Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Blood ; 143(6): 548-560, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-37944157

ABSTRACT

ABSTRACT: Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbß3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.


Subject(s)
Induced Pluripotent Stem Cells , Thrombosis , Humans , Megakaryocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Blood Platelets/metabolism , Thrombosis/metabolism , Fibrin/metabolism , Plasma
2.
Blood ; 139(12): 1892-1902, 2022 03 24.
Article in English | MEDLINE | ID: mdl-34890454

ABSTRACT

Rebalancing the hemostatic system by targeting endogenous anticoagulant pathways, like the protein C (PC) system, is being tested as a means of improving hemostasis in patients with hemophilia. Recent intravital studies of hemostasis demonstrated that, in some vascular contexts, thrombin activity is sequestered in the extravascular compartment. These findings raise important questions about the context-dependent contribution of activated PC (APC) to the hemostatic response, because PC activation occurs on the surface of endothelial cells. We used a combination of pharmacologic, genetic, imaging, and computational approaches to examine the relationships among thrombin spatial distribution, PC activation, and APC anticoagulant function. We found that inhibition of APC activity, in mice either harboring the factor V Leiden mutation or infused with an APC-blocking antibody, significantly enhanced fibrin formation and platelet activation in a microvascular injury model, consistent with the role of APC as an anticoagulant. In contrast, inhibition of APC activity had no effect on hemostasis after penetrating injury of the mouse jugular vein. Computational studies showed that differences in blood velocity, injury size, and vessel geometry determine the localization of thrombin generation and, consequently, the extent of PC activation. Computational predictions were tested in vivo and showed that when thrombin generation occurred intravascularly, without penetration of the vessel wall, inhibition of APC significantly increased fibrin formation in the jugular vein. Together, these studies show the importance of thrombin spatial distribution in determining PC activation during hemostasis and thrombosis.


Subject(s)
Hemostatics , Thrombosis , Animals , Anticoagulants/pharmacology , Endothelial Cells/metabolism , Fibrin/metabolism , Hemostasis , Humans , Mice , Protein C/pharmacology , Thrombin/metabolism , Thrombosis/metabolism
3.
Proc Natl Acad Sci U S A ; 117(39): 24316-24325, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929010

ABSTRACT

Platelets are best known for their vasoprotective responses to injury and inflammation. Here, we have asked whether they also support vascular integrity when neither injury nor inflammation is present. Changes in vascular barrier function in dermal and meningeal vessels were measured in real time in mouse models using the differential extravasation of fluorescent tracers as a biomarker. Severe thrombocytopenia produced by two distinct methods caused increased extravasation of 40-kDa dextran from capillaries and postcapillary venules but had no effect on extravasation of 70-kDa dextran or albumin. This reduction in barrier function required more than 4 h to emerge after thrombocytopenia was established, reverting to normal as the platelet count recovered. Barrier dysfunction was also observed in mice that lacked platelet-dense granules, dense granule secretion machinery, glycoprotein (GP) VI, or the GPVI signaling effector phospholipase C (PLC) γ2. It did not occur in mice lacking α-granules, C type lectin receptor-2 (CLEC-2), or protease activated receptor 4 (PAR4). Notably, although both meningeal and dermal vessels were affected, intracerebral vessels, which are known for their tighter junctions between endothelial cells, were not. Collectively, these observations 1) highlight a role for platelets in maintaining vascular homeostasis in the absence of injury or inflammation, 2) provide a sensitive biomarker for detecting changes in platelet-dependent barrier function, 3) identify which platelet processes are required, and 4) suggest that the absence of competent platelets causes changes in the vessel wall itself, accounting for the time required for dysfunction to emerge.


Subject(s)
Blood Platelets/immunology , Blood Vessels/immunology , Hemostasis , Homeostasis , Animals , Blood Vessels/injuries , Blood Vessels/physiopathology , Female , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Male , Meninges/blood supply , Meninges/immunology , Mice , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Skin/blood supply , Skin/immunology
4.
Blood ; 136(15): 1773-1782, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32542378

ABSTRACT

G protein-coupled receptors are critical mediators of platelet activation whose signaling can be modulated by members of the regulator of G protein signaling (RGS) family. The 2 most abundant RGS proteins in human and mouse platelets are RGS10 and RGS18. While each has been studied individually, critical questions remain about the overall impact of this mode of regulation in platelets. Here, we report that mice missing both proteins show reduced platelet survival and a 40% decrease in platelet count that can be partially reversed with aspirin and a P2Y12 antagonist. Their platelets have increased basal (TREM)-like transcript-1 expression, a leftward shift in the dose/response for a thrombin receptor-activating peptide, an increased maximum response to adenosine 5'-diphosphate and TxA2, and a greatly exaggerated response to penetrating injuries in vivo. Neither of the individual knockouts displays this constellation of findings. RGS10-/- platelets have an enhanced response to agonists in vitro, but platelet count and survival are normal. RGS18-/- mice have a 15% reduction in platelet count that is not affected by antiplatelet agents, nearly normal responses to platelet agonists, and normal platelet survival. Megakaryocyte number and ploidy are normal in all 3 mouse lines, but platelet recovery from severe acute thrombocytopenia is slower in RGS18-/- and RGS10-/-18-/- mice. Collectively, these results show that RGS10 and RGS18 have complementary roles in platelets. Removing both at the same time discloses the extent to which this regulatory mechanism normally controls platelet reactivity in vivo, modulates the hemostatic response to injury, promotes platelet production, and prolongs platelet survival.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/genetics , RGS Proteins/genetics , Thrombopoiesis/genetics , Animals , Blood Platelets/drug effects , Cell Survival/genetics , Mice , Mice, Knockout , Phosphorylation , Platelet Activating Factor/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count , RGS Proteins/metabolism , Thrombopoiesis/drug effects
5.
Proc Natl Acad Sci U S A ; 116(6): 2243-2252, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674670

ABSTRACT

Extensive studies have detailed the molecular regulation of individual components of the hemostatic system, including platelets, coagulation factors, and regulatory proteins. Questions remain, however, about how these elements are integrated at the systems level within a rapidly changing physical environment. To answer some of these questions, we developed a puncture injury model in mouse jugular veins that combines high-resolution, multimodal imaging with functional readouts in vivo. The results reveal striking spatial regulation of platelet activation and fibrin formation that could not be inferred from studies performed ex vivo. As in the microcirculation, where previous studies have been performed, gradients of platelet activation are readily apparent, as is an asymmetrical distribution of fibrin deposition and thrombin activity. Both are oriented from the outer to the inner surface of the damaged vessel wall, with a greater extent of platelet activation and fibrin accumulation on the outside than the inside. Further, we show that the importance of P2Y12 signaling in establishing a competent hemostatic plug is related to the size of the injury, thus limiting its contribution to hemostasis to specific physiologic contexts. Taken together, these studies offer insights into the organization of hemostatic plugs, provide a detailed understanding of the adverse bleeding associated with a widely prescribed class of antiplatelet agents, and highlight differences between hemostasis and thrombosis that may suggest alternative therapeutic approaches.


Subject(s)
Blood Coagulation , Hemostasis , Wounds and Injuries/blood , Animals , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Disease Models, Animal , Fibrin/metabolism , Male , Mice , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thrombosis/metabolism , Thrombosis/pathology , Veins/injuries , Wounds and Injuries/etiology
6.
Small ; 17(15): e2004889, 2021 04.
Article in English | MEDLINE | ID: mdl-33150735

ABSTRACT

Hemostasis is an innate protective mechanism that plays a central role in maintaining the homeostasis of the vascular system during vascular injury. Studying this essential physiological process is often challenged by the difficulty of modeling and probing the complex dynamics of hemostatic responses in the native context of human blood vessels. To address this major challenge, this paper describes a microengineering approach for in vitro modeling of hemostasis. This microphysiological model replicates the living endothelium, multilayered microarchitecture, and procoagulant activity of human blood vessels, and is also equipped with a microneedle that is actuated with spatial precision to simulate penetrating vascular injuries. The system recapitulates key features of the hemostatic response to acute vascular injury as observed in vivo, including i) thrombin-driven accumulation of platelets and fibrin, ii) formation of a platelet- and fibrin-rich hemostatic plug that halts blood loss, and iii) matrix deformation driven by platelet contraction for wound closure. Moreover, the potential use of this model for drug testing applications is demonstrated by evaluating the effects of anticoagulants and antiplatelet agents that are in current clinical use. The vascular injury-on-a-chip may serve as an enabling platform for preclinical investigation of hematological disorders and emerging therapeutic approaches against them.


Subject(s)
Thrombosis , Vascular System Injuries , Fibrin , Hemostasis , Humans , Lab-On-A-Chip Devices
7.
Blood ; 127(12): 1598-605, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26738537

ABSTRACT

Previous studies have shown that hemostatic thrombi formed in response to penetrating injuries have a core of densely packed, fibrin-associated platelets overlaid by a shell of less-activated, loosely packed platelets. Here we asked, first, how the diverse elements of this structure combine to stem the loss of plasma-borne molecules and, second, whether antiplatelet agents and anticoagulants that perturb thrombus structure affect the re-establishment of a tight vascular seal. The studies combined high-resolution intravital microscopy with a photo-activatable fluorescent albumin marker to simultaneously track thrombus formation and protein transport following injuries to mouse cremaster muscle venules. The results show that protein loss persists after red cell loss has ceased. Blocking platelet deposition with an αIIbß3antagonist delays vessel sealing and increases extravascular protein accumulation, as does either inhibiting adenosine 5'-diphosphate (ADP) P2Y12receptors or reducing integrin-dependent signaling and retraction. In contrast, sealing was unaffected by introducing hirudin to block fibrin accumulation or a Gi2α gain-of-function mutation to expand the thrombus shell. Collectively, these observations describe a novel approach for studying vessel sealing after injury in real time in vivo and show that (1) the core/shell architecture previously observed in arterioles also occurs in venules, (2) plasma leakage persists well beyond red cell escape and mature thrombus formation, (3) the most critical events for limiting plasma extravasation are the stable accumulation of platelets, ADP-dependent signaling, and the emergence of a densely packed core, not the accumulation of fibrin, and (4) drugs that affect platelet accumulation and packing can delay vessel sealing, permitting protein escape to continue.


Subject(s)
Blood Proteins/metabolism , Hemostasis , Microvessels/injuries , Microvessels/pathology , Thrombosis/pathology , Adenosine Diphosphate/metabolism , Animals , Blood Proteins/analysis , Fibrin/analysis , Fibrin/metabolism , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Platelet Activation , Platelet Count , Thrombosis/blood , Thrombosis/metabolism
9.
Blood ; 126(24): 2611-20, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26407691

ABSTRACT

Most platelet agonists activate platelets by binding to G-protein-coupled receptors. We have shown previously that a critical node in the G-protein signaling network in platelets is formed by a scaffold protein, spinophilin (SPL), the tyrosine phosphatase, Src homology region 2 domain-containing phosphatase-1 (SHP-1), and the regulator of G-protein signaling family member, RGS18. Here, we asked whether SPL and other RGS18 binding proteins such as 14-3-3γ regulate platelet reactivity by sequestering RGS18 and, if so, how this is accomplished. The results show that, in resting platelets, free RGS18 levels are relatively low, increasing when platelets are activated by thrombin. Free RGS18 levels also rise when platelets are rendered resistant to activation by exposure to prostaglandin I2 (PGI2) or forskolin, both of which increase platelet cyclic adenosine monophosphate (cAMP) levels. However, the mechanism for raising free RGS18 is different in these 2 settings. Whereas thrombin activates SHP-1 and causes dephosphorylation of SPL tyrosine residues, PGI2 and forskolin cause phosphorylation of SPL Ser94 without reducing tyrosine phosphorylation. Substituting alanine for Ser94 blocks cAMP-induced dissociation of the SPL/RGS/SHP-1 complex. Replacing Ser94 with aspartate prevents formation of the complex and produces a loss-of-function phenotype when expressed in mouse platelets. Together with the defect in platelet function we previously observed in SPL(-/-) mice, these data show that (1) regulated sequestration and release of RGS18 by intracellular binding proteins provides a mechanism for coordinating activating and inhibitory signaling networks in platelets, and (2) differential phosphorylation of SPL tyrosine and serine residues provides a key to understanding both.


Subject(s)
Platelet Activation/physiology , RGS Proteins/physiology , Animals , Blood Platelets/drug effects , CHO Cells , Colforsin/pharmacology , Cricetinae , Cricetulus , Cyclic AMP/physiology , Epoprostenol/pharmacology , Fetal Tissue Transplantation , Liver/embryology , Liver Transplantation , Mice , Microfilament Proteins/deficiency , Microfilament Proteins/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , Phosphorylation , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Radiation Chimera , Receptors, Thrombin/agonists , Second Messenger Systems/physiology , Signal Transduction/physiology , Thrombin/pharmacology
10.
Blood ; 125(10): 1623-32, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25477496

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is characterized by oculocutaneous albinism, bleeding diathesis, and other variable symptoms. The bleeding diathesis has been attributed to δ storage pool deficiency, reflecting the malformation of platelet dense granules. Here, we analyzed agonist-stimulated secretion from other storage granules in platelets from mouse HPS models that lack adaptor protein (AP)-3 or biogenesis of lysosome-related organelles complex (BLOC)-3 or BLOC-1. We show that α granule secretion elicited by low agonist doses is impaired in all 3 HPS models. High agonist doses or supplemental adenosine 5'-diphosphate (ADP) restored normal α granule secretion, suggesting that the impairment is secondary to absent dense granule content release. Intravital microscopy following laser-induced vascular injury showed that defective hemostatic thrombus formation in HPS mice largely reflected reduced total platelet accumulation and affirmed a reduced area of α granule secretion. Agonist-induced lysosome secretion ex vivo was also impaired in all 3 HPS models but was incompletely rescued by high agonist doses or excess ADP. Our results imply that (1) AP-3, BLOC-1, and BLOC-3 facilitate protein sorting to lysosomes to support ultimate secretion; (2) impaired secretion of α granules in HPS, and to some degree of lysosomes, is secondary to impaired dense granule secretion; and (3) diminished α granule and lysosome secretion might contribute to pathology in HPS.


Subject(s)
Blood Platelets/physiology , Hermanski-Pudlak Syndrome/blood , Adaptor Protein Complex 3/deficiency , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/physiology , Adenosine Diphosphate/pharmacology , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Degranulation/physiology , Disease Models, Animal , Guanine Nucleotide Exchange Factors , Hermanski-Pudlak Syndrome/etiology , Hermanski-Pudlak Syndrome/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lectins/deficiency , Lectins/genetics , Lectins/physiology , Lysosomes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/blood , SNARE Proteins/blood , Secretory Vesicles/physiology , Thrombin/pharmacology , Thrombosis/blood , Thrombosis/etiology , Vesicular Transport Proteins/deficiency , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/physiology
11.
Blood ; 124(11): 1808-15, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-24951424

ABSTRACT

Hemostatic thrombi develop a characteristic architecture in which a core of highly activated platelets is covered by a shell of less-activated platelets. Here we have used a systems biology approach to examine the interrelationship of this architecture with transport rates and agonist distribution in the gaps between platelets. Studies were performed in mice using probes for platelet accumulation, packing density, and activation plus recently developed transport and thrombin activity probes. The results show that intrathrombus transport within the core is much slower than within the shell. The region of slowest transport coincides with the region of greatest packing density and thrombin activity, and appears prior to full platelet activation. Deleting the contact-dependent signaling molecule, Sema4D, delays platelet activation, but not the emergence of the low transport region. Collectively, these results suggest a timeline in which initial platelet accumulation and the narrowing gaps between platelets create a region of reduced transport that facilitates local thrombin accumulation and greater platelet activation, whereas faster transport rates within the shell help to limit thrombin accumulation and growth of the core. Thus, from a systems perspective, platelet accumulation produces an altered microenvironment that shapes thrombus architecture, which in turn affects agonist distribution and subsequent thrombus growth.


Subject(s)
Blood Coagulation , Models, Cardiovascular , Platelet Activation , Thrombin/metabolism , Animals , Humans , Mice , Protein Transport
12.
Blood ; 124(11): 1816-23, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-24951425

ABSTRACT

Hemostatic thrombi formed after a penetrating injury have a heterogeneous architecture in which a core of highly activated, densely packed platelets is covered by a shell of less-activated, loosely packed platelets. In the first manuscript in this series, we show that regional differences in intrathrombus protein transport rates emerge early in the hemostatic response and are preserved as the thrombus develops. Here, we use a theoretical approach to investigate this process and its impact on agonist distribution. The results suggest that hindered diffusion, rather than convection, is the dominant mechanism responsible for molecular movement within the thrombus. The analysis also suggests that the thrombus core, as compared with the shell, provides an environment for retaining soluble agonists such as thrombin, affecting the extent of platelet activation by establishing agonist-specific concentration gradients radiating from the site of injury. This analysis accounts for the observed weaker activation and relative instability of platelets in the shell and predicts that a failure to form a tightly packed thrombus core will limit thrombin accumulation, a prediction tested by analysis of data from mice with a defect in clot retraction.


Subject(s)
Blood Coagulation , Computer Simulation , Models, Cardiovascular , Platelet Activation , Thrombin/metabolism , Animals , Humans , Mice , Protein Transport
13.
Blood ; 124(11): 1824-31, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-24951426

ABSTRACT

Hemostatic thrombi formed after a penetrating injury have a distinctive structure in which a core of highly activated, closely packed platelets is covered by a shell of less-activated, loosely packed platelets. We have shown that differences in intrathrombus molecular transport emerge in parallel with regional differences in platelet packing density and predicted that these differences affect thrombus growth and stability. Here we test that prediction in a mouse vascular injury model. The studies use a novel method for measuring thrombus contraction in vivo and a previously characterized mouse line with a defect in integrin αIIbß3 outside-in signaling that affects clot retraction ex vivo. The results show that the mutant mice have a defect in thrombus consolidation following vascular injury, resulting in an increase in intrathrombus transport rates and, as predicted by computational modeling, a decrease in thrombin activity and platelet activation in the thrombus core. Collectively, these data (1) demonstrate that in addition to the activation state of individual platelets, the physical properties of the accumulated mass of adherent platelets is critical in determining intrathrombus agonist distribution and platelet activation and (2) define a novel role for integrin signaling in the regulation of intrathrombus transport rates and localization of thrombin activity.


Subject(s)
Blood Coagulation , Models, Cardiovascular , Platelet Activation , Thrombin/metabolism , Animals , Humans , Mice , Protein Transport
15.
Arterioscler Thromb Vasc Biol ; 35(3): 645-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614284

ABSTRACT

OBJECTIVE: Biological and physical factors interact to modulate blood response in a wounded vessel, resulting in a hemostatic clot or an occlusive thrombus. Flow and pressure differential (ΔP) across the wound from the lumen to the extravascular compartment may impact hemostasis and the observed core/shell architecture. We examined physical and biological factors responsible for regulating thrombin-mediated clot growth. APPROACH AND RESULTS: Using factor XIIa-inhibited human whole blood perfused in a microfluidic device over collagen/tissue factor at controlled wall shear rate and ΔP, we found thrombin to be highly localized in the P-selectin(+) core of hemostatic clots. Increasing ΔP from 9 to 29 mm Hg (wall shear rate=400 s(-1)) reduced P-selectin(+) core size and total clot size because of enhanced extravasation of thrombin. Blockade of fibrin polymerization with 5 mmol/L Gly-Pro-Arg-Pro dysregulated hemostasis by enhancing both P-selectin(+) core size and clot size at 400 s(-1) (20 mm Hg). For whole-blood flow (no Gly-Pro-Arg-Pro), the thickness of the P-selectin-negative shell was reduced under arterial conditions (2000 s(-1), 20 mm Hg). Consistent with the antithrombin-1 activity of fibrin implicated with Gly-Pro-Arg-Pro, anti-γ'-fibrinogen antibody enhanced core-localized thrombin, core size, and overall clot size, especially at venous (100 s(-1)) but not arterial wall shear rates (2000 s(-1)). Pathological shear (15 000 s(-1)) and Gly-Pro-Arg-Pro synergized to exacerbate clot growth. CONCLUSIONS: Hemostatic clotting was dependent on core-localized thrombin that (1) triggered platelet P-selectin display and (2) was highly regulated by fibrin and the transclot ΔP. Also, γ'-fibrinogen had a role in venous but not arterial conditions.


Subject(s)
Collagen Type I/blood , Fibrin/metabolism , Fibrinogens, Abnormal/metabolism , Hemostasis , Thrombin/metabolism , Thromboplastin/metabolism , Thrombosis/blood , Vascular System Injuries/blood , Animals , Arteries/metabolism , Arteries/pathology , Arteries/physiopathology , Blood Flow Velocity , Disease Models, Animal , Humans , Lab-On-A-Chip Devices , Male , Mechanotransduction, Cellular , Mice , P-Selectin/blood , Polymerization , Pressure , Regional Blood Flow , Stress, Mechanical , Thrombosis/pathology , Thrombosis/physiopathology , Time Factors , Vascular System Injuries/pathology , Vascular System Injuries/physiopathology , Veins/metabolism , Veins/pathology , Veins/physiopathology
16.
Blood ; 121(20): 4221-30, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23564909

ABSTRACT

Semaphorin 4D (Sema4D) is a transmembrane protein that supports contact-dependent amplification of platelet activation by collagen before being gradually cleaved by the metalloprotease ADAM17, as we have previously shown. Cleavage releases a soluble 120-kDa exodomain fragment for which receptors exist on platelets and endothelial cells. Here we have examined the mechanism that regulates Sema4D exodomain cleavage. The results show that the membrane-proximal cytoplasmic domain of Sema4D contains a binding site for calmodulin within the polybasic region Arg762-Lys779. Coprecipitation studies show that Sema4D and calmodulin are associated in resting platelets, forming a complex that dissociates upon platelet activation by the agonists that trigger Sema4D cleavage. Inhibiting calmodulin with W7 or introducing a membrane-permeable peptide corresponding to the calmodulin-binding site is sufficient to trigger the dissociation of Sema4D from calmodulin and initiate cleavage. Conversely, deletion of the calmodulin-binding site causes constitutive shedding of Sema4D. These results show that (1) Sema4D is a calmodulin-binding protein with a site of interaction in its membrane-proximal cytoplasmic domain, (2) platelet agonists cause dissociation of the calmodulin-Sema4D complex, and (3) dissociation of the complex is sufficient to trigger ADAM17-dependent cleavage of Sema4D, releasing a bioactive fragment.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Blood Platelets/metabolism , Calmodulin/metabolism , Protein Interaction Domains and Motifs/physiology , Semaphorins/chemistry , Semaphorins/metabolism , Amino Acid Sequence , Animals , Antigens, CD/genetics , Blood Platelets/drug effects , Blood Platelets/physiology , CHO Cells , Calmodulin/antagonists & inhibitors , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Humans , Molecular Sequence Data , Platelet Activation/drug effects , Platelet Activation/genetics , Protein Binding/drug effects , Protein Processing, Post-Translational/drug effects , Semaphorins/genetics , Sulfonamides/pharmacology
17.
Blood ; 121(10): 1875-85, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23303817

ABSTRACT

Achieving hemostasis following vascular injury requires the rapid accumulation of platelets and fibrin. Here we used a combination of confocal intravital imaging, genetically engineered mice, and antiplatelet agents to determine how variations in the extent of platelet activation following vascular injury arise from the integration of different elements of the platelet-signaling network. Two forms of penetrating injury were used to evoke the hemostatic response. Both produced a hierarchically organized structure in which a core of fully activated platelets was overlaid with an unstable shell of less-activated platelets. This structure emerged as hemostasis was achieved and persisted for at least 60 minutes following injury, its organization at least partly reflecting agonist concentration gradients. Thrombin activity and fibrin formation were found primarily in the innermost core. As proposed previously, greater packing density in the core facilitated contact-dependent signaling and limited entry of plasma-borne molecules visualized with fluorophores coupled to dextran and albumin. Blocking contact-dependent signaling or inhibiting thrombin reduced the size of the core, while the shell was heavily influenced by adenosine 5'-diphosphate and regulators of Gi2-mediated signaling. Thus, the hemostatic response is shown to produce a hierarchical structure arising, in part, from distinct elements of the platelet-signaling network.


Subject(s)
Blood Platelets/physiology , Hemostasis/physiology , Muscle, Skeletal/metabolism , Signal Transduction , Thrombin/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Animals , Antigens, CD/physiology , Blood Platelets/ultrastructure , Fibrin/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Hemostasis/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/metabolism , Semaphorins/physiology , Thrombin/antagonists & inhibitors
18.
Curr Opin Hematol ; 21(5): 410-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25023471

ABSTRACT

PURPOSE OF REVIEW: Several decades of work by many investigators have elucidated the major signaling pathways responsible for platelet activation. Still to be fully understood is how these pathways are integrated into a single network and how changing conditions within a growing thrombus affect that network. In this review we will consider some of the recent studies that address these issues and describe a model that provides insights into platelet activation as it occurs in vivo. RECENT FINDINGS: Genetic and pharmacologic studies performed in vivo have demonstrated that platelet activation during hemostasis and thrombosis is heterogeneous. Those studies indicate that distinct platelet activation pathways are not merely redundant, but are coordinated in time and space to achieve an optimal response. This coordination is achieved at least in part by the evolving distribution of platelet agonists and changes in solute transport within a hemostatic plug. SUMMARY: Studies examining the coordination of platelet signaling in time and space continue to increase our understanding of hemostasis and thrombosis. In addition to helping to decipher platelet biology, the results have implications for the understanding of new and existing antiplatelet agents and their potential risks.


