Vessel wall BAMBI contributes to hemostasis and thrombus stability

Slides:



Advertisements
Similar presentations
Red blood cells mediate the onset of thrombosis in the ferric chloride murine model by Justin D. Barr, Anil K. Chauhan, Gilbert V. Schaeffer, Jessica K.
Advertisements

Human platelets produced in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice upon transplantation of human cord blood CD34+ cells are.
Arterioscler Thromb Vasc Biol
Thymidine Phosphorylase Participates in Platelet Signaling and Promotes ThrombosisNovelty and Significance by Wei Li, Alba Gigante, Maria-Jesus Perez-Perez,
The distal carboxyl-terminal domains of ADAMTS13 are required for regulation of in vivo thrombus formation by Fumiaki Banno, Anil K. Chauhan, Koichi Kokame,
Factor XIII in plasma, but not in platelets, mediates red blood cell retention in clots and venous thrombus size in mice by Sravya Kattula, James R. Byrnes,
Reduced thrombus stability in mice lacking the α2A-adrenergic receptor
Activation of αIIbβ3 is a sufficient but also an imperative prerequisite for activation of α2β1 on platelets by Gerlinde R. Van de Walle, Anne Schoolmeester,
by Matt W. Goschnick, Lai-Man Lau, Janet L. Wee, Yong S. Liu, P
Occlusive thrombi arise in mammals but not birds in response to arterial injury: evolutionary insight into human cardiovascular disease by Alec A. Schmaier,
Tissue-Specific Expression of Functional Platelet Factor XI Is Independent of Plasma Factor XI Expression by Chang-jun Hu, Frank A. Baglia, David C.B.
Defective release of α granule and lysosome contents from platelets in mouse Hermansky-Pudlak syndrome models by Ronghua Meng, Jie Wu, Dawn C. Harper,
Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI by Erik I. Tucker, Ulla M. Marzec, Tara C.
Intracoronary shear-related up-regulation of platelet P-selectin and platelet-monocyte aggregation despite the use of aspirin and clopidogrel by Andy S.
Kinetics and mechanics of clot contraction are governed by the molecular and cellular composition of the blood by Valerie Tutwiler, Rustem I. Litvinov,
by Kesheng Dai, Richard Bodnar, Michael C. Berndt, and Xiaoping Du
Essential role for phosphoinositide 3-kinase in shear-dependent signaling between platelet glycoprotein Ib/V/IX and integrin αIIbβ3 by Cindy L. Yap, Karen.
by Simon Stritt, Inga Birkholz, Sarah Beck, Simona Sorrentino, K
Recombinant factor VIIa restores aggregation of αIIbβ3-deficient platelets via tissue factor–independent fibrin generation by Ton Lisman, Jelle Adelmeijer,
Effects of inflammatory cytokines on the release and cleavage of the endothelial cell–derived ultralarge von Willebrand factor multimers under flow by.
Necrotic platelets provide a procoagulant surface during thrombosis
by Zhengyan Wang, Tina M. Leisner, and Leslie V. Parise
Angiotensin 1-7 and Mas decrease thrombosis in Bdkrb2−/− mice by increasing NO and prostacyclin to reduce platelet spreading and glycoprotein VI activation.
Inhibition of collagen-induced platelet aggregation by anopheline antiplatelet protein, a saliva protein from a malaria vector mosquito by Shigeto Yoshida,
Alterations in platelet secretion differentially affect thrombosis and hemostasis by Smita Joshi, Meenakshi Banerjee, Jinchao Zhang, Akhil Kesaraju, Irina.
Exposure to fluid shear stress modulates the ability of endothelial cells to recruit neutrophils in response to tumor necrosis factor-α: a basis for local.
BCL2/BCL-XL inhibition induces apoptosis, disrupts cellular calcium homeostasis, and prevents platelet activation by Meike Vogler, Hassan A. Hamali, Xiao-Ming.
A role for the thiol isomerase protein ERP5 in platelet function
The Modifier of hemostasis (Mh) locus on chromosome 4 controls in vivo hemostasis of Gp6−/− mice by Yann Cheli, Deborah Jensen, Patrizia Marchese, David.
Rap1-GTP–interacting adaptor molecule (RIAM) is dispensable for platelet integrin activation and function in mice by Simon Stritt, Karen Wolf, Viola Lorenz,
Two-wavelength near-infrared fluorescence for the quantitation of drug antiplatelet effects in large animal model systems  Yoshitomo Ashitate, MD, Soon.
Β1 integrin−mediated signals are required for platelet granule secretion and hemostasis in mouse by Tobias Petzold, Raphael Ruppert, Dharmendra Pandey,
Impaired activation of platelets lacking protein kinase C-θ isoform
Brain-derived microparticles induce systemic coagulation in a murine model of traumatic brain injury by Ye Tian, Breia Salsbery, Min Wang, Hengjie Yuan,
Coordination of platelet agonist signaling during the hemostatic response in vivo by Jian Shen, Sara Sampietro, Jie Wu, Juan Tang, Shuchi Gupta, Chelsea.
Apoptotic Vascular Endothelial Cells Become Procoagulant
C1qTNF–related protein-1 (CTRP-1): a vascular wall protein that inhibits collagen-induced platelet aggregation by blocking VWF binding to collagen by Gerald.
Cobalt hematoporphyrin inhibits CLEC-2–podoplanin interaction, tumor metastasis, and arterial/venous thrombosis in mice by Nagaharu Tsukiji, Makoto Osada,
IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation by Melanie Antl, Marie-Luise von Brühl, Christina Eiglsperger,
Interaction of kindlin-3 and β2-integrins differentially regulates neutrophil recruitment and NET release in mice by Zhen Xu, Jiayi Cai, Juan Gao, Gilbert.
The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia by Christine Le Roy, Pierre-Antoine Deglesne, Nathalie.
Diverging signaling events control the pathway of GPVI down-regulation in vivo by Tamer Rabie, David Varga-Szabo, Markus Bender, Rastislav Pozgaj, Francois.
Volume 102, Issue 10, Pages (May 2012)
RGS10 shapes the hemostatic response to injury through its differential effects on intracellular signaling by platelet agonists by Peisong Ma, Shuchi Gupta,
Causal relationship between hyperfibrinogenemia, thrombosis, and resistance to thrombolysis in mice by Kellie R. Machlus, Jessica C. Cardenas, Frank C.
Dichloroacetate, an inhibitor of pyruvate dehydrogenase kinases, inhibits platelet aggregation and arterial thrombosis by Manasa K. Nayak, Nirav Dhanesha,
Enhancing functional platelet release in vivo from in vitro–grown megakaryocytes using small molecule inhibitors by Danuta Jarocha, Karen K. Vo, Randolph.
Volume 112, Issue 1, Pages (January 2003)
Humanized GPIbα–von Willebrand factor interaction in the mouse
Platelet-specific deletion of SNAP23 ablates granule secretion, substantially inhibiting arterial and venous thrombosis in mice by Christopher M. Williams,
Hierarchical organization in the hemostatic response and its relationship to the platelet-signaling network by Timothy J. Stalker, Elizabeth A. Traxler,
A function-blocking PAR4 antibody is markedly antithrombotic in the face of a hyperreactive PAR4 variant by Shauna L. French, Claudia Thalmann, Paul F.
by Silvia Mele, Stephen Devereux, Andrea G
Rap1 binding to the talin 1 F0 domain makes a minimal contribution to murine platelet GPIIb-IIIa activation by Frederic Lagarrigue, Alexandre R. Gingras,
Specific Synergy of Multiple Substrate–Receptor Interactions in Platelet Thrombus Formation under Flow  Brian Savage, Fanny Almus-Jacobs, Zaverio M Ruggeri 
Platelet heterogeneity in activation-induced glycoprotein shedding: functional effects by Constance C. F. M. J. Baaten, Frauke Swieringa, Tomasz Misztal,
Comparison of reversal activity and mechanism of action of UHRA, andexanet, and PER977 on heparin and oral FXa inhibitors by Manu T. Kalathottukaren, A.
by Changjie Zhang, Anju Kelkar, and Sriram Neelamegham
Platelet MEKK3 regulates arterial thrombosis and myocardial infarct expansion in mice by Xuemei Fan, Conghui Wang, Panlai Shi, Wen Gao, Jianmin Gu, Yan.
Severe platelet dysfunction in NHL patients receiving ibrutinib is absent in patients receiving acalabrutinib by Alexander P. Bye, Amanda J. Unsworth,
The endogenous antimicrobial cathelicidin LL37 induces platelet activation and augments thrombus formation by Maryam F. Salamah, Divyashree Ravishankar,
Platelet HMGB1 is required for efficient bacterial clearance in intra-abdominal bacterial sepsis in mice by Hui Zhou, Meihong Deng, Yingjie Liu, Chenxuan.
ELMO1 deficiency enhances platelet function
by Megan S. Rost, Ilya Shestopalov, Yang Liu, Andy H. Vo, Catherine E
Deletion of the Arp2/3 complex in megakaryocytes leads to microthrombocytopenia in mice by David S. Paul, Caterina Casari, Congying Wu, Raymond Piatt,
TREM-like transcript 1: a more sensitive marker of platelet activation than P-selectin in humans and mice by Christopher W. Smith, Zaher Raslan, Lola Parfitt,
Complement C5 but not C3 is expendable for tissue factor activation by cofactor-independent antiphospholipid antibodies by Nadine Müller-Calleja, Svenja.
Therapeutic efficacy of the platelet glycoprotein Ib antagonist anfibatide in murine models of thrombotic thrombocytopenic purpura by Liang Zheng, Yingying.
by Alyssa J. Moroi, Nicole M. Zwifelhofer, Matthew J. Riese, Debra K
by Sarah M. Nordstrom, Brian A. Holliday, Brandon C. Sos, James W
Presentation transcript:

Vessel wall BAMBI contributes to hemostasis and thrombus stability by Isabelle I. Salles-Crawley, James H. Monkman, Josefin Ahnström, David A. Lane, and James T. B. Crawley Blood Volume 123(18):2873-2881 May 1, 2014 ©2014 by American Society of Hematology

Genetic ablation of Bambi leads to increased tail-bleeding time. Genetic ablation of Bambi leads to increased tail-bleeding time. (A) PCR was performed on DNA isolated from ear biopsies using previously described primers35 leading to the amplification of the wild-type allele (top; 225bp), and the Bambi-deleted allele (bottom; 316bp). (B) RT-PCR analysis of BMCs isolated from wild-type and Bambi−/− mice using primers specific to Bambi transcript (1:587 bp; 2:329 bp; 3:812 bp) or Gapdh (Ctrl; 452bp). (C-D) Analysis of tail-bleeding experiments on Bambi+/+Bambi+/− and Bambi−/− mice. (C) Graph showing the time to cessation in bleeding for all animals according to genotype. The percentage of mice that were still bleeding after 12 minutes (dotted line) was significantly higher in Bambi-deficient mice compared with wild-type littermates. Statistical analysis was performed using the χ2 test for linear trend (P < .02). Each symbol represents 1 animal. (D) Bambi-deficient mice exhibited a significant increase in blood loss (determined after 10 minutes). Each symbol represents 1 animal. Horizontal lines intersecting data sets represent the median. Statistical analysis was performed using ANOVA (Kruskal-Wallis). *P < .05. Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology

Impaired thrombus formation in Bambi-deficient mice after FeCl3 injury in vivo. Impaired thrombus formation in Bambi-deficient mice after FeCl3 injury in vivo. (A-B) Mesenteric arterioles were injured with FeCl3 and adhesion and thrombus formation of fluorescently labeled platelets was followed by intravital microscopy. (A) Representative images of thrombus formation at indicated times after removal of the filter paper. Scale bar represents 50 µm. A white asterisk indicates occlusion of the vessel for the wild-type mouse. (B) Bambi+/− and Bambi−/− mice displayed a delayed vascular occlusion compared with Bambi+/+ littermates. Each symbol represents 1 animal. Horizontal lines intersecting data sets represent the mean. Statistical analysis was performed using ANOVA (Kruskal-Wallis). **P < .01. Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology

Decreased thrombus stability in Bambi-deficient mice after laser-induced thrombosis model in vivo. Decreased thrombus stability in Bambi-deficient mice after laser-induced thrombosis model in vivo. (A) Representative composite fluorescence and brightfield images of laser-induced thrombus formation in arterioles of Bambi+/+ and Bambi−/− mice. Scale bar represents 10 µm. (B) Median IFI is depicted (AU) as a function of time after the injury. Distribution of the time to maximal thrombus size (C) and maximal thrombus size expressed in IFI AU (D) in wild-type and Bambi-deficient mice. (E) Thrombus stability was assessed by determining the time taken for each thrombus at maximal size to fall by 50% of maximal IFI. See supplemental Videos 4-6 for better visualization of the differences in thrombus stability between the different animal groups. Each symbol represents 1 thrombus: Bambi+/+ (n = 27), Bambi+/− (n = 26), and Bambi−/− mice (n = 22). Horizontal lines intersecting data sets represent the median. Statistical analysis was performed using ANOVA (Kruskal-Wallis). *P < .05. AU, arbitrary unit. Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology

Bambi−/− mice display normal platelet function. Bambi−/− mice display normal platelet function. (A-D) Representative aggregation traces of washed platelets isolated from Bambi+/+ and Bambi−/− mice and stimulated with the indicated concentrations of agonists. Light transmission was recorded on a Chrono-log 2 channel aggregometer over 5 minutes. (E) Bar graphs of results obtained by aggregometry. Results are given as the mean percentage of aggregation ± SEM (n ≥ 3 mice for each condition). Statistical analysis was performed using the unpaired Student t test (P > .05). (F-G) Flow cytometry analysis of αIIbβ3 integrin activation (F) and P-selectin expression (G) in response to the indicated concentrations of agonists in platelets from Bambi+/+ (n = 8) and Bambi−/− mice (n = 11). Data represent mean fluorescence intensities ± SEM. Statistical analysis was performed using the unpaired Student t test (P > .05). (H) Whole blood from Bambi+/+ and Bambi−/− mice was perfused over a collagen-coated surface (0.1 mg/mL) at a shear rate of 1500 s−1. (Left) Representative fluorescence images of aggregate formation on collagen after 3 minutes of perfusion time. Scale bar equals 60 µm. (Right) Surface covered by fluorescently labeled platelets expressed as the percentage of the total surface observed was calculated at the indicated perfusion times. Data represent mean ± SEM (n > 6 for each group). Statistical analysis was performed using the unpaired Student t test (P > .05). Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology

Bambi−/− platelets express PS normally and retain their procoagulant ability. Bambi−/− platelets express PS normally and retain their procoagulant ability. Thrombin generation was measured in mouse citrate-anticoagulated plasma supplemented with 1pM TF, 16.6mM CaCl2, fluorogenic substrate Z-GGR-AMC, and 4µM phospholipids (A) or PRP (108/mL final concentration) (B) in the presence of corn trypsin inhibitor. Representative traces of thrombin generation in PPP (A) or PRP (B) from Bambi+/+ (n = 4) and Bambi−/− mice (n = 3). Thrombin generation was determined from the accumulation of fluorescent product and calculated relative to a thrombin calibrator. Each PPP and PRP sample was run in duplicate. For peak height values, see supplemental Table 3. (C) Exposure of PS was detected with Annexin V–FITC by flow cytometry using Bambi+/+ and Bambi−/− washed platelets stimulated with thrombin (1 U/mL), or a combination of thrombin and CRP (10 µg/mL) for 15 minutes. Data represent mean ± SEM (n = 3). Statistical analysis was performed using the unpaired Student t test (P > .05). (D) Levels of TAT complexes in the plasma of Bambi+/+ and Bambi−/− mice. Values are given as mean ± SEM of n = 5 animals for each group with technical replicates >2 for each animal. Statistical analysis was performed using the unpaired Student t test (P > .05). FITC, fluorescein isothiocyanate; PS, phosphatidylserine. Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology

Endothelial BAMBI is responsible for the defect in thrombus stability in vivo. Endothelial BAMBI is responsible for the defect in thrombus stability in vivo. (A) Representative composite fluorescence and brightfield images of laser-induced thrombus formation in arterioles of Bambi+/+ and Bambi−/− mice receiving BMCs from Bambi−/− and Bambi+/+ mice, respectively. Scale bar represents 10 µm. (B) IFI is depicted (AU) as a function of time after the injury. Distribution of the time to maximal thrombus size (C) and maximal thrombus size expressed in IFI AU (D) in Bambi+/+/Bambi−/− and Bambi−/−/Bambi+/+ mice. (E) Thrombus stability was assessed by determining the time for each thrombus after reaching maximal size to reduce by 50% of maximal IFI. See supplemental Videos 7-8 for better visualization of the differences in thrombus stability between the different animal groups. Each symbol represents 1 thrombus: Bambi+/+/Bambi−/− (n = 19) and Bambi−/−/Bambi+/+ mice (n = 16). Horizontal lines intersecting data sets represent the median. Statistical analysis was performed using the unpaired Mann-Whitney test. *P < .05. Isabelle I. Salles-Crawley et al. Blood 2014;123:2873-2881 ©2014 by American Society of Hematology