (4%)8/91: (9%)Presence Of ³ 1 symptom 17/33: (52%)33/58: (57%)50/91: (55%)DIPSS risk intermediate 2 or higher13/33: (39%)17/58: (29%)30/91: (33%)2 or more prior therapies9/33: (27%)36/58: (62%)45/91: (49%)Vital Status (Alive)25/33: (76%)53/58: (91%)78/90: (86%)Median follow up time (yrs)1.9 (range 0.1-9.8)5.7 (range 0-34.5)3.1 (range 0-34.5) An increased dose of 100 mg/day for at least 7 days overcame this biological resistance in 8 out of 8 re-tested patients. The 2 remaining 75mg resistant patients received an increased dose of aspirin but were not retested. Out of the 24 patients under 100 mg/day, only two (8%) were resistant to aspirin. In these patients, increasing the dose to 160 mg/day did not modify their biological response. However a 75 mgx2 /day was effective to overcome biological resistance. No thrombotic or bleeding event was observed during the 6-month follow-up regardless of the aspirin dose. Conclusions: This is the first study to measure in standardized conditions the biological resistance of aspirin in 77 well-characterized Philadelphia-negative MPN patients. Although this does not reach statistical significance a higher proportion of patients treated with aspirin 75mg/day was resistant to anti-platelet therapy compared to patients treated with aspirin 100 mg/day. Increasing the dose of aspirin from 75 to 100 mg once daily overcomes this biological resistance without increasing bleeding side effects and seems the best compromise. Interestingly in rare cases of extreme resistance to 100mg doubling the dose twice daily (75mgX2) was better than an increase of 160mg in one take. This pilot study on a small number of patients with a limited 6-month follow up compared to the low annual incidence of thromboses in these MPNs will be followed by a prospective study on a larger number of patients with an extended follow-up period to determine if biological resistance to aspirin is correlated to the occurrence of thrombotic events. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1941-1941
    Abstract: Myeloproliferative neoplasms (MPN) are associated with an increased risk of arterial and venous thrombosis. Besides clinical factors (previous history of thrombosis, age 〉 60 years, cardiovascular risk factors), procoagulant phenotype, proadhesive properties of the endothelium and secretion of inflammatory cytokines, contribute to the pathogenesis of thrombosis in MPN. Pegylated interferon alpha (IFN) has obviously a great therapeutic efficacy on hematopoietic cell proliferation but its other biological effects particularly on hemostasis and inflammation have not been determined. The aim of the study was to determine whether IFN impacts the biological profile including endothelial and platelet markers compared to MPN patients treated by hydroxyurea (HU) and non-treated (NT) patients. Patients with polycythemia vera (PV) or essential thrombocythemia (ET) treated with IFN or HU or without any cytostatic drug were included. Platelet membrane glycoproteins and platelet activation were measured by flow cytometry. We evaluated shear-induced platelet aggregation (SIPA) at 4000 sec-1 that mimics stenotic arteries. We measured coagulation proteins by their activity and endothelial parameters (von Willebrand factor (VWF) antigen and activity, soluble thrombomodulin). Thrombin generation was measured in platelet poor plasma and platelet rich plasma. Platelet accumulation on immobilized collagen was measured using whole blood perfused at 1500 sec-1 in flow chambers. Statistical analysis was performed by ANOVA to compare the 3 groups of patients (p 〈 0.05) and post-test Newman-Keuls to compare groups side-by-side (p 〈 0.01). Eighty-eight patients were included: 28 treated by IFN (20 PV and 8 ET), 38 treated by HU (27 PV and 11 ET) and 22 NT (6 PV and 16 ET). All patients were treated with aspirin 75 or 100 mg/day. Prior history of thrombosis occurred in 25.7% of ET and in 31.5% of PV. During the follow up ( 〉 6months), only 1 arterial and 2 venous thrombosis were observed. Hematological parameters and JAK2 V617F status was in the expected range in ET and PV patients. Surprisingly, we observed some significant effect of IFN on 1) platelets with a decreased GPVI expression compared to HU- and non-treated patients and increased levels of SIPA at 4000 sec-1. 2) endothelial cells with significantly increased levels of Willebrand antigen and activity. 3) coagulation parameters with decreased protein S activity and increased levels of factor VIII:C and fibrinogen (table 1). All the other parameters tested were similar in the 3 groups of patients. We had the opportunity to test 10 patients at least 6 months after IFN discontinuation. IFN was stopped for side effects in 6 patients, for complete molecular response in 2 and for normalization of hematological parameters in 2 others. GPVI expression, SIPA, VWF activity and antigen, protein S activity, FVIIIC and fibrinogen returned to levels similar to those of non-treated patients and were significantly different from the levels in IFN-treated patients (table 2). These results indicate that IFN was responsible for these biological modifications. In conclusion, these unexpected data showing an increase of procoagulant and inflammatory markers in IFN-treated MPN patients could be considered as a part of IFN biological activity. Though minimal, these alterations could require more intensive preventive anticoagulation than aspirin in patients with heterozygous FV Leiden or FII G20210A mutation. Table 1 comparison of biological parameters in the 3 groups of patients. *p 〈 0.01 compared to NT Table 1. comparison of biological parameters in the 3 groups of patients. *p 〈 0.01 compared to NT Table 2 comparison of biological parameters before and after IFN discontinuation in 10 patients. *Wilcoxon test p 〈 0.05 Table 2. comparison of biological parameters before and after IFN discontinuation in 10 patients. *Wilcoxon test p 〈 0.05 Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 7 ( 2014-08-14), p. 1136-1145
    Abstract: Mice constitutively developing a JAK2V617F-induced PV exhibit a bleeding tendency combined with the accelerated formation of unstable clots. Hemostatic defects are not concomitant with JAK2V617F expression, suggesting they are not directly caused by the mutation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 126, No. 8 ( 2015-08-20), p. 1017-1026
    Abstract: GPVI-dependent platelet binding and activation contribute to seal neutrophil-induced vascular damage in IC-mediated inflammation. Inflammation represents an uncommon hemostatic situation in which adhesion and activation of single platelets prevent bleeding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancers, MDPI AG, Vol. 12, No. 4 ( 2020-04-17), p. 992-
    Abstract: Myeloproliferative neoplasms (MPN) are associated with an increased risk of arterial and venous thrombosis. Pegylated-interferon alpha (IFN) and hydroxyurea (HU) are commonly used to treat MPN, but their effect on hemostasis has not yet been studied. The aim of our study was to determine whether IFN and HU impact the biological hemostatic profile of MPN patients by studying markers of endothelial, platelet, and coagulation activation. A total of 85 patients (50 polycythemia vera and 35 essential thrombocythemia) were included: 28 treated with IFN, 35 with HU, and 22 with no cytoreductive drug (non-treated, NT). Von Willebrand factor, shear-induced platelet aggregation, factor VIII coagulant activity (FVIII:C), fibrinogen, and thrombin generation with and without exogenous thrombomodulin were significantly higher in IFN-treated patients compared to NT patients, while protein S anticoagulant activity was lower. In 10 patients in whom IFN therapy was discontinued, these hemostatic biomarkers returned to the values observed in NT patients, strongly suggesting an impact of IFN therapy on endothelial and coagulation activation. Overall, our study shows that treatment with IFN is associated with significant and reversible effects on the biological hemostatic profile of MPN patients. Whether they could be associated with an increased thrombotic risk remains to be determined in further randomized clinical studies.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages