ELMO1 deficiency enhances platelet function

Slides:



Advertisements
Similar presentations
Acalabrutinib and ibrutinib therapy cause dysfunctional GPVI-mediated platelet aggregation. Acalabrutinib and ibrutinib therapy cause dysfunctional GPVI-mediated.
Advertisements

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,
by Miguel Vicente-Manzanares, Aranzazu Cruz-Adalia, Noa B
Roles of RabGEF1/Rabex-5 domains in regulating FcϵRI surface expression and FcϵRI-dependent responses in mast cells by Janet Kalesnikoff, Eon J. Rios,
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,
Defective release of α granule and lysosome contents from platelets in mouse Hermansky-Pudlak syndrome models by Ronghua Meng, Jie Wu, Dawn C. Harper,
PRT , a novel Syk inhibitor, prevents heparin-induced thrombocytopenia and thrombosis in a transgenic mouse model by Michael P. Reilly, Uma Sinha,
Vessel wall BAMBI contributes to hemostasis and thrombus stability
by Ji-Long Chen, Andre Limnander, and Paul B. Rothman
S6K1 and mTOR regulate Rac1-driven platelet activation and aggregation
by Kesheng Dai, Richard Bodnar, Michael C. Berndt, and Xiaoping Du
by Hong Yin, Aleksandra Stojanovic, Nissim Hay, 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,
Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway by Kevin M. Elias, Arian Laurence,
by Nayna Mistry, Susan L. Cranmer, Yuping Yuan, Pierre Mangin, Sacha M
ADAP interactions with talin and kindlin promote platelet integrin αIIbβ3 activation and stable fibrinogen binding by Ana Kasirer-Friede, Jian Kang, Bryan.
Agonist-induced aggregation of Chinese hamster ovary cells coexpressing the human receptors for fibrinogen (integrin αIIbβ3) and the platelet-activating.
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.
Negative regulation of platelet function by a secreted cell repulsive protein, semaphorin 3A by Hirokazu Kashiwagi, Masamichi Shiraga, Hisashi Kato, Tsuyoshi.
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.
Identification of FcγRIIa as the ITAM-bearing receptor mediating αIIbβ3 outside-in integrin signaling in human platelets by Brian Boylan, Cunji Gao, Vipul.
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.
Inhibition of Platelet GPIbα and Promotion of Melanoma Metastasis
Rap1-GTP–interacting adaptor molecule (RIAM) is dispensable for platelet integrin activation and function in mice by Simon Stritt, Karen Wolf, Viola Lorenz,
by Bruno Bernardi, Gianni F. Guidetti, Francesca Campus, Jill R
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
IMiD compounds affect CD34+ cell fate and maturation via CRBN-induced IKZF1 degradation by Shirong Li, Jing Fu, Hui Wang, Huihui Ma, Xiaoming Xu, Yong-Guang.
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,
Phosphorylation of CLEC-2 is dependent on lipid rafts, actin polymerization, secondary mediators, and Rac by Alice Y. Pollitt, Beata Grygielska, Bertrand.
IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation by Melanie Antl, Marie-Luise von Brühl, Christina Eiglsperger,
Rapid ubiquitination of Syk following GPVI activation in platelets
Interaction of kindlin-3 and β2-integrins differentially regulates neutrophil recruitment and NET release in mice by Zhen Xu, Jiayi Cai, Juan Gao, Gilbert.
Diverging signaling events control the pathway of GPVI down-regulation in vivo by Tamer Rabie, David Varga-Szabo, Markus Bender, Rastislav Pozgaj, Francois.
RGS10 shapes the hemostatic response to injury through its differential effects on intracellular signaling by platelet agonists by Peisong Ma, Shuchi Gupta,
by Thomas D. Nightingale, Krupa Pattni, Alistair N. Hume, Miguel C
Dasatinib enhances megakaryocyte differentiation but inhibits platelet formation by Alexandra Mazharian, Cedric Ghevaert, Lin Zhang, Steffen Massberg,
In-depth PtdIns(3,4,5)P3 signalosome analysis identifies DAPP1 as a negative regulator of GPVI-driven platelet function by Tom N. Durrant, James L. Hutchinson,
Dichloroacetate, an inhibitor of pyruvate dehydrogenase kinases, inhibits platelet aggregation and arterial thrombosis by Manasa K. Nayak, Nirav Dhanesha,
Mature murine megakaryocytes present antigen-MHC class I molecules to T cells and transfer them to platelets by Anne Zufferey, Edwin R. Speck, Kellie R.
Platelet-specific deletion of SNAP23 ablates granule secretion, substantially inhibiting arterial and venous thrombosis in mice by Christopher M. Williams,
A function-blocking PAR4 antibody is markedly antithrombotic in the face of a hyperreactive PAR4 variant by Shauna L. French, Claudia Thalmann, Paul F.
Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling by Jeff S. Isenberg, Martin J.
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,
Volume 47, Issue 4, Pages e3 (October 2017)
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.
by Caroline Kardeby, Knut Fälker, Elizabeth J
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,
Secretion is ablated in the absence of SNAP23.
by Kelly E. Johnson, Julia R. Ceglowski, Harvey G. Roweth, Jodi A
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
Volume 23, Issue 2, Pages (August 2005)
Presentation transcript:

ELMO1 deficiency enhances platelet function by Akruti Patel, John Kostyak, Carol Dangelmaier, Rachit Badolia, Dheeraj Bhavanasi, Joseph E. Aslan, Salim Merali, Soochong Kim, Johannes A. Eble, Lawrence Goldfinger, and Satya Kunapuli BloodAdv Volume 3(4):575-587 February 26, 2019 © 2019 by The American Society of Hematology

Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 associates with PIP3 in platelets. ELMO1 associates with PIP3in platelets. (A) Resting human platelets (1 × 109 cells/mL) were lysed with NP-40 lysis buffer and incubated with PI(3,4,5)P3 PIP beads or control beads overnight at 4°C. Pull-downs were analyzed by western blot and probed with ELMO1 or Btk antibody. Blots are representative of at least 3 independent experiments. (B) The proteins from resting washed human, WT littermate control, and ELMO1−/− murine platelets were precipitated and analyzed by western blot. The blots were probed for ELMO1 and β-actin as a loading control. Blots are representative of at least 3 independent experiments. MW, molecular weight marker. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

Enhanced aggregation and granular secretion following GPVI stimulation in ELMO1−/− platelets. Enhanced aggregation and granular secretion following GPVI stimulation in ELMO1−/−platelets. Representative aggregation (A) and dense granule secretion tracings (B) of washed platelets from ELMO1−/− or WT littermate controls activated with the indicated agonists for 4 minutes. Platelet aggregation and dense granule secretion was detected by lumiaggregometry under stirring conditions at 37°C. Quantification of extent of aggregation (C) and dense granule secretion (D) of at least 3 independent experiments from panels A and B, respectively. Data are presented as mean ± SE and were analyzed by Student t test (*P < .05). Flow cytometry analysis of P-selectin exposure (E) and activated integrin αIIbβ3 (F) of washed murine platelets from ELMO1−/− or WT littermate controls activated with the indicated concentrations of CRP for 10 minutes at 37°C. Data are presented as fold increase over basal ± SE from at least 3 independent experiments and were analyzed by Student t test (*P < .05). A.U, arbitrary units. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

