We previously showed that platelet aggregation and thrombus formation occurred in mice lacking both fibrinogen (Fg) and von Willebrand factor (VWF) and that plasma fibronectin (pFn) promoted thrombus growth and stability in injured arterioles in wild-type mice. To examine whether pFn is required for Fg/VWF-independent thrombosis, we generated Fg/VWF/conditional pFn triple-deficient (TKO; Cre+, Fnflox/flox, Fg/VWF−/−) mice and littermate control (Cre, Fnflox/flox, Fg/VWF−/−) mice. Surprisingly, TKO platelet aggregation was not abolished, but instead was enhanced in both heparinized platelet-rich plasma and gel-filtered platelets. This enhancement was diminished when TKO platelets were aggregated in pFn-positive control platelet-poor plasma (PPP), whereas aggregation was enhanced when control platelets were aggregated in pFn-depleted TKO PPP. The TKO platelet aggregation can be completely inhibited by our newly developed mouse anti–mouse β3 integrin antibodies but was not affected by anti–mouse GPIbα antibodies. Enhanced platelet aggregation was also observed when heparinized TKO blood was perfused in collagen-coated perfusion chambers. Using intravital microscopy, we further showed that thrombogenesis in TKO mice was enhanced in both FeCl3-injured mesenteric arterioles and laser-injured cremaster arterioles. Our data indicate that pFn is not essential for Fg/VWF-independent thrombosis and that soluble pFn is probably an important inhibitory factor for platelet aggregation.

Platelet adhesion and subsequent aggregation at the site of vascular injury are key events required for hemostasis. However, excessive platelet accumulation and thrombus formation may result in myocardial infarction or stroke, the 2 leading causes of morbidity and mortality worldwide.1,2  It has been shown that fibrinogen (Fg) and von Willebrand factor (VWF) are the 2 key molecules required for platelet adhesion and aggregation.3  Interestingly, we found that platelet aggregation and thrombus formation still occur in mice lacking Fg or VWF or both,4,5  suggesting that additional molecule(s) can promote platelet aggregation and thrombus formation.

We subsequently found that platelet fibronectin (Fn) content was markedly increased in Fg-deficient mice4  and a severe hypofibrinogenemic human patient,6  although no alteration of plasma Fn (pFn) was observed in their Fg-deficient blood. The increase in platelet Fn content was found to be due to enhanced pFn internalization via β3 integrin.7  Further experiments with pFn conditional deficient mice and Fn heterozygous mice showed that pFn promoted thrombus growth and stability in injured arterioles.8,9  These data are consistent with recent in vitro studies that showed that Fn assembly on the platelet surface,10,11  supported thrombus growth.12-15  However, the role of pFn in Fg/VWF-independent platelet aggregation and thrombus formation has not been studied. It was expected that pFn may be required for these processes.

To address this question, we generated Fg, VWF, and pFn triple-deficient (TKO) mice by breeding Fg and VWF double-deficient (Fg/VWF−/−) mice4  with pFn conditional-deficient (Fnflox/flox, Cre+/−) mice.16  pFn depletion was induced by intraperitoneal injections of polyinonic-polycytidylic acid in Cre+ TKO (Cre+, Fnflox/flox, Fg/VWF−/−) mice but not Cre control (Cre, Fnflox/flox, Fg/VWF−/−) littermates. Unexpectedly, we found that platelet aggregation and thrombus growth were not abolished, but rather they were enhanced in these TKO mice compared with their littermate controls. These data indicate that pFn is not required for, but inhibits, platelet aggregation and thrombogenesis in the absence of Fg and VWF. We also found that pFn is able to inhibit wild-type mouse platelet aggregation. We propose that soluble pFn is an important inhibitory factor in platelet aggregation; however, insoluble pFn (formed either by covalent linkage between pFn and fibrin or VWF or by self-assembly) may support thrombus growth. The possible mechanisms that control this pFn functional switching and its implications in clinical patients are discussed.

Mice

Fg and VWF double-deficient (Fg/VWF−/−) mice, pFn conditional deficient (Fnflox/flox, Cre+/−) mice, β3 integrin–deficient (β3−/−), and GPIbα-deficient (GPIbα−/−) mice were described before.4,16-18  β3−/− mice and GPIbα−/− mice were kindly provided by Dr Richard O. Hynes (Howard Hughes Medical Institute, Massachusetts Institute of Technology, Boston, MA), Dr Jerry Ware, and Dr Zaverio M. Ruggeri (The Scripps Research Institute, La Jolla, CA). Syngeneic BALB/c mice and C57BL/6J mice were purchased from The Jackson Laboratory (Bar Harbor, ME).

All mice were housed in the research vivarium of St Michael's Hospital in Toronto, and the experimental procedures were approved by the Animal Care Committee.

Generation of mouse anti–mouse β3 integrin and GPIbα antibodies

β3−/− mice and GPIbα−/− mice were backcrossed onto the BALB/c background at the Massachusetts Institute of Technology and St Michael's Hospital, respectively. Polyclonal antibodies against β3 integrin and GPIbα were generated in these mice by transfusion of syngeneic wild-type platelets as previously described.19  The hybridoma of M1 anti–β3 integrin monoclonal antibody was generated by fusion of immunized β3−/− splenocytes with Ag8.653 myeloma cells (ATCC, Manassas, VA). The effect of monoclonal antibody M1 (8 μg/mL) on platelet aggregation was determined by aggregometry as previously described.5 

Generation of Fg/VWF/pFn−/− TKO mice

The Fg/VWF/pFn−/− TKO mice were generated by breeding male Fg/VWF−/− mice with pFn conditional knockout (Fnflox/flox, Cre+/−) female mice. Because 4 genes are involved in the generation of these TKO mice, heterozygous mice generated from the first round of breeding were bred, generating Cre+/−, Fnflox/flox, Fg/VWF−/− background male mice and Cre+/−, Fnflox/flox, Fg+/−, VWF−/− female mice. The Fg heterozygous female (Cre+/−, Fnflox/flox, Fg+/−, and VWF−/−) mice were used for breeding because female Fg-deficient (Fg−/−) mice die from uterine bleeding during pregnancy.20  These mice were then bred to generate Cre+, Fnflox/flox, Fg/VWF−/− mice and Cre, Fnflox/flox, Fg/VWF−/− littermates (negative controls). pFn depletion was induced by 3 intraperitoneal injections of 250 μg of polyinonic-polycytidylic acid (polyI-polyC; Sigma-Aldrich, St Louis, MO) into both the Cre+ and Cre control mice at 2-day intervals as previously described.8,16  Mouse genotypes were detected by polymerase chain reaction and confirmed by Western blot, immunofluorescence microscopy, or both.

Platelet and plasma preparation

Gel-filtered platelets were prepared as previously described.4,5,7,21  Mice (6-8 weeks old) were anesthetized and bled from the retroorbital plexus with the use of heparin-coated glass capillary tubes. The blood was collected into a tube containing 3% ACD (1/9, vol/vol). Platelet-rich plasma (PRP) was obtained by centrifugation at 300g for 7 minutes. Platelets were then isolated from the PRP with the use of a Sepharose 2B column in PIPES buffer (PIPES 5 mM, NaCl 1.37 mM, KCl 4 mM, glucose 0.1%, pH 7.0). Platelet-poor plasma (PPP; from either citrated or heparinized PRP) was prepared by centrifugation at 1500g for 20 minutes. The PPP was further centrifuged at 10 000g for 5 minutes to remove the remaining cells.

Detection of Fn in plasma and platelets

The plasma and platelet Fn levels in Cre+ TKO and Cre control mice were determined by Western blot analysis with the use of a rabbit anti–human Fn antibody, which cross reacts with mouse Fn as previously described.8  Briefly, 1 μL of plasma samples or 1 × 108 gel-filtered platelet lysates were separated on a 7% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) gel and transferred to Immobilon-P PVDF membrane (Millipore, Bedford, MA). Fn was detected by using a 1:500 dilution of the rabbit anti-Fn antibody followed by a 1:20 000 dilution of alkaline phosphatase–conjugated goat anti–rabbit polyvalent IgG (StressGen, Victoria, BC). Membranes were developed with the use of 5-bromo-4-chloro-3-indolyl phosphate nitroblue tetrazolium purple substrate (Sigma-Aldrich) to visualize immonoreactive bands. Results were scanned using a Canoscan D2400U Photo scanner (Canon, Beijing, China). Fn depletion was determined by comparing the density of the Fn bands using Adobe Photoshop 7.0 Limited Edition (Adobe Systems, San Jose, CA).

Detection of platelet surface adhesive proteins and peripheral blood cell count

Cre+ and Cre platelets were examined for platelet surface expression of β1 integrin, β3 integrin, GPIbα, and P-selectin. Resting gel-filtered platelets (106) from Cre+ TKO and Cre mice were incubated separately with a 1:100 dilution of FITC-conjugated hamster anti–rat CD29 (anti–β1 integrin; BD PharMingen, San Jose, CA) or a 1:100 dilution of PE-conjugated rat anti–mouse CD61 (anti–β3 integrin; BD PharMingen, Missisauga, ON). GPIbα was detected by using 10 μg/mL rat anti–mouse GPIbα antibody p0p5 and subsequently incubated with a 1:100 dilution of FITC-conjugated anti–rat IgG (Sigma-Aldrich). Platelet surface P-selectin expression was detected on resting or thrombin (1 U/mL)–activated platelets after incubation with FITC-conjugated rat anti–mouse CD62P (P-selectin; BD PharMingen Canada). To examine the activation of β3 integrin, thrombin (1 U/mL)– or ADP (20 μM)–treated platelets were incubated with R-phycoerythrin–conjugated JON/A antibody (JON/A-PE; EMFRET Analytics, Eibelstadt, Germany), an antibody that recognizes only activated mouse αIIbβ3.22  All samples were analyzed by a FACScan flow cytometer (Becton Dickinson, San Jose, CA).

To detect peripheral blood cell counts in mice, 20 μL of whole blood was collected into an EDTA-coated Microvette from each polyI-polyC–treated Cre+ TKO and Cre control mouse. Peripheral blood cell counts were analyzed with the use of a HEMAVET HV950FS (Drew Scientific, Oxford, CT).

In vitro platelet aggregation

Platelets were prepared and aggregation was assayed as described previously.5,21  TKO and Cre control PRP and PPP were obtained by centrifugation of heparin (25 IU/mL) anticoagulated whole blood. Platelet concentration in PRP was adjusted to 3 × 108 platelets/mL with the use of autologous PPP, and aggregation was induced by various agonists, including 500 μM thrombin receptor activating peptide (TRAP; AYPGKF-NH2; Peptides Int, Louisville, KY), 20 μg/mL collagen (Nycomed Pharma, Ismaning, Germany), 20 μM ADP (Sigma-Aldrich), 20 μM U46619 (Cayman Chemical, Ann Arbor, MI). We also tested TKO and Cre control platelet aggregation after switched plasma from the opposite genotype. Briefly, gel-filtered TKO and Cre control platelets were suspended in PPP (3 × 108 platelets/mL) from either the same genotype or from the opposite genotype. Platelet aggregation was then induced by 500 μM TRAP. In another set of experiments, gel-filtered platelet aggregation in PIPES buffer was induced by ADP, U46619, and different doses of TRAP, murine thrombin (Sigma-Aldrich), or collagen, as previously described.5  The change in light transmission was monitored and recorded with the use of a computerized Chrono-log aggregometer (Chrono-Log, Havertown, PA) with the stir bar rate set to 1000 rpm at 37°C. The images of platelet aggregates were recorded using a digital camera (DP70; Olympus, Tokyo, Japan) under a Zeiss Axiovert 135 inverted fluorescent microscope (Zeiss, Oberkochen, Germany).

We also performed platelet aggregation after adding purified murine plasma Fn (50-200 μg/mL; Molecular Innovations, Southfield, MI), or our newly developed mouse anti–mouse β3 integrin antisera and monoclonal antibody (8 μg/mL; clone M1),23  and mouse anti–mouse GPIbα antisera. A functional blocking rat anti–mouse GPIbα monoclonal antibody (40 μg/mL; clone Xia.B2; EMFRET Analytics) was also used to test its effect on TKO platelet aggregation.

Perfusion chamber analysis

A 35-mm Petri dish was coated with acid-soluble human fibrillar type III (type X) collagen (1 mg/mL; Sigma-Aldrich) overnight at 4°C as previously described.4  A parallel plate flow chamber (Glycotech, Rockville, MD) with a silicone rubber gasket was placed on the collagen-coated Petri dish. Heparin (25 IU/mL) anticoagulated whole blood was fluorescently labeled with the use of DiOC6 (1 μM; Sigma-Aldrich) for 10 minutes at 37°C and 1 mL of the labeled blood was perfused over the collagen-coated surface under a controlled flow rate (shear rate of 500 seconds−1) with the use of a syringe pump (Harvard Apparatus, Holliston, MA). The blood was perfused for 4 minutes followed by 2 minutes of perfusion with a rinsing buffer (NaCl 130 mM, KCl 2 mM, NaHCO3 12 mM, CaCl2 2.5 mM, MgCl2 0.9 mM, glucose 5 mM, pH 7.4) at 37°C. Platelet adhesion, aggregation, and thrombus formation were monitored under Zeiss Axiovert 135 inverted fluorescent microscope (32×/0.4 NA; Zeiss), and pictures were taken using the DP70 digital camera (Olympus). Three randomly selected fields were recorded in each experiment. The microscope settings, including contrast, brightness, and magnification, were kept constant to facilitate comparison between different experiments. The images of adherent platelets were recorded, and the percentage of surface coverage by platelets was analyzed by computer (IBM IntelliStation Z Pro; IBM, New York, NY) with the use of Slidebook software (Intelligent Imaging Innovations, Denver, CO).

The ex vivo perfusion chamber thrombosis model was also performed at high shear rate (1800 seconds−1) with the use of a method previously described.24  Briefly, rectangular glass microcapillary tubes (height 0.1 × width 1.0 × length 100 mm; Vitro Dynamics, Rockaway, NJ) were coated with 100 μg/mL type I collagen fibrils (equine collagen Horm; Nycomed, Roskilde, Denmark) overnight at 4°C. Fluorescently labeled whole blood was perfused through the collagen-coated microcapillary tubes for 4 minutes. Platelet adhesion, aggregation, and thrombus formation were recorded in real time over the course of perfusion under a Zeiss Axiovert 135 inverted fluorescent microscope by a computer (IBM IntelliStation Z Pro) using the Slidebook program (Intelligent Imaging Innovations). After 4 minutes of perfusion, 3-dimensional images of thrombi were captured with the use of a confocal cell imaging system (CARV; Atto Bioscience, Rockville, MD) attached to the microscope, and volumes of thrombi were analyzed using the Slidebook program (Intelligent Imaging Innovations).

Intravital microscopy thrombosis models

Mesenteric arterial thrombosis model.

The whole process of thrombus formation in arterioles (shear rates are 1466 ± 74 seconds−1 in TKO mice and 1441 ± 62 seconds−1 in Cre littermates; P > .05) was monitored in 3- to 4-week-old Cre+ TKO and Cre control mice injected with donor-matched fluorescently labeled platelets under a Zeiss Axiovert 135-inverted fluorescent microscope (Zeiss).4,21  Injury was induced by topical application of 30 μL of 250 mM FeCl3, and the characteristics of thrombus formation were compared based on (1) number of fluorescent platelets deposited on the vessel wall during the first 3 to 5 minutes after injury, (2) time required for the formation of the first 20-μm thrombus, and (3) time to complete vessel occlusion.4,7,21,25 

Cremaster arterial thrombosis model.

To quantitatively study platelet accumulation within growing thrombi after the vascular injury in vivo, we used a laser to induce an arteriole thrombus in the cremaster muscle as previously described.21,26  Briefly, male adult mice were anesthetized, and a trachea tube was inserted to facilitate breathing. Antibodies and anesthetic reagent (pentobarbital; Abbott Laboratories, Toronto, ON; 0.05 mg/kg) were administered by a jugular vein cannula. The cremaster muscle was prepared under a dissecting microscope and superfused throughout the experiment with preheated bicarbonate-buffered saline. Platelets were labeled by injecting a rat anti–mouse CD41 antibody (Leo.A1; EMFRET Analytics; 0.1 μg/g) detected with Alexa-660–conjugated goat anti–rat IgG (Invitrogen, Carlsbad, CA; 2 mg/mL). Multiple independent upstream injuries were performed on a cremaster arteriole with the use of an Olympus BX51WI microscope with a pulsed nitrogen dye laser. A total of 20 thrombi in 2 Cre+ TKO mice and 21 thrombi in 2 Cre control mice were studied. The dynamic accumulation of fluorescently labeled platelets within the growing thrombus was captured and analyzed using Slidebook software (Intelligent Imaging Innovations).

Statistical analysis

Data are presented as mean plus or minus SEM. Statistical significance was assessed by Student unpaired t test and χ2 test.

Fg/VWF/pFn−/− triple-deficient mice have markedly decreased levels of Fn in both plasma and platelets

To examine whether pFn is the key molecule mediating Fg/VWF-independent platelet aggregation and thrombosis, we generated Cre+, Fnflox/flox, Fg/VWF−/− and Cre, Fnflox/flox, Fg/VWF−/− mice. To minimize genetic background differences, mice were delivered from Cre+/− parents, and both Cre+ and Cre littermates were injected with polyI-polyC. Depletion of the Fnflox/flox gene by Cre only occurred in Cre+, Fnflox/flox, Fg/VWF−/− mice but not in the Cre, Fnflox/flox, Fg/VWF−/− littermate controls. As shown in Figure 1A, Western blot analysis showed that the pFn in Cre+ TKO mice was decreased greater than 98.8% and was nearly undetectable after 3 injections of polyI-polyC. Absence of Fg and VWF was also confirmed by both DNA genotyping (polymerase chain reaction) and Western blot (data not shown).

Figure 1

Determination of Fn in plasma and platelets by Western blot analysis. (A) Plasma Fn was depleted in Cre+ TKO mice but not altered in the plasma of Cre littermates after polyI-polyC treatment. (B) Platelet Fn content in Cre+ TKO mice was also markedly decreased compared with Cre control platelets. Vinculin is shown as an internal loading control.

Figure 1

Determination of Fn in plasma and platelets by Western blot analysis. (A) Plasma Fn was depleted in Cre+ TKO mice but not altered in the plasma of Cre littermates after polyI-polyC treatment. (B) Platelet Fn content in Cre+ TKO mice was also markedly decreased compared with Cre control platelets. Vinculin is shown as an internal loading control.

Close modal

We previously showed that Fg is a competitive ligand of platelet β3 integrin during pFn internalization.7  In the absence of Fg or both Fg and VWF, platelet Fn content is increased 3- to 5-fold.4,6  It is therefore interesting to examine whether platelet β3 integrins, in the absence of both Fg and VWF, are still able to capture and internalize the remaining pFn in TKO mice. As shown in Figure 1B, although some residual platelet Fn is still detectable, the platelet Fn content in Cre+ mice was markedly decreased (>80%) compared with Cre littermates. The residual of platelet Fn (probably a cellular form of Fn originating from megakaryocytes) in TKO mice is similar to that we observed in pFn single-deficient mice.8,9  There is no significant difference in platelet Fn levels between Cre+ and Cre control platelets before injections of polyI-polyC (data not shown). This further showed that depletion of pFn was successful in the Cre+ mice, and platelet β3 integrins are not able to internalize the trace amounts of pFn in TKO blood even without Fg competition. Therefore, we defined the Cre+, Fnflox/flox, Fg/VWF−/− mice as triple deficient (TKO) mice and Cre, Fnflox/flox, Fg/VWF−/− littermates with normal plasma Fn levels as negative controls throughout this study.

It is notable that there was no detectable alteration in platelet content of vitronectin and thrombospondin-1 in TKO platelets compared with platelets from Cre control littermates after polyI-polyC treatment (data not shown).

No significant alteration in platelet surface adhesive molecules, platelet integrin activation, and peripheral blood cell counts were observed after depletion of pFn in Cre+ TKO mice

To exclude intrinsic changes in platelet function after depletion of pFn, we examined several major adhesion receptors on Cre+ TKO and Cre control platelets by flow cytometry. As shown in Figure 2A, the expression levels of GPIbα and β1(CD29) and β3(CD61) integrins are almost identical. We also examined P-selectin (CD62P) expression (Figure 2A) and the active conformation of αIIbβ3 integrin in resting and ADP (20 μM)– and thrombin (1 U/mL)–activated platelets (Figure 2B). No difference was found between Cre+ TKO platelets and the Cre control platelets. There is also no detectable difference in expression levels of these adhesion molecules on platelets between wild-type and TKO mice, except for P-selectin, which is decreased when Fg is absent (H.Y., S. Lang, Z. Zhai, L. Li, W. Kahr, P. Chen, C.M.S., A.R., M. Flick, J. Degen, J.F., and H.N., manuscript submitted). We did not observe significant alterations (data not shown) in peripheral blood cell counts in Cre+ TKO mice recruited in this study compared with their Cre littermates.

Figure 2

Platelet surface expression of adhesion molecules and integrin activation is similar between Cre+ TKO and Cre control platelets. (A) Platelet surface expression of adhesion molecules was detected by flow cytometry. GPIbα, β3 integrin (CD61) and β1 integrin (CD29) on Cre+ TKO platelets (gray line) and Cre control platelets (black line) were analyzed. Negative controls are shown as filled peak. For P-selectin (CD62P) expression, resting platelets (filled peak) or thrombin (1 U/mL)–stimulated Cre+ TKO (gray line) and Cre (black line) platelets were analyzed. No difference in surface expression was observed. (B) β3 Integrin activation on Cre+ TKO platelets (gray line) and Cre control platelets (black line) by thrombin (1 U/mL) and ADP (20 μM) were tested with the use of JON/A-PE antibody. There were no detectible differences in β3 integrin activation between the 2 groups of platelets. Representative data are shown from 6 independent experiments.

Figure 2

Platelet surface expression of adhesion molecules and integrin activation is similar between Cre+ TKO and Cre control platelets. (A) Platelet surface expression of adhesion molecules was detected by flow cytometry. GPIbα, β3 integrin (CD61) and β1 integrin (CD29) on Cre+ TKO platelets (gray line) and Cre control platelets (black line) were analyzed. Negative controls are shown as filled peak. For P-selectin (CD62P) expression, resting platelets (filled peak) or thrombin (1 U/mL)–stimulated Cre+ TKO (gray line) and Cre (black line) platelets were analyzed. No difference in surface expression was observed. (B) β3 Integrin activation on Cre+ TKO platelets (gray line) and Cre control platelets (black line) by thrombin (1 U/mL) and ADP (20 μM) were tested with the use of JON/A-PE antibody. There were no detectible differences in β3 integrin activation between the 2 groups of platelets. Representative data are shown from 6 independent experiments.

Close modal

Plasma Fn depletion enhanced Fg/VWF-independent platelet aggregation in vitro

Surprisingly, we found that platelet aggregation in TKO (Cre+) PRP was not abolished, but rather it was significantly enhanced compared with Cre control PRP when stimulated with TRAP (P < .05). Gel-filtered Cre+ TKO platelet aggregation in PIPES buffer was also enhanced when stimulated with either thrombin (1 U/mL; P < .05; Figure 3A) or TRAP (500 μM; data not shown). The TKO platelet aggregates were larger with fewer individual platelets remaining in the PRP or PIPES buffer (Figure 3A). The trend toward enhancement of platelet aggregation was also reproduced when various concentrations of TRAP (from 125 μM to 1 mM) and thrombin (from 0.25 IU to 5 IU) were used (data not shown; no aggregation was induced when ≤ 62.5 μM TRAP or ≤ 0.125 IU thrombin was used in this study). The observed enhanced platelet aggregation was reproducible when collagen was used to induce platelet activation (Figure 3B). However, neither ADP nor U46619 was able to induce TKO or Cre control platelet aggregation in PRP or gel-filtered platelets in PIPES buffer (Figure 3B). This is consistent with our previous study, which indicated that Fg is required for ADP-induced platelet aggregation in routine anticoagulated blood, and released platelet granule proteins are essential for Fg-independent platelet aggregation.5 

Figure 3

Plasma Fn inhibited Fg/VWF-independent platelet aggregation in vitro. (A top left) Platelet aggregation was enhanced in plasma Fn-depleted Cre+ TKO PRP compared with Cre control PRP when stimulated by 500 μM TRAP (P < .05). (Top right) Gel-filtered platelet aggregation, induced by 1 U/mL thrombin, was also enhanced in Cre+ TKO mice (P < .05). (Bottom) The Cre+ TKO platelets formed larger aggregates than did Cre control platelets when examined under a microscope. Pictures of aggregates were taken under a 10×/0.30 NA objective. (B) Platelet aggregation was induced in both Cre+ TKO PRP and gel-filtered platelets by collagen (5-10 μg/mL) but failed to be induced by ADP (20 μM) and U46619 (20 μM). No platelet aggregates were found in ADP- and U46619-treated Cre+ TKO samples when examined under a microscope (32×/0.40 NA objective). (C left) The enhancement of platelet aggregation in TKO conditions was diminished when Cre+ TKO platelets were aggregated in pFn-positive Cre control PPP in a parallel aggregation assay (top panel). (Right) The enhancement of platelet aggregation by pFn was partially restored in Cre control platelets when they were aggregated in pFn-depleted Cre+ TKO PPP. (D) Exogenous pFn significantly inhibited wild-type gel-filtered mouse platelet aggregation in a dose-dependent manner. (E top) Cre+ TKO platelet aggregation in PRP was inhibited by our newly generated mouse anti–mouse β3 integrin monoclonal antibody M1 (left). Cre+ TKO platelet aggregation with gel-filtered platelets in PIPES buffer was also inhibited by polyclonal anti-β3 integrin sera (right). (Bottom) Cre+ TKO platelet aggregation was not affected by a monoclonal antibody against GPIbα (left) and our mouse anti–mouse anti-GPIbα sera (right). All representative traces of platelet aggregation are shown from at least 3 independent experiments.

Figure 3

Plasma Fn inhibited Fg/VWF-independent platelet aggregation in vitro. (A top left) Platelet aggregation was enhanced in plasma Fn-depleted Cre+ TKO PRP compared with Cre control PRP when stimulated by 500 μM TRAP (P < .05). (Top right) Gel-filtered platelet aggregation, induced by 1 U/mL thrombin, was also enhanced in Cre+ TKO mice (P < .05). (Bottom) The Cre+ TKO platelets formed larger aggregates than did Cre control platelets when examined under a microscope. Pictures of aggregates were taken under a 10×/0.30 NA objective. (B) Platelet aggregation was induced in both Cre+ TKO PRP and gel-filtered platelets by collagen (5-10 μg/mL) but failed to be induced by ADP (20 μM) and U46619 (20 μM). No platelet aggregates were found in ADP- and U46619-treated Cre+ TKO samples when examined under a microscope (32×/0.40 NA objective). (C left) The enhancement of platelet aggregation in TKO conditions was diminished when Cre+ TKO platelets were aggregated in pFn-positive Cre control PPP in a parallel aggregation assay (top panel). (Right) The enhancement of platelet aggregation by pFn was partially restored in Cre control platelets when they were aggregated in pFn-depleted Cre+ TKO PPP. (D) Exogenous pFn significantly inhibited wild-type gel-filtered mouse platelet aggregation in a dose-dependent manner. (E top) Cre+ TKO platelet aggregation in PRP was inhibited by our newly generated mouse anti–mouse β3 integrin monoclonal antibody M1 (left). Cre+ TKO platelet aggregation with gel-filtered platelets in PIPES buffer was also inhibited by polyclonal anti-β3 integrin sera (right). (Bottom) Cre+ TKO platelet aggregation was not affected by a monoclonal antibody against GPIbα (left) and our mouse anti–mouse anti-GPIbα sera (right). All representative traces of platelet aggregation are shown from at least 3 independent experiments.

Close modal

The enhancement of platelet aggregation in TKO mice was diminished when Cre+ TKO platelets were aggregated in pFn-positive control PPP, whereas enhanced aggregation was observed when control platelets were aggregated in pFn-depleted TKO plasma (Figure 3C). These data exclude the possibility of TKO platelet hyperactivity during platelet aggregation. Only partial correction was observed with respect to the enhanced platelet aggregation in these plasma switch experiments, potentially reflecting the inhibitory effect of α-granule pFn in Cre control platelets. This internalized platelet pFn may be released from platelets and inhibit platelet aggregation, as observed in gel-filtered Cre platelet aggregation (Figure 3A). In addition to inhibiting platelet aggregation in TKO mice, we also found that exogenous pFn significantly inhibited both thrombin-induced (Figure 3D) and TRAP-induced (data not shown) gel-filtered wild-type mouse platelet aggregation in a dose-dependent manner. This is consistent with earlier studies in wild-type rat and pig and in healthy human platelets.27-30 

To examine whether β3 integrin is essential for TKO platelet aggregation, we generated mouse anti–mouse β3 integrin antisera and developed a mouse anti–mouse β3 integrin monoclonal antibody (M1) with the use of β3 integrin−/− mice. High-titer (> 1:1600) mouse anti–mouse GPIbα antisera were also generated in GPIbα−/− mice. The anti–β3 integrin antisera and M1 are specific to β3 integrin and are able to inhibit wild-type mouse platelet aggregation (data not shown). We found that TKO platelet aggregation can be completely blocked by this anti–mouse β3 integrin antisera and the monoclonal antibody M1, but it was not affected by polyclonal anti–mouse GPIbα antisera and a functional blocking rat anti–mouse GPIbα monoclonal antibody (Figure 3E) in both PRP and gel-filtered platelets in PIPES buffer. This indicates that β3 integrin, but not GPIbα, is required and that other ligands of β3 integrin may mediate Fg/VWF/pFn-independent platelet aggregation.

Plasma Fn depletion enhanced Fg/VWF-independent platelet aggregation in ex vivo laminar flow chambers

To examine platelet adhesion and aggregation under flow conditions, heparin-anticoagulated whole blood from TKO mice or Cre control mice was perfused over collagen-coated surfaces in laminar perfusion chambers. Consistent with data from in vitro platelet aggregation, significantly more platelet thrombi formed when TKO blood was perfused at a shear rate of 500 seconds−1. Platelet surface coverage was significantly higher in TKO blood (TKO, 14.50% ± 1.8%, Cre control, 6.2% ± 0.8%; P < .001; Figure 4A,). Because of the absence of VWF, platelet adhesion and aggregation at high shear (1800 seconds−1) was significantly decreased in both TKO and Cre controls, but enhancement of platelet aggregation in TKO mice was still evident (TKO, 3.9 ± 0.3, Cre control, 2.2 ± 0.3; P < .01; Figure 4B,).

Figure 4

Significantly more Cre+ TKO platelets adhered and aggregated on collagen matrices under physiologic flow conditions. Heparinized whole blood from Cre+ TKO and Cre control mice was perfused over a collagen type III surface at a shear rate of 500 seconds−1 (A; 32×/0.40 NA objective) or a collagen type I surface at a shear rate of 1800 seconds−1(B; 60×/0.90 NA water objective). The percentage of surface coverage was significantly higher in perfusion at both lower and higher shear rates in TKO whole blood compared with Cre controls. Representative pictures of platelet adhesion/aggregation in whole blood (left; n = 9).

Figure 4

Significantly more Cre+ TKO platelets adhered and aggregated on collagen matrices under physiologic flow conditions. Heparinized whole blood from Cre+ TKO and Cre control mice was perfused over a collagen type III surface at a shear rate of 500 seconds−1 (A; 32×/0.40 NA objective) or a collagen type I surface at a shear rate of 1800 seconds−1(B; 60×/0.90 NA water objective). The percentage of surface coverage was significantly higher in perfusion at both lower and higher shear rates in TKO whole blood compared with Cre controls. Representative pictures of platelet adhesion/aggregation in whole blood (left; n = 9).

Close modal

Plasma Fn depletion enhanced Fg/VWF-independent thrombus formation in vivo

To test the effects of pFn depletion on Fg/VWF−/− thrombus formation in vivo, we used 2 intravital microscopy models in different vascular beds. In the FeCl3-injury mesenteric arteriole thrombus model, we studied thrombosis in response to injury in a total of 9 Cre+ TKO mice and 10 Cre Fg/VWF−/− control mice. As shown in Figure 5A and B, the number of single fluorescently labeled adherent platelets per minute, determined at 3 to 5 minutes after vessel injury, was not significantly different between TKO and Cre control mice (Cre+ TKO, 80.75 ± 25.1 minute−1; Cre control, 61.5 ± 24.9 minute−1; P > .05). However, in TKO mice, the formation of the first thrombus greater than 20 μm in diameter was faster (Cre+ TKO, 10 ± 0.5 minutes; Cre control, 13.1 ± 1.1 minute; P < .05) and more stable, and the time required for complete vessel occlusion was significantly shorter (Cre+ TKO, 25.9 ± 2.8 minutes; Cre control, 34.1 ± 2.3 minutes; P < .05) compared with Cre controls. The thrombus did not result in vessel occlusion in 5 (50%) of 10 Cre control mice within a 40-minute recording time after vascular injury, whereas vessel occlusion occurred in 7 (78%) of 9 TKO mice, although a statistically significant difference was not reached (χ2 test = 1.569; P > .05).

Figure 5

Depletion of pFn enhanced thrombus formation in Cre+ TKO mice in a FeCl3-injury mesenteric arteriole thrombosis model. (A) No significant difference was observed in early platelet deposition (shown at 4 minutes), but thrombus growth (shown at 15 minutes) and vessel occlusion (27 minutes) were accelerated. The arrow in the Cre panel (27 minutes) shows blood flow from right to left in the arteriole. (B) Quantitative analysis of thrombus formation in Cre+ TKO mice () and Cre control (■) mice. Cre+ TKO mice, n = 9; Cre control mice, n = 10.

Figure 5

Depletion of pFn enhanced thrombus formation in Cre+ TKO mice in a FeCl3-injury mesenteric arteriole thrombosis model. (A) No significant difference was observed in early platelet deposition (shown at 4 minutes), but thrombus growth (shown at 15 minutes) and vessel occlusion (27 minutes) were accelerated. The arrow in the Cre panel (27 minutes) shows blood flow from right to left in the arteriole. (B) Quantitative analysis of thrombus formation in Cre+ TKO mice () and Cre control (■) mice. Cre+ TKO mice, n = 9; Cre control mice, n = 10.

Close modal

We also used a laser-injury cremaster muscle arteriole thrombosis model to quantitatively compare platelet accumulation within growing thrombi in TKO and Cre control mice. We found that thrombi in Cre control mice appeared fragile and unstable; frequent embolization and detachment of whole thrombi was evident. In sharp contrast, thrombi in TKO mice exhibited rapid platelet accumulation within the growing thrombus. The total amount of platelets within the Cre+ TKO thrombus was significantly greater and the rate of decrease in amount of platelets after maximal platelet deposition was lower compared with Cre control thrombi (P < .05; Figure 6).

Figure 6

Plasma Fn depletion enhanced the incorporation of platelets into arterial thrombi in the cremaster muscle arteriole thrombosis model. (A) Significantly more platelets were recruited into Cre+ TKO thrombi compared with Cre control thrombi. The kinetic curves represent platelet mean fluorescence intensity, and the shaded regions are representative of SEM. (B) The total area under the curve (AUC) was greater in Cre+ TKO compared with Cre controls. (Cre+ TKO thrombi, n = 20; Cre control thrombi, n = 21, P < .05.)

Figure 6

Plasma Fn depletion enhanced the incorporation of platelets into arterial thrombi in the cremaster muscle arteriole thrombosis model. (A) Significantly more platelets were recruited into Cre+ TKO thrombi compared with Cre control thrombi. The kinetic curves represent platelet mean fluorescence intensity, and the shaded regions are representative of SEM. (B) The total area under the curve (AUC) was greater in Cre+ TKO compared with Cre controls. (Cre+ TKO thrombi, n = 20; Cre control thrombi, n = 21, P < .05.)

Close modal

It has been suspected for decades that pFn may contribute to thrombosis and hemostasis. However, its precise role in these processes has not been well developed. We previously showed that platelet aggregation and occlusive thrombi occurred in mice lacking Fg or VWF or both.4,5  In these Fg-deficient animals and a severely hypofibrinogenemic human patient,6  platelet Fn content was markedly increased. It was speculated that pFn may be required for platelet aggregation and thrombus formation in the absence of Fg and VWF. To address this question, we generated TKO (Fg/VWF/pFn−/−) mice in the present studies. We found these mice are viable. Interestingly, TKO platelets did not fail to aggregate, but they exhibited enhanced aggregation in both PRP and gel-filtered platelets in PIPES buffer. The enhancement of platelet aggregation was reproducible in perfusion chambers at different shear stresses. Furthermore, enhanced thrombogenesis in TKO mice was shown in both FeCl3-injured mesenteric arterioles and laser-injured cremaster arterioles with the use of intravital microscopy thrombosis models. These data suggest that pFn is not required for, but inhibits Fg/VWF-independent platelet aggregation and thrombus formation.

The role of Fn in platelet aggregation is inconclusive and controversial, although pFn is a dimer that contains 2 RGD sites that could potentially cross-link adjacent platelets. Early experiments suggested that pFn may support platelet aggregation,31  and an anti-Fn monoclonal antibody decreased platelet aggregation.32  Subsequent studies, however, showed that pFn may have either no effect or an inhibitory effect on platelet aggregation.27-29  The inhibitory effect was thought to be due to pFn competition with more potent integrin ligands such as Fg and VWF.

It has been speculated that pFn may be the bridging molecule for platelet aggregation in the absence of Fg or of both Fg and VWF. In Fg/VWF−/− mice, we found that (1) platelet Fn content increased 3- to 5-fold, (2) platelet-rich thrombi still formed in vivo after vascular injury, and (3) platelet aggregation can be induced in vitro under more physiologic conditions by strong agonists such as thrombin, TRAP, collagen, A23187, and phorbol myristyl acetate,5  indicating that pFn may play a supportive role in platelet aggregation. This hypothesis was further supported by our in vivo study showing that pFn promotes thrombus growth8  and studies showing that Fn assembly on the platelet surface10,11  supports thrombus growth in ex vivo perfusion chamber experiments.12-15,33  However, there is no direct evidence indicating that pFn supports Fg/VWF-independent platelet aggregation. The question cannot be addressed simply by removing Fn from the plasma during aggregation assays because Fn released from platelets may also contribute to platelet aggregation, and greater than 80% of platelet Fn comes from pFn internalization by β3 integrin.7  TKO (Fg/VWF/pFn−/−) mice are therefore required to answer this question.

The enhancement of TKO platelet aggregation both in the presence and absence of plasma was unexpected. After blindly repeating these results several times, we asked whether adhesion molecules potentially involved in platelet aggregation were up-regulated on TKO platelets. However, this is probably not the answer because the expression levels of β3 and β1 integrins, GPIbα, and P-selectin are almost identical between TKO and Cre control platelets (Figure 2A). The active conformations of αIIbβ3 integrin, detected by a conformation sensitive monoclonal antibody JON/A, in resting and ADP- and thrombin-activated platelets are also comparable (Figure 2B). We further performed platelet aggregation after switching their plasma and found that aggregation of Cre littermate platelets was enhanced in TKO PPP, and aggregation of TKO platelets was decreased in pFn-positive plasma (Figure 3C). These results indicate that the observed enhanced platelet aggregation in TKO mice is unlikely due to an alteration in platelet surface adhesive receptors or other possible mechanisms that lead to hyperactivation of TKO platelets. Because enhanced platelet aggregation was also found in gel-filtered platelets in PIPES buffer, we therefore conclude that pFn, including internalized platelets pFn, may not be the bridging molecule required for Fg/VWF-independent platelet aggregation, but rather act as an inhibitory factor in this process.

Note that we also found that pFn inhibited wild-type gel-filtered platelet aggregation, which is consistent with earlier studies from other species, including healthy human platelets.27-30  Although the mechanism is not fully understood, it is possible that pFn may inhibit β3 integrin–mediated platelet aggregation by the same pathway as plasma vitronectin21  (ie, probably by RGD occupancy of the β3 integrin ligand binding site). This occupancy may sterically prevent the bridging ligand(s) from cross-linking adjacent platelets. In addition, it is currently unknown whether soluble pFn can also deliver negative signals to platelets or attenuate platelet activation through blocking integrin outside-in signals delivered by more potent β3 integrin ligand(s). We also cannot exclude the possibility that thrombospondin-1 is able to more efficiently block the antithrombotic activity of nitric oxide/cGMP signaling34  in the absence of pFn. These questions should be of interest for further studies.

TKO platelet aggregation was completely blocked by our newly developed mouse anti–mouse β3 integrin antisera and monoclonal antibody M1 (Figure 3E). However, aggregation was not affected by our anti–mouse GPIbα antisera and a functional blocking rat anti–mouse GPIbα monoclonal antibody. These data indicate that β3 integrin, but not GPIbα, is essential for TKO platelet aggregation. They also exclude the possibility that mutual interactions, including indirect binding, between these 2 most abundant glyco-proteins (ie, β3 integrin and GPIbα) on the platelet surface may mediate this aggregation, although engagement between β2 integrin (Mac-1) and GPIbα occurs under certain circumstances.35  Because TKO platelet aggregation cannot be induced by either ADP or U46619 (Figure 3B), other ligand(s) of β3 integrin released from platelets may mediate Fg/VWF/pFn-independent platelet aggregation. However, it is currently unknown whether EDA domain-positive cellular Fn36  (a megakaryocyte synthesized form of Fn present in platelets in a small proportion; 10%-20%) is necessary for this process. It is probably that the cellular Fn and other adhesion ligands (such as thrombospondin-1, vitronectin, and CD40L or combinations thereof)21,34,37-42  synergistically contribute to this Fg/VWF/pFn-independent platelet aggregation.

Our data from ex vivo perfusion chambers and the 2 intravital microscopy models are consistent with our results from in vitro platelet aggregation assays (Figure 3). However, the enhancement of thrombus growth in these ex vivo and in vivo models may not only be due to the enhanced platelet aggregation, but also to enhanced platelet adhesion. There are significantly more adhesive platelets in the perfusion chamber experiments (Figure 4A), and there is also a trend toward increased platelet deposition at the site of vessel wall injury at the early stages of thrombus formation (Figure 5B). It is currently unknown whether this is due to pFn inhibition of platelet interactions with either collagen,28  or to other subendothelial matrix proteins.

It is interesting that our results from the present studies contrast with our previous studies that used conditionally deficient pFn mice (single deficiency)8  and studies of Fn assembly.11-13,15  One possible explanation is that Fg and VWF may be required for pFn to promote thrombus growth. Our recent finding that fibrin formation is important for Fn retention on the human platelet surface,6  together with findings from perfusion chamber studies that thrombogenesis is enhanced by Fn assembly in platelet thrombi by cross-linking to fibrin,13  supports this explanation. It is currently unclear whether Fg or VWF or both (in plasma or that released from activated platelets) are required for Fn assembly on aggregated platelets in vivo. However, it is likely that fibrin and subsequent covalently linked pFn-fibrin matrix formation at the site of vascular injury may promote thrombus growth. This may explain why impaired thrombus formation was observed in pFn single-deficient mice with the use of the intravital microscopy model8  because significant thrombin and fibrin were generated after vascular injury in the system.4,7,21,25  The levels of severity of vascular injury, local concentration of thrombin, as well as other enzymes, including factor XIIIa and the possible protein disulfide isomerases (PDIs)43,44  that enhance insoluble pFn matrix formation, may contribute to switching the function of pFn from an inhibitory role in platelet aggregation to a supportive effect on thrombus growth. The intrinsic PDI activities of both VWF propolypetide and pFn may also facilitate pFn insolubilization.45,46  Understanding the mechanisms of this pFn functional “switch” may provide insights into the elevated pFn levels in patients with acute myocardial infarction and venous thromboembolism47-51  and elucidate whether increasing pFn is a potential protective mechanism in these common and critical human diseases.

Another challenging question is whether increased platelet Fn content in Fg-deficient platelets plays a supportive role in hemostasis. Here, we found that pFn, including platelet pFn internalized from the plasma, inhibits platelet aggregation (an important hemostatic event). However, it is possible that high amounts of Fn released from adherent platelets at the site of vascular injury may dramatically boost the local pFn concentration, which facilitates Fn self-and nonself assembly on the injured vessel wall. These pFn-mediated extracellular matrix-like fibrils may significantly contribute to hemostasis.

In summary, we generated Fg/VWF/pFn−/− triple-deficient mice. We clearly show that pFn is unlikely the key molecule that mediates Fg/VWF-independent platelet aggregation. In contrast, soluble pFn is a significant inhibitory factor during thrombus formation. We propose that pFn may play dual roles in thrombosis and hemostasis. By switching its soluble form (inhibitory) to insoluble extracellular matrix-like fibrils (supportive) by either self-assembly or nonself covalent linkage with fibrin or other matrix proteins, pFn sophisticatedly controls the vital physiologic and pathologic processes (ie, hemostasis and thrombosis).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Drs Reinhard Fässler, Jay L. Degen, Denisa D. Wagner, and Cecile V. Denis for their earlier work preparing pFn conditional knockout mice, Fg and VWF gene-deficient mice. We thank Dr Pingguo Chen for his assistance with genotyping and backcrossing GPIbα−/− and β3−/− mice onto BALB/c background and Michelle Lee Webster and Sean Lang for assistance with preparation of the manuscript.

This work was supported by the Heart and Stroke Foundation of Canada (Ontario), Canadian Blood Services and Canadian Institutes of Health Research, and by equipment funds from St Michael's Hospital, Canadian Blood Services, and Canada Foundation for Innovation. H.Y. is a recipient of the Heart and Stroke/Richard Lewar Excellence Award and a Canadian Blood Services postdoctoral fellowship award.

Contribution: A.R. designed the experiments, performed the research, analyzed the data, and wrote part of the manuscript; H.Y., G.Z., W.J., F.H., and X.B. performed the research; C.M.S. performed the research and edited the manuscript; P.L.G. analyzed the data and edited the manuscript; J.F. contributed a vital analytical tool (flow cytometer), analyzed the data, and edited the manuscript; and H.N. is the principal investigator who designed the experiments, analyzed the data, and wrote the manuscript.

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

Correspondence: Heyu Ni, Canadian Blood Services and Department of Laboratory Medicine and Pathobiology, St Michael's Hospital, University of Toronto, 30 Bond Street, Room 2-006, Bond Wing, Toronto, ON, Canada M5B 1W8; e-mail: nih@smh.toronto.on.ca.

1
Ruggeri
 
ZM
Platelets in atherothrombosis.
Nat Med
2002
, vol. 
8
 (pg. 
1227
-
1234
)
2
Sachs
 
UJ
Nieswandt
 
B
In vivo thrombus formation in murine models.
Circ Res
2007
, vol. 
100
 (pg. 
979
-
991
)
3
Ruggeri
 
ZM
Mechanisms initiating platelet thrombus formation.
Thromb Haemost
1997
, vol. 
78
 (pg. 
611
-
616
)
4
Ni
 
H
Denis
 
CV
Subbarao
 
S
, et al. 
Persistence of platelet thrombus formation in arterioles of mice lacking both von Willebrand factor and fibrinogen.
J Clin Invest
2000
, vol. 
106
 (pg. 
385
-
392
)
5
Yang
 
H
Reheman
 
A
Chen
 
P
, et al. 
Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
2230
-
2237
)
6
Zhai
 
Z
Wu
 
J
Xu
 
X
, et al. 
Fibrinogen controls human platelet fibronectin internalization and cell-surface retention.
J Thromb Haemost
2007
, vol. 
5
 (pg. 
1740
-
1746
)
7
Ni
 
H
Papalia
 
JM
Degen
 
JL
Wagner
 
DD
Control of thrombus embolization and fibronectin internalization by integrin alpha IIb beta 3 engagement of the fibrinogen gamma chain.
Blood
2003
, vol. 
102
 (pg. 
3609
-
3614
)
8
Ni
 
H
Yuen
 
PS
Papalia
 
JM
, et al. 
Plasma fibronectin promotes thrombus growth and stability in injured arterioles.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
2415
-
2419
)
9
Matuskova
 
J
Chauhan
 
AK
Cambien
 
B
, et al. 
Decreased plasma fibronectin leads to delayed thrombus growth in injured arterioles.
Arterioscler Thromb Vasc Biol
2006
, vol. 
26
 (pg. 
1391
-
1396
)
10
Olorundare
 
OE
Peyruchaud
 
O
Albrecht
 
RM
Mosher
 
DF
Assembly of a fibronectin matrix by adherent platelets stimulated by lysophosphatidic acid and other agonists.
Blood
2001
, vol. 
98
 (pg. 
117
-
124
)
11
Cho
 
J
Degen
 
JL
Coller
 
BS
Mosher
 
DF
Fibrin but not adsorbed fibrinogen supports fibronectin assembly by spread platelets. Effects of the interaction of alphaIIb beta3 with the C terminus of the fibrinogen gamma-chain.
J Biol Chem
2005
, vol. 
280
 (pg. 
35490
-
35498
)
12
Cho
 
J
Mosher
 
DF
Impact of fibronectin assembly on platelet thrombus formation in response to type I collagen and von Willebrand factor.
Blood
2006
, vol. 
108
 (pg. 
2229
-
2236
)
13
Cho
 
J
Mosher
 
DF
Enhancement of thrombogenesis by plasma fibronectin cross-linked to fibrin and assembled in platelet thrombi.
Blood
2006
, vol. 
107
 (pg. 
3555
-
3563
)
14
Ni
 
H
Unveiling the new face of fibronectin in thrombosis and hemostasis.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
940
-
942
)
15
Cho
 
J
Mosher
 
DF
Role of fibronectin assembly in platelet thrombus formation.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
1461
-
1469
)
16
Sakai
 
T
Johnson
 
KJ
Murozono
 
M
, et al. 
Plasma fibronectin supports neuronal survival and reduces brain injury following transient focal cerebral ischemia but is not essential for skin-wound healing and hemostasis.
Nat Med
2001
, vol. 
7
 (pg. 
324
-
330
)
17
Hodivala-Dilke
 
KM
McHugh
 
KP
Tsakiris
 
DA
, et al. 
Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival.
J Clin Invest
1999
, vol. 
103
 (pg. 
229
-
238
)
18
Ware
 
J
Russell
 
S
Ruggeri
 
ZM
Generation and rescue of a murine model of platelet dysfunction: the Bernard-Soulier syndrome.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 (pg. 
2803
-
2808
)
19
Ni
 
H
Chen
 
P
Spring
 
CM
, et al. 
A novel murine model of fetal and neonatal alloimmune thrombocytopenia: response to intravenous IgG therapy.
Blood
2006
, vol. 
107
 (pg. 
2976
-
2983
)
20
Suh
 
TT
Holmback
 
K
Jensen
 
NJ
, et al. 
Resolution of spontaneous bleeding events but failure of pregnancy in fibrinogen-deficient mice.
Genes Dev
1995
, vol. 
9
 (pg. 
2020
-
2033
)
21
Reheman
 
A
Gross
 
P
Yang
 
H
, et al. 
Vitronectin stabilizes thrombi and vessel occlusion but plays a dual role in platelet aggregation.
J Thromb Haemost
2005
, vol. 
3
 (pg. 
875
-
883
)
22
Bergmeier
 
W
Schulte
 
V
Brockhoff
 
G
Bier
 
U
Zirngibl
 
H
Nieswandt
 
B
Flow cytometric detection of activated mouse integrin alphaIIbbeta3 with a novel monoclonal antibody.
Cytometry
2002
, vol. 
48
 (pg. 
80
-
86
)
23
Zhu
 
G
Webster
 
M
Reheman
 
A
, et al. 
Novel mouse anti-mouse beta 3 integrin monoclonal antibodies: development and characterization of new reagents for research in thrombosis and thrombocytopenia [abstract].
Blood
2007
, vol. 
118
 pg. 
627a
 
24
Jones
 
KL
Hughan
 
SC
Dopheide
 
SM
Farndale
 
RW
Jackson
 
SP
Jackson
 
DE
Platelet endothelial cell adhesion molecule-1 is a negative regulator of platelet-collagen interactions.
Blood
2001
, vol. 
98
 (pg. 
1456
-
1463
)
25
Ni
 
H
Ramakrishnan
 
V
Ruggeri
 
ZM
Papalia
 
JM
Phillips
 
DR
Wagner
 
DD
Increased thrombogenesis and embolus formation in mice lacking glycoprotein V.
Blood
2001
, vol. 
98
 (pg. 
368
-
373
)
26
Falati
 
S
Gross
 
P
Merrill-Skoloff
 
G
Furie
 
BC
Furie
 
B
Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse.
Nat Med
2002
, vol. 
8
 (pg. 
1175
-
1181
)
27
Santoro
 
SA
Inhibition of platelet aggregation by fibronectin.
Biochem Biophys Res Commun
1983
, vol. 
116
 (pg. 
135
-
140
)
28
Moon
 
DG
Kaplan
 
JE
Mazurkewicz
 
JE
The inhibitory effect of plasma fibronectin on collagen-induced platelet aggregation.
Blood
1986
, vol. 
67
 (pg. 
450
-
457
)
29
Hynes
 
R
Fibronectin
1990
New York, NY
Springer Verlag
pg. 
546
 
30
Moon
 
DG
Matayoshi
 
BM
Weston
 
LK
Minnear
 
FL
Kaplan
 
JE
Fibronectin inhibition of platelet thrombus formation in an in vivo porcine model of vascular injury.
Thromb Res
1994
, vol. 
76
 (pg. 
343
-
351
)
31
Arneson
 
MA
Hammerschmidt
 
DE
Furcht
 
LT
King
 
RA
A new form of Ehlers-Danlos syndrome. Fibronectin corrects defective platelet function.
JAMA
1980
, vol. 
244
 (pg. 
144
-
147
)
32
Dixit
 
VM
Haverstick
 
DM
O'Rourke
 
K
, et al. 
Inhibition of platelet aggregation by a monoclonal antibody against human fibronectin.
Proc Natl Acad Sci U S A
1985
, vol. 
82
 (pg. 
3844
-
3848
)
33
Cho
 
J
Mosher
 
DF
Characterization of fibronectin assembly by platelets adherent to adsorbed laminin-111.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
943
-
951
)
34
Isenberg
 
JS
Romeo
 
MJ
Yu
 
C
, et al. 
Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling.
Blood
2008
, vol. 
111
 (pg. 
613
-
623
)
35
Hoffmeister
 
KM
Felbinger
 
TW
Falet
 
H
, et al. 
The clearance mechanism of chilled blood platelets.
Cell
2003
, vol. 
112
 (pg. 
87
-
97
)
36
Chauhan
 
AK
Kisucka
 
J
Cozzi
 
MR
, et al. 
Prothrombotic effects of fibronectin isoforms containing the EDA domain.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
296
-
301
)
37
Ni
 
H
Freedman
 
J
Platelets in hemostasis and thrombosis: role of integrins and their ligands.
Transfus Apher Sci
2003
, vol. 
28
 (pg. 
257
-
264
)
38
Jackson
 
SP
The growing complexity of platelet aggregation.
Blood
2007
, vol. 
109
 (pg. 
5087
-
5095
)
39
Bonnefoy
 
A
Hantgan
 
R
Legrand
 
C
Frojmovic
 
MM
A model of platelet aggregation involving multiple interactions of thrombospondin-1, fibrinogen, and GPIIbIIIa receptor.
J Biol Chem
2001
, vol. 
276
 (pg. 
5605
-
5612
)
40
Wu
 
YP
Bloemendal
 
HJ
Voest
 
EE
, et al. 
Fibrin-incorporated vitronectin is involved in platelet adhesion and thrombus formation through homotypic interactions with platelet-associated vitronectin.
Blood
2004
, vol. 
104
 (pg. 
1034
-
1041
)
41
Andre
 
P
Prasad
 
KS
Denis
 
CV
, et al. 
CD40L stabilizes arterial thrombi by a beta3 integrin–dependent mechanism.
Nat Med
2002
, vol. 
8
 (pg. 
247
-
252
)
42
Plow
 
EF
Cierniewski
 
CS
Xiao
 
Z
Haas
 
TA
Byzova
 
TV
AlphaIIbbeta3 and its antagonism at the new millennium.
Thromb Haemost
2001
, vol. 
86
 (pg. 
34
-
40
)
43
Chen
 
VM
Hogg
 
PJ
Allosteric disulfide bonds in thrombosis and thrombolysis.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
2533
-
2541
)
44
Cho
 
J
Furie
 
BC
Coughlin
 
SR
Furie
 
B
A critical role for extracellular protein disulfide isomerase during thrombus formation in mice.
J Clin Invest
2008
, vol. 
118
 (pg. 
1123
-
1131
)
45
Langenbach
 
KJ
Sottile
 
J
Identification of protein-disulfide isomerase activity in fibronectin.
J Biol Chem
1999
, vol. 
274
 (pg. 
7032
-
7038
)
46
Mayadas
 
TN
Wagner
 
DD
Vicinal cysteines in the prosequence play a role in von Willebrand factor multimer assembly.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
3531
-
3535
)
47
Mosher
 
DF
Plasma fibronectin concentration: a risk factor for arterial thrombosis?
Arterioscler Thromb Vasc Biol
2006
, vol. 
26
 (pg. 
1193
-
1195
)
48
Orem
 
C
Celik
 
S
Orem
 
A
Calapoglu
 
M
Erdol
 
C
Increased plasma fibronectin levels in patients with acute myocardial infarction complicated with left ventricular thrombus.
Thromb Res
2002
, vol. 
105
 (pg. 
37
-
41
)
49
Orem
 
C
Durmus
 
I
Kilinc
 
K
, et al. 
Plasma fibronectin level and its association with coronary artery disease and carotid intima-media thickness.
Coron Artery Dis
2003
, vol. 
14
 (pg. 
219
-
224
)
50
Song
 
KS
Kim
 
HK
Shim
 
W
Jee
 
SH
Plasma fibronectin levels in ischemic heart disease.
Atherosclerosis
2001
, vol. 
154
 (pg. 
449
-
453
)
51
Pecheniuk
 
NM
Elias
 
DJ
Deguchi
 
H
Averell
 
PM
Griffin
 
JH
Elevated plasma fibronectin levels associated with venous thromboembolism.
Thromb Haemost
2008
, vol. 
100
 (pg. 
224
-
228
)
Sign in via your Institution