Subject(s)
Blood Platelets/cytology , Vascular System Injuries/pathology , Animals , Blood Platelets/metabolism , Cell Shape , Humans , Platelet Activation , Signal Transduction , Vascular System Injuries/metabolism
19.
Blood ; 119(14): 3352-60, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22271446

ABSTRACT

Mounting evidence suggests that agonist-initiated signaling in platelets is closely regulated to avoid excessive responses to injury. A variety of physiologic agonists induce a cascade of signaling events termed as inside-out signaling that culminate in exposure of high-affinity binding sites on integrin α(IIb)ß(3). Once platelet activation has occurred, integrin α(IIb)ß(3) stabilizes thrombus formation by providing agonist-independent "outside-in" signals mediated in part by contractile signaling. Junctional adhesion molecule A (JAM-A), a member of the cortical thymocyte marker of the Xenopus (CTX) family, was initially identified as a receptor for a platelet stimulatory mAb. Here we show that JAM-A in resting platelets functions as an endogenous inhibitor of platelet function. Genetic ablation of Jam-A in mice enhances thrombotic function of platelets in vivo. The absence of Jam-A results in increase in platelet aggregation ex vivo. This gain of function is not because of enhanced inside-out signaling because granular secretion, Thromboxane A2 (TxA2) generation, as well as fibrinogen receptor activation, are normal in the absence of Jam-A. Interestingly, integrin outside-in signaling such as platelet spreading and clot retraction is augmented in Jam-A-deficient platelets. We conclude that JAM-A normally limits platelet accumulation by inhibiting integrin outside-in signaling thus preventing premature platelet activation.


Subject(s)
Blood Platelets/metabolism , Cell Adhesion Molecules/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Cell Surface/metabolism , Thrombosis/etiology , Animals , Bleeding Time , Cell Adhesion Molecules/genetics , Clot Retraction/genetics , Gene Knockout Techniques , Genetic Association Studies , Humans , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Adhesiveness/genetics , Pulmonary Embolism/genetics , Pulmonary Embolism/mortality , Pulmonary Embolism/pathology , Receptors, Cell Surface/genetics , Signal Transduction , Thrombosis/genetics , Thrombosis/prevention & control
20.
Blood ; 120(1): 190-8, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22517902

ABSTRACT

During thrombotic or hemostatic episodes, platelets bind collagen and release ADP and thromboxane A(2), recruiting additional platelets to a growing deposit that distorts the flow field. Prediction of clotting function under hemodynamic conditions for a patient's platelet phenotype remains a challenge. A platelet signaling phenotype was obtained for 3 healthy donors using pairwise agonist scanning, in which calcium dye-loaded platelets were exposed to pairwise combinations of ADP, U46619, and convulxin to activate the P2Y(1)/P2Y(12), TP, and GPVI receptors, respectively, with and without the prostacyclin receptor agonist iloprost. A neural network model was trained on each donor's pairwise agonist scanning experiment and then embedded into a multiscale Monte Carlo simulation of donor-specific platelet deposition under flow. The simulations were compared directly with microfluidic experiments of whole blood flowing over collagen at 200 and 1000/s wall shear rate. The simulations predicted the ranked order of drug sensitivity for indomethacin, aspirin, MRS-2179 (a P2Y(1) inhibitor), and iloprost. Consistent with measurement and simulation, one donor displayed larger clots and another presented with indomethacin resistance (revealing a novel heterozygote TP-V241G mutation). In silico representations of a subject's platelet phenotype allowed prediction of blood function under flow, essential for identifying patient-specific risks, drug responses, and novel genotypes.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/physiology , Microfluidic Analytical Techniques/methods , Models, Biological , Platelet Function Tests/methods , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Calcium/metabolism , Crotalid Venoms/pharmacology , HEK293 Cells , Humans , Lectins, C-Type , Male , Microfluidic Analytical Techniques/standards , Platelet Activating Factor/physiology , Platelet Function Tests/standards , Predictive Value of Tests , Receptors, Thromboxane/genetics , Receptors, Thromboxane/metabolism , Reference Values , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombosis/physiopathology , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL