scholarly journals Platelet-derived microparticles associate with fibrin during thrombosis

Blood ◽  
1996 ◽  
Vol 87 (11) ◽  
pp. 4651-4663 ◽  
Author(s):  
P Siljander ◽  
O Carpen ◽  
R Lassila

Platelet-derived microparticles (MP) are reported to express both pro- and anticoagulant activities. Nevertheless, their functional significance has remained unresolved. The present study monitored the generation and fate of MP in an experimental model of thrombosis with costimulation of platelets by collagen and thrombin. When minimally anticoagulated (0.5 micromol/L PPACK) blood was perfused over immobilized fibrillar type I collagen in a flow chamber at a low shear rate (300 s(-1)), endogenous thrombin was generated, as evidenced by thrombin-antithrombin III complex. In contrast to full anticoagulation 150 micromol/L PPACK) and the absence of collagen, large platelet aggregates and fibrin ensued during perfusions over collagen in the presence of thrombin. In these thrombi, MP, defined as GPIIbIIIa- and P- selectin-positive vesicles (<1 micron), were found to align fibrin in immunofluorescence and scanning immunoelectron microscopy. Moreover, in sections of embolectomized thromboemboli from patients GPIIbIIIa- and P- selectin-positive material compatible with MP was detected in a fibrin strand-like pattern. In vitro binding studies showed that MP bound to fibrin and acted there as procoagulants. In summary, we show that MP generated during thrombus formation associate with local fibrin. This adhesive function fibrin could imply a sustained modulatory role for MP in evolving thrombi.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3964-3964
Author(s):  
Volker R. Stoldt ◽  
Jan Peveling ◽  
Robert Loncar ◽  
Andreas Beck ◽  
Volker Aurich ◽  
...  

Abstract We have used confocal laser microscopy and a novel “voxel”-based imaging software to study the dynamics of platelet aggregation and thrombus formation when anticoagulated blood was perfused over collagen-coated surfaces at shear rates simulating arterial flow. The objective was to evaluate the three-dimensional growth of platelet thrombi over time (“4-D” imaging). Blood from healthy donors, anticoagulated with either PPACK (80 μM) or, depending on the type of experiment, with trisodium citrate (11 mM), was incubated with mepacrine (10 μM) to render platelets fluorescent. Blood was aspirated with a syringe pump through a rectangular perfusion chamber (flow path height of 80 μm) at a flow rate of 160 or 480 μl per min to provide initial shear rates of 500 or 1,500 sec−1, respectively. Prior to perfusion, glass coverslips were coated with fibrillar type I collagen (Roche Diagnostics, Mannheim, Germany) prepared in 0.5 M acedic acid, pH 2.8, and blocked with 2 % BSA. The chamber was mounted on a Zeiss Axiovert 100M/LSM 510 invert laser scanning confocal microscope (Carl Zeiss, Oberkochem, Germany). Upon perfusion, a series of stacks, i.e. 30 confocal optical sections, from the bottom to the apex of the forming platelet aggregate or thrombus, were obtained every 25 sec with a 488-nm laser and a scanning time of &lt; 500 msec on an area of 26,450 μm2. Images corresponding to an area of 0.202 μm2 were analyzed by a “voxel”-based procedure, whereby a voxel is defined by a volume of 0.202 μm3 (0.45 μm x 0.45 μm x 1 μm). For calibration, fluorescent beads (Invitrogen, Carlsbad, CA, USA) were used, and the volume coresponding to a 1.0 μm thick stack was calculated pursuant to the voxel technique. A threshold was applied to distinguish adherent platelets from the background. Using these procedures, a uniform profile of thrombus formation and volume was observed (n=7). With citrate anticoagulated blood at an initial shear rate of 500 sec−1, thrombus growth begun after a lag phase of 220 sec, and, after 420 sec, thrombus volume reached a maximum (mean ± SD, 5x104 ± 4.9x103 μm3). Thrombus progression occurred in a two-step way with an apical growth (height extension) at the interval of 220 and 300 sec, and a further growth in the plane section at the interval of 300 and 420 sec after perfusion. Prolonged perfusion resulted in markedly abnormal flow pattern due to thrombus growth and increased shear rates. Again at an initial shear rate of 500 sec−1, platelet aggregate formation and thrombus progression were completely suppressed in the presence of anti-αIIbβ3 antibody (abciximab, 4 μg/ml). Interestingly, the polymorphism (HPA-1, PlA) of the β subunit of αIIbβ3 had a dramatic effect on thrombus growth. Thus, when comparing blood from homozygous carriers of HPA-1b (n=8) and HPA-1a (n=8), thrombus formation and progression occurred more rapidly with HPA-1b than with HPA-1a platelets, resulting in significantly larger thrombi from HPA-1b than from HPA-1a individuals (p=0.001). In conclusion, the voxel-based determination of thrombus formation and progression in vitro provides an appropriate technique to assess volumina of thrombi. Moreover, this technique can detect phenotypic differences related to an αIIbβ3 polymorphism which is postulated to modulate platelet thrombogenicity.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 383-383
Author(s):  
Shuju Feng ◽  
Xin Lu ◽  
Michael H. Kroll

Abstract von Willebrand factor (VWF) binding to platelet glycoprotein (Gp) Ib-IX-V triggers platelet activation. Under conditions of pathologically elevated arterial wall shear stress, VWF-dependent platelet adhesion is coupled to aggregation and thrombus formation principally through GpIbα-induced signaling to α IIbβ 3. To elucidate the mechanism of GpIbα signaling to α IIbβ 3, we have examined molecular interactions involving structural proteins that bind to the cytoplasmic domains of GpIbα and β 3. In CHO cells co-expressing human GpIb-IX and α IIbβ 3, the activation of α IIbβ 3 as reported by monoclonal antibody PAC-1 binding is stimulated by ristocetin (1 mg/ml) + purified human VWF(5 μg/ml). When filamin binding to the cytoplasmic domain of GpIbα is eliminated by deleting GpIbα residues 560-570, PAC-1 binding is eliminated. When human platelets in reconstituted whole blood are treated with a peptide that interferes with filamin binding to GpIbα (as reported in Blood2003;102:2122–2129), shear-dependent (1500 sec−1 shear rate or 60 dynes/cm2 shear stress) platelet deposition onto bovine type I collagen is inhibited. In washed resting platelets and platelets activated by 120 dynes/cm2 shear stress, filamin co-immunoprecipitates with both GpIbα and α IIbβ 3; only its association with β 3 is eliminated by DNaseI (1 mg/ml), demonstrating that filamin binds indirectly to α IIbβ 3 through other cytoskeletal elements. One such element is observed to be talin, which co-immunoprecipitates with filamin, α IIbβ 3 and small amounts of GpIbα in resting platelets. When platelets are sheared for two minutes at 120 dynes/cm2, talin’s DNaseI-resistant association with filamin is decreased and its DNaseI-sensitive association with α IIbβ 3 is increased. These changes are prevented when shear-dependent VWF binding to GpIb-IX-V is blocked by monoclonal antibody AK2. Shear-dependent VWF binding to GpIb-IX-V also results in the proteolysis of talin, which is considered to be one mechanism by which the N-terminal head domain of talin regulates α IIbβ 3 activation. Shear-dependent talin proteolysis is not affected by blocking VWF binding to α IIbβ 3 with a RGD peptide. These results demonstrate that the cytoplasmic domain of GpIbα transduces signals to activate α IIbβ 3 through its interactions with filamin. These signals depend only on VWF binding to GpIb-IX-V. Under pathologically elevated wall shear stress in vitro, the mechanism of signaling may be the release of talin by filamin, thus permitting the proteolysis of talin and enhancing talin’s interaction with α IIbβ 3.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 141-141 ◽  
Author(s):  
Catherine C Smith ◽  
Elisabeth Lasater ◽  
Melissa Mccreery ◽  
Kimberly Lin ◽  
Whitney Stewart ◽  
...  

Abstract Abstract 141 Background: The clinically active FLT3 inhibitor AC220 is vulnerable in vitro to resistance-conferring mutations at 3 residues in the FLT3-ITD kinase domain: the “gatekeeper” residue F691, and activation loop (AL) residues D835 and Y842. Mutations at 2 of these residues, F691 and D835, were identified in 8/8 FLT3-ITD+ acute myeloid leukemia (AML) patients who relapsed on AC220, including mutations at residue D835 in 6/8 patients. A molecular docking analysis suggests that mutations at D835 favor the active “DFG-in” kinase conformation and thereby impair binding of AC220, which presumably requires an inactive conformation for binding (Smith, et al, Nature 2012). Therefore, we predict that a type I inhibitor capable of binding the “DFG-in” active conformation of FLT3 will be required to inhibit AC220-resistant FLT3-ITD AL mutants. Crenolanib (CP-868,596) is a potent, selective ATP-competitive inhibitor of the FLT3-related receptor tyrosine kinases PDGFR-a and -b. Notably, crenolanib retains activity against the imatinib-resistant PDGFR-a D842V mutation, which is analogous to the AC220-resistant FLT3-ITD/D835V mutation. Low micromolar concentrations of crenolanib have been safely achieved in a phase I study of solid tumor patients with a half-life of ∼14 hours (Lewis et al, JCO 2009). We hypothesized that crenolanib may be a Type I inhibitor of FLT3 that retains activity against FLT3 mutant isoforms, including AC220-resistant FLT3 AL mutants, which are highly cross-resistant to multiple FLT3 TKIs. Results: In vitro binding studies demonstrate that crenolanib binds preferentially to the phosphorylated form of ABL (Kd =140nM vs Kd=440nM for non-phosphorylated ABL), confirming crenolanib is a Type I inhibitor. Additionally, crenolanib potently binds native FLT3 in vitro (Kd=0.26nM) and retains affinity for FLT3 harboring substitutions at D835 (H/V/Y; Kd= 0.24, 0.048 and 0.26nM respectively). Crenolanib demonstrates substantially more potent in vitro binding affinity for the compound FLT3-ITD/D835V mutant than AC220 (Kd=0.05nM vs Kd=210nM). In cellular assays, crenolanib induces apoptosis and inhibits the proliferation of the patient-derived FLT3-ITD+ cell lines MV4;11 and Molm14 with an inhibitory concentration 50 (IC50) of 5.2 and 9nM, respectively. FLT3 autophosphorylation and downstream signaling in MV4;11 and Molm14 cells were inhibited at similar concentrations. Treatment with crenolanib prolonged survival in a murine bone marrow transplant model of FLT3-ITD+ leukemia. Crenolanib inhibits the proliferation of BaF/3 cells transformed with FLT3-ITD (IC50 7.8 nM), and retains activity in BaF/3 cells harboring highly AC220-resistant FLT3-ITD/D835V/Y/F and FLT3-ITD/Y842C/H mutants (IC50 15–19nM). Crenolanib also potently suppresses the growth of BaF/3 cell lines containing the FLT3-activating point mutations D835V and D835Y in the absence of ITD (IC50 3.1nM), which we have recently found to be associated with AC220 resistance (Smith et al, ASH 2012, submitted). The FLT3-ITD/F691L mutation confers modest resistance to crenolanib (IC50 49.7nM). Western blot analysis reveals dose-dependent decrease in FLT3 autophosphorylation and downstream signaling. Crenolanib potently inhibits the proliferation of an AC220-resistant Molm14 subclone that harbors a D835Y mutation (IC50 15.4nM). In these cells and native Molm14 cells, crenolanib appears to retain maximal biochemical inhibition of FLT3 autophosphorylation and downstream signaling at nanomolar concentrations in human plasma, indicating relatively low plasma protein binding. Finally, treatment with crenolanib inhibited FLT3 autophosphorylation in human primary FLT3-ITD+ AML cells, including those from a patient who developed resistance to AC220 associated with a D835 mutation. Conclusions: Crenolanib is a Type I inhibitor of FLT3 that retains activity in the low nanomolar range against native and AC220-resistant FLT3-ITD mutant isoforms in in vitro binding studies, cell line and murine leukemia models, as well as in primary human AML cells. Crenolanib therefore has the potential to be clinically active in AML patients with activating FLT3-ITD or AL mutations, and to recapture clinical response in patients with acquired AC220-resistant kinase domain mutations. Clinical trials of crenolanib in TKI-naïve and TKI-pretreated FLT3-mutant AML are currently being planned or have recently been initiated. Disclosures: Perl: Astellas Pharmaceuticals: Consultancy. Carroll:GlaxoSmithKlein: Research Funding. Shah:Ariad: Consultancy, Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1126-1126
Author(s):  
Gasim Dobie ◽  
Daniel Man-yuen Sze ◽  
Constantine Tam ◽  
Denise Jackson

Abstract Introduction The Btk inhibitor, Ibrutinib (Imbruvica) which has proven to be efficacious in achieving remission of lymphocytosis and lymph node enlargement in B-CLL, it does have adverse side effects of bleeding, including major haemorrhages. The bleeding associated with Ibrutinib use is thought to be due to a combination of on-target Btk inhibition (as Btk is a key component of platelet GPVI signalling) as well as off targeted inhibition of other kinases including EGFR, ITK, JAK3 and Tec kinase. The major next generation Btk inhibitors in clinical development include Zanubrutinib (BGB-3111). Zanubrutinib shows improved selectivity for Btk compared with Ibrutinib, and thus may have reduced bleeding effects. Our study aims to determine in detail differential platelet effects between Ibrutinib and Zanubrutinib in human and mouse models using in vitro, exvivo and in vivo approaches. Methods Intravital microscopy was used to determine thrombus formation and growth after Btk inhibitors treatment in vitro and ex vivo using micro-slides or inside the mesenteric arterioles after injury by ferric chloride (FeCl3). Z-stack digital Axiocam mRm camera (Carl Zeiss) and Zeiss Axiovision software was used to capture images. Three dimensional (3D) deconvolved reconstructions of thrombi formed were analysed for surface coverage of platelet aggregates (μm2), thrombus height (μm) and thrombus volume (μm3). Flow cytometry analysis was also used to determine the release of agonist-induced platelet P-selectin exposure and dense granule after treatment with Btk inhibitors. Results In vitro experiments demonstrated that Btk inhibitors did not affect alpha or dense granule secretion mediated by GPCRs agonists, thrombin, PAR1 or PAR4. However, they inhibited alpha granule secretion mediated by GPVI selective agonists, CRP-XL or Rhodocytin. Ibrutinib inhibited human thrombus formation on type I collagen, fibrinogen or von Willebrand factor under arterial shear with 3 fold reduction whereas Zanubrutinib had no effect over a dose dependent range of concentrations. Ibrutinib treated PRP significantly delayed the kinetics of clot retraction at all-time points over the 2 hour time frame compared to Zanubrutinib treated and vehicle control. The studies also showed that Ibrutinib but not Zanubrutinib inhibited ex vivo human thrombus formation on type I collagen under arterial shear using B-CLL patient samples. The data demonstrated that treatment of C57BL/6 mouse whole blood with 0.5-2.0 µM of ibrutinib significantly inhibited thrombus growth on type I collagen under in vitro flow conditions whereas Zanubrutinib was comparable to the vehicle control. Consequently, pre-treatment of C57BL/6 mice with ibrutinib (10 mg/kg), but not Zanubrutinib (10 mg/kg) markedly inhibited platelet thrombus growth and formation on type I collagen under ex vivo arterial flow conditions. Intravital microscopy of vascular injury of mesenteric arterioles induced by ferric chloride (FeCl3) demonstrated that Ibrutinib (10 mg/kg), but not Zanubrutinib (10 mg/kg) inhibited in vivo murine thrombus formation and growth over time. Conclusion Btk inhibitors used in the treatment of B-cell malignancies have differential effects on platelet function and thrombosis. Zanubrutinib is superior to ibrutinib as it showed no effect on platelet thrombus formation, thus reduces risk of bleeding. Disclosures Tam: AbbVie: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Beigene: Honoraria.


Author(s):  
Arthur J. Wasserman ◽  
Kathy C. Kloos ◽  
David E. Birk

Type I collagen is the predominant collagen in the cornea with type V collagen being a quantitatively minor component. However, the content of type V collagen (10-20%) in the cornea is high when compared to other tissues containing predominantly type I collagen. The corneal stroma has a homogeneous distribution of these two collagens, however, immunochemical localization of type V collagen requires the disruption of type I collagen structure. This indicates that these collagens may be arranged as heterpolymeric fibrils. This arrangement may be responsible for the control of fibril diameter necessary for corneal transparency. The purpose of this work is to study the in vitro assembly of collagen type V and to determine whether the interactions of these collagens influence fibril morphology.


2021 ◽  
Vol 10 (14) ◽  
pp. 3141
Author(s):  
Hyerin Jung ◽  
Yeri Alice Rim ◽  
Narae Park ◽  
Yoojun Nam ◽  
Ji Hyeon Ju

Osteogenesis imperfecta (OI) is a genetic disease characterized by bone fragility and repeated fractures. The bone fragility associated with OI is caused by a defect in collagen formation due to mutation of COL1A1 or COL1A2. Current strategies for treating OI are not curative. In this study, we generated induced pluripotent stem cells (iPSCs) from OI patient-derived blood cells harboring a mutation in the COL1A1 gene. Osteoblast (OB) differentiated from OI-iPSCs showed abnormally decreased levels of type I collagen and osteogenic differentiation ability. Gene correction of the COL1A1 gene using CRISPR/Cas9 recovered the decreased type I collagen expression in OBs differentiated from OI-iPSCs. The osteogenic potential of OI-iPSCs was also recovered by the gene correction. This study suggests a new possibility of treatment and in vitro disease modeling using patient-derived iPSCs and gene editing with CRISPR/Cas9.


2021 ◽  
Vol 19 ◽  
pp. 228080002198969
Author(s):  
Min-Xia Zhang ◽  
Wan-Yi Zhao ◽  
Qing-Qing Fang ◽  
Xiao-Feng Wang ◽  
Chun-Ye Chen ◽  
...  

The present study was designed to fabricate a new chitosan-collagen sponge (CCS) for potential wound dressing applications. CCS was fabricated by a 3.0% chitosan mixture with a 1.0% type I collagen (7:3(w/w)) through freeze-drying. Then the dressing was prepared to evaluate its properties through a series of tests. The new-made dressing demonstrated its safety toward NIH3T3 cells. Furthermore, the CCS showed the significant surround inhibition zone than empty controls inoculated by E. coli and S. aureus. Moreover, the moisture rates of CCS were increased more rapidly than the collagen and blank sponge groups. The results revealed that the CCS had the characteristics of nontoxicity, biocompatibility, good antibacterial activity, and water retention. We used a full-thickness excisional wound healing model to evaluate the in vivo efficacy of the new dressing. The results showed remarkable healing at 14th day post-operation compared with injuries treated with collagen only as a negative control in addition to chitosan only. Our results suggest that the chitosan-collagen wound dressing were identified as a new promising candidate for further wound application.


1991 ◽  
Vol 274 (2) ◽  
pp. 615-617 ◽  
Author(s):  
P Kern ◽  
M Menasche ◽  
L Robert

The biosynthesis of type I, type V and type VI collagens was studied by incubation of calf corneas in vitro with [3H]proline as a marker. Pepsin-solubilized collagen types were isolated by salt fractionation and quantified by SDS/PAGE. Expressed as proportions of the total hydroxyproline solubilized, corneal stroma comprised 75% type I, 8% type V and 17% type VI collagen. The rates of [3H]proline incorporation, linear up to 24 h for each collagen type, were highest for type VI collagen and lowest for type I collagen. From pulse-chase experiments, the calculated apparent half-lives for types I, V and VI collagens were 36 h, 10 h and 6 h respectively.


2016 ◽  
Vol 12 ◽  
pp. P144-P144
Author(s):  
Zhizhen Zeng ◽  
Patricia J. Miller ◽  
Brett M. Connolly ◽  
Stacey S. O’Malley ◽  
Idriss Bennacef ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document