Abstract
Background:
Protease-activated receptor 4 (PAR4) is expressed by a wide variety of cells, including megakaryocytes/platelets, immune cells, cardiomyocytes and lung epithelial cells. It is the only functional thrombin receptor on murine platelets. A global deficiency of PAR4 is associated with impaired hemostasis and reduced thrombosis.
Objective:
Generation of a mouse line with a megakaryocyte/platelet-specific deletion of PAR4 (PAR4fl/fl;PF4Cre+). Use the mouse line to investigate the role of platelet PAR4 in hemostasis and thrombosis in mice.
Methods:
Platelets from PAR4fl/fl;PF4Cre+ were characterized in vitro. Arterial and venous thrombosis was analyzed. Hemostatic plug formation was analyzed using a saphenous vein laser injury model in global PAR4 and megakaryocyte/platelet-specific deletion of PAR4, wild-type mice treated with thrombin or GPVI inhibitors.
Results:
PAR4fl/fl;PF4Cre+ platelets were unresponsive to thrombin or specific PAR4 stimulation but not to other agonists. PAR4−/− and PAR4fl/fl;PF4Cre+ mice both exhibited a similar reduction in arterial thrombosis compared to their respective controls. More importantly, we show for the first time that platelet PAR4 is critical for venous thrombosis in mice. In addition, PAR4−/− mice and PAR4fl/fl;PF4Cre+ mice exhibited a similar impairment in hemostatic plug stability in a saphenous vein laser injury model. Inhibition of thrombin in wild-type mice gave a similar phenotype. Combined PAR4 deficiency on platelets with GPVI inhibition did not impair hemostatic plug formation but further reduced plug stability.
Conclusion:
We generated a novel PAR4fl/fl;PF4Cre+ mouse line. We used this mouse line to show that PAR4 signaling in platelets is critical for arterial and venous thrombosis and hemostatic plug stability.
Keywords: Protease-activated receptor 4, platelets, thrombosis, hemostasis, saphenous vein injury
Introduction
Protease-activated receptor 4 (PAR4) has previously been studied in the context of thrombin-mediated platelet activation in humans and mice.1, 2 Human platelets express PAR1 and PAR4 whereas mouse platelets PAR3 and PAR4. Both, human PAR1 and PAR4 are druggable targets to reduce pathologic platelet activation in cardiovascular disease.1, 3 Since PAR4 on human platelets responds to higher thrombin concentrations, it is thought to contribute to pathologic thrombus formation but is not essential for platelet-mediated hemostasis in humans.1 Currently, only global PAR4-deficient mice are available to investigate the role of PAR4 in vivo.4 PAR4 deficiency in mice results in platelets that are unresponsive to thrombin but respond normally to thromboxane receptor and glycoprotein (GP) VI stimulation.4 In addition, PAR4−/− mice exhibited prolonged bleeding times and increased blood loss in a tail transection model compared to the PAR4+/+ controls.4, 5 Moreover, PAR4−/− mice or PAR4 inhibition in mice was associated with reduced thrombosis in mouse models of arterial thrombosis.4–9
In addition to platelets, PAR4 is expressed by a variety of cells, including smooth muscle cells, endothelial cells (ECs), cardiomyocytes, immune cells, neuronal cells and lung epithelial cells.10–16 Studies by us and others suggested that non-platelet PAR4 is protective in mouse models of myocardial infarction and virus infection.11, 17, 18 Thus, studies in PAR4−/− mice have to be interpreted with caution as impaired function of more than one cell type may be responsible for the observed phenotype.
Here, we describe the generation of PAR4 floxed (PAR4fl/fl) mice. These mice were crossed with mice expressing Cre recombinase under the platelet factor 4 (PF4) promoter to selectively delete PAR4 in megakaryocytes/platelets (PAR4fl/fl;PF4Cre+). Platelet function, hemostasis, and arterial and venous thrombosis were impaired in PAR4fl/fl;PF4Cre+ and PAR4−/− mice, demonstrating that PAR4 on platelets is critical in hemostasis and thrombosis.
Methods
Mice
To generate mice with megakaryocyte/platelet-specific deletion of PAR4, female PAR4fl/fl mice were crossed with male PAR4fl/fl mice expressing the Cre recombinase under control of the PF4 promoter to generate PAR4fl/fl;PF4Cre+ mice.19 PAR4−/−, PAR4+/+ and wild-type (WT) mice on a C57Bl/6J background were also used.4 Adult mice (8–12 weeks old) of both sexes (equally distributed) were used for the study if not indicated otherwise. All animal experiments were performed in accordance with the guidelines of the animal care and use committee of the University of North Carolina at Chapel Hill and complies with National Institutes of Health guidelines.
Platelet function analysis
Venous blood was collected into low molecular weight heparin (Lovenox, Sanofi, France).20 Platelet counts were determined in whole blood by BD Accuri C6 flow cytometer (BD Biosciences, San Jose, CA) using anti-GPIX-AlexaFluor488 (clone Xia.B4, Emfret Analytics, Würzburg, Germany).20 α-granule release (anti-P-selectin-AlexaFluor647, clone RB40.34, BD Biosciences) and αIIbβ3 integrin activation (JON/A-PE, Emfret Analytics) in the absence or presence of agonists (PAR4 agonist peptide [PAR4 AP, 800μM], ADP [10 μM], convulxin [Cvx, 100 ng/mL]) was analyzed.20 For aggregometry studies, washed platelets (for α-thrombin [Enzyme Research Laboratories, South Bend, IN], PAR4-AP and Cvx) or platelet-rich plasma (for ADP) were resuspended in modified Tyrode’s buffer at a final concentration of 2.5 × 108 platelets/mL in a Chrono-log 4-channel optical aggregation system (Chrono-log, Havertown, PA).20, 21
Mouse arterial thrombosis model
Male mice were anesthetized with 2% isoflurane. Mice were placed in a supine position and the right carotid artery was isolated by blunt dissection.7 A one mm2 piece of filter paper was soaked with FeCl3 (8%) and applied to ventral surface of the exposed carotid artery for 3 min.22 A microvascular ultrasonic flow probe (MA0.5PSB, Transonic Systems Inc, Ithaca, NY, USA) was used to detect blood flow for 30 min after removal of the filter paper. Occlusion was defined as a cessation of blood flow (0 ml/min) for ≥2 min.5, 7
Mouse venous thrombosis model
Male mice were anesthetized with isoflurane (1.5–3%) and body temperature maintained using a heated pad for the duration of the surgery procedure. All following procedures were performed under aseptic conditions. The abdomen was opened by midline laparotomy and the bowel externalized and wrapped in wetted gauze. The inferior vena cava (IVC) was stenosed as described.23 At 48 hours post-stenosis induction, the IVC was inspected for the presence of thrombus with any resultant thrombus removed from the IVC and weighed using a microbalance.
Saphenous vein laser injury hemostasis model
Mice were anesthetized and the saphenous vein was exposed.24 Mice were injected (intravenously, i.v.) with 2.5 μg of an anti-GPIX-AlexaFluor488 antibody to label platelets, and 2 μg of an anti-fibrin-AlexaFluor647 antibody to visualize fibrin formation. To test the effects of recombinant hirudin, mice were injected i.v. with lepirudin (resuspended in sterile saline, 50 mg/kg) immediately before laser injury. To test the effects of the direct thrombin inhibitor dabigatran etexilate, mice were fed custom-made AIN-93M chow (Dyets Inc., Bethlehem, Pa., USA) with peanut flavoring (2 g/kg chow) with or without dabigatran etexilate (10 mg/g chow) for 5 days.25–27 To test the effects of inhibition of platelet GPVI signaling, mice were injected i.p. with ibrutinib (PCI-32765, SelleckChem, 12.5 mg/kg) 1 hour prior to experiment. The saphenous vein was injured by 3 532-nm laser pulses at 0.5, 5.5 and 10.5 mins using Ablate! photoablation system (Intelligent Imaging Innovations, Denver, CO, USA).24 The 1st pulse generated an initial injury, and the 2nd and 3rd pulses reopened the existing platelet plug. If active bleeding was occurring at the injury site at the time of the next laser pulse, the pulse was omitted. Fluorescence images were captured on a Zeiss Axio Examiner Z1 microscope (Zeiss, Jena, Germany) with multicolor LED light source (Lumencor, Beaverton, OR, USA) and an Orca Flash 4.0 camera (Hammamatsu, Bridgewater, NJ, USA) for a total of 15.5 mins.24 Data were analyzed with SLIDEBOOK 6.0 (Intelligent Imaging Innovations).
Statistics
Statistical analyses were performed with GraphPad Prism 9.2 (GraphPad Software Inc., La Jolla, CA). All data are represented as mean ± SEM. For 2-group comparison, the 2-tailed Student’s t test was used for normally distributed data, and the Mann Whitney test for non-normally distributed data. For multiple-group comparison, normally distributed data were analyzed by 1-way or 2-way ANOVA with Bonferroni correction for repeated measures. P-value ≤ 0.05 was regarded as significant.
Results
Generation of conditional PAR4 (F2lr3) gene expressing mice
A PAR4 (F2rl3) floxed allele was generated in C57BL/6J mice by CRISPR-mediated genome editing using the UNC Animal Models Core. GuideScan and CCTop websites were used to identify potential Cas9 guide RNAs targeting the 5’ and 3’ flanking regions of the F2lr3 gene.28, 29 The entire F2rl3 gene was flanked with loxP sites, with the 5’ loxP site inserted ~1 kb upstream of exon 1 and the 3’ loxP site inserted ~1.6 kb 3’ downstream of exon 2. The floxed allele was generated by microinjecting embryos with Cas9 protein (400 nM), guide RNAs (25 ng/μL) targeting the two loxP insertion sites and a circular donor vector (20 ng/μL) designed to insert a loxP and a flippase (Flp) recognition target (FRT) site at the 5’ guide RNA cut site and a loxP site at the 3’ guide RNA cut site. Injected embryos were implanted in pseudo-pregnant B6D2F1/J recipient females. We obtained two founder animals with multicopy tandem integration of the donor vector at the target locus. The founders were mated to animals with a C57BL/6J-Rosa26-Flp recombinase transgene. The presence of a single FRT site in each copy of the knock-in event in a tandem array allows the array to be reduced to a single copy by Flp recombinase expression. F1 animals harboring the tandem integration conditional allele and the Flp transgene were subsequently mated to WT C57BL/6J animals (#000664, Jackson Laboratory, Bar Harbor, ME, USA) to obtain animals with a single-copy conditional allele without the Flp transgene.
Generation of PAR4fl/fl,PF4Cre+ mice
We generated PAR4fl/fl;PF4Cre+ mice in 3 steps: 1/ female PAR4fl/fl mice were crossed with male WT mice expressing PF4 promotor driven Cre recombinase, 2/ female PAR4+/fl were crossed with male PAR4+/fl;PF4Cre+ and 3/ female PAR4fl/fl were crossed with male PAR4fl/fl;PF4Cre+.
Characterization of platelets from PAR4fl/fl;PF4Cre+ mice
Circulating platelet counts were similar between controls (PAR4fl/fl) and PAR4fl/fl;PF4Cre+ littermates (Figure 1A). This is consistent with data from global PAR4−/− mice that loss of PAR4 does not affect platelet count.4 Next, we assessed markers of platelet activation in whole blood by flow cytometry using antibodies recognizing the active conformation of αIIbβ3 integrin (JON/A-PE)30 and P-selectin, a marker of α-granule secretion. No differences were observed in resting platelets (Figure 1B, C) from either genotype. As expected, PAR4fl/fl;PF4Cre+ platelets were unresponsive to thrombin or PAR4 activating peptide (PAR4-AP) (Figure 1B, C). However, no differences in integrin activation (Figure 1B) were observed between control and PAR4fl/fl;PF4Cre+ platelets when stimulated via P2Y1/P2Y12 (ADP) or GPVI (convulxin, Cvx), respectively. A small but significant increase in surface P-selectin expression was observed in PAR4fl/fl;PF4Cre+ platelets activated with Cvx compared to control platelets (Figure 1C). Next, platelet function was assessed by light transmission aggregometry. PAR4fl/fl;PF4Cre+ platelets were unresponsive to increasing concentrations of α-thrombin and high dose PAR4-AP but aggregated normally in response to ADP or Cvx when compared to control platelets (Figure 1D). These results demonstrate that platelets from PAR4fl/fl;PF4Cre+ mice are selectively unresponsive to stimulation via the PAR4 receptor in vitro.
Figure 1: Complete absence of activation response to PAR4 receptor stimulation in PAR4fl/fl;PF4Cre+ platelets.
(A) Platelet counts determined by flow cytometry in whole blood (n=3–4). (B,C) Flow cytometric analysis of αIIbβ3 integrin activation [(B); JON/A-PE] and α-granule secretion [(C); anti-P-selectin]. Diluted whole blood samples from PAR4fl/fl;PF4Cre+ and PAR4fl/fl littermates were incubated for 10 minutes in the presence of antibodies, agonist (800 μM PAR4 activating peptide (PAR4-AP), 10 μM ADP, 100 ng/ml Cvx, PBS for resting controls) and Ca2+, before being further diluted in PBS for analysis of mean fluorescence intensity (MFI) by flow cytometry. Platelets were gated by GPIX expression. Data shown as mean ± SEM (n=3–4). (D) Aggregation responses of washed platelets (α-thrombin, PAR4-AP, Cvx) or PRP (ADP) from PAR4fl/fl;PF4Cre+ and PAR4fl/fl littermates, representative of at least 2 experiments. Statistical significance was determined by unpaired t-test (A) or ANOVA with Bonferroni post-test (B,C).
Platelet PAR4 contributes to FeCl3-induced arterial thrombosis.
Global PAR4 deficiency was shown to be associated with reduced thrombosis in a FeCl3-induced carotid artery thrombosis model in mice.5, 7 Here, we confirmed the findings that global PAR4 deficient mice had prolonged mean time of carotid artery occlusion of 22.6 vs. 6.7 min (Figure 2A) after FeCl3 application. Next, mice with megakaryocytes/platelets PAR4 deletion were subjected to the arterial thrombosis model and we showed that platelet PAR4 contributes to occlusive thrombus formation. The mean occlusion time of the carotid artery was in PAR4fl/fl;PF4Cre+ longer compared to the PAR4fl/fl control mice with 25.1 vs 11.5 min (Figure 2B).
Figure 2: Effect of a global PAR4 deficiency or megakaryocyte/platelet PAR4 deficiency on arterial thrombosis.
Mice with a global or megakaryocyte/platelet specific PAR4 deletion were subjected to a ferric chloride carotid artery thrombosis model. A) Mean time to occlusion in PAR4+/+ and PAR4−/− mice and B) PAR4fl/fl and PAR4fl/fl;PF4Cre+ mice after 8% FeCl3 for 3 min. Occlusion was defined as no blood flow for ≥2 min over a observational period of 30 min. One PAR4+/+ mouse reopened approximal 3 min after our defined occlusion of no blood flow for 2 min. Data shown as mean. **P<0.01, ****P<0.0001. Statistical significance was determined by Mann-Whitney U test.
Platelet PAR4 contributes to venous thrombosis.
Platelets have been shown to contribute to venous thrombosis in the murine IVC stenosis model.31 Here, we investigated the effect of a global PAR4 deficiency or megakaryocytes/platelets PAR4 deficiency on venous thrombosis. PAR4−/− mice exhibited reduced thrombus weight 48 hours after stenosis surgery compared to the WT controls (1.93±1.23mg vs. 12.29±2.57mg, Figure 3A). Global PAR4 deficiency was further associated with reduced overall thrombus incidence compared to the WT controls (Figure 3B). Similar data were obtained from PAR4fl/fl;PF4Cre+ mice which had thrombus weight of 0.40±0.31mg and the PAR4fl/fl control of 6.89±2.06mg (Figure 3C). PAR4fl/fl;PF4Cre+ mice also had reduced thrombus incidence compared to the PAR4fl/fl controls (Figure 3D).
Figure 3: Effect of a global PAR4 deficiency or and megakaryocyte/platelet PAR4 deficiency on venous thrombosis.
Mice with a global or megakaryocyte/platelet specific PAR4 deletion were subjected to IVC stenosis thrombosis model. Thrombus weight (A, C) and incidence of venous thrombus formation (B, D) in PAR4+/+ and PAR4−/− and PAR4fl/fl and PAR4fl/fl;PF4Cre+ mice, respectively, at 48 hours postinduction. Data shown as mean. *P<0.05, **P<0.01. Statistical significance was determined by Mann-Whitney U test.
Effect of global or platelet-specific PAR4 deficiency on hemostatic plug formation in mice.
The saphenous vein laser injury model24, 32 was used to determine bleeding times and quantify platelet and fibrin accumulation during hemostatic plug formation in PAR4 mutant mice. First, we assessed hemostasis in mice with a global PAR4 deficiency. Bar graph representation of bleeding times shows that WT littermates and PAR4−/− mice rapidly formed a hemostatic plug after injury (Figure 4A). All injuries in WT mice (7/7) formed a stable hemostatic plug after each successive injury (Figure 4A). In contrast, injuries in PAR4−/− mice exhibited 2 distinct phenotypes; one third of the injury sites (4/12) formed a stable hemostatic plug after each successive laser ablation, like the WT controls, whereas the majority of the PAR4−/− injury sites (8/12) formed unstable hemostatic plugs that re-opened several minutes after the first ablation leading to extended bleeding (Figure 4A). When bleeding times were calculated, WT littermates exhibited median bleeding times of 16, 20 and 20 secs after ablation (“shot”) #1, 2 and 3, respectively (Figure 4B). PAR4−/− mice demonstrated severely prolonged bleeding (median bleeding time 731 secs after shot #1) despite rapidly forming an initial hemostatic plug (Figure 4C). Injury sites with prolonged bleeding (“bleeding injuries”) also had reduced platelet accumulation beginning around the time of hemostatic plug re-opening (~180 sec post-ablation; Figure 4D). Fibrin formation was comparable to PAR4+/+ controls in PAR4−/− “hemostatic injuries”, and also comparable at PAR4−/− “bleeding injuries” after shot #1 (Figure 4E). Epifluorescence intravital microscopy still-frames from injury sites ~4 mins post-ablation are shown for PAR4+/+ and PAR4−/− mice demonstrating a stable platelet plug in the PAR4+/+ and PAR4−/− hemostatic injury, and active bleeding in the PAR4−/− bleeding injury (Figure 4F). Corresponding movies for still-frames in figures 4–7 are available in the online supplement.
Figure 4: Impaired hemostasis in large subset of global PAR4-deficient mice.
(A,B,C) Hemostasis was monitored by intravital microscopy following laser ablation of the saphenous vein. (A) Bleeding duration at individual injury sites represented in bar graph form. After 5 and 10 minutes, if hemostasis occurred, the platelet plug was disrupted by subsequent laser ablations (arrows/X’s; as in PAR4+/+ and PAR4−/− hemostatic injuries). If bleeding was ongoing at the 5 and/or 10 minute timepoint, subsequent laser ablation was omitted (as in PAR4−/− bleeding injuries). Bleeding time was quantified as total bleeding time for each ablation (“shot”) (B) or time to formation of the first platelet plug (C). Data shown is for 7 injuries from 3 PAR4+/+ mice and 12 injuries (4 hemostatic injuries, 8 bleeding injuries) from 4 PAR4−/− mice. (D) Sum fluorescence intensity for platelets (anti-GPIX-AlexaFluor488) and (E) fibrin (anti-fibrin-AlexaFluor647) was quantified from intravital microscopy videos. PAR4−/− intensity data was segregated between hemostatic and bleeding injuries. Data shown as mean. (F) Still frames from intravital microscopy videos at t ~4 minutes showing hemostasis at a PAR4+/+ injury and PAR4−/− hemostatic and bleeding injuries. Single channels in greyscale are shown for platelets and fibrin with the colored merged image on the far right. Scale bar represents 25 μm. Statistical significance was determined by Mann-Whitney U test for each shot (B) or unpaired t-test (C).
Figure 7: Further impaired hemostasis in PAR4fl/fl;PF4Cre+ mice treated with ibrutinib.
(A) Bleeding duration at individual injury sites represented in bar graph form. Bleeding time was quantified as total bleeding time for each ablation (“shot”) (B) or time to formation of the first platelet plug (C). Data shown is for 8 injuries from 4 PAR4fl/fl ibrutinib-treated mice and 7 injuries from 3 PAR4fl/fl;PF4Cre+ ibrutinib-treated mice. (D) Sum fluorescence intensity for platelets (anti-GPIX-AlexaFluor488) and (E) fibrin (anti-fibrin-AlexaFluor647) was quantified from intravital microscopy videos. Data shown as mean. (F) Still frames from intravital microscopy videos at t ~4 minutes showing hemostasis at a PAR4fl/fl injury and bleeding at a PAR4fl/fl;PF4Cre+ injury. Single channels in greyscale are shown for platelets and fibrin with the colored merged image on the far right. Scale bar represents 25 μm. Statistical significance was determined by Mann-Whitney U test for shot1 (B) or unpaired t-test (C).
Next, we subjected PAR4fl/fl;PF4Cre+ mice and their littermate controls (PAR4fl/fl) to the laser injury hemostasis model. As seen for PAR4+/+ mice, PAR4fl/fl mice formed a stable hemostatic plug after each successive laser ablation (Figure 5A) with median bleeding times of 12, 21 and 22 secs after shot #1, 2 and 3, respectively (Figure 5B). In contrast, almost all of the injury sites in PAR4fl/fl;PF4Cre+ mice (7/8) demonstrated prolonged bleeding times (median bleeding time 737 sec) (Figures 5A, B), despite time to initial hemostasis being similar to that in PAR4fl/fl mice (Figure 5C). Moreover, platelet accumulation after re-opening was significantly reduced at PAR4fl/fl;PF4Cre+ injuries (~150 sec post-ablation), while fibrin formation was similar to the control injuries after shot #1 (Figure 5E). Representative still-frames and movies from intravital video recordings show on-going bleeding at the injury site in a PAR4fl/fl;PF4Cre+ mouse and stable hemostatic plug formation in a PAR4fl/fl control mouse (Figure 5F and online supplement).
Figure 5: Impaired hemostasis in mice with megakaryocyte/platelet-specific deficiency in PAR4.
(A) Bleeding duration at individual injury sites represented in bar graph form. Bleeding time was quantified as total bleeding time for each ablation (“shot”) (B) or time to formation of the first platelet plug (C). Data shown is for 6 injuries from 3 PAR4fl/fl mice and 8 injuries from 3 PAR4fl/fl;PF4Cre+ mice. (D) Sum fluorescence intensity for platelets (anti-GPIX-AlexaFluor488) and (E) fibrin (anti-fibrin-AlexaFluor647) was quantified from intravital microscopy videos. Data shown as mean + SEM. (F) Still frames from intravital microscopy videos at t ~4 minutes showing hemostasis at a PAR4fl/fl injury and bleeding at a PAR4fl/fl;PF4Cre+ injury. Single channels in greyscale are shown for platelets and fibrin with the colored merged image on the far right. Scale bar represents 25 μm. Statistical significance was determined by Mann-Whitney U test for shot1 (B) or unpaired t-test (C).
Our data demonstrate that PAR4 signaling is required for hemostatic plug stability following a small penetrating injury to blood vessels in mice, and that the bleeding phenotype is comparable between mice with either global or megakaryocyte/platelet-specific PAR4 deficiency.
Interestingly, our results showed that platelet PAR4 was not required for initial hemostatic plug formation but was required for maintaining hemostatic plug stability.
Effect of thrombin inhibition on hemostatic plug formation in mice
Recently, we reported reduced expression of tissue factor (TF) in TFlow mice was associated with reduced platelet accumulation and fibrin generation but minimally impacted bleeding in the saphenous vein laser injury model.24 Because complete loss of TF33–35 or prothrombin36, 37 is embryonic lethal, we used a pharmacological approach to inhibit thrombin activity by treating mice with either dabigatran etexilate or recombinant hirudin. Thrombin inhibition prior to the saphenous vein injury was performed to test if the observed phenotype in global PAR4 and megakaryocyte/platelet deficient mice can be reproduced by an inhibition of thrombin.
We first determined if anticoagulation with dabigatran etexilate (10 mg/g chow)25–27 influenced hemostatic plug formation. Dabigatran-fed WT mice had relatively normal bleeding times (Figure 6A–C) and platelet accumulation (Figure 6D), and reduced fibrin generation after shot #1 although fibrin intensity increased following subsequent injuries (Figure 6D). We then determined if more robust thrombin inhibition with i.v. recombinant hirudin would prolong bleeding to a greater extent. In WT mice injected with hirudin, only a third of the injury sites (3/9) demonstrated severely prolonged bleeding, and platelet accumulation was comparable to injury sites in placebo-treated mice despite the near complete loss of fibrin generation at sites of ablation (Figure 6A–D). Representative still frames and movies from intravital video recordings show comparable hemostatic plug formation and reduced or absent fibrin generation at the injury site of a dabigatran- or hirudin-treated mouse, respectively, compared to placebo-treated mice (Figure 6F and online supplement).
Figure 6: Partially impaired hemostasis in mice treated with thrombin inhibitors.
(A) Bleeding duration at individual injury sites represented in bar graph form. Bleeding time was quantified as total bleeding time for each ablation (“shot”) (B) or time to formation of the first platelet plug (C). Data shown is for 5 injuries from 2 placebo-treated mice, 11 injuries from 4 dabigatran etexilate chow-fed mice and 9 injuries from 4 hirudin-injected mice. (D) Sum fluorescence intensity for platelets (anti-GPIX-AlexaFluor488) and (E) fibrin (anti-fibrin-AlexaFluor647) was quantified from intravital microscopy videos. Data shown as mean. (F) Still frames from intravital microscopy videos ~4 minutes showing hemostatic plug formation in all mice and absent and reduced fibrin formation in hirudin- and dabigatran-treated mice, respectively. Single channels in greyscale are shown for platelets and fibrin with the colored merged image on the far right. Scale bar represents 25 μm. Statistical significance was determined by 2-way (B) or 1-way (C) ANOVA with Bonferroni post-test.
The data show that direct inhibition of thrombin, the main PAR4 activating protease, was able to partly reproduce the finding observed with the genetic approach of platelet-specific deletion of PAR4.
Effect of combined loss of PAR4/GPVI signaling on hemostatic plug formation
The ability of PAR4-deficient mice to rapidly form an initial hemostatic plug suggests that other ligand/receptor partners can mediate initial platelet plug formation after small vessel injuries. We have previously demonstrated that mice lacking GPIbα binding to von Willebrand Factor (vWF) have severely prolonged initial hemostatic plug formation.24 The collagen receptor GPVI is an additional candidate to mediate rapid platelet adhesion and activation. We thus treated PAR4fl/fl;PF4Cre+ mice or littermate controls with the Bruton’s tyrosine kinase (Btk) inhibitor, ibrutinib, which inhibits signaling downstream of GPVI (Supplemental Figure 1).38 Ibrutinib-treated PAR4fl/fl mice demonstrated normal bleeding times (median bleeding times 18, 20, and 18 sec after shots #1, 2 and 3, Figure 7A–C) and platelet/fibrin accumulation (Figure 7D,E), in agreement with historical data demonstrating that GPVI plays a minimal role in hemostatic plug formation.39 However, despite retaining the ability to form an initial hemostatic plug, ibrutinib-treated PAR4fl/fl;PAR4Cre+ mice demonstrated faster plug re-opening and longer bleeding times than PAR4fl/fl;PAR4Cre+ mice (median bleeding time 876 secs, Figure 7B). Platelet accumulation was also severely impaired although fibrin formation was increased, likely due to excessive bleeding beginning earlier on (Figure 7D,E).24 Still-frames demonstrate stable plug formation in ibrutinib-treated control mice and active bleeding in ibrutinib-treated PAR4fl/fl;PF4Cre+ mice (Figure 7F). These results suggest that GPVI activation contributes to early plug stability in the absence of PAR4, but even with loss of both PAR4 and GPVI signaling, platelets are able to transiently bridge small injuries likely via GPIbα/vWF.
Discussion
Here, we present the generation of a mouse carrying a floxed PAR4 allele. Crossing these mice with mice expressing Cre recombinase under control of the PF4 promoter (megakaryocyte/platelet-specific gene)40 allowed us to investigate, for the first time, the specific role of platelet PAR4 in arterial and venous thrombosis as well as hemostatic plug formation in mice. We show that platelet PAR4 is critical for arterial thrombus formation. We also showed that a lack of PAR4 on platelets reduced thrombosis in a murine IVC stenosis model. In addition, we found that platelet-specific PAR4 deletion did not impact initial hemostatic platelet plug formation after saphenous vein laser ablation, but the hemostatic plugs were unstable and reopened, leading to significantly prolonged bleeding times. Inhibition of GPVI signaling further impaired plug stability in PAR4fl/fl;PF4Cre+ mice, and inhibition of thrombin, the main activator of PAR4 on platelets, reproduced, in part, the bleeding phenotype of mice with global or megakaryocyte/platelet-specific PAR4 deficiency.
The role of PAR4 for thrombin-mediated platelet activation in mice was first shown by Sambrano et al..4 In that study, global PAR4 deficiency resulted in platelets unresponsive to thrombin or PAR4-AP, prolonged bleeding times/increased blood loss and partial protection from arterial thrombosis.4 Thrombin-mediated platelet activation at sites of vascular injury contributes to thrombosis that can cause myocardial infarction and a subset of stroke.41, 42 We confirmed that a lack of thrombin-mediated platelet activation prolonged time to occlusion in an artery injury model. In addition, we showed that specific platelet PAR4 was responsible for the increased arterial thrombosis in the model. Platelets were indicated to contribute to venous thrombosis in mice31, we show here that platelet PAR4 is one of the important mediators of venous thrombosis in mice. Global PAR4 deficient mice and mice with a megakaryocyte/platelet PAR4 deficiency showed similar results in the used arterial and venous thrombosis models suggesting that the major contributor for thrombus formation is platelet PAR4.
Using the saphenous vein laser injury model24, which combines a small injury hemostasis model with intravital fluorescence microscopy, we confirmed that PAR4 on platelets is critical for hemostatic plug formation in mice. Owing to the small injury size (~50 μm diameter) and the use of real-time intravital fluorescence microscopy, we are able to detect more subtle roles for platelet receptors and signaling proteins in hemostatic plug formation. In the large injury tail bleed assay, PAR4 deficiency results in injuries that are entirely unable to close.4 However, in a small laser injury thrombosis model, PAR4 deficiency resulted in reduced platelet accumulation with no impact on fibrin generation6, similar to our findings. Interestingly, we observed that initial plug formation was normal in the absence of platelet PAR4 (a finding also observed by Vandendries et al.6), likely mediated by platelet adhesion and activation by GPIbα/vWF and GPVI/collagen interactions. Indeed, mice with platelets lacking the vWF-binding domain of GPIbα had drastically prolonged bleeding times caused by an inability to form an initial hemostatic plug.24 Additionally, treatment of PAR4fl/fl;PF4Cre+ mice with the Btk inhibitor ibrutinib, to inhibit GPVI-mediated platelet activation, led to more rapid re-opening of hemostatic plugs and further prolonged bleeding times compared to untreated PAR4fl/fl;PF4Cre+ mice. While loss of expression or function of GPVI by itself does not significantly impair hemostasis43, we also observed prolonged bleeding in P2Y12-deficient mice treated with ibrutinib38, suggesting GPVI plays a supporting role in hemostasis but becomes more critical in the absence of other major platelet signaling receptors. Our results also confirm our previous findings24 that in small injuries, platelets are critical for hemostatic plug formation while fibrin plays a less important role, as exemplified by hemostatic injuries in hirudin-treated mice where fibrin was nearly undetectable. Normal hemostasis in dabigatran etexilate-fed mice could be explained at least in part by incomplete inhibition of thrombin activity. The dose used was shown not to cause spontaneous bleeding due to impairment of hemostasis.26, 27, 44 In addition, we believe the level of inhibition with hirudin was higher than dabigatran etexilate because it is injected rather than delivered via the chow. Moreover, the partial phenotype in hirudin-treated mice could be explained by a role for additional proteases in PAR4 cleavage such as cathepsin G.45 Indeed, neutrophils, a primary source of cathepsin G in blood, are recruited to the hemostatic plug in our model (unpublished observation). Importantly, our data demonstrate that for small injuries, PAR4-mediated platelet activation is not required for initial formation of the hemostatic plug but instead plays a critical role in platelet activation for plug stability. These findings may suggest that the initial TF-dependent thrombin burst plays a lesser role in small injuries and the cell surface-generated second wave of thrombin mediates the majority of PAR4 cleavage.46 Finally, we have previously demonstrated that mice lacking the Rap1 GTPase activator CalDAG-GEFI are unable to form a hemostatic plug at least five minutes post-ablation47, suggesting platelet activation is required to bridge even small injuries. Therefore, it was surprising to observe initial plug formation with platelets lacking both PAR4 and GPVI signaling. Additional candidates to induce rapid platelet signaling are ADP/P2Y12 and TxA2/TP, and GPIbα/vWF (or other ligands). In fact, GPIbα is not only critical for platelet tethering but can also induce platelet activation and release of second wave mediators.48 The contribution of GPIbα activation signaling during initial plug formation in our model will need to be explored.
While we showed here the effect of PAR4-mediated signaling on mouse platelets and its role in arterial and venous thrombosis and the saphenous vein laser injury model for hemostatic plug formation and stability, the generation of a mouse carrying the floxed PAR4 allele will help to delineate the cell-specific role of PAR4 in different disease models in vivo. For instance, we showed that PAR4 limits RNA virus infection in mice.17 However, by using PAR4−/− mice we were unable to distinguish between platelet PAR4, immune cell PAR4 or other resident cell PAR4 in their specific contribution to the anti-viral responses.17 The here described PAR4fl/fl mice will be an important tool to investigate those cell-specific roles of PAR4. Others indicated a role for PAR4 in cardiomyocyte, neuron, neutrophil, or regulatory T cell functions.11, 12, 14, 18, 49
In conclusion, we described the generation of PAR4fl/fl mice. We showed that PAR4 on platelets contribute to both arterial and venous thrombosis in mice. In addition, platelet PAR4 contributes to the stability of the hemostatic plug but not initial hemostatic plug formation in the murine saphenous vein after laser injury. Further, we showed that lack of PAR4 in combination with GPVI inhibition did not change initial hemostatic plug formation but further destabilized the hemostatic plug.
Supplementary Material
Supplemental Figure 1: Ibrutinib inhibits platelet GPVI signaling. Blood samples were collected from control mice or mice 1 hour after ibrutinib treatment. Platelets in diluted whole blood were activated for 10 mins with Cvx (50 or 200 ng/ml) or PAR4-AP (400 μM) in the presence of JON/A-PE and anti-P-selectin-AlexaFluor647 antibodies, and samples were then diluted in PBS for flow cytometric analysis. Platelets were gated by GPIX expression.
Supplemental video 1: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type (PAR4+/+) mouse. See figure 4A+F, injury #3 in main text.
Supplemental video 2: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4−/− mouse. See figure 4A+F, hemostatic injury #1 in main text.
Supplemental video 3: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4−/− mouse. See figure 4A+F, bleeding injury #7 in main text.
Supplemental video 4: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl (control) mouse. See figure 5A+F, injury #6 in main text.
Supplemental video 5: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl;PF4Cre+ mouse. See figure 5A+F, injury #3 in main text.
Supplemental video 7: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with dabigatran etexilate chow. See figure 6A+F, injury #6 in main text.
Supplemental video 6: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with placebo chow. See figure 5A+F, injury #2 in main text.
Supplemental video 8: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with hirudin. See figure 6A+F, injury #7 in main text.
Supplemental video 10: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl;PF4Cre+ mouse treated with ibrutinib. See figure 7A+F, injury #2 in main text.
Supplemental video 9: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl (control) mouse treated with ibrutinib. See figure 7A+F, injury #5 in main text.
Essentials.
Global PAR4 deficiency was shown to reduce thrombosis and hemostasis in mice.
Generation of a new PAR4fl/fl;PF4Cre+ mouse line.
Platelet PAR4 contributes to arterial and venous thrombosis.
Platelet PAR4 and GPVI are required to maintain hemostatic plug stability.
Acknowledgement
We thank the staff of the UNC Animal Models Core for their work to generate the PAR4fl,fl mice. We want to thank Ying Zhang for excellent technical assistance. The presented study was supported by grants from ASH (ASH Scholar Award) and NBF (NBF Early Career Grant) to RHL, and from the NHLBI to WB (HL144976) and SA (HL142799).
Footnotes
Disclosure of Interests
D. O. Cowley is employed by, has equity ownership in and serves on the board of directors of TransViragen, the company which has been contracted by UNC-Chapel Hill to manage its Animal Models Core Facility. The other authors declare no competing interests.
References
- 1.Han X and Nieman MT. PAR4 (Protease-Activated Receptor 4): PARticularly Important 4 Antiplatelet Therapy. Arteriosclerosis, thrombosis, and vascular biology. 2018;38:287–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.French SL and Hamilton JR. Protease-activated receptor 4: from structure to function and back again. British journal of pharmacology. 2016;173:2952–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wilson SJ, Ismat FA, Wang Z, Cerra M, Narayan H, Raftis J, Gray TJ, Connell S, Garonzik S, Ma X, Yang J and Newby DE. PAR4 (Protease-Activated Receptor 4) Antagonism With BMS-986120 Inhibits Human Ex Vivo Thrombus Formation. Arteriosclerosis, thrombosis, and vascular biology. 2018;38:448–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sambrano GR, Weiss EJ, Zheng YW, Huang W and Coughlin SR. Role of thrombin signalling in platelets in haemostasis and thrombosis. Nature. 2001;413:74–8. [DOI] [PubMed] [Google Scholar]
- 5.Bynagari-Settipalli YS, Cornelissen I, Palmer D, Duong D, Concengco C, Ware J and Coughlin SR. Redundancy and interaction of thrombin- and collagen-mediated platelet activation in tail bleeding and carotid thrombosis in mice. Arteriosclerosis, thrombosis, and vascular biology. 2014;34:2563–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vandendries ER, Hamilton JR, Coughlin SR, Furie B and Furie BC. Par4 is required for platelet thrombus propagation but not fibrin generation in a mouse model of thrombosis. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:288–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cornelissen I, Palmer D, David T, Wilsbacher L, Concengco C, Conley P, Pandey A and Coughlin SR. Roles and interactions among protease-activated receptors and P2ry12 in hemostasis and thrombosis. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:18605–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lee H, Sturgeon SA, Mountford JK, Jackson SP and Hamilton JR. Safety and efficacy of targeting platelet proteinase-activated receptors in combination with existing anti-platelet drugs as antithrombotics in mice. British journal of pharmacology. 2012;166:2188–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lee H, Sturgeon SA, Jackson SP and Hamilton JR. The contribution of thrombin-induced platelet activation to thrombus growth is diminished under pathological blood shear conditions. Thrombosis and haemostasis. 2012;107:328–37. [DOI] [PubMed] [Google Scholar]
- 10.Li S, Tarlac V and Hamilton JR. Using PAR4 Inhibition as an Anti-Thrombotic Approach: Why, How, and When? International journal of molecular sciences. 2019;20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Antoniak S, Pawlinski R and Mackman N. Protease-activated receptors and myocardial infarction. IUBMB life. 2011;63:383–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Peng Q, Ratnasothy K, Boardman DA, Jacob J, Tung SL, McCluskey D, Smyth LA, Lechler RI, Dorling A and Lombardi G. Protease Activated Receptor 4 as a Novel Modulator of Regulatory T Cell Function. Frontiers in immunology. 2019;10:1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pavic G, Grandoch M, Dangwal S, Jobi K, Rauch BH, Doller A, Oberhuber A, Akhyari P, Schror K, Fischer JW and Fender AC. Thrombin receptor protease-activated receptor 4 is a key regulator of exaggerated intimal thickening in diabetes mellitus. Circulation. 2014;130:1700–11. [DOI] [PubMed] [Google Scholar]
- 14.Sessenwein JL, Baker CC, Pradhananga S, Maitland ME, Petrof EO, Allen-Vercoe E, Noordhof C, Reed DE, Vanner SJ and Lomax AE. Protease-Mediated Suppression of DRG Neuron Excitability by Commensal Bacteria. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2017;37:11758–11768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hollenberg MD, Saifeddine M, Al-Ani B and Gui Y. Proteinase-activated receptor 4 (PAR4): action of PAR4-activating peptides in vascular and gastric tissue and lack of cross-reactivity with PAR1 and PAR2. Can J Physiol Pharmacol. 1999;77:458–64. [PubMed] [Google Scholar]
- 16.Syeda F, Grosjean J, Houliston RA, Keogh RJ, Carter TD, Paleolog E and Wheeler-Jones CP. Cyclooxygenase-2 induction and prostacyclin release by protease-activated receptors in endothelial cells require cooperation between mitogen-activated protein kinase and NF-kappaB pathways. The Journal of biological chemistry. 2006;281:11792–804. [DOI] [PubMed] [Google Scholar]
- 17.Tatsumi K, Schmedes CM, Houston ER, Butler E, Mackman N and Antoniak S. Protease-activated receptor 4 protects mice from Coxsackievirus B3 and H1N1 influenza A virus infection. Cellular immunology. 2019;344:103949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kolpakov MA, Guo X, Rafiq K, Vlasenko L, Hooshdaran B, Seqqat R, Wang T, Fan X, Tilley DG, Kostyak JC, Kunapuli SP, Houser SR and Sabri A. Loss of Protease-Activated Receptor 4 Prevents Inflammation Resolution and Predisposes the Heart to Cardiac Rupture After Myocardial Infarction. Circulation. 2020;142:758–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pawlinski R, Wang JG, Owens AP 3rd, Williams J, Antoniak S, Tencati M, Luther T, Rowley JW, Low EN, Weyrich AS and Mackman N. Hematopoietic and nonhematopoietic cell tissue factor activates the coagulation cascade in endotoxemic mice. Blood. 2010;116:806–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Piatt R, Paul DS, Lee RH, McKenzie SE, Parise LV, Cowley DO, Cooley BC and Bergmeier W. Mice Expressing Low Levels of CalDAG-GEFI Exhibit Markedly Impaired Platelet Activation With Minor Impact on Hemostasis. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1838–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Stefanini L, Boulaftali Y, Ouellette TD, Holinstat M, Desire L, Leblond B, Andre P, Conley PB and Bergmeier W. Rap1-Rac1 circuits potentiate platelet activation. Arteriosclerosis, thrombosis, and vascular biology. 2012;32:434–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Owens AP 3rd, Lu Y, Whinna HC, Gachet C, Fay WP and Mackman N. Towards a standardization of the murine ferric chloride-induced carotid arterial thrombosis model. Journal of thrombosis and haemostasis : JTH. 2011;9:1862–3. [DOI] [PubMed] [Google Scholar]
- 23.Grover SP, Olson TM, Cooley BC and Mackman N. Model-dependent contributions of FXII and FXI to venous thrombosis in mice. Journal of thrombosis and haemostasis : JTH. 2020;18:2899–2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Getz TM, Piatt R, Petrich BG, Monroe D, Mackman N and Bergmeier W. Novel mouse hemostasis model for real-time determination of bleeding time and hemostatic plug composition. Journal of thrombosis and haemostasis : JTH. 2015;13:417–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Antoniak S, Tatsumi K, Bode M, Vanja S, Williams JC and Mackman N. Protease-Activated Receptor 1 Enhances Poly I:C Induction of the Antiviral Response in Macrophages and Mice. Journal of innate immunity. 2017;9:181–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tatsumi K, Antoniak S, Subramaniam S, Gondouin B, Neidich SD, Beck MA, Mickelson J, Monroe DM 3rd, Bastarache JA and Mackman N. Anticoagulation increases alveolar hemorrhage in mice infected with influenza A. Physiol Rep. 2016;4:e13071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Antoniak S, Owens AP 3rd, Baunacke M, Williams JC, Lee RD, Weithauser A, Sheridan PA, Malz R, Luyendyk JP, Esserman DA, Trejo J, Kirchhofer D, Blaxall BC, Pawlinski R, Beck MA, Rauch U and Mackman N. PAR-1 contributes to the innate immune response during viral infection. The Journal of clinical investigation. 2013;123:1310–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Perez AR, Pritykin Y, Vidigal JA, Chhangawala S, Zamparo L, Leslie CS and Ventura A. GuideScan software for improved single and paired CRISPR guide RNA design. Nat Biotechnol. 2017;35:347–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Stemmer M, Thumberger T, Del Sol Keyer M, Wittbrodt J and Mateo JL. CCTop: An Intuitive, Flexible and Reliable CRISPR/Cas9 Target Prediction Tool. PloS one. 2015;10:e0124633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bergmeier W, Schulte V, Brockhoff G, Bier U, Zirngibl H and Nieswandt B. Flow cytometric detection of activated mouse integrin alphaIIbbeta3 with a novel monoclonal antibody. Cytometry. 2002;48:80–6. [DOI] [PubMed] [Google Scholar]
- 31.von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B and Massberg S. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. The Journal of experimental medicine. 2012;209:819–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Neeves KB. Down in a hole: a new laser ablation model of hemostasis. Journal of thrombosis and haemostasis : JTH. 2015;13:414–6. [DOI] [PubMed] [Google Scholar]
- 33.Carmeliet P, Mackman N, Moons L, Luther T, Gressens P, Van Vlaenderen I, Demunck H, Kasper M, Breier G, Evrard P, Muller M, Risau W, Edgington T and Collen D. Role of tissue factor in embryonic blood vessel development. Nature. 1996;383:73–5. [DOI] [PubMed] [Google Scholar]
- 34.Bugge TH, Xiao Q, Kombrinck KW, Flick MJ, Holmback K, Danton MJ, Colbert MC, Witte DP, Fujikawa K, Davie EW and Degen JL. Fatal embryonic bleeding events in mice lacking tissue factor, the cell-associated initiator of blood coagulation. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:6258–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Toomey JR, Kratzer KE, Lasky NM, Stanton JJ and Broze GJ Jr., Targeted disruption of the murine tissue factor gene results in embryonic lethality. Blood. 1996;88:1583–7. [PubMed] [Google Scholar]
- 36.Sun WY, Witte DP, Degen JL, Colbert MC, Burkart MC, Holmback K, Xiao Q, Bugge TH and Degen SJ. Prothrombin deficiency results in embryonic and neonatal lethality in mice. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:7597–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Xue J, Wu Q, Westfield LA, Tuley EA, Lu D, Zhang Q, Shim K, Zheng X and Sadler JE. Incomplete embryonic lethality and fatal neonatal hemorrhage caused by prothrombin deficiency in mice. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:7603–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lee RH, Piatt R, Conley PB and Bergmeier W. Effects of ibrutinib treatment on murine platelet function during inflammation and in primary hemostasis. Haematologica. 2017;102:e89–e92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kato K, Kanaji T, Russell S, Kunicki TJ, Furihata K, Kanaji S, Marchese P, Reininger A, Ruggeri ZM and Ware J. The contribution of glycoprotein VI to stable platelet adhesion and thrombus formation illustrated by targeted gene deletion. Blood. 2003;102:1701–7. [DOI] [PubMed] [Google Scholar]
- 40.Tiedt R, Schomber T, Hao-Shen H and Skoda RC. Pf4-Cre transgenic mice allow the generation of lineage-restricted gene knockouts for studying megakaryocyte and platelet function in vivo. Blood. 2007;109:1503–6. [DOI] [PubMed] [Google Scholar]
- 41.Kollikowski AM, Pham M, Marz AG, Papp L, Nieswandt B, Stoll G and Schuhmann MK. Platelet Activation and Chemokine Release Are Related to Local Neutrophil-Dominant Inflammation During Hyperacute Human Stroke. Translational stroke research. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mackman N. Triggers targets and treatments for thrombosis. Nature. 2008;451:914–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bender M, May F, Lorenz V, Thielmann I, Hagedorn I, Finney BA, Vogtle T, Remer K, Braun A, Bosl M, Watson SP and Nieswandt B. Combined in vivo depletion of glycoprotein VI and C-type lectin-like receptor 2 severely compromises hemostasis and abrogates arterial thrombosis in mice. Arteriosclerosis, thrombosis, and vascular biology. 2013;33:926–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Posma JJ, Grover SP, Hisada Y, Owens AP 3rd, Antoniak S, Spronk HM and Mackman N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arteriosclerosis, thrombosis, and vascular biology. 2019;39:13–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Han X, Nieman MT and Kerlin BA. Protease-activated receptors: An illustrated review. Res Pract Thromb Haemost. 2021;5:17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Monroe DM and Hoffman M. What does it take to make the perfect clot? Arteriosclerosis, thrombosis, and vascular biology. 2006;26:41–8. [DOI] [PubMed] [Google Scholar]
- 47.Stefanini L, Paul DS, Robledo RF, Chan ER, Getz TM, Campbell RA, Kechele DO, Casari C, Piatt R, Caron KM, Mackman N, Weyrich AS, Parrott MC, Boulaftali Y, Adams MD, Peters LL and Bergmeier W. RASA3 is a critical inhibitor of RAP1-dependent platelet activation. The Journal of clinical investigation. 2015;125:1419–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Estevez B, Stojanovic-Terpo A, Delaney MK, O’Brien KA, Berndt MC, Ruan C and Du X. LIM kinase-1 selectively promotes glycoprotein Ib-IX-mediated TXA2 synthesis, platelet activation, and thrombosis. Blood. 2013;121:4586–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gomides LF, Lima OC, Matos NA, Freitas KM, Francischi JN, Tavares JC and Klein A. Blockade of proteinase-activated receptor 4 inhibits neutrophil recruitment in experimental inflammation in mice. Inflammation research : official journal of the European Histamine Research Society [et al]. 2014;63:935–41. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental Figure 1: Ibrutinib inhibits platelet GPVI signaling. Blood samples were collected from control mice or mice 1 hour after ibrutinib treatment. Platelets in diluted whole blood were activated for 10 mins with Cvx (50 or 200 ng/ml) or PAR4-AP (400 μM) in the presence of JON/A-PE and anti-P-selectin-AlexaFluor647 antibodies, and samples were then diluted in PBS for flow cytometric analysis. Platelets were gated by GPIX expression.
Supplemental video 1: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type (PAR4+/+) mouse. See figure 4A+F, injury #3 in main text.
Supplemental video 2: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4−/− mouse. See figure 4A+F, hemostatic injury #1 in main text.
Supplemental video 3: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4−/− mouse. See figure 4A+F, bleeding injury #7 in main text.
Supplemental video 4: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl (control) mouse. See figure 5A+F, injury #6 in main text.
Supplemental video 5: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl;PF4Cre+ mouse. See figure 5A+F, injury #3 in main text.
Supplemental video 7: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with dabigatran etexilate chow. See figure 6A+F, injury #6 in main text.
Supplemental video 6: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with placebo chow. See figure 5A+F, injury #2 in main text.
Supplemental video 8: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a wild-type mouse treated with hirudin. See figure 6A+F, injury #7 in main text.
Supplemental video 10: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl;PF4Cre+ mouse treated with ibrutinib. See figure 7A+F, injury #2 in main text.
Supplemental video 9: Platelet (green) and fibrin (red) accumulation after saphenous vein laser injury of a PAR4fl/fl (control) mouse treated with ibrutinib. See figure 7A+F, injury #5 in main text.