Enhanced thromboxane generation in response to GPVI agonists in ELMO1−/− platelets. Enhanced thromboxane generation in response to GPVI agonists in ELMO1−/−platelets. (A) Washed murine platelets were activated with the indicated concentrations of CRP or collagen for 4 minutes, and the reaction was stopped via flash-freezing. Thromboxane B2 generation was determined using an ELISA kit as per the manufacturer’s instructions. Data are presented as mean ± SE of at least 3 independent experiments and were analyzed by Student t test (*P < .05). (B) Representative aggregation tracings and dense granule secretion of washed platelets from ELMO1−/− or WT littermate controls preincubated with the feedback inhibitors (FBI’s) 10 μM indomethacin, 10 μM MRS-2179, and 100 nM AR-C69931MX for 5 minutes and activated with indicated concentration of CRP for 3 minutes. (C) Quantitation of aggregation and ATP secretion showing the mean ± standard error of the mean and analyzed by Student t test (*P < .05). Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 regulates integrin-mediated platelet spreading. ELMO1 regulates integrin-mediated platelet spreading. (A) Representative images of murine platelets incubated on fibrinogen-coated plates for the times indicated and fixed and stained with FITC-phalloidin. The images were obtained using confocal microscope. Scale bars, 5 μM. (B) Quantification of platelets with a filopodial phenotype expressed as a percentage of adhered platelets. At least 3 fields containing at least 25 platelets were analyzed per replicate (at least 2 replicates per experiment). Data were analyzed via Student t test (*P < .05). WT and ELMO1−/−, n = 4. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 regulates thrombus formation in vitro. ELMO1 regulates thrombus formation in vitro. Representative light microscopic image of thrombus formation on a collagen-coated surface (A) and percent thrombus area (B). Whole blood from WT littermate controls and ELMO1−/− mice was perfused over a collagen (50 μg/mL)–coated surface at arterial shear rate 1000 s−1 or venous shear rate 200 s−1 for 4 minutes. The direction of flow was right to left. (B) Images were analyzed using ImageJ, and data are presented as percent thrombus area ± SE. Data were analyzed by Student t test (*P < .05). WT and ELMO1−/−, n = 3. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 regulates thrombus formation and hemostasis in vivo. ELMO1 regulates thrombus formation and hemostasis in vivo. (A) Survival curves of WT and ELMO1−/− mice in a pulmonary thromboembolism model. Time to cessation of respiration was recorded after IV administration of 400 μg/kg collagen and 60 μg/kg epinephrine or phosphate-buffered saline. WT, n = 7; ELMO1−/−, n = 6. (B) Representative carotid artery blood flow tracings (i) and average time to occlusion (ii) from WT and ELMO1−/− mice. A carotid artery was isolated from the indicated mice and was exposed to 7.5% FeCl3 injury for 90 seconds. Data are presented as mean ± SE. WT, n = 12; ELMO1−/−, n = 14. Statistical analysis was performed using a Mann-Whitney U test (*P < .05). (C) Tail bleeding times of WT and ELMO1−/− mice. The distal 3 mm of the tail was cut, and the tail was immersed in 37°C saline. The time it took for bleeding to stop was recorded. WT, n = 38; ELMO1−/−, n = 18. Statistical analysis was performed using a Student t test (*P < .05). Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 regulates RhoG activity in the GPVI pathway. ELMO1 regulates RhoG activity in the GPVI pathway. (A) Rac1 activation in response to CRP in the absence of ELMO1. Washed platelets were stimulated with 1.5 μg/mL CRP for the indicated times and lysed with NP-40 lysis buffer, and active Rac was pulled down using GST-PAK-RBD. Active Rac1 was detected via western blot analysis using a specific antibody to Rac1. (B-C) Time course of ELMO1−/− and WT platelets activated with 1.25 μg/mL CRP at the indicated time points under stirring conditions at 37°C. Proteins were precipitated and analyzed by western blot using indicated antibodies. (D) Surface expression of GPVI in washed murine platelets from WT and ELMO1−/− mice assessed by flow cytometry. GPVI surface expression is a representative of at least 3 independent experiments. (E) RhoG activity in ELMO1−/− and WT murine platelets in response to CRP. Washed murine platelets were activated with 10 μg/mL CRP for 1 minute and lysed with NP-40 lysis buffer. Active RhoG was pulled down using GST-ELMO1 fusion protein, and RhoG activity was detected by western blot. (F) Model of GPVI signaling by ELMO1 in platelets. All western blot images are representative of at least 3 independent experiments. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology

ELMO1 deficiency is dispensable for CLEC-2–mediated platelet reactivity. ELMO1 deficiency is dispensable for CLEC-2–mediated platelet reactivity. Washed platelets isolated from WT, ELMO1−/−, and RhoG−/− mice were diluted to 1.5 × 108 and stimulated with either 5 nM (A) or 10 nM (C) rhodocytin, and subsequent aggregations and secretions were recorded. (B,D) Quantification of ATP secretion expressed as mean ± standard error of the mean following platelet stimulation with either 5 nM or 10 nM rhodocytin. Akruti Patel et al. Blood Adv 2019;3:575-587 © 2019 by The American Society of Hematology